1
|
Xue C, Ko HK, Shi K, Pittsenbarger J, Dao LV, Shi K, Libmann M, Geng H, Qian DZ. Understanding the development of enzalutamide resistance based on a functional single-cell approach. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.20.619319. [PMID: 39484437 PMCID: PMC11527018 DOI: 10.1101/2024.10.20.619319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Most metastatic prostate cancers (PCa) initially depend on androgen for survival and proliferation. Thus, anti-androgen or castration therapies are the mainstay treatment. Although effective at first, androgen-dependent PCa (ADPC) universally develops therapy resistance, thereby evolving to the incurable disease, called castration resistant PCa (CRPC). Currently, mechanisms underlying the emergence of CRPC from ADPC are largely unclear. We used single-cell RNA-sequencing (scRNA-Seq) to determine how a therapy-naïve ADPC cell line - LNCaP responds to the anti-androgen drug, enzalutamide. We found that most cells expressed the drug-target androgen receptor (AR+), while a small subpopulation (∼12%) expressed low or no AR (AR low/- ). Gene set enrichment analysis (GSEA) revealed that AR+ and AR low/- cells were enriched with significantly different gene expressions and signaling pathways. Unexpectedly, AR low/- cells displayed robust transcriptional response, including upregulations of genes and pathways involved in clinical CRPC. Next, we isolate AR low/- and AR+ cells from the LNCaP cell line, and functionally confirmed the enzalutamide resistant phenotype of AR low/- cells in vitro and in xenograft models in vivo. Finally, to explore a therapeutic option for AR low/- cells, we found that AR low/- cells expressed low levels of NAD+ biosynthesis genes, notably NAPRT, indicating a possible vulnerability to inhibitors blocking NAD+ synthesis. Indeed, treating AR low/- cells with NAD+ synthesis inhibitors, FK866 and OT-82, significantly inhibited the survival and proliferation of AR low/- cells, thus suggesting a possible novel therapeutic option for ADT and enzalutamide resistant PCa. SUMMARY Single-cell RNA-Sequencing reveals heterogeneities of tumor cell populations. In most cases, however, the functional significance of the observed heterogeneity is not tested. In this study, we first identified a possible therapy-resistant prostate cancer cell subpopulation with scRNA-Seq, then confirmed the resistant phenotype with single cell and colony - based cloning and functional testing. In addition, we also identified a therapeutic vulnerability of the resistant cells.
Collapse
|
2
|
Parolia A, Eyunni S, Verma BK, Young E, Liu Y, Liu L, George J, Aras S, Das CK, Mannan R, Ur Rasool R, Mitchell-Velasquez E, Mahapatra S, Luo J, Carson SE, Xiao L, Gajjala PR, Venkatesh S, Jaber M, Wang X, He T, Qiao Y, Pang M, Zhang Y, Tien JCY, Louw M, Alhusayan M, Cao X, Su F, Tavana O, Hou C, Wang Z, Ding K, Chinnaiyan AM, Asangani IA. NSD2 is a requisite subunit of the AR/FOXA1 neo-enhanceosome in promoting prostate tumorigenesis. Nat Genet 2024; 56:2132-2143. [PMID: 39251788 PMCID: PMC11525188 DOI: 10.1038/s41588-024-01893-6] [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: 10/31/2023] [Accepted: 08/01/2024] [Indexed: 09/11/2024]
Abstract
Androgen receptor (AR) is a ligand-responsive transcription factor that drives terminal differentiation of the prostatic luminal epithelia. By contrast, in tumors originating from these cells, AR chromatin occupancy is extensively reprogrammed to activate malignant phenotypes, the molecular mechanisms of which remain unknown. Here, we show that tumor-specific AR enhancers are critically reliant on H3K36 dimethyltransferase activity of NSD2. NSD2 expression is abnormally induced in prostate cancer, where its inactivation impairs AR transactivation potential by disrupting over 65% of its cistrome. NSD2-dependent AR sites distinctively harbor the chimeric FOXA1:AR half-motif, which exclusively comprise tumor-specific AR enhancer circuitries defined from patient specimens. NSD2 inactivation also engenders increased dependency on the NSD1 paralog, and a dual NSD1/2 PROTAC degrader is preferentially cytotoxic in AR-dependent prostate cancer models. Altogether, we characterize NSD2 as an essential AR neo-enhanceosome subunit that enables its oncogenic activity, and position NSD1/2 as viable co-targets in advanced prostate cancer.
Collapse
Affiliation(s)
- Abhijit Parolia
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Department of Urology, University of Michigan, Ann Arbor, MI, USA.
| | - Sanjana Eyunni
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Molecular and Cellular Pathology Program, University of Michigan, Ann Arbor, MI, USA
| | - Brijesh Kumar Verma
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eleanor Young
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yihan Liu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Cancer Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Lianchao Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - James George
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Shweta Aras
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chandan Kanta Das
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Reyaz Ur Rasool
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erick Mitchell-Velasquez
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Somnath Mahapatra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jie Luo
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Sandra E Carson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Lanbo Xiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Prathibha R Gajjala
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Sharan Venkatesh
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mustapha Jaber
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoju Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tongchen He
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew Pang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jean Ching-Yi Tien
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Micheala Louw
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mohammed Alhusayan
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Omid Tavana
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Caiyun Hou
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Department of Urology, University of Michigan, Ann Arbor, MI, USA.
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Irfan A Asangani
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Afshan S, Kim YG, Mattsson J, Åkerfelt M, Härkönen P, Baumgartner M, Nees M. Targeting the cancer cells and cancer-associated fibroblasts with next-generation FGFR inhibitors in prostate cancer co-culture models. Cancer Med 2024; 13:e70240. [PMID: 39300962 PMCID: PMC11413502 DOI: 10.1002/cam4.70240] [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: 06/06/2024] [Revised: 08/06/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Inhibition of androgen receptor (AR) signaling is the main treatment strategy in advanced prostate cancer (PCa). A subset of castration resistant prostate cancer (CRPC) bypasses the AR blockade by increased fibroblast growth factor receptor (FGFR) signaling. The first- and second-generation, non-covalent FGFR inhibitors (FGFRis) have largely failed in the clinical trials against PCa. PURPOSE In this study, we tested the drug sensitivity of LNCaP, VCaP, and CWR-R1PCa cell lines to second-generation, covalent FGFRis (FIIN1, FIIN2) and a novel FGFR downstream molecule inhibitor (FRS2αi). METHODS 2D and 3D mono- and co-cultures of cancer cells, and cancer-associated fibroblasts (CAFs) were used to mimic tumor-stroma interactions in the extracellular matrix (ECM). The treatment responses of the FGFR signaling molecules, the viability and proliferation of cancer cells, and CAFs were determined through immunoblotting, migration assay, cell viability assay, and real-time imaging. Immunofluorescent and confocal microscopy images of control and treated cultures of cancer cells and CAFs, and their morphometric data were deduced. RESULTS The FGFRis were more effective in mono-cultures of the cancer cells compared with co-cultures with CAFs. The FRS2αi was specifically effective in co-cultures with CAFs but was not cytotoxic to CAF mono-cultures as in the case of FIIN1 and FIIN2. At the molecular level, FRS2αi decreased p-FRS2α, p-ERK1/2, and activated apoptosis as monitored by cleaved caspase-3 activity in a concentration-dependent manner in the co-cultures. We observed no synergistic drug efficacy in the combination treatment of the FGFRi with ARi, enzalutamide, and darolutamide. The FRS2αi treatment led to a decrease in proliferation of cancer cell clusters in co-cultures as indicated by their reduced size and Ki67 expression. CONCLUSIONS CAFs exert a protective effect on cancer cells and should be included in the in vitro models to make them physiologically more relevant in screening and testing of FGFRis. The FRS2αi was the most potent agent in reducing the viability and proliferation of the 3D organotypic co-cultures, mainly by disrupting the contact between CAFs and cancer cell clusters. The next-generation FGFRi, FRS2αi, may be a better alternative treatment option for overcoming ARi treatment resistance in advanced PCa.
Collapse
Affiliation(s)
- Syeda Afshan
- FICAN West Cancer CentreInstitute of Biomedicine, University of TurkuTurkuFinland
| | - Yu Gang Kim
- FICAN West Cancer CentreInstitute of Biomedicine, University of TurkuTurkuFinland
- Present address:
Korea Mouse Phenotyping Center (KMPC)Seoul National UniversitySeoulSouth Korea
| | - Jesse Mattsson
- FICAN West Cancer CentreInstitute of Biomedicine, University of TurkuTurkuFinland
- Present address:
DelSiTech LtdTurkuFinland
| | - Malin Åkerfelt
- FICAN West Cancer CentreInstitute of Biomedicine, University of TurkuTurkuFinland
- Cell Biology, Faculty of Science and EngineeringÅbo Akademi UniversityTurkuFinland
| | - Pirkko Härkönen
- FICAN West Cancer CentreInstitute of Biomedicine, University of TurkuTurkuFinland
| | - Martin Baumgartner
- Pediatric Molecular Neuro‐Oncology Research LaboratoryUniversity Children's Hospital ZurichZurichSwitzerland
| | - Matthias Nees
- FICAN West Cancer CentreInstitute of Biomedicine, University of TurkuTurkuFinland
- Department of Biochemistry and Molecular BiologyMedical University of LublinLublinPoland
| |
Collapse
|
4
|
Hantusch B, Kenner L, Stanulović VS, Hoogenkamp M, Brown G. Targeting Androgen, Thyroid Hormone, and Vitamin A and D Receptors to Treat Prostate Cancer. Int J Mol Sci 2024; 25:9245. [PMID: 39273194 PMCID: PMC11394715 DOI: 10.3390/ijms25179245] [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: 07/11/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The nuclear hormone family of receptors regulates gene expression. The androgen receptor (AR), upon ligand binding and homodimerization, shuttles from the cytosol into the nucleus to activate gene expression. Thyroid hormone receptors (TRs), retinoic acid receptors (RARs), and the vitamin D receptor (VDR) are present in the nucleus bound to chromatin as a heterodimer with the retinoid X receptors (RXRs) and repress gene expression. Ligand binding leads to transcription activation. The hormonal ligands for these receptors play crucial roles to ensure the proper conduct of very many tissues and exert effects on prostate cancer (PCa) cells. Androgens support PCa proliferation and androgen deprivation alone or with chemotherapy is the standard therapy for PCa. RARγ activation and 3,5,3'-triiodo-L-thyronine (T3) stimulation of TRβ support the growth of PCa cells. Ligand stimulation of VDR drives growth arrest, differentiation, and apoptosis of PCa cells. Often these receptors are explored as separate avenues to find treatments for PCa and other cancers. However, there is accumulating evidence to support receptor interactions and crosstalk of regulatory events whereby a better understanding might lead to new combinatorial treatments.
Collapse
Affiliation(s)
- Brigitte Hantusch
- Department of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, 1010 Vienna, Austria;
- Comprehensive Cancer Center, Medical University Vienna, 1090 Vienna, Austria
| | - Lukas Kenner
- Department of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, 1010 Vienna, Austria;
- Comprehensive Cancer Center, Medical University Vienna, 1090 Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
- Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden
- Christian Doppler Laboratory for Applied Metabolomics, Medical University Vienna, 1090 Vienna, Austria
- Center for Biomarker Research in Medicine (CBmed), 8010 Graz, Austria
| | - Vesna S. Stanulović
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (V.S.S.); (M.H.)
| | - Maarten Hoogenkamp
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (V.S.S.); (M.H.)
| | - Geoffrey Brown
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
5
|
Zhong M, Xu W, Tian P, Zhang Q, Wang Z, Liang L, Zhang Q, Yang Y, Lu Y, Wei G. An Inherited Allele Confers Prostate Cancer Progression and Drug Resistance via RFX6/HOXA10-Orchestrated TGFβ Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401492. [PMID: 38932472 PMCID: PMC11348203 DOI: 10.1002/advs.202401492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/01/2024] [Indexed: 06/28/2024]
Abstract
Genetic and epigenetic alterations are cancer hallmark characteristics. However, the role of inherited cancer predisposition alleles in co-opting lineage factor epigenetic reprogramming and tumor progression remains elusive. Here the FinnGen cohort phenome-wide analysis, along with multiple genome-wide association studies, has consistently identified the rs339331-RFX6/6q22 locus associated with prostate cancer (PCa) risk across diverse populations. It is uncovered that rs339331 resides in a reprogrammed androgen receptor (AR) binding site in PCa tumors, with the T risk allele enhancing AR chromatin occupancy. RFX6, an AR-regulated gene linked to rs339331, exhibits synergistic prognostic value for PCa recurrence and metastasis. This comprehensive in vitro and in vivo studies demonstrate the oncogenic functions of RFX6 in promoting PCa cell proliferation and metastasis. Mechanistically, RFX6 upregulates HOXA10 that profoundly correlates with adverse PCa outcomes and is pivotal in RFX6-mediated PCa progression, facilitating the epithelial-mesenchymal transition (EMT) and modulating the TGFβ/SMAD signaling axis. Clinically, HOXA10 elevation is associated with increased EMT scores, tumor advancement and PCa recurrence. Remarkably, reducing RFX6 expression restores enzalutamide sensitivity in resistant PCa cells and tumors. This findings reveal a complex interplay of genetic and epigenetic mechanisms in PCa pathogenesis and drug resistance, centered around disrupted prostate lineage AR signaling and abnormal RFX6 expression.
Collapse
Affiliation(s)
- Mengjie Zhong
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Wenjie Xu
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Pan Tian
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Qin Zhang
- Disease Networks Research UnitFaculty of Biochemistry and Molecular MedicineBiocenter OuluUniversity of OuluOulu90220Finland
| | - Zixian Wang
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Limiao Liang
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Qixiang Zhang
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Yuehong Yang
- Disease Networks Research UnitFaculty of Biochemistry and Molecular MedicineBiocenter OuluUniversity of OuluOulu90220Finland
| | - Ying Lu
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Gong‐Hong Wei
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
- Disease Networks Research UnitFaculty of Biochemistry and Molecular MedicineBiocenter OuluUniversity of OuluOulu90220Finland
| |
Collapse
|
6
|
Karan D, Dubey S, Gunewardena S, Iczkowski KA, Singh M, Liu P, Poletti A, Choo Y, Chen H, Hamann MT. Manzamine A reduces androgen receptor transcription and synthesis by blocking E2F8-DNA interactions and effectively inhibits prostate tumor growth in mice. Mol Oncol 2024; 18:1966-1979. [PMID: 38605607 PMCID: PMC11306517 DOI: 10.1002/1878-0261.13637] [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: 11/03/2023] [Revised: 02/14/2024] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
The androgen receptor (AR) is the main driver in the development of castration-resistant prostate cancer, where the emergence of AR splice variants leads to treatment-resistant disease. Through detailed molecular studies of the marine alkaloid manzamine A (MA), we identified transcription factor E2F8 as a previously unknown regulator of AR transcription that prevents AR synthesis in prostate cancer cells. MA significantly inhibited the growth of various prostate cancer cell lines and was highly effective in inhibiting xenograft tumor growth in mice without any pathophysiological perturbations in major organs. MA suppressed the full-length AR (AR-FL), its spliced variant AR-V7, and the AR-regulated prostate-specific antigen (PSA; also known as KLK3) and human kallikrein 2 (hK2; also known as KLK2) genes. RNA sequencing (RNA-seq) analysis and protein modeling studies revealed E2F8 interactions with DNA as a potential novel target of MA, suppressing AR transcription and its synthesis. This novel mechanism of blocking AR biogenesis via E2F8 may provide an opportunity to control therapy-resistant prostate cancer over the currently used AR antagonists designed to target different parts of the AR gene.
Collapse
Affiliation(s)
- Dev Karan
- Department of Pathology, and MCW Cancer CenterMedical College of WisconsinMilwaukeeWIUSA
| | - Seema Dubey
- Department of Pathology, and MCW Cancer CenterMedical College of WisconsinMilwaukeeWIUSA
| | - Sumedha Gunewardena
- Department of Cell Biology and PhysiologyUniversity of Kansas Medical CenterKSUSA
| | - Kenneth A. Iczkowski
- Department of Pathology, and MCW Cancer CenterMedical College of WisconsinMilwaukeeWIUSA
| | - Manohar Singh
- Department of Pathology, and MCW Cancer CenterMedical College of WisconsinMilwaukeeWIUSA
| | - Pengyuan Liu
- Department of Physiology and Center of Systems Molecular MedicineMedical College of WisconsinMilwaukeeWIUSA
| | - Angelo Poletti
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanItaly
| | - Yeun‐Mun Choo
- Department of ChemistryUniversity of MalayaKuala LumpurMalaysia
| | - Hui‐Zi Chen
- Department of MedicineMedical College of WisconsinMilwaukeeWIUSA
| | - Mark T. Hamann
- Department of Drug Discovery and Biomedical Sciences and Public Health, Colleges of Pharmacy and Medicine, Hollings Cancer CenterMedical University of South CarolinaCharlestonSCUSA
| |
Collapse
|
7
|
Abdelaal AM, Sohal IS, Iyer SG, Sudarshan K, Orellana EA, Ozcan KE, dos Santos AP, Low PS, Kasinski AL. Selective targeting of chemically modified miR-34a to prostate cancer using a small molecule ligand and an endosomal escape agent. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102193. [PMID: 38745855 PMCID: PMC11091501 DOI: 10.1016/j.omtn.2024.102193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 04/18/2024] [Indexed: 05/16/2024]
Abstract
Use of tumor-suppressive microRNAs (miRNAs) as anti-cancer agents is hindered by the lack of effective delivery vehicles, entrapment of the miRNA within endocytic compartments, and rapid degradation of miRNA by nucleases. To address these issues, we developed a miRNA delivery strategy that includes (1) a targeting ligand, (2) an endosomal escape agent, nigericin and (3) a chemically modified miRNA. The delivery ligand, DUPA (2-[3-(1,3-dicarboxy propyl) ureido] pentanedioic acid), was selected based on its specificity for prostate-specific membrane antigen (PSMA), a receptor routinely upregulated in prostate cancer-one of the leading causes of cancer death among men. DUPA was conjugated to the tumor suppressive miRNA, miR-34a (DUPA-miR-34a) based on the ability of miR-34a to inhibit prostate cancer cell proliferation. To mediate endosomal escape, nigericin was incorporated into the complex, resulting in DUPA-nigericin-miR-34a. Both DUPA-miR-34a and DUPA-nigericin-miR-34a specifically bound to, and were taken up by, PSMA-expressing cells in vitro and in vivo. And while both DUPA-miR-34a and DUPA-nigericin-miR-34a downregulated miR-34a target genes, only DUPA-nigericin-miR-34a decreased cell proliferation in vitro and delayed tumor growth in vivo. Tumor growth was further reduced using a fully modified version of miR-34a that has significantly increased stability.
Collapse
Affiliation(s)
- Ahmed M. Abdelaal
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Ikjot S. Sohal
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Shreyas G. Iyer
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | | - Esteban A. Orellana
- Department of Molecular and Systems Biology, Dartmouth Geisel School of Medicine, Hanover, NH 03755, USA
| | - Kenan E. Ozcan
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Andrea P. dos Santos
- Department of Comparative Pathology, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Philip S. Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Andrea L. Kasinski
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
8
|
Chen H, Zhou Y, Wang X, Chai X, Wang Z, Wang E, Xu L, Hou T, Li D, Duan M. Discovery of Novel Anti-Resistance AR Antagonists Guided by Funnel Metadynamics Simulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309261. [PMID: 38481034 PMCID: PMC11109662 DOI: 10.1002/advs.202309261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/18/2024] [Indexed: 05/23/2024]
Abstract
Androgen receptor (AR) antagonists are widely used for the treatment of prostate cancer (PCa), but their therapeutic efficacy is usually compromised by the rapid emergence of drug resistance. However, the lack of the detailed interaction between AR and its antagonists poses a major obstacle to the design of novel AR antagonists. Here, funnel metadynamics is employed to elucidate the inherent regulation mechanisms of three AR antagonists (hydroxyflutamide, enzalutamide, and darolutamide) on AR. For the first time it is observed that the binding of antagonists significantly disturbed the C-terminus of AR helix-11, thereby disrupting the specific internal hydrophobic contacts of AR-LBD and correspondingly the communication between AR ligand binding pocket (AR-LBP), activation function 2 (AF2), and binding function 3 (BF3). The subsequent bioassays verified the necessity of the hydrophobic contacts for AR function. Furthermore, it is found that darolutamide, a newly approved AR antagonist capable of fighting almost all reported drug resistant AR mutants, can induce antagonistic binding structure. Subsequently, docking-based virtual screening toward the dominant binding conformation of AR for darolutamide is conducted, and three novel AR antagonists with favorable binding affinity and strong capability to combat drug resistance are identified by in vitro bioassays. This work provides a novel rational strategy for the development of anti-resistant AR antagonists.
Collapse
Affiliation(s)
- Haiyi Chen
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
- National Centre for Magnetic Resonance in WuhanState Key Laboratory of Magnetic Resonance and Atomic and Molecular PhysicsInnovation Academy for Precision Measurement Science and TechnologyChinese Academy of SciencesWuhanHubei430071China
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouZhejiang311121China
| | - Yuxin Zhou
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Xinyue Wang
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Xin Chai
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouZhejiang311121China
| | - Zhe Wang
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | | | - Lei Xu
- Institute of Bioinformatics and Medical EngineeringSchool of Electrical and Information EngineeringJiangsu University of TechnologyChangzhou213001China
| | - Tingjun Hou
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Dan Li
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Mojie Duan
- National Centre for Magnetic Resonance in WuhanState Key Laboratory of Magnetic Resonance and Atomic and Molecular PhysicsInnovation Academy for Precision Measurement Science and TechnologyChinese Academy of SciencesWuhanHubei430071China
- NMR and Molecular Sciences, School of Chemistry and Chemical Engineering, The State Key Laboratory of Refractories and MetallurgyWuhan University of Science and TechnologyWuhan430081China
| |
Collapse
|
9
|
Smith SF, Brewer DS, Hurst R, Cooper CS. Applications of Urinary Extracellular Vesicles in the Diagnosis and Active Surveillance of Prostate Cancer. Cancers (Basel) 2024; 16:1717. [PMID: 38730670 PMCID: PMC11083542 DOI: 10.3390/cancers16091717] [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: 04/14/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Prostate cancer is the most common non-cutaneous cancer among men in the UK, causing significant health and economic burdens. Diagnosis and risk prognostication can be challenging due to the genetic and clinical heterogeneity of prostate cancer as well as uncertainties in our knowledge of the underlying biology and natural history of disease development. Urinary extracellular vesicles (EVs) are microscopic, lipid bilayer defined particles released by cells that carry a variety of molecular cargoes including nucleic acids, proteins and other molecules. Urine is a plentiful source of prostate-derived EVs. In this narrative review, we summarise the evidence on the function of urinary EVs and their applications in the evolving field of prostate cancer diagnostics and active surveillance. EVs are implicated in the development of all hallmarks of prostate cancer, and this knowledge has been applied to the development of multiple diagnostic tests, which are largely based on RNA and miRNA. Common gene probes included in multi-probe tests include PCA3 and ERG, and the miRNAs miR-21 and miR-141. The next decade will likely bring further improvements in the diagnostic accuracy of biomarkers as well as insights into molecular biological mechanisms of action that can be translated into opportunities in precision uro-oncology.
Collapse
Affiliation(s)
- Stephanie F. Smith
- Metabolic Health Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK (C.S.C.)
- Department of Urology, Norfolk and Norwich University Hospitals, Norwich NR4 7UY, UK
| | - Daniel S. Brewer
- Metabolic Health Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK (C.S.C.)
| | - Rachel Hurst
- Metabolic Health Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK (C.S.C.)
| | - Colin S. Cooper
- Metabolic Health Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK (C.S.C.)
| |
Collapse
|
10
|
Parolia A, Eyunni S, Verma BK, Young E, Liu L, George J, Aras S, Das CK, Mannan R, Rasool RU, Luo J, Carson SE, Mitchell-Velasquez E, Liu Y, Xiao L, Gajjala PR, Jaber M, Wang X, He T, Qiao Y, Pang M, Zhang Y, Alhusayan M, Cao X, Tavana O, Hou C, Wang Z, Ding K, Chinnaiyan AM, Asangani IA. NSD2 is a requisite subunit of the AR/FOXA1 neo-enhanceosome in promoting prostate tumorigenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.22.581560. [PMID: 38464251 PMCID: PMC10925163 DOI: 10.1101/2024.02.22.581560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The androgen receptor (AR) is a ligand-responsive transcription factor that binds at enhancers to drive terminal differentiation of the prostatic luminal epithelia. By contrast, in tumors originating from these cells, AR chromatin occupancy is extensively reprogrammed to drive hyper-proliferative, metastatic, or therapy-resistant phenotypes, the molecular mechanisms of which remain poorly understood. Here, we show that the tumor-specific enhancer circuitry of AR is critically reliant on the activity of Nuclear Receptor Binding SET Domain Protein 2 (NSD2), a histone 3 lysine 36 di-methyltransferase. NSD2 expression is abnormally gained in prostate cancer cells and its functional inhibition impairs AR trans-activation potential through partial off-loading from over 40,000 genomic sites, which is greater than 65% of the AR tumor cistrome. The NSD2-dependent AR sites distinctly harbor a chimeric AR-half motif juxtaposed to a FOXA1 element. Similar chimeric motifs of AR are absent at the NSD2-independent AR enhancers and instead contain the canonical palindromic motifs. Meta-analyses of AR cistromes from patient tumors uncovered chimeric AR motifs to exclusively participate in tumor-specific enhancer circuitries, with a minimal role in the physiological activity of AR. Accordingly, NSD2 inactivation attenuated hallmark cancer phenotypes that were fully reinstated upon exogenous NSD2 re-expression. Inactivation of NSD2 also engendered increased dependency on its paralog NSD1, which independently maintained AR and MYC hyper-transcriptional programs in cancer cells. Concordantly, a dual NSD1/2 PROTAC degrader, called LLC0150, was preferentially cytotoxic in AR-dependent prostate cancer as well as NSD2-altered hematologic malignancies. Altogether, we identify NSD2 as a novel subunit of the AR neo-enhanceosome that wires prostate cancer gene expression programs, positioning NSD1/2 as viable paralog co-targets in advanced prostate cancer.
Collapse
Affiliation(s)
- Abhijit Parolia
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Sanjana Eyunni
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Molecular and Cellular Pathology Program, University of Michigan, Ann Arbor, MI, USA
- These authors contributed equally
| | - Brijesh Kumar Verma
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- These authors contributed equally
| | - Eleanor Young
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Lianchao Liu
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - James George
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Shweta Aras
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chandan Kanta Das
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Reyaz ur Rasool
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jie Luo
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Sandra E. Carson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Erick Mitchell-Velasquez
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yihan Liu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Cancer Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Lanbo Xiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Prathibha R. Gajjala
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mustapha Jaber
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoju Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tongchen He
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew Pang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mohammed Alhusayan
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Omid Tavana
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | - Caiyun Hou
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zhen Wang
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ke Ding
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Irfan A. Asangani
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Zhou C, Zhang X, Ma H, Zhou Y, Meng Y, Chen C, Shi G, Yu W, Zhang J. USP54 is a potential therapeutic target in castration-resistant prostate cancer. BMC Urol 2024; 24:32. [PMID: 38321455 PMCID: PMC10845770 DOI: 10.1186/s12894-024-01418-7] [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: 11/20/2023] [Accepted: 01/25/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND USP54, a ubiquitin-specific protease in the deubiquitinase (DUB) family, facilitates the malignant progression of several types of cancer. However, the role of USP54 in prostate cancer (PCa), especially castration-resistant prostate cancer (CRPC), remains unknown. METHODS We established the CRPC LNCaP-AI cell line from the hormone-sensitive prostate cancer (HSPC) LNCaP cell line. RNA-Seq was utilized to explore DUB expression levels in LNCaP and LNCaP-AI. USP54 was knocked down, and its effects on cell growth were evaluated in vitro and in vivo. Bioinformatics analyses were conducted to explore signaling pathways affected by USP54 in PCa. Quantitative polymerase chain reaction was used to confirm key signaling pathways involved. RESULTS USP54 was the most strongly upregulated DUB in LNCaP-AI cells compared with LNCaP cells. USP54 levels were higher in PCa than in normal tissues. USP54 silencing suppressed the proliferation of PCa cell lines, both in vitro and in vivo. USP54 expression was positively correlated with the androgen receptor (AR) signaling level in PCa samples, and USP54 knockdown inhibited AR signaling in PCa cells. CONCLUSIONS USP54 was upregulated during HSPC progression to CRPC. USP54 depletion suppressed CRPC cell proliferation both in vitro and in vivo. USP54 may facilitate PCa progression by regulating AR signaling.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Xuehu Zhang
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Hangbin Ma
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Yinghao Zhou
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Yibo Meng
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Chenchen Chen
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Guowei Shi
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Wandong Yu
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, P.R. China.
| | - Jun Zhang
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, P.R. China.
| |
Collapse
|
12
|
Chen Y, Han L, Dufour CR, Alfonso A, Giguère V. Canonical and Nuclear mTOR Specify Distinct Transcriptional Programs in Androgen-Dependent Prostate Cancer Cells. Mol Cancer Res 2024; 22:113-124. [PMID: 37889103 DOI: 10.1158/1541-7786.mcr-23-0087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 09/21/2023] [Accepted: 10/24/2023] [Indexed: 10/28/2023]
Abstract
mTOR is a serine/threonine kinase that controls prostate cancer cell growth in part by regulating gene programs associated with metabolic and cell proliferation pathways. mTOR-mediated control of gene expression can be achieved via phosphorylation of transcription factors, leading to changes in their cellular localization and activities. mTOR also directly associates with chromatin in complex with transcriptional regulators, including the androgen receptor (AR). Nuclear mTOR (nmTOR) has been previously shown to act as a transcriptional integrator of the androgen signaling pathway in association with the chromatin remodeling machinery, AR, and FOXA1. However, the contribution of cytoplasmic mTOR (cmTOR) and nmTOR and the role played by FOXA1 in this process remains to be explored. Herein, we engineered cells expressing mTOR tagged with nuclear localization and export signals dictating mTOR localization. Transcriptome profiling in AR-positive prostate cancer cells revealed that nmTOR generally downregulates a subset of the androgen response pathway independently of its kinase activity, while cmTOR upregulates a cell cycle-related gene signature in a kinase-dependent manner. Biochemical and genome-wide transcriptomic analyses demonstrate that nmTOR functionally interacts with AR and FOXA1. Ablation of FOXA1 reprograms the nmTOR cistrome and transcriptome of androgen responsive prostate cancer cells. This works highlights a transcriptional regulatory pathway in which direct interactions between nmTOR, AR and FOXA1 dictate a combinatorial role for these factors in the control of specific gene programs in prostate cancer cells. IMPLICATIONS The finding that canonical and nuclear mTOR signaling pathways control distinct gene programs opens therapeutic opportunities to modulate mTOR activity in prostate cancer cells.
Collapse
Affiliation(s)
- Yonghong Chen
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Lingwei Han
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | | | - Anthony Alfonso
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Vincent Giguère
- Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| |
Collapse
|
13
|
Huang JL, Yan XL, Huang D, Gan L, Gao H, Fan RZ, Li S, Yuan FY, Zhu X, Tang GH, Chen HW, Wang J, Yin S. Discovery of a highly potent and orally available importin- β1 inhibitor that overcomes enzalutamide-resistance in advanced prostate cancer. Acta Pharm Sin B 2023; 13:4934-4944. [PMID: 38045040 PMCID: PMC10692375 DOI: 10.1016/j.apsb.2023.07.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/27/2023] [Accepted: 07/11/2023] [Indexed: 12/05/2023] Open
Abstract
Nuclear transporter importin-β1 is emerging as an attractive target by virtue of its prevalence in many cancers. However, the lack of druggable inhibitors restricts its therapeutic proof of concept. In the present work, we optimized a natural importin-β1 inhibitor DD1 to afford an improved analog DD1-Br with better tolerability (>25 folds) and oral bioavailability. DD1-Br inhibited the survival of castration-resistant prostate cancer (CRPC) cells with sub-nanomolar potency and completely prevented tumor growth in resistant CRPC models both in monotherapy (0.5 mg/kg) and in enzalutamide-combination therapy. Mechanistic study revealed that by targeting importin-β1, DD1-Br markedly inhibited the nuclear accumulation of multiple CRPC drivers, particularly AR-V7, a main contributor to enzalutamide resistance, leading to the integral suppression of downstream oncogenic signaling. This study provides a promising lead for CRPC and demonstrates the potential of overcoming drug resistance in advanced CRPC via targeting importin-β1.
Collapse
Affiliation(s)
- Jia-Luo Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xue-Long Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou 550025, China
| | - Dong Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Lu Gan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Huahua Gao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Run-Zhu Fan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shen Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fang-Yu Yuan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xinying Zhu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Gui-Hua Tang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hong-Wu Chen
- School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Junjian Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Sheng Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
14
|
Jiang H, Li G. Transcription factors direct epigenetic reprogramming at specific loci in human cancers. Front Genet 2023; 14:1234515. [PMID: 37876590 PMCID: PMC10591108 DOI: 10.3389/fgene.2023.1234515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
The characterization of epigenetic changes during cancer development and progression led to notable insights regarding the roles of cancer-specific epigenetic reprogramming. Recent studies showed that transcription factors (TFs) are capable to regulate epigenetic reprogramming at specific loci in different cancer types through their DNA-binding activities. However, the causal association of dynamic histone modification change mediated by TFs is still not well elucidated. Here we evaluated the impacts of 636 transcription factor binding activities on histone modification in 24 cancer types. We performed Instrumental Variables analysis by using genetic lesions of TFs as our instrumental proxies, which previously discovered to be associated with histone mark activities. As a result, we showed a total of 6 EpiTFs as strong directors of epigenetic reprogramming of histone modification in cancers, which alters the molecular and clinical phenotypes of cancer. Together our findings highlight a causal mechanism driven by the TFs and genome-wide histone modification, which is relevant to multiple status of oncogenesis.
Collapse
Affiliation(s)
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Lam W, Arammash M, Cai W, Guan F, Jiang Z, Liu SH, Cheng P, Cheng YC. YIV-818-A: a novel therapeutic agent in prostate cancer management through androgen receptor downregulation, glucocorticoid receptor inhibition, epigenetic regulation, and enhancement of apalutamide, darolutamide, and enzalutamide efficacy. Front Pharmacol 2023; 14:1244655. [PMID: 37860121 PMCID: PMC10582333 DOI: 10.3389/fphar.2023.1244655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023] Open
Abstract
Introduction: Prostate cancer is the second leading cause of cancer death among men in the United States. Castration-Resistant Prostate Cancer (CRPC) often develops resistance to androgen deprivation therapy. Resistance in CRPC is often driven by AR variants and glucocorticoid receptor (GR). Thus, drugs that target both could be vital in overcoming resistance. Methods: Utilizing the STAR Drug Discovery Platform, three hundred medicinal plant extracts were examined across 25 signaling pathways to identify potential drug candidates. Effects of the botanical drug YIV-818-A, derived from optimized water extracts of Rubia cordifolia (R.C.), on Dihydrotestosterone (DHT) or Dexamethasone (DEX) induced luciferase activity were assessed in 22RV1 cells harboring the ARE luciferase reporter. Furthermore, the key active compounds in YIV-818-A were identified through activity guided purification. The inhibitory effects of YIV-818-A, RA-V, and RA-VII on AR and GR activities, their impact on AR target genes, and their roles in modifying epigenetic status were investigated. Finally, the synergistic effects of these compounds with established CRPC drugs were evaluated both in vitro and in vivo. Results: YIV-818-A was found to effectively inhibit DHT or DEX induced luciferase activity in 22RV1 cells. Deoxybouvardin (RA-V) was identified as the key active compound responsible for inhibiting AR and GR activities. Both YIV-818-A and RA-V, along with RA-VII, effectively downregulated AR and AR-V proteins through inhibiting protein synthesis, impacted the expression of AR target genes, and modified the epigenetic status by reducing levels of Bromodomain and Extra-Terminal proteins (Brd2/Brd4) and H3K27Ac. Furthermore, these compounds exhibited synergistic effects with apalutamide, darolutamide, or enzalutamide, and suppressed AR mediated luciferase activity of 22RV1 cells. Co-administration of YIV-818-A and enzalutamide led to a significant reduction of 22RV1 tumor growth in vivo. Different sources of R.C. had variable levels of RA-V, correlating with their potency in AR inhibition. Discussion: YIV-818-A, RA-V, and RA-VII show considerable promise in addressing drug resistance in CRPC by targeting both AR protein and GR function, along with modulation of vital epigenetic markers. Given the established safety profile of YIV-818-A, these findings suggest its potential as a chemopreventive agent and a robust anti-prostate cancer drug.
Collapse
Affiliation(s)
- Wing Lam
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
| | - Mohammad Arammash
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
| | - Wei Cai
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Fulan Guan
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
| | - Zaoli Jiang
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
| | | | | | - Yung-Chi Cheng
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
16
|
Bhat TA, Dheeraj A, Nambiar DK, Singh SP, Yim DS, Singh RP. Decursin inhibits EGFR-ERK1/2 signaling axis in advanced human prostate carcinoma cells. Prostate 2023; 83:534-546. [PMID: 36597263 DOI: 10.1002/pros.24482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/23/2020] [Indexed: 01/05/2023]
Abstract
We have shown that decursin, a coumarin compound, induces cell cycle arrest and apoptosis in human prostate cancer cells (PCa); however, its molecular mechanisms are largely unexplored. We studied the mechanisms associated with its anticancer activity in advanced human prostate carcinoma cells. We found that decursin inhibited epidermal growth factor receptor (EGFR) signaling by inhibiting its activating phosphorylation at tyrosine 1068 residue in DU145 and 22Rv1 cells. This inhibition of EGFR was associated with the downregulation of ERK1/2 phosphorylation. Both EGFR and ERK1/2 are known to be deregulated/activated in many human malignancies. Consistent with our earlier study, decursin (25-100 µM) treatment for 24-72 h inhibited DU145 cell proliferation by 49%-87% (p < 0.001) which was associated with strong G1 phase arrest and cell death. It also decreased (p < 0.001) the number of surviving colonies. Decursin moderately increased the expression of Rb-related proteins p107 and p130 but decreased the levels of E2F family transcription factors including E2F-3, E2F-4 and E2F-5. Further, decursin strongly inhibited the growth of androgen-dependent prostate carcinoma 22Rv1 cells from 61% to 79% (p < 0.001) and arrested these cells at G1 phase via induction of cyclin-dependent kinase inhibitor p27/Kip1 and downregulation of CDK2 and CDK4 protein expression. Additionally, EGFR inhibitor erlotinib- and EGF ligand-modulated EGFR activation validated EGFR signaling as a target of decursin-mediated cell growth inhibition and cytotoxicity. Decursin decreased EGF ligand-induced phosphorylation of EGFR (Y-1068) as well as activation of its downstream mediator, ERK1/2. Furthermore, inhibitory targeting of EGFR-ERK1/2 axis by combinatorial treatment of decursin and erlotinib further sensitized DU145 cells for the decursin-induced growth inhibition and cell death. Overall, these findings strongly suggest that anticancer efficacy of decursin against human PCa involves inhibitory targeting of EGFR-ERK1/2 signaling axis, a pathway constitutively active in advanced PCa.
Collapse
Affiliation(s)
- Tariq A Bhat
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Arpit Dheeraj
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Dhanya K Nambiar
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Surya Pratap Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Dong Sool Yim
- Department of Pharmacy, Sahmyook University, Seoul, Korea
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
17
|
Zhang S, Li H, Shen C, Cao F, Kang S. HP1α promotes the progression of prostate cancer. Mol Biol Rep 2023; 50:4459-4468. [PMID: 37014569 DOI: 10.1007/s11033-023-08373-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 03/02/2023] [Indexed: 04/05/2023]
Abstract
PURPOSE Patients who have been diagnosed with prostate cancer (PCa) typically have a dismal outlook and few therapeutic choices available to them, because the precise pathogenesis of the disease is not yet fully understood. The presence of HP1α, also known as the heterochromatin protein 1α, is required for the creation of higher-order chromatin structures. However, little is known about HP1α that serves roles in the pathogenesis of PCa. The primary purpose of our research was to investigate alterations in the levels of HP1α expression and to plan a series of tests to validate the function of HP1α in PCa. METHOD Information on HP1α expression in PCa and benign prostatic hyperplasia (BPH) tissues were gathered using the Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis (GEPIA) databases. RT-qPCR, western blotting, and immunohistochemistry (IHC) were used to assess HP1α mRNA and protein expression in several human PCa tissues and cell lines. The CCK8 assay, clone formation assay, and transwell assay were used to examine biological activities including cell proliferation, migration, and invasion. The expression of proteins connected to apoptosis and the epithelial-mesenchymal transition (EMT) was examined using Western blot. The tumorigenic effect of HP1α was also verified by in vivo experiments. RESULT HP1α expression was much higher in PCa than in BPH tissues and cells, and was positively correlated with the Gleason score of PCa. In vitro experiments showed that HP1α knockdown could inhibit the ability of proliferation, invasion, and migration of PC3 and LNCaP cells, and promote cell apoptosis and EMT. In vivo experiments showed that HP1α knockdown inhibited tumorigenesis in mice. CONCLUSION Our findings indicate that HP1α expression promotes PCa development and might be a novel therapeutic target for the diagnosis or treatment of PCa.
Collapse
Affiliation(s)
- Siyang Zhang
- Department of Urology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, 366 Taihu Road, 225300, Taizhou, Jiangsu, China.
| | - Hengran Li
- Department of Urology, North China University of Science and Technology Affiliated Hospital, 73 Jianshe South Road, 063000, Tangshan, Hebei, China
| | - Chong Shen
- Department of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang road, 300211, Tianjin, China
| | - Fenghong Cao
- Department of Urology, North China University of Science and Technology Affiliated Hospital, 73 Jianshe South Road, 063000, Tangshan, Hebei, China
| | - Shaosan Kang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, 73 Jianshe South Road, 063000, Tangshan, Hebei, China
| |
Collapse
|
18
|
Yang F, Xiao Y, Ding JH, Jin X, Ma D, Li DQ, Shi JX, Huang W, Wang YP, Jiang YZ, Shao ZM. Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy. Cell Metab 2023; 35:84-100.e8. [PMID: 36257316 DOI: 10.1016/j.cmet.2022.09.021] [Citation(s) in RCA: 158] [Impact Index Per Article: 158.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/24/2022] [Accepted: 09/21/2022] [Indexed: 01/20/2023]
Abstract
Treatment of triple-negative breast cancer (TNBC) remains challenging. Deciphering the orchestration of metabolic pathways in regulating ferroptosis will provide new insights into TNBC therapeutic strategies. Here, we integrated the multiomics data of our large TNBC cohort (n = 465) to develop the ferroptosis atlas. We discovered that TNBCs had heterogeneous phenotypes in ferroptosis-related metabolites and metabolic pathways. The luminal androgen receptor (LAR) subtype of TNBC was characterized by the upregulation of oxidized phosphatidylethanolamines and glutathione metabolism (especially GPX4), which allowed the utilization of GPX4 inhibitors to induce ferroptosis. Furthermore, we verified that GPX4 inhibition not only induced tumor ferroptosis but also enhanced antitumor immunity. The combination of GPX4 inhibitors and anti-PD1 possessed greater therapeutic efficacy than monotherapy. Clinically, higher GPX4 expression correlated with lower cytolytic scores and worse prognosis in immunotherapy cohorts. Collectively, this study demonstrated the ferroptosis landscape of TNBC and revealed an innovative immunotherapy combination strategy for refractory LAR tumors.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Jia-Han Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xi Jin
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ding Ma
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Da-Qiang Li
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jin-Xiu Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai 201203, China
| | - Wei Huang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai 201203, China
| | - Yi-Ping Wang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
19
|
Castration-resistant prostate cancer cells are dependent on the high activity of CDK7. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04475-3. [DOI: 10.1007/s00432-022-04475-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022]
Abstract
Abstract
Purpose
Prostate cancer (PC) is successfully treated with anti-androgens; however, a significant proportion of patients develop resistance against this therapy. Anti-androgen-resistant disease (castration-resistant prostate cancer; CRPC) is currently incurable. Cyclin-dependent kinase 7 (CDK7) is positioned to positively regulate both cell cycle and transcription, the two features critical for the rapid proliferation of the CRPC cells. Here, we assess if CDK7 is a viable target to halt the proliferation of CRPC cells.
Methods
We use recently developed clinically relevant compounds targeting CDK7 and multiple cell proliferation assays to probe the importance of this kinase for the proliferation of normal, androgen-dependent, and CRPC cells. PC patient data were used to evaluate expression of CDK7 at different disease-stages. Finally, comprehensive glycoproteome-profiling was performed to evaluate CDK7 inhibitor effects on androgen-dependent and CRPC cells.
Results
We show that CDK7 is overexpressed in PC patients with poor prognosis, and that CRPC cells are highly sensitive to compounds targeting CDK7. Inhibition of O-GlcNAc transferase sensitizes the CRPC, but not androgen-dependent PC cells, to CDK7 inhibitors. Glycoproteome-profiling revealed that CDK7 inhibition induces hyper-O-GlcNAcylation of the positive transcription elongation complex (pTEFB: CDK9 and CCNT1) in the CRPC cells. Accordingly, co-targeting of CDK7 and CDK9 synergistically blocks the proliferation of the CRPC cells but does not have anti-proliferative effects in the normal prostate cells.
Conclusion
We show that CRPC cells, but not normal prostate cells, are addicted on the high activity of the key transcriptional kinases, CDK7 and CDK9.
Collapse
|
20
|
Bai T, Chen Y, Beck S, Stanelle-Bertram S, Mounogou NK, Chen T, Dong J, Schneider B, Jia T, Yang J, Wang L, Meinhardt A, Zapf A, Kreienbrock L, Wang D, Shu Y, Gabriel G. H7N9 avian influenza virus infection in men is associated with testosterone depletion. Nat Commun 2022; 13:6936. [PMID: 36376288 PMCID: PMC9662777 DOI: 10.1038/s41467-022-34500-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/27/2022] [Indexed: 11/16/2022] Open
Abstract
Human infections with H7N9 avian influenza A virus that emerged in East China in 2013 and caused high morbidity rates were more frequently detected in men than in women over the last five epidemic waves. However, molecular markers associated with poor disease outcomes in men are still unknown. In this study, we systematically analysed sex hormone and cytokine levels in males and females with laboratory-confirmed H7N9 influenza in comparison to H7N9-negative control groups as well as laboratory-confirmed seasonal H1N1/H3N2 influenza cases (n = 369). Multivariable analyses reveal that H7N9-infected men present with considerably reduced testosterone levels associated with a poor outcome compared to non-infected controls. Regression analyses reveal that testosterone levels in H7N9-infected men are negatively associated with the levels of several pro-inflammatory cytokines, such as IL-6 and IL-15. To assess whether there is a causal relationship between low testosterone levels and avian H7N9 influenza infection, we used a mouse model. In male mice, we show that respiratory H7N9 infection leads to a high viral load and inflammatory cytokine response in the testes as well as a reduction in pre-infection plasma testosterone levels. Collectively, these findings suggest that monitoring sex hormone levels may support individualized management for patients with avian influenza infections.
Collapse
Affiliation(s)
- Tian Bai
- Viral Zoonoses-One Health, Leibniz Institute for Virology (LIV), Hamburg, Germany ,grid.198530.60000 0000 8803 2373Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206 P.R. China
| | - Yongkun Chen
- grid.12981.330000 0001 2360 039XSchool of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, 510275 P.R. China ,grid.12981.330000 0001 2360 039XSchool of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107 P.R. China
| | - Sebastian Beck
- Viral Zoonoses-One Health, Leibniz Institute for Virology (LIV), Hamburg, Germany
| | | | | | - Tao Chen
- grid.198530.60000 0000 8803 2373Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206 P.R. China
| | - Jie Dong
- grid.198530.60000 0000 8803 2373Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206 P.R. China
| | - Bettina Schneider
- grid.412970.90000 0001 0126 6191Department of Biometry, Epidemiology and Information Processing, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Tingting Jia
- grid.12981.330000 0001 2360 039XSchool of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, 510275 P.R. China ,grid.12981.330000 0001 2360 039XSchool of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107 P.R. China
| | - Jing Yang
- grid.198530.60000 0000 8803 2373Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206 P.R. China
| | - Lijie Wang
- grid.198530.60000 0000 8803 2373Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206 P.R. China
| | - Andreas Meinhardt
- grid.8664.c0000 0001 2165 8627Institute for Anatomy and Cell Biology, Justus-Liebig University of Gießen, Gießen, Germany
| | - Antonia Zapf
- grid.13648.380000 0001 2180 3484Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lothar Kreienbrock
- grid.412970.90000 0001 0126 6191Department of Biometry, Epidemiology and Information Processing, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Dayan Wang
- grid.198530.60000 0000 8803 2373Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206 P.R. China
| | - Yuelong Shu
- grid.198530.60000 0000 8803 2373Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206 P.R. China ,grid.12981.330000 0001 2360 039XSchool of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, 510275 P.R. China ,grid.12981.330000 0001 2360 039XSchool of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107 P.R. China ,grid.506261.60000 0001 0706 7839Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Gülsah Gabriel
- Viral Zoonoses-One Health, Leibniz Institute for Virology (LIV), Hamburg, Germany ,grid.412970.90000 0001 0126 6191Institute of Virology, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
21
|
Wu Q, Qian W, Sun X, Jiang S. Small-molecule inhibitors, immune checkpoint inhibitors, and more: FDA-approved novel therapeutic drugs for solid tumors from 1991 to 2021. J Hematol Oncol 2022; 15:143. [PMID: 36209184 PMCID: PMC9548212 DOI: 10.1186/s13045-022-01362-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/02/2022] [Indexed: 11/10/2022] Open
Abstract
The United States Food and Drug Administration (US FDA) has always been a forerunner in drug evaluation and supervision. Over the past 31 years, 1050 drugs (excluding vaccines, cell-based therapies, and gene therapy products) have been approved as new molecular entities (NMEs) or biologics license applications (BLAs). A total of 228 of these 1050 drugs were identified as cancer therapeutics or cancer-related drugs, and 120 of them were classified as therapeutic drugs for solid tumors according to their initial indications. These drugs have evolved from small molecules with broad-spectrum antitumor properties in the early stage to monoclonal antibodies (mAbs) and antibody‒drug conjugates (ADCs) with a more precise targeting effect during the most recent decade. These drugs have extended indications for other malignancies, constituting a cancer treatment system for monotherapy or combined therapy. However, the available targets are still mainly limited to receptor tyrosine kinases (RTKs), restricting the development of antitumor drugs. In this review, these 120 drugs are summarized and classified according to the initial indications, characteristics, or functions. Additionally, RTK-targeted therapies and immune checkpoint-based immunotherapies are also discussed. Our analysis of existing challenges and potential opportunities in drug development may advance solid tumor treatment in the future.
Collapse
Affiliation(s)
- Qing Wu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| | - Wei Qian
- Department of Radiology, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310009 Zhejiang China
| | - Xiaoli Sun
- Department of Radiation Oncology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310003 Zhejiang China
| | - Shaojie Jiang
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| |
Collapse
|
22
|
AR-regulated ZIC5 contributes to the aggressiveness of prostate cancer. Cell Death Dis 2022; 8:393. [PMID: 36127329 PMCID: PMC9489711 DOI: 10.1038/s41420-022-01181-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/23/2022]
Abstract
The mechanisms by which prostate cancer (PCa) progresses to the aggressive castration-resistant stage remain uncertain. Zinc finger of the cerebellum 5 (ZIC5), a transcription factor belonging to the ZIC family, is involved in the pathology of various cancers. However, the potential effect of ZIC5 on PCa malignant progression has not been fully defined. Here, we show that ZIC5 is upregulated in PCa, particularly in metastatic lesions, in positive association with poor prognosis. Genetic inhibition of ZIC5 in PCa cells obviously attenuated invasion and metastasis and blunted the oncogenic properties of colony formation. Mechanistically, ZIC5 functioned as a transcription factor to promote TWIST1-mediated EMT progression or as a cofactor to strengthen the β-catenin-TCF4 association and stimulate Wnt/β-catenin signaling. Importantly, ZIC5 and the androgen receptor (AR) form a positive feed-forward loop to mutually stimulate each other’s expression. AR, in cooperation with its steroid receptor coactivator 3 (SRC-3), increased ZIC5 expression through binding to the miR-27b-3p promoter and repressing miR-27b-3p transcription. In turn, ZIC5 potentiated AR, AR-V7, and AR targets’ expression. Besides, ZIC5 inhibition reduced AR and AR-V7 protein expression and enhanced the sensitivity of PCa to enzalutamide (Enz) treatment, both in vitro and in vivo. These findings indicate that the reciprocal activation between AR and ZIC5 promotes metastasis and Enz resistance of PCa and suggest the therapeutic value of cotargeting ZIC5 and AR for the treatment of advanced PCa.
Collapse
|
23
|
Huang JL, Yan XL, Li W, Fan RZ, Li S, Chen J, Zhang Z, Sang J, Gan L, Tang GH, Chen H, Wang J, Yin S. Discovery of Highly Potent Daphnane Diterpenoids Uncovers Importin-β1 as a Druggable Vulnerability in Castration-Resistant Prostate Cancer. J Am Chem Soc 2022; 144:17522-17532. [DOI: 10.1021/jacs.2c06449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Jia-Luo Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Xue-Long Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
- School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou 550025, P.R. China
| | - Wei Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Run-Zhu Fan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Shen Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Jianghe Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Zhenhua Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Jun Sang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Lu Gan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Gui-Hua Tang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Hongwu Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California 95817, United States
| | - Junjian Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Sheng Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
24
|
Caggiano C, Pieraccioli M, Pitolli C, Babini G, Zheng D, Tian B, Bielli P, Sette C. The androgen receptor couples promoter recruitment of RNA processing factors to regulation of alternative polyadenylation at the 3' end of transcripts. Nucleic Acids Res 2022; 50:9780-9796. [PMID: 36043441 PMCID: PMC9508809 DOI: 10.1093/nar/gkac737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 07/20/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer (PC) relies on androgen receptor (AR) signaling. While hormonal therapy (HT) is efficacious, most patients evolve to an incurable castration-resistant stage (CRPC). To date, most proposed mechanisms of acquired resistance to HT have focused on AR transcriptional activity. Herein, we uncover a new role for the AR in alternative cleavage and polyadenylation (APA). Inhibition of the AR by Enzalutamide globally regulates APA in PC cells, with specific enrichment in genes related to transcription and DNA topology, suggesting their involvement in transcriptome reprogramming. AR inhibition selects promoter-distal polyadenylation sites (pAs) enriched in cis-elements recognized by the cleavage and polyadenylation specificity factor (CPSF) complex. Conversely, promoter-proximal intronic pAs relying on the cleavage stimulation factor (CSTF) complex are repressed. Mechanistically, Enzalutamide induces rearrangement of APA subcomplexes and impairs the interaction between CPSF and CSTF. AR inhibition also induces co-transcriptional CPSF recruitment to gene promoters, predisposing the selection of pAs depending on this complex. Importantly, the scaffold CPSF160 protein is up-regulated in CRPC cells and its depletion represses HT-induced APA patterns. These findings uncover an unexpected role for the AR in APA regulation and suggest that APA-mediated transcriptome reprogramming represents an adaptive response of PC cells to HT.
Collapse
Affiliation(s)
- Cinzia Caggiano
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome 00168, Italy.,IRCCS Fondazione Policlinico A. Gemelli, Rome 00168, Italy
| | - Marco Pieraccioli
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome 00168, Italy.,IRCCS Fondazione Policlinico A. Gemelli, Rome 00168, Italy
| | - Consuelo Pitolli
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome 00168, Italy.,IRCCS Fondazione Policlinico A. Gemelli, Rome 00168, Italy
| | | | - Dinghai Zheng
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Bin Tian
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Pamela Bielli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome 00133, Italy.,IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| | - Claudio Sette
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome 00168, Italy.,IRCCS Fondazione Policlinico A. Gemelli, Rome 00168, Italy
| |
Collapse
|
25
|
Chen M, Lingadahalli S, Narwade N, Lei KMK, Liu S, Zhao Z, Zheng Y, Lu Q, Tang AHN, Poon TCW, Cheung E. TRIM33 drives prostate tumor growth by stabilizing androgen receptor from Skp2-mediated degradation. EMBO Rep 2022; 23:e53468. [PMID: 35785414 DOI: 10.15252/embr.202153468] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 05/13/2022] [Accepted: 06/07/2022] [Indexed: 12/23/2022] Open
Abstract
Androgen receptor (AR) is a master transcription factor that drives prostate cancer (PCa) development and progression. Alterations in the expression or activity of AR coregulators significantly impact the outcome of the disease. Using a proteomics approach, we identified the tripartite motif-containing 33 (TRIM33) as a novel transcriptional coactivator of AR. We demonstrate that TRIM33 facilitates AR chromatin binding to directly regulate a transcription program that promotes PCa progression. TRIM33 further stabilizes AR by protecting it from Skp2-mediated ubiquitination and proteasomal degradation. We also show that TRIM33 is essential for PCa tumor growth by avoiding cell-cycle arrest and apoptosis, and TRIM33 knockdown sensitizes PCa cells to AR antagonists. In clinical analyses, we find TRIM33 upregulated in multiple PCa patient cohorts. Finally, we uncover an AR-TRIM33-coactivated gene signature highly expressed in PCa tumors and predict disease recurrence. Overall, our results reveal that TRIM33 is an oncogenic AR coactivator in PCa and a potential therapeutic target for PCa treatment.
Collapse
Affiliation(s)
- Mi Chen
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Shreyas Lingadahalli
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Nitin Narwade
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Kate Man Kei Lei
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Pilot Laboratory, University of Macau, Taipa, Macau SAR.,Institute of Translational Medicine, University of Macau, Taipa, Macau SAR
| | | | - Zuxianglan Zhao
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Yimin Zheng
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Qian Lu
- Xuzhou Medical University, Xuzhou, China
| | | | - Terence Chuen Wai Poon
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR.,Pilot Laboratory, University of Macau, Taipa, Macau SAR.,Institute of Translational Medicine, University of Macau, Taipa, Macau SAR
| | - Edwin Cheung
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| |
Collapse
|
26
|
Gan X, Huang H, Wen J, Liu K, Yang Y, Li X, Fang G, Liu Y, Wang X. α-Terthienyl induces prostate cancer cell death through inhibiting androgen receptor expression. Biomed Pharmacother 2022; 152:113266. [PMID: 35691152 DOI: 10.1016/j.biopha.2022.113266] [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: 02/03/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022] Open
Abstract
Prostate cancer is a disease that often occurs in elderly men. Androgen receptor signaling pathway runs through the occurrence and development of prostate cancer. Thereby, targeting androgen receptor is a crucial strategy for the treatment of prostate cancer. α-Terthienyl, which has been used as photosensitive activator and insecticide, is a natural compound rich in marigold. In the present study, we found α- terthienyl could inhibit the cell viability of four prostate cancer cell lines, especially on LNCaP and 22Rv1 cells which endogenously express androgen receptor. Then we proved that it could inhibit the proliferation of prostate cancer cells and induce apoptosis of prostate cancer cells by plate clone formation assay and flow cytometry respectively. Furthermore, we found α-terthienyl could inhibit androgen receptor nuclear translocation, reduce androgen receptor expression, reduce the mRNA and protein expression of androgen receptor target genes (KLK3, TMPRSS2, PCA3) and nuclear proliferation antigen Ki67 and PCNA. In addition, it inhibited the expression and phosphorylation of Akt protein while increasing the expression of tumor suppressor p27. Besides, we constructed a mouse xenograft prostate cancer model and confirmed that α-terthienyl also inhibited the growth of prostate cancer in vivo. In conclusively, α-terthienyl played an anti-prostate cancer role by inhibiting both the expression of androgen receptor and the transduction of its signal pathway, suggesting that it is a promising natural small molecule for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Xia Gan
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning 530200, China; Institute of Marine Drugs, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning 530200, China
| | - Hailing Huang
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning 530200, China
| | - Jing Wen
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning 530200, China
| | - Kai Liu
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning 530200, China
| | - Yuting Yang
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning 530200, China
| | - Xiaoning Li
- School of Foreign Languages, Nanning Normal University, 3 Hexing Road, Qingxiu District, Nanning 530299, China
| | - Gang Fang
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning 530200, China.
| | - Yonghong Liu
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning 530200, China; CAS Key Laboratory of Tropical Marine Bio-resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China.
| | - Xueni Wang
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning 530200, China; CAS Key Laboratory of Tropical Marine Bio-resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China.
| |
Collapse
|
27
|
Hu R, Huang JL, Yuan FY, Wei X, Zou MF, Tang GH, Li W, Yin S. Crotonianoids A-C, Three Unusual Tigliane Diterpenoids from the Seeds of Croton tiglium and Their Anti-Prostate Cancer Activity. J Org Chem 2022; 87:9301-9306. [PMID: 35758034 DOI: 10.1021/acs.joc.2c01024] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Crotonianoids A-C (1-3), three unusual tigliane diterpenoids, were isolated from the seeds of Croton tiglium. Compound 1 is a 13,14:13,15-diseco-tigliane featuring a unique spiro[bicyclo[5.3.0]decane-2,5'-2'(3'H,4'H)-furanone] core; 2 is a 13,15-seco-tigliane incorporating a rare peroxide bridge between C-13 and C-15; and 3 is the first example of a phorbol ester with a 10R-configuration. Their structures were determined by spectroscopic, computational, and X-ray diffraction methods. Compounds 1 and 2 markedly inhibited the growth and survival of prostate cancer cell C4-2B at micromolar concentrations and induced cell apoptosis. Mechanistic study revealed that 1 and 2 could suppress androgen receptor (AR) signaling pathway by promoting the degradation of AR protein.
Collapse
Affiliation(s)
- Rong Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Jia-Luo Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Fang-Yu Yuan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Xun Wei
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Ming-Feng Zou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Gui-Hua Tang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Wei Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Sheng Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| |
Collapse
|
28
|
Azeem W, Olsen JR, Hellem MR, Hua Y, Marvyin K, Ke X, Øyan AM, Kalland KH. Proteasome-Mediated Regulation of GATA2 Expression and Androgen Receptor Transcription in Benign Prostate Epithelial Cells. Biomedicines 2022; 10:biomedicines10020473. [PMID: 35203681 PMCID: PMC8962351 DOI: 10.3390/biomedicines10020473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
GATA2 has been shown to be an important transcription factor together with androgen receptor (AR) in prostate cancer cells. Less is known about GATA2 in benign prostate epithelial cells. We have investigated if GATA2 exogenous expression in prostate epithelial basal-like cells could induce AR transcription or luminal differentiation. Prostate epithelial basal-like (transit amplifying) cells were transduced with lentiviral vector expressing GATA2. Luminal differentiation markers were assessed by RT-qPCR, Western blot and global gene expression microarrays. We utilized our previously established AR and androgen-dependent fluorescence reporter assay to investigate AR activity at the single-cell level. Exogenous GATA2 protein was rapidly and proteasome-dependently degraded. GATA2 protein expression was rescued by the proteasome inhibitor MG132 and partly by mutating the target site of the E3 ligase FBXW7. Moreover, MG132-mediated proteasome inhibition induced AR mRNA and additional luminal marker gene transcription in the prostate transit amplifying cells. Different types of intrinsic mechanisms restricted GATA2 expression in the transit amplifying cells. The appearance of AR mRNA and additional luminal marker gene expression changes following proteasome inhibition suggests control of essential cofactor(s) of AR mRNA expression and luminal differentiation at this proteolytic level.
Collapse
Affiliation(s)
- Waqas Azeem
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
- Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence: (W.A.); (K.-H.K.)
| | - Jan Roger Olsen
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
| | - Margrete Reime Hellem
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
| | - Yaping Hua
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
| | - Kristo Marvyin
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
| | - Xisong Ke
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
| | - Anne Margrete Øyan
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Karl-Henning Kalland
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (J.R.O.); (M.R.H.); (Y.H.); (K.M.); (X.K.); (A.M.Ø.)
- Centre for Cancer Biomarkers, University of Bergen, 5021 Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence: (W.A.); (K.-H.K.)
| |
Collapse
|
29
|
Peng Y, Song Y, Wang H. Systematic Elucidation of the Aneuploidy Landscape and Identification of Aneuploidy Driver Genes in Prostate Cancer. Front Cell Dev Biol 2022; 9:723466. [PMID: 35127694 PMCID: PMC8814427 DOI: 10.3389/fcell.2021.723466] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
Aneuploidy is widely identified as a remarkable feature of malignancy genomes. Increasing evidences suggested aneuploidy was involved in the progression and metastasis of prostate cancer (PCa). Nevertheless, no comprehensive analysis was conducted in PCa about the effects of aneuploidy on different omics and, especially, about the driver genes of aneuploidy. Here, we validated the association of aneuploidy with the progression and prognosis of PCa and performed a systematic analysis in mutation profile, methylation profile, and gene expression profile, which detailed the molecular process aneuploidy implicated. By multi-omics analysis, we managed to identify 11 potential aneuploidy driver genes (GSTM2, HAAO, C2orf88, CYP27A1, FAXDC2, HFE, C8orf88, GSTP1, EFS, HIF3A, and WFDC2), all of which were related to the development and metastasis of PCa. Meanwhile, we also found aneuploidy and its driver genes were correlated with the immune microenvironment of PCa. Our findings could shed light on the tumorigenesis of PCa and provide a better understanding of the development and metastasis of PCa; additionally, the driver genes could be promising and actionable therapeutic targets pointing to aneuploidy.
Collapse
Affiliation(s)
- Yun Peng
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Yuxuan Song
- Department of Urology, Peking University People’s Hospital, Beijing, China
| | - Haitao Wang
- Department of Oncology, the 2nd Hospital of Tianjin Medical University, Tianjin, China
- *Correspondence: Haitao Wang,
| |
Collapse
|
30
|
Erdmann É, Ould Madi Berthélémy P, Cottard F, Angel CZ, Schreyer E, Ye T, Morlet B, Negroni L, Kieffer B, Céraline J. Androgen receptor-mediated transcriptional repression targets cell plasticity in prostate cancer. Mol Oncol 2021; 16:2518-2536. [PMID: 34919781 PMCID: PMC9462842 DOI: 10.1002/1878-0261.13164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/16/2021] [Accepted: 12/15/2021] [Indexed: 11/24/2022] Open
Abstract
Androgen receptor (AR) signaling remains the key therapeutic target in the management of hormone‐naïve‐advanced prostate cancer (PCa) and castration‐resistant PCa (CRPC). Recently, landmark molecular features have been reported for CRPC, including the expression of constitutively active AR variants that lack the ligand‐binding domain. Besides their role in CRPC, AR variants lead to the expression of genes involved in tumor progression. However, little is known about the specificity of their mode of action compared with that of wild‐type AR (AR‐WT). We performed AR transcriptome analyses in an androgen‐dependent PCa cell line as well as cross‐analyses with publicly available RNA‐seq datasets and established that transcriptional repression capacity that was marked for AR‐WT was pathologically lost by AR variants. Functional enrichment analyses allowed us to associate AR‐WT repressive function to a panel of genes involved in cell adhesion and epithelial‐to‐mesenchymal transition. So, we postulate that a less documented AR‐WT normal function in prostate epithelial cells could be the repression of a panel of genes linked to cell plasticity and that this repressive function could be pathologically abrogated by AR variants in PCa.
Collapse
Affiliation(s)
- Éva Erdmann
- CNRS, UMR 7104, INSERM U1258 - IGBMC - University de Strasbourg, France
| | | | - Félicie Cottard
- University of Freiburg - Albert - Ludwigs - Universität Freiburg, Germany
| | | | - Edwige Schreyer
- CNRS, UMR 7104, INSERM U1258 - IGBMC - University de Strasbourg, France
| | - Tao Ye
- CNRS, UMR 7104, INSERM U1258 - IGBMC - University de Strasbourg, France
| | - Bastien Morlet
- CNRS, UMR 7104, INSERM U1258 - IGBMC - University de Strasbourg, France
| | - Luc Negroni
- CNRS, UMR 7104, INSERM U1258 - IGBMC - University de Strasbourg, France
| | - Bruno Kieffer
- CNRS, UMR 7104, INSERM U1258 - IGBMC - University de Strasbourg, France
| | - Jocelyn Céraline
- CNRS, UMR 7104, INSERM U1258 - IGBMC - University de Strasbourg, France.,Institut de Cancérologie de Strasbourg Europe (ICANS), Hôpitaux Universitaires de Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg - FMTS - Faculté de Médecine, Université de Strasbourg, France
| |
Collapse
|
31
|
Single-cell analysis reveals androgen receptor regulates the ER-to-Golgi trafficking pathway with CREB3L2 to drive prostate cancer progression. Oncogene 2021; 40:6479-6493. [PMID: 34611310 DOI: 10.1038/s41388-021-02026-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/29/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023]
Abstract
Androgen receptor (AR) plays a central role in driving prostate cancer (PCa) progression. How AR promotes this process is still not completely clear. Herein, we used single-cell transcriptome analysis to reconstruct the transcriptional network of AR in PCa. Our work shows AR directly regulates a set of signature genes in the ER-to-Golgi protein vesicle-mediated transport pathway. The expression of these genes is required for maximum androgen-dependent ER-to-Golgi trafficking, cell growth, and survival. Our analyses also reveal the signature genes are associated with PCa progression and prognosis. Moreover, we find inhibition of the ER-to-Golgi transport process with a small molecule enhanced antiandrogen-mediated tumor suppression of hormone-sensitive and insensitive PCa. Finally, we demonstrate AR collaborates with CREB3L2 in mediating ER-to-Golgi trafficking in PCa. In summary, our findings uncover a critical role for dysregulation of ER-to-Golgi trafficking expression and function in PCa progression, provide detailed mechanistic insights for how AR tightly controls this process, and highlight the prospect of targeting the ER-to-Golgi pathway as a therapeutic strategy for advanced PCa.
Collapse
|
32
|
Pungsrinont T, Kallenbach J, Baniahmad A. Role of PI3K-AKT-mTOR Pathway as a Pro-Survival Signaling and Resistance-Mediating Mechanism to Therapy of Prostate Cancer. Int J Mol Sci 2021; 22:11088. [PMID: 34681745 PMCID: PMC8538152 DOI: 10.3390/ijms222011088] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/27/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Androgen deprivation therapy (ADT) and androgen receptor (AR)-targeted therapy are the gold standard options for treating prostate cancer (PCa). These are initially effective, as localized and the early stage of metastatic disease are androgen- and castration-sensitive. The tumor strongly relies on systemic/circulating androgens for activating AR signaling to stimulate growth and progression. However, after a certain point, the tumor will eventually develop a resistant stage, where ADT and AR antagonists are no longer effective. Mechanistically, it seems that the tumor becomes more aggressive through adaptive responses, relies more on alternative activated pathways, and is less dependent on AR signaling. This includes hyperactivation of PI3K-AKT-mTOR pathway, which is a central signal that regulates cell pro-survival/anti-apoptotic pathways, thus, compensating the blockade of AR signaling. The PI3K-AKT-mTOR pathway is well-documented for its crosstalk between genomic and non-genomic AR signaling, as well as other signaling cascades. Such a reciprocal feedback loop makes it more complicated to target individual factor/signaling for treating PCa. Here, we highlight the role of PI3K-AKT-mTOR signaling as a resistance mechanism for PCa therapy and illustrate the transition of prostate tumor from AR signaling-dependent to PI3K-AKT-mTOR pathway-dependent. Moreover, therapeutic strategies with inhibitors targeting the PI3K-AKT-mTOR signal used in clinic and ongoing clinical trials are discussed.
Collapse
Affiliation(s)
| | | | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, 07747 Jena, Germany; (T.P.); (J.K.)
| |
Collapse
|
33
|
Hałubiec P, Łazarczyk A, Szafrański O, Bohn T, Dulińska-Litewka J. Synthetic Retinoids as Potential Therapeutics in Prostate Cancer-An Update of the Last Decade of Research: A Review. Int J Mol Sci 2021; 22:10537. [PMID: 34638876 PMCID: PMC8508817 DOI: 10.3390/ijms221910537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/16/2022] Open
Abstract
Prostate cancer (PC) is the second most common tumor in males. The search for appropriate therapeutic options against advanced PC has been in process for several decades. Especially after cessation of the effectiveness of hormonal therapy (i.e., emergence of castration-resistant PC), PC management options have become scarce and the prognosis is poor. To overcome this stage of disease, an array of natural and synthetic substances underwent investigation. An interesting and promising class of compounds constitutes the derivatives of natural retinoids. Synthesized on the basis of the structure of retinoic acid, they present unique and remarkable properties that warrant their investigation as antitumor drugs. However, there is no up-to-date compilation that consecutively summarizes the current state of knowledge about synthetic retinoids with regard to PC. Therefore, in this review, we present the results of the experimental studies on synthetic retinoids conducted within the last decade. Our primary aim is to highlight the molecular targets of these compounds and to identify their potential promise in the treatment of PC.
Collapse
Affiliation(s)
- Przemysław Hałubiec
- Medical Biochemistry Medical College, Jagiellonian University, 31-034 Cracow, Poland; (P.H.); (A.Ł.); (O.S.)
| | - Agnieszka Łazarczyk
- Medical Biochemistry Medical College, Jagiellonian University, 31-034 Cracow, Poland; (P.H.); (A.Ł.); (O.S.)
| | - Oskar Szafrański
- Medical Biochemistry Medical College, Jagiellonian University, 31-034 Cracow, Poland; (P.H.); (A.Ł.); (O.S.)
| | - Torsten Bohn
- Nutrition and Health Research Group 1 A-B, Department of Population Health, Luxembourg Institute of Health, 1 A-B, rue Thomas Edison, L-23 1445 Strassen, Luxembourg;
| | - Joanna Dulińska-Litewka
- Medical Biochemistry Medical College, Jagiellonian University, 31-034 Cracow, Poland; (P.H.); (A.Ł.); (O.S.)
| |
Collapse
|
34
|
Offermann A, Kang D, Watermann C, Weingart A, Hupe MC, Saraji A, Stegmann-Frehse J, Kruper R, Schüle R, Pantel K, Taubert H, Duensing S, Culig Z, Aigner A, Klapper W, Jonigk D, Philipp Kühnel M, Merseburger AS, Kirfel J, Sailer V, Perner S. Manuscript Title: Analysis of tripartite motif (TRIM) family gene expression in prostate cancer bone metastases. Carcinogenesis 2021; 42:1475-1484. [PMID: 34487169 DOI: 10.1093/carcin/bgab083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/29/2021] [Accepted: 09/04/2021] [Indexed: 12/27/2022] Open
Abstract
Tripartite motif (TRIM) family proteins are post-translational protein modifiers with E3-ubiquitin ligase activity, thereby involved in various biological processes. The molecular mechanisms driving prostate cancer (PCa) bone metastasis (BM) are incompletely understood, and targetable genetic alterations are lacking in the majority of cases. Therefore, we aimed to explore the expression and potential functional relevance of 71 TRIM members in bone metastatic PCa. We performed transcriptome analysis of all human TRIM family members and 770 cancer-related genes in 29 localized PCa and 30 PCa BM using Nanostring. KEGG, STRING and Ubibrowser were used for further bioinformatic gene correlation and pathway enrichment analyses. Compared to localized tumors, six TRIMs are under-expressed while nine TRIMs are over-expressed in BM. The differentially expressed TRIM proteins are linked to TNF-, TGFβ-, PI3K/AKT- and HIF-1-signaling, and to features such as proteoglycans, platelet activation, adhesion and ECM-interaction based on correlation to cancer-related genes. The identification of TRIM-specific E3-ligase-substrates revealed insight into functional connections to oncogenes, tumor suppressors and cancer-related pathways including androgen receptor- and TGFβ signaling, cell cycle regulation and splicing. In summary, this is the first study that comprehensively and systematically characterizes the expression of all TRIM members in PCa BM. Our results describe post-translational protein modification as an important regulatory mechanism of oncogenes, tumor suppressors, and pathway molecules in PCa progression. Therefore, this study may provide evidence for novel therapeutic targets, in particular for the treatment or prevention of BM.
Collapse
Affiliation(s)
- Anne Offermann
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Duan Kang
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Christian Watermann
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Anika Weingart
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Marie C Hupe
- Department of Urology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Alireza Saraji
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Janine Stegmann-Frehse
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | | | - Roland Schüle
- Klinik für Urologie und Zentrale Klinische Forschung, Klinikum der Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Klaus Pantel
- Institute for Tumor Biology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Helge Taubert
- Department of Urology and Paediatric Urology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| | - Zoran Culig
- Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, University of Leipzig, Germany
| | - Wolfram Klapper
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, Hannover, Germany
| | - Mark Philipp Kühnel
- Institute of Pathology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, Hannover, Germany
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Jutta Kirfel
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Verena Sailer
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Sven Perner
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| |
Collapse
|
35
|
Xu Z, Huang L, Dai T, Pei X, Xia L, Zeng G, Ye M, Liu K, Zeng F, Han W, Jiang S. SQLE Mediates Metabolic Reprogramming to Promote LN Metastasis in Castration-Resistant Prostate Cancer. Onco Targets Ther 2021; 14:4285-4295. [PMID: 34335030 PMCID: PMC8318010 DOI: 10.2147/ott.s315813] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/13/2021] [Indexed: 12/21/2022] Open
Abstract
Background Almost all metastatic hormone-sensitive prostate cancers (mHSPC) will develop into metastatic castration-resistant prostate cancer (mCRPC) after androgen deprivation therapy (ADT). The expression level of squalene monooxygenase (SQLE) is increased in CRPC cells and regulates cholesterol metabolism. This study verified the biological function and mechanisms of SQLE in CRPC. Methods The expression of SQLE in human prostate cancer cells was overexpressed or silenced and its efficacy on cell survival was determined by the MTS test. Energy metabolism phenotype test was evaluated by XF real-time ATP rate assay, XF cell mitochondrial stress test, XF glycolysis stress test and XF mito fuel flex test. Cell migration and invasion were evaluated by colony formation assays and transwell assays; the expression of mRNA and protein was assessed by RT-qPCR and Western blot, respectively. Moreover, BALB/c nude mice model was performed to evaluate the lymph node metastasis. Results In our study, we found that the expression level of SQLE was significantly increased in bicalutamide-resistant-C4-2B cells compared to LNCaP cells. SQLE knockdown partly restored the sensitivity of drug-resistant cells to bicalutamide and reduced lymph node metastasis by inhibiting fatty acid oxidation in mitochondria. We also found that terbinafine, the specific inhibitor of SQLE, can enhance the sensitivity of prostate cancer cells to bicalutamide. Conclusion Our study revealed that SQLE is involved in the progression of castration resistance in CRPC through mediating metabolic reprogramming, presenting SQLE as a new target for the treatment of mCRPC.
Collapse
Affiliation(s)
- Zhenzhou Xu
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Liang Huang
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Tao Dai
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Xiaming Pei
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Longzheng Xia
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Gongqian Zeng
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Mingji Ye
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Kan Liu
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Fuhua Zeng
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Weiqing Han
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Shusuan Jiang
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| |
Collapse
|
36
|
Glycosylation: Rising Potential for Prostate Cancer Evaluation. Cancers (Basel) 2021; 13:cancers13153726. [PMID: 34359624 PMCID: PMC8345048 DOI: 10.3390/cancers13153726] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Aberrant protein glycosylation is a well-known hallmark of cancer and is associated with differential expression of enzymes such as glycosyltransferases and glycosidases. The altered expression of the enzymes triggers cancer cells to produce glycoproteins with specific cancer-related aberrations in glycan structures. Increasing number of data indicate that glycosylation patterns of PSA and other prostate-originated proteins exert a potential to distinguish between benign prostate disease and cancer as well as among different stages of prostate cancer development and aggressiveness. This review summarizes the alterations in glycan sialylation, fucosylation, truncated O-glycans, and LacdiNAc groups outlining their potential applications in non-invasive diagnostic procedures of prostate diseases. Further research is desired to develop more general algorithms exploiting glycobiology data for the improvement of prostate diseases evaluation. Abstract Prostate cancer is the second most commonly diagnosed cancer among men. Alterations in protein glycosylation are confirmed to be a reliable hallmark of cancer. Prostate-specific antigen is the biomarker that is used most frequently for prostate cancer detection, although its lack of sensitivity and specificity results in many unnecessary biopsies. A wide range of glycosylation alterations in prostate cancer cells, including increased sialylation and fucosylation, can modify protein function and play a crucial role in many important biological processes in cancer, including cell signalling, adhesion, migration, and cellular metabolism. In this review, we summarize studies evaluating the prostate cancer associated glycosylation related alterations in sialylation, mainly α2,3-sialylation, core fucosylation, branched N-glycans, LacdiNAc group and presence of truncated O-glycans (sTn, sT antigen). Finally, we discuss the great potential to make use of glycans as diagnostic and prognostic biomarkers for prostate cancer.
Collapse
|
37
|
El-Kenawi A, Dominguez-Viqueira W, Liu M, Awasthi S, Abraham-Miranda J, Keske A, Steiner KK, Noel L, Serna AN, Dhillon J, Gillies RJ, Yu X, Koomen JM, Yamoah K, Gatenby RA, Ruffell B. Macrophage-derived cholesterol contributes to therapeutic resistance in prostate cancer. Cancer Res 2021; 81:5477-5490. [PMID: 34301759 DOI: 10.1158/0008-5472.can-20-4028] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 06/16/2021] [Accepted: 07/21/2021] [Indexed: 11/16/2022]
Abstract
Castration-resistant prostate cancer (CRPC) is a lethal stage of disease in which androgen receptor (AR) signaling is persistent despite androgen deprivation therapy (ADT). Most studies have focused on investigating cell-autonomous alterations in CRPC, while the contributions of the tumor microenvironment are less well understood. Here we sought to determine the role of tumor-associated macrophages in CRPC, based upon their role in cancer progression and therapeutic resistance. In a syngeneic model that reflected the mutational landscape of CRPC, macrophage depletion resulted in a reduced transcriptional signature for steroid and bile acid synthesis, indicating potential perturbation of cholesterol metabolism. As cholesterol is the precursor of the five major types of steroid hormones, we hypothesized that macrophages were regulating androgen biosynthesis within the prostate tumor microenvironment. Macrophage depletion reduced androgen levels within prostate tumors and restricted androgen receptor (AR) nuclear localization in vitro and in vivo. Macrophages were also cholesterol-rich and were able to transfer cholesterol to tumor cells in vitro. AR nuclear translocation was inhibited by activation of Liver X Receptor (LXR)-β, the master regulator of cholesterol homeostasis. Consistent with these data, macrophage depletion extended survival during ADT and the presence of macrophages correlated with therapeutic resistance in patient-derived explants. Taken together, these findings support the therapeutic targeting of macrophages in CRPC.
Collapse
Affiliation(s)
- Asmaa El-Kenawi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | - Min Liu
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Shivanshu Awasthi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Julieta Abraham-Miranda
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aysenur Keske
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - KayLee K Steiner
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Leenil Noel
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Amparo N Serna
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jasreman Dhillon
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - John M Koomen
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kosj Yamoah
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Robert A Gatenby
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
38
|
Suppression of bone metastatic castration-resistant prostate cancer cell growth by a suicide gene delivered by JC polyomavirus-like particles. Gene Ther 2021:10.1038/s41434-021-00280-8. [PMID: 34285388 DOI: 10.1038/s41434-021-00280-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 11/08/2022]
Abstract
Prostate cancer is one of the most common cancers in men. The heterogeneity and mutations exhibited by prostate cancer cells often results in the progression to incurable metastatic castration-resistant prostate cancer (mCRPC). Our previous investigations demonstrated that the virus-like particles (VLPs) of JC polyomavirus (JCPyV) can deliver exogenous genes to prostate cancer cells for expression. JCPyV VLPs packaging pPSAtk (PSAtk-VLPs) possess the ability to transcriptionally target and selectively induce cytotoxicity in prostate cancer cells in vitro and in vivo, as pPSAtk can only express the thymidine kinase gene, a suicide gene, in androgen receptor-positive cells. To further investigate whether PSAtk-VLPs inhibit the growth of metastasized prostate cancer cells, we established an animal model of bone-metastatic prostate cancer to compare PSAtk-VLPs with leuprorelin acetate and enzalutamide, hormonal agents commonly used in clinical settings, and investigated the effectiveness of PSAtk-VLPs. In the present study, we observed that PSAtk-VLPs effectively inhibited the growth of prostate cancer cells that had metastasized to the bone in the metastatic animal model. In addition, PSAtk-VLPs showed a higher effectiveness than hormone therapy in this animal model study. These results suggest that PSAtk-VLPs may serve as a treatment option for mCRPC therapy in the future.
Collapse
|
39
|
Kukkonen K, Taavitsainen S, Huhtala L, Uusi-Makela J, Granberg KJ, Nykter M, Urbanucci A. Chromatin and Epigenetic Dysregulation of Prostate Cancer Development, Progression, and Therapeutic Response. Cancers (Basel) 2021; 13:3325. [PMID: 34283056 PMCID: PMC8268970 DOI: 10.3390/cancers13133325] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023] Open
Abstract
The dysregulation of chromatin and epigenetics has been defined as the overarching cancer hallmark. By disrupting transcriptional regulation in normal cells and mediating tumor progression by promoting cancer cell plasticity, this process has the ability to mediate all defined hallmarks of cancer. In this review, we collect and assess evidence on the contribution of chromatin and epigenetic dysregulation in prostate cancer. We highlight important mechanisms leading to prostate carcinogenesis, the emergence of castration-resistance upon treatment with androgen deprivation therapy, and resistance to antiandrogens. We examine in particular the contribution of chromatin structure and epigenetics to cell lineage commitment, which is dysregulated during tumorigenesis, and cell plasticity, which is altered during tumor progression.
Collapse
Affiliation(s)
- Konsta Kukkonen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Sinja Taavitsainen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Laura Huhtala
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Joonas Uusi-Makela
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Kirsi J. Granberg
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Alfonso Urbanucci
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
40
|
Martinez RS, Salji MJ, Rushworth L, Ntala C, Rodriguez Blanco G, Hedley A, Clark W, Peixoto P, Hervouet E, Renaude E, Kung SHY, Galbraith LCA, Nixon C, Lilla S, MacKay GM, Fazli L, Gaughan L, Sumpton D, Gleave ME, Zanivan S, Blomme A, Leung HY. SLFN5 Regulates LAT1-Mediated mTOR Activation in Castration-Resistant Prostate Cancer. Cancer Res 2021; 81:3664-3678. [PMID: 33985973 DOI: 10.1158/0008-5472.can-20-3694] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/15/2021] [Accepted: 05/11/2021] [Indexed: 11/16/2022]
Abstract
Androgen deprivation therapy (ADT) is the standard of care for treatment of nonresectable prostate cancer. Despite high treatment efficiency, most patients ultimately develop lethal castration-resistant prostate cancer (CRPC). In this study, we performed a comparative proteomic analysis of three in vivo, androgen receptor (AR)-responsive orthograft models of matched hormone-naïve prostate cancer and CRPC. Differential proteomic analysis revealed that distinct molecular mechanisms, including amino acid (AA) and fatty acid metabolism, are involved in the response to ADT in the different models. Despite this heterogeneity, Schlafen family member 5 (SLFN5) was identified as an AR-regulated protein in CRPC. SLFN5 expression was high in CRPC tumors and correlated with poor patient outcome. In vivo, SLFN5 depletion strongly impaired tumor growth in castrated conditions. Mechanistically, SLFN5 interacted with ATF4 and regulated the expression of LAT1, an essential AA transporter. Consequently, SLFN5 depletion in CRPC cells decreased intracellular levels of essential AA and impaired mTORC1 signaling in a LAT1-dependent manner. These results confirm that these orthograft models recapitulate the high degree of heterogeneity observed in patients with CRPC and further highlight SLFN5 as a clinically relevant target for CRPC. SIGNIFICANCE: This study identifies SLFN5 as a novel regulator of the LAT1 amino acid transporter and an essential contributor to mTORC1 activity in castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Rafael S Martinez
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Mark J Salji
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Linda Rushworth
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Chara Ntala
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | | | - Ann Hedley
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - William Clark
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Paul Peixoto
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- EPIGENExp, (EPIgenetics and GENe EXPression Technical Platform), Besançon, France
| | - Eric Hervouet
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- EPIGENExp, (EPIgenetics and GENe EXPression Technical Platform), Besançon, France
| | - Elodie Renaude
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- EPIGENExp, (EPIgenetics and GENe EXPression Technical Platform), Besançon, France
| | - Sonia H Y Kung
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Laura C A Galbraith
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Colin Nixon
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Sergio Lilla
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Gillian M MacKay
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Ladan Fazli
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Luke Gaughan
- Northern Institute for Cancer Research, The Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David Sumpton
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Martin E Gleave
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Sara Zanivan
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Arnaud Blomme
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom.
| | - Hing Y Leung
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom.
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| |
Collapse
|
41
|
Cangiano M, Grudniewska M, Salji MJ, Nykter M, Jenster G, Urbanucci A, Granchi Z, Janssen B, Hamilton G, Leung HY, Beumer IJ. Gene Regulation Network Analysis on Human Prostate Orthografts Highlights a Potential Role for the JMJD6 Regulon in Clinical Prostate Cancer. Cancers (Basel) 2021; 13:cancers13092094. [PMID: 33925994 PMCID: PMC8123677 DOI: 10.3390/cancers13092094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/09/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Prostate cancer is a very common malignancy worldwide. Treatment resistant prostate cancer poses a big challenge to clinicians and is the second most common cause of premature death in men with cancer. Gene expression analysis has been performed on clinical tumours but to date none of the gene expression-based biomarkers for prostate cancer have been successfully integrated to into clinical practice to improve patient management and treatment choice. We applied a novel laboratory prostate cancer model to mimic clinical hormone responsive and resistant prostate cancer and tested whether a network of genes similarly regulated by transcription factors (gene products that control the expression of target genes) are associated with patient outcome. We identified regulons (networks of genes similarly regulated) from our preclinical prostate cancer models and further evaluated the top ranked JMJD6 gene related regulated network in three independent clinical patient cohorts. Abstract Background: Prostate cancer (PCa) is the second most common tumour diagnosed in men. Tumoral heterogeneity in PCa creates a significant challenge to develop robust prognostic markers and novel targets for therapy. An analysis of gene regulatory networks (GRNs) in PCa may provide insight into progressive PCa. Herein, we exploited a graph-based enrichment score to integrate data from GRNs identified in preclinical prostate orthografts and differentially expressed genes in clinical resected PCa. We identified active regulons (transcriptional regulators and their targeted genes) associated with PCa recurrence following radical prostatectomy. Methods: The expression of known transcription factors and co-factors was analysed in a panel of prostate orthografts (n = 18). We searched for genes (as part of individual GRNs) predicted to be regulated by the highest number of transcriptional factors. Using differentially expressed gene analysis (on a per sample basis) coupled with gene graph enrichment analysis, we identified candidate genes and associated GRNs in PCa within the UTA cohort, with the most enriched regulon being JMJD6, which was further validated in two additional cohorts, namely EMC and ICGC cohorts. Cox regression analysis was performed to evaluate the association of the JMJD6 regulon activity with disease-free survival time in the three clinical cohorts as well as compared to three published prognostic gene signatures (TMCC11, BROMO-10 and HYPOXIA-28). Results: 1308 regulons were correlated to transcriptomic data from the three clinical prostatectomy cohorts. The JMJD6 regulon was identified as the top enriched regulon in the UTA cohort and again validated in the EMC cohort as the top-ranking regulon. In both UTA and EMC cohorts, the JMJD6 regulon was significantly associated with cancer recurrence. Active JMJD6 regulon also correlated with disease recurrence in the ICGC cohort. Furthermore, Kaplan–Meier analysis confirmed shorter time to recurrence in patients with active JMJD6 regulon for all three clinical cohorts (UTA, EMC and ICGC), which was not the case for three published prognostic gene signatures (TMCC11, BROMO-10 and HYPOXIA-28). In multivariate analysis, the JMJD6 regulon status significantly predicted disease recurrence in the UTA and EMC, but not ICGC datasets, while none of the three published signatures significantly prognosticate for cancer recurrence. Conclusions: We have characterised gene regulatory networks from preclinical prostate orthografts and applied transcriptomic data from three clinical cohorts to evaluate the prognostic potential of the JMJD6 regulon.
Collapse
Affiliation(s)
- Mario Cangiano
- GenomeScan B.V. Plesmanlaan 1D, 2333 BZ Leiden, The Netherlands; (M.C.); (M.G.); (Z.G.); (B.J.)
| | - Magda Grudniewska
- GenomeScan B.V. Plesmanlaan 1D, 2333 BZ Leiden, The Netherlands; (M.C.); (M.G.); (Z.G.); (B.J.)
| | - Mark J. Salji
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK;
- CRUK Beatson Institute, Glasgow G61 1BD, UK
| | - Matti Nykter
- Laboratory of Computational Biology, Institute of Biomedical Technology, Arvo Ylpön katu 34, 33520 Tampere, Finland;
| | - Guido Jenster
- Department of Urology, Erasmus Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands;
| | - Alfonso Urbanucci
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway;
| | - Zoraide Granchi
- GenomeScan B.V. Plesmanlaan 1D, 2333 BZ Leiden, The Netherlands; (M.C.); (M.G.); (Z.G.); (B.J.)
| | - Bart Janssen
- GenomeScan B.V. Plesmanlaan 1D, 2333 BZ Leiden, The Netherlands; (M.C.); (M.G.); (Z.G.); (B.J.)
| | - Graham Hamilton
- Glasgow Polyomics, University of Glasgow, Glasgow G61 1QH, UK;
| | - Hing Y. Leung
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK;
- CRUK Beatson Institute, Glasgow G61 1BD, UK
- Correspondence: (H.Y.L.); (I.J.B.)
| | - Inès J. Beumer
- GenomeScan B.V. Plesmanlaan 1D, 2333 BZ Leiden, The Netherlands; (M.C.); (M.G.); (Z.G.); (B.J.)
- Correspondence: (H.Y.L.); (I.J.B.)
| |
Collapse
|
42
|
Xu F, Shangguan X, Pan J, Yue Z, Shen K, Ji Y, Zhang W, Zhu Y, Sha J, Wang Y, Fan L, Dong B, Wang Q, Xue W. HOXD13 suppresses prostate cancer metastasis and BMP4-induced epithelial-mesenchymal transition by inhibiting SMAD1. Int J Cancer 2021; 148:3060-3070. [PMID: 33521930 DOI: 10.1002/ijc.33494] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/20/2022]
Abstract
The HOX genes are a group of highly conserved Homeobox-containing genes that control the body plan organization during development. However, their contributions to tumorigenesis and tumor progression remain uncertain and controversial. Here we provided evidence of tumor-suppressive activity of HOXD13 in prostate cancer. HOXD13 depletion contributes to more aggressiveness of prostate cancer cells in vitro and in vivo. These effects were corroborated in a metastatic mice model, where we observed more bone metastatic lesions formed by prostate cancer cells with HOXD13 ablation. Mechanistically, HOXD13 prevents BMP4-induced epithelial-mesenchymal transition (EMT) by inhibiting mothers against decapentaplegic homolog 1 (SMAD1) transcription. Both bioinformation and our tissue microarray cohort data show that HOXD13 expression inversely correlated in advanced prostate cancer patient specimens. Our findings establish HOXD13 as a negative regulator of prostate cancer progression and metastasis by preventing BMP4/SMAD1 signaling, and potentially suggest new strategies for targeting metastatic prostate cancer.
Collapse
Affiliation(s)
- Fan Xu
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xun Shangguan
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiahua Pan
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiying Yue
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Shen
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiyi Ji
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weiwei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yinjie Zhu
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Sha
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanqing Wang
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liancheng Fan
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Baijun Dong
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Wang
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Xue
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
43
|
Rebello RJ, Oing C, Knudsen KE, Loeb S, Johnson DC, Reiter RE, Gillessen S, Van der Kwast T, Bristow RG. Prostate cancer. Nat Rev Dis Primers 2021. [PMID: 33542230 DOI: 10.1038/s41572-020-0024.3-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2023]
Abstract
Prostate cancer is a complex disease that affects millions of men globally, predominantly in high human development index regions. Patients with localized disease at a low to intermediate risk of recurrence generally have a favourable outcome of 99% overall survival for 10 years if the disease is detected and treated at an early stage. Key genetic alterations include fusions of TMPRSS2 with ETS family genes, amplification of the MYC oncogene, deletion and/or mutation of PTEN and TP53 and, in advanced disease, amplification and/or mutation of the androgen receptor (AR). Prostate cancer is usually diagnosed by prostate biopsy prompted by a blood test to measure prostate-specific antigen levels and/or digital rectal examination. Treatment for localized disease includes active surveillance, radical prostatectomy or ablative radiotherapy as curative approaches. Men whose disease relapses after prostatectomy are treated with salvage radiotherapy and/or androgen deprivation therapy (ADT) for local relapse, or with ADT combined with chemotherapy or novel androgen signalling-targeted agents for systemic relapse. Advanced prostate cancer often progresses despite androgen ablation and is then considered castration-resistant and incurable. Current treatment options include AR-targeted agents, chemotherapy, radionuclides and the poly(ADP-ribose) inhibitor olaparib. Current research aims to improve prostate cancer detection, management and outcomes, including understanding the fundamental biology at all stages of the disease.
Collapse
Affiliation(s)
- Richard J Rebello
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK
| | - Christoph Oing
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK
- Department of Oncology, Haematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Centre Eppendorf, Hamburg, Germany
| | - Karen E Knudsen
- Sidney Kimmel Cancer Center at Jefferson Health and Thomas Jefferson University, Philadelphia, PA, USA
| | - Stacy Loeb
- Department of Urology and Population Health, New York University and Manhattan Veterans Affairs, Manhattan, NY, USA
| | - David C Johnson
- Department of Urology, University of North Carolina, Chapel Hill, NC, USA
| | - Robert E Reiter
- Department of Urology, Jonssen Comprehensive Cancer Center UCLA, Los Angeles, CA, USA
| | | | - Theodorus Van der Kwast
- Laboratory Medicine Program, Princess Margaret Cancer Center, University Health Network, Toronto, Canada
| | - Robert G Bristow
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK.
| |
Collapse
|
44
|
Abstract
Prostate cancer is a complex disease that affects millions of men globally, predominantly in high human development index regions. Patients with localized disease at a low to intermediate risk of recurrence generally have a favourable outcome of 99% overall survival for 10 years if the disease is detected and treated at an early stage. Key genetic alterations include fusions of TMPRSS2 with ETS family genes, amplification of the MYC oncogene, deletion and/or mutation of PTEN and TP53 and, in advanced disease, amplification and/or mutation of the androgen receptor (AR). Prostate cancer is usually diagnosed by prostate biopsy prompted by a blood test to measure prostate-specific antigen levels and/or digital rectal examination. Treatment for localized disease includes active surveillance, radical prostatectomy or ablative radiotherapy as curative approaches. Men whose disease relapses after prostatectomy are treated with salvage radiotherapy and/or androgen deprivation therapy (ADT) for local relapse, or with ADT combined with chemotherapy or novel androgen signalling-targeted agents for systemic relapse. Advanced prostate cancer often progresses despite androgen ablation and is then considered castration-resistant and incurable. Current treatment options include AR-targeted agents, chemotherapy, radionuclides and the poly(ADP-ribose) inhibitor olaparib. Current research aims to improve prostate cancer detection, management and outcomes, including understanding the fundamental biology at all stages of the disease.
Collapse
|
45
|
Estrogen Receptor on the move: Cistromic plasticity and its implications in breast cancer. Mol Aspects Med 2020; 78:100939. [PMID: 33358533 DOI: 10.1016/j.mam.2020.100939] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 01/27/2023]
Abstract
Estrogen Receptor (ERα) is a hormone-driven transcription factor, critically involved in driving tumor cell proliferation in the vast majority of breast cancers (BCas). ERα binds the genome at cis-regulatory elements, dictating the expression of a large spectrum of responsive genes in 3D genomic space. While initial reports described a rather static ERα chromatin binding repertoire, we now know that ERα DNA interactions are highly versatile, altered in breast tumor development and progression, and deviate between tumors from patients with differential outcome. Multiple cellular signaling cascades are known to impinge on ERα genomic function, changing its cistrome to retarget the receptor to other regions of the genome and reprogram its impact on breast cell biology. This review describes the current state-of-the-art on which factors manipulate the ERα cistrome and how this alters the response to both endogenous and exogenous hormonal stimuli, ultimately impacting BCa cell progression and response to commonly used therapeutic interventions. Novel insights in ERα cistrome dynamics may pave the way for better patient diagnostics and the development of novel therapeutic interventions, ultimately improving cancer care and patient outcome.
Collapse
|
46
|
Smith R, Liu M, Liby T, Bayani N, Bucher E, Chiotti K, Derrick D, Chauchereau A, Heiser L, Alumkal J, Feiler H, Carroll P, Korkola JE. Enzalutamide response in a panel of prostate cancer cell lines reveals a role for glucocorticoid receptor in enzalutamide resistant disease. Sci Rep 2020; 10:21750. [PMID: 33303959 PMCID: PMC7729982 DOI: 10.1038/s41598-020-78798-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 11/22/2020] [Indexed: 01/04/2023] Open
Abstract
Representative in vitro model systems that accurately model response to therapy and allow the identification of new targets are important for improving our treatment of prostate cancer. Here we describe molecular characterization and drug testing in a panel of 20 prostate cancer cell lines. The cell lines cluster into distinct subsets based on RNA expression, which is largely driven by functional Androgen Receptor (AR) expression. KLK3, the AR-responsive gene that encodes prostate specific antigen, shows the greatest variability in expression across the cell line panel. Other common prostate cancer associated genes such as TMPRSS2 and ERG show similar expression patterns. Copy number analysis demonstrates that many of the most commonly gained (including regions containing TERC and MYC) and lost regions (including regions containing TP53 and PTEN) that were identified in patient samples by the TCGA are mirrored in the prostate cancer cell lines. Assessment of response to the anti-androgen enzalutamide shows a distinct separation of responders and non-responders, predominantly related to status of wild-type AR. Surprisingly, several AR-null lines responded to enzalutamide. These AR-null, enzalutamide-responsive cells were characterized by high levels of expression of glucocorticoid receptor (GR) encoded by NR3C1. Treatment of these cells with the anti-GR agent mifepristone showed that they were more sensitive to this drug than enzalutamide, as were several of the enzalutamide non-responsive lines. This is consistent with several recent reports that suggest that GR expression is an alternative signaling mechanism that can bypass AR blockade. This study reinforces the utility of large cell line panels for the study of cancer and identifies several cell lines that represent ideal models to study AR-null cells that have upregulated GR to sustain growth.
Collapse
Affiliation(s)
- Rebecca Smith
- Department of Biomedical Engineering, Oregon Health & Science University, 2730 SW Moody Ave CLSB Rm 3N018, Portland, OR, 97201, USA
| | - Moqing Liu
- Department of Biomedical Engineering, Oregon Health & Science University, 2730 SW Moody Ave CLSB Rm 3N018, Portland, OR, 97201, USA
| | - Tiera Liby
- Department of Biomedical Engineering, Oregon Health & Science University, 2730 SW Moody Ave CLSB Rm 3N018, Portland, OR, 97201, USA
| | - Nora Bayani
- Life Sciences Division, Lawrence Berkeley National Laboratories, Berkeley, CA, 94720, USA
| | - Elmar Bucher
- Department of Biomedical Engineering, Oregon Health & Science University, 2730 SW Moody Ave CLSB Rm 3N018, Portland, OR, 97201, USA
| | - Kami Chiotti
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Daniel Derrick
- Department of Biomedical Engineering, Oregon Health & Science University, 2730 SW Moody Ave CLSB Rm 3N018, Portland, OR, 97201, USA
| | - Anne Chauchereau
- INSERM U981, Gustave Roussy Institute, Paris-Saclay University, 94800, Villejuif, France
| | - Laura Heiser
- Department of Biomedical Engineering, Oregon Health & Science University, 2730 SW Moody Ave CLSB Rm 3N018, Portland, OR, 97201, USA
- OHSU Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Joshi Alumkal
- Department of Medicine, Oregon Health & Science University, Portland, OR, 97201, USA
- Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Heidi Feiler
- Department of Biomedical Engineering, Oregon Health & Science University, 2730 SW Moody Ave CLSB Rm 3N018, Portland, OR, 97201, USA
- OHSU Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Peter Carroll
- Department of Urology, UCSF, San Francisco, CA, 94158, USA
| | - James E Korkola
- Department of Biomedical Engineering, Oregon Health & Science University, 2730 SW Moody Ave CLSB Rm 3N018, Portland, OR, 97201, USA.
- OHSU Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, OR, 97201, USA.
| |
Collapse
|
47
|
Costa-Pinheiro P, Heher A, Raymond MH, Jividen K, Shaw JJ, Paschal BM, Walker SJ, Fox TE, Kester M. Role of SPTSSB-Regulated de Novo Sphingolipid Synthesis in Prostate Cancer Depends on Androgen Receptor Signaling. iScience 2020; 23:101855. [PMID: 33313495 PMCID: PMC7721643 DOI: 10.1016/j.isci.2020.101855] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/23/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023] Open
Abstract
Anti-androgens are a common therapy in prostate cancer (PCa) targeting androgen receptor (AR) signaling. However, these therapies fail due to selection of highly aggressive AR-negative cancer cells that have no therapeutic options available. We demonstrate that elevating endogenous ceramide levels with administration of exogenous ceramide nanoliposomes (CNLs) was efficacious in AR-negative cell lines with limited efficacy in AR-positive cells. This effect is mediated through reduced de novo sphingolipid synthesis in AR-positive cells. We show that anti-androgens elevate de novo generation of sphingolipids via SPTSSB, a rate-limiting mediator of sphingolipid generation. Moreover, pharmacological inhibition of AR increases the efficacy of CNL in AR-positive cells through de novo synthesis, while SPTSSB knockdown limited CNL's efficacy in AR-negative cells. Alluding to clinical relevance, SPTSSB is upregulated in patients with advanced PCa after anti-androgens treatment. These findings emphasize the relevance of AR regulation upon sphingolipid metabolism and the potential of CNL as a PCa therapeutic. AR-negative PCa cells are more susceptible to CNL than AR-positive cells Combination of anti-androgens and CNL results in enhanced efficacy for AR-positive PCa AR negatively regulates the de novo synthesis of sphingolipids through SPTSSB SPTSSB is crucial for CNL effect in AR-negative PCa and is upregulated in neuroendocrine tumors
Collapse
Affiliation(s)
| | - Abigail Heher
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Michael H Raymond
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Kasey Jividen
- Center for Cell Signaling, University of Virginia, Charlottesville, VA 22903, USA
| | - Jeremy Jp Shaw
- Department of Pathology, University of Virginia, Charlottesville, VA 22903, USA
| | - Bryce M Paschal
- Center for Cell Signaling, University of Virginia, Charlottesville, VA 22903, USA.,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Susan J Walker
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903, USA
| | - Todd E Fox
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903, USA
| | - Mark Kester
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903, USA.,nanoSTAR Institute, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
48
|
Lounis MA, Péant B, Leclerc-Desaulniers K, Ganguli D, Daneault C, Ruiz M, Zoubeidi A, Mes-Masson AM, Saad F. Modulation of de Novo Lipogenesis Improves Response to Enzalutamide Treatment in Prostate Cancer. Cancers (Basel) 2020; 12:cancers12113339. [PMID: 33187317 PMCID: PMC7698241 DOI: 10.3390/cancers12113339] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Prostate cancer cells produce lipids via the activation of a specific pathway called fatty acid synthesis, also known as De novo lipogenesis. This pathway is essential for the survival and growth of most types of cancer cells, including prostate cancer. In our study, we showed that prostate cancer cells activate this lipid synthesis pathway to become more aggressive and develop resistance to commonly used therapeutic agents for advanced prostate cancer such as enzalutamide, an effective and commonly used androgen receptor (AR) targeted agent. Interestingly, by combining enzalutamide with a lipid synthesis pathway inhibitor, we were able to show that growth of prostate cancer tumors was more effectively reduced than with either agent alone. We also showed that this combination led to cell stress and death by changing the lipid content in the cell. These important findings could lead to new therapeutic strategies combining effective AR targeted therapies with lipid synthesis inhibitors for the treatment of advanced prostate cancer. Abstract De novo lipogenesis (DNL) is now considered as a hallmark of cancer. The overexpression of key enzymes of DNL is characteristic of both primary and advanced disease and may play an important role in resistance to therapies. Here, we showed that DNL is highly enhanced in castrate resistant prostate cancer (CRPC) cells compared to hormone sensitive and enzalutamide resistant cells. This observation suggests that this pathway plays an important role in the initiation of aggressive prostate cancer and in the development of enzalutamide resistance. Importantly, here we show that both prostate cancer cells sensitive and resistant to enzalutamide are dependent on DNL to proliferate. We next combined enzalutamide with an inhibitor of Stearoyl CoA Desaturase 1 (SCD1), an important enzyme in DNL, and observed significantly reduced tumor growth caused by the important change in tumoral lipid desaturation. Our findings suggest that the equilibrium between monounsaturated fatty acids and saturated fatty acids is essential in the establishment of the more aggressive prostate cancer phenotype and that the combination therapy induces a disruption of this equilibrium leading to an important decrease of cell proliferation. These findings provide new insights into the role of DNL in the progression of prostate cancer cells. The study also provides the rationale for the use of an inhibitor of SCD1 in combination with enzalutamide to improve response, delay enzalutamide resistance and improve disease free progression.
Collapse
Affiliation(s)
- Mohamed Amine Lounis
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada; (M.A.L.); (B.P.); (K.L.-D.); (A.-M.M.-M.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Benjamin Péant
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada; (M.A.L.); (B.P.); (K.L.-D.); (A.-M.M.-M.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Kim Leclerc-Desaulniers
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada; (M.A.L.); (B.P.); (K.L.-D.); (A.-M.M.-M.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Dwaipayan Ganguli
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada; (D.G.); (A.Z.)
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Caroline Daneault
- Institut de Cardiologie de Montréal, Montreal, QC H1T 1C8, Canada; (C.D.); (M.R.)
| | - Matthieu Ruiz
- Institut de Cardiologie de Montréal, Montreal, QC H1T 1C8, Canada; (C.D.); (M.R.)
- Département de Nutrition, Université de Montréal (UdeM), Montreal, QC H3C 3J7, Canada
| | - Amina Zoubeidi
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada; (D.G.); (A.Z.)
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Anne-Marie Mes-Masson
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada; (M.A.L.); (B.P.); (K.L.-D.); (A.-M.M.-M.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
- Département de Médecine, Université de Montréal (UdeM), Montreal, QC H3C 3J7, Canada
| | - Fred Saad
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada; (M.A.L.); (B.P.); (K.L.-D.); (A.-M.M.-M.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
- Département de Chirurgie, Université de Montréal (UdeM), Montreal, QC H3C 3J7, Canada
- Correspondence:
| |
Collapse
|
49
|
Tang S, Lian X, Jiang J, Cheng H, Guo J, Huang C, Meng H, Li X. Tumor Suppressive Maspin-Sensitized Prostate Cancer to Drug Treatment Through Negative Regulating Androgen Receptor Expression. Front Cell Dev Biol 2020; 8:573820. [PMID: 33195208 PMCID: PMC7649228 DOI: 10.3389/fcell.2020.573820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022] Open
Abstract
Overactivation of androgen receptor (AR)-mediated signal has been extensively implicated in prostate cancer (CaP) development, progression, and recurrence, which makes it an attractive therapeutic target. Meanwhile, as an endogenous inhibitor of histone deacetylase 1 (HDAC 1), tumor-suppressive mammary serine protease inhibitor (maspin) was reported to sensitize drug-induced apoptosis with a better therapeutic outcome in CaP, but the relationship between AR and maspin remains unclear. In the current study, treatment of 5'-Aza or MS-275/enzalutamide induced poly (ADP-ribose) polymerase (PARP) cleavage and p-H2A.X in CaP cells with an increase of maspin expression but a decrease of AR. Then, treatment with protease inhibitor MG132 did not rescue the above drug-induced loss of AR. In addition, modulation of maspin expression by gene recombinant or siRNA technology showed an inverse correlation between expression of maspin and AR, consequently affecting the AR-regulated downstream gene transcription (e.g., NKX3.1 and TMPRSS2). Bioinformatics analysis of the data extracted from the National Center for Biotechnology Information Gene Expression Omnibus (NCBI GEO) database also revealed an inverse correlation between low maspin expression and high AR level in advanced CaP. Furthermore, chromatin immunoprecipitation (ChIP) assay using anti-maspin antibody identified that a portion of AR promoter sequence was co-precipitated and presented in the immunoprecipitated complex. Finally, maspin-mediated repression of AR was induced by treatment of MS-275, which promoted enzalutamide treatment efficacy with decrease of prostate-specific antigen (PSA) expression in LNCaP and 22RV1 cells. Taken together, the data not only demonstrated maspin-mediated repression of AR to augment drug anti-tumor activity but also provided in-depth support for combination of HDAC inhibitors with AR antagonist in CaP therapy.
Collapse
Affiliation(s)
- Sijie Tang
- The AoYang Cancer Institute, Jiangsu University, Suzhou, China
| | - Xueqi Lian
- The AoYang Cancer Institute, Jiangsu University, Suzhou, China
| | - Jiajia Jiang
- The AoYang Cancer Institute, Jiangsu University, Suzhou, China
| | - Huiying Cheng
- The AoYang Cancer Institute, Jiangsu University, Suzhou, China
| | - Jiaqian Guo
- The AoYang Cancer Institute, Jiangsu University, Suzhou, China
| | - Can Huang
- The AoYang Cancer Institute, Jiangsu University, Suzhou, China
| | - Hong Meng
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, United States
| | - Xiaohua Li
- The AoYang Cancer Institute, Jiangsu University, Suzhou, China
- The Laboratory of Clinical Genomics, Hefei KingMed Diagnostics Laboratory, Hefei, China
- National Center for Gene Testing Technology Application & Demonstration (Anhui), Hefei, China
| |
Collapse
|
50
|
Jiang SH, Zhang XX, Hu LP, Wang X, Li Q, Zhang XL, Li J, Gu JR, Zhang ZG. Systemic Regulation of Cancer Development by Neuro-Endocrine-Immune Signaling Network at Multiple Levels. Front Cell Dev Biol 2020; 8:586757. [PMID: 33117814 PMCID: PMC7561376 DOI: 10.3389/fcell.2020.586757] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022] Open
Abstract
The overarching view of current tumor therapies simplifies cancer to a cell-biology problem in which neoplasms are caused solely by malignant cells and the exploration of carcinogenesis and tumor progression largely focuses on somatic mutations and other genetic abnormalities of cancer cells. The limited therapeutic response indicates that cancer is driven not only by endogenous oncogenic factors and reciprocal interactions within the tumor microenvironment, but also by complex systemic processes. Homeostasis is the fundamental premise of health, and is maintained by systemic regulation of neuro-endocrine-immune axis. Cancer is also a systemic disease that manifested by dysfunction of the nervous, endocrine, and immune systems. Multiple axes of regulation exist in cancer, including central-, organ-, and microenvironment-level manipulation. At each specific regulatory level, the tridirectional communication among the nervous, endocrine, and immune factors transmit flexible signaling to induce proliferation, invasion, reprogrammed metabolism, therapeutic resistance, and other malignant phenotypes of cancer cells, resulting in the extremely poor prognosis of this lethal disease. Understanding this coordinated signaling network will enable the development of new approaches for cancer treatment via behavioral and pharmacological interventions.
Collapse
Affiliation(s)
- Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Xin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Ren Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|