1
|
Rajabi F, Smith R, Liu-Bordes WY, Schertzer M, Huet S, Londoño-Vallejo A. DNA damage-induced EMT controlled by the PARP-dependent chromatin remodeler ALC1 promotes DNA repair efficiency through RAD51 in tumor cells. Mol Biol Cell 2024; 35:ar151. [PMID: 39504452 PMCID: PMC11656468 DOI: 10.1091/mbc.e24-08-0370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) allows cancer cells to metastasize while acquiring resistance to apoptosis and chemotherapeutic agents with significant implications for patients' prognosis and survival. Despite its clinical relevance, the mechanisms initiating EMT during cancer progression remain poorly understood. We demonstrate that DNA damage triggers EMT and that activation of poly (ADP-ribose) polymerase (PARP) and the PARP-dependent chromatin remodeler ALC1 (CHD1L) was required for this response. Our results suggest that this activation directly facilitates access to the chromatin of EMT transcriptional factors (TFs) which then initiate cell reprogramming. We also show that EMT-TFs bind to the RAD51 promoter to stimulate its expression and to promote DNA repair by homologous recombination. Importantly, a clinically relevant PARP inhibitor reversed or prevented EMT in response to DNA damage while resensitizing tumor cells to other genotoxic agents. Overall, our observations shed light on the intricate relationship between EMT, DNA damage response, and PARP inhibitors, providing potential insights for in cancer therapeutics.
Collapse
Affiliation(s)
- Fatemeh Rajabi
- Institut Curie, CNRS-UMR3244, Sorbonne University, 75005 Paris, France
- Present addresses: Cancer Genomics lab, Inserm-U981, Gustave Roussy Cancer Center Grand Paris, Villejuif, 94805, France
| | - Rebecca Smith
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, BIOSIT – UMS3480, F- 35000 Rennes, France
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, United Kingdom
| | | | - Michael Schertzer
- Institut Curie, CNRS-UMR3244, Sorbonne University, 75005 Paris, France
| | - Sebastien Huet
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, BIOSIT – UMS3480, F- 35000 Rennes, France
| | - Arturo Londoño-Vallejo
- Institut Curie, CNRS-UMR3244, Sorbonne University, 75005 Paris, France
- Institut Curie, Inserm U1021-CNRS UMR 3347, Paris Saclay University, Centre Universitaire, 91405 Orsay Cedex, France
| |
Collapse
|
2
|
Tamarindo GH, Ribeiro CF, Silva ADT, Castro A, Caruso ÍP, Souza FP, Taboga SR, Loda M, Góes RM. The polyunsaturated fatty acid docosahexaenoic affects mitochondrial function in prostate cancer cells. Cancer Metab 2024; 12:24. [PMID: 39113152 PMCID: PMC11308158 DOI: 10.1186/s40170-024-00348-0] [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: 12/19/2023] [Accepted: 06/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) shows a rewired metabolism featuring increased fatty acid uptake and synthesis via de novo lipogenesis, both sharply related to mitochondrial physiology. The docosahexaenoic acid (DHA) is an omega-3 polyunsaturated fatty acid (PUFA) that exerts its antitumoral properties via different mechanisms, but its specific action on mitochondria in PCa is not clear. Therefore, we investigated whether the DHA modulates mitochondrial function in PCa cell lines. METHODS Here, we evaluated mitochondrial function of non-malignant PNT1A and the castration-resistant (CRPC) prostate 22Rv1 and PC3 cell lines in response to DHA incubation. For this purpose, we used Seahorse extracellular flux assay to assess mitochondria function, [14C]-glucose to evaluate its oxidation as well as its contribution to fatty acid synthesis, 1H-NMR for metabolite profile determination, MitoSOX for superoxide anion production, JC-1 for mitochondrial membrane polarization, mass spectrometry for determination of phosphatidylglycerol levels and composition, staining with MitoTracker dye to assess mitochondrial morphology under super-resolution in addition to Transmission Electron Microscopy, In-Cell ELISA for COX-I and SDH-A protein expression and flow cytometry (Annexin V and 7-AAD) for cell death estimation. RESULTS In all cell lines DHA decreased basal respiratory activity, ATP production, and the spare capacity in mitochondria. Also, the omega-3 induced mitochondrial hyperpolarization, ROS overproduction and changes in membrane phosphatidylglycerol composition. In PNT1A, DHA led to mitochondrial fragmentation and it increased glycolysis while in cancer cells it stimulated glucose oxidation, but decreased de novo lipogenesis specifically in 22Rv1, indicating a metabolic shift. In all cell lines, DHA modulated several metabolites related to energy metabolism and it was incorporated in phosphatidylglycerol, a precursor of cardiolipin, increasing the unsaturation index in the mitochondrial membrane. Accordingly, DHA triggered cell death mainly in PNT1A and 22Rv1. CONCLUSION In conclusion, mitochondrial metabolism is significantly affected by the PUFA supplementation to the point that cells are not able to proliferate or survive under DHA-enriched condition. Moreover, combination of DHA supplementation with inhibition of metabolism-related pathways, such as de novo lipogenesis, may be synergistic in castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Guilherme Henrique Tamarindo
- Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | | | - Alana Della Torre Silva
- Department of Biological Sciences, IBILCE - UNESP. Rua Cristovão Colombo, 2265 Jardim Nazareth, São José Do Rio Preto, São Paulo, 15054-000, Brazil
| | - Alex Castro
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | - Ícaro Putinhon Caruso
- Department of Biophysics, Institute of Biosciences, Humanities and Exact Science, São Paulo State University, São José Do Rio Preto, São Paulo, Brazil
- Institute of Medical Biochemistry and National Center for Structure Biology and Bioimaging (CENABIO), National Center for Nuclear Magnetic Resonance of Macromolecules, Federal University of Rio de Janeiro, Ilha Do Fundão, Rio de Janeiro, Brazil
| | - Fátima Pereira Souza
- Department of Biophysics, Institute of Biosciences, Humanities and Exact Science, São Paulo State University, São José Do Rio Preto, São Paulo, Brazil
| | - Sebastião Roberto Taboga
- Department of Biological Sciences, IBILCE - UNESP. Rua Cristovão Colombo, 2265 Jardim Nazareth, São José Do Rio Preto, São Paulo, 15054-000, Brazil
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Rejane Maira Góes
- Department of Biological Sciences, IBILCE - UNESP. Rua Cristovão Colombo, 2265 Jardim Nazareth, São José Do Rio Preto, São Paulo, 15054-000, Brazil.
| |
Collapse
|
3
|
Rajabi F, Liu-Bordes WY, Pinskaya M, Dominika F, Kratassiouk G, Pinna G, Nanni S, Farsetti A, Gespach C, Londoño-Vallejo A, Groisman I. CPEB1 orchestrates a fine-tuning of miR-145-5p tumor-suppressive activity on TWIST1 translation in prostate cancer cells. Oncotarget 2020; 11:4155-4168. [PMID: 33227047 PMCID: PMC7665230 DOI: 10.18632/oncotarget.27806] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/27/2020] [Indexed: 11/25/2022] Open
Abstract
TWIST1 is a basic helix-loop-helix transcription factor, and one of the master Epithelial-to-Mesenchymal Transition (EMT) regulators. We show that tumor suppressor miR-145-5p controls TWIST1 expression in an immortalized prostate epithelial cell line and in a tumorigenic prostate cancer-derived cell line. Indeed, shRNA-mediated miR-145-5p silencing enhanced TWIST1 expression and induced EMT-associated malignant properties in these cells. However, we discovered that the translational inhibitory effect of miR-145-5p on TWIST1 is lost in 22Rv1, another prostate cancer cell line that intrinsically expresses high levels of the CPEB1 cytoplasmic polyadenylation element binding protein. This translational regulator typically reduces TWIST1 translation efficiency by shortening the TWIST1 mRNA polyA tail. However, our results indicate that the presence of CPEB1 also interferes with the binding of miR-145-5p to the TWIST1 mRNA 3′UTR. Mechanistically, CPEB1 binding to its first cognate site either directly hampers the access to the miR-145-5p response element or redirects the cleavage/polyadenylation machinery to an intermediate polyadenylation site, resulting in the elimination of the miR-145-5p binding site. Taken together, our data support the notion that the tumor suppressive activity of miR-145-5p on TWIST1 translation, consequently on EMT, self-renewal, and migration, depends on the CPEB1 expression status of the cancer cell. A preliminary prospective study using clinical samples suggests that reconsidering the relative status of miR-145-5p/TWIST1 and CPEB1 in the tumors of prostate cancer patients may bear prognostic value.
Collapse
Affiliation(s)
- Fatemeh Rajabi
- Telomeres and Cancer Laboratory, CNRS, Sorbonne Université, Université PSL, Institut Curie, Paris, France
| | - Win-Yan Liu-Bordes
- Telomeres and Cancer Laboratory, CNRS, Sorbonne Université, Université PSL, Institut Curie, Paris, France
| | - Marina Pinskaya
- Non-Coding RNA, Epigenetic and Genome Fluidity, Sorbonne Université, Université PSL, Institut Curie, Paris, France
| | - Foretek Dominika
- Non-Coding RNA, Epigenetic and Genome Fluidity, Sorbonne Université, Université PSL, Institut Curie, Paris, France
| | - Gueorgui Kratassiouk
- Plateforme ARN Interférence, Service de Biologie Intégrative et de Génétique Moléculaire (SBIGeM), Gif-sur-Yvette, France
| | - Guillaume Pinna
- Plateforme ARN Interférence, Service de Biologie Intégrative et de Génétique Moléculaire (SBIGeM), Gif-sur-Yvette, France
| | - Simona Nanni
- Istituto di Patologia Medica, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Antonella Farsetti
- Istituto di Biologia Cellulare e Neurobiologia, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Christian Gespach
- Sorbonne Université, Inserm U938, Team TGFβ Signaling in Cellular Plasticity and Cancer, Centre de Recherche Saint-Antoine, Paris, France
| | - Arturo Londoño-Vallejo
- Telomeres and Cancer Laboratory, CNRS, Sorbonne Université, Université PSL, Institut Curie, Paris, France
| | - Irina Groisman
- Telomeres and Cancer Laboratory, CNRS, Sorbonne Université, Université PSL, Institut Curie, Paris, France
| |
Collapse
|
4
|
Tamarindo GH, Góes RM. Docosahexaenoic acid differentially modulates the cell cycle and metabolism- related genes in tumor and pre-malignant prostate cells. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158766. [PMID: 32712248 DOI: 10.1016/j.bbalip.2020.158766] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/13/2020] [Accepted: 07/19/2020] [Indexed: 12/11/2022]
Abstract
Prostate cancer (PCa) has different molecular features along progression, including androgen profile, which is associated to therapy inefficiency leading to more aggressive phenotype. Docosahexaenoic acid (DHA) has antiproliferative and pro-apoptotic properties in different cancers associated to cell metabolism modulation. The latter is of particular interest since metabolic reprogramming is one of PCa hallmarks, but is not clear how this occurs among disease progression. Therefore, we evaluated DHA antiproliferative potential in distinct androgenic backgrounds associated to metabolism modulation and androgen-regulated genes. For this purpose, pre-malignant PNT1A and tumor AR-positive 22rv1, and AR-negative PC3 cells were incubated with DHA at 100 μM-48 h. DHA reduced at least 26% cell number for all lineages due to S-phase decrease in AR-positive and G2/M arrest in AR-negative. Mitochondrial metabolic rate decreased in PNT1A (~38%) and increased in tumor cells (at least 40%). This was associated with ROS overproduction (1.6-fold PNT1A; 2.1 22rv1; 2.2 PC3), lipid accumulation (3-fold PNT1A; 1.8 22rv1; 3.6 PC3) and mitochondria damage in all cell lines. AKT, AMPK and PTEN were not activated in any cell line, but p-ERK1/2 increased (1.5-fold) in PNT1A. Expression of androgen-regulated and nuclear receptors genes showed that DHA affected them in a distinct pattern in each cell line, but most converged to metabolism regulation, response to hormones, lipids and stress. In conclusion, regardless of androgenic or PTEN background DHA exerted antiproliferative effect associated to cell cycle impairment, lipid deregulation and oxidative stress, but differentially regulated gene expression probably due to distinct molecular features of each pathologic stage.
Collapse
Affiliation(s)
| | - Rejane Maira Góes
- Institute of Biology, University of Campinas, Campinas, SP, Brazil; Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP, Brazil.
| |
Collapse
|
5
|
Elevated miR-615-3p Expression Predicts Adverse Clinical Outcome and Promotes Proliferation and Migration of Prostate Cancer Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2377-2388. [DOI: 10.1016/j.ajpath.2019.08.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 12/11/2022]
|
6
|
Katsogiannou M, Boyer JB, Valdeolivas A, Remy E, Calzone L, Audebert S, Rocchi P, Camoin L, Baudot A. Integrative proteomic and phosphoproteomic profiling of prostate cell lines. PLoS One 2019; 14:e0224148. [PMID: 31675377 PMCID: PMC6824562 DOI: 10.1371/journal.pone.0224148] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/06/2019] [Indexed: 12/15/2022] Open
Abstract
Background Prostate cancer is a major public health issue, mainly because patients relapse after androgen deprivation therapy. Proteomic strategies, aiming to reflect the functional activity of cells, are nowadays among the leading approaches to tackle the challenges not only of better diagnosis, but also of unraveling mechanistic details related to disease etiology and progression. Methods We conducted here a large SILAC-based Mass Spectrometry experiment to map the proteomes and phosphoproteomes of four widely used prostate cell lines, namely PNT1A, LNCaP, DU145 and PC3, representative of different cancerous and hormonal status. Results We identified more than 3000 proteins and phosphosites, from which we quantified more than 1000 proteins and 500 phosphosites after stringent filtering. Extensive exploration of this proteomics and phosphoproteomics dataset allowed characterizing housekeeping as well as cell-line specific proteins, phosphosites and functional features of each cell line. In addition, by comparing the sensitive and resistant cell lines, we identified protein and phosphosites differentially expressed in the resistance context. Further data integration in a molecular network highlighted the differentially expressed pathways, in particular migration and invasion, RNA splicing, DNA damage repair response and transcription regulation. Conclusions Overall, this study proposes a valuable resource toward the characterization of proteome and phosphoproteome of four widely used prostate cell lines and reveals candidates to be involved in prostate cancer progression for further experimental validation.
Collapse
Affiliation(s)
- Maria Katsogiannou
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
- Obstetrics and Gynecology department, Hôpital Saint Joseph, Marseille, France
| | - Jean-Baptiste Boyer
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Alberto Valdeolivas
- Aix Marseille Univ, CNRS, Centrale Marseille, I2M, Marseille, France
- Aix Marseille Univ, INSERM, MMG, Marseille, France
- ProGeLife, Marseille, France
| | - Elisabeth Remy
- Aix Marseille Univ, CNRS, Centrale Marseille, I2M, Marseille, France
| | - Laurence Calzone
- Mines Paris Tech, Institut Curie, PSL Research University, Paris, France
| | - Stéphane Audebert
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Palma Rocchi
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
- * E-mail: (PR); (LC); (AB)
| | - Luc Camoin
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
- * E-mail: (PR); (LC); (AB)
| | - Anaïs Baudot
- Aix Marseille Univ, CNRS, Centrale Marseille, I2M, Marseille, France
- Aix Marseille Univ, INSERM, MMG, Marseille, France
- * E-mail: (PR); (LC); (AB)
| |
Collapse
|
7
|
Melatonin and Docosahexaenoic Acid Decrease Proliferation of PNT1A Prostate Benign Cells via Modulation of Mitochondrial Bioenergetics and ROS Production. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5080798. [PMID: 30728886 PMCID: PMC6343140 DOI: 10.1155/2019/5080798] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/20/2018] [Accepted: 09/18/2018] [Indexed: 12/17/2022]
Abstract
Prostate cancer development has been associated with changes in mitochondrial activity and reactive oxygen species (ROS) production. Melatonin (MLT) and docosahexaenoic acid (DHA) have properties to modulate both, but their protective role, mainly at early stages of prostate cancer, remains unclear. In this study, the effects of MLT and DHA, combined or not, on PNT1A cells with regard to mitochondria bioenergetics, ROS production, and proliferation-related pathways were examined. Based on dose response and lipid accumulation assays, DHA at 100 μM and MLT at 1 μM for 48 h were chosen. DHA doubled and MLT reduced (40%) superoxide anion production, but coincubation (DM) did not normalize to control. Hydrogen peroxide production decreased after MLT incubation only (p < 0.01). These alterations affected the area and perimeter of mitochondria, since DHA increased whereas MLT decreased, but such hormone has no effect on coincubation. DHA isolated did not change the oxidative phosphorylation rate (OXPHOS), but decreased (p < 0.001) the mitochondrial bioenergetic reserve capacity (MBRC) which is closely related to cell responsiveness to stress conditions. MLT, regardless of DHA, ameliorated OXPHOS and recovered MBRC after coincubation. All incubations decreased AKT phosphorylation; however, only MLT alone inhibited p-mTOR. MLT increased p-ERK1/2 and, when combined to DHA, increased GSTP1 expression (p < 0.01). DHA did not change the testosterone levels in the medium, whereas MLT alone or coincubated decreased by about 20%; however, any incubation affected AR expression. Moreover, incubation with luzindole revealed that MLT effects were MTR1/2-independent. In conclusion, DHA increased ROS production and impaired mitochondrial function which was probably related to AKT inactivation; MLT improved OXPHOS and decreased ROS which was related to AKT/mTOR dephosphorylation, and when coincubated, the antiproliferative action was related to mitochondrial bioenergetic modulation associated to AKT and ERK1/2 regulation. Together, these findings point to the potential application of DHA and MLT towards the prevention of proliferative prostate diseases.
Collapse
|
8
|
Domińska K, Ochędalski T, Kowalska K, Matysiak-Burzyńska ZE, Płuciennik E, Piastowska-Ciesielska AW. A common effect of angiotensin II and relaxin 2 on the PNT1A normal prostate epithelial cell line. J Physiol Biochem 2016; 72:381-92. [PMID: 27119161 DOI: 10.1007/s13105-016-0489-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/15/2016] [Indexed: 12/12/2022]
Abstract
The prostate gland is a part of the male reproductive tract which produces both angiotensin II (Ang II) and relaxin 2 (RLN2). The present study analyzes the effect of both these peptide hormones at concentration 10(-8)M on viability, proliferation, adhesion, migration, and invasion of normal prostate epithelial cells (PNT1A). Improved survival in two- and three-dimensional cell cultures was noted as well as visual changes in colony size and structure in Geltrex™. Stimulatory influence on cell viability of each peptide applied single was lower than in combination. Enhanced survival of PNT1A cells appears to be associated with increased BCL2/BAX messenger RNA (mRNA) expression ratio. Modulation of cell spreading and cell-extracellular matrix adhesion dynamics were also altered as an influence of tested hormone application. However, long-term Ang II and RLN2 effects may lead to an increase of normal prostate cell migration and invasion abilities. Moreover, gelatin zymography revealed that both gelatinases A and B were augmented by Ang II treatment, whereas RLN2 significantly stimulated only MMP-9 secretion. These results support the hypothesis that deregulation of locally secreted peptide hormones such as Ang II and RLN2 may take part in the development of certain cancers, including prostate cancer. Moreover, the observed ability of relaxin 2 to act as a regulator of mRNA expression levels not only LGR7 but also classic angiotensin receptors suggested that renin-angiotensin system and relaxin family peptide system are functionally linked.
Collapse
Affiliation(s)
- Kamila Domińska
- Department of Comparative Endocrinology, Medical University of Lodz, ul. Zeligowskiego 7/9, 90-752, Lodz, Poland.
| | - Tomasz Ochędalski
- Department of Comparative Endocrinology, Medical University of Lodz, ul. Zeligowskiego 7/9, 90-752, Lodz, Poland
| | - Karolina Kowalska
- Department of Comparative Endocrinology, Medical University of Lodz, ul. Zeligowskiego 7/9, 90-752, Lodz, Poland
| | - Zuzanna E Matysiak-Burzyńska
- Department of Comparative Endocrinology, Medical University of Lodz, ul. Zeligowskiego 7/9, 90-752, Lodz, Poland
| | - Elżbieta Płuciennik
- Department of Molecular Cancerogenesis, Medical University of Lodz, ul. Zeligowskiego 7/9, 90-752, Lodz, Poland
| | | |
Collapse
|
9
|
Zhang L, Shao L, Creighton CJ, Zhang Y, Xin L, Ittmann M, Wang J. Function of phosphorylation of NF-kB p65 ser536 in prostate cancer oncogenesis. Oncotarget 2016; 6:6281-94. [PMID: 25749044 PMCID: PMC4467437 DOI: 10.18632/oncotarget.3366] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/12/2015] [Indexed: 01/11/2023] Open
Abstract
Majority of prostate cancer (PCa) patients carry TMPRSS2/ERG (T/E) fusion genes and there has been tremendous interest in understanding how the T/E fusion may promote progression of PCa. We showed that T/E fusion can activate NF-kB pathway by increasing phosphorylation of NF-kB p65 Ser536 (p536), but the function of p536 has never been studied in PCa. We report here that active p536 can significantly increase cell motility and transform PNT1a cells (an immortalized normal cell line), suggesting p536 plays a critical role in promoting PCa tumorigenesis. We have discovered a set of p536 regulated genes, among which we validated the regulation of CCL2 by p536. Based on all evidence, we favor that T/E fusion, NF-kB p536 and CCL2 form a signaling chain. Finally, PNT1a cells (not tumorigenic) can form tumors in SCID mice when overexpressing of either wild type or active p65 in the presence of activated AKT, demonstrating synergistic activities of NF-kB and AKT signals in promoting PCa tumorigenesis. These findings indicate that combination therapies targeting T/E fusion, NF-kB, CCL2 and/or AKT pathways may have efficacy in T/E fusion gene expressing PCa. If successful, such targeted therapy will benefit more than half of PCa patients who carry T/E fusions.
Collapse
Affiliation(s)
- Li Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China.,Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Deptartment of Veterans Affairs Medical Center, Houston, Texas, USA
| | - Longjiang Shao
- Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Deptartment of Veterans Affairs Medical Center, Houston, Texas, USA
| | - Chad J Creighton
- Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Yiqun Zhang
- Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas, USA
| | - Li Xin
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Michael Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Deptartment of Veterans Affairs Medical Center, Houston, Texas, USA
| | - Jianghua Wang
- Department of Pathology and Immunology, Baylor College of Medicine and Michael E. DeBakey Deptartment of Veterans Affairs Medical Center, Houston, Texas, USA
| |
Collapse
|
10
|
Amirbeagi F, Thulin P, Pullerits R, Pedersen B, Andersson BA, Dahlgren C, Welin A, Bylund J. Olfactomedin-4 autoantibodies give unusual c-ANCA staining patterns with reactivity to a subpopulation of neutrophils. J Leukoc Biol 2015; 97:181-189. [PMID: 25387833 DOI: 10.1189/jlb.5a0614-311r] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2024] Open
Abstract
Testing for the presence of ANCAs in circulation is part of the clinical examinations routinely performed upon suspected autoimmune disorders, mainly vasculitis. The autoantibodies are typically directed toward neutrophil MPO or PR3. These are major granule-localized proteins, and similar to all hitherto-described ANCA antigens, they are expressed by all neutrophils, and ANCA-containing sera thus give rise to uniform reactivity toward all neutrophils in a sample. In this paper, we describe sera from 2 unrelated patients with diffuse inflammatory symptoms that gave rise to peculiar c-ANCA patterns, only reacting with a subpopulation (roughly 30%) of human neutrophils. By immunoblotting, both sera reacted to the same antigen, which was expressed in intracellular granules. The antigen could be released to the extracellular milieu through secretion but also through the formation of NETs. Neutrophils have long been considered a homogenous cell population, but it is becoming increasingly clear that distinct subpopulations, defined by the presence or absence of certain proteins, exist. One such marker that defines a neutrophil subset is the granule protein OLFM4. The unusual, subset-restricted c-ANCA sera reacted only with OLFM4-positive neutrophils, and MS analysis revealed that the autoantigen was, in fact, OLFM4. These data describe for the first time a c-ANCA pattern reactive to only a subpopulation of neutrophils and identify the granule protein OLFM4 as a novel autoantigen.
Collapse
Affiliation(s)
- Firoozeh Amirbeagi
- Departments of *Rheumatology and Inflammation Research and Oral Microbiology and Immunology, Sahlgrenska Academy at University of Gothenburg, Sweden; Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden; and Department of Pulmonary Medicine, Norra Älvsborg County Hospital, Trollhättan, Sweden
| | - Pontus Thulin
- Departments of *Rheumatology and Inflammation Research and Oral Microbiology and Immunology, Sahlgrenska Academy at University of Gothenburg, Sweden; Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden; and Department of Pulmonary Medicine, Norra Älvsborg County Hospital, Trollhättan, Sweden
| | - Rille Pullerits
- Departments of *Rheumatology and Inflammation Research and Oral Microbiology and Immunology, Sahlgrenska Academy at University of Gothenburg, Sweden; Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden; and Department of Pulmonary Medicine, Norra Älvsborg County Hospital, Trollhättan, Sweden
| | - Bo Pedersen
- Departments of *Rheumatology and Inflammation Research and Oral Microbiology and Immunology, Sahlgrenska Academy at University of Gothenburg, Sweden; Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden; and Department of Pulmonary Medicine, Norra Älvsborg County Hospital, Trollhättan, Sweden
| | - Bengt A Andersson
- Departments of *Rheumatology and Inflammation Research and Oral Microbiology and Immunology, Sahlgrenska Academy at University of Gothenburg, Sweden; Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden; and Department of Pulmonary Medicine, Norra Älvsborg County Hospital, Trollhättan, Sweden
| | - Claes Dahlgren
- Departments of *Rheumatology and Inflammation Research and Oral Microbiology and Immunology, Sahlgrenska Academy at University of Gothenburg, Sweden; Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden; and Department of Pulmonary Medicine, Norra Älvsborg County Hospital, Trollhättan, Sweden
| | - Amanda Welin
- Departments of *Rheumatology and Inflammation Research and Oral Microbiology and Immunology, Sahlgrenska Academy at University of Gothenburg, Sweden; Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden; and Department of Pulmonary Medicine, Norra Älvsborg County Hospital, Trollhättan, Sweden
| | - Johan Bylund
- Departments of *Rheumatology and Inflammation Research and Oral Microbiology and Immunology, Sahlgrenska Academy at University of Gothenburg, Sweden; Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden; and Department of Pulmonary Medicine, Norra Älvsborg County Hospital, Trollhättan, Sweden
| |
Collapse
|
11
|
Borkowska A, Knap N, Antosiewicz J. Diallyl trisulfide is more cytotoxic to prostate cancer cells PC-3 than to noncancerous epithelial cell line PNT1A: a possible role of p66Shc signaling axis. Nutr Cancer 2014; 65:711-7. [PMID: 23859039 DOI: 10.1080/01635581.2013.789115] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Diallyl trisulfide (DATS) is an organosulfur compound isolated from garlic, and has been shown to have anticancer activity both in vitro and in vivo. The aim of this study was to compare cytotoxic effects of DATS on prostate cancer cells PC-3 and noncancerous human prostate epithelial cells PNT1A. PC-3 prostate cancer and noncancerous human prostate epithelial cells PNT1A were used in the study. We observed that PNT1A cells had higher resistance to DATS-induced cell death than PC-3 cells. Investigating signaling pathways involved in the cell death we observed that p66Shc phosphorylation at serine 36 and extracellular signal-regulated kinase 1/2 activation induced by DATS, were significantly attenuated in PNT1A cells as compared to PC-3 cells. Moreover, DATS-induced Akt inactivation was also significantly reduced in PNT1A cells. In addition to that, DATS-induced reactive oxygen species generation was nearly completely abolished in PNT1A cells. Interestingly, DATS induced only slight decrease in the level of ferritin H, whereas ferritin L was elevated. These data suggest that cytotoxicity of DATS toward PNT1A cells is strongly reduced as opposed to PC-3 cancer cells, which corresponds to the lower activation of prodeath signaling pathway mediated by the adaptor protein p66Shc in the noncancerous PNT1A cells.
Collapse
Affiliation(s)
- Andzelika Borkowska
- Department of Bioenergetics and Physiology of Exercise, Medical University of Gdansk, Gdansk, Poland
| | | | | |
Collapse
|
12
|
Huang M, Narita S, Inoue T, Tsuchiya N, Satoh S, Nanjo H, Sasaki T, Habuchi T. Diet-induced macrophage inhibitory cytokine 1 promotes prostate cancer progression. Endocr Relat Cancer 2014; 21:39-50. [PMID: 24344250 DOI: 10.1530/erc-13-0227] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Recent studies have indicated that a high-fat diet (HFD) plays an important role in prostate cancer (PCa) progression. Palmitic acid (PA) is one of the most abundant saturated free fatty acids (FAs) and is associated with carcinogenesis. In this study, we investigated the mechanism underlying the association of dietary fat, including PA, with PCa progression. In four PCa cell lines, in vitro PA administration stimulated the expression of macrophage inhibitory cytokine 1 (MIC1), which is a divergent member of the transforming growth factor-β family. In vivo, LNCaP xenograft tumor growth, serum MIC1 levels, and FA levels in xenograft tumors were significantly higher in mice receiving an HFD containing high amounts of PA than in those receiving a low-fat diet (LFD). In addition, tumor cells with high MIC1 expression invaded to venules and lymph vessels in the LNCaP xenograft. In vitro studies showed that proliferation and invasive capacity were significantly higher in PCa cells cultured with serum from HFD-fed mice than in those cultured with the serum from LFD-fed mice. This effect was attenuated by the addition of neutralizing antibodies against MIC1, but not by isotype control antibodies. Clinically, serum MIC1 levels were significantly higher in PCa patients than in healthy controls, and higher levels were associated with higher pathological grade and obesity. In conclusion, our results indicate that an HFD containing PA may promote growth and invasiveness of PCa cells through the upregulation of MIC1 expression.
Collapse
Affiliation(s)
- Mingguo Huang
- Department of Urology Research Center for Biosignal Department of Clinical Pathology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan CREST, Japan Science and Technology Agency (JST), Tokyo 102-0076, Japan
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Rizzi F, Naponelli V, Silva A, Modernelli A, Ramazzina I, Bonacini M, Tardito S, Gatti R, Uggeri J, Bettuzzi S. Polyphenon E(R), a standardized green tea extract, induces endoplasmic reticulum stress, leading to death of immortalized PNT1a cells by anoikis and tumorigenic PC3 by necroptosis. Carcinogenesis 2013; 35:828-39. [PMID: 24343359 DOI: 10.1093/carcin/bgt481] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Increasing doses of Polyphenon E®, a standardized green tea extract, were given to PNT1a and PC3 prostate epithelial cells mimicking initial and advanced stages of prostate cancer (PCa), respectively. Cell death occurred in both cell lines, with PNT1a being more sensitive [half-maximal inhibitory concentration (IC50) = 35 μg/ml] than PC3 (IC50 = 145 μg/ml) to Polyphenon E®. Cell cycle arrest occurred at G0/G1 checkpoint for PNT1a, and G2/M for PC3 cells. Endoplasmic reticulum stress (ERS) and unfolded protein response (UPR) occurred in both cell lines, with each exhibiting different timing in response to Polyphenon E®. Autophagy was transiently activated in PNT1a cells within 12 h after treatment as a survival response to overcome ERS; then activation of caspases and cleavage of poly (ADP ribose) polymerase 1 occurred, committing cells to anoikis death. Polyphenon E® induced severe ERS in PC3 cells, causing a dramatic enlargement of the ER; persistent activation of UPR produced strong upregulation of GADD153/CHOP, a key protein of ERS-mediated cell death. Thereafter, GADD153/CHOP activated Puma, a BH3-only protein, committing cells to necroptosis, a programmed caspase-independent mechanism of cell death. Our results provide a foundation for the identification of novel targets and strategies aimed at sensitizing apoptosis-resistant cells to alternative death pathways.
Collapse
Affiliation(s)
- Federica Rizzi
- Department of Biomedicine, Biotechnology and Translational Research and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Four microRNAs promote prostate cell proliferation with regulation of PTEN and its downstream signals in vitro. PLoS One 2013; 8:e75885. [PMID: 24098737 PMCID: PMC3787937 DOI: 10.1371/journal.pone.0075885] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/19/2013] [Indexed: 01/03/2023] Open
Abstract
Background Phosphatase and tensin homologue (PTEN), as a tumor suppressor, plays vital roles in tumorigenesis and progression of prostate cancer. However, the mechanisms of PTEN regulation still need further investigation. We here report that a combination of four microRNAs (miR-19b, miR-23b, miR-26a and miR-92a) promotes prostate cell proliferation by regulating PTEN and its downstream signals invitro. Methodology/Principal Findings We found that the four microRNAs (miRNAs) could effectively suppress PTEN expression by directly interacting with its 3’ UTR in prostate epithelial and cancer cells. Under-expression of the four miRNAs by antisense neutralization up-regulates PTEN expression, while overexpression of the four miRNAs accelerates epithelial and prostate cancer cell proliferation. Furthermore, the expression of the four miRNAs could, singly or jointly, alter the expression of the key components in the phosphoinositide 3-kinase (PI3K)/Akt pathway, including PIK3CA, PIK3CD, PIK3R1 and Akt, along with their downstream signal, cyclin D1. Conclusions These results suggested that the four miRNAs could promote prostate cancer cell proliferation by co-regulating the expression of PTEN, PI3K/Akt pathway and cyclin D1 invitro. These findings increase understanding of the molecular mechanisms of prostate carcinogenesis and progression, even provide valuable insights into the diagnosis, prognosis, and rational design of novel therapeutics for prostate cancer.
Collapse
|
15
|
TRPM8 ion channels differentially modulate proliferation and cell cycle distribution of normal and cancer prostate cells. PLoS One 2012; 7:e51825. [PMID: 23251635 PMCID: PMC3522609 DOI: 10.1371/journal.pone.0051825] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 11/06/2012] [Indexed: 11/19/2022] Open
Abstract
Overexpression of the cation-permeable channel TRPM8 in prostate cancers might represent a novel opportunity for their treatment. Inhibitors of TRPM8 reduce the growth of prostate cancer cells. We have used two recently described and highly specific blockers, AMTB and JNJ41876666, and RNAi to determine the relevance of TRPM8 expression in the proliferation of non-tumor and tumor cells. Inhibition of the expression or function of the channel reduces proliferation rates and proliferative fraction in all tumor cells tested, but not of non-tumor prostate cells. We observed no consistent acceleration of growth after stimulation of the channel with menthol or icilin, indicating that basal TRPM8 expression is enough to sustain growth of prostate cancer cells.
Collapse
|
16
|
Gingaras C, Danielson BP, Vigil KJ, Vey E, Arduino RC, Kimata JT. Absence of XMRV in peripheral blood mononuclear cells of ARV-treatment naïve HIV-1 infected and HIV-1/HCV coinfected individuals and blood donors. PLoS One 2012; 7:e31398. [PMID: 22348082 PMCID: PMC3278456 DOI: 10.1371/journal.pone.0031398] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 01/06/2012] [Indexed: 11/19/2022] Open
Abstract
Background Xenotropic murine leukemia virus-related virus (XMRV) has been found in the prostatic tissue of prostate cancer patients and in the blood of chronic fatigue syndrome patients. However, numerous studies have found little to no trace of XMRV in different human cohorts. Based on evidence suggesting common transmission routes between XMRV and HIV-1, HIV-1 infected individuals may represent a high-risk group for XMRV infection and spread. Methodology/Principal Findings DNA was isolated from the peripheral blood mononuclear cells (PBMCs) of 179 HIV-1 infected treatment naïve patients, 86 of which were coinfected with HCV, and 54 healthy blood donors. DNA was screened for XMRV provirus with two sensitive, published PCR assays targeting XMRV gag and env and one sensitive, published nested PCR assay targeting env. Detection of XMRV was confirmed by DNA sequencing. One of the 179 HIV-1 infected patients tested positive for gag by non-nested PCR whereas the two other assays did not detect XMRV in any specimen. All healthy blood donors were negative for XMRV proviral sequences. Sera from 23 HIV-1 infected patients (15 HCV+) and 12 healthy donors were screened for the presence of XMRV-reactive antibodies by Western blot. Thirteen sera (57%) from HIV-1+ patients and 6 sera (50%) from healthy donors showed reactivity to XMRV-infected cell lysate. Conclusions/Significance The virtual absence of XMRV in PBMCs suggests that XMRV is not associated with HIV-1 infected or HIV-1/HCV coinfected patients, or blood donors. Although we noted isolated incidents of serum reactivity to XMRV, we are unable to verify the antibodies as XMRV specific.
Collapse
Affiliation(s)
- Cosmina Gingaras
- Section of Retrovirology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Baylor International Pediatric AIDS Initiative, Baylor College of Medicine, Houston, Texas
| | - Bryan P. Danielson
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Karen J. Vigil
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center, Houston, Texas
| | - Elana Vey
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center, Houston, Texas
| | - Roberto C. Arduino
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center, Houston, Texas
| | - Jason T. Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
- * E-mail:
| |
Collapse
|
17
|
Ye X, Lee YC, Choueiri M, Chu K, Huang CF, Tsai WW, Kobayashi R, Logothetis CJ, Yu-Lee LY, Lin SH. Aberrant expression of katanin p60 in prostate cancer bone metastasis. Prostate 2012; 72:291-300. [PMID: 21681775 PMCID: PMC3179562 DOI: 10.1002/pros.21431] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 05/09/2011] [Indexed: 12/27/2022]
Abstract
BACKGROUND Katanin p60 is a microtubule-severing protein and is involved in microtubule cytoskeleton organization in both mitotic and non-mitotic processes. Its role in cancer metastasis is unknown. METHODS Differential protein profiles of bone marrow aspirates were analyzed by chromatography, electrophoresis, and mass spectrometry. Expression of katanin p60 in primary and metastatic prostate cancer was examined by immunohistochemistry. Cellular function of katanin p60 was further examined in prostate cell lines. RESULTS In a proteomic profiling of bone marrow aspirates from men with prostate cancer, we found that katanin p60 was one of the proteins differentially expressed in bone metastasis samples. Immunohistochemical staining showed that katanin p60 was expressed in the basal cells in normal human prostate glands. In prostatic adenocarcinomas, in which the basal cells were absent, katanin p60 was expressed in the prostate cancer cells. In the specimens from bone metastasis, katanin p60 was detectable in the metastatic cancer cells. Strikingly, some of the metastatic cancer cells also co-expressed basal cell biomarkers including the tumor suppressor p53-homologous protein p63 and the high molecular weight cytokeratins, suggesting that the metastatic prostate cancer cells may have a basal cell-like phenotype. Moreover, overexpression of katanin p60 inhibited prostate cancer cell proliferation but enhanced cell migration activity. CONCLUSIONS Katanin p60 was aberrantly expressed during prostate cancer progression. Its expression in the metastatic cells in bone was associated with the re-emergence of a basal cell-like phenotype. The elevated katanin p60 expression may contribute to cancer cell metastasis via a stimulatory effect on cell motility.
Collapse
Affiliation(s)
- Xiangcang Ye
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Yu-Chen Lee
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Michel Choueiri
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Khoi Chu
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Chih-Fen Huang
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Wen-Wei Tsai
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Ryuji Kobayashi
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Christopher J. Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Li-Yuan Yu-Lee
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Sue-Hwa Lin
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
- Department of Genitourinary Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
18
|
Wedel S, Hudak L, Seibel JM, Makarević J, Juengel E, Tsaur I, Waaga-Gasser A, Haferkamp A, Blaheta RA. Molecular targeting of prostate cancer cells by a triple drug combination down-regulates integrin driven adhesion processes, delays cell cycle progression and interferes with the cdk-cyclin axis. BMC Cancer 2011; 11:375. [PMID: 21867506 PMCID: PMC3170298 DOI: 10.1186/1471-2407-11-375] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 08/25/2011] [Indexed: 11/24/2022] Open
Abstract
Background Single drug use has not achieved satisfactory results in the treatment of prostate cancer, despite application of increasingly widespread targeted therapeutics. In the present study, the combined impact of the mammalian target of rapamycin (mTOR)-inhibitor RAD001, the dual EGFr and VGEFr tyrosine kinase inhibitor AEE788 and the histone deacetylase (HDAC)-inhibitor valproic acid (VPA) on prostate cancer growth and adhesion in vitro was investigated. Methods PC-3, DU-145 and LNCaP cells were treated with RAD001, AEE788 or VPA or with a RAD-AEE-VPA combination. Tumor cell growth, cell cycle progression and cell cycle regulating proteins were then investigated by MTT-assay, flow cytometry and western blotting, respectively. Furthermore, tumor cell adhesion to vascular endothelium or to immobilized extracellular matrix proteins as well as migratory properties of the cells was evaluated, and integrin α and β subtypes were analyzed. Finally, effects of drug treatment on cell signaling pathways were determined. Results All drugs, separately applied, reduced tumor cell adhesion, migration and growth. A much stronger anti-cancer effect was evoked by the triple drug combination. Particularly, cdk1, 2 and 4 and cyclin B were reduced, whereas p27 was elevated. In addition, simultaneous application of RAD001, AEE788 and VPA altered the membranous, cytoplasmic and gene expression pattern of various integrin α and β subtypes, reduced integrin-linked kinase (ILK) and deactivated focal adhesion kinase (FAK). Signaling analysis revealed that EGFr and the downstream target Akt, as well as p70S6k was distinctly modified in the presence of the drug combination. Conclusions Simultaneous targeting of several key proteins in prostate cancer cells provides an advantage over targeting a single pathway. Since strong anti-tumor properties became evident with respect to cell growth and adhesion dynamics, the triple drug combination might provide progress in the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Steffen Wedel
- Department of Urology, Goethe-University, Frankfurt am Main, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Morgan K, Stavrou E, Leighton SP, Miller N, Sellar R, Millar RP. Elevated GnRH receptor expression plus GnRH agonist treatment inhibits the growth of a subset of papillomavirus 18-immortalized human prostate cells. Prostate 2011; 71:915-28. [PMID: 21541969 DOI: 10.1002/pros.21308] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 10/18/2010] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND AIMS Human metastatic prostate cancer cell growth can be inhibited by GnRH analogs but effects on virus-immortalized prostate cells have not been investigated. METHODS Virus-immortalized prostate cells were stably transfected with rat GnRH receptor cDNA and levels of GnRH binding were correlated with GnRH effects on signaling, cell cycle, growth, exosome production, and apoptosis. RESULTS High levels of cell surface GnRH receptor occurred in transfected papillomavirus-immortalized WPE-1-NB26 epithelial cells but not in non-tumourigenic RWPE-1, myoepithelial WPMY-1 cells, or SV40-immortalized PNT1A. Endogenous cell surface GnRH receptor was undetectable in non-transfected cells or cancer cell lines LNCaP, PC3, and DU145. GnRH receptor levels correlated with induction of inositol phosphates, elevation of intracellular Ca(2+) , cytoskeletal actin reorganization, modulation of ERK activation and cell growth-inhibition with GnRH agonists. Hoechst 33342 DNA staining-cell sorting indicated accumulation of cells in G2 following agonist treatment. Release of exosomes from transfected WPE-1-NB26 was unaffected by agonists, unlike induction observed in HEK293([SCL60]) cells. Increased PARP cleavage and apoptotic body production were undetectable during growth-inhibition in WPE-1-NB26 cells, contrasting with HEK293([SCL60]) . EGF receptor activation inhibited GnRH-induced ERK activation in WPE-1-NB26 but growth-inhibition was not rescued by EGF or PKC inhibitor Ro320432. Growth of cells expressing low levels of GnRH receptor was not affected by agonists. CONCLUSIONS Engineered high-level GnRH receptor activation inhibits growth of a subset of papillomavirus-immortalized prostate cells. Elucidating mechanisms leading to clone-specific differences in cell surface GnRH receptor levels is a valuable next step in developing strategies to exploit prostate cell anti-proliferation using GnRH agonists.
Collapse
Affiliation(s)
- Kevin Morgan
- Medical Research Council Human Reproductive Sciences Unit, The Queen's Medical Research Institute, Little France Crescent, Edinburgh, UK.
| | | | | | | | | | | |
Collapse
|
20
|
Vestergaard EM, Nexø E, Tørring N, Borre M, Ørntoft TF, Sørensen KD. Promoter hypomethylation and upregulation of trefoil factors in prostate cancer. Int J Cancer 2010; 127:1857-65. [PMID: 20112343 DOI: 10.1002/ijc.25209] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Trefoil factors, mucin-associated peptides, are overexpressed in prostate cancer (PC). We hypothesized that promoter methylation contributes to the regulation of trefoil factors (TFF1, TFF2 and TFF3) in human prostate cells. Here we show hypomethylation of promoter regions of TFF1 and TFF3 in PC cell lines with significant TFF expression as compared to benign immortalized prostate cell lines and PC cell lines not expressing trefoil factor. The most striking difference was observed for CpG sites located close to the AUG start codon overlapping several putative binding sites for cellular transcription factors. TFF2 was hypermethylated and had no or very low expression in all prostate cell lines investigated. Treatment of methylated cell lines with 5-aza-2'-deoxycytidine restored TFF expression in cell lines not expressing TFF and increased expression significantly in low-expressing cell lines. In clinical samples, methylation of the promoter/enhancer regions of TFF1 and TFF3 was significantly lower in PC compared to benign prostatic hyperplasia. The present study shows an inverse relation between promoter methylation and expression of trefoil factors. Preliminary analysis on clinical samples suggests that this regulatory mechanism is responsible for the increased levels of TFF1 and TFF3 observed in PC. The overexpression and promoter hypomethylation of trefoil factors may serve as biomarkers in PC.
Collapse
Affiliation(s)
- Else Marie Vestergaard
- Department of Clinical Biochemistry, Aarhus University Hospital Skejby, Brendstrupgaardsvej, Aarhus N, Denmark.
| | | | | | | | | | | |
Collapse
|
21
|
Transcriptional and translational dual-regulated oncolytic herpes simplex virus type 1 for targeting prostate tumors. Mol Ther 2010; 18:929-35. [PMID: 20179676 DOI: 10.1038/mt.2010.26] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The aim of this project was to demonstrate that an oncolytic herpes simplex virus type 1 (HSV-1) can replicate in a tissue- and tumor-specific fashion through both transcriptional (prostate-specific promoter, ARR(2)PB) and translational (5'-untranslated regions (5'UTRs) of rFGF-2) regulation of an essential viral gene, ICP27. We generated two recombinant viruses, ARR(2)PB-ICP27 (A27) and ARR(2)PB-5'UTR-ICP27 (AU27) and tested their efficacy and toxicity both in vitro and in vivo. The ARR(2)PB promoter caused overexpression of ICP27 gene in the presence of activated androgen receptors (ARs) and increased viral replication in prostate cells. However, this transcriptional upregulation was effectively constrained by the 5'UTR-mediated translational regulation. Mice bearing human prostate LNCaP tumors, treated with a single intravenous injection of 5 x 10(7) plaque-forming units (pfu) of AU27 virus exhibited a >85% reduction in tumor size at day 28 after viral injection. Although active viral replication was readily evident in the tumors, no viral DNA was detectable in normal organs as measured by real-time PCR analyses. In conclusion, a transcriptional and translational dual-regulated (TTDR) viral essential gene expression can increase both viral lytic activity and tumor specificity, and this provides a basis for the development of a novel tumor-specific oncolytic virus for systemic treatment of locally advanced and metastatic prostate cancers.
Collapse
|
22
|
Heebøll S, Borre M, Ottosen PD, Dyrskjøt L, Orntoft TF, Tørring N. Snail1 is over-expressed in prostate cancer. APMIS 2009; 117:196-204. [PMID: 19245592 DOI: 10.1111/j.1600-0463.2008.00007.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transcription factor Snail1 is a mediator of cell migration and survival, and expression is elevated in several cancer types. The Snail1 gene is reportedly amplified in prostate cancer (PC), and we investigated Snail1 expression in PC. Immunohistochemical Snail1 staining was determined on a tissue microarray which includes 327 specimens of PC, 30 specimens from patients with benign prostatic hyperplasia (BPH), benign tissue from 30 PC patients and 15 high-grade prostate intraepithelial neoplasia (high-grade PIN) specimens. Clinicopathological and follow-up data were available for all patients. No BPH specimen and only 21% of benign tissue from PC patients showed high expression of Snail1. Only 7% of high-grade PIN patients expressed a high level of Snail1. In contrast, approximately 50% of PC tissue from patients with PC showed marked nuclear immunostaining. Snail1 immunostaining was significantly associated with Gleason score (p<0.05). Snail1 expression was not correlated to T stage, metastasis at time of diagnosis, risk of or time to recurrence. Snail1 expression was significantly increased in PC with a positive correlation to dedifferentiation, but not to cancer progression or prognosis. The presented data indicate that Snail1 expression is upregulated from the early stages of PC.
Collapse
Affiliation(s)
- Sara Heebøll
- Molecular Diagnostic Laboratory, Department of Clinical Biochemistry, Aarhus University Hospital, Skejby, Brenstrupgaardsvej 100, Aarhus N, Denmark.
| | | | | | | | | | | |
Collapse
|
23
|
Lechner D, Kállay E, Cross HS. 1alpha,25-dihydroxyvitamin D3 downregulates CYP27B1 and induces CYP24A1 in colon cells. Mol Cell Endocrinol 2007; 263:55-64. [PMID: 17029768 DOI: 10.1016/j.mce.2006.08.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Revised: 08/21/2006] [Accepted: 08/21/2006] [Indexed: 12/11/2022]
Abstract
The antimitotic and prodifferentiating 1alpha,25-dihydroxyvitamin D3 (1alpha,25-(OH)2D3), synthesized at various extrarenal sites could potentially prevent sporadic tumor development. Physiological regulation of extrarenal Vitamin D hydroxylases following tissue accumulation of 1alpha,25-(OH)2D3 is unknown. We therefore investigated basal and Vitamin D-regulated expression and activity of the synthesizing (CYP27B1) and metabolizing (CYP24A1) hydroxylase in three cell lines derived from the colon, and compared this to cells from the prostate and mammary gland. Our results show that all cells, irrespective of origin and differentiation, express CYP27B1 mRNA, whereas basal CYP24A1 mRNA is highly expressed only in undifferentiated cells. Treatment with 1alpha,25-(OH)2D3 diminishes CYP27B1 and Vitamin D receptor mRNA expression, but elevates CYP24A1 mRNA to equal levels in all cells. As shown by HPLC, CYP27B1 is active only if basal 24-hydroxylation is not maximally functional. In turn, accumulation of 1alpha,25-(OH)2D3 will induce 24-hydroxylation. We conclude that, although extrarenal and renal metabolic pathways for Vitamin D are similar, malignancy of tumor cells determines extent of Vitamin D catabolism.
Collapse
Affiliation(s)
- Daniel Lechner
- Department of Pathophysiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | | | | |
Collapse
|
24
|
Sørensen KD, Borre M, Ørntoft TF, Dyrskjøt L, Tørring N. Chromosomal deletion, promoter hypermethylation and downregulation ofFYN in prostate cancer. Int J Cancer 2007; 122:509-19. [DOI: 10.1002/ijc.23136] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
25
|
Moro L, Arbini AA, Marra E, Greco M. Down-regulation of BRCA2 expression by collagen type I promotes prostate cancer cell proliferation. J Biol Chem 2005; 280:22482-91. [PMID: 15805113 DOI: 10.1074/jbc.m414091200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BRCA2 is a tumor suppressor gene that when mutated confers an increased susceptibility to developing breast and prostate carcinoma. Besides its role in mediating DNA repair, new evidence suggests that BRCA2 may also play a role in suppressing cancer cell growth. Because altered interactions between neoplastic cells and the surrounding extracellular matrix (ECM) play a pivotal role in unchecked cancer cell proliferation and metastatic progression, we hypothesized that the ECM may have an effect in BRCA2 expression. By using normal and prostate carcinoma cell lines, we demonstrated that although normal cells transiently increase BRCA2 protein levels when adhering to the ECM protein collagen type I (COL1), carcinoma cells exhibit a significant reduction in BRCA2 protein. This aberrant effect is independent from de novo protein synthesis and results from COL1-beta(1) integrin signaling through phosphatidylinositol (PI) 3-kinase leading to BRCA2 ubiquitination and degradation in the proteasome. BRCA2 protein depletion after cancer cell adhesion to COL1 or in small RNA interference assays triggers new DNA synthesis, a trophic effect that is abrogated by recombinant BRCA2 expression. Blocking or inhibiting beta(1) integrin, PI 3-kinase, or proteasome activity all have a negative effect on COL1-mediated DNA synthesis in cancer cells. In normal cells, the transient increase in BRCA2 expression is independent from beta(1) integrin or PI 3-kinase and has no effect in cell proliferation. In summary, these results unravel a novel mechanism whereby prostate carcinoma cell proliferation is enhanced by the down-regulation of BRCA2 expression when interacting with COL1, a major component of the ECM at osseous metastatic sites.
Collapse
Affiliation(s)
- Loredana Moro
- Institute of Biomembranes and Bioenergetics, National Research Council (Consiglio Nazionale delle Ricerche), Via Amendola 165/A, Bari 70126, Italy.
| | | | | | | |
Collapse
|
26
|
Sheikh SSE, Domin J, Abel P, Stamp G, Lalani EN. Phosphorylation of both EGFR and ErbB2 is a reliable predictor of prostate cancer cell proliferation in response to EGF. Neoplasia 2004; 6:846-53. [PMID: 15720812 PMCID: PMC1531689 DOI: 10.1593/neo.04379] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2004] [Revised: 06/08/2004] [Accepted: 06/18/2004] [Indexed: 01/08/2023]
Abstract
Despite multiple reports of overexpression in prostate cancer (PC), the reliance of PC cells on activated epidermal growth factor receptor (EGFR) and its downstream signaling to phosphoinositide 3'-kinase/Akt (PI3K/Akt/PTEN) and/or mitogen-activated protein kinase (MAPK/ERK) pathways has not been fully elucidated. In this study, we compared the role of EGF-mediated signaling in nonmalignant (BPH-1, PNT1A, and PNT1B) and PC cell lines (DU145, PC3, LNCaP, and CWR22Rv1). EGF-induced proliferation was observed in all EGFR-expressing PC cells except PC3, indicating that EGFR expression does not unequivocally trigger proliferation following EGF stimulation. ErbB2 recruitment potentiated EGF-induced signals and was associated with the most pronounced effects of EGF despite low EGFR expression. In this way, the sum of EGFR and ErbB2 receptor phosphorylation proved to be a more sensitive indicator of EGF-induced proliferation than quantification of the expression of either receptor alone. Both Akt and ERK were rapidly phosphorylated in response to EGF, with ERK phosphorylation being the weakest in PC3 cells. Extrapolation of these findings to clinical PC suggests that assessment of phosphorylated EGFR + ErbB2 together could serve as a marker for sensitivity to anti-EGFR-targeted therapies.
Collapse
Affiliation(s)
- Soha Salama El Sheikh
- Department of Histopathology Imperial College, Hammersmith Hospital Campus, Ducane Road, London W12 0NN, UK
| | - Jan Domin
- Department of Renal Medicine Imperial College Hammersmith Hospital Campus, Ducane Road, London W12 0NN, UK
| | - Paul Abel
- Department of Surgery Imperial College, Hammersmith Hospital Campus, Ducane Road, London W12 0NN, UK
| | - Gordon Stamp
- Department of Histopathology Imperial College, Hammersmith Hospital Campus, Ducane Road, London W12 0NN, UK
| | - El-Nasir Lalani
- Department of Histopathology Imperial College, Hammersmith Hospital Campus, Ducane Road, London W12 0NN, UK
| |
Collapse
|
27
|
Li L, Ren CH, Tahir SA, Ren C, Thompson TC. Caveolin-1 maintains activated Akt in prostate cancer cells through scaffolding domain binding site interactions with and inhibition of serine/threonine protein phosphatases PP1 and PP2A. Mol Cell Biol 2003; 23:9389-404. [PMID: 14645548 PMCID: PMC309640 DOI: 10.1128/mcb.23.24.9389-9404.2003] [Citation(s) in RCA: 234] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2003] [Revised: 06/04/2003] [Accepted: 09/09/2003] [Indexed: 12/28/2022] Open
Abstract
Previously it has been reported that caveolin-1 (cav-1) has antiapoptotic activities in prostate cancer cells and functions downstream of androgenic stimulation. In this study, we demonstrate that cav-1 overexpression significantly reduced thapsigargin (Tg)-stimulated apoptosis. Examination of the phosphatidylinositol 3-kinase (PI3-K)/Akt signaling cascade revealed higher activities of PDK1 and Akt but not PI3-K in cav-1-stimulated cells compared to control cells. We subsequently found that cav-1 interacts with and inhibits serine/threonine protein phosphatases PP1 and PP2A through scaffolding domain binding site interactions. Deletion of the cav-1 scaffolding domain significantly reduces phosphorylated Akt and cell viability compared with wild-type cav-1. Analysis of potential substrates for PP1 and PP2A revealed that cav-1-mediated inhibition of PP1 and PP2A leads to increased PDK1, Akt, and ERK1/2 activities. We demonstrate that increased Akt activities are largely responsible for cav-1-mediated cell survival using dominant-negative Akt mutants and specific inhibitors to MEK1/MEK and show that cav-1 increases the half-life of phosphorylated PDK1 and Akt after inhibition of PI3-K by LY294002. We further demonstrate that cav-1-stimulated Akt activities lead to increased phosphorylation of multiple Akt substrates, including GSK3, FKHR, and MDM2. In addition, overexpression of cav-1 significantly increases translocation of phosphorylated androgen receptor to nucleus. Our studies therefore reveal a novel mechanism of Akt activation in prostate cancer and potentially other malignancies.
Collapse
Affiliation(s)
- Likun Li
- Scott Department of Urology, Baylor College of Medicine, 6560 Fannin, Suite 2100, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
28
|
Moro L, Perlino E, Marra E, Languino LR, Greco M. Regulation of beta1C and beta1A integrin expression in prostate carcinoma cells. J Biol Chem 2003; 279:1692-702. [PMID: 14585844 DOI: 10.1074/jbc.m307857200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
beta(1C) and beta(1A) integrins are two splice variants of the human beta(1) integrin subfamily that act as an inhibitor and a stimulator of cell proliferation, respectively. In neoplastic prostate epithelium, both these variants are down-regulated at the mRNA level, but only beta(1C) protein levels are reduced. We used an experimental model consisting of PNT1A, a normal immortalized prostate cell line, and LNCaP and PC-3, two prostate carcinoma cell lines, to investigate both the transcription/post-transcription and translation/post-translation processes of beta(1C) and beta(1A). Transcriptional regulation played the key role for the reduction in beta(1C) and beta(1A) mRNA expression in cancer cells, as beta(1C) and beta(1A) mRNA half-lives were comparable in normal and cancer cells. beta(1C) translation rate decreased in cancer cells in agreement with the decrease in mRNA levels, whereas beta(1A) translation rate increased more than 2-fold, despite the reduction in mRNA levels. Both beta(1C) and beta(1A) proteins were degraded more rapidly in cancer than in normal cells, and pulse-chase experiments showed that intermediates and/or rates of beta(1C) and beta(1A) protein maturation differ in cancer versus normal cells. Inhibition of either calpain- or lysosomal-mediated proteolysis increased both beta(1C) and beta(1A) protein levels, the former in normal but not in cancer cells and the latter in both cell types, albeit at a higher extent in cancer than in normal cells. Interestingly, inhibition of the ubiquitin proteolytic pathway increased expression of ubiquitinated beta(1C) protein without affecting beta(1A) protein levels in cancer cells. These results show that transcriptional, translational, and post-translational processes, the last involving the ubiquitin proteolytic pathway, contribute to the selective loss of beta(1C) integrin, a very efficient inhibitor of cell proliferation, in prostate malignant transformation.
Collapse
Affiliation(s)
- Loredana Moro
- Institute of Biomembranes and Bioenergetics, National Research Council (C.N.R.), Via Amendola 165/A, 70126 Bari, Italy.
| | | | | | | | | |
Collapse
|
29
|
Lehrer S, Diamond EJ, Stone NN, Droller MJ, Stock RG, Stone M, Bajwa A, Kornreich R. Elevated serum triiodothyronine (t3) in Ashkenazi Jewish prostate cancer patients carrying the I1307k allele of the APC (adenopolyposis coli) gene. Urol Oncol 2003; 21:101-4. [PMID: 12856637 DOI: 10.1016/s1078-1439(02)00210-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE The risk of developing any cancer in carriers of the I1307K mutation of the adenopolyposis coli (APC) gene is significantly increased (odds ratio 1.5, P = 0.01). One of the cancers associated with the I1307K mutation is prostate cancer (odds ratio 2.0, P = 0.14). Also, there is an association of APC mutations with thyroid cancer. In this study, we measured triiodothyronine (t3) levels in Ashkenazi Jewish prostate cancer patients, with and without the I1307K mutation of the APC gene. MATERIALS AND METHODS Participants in our study were found through urology and radiation oncology clinics in 1999 and 2000. All eligible patients were asked to take part. All patients had been initially diagnosed on the basis of rising PSA or abnormal physical examination. Histological confirmation of diagnosis was obtained for all subjects. Ethnic background was confirmed for all subjects by self-report or interview. The I1307K allele of the APC gene was detected by amplification of DNA isolated from peripheral blood according to standard polymerase chain reaction (PCR) and dot blot procedures. Serum t3 level was determined by fluorescent immunoassay with a standard, commercially available instrument. RESULTS We studied 77 patients. The youngest patient was 46, the oldest 88, average age 67 +/- 7.2 (mean +/- SD). Eleven males carrying the APCI 1307K allele had significantly higher serum t3 levels than 66 males carrying the wild type allele. There were no homozygotes for the I1307K allele. None of the males had a t3 level that was above the normal range for our laboratory (137 ng/dl). CONCLUSIONS Our findings of increased serum t3 level with the APC I1307K allele in prostate cancer patients is not surprising, given the mitogenic potential of t3. Further studies may clarify whether t3 elevation is the mechanism whereby APC gene mutations increase the risk of prostate cancer, or whether other pathophysiologic abnormalities are involved.
Collapse
Affiliation(s)
- Steven Lehrer
- Department of Radiation Oncology, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Jackson-Cook C, Zou Y, Turner K, Astbury C, Ware J. A novel tumorigenic human prostate epithelial cell line (M2205): molecular cytogenetic characterization demonstrates C-MYC amplification and jumping translocations. CANCER GENETICS AND CYTOGENETICS 2003; 141:56-64. [PMID: 12581899 DOI: 10.1016/s0165-4608(02)00665-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The paucity of cell lines from early-stage prostate cancer tumors has hindered the recognition of genetic and cellular changes that are associated with the acquisition of tumorigenesis. We describe the chromosomal complement of a novel tumorigenic prostate epithelial cell subline, called M2205, that acquired only three new, consistent chromosomal changes (from those present in the SV40T antigen immortalized parental cell line, P69SV40TAg) when it attained tumor-forming potential. The consistent changes, which were fully characterized using GTG-banding, CBG-banding, silver staining, fluorescence in situ hybridization (FISH), comparative genomic hybridization (CGH), and spectral karyotyping (SKY), involved segmental jumping translocations and resulted in gains in the copy number of genes located on the distal long arm of chromosome 8 (8q22 to 8q24.3), including c-myc. Furthermore, the jumping translocations also resulted in ribosomal genes being present in multiple, tandem copies next to the chromatin from 8q. Given the relatively small number of cytogenetic changes present, this subline provides a means for better understanding the cellular changes associated with the acquired chromosomal imbalances. Further studies of this subline could also provide insight as to the mechanism or mechanisms leading to the formation of jumping translocations, as well as potential position effects resulting from the relocation of ribosomal genes next to other cellular genes or oncogenes.
Collapse
Affiliation(s)
- Colleen Jackson-Cook
- Department of Human Genetics, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond, VA 23298-0033, USA.
| | | | | | | | | |
Collapse
|
31
|
Maillard M, Cadot B, Ball RY, Sethia K, Edwards DR, Perbal B, Tatoud R. Differential expression of the ccn3 (nov) proto-oncogene in human prostate cell lines and tissues. Mol Pathol 2001; 54:275-80. [PMID: 11477145 PMCID: PMC1187081 DOI: 10.1136/mp.54.4.275] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIMS To investigate the expression of the human ccn3 (hccn3; nov) proto-oncogene, a member of the CCN family of proteins, in prostate epithelial cells and prostate tissue samples. METHODS Normal adult prostate luminal epithelial cells immortalised by SV40 large T (PNT1A and PNT1B), metastatic tumours (LNCaP, DU-145, and PC-3), and prostate tissue samples from patients with benign prostatic hyperplasia (BPH) and prostatic adenocarcinoma were used. hccn3 (nov) mRNA was measured by the reverse transcription polymerase chain reaction (RT-PCR) and hCCN3 (NOV) protein expression was determined by immunochemistry. RESULTS hccn3 (nov) RNA values were higher in PC-3 cells than in the other prostate cell lines. The immortalised normal cell lines either did not express hccn3 (nov) RNA (PNT1B) or expressed very low amounts (PNT1A). BPH samples expressed variable amounts of hccn3 (nov) RNA. With the use of immunocytochemistry, all cell lines except PNT1A and PNT1B were shown to contain hCCN3 (NOV) protein. hCCN3 (NOV) was localised mainly in the epithelial compartment of BPH and adenocarcinoma samples, and there was evidence of luminal secretion. CONCLUSION The overexpression of hccn3 (nov) RNA in cancer cell lines compared with other cell lines and its epithelial localisation in human prostate samples are consistent with a role for hCCN3 (NOV) in prostatic tumorigenesis.
Collapse
Affiliation(s)
- M Maillard
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | | | | | | | | | | | | |
Collapse
|
32
|
Yeh CC, Lee C, Huang MC, Dahiya R. Loss of mismatch repair activity in simian virus 40 large T antigen-immortalized BPH-1 human prostatic epithelial cell line. Mol Carcinog 2001; 31:145-51. [PMID: 11479923 DOI: 10.1002/mc.1049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Simian virus 40 large T antigen (SVLTAg) has been used to immortalize cells; however, the mechanism leading to immortalization is still unclear. We hypothesize that DNA mismatch repair (MMR) activity is important during SVLTAg-induced immortalization. To test this hypothesis, we used the SVLTAg-immortalized cell line BPH-1 derived from human benign prostate epithelial cells to analyze MMR activity and the expression of MMR genes (hMLH1, hPMS1, hPMS2, hMSH2, hMSH3, and hMSH6). The results demonstrated that BPH-1 cells were deficient in repairing G:T, A:C, and G:G mispairs in bacteriophage M13mp2. Reverse-transcription polymerase chain reaction experiments indicated MMR genes (hMSH3, hMSH6, and hPMS1) were expressed at a low level in BPH-1 cells. In contrast, all six MMR genes were expressed in human benign prostate hyperplasia tissues. Downregulation of hMSH3, hMSH6, and hPMS1 genes is not a result of the hypermethylation mechanism because demethylation with 5-aza-2'-deoxycytidine did not restore expression of these genes. Although the hMLH1 gene is expressed in BPH-1 cells, western blotting and exon analyses demonstrated that hMLH1 was mutated and/or deleted in BPH-1 cells.
Collapse
Affiliation(s)
- C C Yeh
- Department of Urology, Veterans Affairs Medical Center and University of California, San Francisco, California, USA
| | | | | | | |
Collapse
|
33
|
Downing SR, Jackson P, Russell PJ. Mutations within the tumour suppressor gene p53 are not confined to a late event in prostate cancer progression. a review of the evidence. Urol Oncol 2001; 6:103-110. [PMID: 11344000 DOI: 10.1016/s1078-1439(00)00119-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mutations in the p53 tumour suppressor gene are generally believed to be a late event in the progression of prostate cancer, and are associated with androgen independence, metastasis, and a worse prognosis. In this review, we examine the current literature available on p53 mutations and focus on stages A (T1) and B (T2) of prostate cancer. We report here that p53 mutations can be found in approximately one third of prostate cancers that are clinically localized to the prostate. In addition, high levels of p53 mutation are found in normal prostate tissue of prostate cancer patients, prostatic intraepithelial neoplasia, and benign prostatic hyperplasia. The limitations of techniques used to determine p53 mutations are discussed, as well as other modes of p53 loss in early stage prostate cancer.
Collapse
Affiliation(s)
- S R. Downing
- Oncology Research Centre, Level 2 Clinical Sciences Building, Prince of Wales Hospital, Barker Street, Randwick, NSW 2031, Australia, and Faculty of Medicine, University of New South Wales, NSW 2033, Kensington, Australia
| | | | | |
Collapse
|
34
|
Yin Z, Babaian RJ, Troncoso P, Strom SS, Spitz MR, Caudell JJ, Stein JD, Kagan J. Limiting the location of putative human prostate cancer tumor suppressor genes on chromosome 18q. Oncogene 2001; 20:2273-80. [PMID: 11402322 DOI: 10.1038/sj.onc.1204310] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2000] [Revised: 09/19/2000] [Accepted: 01/29/2001] [Indexed: 11/09/2022]
Abstract
We studied loss of heterozygosity (LOH) on the long arm of human chromosome 18 in prostate cancer to determine the location of a putative tumor suppressor gene (TSG) and to correlate these losses with the pathological grade and stage of the cancer. Of 48 specimens analysed 17 (35.4%) lost at least one allele on chromosome 18q. All the specimens with allelic losses lost at least one allele within chromosomal region 18q21. Allelic losses picked at D18S51 (19%) and D18S858 (17%). A 0.58 cM DNA segment that includes the D18S858 locus and is flanked by the microsatellite loci D18S41 and D18S381, was lost in eight (47%) of 17 specimens with allelic losses. This segment was designated as a LOH cluster region 1 (LCR 1). Although Smad2 resides within LCR 1, it was not mutated in any of the six prostate cell lines (five prostate cancer cell lines and one immortalized prostate epithelial cell line) analysed, suggesting that it is not a candidate TSG in prostate cancer. A second LCR at 18q21, LCR 2, includes the D18S51 locus and is flanked by the D18S1109 and D18S68 loci, which are separated by 7.64 cM. LCR 2 was lost in six (35%) of the 17 specimens with chromosome 18q losses. These results suggest that chromosome 18q21 may harbor two candidate prostate cancer TSGs. The candidate TSGs DCC and Smad4 are located centromeric to the LCRs. No alleles were lost within or in close proximity to these genes, suggesting that they are not targets for inactivation by allelic losses in prostate cancer. Although there was no obvious correlation between chromosome 18q LOH and the pathological grade or stage, three (37.5%) of eight low-grade cancers and nine (32.1%) of 28 organ-confined cancers lost alleles at 18q21, suggesting that allelic losses are relatively early events in the development of invasive prostate cancer.
Collapse
Affiliation(s)
- Z Yin
- Division of Pathology and Laboratory Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, TX 77030-4095, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Aurich-Costa J, Vannier A, Grégoire E, Nowak F, Cherif D. IPM-FISH, a new M-FISH approach using IRS-PCR painting probes: Application to the analysis of seven human prostate cell lines. Genes Chromosomes Cancer 2001. [DOI: 10.1002/1098-2264(2000)9999:9999<::aid-gcc1076>3.0.co;2-e] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
36
|
Lehrer S, McCurdy LD, Stock RG, Kornreich R, Stone NN, Eng C. Body mass, age, and the APC I1307K allele in Ashkenazi Jewish prostate cancer patients. CANCER GENETICS AND CYTOGENETICS 2000; 122:131-3. [PMID: 11106824 DOI: 10.1016/s0165-4608(00)00291-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The I1307K mutation of the adenopolyposis coli gene (APC), located on chromosome 5q21-q22, is associated with an increased risk of cancer in Ashkenazi Jews. In the present study, we analyzed age and body mass of Ashkenazi Jewish prostate cancer patients, with and without the APC I1307K mutation. Participants in our study were found through urology and radiation oncology clinics, and all eligible patients were asked to take part. A familial history was obtained by interview or self-report questionnaire. Histological confirmation of diagnosis was obtained for all subjects. The I1307K allele of the APC gene was detected by amplification of lymphocyte DNA from peripheral blood according to standard polymerase chain reaction (PCR) and dot blot procedures. We studied 135 Ashkenazi Jewish men with prostate cancer. The youngest was 49, the oldest 80, average age 68 +/- 6.88 (mean +/- SD). The older patients carrying the wild type APC allele tended to have a lower body mass than the younger ones (r = -.27, P =.002). Of 71 patients under 70 years old, 65 carried the wild type APC allele, and had a body mass index of 28. 7 +/- 4.23 kg/m(2). The six men under age 70 carrying the I1307K APC allele had a body mass index of 26.87 +/- 1.44 kg/m(2). The difference in body mass index of the two groups is significant (P =. 032, t test for unequal variance). Increased body mass is a prostate cancer risk factor, and hereditary prostate cancer is associated with younger patients. Therefore, our finding, that patients under age 70 carrying the I1307K allele are significantly thinner than those carrying the wild type allele, suggests that the APC I1307K allele is also a prostate cancer risk factor. Our results are in accord with other studies indicating that APC mutations increase the risk of prostate cancer.
Collapse
Affiliation(s)
- S Lehrer
- Department of Radiation Oncology, Mount Sinai School of Medicine, New York and Veterans Affairs Medical Center, Bronx, NY, USA
| | | | | | | | | | | |
Collapse
|
37
|
Mitchell S, Abel P, Ware M, Stamp G, Lalani E. Phenotypic and genotypic characterization of commonly used human prostatic cell lines. BJU Int 2000; 85:932-44. [PMID: 10792179 DOI: 10.1046/j.1464-410x.2000.00606.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To investigate and catalogue systematically the phenotypic and genotypic characteristics of the commonly used prostatic cell lines using immunocytochemistry and polymerase chain reaction (PCR) of hypervariable sequences within the genome to provide a 'fingerprint' characteristic of each cell line. Materials and methods Malignant (LNCaP, LNCaP-r, PC-3, DU-145) and benign immortalized prostatic cell lines (PNT-1A, PNT-1B, BPH-1) were grown on four-well slides, fixed and subjected to indirect streptavidin-biotin immunocytochemistry. Twenty-three antibodies were used in the following groups: cytoskeletal elements: cytokeratins (CK)-5, -7, -8, -14 (two), -16, -18, -19 (three), -20, vimentin and desmin; MUC1 (three); cell adhesion molecules (E-cadherin, alpha-beta-and gamma-catenin); and prostatic associated proteins: prostate specific antigen (PSA), prostatic acid phosphatase (PAP) and androgen receptor (AR). For the PCR, genomic DNA was extracted from the cell lines and from SKOV3 and MCF7 (positive controls). PCR was performed on three variable regions which were then sequenced: AR exon 1 (CAG repeat polymorphism), and two areas of microsatellite instability (MSI): AR exon 8 and hypoxanthine-guanine phosphoribosyl transferase (HPRT) exon 3. RESULTS All cell lines were CK-8/18 positive and most also expressed CK-7 and -19. Heterogeneous CK-20 expression was detected for the first time in prostatic cell lines. All lines were positive for vimentin and negative for desmin. MUC1 was expressed in one malignant (DU-145) and all immortalized cell lines. E-cadherin expression was low or absent in three lines: PNT1A, 1B and PC-3. Only PC-3 failed to express alpha-catenin; beta- and gamma-catenin were expressed by all lines. PSA, PAP and AR were only expressed by LNCaP and LNCaP-r. On PCR, the CAG repeat lengths in exon 1 of the AR ranged from 19 to 27. Three pairs of cell lines had the same exon 1 CAG repeat length: LNCaP/PC-3 (26 repeats), BPH-1/DU-145 (19 repeats) and PNT1 A/1B (20 repeats). Exon 8 sequences were identical except for LNCaP, which showed a single base mutation, and HPRT exon 3 sequences were all identical. There was no evidence of generalized MSI in any of the cell lines examined. CONCLUSIONS The cell lines studied fell into three broad groups according to their phenotypic characteristics: (i) prostatic marker positive (LNCaP and LNCaP-r); (ii) high expression of most antigens (DU-145, PC-3 and BPH-1); and (iii) low or absent expression of most antigens (PNT1 A and 1B). Each of the cell lines derived from PC could be identified on the basis of exon 1 and 8 AR sequence variability. DU145 and BPH-1 had identical profiles of the three areas studied, but these cell lines are easily distinguished by their different phenotypic characteristics. PNT1A and 1B had identical genetic and similar phenotypic profiles, which is unsurprising given that they are subclones derived from the same parental line. Even so, these were separable on the basis of CK-19 immunostaining. Using a combination of geno- and phenotypic markers it was possible to derive a 'fingerprint' for each of the cell lines assessed, which will allow meaningful comparison between similar cell lines held in other laboratories.
Collapse
Affiliation(s)
- S Mitchell
- Departments of Pathology and Surgery, Imperial College of Science, Technology and Medicine, Hammersmith Campus, London, UK
| | | | | | | | | |
Collapse
|
38
|
Ozen M, Navone NM, Multani AS, Troncoso P, Logothetis CJ, Chung LW, von Eschenbach AC, Pathak S. Structural alterations of chromosome 5 in twelve human prostate cancer cell lines. CANCER GENETICS AND CYTOGENETICS 1998; 106:105-9. [PMID: 9797773 DOI: 10.1016/s0165-4608(98)00051-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Neoplastic transformation, cancer progression, and metastasis are determined by a series of well-defined changes that take place in target tissue cells. Genetic alterations associated with human prostate carcinogenesis are not well defined. Some chromosomal changes, including gain of chromosomes 7, 12, 17, and X and loss of heterozygosity in chromosomes 8p, 10q, 16q, 17p, and 18q, have been reported. We examined five newly established and eight previously established prostate cancer cell lines before and after subcutis and orthotopic injection into nude mice and observed that structural alterations of chromosome 5 were present in all of the cell lines except the parental LNCaP. The fluorescence in situ hybridization preparations with the use of whole chromosome 5 DNA painting probe confirmed our Giemsa-banding data. Alterations of chromosome 5 consisted of t(1;5)(p36;q15), t(5;?)(p11;?), del(5)(q23q35) in the SP2964(= ARCaP) cell line; t(5;8)(p15;q12), i(5)(p10), t(5;15)(q11;p11) in the SP3031 cell line; t(5;?;15) (q15;?;p11), t(5;7;14)(q31;p11-q32;q11), in the SP3173 and SP3241 cell lines (derived from the same patient); del(5)(q23-33) and t(5;7;14)(q31;p11-q32;q11) in the SP3316 cell line; t(3;5)(q21;q35) in the SP2884 cell line; t(5;5)(p15;q11) in the SP2356 cell line; i(5)(p10),t(5;?)(q23;?) in the DU-145 cell line; and i(5)(p10), t(5;?)(q11;?) and t(2;5)(q15;q15) in the PC-3 cell line. Because, in most cases, alteration of chromosome 5 resulted in the partial or complete loss of 5q, we conjectured that 5q might contain one or more tumor-suppressor genes for human prostate cancer development.
Collapse
Affiliation(s)
- M Ozen
- Department of Cell Biology, University of Texas M. D. Anderson Cancer Center 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Chin RK, Hawkins AL, Isaacs WB, Griffin CA. E1A transformed normal human prostate epithelial cells contain a 16q deletion. CANCER GENETICS AND CYTOGENETICS 1998; 103:155-63. [PMID: 9614916 DOI: 10.1016/s0165-4608(97)00407-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The difficulty of maintaining long-term prostate cell cultures has hindered the development of essential models for understanding prostate cancer. We report here the establishment of two 12S E1A transformed non-tumorigenic prostate epithelial cell strains, and their characterization. The two clonal cell strains, TP2 and TP4, proliferated for approximately 40 passages before senescence. Both exhibited a strong dependence on exogenous peptide growth factors and an immunophenotype characteristic of their prostate epithelial origin. Cytogenetic analysis revealed a consistent deletion on the q arm of chromosome 16 in TP2 with an otherwise normal karyotype. Band-specific microdissection generated region-specific probes from 16q23, which when used in fluorescence in situ hybridization (FISH) revealed that the region was deleted in 83% of metaphases analyzed. By cytogenetic analysis and FISH, the q arm of 16 was found deleted from the genome of TP4 in 60% of cells analyzed. Lost sequences on 16q-16q23 in particular--in prostate cancer have been observed by a variety of methods. Localization of common region of deletion has been determined from these studies to be distal to 16q23. Our findings suggest that 16q23 may be of major importance in the development of prostate cancer, and may harbor tumor suppressor elements.
Collapse
Affiliation(s)
- R K Chin
- Johns Hopkins School of Medicine, Department of Pathology, Baltimore, MD, USA
| | | | | | | |
Collapse
|
40
|
Khaira P, James CD, Leffak M. Amplification of the translocated c-myc genes in three Burkitt lymphoma cell lines. Gene 1998; 211:101-8. [PMID: 9643977 DOI: 10.1016/s0378-1119(98)00104-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Translocations of the coding exons of the human c-myc gene are consistent features of human Burkitt lymphomas (BL). In the BL cell lines CA46, JD40, and ST486, the second and third c-myc exons have been translocated into the immunoglobulin heavy chain locus. In addition to this rearrangement, in all three cell lines, we have found that the translocated c-myc exons show low-level amplification relative to restriction fragments from the germ-line c-myc gene. The patterns of hybridization of an IgM switch region probe suggest that immunoglobulin heavy chain sequences have been co-amplified with the translocated c-myc sequences. Differential sedimentation was used to determine whether the amplified sequences reside in high-molecular-weight chromosomes or low-molecular-weight extrachromosomal DNA. In JD40 and ST486 cells, the amplified c-myc sequences were found on high-molecular-weight chromosomes ST486 cells also contained translocated C-myc sequences in low-molecular-weight, extrachromosomal DNA, as did CA46 cells. These conclusions were corroborated by fluorescence in-situ hybridization (FISH) of HeLa, CA46, ST486 and JD40 metaphase chromosomes. These results suggest that there is ongoing selection for cells containing amplified copies of the expressed c-myc sequences. and that there is continuous generation of extrachromosomal copies of the translocated c-myc sequences in ST486 and CA46 cells.
Collapse
Affiliation(s)
- P Khaira
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH 45435, USA
| | | | | |
Collapse
|
41
|
|
42
|
Degeorges A, Tatoud R, Fauvel-Lafeve F, Podgorniak MP, Millot G, de Cremoux P, Calvo F. Stromal cells from human benign prostate hyperplasia produce a growth-inhibitory factor for LNCaP prostate cancer cells, identified as interleukin-6. Int J Cancer 1996; 68:207-14. [PMID: 8900430 DOI: 10.1002/(sici)1097-0215(19961009)68:2<207::aid-ijc12>3.0.co;2-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
To understand specific interactions between stromal cells and epithelial cells in benign prostatic hyperplasia (BPH) and prostatic adenocarcinoma, we developed stromal-cell cultures from normal human prostate (PNX) and BPH (BH101), composed of fibroblasts and myofibroblasts. Their role in epithelial-cell growth was studied using the established cancer cell lines LNCaP, PC3 and DU145 and an SV40 large T-immortalized normal epithelial-cell line, PNT1A, in double-diffusion co-culture chambers. PNT1A was stimulated by PNX (x1.6) and more strongly by BH101 stromal cells (x2.7). Conversely, LNCaP growth decreased by 50% in the presence of BH101 stromal cells (stromal/epithelial ratio: 10). A BH101-conditioned medium (CM), obtained in serum-free conditions, induced 90% inhibition of [3H]thymidine incorporation of the LNCaP androgen-sensitive cell line. Two other androgen-independent prostate cancer cell lines were either insensitive to BH101 CM (PC3) or slightly inhibited (40% for DU145). BH101 produced large amounts of IL-1beta, IL-6 and IL-8. HPLC gel filtration enabled separation of an inhibitory fraction which contained IL-6. IL-6 was demonstrated to be responsible for the strong inhibitory effect since an IL-6-neutralizing antibody abolished this inhibition, which was reproduced by human recombinant IL-6. Recombinant IL-6 growth inhibition was observed only on LNCaP prostate cancer androgen-sensitive cells.
Collapse
Affiliation(s)
- A Degeorges
- Laboratoire de Pharmacologie, Institut de Génétique Moléculaire, Hôpital Saint-Louis, Paris, France
| | | | | | | | | | | | | |
Collapse
|