1
|
Gülyüz S, Sessevmez M, Ukuser G, Khalily MP, Tiryaki S, Sipahioglu T, Birgül K, Ömeroğlu İ, Özçubukçu S, Telci D, Küçükgüzel ŞG, Durmuş M, Cevher E, Yılmaz Ö. A Novel PEtOx-Based Nanogel Targeting Prostate Cancer Cells for Drug Delivery. Macromol Biosci 2024; 24:e2300324. [PMID: 37827519 DOI: 10.1002/mabi.202300324] [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: 07/12/2023] [Revised: 09/28/2023] [Indexed: 10/14/2023]
Abstract
This study focuses on creating a specialized nanogel for targeted drug delivery in cancer treatment, specifically targeting prostate cancer. This nanogel (referred to as SGK 636/Peptide 563/PEtOx nanogel) is created using hydrophilic poly(2-ethyl-2-oxazoline) (PEtOx) through a combination of living/cationic ring-opening polymerization (CROP) and alkyne-azide cycloaddition (CuAAC) "click" chemical reactions. A fluorescent probe (BODIPY) is also conjugated with the nanogel to monitor drug delivery. The characterizations through 1 H-NMR, and FT-IR, SEM, TEM, and DLS confirm the successful production of uniform, and spherical nanogels with controllable sizes (100 to 296 nm) and stability in physiological conditions. The biocompatibility of nanogels is evaluated using MTT cytotoxicity assays, revealing dose-dependent cytotoxicity. Drug-loaded nanogels exhibited significantly higher cytotoxicity against cancer cells in vitro compared to drug-free nanogels. Targeting efficiency is examined using both peptide-conjugated and peptide-free nanogels, with the intracellular uptake of peptide 563-conjugated nanogels by tumor cells being 60-fold higher than that of nanogels without the peptide. The findings suggest that the prepared nanogel holds great potential for various drug delivery applications due to its ease of synthesis, tunable functionality, non-toxicity, and enhanced intracellular uptake in the tumor region.
Collapse
Affiliation(s)
- Sevgi Gülyüz
- Material Technologies, Marmara Research Center, TUBITAK, Gebze, Kocaeli, 41470, Turkey
| | - Melike Sessevmez
- Department of Pharmaceutical Technology, Istanbul University, Istanbul, 34116, Turkey
| | - Gokcen Ukuser
- Material Technologies, Marmara Research Center, TUBITAK, Gebze, Kocaeli, 41470, Turkey
| | - Melek Parlak Khalily
- Department of Basic Science and Health, Cannabis Research Institute, Yozgat Bozok University, Yozgat, 66100, Turkey
| | - Selen Tiryaki
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, 34755, Turkey
| | - Tarik Sipahioglu
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, 34755, Turkey
| | - Kaan Birgül
- Department of Pharmaceutical Chemistry, School of Pharmacy, Bahçeşehir University, Beşiktaş, Istanbul, 34353, Turkey
| | - İpek Ömeroğlu
- Department of Chemistry, Gebze Technical University, Gebze, Kocaeli, 41400, Turkey
| | - Salih Özçubukçu
- Department of Chemistry, Middle East Technical University, Ankara, 06800, Turkey
| | - Dilek Telci
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, 34755, Turkey
| | - Ş Güniz Küçükgüzel
- Department of Pharmaceutical Chemistry, Fenerbahçe University, Ataşehir, Istanbul, 34758, Turkey
| | - Mahmut Durmuş
- Department of Chemistry, Gebze Technical University, Gebze, Kocaeli, 41400, Turkey
| | - Erdal Cevher
- Department of Pharmaceutical Technology, Istanbul University, Istanbul, 34116, Turkey
| | - Özgür Yılmaz
- Material Technologies, Marmara Research Center, TUBITAK, Gebze, Kocaeli, 41470, Turkey
| |
Collapse
|
2
|
Jin H, Zhang C, Zwahlen M, von Feilitzen K, Karlsson M, Shi M, Yuan M, Song X, Li X, Yang H, Turkez H, Fagerberg L, Uhlén M, Mardinoglu A. Systematic transcriptional analysis of human cell lines for gene expression landscape and tumor representation. Nat Commun 2023; 14:5417. [PMID: 37669926 PMCID: PMC10480497 DOI: 10.1038/s41467-023-41132-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 08/24/2023] [Indexed: 09/07/2023] Open
Abstract
Cell lines are valuable resources as model for human biology and translational medicine. It is thus important to explore the concordance between the expression in various cell lines vis-à-vis human native and disease tissues. In this study, we investigate the expression of all human protein-coding genes in more than 1,000 human cell lines representing 27 cancer types by a genome-wide transcriptomics analysis. The cell line gene expression is compared with the corresponding profiles in various tissues, organs, single-cell types and cancers. Here, we present the expression for each cell line and give guidance for the most appropriate cell line for a given experimental study. In addition, we explore the cancer-related pathway and cytokine activity of the cell lines to aid human biology studies and drug development projects. All data are presented in an open access cell line section of the Human Protein Atlas to facilitate the exploration of all human protein-coding genes across these cell lines.
Collapse
Affiliation(s)
- Han Jin
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Martin Zwahlen
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Kalle von Feilitzen
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Max Karlsson
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mengnan Shi
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Meng Yuan
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Xiya Song
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Xiangyu Li
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Hong Yang
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Linn Fagerberg
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden.
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden.
| | - Adil Mardinoglu
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden.
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK.
| |
Collapse
|
3
|
18F-DCFPyL (PSMA) PET as a radiotherapy response assessment tool in metastatic prostate cancer. Clin Transl Radiat Oncol 2023; 39:100583. [PMID: 36713978 PMCID: PMC9879766 DOI: 10.1016/j.ctro.2023.100583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/19/2023] Open
Abstract
Background Prostate Specific Membrane Antigen (PSMA) - positron emission tomography (PET) guides metastasis-directed radiotherapy (MDRT) in prostate cancer (PrCa). However, its value as a treatment response assessment tool after MDRT remains unclear. Importantly, there is limited understanding of the potential of radiotherapy (RT) to alter PSMA gene (folate hydrolase 1; FOLH1) expression. Methodology We reviewed a series of 11 men with oligo-metastatic PrCa (25 metastasis sites) treated with MDRT before re-staging with 18F-DCFPyL (PSMA) PET upon secondary recurrence. Acute effects of RT on PSMA protein and mRNA levels were examined with qPCR and immunoblotting in human wild-type androgen-sensitive (LNCap), castrate-resistant (22RV1) and castrate-resistant neuroendocrine (PC3 and DU145) PrCa cell lines. Xenograft tumors were analyzed with immunohistochemistry. Further, we examined PSMA expression in untreated and irradiated radio-resistant (RR) 22RV1 (22RV1-RR) and DU145 (DU145-RR) cells and xenografts selected for survival after high-dose RT. Results The majority of MDRT-treated lesions showed lack of PSMA-PET/CT avidity, suggesting treatment response even after low biological effective dose (BED) MDRT. We observed similar high degree of heterogeneity of PSMA expression in both human specimens and in xenograft tumors. PSMA was highly expressed in LNCap and 22RV1 cells and tumors but not in the neuroendocrine PC3 and DU145 models. Single fraction RT caused detectable reduction in PSMA protein but not in mRNA levels in LNCap cells and did not significantly alter PSMA protein or mRNA levels in tissue culture or xenografts of the other cell lines. However, radio-resistant 22RV1-RR cells and tumors demonstrated marked decrease of PSMA transcript and protein expression over their parental counterparts. Conclusions PSMA-PET may be a promising tool to assess RT response in oligo-metastatic PrCa. However, future systematic investigation of this concept should recognize the high degree of heterogeneity of PSMA expression within prostate tumors and the risk for loss of PSMA expression in tumor surviving curative courses of RT.
Collapse
Key Words
- ADT, Androgen Deprivation Therapy
- AMACR, Alpha-Methylacyl-CoA Racemase
- ARAT, Androgen Receptor Axis-Targeted
- BED, Biological Effective Dose
- CRPC, Castration Resistant Prostate Cancer
- FOLH1 expression
- FOLH1, Folate Hydrolase 1
- H&E, Hematoxylin and Eosin
- H-Score, Histologic Score
- HSPC, Hormone Sensitive Prostate Cancer
- IHC, Immunohistochemistry
- Immunohistochemistry
- LHRH, Luteinizing Hormone Releasing Hormone
- MDRT, Metastasis Directed Radiotherapy
- NH, Hormone Naïve
- P-H3, Phosphorylated Histone-H3
- PET, Positron Emission Tomography
- PSA, Prostate Specific Antigen
- PSMA, Prostate Specific Membrane Antigen
- PSMA-PET
- PrCa, Prostate Cancer
- RP, Radical Prostatectomy
- RT, Radiation Therapy
- Radio-resistance
- Rec, Recurrence
- SUV, Standardized Uptake Value
- Tumor heterogeneity
- mCRPC, Metastatic Castration Resistant Prostate Cancer
- mRNA, Messenger Ribonucleic Acid
- qPCR
- qPCR, Quantitative Polymerase Chain Reaction
Collapse
|
4
|
Filon MJ, Gillette AA, Yang B, Khemees TA, Skala MC, Jarrard DF. Prostate cancer cells demonstrate unique metabolism and substrate adaptability acutely after androgen deprivation therapy. Prostate 2022; 82:1547-1557. [PMID: 35980831 PMCID: PMC9804183 DOI: 10.1002/pros.24428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/04/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Androgen deprivation therapy (ADT) has been the standard of care for advanced hormone-sensitive prostate cancer (PC), yet tumors invariably develop resistance resulting in castrate-resistant PC. The acute response of cancer cells to ADT includes apoptosis and cell death, but a large fraction remains arrested but viable. In this study, we focused on intensively characterizing the early metabolic changes that result after ADT to define potential metabolic targets for treatment. METHODS A combination of mass spectrometry, optical metabolic imaging which noninvasively measures drug responses in cells, oxygen consumption rate, and protein expression analysis was used to characterize and block metabolic pathways over several days in multiple PC cell lines with variable hormone response status including ADT sensitive lines LNCaP and VCaP, and resistant C4-2 and DU145. RESULTS Mass spectrometry analysis of LNCaP pre- and postexposure to ADT revealed an abundance of glycolytic intermediates after ADT. In LNCaP and VCaP, a reduction in the optical redox ratio [NAD(P)H/FAD], extracellular acidification rate, and a downregulation of key regulatory enzymes for fatty acid and glutamine utilization was acutely observed after ADT. Screening several metabolic inhibitors revealed that blocking fatty acid oxidation and synthesis reversed this stress response in the optical redox ratio seen with ADT alone in LNCaP and VCaP. In contrast, both cell lines demonstrated increased sensitivity to the glycolytic inhibitor 2-Deoxy- d-glucose(2-DG) and maintained sensitivity to electron transport chain inhibitor Malonate after ADT exposure. ADT followed by 2-DG results in synergistic cell death, a result not seen with simultaneous administration. CONCLUSIONS Hormone-sensitive PC cells displayed altered metabolic profiles early after ADT including an overall depression in energy metabolism, induction of a quiescent/senescent phenotype, and sensitivity to selected metabolic inhibitors. Glycolytic blocking agents (e.g., 2-DG) as a sequential treatment after ADT may be promising.
Collapse
Affiliation(s)
- Mikolaj J. Filon
- Department of Urology, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
| | - Amani A. Gillette
- Department of Biomedical EngineeringUniversity of WisconsinMadisonWisconsinUSA
- Morgridge Institute for ResearchMadisonWisconsinUSA
| | - Bing Yang
- Department of Urology, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
| | - Tariq A. Khemees
- Department of Urology, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
| | - Melissa C. Skala
- Department of Biomedical EngineeringUniversity of WisconsinMadisonWisconsinUSA
- Morgridge Institute for ResearchMadisonWisconsinUSA
- Carbone Comprehensive Cancer CenterUniversity of WisconsinMadisonWisconsinUSA
| | - David F. Jarrard
- Department of Urology, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
- Carbone Comprehensive Cancer CenterUniversity of WisconsinMadisonWisconsinUSA
- Molecular and Environmental Toxicology ProgramUniversity of WisconsinMadisonWisconsinUSA
| |
Collapse
|
5
|
Barysevich MV, Laktsevich-Iskryk MV, Scherbakov AM, Salnikova DI, Andreeva OE, Sorokin DV, Shchegolev YY, Hurski AL, Zhabinskii VN, Khripach VA. Synthesis and biological activity of 21,22-cyclosteroids and their derivatives. Steroids 2022; 188:109135. [PMID: 36336105 DOI: 10.1016/j.steroids.2022.109135] [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: 08/03/2021] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Synthesis of 21,22-cyclosteroids has been achieved starting from pregnenolone acetate. The key transformation was the Kulinkovich reaction of 17-vinyl steroids with esters. The resulting cyclopropanols were further subjected to three-membered ring-opening under various conditions including to base-, palladium or visible light-promoted isomerization and cross-coupling reaction. A number of steroidal Δ2-6-ketones and 3β-hydroxy-Δ5-enes with functional groups at C-21 - C-23 have been synthesized via the 21,22-cyclosteroids. The antiproliferative and antihormonal activity of the obtained compounds on the cell lines of prostate (22Rv1) and breast (MCF-7) cancer was studied. The androgen receptor activity was assessed by reporter assay when the expression of signalling proteins was evaluated by immunoblotting. (20S,22R)-22-Acetoxy-21,22-cyclo-5α-cholest-5-ene with the moderate antiandrogenic potency revealed IC50 values of 18.4 ± 1.2 and 14.6 ± 1.4 µM against MCF-7 and 22Rv1 cells, respectively, and its effects on the expression of AR-V7, cyclin D1 and BCL2 were explored.
Collapse
Affiliation(s)
- Maryia V Barysevich
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich st, 5/2, 220141 Minsk, Belarus
| | - Marharyta V Laktsevich-Iskryk
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich st, 5/2, 220141 Minsk, Belarus
| | - Alexander M Scherbakov
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Ministry of Health of Russia, Kashirskoe shosse, 24, 115522 Moscow, Russia
| | - Diana I Salnikova
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Ministry of Health of Russia, Kashirskoe shosse, 24, 115522 Moscow, Russia
| | - Olga E Andreeva
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Ministry of Health of Russia, Kashirskoe shosse, 24, 115522 Moscow, Russia
| | - Danila V Sorokin
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Ministry of Health of Russia, Kashirskoe shosse, 24, 115522 Moscow, Russia
| | - Yuri Y Shchegolev
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Ministry of Health of Russia, Kashirskoe shosse, 24, 115522 Moscow, Russia
| | - Alaksiej L Hurski
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich st, 5/2, 220141 Minsk, Belarus
| | - Vladimir N Zhabinskii
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich st, 5/2, 220141 Minsk, Belarus.
| | - Vladimir A Khripach
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich st, 5/2, 220141 Minsk, Belarus
| |
Collapse
|
6
|
Moraes de Farias K, Rosa-Ribeiro R, Souza EE, Kobarg J, Banwell MG, de Brito Vieira Neto J, Leyenne Alves Sales S, Roberto Ribeiro Costa P, Cavalcante Dos Santos R, Vilaça Gaspar F, Gomes Barreto Junior A, da Conceição Ferreira Oliveira M, Odorico de Moraes M, Libardi M Furtado C, Carvalho HF, Pessoa C. The Isoflavanoid (+)-PTC Regulates Cell-Cycle Progression and Mitotic Spindle Assembly in a Prostate Cancer Cell Line. Chem Biodivers 2022; 19:e202200102. [PMID: 35362194 DOI: 10.1002/cbdv.202200102] [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: 01/31/2022] [Accepted: 03/31/2022] [Indexed: 12/24/2022]
Abstract
Prostate cancer is the second most common malignancy in men and the development of effective therapeutic strategies remains challenging when more advanced, androgen-independent or insensitive forms are involved. Accordingly, we have evaluated, using flow cytometry, confocal microscopy and image analysis, the anti-proliferative effects of (+)-2,3,9-trimethoxypterocarpan [(+)-PTC, 1] on relevant human prostate cancer cells as well as its capacity to control mitosis within them. In particular, the studies reported herein reveal that (+)-PTC exerts anti-proliferative activity against the PC-3 cell lines by regulating cell-cycle progression with mitosis being arrested in the prophase or prometaphase. Furthermore, it emerges that treatment of the target cells with this compound results in the formation of monopolar spindles, disorganized centrosomes and extensively disrupted γ-tubulin distributions while centriole replication remains unaffected. Such effects suggest (+)-PTC should be considered as a possible therapy for androgen-insensitive/independent prostate cancer.
Collapse
Affiliation(s)
- Kaio Moraes de Farias
- Programa de Pós-Graduação em Biotecnologia - RENORBIO - Rede Nordeste de Biotecnologia, Federal University of Ceará - UFC, 60020-181, Fortaleza, CE, Brazil.,Núcleo de Pesquisa e Desenvolvimento de Medicamentos - NPDM, Federal University of Ceará - UFC, Fortaleza, CE 60430-275, Brazil
| | - Rafaela Rosa-Ribeiro
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Campinas, 13083-970, SP, Brazil
| | - Edmarcia E Souza
- Faculdade de Ciências Farmacêuticas, State University of Campinas, Campinas, 13083-859, SP, Brazil
| | - Jörg Kobarg
- Faculdade de Ciências Farmacêuticas, State University of Campinas, Campinas, 13083-859, SP, Brazil
| | - Martin G Banwell
- Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou, 510632, China
| | - José de Brito Vieira Neto
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos - NPDM, Federal University of Ceará - UFC, Fortaleza, CE 60430-275, Brazil
| | - Sarah Leyenne Alves Sales
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos - NPDM, Federal University of Ceará - UFC, Fortaleza, CE 60430-275, Brazil
| | - Paulo Roberto Ribeiro Costa
- Laboratório de Química Bioorgânica (LQB), Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-909, RJ, Brazil
| | - Rafael Cavalcante Dos Santos
- Engenharia de Processos Químicos e Bioquímicos (EPQB), Escola de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-909, RJ, Brazil
| | - Francisco Vilaça Gaspar
- Laboratório de Química Bioorgânica (LQB), Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-909, RJ, Brazil
| | - Amaro Gomes Barreto Junior
- Engenharia de Processos Químicos e Bioquímicos (EPQB), Escola de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-909, RJ, Brazil
| | | | - Manoel Odorico de Moraes
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos - NPDM, Federal University of Ceará - UFC, Fortaleza, CE 60430-275, Brazil
| | - Cristiana Libardi M Furtado
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos - NPDM, Federal University of Ceará - UFC, Fortaleza, CE 60430-275, Brazil.,Experimental Biology Center - NUBEX, University of Fortaleza, UNIFOR, Fortaleza, CE 60811-905, Brazil
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Campinas, 13083-970, SP, Brazil
| | - Claudia Pessoa
- Programa de Pós-Graduação em Biotecnologia - RENORBIO - Rede Nordeste de Biotecnologia, Federal University of Ceará - UFC, 60020-181, Fortaleza, CE, Brazil.,Núcleo de Pesquisa e Desenvolvimento de Medicamentos - NPDM, Federal University of Ceará - UFC, Fortaleza, CE 60430-275, Brazil
| |
Collapse
|
7
|
Dyshlovoy SA, Pelageev DN, Jakob LS, Borisova KL, Hauschild J, Busenbender T, Kaune M, Khmelevskaya EA, Graefen M, Bokemeyer C, Anufriev VP, von Amsberg G. Activity of New Synthetic (2-Chloroethylthio)-1,4-naphthoquinones in Prostate Cancer Cells. Pharmaceuticals (Basel) 2021; 14:ph14100949. [PMID: 34681173 PMCID: PMC8540265 DOI: 10.3390/ph14100949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/14/2022] Open
Abstract
Development of resistance to currently available standard therapies in advanced prostate cancer (PCa) emphasizes the need for novel therapeutic options. Here, we report the synthesis of new hybrid molecules consisting of 2-chloroethylthio and 1,4-naphthoquinone pharmacophores and describe their activity in PCa. In screening analyses, the introduction of one 2-chloroethylthio group improved the anticancer properties of 1,4-naphthoquinones, whereas the introduction of a second 2-chloroethylthio moiety rather decreased activity. Two most promising of the synthesized compounds, 30 and 32, were highly active in different human PCa cell lines harboring varying resistance profiles at nanomolar concentrations. The generated data suggest that the compounds are capable of mitochondria targeting, cytotoxic ROS induction, and DNA damage, which resulted in apoptosis presumably executed in a caspase-dependent manner. The substances synergized with the clinically approved PARP inhibitor olaparib and resensitized AR-V7-expressing PCa cells to antiandrogen enzalutamide, as well as to a combination of enzalutamide and an AKT inhibitor. This was at least in part exerted via down-regulation of AR-V7 expression and inhibition of AR signaling. Mild antagonism was observed in combination with platinum- or taxane-based chemotherapy, which was putatively related to treatment-induced activation of p38, JNK1/2, ERK1/2, MEK1/2, and AKT, functioning as potential pro-survival factors. Thus, the synthesized (2-chloroethylthio)-1,4-naphthoquinone derivatives exhibit promising anticancer properties in vitro, suggesting their further development as potential therapeutics for the treatment of castration-resistant PCa.
Collapse
Affiliation(s)
- Sergey A. Dyshlovoy
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany; (L.S.J.); (J.H.); (T.B.); (M.K.); (C.B.); (G.v.A.)
- Martini-Klinik, Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany;
- School of Natural Sciences, FEFU Campus, Far Eastern Federal University, Ajax Bay 10, Russky Island, 690922 Vladivostok, Russia; (D.N.P.); (E.A.K.)
- Correspondence: or ; Tel.: +49-40-7410-51950
| | - Dmitry N. Pelageev
- School of Natural Sciences, FEFU Campus, Far Eastern Federal University, Ajax Bay 10, Russky Island, 690922 Vladivostok, Russia; (D.N.P.); (E.A.K.)
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (K.L.B.); (V.P.A.)
| | - Lea S. Jakob
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany; (L.S.J.); (J.H.); (T.B.); (M.K.); (C.B.); (G.v.A.)
| | - Ksenia L. Borisova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (K.L.B.); (V.P.A.)
| | - Jessica Hauschild
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany; (L.S.J.); (J.H.); (T.B.); (M.K.); (C.B.); (G.v.A.)
| | - Tobias Busenbender
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany; (L.S.J.); (J.H.); (T.B.); (M.K.); (C.B.); (G.v.A.)
| | - Moritz Kaune
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany; (L.S.J.); (J.H.); (T.B.); (M.K.); (C.B.); (G.v.A.)
| | - Ekaterina A. Khmelevskaya
- School of Natural Sciences, FEFU Campus, Far Eastern Federal University, Ajax Bay 10, Russky Island, 690922 Vladivostok, Russia; (D.N.P.); (E.A.K.)
| | - Markus Graefen
- Martini-Klinik, Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany;
| | - Carsten Bokemeyer
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany; (L.S.J.); (J.H.); (T.B.); (M.K.); (C.B.); (G.v.A.)
| | - Victor Ph. Anufriev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia; (K.L.B.); (V.P.A.)
| | - Gunhild von Amsberg
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany; (L.S.J.); (J.H.); (T.B.); (M.K.); (C.B.); (G.v.A.)
- Martini-Klinik, Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany;
| |
Collapse
|
8
|
Interplay of Epidermal Growth Factor Receptor and Signal Transducer and Activator of Transcription 3 in Prostate Cancer: Beyond Androgen Receptor Transactivation. Cancers (Basel) 2021; 13:cancers13143452. [PMID: 34298665 PMCID: PMC8307975 DOI: 10.3390/cancers13143452] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 01/16/2023] Open
Abstract
Prostate cancer (PCa) is one of the most common cancers in the world and causes thousands of deaths every year. Conventional therapy for PCa includes surgery and androgen deprivation therapy (ADT). However, about 10-20% of all PCa cases relapse; there is also the further development of castration resistant adenocarcinoma (CRPC-Adeno) or neuroendocrine (NE) PCa (CRPC-NE). Due to their androgen-insensitive properties, both CRPC-Adeno and CRPC-NE have limited therapeutic options. Accordingly, this study reveals the inductive mechanisms of CRPC (for both CRPC-Adeno and CRPC-NE) and fulfils an urgent need for the treatment of PCa patients. Although previous studies have illustrated the emerging roles of epidermal growth factor receptors (EGFR), signal transducer, and activator of transcription 3 (STAT3) signaling in the development of CRPC, the regulatory mechanisms of this interaction between EGFR and STAT3 is still unclear. Our recent studies have shown that crosstalk between EGFR and STAT3 is critical for NE differentiation of PCa. In this review, we have collected recent findings with regard to the involvement of EGFR and STAT3 in malignancy progression and discussed their interactions during the development of therapeutic resistance for PCa.
Collapse
|
9
|
Su CY, Huang GC, Chang YC, Chen YJ, Fang HW. Analyzing the Expression of Biomarkers in Prostate Cancer Cell Lines. In Vivo 2021; 35:1545-1548. [PMID: 33910833 DOI: 10.21873/invivo.12408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND/AIM CD44 and CD133 have been implicated as biomarkers of cancer cells and their expression could be analyzed to identify circulating tumor cells. Although CD44 and CD133 have been shown to be expressed in prostate cancer cells, a differential expression pattern has been reported depending on the tumor stage and cell line examined. We further investigated CD44 and CD133 expression in different prostate cancer cell lines to confirm whether their expression is distinguishable among patients with various tumor stages. MATERIALS AND METHODS CWR22Rv1, PC3, LNCaP, and DU145 cell lines were cultured and the cell morphology was observed for three days. The single expression of CD44 or CD133 and their combined expression were analyzed by flow cytometry. RESULTS We report that the single expression of CD133 was less than 5% in all cell lines examined here. PC3 and DU145 cells displayed a high expression of CD44 (>93%), while the expression of CD44 was less than 4% in CWR22Rv1 and LNCaP cells. CWR22Rv1 was the only cell line that demonstrated a high co-expression of both CD44 and CD133. CONCLUSION Both single and combined expression of CD44 and CD133 should be considered when validating the detection of prostate cancer cells in circulating tumor cells.
Collapse
Affiliation(s)
- Chen-Ying Su
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan, R.O.C
| | - Gwo-Che Huang
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei, Taiwan, R.O.C
| | - You-Cheng Chang
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan, R.O.C
| | - Yu-Jen Chen
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei, Taiwan, R.O.C
| | - Hsu-Wei Fang
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan, R.O.C.; .,Institute of Biomedical Engineering and Nanomedicine, National Health Research Institute, Zhunan, Taiwan, R.O.C
| |
Collapse
|
10
|
Sodium bicarbonate transporter NBCe1 regulates proliferation and viability of human prostate cancer cells LNCaP and PC3. Oncol Rep 2021; 46:129. [PMID: 34013380 PMCID: PMC8144930 DOI: 10.3892/or.2021.8080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 03/29/2021] [Indexed: 12/16/2022] Open
Abstract
Studies on cultured cancer cells or cell lines have revealed multiple acid extrusion mechanisms and their involvement in cancer cell growth and progression. In the present study, the role of the sodium bicarbonate transporters (NBCs) in prostate cancer cell proliferation and viability was examined. qPCR revealed heterogeneous expression of five NBC isoforms in human prostate cancer cell lines LNCaP, PC3, 22RV1, C4-2, DU145, and the prostate cell line RWPE-1. In fluorescence pH measurement of LNCaP cells, which predominantly express NBCe1, Na+ and HCO3–-mediated acid extrusion was identified by bath ion replacement and sensitivity to the NBC inhibitor S0859. NBCe1 knockdown using siRNA oligonucleotides decreased the number of viable cells, and pharmacological inhibition with S0859 (50 µM) resulted in a similar decrease. NBCe1 knockdown and inhibition also increased cell death, but this effect was small and slow. In PC3 cells, which express all NBC isoforms, NBCe1 knockdown decreased viable cell number and increased cell death. The effects of NBCe1 knockdown were comparable to those by S0859, indicating that NBCe1 among NBCs primarily contributes to PC3 cell proliferation and viability. S0859 inhibition also decreased the formation of cell spheres in 3D cultures. Immunohistochemistry of human prostate cancer tissue microarrays revealed NBCe1 localization to the glandular epithelial cells in prostate tissue and robust expression in acinar and duct adenocarcinoma. In conclusion, our study demonstrates that NBCe1 regulates acid extrusion in prostate cancer cells and inhibiting or abolishing this transporter decreases cancer cell proliferation.
Collapse
|
11
|
Demirci S, Hayal TB, Kıratlı B, Şişli HB, Demirci S, Şahin F, Doğan A. Design and synthesis of phenylpiperazine derivatives as potent anticancer agents for prostate cancer. Chem Biol Drug Des 2019; 94:1584-1595. [PMID: 31148379 DOI: 10.1111/cbdd.13575] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/23/2019] [Accepted: 05/26/2019] [Indexed: 12/19/2022]
Abstract
Novel thiourea (5a, 5b) and thiazolidinone derivatives (6a, 6b) were synthesized by hybridizing molecules starting from the compound 6-(4-phenylpiperazin-1-yl)pyridin-3-amine (4) which is known to show anticancer activity. The synthesis of the leading compound was carried out by using 1-(5-nitropyridin-2-yl)-4-phenylpiperazine (3) which was obtained by a novel method of the reaction of 2-chloro-5-nitropyridine (1) and N-phenylpiperazine (2). The structures of the compounds were confirmed using FTIR, 1 H NMR, 13 C NMR, HRMS spectroscopic methods and elemental analysis. The organic molecules were tested for their anticancer activities against prostate cancer (PC) cell lines: DU 145, PC-3 and LNCaP. As the compound 5a exerted the highest cytotoxic activity, IC50 concentrations of compound 5a were further investigated in terms of morphology, colony-forming ability, RNA expression, fragmented DNA and cell cycle distributions of PC cell lines. Overall data revealed that compound 5a treatment induces apoptosis and DNA fragmentation in PC cell lines and inhibits cell cycle progression resulting in the accumulation of cells in either the G1 or the S phases.
Collapse
Affiliation(s)
- Serpil Demirci
- Department of Medical Services and Techniques, Vocational High School of Health Services, Giresun University, Giresun, Turkey
| | - Taha Bartu Hayal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Binnur Kıratlı
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Hatice Burcu Şişli
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Selami Demirci
- Cellular and Molecular Therapeutics, Sickle Cell Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Ayşegül Doğan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
12
|
Chen X, Shi H, Bi X, Li Y, Huang Z. Targeting the deubiquitinase STAMBPL1 triggers apoptosis in prostate cancer cells by promoting XIAP degradation. Cancer Lett 2019; 456:49-58. [PMID: 31004702 DOI: 10.1016/j.canlet.2019.04.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/14/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022]
Abstract
The zinc metalloprotease STAM-binding protein-like 1 (STAMBPL1) has been identified as a deubiquitinase by specifically cleaving Lys-63-linked polyubiquitin chains, but its cellular function remains unclear. Here we described the potential role of STAMBPL1 in suppression of the intrinsic apoptosis. We observed substantially high amounts of STAMBPL1 proteins in androgen-independent prostate cancer PC3 and DU145 cell lines. STAMBPL1 RNAi depletion triggered caspase-3/-7-dependent apoptosis in PC3 and DU145 cells. STAMBPL1 knockdown-induced apoptosis was accompanied by accumulation of cellular ROS and a decrease in endogenous caspase inhibitor XIAP protein content. Treatment cells with antioxidant NAC delayed STAMBPL1 silencing-induced apoptosis, whereas ectopic expression of XIAP almost completely abrogated apoptosis. We further elucidated that STAMBPL1 knockdown diverted XIAP protein to lysosomal degradation pathway. Taken together, these studies show that STAMBPL1 depletion induces apoptosis by promoting XIAP lysosomal degradation, and suggest that targeting deubiquitinase STAMBPL1 might offer promising therapeutic strategy for prostate cancer.
Collapse
Affiliation(s)
- Xi Chen
- Department of Urology, National Cancer Center, National Clinical Research Center For Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hongzhe Shi
- Department of Urology, National Cancer Center, National Clinical Research Center For Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xingang Bi
- Department of Urology, National Cancer Center, National Clinical Research Center For Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yajian Li
- Department of Urology, National Cancer Center, National Clinical Research Center For Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhenhua Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
13
|
Miah S, Tharakan T, Gallagher KA, Shah TT, Winkler M, Jayasena CN, Ahmed HU, Minhas S. The effects of testosterone replacement therapy on the prostate: a clinical perspective. F1000Res 2019; 8:F1000 Faculty Rev-217. [PMID: 30828436 PMCID: PMC6392157 DOI: 10.12688/f1000research.16497.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2019] [Indexed: 12/28/2022] Open
Abstract
Male hypogonadism is a clinical syndrome characterized by low testosterone and symptoms of androgen deficiency. Prostate cancer remains a significant health burden and cause of male mortality worldwide. The use of testosterone replacement therapy drugs is rising year-on-year for the treatment of androgen deficiency and has reached global proportions. As clinicians, we must be well versed and provide appropriate counseling for men prior to the commencement of testosterone replacement therapy. This review summarizes the current clinical and basic science evidence in relation to this commonly encountered clinical scenario. There is gathering evidence that suggests, from an oncological perspective, that it is safe to commence testosterone replacement therapy for men who have a combination of biochemically confirmed androgen deficiency and who have either had definitive treatment of their prostate cancer or no previous history of this disease. However, patients must be made aware and cautioned that there is a distinct lack of level 1 evidence. Calls for such studies have been made throughout the urological and andrological community to provide a definitive answer. For those with a diagnosis of prostate cancer that remains untreated, there is a sparsity of evidence and therefore clinicians are "pushing the limits" of safety when considering the commencement of testosterone replacement therapy.
Collapse
Affiliation(s)
- Saiful Miah
- Department of Urology, Imperial College Healthcare NHS Trust, Charing Cross Hospital, Fulham Palace Road, London, W6 8RF, UK
- Division of Surgery and Interventional Science, University College London Medical School, 21 University Street, London, WC1E 6AU, UK
| | - Tharu Tharakan
- Department of Urology, Imperial College Healthcare NHS Trust, Charing Cross Hospital, Fulham Palace Road, London, W6 8RF, UK
| | - Kylie A Gallagher
- Department of Urology, Imperial College Healthcare NHS Trust, Charing Cross Hospital, Fulham Palace Road, London, W6 8RF, UK
| | - Taimur T Shah
- Department of Urology, Imperial College Healthcare NHS Trust, Charing Cross Hospital, Fulham Palace Road, London, W6 8RF, UK
- Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Mathias Winkler
- Department of Urology, Imperial College Healthcare NHS Trust, Charing Cross Hospital, Fulham Palace Road, London, W6 8RF, UK
| | - Channa N Jayasena
- Section of Investigative Medicine, Department of Medicine, Imperial College London, London, W12 0NN, UK
| | - Hashim U Ahmed
- Department of Urology, Imperial College Healthcare NHS Trust, Charing Cross Hospital, Fulham Palace Road, London, W6 8RF, UK
- Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Suks Minhas
- Department of Urology, Imperial College Healthcare NHS Trust, Charing Cross Hospital, Fulham Palace Road, London, W6 8RF, UK
| |
Collapse
|
14
|
Puca L, Vlachostergios PJ, Beltran H. Neuroendocrine Differentiation in Prostate Cancer: Emerging Biology, Models, and Therapies. Cold Spring Harb Perspect Med 2019; 9:a030593. [PMID: 29844220 PMCID: PMC6360865 DOI: 10.1101/cshperspect.a030593] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although a de novo clinical presentation of small cell neuroendocrine carcinoma of the prostate is rare, a subset of patients previously diagnosed with prostate adenocarcinoma may develop neuroendocrine features in later stages of castration-resistant prostate cancer (CRPC) progression as a result of treatment resistance. Despite sharing clinical, histologic, and some molecular features with other neuroendocrine carcinomas, including small cell lung cancer, castration-resistant neuroendocrine prostate cancer (CRPC-NE) is clonally derived from prostate adenocarcinoma. CRPC-NE therefore retains early prostate cancer genomic alterations and acquires new molecular changes making them resistant to traditional CRPC therapies. This review focuses on recent advances in our understanding of CRPC-NE biology, the transdifferentiation/plasticity process, and development and characterization of relevant CRPC-NE preclinical models.
Collapse
Affiliation(s)
- Loredana Puca
- Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York 10021
- Division of Medical Oncology, Weill Cornell Medicine, New York, New York 10021
| | | | - Himisha Beltran
- Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York 10021
- Division of Medical Oncology, Weill Cornell Medicine, New York, New York 10021
| |
Collapse
|
15
|
Namekawa T, Ikeda K, Horie-Inoue K, Inoue S. Application of Prostate Cancer Models for Preclinical Study: Advantages and Limitations of Cell Lines, Patient-Derived Xenografts, and Three-Dimensional Culture of Patient-Derived Cells. Cells 2019; 8:cells8010074. [PMID: 30669516 PMCID: PMC6357050 DOI: 10.3390/cells8010074] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
Various preclinical models have been developed to clarify the pathophysiology of prostate cancer (PCa). Traditional PCa cell lines from clinical metastatic lesions, as exemplified by DU-145, PC-3, and LNCaP cells, are useful tools to define mechanisms underlying tumorigenesis and drug resistance. Cell line-based experiments, however, have limitations for preclinical studies because those cells are basically adapted to 2-dimensional monolayer culture conditions, in which the majority of primary PCa cells cannot survive. Recent tissue engineering enables generation of PCa patient-derived xenografts (PDXs) from both primary and metastatic lesions. Compared with fresh PCa tissue transplantation in athymic mice, co-injection of PCa tissues with extracellular matrix in highly immunodeficient mice has remarkably improved the success rate of PDX generation. PDX models have advantages to appropriately recapitulate the molecular diversity, cellular heterogeneity, and histology of original patient tumors. In contrast to PDX models, patient-derived organoid and spheroid PCa models in 3-dimensional culture are more feasible tools for in vitro studies for retaining the characteristics of patient tumors. In this article, we review PCa preclinical model cell lines and their sublines, PDXs, and patient-derived organoid and spheroid models. These PCa models will be applied to the development of new strategies for cancer precision medicine.
Collapse
Affiliation(s)
- Takeshi Namekawa
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan.
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Chiba 260-8677, Japan.
| | - Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan.
| | - Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan.
| | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan.
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan.
| |
Collapse
|
16
|
DiNatale A, Fatatis A. The Bone Microenvironment in Prostate Cancer Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:171-184. [PMID: 31900910 DOI: 10.1007/978-3-030-32656-2_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The propensity of prostate cancer cells to seed the skeleton and then progress into clinically relevant metastatic tumors is widely recognized and a major cause of morbidity and mortality for patients. The natural history of prostate adenocarcinoma most frequently begins with a tumor diagnosed at a localized stage, which is successfully treated by surgical and/or radiation therapy modalities. A relevant percentage of patients are clinically cured but approximately 20-30% will develop biochemical signs of recurrence, which respond to the inhibition of androgen receptor (AR) signaling by hormone-deprivation and receptor antagonists, before the inevitable transition into castration-resistant prostate cancer (CRPC). This stage simultaneously presents with or is rapidly followed by secondary tumors, which involve the skeleton in more than 90% of cases (mCRPC). While generalization in clinical practice is always unwise, it is indisputable that bone-metastatic prostate cancer is virtually incurable. Decades of research have revealed that the tissue microenvironment provided by the bone marrow is as important as the cell-autonomous features of tumor cells in fostering the right conditions that lead to establishment and progression of metastatic tumors in the skeleton.
Collapse
Affiliation(s)
- Anthony DiNatale
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA.,Program in Prostate Cancer, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Alessandro Fatatis
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA. .,Program in Prostate Cancer, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Genc GE, Hipolito VEB, Botelho RJ, Gumuslu S. Lysophosphatidic acid represses autophagy in prostate carcinoma cells. Biochem Cell Biol 2018; 97:387-396. [PMID: 30403494 DOI: 10.1139/bcb-2018-0164] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Lysophosphatidic acid (LPA) is a small signaling phospholipid that mediates diverse functions including cell proliferation, migration, and survival by engaging LPA-agonized G-protein coupled receptors. Autophagy is a survival mechanism in response to nutrient depletion or organellar damage that encloses idle or damaged organelles within autophagosomes that are then delivered to lysosomes for degradation. However, the relationship between LPA and autophagy is largely unknown. The purpose of this study is to elucidate whether LPA affects autophagy through the ERK1/2 and (or) the Akt-mTOR signaling pathways. In this study, we investigated the effect of LPA on autophagy-regulating pathways in various prostate-derived cancer cells including PC3, LNCaP, and Du145 cells grown in complete medium and exposed to serum-free medium. Using Western blotting and ELISA, we determined that LPA stimulates the ERK and mTOR pathways in complete and serum-free medium. The mTOR pathway led to phosphorylation of S6K and ULK, which respectively stimulates protein synthesis and arrests autophagy. Consistent with this, LPA exposure suppressed autophagy as measured by LC3 maturation and formation of GFP-LC3 puncta. Altogether, these results suggest that LPA suffices to activate mTORC1 and suppress autophagy in prostate cancer cells.
Collapse
Affiliation(s)
- Gizem E Genc
- a Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Victoria E B Hipolito
- b Department of Chemistry and Biology and the Graduate Program in Molecular Science, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Roberto J Botelho
- b Department of Chemistry and Biology and the Graduate Program in Molecular Science, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Saadet Gumuslu
- a Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| |
Collapse
|
18
|
Manca S, Frisbie CP, LaGrange CA, Casey CA, Riethoven JJM, Petrosyan A. The Role of Alcohol-Induced Golgi Fragmentation for Androgen Receptor Signaling in Prostate Cancer. Mol Cancer Res 2018; 17:225-237. [PMID: 30224543 DOI: 10.1158/1541-7786.mcr-18-0577] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/23/2018] [Accepted: 08/22/2018] [Indexed: 01/18/2023]
Abstract
Multiple epidemiologic observations and meta-analysis clearly indicate the link between alcohol abuse and the incidence and progression of prostate cancer; however, the mechanism remains enigmatic. Recently, it was found that ethanol (EtOH) induces disorganization of the Golgi complex caused by impaired function of the largest Golgi matrix protein, giantin (GOLGB1), which, in turn, alters the Golgi docking of resident Golgi proteins. Here, it is determined that in normal prostate cells, histone deacetylase 6 (HDAC6), the known regulator of androgen receptor (AR) signaling, localizes in the cytoplasm and nucleus, while its kinase, glycogen synthase kinase β (GSK3β), primarily resides in the Golgi. Progression of prostate cancer is accompanied by Golgi scattering, translocation of GSK3β from the Golgi to the cytoplasm, and the cytoplasmic shift in HDAC6 localization. Alcohol dehydrogenase-generated metabolites induces Golgi disorganization in androgen-responsive LNCaP and 22Rv1 cells, facilitates tumor growth in a mouse xenograft model and activates anchorage-independent proliferation, migration, and cell adhesion. EtOH-treated cells demonstrate reduced giantin and subsequent cytoplasmic GSK3β; this phenomenon was validated in giantin-depleted cells. Redistribution of GSK3β to the cytoplasm results in phosphorylation of HDAC6 and its retention in the cytoplasm, which, in turn, stimulates deacetylation of HSP90, AR import into the nucleus, and secretion of prostate-specific antigen (PSA). Finally, the relationship between Golgi morphology, HDAC6 cytoplasmic content, and clinicopathologic features was assessed in human prostate cancer patient specimens with and without a history of alcohol dependence. IMPLICATIONS: This study demonstrates the importance of alcohol-induced Golgi fragmentation in the activation of AR-mediated proliferation.
Collapse
Affiliation(s)
- Sonia Manca
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Cole P Frisbie
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Chad A LaGrange
- Division of Urologic Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Carol A Casey
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jean-Jack M Riethoven
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, Nebraska.,The Nebraska Center for Integrated Biomolecular Communication, Lincoln, Nebraska
| | - Armen Petrosyan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska. .,The Nebraska Center for Integrated Biomolecular Communication, Lincoln, Nebraska.,The Fred and Pamela Buffett Cancer Center, Omaha, Nebraska
| |
Collapse
|
19
|
Wu CL, Chen CL, Huang HS, Yu DS. A new niclosamide derivatives-B17 can inhibit urological cancers growth through apoptosis-related pathway. Cancer Med 2018; 7:3945-3954. [PMID: 29953738 PMCID: PMC6089145 DOI: 10.1002/cam4.1635] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/04/2018] [Accepted: 06/04/2018] [Indexed: 12/16/2022] Open
Abstract
The incidence and mortality rate of urological cancers is increasing yearly. Niclosamide has been repurposed as an anti‐cancer drug in recent years. Synthesized derivative of niclosamide was testified for its anti‐cancer activity in urological cancers. MTT assay was used to measure the cytotoxicity effect of niclosamide and its derivatives in urological cancer cell lines. Migratory ability was monitored by scratch migration assay. Apoptosis and cell cycle changes were analyzed by annexin V and PI staining. The apoptosis‐related signal proteins were evaluated by western blotting. T24 had the best drug sensitivity with the lowest IC50 in niclosamide and B17 treatment than DU145 and Caki‐1 cells. After niclosamide and B17 treatment, the mitotic cells were decreased, but apoptotic bodies and morphology changes were not prominent in T24, Caki‐1, and DU145 cells. The migratory ability was inhibited in niclosamide treatment than control group on Caki‐1 cells and niclosamide and B17 treatment than control group on DU145 cells. Early apoptosis cells were increased after niclosamide and B17 treatment than control group without cell cycle changes in T24, Caki‐1, and DU145 cells. Programmed cell death was activated majorly through PAPR and bcl‐2 in T24 and caspase‐3 in Caki‐1 cells, respectively. Niclosamide and B17 derivative had good ability in inhibition proliferation and migratory ability in T24, Caki‐1, and DU145 cells without prominent morphology and apoptotic body changes. UCC cells are more sensitive to niclosamide and B17 treatment. Early apoptosis was induced after niclosamide and B17 treatment through different mechanisms in T24, Caki‐1, and DU145 cells.
Collapse
Affiliation(s)
- Chia-Lun Wu
- Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan.,Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Liang Chen
- Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan.,Graduate Institutes for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hsu-Shan Huang
- Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan.,Graduate Institutes for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Dah-Shyong Yu
- Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan.,Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
20
|
Lin TP, Chang YT, Lee SY, Campbell M, Wang TC, Shen SH, Chung HJ, Chang YH, Chiu AW, Pan CC, Lin CH, Chu CY, Kung HJ, Cheng CY, Chang PC. REST reduction is essential for hypoxia-induced neuroendocrine differentiation of prostate cancer cells by activating autophagy signaling. Oncotarget 2018; 7:26137-51. [PMID: 27034167 PMCID: PMC5041970 DOI: 10.18632/oncotarget.8433] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 03/10/2016] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer (PCa) with neuroendocrine differentiation (NED) is tightly associated with hormone refractory PCa (HRPC), an aggressive form of cancer that is nearly impossible to treat. Determining the mechanism of the development of NED may yield novel therapeutic strategies for HRPC. Here, we first demonstrate that repressor element-1 silencing transcription factor (REST), a transcriptional repressor of neuronal genes that has been implicated in androgen-deprivation and IL-6 induced NED, is essential for hypoxia-induced NED of PCa cells. Bioinformatics analysis of transcriptome profiles of REST knockdown during hypoxia treatment demonstrated that REST is a master regulator of hypoxia-induced genes. Gene set enrichment analysis (GSEA) of hypoxia and REST knockdown co-upregulated genes revealed their correlation with HRPC. Consistently, gene ontology (GO) analysis showed that REST reduction potential associated with hypoxia-induced tumorigenesis, NE development, and AMPK pathway activation. Emerging reports have revealed that AMPK activation is a potential mechanism for hypoxia-induced autophagy. In line with this, we demonstrate that REST knockdown alone is capable of activating AMPK and autophagy activation is essential for hypoxia-induced NED of PCa cells. Here, making using of in vitro cell-based assay for NED, we reveal a new role for the transcriptional repressor REST in hypoxia-induced NED and characterized a sequential molecular mechanism downstream of REST resulting in AMPK phosphorylation and autophagy activation, which may be a common signaling pathway leading to NED of PCa.
Collapse
Affiliation(s)
- Tzu-Ping Lin
- Institute of Clinical Medicine, National Yang Ming University, Taipei, Taiwan, R.O.C.,Department of Urology, School of Medicine, and Shu-Tien Urological Research Center, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Yi-Ting Chang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Sung-Yuan Lee
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Mel Campbell
- UC Davis Cancer Center, University of California, Davis, CA, USA
| | - Tien-Chiao Wang
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C.,Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Shu-Huei Shen
- Department of Radiology, Taipei Veterans General Hospital, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Hsiao-Jen Chung
- Department of Urology, School of Medicine, and Shu-Tien Urological Research Center, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Yen-Hwa Chang
- Department of Urology, School of Medicine, and Shu-Tien Urological Research Center, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Allen W Chiu
- Institute of Clinical Medicine, National Yang Ming University, Taipei, Taiwan, R.O.C.,Department of Urology, School of Medicine, and Shu-Tien Urological Research Center, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Chin-Chen Pan
- Department of Pathology, Taipei Veterans General Hospital, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Chi-Hung Lin
- Institute of Clinical Medicine, National Yang Ming University, Taipei, Taiwan, R.O.C.,Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Cheng-Ying Chu
- Institute for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei City, Taiwan, R.O.C
| | - Hsing-Jien Kung
- UC Davis Cancer Center, University of California, Davis, CA, USA.,Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA.,Institute for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei City, Taiwan, R.O.C.,Division of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, R.O.C
| | - Chia-Yang Cheng
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan, R.O.C.,Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Pei-Ching Chang
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan, R.O.C.,Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, R.O.C
| |
Collapse
|
21
|
Gao Q, Zheng J. Ginsenoside Rh2 inhibits prostate cancer cell growth through suppression of microRNA-4295 that activates CDKN1A. Cell Prolif 2018; 51:e12438. [PMID: 29457293 DOI: 10.1111/cpr.12438] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 12/23/2017] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Ginsenoside Rh2 (GRh2) has demonstrative therapeutic effects on a variety of diseases, including some tumours. However, the effects of GRh2 on prostate cancer (PC) cell growth remain unknown, and were, thus, addressed in the present study. MATERIALS AND METHODS PC3 and DU145 PC cell lines were exposed to GRh2. Cell proliferation was assessed in an MTT assay and by BrdU incorporation. Apoptosis of the cells were assessed by TUNEL staining. Total RNA was assessed by RT-qPCR. Protein levels were assessed by Western blotting. Bioinformatics and dual luciferase reporter assay were applied to determine the functional binding of miRNA to mRNA of target gene. RESULTS GRh2 dose-dependently decreased PC cell proliferation, but did not alter cell apoptosis. Mechanistically, GRh2 dose-dependently increased the protein, but not mRNA of a cell-cycle suppressor CDKN1A in PC cells, suggesting the presence of microRNA (miRNA)-mediated protein translation control of CDKN1A by GRh2. In all candidate miRNAs that bind to 3'-UTR of CDKN1A, miR-4295 was specifically found to be suppressed dose-dependently by GRh2 in PC cells. Moreover, miR-4295 bound CDKN1A to suppress its protein translation. Furthermore, cell proliferation in PC cells that overexpressed miR-4295 did not alter in response to GRh2. CONCLUSIONS GRh2 may inhibit PC cell growth through suppression of microRNA-4295 that activates CDKN1A.
Collapse
Affiliation(s)
- Qiruo Gao
- Department of Urology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junhua Zheng
- Department of Urology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Synthesis and anticancer activity of novel water soluble benzimidazole carbamates. Eur J Med Chem 2018; 144:372-385. [DOI: 10.1016/j.ejmech.2017.11.037] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/06/2017] [Accepted: 11/17/2017] [Indexed: 02/07/2023]
|
23
|
Domińska K, Kowalski A, Ochędalski T, Rębas E. Effects of testosterone and 17β‑estradiol on angiotensin‑induced changes in tyrosine kinase activity in the androgen‑independent human prostate cancer cell line, DU145. Int J Mol Med 2017; 40:1573-1581. [PMID: 28949385 DOI: 10.3892/ijmm.2017.3149] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/24/2017] [Indexed: 11/06/2022] Open
Abstract
Angiotensin II (AngII), the main peptide of the renin‑angiotensin system (RAS), is involved in the proliferation of different types of cells, normal and pathological as well. The protein tyrosine kinases (PTKs) play an important role in the growth, differentiation and apoptosis of cells. AngII action depends on the hormonal milieu of the cell, and on sex steroid influence. Angiotensin 1‑7 (Ang1‑7), metabolite of AngII, shows opposite action to AngII in cells. The present study aimed to examine the influence of 17β‑estradiol and testosterone on AngII and Ang1‑7 action on PTK activity in androgen‑independent humane prostate cancer cell line DU145. Cell cultures of human prostate cancer DU145 cells were used as a source of PTKs. Cultures were exposed to different concentrations of AngII (5x10‑11 to 5x10‑9 M). The incubation with hormones lasted 15 min to limit the genomic effects of steroids. In the phosphorylation reaction, we used γ32P‑ATP as a donor of phosphate and a synthetic peptide, Poly(Glu, Tyr) (4:1), as a substrate. The specific activities of PTKs were defined as pmol of 32P incorporated into 1 mg of exogenous Poly(Glu, Tyr) per minute (pmol/mg/min). Our findings suggest that testosterone and 17β‑estradiol may change the effects of angiotensins in a rapid non‑genomic way, probably via membrane‑located receptors. The most significant change was caused by testosterone, whose effect was most significant on changes caused by Ang1‑7. AngII‑induced changes in phosphorylation appeared to be insensitive to the presence of testosterone, but were modified by 17β‑estradiol.
Collapse
Affiliation(s)
- Kamila Domińska
- Department of Comparative Endocrinology, Medical University of Lodz, 92‑215 Lodz, Poland
| | - Antoni Kowalski
- Department of Molecular Neurochemistry, Medical University of Lodz, 92‑215 Lodz, Poland
| | - Tomasz Ochędalski
- Department of Comparative Endocrinology, Medical University of Lodz, 92‑215 Lodz, Poland
| | - Elżbieta Rębas
- Department of Molecular Neurochemistry, Medical University of Lodz, 92‑215 Lodz, Poland
| |
Collapse
|
24
|
Skp2 deficiency restricts the progression and stem cell features of castration-resistant prostate cancer by destabilizing Twist. Oncogene 2017; 36:4299-4310. [PMID: 28346424 PMCID: PMC5532065 DOI: 10.1038/onc.2017.64] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 12/13/2022]
Abstract
Castration-resistant prostate cancer (CRPC) remains a major clinical challenge because of the lack of effective targeted therapy for its treatment. The mechanism underlying how CRPC gains resistance toward hormone depletion and other forms of chemotherapy is poorly understood. Research on understanding the factors that drive these processes is desperately needed to generate new therapies to cure the disease. Here, we discovered a fundamental role of S-phase protein kinase 2 (Skp2) in the formation and progression of CRPC. In transgenic adenocarcinoma mouse prostate model, Skp2 depletion leads to a profound repression of prostate tumor growth and distal metastasis and substantially prolonged overall survival. We revealed that Skp2 regulates CRPC through Twist-mediated oncogenic functions including epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) acquisitions. Mechanistically, Skp2 interacted with Twist and promoted the non-degradative ubiquitination of Twist. Consequently, Skp2 stabilized Twist protein expression by preventing proteasomal degradation of Twist by β-TrCP. We found that Twist overexpression augments CSC self-renewal and population and that Skp2 inhibition reverts Twist's effects on CSC regulation. Furthermore, genetically depleting or pharmacologically inactivating Skp2 synergistically re-sensitized CRPC cells toward chemotherapies such as paclitaxel or doxorubicin. Together, this study uncovering Skp2-mediated Twist stabilization and oncogenic functions in CRPC offers new knowledge on how CRPC progresses and acquires chemoresistance during tumor progression. It provides proof of principle that Skp2 targeting is a promising approach to combat metastatic CRPC by targeting Twist and CSCs.
Collapse
|
25
|
REST is a crucial regulator for acquiring EMT-like and stemness phenotypes in hormone-refractory prostate cancer. Sci Rep 2017; 7:42795. [PMID: 28256535 PMCID: PMC5335619 DOI: 10.1038/srep42795] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 01/17/2017] [Indexed: 01/06/2023] Open
Abstract
Castration-resistance prostate cancer (CRPC), also known as hormone-refractory prostate cancer (HRPC), requires immediate attention since it is not only resistant to androgen ablation, chemo- and radiotherapy, but also highly metastatic. Increasing evidence suggests that enrichment of neuroendocrine (NE) cells is associated with CRPC. Here, combined RNA-seq and ChIP-seq analysis reveals that REST is involved in epithelial-mesenchymal transition (EMT) and stemness acquisition in NE differentiated prostate cancer (PCa) cells via direct transcriptional repression of Twist1 and CD44. Specifically we show that short-term knockdown of REST induces NE differentiation of LNCaP cells. Long-term REST knockdown enhanced the expression of Twist1 and CD44, cell migration and sphere formation. Overexpression of REST in hormone-refractory CWR22Rv1 PCa cells significantly reduces Twist1 and CD44 expression, cell migration and sphere formation. Collectively, our study uncovers REST in regulating EMT and stemness properties of NE PCa cells and suggests that REST is a potential therapeutic target for CRPC.
Collapse
|
26
|
Maina PK, Shao P, Liu Q, Fazli L, Tyler S, Nasir M, Dong X, Qi HH. c-MYC drives histone demethylase PHF8 during neuroendocrine differentiation and in castration-resistant prostate cancer. Oncotarget 2016; 7:75585-75602. [PMID: 27689328 PMCID: PMC5342763 DOI: 10.18632/oncotarget.12310] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 09/20/2016] [Indexed: 02/06/2023] Open
Abstract
Epigenetic factors play critical roles in prostate cancer (PCa) development. However, how they contribute to neuroendocrine differentiation (NED) and castration-resistant PCa (CRPC) is not fully understood. Using bioinformatics and biochemical approaches to analyze cell-based models of NED and CRPC, we found a cluster of epigenetic factors whose expression is downregulated during NED and upregulated in CRPC (i.e. follow a Down-Up pattern). Two histone demethylases within this cluster, PHF8 and KDM3A, are post-transcriptionally regulated by c-MYC through miR-22, which targets both PHF8 and KDM3A. We also found that the c-MYC/miR-22/PHF8 axis is downstream of androgen receptor (AR) signaling in CRPC cells. The co-expression of PHF8 with AR in clinical CRPC samples, normal mouse prostate, and adenocarcinomas of the prostate during PCa progression in a transgenic (TRAMP) mouse model supports the connection between PHF8 and AR. Knockdown of PHF8 impedes cell cycle progression in CRPC cells and has more profound effects on their growth than on the parental LNCaP cell line. Furthermore, PHF8 knockdown sensitizes LNCaP-Abl cells to the AR antagonist enzalutamide. Our data reveal novel mechanisms that underlie the regulation of PHF8 and KDM3A during NED and in CRPC, and support the candidacy of PHF8 as a therapeutic target in CRPC.
Collapse
MESH Headings
- 3' Untranslated Regions
- Animals
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Cycle Checkpoints/genetics
- Cell Line, Tumor
- Cell Proliferation/genetics
- Cell Survival/genetics
- Epigenesis, Genetic
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, myc
- Histone Demethylases/genetics
- Humans
- Interleukin-6/metabolism
- Interleukin-6/pharmacology
- Jumonji Domain-Containing Histone Demethylases/genetics
- Jumonji Domain-Containing Histone Demethylases/metabolism
- Male
- Mice
- MicroRNAs/genetics
- Models, Biological
- Neoplasm Grading
- Neuroendocrine Tumors/genetics
- Neuroendocrine Tumors/metabolism
- Neuroendocrine Tumors/pathology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- RNA Interference
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Transcription Factors/genetics
- Transcriptome
Collapse
Affiliation(s)
- Peterson Kariuki Maina
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Peng Shao
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Qi Liu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Ladan Fazli
- Vancouver Prostate Center, Department of Urology Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Scott Tyler
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Moman Nasir
- Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - Xuesen Dong
- Vancouver Prostate Center, Department of Urology Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Hank Heng Qi
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| |
Collapse
|
27
|
Delgado-González E, Sánchez-Tusie AA, Morales G, Aceves C, Anguiano B. Triiodothyronine Attenuates Prostate Cancer Progression Mediated by β-Adrenergic Stimulation. Mol Med 2016; 22:1-11. [PMID: 26928389 DOI: 10.2119/molmed.2015.00047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 02/19/2016] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer cells are responsive to adrenergic and thyroid stimuli. It is well established that β-adrenergic activation (protein kinase A [PKA]/cAMP response element binding protein [CREB]) promotes cancer progression, but the role of thyroid hormones is poorly understood. We analyzed the effects of β-adrenergic stimulation (isoproterenol [ISO]) and/or thyroid hormone on neuroendocrine (NE) differentiation and cell invasion, using in vivo (LNCaP tumor) and in vitro models (LNCaP and DU145 human cells). Nude mice were inoculated with LNCaP cells and were treated for 6 wks with ISO (200 μg/d), triiodothyronine (T3, 2.5 μg/d) or both. ISO alone reduced tumor growth but increased tumor expression of cAMP response element (CRE)-dependent genes (real-time polymerase chain reaction, chromogranin A, neuron-specific enolase, survivin, vascular endothelial growth factor [VEGF], urokinase plasmin activator [uPA] and metalloproteinase-9 [MMP-9]) and some proteins related to NE differentiation and/or invasiveness (synaptophysin, VEGF, pCREB). T3 reduced tumor growth and prevented the overexpression of ISO-stimulated factors through a pCREB-independent mechanism. In low invasive LNCaP cells, 50 μmol/L ISO or 100 nmol/L thyroxine (T4) induced the acquisition of NE-like morphology (phase-contrast microscopy), increased VEGF secretion (ELISA) and invasive capacity (Transwell assay), but no synergistic effects were observed after the coadministration of ISO + T4. In contrast, 10 nmol/L T3 alone had no effect, but it prevented the NE-like morphology and invasiveness stimulated by ISO. None of these treatments had any effect on highly invasive DU145 cells. In summary, this study showed that ISO and T4 increase cancer progression, and T3 attenuates ISO-stimulated progression. Further studies are required to determine if changes in the ratio of T4/T3 could be relevant for prostate cancer progression.
Collapse
Affiliation(s)
- Evangelina Delgado-González
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Ana Alicia Sánchez-Tusie
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Giapsy Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Carmen Aceves
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Brenda Anguiano
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| |
Collapse
|
28
|
Moritz T, Venz S, Junker H, Kreuz S, Walther R, Zimmermann U. Isoform 1 of TPD52 (PC-1) promotes neuroendocrine transdifferentiation in prostate cancer cells. Tumour Biol 2016; 37:10435-46. [PMID: 26846108 DOI: 10.1007/s13277-016-4925-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/28/2016] [Indexed: 11/24/2022] Open
Abstract
The tumour protein D52 isoform 1 (PC-1), a member of the tumour protein D52 (TPD52) protein family, is androgen-regulated and prostate-specific expressed. Previous studies confirmed that PC-1 contributes to malignant progression in prostate cancer with an important role in castration-resistant stage. In the present work, we identified its impact in mechanisms leading to neuroendocrine (NE) transdifferentiation. We established for long-term PC-1 overexpression an inducible expression system derived from the prostate carcinoma cell line LNCaP. We observed that PC-1 overexpression itself initiates characteristics of neuroendocrine cells, but the effect was much more pronounced in the presence of the cytokine interleukin-6 (IL-6). Moreover, to our knowledge, this is the first report that treatment with IL-6 leads to a significant upregulation of PC-1 in LNCaP cells. Other TPD52 isoforms were not affected. Proceeding from this result, we conclude that PC-1 overexpression enhances the IL-6-mediated differentiation of LNCaP cells into a NE-like phenotype, noticeable by morphological changes and increased expression of typical NE markers, like chromogranin A, synaptophysin or beta-3 tubulin. Immunofluorescent staining of IL-6-treated PC-1-overexpressing LNCaP cells indicates a considerable PC-1 accumulation at the end of the long-branched neuron-like cell processes, which are typically formed by NE cells. Additionally, the experimentally initiated NE transdifferentiation correlates with the androgen receptor status, which was upregulated additively. In summary, our data provide evidence for an involvement of PC-1 in NE transdifferentiation, frequently associated with castration resistance, which is a major therapeutic challenge in the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Tom Moritz
- Department of Medical Biochemistry and Molecular Biology, Ernst Moritz Arndt University of Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Simone Venz
- Department of Medical Biochemistry and Molecular Biology, Ernst Moritz Arndt University of Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Heike Junker
- Department of Medical Biochemistry and Molecular Biology, Ernst Moritz Arndt University of Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany.
| | - Sarah Kreuz
- Department of Medical Biochemistry and Molecular Biology, Ernst Moritz Arndt University of Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany.,Laboratory of Chromatin Biochemistry, BESE Division, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Reinhard Walther
- Department of Medical Biochemistry and Molecular Biology, Ernst Moritz Arndt University of Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany.
| | - Uwe Zimmermann
- Department of Urology, Ernst Moritz Arndt University Greifswald, Greifswald, Germany
| |
Collapse
|
29
|
Dyshlovoy SA, Menchinskaya ES, Venz S, Rast S, Amann K, Hauschild J, Otte K, Kalinin VI, Silchenko AS, Avilov SA, Alsdorf W, Madanchi R, Bokemeyer C, Schumacher U, Walther R, Aminin DL, Fedorov SN, Shubina LK, Stonik VA, Balabanov S, Honecker F, von Amsberg G. The marine triterpene glycoside frondoside A exhibits activity in vitro and in vivo in prostate cancer. Int J Cancer 2016; 138:2450-65. [PMID: 26695519 DOI: 10.1002/ijc.29977] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 12/09/2015] [Indexed: 12/17/2022]
Abstract
Despite recent advances in the treatment of metastatic castration-resistant prostate cancer (CRPC), outcome of patients remains poor due to the development of drug resistance. Thus, new drugs are urgently needed. We investigated efficacy, toxicity and mechanism of action of marine triterpene glycoside frondoside A (FrA) using CRPC cell lines in vitro and in vivo. FrA revealed high efficacy in human prostate cancer cells, while non-malignant cells were less sensitive. Remarkably, proliferation and colony formation of cells resistant to enzalutamide and abiraterone (due to the androgen receptor splice variant AR-V7) were also significantly inhibited by FrA. The marine compound caused cell type specific cell cycle arrest and induction of caspase-dependent or -independent apoptosis. Up-regulation or induction of several pro-apoptotic proteins (Bax, Bad, PTEN), cleavage of PARP and caspase-3 and down-regulation of anti-apoptotic proteins (survivin and Bcl-2) were detected in treated cells. Global proteome analysis revealed regulation of proteins involved in formation of metastases, tumor cell invasion, and apoptosis, like keratin 81, CrkII, IL-1β and cathepsin B. Inhibition of pro-survival autophagy was observed following FrA exposure. In vivo, FrA inhibited tumor growth of PC-3 and DU145 cells with a notable reduction of lung metastasis, as well as circulating tumor cells in the peripheral blood. Increased lymphocyte counts of treated animals might indicate an immune modulating effect of FrA. In conclusion, our results suggest that FrA is a promising new drug for the treatment of mCRPC. Induction of apoptosis, inhibition of pro-survival autophagy, and immune modulatory effects are suspected modes of actions.
Collapse
Affiliation(s)
- Sergey A Dyshlovoy
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation.,Laboratory of bioactive compounds, Department of bioorganic chemistry and biotechnology, School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russian Federation
| | - Ekaterina S Menchinskaya
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Simone Venz
- Department of Medical Biochemistry and Molecular Biology, University of Greifswald, Greifswald, Germany.,Department of Functional Genomics, Interfacultary Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Stefanie Rast
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Amann
- Nephropathology Department, University Medical Center Erlangen, Erlangen, Germany
| | - Jessica Hauschild
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Otte
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vladimir I Kalinin
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Alexandra S Silchenko
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Sergey A Avilov
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Winfried Alsdorf
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ramin Madanchi
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Reinhard Walther
- Department of Medical Biochemistry and Molecular Biology, University of Greifswald, Greifswald, Germany
| | - Dmitry L Aminin
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Sergey N Fedorov
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Larisa K Shubina
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Valentin A Stonik
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Stefan Balabanov
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Friedemann Honecker
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Tumor and Breast Center ZeTuP St. Gallen, St. Gallen, Switzerland
| | - Gunhild von Amsberg
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
30
|
Weaver EM, Zamora FJ, Hearne JL, Martin-Caraballo M. Posttranscriptional regulation of T-type Ca 2+ channel expression by interleukin-6 in prostate cancer cells. Cytokine 2015. [DOI: 10.1016/j.cyto.2015.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
Savelli G, Muni A, Falchi R, Zaniboni A, Barbieri R, Valmadre G, Minari C, Casi C, Rossini P. Somatostatin receptors over-expression in castration resistant prostate cancer detected by PET/CT: preliminary report of in six patients. ANNALS OF TRANSLATIONAL MEDICINE 2015. [PMID: 26207238 DOI: 10.3978/j.issn.2305-5839.2015.06.10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PC) is usually characterized by an excellent prognosis, largely due to little biological aggressiveness and the power of hormonal deprivation therapy. In spite of these favorable characteristics, however, a significant quota of patients does not respond to androgen deprivation therapy (ADT) and develop a progressive disease. Castration-resistant prostate cancer (CRPC) is defined by disease progression in spite of ADT. This progression may show any combination of a rise in serum prostate-specific antigen (PSA), clinical and radiological progression of pre-existing disease, and appearance of new metastases. This event is a striking change in the clinical scenario, since the power of treatment for CRPC patients with distant metastases is very limited. Somatostatin is a hormone produced by neuroendocrine cells. Its distant effects are mediated by the binding to five specific receptors, which are the most striking parameter for neuroendocrine. Various synthetic somatostatin agonists able to bind to the receptors have been synthesized during the past two decades for diagnostic and therapeutic purposes. Octreotide, the most popular of these, is widely used to treat patients affected by neuroendocrine tumors. A number of researches carried out in the past evaluated the possible neuroendocrine differentiation (NED) of PC cells in the castration resistant phase. If proved, the presence of a specific class of receptor on cell's surfaces should give a potentially biological target to be used for therapy. However, these studies led to contradictory results. Aim of our phase III diagnostic trial was to study "in vivo" the over-expression of somatostatin receptors (SSTRs) in CRPC patients by PET/CT after the administration of the somatostatin analog [(68)Ga-DOTANOC,1-Nal(3)]-octreotide labeled with (68)Ga. Every area of increased uptake corresponding to a metastasis detected with other methods was considered as SSTRs expressing. False positivity to SSTRs expression was considered those localizations with a suspicious uptake not confirmed by other radiologic procedures. On the other hand, metastatic lesions lacking the radiopharmaceutical's uptake were considered not SSTRs expressing metastases. The preliminary results in 6 of the 67 patients scheduled by our phase III trial showed metastases with a variable SSTRs expression in 2 patients.
Collapse
Affiliation(s)
- Giordano Savelli
- 1 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 2 Nuclear Medicine Division, SS. Antonio e Biagio e C.Arrigo City Hospital, Alessandria, Italy ; 3 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 4 Medical Oncology Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 5 Oncology Unit, "Carlo Poma" Hospital, Mantua, Italy ; 6 Medical Oncology Division, Presidio Ospedaliero E. Morelli AOVV, Sondrio, Italy ; 7 Medical Physiscs Division, "Carlo Poma" Hospital, Mantua, Italy ; 8 Medical Oncology Division, Medical Oncology Department, Val d'Elsa Hospital, Siena, Italy ; 9 Nuclear Medicine Division, "Carlo Poma" Hospital, Mantua, Italy
| | - Alfredo Muni
- 1 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 2 Nuclear Medicine Division, SS. Antonio e Biagio e C.Arrigo City Hospital, Alessandria, Italy ; 3 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 4 Medical Oncology Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 5 Oncology Unit, "Carlo Poma" Hospital, Mantua, Italy ; 6 Medical Oncology Division, Presidio Ospedaliero E. Morelli AOVV, Sondrio, Italy ; 7 Medical Physiscs Division, "Carlo Poma" Hospital, Mantua, Italy ; 8 Medical Oncology Division, Medical Oncology Department, Val d'Elsa Hospital, Siena, Italy ; 9 Nuclear Medicine Division, "Carlo Poma" Hospital, Mantua, Italy
| | - Roberta Falchi
- 1 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 2 Nuclear Medicine Division, SS. Antonio e Biagio e C.Arrigo City Hospital, Alessandria, Italy ; 3 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 4 Medical Oncology Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 5 Oncology Unit, "Carlo Poma" Hospital, Mantua, Italy ; 6 Medical Oncology Division, Presidio Ospedaliero E. Morelli AOVV, Sondrio, Italy ; 7 Medical Physiscs Division, "Carlo Poma" Hospital, Mantua, Italy ; 8 Medical Oncology Division, Medical Oncology Department, Val d'Elsa Hospital, Siena, Italy ; 9 Nuclear Medicine Division, "Carlo Poma" Hospital, Mantua, Italy
| | - Alberto Zaniboni
- 1 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 2 Nuclear Medicine Division, SS. Antonio e Biagio e C.Arrigo City Hospital, Alessandria, Italy ; 3 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 4 Medical Oncology Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 5 Oncology Unit, "Carlo Poma" Hospital, Mantua, Italy ; 6 Medical Oncology Division, Presidio Ospedaliero E. Morelli AOVV, Sondrio, Italy ; 7 Medical Physiscs Division, "Carlo Poma" Hospital, Mantua, Italy ; 8 Medical Oncology Division, Medical Oncology Department, Val d'Elsa Hospital, Siena, Italy ; 9 Nuclear Medicine Division, "Carlo Poma" Hospital, Mantua, Italy
| | - Roberto Barbieri
- 1 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 2 Nuclear Medicine Division, SS. Antonio e Biagio e C.Arrigo City Hospital, Alessandria, Italy ; 3 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 4 Medical Oncology Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 5 Oncology Unit, "Carlo Poma" Hospital, Mantua, Italy ; 6 Medical Oncology Division, Presidio Ospedaliero E. Morelli AOVV, Sondrio, Italy ; 7 Medical Physiscs Division, "Carlo Poma" Hospital, Mantua, Italy ; 8 Medical Oncology Division, Medical Oncology Department, Val d'Elsa Hospital, Siena, Italy ; 9 Nuclear Medicine Division, "Carlo Poma" Hospital, Mantua, Italy
| | - Giuseppe Valmadre
- 1 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 2 Nuclear Medicine Division, SS. Antonio e Biagio e C.Arrigo City Hospital, Alessandria, Italy ; 3 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 4 Medical Oncology Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 5 Oncology Unit, "Carlo Poma" Hospital, Mantua, Italy ; 6 Medical Oncology Division, Presidio Ospedaliero E. Morelli AOVV, Sondrio, Italy ; 7 Medical Physiscs Division, "Carlo Poma" Hospital, Mantua, Italy ; 8 Medical Oncology Division, Medical Oncology Department, Val d'Elsa Hospital, Siena, Italy ; 9 Nuclear Medicine Division, "Carlo Poma" Hospital, Mantua, Italy
| | - Chiara Minari
- 1 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 2 Nuclear Medicine Division, SS. Antonio e Biagio e C.Arrigo City Hospital, Alessandria, Italy ; 3 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 4 Medical Oncology Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 5 Oncology Unit, "Carlo Poma" Hospital, Mantua, Italy ; 6 Medical Oncology Division, Presidio Ospedaliero E. Morelli AOVV, Sondrio, Italy ; 7 Medical Physiscs Division, "Carlo Poma" Hospital, Mantua, Italy ; 8 Medical Oncology Division, Medical Oncology Department, Val d'Elsa Hospital, Siena, Italy ; 9 Nuclear Medicine Division, "Carlo Poma" Hospital, Mantua, Italy
| | - Camilla Casi
- 1 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 2 Nuclear Medicine Division, SS. Antonio e Biagio e C.Arrigo City Hospital, Alessandria, Italy ; 3 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 4 Medical Oncology Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 5 Oncology Unit, "Carlo Poma" Hospital, Mantua, Italy ; 6 Medical Oncology Division, Presidio Ospedaliero E. Morelli AOVV, Sondrio, Italy ; 7 Medical Physiscs Division, "Carlo Poma" Hospital, Mantua, Italy ; 8 Medical Oncology Division, Medical Oncology Department, Val d'Elsa Hospital, Siena, Italy ; 9 Nuclear Medicine Division, "Carlo Poma" Hospital, Mantua, Italy
| | - Pierluigi Rossini
- 1 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 2 Nuclear Medicine Division, SS. Antonio e Biagio e C.Arrigo City Hospital, Alessandria, Italy ; 3 Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 4 Medical Oncology Division, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy ; 5 Oncology Unit, "Carlo Poma" Hospital, Mantua, Italy ; 6 Medical Oncology Division, Presidio Ospedaliero E. Morelli AOVV, Sondrio, Italy ; 7 Medical Physiscs Division, "Carlo Poma" Hospital, Mantua, Italy ; 8 Medical Oncology Division, Medical Oncology Department, Val d'Elsa Hospital, Siena, Italy ; 9 Nuclear Medicine Division, "Carlo Poma" Hospital, Mantua, Italy
| |
Collapse
|
32
|
Han J, Sohn EJ, Kim B, Kim S, Won G, Yoon S, Lee J, Kim MJ, Lee H, Chung K, Kim SH. Upregulation of death receptor 5 and activation of caspase 8/3 play a critical role in ergosterol peroxide induced apoptosis in DU 145 prostate cancer cells. Cancer Cell Int 2014; 14:117. [PMID: 25506265 PMCID: PMC4265345 DOI: 10.1186/s12935-014-0117-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 10/27/2014] [Indexed: 01/13/2023] Open
Abstract
Background Though ergosterol peroxide (EP) derived from Neungyi mushrooms (Sarcodon aspratus) was known to have cytotoxic, apoptotic, anti-inflammatory and antimycobacterial effects, the underlying molecular mechanism of EP still remains unclear. Thus, in the present study, the apoptotic mechanism of EP was elucidated in DU 145 prostate cancer cells. Methods Cell viability of prostate cancer cells was measured by MTT assay. To see whether EP induces the apoptosis, FACS, western blot and TUNEL assay were performed. To determine the role of Death receptor (DR) 5 molecules in EP-induced apoptosis in DU 145 prostate cancer cells, the silencing of DR 5 was performed by using siRNAs. Results EP showed significant cytotoxicity against DU 145, PC 3, M2182 prostate cancer cells. Also, EP effectively increased the sub G1 population and terminal deoxynucleotidyl transferase DUTP nick end labeling (TUNEL) positive cells in DU 145 prostate cancer cells. Furthermore, western blotting revealed that EP cleaved poly (ADP-ribose) polymerase (PARP) and caspase 8/3, attenuated the expression of fluorescence loss in photobleaching (FLIP), Bcl-XL and Bcl-2 as well as activated Bax, Fas-associated death domain (FADD) and DR 5 in a concentration dependent manner in DU 145 prostate cancer cells. Conversely, caspase 8 inhibitor Z-IETD-FMK blocked the apoptotic ability of EP to cleave PARP and an increase of sub G1 population in DU 145 prostate cancer cells. Likewise, the silencing of DR 5 suppressed the cleavages of PARP induced by EP in DU 145 prostate cancer cells. Conclusion Overall, our findings suggest that ergosterol peroxide induces apoptosis via activation of death receptor 5 and caspase 8/3 in DU 145 prostate cancer cells as a cancer chemopreventive agent or dietary factor.
Collapse
Affiliation(s)
- Jonghyun Han
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| | - Eun Jung Sohn
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| | - Bonglee Kim
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| | - Sunhee Kim
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| | - Gunho Won
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| | - Sangwook Yoon
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| | - Jihyun Lee
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| | - Moon Joon Kim
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| | - Hojin Lee
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| | - Kyujin Chung
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| | - Sung-Hoon Kim
- Cancer Preventive Material Development Research Center, College of Oriental Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 130-701 Republic of Korea
| |
Collapse
|
33
|
Courel M, El Yamani FZ, Alexandre D, El Fatemi H, Delestre C, Montero-Hadjadje M, Tazi F, Amarti A, Magoul R, Chartrel N, Anouar Y. Secretogranin II is overexpressed in advanced prostate cancer and promotes the neuroendocrine differentiation of prostate cancer cells. Eur J Cancer 2014; 50:3039-49. [PMID: 25307750 DOI: 10.1016/j.ejca.2014.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/11/2014] [Accepted: 09/15/2014] [Indexed: 11/28/2022]
Abstract
AIM In prostate cancer (PCa), neuroendocrine differentiation (NED) is commonly observed in relapsing, hormone therapy-resistant tumours after androgen deprivation. However, the molecular mechanisms involved in the NED of PCa cells remain poorly understood. In this study, we investigated the expression of the neuroendocrine secretory protein secretogranin II (SgII) in PCa, and its potential involvement in the progression of this cancer as a granulogenic factor promoting NED. METHODS We have examined SgII immunoreactivity in 25 benign prostate hyperplasia and 32 PCa biopsies. In vitro experiments were performed to investigate the involvement of SgII in the neuroendocrine differentiation and the proliferation of PCa cell lines. RESULTS We showed that immunoreactive SgII intensity correlates with tumour grade in PCa patients. Using the androgen-dependent lymph node cancer prostate cells (LNCaP) cells, we found that NED triggered by androgen deprivation is associated with the induction of SgII expression. In addition, forced expression of SgII in LNCaP cells implemented a regulated secretory pathway by triggering the formation of secretory granule-like structures competent for hormone storage and regulated release. Finally, we found that SgII promotes prostate cancer (CaP) cell proliferation. CONCLUSION The present data show that SgII is highly expressed in advanced PCa and may contribute to the neuroendocrine differentiation by promoting the formation of secretory granules and the proliferation of PCa cells.
Collapse
Affiliation(s)
- Maïté Courel
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France
| | - Fatima-Zohra El Yamani
- Laboratory of Neuroendocrinology and Nutritional and Climatic Environment, Faculty of Sciences Dhar-El Mahraz, University Sidi Mohamed Ben Abdellah, 30000 Fez, Morocco
| | - David Alexandre
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France
| | - Hinde El Fatemi
- Department of Pathology, University Hospital Hassan II, University Sidi Mohamed Ben Abdellah, 30000 Fez, Morocco
| | - Charlène Delestre
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France
| | - Maité Montero-Hadjadje
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France
| | - Fadl Tazi
- Department of Pathology, University Hospital Hassan II, University Sidi Mohamed Ben Abdellah, 30000 Fez, Morocco
| | - Afaf Amarti
- Department of Pathology, University Hospital Hassan II, University Sidi Mohamed Ben Abdellah, 30000 Fez, Morocco
| | - Rabia Magoul
- Laboratory of Neuroendocrinology and Nutritional and Climatic Environment, Faculty of Sciences Dhar-El Mahraz, University Sidi Mohamed Ben Abdellah, 30000 Fez, Morocco
| | - Nicolas Chartrel
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France
| | - Youssef Anouar
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France.
| |
Collapse
|
34
|
Li P, Wang J, Chu M, Zhang K, Yang R, Gao WQ. Zeb1 promotes androgen independence of prostate cancer via induction of stem cell-like properties. Exp Biol Med (Maywood) 2014; 239:813-822. [PMID: 24912507 DOI: 10.1177/1535370214538727] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Current androgen deprivation therapy often leads to androgen independence. However, mechanism of the therapeutic failure is still not well understood. Here, we demonstrate elevated expression of Zeb1 in androgen-independent prostate cancer cells and prostate tumors of castrated PTEN conditional knockout mice. While Zeb1 shRNA resulted in a sensitization of androgen-independent prostate cancer cells, forced Zeb1 expression caused androgen-dependent prostate cancer cells to be more resistant to androgen deprivation. Moreover, such effects appeared to be mediated by induction of pluripotent genes or stem cell-like properties. Collectively, these findings suggest that inhibition of Zeb1 might be a potential therapeutic strategy for treatment of androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Ping Li
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jia Wang
- Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Mingliang Chu
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Kaiqing Zhang
- Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ru Yang
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
35
|
Pernicová Z, Slabáková E, Fedr R, Šimečková Š, Jaroš J, Suchánková T, Bouchal J, Kharaishvili G, Král M, Kozubík A, Souček K. The role of high cell density in the promotion of neuroendocrine transdifferentiation of prostate cancer cells. Mol Cancer 2014; 13:113. [PMID: 24884804 PMCID: PMC4229954 DOI: 10.1186/1476-4598-13-113] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 05/13/2014] [Indexed: 11/13/2022] Open
Abstract
Background Tumor heterogeneity and the plasticity of cancer cells present challenges for effective clinical diagnosis and therapy. Such challenges are epitomized by neuroendocrine transdifferentiation (NED) and the emergence of neuroendocrine-like cancer cells in prostate tumors. This phenomenon frequently arises from androgen-depleted prostate adenocarcinoma and is associated with the development of castration-resistant prostate cancer and poor prognosis. Results In this study, we showed that NED was evoked in both androgen receptor (AR)-positive and AR-negative prostate epithelial cell lines by growing the cells to a high density. Androgen depletion and high-density cultivation were both associated with cell cycle arrest and deregulated expression of several cell cycle regulators, such as p27Kip1, members of the cyclin D protein family, and Cdk2. Dual inhibition of Cdk1 and Cdk2 using pharmacological inhibitor or RNAi led to modulation of the cell cycle and promotion of NED. We further demonstrated that the cyclic adenosine 3′, 5′-monophosphate (cAMP)-mediated pathway is activated in the high-density conditions. Importantly, inhibition of cAMP signaling using a specific inhibitor of adenylate cyclase, MDL-12330A, abolished the promotion of NED by high cell density. Conclusions Taken together, our results imply a new relationship between cell cycle attenuation and promotion of NED and suggest high cell density as a trigger for cAMP signaling that can mediate reversible NED in prostate cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, v,v,i, Královopolská 135, CZ-612 65 Brno, Czech Republic.
| |
Collapse
|
36
|
Arenas Reyes NJ, Manuel Moreno LA, Carrillo Rodríguez AP, Fonseca Buitrago CL, Pompilio Daza Almendrales F. Diferenciación neuroendocrina en cáncer de próstata. Revisión de la literatura. UROLOGÍA COLOMBIANA 2014. [DOI: 10.1016/s0120-789x(14)50007-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
37
|
Gacci M, Baldi E, Tamburrino L, Detti B, Livi L, De Nunzio C, Tubaro A, Gravas S, Carini M, Serni S. Quality of Life and Sexual Health in the Aging of PCa Survivors. Int J Endocrinol 2014; 2014:470592. [PMID: 24744780 PMCID: PMC3976934 DOI: 10.1155/2014/470592] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 02/02/2014] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is the most common malignancy in elderly men. The progressive ageing of the world male population will further increase the need for tailored assessment and treatment of PCa patients. The determinant role of androgens and sexual hormones for PCa growth and progression has been established. However, several trials on androgens and PCa are recently focused on urinary continence, quality of life, and sexual function, suggesting a new point of view on the whole endocrinological aspect of PCa. During aging, metabolic syndrome, including diabetes, hypertension, dyslipidemia, and central obesity, can be associated with a chronic, low-grade inflammation of the prostate and with changes in the sex steroid pathways. These factors may affect both the carcinogenesis processes and treatment outcomes of PCa. Any treatment for PCa can have a long-lasting negative impact on quality of life and sexual health, which should be assessed by validated self-reported questionnaires. In particular, sexual health, urinary continence, and bowel function can be worsened after prostatectomy, radiotherapy, or hormone treatment, mostly in the elderly population. In the present review we summarized the current knowledge on the role of hormones, metabolic features, and primary treatments for PCa on the quality of life and sexual health of elderly Pca survivors.
Collapse
Affiliation(s)
- Mauro Gacci
- Department of Urology, University of Florence, Careggi Hospital, Viale Gramsci 7, 50121 Florence, Italy
| | - Elisabetta Baldi
- Department of Experimental and Clinical Biomedical Sciences, Section of Clinical Pathophysiology, University of Florence, Italy
| | - Lara Tamburrino
- Department of Experimental and Clinical Biomedical Sciences, Section of Clinical Pathophysiology, University of Florence, Italy
| | - Beatrice Detti
- Radiotherapy, University Hospital Careggi, University of Florence, Italy
| | - Lorenzo Livi
- Radiotherapy, University Hospital Careggi, University of Florence, Italy
| | - Cosimo De Nunzio
- Department of Urology, Sant'Andrea Hospital, University “La Sapienza”, Rome, Italy
| | - Andrea Tubaro
- Department of Urology, Sant'Andrea Hospital, University “La Sapienza”, Rome, Italy
| | - Stavros Gravas
- Department of Urology, University Hospital of Larissa, Larissa, Greece
| | - Marco Carini
- Department of Urology, University of Florence, Careggi Hospital, Viale Gramsci 7, 50121 Florence, Italy
| | - Sergio Serni
- Department of Urology, University of Florence, Careggi Hospital, Viale Gramsci 7, 50121 Florence, Italy
| |
Collapse
|
38
|
De Petrocellis L, Ligresti A, Schiano Moriello A, Iappelli M, Verde R, Stott CG, Cristino L, Orlando P, Di Marzo V. Non-THC cannabinoids inhibit prostate carcinoma growth in vitro and in vivo: pro-apoptotic effects and underlying mechanisms. Br J Pharmacol 2014; 168:79-102. [PMID: 22594963 DOI: 10.1111/j.1476-5381.2012.02027.x] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Cannabinoid receptor activation induces prostate carcinoma cell (PCC) apoptosis, but cannabinoids other than Δ(9) -tetrahydrocannabinol (THC), which lack potency at cannabinoid receptors, have not been investigated. Some of these compounds antagonize transient receptor potential melastatin type-8 (TRPM8) channels, the expression of which is necessary for androgen receptor (AR)-dependent PCC survival. EXPERIMENTAL APPROACH We tested pure cannabinoids and extracts from Cannabis strains enriched in particular cannabinoids (BDS), on AR-positive (LNCaP and 22RV1) and -negative (DU-145 and PC-3) cells, by evaluating cell viability (MTT test), cell cycle arrest and apoptosis induction, by FACS scans, caspase 3/7 assays, DNA fragmentation and TUNEL, and size of xenograft tumours induced by LNCaP and DU-145 cells. KEY RESULTS Cannabidiol (CBD) significantly inhibited cell viability. Other compounds became effective in cells deprived of serum for 24 h. Several BDS were more potent than the pure compounds in the presence of serum. CBD-BDS (i.p.) potentiated the effects of bicalutamide and docetaxel against LNCaP and DU-145 xenograft tumours and, given alone, reduced LNCaP xenograft size. CBD (1-10 µM) induced apoptosis and induced markers of intrinsic apoptotic pathways (PUMA and CHOP expression and intracellular Ca(2+)). In LNCaP cells, the pro-apoptotic effect of CBD was only partly due to TRPM8 antagonism and was accompanied by down-regulation of AR, p53 activation and elevation of reactive oxygen species. LNCaP cells differentiated to androgen-insensitive neuroendocrine-like cells were more sensitive to CBD-induced apoptosis. CONCLUSIONS AND IMPLICATIONS These data support the clinical testing of CBD against prostate carcinoma.
Collapse
Affiliation(s)
- Luciano De Petrocellis
- Istituto di Cibernetica, Endocannabinoid Research Group, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
A Comprehensive Gene Expression Analysis of Resistance Formation upon Metronomic Cyclophosphamide Therapy. Transl Oncol 2013; 6:1-9. [PMID: 23418611 DOI: 10.1593/tlo.12295] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 12/18/2012] [Accepted: 12/24/2012] [Indexed: 01/12/2023] Open
Abstract
Resistance formation is one of the major hurdles in cancer therapy. Metronomic anti-angiogenic treatment of xenografted prostate cancer tumors in severe combined-immunodeficiency (SCID) mice with cyclophosphamide (CPA) results in the appearance of resistant tumors. To investigate the complex molecular changes occurring during resistance formation, we performed a comprehensive gene expression analysis of the resistant tumors in vivo. We observed a multitude of differentially expressed genes, e.g., PAS domain containing protein 1, annexin A3 (ANXA3), neurotensin, or plasminogen activator tissue (PLAT), when comparing resistant to in vivo passaged tumor samples. Furthermore, tumor cells from in vivo and in vitro conditions showed a significant difference in target gene expression. We assigned the differentially expressed genes to functional pathways like axon guidance, steroid biosynthesis, and complement and coagulation cascades. Most of these genes were involved in anti-coagulation. Up-regulation of anticoagulatory ANXA3 and PLAT and down-regulation of PLAT inhibitor serpin peptidase inhibitor clade A were validated by quantitative real-time polymerase chain reaction. In contrast, coagulation factor F3 was upregulated, accompanied by the expression of an altered gene product. These findings give insights into the resistance mechanisms of metronomic CPA treatment, suggesting an important role of anti-coagulation in resistance formation.
Collapse
|
40
|
Tamburrino L, Salvianti F, Marchiani S, Pinzani P, Nesi G, Serni S, Forti G, Baldi E. Androgen receptor (AR) expression in prostate cancer and progression of the tumor: Lessons from cell lines, animal models and human specimens. Steroids 2012; 77:996-1001. [PMID: 22289337 DOI: 10.1016/j.steroids.2012.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 01/10/2012] [Accepted: 01/11/2012] [Indexed: 11/18/2022]
Abstract
Prostate cancer (PC) is among the most frequent causes of death for cancer in men in western countries. In about 30% of cases, the disease is very aggressive rapidly leading to a metastatic disease. In these cases, prostatectomy is not possible and the patient is usually directed to androgen deprivation therapy (ADT) which is only palliative as a castration resistant PC (CRPC) usually develops within 2-3 years of treatment. At present there are no prognostic markers of PC progression. The role of the androgen receptor (AR) in initiation and development of PC is well established and documented. In particular, it is now recognized that androgens actions are mediated by an integration of classical (genomic) and non-classical (extragenomic) activity of AR. The picture about AR and PC become less clear when CRPC is considered. Indeed, the role of AR in the progression of PC and in CRPC is controversial. Results of studies on the role of AR in the progression of PC in cell lines, xenografts, animal models and even clinical specimens are conflicting reflecting the high heterogeneity of PC. Recent evidence in AR conditional KO in mouse models of PC shows possible contrasting roles of AR depending on its location in the two (epithelial or stromal) compartments of PC. Here, we review this evidence and report preliminary data of a study performed in microdissected areas of epithelia and stromal compartments of human PC.
Collapse
Affiliation(s)
- Lara Tamburrino
- Dept. of Clinical Physiopathology, Center of Excellence for Research, Transfer and High Education DeNothe, University of Florence, Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sagnak L, Topaloglu H, Ozok U, Ersoy H. Prognostic significance of neuroendocrine differentiation in prostate adenocarcinoma. Clin Genitourin Cancer 2012; 9:73-80. [PMID: 22035833 DOI: 10.1016/j.clgc.2011.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 07/20/2011] [Accepted: 07/27/2011] [Indexed: 01/04/2023]
Abstract
Much progress has been made toward an understanding of the development and progression of prostate cancer (PC) and the factors that drive the development of androgen independence. Neuroendocrine (NE) cells may provide an intriguing link between NE cell differentiation (NED) and tumor progression in PC. NED in PC generally confers a more aggressive clinical behavior and less favorable prognosis than conventional PC. In this article, we review the known functions of NE cells in PC and discuss the current knowledge on stimulation of cancer proliferation, invasion, apoptosis resistance, serum and immunohistochemical markers, and the prognostic significance of NED in human PC.
Collapse
Affiliation(s)
- Levent Sagnak
- Ministry of Health, Diskapi Yildirim Beyazit Education and Research Hospital, 3rd Urology Clinic, Ankara, Turkey.
| | | | | | | |
Collapse
|
42
|
Danza G, Di Serio C, Rosati F, Lonetto G, Sturli N, Kacer D, Pennella A, Ventimiglia G, Barucci R, Piscazzi A, Prudovsky I, Landriscina M, Marchionni N, Tarantini F. Notch signaling modulates hypoxia-induced neuroendocrine differentiation of human prostate cancer cells. Mol Cancer Res 2011; 10:230-8. [PMID: 22172337 DOI: 10.1158/1541-7786.mcr-11-0296] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
UNLABELLED Prostate carcinoma is among the most common causes of cancer-related death in men, representing 15% of all male malignancies in developed countries. Neuroendocrine differentiation (NED) has been associated with tumor progression, poor prognosis, and with the androgen-independent status. Currently, no successful therapy exists for advanced, castration-resistant disease. Because hypoxia has been linked to prostate cancer progression and unfavorable outcome, we sought to determine whether hypoxia would impact the degree of neuroendocrine differentiation of prostate cancer cells in vitro. RESULTS Exposure of LNCaP cells to low oxygen tension induced a neuroendocrine phenotype, associated with an increased expression of the transcription factor neurogenin3 and neuroendocrine markers, such as neuron-specific enolase, chromogranin A, and β3-tubulin. Moreover, hypoxia triggered a significant decrease of Notch 1 and Notch 2 mRNA and protein expression, with subsequent downregulation of Notch-mediated signaling, as shown by reduced levels of the Notch target genes, Hes1 and Hey1. NED was promoted by attenuation of Hes1 transcription, as cells expressing a dominant-negative form of Hes1 displayed increased levels of neuroendocrine markers under normoxic conditions. Although hypoxia downregulated Notch 1 and Notch 2 mRNA transcription and receptor activation also in the androgen-independent cell lines, PC-3 and Du145, it did not change the extent of NED in these cultures, suggesting that androgen sensitivity may be required for transdifferentiation to occur. CONCLUSIONS Hypoxia induces NED of LNCaP cells in vitro, which seems to be driven by the inhibition of Notch signaling with subsequent downregulation of Hes1 transcription.
Collapse
Affiliation(s)
- Giovanna Danza
- Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tai S, Sun Y, Squires JM, Zhang H, Oh WK, Liang CZ, Huang J. PC3 is a cell line characteristic of prostatic small cell carcinoma. Prostate 2011; 71:1668-79. [PMID: 21432867 PMCID: PMC3426349 DOI: 10.1002/pros.21383] [Citation(s) in RCA: 339] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 02/24/2011] [Indexed: 11/11/2022]
Abstract
BACKGROUND The majority of the prostatic cancers are adenocarcinomas characterized by glandular formation and the expression of luminal differentiation markers androgen receptor (AR) and prostate-specific antigen (PSA). Most adenocarcinomas are indolent and androgen-dependent. Hormonal therapy that inhibits AR signaling produces symptomatic relief in patients with advanced and metastatic adenocarcinomas. Prostatic small cell neuroendocrine carcinoma (SCNC) is a variant form of prostate cancer (PC). In contrast to adenocarcinoma, the tumor cells of SCNC do not form glands and are negative for AR and PSA. SCNC is extremely aggressive and does not respond to hormonal therapy. The purpose of this study was to compare the important and relevant features of two most commonly used PC cell lines, LNCaP and PC3, with prostatic adenocarcinoma and SCNC. METHODS Xenograft tumors of LNCaP and PC3 were prepared and compared with human prostatic adenocarcinoma and SCNC for the expression of key signaling molecules by immunohistochemistry and Western blot analysis. RESULTS LNCaP cells express AR and PSA and their growth is inhibited by androgen withdrawal, similar to human prostatic adenocarcinoma. PC3 cells do not express AR and PSA and their proliferation is independent of androgen, similar to SCNC. Adenocarcinoma cells and LNCaP cells are negative for neuroendocrine markers and stem cell-associated marker CD44 while SCNC and PC3 cells are positive. LNCaP cells have identical cytokeratin profiles to adenocarcinoma while PC3 cells have cytokeratin profiles similar to SCNC. CONCLUSION LNCaP cells share common features with adenocarcinoma while PC3 cells are characteristic of SCNC.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Animals
- Blotting, Western
- Carcinoma, Neuroendocrine/metabolism
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Small Cell/metabolism
- Carcinoma, Small Cell/pathology
- Cell Line, Tumor
- Humans
- Immunohistochemistry
- Male
- Mice
- Mice, SCID
- Neoplasm Transplantation
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Prostate-Specific Antigen/biosynthesis
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- RNA, Neoplasm/chemistry
- RNA, Neoplasm/genetics
- Receptors, Androgen/biosynthesis
- Reverse Transcriptase Polymerase Chain Reaction
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Sheng Tai
- Department of Pathology, Jonsson Comprehensive Cancer Center and Broad Center for Regenerative Medicine and Stem Cell Biology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Urology, the Geriatrics Research Institute, the First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Yin Sun
- Department of Pathology, Jonsson Comprehensive Cancer Center and Broad Center for Regenerative Medicine and Stem Cell Biology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jill M. Squires
- Department of Pathology, Jonsson Comprehensive Cancer Center and Broad Center for Regenerative Medicine and Stem Cell Biology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Hong Zhang
- Department of Pathology, Jonsson Comprehensive Cancer Center and Broad Center for Regenerative Medicine and Stem Cell Biology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Anhui, China
| | - William K. Oh
- Department of Medicine and Urology, the Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York
| | - Chao-Zhao Liang
- Department of Urology, the Geriatrics Research Institute, the First Affiliated Hospital of Anhui Medical University, Anhui, China
- Correspondence to: Dr. Jiaoti Huang, MD, PhD, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave., 13-229 CHS, Los Angeles, CA 90095-1732.
| | - Jiaoti Huang
- Department of Pathology, Jonsson Comprehensive Cancer Center and Broad Center for Regenerative Medicine and Stem Cell Biology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Correspondence to: Dr. Jiaoti Huang, MD, PhD, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave., 13-229 CHS, Los Angeles, CA 90095-1732.
| |
Collapse
|
44
|
Abstract
BACKGROUND Androgens play an essential role in the development and differentiation of the prostate gland; their contribution to pathological conditions, such as benign prostatic hyperplasia and prostate cancer (PC), remains unclear. AIM We reviewed relationships between androgens and the prostate both in physiological and pathological conditions. MATERIAL AND METHODS A systematic search of published evidence was performed using Medline (1969 to September 2010). RESULTS Androgen-dependency of prostate growth is evident only in the hypogonadal condition, but not in the eugonadal state (the "saturation hypothesis"). There is unequivocal evidence that reducing androgen signaling to the hypogonadal range can reduce PC growth and patient symptoms. At physiological testosterone concentration there is no link between androgen levels and PC risk. In addition, different strategies of androgen deprivation (ADT) for advanced PC are only palliative and rarely cure patients. Preliminary evidence indicates that a low androgen milieu is associated with tumor aggressiveness. Transition to androgen-independence is complex and involves both selection and outgrowth of preexisting androgen resistant clones, as well as adaptative upregulation of genes that help the cancer cells to survive and grow after ADT. Because androgens are essential for the regulation of fat distribution, insulin sensitivity, and lipid and bone metabolism, recent publications have highlighted the concept that ADT may also be involved with an increase in overall, as well as cardiovascular, morbidity and mortality. CONCLUSIONS While ADT still represents a cornerstone for the palliative therapy of a small fraction of aggressive PC, a "misuse and/or abuse" of ADT should be avoided.
Collapse
Affiliation(s)
- G Corona
- Sexual Medicine and Andrology Unit, Department of Clinical Physiopathology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | | | | |
Collapse
|
45
|
Søkilde R, Kaczkowski B, Podolska A, Cirera S, Gorodkin J, Møller S, Litman T. Global microRNA analysis of the NCI-60 cancer cell panel. Mol Cancer Ther 2011; 10:375-84. [PMID: 21252286 DOI: 10.1158/1535-7163.mct-10-0605] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
MicroRNAs (miRNA) are a group of short noncoding RNAs that regulate gene expression at the posttranscriptional level. They are involved in many biological processes, including development, differentiation, apoptosis, and carcinogenesis. Because miRNAs may play a role in the initiation and progression of cancer, they comprise a novel class of promising diagnostic and prognostic molecular markers and potential drug targets. By applying an LNA-enhanced microarray platform, we studied the expression profiles of 955 miRNAs in the NCI-60 cancer cell lines and identified tissue- and cell-type-specific miRNA patterns by unsupervised hierarchical clustering and statistical analysis. A comparison of our data to three previously published miRNA expression studies on the NCI-60 panel showed a remarkably high correlation between the different technical platforms. In addition, the current work contributes expression data for 369 miRNAs that have not previously been profiled. Finally, by matching drug sensitivity data for the NCI-60 cells to their miRNA expression profiles, we found numerous drug-miRNAs pairs, for which the miRNA expression and drug sensitivity profiles were highly correlated and thus represent potential candidates for further investigation of drug resistance and sensitivity mechanisms.
Collapse
Affiliation(s)
- Rolf Søkilde
- Department of Biomarker Discovery, Exiqon A/S, Bygstubben 9, DK-2950 Vedbk, Denmark
| | | | | | | | | | | | | |
Collapse
|