1
|
Tao T, Chen H, Xu Q, Li Z, Chen X, Zhou X, Luo W. NKX2-1-AS1 promotes the lymphangiogenesis of lung adenocarcinoma through regulation of ERG-mediated FABP4. Tissue Cell 2024; 87:102314. [PMID: 38309204 DOI: 10.1016/j.tice.2024.102314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
Lymphatic metastasis is a common metastasis of lung adenocarcinoma (LUAD). The current study illustrated the action of lncRNA NKX2-1-AS1 in lymphangiogenesis in LUAD and the underlying mechanisms. Clinical tissue samples were collected for determining NKX2-1-AS1 expression. Then, H441 and H661 cells were selected to perform gain- and loss-of-function assays for dissecting the roles of NKX2-1-AS1 in LUAD cell proliferation and migration. Besides, H441 and H661 cell supernatant was harvested to stimulate HLECs for assessing tube formation ability. Interaction among NKX2-1-AS1, ERG, and fatty acid binding protein 4 (FABP4) was validated through luciferase and RIP assays. NKX2-1-AS1 was highly-expressed in LUAD tissues. Silencing NKX2-1-AS1 suppressed H441 and H661 cell proliferation and migration, reduced expression levels of lymphangiogenesis-related factors (LYVE-1, VEGF-C, VEGFR3, VEGF-A, VEGFR2, and CCR7), and inhibited HLEC tube formation. Interaction validation demonstrated that NKX2-1-AS1 regulated FABP4 transcription by binding to ERG. Overexpression of FABP4 could effectively block the inhibition role of NKX2-1-AS1 silencing in lymphangiogenesis in H441 and H661 cells. This study provided evidence that NKX2-1-AS1 regulated FABP4 transcription by binding to ERG to facilitate the proliferation and migration of LUAD cells and tube formation of HLECs, thus participating in lymphangiogenesis.
Collapse
Affiliation(s)
- Ting Tao
- Department of Pathology, the First Hospital of Changsha, Changsha, Hunan 410005, PR China
| | - Hui Chen
- Department of Pathology, the First Hospital of Changsha, Changsha, Hunan 410005, PR China
| | - Qimei Xu
- Department of Pathology, the First Hospital of Changsha, Changsha, Hunan 410005, PR China
| | - Zhen Li
- Department of Pathology, the First Hospital of Changsha, Changsha, Hunan 410005, PR China
| | - Xuelian Chen
- Department of Respiratory Medicine, the First Hospital of Changsha, Changsha, Hunan 410005, PR China
| | - Xunjian Zhou
- Department of Pathology, the First Hospital of Changsha, Changsha, Hunan 410005, PR China
| | - Wu Luo
- Laboratory Medicine, the First Hospital of Changsha, Changsha, Hunan 410005, PR China.
| |
Collapse
|
2
|
Ma T, Jin L, Bai S, Liu Z, Wang S, Shen B, Cho Y, Cao S, Sun MJS, Fazli L, Zhang D, Wedderburn C, Zhang DY, Mugon G, Ungerleider N, Baddoo M, Zhang K, Schiavone LH, Burkhardt BR, Fan J, You Z, Flemington EK, Dong X, Dong Y. Loss of feedback regulation between FAM3B and androgen receptor driving prostate cancer progression. J Natl Cancer Inst 2024; 116:421-433. [PMID: 37847647 PMCID: PMC10919334 DOI: 10.1093/jnci/djad215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Although the fusion of the transmembrane serine protease 2 gene (TMPRSS2) with the erythroblast transformation-specific-related gene (ERG), or TMPRSS2-ERG, occurs frequently in prostate cancer, its impact on clinical outcomes remains controversial. Roughly half of TMPRSS2-ERG fusions occur through intrachromosomal deletion of interstitial genes and the remainder via insertional chromosomal rearrangements. Because prostate cancers with deletion-derived TMPRSS2-ERG fusions are more aggressive than those with insertional fusions, we investigated the impact of interstitial gene loss on prostate cancer progression. METHODS We conducted an unbiased analysis of transcriptome data from large collections of prostate cancer samples and employed diverse in vitro and in vivo models combined with genetic approaches to characterize the interstitial gene loss that imposes the most important impact on clinical outcome. RESULTS This analysis identified FAM3B as the top-ranked interstitial gene whose loss is associated with a poor prognosis. The association between FAM3B loss and poor clinical outcome extended to fusion-negative prostate cancers where FAM3B downregulation occurred through epigenetic imprinting. Importantly, FAM3B loss drives disease progression in prostate cancer. FAM3B acts as an intermediator of a self-governing androgen receptor feedback loop. Specifically, androgen receptor upregulates FAM3B expression by binding to an intronic enhancer to induce an enhancer RNA and facilitate enhancer-promoter looping. FAM3B, in turn, attenuates androgen receptor signaling. CONCLUSION Loss of FAM3B in prostate cancer, whether through the TMPRSS2-ERG translocation or epigenetic imprinting, causes an exit from this autoregulatory loop to unleash androgen receptor activity and prostate cancer progression. These findings establish FAM3B loss as a new driver of prostate cancer progression and support the utility of FAM3B loss as a biomarker to better define aggressive prostate cancer.
Collapse
Affiliation(s)
- Tianfang Ma
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - Lianjin Jin
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - Shanshan Bai
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Zhan Liu
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Shuo Wang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Beibei Shen
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yeyoung Cho
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - Subing Cao
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Meijuan J S Sun
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ladan Fazli
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - David Zhang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Duke University, Durham, NC, USA
| | - Chiyaro Wedderburn
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Derek Y Zhang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- University of Southern California, Los Angeles, CA, USA
| | - Gavisha Mugon
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Nathan Ungerleider
- Department of Pathology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Melody Baddoo
- Department of Pathology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Kun Zhang
- Department of Computer Science, Bioinformatics Facility of Xavier RCMI Center of Cancer Research, Xavier University of Louisiana, New Orleans, LA, USA
| | | | - Brant R Burkhardt
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Jia Fan
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Zongbing You
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - Erik K Flemington
- Department of Pathology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Xuesen Dong
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Yan Dong
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| |
Collapse
|
3
|
Pakula H, Omar M, Carelli R, Pederzoli F, Fanelli GN, Pannellini T, Socciarelli F, Van Emmenis L, Rodrigues S, Fidalgo-Ribeiro C, Nuzzo PV, Brady NJ, Dinalankara W, Jere M, Valencia I, Saladino C, Stone J, Unkenholz C, Garner R, Alexanderani MK, Khani F, de Almeida FN, Abate-Shen C, Greenblatt MB, Rickman DS, Barbieri CE, Robinson BD, Marchionni L, Loda M. Distinct mesenchymal cell states mediate prostate cancer progression. Nat Commun 2024; 15:363. [PMID: 38191471 PMCID: PMC10774315 DOI: 10.1038/s41467-023-44210-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
In the complex tumor microenvironment (TME), mesenchymal cells are key players, yet their specific roles in prostate cancer (PCa) progression remain to be fully deciphered. This study employs single-cell RNA sequencing to delineate molecular changes in tumor stroma that influence PCa progression and metastasis. Analyzing mesenchymal cells from four genetically engineered mouse models (GEMMs) and correlating these findings with human tumors, we identify eight stromal cell populations with distinct transcriptional identities consistent across both species. Notably, stromal signatures in advanced mouse disease reflect those in human bone metastases, highlighting periostin's role in invasion and differentiation. From these insights, we derive a gene signature that predicts metastatic progression in localized disease beyond traditional Gleason scores. Our results illuminate the critical influence of stromal dynamics on PCa progression, suggesting new prognostic tools and therapeutic targets.
Collapse
Affiliation(s)
- Hubert Pakula
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Mohamed Omar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY, 10021, USA
| | - Ryan Carelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Filippo Pederzoli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Giuseppe Nicolò Fanelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Department of Laboratory Medicine, Pisa University Hospital, Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, 56126, Italy
| | - Tania Pannellini
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Fabio Socciarelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Lucie Van Emmenis
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Silvia Rodrigues
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Caroline Fidalgo-Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Pier Vitale Nuzzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Nicholas J Brady
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Wikum Dinalankara
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Madhavi Jere
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Itzel Valencia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Christopher Saladino
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jason Stone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Caitlin Unkenholz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Richard Garner
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Mohammad K Alexanderani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Francesca Khani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Francisca Nunes de Almeida
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Cory Abate-Shen
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Urology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - David S Rickman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Christopher E Barbieri
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY, 10021, USA
- Department of Urology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY, 10021, USA
- Department of Urology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY, 10021, USA.
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA, 02215, USA.
- University of Oxford, Nuffield Department of Surgical Sciences, Oxford, UK.
| |
Collapse
|
4
|
Schimmelpfennig C, Rade M, Füssel S, Löffler D, Blumert C, Bertram C, Borkowetz A, Otto DJ, Puppel SH, Hönscheid P, Sommer U, Baretton GB, Köhl U, Wirth M, Thomas C, Horn F, Kreuz M, Reiche K. Characterization and evaluation of gene fusions as a measure of genetic instability and disease prognosis in prostate cancer. BMC Cancer 2023; 23:575. [PMID: 37349736 DOI: 10.1186/s12885-023-11019-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/27/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) is one of the most prevalent cancers worldwide. The clinical manifestations and molecular characteristics of PCa are highly variable. Aggressive types require radical treatment, whereas indolent ones may be suitable for active surveillance or organ-preserving focal therapies. Patient stratification by clinical or pathological risk categories still lacks sufficient precision. Incorporating molecular biomarkers, such as transcriptome-wide expression signatures, improves patient stratification but so far excludes chromosomal rearrangements. In this study, we investigated gene fusions in PCa, characterized potential novel candidates, and explored their role as prognostic markers for PCa progression. METHODS We analyzed 630 patients in four cohorts with varying traits regarding sequencing protocols, sample conservation, and PCa risk group. The datasets included transcriptome-wide expression and matched clinical follow-up data to detect and characterize gene fusions in PCa. With the fusion calling software Arriba, we computationally predicted gene fusions. Following detection, we annotated the gene fusions using published databases for gene fusions in cancer. To relate the occurrence of gene fusions to Gleason Grading Groups and disease prognosis, we performed survival analyses using the Kaplan-Meier estimator, log-rank test, and Cox regression. RESULTS Our analyses identified two potential novel gene fusions, MBTTPS2,L0XNC01::SMS and AMACR::AMACR. These fusions were detected in all four studied cohorts, providing compelling evidence for the validity of these fusions and their relevance in PCa. We also found that the number of gene fusions detected in a patient sample was significantly associated with the time to biochemical recurrence in two of the four cohorts (log-rank test, p-value < 0.05 for both cohorts). This was also confirmed after adjusting the prognostic model for Gleason Grading Groups (Cox regression, p-values < 0.05). CONCLUSIONS Our gene fusion characterization workflow revealed two potential novel fusions specific for PCa. We found evidence that the number of gene fusions was associated with the prognosis of PCa. However, as the quantitative correlations were only moderately strong, further validation and assessment of clinical value is required before potential application.
Collapse
Affiliation(s)
- Carolin Schimmelpfennig
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Michael Rade
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Susanne Füssel
- Department of Urology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Dennis Löffler
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Conny Blumert
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Catharina Bertram
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Angelika Borkowetz
- Department of Urology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Dominik J Otto
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Sven-Holger Puppel
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Pia Hönscheid
- Institute of Pathology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ulrich Sommer
- Institute of Pathology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Gustavo B Baretton
- Institute of Pathology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Köhl
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Institute of Clinical Immunology, Medical Faculty, University Hospital, University of Leipzig, Leipzig, Germany
| | - Manfred Wirth
- Department of Urology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christian Thomas
- Department of Urology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Friedemann Horn
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Markus Kreuz
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Kristin Reiche
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.
- Institute of Clinical Immunology, Medical Faculty, University Hospital, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
5
|
Kong Y, Jiang C, Wei G, Sun K, Wang R, Qiu T. Small Molecule Inhibitors as Therapeutic Agents Targeting Oncogenic Fusion Proteins: Current Status and Clinical. Molecules 2023; 28:4672. [PMID: 37375228 DOI: 10.3390/molecules28124672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/30/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Oncogenic fusion proteins, arising from chromosomal rearrangements, have emerged as prominent drivers of tumorigenesis and crucial therapeutic targets in cancer research. In recent years, the potential of small molecular inhibitors in selectively targeting fusion proteins has exhibited significant prospects, offering a novel approach to combat malignancies harboring these aberrant molecular entities. This review provides a comprehensive overview of the current state of small molecular inhibitors as therapeutic agents for oncogenic fusion proteins. We discuss the rationale for targeting fusion proteins, elucidate the mechanism of action of inhibitors, assess the challenges associated with their utilization, and provide a summary of the clinical progress achieved thus far. The objective is to provide the medicinal community with current and pertinent information and to expedite the drug discovery programs in this area.
Collapse
Affiliation(s)
- Yichao Kong
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Caihong Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Guifeng Wei
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Kai Sun
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Ruijie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Ting Qiu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
6
|
Warli SM, Warli MH, Prapiska FF. PCA3 and TMPRSS2: ERG Urine Level as Diagnostic Biomarker of Prostate Cancer. Res Rep Urol 2023; 15:149-155. [PMID: 37181497 PMCID: PMC10167967 DOI: 10.2147/rru.s401131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Background Prostate cancer is a highly prevalent urological carcinoma with an increasing incidence in Indonesia and all around the world. Early diagnosis can greatly affect treatment outcomes and increase life expectancy. Several biomarkers for detecting prostate cancer have been studied and showed great promise. Purpose This study aims to analyze prostate cancer antigen 3 (PCA3) as well as transmembrane serine protease 2:ERG (TMPRSS2:ERG) for diagnosing and serving as urine biomarkers in predicting prostate cancer incidences. Methods We conducted an analytical study to assess the utility of PCA3 and TMPRSS2:ERG for detecting prostate cancer. Thirty samples were included in this study to see the utilization of PCA3 and TMPRSS2:ERG as diagnostic biomarkers of prostate cancer. A urine sample was taken and the PCA3 test was performed using the PCA3 PROGENSA test, while the TMPRSS2:ERG was performed using the chemiluminescent DNA probe method with a hybridization protection test. Results The average age of the subject was 61.07±8.3 years. Based on calculations using the Mann-Whitney test, there was a significant relationship between prostate-Specific Antigen (PSA) overexpression (p<0.001), TMPRSS2:ERG (p=0.001), and PCA3 (p=0.003) with prostate cancer incidence. The sensitivity of PCA3 and TMPRSS2:ERG in detecting prostate cancer was 76.9% and 92.3%, respectively. Hence, TMPRSS2:ERG and PCA3 can be used as biomarkers for the occurrence of prostate cancer. We also performed a Kruskal-Wallis test; however, there was no significant relationship between PSA (p=0.236), TMPRSS2:ERG (p=0.801), and PCA3 (p=0.091) with the Gleason score. Conclusion There is a significant correlation between overexpression of PSA, TMPRSS2:ERG and PCA3 with the incidence of prostate cancer, and TMPRSS2:ERG and PCA3 can be used as biomarkers of prostate cancer.
Collapse
Affiliation(s)
- Syah Mirsya Warli
- Department of Urology, Faculty of Medicine Universitas Sumatera Utara – Universitas Sumatera Utara Hospital, Medan, North Sumatera, Indonesia
- Department of Surgery Urology Division, Faculty of Medicine Universitas Sumatera Utara – Haji Adam Malik General Hospital, Medan, North Sumatera, Indonesia
| | - Muhammad Haritsyah Warli
- Department of Urology, Faculty of Medicine Universitas Indonesia – Haji Adam Malik General Hospital, Medan, North Sumatera, Indonesia
| | - Fauriski Febrian Prapiska
- Department of Surgery Urology Division, Faculty of Medicine Universitas Sumatera Utara – Haji Adam Malik General Hospital, Medan, North Sumatera, Indonesia
| |
Collapse
|
7
|
Acosta-Vega NL, Varela R, Mesa JA, Garai J, Baddoo MC, Gómez-Gutiérrez A, Serrano-Gómez SJ, Lemus MN, Serrano ML, Zabaleta J, Combita AL, Sanabria-Salas MC. Metabolic pathways enriched according to ERG status are associated with biochemical recurrence in Hispanic/Latino patients with prostate cancer. Cancer Med 2023; 12:4306-4320. [PMID: 36329628 PMCID: PMC9972164 DOI: 10.1002/cam4.5301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/28/2022] [Accepted: 09/15/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The role of ERG-status molecular subtyping in prognosis of prostate cancer (PCa) is still under debate. In this study, we identified differentially expressed genes (DEGs) according to ERG-status to explore their enriched pathways and implications in prognosis in Hispanic/Latino PCa patients. METHODS RNA from 78 Hispanic PCa tissues from radical prostatectomies (RP) were used for RNA-sequencing. ERGhigh /ERGlow tumor groups were determined based on the 1.5-fold change median expression in non-tumor samples. DEGs with a False Discovery Rate (FDR) < 0.01 and a fold change >2 were identified between ERGhigh and ERGlow tumors and submitted to enrichment analysis in MetaCore. Survival and association analyses were performed to evaluate biochemical recurrence (BCR)-free survival. RESULTS The identification of 150 DEGs between ERGhigh and ERGlow tumors revealed clustering of most of the non-BCR cases (60%) into de ERGhigh group and most of the BCR cases (60.8%) in ERGlow group. Kaplan-Meier survival curves showed a worst BCR-free survival for ERGlow patients, and a significant reduced risk of BCR was observed for ERGhigh cases (OR = 0.29 (95%CI, 0.10-0.8)). Enrichment pathway analysis identified metabolic-related pathways, such as the renin-angiotensin system and angiotensin maturation system, the linoleic acid metabolism, and polyamines metabolism in these ERG groups. CONCLUSIONS ERGlow tumor cases were associated with poor BCR-free survival in our Hispanic/Latino patients, with metabolism-related pathways altered in the BCR progression. IMPACT Our findings suggest the need to dissect the role of diet, metabolism, and lifestyle as risk factors for more aggressive PCa subtypes.
Collapse
Affiliation(s)
- Natalia L Acosta-Vega
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología de Colombia, Bogotá, DC, Colombia.,Programa de doctorado en Ciencias Biológicas, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Rodolfo Varela
- Departamento de Urología, Instituto Nacional de Cancerología de Colombia, Bogotá, DC, Colombia
| | - Jorge Andrés Mesa
- Departamento de Patología Oncológica, Instituto Nacional de Cancerología de Colombia, Bogotá, DC, Colombia
| | - Jone Garai
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Melody C Baddoo
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Alberto Gómez-Gutiérrez
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Silvia J Serrano-Gómez
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología de Colombia, Bogotá, DC, Colombia
| | - Marcela Nuñez Lemus
- Grupo de Apoyo y Seguimiento para la Investigación, Instituto Nacional de Cancerología de Colombia, Bogotá, DC, Colombia
| | - Martha Lucía Serrano
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología de Colombia, Bogotá, DC, Colombia.,Departamento de Química, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, DC, Colombia
| | - Jovanny Zabaleta
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Department of Interdisciplinary Oncology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Alba L Combita
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología de Colombia, Bogotá, DC, Colombia.,Departamento de Microbiología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, DC, Colombia
| | | |
Collapse
|
8
|
Lasorsa F, di Meo NA, Rutigliano M, Ferro M, Terracciano D, Tataru OS, Battaglia M, Ditonno P, Lucarelli G. Emerging Hallmarks of Metabolic Reprogramming in Prostate Cancer. Int J Mol Sci 2023; 24:ijms24020910. [PMID: 36674430 PMCID: PMC9863674 DOI: 10.3390/ijms24020910] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Prostate cancer (PCa) is the most common male malignancy and the fifth leading cause of cancer death in men worldwide. Prostate cancer cells are characterized by a hybrid glycolytic/oxidative phosphorylation phenotype determined by androgen receptor signaling. An increased lipogenesis and cholesterogenesis have been described in PCa cells. Many studies have shown that enzymes involved in these pathways are overexpressed in PCa. Glutamine becomes an essential amino acid for PCa cells, and its metabolism is thought to become an attractive therapeutic target. A crosstalk between cancer and stromal cells occurs in the tumor microenvironment because of the release of different cytokines and growth factors and due to changes in the extracellular matrix. A deeper insight into the metabolic changes may be obtained by a multi-omic approach integrating genomics, transcriptomics, metabolomics, lipidomics, and radiomics data.
Collapse
Affiliation(s)
- Francesco Lasorsa
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Nicola Antonio di Meo
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Monica Rutigliano
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy
| | - Octavian Sabin Tataru
- The Institution Organizing University Doctoral Studies (I.O.S.U.D.), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, 540142 Târgu Mureș, Romania
| | - Michele Battaglia
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Pasquale Ditonno
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy
- Correspondence: or
| |
Collapse
|
9
|
Hiregange DG, Rivalta A, Yonath A, Zimmerman E, Bashan A, Yonath H. Mutations in RPS19 may affect ribosome function and biogenesis in Diamond Blackfan Anemia. FEBS Open Bio 2022; 12:1419-1434. [PMID: 35583751 PMCID: PMC9249338 DOI: 10.1002/2211-5463.13444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/04/2022] [Accepted: 05/17/2022] [Indexed: 11/12/2022] Open
Abstract
Ribosomes, the cellular organelles translating the genetic code to proteins, are assemblies of RNA chains and many proteins (RPs) arranged in precise fine-tuned interwoven structures. Mutated ribosomal genes cause ribosomopathies, including Diamond Blackfan Anemia (DBA, a rare heterogeneous red-cell aplasia connected to ribosome malfunction) or failed biogenesis. Combined bioinformatical, structural, and predictive analyses of potential consequences of possibly expressed mutations in eS19, the protein product of the highly mutated RPS19, suggests that mutations in its exposed surface could alter its positioning during assembly and consequently prevent biogenesis, implying a natural selective strategy to avoid malfunctions in ribosome assembly. A search for RPS19 pseudogenes indicated >90% sequence identity with the wild type, hinting at its expression in cases of absent or truncated gene products.
Collapse
Affiliation(s)
| | - Andre Rivalta
- The Department of Chemical and Structural Biology, Weizmann Institute of Science, Israel
| | - Ada Yonath
- The Department of Chemical and Structural Biology, Weizmann Institute of Science, Israel
| | - Ella Zimmerman
- The Department of Chemical and Structural Biology, Weizmann Institute of Science, Israel
| | - Anat Bashan
- The Department of Chemical and Structural Biology, Weizmann Institute of Science, Israel
| | - Hagith Yonath
- Internal Medicine A and Genetics Institute Sheba Medical Center, and Sackler School of Medicine, Tel Aviv University, Israel
| |
Collapse
|
10
|
Mangolini A, Rocca C, Bassi C, Ippolito C, Negrini M, Dell'Atti L, Lanza G, Gafà R, Bianchi N, Pinton P, Aguiari G. DETECTION OF DISEASE‐CAUSING MUTATIONS IN PROSTATE CANCER BY NGS SEQUENCING. Cell Biol Int 2022; 46:1047-1061. [PMID: 35347810 PMCID: PMC9320837 DOI: 10.1002/cbin.11803] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/27/2022] [Indexed: 11/11/2022]
Abstract
Gene mutations may affect the fate of many tumors including prostate cancer (PCa); therefore, the research of specific mutations associated with tumor outcomes might help the urologist to identify the best therapy for PCa patients such as surgical resection, adjuvant therapy or active surveillance. Genomic DNA (gDNA) was extracted from 48 paraffin‐embedded PCa samples and normal paired tissues. Next, gDNA was amplified and analyzed by next‐generation sequencing (NGS) using a specific gene panel for PCa. Raw data were refined to exclude false‐positive mutations; thus, variants with coverage and frequency lower than 100× and 5%, respectively were removed. Mutation significance was processed by Genomic Evolutionary Rate Profiling, ClinVar, and Varsome tools. Most of 3000 mutations (80%) were single nucleotide variants and the remaining 20% indels. After raw data elaboration, 312 variants were selected. Most mutated genes were KMT2D (26.45%), FOXA1 (16.13%), ATM (15.81%), ZFHX3 (9.35%), TP53 (8.06%), and APC (5.48%). Hot spot mutations in FOXA1, ATM, ZFHX3, SPOP, and MED12 were also found. Truncating mutations of ATM, lesions lying in hot spot regions of SPOP and FOXA1 as well as mutations of TP53 correlated with poor prognosis. Importantly, we have also found some germline mutations associated with hereditary cancer‐predisposing syndrome. gDNA sequencing of 48 cancer tissues by NGS allowed to detect new tumor variants as well as confirmed lesions in genes linked to prostate cancer. Overall, somatic and germline mutations linked to good/poor prognosis could represent new prognostic tools to improve the management of PCa patients.
Collapse
Affiliation(s)
- Alessandra Mangolini
- Department of Neuroscience and RehabilitationUniversity of Ferraravia fossato di mortara, 7444121FerraraItaly
| | - Christian Rocca
- UO Urology, St Anna Hospital, via Aldo Moro 844124FerraraItaly
| | - Cristian Bassi
- Department of Translational MedicineUniversity of Ferraravia Luigi Borsari 4644121FerraraItaly
| | | | - Massimo Negrini
- Department of Translational MedicineUniversity of Ferraravia Luigi Borsari 4644121FerraraItaly
| | - Lucio Dell'Atti
- Division of Urology, Department of Clinical, Special and Dental Science, University Hospital "Ospedali Riuniti", Marche Polytechnic University, 71 Conca Street60126AnconaItaly
| | - Giovanni Lanza
- Department of Translational MedicineUniversity of Ferraravia Luigi Borsari 4644121FerraraItaly
| | - Roberta Gafà
- Department of Translational MedicineUniversity of Ferraravia Luigi Borsari 4644121FerraraItaly
| | - Nicoletta Bianchi
- Department of Translational MedicineUniversity of Ferraravia Luigi Borsari 4644121FerraraItaly
| | - Paolo Pinton
- Department of Medical SciencesUniversity of Ferraravia fossato di mortara, 64/B44121FerraraItaly
| | - Gianluca Aguiari
- Department of Neuroscience and RehabilitationUniversity of Ferraravia fossato di mortara, 7444121FerraraItaly
| |
Collapse
|
11
|
Chimeric RNA Design Principles for RNA-Mediated Gene Fusion. Cells 2022; 11:cells11061002. [PMID: 35326453 PMCID: PMC8947500 DOI: 10.3390/cells11061002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 12/02/2022] Open
Abstract
One common genetic alteration in cancer is gene fusion resulting from chromosomal translocations. The mechanisms that create such oncogenic fusion genes are not well understood. Previously, we provided the direct evidence that expression of a designed chimeric RNA can drive the formation of TMPRSS2-ERG gene fusion. Central to this RNA-mediated gene fusion mechanism is a proposed three-way junction formed by RNA/DNA hybrid and the intergenic DNA stem formed by target genes. In this study, we determined the important parameters for chimeric RNA-mediated gene fusion using TMPRSS2-ERG fusion gene as the model. Our results indicate that both the chimeric RNA lengths and the sizes of unpaired bulges play important roles in inducing TMPRSS2-ERG gene fusion. The optimal length of unpaired bulges was about 35 nt, while the optimal chimeric RNA length was about 50 nt for targeting. These observations were consistent regardless of the target locations within TMPRSS2 and ERG genes. These empirically determined parameters provide important insight for searching cellular RNAs that may initiate oncogenic fusion genes. The knowledge could also facilitate the development of useful genomic technology for manipulating mammalian genomes.
Collapse
|
12
|
Liao C, Wang Q, An J, Zhang M, Chen J, Li X, Xiao L, Wang J, Long Q, Liu J, Guan X. SPINKs in Tumors: Potential Therapeutic Targets. Front Oncol 2022; 12:833741. [PMID: 35223512 PMCID: PMC8873584 DOI: 10.3389/fonc.2022.833741] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
The serine protease inhibitor Kazal type (SPINK) family includes SPINK1-14 and is the largest branch in the serine protease inhibitor family. SPINKs play an important role in pancreatic physiology and disease, sperm maturation and capacitation, Nager syndrome, inflammation and the skin barrier. Evidence shows that the unregulated expression of SPINK1, 2, 4, 5, 6, 7, and 13 is closely related to human tumors. Different SPINKs exhibit various regulatory modes in different tumors and can be used as tumor prognostic markers. This article reviews the role of SPINK1, 2, 4, 5, 6, 7, and 13 in different human cancer processes and helps to identify new cancer treatment targets.
Collapse
Affiliation(s)
- Chengcheng Liao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Qian Wang
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, Zunyi, China
| | - Jiaxing An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Minglin Zhang
- Department of Gastroenterology, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, China
| | - Jie Chen
- Department of Urology, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaolan Li
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, Zunyi, China
| | - Linlin Xiao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Jiajia Wang
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Qian Long
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Qian Long, ; Xiaoyan Guan, ; Jianguo Liu,
| | - Jianguo Liu
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Qian Long, ; Xiaoyan Guan, ; Jianguo Liu,
| | - Xiaoyan Guan
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Qian Long, ; Xiaoyan Guan, ; Jianguo Liu,
| |
Collapse
|
13
|
Grossmann S, Hooks Y, Wilson L, Moore L, O'Neill L, Martincorena I, Voet T, Stratton MR, Heer R, Campbell PJ. Development, maturation, and maintenance of human prostate inferred from somatic mutations. Cell Stem Cell 2021; 28:1262-1274.e5. [PMID: 33657416 PMCID: PMC8260206 DOI: 10.1016/j.stem.2021.02.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 11/19/2020] [Accepted: 02/02/2021] [Indexed: 01/23/2023]
Abstract
Clonal dynamics and mutation burden in healthy human prostate epithelium are relevant to prostate cancer. We sequenced whole genomes from 409 microdissections of normal prostate epithelium across 8 donors, using phylogenetic reconstruction with spatial mapping in a 59-year-old man's prostate to reconstruct tissue dynamics across the lifespan. Somatic mutations accumulate steadily at ∼16 mutations/year/clone, with higher rates in peripheral than peri-urethral regions. The 24-30 independent glandular subunits are established as rudimentary ductal structures during fetal development by 5-10 embryonic cells each. Puberty induces formation of further side and terminal branches by local stem cells disseminated throughout the rudimentary ducts during development. During adult tissue maintenance, clonal expansions have limited geographic scope and minimal migration. Driver mutations are rare in aging prostate epithelium, but the one driver we did observe generated a sizable intraepithelial clonal expansion. Leveraging unbiased clock-like mutations, we define prostate stem cell dynamics through fetal development, puberty, and aging.
Collapse
Affiliation(s)
- Sebastian Grossmann
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Yvette Hooks
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Laura Wilson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Luiza Moore
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Laura O'Neill
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Iñigo Martincorena
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Thierry Voet
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK; Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Michael R Stratton
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Rakesh Heer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK.
| | - Peter J Campbell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK; Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK.
| |
Collapse
|
14
|
Kobelyatskaya AA, Pudova EA, Snezhkina AV, Fedorova MS, Pavlov VS, Guvatova ZG, Savvateeva MV, Melnikova NV, Dmitriev AA, Trofimov DY, Sukhikh GT, Nyushko KM, Alekseev BY, Razin SV, Krasnov GS, Kudryavtseva AV. Impact TMPRSS2-ERG Molecular Subtype on Prostate Cancer Recurrence. Life (Basel) 2021; 11:588. [PMID: 34205581 PMCID: PMC8234735 DOI: 10.3390/life11060588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/07/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, seven molecular subtypes of prostate cancer (PCa) are known, the most common of which being the subtype characterized by the presence of the TMPRSS2-ERG fusion transcript. While there is a considerable amount of work devoted to the influence of this transcript on the prognosis of the disease, data on its role in the progression and prognosis of PCa remain controversial. The present study is devoted to the analysis of the association between the TMPRSS2-ERG transcript and the biochemical recurrence of PCa. The study included two cohorts: the RNA-Seq sample of Russian patients with PCa (n = 72) and the TCGA-PRAD data (n = 203). The results of the analysis of the association between the TMPRSS2-ERG transcript and biochemical recurrence were contradictory. The differential expression analysis (biochemical recurrence cases versus biochemical recurrence-free) and the gene set enrichment analysis revealed a list of genes involved in major cellular pathways. The GNL3, QSOX2, SSPO, and SYS1 genes were selected as predictors of the potential prognostic model (AUC = 1.000 for a cohort of Russian patients with PCa and AUC = 0.779 for a TCGA-PRAD cohort).
Collapse
Affiliation(s)
- Anastasiya A. Kobelyatskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
- Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Elena A. Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
| | - Anastasiya V. Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
| | - Maria S. Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
| | - Vladislav S. Pavlov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
| | - Zulfiya G. Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
| | - Maria V. Savvateeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
| | - Nataliya V. Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
| | - Alexey A. Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
| | - Dmitry Y. Trofimov
- Gynecology and Perinatology named after Academician V.I. Kulakov, National Medical Research Center for Obstetrics, Ministry of Health of the Russian Federation, 117997 Moscow, Russia; (D.Y.T.); (G.T.S.)
| | - Gennady T. Sukhikh
- Gynecology and Perinatology named after Academician V.I. Kulakov, National Medical Research Center for Obstetrics, Ministry of Health of the Russian Federation, 117997 Moscow, Russia; (D.Y.T.); (G.T.S.)
| | - Kirill M. Nyushko
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, 125284 Moscow, Russia; (K.M.N.); (B.Y.A.)
| | - Boris Y. Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, 125284 Moscow, Russia; (K.M.N.); (B.Y.A.)
| | - Sergey V. Razin
- Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - George S. Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
| | - Anna V. Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.A.P.); (A.V.S.); (M.S.F.); (V.S.P.); (Z.G.G.); (M.V.S.); (N.V.M.); (A.A.D.); (G.S.K.)
| |
Collapse
|
15
|
Videm P, Kumar A, Zharkov O, Grüning BA, Backofen R. ChiRA: an integrated framework for chimeric read analysis from RNA-RNA interactome and RNA structurome data. Gigascience 2021; 10:giaa158. [PMID: 33511995 PMCID: PMC7844879 DOI: 10.1093/gigascience/giaa158] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/26/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND With the advances in next-generation sequencing technologies, it is possible to determine RNA-RNA interaction and RNA structure predictions on a genome-wide level. The reads from these experiments usually are chimeric, with each arm generated from one of the interaction partners. Owing to short read lengths, often these sequenced arms ambiguously map to multiple locations. Thus, inferring the origin of these can be quite complicated. Here we present ChiRA, a generic framework for sensitive annotation of these chimeric reads, which in turn can be used to predict the sequenced hybrids. RESULTS Grouping reference loci on the basis of aligned common reads and quantification improved the handling of the multi-mapped reads in contrast to common strategies such as the selection of the longest hit or a random choice among all hits. On benchmark data ChiRA improved the number of correct alignments to the reference up to 3-fold. It is shown that the genes that belong to the common read loci share the same protein families or similar pathways. In published data, ChiRA could detect 3 times more new interactions compared to existing approaches. In addition, ChiRAViz can be used to visualize and filter large chimeric datasets intuitively. CONCLUSION ChiRA tool suite provides a complete analysis and visualization framework along with ready-to-use Galaxy workflows and tutorials for RNA-RNA interactome and structurome datasets. Common read loci built by ChiRA can rescue multi-mapped reads on paralogous genes without requiring any information on gene relations. We showed that ChiRA is sensitive in detecting new RNA-RNA interactions from published RNA-RNA interactome datasets.
Collapse
Affiliation(s)
- Pavankumar Videm
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Georges-Koehler-Allee 106, 79110 Freiburg, Germany
| | - Anup Kumar
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Georges-Koehler-Allee 106, 79110 Freiburg, Germany
| | - Oleg Zharkov
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Georges-Koehler-Allee 106, 79110 Freiburg, Germany
| | - Björn Andreas Grüning
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Georges-Koehler-Allee 106, 79110 Freiburg, Germany
| | - Rolf Backofen
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Georges-Koehler-Allee 106, 79110 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schaenzlestr. 18, 79104 Freiburg, Germany
| |
Collapse
|
16
|
Kalkanli A, Kirkik D, Bostanci E, Tas SK. The Important Role of TMPRSS2 Gene in Covid-19 and Prostate Cancer: In Silico Approach. BRAZILIAN ARCHIVES OF BIOLOGY AND TECHNOLOGY 2021; 64. [DOI: 10.1590/1678-4324-2021200785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
|
17
|
Chakravarty D, Huang L, Kahn M, Tewari AK. Immunotherapy for Metastatic Prostate Cancer: Current and Emerging Treatment Options. Urol Clin North Am 2020; 47:487-510. [PMID: 33008499 DOI: 10.1016/j.ucl.2020.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The advent of immunotherapy has revolutionized cancer treatment. Prostate cancer has an immunosuppressive microenvironment and a low tumor mutation burden, resulting in low neoantigen expression. The consensus was that immunotherapy would be less effective in prostate cancer. However, recent studies have reported that prostate cancer does have a high number of DNA damage and repair gene defects. Immunotherapies that have been tested in prostate cancer so far have been mainly vaccines and checkpoint inhibitors. A combination of genomically targeted therapies, with approaches to alleviate immune response and thereby make the tumor microenvironment immunologically hot, is promising.
Collapse
Affiliation(s)
- Dimple Chakravarty
- Department of Urology and the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Li Huang
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Matthew Kahn
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ashutosh K Tewari
- Department of Urology and the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
18
|
Zhang W, Dong Y, Sartor O, Flemington EK, Zhang K. SEER and Gene Expression Data Analysis Deciphers Racial Disparity Patterns in Prostate Cancer Mortality and the Public Health Implication. Sci Rep 2020; 10:6820. [PMID: 32321981 PMCID: PMC7176737 DOI: 10.1038/s41598-020-63764-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 03/30/2020] [Indexed: 01/13/2023] Open
Abstract
A major racial disparity in prostate cancer (PCa) is that African American (AA) patients have a higher mortality rate than European American (EA) patients. We filtered the SEER 2009-2011 records and divided them into four groups regarding patient races and cancer grades. On such a partition, we performed a series of statistical analyses to further clarify the aforementioned disparity. Molecular evidence for a primary result of the epidemiological analysis was obtained from gene expression data. The results include: (1) Based on the registry-specific measures, a significant linear regression of total mortality rate (as well as PCa specific mortality rate) on the percentage of (Gleason pattern-based) high-grade cancers (PHG) is demonstrated in EAs (p < 0.01) but not in AAs; (2) PHG and its racial disparity are differentiated across ages and the groups defined by patient outcomes; (3) For patients with cancers in the same grade category, i.e. the high or low grade, the survival stratification between races is not significant in most geographical areas; and (4) The genes differentially expressed between AAs' and EAs' tumors of the same grade category are relatively rare. The perception that prostate tumors are more lethal in AAs than in EAs is reasonable regarding AAs' higher PHG, while high grade alone could not imply aggressiveness. However, this perception is questionable when the comparison is focused on cases within the same grade category. Supporting observations for this conclusion hold a remarkable implication for erasing racial disparity in PCa. That is, "Equal grade, equal outcomes" is not only a verifiable hypothesis but also an achievable public health goal.
Collapse
Affiliation(s)
- Wensheng Zhang
- Bioinformatics Core of Xavier NIH RCMI Center of Cancer Research; Department of Computer Science, Xavier University of Louisiana, New Orleans, 70125, LA, USA
| | - Yan Dong
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, 70112, LA, USA
| | - Oliver Sartor
- Department of Medicine, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, 70112, LA, USA
| | - Erik K Flemington
- Department of Pathology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, 70112, LA, USA
| | - Kun Zhang
- Bioinformatics Core of Xavier NIH RCMI Center of Cancer Research; Department of Computer Science, Xavier University of Louisiana, New Orleans, 70125, LA, USA.
| |
Collapse
|
19
|
Identifying FL11 subtype by characterizing tumor immune microenvironment in prostate adenocarcinoma via Chou's 5-steps rule. Genomics 2020; 112:1500-1515. [DOI: 10.1016/j.ygeno.2019.08.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/03/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022]
|
20
|
Circulating cell-free DNA: Translating prostate cancer genomics into clinical care. Mol Aspects Med 2020; 72:100837. [PMID: 31954523 DOI: 10.1016/j.mam.2019.100837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/12/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
Abstract
Only in the past decade tremendous advances have been made in understanding prostate cancer genomics and consequently in applying new treatment strategies. As options regarding treatments are increasing so are the challenges in selecting the right treatment option for each patient and not the least, understanding the optimal time-point and sequence of applying available treatments. Critically, without reliable methods that enable sequential monitoring of evolving genotypes in individual patients, we will never reach effective personalised driven treatment approaches. This review focuses on the clinical implications of prostate cancer genomics and the potential of cfDNA in facilitating treatment management.
Collapse
|
21
|
Chen X, Wan L, Wang W, Xi WJ, Yang AG, Wang T. Re-recognition of pseudogenes: From molecular to clinical applications. Theranostics 2020; 10:1479-1499. [PMID: 32042317 PMCID: PMC6993246 DOI: 10.7150/thno.40659] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022] Open
Abstract
Pseudogenes were initially regarded as "nonfunctional" genomic elements that did not have protein-coding abilities due to several endogenous inactivating mutations. Although pseudogenes are widely expressed in prokaryotes and eukaryotes, for decades, they have been largely ignored and classified as gene "junk" or "relics". With the widespread availability of high-throughput sequencing analysis, especially omics technologies, knowledge concerning pseudogenes has substantially increased. Pseudogenes are evolutionarily conserved and derive primarily from a mutation or retrotransposon, conferring the pseudogene with a "gene repository" role to store and expand genetic information. In contrast to previous notions, pseudogenes have a variety of functions at the DNA, RNA and protein levels for broadly participating in gene regulation to influence the development and progression of certain diseases, especially cancer. Indeed, some pseudogenes have been proven to encode proteins, strongly contradicting their "trash" identification, and have been confirmed to have tissue-specific and disease subtype-specific expression, indicating their own value in disease diagnosis. Moreover, pseudogenes have been correlated with the life expectancy of patients and exhibit great potential for future use in disease treatment, suggesting that they are promising biomarkers and therapeutic targets for clinical applications. In this review, we summarize the natural properties, functions, disease involvement and clinical value of pseudogenes. Although our knowledge of pseudogenes remains nascent, this field deserves more attention and deeper exploration.
Collapse
|
22
|
Malik A, Srinivasan S, Batra J. A New Era of Prostate Cancer Precision Medicine. Front Oncol 2019; 9:1263. [PMID: 31850193 PMCID: PMC6901987 DOI: 10.3389/fonc.2019.01263] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/31/2019] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer is the second most common male cancer affecting Western society. Despite substantial advances in the exploration of prostate cancer biomarkers and treatment strategies, men are over diagnosed with inert prostate cancer, while there is also a substantial mortality from the invasive disease. Precision medicine is the management of treatment profiles across different cancers predicting therapies for individual cancer patients. With strategies including individual genomic profiling and targeting specific cancer pathways, precision medicine for prostate cancer has the potential to impose changes in clinical practices. Some of the recent advances in prostate cancer precision medicine comprise targeting gene fusions, genome editing tools, non-coding RNA biomarkers, and the promise of liquid tumor profiling. In this review, we will discuss these recent scientific advances to scale up these approaches and endeavors to overcome clinical barriers for prostate cancer precision medicine.
Collapse
Affiliation(s)
- Adil Malik
- School of Biomedical Sciences, Queensland University of Technology, Institute of Health and Biomedical Innovation, Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre–Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Srilakshmi Srinivasan
- School of Biomedical Sciences, Queensland University of Technology, Institute of Health and Biomedical Innovation, Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre–Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Queensland University of Technology, Institute of Health and Biomedical Innovation, Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre–Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| |
Collapse
|
23
|
Prostatic carcinoma with neuroendocrine differentiation harboring the EWSR1-FEV fusion transcript in a man with the WRN G327X germline mutation: A new variant of prostatic carcinoma or a member of the Ewing sarcoma family of tumors? Pathol Res Pract 2019; 216:152758. [PMID: 31831298 DOI: 10.1016/j.prp.2019.152758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 01/23/2023]
Abstract
Since the discovery of the TMPRSS2-ERG fusion transcript in prostatic carcinoma (PCa) more than ten years ago, a long list of recurrent genomic rearrangements involving other transcription factors of the ETS family has been described. Fusions of ETS with the EWSR1 partner gene define many members of the Ewing family of tumors, including primitive neuroectodermal tumor (PNET). Although the expression of EWSR1 appears to be necessary for the oncogenic effects of ETS factors, the EWSR1-ETS rearrangement has never been reported in PCa. Herein, we discuss the pathologic diagnosis of a prostatic tumor in a 44 year-old man, recently treated with finasteride, with the EWSR1-FEV fusion (exon 7: exon 2, join in-frame) discovered by RNA-sequencing and fluorescence in situ hybridization. The tumor was morphologically and immunophenotypically equivocal for a Ewing sarcoma/PNET, and most consistent with a PCa with neuroendocrine differentiation. The patient's family history of PCa led to germline mutation testing by next-generation sequencing showing heterozygosity for the WRNG327X mutation. The WRN protein along with ATM, BRCA1, BRCA2, and RAD51 among others, comprise a DNA repair system by homologous recombination, and its alterations are associated with forms of hereditary PCa. We dispute whether the detection of EWSR1-FEV mandates one to diagnose the patient's tumor as a member of the Ewing sarcoma family.
Collapse
|
24
|
Alvarez CS, Virani S, Meza R, Rozek LS, Sriplung H, Mondul AM. Current and Future Burden of Prostate Cancer in Songkhla, Thailand: Analysis of Incidence and Mortality Trends From 1990 to 2030. J Glob Oncol 2019; 4:1-11. [PMID: 30241231 PMCID: PMC6223432 DOI: 10.1200/jgo.17.00128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Prostate cancer is the second most common malignancy among men worldwide, and it poses a significant public health burden that has traditionally been limited mostly to developed countries. However, the burden of the disease is expected to increase, affecting developing countries, including Thailand. We undertook an analysis to investigate current and future trends of prostate cancer in the province of Songkhla, Thailand, using data from the Songkhla Cancer Registry from 1990 to 2013. METHODS Joinpoint regression analysis was used to examine trends in age-adjusted incidence and mortality rates of prostate cancer and provide estimated annual percent change (EAPC) with 95% CIs. Age-period-cohort (APC) models were used to assess the effect of age, calendar year, and birth cohort on incidence and mortality rates. Three different methods (Joinpoint, Nordpred, and APC) were used to project trends from 2013 to 2030. RESULTS Eight hundred fifty-five cases of prostate cancer were diagnosed from 1990 to 2013 in Songkhla, Thailand. The incidence rates of prostate cancer significantly increased since 1990 at an EAPC of 4.8% (95% CI, 3.6% to 5.9%). Similarly, mortality rates increased at an EAPC of 5.3% (95% CI, 3.4% to 7.2%). The APC models suggest that birth cohort is the most important factor driving the increased incidence and mortality rates of prostate cancer. Future incidence and mortality of prostate cancer are projected to continue to increase, doubling the rates observed in 2013 by 2030. CONCLUSION It is critical to allocate resources to provide care for the men who will be affected by this increase in prostate cancer incidence in Songkhla, Thailand, and to design context-appropriate interventions to prevent its increasing burden.
Collapse
Affiliation(s)
- Christian S Alvarez
- Christian S. Alvarez, Shama Virani, Rafael Meza, Laura S. Rozek, and Alison M. Mondul, University of Michigan School of Public Health, Ann Arbor, MI; and Shama Virani and Hutcha Sriplung, Prince of Songkla University, Songkhla, Thailand
| | - Shama Virani
- Christian S. Alvarez, Shama Virani, Rafael Meza, Laura S. Rozek, and Alison M. Mondul, University of Michigan School of Public Health, Ann Arbor, MI; and Shama Virani and Hutcha Sriplung, Prince of Songkla University, Songkhla, Thailand
| | - Rafael Meza
- Christian S. Alvarez, Shama Virani, Rafael Meza, Laura S. Rozek, and Alison M. Mondul, University of Michigan School of Public Health, Ann Arbor, MI; and Shama Virani and Hutcha Sriplung, Prince of Songkla University, Songkhla, Thailand
| | - Laura S Rozek
- Christian S. Alvarez, Shama Virani, Rafael Meza, Laura S. Rozek, and Alison M. Mondul, University of Michigan School of Public Health, Ann Arbor, MI; and Shama Virani and Hutcha Sriplung, Prince of Songkla University, Songkhla, Thailand
| | - Hutcha Sriplung
- Christian S. Alvarez, Shama Virani, Rafael Meza, Laura S. Rozek, and Alison M. Mondul, University of Michigan School of Public Health, Ann Arbor, MI; and Shama Virani and Hutcha Sriplung, Prince of Songkla University, Songkhla, Thailand
| | - Alison M Mondul
- Christian S. Alvarez, Shama Virani, Rafael Meza, Laura S. Rozek, and Alison M. Mondul, University of Michigan School of Public Health, Ann Arbor, MI; and Shama Virani and Hutcha Sriplung, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
25
|
Elliott B, Millena AC, Matyunina L, Zhang M, Zou J, Wang G, Zhang Q, Bowen N, Eaton V, Webb G, Thompson S, McDonald J, Khan S. Essential role of JunD in cell proliferation is mediated via MYC signaling in prostate cancer cells. Cancer Lett 2019; 448:155-167. [PMID: 30763715 PMCID: PMC6414252 DOI: 10.1016/j.canlet.2019.02.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/12/2022]
Abstract
JunD, a member of the AP-1 family, is essential for cell proliferation in prostate cancer (PCa) cells. We recently demonstrated that JunD knock-down (KD) in PCa cells results in cell cycle arrest in G1-phase concomitant with a decrease in cyclin D1, Ki67, and c-MYC, but an increase in p21 levels. Furthermore, the over-expression of JunD significantly increased proliferation suggesting JunD regulation of genes required for cell cycle progression. Here, employing gene expression profiling, quantitative proteomics, and validation approaches, we demonstrate that JunD KD is associated with distinct gene and protein expression patterns. Comparative integrative analysis by Ingenuity Pathway Analysis (IPA) identified 1) cell cycle control/regulation as the top canonical pathway whose members exhibited a significant decrease in their expression following JunD KD including PRDX3, PEA15, KIF2C, and CDK2, and 2) JunD dependent genes are associated with cell proliferation, with MYC as the critical downstream regulator. Conversely, JunD over-expression induced the expression of the above genes including c-MYC. We conclude that JunD is a crucial regulator of cell cycle progression and inhibiting its target genes may be an effective approach to block prostate carcinogenesis.
Collapse
Affiliation(s)
- Bethtrice Elliott
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Ana Cecilia Millena
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Lilya Matyunina
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA, 30309, USA
| | - Mengnan Zhang
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA, 30309, USA
| | - Jin Zou
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Guangdi Wang
- Department of Chemistry, RCMI Cancer Research Center, Xavier University, 1 Drexel Drive, New Orleans, LA, 70125, USA
| | - Qiang Zhang
- Department of Chemistry, RCMI Cancer Research Center, Xavier University, 1 Drexel Drive, New Orleans, LA, 70125, USA
| | - Nathan Bowen
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Vanessa Eaton
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Gabrielle Webb
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Shadyra Thompson
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - John McDonald
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA, 30309, USA
| | - Shafiq Khan
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA.
| |
Collapse
|
26
|
Segalés L, Juanpere N, Lorenzo M, Albero-González R, Fumadó L, Cecchini L, Bellmunt J, Lloreta-Trull J, Hernández-Llodrà S. Strong cytoplasmic ETV1 expression has a negative impact on prostate cancer outcome. Virchows Arch 2019; 475:457-466. [DOI: 10.1007/s00428-019-02573-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/14/2019] [Accepted: 04/04/2019] [Indexed: 01/28/2023]
|
27
|
Wang H, Zhang H, Zeng J, Tan Y. ceRNA network analysis reveals prognostic markers for glioblastoma. Oncol Lett 2019; 17:5545-5557. [PMID: 31186776 PMCID: PMC6507312 DOI: 10.3892/ol.2019.10275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 12/13/2018] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma (GBM) is a common aggressive cancer that originates in the brain, which has a poor prognosis. It is therefore crucial to understand its underlying genetic mechanisms in order to develop novel therapies. The present study aimed to identify some prognostic markers and candidate therapeutic targets for GBM. To do so, RNA expression levels in tumor and normal tissues were compared by microarray analysis. The differential expression of RNAs in normal and cancer tissues was analyzed, and a competing endogenous RNA (ceRNA) network was constructed for pathway analysis. The results revealed that RNA expression patterns were considerably different between normal and tumor samples. A ceRNA network was therefore constructed with the differentially expressed RNAs. ETS variant 5 (ETV5), myocyte enhancer factor 2C and ETS transcription factor (ELK4) were considerably enriched in the significant pathway of ‘transcriptional misregulation in cancer’. In addition, prognostic analysis demonstrated that ETV5 and ELK4 expression levels were associated with the survival time of patients with GBM. These results suggested that ELK4 and ETV5 may be prognostic markers for GBM, and that their microRNAs may be candidate therapeutic targets.
Collapse
Affiliation(s)
- Hao Wang
- Department of Neurosurgery, Luhe Hospital, Capital Medical University, Tongzhou, Beijing 101149, P.R. China
| | - Heying Zhang
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Juan Zeng
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yonggang Tan
- Department of Oncology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
28
|
Evaluating gene fusions in solid tumors – Clinical experience using an RNA based 53 gene next-generation sequencing panel. Cancer Genet 2019; 233-234:32-42. [DOI: 10.1016/j.cancergen.2019.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 01/20/2023]
|
29
|
Verma S, Shukla S, Pandey M, MacLennan GT, Gupta S. Differentially Expressed Genes and Molecular Pathways in an Autochthonous Mouse Prostate Cancer Model. Front Genet 2019; 10:235. [PMID: 30972102 PMCID: PMC6445055 DOI: 10.3389/fgene.2019.00235] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/04/2019] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer remains a major public health problem and the second leading cause of cancer-related deaths in men in the United States. The present study aims to understand the molecular pathway(s) of prostate cancer which is essential for early detection and treatment. Dorsolateral prostate from 20 week transgenic adenocarcinoma of the mouse prostate (TRAMP) mice, which spontaneously develops prostate cancer and recapitulates human disease and age-matched non-transgenic littermates were utilized for microarray analysis. Mouse genome network and pathway analyses were mapped to the human genome using the Ingenuity Pathway Analysis (IPA) database for annotation, visualization, and integrated discovery. In total, 136 differentially expressed genes, including 32 downregulated genes and 104 upregulated genes were identified in the dorsolateral prostate of TRAMP, compared to non-transgenic mice. A subset of differentially expressed genes were validated by qRT-PCR. Alignment with human genome database identified 18 different classes of proteins, among these, 36% were connected to the nucleic acid binding, including ribosomal proteins, which play important role in protein synthesis-the most enriched pathway in the development of prostate cancer. Furthermore, the results suggest deregulation of signaling molecules (9%) and enzyme modulators (8%) affect various pathways. An imbalance in other protein classes, including transporter proteins (7%), hydrolases (6%), oxidoreductases, and cytoskeleton proteins (5%), contribute to cancer progression. Our study evaluated the underlying pathways and its connection to human prostate cancer, which may further help assess the risk of disease development and progression and identify potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Shiv Verma
- Department of Urology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sanjeev Shukla
- Department of Urology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Mitali Pandey
- Department of Urology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Vancouver Prostate Center, Vancouver, BC, Canada
| | - Gregory T MacLennan
- Department of Urology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Sanjay Gupta
- Department of Urology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
- Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH, United States
| |
Collapse
|
30
|
Eid W, Abdel-Rehim W. Genome-wide analysis of ETV1 targets: Insights into the role of ETV1 in tumor progression. J Cell Biochem 2019; 120:8983-8991. [PMID: 30629294 DOI: 10.1002/jcb.28169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/08/2018] [Indexed: 12/15/2022]
Abstract
ETS variant 1 (ETV1) is a key player in metastatic progression in several types of human cancers, yet the direct target genes of ETV1 and the mechanisms by which ETV1 exerts its deleterious function remain largely elusive. Here, we performed large-scale mapping and analysis of target loci of ETV1 in the prostate cancer cells LNCaP using the DNA adenine methyltransferase identification technique, we identified close to 800 direct targets for ETV1. Expression analysis using quantitative reverse transcription polymerase chain reaction confirmed a positive regulation by ETV1 in most of the genes examined. Furthermore, gene and pathway analysis unraveled new signaling pathways and biological networks that interact with ETV1. Our findings cast light on genes and networks regulated by ETV1, it also opens new fronts for studying the role of ETV1 and its target genes in tumorigenesis.
Collapse
Affiliation(s)
- Wassim Eid
- Department of Biochemistry, Medical Research Institute, University of Alexandria, Alexandria, Egypt
| | - Wafaa Abdel-Rehim
- Department of Biochemistry, Medical Research Institute, University of Alexandria, Alexandria, Egypt
| |
Collapse
|
31
|
Abstract
Prostatic adenocarcinoma (PCa) remains a significant health concern. Although localized PCa can be effectively treated, disseminated disease remains uniformly fatal. PCa is reliant on androgen receptor (AR); as such, first-line therapy for metastatic PCa entails suppression of AR signaling. Although initially effective, recurrent tumors reactivate AR function, leading to a lethal stage of disease termed castration-resistant PCa (CRPC). Recent findings implicate AR signaling in control of DNA repair and show that alterations in DNA damage repair pathways are strongly associated with disease progression and poor outcome. This review will address the DNA repair alterations observed in the clinical setting, explore the anticipated molecular and cellular consequence of DNA repair dysfunction, and consider clinical strategies for targeting tumors with altered DNA repair.
Collapse
Affiliation(s)
- Matthew J Schiewer
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania,19107.,The Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Karen E Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania,19107.,Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107.,Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107.,Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107.,The Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| |
Collapse
|
32
|
Characterize the difference between TMPRSS2-ERG and non-TMPRSS2-ERG fusion patients by clinical and biological characteristics in prostate cancer. Gene 2018; 679:186-194. [PMID: 30195632 DOI: 10.1016/j.gene.2018.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/10/2018] [Accepted: 09/05/2018] [Indexed: 11/23/2022]
Abstract
The TMPRSS2-ERG gene fusion were frequently found in prostate cancer, and thought to play some fundamental mechanisms for the development of prostate cancer. However, until now, the clinical and prognostic significance of TMPRSS2-ERG gene fusion was not fully understood. In this study, based on the 281 prostate cancers that constructed from a historical watchful waiting cohort, the statistically significant associations between TMPRSS2-ERG gene fusion and clinicopathologic characteristics were identified. In addition, the Elastic Net algorithm was used to predict the patients with TMPRSS2-ERG fusion status, and good predictive results were obtained, indicating that this algorithm was suitable to this prediction problem. The differential gene network was constructed from the network, and the KEGG enrichment analysis demonstrated that the module genes were significantly enriched in several important pathways.
Collapse
|
33
|
Systematic analysis reveals molecular characteristics of ERG-negative prostate cancer. Sci Rep 2018; 8:12868. [PMID: 30150711 PMCID: PMC6110738 DOI: 10.1038/s41598-018-30325-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 07/27/2018] [Indexed: 01/18/2023] Open
Abstract
The TMPRSS2:ERG gene fusion is the most prevalent early driver gene activation in prostate cancers of European ancestry, while the fusion frequency is much lower in Africans and Asians. The genomic characteristics and mechanisms for patients lacking ERG fusion are still unclear. In this study, we systematically compared the characteristics of gene fusions, somatic mutations, copy number alterations and gene expression signatures between 201 ERG fusion positive and 296 ERG fusion negative prostate cancer samples. Both common and group-specific genomic alterations were observed, suggesting shared and different mechanisms of carcinogenesis in prostate cancer samples with or without ERG fusion. The genomic alteration patterns detected in ERG-negative group showed similarities with 77.5% of tumor samples of African American patients. These results emphasize that genomic and gene expression features of the ERG-negative group may provide a reference for populations with lower ERG fusion frequency. While the overall expression patterns were comparable between ERG-negative and ERG-positive tumors, we found that genomic alterations could affect the same pathway through distinct genes in the same pathway in both groups of tumor types. Altogether, the genomic and molecular characteristics revealed in our study may provide new opportunities for molecular stratification of ERG-negative prostate cancers.
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW This review will examine the taxonomy of PCa subclasses across disease states, explore the relationship among specific alterations, and highlight current clinical relevance. RECENT FINDINGS Prostate cancer (PCa) is driven by multiple genomic alterations, with distinct patterns and clinical implications. Alterations occurring early in the timeline of the disease define core subtypes of localized, treatment-naive PCa. With time, an increase in number and severity of genomic alterations adds molecular complexity and is associated with progression to metastasis. These later events are not random and are influenced by the underlying subclasses. All the subclasses of localized disease initially respond to androgen deprivation therapy (ADT), but with progression to castrate-resistant PCa (CRPC), mechanisms of resistance against ADT shift the molecular landscape. In CRPC, resistance mechanisms largely define the biology and sub-classification of these cancers, while clinical relevance and opportunities for precision therapy are still being defined.
Collapse
Affiliation(s)
- Kaveri Arora
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, BRB 1452, 413 East 69th Street, New York, NY, 10021, USA.,Department of Urology, Weill Cornell Medicine, New York, NY, USA
| | - Christopher E Barbieri
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, BRB 1452, 413 East 69th Street, New York, NY, 10021, USA. .,Department of Urology, Weill Cornell Medicine, New York, NY, USA. .,Englander Institute for Precision Medicine of Weill Cornell Medicine and NewYork-Presbyterian Hospital, New York, NY, USA.
| |
Collapse
|
35
|
Prostate-Derived Ets Factor (PDEF) Inhibits Metastasis by Inducing Epithelial/Luminal Phenotype in Prostate Cancer Cells. Mol Cancer Res 2018; 16:1430-1440. [DOI: 10.1158/1541-7786.mcr-18-0010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/15/2018] [Accepted: 05/15/2018] [Indexed: 11/16/2022]
|
36
|
Nevedomskaya E, Baumgart SJ, Haendler B. Recent Advances in Prostate Cancer Treatment and Drug Discovery. Int J Mol Sci 2018; 19:ijms19051359. [PMID: 29734647 PMCID: PMC5983695 DOI: 10.3390/ijms19051359] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 02/08/2023] Open
Abstract
Novel drugs, drug sequences and combinations have improved the outcome of prostate cancer in recent years. The latest approvals include abiraterone acetate, enzalutamide and apalutamide which target androgen receptor (AR) signaling, radium-223 dichloride for reduction of bone metastases, sipuleucel-T immunotherapy and taxane-based chemotherapy. Adding abiraterone acetate to androgen deprivation therapy (ADT) in order to achieve complete androgen blockade has proven highly beneficial for treatment of locally advanced prostate cancer and metastatic hormone-sensitive prostate cancer (mHSPC). Also, ADT together with docetaxel treatment showed significant benefit in mHSPC. Ongoing clinical trials for different subgroups of prostate cancer patients include the evaluation of the second-generation AR antagonists enzalutamide, apalutamide and darolutamide, of inhibitors of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) pathway, of inhibitors of DNA damage response, of targeted alpha therapy and of prostate-specific membrane antigen (PSMA) targeting approaches. Advanced clinical studies with immune checkpoint inhibitors have shown limited benefits in prostate cancer and more trials are needed to demonstrate efficacy. The identification of improved, personalized treatments will be much supported by the major progress recently made in the molecular characterization of early- and late-stage prostate cancer using “omics” technologies. This has already led to novel classifications of prostate tumors based on gene expression profiles and mutation status, and should greatly help in the choice of novel targeted therapies best tailored to the needs of patients.
Collapse
Affiliation(s)
- Ekaterina Nevedomskaya
- Therapeutic Research Groups, Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany.
| | - Simon J Baumgart
- Therapeutic Research Groups, Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany.
| | - Bernard Haendler
- Therapeutic Research Groups, Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany.
| |
Collapse
|
37
|
Böttcher-Friebertshäuser E, Garten W, Klenk HD. Membrane-Anchored Serine Proteases: Host Cell Factors in Proteolytic Activation of Viral Glycoproteins. ACTIVATION OF VIRUSES BY HOST PROTEASES 2018. [PMCID: PMC7122464 DOI: 10.1007/978-3-319-75474-1_8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over one third of all known proteolytic enzymes are serine proteases. Among these, the trypsin-like serine proteases comprise one of the best characterized subfamilies due to their essential roles in blood coagulation, food digestion, fibrinolysis, or immunity. Trypsin-like serine proteases possess primary substrate specificity for basic amino acids. Most of the well-characterized trypsin-like proteases such as trypsin, plasmin, or urokinase are soluble proteases that are secreted into the extracellular environment. At the turn of the millennium, a number of novel trypsin-like serine proteases have been identified that are anchored in the cell membrane, either by a transmembrane domain at the N- or C-terminus or via a glycosylphosphatidylinositol (GPI) linkage. Meanwhile more than 20 membrane-anchored serine proteases (MASPs) have been identified in human and mouse, and some of them have emerged as key regulators of mammalian development and homeostasis. Thus, the MASP corin and TMPRSS6/matriptase-2 have been demonstrated to be the activators of the atrial natriuretic peptide (ANP) and key regulator of hepcidin expression, respectively. Furthermore, MASPs have been recognized as host cell factors activating respiratory viruses including influenza virus as well as severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) coronaviruses. In particular, transmembrane protease serine S1 member 2 (TMPRSS2) has been shown to be essential for proteolytic activation and consequently spread and pathogenesis of a number of influenza A viruses in mice and as a factor associated with severe influenza virus infection in humans. This review gives an overview on the physiological functions of the fascinating and rapidly evolving group of MASPs and a summary of the current knowledge on their role in proteolytic activation of viral fusion proteins.
Collapse
Affiliation(s)
| | - Wolfgang Garten
- 0000 0004 1936 9756grid.10253.35Institut für Virologie, Philipps Universität, Marburg, Germany
| | - Hans Dieter Klenk
- 0000 0004 1936 9756grid.10253.35Institut für Virologie, Philipps-Universität, Marburg, Germany
| |
Collapse
|
38
|
The molecular biology of prostate cancer: current understanding and clinical implications. Prostate Cancer Prostatic Dis 2017; 21:22-36. [PMID: 29282359 DOI: 10.1038/s41391-017-0023-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/11/2017] [Accepted: 11/02/2017] [Indexed: 01/07/2023]
Abstract
BACKGROUND With continuous progress over the past few decades in understanding diagnosis, treatment, and genetics, much has been learned about the prostate cancer-diagnosed genome. METHODS A comprehensive MEDLINE® and Google scholar literature search was conducted using keyword variations relating to the genetics of prostate cancer such as chromosomal alterations, androgen receptor, castration-resistant, inheritance, polymorphisms, oncogenes, metastasis, biomarkers, and immunotherapy. RESULTS Traditionally, androgen receptors (AR) have been the focus of research. Recently, identification of recurrent chromosomal alterations that lead to either multiplication of regions (gain-of-function) or deletion of regions (loss-of-function) has opened the door to greater genetic accessibility. These chromosomal aberrations lead to variation in copy number and gene expression. Some of these chromosomal alterations are inherited, while others undergo somatic mutations during disease progression. Inherited gene mutations that make one susceptible to prostate cancer have been identified with familial-linked studies. Somatic genes that progress tumorigenesis have also been identified. Research on the molecular biology of prostate cancer has characterized these genes into tumor suppressor genes or oncogenes. Additionally, genome-wide assay studies have identified many high-risk single-nucleotide polymorphisms recurrent throughout the prostate cancer-diagnosed genome. Castration-resistant prostate cancer is the most aggressive form of prostate cancer, and its research has elucidated many types of mutations associated with AR itself, including enhanced expression and amplification, point mutations, and alternative splicing. Understanding the molecular biology of prostate cancer has permitted more accurate identification using advanced biomarkers and therapy for aggressive forms using immunotherapy. CONCLUSIONS An age-related disease, prostate cancer commands profound attention. With increasing life expectancy and the continuous pursuit of it, prostate cancer is a powerful obstacle best defeated using targeted therapies specifically designed for the unique molecular profile of the malignancy.
Collapse
|
39
|
Gu JL, Chukhman M, Lu Y, Liu C, Liu SY, Lu H. RNA-seq Based Transcription Characterization of Fusion Breakpoints as a Potential Estimator for Its Oncogenic Potential. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9829175. [PMID: 29181411 PMCID: PMC5664375 DOI: 10.1155/2017/9829175] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 08/23/2017] [Indexed: 12/20/2022]
Abstract
Based on high-throughput sequencing technology, the detection of gene fusions is no longer a big challenge but estimating the oncogenic potential of fusion genes remains challenging. Recent studies successfully applied machine learning methods and gene structural and functional features of fusion mutation to predict their oncogenic potentials. However, the transcription characterizations features of fusion genes have not yet been studied. In this study, based on the clonal evolution theory, we hypothesized that a fusion gene is more likely to be an oncogenic genomic alteration, if the neoplastic cells harboring this fusion mutation have larger clonal size than other neoplastic cells in a tumor. We proposed a novel method, called iFCR (internal Fusion Clone Ratio), given an estimation of oncogenic potential for fusion mutations. We have evaluated the iFCR method in three public cancer transcriptome sequencing datasets; the results demonstrated that the fusion mutations occurring in tumor samples have higher internal fusion clone ratio than normal samples. And the most frequent prostate cancer fusion mutation, TMPRSS2-ERG, appears to have a remarkably higher iFCR value in all three independent patients. The preliminary results suggest that the internal fusion clone ratio might potentially advantage current fusion mutation oncogenic potential prediction methods.
Collapse
Affiliation(s)
- Jian-lei Gu
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China
- Department of Bioinformatics, SJTU-Yale Joint Center for Biostatistics, Shanghai Jiao Tong University, Shanghai 200240, China
- Key Laboratory of Molecular Embryology, Ministry of Health and Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200040, China
| | - Morris Chukhman
- Department of Bioengineering, Bioinformatics Program, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Yao Lu
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China
- Department of Bioinformatics, SJTU-Yale Joint Center for Biostatistics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cong Liu
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China
- Department of Bioinformatics, SJTU-Yale Joint Center for Biostatistics, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Bioengineering, Bioinformatics Program, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Shi-yi Liu
- Department of Bioinformatics, SJTU-Yale Joint Center for Biostatistics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hui Lu
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China
- Department of Bioinformatics, SJTU-Yale Joint Center for Biostatistics, Shanghai Jiao Tong University, Shanghai 200240, China
- Key Laboratory of Molecular Embryology, Ministry of Health and Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200040, China
- Department of Bioengineering, Bioinformatics Program, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
40
|
Wang Z, Wang Y, Zhang J, Hu Q, Zhi F, Zhang S, Mao D, Zhang Y, Liang H. Significance of the TMPRSS2:ERG gene fusion in prostate cancer. Mol Med Rep 2017; 16:5450-5458. [PMID: 28849022 PMCID: PMC5647090 DOI: 10.3892/mmr.2017.7281] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 06/28/2017] [Indexed: 12/31/2022] Open
Abstract
The transmembrane protease serine 2:v-ets erythroblastosis virus E26 oncogene homolog (TMPRSS2:ERG) gene fusion is common in prostate cancer, while its functional role is not fully understood. The present study aimed to investigate the significance of the TMPRSS2:ERG gene fusion in human prostate cancers using bioinformatics tools. Comprehensive alteration analysis of TMPRSS2 and ERG in 148 different human cancer studies was performed by cBioPortal, and the mRNA expression level of the ERG gene was evaluated using Oncomine analysis. Furthermore, lentiviral short hairpin (sh)RNA-mediated knockdown of TMPRSS2:ERG was performed to study the impact of ERG silencing on cell proliferation and cell cycle distribution in prostate cancer cells. The results demonstrated that the TMPRSS2 and ERG genes were mostly altered in prostate cancer, and the most frequent alteration was gene fusion. Oncomine analysis demonstrated that the ERG gene was significantly upregulated in prostate clinical samples compared with the normal prostate gland in four independent datasets, and a positive association was observed between potassium inwardly-rectifying channel subfamily J member 15, down syndrome critical region gene 4, potassium inwardly-rectifying channel subfamily J member 6 and ERG gene expression. There were 272 mutations of the ERG gene identified in the cBioPortal database; among the mutations, 2 missense mutations (R367C and P401H) were regarded as functional mutations (functional impact score >1.938). Furthermore, the present study successfully knocked down ERG gene expression through a lentiviral-mediated gene silencing approach in VCaP prostate cancer cells. The ERG mRNA and protein expression levels were both suppressed significantly, and a cell-cycle arrest at G0/G1 phase was observed after ERG gene silencing. In conclusion, these bioinformatics analyses provide novel insights for TMPRSS2:ERG fusion gene study in prostate cancer. Target inhibition of ERG expression could significantly cause cell growth arrest in prostate cancer cells, which could be a potentially valuable target for prostate cancer treatment. However, the precise mechanism of these results remains unclear; therefore, further studies are required.
Collapse
Affiliation(s)
- Zhu Wang
- Department of Urology, People's Hospital of Longhua New District of Shenzhen and Affiliated Shenzhen Longhua Hospital of Southern Medical University, Shenzhen, Guangdong 518109, P.R. China
| | - Yuliang Wang
- Department of Urology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Jianwen Zhang
- Department of Urology, People's Hospital of Longhua New District of Shenzhen and Affiliated Shenzhen Longhua Hospital of Southern Medical University, Shenzhen, Guangdong 518109, P.R. China
| | - Qiyi Hu
- Department of Urology, People's Hospital of Longhua New District of Shenzhen and Affiliated Shenzhen Longhua Hospital of Southern Medical University, Shenzhen, Guangdong 518109, P.R. China
| | - Fan Zhi
- Department of Urology, People's Hospital of Longhua New District of Shenzhen and Affiliated Shenzhen Longhua Hospital of Southern Medical University, Shenzhen, Guangdong 518109, P.R. China
| | - Shengping Zhang
- Department of Urology, People's Hospital of Longhua New District of Shenzhen and Affiliated Shenzhen Longhua Hospital of Southern Medical University, Shenzhen, Guangdong 518109, P.R. China
| | - Dengqi Mao
- Department of Urology, People's Hospital of Longhua New District of Shenzhen and Affiliated Shenzhen Longhua Hospital of Southern Medical University, Shenzhen, Guangdong 518109, P.R. China
| | - Ying Zhang
- Department of Urology, People's Hospital of Longhua New District of Shenzhen and Affiliated Shenzhen Longhua Hospital of Southern Medical University, Shenzhen, Guangdong 518109, P.R. China
| | - Hui Liang
- Department of Urology, People's Hospital of Longhua New District of Shenzhen and Affiliated Shenzhen Longhua Hospital of Southern Medical University, Shenzhen, Guangdong 518109, P.R. China
| |
Collapse
|
41
|
Kakehi Y, Sugimoto M, Taoka R. Evidenced-based clinical practice guideline for prostate cancer (summary: Japanese Urological Association, 2016 edition). Int J Urol 2017; 24:648-666. [PMID: 28667698 DOI: 10.1111/iju.13380] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/11/2017] [Indexed: 12/18/2022]
Abstract
These guidelines cover a wide range of topics from prostate cancer epidemiology to palliative care. Questions arising in daily clinical practice have been extracted and formulated as clinical questions. In the 4 years since the previous edition, there have been major changes - for example, robot-assisted prostatectomy has rapidly come into widespread use, and new hormones and anticancer drugs have been developed for castration-resistant prostate cancer. In response to these developments, the number of fields included in this guideline was increased from 11 in the 2012 edition to 16, and the number of clinical questions was increased from 63 to 70. The number of papers identified in searches of the existing literature increased from 4662 in the first edition, published in 2006, to 10 490 in the 2012 edition. The number of references has reached 29 448 just during this review period, indicating the exponential increase in research on the topic of prostate cancer. Clinical answers have been prepared based on the latest evidence. Recommendation grades for the clinical answers were determined by radiologists, pathologists, and other specialists in addition to urologists in order to reflect the recent advances and diversity of prostate cancer treatment. Here, we present a short English version of the original guideline, and overview its key clinical issues.
Collapse
Affiliation(s)
- Yoshiyuki Kakehi
- Department of Urology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Mikio Sugimoto
- Department of Urology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Rikiya Taoka
- Department of Urology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | | |
Collapse
|
42
|
Abstract
Background Genome rearrangements are critical oncogenic driver events in many malignancies. However, the identification and resolution of the structure of cancer genomic rearrangements remain challenging even with whole genome sequencing. Methods To identify oncogenic genomic rearrangements and resolve their structure, we analyzed linked read sequencing. This approach relies on a microfluidic droplet technology to produce libraries derived from single, high molecular weight DNA molecules, 50 kb in size or greater. After sequencing, the barcoded sequence reads provide long range genomic information, identify individual high molecular weight DNA molecules, determine the haplotype context of genetic variants that occur across contiguous megabase-length segments of the genome and delineate the structure of complex rearrangements. We applied linked read sequencing of whole genomes to the analysis of a set of synchronous metastatic diffuse gastric cancers that occurred in the same individual. Results When comparing metastatic sites, our analysis implicated a complex somatic rearrangement that was present in the metastatic tumor. The oncogenic event associated with the identified complex rearrangement resulted in an amplification of the known cancer driver gene FGFR2. With further investigation using these linked read data, the FGFR2 copy number alteration was determined to be a deletion-inversion motif that underwent tandem duplication, with unique breakpoints in each metastasis. Using a three-dimensional organoid tissue model, we functionally validated the metastatic potential of an FGFR2 amplification in gastric cancer. Conclusions Our study demonstrates that linked read sequencing is useful in characterizing oncogenic rearrangements in cancer metastasis. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0447-8) contains supplementary material, which is available to authorized users.
Collapse
|
43
|
Abstract
Prostate cancer is the second most common cause of cancer mortality among men in the United States. While many prostate cancers are indolent, an important subset of patients experiences disease recurrence after conventional therapy and progresses to castration-resistant prostate cancer (CRPC), which is currently incurable. Thus, there is a critical need to identify biomarkers that will distinguish indolent from aggressive disease, as well as novel therapeutic targets for the prevention or treatment of CRPC. In recent years, long noncoding RNAs (lncRNAs) have emerged as an important class of biological molecules. LncRNAs are polyadenylated RNA species that share many similarities with protein-coding genes despite the fact that they are noncoding (not translated into proteins). They are usually transcribed by RNA polymerase II and exhibit the same epigenetic signatures as protein-coding genes. LncRNAs have also been implicated in the development and progression of variety of cancers, including prostate cancer. While a large number of lncRNAs exhibit tissue- and cancer-specific expression, their utility as diagnostic and prognostic biomarkers is just starting to be explored. In this review, we highlight recent findings on the functional role and molecular mechanisms of lncRNAs in the progression of prostate cancer and evaluate their use as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Bhavna Malik
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Felix Y Feng
- Department of Radiation Oncology, Urology, and Medicine, University of California at San Francisco; Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, USA
| |
Collapse
|
44
|
Geybels MS, McCloskey KD, Mills IG, Stanford JL. Calcium Channel Blocker Use and Risk of Prostate Cancer by TMPRSS2:ERG Gene Fusion Status. Prostate 2017; 77:282-290. [PMID: 27753122 PMCID: PMC5668682 DOI: 10.1002/pros.23267] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Calcium channel blockers (CCBs) may affect prostate cancer (PCa) growth by various mechanisms including those related to androgens. The fusion of the androgen-regulated gene TMPRSS2 and the oncogene ERG (TMPRSS2:ERG or T2E) is common in PCa, and prostate tumors that harbor the gene fusion are believed to represent a distinct disease subtype. We studied the association of CCB use with the risk of PCa, and molecular subtypes of PCa defined by T2E status. METHODS Participants were residents of King County, Washington, recruited for population-based case-control studies (1993-1996 or 2002-2005). Tumor T2E status was determined by fluorescence in situ hybridization using tumor tissue specimens from radical prostatectomy. Detailed information on use of CCBs and other variables was obtained through in-person interviews. Binomial and polytomous logistic regression were used to generate odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS The study included 1,747 PCa patients and 1,635 age-matched controls. A subset of 563 patients treated with radical prostatectomy had T2E status determined, of which 295 were T2E positive (52%). Use of CCBs (ever vs. never) was not associated with overall PCa risk. However, among European-American men, users had a reduced risk of higher-grade PCa (Gleason scores ≥7: adjusted OR = 0.64; 95% CI: 0.44-0.95). Further, use of CCBs was associated with a reduced risk of T2E positive PCa (adjusted OR = 0.38; 95% CI: 0.19-0.78), but was not associated with T2E negative PCa. CONCLUSIONS This study found suggestive evidence that use of CCBs is associated with reduced relative risks for higher Gleason score and T2E positive PCa. Future studies of PCa etiology should consider etiologic heterogeneity as PCa subtypes may develop through different causal pathways. Prostate 77:282-290, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Milan S. Geybels
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
- Corresponding authors: ;
| | - Karen D. McCloskey
- Centre for Cancer Research and Cell Biology, Queen’s University of Belfast, Belfast, UK
| | - Ian G. Mills
- Prostate Cancer Research Group, Centre for Molecular Medicine, University of Oslo and Oslo University Hospitals, Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospitals-Radium Hospital, Montebello, Oslo, Norway
| | - Janet L. Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Epidemiology, University of Washington, School of Public Health, Seattle, Washington
- Corresponding authors: ;
| |
Collapse
|
45
|
Syvälä H, Pennanen P, Bläuer M, Tammela TL, Murtola TJ. Additive inhibitory effects of simvastatin and enzalutamide on androgen-sensitive LNCaP and VCaP prostate cancer cells. Biochem Biophys Res Commun 2016; 481:46-50. [DOI: 10.1016/j.bbrc.2016.11.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 11/05/2016] [Indexed: 01/09/2023]
|
46
|
Kim H, Datta A, Talwar S, Saleem SN, Mondal D, Abdel-Mageed AB. Estradiol-ERβ2 signaling axis confers growth and migration of CRPC cells through TMPRSS2-ETV5 gene fusion. Oncotarget 2016; 8:62820-62833. [PMID: 28968951 PMCID: PMC5609883 DOI: 10.18632/oncotarget.11355] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 07/26/2016] [Indexed: 12/12/2022] Open
Abstract
Estrogen receptor beta (ERβ) splice variants are implicated in prostate cancer (PC) progression; however their underlying mechanisms remain elusive. We report that non-canonical activation of estradiol (E2)-ERβ2 signaling axis primes growth, colony-forming ability and migration of the androgen receptor (AR)-null castration-resistant PC (CRPC) cells under androgen-deprived conditions (ADC). The non-classical E2-ERβ2 mediates phosphorylation and activation of Src-IGF-1R complex, which in turn triggers p65-dependent transcriptional upregulation of the androgen-regulated serine protease TMPRSS2:ETV5a/TMPRSS2:ETV5b gene fusions under ADC. siRNA silencing of TMPRSS2 and/or ETV5 suggests that TMPRSS2:ETV5 fusions facilitates the E2-ERβ induced growth and migration effects via NF-κB-dependent induction of cyclin D1 and MMP2 and MMP9 in PC-3 cells. Collectively, our results unravel the functional significance of oncogenic TMPRSS2:ETV5 fusions in mediating growth and migration of E2-ERβ2 signaling axis in CRPC cells. E2-ERβ2 signaling axis may have significant therapeutic and prognostic implications in patients with CRPC.
Collapse
Affiliation(s)
- Hogyoung Kim
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, 70112, USA
| | - Amrita Datta
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, 70112, USA
| | - Sudha Talwar
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, 70112, USA
| | - Sarmad N Saleem
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, 70112, USA
| | - Debasis Mondal
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, 70112, USA
| | - Asim B Abdel-Mageed
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, 70112, USA.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, 70112, USA.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, 70112, USA
| |
Collapse
|
47
|
Stelloo S, Nevedomskaya E, van der Poel HG, de Jong J, van Leenders GJLH, Jenster G, Wessels LFA, Bergman AM, Zwart W. Androgen receptor profiling predicts prostate cancer outcome. EMBO Mol Med 2016; 7:1450-64. [PMID: 26412853 PMCID: PMC4644377 DOI: 10.15252/emmm.201505424] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Prostate cancer is the second most prevalent malignancy in men. Biomarkers for outcome prediction are urgently needed, so that high-risk patients could be monitored more closely postoperatively. To identify prognostic markers and to determine causal players in prostate cancer progression, we assessed changes in chromatin state during tumor development and progression. Based on this, we assessed genomewide androgen receptor/chromatin binding and identified a distinct androgen receptor/chromatin binding profile between primary prostate cancers and tumors with an acquired resistance to therapy. These differential androgen receptor/chromatin interactions dictated expression of a distinct gene signature with strong prognostic potential. Further refinement of the signature provided us with a concise list of nine genes that hallmark prostate cancer outcome in multiple independent validation series. In this report, we identified a novel gene expression signature for prostate cancer outcome through generation of multilevel genomic data on chromatin accessibility and transcriptional regulation and integration with publically available transcriptomic and clinical datastreams. By combining existing technologies, we propose a novel pipeline for biomarker discovery that is easily implementable in other fields of oncology.
Collapse
Affiliation(s)
- Suzan Stelloo
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ekaterina Nevedomskaya
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Henk G van der Poel
- Division of Urology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeroen de Jong
- Division of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Geert J L H van Leenders
- Department of Pathology, Josephine Nefkens Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Guido Jenster
- Department of Urology, Josephine Nefkens Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andries M Bergman
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Geybels MS, Alumkal JJ, Luedeke M, Rinckleb A, Zhao S, Shui IM, Bibikova M, Klotzle B, van den Brandt PA, Ostrander EA, Fan JB, Feng Z, Maier C, Stanford JL. Epigenomic profiling of prostate cancer identifies differentially methylated genes in TMPRSS2:ERG fusion-positive versus fusion-negative tumors. Clin Epigenetics 2015; 7:128. [PMID: 26692910 PMCID: PMC4676897 DOI: 10.1186/s13148-015-0161-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/03/2015] [Indexed: 12/17/2022] Open
Abstract
Background About half of all prostate cancers harbor the TMPRSS2:ERG (T2E) gene fusion. While T2E-positive and T2E-negative tumors represent specific molecular subtypes of prostate cancer (PCa), previous studies have not yet comprehensively investigated how these tumor subtypes differ at the epigenetic level. We therefore investigated epigenome-wide DNA methylation profiles of PCa stratified by T2E status. Results The study included 496 patients with clinically localized PCa who had a radical prostatectomy as primary treatment for PCa. Fluorescence in situ hybridization (FISH) “break-apart” assays were used to determine tumor T2E-fusion status, which showed that 266 patients (53.6 %) had T2E-positive PCa. The study showed global DNA methylation differences between tumor subtypes. A large number of differentially methylated CpG sites were identified (false-discovery rate [FDR] Q-value <0.00001; n = 27,876) and DNA methylation profiles accurately distinguished between tumor T2E subgroups. A number of top-ranked differentially methylated CpGs in genes (FDR Q-values ≤1.53E−29) were identified: C3orf14, CACNA1D, GREM1, KLK10, NT5C, PDE4D, RAB40C, SEPT9, and TRIB2, several of which had a corresponding alteration in mRNA expression. These genes may have various roles in the pathogenesis of PCa, and the calcium-channel gene CACNA1D is a known ERG-target. Analysis of The Cancer Genome Atlas (TCGA) data provided confirmatory evidence for our findings. Conclusions This study identified substantial differences in DNA methylation profiles of T2E-positive and T2E-negative tumors, thereby providing further evidence that different underlying oncogenic pathways characterize these molecular subtypes. Electronic supplementary material The online version of this article (doi:10.1186/s13148-015-0161-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Milan S Geybels
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Joshi J Alumkal
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR USA
| | - Manuel Luedeke
- Institute of Human Genetics and Department of Urology, Faculty of Medicine, University of Ulm, Ulm, Germany
| | - Antje Rinckleb
- Institute of Human Genetics and Department of Urology, Faculty of Medicine, University of Ulm, Ulm, Germany
| | - Shanshan Zhao
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NC Research Triangle Park, USA
| | - Irene M Shui
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | | | | | - Piet A van den Brandt
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Elaine A Ostrander
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
| | - Jian-Bing Fan
- Illumina, Inc., San Diego, CA USA ; Present Address: AnchorDx Corp., Guangzhou, 510300 People's Republic of China
| | | | - Christiane Maier
- Institute of Human Genetics and Department of Urology, Faculty of Medicine, University of Ulm, Ulm, Germany
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA USA
| |
Collapse
|
49
|
Abstract
Drugs that target intracellular signalling pathways have markedly improved progression-free survival of patients with cancers who were previously regarded as untreatable. However, the rapid emergence of therapeutic resistance, as a result of bypass signalling or downstream mutation within kinase-mediated signalling cascades, has curtailed the benefit gained from these therapies. Such resistance mechanisms are facilitated by the linearity and redundancy of kinase signalling pathways. We argue that, in each cancer, the dysregulation of key transcriptional regulators not only defines the cancer phenotype but is essential for its development and maintenance. Furthermore, we propose that, as therapeutic targets, these transcriptional regulators are less prone to bypass by alternative mutational events or clonal heterogeneity, and therefore we must rekindle our efforts to directly target transcriptional regulation across a broad range of cancers.
Collapse
Affiliation(s)
- Thomas J Gonda
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence (PACE), 20 Cornwall Street, Woolloongabba, Queensland 4102, Australia
| | - Robert G Ramsay
- Peter MacCallum Cancer Centre and the Sir Peter MacCallum Oncology Department and the Pathology Department, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
50
|
Vlaeminck-Guillem V. When Prostate Cancer Circulates in the Bloodstream. Diagnostics (Basel) 2015; 5:428-74. [PMID: 26854164 PMCID: PMC4728468 DOI: 10.3390/diagnostics5040428] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/14/2015] [Accepted: 10/22/2015] [Indexed: 12/14/2022] Open
Abstract
Management of patients with prostate cancer is currently based on imperfect clinical, biological, radiological and pathological evaluation. Prostate cancer aggressiveness, including metastatic potential, remains difficult to accurately estimate. In an attempt to better adapt therapeutics to an individual (personalized medicine), reliable evaluation of the intrinsic molecular biology of the tumor is warranted, and particularly for all tumor sites (primary tumors and secondary sites) at any time of the disease progression. As a consequence of their natural tendency to grow (passive invasion) or as a consequence of an active blood vessel invasion by metastase-initiating cells, tumors shed various materials into the bloodstream. Major efforts have been recently made to develop powerful and accurate methods able to detect, quantify and/or analyze all these circulating tumor materials: circulating tumors cells, disseminating tumor cells, extracellular vesicles (including exosomes), nucleic acids, etc. The aim of this review is to summarize current knowledge about these circulating tumor materials and their applications in translational research.
Collapse
Affiliation(s)
- Virginie Vlaeminck-Guillem
- Cancer Research Centre of Lyon, U1052 INSERM, CNRS 5286, Léon Bérard Centre, Lyon I University, 28 rue Laennec, Lyon 69008, France.
- Medical Unit of Molecular Oncology and Transfer, Department of Biochemistry and Molecular Biology, University Hospital of Lyon-Sud, Hospices Civils of Lyon, Lyon 69008, France.
| |
Collapse
|