1
|
Siegelmann-Danieli N, Neiman V, Bareket-Samish A, Berger R, Peretz A, Alapi H, Tsur E, Patalon T, Beller D, Rimler G, Chodick G, Shohat M. Whole exome germline sequencing in early-onset prostate cancer patients: Genomic findings and clinical outcomes. Prostate 2024; 84:39-46. [PMID: 37842866 DOI: 10.1002/pros.24622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/13/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Whole exome sequencing (WES) furthered our understanding of various tumors. We assessed the occurrence of germline likely pathogenic/pathogenic (LP/P) variants, disease features, and clinical outcomes in early-onset prostate cancer. METHODS This retrospective study (N = 134) included consecutive prostate cancer patients who donated blood samples for research purposes to the Kahn-Sagol-Maccabi biobank. Patients diagnosed at >65 years were excluded. Clinical characteristics were extracted from the medical records. Germline WES was performed with analysis reporting on oncogenetic, two immunogenic, and a secondary minimum list panels (121, 468, 76, and 59 genes, respectively). RESULTS Median age at diagnosis was 61 (range 46-65) years; 131 (98%) were diagnosed with local disease. The median follow-up time from diagnosis was 14 (range <1-25) years. Of the patients with local disease, 32 (24%) and 10 (8%) had biochemical and distant recurrences, respectively. Twenty-five patients (19%) had ≥1 additional cancer (excluding non-melanoma skin cancer), most frequently bladder (6), colorectal (5), and lymphoma (5). Seven (5%) deaths were reported, with only one related to prostate cancer. LP/P variants were identified in 8 patients (6%), all in genes from the oncogenetic panel: ATM, BRCA1 (in two patients), BRCA2 (in two patients), HOXB13, MUTYH, and MYH7. Of these eight patients, with a median follow-up of 7 years (range <1-15), two (25%) had biochemical recurrences, one had (12.5%) distant recurrence, and no deaths were reported. CONCLUSIONS In this cohort of 134 early-onset prostate cancer patients, we identified germline LP/P variants in an oncogenetic panel in 6% of participants, with no unique clinical outcome.
Collapse
Affiliation(s)
- Nava Siegelmann-Danieli
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Professional Medicine, Maccabi Healthcare Services, Tel Aviv, Israel
- KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Victoria Neiman
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | | | - Racheli Berger
- Genetic Institute, Maccabi Healthcare Services, Rehovot, Israel
| | - Asaf Peretz
- KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
- Tipa Biobank, KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Hillel Alapi
- KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Erez Tsur
- Genetic Institute, Maccabi Healthcare Services, Rehovot, Israel
| | - Tal Patalon
- KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
- Tipa Biobank, KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Daniella Beller
- Tipa Biobank, KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Galit Rimler
- Tipa Biobank, KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Gabriel Chodick
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- KSM Research and Innovation Center, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Mordechai Shohat
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Genetic Institute, Maccabi Healthcare Services, Rehovot, Israel
- The Genomics Unit, Sheba Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| |
Collapse
|
2
|
Seo E, Kang M. Current status and clinical application of patient-derived tumor organoid model in kidney and prostate cancers. BMB Rep 2023; 56:24-31. [PMID: 36476272 PMCID: PMC9887101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Indexed: 01/28/2023] Open
Abstract
Urological cancers such as kidney, bladder, prostate, and testicular cancers are the most common types of cancers worldwide with high mortality and morbidity. To date, traditional cell lines and animal models have been broadly used to study pre-clinical applications and underlying molecular mechanisms of urological cancers. However, they cannot reflect biological phenotypes of real tissues and clinical diversities of urological cancers in vitro system. In vitro models cannot be utilized to reflect the tumor microenvironment or heterogeneity. Cancer organoids in three-dimensional culture have emerged as a promising platform for simulating tumor microenvironment and revealing heterogeneity. In this review, we summarize recent advances in prostate and kidney cancer organoids regarding culture conditions, advantages, and applications of these cancer organoids. [BMB Reports 2023; 56(1): 24-31].
Collapse
Affiliation(s)
- Eunjeong Seo
- Molecular Pharmacology, OliPass Corporation, Yongin 17015, Korea
| | - Minyong Kang
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea,Department of Health Sciences and Technology, SAIHST, Seoul 06351, Korea,Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea,Corresponding author. Tel: +82-2-3410-1138; Fax: +82-2-3410-6992; E-mail:
| |
Collapse
|
3
|
Seo E, Kang M. Current status and clinical application of patient-derived tumor organoid model in kidney and prostate cancers. BMB Rep 2023; 56:24-31. [PMID: 36476272 PMCID: PMC9887101 DOI: 10.5483/bmbrep.2022-0200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 11/30/2023] Open
Abstract
Urological cancers such as kidney, bladder, prostate, and testicular cancers are the most common types of cancers worldwide with high mortality and morbidity. To date, traditional cell lines and animal models have been broadly used to study pre-clinical applications and underlying molecular mechanisms of urological cancers. However, they cannot reflect biological phenotypes of real tissues and clinical diversities of urological cancers in vitro system. In vitro models cannot be utilized to reflect the tumor microenvironment or heterogeneity. Cancer organoids in three-dimensional culture have emerged as a promising platform for simulating tumor microenvironment and revealing heterogeneity. In this review, we summarize recent advances in prostate and kidney cancer organoids regarding culture conditions, advantages, and applications of these cancer organoids. [BMB Reports 2023; 56(1): 24-31].
Collapse
Affiliation(s)
- Eunjeong Seo
- Molecular Pharmacology, OliPass Corporation, Yongin 17015, Korea
| | - Minyong Kang
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Department of Health Sciences and Technology, SAIHST, Seoul 06351, Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea
| |
Collapse
|
4
|
Discovery of a Novel Bloom's Syndrome Protein (BLM) Inhibitor Suppressing Growth and Metastasis of Prostate Cancer. Int J Mol Sci 2022; 23:ijms232314798. [PMID: 36499126 PMCID: PMC9736344 DOI: 10.3390/ijms232314798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Prostate cancer (PCa) is a common cancer and a major cause of cancer-related death worldwide in men, necessitating novel targets for cancer therapy. High expression of Bloom's syndrome protein (BLM) helicase is associated with the occurrence and development of PCa. Therefore, the identification and development of new BLM inhibitors may be a new direction for the treatment of PCa. Here, we identified a novel inhibitor by molecular docking and put it to systematic evaluation via various experiments, AO/854, which acted as a competitive inhibitor that blocked the BLM-DNA interaction. Cellular evaluation indicated that AO/854-suppressed tumor growth and metastasis in PC3 cells by enhancing DNA damage, phosphorylating Chk1/Chk2, and altering the p53 signaling pathway. Collectively, the study highlights the potential of BLM as a therapeutic target in PCa and reveals a distinct mechanism by which AO/854 competitively inhibits the function of BLM.
Collapse
|
5
|
Goetze S, Schüffler P, Athanasiou A, Koetemann A, Poyet C, Fankhauser CD, Wild PJ, Schiess R, Wollscheid B. Use of MS-GUIDE for identification of protein biomarkers for risk stratification of patients with prostate cancer. Clin Proteomics 2022; 19:9. [PMID: 35477343 PMCID: PMC9044739 DOI: 10.1186/s12014-022-09349-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 04/05/2022] [Indexed: 11/25/2022] Open
Abstract
Background Non-invasive liquid biopsies could complement current pathological nomograms for risk stratification of prostate cancer patients. Development and testing of potential liquid biopsy markers is time, resource, and cost-intensive. For most protein targets, no antibodies or ELISAs for efficient clinical cohort pre-evaluation are currently available. We reasoned that mass spectrometry-based prescreening would enable the cost-effective and rational preselection of candidates for subsequent clinical-grade ELISA development. Methods Using Mass Spectrometry-GUided Immunoassay DEvelopment (MS-GUIDE), we screened 48 literature-derived biomarker candidates for their potential utility in risk stratification scoring of prostate cancer patients. Parallel reaction monitoring was used to evaluate these 48 potential protein markers in a highly multiplexed fashion in a medium-sized patient cohort of 78 patients with ground-truth prostatectomy and clinical follow-up information. Clinical-grade ELISAs were then developed for two of these candidate proteins and used for significance testing in a larger, independent patient cohort of 263 patients. Results Machine learning-based analysis of the parallel reaction monitoring data of the liquid biopsies prequalified fibronectin and vitronectin as candidate biomarkers. We evaluated their predictive value for prostate cancer biochemical recurrence scoring in an independent validation cohort of 263 prostate cancer patients using clinical-grade ELISAs. The results of our prostate cancer risk stratification test were statistically significantly 10% better than results of the current gold standards PSA alone, PSA plus prostatectomy biopsy Gleason score, or the National Comprehensive Cancer Network score in prediction of recurrence. Conclusion Using MS-GUIDE we identified fibronectin and vitronectin as candidate biomarkers for prostate cancer risk stratification. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-022-09349-x.
Collapse
Affiliation(s)
- Sandra Goetze
- Department of Health Sciences and Technology, Institute of Translational Medicine, Swiss Federal Institute of Technology, ETH Zurich, 8093, Zurich, Switzerland.,Swiss Institute of Bioinformatics (SIB), 1015, Lausanne, Switzerland.,ETH PHRT Swiss Multi-Omics Center (SMOC), 8093, Zurich, Switzerland
| | - Peter Schüffler
- Institute of General and Surgical Pathology, Technical University of Munich, 81675, Munich, Germany
| | | | - Anika Koetemann
- Department of Health Sciences and Technology, Institute of Translational Medicine, Swiss Federal Institute of Technology, ETH Zurich, 8093, Zurich, Switzerland
| | - Cedric Poyet
- Clinic of Urology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
| | | | - Peter J Wild
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland. .,Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, 60590, Frankfurt, Germany. .,Frankfurt Institute for Advanced Studies (FIAS), 60438, Frankfurt, Germany. .,WILDLAB, University Hospital Frankfurt MVZ GmbH, 60590, Frankfurt, Germany.
| | | | - Bernd Wollscheid
- Department of Health Sciences and Technology, Institute of Translational Medicine, Swiss Federal Institute of Technology, ETH Zurich, 8093, Zurich, Switzerland. .,Swiss Institute of Bioinformatics (SIB), 1015, Lausanne, Switzerland. .,ETH PHRT Swiss Multi-Omics Center (SMOC), 8093, Zurich, Switzerland.
| |
Collapse
|
6
|
Mancini M, Grasso M, Muccillo L, Babbio F, Precazzini F, Castiglioni I, Zanetti V, Rizzo F, Pistore C, De Marino MG, Zocchi M, Del Vescovo V, Licursi V, Giurato G, Weisz A, Chiarugi P, Sabatino L, Denti MA, Bonapace IM. DNMT3A epigenetically regulates key microRNAs involved in epithelial-to-mesenchymal transition in prostate cancer. Carcinogenesis 2021; 42:1449-1460. [PMID: 34687205 DOI: 10.1093/carcin/bgab101] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 08/17/2021] [Accepted: 10/21/2021] [Indexed: 11/14/2022] Open
Abstract
Epithelial-to-Mesenchymal Transition (EMT) is involved in prostate cancer metastatic progression, and its plasticity suggests epigenetic implications. Deregulation of DNMTs and several miRNAs plays a relevant role in EMT, but their interplay has not been clarified yet. In this study we provide evidence that DNMT3A interaction with several miRNAs has a central role in an ex-vivo EMT prostate cancer model obtained via exposure of PC3 cells to conditioned media from cancer-associated fibroblasts (CM-CAFs). The analysis of the alterations of the miRNA profile shows that miR-200 family (miR-200a/200b/429, miR-200c/141), miR-205, and miR-203, known to modulate key EMT factors, are downregulated and hyper-methylated at their promoters. DNMT3A (mainly isoform a) is recruited onto these miRNA promoters, coupled with the increase of H3K27me3/H3K9me3 and/or the decrease of H3K4me3/H3K36me3. Most interestingly, our results reveal the differential expression of two DNMT3A isoforms (a and b) during ex-vivo EMT and a regulatory feedback loop between miR-429 and DNMT3A that can promote and sustain the transition toward a more mesenchymal phenotype. We demonstrate the ability of miR-429 to target DNMT3A 3'UTR and modulate the expression of EMT factors, in particular ZEB1. Survey of the PRAD-TCGA data set shows that patients expressing an EMT-like signature are indeed characterized by down-regulation of the same miRNAs with a diffused hyper-methylation at miR-200c/141 and miR-200a/200b/429 promoters. Finally, we show that miR-1260a also targets DNMT3A, although it does not seem involved in EMT in prostate cancer.
Collapse
Affiliation(s)
- Monica Mancini
- Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (VA), Italy
| | - Margherita Grasso
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Povo (TN), Italy
| | - Livio Muccillo
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Federica Babbio
- Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (VA), Italy
| | - Francesca Precazzini
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Povo (TN), Italy
| | - Ilaria Castiglioni
- Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (VA), Italy
| | - Valentina Zanetti
- Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (VA), Italy
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081 Baronissi, Italy.,Genome Research Center for Health, c/o University of Salerno Campus of Medicine, 84081 Baronissi (SA), Italy
| | - Christian Pistore
- Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (VA), Italy
| | - Maria Giovanna De Marino
- Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (VA), Italy
| | - Michele Zocchi
- Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (VA), Italy
| | - Valerio Del Vescovo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Povo (TN), Italy
| | - Valerio Licursi
- Department of Biology and Biotechnology "Charles Darwin", "Sapienza" University of Rome, Rome, Italy
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081 Baronissi, Italy.,Genome Research Center for Health, c/o University of Salerno Campus of Medicine, 84081 Baronissi (SA), Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081 Baronissi, Italy.,Genome Research Center for Health, c/o University of Salerno Campus of Medicine, 84081 Baronissi (SA), Italy
| | - Paola Chiarugi
- Department of Biomedical, Experimental and Clinical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Lina Sabatino
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Michela Alessandra Denti
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Povo (TN), Italy
| | - Ian Marc Bonapace
- Department of Biotechnology and Life Sciences, University of Insubria, 21052 Busto Arsizio (VA), Italy
| |
Collapse
|
7
|
BRCA Germline Mutations in Prostate Cancer: The Future Is Tailored. Diagnostics (Basel) 2021; 11:diagnostics11050908. [PMID: 34069669 PMCID: PMC8161324 DOI: 10.3390/diagnostics11050908] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PCa) is the second most common neoplasm in men and the fifth leading cause of death worldwide [...].
Collapse
|
8
|
Ruan Y, Xu H, Ji X, Zhao J. BLM interaction with EZH2 regulates MDM2 expression and is a poor prognostic biomarker for prostate cancer. Am J Cancer Res 2021; 11:1347-1368. [PMID: 33948362 PMCID: PMC8085859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/20/2021] [Indexed: 06/12/2023] Open
Abstract
Prostate cancer (PCa) is one of the major causes of cancer death among males worldwide. Our previous studies indicated that the proliferation of prostate cancer cells was reduced after BLM knockdown, however, the mechanism is still not clear. In this study, we identified a direct interaction between BLM and EZH2, which had extremely significantly positive correlations (P<0.001). In vitro, our research revealed that tumor growth was inhibited after EZH2 knockdown and that inhibition could be reversed by BLM overexpression; conversely, tumor growth was promoted after EZH2 overexpression, and promotion could be reversed by BLM knockdown. This suggests that BLM and EZH2 play important roles in the progression of prostate cancer cells. In vivo, the impact of BLM and EZH2 was investigated in mouse xenograft models, and the results showed that EZH2 could be regulated by BLM, which was consistent with our in vitro observations. Our results demonstrated that the expression of P53 is affected by the binding of BLM and EZH2 to the MDM2 promoter region. This finding indicated that EZH2 regulates the expression of MDM2 at the transcriptional level by interacting with BLM.
Collapse
Affiliation(s)
- Yong Ruan
- Guizhou University School of MedicineGuiyang, Guizhou, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of EducationGuiyang, Guizhou, China
- Guizhou Key Laboratory of Animal Heredity, Breeding and ReproductionGuizhou, China
- College of Animal Science, Guizhou UniversityGuiyang, Guizhou, China
| | - Houqiang Xu
- Guizhou University School of MedicineGuiyang, Guizhou, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of EducationGuiyang, Guizhou, China
- Guizhou Key Laboratory of Animal Heredity, Breeding and ReproductionGuizhou, China
- College of Animal Science, Guizhou UniversityGuiyang, Guizhou, China
| | - Xinqin Ji
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of EducationGuiyang, Guizhou, China
- Guizhou Key Laboratory of Animal Heredity, Breeding and ReproductionGuizhou, China
- College of Animal Science, Guizhou UniversityGuiyang, Guizhou, China
| | - Jiafu Zhao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of EducationGuiyang, Guizhou, China
- Guizhou Key Laboratory of Animal Heredity, Breeding and ReproductionGuizhou, China
- College of Animal Science, Guizhou UniversityGuiyang, Guizhou, China
| |
Collapse
|
9
|
Abstract
High-risk prostate cancer is a heterogeneous disease that lacks clear consensus on its ideal management. Historically, non-surgical treatment was the preferred strategy, and several studies demonstrated improved survival among men with high-risk disease managed with the combination of radiotherapy and androgen deprivation therapy (ADT) compared with ADT alone. However, practice trends in the past 10-15 years have shown increased use of radical prostatectomy with pelvic lymph node dissection for primary management of high-risk, localized disease. Radical prostatectomy, as a primary monotherapy, offers the potential benefits of avoiding ADT, reducing rates of symptomatic local recurrence, enabling full pathological tumour staging and potentially reducing late adverse effects such as secondary malignancy compared with radiation therapy. Retrospective studies have reported wide variability in short-term (pathological) and long-term (oncological) outcomes of radical prostatectomy. Surgical monotherapy continues to be appropriate for selected patients, whereas in others the best treatment strategy probably involves a multimodal approach. Appropriate risk stratification utilizing clinical, pathological and potentially also genomic risk data is imperative in the initial management of men with prostate cancer. However, data from ongoing and planned prospective trials are needed to identify the optimal management strategy for men with high-risk, localized prostate cancer.
Collapse
|
10
|
Angeles AK, Heckmann D, Flosdorf N, Duensing S, Sültmann H. The ERG-Regulated LINC00920 Promotes Prostate Cancer Cell Survival via the 14-3-3ε-FOXO Pathway. Mol Cancer Res 2020; 18:1545-1559. [PMID: 32646965 DOI: 10.1158/1541-7786.mcr-20-0021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/04/2020] [Accepted: 06/29/2020] [Indexed: 11/16/2022]
Abstract
Numerous noncoding transcripts have been reported to correlate with cancer development and progression. Nevertheless, there remains a paucity of long noncoding RNAs (lncRNA) with well-elucidated functional roles. Here, we leverage the International Cancer Genome Consortium-Early Onset Prostate Cancer transcriptome and identify the previously uncharacterized lncRNA LINC00920 to be upregulated in prostate tumors. Phenotypic characterization of LINC00920 revealed its positive impact on cellular proliferation, colony formation, and migration. We demonstrate that LINC00920 transcription is directly activated by ERG, an oncogenic transcription factor overexpressed in 50% of prostate cancers. Chromatin isolation by RNA purification-mass spectrometry revealed the interaction of LINC00920 with the 14-3-3ε protein, leading to enhanced sequestration of tumor suppressive FOXO1. Altogether, our results provide a rationale on how ERG overexpression, partly by driving LINC00920 transcription, could confer survival advantage to prostate cancer cells and potentially prime PTEN-intact prostate cells for cellular transformation through FOXO inactivation. IMPLICATIONS: The study describes a novel lncRNA-mediated mechanism of regulating the FOXO signaling pathway and provides additional insight into the role of ERG in prostate cancer cells.
Collapse
Affiliation(s)
- Arlou Kristina Angeles
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Doreen Heckmann
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Niclas Flosdorf
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Stefan Duensing
- Department of Urology, Section of Molecular Urooncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| |
Collapse
|
11
|
Couñago F, López-Campos F, Díaz-Gavela AA, Almagro E, Fenández-Pascual E, Henríquez I, Lozano R, Linares Espinós E, Gómez-Iturriaga A, de Velasco G, Quintana Franco LM, Rodríguez-Melcón I, López-Torrecilla J, Spratt DE, Guerrero LL, Martínez-Salamanca JI, del Cerro E. Clinical Applications of Molecular Biomarkers in Prostate Cancer. Cancers (Basel) 2020; 12:E1550. [PMID: 32545454 PMCID: PMC7352850 DOI: 10.3390/cancers12061550] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
There is clinically relevant molecular heterogeneity in prostate cancer (PCa), but this biological diversity has had only a minimal impact on clinical practice. Treatment outcomes in patients with localised PCa are often highly variable, even among patients stratified to the same risk group or disease state based on standard clinical and pathological parameters. In recent years, the development of gene panels has provided valuable data on the differential expression of genes in patients with PCa. Nevertheless, there is an urgent need to identify and validate prognostic and predictive biomarkers that can be applied across clinical scenarios, ranging from localised disease to metastatic castration-resistant PCa. The availability of such tools would allow for precision medicine to finally reach PCa patients. In this review, we evaluate current data on molecular biomarkers for PCa, with an emphasis on the biomarkers and gene panels with the most robust evidence to support their application in routine clinical practice.
Collapse
Affiliation(s)
- Felipe Couñago
- Radiation Oncology, Hospital Universitario Quirónsalud Madrid, 28223 Madrid, Spain; (A.A.D.-G.); (L.L.G.); (E.d.C.)
- Radiation Oncology, Hospital La Luz, 28003 Madrid, Spain
- Clinical Department, Faculty of Biomedicine. Universidad Europea de Madrid, 28670 Madrid, Spain
| | | | - Ana Aurora Díaz-Gavela
- Radiation Oncology, Hospital Universitario Quirónsalud Madrid, 28223 Madrid, Spain; (A.A.D.-G.); (L.L.G.); (E.d.C.)
- Radiation Oncology, Hospital La Luz, 28003 Madrid, Spain
- Clinical Department, Faculty of Biomedicine. Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Elena Almagro
- Medical Oncology, Hospital Universitario Quirónsalud Madrid, 28223 Madrid, Spain;
| | - Esaú Fenández-Pascual
- Lyx Institute of Urology, Universidad Francisco de Vitoria, 28006 Madrid, Spain; (E.F.-P.); (E.L.E.)
- Department of Urology, Hospital Universitario La Paz, 28046 Madrid, Spain;
| | - Iván Henríquez
- Radiation Oncology, Hospital Universitario Sant Joan, 43204 Reus, Spain;
| | - Rebeca Lozano
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, 28029 Madrid, Spain;
- Genitourinary Cancer Traslational Research Group, Institute of Biomedical Research, 29010 Málaga, Spain
| | - Estefanía Linares Espinós
- Lyx Institute of Urology, Universidad Francisco de Vitoria, 28006 Madrid, Spain; (E.F.-P.); (E.L.E.)
- Department of Urology, Hospital Universitario La Paz, 28046 Madrid, Spain;
| | | | | | | | - Ignacio Rodríguez-Melcón
- Radiation Oncology, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Spain;
| | - José López-Torrecilla
- Radiation Oncology-ERESA, Hospital General Universitario de Valencia, 46014 Valencia, Spain;
| | - Daniel E. Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Luis Leonardo Guerrero
- Radiation Oncology, Hospital Universitario Quirónsalud Madrid, 28223 Madrid, Spain; (A.A.D.-G.); (L.L.G.); (E.d.C.)
- Radiation Oncology, Hospital La Luz, 28003 Madrid, Spain
- Clinical Department, Faculty of Biomedicine. Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Juan Ignacio Martínez-Salamanca
- Lyx Institute of Urology, Universidad Francisco de Vitoria, 28006 Madrid, Spain; (E.F.-P.); (E.L.E.)
- Department of Urology, Hospital Universitario Puerta de Hierro, 28222 Madrid, Spain
| | - Elia del Cerro
- Radiation Oncology, Hospital Universitario Quirónsalud Madrid, 28223 Madrid, Spain; (A.A.D.-G.); (L.L.G.); (E.d.C.)
- Radiation Oncology, Hospital La Luz, 28003 Madrid, Spain
- Clinical Department, Faculty of Biomedicine. Universidad Europea de Madrid, 28670 Madrid, Spain
| |
Collapse
|
12
|
Yuan Y, Chen X, Huang E. Upregulation of Circular RNA Itchy E3 Ubiquitin Protein Ligase Inhibits Cell Proliferation and Promotes Cell Apoptosis Through Targeting MiR-197 in Prostate Cancer. Technol Cancer Res Treat 2020; 18:1533033819886867. [PMID: 31694481 PMCID: PMC6997844 DOI: 10.1177/1533033819886867] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Objective: This study aimed to investigate the effect of circular RNA itchy E3 ubiquitin protein
ligase on cell proliferation and apoptosis and to explore its target micro-RNAs in
prostate cancer cells. Methods: Circular RNA itchy E3 ubiquitin protein ligase expression in human prostate cancer
cells and normal prostate epithelial cells was determined by real time-quantitative
polymerase chain reaction assay. Circular RNA itchy E3 ubiquitin protein ligase
overexpression plasmids (circular RNA itchy E3 ubiquitin protein ligase(+) group and
control overexpression plasmids group were transfected with PC-3 cells. Rescue
experiment was performed by transfection of circular RNA itchy E3 ubiquitin protein
ligase overexpression and micro-197 overexpression plasmids (circular RNA itchy E3
ubiquitin protein ligase overexpression plasmids/micro RNA (+) group) into PC-3 cells.
Cell Counting Kit-8 and annexin V/propidium iodide assays were conducted to evaluate
cell proliferation and apoptosis, respectively. Western blot was performed to determine
the expressions of apoptotic-related markers. Results: Circular RNA itchy E3 ubiquitin protein ligase expression was decreased in DU 145,
22RV1, VCaP, and PC-3 cells compared to RWPE cells. In PC-3 cells, cell proliferation
rate was reduced in circular RNA itchy E3 ubiquitin protein ligase overexpression
plasmids group compared to control overexpression plasmids group at 48 hours and 72
hours. Cell apoptosis rate was elevated in circular RNA itchy E3 ubiquitin protein
ligase overexpression plasmids group compared to control overexpression plasmids group
at 48 hours, and Western blot showed the similar results. Micro RNA-197 but not micro
RNA-31 or micro RNA-432 was the target micro-RNA of circular RNA itchy E3 ubiquitin
protein ligase. In rescue experiments, cell proliferation rate was elevated, but
apoptosis rate was reduced in circular RNA itchy E3 ubiquitin protein ligase
overexpression plasmids/micro RNA (+) group compared to circular RNA itchy E3 ubiquitin
protein ligase overexpression plasmids group, indicating that circular RNA itchy E3
ubiquitin protein ligase upregulation inhibited cell proliferation but promoted
apoptosis through downregulating micro RNA-197. Conclusion: Circular RNA itchy E3 ubiquitin protein ligase upregulation suppresses cell
proliferation but promotes apoptosis through targeting micro RNA-197 in prostate cancer.
Our study may provide a new insight for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Urology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China.,Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, China
| | - Xiaogang Chen
- Department of Urology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China.,Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, China
| | - Enying Huang
- Department of Urology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China.,Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, China
| |
Collapse
|
13
|
van Dessel LF, van Riet J, Smits M, Zhu Y, Hamberg P, van der Heijden MS, Bergman AM, van Oort IM, de Wit R, Voest EE, Steeghs N, Yamaguchi TN, Livingstone J, Boutros PC, Martens JWM, Sleijfer S, Cuppen E, Zwart W, van de Werken HJG, Mehra N, Lolkema MP. The genomic landscape of metastatic castration-resistant prostate cancers reveals multiple distinct genotypes with potential clinical impact. Nat Commun 2019; 10:5251. [PMID: 31748536 PMCID: PMC6868175 DOI: 10.1038/s41467-019-13084-7] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 10/17/2019] [Indexed: 12/22/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) has a highly complex genomic landscape. With the recent development of novel treatments, accurate stratification strategies are needed. Here we present the whole-genome sequencing (WGS) analysis of fresh-frozen metastatic biopsies from 197 mCRPC patients. Using unsupervised clustering based on genomic features, we define eight distinct genomic clusters. We observe potentially clinically relevant genotypes, including microsatellite instability (MSI), homologous recombination deficiency (HRD) enriched with genomic deletions and BRCA2 aberrations, a tandem duplication genotype associated with CDK12-/- and a chromothripsis-enriched subgroup. Our data suggests that stratification on WGS characteristics may improve identification of MSI, CDK12-/- and HRD patients. From WGS and ChIP-seq data, we show the potential relevance of recurrent alterations in non-coding regions identified with WGS and highlight the central role of AR signaling in tumor progression. These data underline the potential value of using WGS to accurately stratify mCRPC patients into clinically actionable subgroups.
Collapse
Affiliation(s)
- Lisanne F van Dessel
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Job van Riet
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Cancer Computational Biology Center, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Minke Smits
- Department of Medical Oncology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Yanyun Zhu
- Division on Oncogenomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Michiel S van der Heijden
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andries M Bergman
- Division on Oncogenomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Inge M van Oort
- Department of Urology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Ronald de Wit
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Emile E Voest
- Oncode Institute, Utrecht, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Neeltje Steeghs
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Takafumi N Yamaguchi
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Canada
| | - Julie Livingstone
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Canada
| | - Paul C Boutros
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
- Department of Human Genetics, University of California Los Angeles, Los Angeles, USA
- Department of Urology, University of California Los Angeles, Los Angeles, USA
- Jonsson Comprehensive Cancer Centre, University of California Los Angeles, Los Angeles, USA
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - Edwin Cuppen
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht, The Netherlands
- Hartwig Medical Foundation, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division on Oncogenomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Harmen J G van de Werken
- Cancer Computational Biology Center, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands.
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands.
| |
Collapse
|
14
|
Dysregulated Transcriptional Control in Prostate Cancer. Int J Mol Sci 2019; 20:ijms20122883. [PMID: 31200487 PMCID: PMC6627928 DOI: 10.3390/ijms20122883] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/24/2022] Open
Abstract
Recent advances in whole-genome and transcriptome sequencing of prostate cancer at different stages indicate that a large number of mutations found in tumors are present in non-protein coding regions of the genome and lead to dysregulated gene expression. Single nucleotide variations and small mutations affecting the recruitment of transcription factor complexes to DNA regulatory elements are observed in an increasing number of cases. Genomic rearrangements may position coding regions under the novel control of regulatory elements, as exemplified by the TMPRSS2-ERG fusion and the amplified enhancer identified upstream of the androgen receptor (AR) gene. Super-enhancers are increasingly found to play important roles in aberrant oncogenic transcription. Several players involved in these processes are currently being evaluated as drug targets and may represent new vulnerabilities that can be exploited for prostate cancer treatment. They include factors involved in enhancer and super-enhancer function such as bromodomain proteins and cyclin-dependent kinases. In addition, non-coding RNAs with an important gene regulatory role are being explored. The rapid progress made in understanding the influence of the non-coding part of the genome and of transcription dysregulation in prostate cancer could pave the way for the identification of novel treatment paradigms for the benefit of patients.
Collapse
|
15
|
Schwarzenbach H, Gahan PB. MicroRNA Shuttle from Cell-To-Cell by Exosomes and Its Impact in Cancer. Noncoding RNA 2019; 5:E28. [PMID: 30901915 PMCID: PMC6468647 DOI: 10.3390/ncrna5010028] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023] Open
Abstract
The identification of exosomes, their link to multivesicular bodies and their potential role as a messenger vehicle between cancer and healthy cells opens up a new approach to the study of intercellular signaling. Furthermore, the fact that their main cargo is likely to be microRNAs (miRNAs) provides the possibility of the transfer of such molecules to control activities in the recipient cells. This review concerns a brief overview of the biogenesis of both exosomes and miRNAs together with the movement of such structures between cells. The possible roles of miRNAs in the development and progression of breast, ovarian and prostate cancers are discussed.
Collapse
Affiliation(s)
- Heidi Schwarzenbach
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Peter B Gahan
- Fondazione "Enrico Puccinelli" Onlus, 06126 Perugia, Italy.
| |
Collapse
|