1
|
Cacciatore A, Shinde D, Musumeci C, Sandrini G, Guarrera L, Albino D, Civenni G, Storelli E, Mosole S, Federici E, Fusina A, Iozzo M, Rinaldi A, Pecoraro M, Geiger R, Bolis M, Catapano CV, Carbone GM. Epigenome-wide impact of MAT2A sustains the androgen-indifferent state and confers synthetic vulnerability in ERG fusion-positive prostate cancer. Nat Commun 2024; 15:6672. [PMID: 39107274 PMCID: PMC11303763 DOI: 10.1038/s41467-024-50908-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/25/2024] [Indexed: 08/09/2024] Open
Abstract
Castration-resistant prostate cancer (CRPC) is a frequently occurring disease with adverse clinical outcomes and limited therapeutic options. Here, we identify methionine adenosyltransferase 2a (MAT2A) as a critical driver of the androgen-indifferent state in ERG fusion-positive CRPC. MAT2A is upregulated in CRPC and cooperates with ERG in promoting cell plasticity, stemness and tumorigenesis. RNA, ATAC and ChIP-sequencing coupled with histone post-translational modification analysis by mass spectrometry show that MAT2A broadly impacts the transcriptional and epigenetic landscape. MAT2A enhances H3K4me2 at multiple genomic sites, promoting the expression of pro-tumorigenic non-canonical AR target genes. Genetic and pharmacological inhibition of MAT2A reverses the transcriptional and epigenetic remodeling in CRPC models and improves the response to AR and EZH2 inhibitors. These data reveal a role of MAT2A in epigenetic reprogramming and provide a proof of concept for testing MAT2A inhibitors in CRPC patients to improve clinical responses and prevent treatment resistance.
Collapse
MESH Headings
- Male
- Humans
- Transcriptional Regulator ERG/genetics
- Transcriptional Regulator ERG/metabolism
- Methionine Adenosyltransferase/genetics
- Methionine Adenosyltransferase/metabolism
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic/drug effects
- Epigenesis, Genetic/drug effects
- Animals
- Androgens/metabolism
- Epigenome
- Mice
- Histones/metabolism
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Enhancer of Zeste Homolog 2 Protein/genetics
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
Collapse
Affiliation(s)
- Alessia Cacciatore
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Dheeraj Shinde
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Carola Musumeci
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Giada Sandrini
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Bioinformatics Core Unit, 6500, Bellinzona, Switzerland
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, 20156, Milano, Italy
| | - Domenico Albino
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Gianluca Civenni
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Elisa Storelli
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Simone Mosole
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Elisa Federici
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Alessio Fusina
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Marta Iozzo
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Matteo Pecoraro
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Roger Geiger
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Marco Bolis
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
- Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, 20156, Milano, Italy
| | - Carlo V Catapano
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Giuseppina M Carbone
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland.
| |
Collapse
|
2
|
Buteyn NJ, Burke CG, Sartori VJ, Deering-Gardner E, DeBruine ZJ, Kamarudin D, Chandler DP, Monovich AC, Perez MW, Yi JS, Ries RE, Alonzo TA, Ryan RJ, Meshinchi S, Triche TJ. EZH2-driven immune evasion defines high-risk pediatric AML with t(16;21) FUS::ERG gene fusion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594150. [PMID: 38798454 PMCID: PMC11118270 DOI: 10.1101/2024.05.14.594150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Despite decades of research, acute myeloid leukemia (AML) remains a remarkably lethal malignancy. While pediatric AML (pAML) carries a more favorable prognosis than adult AML, the past 25 years of large clinical trials have produced few improvements in pAML survival. Nowhere is this more evident than in patients carrying a t(16;21)(p11;q22) translocation, which yields the FUS::ERG fusion transcript. Patients with FUS::ERG-positive AML are often primary refractory, and most responders quickly relapse. In COG clinical trials, allogeneic stem cell transplantation was of no benefit to FUS::ERG pAML patients; 100% of transplanted patients succumbed to their disease. Expression of major histocompatibility complex (MHC) class I & II and costimulatory molecules is absent at diagnosis in FUS::ERG AML, mirroring the epigenetic mechanism of post-transplant relapse seen in adult AML and its associated dismal outcomes. Here we show that this class-defining immune-repressive phenotype is driven by overexpression of the EZH2 histone lysine methyltransferase in vitro and in multiple clinical cohorts. We show that treatment with the FDA-approved EZH2 inhibitor tazemetostat along with IFN-γ reverses this phenotype, re-establishes MHC presentation, and severely impairs the viability of FUS::ERG AML cells. EZH2 inhibitors may thus provide the first targeted therapeutic option for patients with this high-risk subtype of pAML, with particular benefit as a bridge to successful allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Nathaniel J Buteyn
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI
| | - Connor G Burke
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI
| | - Vincent J Sartori
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI
| | | | - Zachary J DeBruine
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI
| | - Dahlya Kamarudin
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI
| | - Darrell P Chandler
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI
| | | | - Monika W Perez
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Joanna S Yi
- Department of Pediatrics, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, TX
| | - Rhonda E Ries
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Todd A Alonzo
- Children's Oncology Group, Monrovia, CA
- Department of Translational Genomics, University of Southern California, Los Angeles, CA
| | - Russell Jh Ryan
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Children's Oncology Group, Monrovia, CA
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Timothy J Triche
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI
- Department of Translational Genomics, University of Southern California, Los Angeles, CA
- Department of Pediatrics, College of Human Medicine, Michigan State University, East Lansing, MI
| |
Collapse
|
3
|
Wang JM, Zhang FH, Liu ZX, Tang YJ, Li JF, Xie LP. Cancer on motors: How kinesins drive prostate cancer progression? Biochem Pharmacol 2024; 224:116229. [PMID: 38643904 DOI: 10.1016/j.bcp.2024.116229] [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: 01/02/2024] [Revised: 04/02/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Prostate cancer causes numerous male deaths annually. Although great progress has been made in the diagnosis and treatment of prostate cancer during the past several decades, much about this disease remains unknown, especially its pathobiology. The kinesin superfamily is a pivotal group of motor proteins, that contains a microtubule-based motor domain and features an adenosine triphosphatase activity and motility characteristics. Large-scale sequencing analyses based on clinical samples and animal models have shown that several members of the kinesin family are dysregulated in prostate cancer. Abnormal expression of kinesins could be linked to uncontrolled cell growth, inhibited apoptosis and increased metastasis ability. Additionally, kinesins may be implicated in chemotherapy resistance and escape immunologic cytotoxicity, which creates a barrier to cancer treatment. Here we cover the recent advances in understanding how kinesins may drive prostate cancer progression and how targeting their function may be a therapeutic strategy. A better understanding of kinesins in prostate cancer tumorigenesis may be pivotal for improving disease outcomes in prostate cancer patients.
Collapse
Affiliation(s)
- Jia-Ming Wang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Feng-Hao Zhang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Zi-Xiang Liu
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, People's Republic of China
| | - Yi-Jie Tang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jiang-Feng Li
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
| | - Li-Ping Xie
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
| |
Collapse
|
4
|
Lu B, Liu Y, Yao Y, Yang T, Zhang H, Yang X, Huang R, Zhou W, Pan X, Cui X. Advances in sequencing and omics studies in prostate cancer: unveiling molecular pathogenesis and clinical applications. Front Oncol 2024; 14:1355551. [PMID: 38800374 PMCID: PMC11116611 DOI: 10.3389/fonc.2024.1355551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/16/2024] [Indexed: 05/29/2024] Open
Abstract
Background Prostate cancer (PCa) is one of the most threatening health problems for the elderly males. However, our understanding of the disease has been limited by the research technology for a long time. Recently, the maturity of sequencing technology and omics studies has been accelerating the studies of PCa, establishing themselves as an essential impetus in this field. Methods We assessed Web of Science (WoS) database for publications of sequencing and omics studies in PCa on July 3rd, 2023. Bibliometrix was used to conduct ulterior bibliometric analysis of countries/affiliations, authors, sources, publications, and keywords. Subsequently, purposeful large amounts of literature reading were proceeded to analyze research hotspots in this field. Results 3325 publications were included in the study. Research associated with sequencing and omics studies in PCa had shown an obvious increase recently. The USA and China were the most productive countries, and harbored close collaboration. CHINNAIYAN AM was identified as the most influential author, and CANCER RESEARCH exhibited huge impact in this field. Highly cited publications and their co-citation relationships were used to filtrate literatures for subsequent literature reading. Based on keyword analysis and large amounts of literature reading, 'the molecular pathogenesis of PCa' and 'the clinical application of sequencing and omics studies in PCa' were summarized as two research hotspots in the field. Conclusion Sequencing technology had a deep impact on the studies of PCa. Sequencing and omics studies in PCa helped researchers reveal the molecular pathogenesis, and provided new possibilities for the clinical practice of PCa.
Collapse
Affiliation(s)
- Bingnan Lu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifan Liu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuntao Yao
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyue Yang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyu Zhang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyue Yang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wang Zhou
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Zhang M, Ceyhan Y, Mei S, Hirz T, Sykes DB, Agoulnik IU. Regulation of EZH2 Expression by INPP4B in Normal Prostate and Primary Prostate Cancer. Cancers (Basel) 2023; 15:5418. [PMID: 38001678 PMCID: PMC10670027 DOI: 10.3390/cancers15225418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
The phosphatases INPP4B and PTEN are tumor suppressors that are lost in nearly half of advanced metastatic cancers. The loss of PTEN in prostate epithelium initially leads to an upregulation of several tumor suppressors that slow the progression of prostate cancer in mouse models. We tested whether the loss of INPP4B elicits a similar compensatory response in prostate tissue and whether this response is distinct from the one caused by the loss of PTEN. Knockdown of INPP4B but not PTEN in human prostate cancer cell lines caused a decrease in EZH2 expression. In Inpp4b-/- mouse prostate epithelium, EZH2 levels were decreased, as were methylation levels of histone H3. In contrast, Ezh2 levels were increased in the prostates of Pten-/- male mice. Contrary to PTEN, there was a positive correlation between INPP4B and EZH2 expression in normal human prostates and early-stage prostate tumors. Analysis of single-cell transcriptomic data demonstrated that a subset of EZH2-positive cells expresses INPP4B or PTEN, but rarely both, consistent with their opposing correlation with EZH2 expression. Unlike PTEN, INPP4B did not affect the levels of SMAD4 protein expression or Pml mRNA expression. Like PTEN, p53 protein expression and phosphorylation of Akt in Inpp4b-/- murine prostates were elevated. Taken together, the loss of INPP4B in the prostate leads to overlapping and distinct changes in tumor suppressor and oncogenic downstream signaling.
Collapse
Affiliation(s)
- Manqi Zhang
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC 27708, USA;
| | - Yasemin Ceyhan
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA;
| | - Shenglin Mei
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (S.M.); (T.H.); (D.B.S.)
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Taghreed Hirz
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (S.M.); (T.H.); (D.B.S.)
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - David B. Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (S.M.); (T.H.); (D.B.S.)
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Irina U. Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Biomolecular Science Institute, Florida International University, Miami, FL 33199, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
6
|
Silvestri R, Nicolì V, Gangadharannambiar P, Crea F, Bootman MD. Calcium signalling pathways in prostate cancer initiation and progression. Nat Rev Urol 2023; 20:524-543. [PMID: 36964408 DOI: 10.1038/s41585-023-00738-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/26/2023]
Abstract
Cancer cells proliferate, differentiate and migrate by repurposing physiological signalling mechanisms. In particular, altered calcium signalling is emerging as one of the most widespread adaptations in cancer cells. Remodelling of calcium signalling promotes the development of several malignancies, including prostate cancer. Gene expression data from in vitro, in vivo and bioinformatics studies using patient samples and xenografts have shown considerable changes in the expression of various components of the calcium signalling toolkit during the development of prostate cancer. Moreover, preclinical and clinical evidence suggests that altered calcium signalling is a crucial component of the molecular re-programming that drives prostate cancer progression. Evidence points to calcium signalling re-modelling, commonly involving crosstalk between calcium and other cellular signalling pathways, underpinning the onset and temporal progression of this disease. Discrete alterations in calcium signalling have been implicated in hormone-sensitive, castration-resistant and aggressive variant forms of prostate cancer. Hence, modulation of calcium signals and downstream effector molecules is a plausible therapeutic strategy for both early and late stages of prostate cancer. Based on this premise, clinical trials have been undertaken to establish the feasibility of targeting calcium signalling specifically for prostate cancer.
Collapse
Affiliation(s)
| | - Vanessa Nicolì
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | | | - Francesco Crea
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Martin D Bootman
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK.
| |
Collapse
|
7
|
Laajala TD, Sreekanth V, Soupir AC, Creed JH, Halkola AS, Calboli FCF, Singaravelu K, Orman MV, Colin-Leitzinger C, Gerke T, Fridley BL, Tyekucheva S, Costello JC. A harmonized resource of integrated prostate cancer clinical, -omic, and signature features. Sci Data 2023; 10:430. [PMID: 37407670 PMCID: PMC10322899 DOI: 10.1038/s41597-023-02335-4] [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: 01/18/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023] Open
Abstract
Genomic and transcriptomic data have been generated across a wide range of prostate cancer (PCa) study cohorts. These data can be used to better characterize the molecular features associated with clinical outcomes and to test hypotheses across multiple, independent patient cohorts. In addition, derived features, such as estimates of cell composition, risk scores, and androgen receptor (AR) scores, can be used to develop novel hypotheses leveraging existing multi-omic datasets. The full potential of such data is yet to be realized as independent datasets exist in different repositories, have been processed using different pipelines, and derived and clinical features are often not provided or not standardized. Here, we present the curatedPCaData R package, a harmonized data resource representing >2900 primary tumor, >200 normal tissue, and >500 metastatic PCa samples across 19 datasets processed using standardized pipelines with updated gene annotations. We show that meta-analysis across harmonized studies has great potential for robust and clinically meaningful insights. curatedPCaData is an open and accessible community resource with code made available for reproducibility.
Collapse
Affiliation(s)
- Teemu D Laajala
- Department of Mathematics and Statistics, University of Turku, Turku, Finland.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Varsha Sreekanth
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alex C Soupir
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Jordan H Creed
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Anni S Halkola
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
| | - Federico C F Calboli
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
- Natural Resources Institute Finland (Luke), F-31600, Jokioinen, Finland
| | | | - Michael V Orman
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Travis Gerke
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Brooke L Fridley
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Svitlana Tyekucheva
- Department of Data Science, Dana-Farber Cancer Institute; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - James C Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
8
|
Segura‐Moreno YY, Sanabria‐Salas MC, Mesa‐López De Mesa JA, Varela‐Ramirez R, Acosta‐Vega NL, Serrano ML. Determination of ERG(+), EZH2, NKX3.1, and SPINK-1 subtypes to evaluate their association with clonal origin and disease progression in multifocal prostate cancer. Cancer Rep (Hoboken) 2023; 6:e1728. [PMID: 36199157 PMCID: PMC9940006 DOI: 10.1002/cnr2.1728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/26/2022] [Accepted: 09/14/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND The prognostic relevance of prostate cancer (PCa) molecular subtypes remains controversial, given the presence of multiple foci with the possibility of different subtypes in the same patient. AIM To determine the clonal origin of heterogeneity in PCa and its association with disease progression, SPOP, ERG(+), EZH2, NKX3.1, and SPINK-1 subtypes were analyzed. METHODS A total of 103 samples from 20 PCa patients were analyzed; foci of adjacent non-tumor prostate tissue, HGPIN, GL3, GL4, GL5, and LN were examined to determine the presence of the TMPRSS2-ERG fusion and ERG, EZH2, NKX3.1, and SPINK-1 expression levels, using RT-PCR. Mutations in exons 6 and 7 of the SPOP gene were determined by sequencing. The presence of subtypes and molecular patterns were identified by combining all subtypes analyzed. To establish the clonal origin of multifocal PCa, molecular concordance between different foci of the same patient was determined. Association of these subtypes with histopathological groups and time to biochemical recurrence (BCR) was assessed. RESULTS No mutation was found in SPOP in any sample. The ERG(+) subtype was the most frequent. The molecular pattern containing all four PCa subtypes was only detected in 3 samples (4%), all LN, but it was the most frequent (40%) in patients. Molecular discordance was the predominant status (55%) when all analyzed molecular characteristics were considered. It was possible to find all subtypes, starting as a preneoplastic lesion, and all but one LN molecular subtype were ERG(+) and NKX3.1 subtypes. Only the expression of the NKX3.1 gene was significantly different among the histopathological groups. No association was found between BCR time in patients and molecular subtypes or molecular concordance or between clinicopathological characteristics and molecular subtypes of ERG, EZH2, and SPINK-1. CONCLUSION The predominance of molecular discordance in prostatic foci per patient, which reflects the multifocal origin of PCa foci, highlights the importance of analyzing multiple samples to establish the prognostic and therapeutic relevance of molecular subtypes in a patient. All the subtypes analyzed here are of early onset, starting from preneoplastic lesions. NKX3.1 gene expression is the only molecular characteristic that shows a progression pattern by sample.
Collapse
Affiliation(s)
- Yenifer Yamile Segura‐Moreno
- Cancer Biology Research GroupInstituto Nacional de CancerologíaBogotáColombia
- Department of ChemistryUniversidad Nacional de Colombia, Ciudad UniversitariaBogotáColombia
| | | | | | - Rodolfo Varela‐Ramirez
- Department of UrologyInstituto Nacional de CancerologíaBogotáColombia
- Department of UrologyUniversidad Nacional de ColombiaBogotáColombia
| | | | - Martha Lucía Serrano
- Cancer Biology Research GroupInstituto Nacional de CancerologíaBogotáColombia
- Department of ChemistryUniversidad Nacional de Colombia, Ciudad UniversitariaBogotáColombia
| |
Collapse
|
9
|
Laajala TD, Sreekanth V, Soupir A, Creed J, Calboli FCF, Singaravelu K, Orman M, Colin-Leitzinger C, Gerke T, Fidley BL, Tyekucheva S, Costello JC. curatedPCaData: Integration of clinical, genomic, and signature features in a curated and harmonized prostate cancer data resource. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.524403. [PMID: 36711769 PMCID: PMC9882125 DOI: 10.1101/2023.01.17.524403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Genomic and transcriptomic data have been generated across a wide range of prostate cancer (PCa) study cohorts. These data can be used to better characterize the molecular features associated with clinical outcomes and to test hypotheses across multiple, independent patient cohorts. In addition, derived features, such as estimates of cell composition, risk scores, and androgen receptor (AR) scores, can be used to develop novel hypotheses leveraging existing multi-omic datasets. The full potential of such data is yet to be realized as independent datasets exist in different repositories, have been processed using different pipelines, and derived and clinical features are often not provided or unstandardized. Here, we present the curatedPCaData R package, a harmonized data resource representing >2900 primary tumor, >200 normal tissue, and >500 metastatic PCa samples across 19 datasets processed using standardized pipelines with updated gene annotations. We show that meta-analysis across harmonized studies has great potential for robust and clinically meaningful insights. curatedPCaData is an open and accessible community resource with code made available for reproducibility.
Collapse
Affiliation(s)
- Teemu D Laajala
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Varsha Sreekanth
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alex Soupir
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Jordan Creed
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Federico CF Calboli
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
- Natural Resources Institute Finland (Luke), F-31600, Jokioinen, Finland
| | | | - Michael Orman
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Travis Gerke
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Brooke L. Fidley
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Svitlana Tyekucheva
- Department of Data Science, Dana-Farber Cancer Institute; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - James C Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
10
|
Su Z, Zhang M, Luo H, Zhong J, Tan J, Xu Y, Pan X, Zeng H, Nie L, Xu M, Chen N, Chen X, Zhou Q. circEZH2 E2 /E3 is a dual suppressor of miR363/miR708 to promote EZH2 expression and prostate cancer progression. Cancer Sci 2022; 114:1378-1395. [PMID: 36519785 PMCID: PMC10067432 DOI: 10.1111/cas.15694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/28/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
The histone methyltransferase enhancer of zeste homolog 2 (EZH2) is overexpressed in a variety of malignancies including prostate cancer (PCa) and may play important roles in tumor progression. Gene copy number gains, enhanced transcription, and a few circRNAs have been reported to upregulate EZH2. It was not known whether EZH2 itself generates circRNAs that promote its own expression. We here report the identification of circEZH2E2/E3 that is derived from exons 2 and 3 of the EZH2 gene and overexpressed in PCa. We show that circEZH2E2/E3 functions as a dual inhibitor for both miR363 and miR708 that target the EZH2 3'UTR and CDS, respectively, resulting in the upregulation of EZH2 expression and hence the downregulation of EZH2-repressed genes (e.g., CDH1 and DAB2IP), and enhancement of PCa cell proliferation, migration, invasion, and xenograft PCa growth. Overexpression of circEZH2E2/E3 is significantly correlated with higher tumor grade, tumor progression, and unfavorable progression-free and disease-specific survival in PCa patients. These findings show a novel autoenhancing EZH2-circEZH2E2/E3 -miR363/miR708-EZH2 regulatory loop, by which circEZH2E2/E3 plays important roles in PCa tumorigenesis and progression by upregulating EZH2, and may have potential diagnostic, prognostic, and therapeutic uses in PCa management.
Collapse
Affiliation(s)
- Zhengzheng Su
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Mengni Zhang
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Han Luo
- Department of Thyroid and Parathyroid Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jinjing Zhong
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Junya Tan
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yunyi Xu
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xiuyi Pan
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hao Zeng
- Department of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ling Nie
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Miao Xu
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ni Chen
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xueqin Chen
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Qiao Zhou
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
The role of transcription factors in the acquisition of the four latest proposed hallmarks of cancer and corresponding enabling characteristics. Semin Cancer Biol 2022; 86:1203-1215. [PMID: 36244529 DOI: 10.1016/j.semcancer.2022.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 01/27/2023]
Abstract
With the recent description of the molecular and cellular characteristics that enable acquisition of both core and new hallmarks of cancer, the consequences of transcription factor dysregulation in the hallmarks scheme has become increasingly evident. Dysregulation or mutation of transcription factors has long been recognized in the development of cancer where alterations in these key regulatory molecules can result in aberrant gene expression and consequential blockade of normal cellular differentiation. Here, we provide an up-to-date review of involvement of dysregulated transcription factor networks with the most recently reported cancer hallmarks and enabling characteristic properties. We present some illustrative examples of the impact of dysregulated transcription factors, specifically focusing on the characteristics of phenotypic plasticity, non-mutational epigenetic reprogramming, polymorphic microbiomes, and senescence. We also discuss how new insights into transcription factor dysregulation in cancer is contributing to addressing current therapeutic challenges.
Collapse
|
12
|
Lin Z, Radaeva M, Cherkasov A, Dong X. Lin28 Regulates Cancer Cell Stemness for Tumour Progression. Cancers (Basel) 2022; 14:4640. [PMID: 36230562 PMCID: PMC9564245 DOI: 10.3390/cancers14194640] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/15/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Tumours develop therapy resistance through complex mechanisms, one of which is that cancer stem cell (CSC) populations within the tumours present self-renewable capability and phenotypical plasticity to endure therapy-induced stress conditions and allow tumour progression to the therapy-resistant state. Developing therapeutic strategies to cope with CSCs requires a thorough understanding of the critical drivers and molecular mechanisms underlying the aforementioned processes. One such hub regulator of stemness is Lin28, an RNA-binding protein. Lin28 blocks the synthesis of let-7, a tumour-suppressor microRNA, and acts as a global regulator of cell differentiation and proliferation. Lin28also targets messenger RNAs and regulates protein translation. In this review, we explain the role of the Lin28/let-7 axis in establishing stemness, epithelial-to-mesenchymal transition, and glucose metabolism reprogramming. We also highlight the role of Lin28 in therapy-resistant prostate cancer progression and discuss the emergence of Lin28-targeted therapeutics and screening methods.
Collapse
Affiliation(s)
- Zhuohui Lin
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Faculty of Food and Land Systems, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Mariia Radaeva
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Artem Cherkasov
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Xuesen Dong
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| |
Collapse
|
13
|
Oyelakin A, Nayak KB, Glathar AR, Gluck C, Wrynn T, Tugores A, Romano RA, Sinha S. EHF is a novel regulator of cellular redox metabolism and predicts patient prognosis in HNSCC. NAR Cancer 2022; 4:zcac017. [PMID: 35664541 PMCID: PMC9155246 DOI: 10.1093/narcan/zcac017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 04/28/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022] Open
Abstract
Head and Neck Squamous Cell Carcinoma (HNSCC) is a heterogeneous disease with relatively high morbidity and mortality rates. The lack of effective therapies, high recurrence rates and drug resistance driven in part, by tumor heterogeneity, contribute to the poor prognosis for patients diagnosed with this cancer. This problem is further exacerbated by the fact that key regulatory factors contributing to the disease diversity remains largely elusive. Here, we have identified EHF as an important member of the ETS family of transcription factors that is highly expressed in normal oral tissues, but lost during HNSCC progression. Interestingly, HNSCC tumors and cell lines exhibited a dichotomy of high and low EHF expression, and patients whose tumors retained EHF expression showed significantly better prognosis, suggesting a potential tumor suppressive role for EHF. To address this, we have performed gain and loss of function studies and leveraged bulk and single-cell cancer genomic datasets to identify global EHF targets by RNA-sequencing (RNA-seq) and Chromatin Immunoprecipitation and next generation sequencing (ChIP-seq) experiments of HNSCC cell lines. These mechanistic studies have revealed that EHF, acts as a regulator of a broad spectrum of metabolic processes, specifically targeting regulators of redox homeostasis such as NRF2 and SOX2. Our immunostaining results confirm the mutually exclusive expression patterns of EHF and SOX2 in HNSCC tumors and suggest a possible role for these two factors in establishing discrete metabolic states within the tumor microenvironment. Taken together, EHF may serve as a novel prognostic marker for classifying HNSCC patients for actionable and targeted therapeutic intervention.
Collapse
Affiliation(s)
- Akinsola Oyelakin
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, USA
| | - Kasturi Bala Nayak
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Alexandra Ruth Glathar
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Christian Gluck
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Theresa Wrynn
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Antonio Tugores
- Unidad de Investigación, Complejo Hospitalario Universitario Insular Materno Infantil Avda Maritima del Sur, Las Palmas de Gran Canaria, Spain
| | - Rose-Anne Romano
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
14
|
Lorenzoni M, De Felice D, Beccaceci G, Di Donato G, Foletto V, Genovesi S, Bertossi A, Cambuli F, Lorenzin F, Savino A, Avalle L, Cimadamore A, Montironi R, Weber V, Carbone FG, Barbareschi M, Demichelis F, Romanel A, Poli V, Del Sal G, Julio MKD, Gaspari M, Alaimo A, Lunardi A. ETS-related gene (ERG) undermines genome stability in mouse prostate progenitors via Gsk3β dependent Nkx3.1 degradation. Cancer Lett 2022; 534:215612. [PMID: 35259458 PMCID: PMC8968219 DOI: 10.1016/j.canlet.2022.215612] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/13/2022]
Abstract
21q22.2–3 deletion is the most common copy number alteration in prostate cancer (PCa). The genomic rearrangement results in the androgen-dependent de novo expression of ETS-related gene (ERG) in prostate cancer cells, a condition promoting tumor progression to advanced stages of the disease. Interestingly, ERG expression characterizes 5–30% of tumor precursor lesions – High Grade Prostatic Intraepithelial Neoplasia (HGPIN) - where its role remains unclear. Here, by combining organoids technology with Click-chemistry coupled Mass Spectrometry, we demonstrate a prominent role of ERG in remodeling the protein secretome of prostate progenitors. Functionally, by lowering autocrine Wnt-4 signaling, ERG represses canonical Wnt pathway in prostate progenitors, and, in turn, promotes the accumulation of DNA double strand breaks via Gsk3β-dependent degradation of the tumor suppressor Nkx3.1. On the other hand, by shaping extracellular paracrine signals, ERG strengthens the pro-oxidative transcriptional signature of inflammatory macrophages, which we demonstrate to infiltrate pre-malignant ERG positive prostate lesions. These findings highlight previously unrecognized functions of ERG in undermining adult prostate progenitor niche through cell autonomous and non-autonomous mechanisms. Overall, by supporting the survival and proliferation of prostate progenitors in the absence of growth stimuli and promoting the accumulation of DNA damage through destabilization of Nkx3.1, ERG could orchestrate the prelude to neoplastic transformation. Expression of ERGM40 in mouse prostate organoids promotes their survival and growth in the absence of Egf. ERGM40 alters the extracellular signaling network of mouse prostate organoids. Canonical Wnt pathway is substantially reduced in ERG + prostate organoids due to decreased autocrine signaling of Wnt4. Gsk3b promotes Nkx3.1 proteolysis and, in turn, accumulation of double strand breaks in ERG + prostate organoids. Paracrine signaling of ERG + prostate organoids modulates Arginase 1 expression in M1-polarized macrophages.
Collapse
Affiliation(s)
- Marco Lorenzoni
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Dario De Felice
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Giulia Beccaceci
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Giorgia Di Donato
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Veronica Foletto
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Sacha Genovesi
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Arianna Bertossi
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Francesco Cambuli
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Francesca Lorenzin
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Aurora Savino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Alessia Cimadamore
- Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| | - Rodolfo Montironi
- Molecular Medicine and Cell Therapy Foundation, Polytechnic University of the Marche Region, Via Tronto, 10, Ancona, Italy
| | - Veronica Weber
- Unit of Surgical Pathology, Santa Chiara Hospital, Trento, Italy
| | | | | | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Alessandro Romanel
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Valeria Poli
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Giannino Del Sal
- University of Trieste Department Life Sciences, ICGEB-Area Science Park Trieste, IFOM, Milan, Italy
| | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, Bern, Switzerland; Translational Organoid Resource CORE, Department for BioMedical Research, University of Bern, Bern, Switzerland; Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland; Department of Urology, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Marco Gaspari
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy.
| | - Alessandro Alaimo
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy.
| | - Andrea Lunardi
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy.
| |
Collapse
|
15
|
Montagud A, Béal J, Tobalina L, Traynard P, Subramanian V, Szalai B, Alföldi R, Puskás L, Valencia A, Barillot E, Saez-Rodriguez J, Calzone L. Patient-specific Boolean models of signalling networks guide personalised treatments. eLife 2022; 11:e72626. [PMID: 35164900 PMCID: PMC9018074 DOI: 10.7554/elife.72626] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/27/2022] [Indexed: 11/22/2022] Open
Abstract
Prostate cancer is the second most occurring cancer in men worldwide. To better understand the mechanisms of tumorigenesis and possible treatment responses, we developed a mathematical model of prostate cancer which considers the major signalling pathways known to be deregulated. We personalised this Boolean model to molecular data to reflect the heterogeneity and specific response to perturbations of cancer patients. A total of 488 prostate samples were used to build patient-specific models and compared to available clinical data. Additionally, eight prostate cell line-specific models were built to validate our approach with dose-response data of several drugs. The effects of single and combined drugs were tested in these models under different growth conditions. We identified 15 actionable points of interventions in one cell line-specific model whose inactivation hinders tumorigenesis. To validate these results, we tested nine small molecule inhibitors of five of those putative targets and found a dose-dependent effect on four of them, notably those targeting HSP90 and PI3K. These results highlight the predictive power of our personalised Boolean models and illustrate how they can be used for precision oncology.
Collapse
Affiliation(s)
- Arnau Montagud
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3BarcelonaSpain
| | - Jonas Béal
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| | - Luis Tobalina
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
| | - Pauline Traynard
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| | - Vigneshwari Subramanian
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
| | - Bence Szalai
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
- Semmelweis University, Faculty of Medicine, Department of PhysiologyBudapestHungary
| | | | | | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3BarcelonaSpain
- ICREA, Pg. Lluís Companys 23BarcelonaSpain
| | - Emmanuel Barillot
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| | - Julio Saez-Rodriguez
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg UniversityHeidelbergGermany
| | - Laurence Calzone
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| |
Collapse
|
16
|
Chromatin-Independent Interplay of NFATc1 and EZH2 in Pancreatic Cancer. Cells 2021; 10:cells10123463. [PMID: 34943970 PMCID: PMC8700089 DOI: 10.3390/cells10123463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/15/2022] Open
Abstract
Background: The Nuclear Factor of Activated T-cells 1 (NFATc1) transcription factor and the methyltransferase Enhancer of Zeste Homolog 2 (EZH2) significantly contribute to the aggressive phenotype of pancreatic ductal adenocarcinoma (PDAC). Herein, we aimed at dissecting the mechanistic background of their interplay in PDAC progression. Methods: NFATc1 and EZH2 mRNA and protein expression and complex formation were determined in transgenic PDAC models and human PDAC specimens. NFATc1 binding on the Ezh2 gene and the consequences of perturbed NFATc1 expression on Ezh2 transcription were explored by Chromatin Immunoprecipitation (ChIP) and upon transgenic or siRNA-mediated interference with NFATc1 expression, respectively. Integrative analyses of RNA- and ChIP-seq data was performed to explore NFATc1-/EZH2-dependent gene signatures. Results: NFATc1 targets the Ezh2 gene for transcriptional activation and biochemically interacts with the methyltransferase in murine and human PDAC. Surprisingly, our genome-wide binding and expression analyses do not link the protein complex to joint gene regulation. In contrast, our findings provide evidence for chromatin-independent functions of the NFATc1:EZH2 complex and reveal posttranslational EZH2 phosphorylation at serine 21 as a prerequisite for robust complex formation. Conclusion: Our findings disclose a previously unknown NFATc1-EZH2 axis operational in the pancreas and provide mechanistic insights into the conditions fostering NFATc1:EZH2 complex formation in PDAC.
Collapse
|
17
|
Dovey ZS, Nair SS, Chakravarty D, Tewari AK. Racial disparity in prostate cancer in the African American population with actionable ideas and novel immunotherapies. Cancer Rep (Hoboken) 2021; 4:e1340. [PMID: 33599076 PMCID: PMC8551995 DOI: 10.1002/cnr2.1340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/22/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND African Americans (AAs) in the United States are known to have a higher incidence and mortality for Prostate Cancer (PCa). The drivers of this epidemiological disparity are multifactorial, including socioeconomic factors leading to lifestyle and dietary issues, healthcare access problems, and potentially tumor biology. RECENT FINDINGS Although recent evidence suggests once access is equal, AA men have equal outcomes to Caucasian American (CA) men, differences in PCa incidence remain, and there is much to do to reverse disparities in mortality across the USA. A deeper understanding of these issues, both at the clinical and molecular level, can facilitate improved outcomes in the AA population. This review first discusses PCa oncogenesis in the context of its diverse hallmarks before benchmarking key molecular and genomic differences for PCa in AA men that have emerged in the recent literature. Studies have emphasized the importance of tumor microenvironment that contributes to both the unequal cancer burden and differences in clinical outcome between the races. Management of comorbidities like obesity, hypertension, and diabetes will provide an essential means of reducing prostate cancer incidence in AA men. Although requiring further AA specific research, several new treatment strategies such as immune checkpoint inhibitors used in combination PARP inhibitors and other emerging vaccines, including Sipuleucel-T, have demonstrated some proven efficacy. CONCLUSION Genomic profiling to integrate clinical and genomic data for diagnosis, prognosis, and treatment will allow physicians to plan a "Precision Medicine" approach to AA men. There is a pressing need for further research for risk stratification, which may allow early identification of AA men with higher risk disease based on their unique clinical, genomic, and immunological profiles, which can then be mapped to appropriate clinical trials. Treatment options are outlined, with a concise description of recent work in AA specific populations, detailing several targeted therapies, including immunotherapy. Also, a summary of current clinical trials involving AA men is presented, and it is important that policies are adopted to ensure that AA men are actively recruited. Although it is encouraging that many of these explore the lifestyle and educational initiatives and therapeutic interventions, there is much still work to be done to reduce incidence and mortality in AA men and equalize current racial disparities.
Collapse
Affiliation(s)
- Zachary S. Dovey
- The Department of UrologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Sujit S. Nair
- The Department of UrologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Dimple Chakravarty
- The Department of UrologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ashutosh K. Tewari
- The Department of UrologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
18
|
Jillson LK, Yette GA, Laajala TD, Tilley WD, Costello JC, Cramer SD. Androgen Receptor Signaling in Prostate Cancer Genomic Subtypes. Cancers (Basel) 2021; 13:3272. [PMID: 34208794 PMCID: PMC8269091 DOI: 10.3390/cancers13133272] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022] Open
Abstract
While many prostate cancer (PCa) cases remain indolent and treatable, others are aggressive and progress to the metastatic stage where there are limited curative therapies. Androgen receptor (AR) signaling remains an important pathway for proliferative and survival programs in PCa, making disruption of AR signaling a viable therapy option. However, most patients develop resistance to AR-targeted therapies or inherently never respond. The field has turned to PCa genomics to aid in stratifying high risk patients, and to better understand the mechanisms driving aggressive PCa and therapy resistance. While alterations to the AR gene itself occur at later stages, genomic changes at the primary stage can affect the AR axis and impact response to AR-directed therapies. Here, we review common genomic alterations in primary PCa and their influence on AR function and activity. Through a meta-analysis of multiple independent primary PCa databases, we also identified subtypes of significantly co-occurring alterations and examined their combinatorial effects on the AR axis. Further, we discussed the subsequent implications for response to AR-targeted therapies and other treatments. We identified multiple primary PCa genomic subtypes, and given their differing effects on AR activity, patient tumor genetics may be an important stratifying factor for AR therapy resistance.
Collapse
Affiliation(s)
- Lauren K. Jillson
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (J.K.L.); (G.A.Y.); (T.D.L.); (J.C.C.)
| | - Gabriel A. Yette
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (J.K.L.); (G.A.Y.); (T.D.L.); (J.C.C.)
| | - Teemu D. Laajala
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (J.K.L.); (G.A.Y.); (T.D.L.); (J.C.C.)
- Department of Mathematics and Statistics, University of Turku, 20500 Turku, Finland
| | - Wayne D. Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia;
- Freemason’s Foundation Centre for Men’s Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - James C. Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (J.K.L.); (G.A.Y.); (T.D.L.); (J.C.C.)
| | - Scott D. Cramer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (J.K.L.); (G.A.Y.); (T.D.L.); (J.C.C.)
| |
Collapse
|
19
|
Segura-Moreno YY, Sanabria-Salas MC, Varela R, Mesa JA, Serrano ML. Decoding the heterogeneous landscape in the development prostate cancer. Oncol Lett 2021; 21:376. [PMID: 33777200 PMCID: PMC7988715 DOI: 10.3892/ol.2021.12637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 06/02/2020] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer (PCa) is characterized as being histologically and molecularly heterogeneous; however, this is not only incorrect among individuals, but also at the multiple foci level, which originates in the prostate gland itself. The reasons for such heterogeneity have not been fully elucidated; however, understanding these may be crucial in determining the course of the disease. PCa is characterized by a complex network of chromosomal rearrangements, which simultaneously deregulate multiple genes; this could explain the appearance of exclusive events associated with molecular subtypes, which have been extensively investigated to establish clinical management and the development of therapies targeted to this type of cancer. From a clinical aspect, the prognosis of the patient has focused on the characteristics of the index lesion (the largest focus in PCa); however, a significant percentage of patients (11%) also exhibit an aggressive secondary foci, which may determine the prognosis of the disease, and could be the determining factor of why, in different studies, the classification of the subtypes does not have an association with prognosis. Due to the aforementioned reasons, the analysis of molecular subtypes in several foci, from the same individual could assist in determining the association between clinical evolution and management of patients with PCa. Castration-resistant PCa (CRPC) has the worst prognosis and develops following androgen ablation therapy. Currently, there are two models to explain the development of CRPC: i) The selection model and ii) the adaptation model; both of which, have been found to include alterations described in the molecular subtypes, such as Enhancer of zeste 2 polycomb repressive complex 2 subunit overexpression, isocitrate dehydrogenase (NAPD+)1 and forkhead box A1 mutations, suggesting that the presence of specific molecular alterations could predict the development of CRPC. This type of analysis could lead to a biological understanding of PCa, to develop personalized medicine strategies, which could improve the response to treatment thus, avoiding the development of resistance. Therefore, the present review discusses the primary molecular factors, to which variable heterogeneity in PCa progress has been attributed.
Collapse
Affiliation(s)
- Yenifer Yamile Segura-Moreno
- Cancer Biology Research Group, National Institute of Cancerology, Bogota 110411, Colombia.,Department of Chemistry, Faculty of Sciences, National University of Colombia, University City, Bogota 111321, Colombia
| | | | - Rodolfo Varela
- Department of Urology, National Institute of Cancerology, Bogota 110411, Colombia.,Department of Urology, National University of Colombia, University City, Bogota 111321, Colombia
| | - Jorge Andrés Mesa
- Department of Pathology, National Institute of Cancerology, Bogota 110411, Colombia
| | - Martha Lucia Serrano
- Cancer Biology Research Group, National Institute of Cancerology, Bogota 110411, Colombia.,Department of Chemistry, Faculty of Sciences, National University of Colombia, University City, Bogota 111321, Colombia
| |
Collapse
|
20
|
Circulating extracellular vesicles release oncogenic miR-424 in experimental models and patients with aggressive prostate cancer. Commun Biol 2021; 4:119. [PMID: 33500545 PMCID: PMC7838273 DOI: 10.1038/s42003-020-01642-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/24/2020] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs) are relevant means for transferring signals across cells and facilitate propagation of oncogenic stimuli promoting disease evolution and metastatic spread in cancer patients. Here, we investigated the release of miR-424 in circulating small EVs or exosomes from prostate cancer patients and assessed the functional implications in multiple experimental models. We found higher frequency of circulating miR-424 positive EVs in patients with metastatic prostate cancer compared to patients with primary tumors and BPH. Release of miR-424 in small EVs was enhanced in cell lines (LNCaPabl), transgenic mice (Pb-Cre4;Ptenflox/flox;Rosa26ERG/ERG) and patient-derived xenograft (PDX) models of aggressive disease. EVs containing miR-424 promoted stem-like traits and tumor-initiating properties in normal prostate epithelial cells while enhanced tumorigenesis in transformed prostate epithelial cells. Intravenous administration of miR-424 positive EVs to mice, mimicking blood circulation, promoted miR-424 transfer and tumor growth in xenograft models. Circulating miR-424 positive EVs from patients with aggressive primary and metastatic tumors induced stem-like features when supplemented to prostate epithelial cells. This study establishes that EVs-mediated transfer of miR-424 across heterogeneous cell populations is an important mechanism of tumor self-sustenance, disease recurrence and progression. These findings might indicate novel approaches for the management and therapy of prostate cancer. Following on reports that miR-424 expression promotes oncogenesis, Domenico Albino et al. find that extracellular vesicles (EVs) in the plasma of prostate cancer patients secrete miR-424. Using cell-based and animal models, they demonstrate that EV-mediated release of miR-424 can transfer oncogenic signals across cells to promote recurrence and metastatic progression.
Collapse
|
21
|
Zhou W, Su Y, Zhang Y, Han B, Liu H, Wang X. Endothelial Cells Promote Docetaxel Resistance of Prostate Cancer Cells by Inducing ERG Expression and Activating Akt/mTOR Signaling Pathway. Front Oncol 2021; 10:584505. [PMID: 33425737 PMCID: PMC7793734 DOI: 10.3389/fonc.2020.584505] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/13/2020] [Indexed: 11/13/2022] Open
Abstract
Docetaxel is a first-line chemotherapy for the treatment of patients with castration-resistant prostate cancer (CRPC). Despite the good initial response of docetaxel, drug resistance will inevitably occur. Mechanisms underlying docetaxel resistance are not well elaborated. Endothelial cells (ECs) have been implicated in the progression and metastasis of prostate cancer. However, little attention has been paid to the role of endothelial cells in the development of docetaxel resistance in prostate cancer. Here, we sought to investigate the function and mechanism of endothelial cells involving in the docetaxel resistance of prostate cancer. We found that endothelial cells significantly promoted the proliferation of prostate cancer cells and decreased their sensitivity to docetaxel. Mechanistically, basic fibroblast growth factor (FGF2) secreted by endothelial cells leads to the upregulation of ETS related gene (ERG) expression and activation of the Akt/mTOR signaling pathway in prostate cancer cells to promote docetaxel resistance. In summary, these findings demonstrate a microenvironment-dependent mechanism mediating chemoresistance of prostate cancer and suggest that targeting FGF/FGFR signaling might represent a promising therapeutic strategy to overcome docetaxel resistance.
Collapse
Affiliation(s)
- Wenhao Zhou
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiming Su
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bangmin Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haitao Liu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohai Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Long Z, Deng L, Li C, He Q, He Y, Hu X, Cai Y, Gan Y. Loss of EHF facilitates the development of treatment-induced neuroendocrine prostate cancer. Cell Death Dis 2021; 12:46. [PMID: 33414441 PMCID: PMC7790822 DOI: 10.1038/s41419-020-03326-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022]
Abstract
The rising of a highly aggressive subtype of castration-resistant prostate cancer (CRPC) named treatment-induced neuroendocrine prostate cancer (t-NEPC) after androgen deprivation therapy (ADT) is well known for its features of the neuroendocrine differentiation (NED) and androgen receptor (AR) independence. However, t-NEPC is still largely unknown. Here, we found that EHF is notably depressed in t-NEPC tumors, patient-derived xenografts, transgenic mice, and cell models. Results from cell lines uncovered that ADT represses EHF expression, which is required for the ADT-induced NED. Mechanism dissection revealed that ADT decreases the EHF transcription via relieving the AR binding to different androgen-responsive elements, which then promotes the expression and enzymatic activity of enhancer of zeste homolog 2 (EZH2), consequently catalyzing tri-methylation lysine 27 of histone H3 for transcriptional repression of its downstream genes to promote the NED. Furthermore, preclinical studies from cell and mice models proved that recovery of EHF expression or using EZH2 inhibitor can attenuate aggressive properties of CRPC cells, hinder the progression of t-NEPC, and promote the response of CPRC cells to enzalutamide. Together, we elucidate that the ADT/AR/EHF/EZH2 signaling is required for the ADT-enhanced NED and plays a critical role in the progression of t-NEPC.
Collapse
Affiliation(s)
- Zhi Long
- Department of Urology, Andrology Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Liang Deng
- Department of Urology, Andrology Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Chao Li
- Department of Urology, Andrology Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Qiangrong He
- Department of Urology, Andrology Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yao He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Xiheng Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Yi Cai
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Yu Gan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| |
Collapse
|
23
|
Kareddula A, Medina DJ, Petrosky W, Dolfi S, Tereshchenko I, Walton K, Aviv H, Sadimin E, Tabakin AL, Singer EA, Hirshfield KM. The role of chromodomain helicase DNA binding protein 1 (CHD1) in promoting an invasive prostate cancer phenotype. Ther Adv Urol 2021; 13:17562872211022462. [PMID: 34408788 PMCID: PMC8365013 DOI: 10.1177/17562872211022462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 05/15/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Prostate cancer (PCa) phenotypes vary from indolent to aggressive. Molecular subtyping may be useful in predicting aggressive cancers and directing therapy. One such subtype involving deletions of chromodomain helicase DNA binding protein 1 (CHD1), a tumor suppressor gene, are found in 10-26% of PCa tumors. In this study, we evaluate the functional cellular effects that follow CHD1 deletion. METHODS CHD1 was knocked out (KO) in the non-tumorigenic, human papillomavirus 16 (HPV16)-immortalized prostate epithelial cell line, RWPE-1, using CRISPR/Cas9. In vitro assays such as T7 endonuclease assay, western blot, and sequencing were undertaken to characterize the CHD1 KO clones. Morphologic and functional assays for cell adhesion and viability were performed. To study expression of extracellular matrix (ECM) and adhesion molecules, a real-time (RT) profiler assay was performed using RWPE-1 parental, non-target cells (NT2) and CHD1 KO cells. RESULT Compared to parental RWPE-1 and non-target cells (NT2), the CHD1 KO cells had a smaller, rounder morphology and were less adherent under routine culture conditions. Compared to parental cells, CHD1 KO cells showed a reduction in ECM and adhesion molecules as well as a greater proportion of viable suspension cells when cultured on standard tissue culture plates and on plates coated with laminin, fibronectin or collagen I. CHD1 KO cells showed a decrease in the expression of secreted protein acidic and rich in cysteine (SPARC), matrix metalloproteinase 2 (MMP2), integrin subunit alpha 2 (ITGA2), integrin subunit alpha 5 (ITGA5), integrin subunit alpha 6 (ITGA6), fibronectin (FN1), laminin subunit beta-3 precursor (LAMB3), collagen, tenascin and vitronectin as compared to parental and NT2 cells. CONCLUSION These data suggest that in erythroblast transformation specific (ETS) fusion-negative, phosphatase and tensin homolog (PTEN) wildtype PCa, deletion of CHD1 alters cell-cell and cell-matrix adhesion dynamics, suggesting an important role for CHD1 in the development and progression of PCa.
Collapse
Affiliation(s)
- Aparna Kareddula
- Department of Medicine, Rutgers Cancer Institute of New Jersey/Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Daniel J. Medina
- Department of Medicine, Rutgers Cancer Institute of New Jersey/Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Whitney Petrosky
- Department of Medicine, Rutgers Cancer Institute of New Jersey/Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Sonia Dolfi
- Department of Medicine, Rutgers Cancer Institute of New Jersey/Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Irina Tereshchenko
- Department of Medicine, Rutgers Cancer Institute of New Jersey/Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Kelly Walton
- Department of Medicine/Division of Medical Oncology, Rutgers Cancer Institute of New Jersey/Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Hana Aviv
- Department of Pathology and Laboratory Medicine, Rutgers -Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Evita Sadimin
- Section of Urologic Pathology, Rutgers Cancer Institute of New Jersey/Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Alexandra L. Tabakin
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey/Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Eric A. Singer
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey/Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | | |
Collapse
|
24
|
Kaundal B, Kushwaha AC, Srivastava AK, Karmakar S, Choudhury SR. A non-viral nano-delivery system targeting epigenetic methyltransferase EZH2 for precise acute myeloid leukemia therapy. J Mater Chem B 2020; 8:8658-8670. [PMID: 32844866 DOI: 10.1039/d0tb01177k] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acute myeloid leukemia (AML), which is common in the elderly population, accounts for poor long-term survival with a high possibility of relapse. The associated lack of currently developed therapeutics is directing the search for new therapeutic targets relating to AML. EZH2 (Enhancer of Zeste Homolog 2) is a histone methyltransferase member of the polycomb-group (PcG) family, and its significant overexpression in AML means it has emerged as a potential epigenetic target. Here, we propose the human serum albumin (HSA) nanoparticle based delivery of small interfering RNA (siRNA), which can target EZH2-expressing genes in AML. EZH2 specific siRNA loaded in a polyethyleneimine (PEI) conjugated HSA nanocarrier can overcome the systemic instability of siRNA and precisely target the AML cell population for increased EZH2 gene silencing. A stable nanosized complex (HSANPs-PEI@EZH2siRNA), achieved via the electrostatic interaction of PEI and EZH2 siRNA, shows increased systemic stability and hemocompatibility, and enhanced EZH2 gene silencing activity in vitro, compared to conventional transfection reagents. HSANPs-PEI@EZH2siRNA-treated AML cells showed downregulated EZH2, which is associated with a reduced level of Bmi-1 protein, and H3K27me3 and H2AK119ub modification. The ubiquitin-mediated proteasomal degradation pathway plays a critical role in the downregulation of associated proteins following HSANPs-PEI@EZH2siRNA exposure to AML cells. c-Myb is the AML-responsive transcription factor that directly binds on the EZH2 promoter and was downregulated in HSANPs-PEI@EZH2siRNA-treated AML cells. The systemic exposure to HSANPs-PEI@EZH2siRNA of AML engrafted immunodeficient nude mice displayed efficient EZH2 gene silencing and a reduced AML cell population in peripheral blood and bone marrow. The present study demonstrates a non-viral siRNA delivery system for epigenetic targeting based superior anti-leukemic therapy.
Collapse
Affiliation(s)
- Babita Kaundal
- Institute of Nano Science and Technology, Mohali, Punjab, India.
| | | | | | - Surajit Karmakar
- Institute of Nano Science and Technology, Mohali, Punjab, India.
| | | |
Collapse
|
25
|
Das P, Taube JH. Regulating Methylation at H3K27: A Trick or Treat for Cancer Cell Plasticity. Cancers (Basel) 2020; 12:E2792. [PMID: 33003334 PMCID: PMC7600873 DOI: 10.3390/cancers12102792] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
Properly timed addition and removal of histone 3 lysine 27 tri-methylation (H3K27me3) is critical for enabling proper differentiation throughout all stages of development and, likewise, can guide carcinoma cells into altered differentiation states which correspond to poor prognoses and treatment evasion. In early embryonic stages, H3K27me3 is invoked to silence genes and restrict cell fate. Not surprisingly, mutation or altered functionality in the enzymes that regulate this pathway results in aberrant methylation or demethylation that can lead to malignancy. Likewise, changes in expression or activity of these enzymes impact cellular plasticity, metastasis, and treatment evasion. This review focuses on current knowledge regarding methylation and de-methylation of H3K27 in cancer initiation and cancer cell plasticity.
Collapse
Affiliation(s)
| | - Joseph H. Taube
- Department of Biology, Baylor University, Waco, TX 76706, USA;
| |
Collapse
|
26
|
Burkhart DL, Morel KL, Wadosky KM, Labbé DP, Galbo PM, Dalimov Z, Xu B, Loda M, Ellis L. Evidence that EZH2 Deregulation is an Actionable Therapeutic Target for Prevention of Prostate Cancer. Cancer Prev Res (Phila) 2020; 13:979-988. [PMID: 32917647 DOI: 10.1158/1940-6207.capr-20-0186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/10/2020] [Accepted: 08/25/2020] [Indexed: 11/16/2022]
Abstract
Chemoprevention trials for prostate cancer by androgen receptor or androgen synthesis inhibition have proven ineffective. Recently, it has been demonstrated that the histone methlytransferase, EZH2 is deregulated in mouse and human high-grade prostatic intraepithelial neoplasia (HG-PIN). Using preclinical mouse and human models of prostate cancer, we demonstrate that genetic and chemical disruption of EZH2 expression and catalytic activity reversed the HG-PIN phenotype. Furthermore, inhibition of EZH2 function was associated with loss of cellular proliferation and induction of Tp53-dependent senescence. Together, these data provide provocative evidence for EZH2 as an actionable therapeutic target toward prevention of prostate cancer.
Collapse
Affiliation(s)
- Deborah L Burkhart
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Katherine L Morel
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kristine M Wadosky
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York
| | - David P Labbé
- Division of Urology, Department of Surgery, McGill University and Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Phillip M Galbo
- Department of Cell Biology, Albert Einstein College of Medicine, New York, New York
| | | | - Bo Xu
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Leigh Ellis
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
27
|
Secreted Frizzled-Related Protein 4 (SFRP4) Is an Independent Prognostic Marker in Prostate Cancers Lacking TMPRSS2: ERG Fusions. Pathol Oncol Res 2020; 26:2709-2722. [PMID: 32677026 PMCID: PMC7471174 DOI: 10.1007/s12253-020-00861-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/23/2020] [Indexed: 12/22/2022]
Abstract
Secreted frizzled-related protein 4 (SFRP4) controls WNT signaling and is thought to play a role for tumor aggressiveness. Here, we analyzed a tissue microarray containing 11,152 prostate cancers with pathological, clinical and molecular data by immunohistochemistry. SFRP4 expression was higher in cancer than in non-neoplastic acinar cells. SFRP4 staining was seen in 64.9% of tumors and classified as weak in 33.2%, moderate in 23.9% and strong in 7.8% of cancers. SFRP4 overexpression was linked to advanced tumor stage, high classical/quantitative Gleason grade (p < 0.0001 each), lymph node metastasis (p = 0.0002), and a positive surgical margin (p = 0.0017). SFRP4 positivity was markedly more frequent in ERG positive (77.4%) than in ERG negative cancers (57.4% p < 0.0001). Subset analyses in 2725 cancers with and 3592 cancers without TMPRSS2:ERG fusion revealed that associations with tumor phenotype and patient outcome were largely driven by the subset of ERG negative tumors. In a multivariate analysis including various postoperative and prognostic clinico-pathological features, SFRP4 protein expression emerged as an independent prognostic parameter in ERG negative cancers. SFRP4 immunostaining was significantly linked with 10 of 11 previously analyzed chromosomal deletions (p < 0.05 each). In conclusion, high SFRP4 immunostaining is associated with poor prognosis and genomic instability in ERG negative prostate cancers.
Collapse
|
28
|
Molecular and Clinical Relevance of ZBTB38 Expression Levels in Prostate Cancer. Cancers (Basel) 2020; 12:cancers12051106. [PMID: 32365491 PMCID: PMC7281456 DOI: 10.3390/cancers12051106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/12/2020] [Accepted: 04/23/2020] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer is one of the most commonly diagnosed cancers in men. A number of genomic and clinical studies have led to a better understanding of prostate cancer biology. Still, the care of patients as well as the prediction of disease aggressiveness, recurrence and outcome remain challenging. Here, we showed that expression of the gene ZBTB38 is associated with poor prognosis in localised prostate cancer and could help discriminate aggressive localised prostate tumours from those who can benefit only from observation. Analysis of different prostate cancer cohorts indicates that low expression levels of ZBTB38 associate with increased levels of chromosomal abnormalities and more aggressive pathological features, including higher rate of biochemical recurrence of the disease. Importantly, gene expression profiling of these tumours, complemented with cellular assays on prostate cancer cell lines, unveiled that tumours with low levels of ZBTB38 expression might be targeted by doxorubicin, a compound generating reactive oxygen species. Our study shows that ZBTB38 is involved in prostate cancer pathogenesis and may represent a useful marker to identify high risk and highly rearranged localised prostate cancer susceptible to doxorubicin.
Collapse
|
29
|
Zhang WF, Li T, Lin SX. Meta-Analysis of steroid-converting enzymes and related receptors in prostate cancer suggesting novel combined therapies. J Steroid Biochem Mol Biol 2020; 198:105559. [PMID: 31783154 DOI: 10.1016/j.jsbmb.2019.105559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023]
Abstract
Androgen receptor (AR) signaling is essential for prostate cancer (PC) progression and treatment. Experiments have demonstrated that the intratumoral androgen levels are not affected by circulating androgen levels, but rather modulated by local steroid-converting enzyme activities. The expression modulation status of human steroid-converting enzymes and nuclear receptors are of great promise to identify novel therapeutic targets. Meta-analysis was performed with 9 cohorts (1093 specimens) from Gene Expression Omnibus, 16 cohorts (933 specimens) from Oncomine and the TCGA cohort (550 specimens). We found significant up regulation of 5α-reductase type 1 and type 3 in both primary and metastatic PC, together with the down regulation of AKR1C2 in primary PC, contributing to the high intratumoral DHT levels. The expression of AR in metastatic PC was up regulated, indicating the importance of AR signaling in the progression of this cancer. The down regulations of HSD11B1 and NR3C1 in primary and metastatic PC may diminish the anti-inflammation and anti-proliferation effects of glucocorticoids signaling. Furthermore, the decrease of progesterone receptor (PGR) expression in primary and metastatic PC was also observed, relieving the suppression effect of PGR on PC proliferation. The clinical evidences of the remarkable expression modulation of steroid-converting enzymes and receptors in PC may indicate novel combined treatment against this highly incident cancer.
Collapse
Affiliation(s)
- Wen-Fa Zhang
- Axe Molecular Endocrinology and Nephrology, CHU Research Center and Department of Molecular Medicine, Laval University, 2705 Boulevard Laurier, Quebec City, Quebec G1V 4G2, Canada.
| | - Tang Li
- Axe Molecular Endocrinology and Nephrology, CHU Research Center and Department of Molecular Medicine, Laval University, 2705 Boulevard Laurier, Quebec City, Quebec G1V 4G2, Canada; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China.
| | - Sheng-Xiang Lin
- Axe Molecular Endocrinology and Nephrology, CHU Research Center and Department of Molecular Medicine, Laval University, 2705 Boulevard Laurier, Quebec City, Quebec G1V 4G2, Canada.
| |
Collapse
|
30
|
Kaundal B, Srivastava AK, Dev A, Mohanbhai SJ, Karmakar S, Roy Choudhury S. Nanoformulation of EPZ011989 Attenuates EZH2–c-Myb Epigenetic Interaction by Proteasomal Degradation in Acute Myeloid Leukemia. Mol Pharm 2020; 17:604-621. [DOI: 10.1021/acs.molpharmaceut.9b01071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Babita Kaundal
- Institute of Nano Science and Technology, Phase-10, Mohali (Habitat Center), Punjab 160062, India
| | - Anup K. Srivastava
- Institute of Nano Science and Technology, Phase-10, Mohali (Habitat Center), Punjab 160062, India
| | - Atul Dev
- Institute of Nano Science and Technology, Phase-10, Mohali (Habitat Center), Punjab 160062, India
| | - Soni Jignesh Mohanbhai
- Institute of Nano Science and Technology, Phase-10, Mohali (Habitat Center), Punjab 160062, India
| | - Surajit Karmakar
- Institute of Nano Science and Technology, Phase-10, Mohali (Habitat Center), Punjab 160062, India
| | - Subhasree Roy Choudhury
- Institute of Nano Science and Technology, Phase-10, Mohali (Habitat Center), Punjab 160062, India
| |
Collapse
|
31
|
Yan W, Jamal M, Tan SH, Song Y, Young D, Chen Y, Katta S, Ying K, Ravindranath L, Woodle T, Kohaar I, Cullen J, Kagan J, Srivastava S, Dobi A, McLeod DG, Rosner IL, Sesterhenn IA, Srinivasan A, Srivastava S, Petrovics G. Molecular profiling of radical prostatectomy tissue from patients with no sign of progression identifies ERG as the strongest independent predictor of recurrence. Oncotarget 2019; 10:6466-6483. [PMID: 31741711 PMCID: PMC6849651 DOI: 10.18632/oncotarget.27294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/19/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND As a major cause of morbidity and mortality among men, prostate cancer is a heterogenous disease, with a vast heterogeneity in the biology of the disease and in clinical outcome. While it often runs an indolent course, local progression or metastasis may eventually develop, even among patients considered "low risk" at diagnosis. Therefore, biomarkers that can discriminate aggressive from indolent disease at an early stage would greatly benefit patients. We hypothesized that tissue specimens from early stage prostate cancers may harbor predictive signatures for disease progression. METHODS We used a cohort of radical prostatectomy patients with longitudinal follow-up, who had tumors with low grade and stage that revealed no signs of future disease progression at surgery. During the follow-up period, some patients either remained indolent (non-BCR) or progressed to biochemical recurrence (BCR). Total RNA was extracted from tumor, and adjacent normal epithelium of formalin-fixed-paraffin-embedded (FFPE) specimens. Differential gene expression in tumors, and in tumor versus normal tissues between BCR and non-BCR patients were analyzed by NanoString using a customized CodeSet of 151 probes. RESULTS After controlling for false discovery rates, we identified a panel of eight genes (ERG, GGT1, HDAC1, KLK2, MYO6, PLA2G7, BICD1 and CACNAID) that distinguished BCR from non-BCR patients. We found a clear association of ERG expression with non-BCR, which was further corroborated by quantitative RT-PCR and immunohistochemistry assays. CONCLUSIONS Our results identified ERG as the strongest predictor for BCR and showed that potential prognostic prostate cancer biomarkers can be identified from FFPE tumor specimens.
Collapse
Affiliation(s)
- Wusheng Yan
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
- These authors contributed equally to this work
| | - Muhammad Jamal
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
- These authors contributed equally to this work
| | - Shyh-Han Tan
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
- These authors contributed equally to this work
| | - Yingjie Song
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Denise Young
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Yongmei Chen
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Shilpa Katta
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Kai Ying
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Lakshmi Ravindranath
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Tarah Woodle
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Indu Kohaar
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Jennifer Cullen
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Jacob Kagan
- Division of Cancer Prevention, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Sudhir Srivastava
- Division of Cancer Prevention, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Albert Dobi
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - David G. McLeod
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Inger L. Rosner
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | | | - Alagarsamy Srinivasan
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Shiv Srivastava
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Gyorgy Petrovics
- Henry Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, USA
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD, USA
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA
| |
Collapse
|
32
|
Civenni G, Albino D, Shinde D, Vázquez R, Merulla J, Kokanovic A, Mapelli SN, Carbone GM, Catapano CV. Transcriptional Reprogramming and Novel Therapeutic Approaches for Targeting Prostate Cancer Stem Cells. Front Oncol 2019; 9:385. [PMID: 31143708 PMCID: PMC6521702 DOI: 10.3389/fonc.2019.00385] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/25/2019] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer is the most common malignancy in men and the second cause of cancer-related deaths in western countries. Despite the progress in the treatment of localized prostate cancer, there is still lack of effective therapies for the advanced forms of the disease. Most patients with advanced prostate cancer become resistant to androgen deprivation therapy (ADT), which remains the main therapeutic option in this setting, and progress to lethal metastatic castration-resistant prostate cancer (mCRPC). Current therapies for prostate cancer preferentially target proliferating, partially differentiated, and AR-dependent cancer cells that constitute the bulk of the tumor mass. However, the subpopulation of tumor-initiating or tumor-propagating stem-like cancer cells is virtually resistant to the standard treatments causing tumor relapse at the primary or metastatic sites. Understanding the pathways controlling the establishment, expansion and maintenance of the cancer stem cell (CSC) subpopulation is an important step toward the development of more effective treatment for prostate cancer, which might enable ablation or exhaustion of CSCs and prevent treatment resistance and disease recurrence. In this review, we focus on the impact of transcriptional regulators on phenotypic reprogramming of prostate CSCs and provide examples supporting the possibility of inhibiting maintenance and expansion of the CSC pool in human prostate cancer along with the currently available methodological approaches. Transcription factors are key elements for instructing specific transcriptional programs and inducing CSC-associated phenotypic changes implicated in disease progression and treatment resistance. Recent studies have shown that interfering with these processes causes exhaustion of CSCs with loss of self-renewal and tumorigenic capability in prostate cancer models. Targeting key transcriptional regulators in prostate CSCs is a valid therapeutic strategy waiting to be tested in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Carlo V. Catapano
- Institute of Oncology (IOR), Università della Svizzera Italiana, Bellinzona, Switzerland
| |
Collapse
|
33
|
Stelloo S, Bergman AM, Zwart W. Androgen receptor enhancer usage and the chromatin regulatory landscape in human prostate cancers. Endocr Relat Cancer 2019; 26:R267-R285. [PMID: 30865928 DOI: 10.1530/erc-19-0032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022]
Abstract
The androgen receptor (AR) is commonly known as a key transcription factor in prostate cancer development, progression and therapy resistance. Genome-wide chromatin association studies revealed that transcriptional regulation by AR mainly depends on binding to distal regulatory enhancer elements that control gene expression through chromatin looping to gene promoters. Changes in the chromatin epigenetic landscape and DNA sequence can locally alter AR-DNA-binding capacity and consequently impact transcriptional output and disease outcome. The vast majority of reports describing AR chromatin interactions have been limited to cell lines, identifying numerous other factors and interacting transcription factors that impact AR chromatin interactions. Do these factors also impact AR cistromics - the genome-wide chromatin-binding landscape of AR - in vivo? Recent technological advances now enable researchers to identify AR chromatin-binding sites and their target genes in human specimens. In this review, we provide an overview of the different factors that influence AR chromatin binding in prostate cancer specimens, which is complemented with knowledge from cell line studies. Finally, we discuss novel perspectives on studying AR cistromics in clinical samples.
Collapse
Affiliation(s)
- Suzan Stelloo
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andries M Bergman
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
34
|
Attwood SW, Edel MJ. iPS-Cell Technology and the Problem of Genetic Instability-Can It Ever Be Safe for Clinical Use? J Clin Med 2019; 8:E288. [PMID: 30823421 PMCID: PMC6462964 DOI: 10.3390/jcm8030288] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022] Open
Abstract
The use of induced Pluripotent Stem Cells (iPSC) as a source of autologous tissues shows great promise in regenerative medicine. Nevertheless, several major challenges remain to be addressed before iPSC-derived cells can be used in therapy, and experience of their clinical use is extremely limited. In this review, the factors affecting the safe translation of iPSC to the clinic are considered, together with an account of efforts being made to overcome these issues. The review draws upon experiences with pluripotent stem-cell therapeutics, including clinical trials involving human embryonic stem cells and the widely transplanted mesenchymal stem cells. The discussion covers concerns relating to: (i) the reprogramming process; (ii) the detection and removal of incompletely differentiated and pluripotent cells from the resulting medicinal products; and (iii) genomic and epigenetic changes, and the evolutionary and selective processes occurring during culture expansion, associated with production of iPSC-therapeutics. In addition, (iv) methods for the practical culture-at-scale and standardization required for routine clinical use are considered. Finally, (v) the potential of iPSC in the treatment of human disease is evaluated in the light of what is known about the reprogramming process, the behavior of cells in culture, and the performance of iPSC in pre-clinical studies.
Collapse
Affiliation(s)
- Stephen W Attwood
- Department of Life Sciences, The Natural History Museum, London SW7 5BD, UK.
| | - Michael J Edel
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.
- Control of Pluripotency Laboratory, Department of Physiological Sciences I, Faculty of Medicine, University of Barcelona, Hospital Clinic, Casanova 143, 08036 Barcelona, Spain.
- Victor Chang Cardiac Research Institute, Sydney, NSW 2145, Australia.
- Harry Perkins Research Institute, Fiona Stanley Hospital, University of Western Australia, PO Box 404, Bull Creek, Western Australia 6149, Australia.
| |
Collapse
|
35
|
Nicholas TR, Strittmatter BG, Hollenhorst PC. Oncogenic ETS Factors in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:409-436. [PMID: 31900919 DOI: 10.1007/978-3-030-32656-2_18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Prostate cancer is unique among carcinomas in that a fusion gene created by a chromosomal rearrangement is a common driver of the disease. The TMPRSS2/ERG rearrangement drives aberrant expression of the ETS family transcription factor ERG in 50% of prostate tumors. Similar rearrangements promote aberrant expression of the ETS family transcription factors ETV1 and ETV4 in another 10% of cases. Together, these three ETS factors are thought to promote tumorigenesis in the majority of prostate cancers. A goal of precision medicine is to be able to apply targeted therapeutics that are specific to disease subtypes. ETS gene rearrangement positive tumors represent the largest molecular subtype of prostate cancer, but to date there is no treatment specific to this marker. In this chapter we will review the latest findings regarding the molecular mechanisms of ETS factor function in the prostate. These molecular details may provide a path towards new therapeutic targets for this subtype of prostate cancer. Further, we will describe efforts to target the oncogenic functions of ETS family transcription factors directly as well as indirectly.
Collapse
Affiliation(s)
| | - Brady G Strittmatter
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Peter C Hollenhorst
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN, USA.
| |
Collapse
|
36
|
Budka JA, Ferris MW, Capone MJ, Hollenhorst PC. Common ELF1 deletion in prostate cancer bolsters oncogenic ETS function, inhibits senescence and promotes docetaxel resistance. Genes Cancer 2018; 9:198-214. [PMID: 30603056 PMCID: PMC6305106 DOI: 10.18632/genesandcancer.182] [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] [Indexed: 12/14/2022] Open
Abstract
ETS family transcription factors play major roles in prostate tumorigenesis with some acting as oncogenes and others as tumor suppressors. ETS factors can compete for binding at some cis-regulatory sequences, but display specific binding at others. Therefore, changes in expression of ETS family members during tumorigenesis can have complex, multimodal effects. Here we show that ELF1 was the most commonly down-regulated ETS factor in primary prostate tumors, and expression decreased further in metastatic disease. Genome-wide mapping in cell lines indicated that ELF1 has two distinct tumor suppressive roles mediated by distinct cis-regulatory sequences. First, ELF1 inhibited cell migration and epithelial-mesenchymal transition by interfering with oncogenic ETS functions at ETS/AP-1 cis-regulatory motifs. Second, ELF1 uniquely targeted and activated genes that promote senescence. Furthermore, knockdown of ELF1 increased docetaxel resistance, indicating that the genomic deletions found in metastatic prostate tumors may promote therapeutic resistance through loss of both RB1 and ELF1.
Collapse
Affiliation(s)
- Justin A Budka
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana, USA
| | - Mary W Ferris
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana, USA
| | - Matthew J Capone
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana, USA
| | - Peter C Hollenhorst
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana, USA
| |
Collapse
|
37
|
Berglund AE, Rounbehler RJ, Gerke T, Awasthi S, Cheng CH, Takhar M, Davicioni E, Alshalalfa M, Erho N, Klein EA, Freedland SJ, Ross AE, Schaeffer EM, Trock BJ, Den RB, Cleveland JL, Park JY, Dhillon J, Yamoah K. Distinct transcriptional repertoire of the androgen receptor in ETS fusion-negative prostate cancer. Prostate Cancer Prostatic Dis 2018; 22:292-302. [PMID: 30367117 PMCID: PMC6760558 DOI: 10.1038/s41391-018-0103-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/27/2018] [Accepted: 09/08/2018] [Indexed: 12/21/2022]
Abstract
Background Prostate cancer (PCa) tumors harboring translocations of ETS family genes with the androgen responsive TMPRSS2 gene (ETS+ tumors) provide a robust biomarker for detecting PCa in approximately 70% of patients. ETS+ PCa express high levels of the androgen receptor (AR), yet PCa tumors lacking ETS fusions (ETS−) also express AR and demonstrate androgen-regulated growth. In this study, we evaluate the differences in the AR-regulated transcriptomes between ETS+ and ETS− PCa tumors. Methods 10,608 patient tumors from three independent PCa datasets classified as ETS+ (samples overexpressing ERG or other ETS family members) or ETS− (all other PCa) were analyzed for differential gene expression using false-discovery-rate adjusted methods and gene-set enrichment analysis (GSEA). Results Based on the expression of AR-dependent genes and an unsupervised Principal Component Analysis (PCA) model, AR-regulated gene expression alone was able to separate PCa samples into groups based on ETS status in all PCa databases. ETS status distinguished several differentially expressed genes in both TCGA (6.9%) and GRID (6.6%) databases, with 413 genes overlapping in both databases. Importantly, GSEA showed enrichment of distinct androgen-responsive genes in both ETS− and ETS+ tumors, and AR ChIP-seq data identified 131 direct AR-target genes that are regulated in an ETS-specific fashion. Notably, dysregulation of ETS-dependent AR-target genes within the metabolic and non-canonical WNT pathways was associated with clinical outcomes. Conclusions ETS status influences the transcriptional repertoire of the AR, and ETS− PCa tumors appear to rely on distinctly different AR-dependent transcriptional programs to drive and sustain tumorigenesis.
Collapse
Affiliation(s)
- Anders E Berglund
- Department of Biostatistics & Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Robert J Rounbehler
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.,Department of Oncological Sciences, University of South Florida, Tampa, FL, USA
| | - Travis Gerke
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Shivanshu Awasthi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Chia-Ho Cheng
- Department of Biostatistics & Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | | | | | | | | | - Eric A Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Stephen J Freedland
- Department of Surgery, Division of Urology, Center for Integrated Research on Cancer and Lifestyle, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - Bruce J Trock
- Department of Urology, Johns Hopkins, Baltimore, MD, USA
| | - Robert B Den
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - John L Cleveland
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jong Y Park
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jasreman Dhillon
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Kosj Yamoah
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA. .,Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.
| |
Collapse
|
38
|
Cariello M, Ducheix S, Maqdasy S, Baron S, Moschetta A, Lobaccaro JMA. LXRs, SHP, and FXR in Prostate Cancer: Enemies or Ménage à Quatre With AR? NUCLEAR RECEPTOR SIGNALING 2018; 15:1550762918801070. [PMID: 30718981 PMCID: PMC6348739 DOI: 10.1177/1550762918801070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 01/03/2018] [Indexed: 12/11/2022]
Abstract
Androgens and androgen receptor (AR, NR3C4) clearly play a crucial role in
prostate cancer progression. Besides, the link between metabolic disorders and
the risk of developing a prostate cancer has been emerging these last years.
Interestingly, “lipid” nuclear receptors such as LXRα/NR1H3 and LXRβ/NR1H2 (as
well as FXRα/NR1H4 and SHP/NR0B2) have been described to decrease the lipid
metabolism, while AR increases it. Moreover, these former orphan nuclear
receptors can regulate androgen levels and modulate AR activity. Thus, it is not
surprising to find such receptors involved in the physiology of prostate. This
review is focused on the roles of liver X receptors (LXRs), farnesoid X receptor
(FXR), and small heterodimeric partner (SHP) in prostate physiology and their
capabilities to interfere with the androgen-regulated pathways by modulating the
levels of active androgen within the prostate. By the use of prostate cancer
cell lines, mice deficient for these nuclear receptors and human tissue
libraries, several authors have pointed out the putative possibility to
pharmacologically target these receptors. These data open a new field of
research for the development of new drugs that could overcome the castration
resistance in prostate cancer, a usual phenomenon in patients.
Collapse
Affiliation(s)
| | - Simon Ducheix
- Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Salwan Maqdasy
- Université Clermont Auvergne, Clermont-Ferrand, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France.,CHU Clermont-Ferrand, France
| | - Silvère Baron
- Université Clermont Auvergne, Clermont-Ferrand, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Antonio Moschetta
- "Aldo Moro" University of Bari, Italy.,Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy.,IRCCS Istituto Oncologico "Giovanni Paolo II," Bari, Italy
| | - Jean-Marc A Lobaccaro
- "Aldo Moro" University of Bari, Italy.,Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy.,Université Clermont Auvergne, Clermont-Ferrand, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| |
Collapse
|
39
|
Luk IY, Reehorst CM, Mariadason JM. ELF3, ELF5, EHF and SPDEF Transcription Factors in Tissue Homeostasis and Cancer. Molecules 2018; 23:molecules23092191. [PMID: 30200227 PMCID: PMC6225137 DOI: 10.3390/molecules23092191] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 08/23/2018] [Accepted: 08/23/2018] [Indexed: 02/07/2023] Open
Abstract
The epithelium-specific ETS (ESE) transcription factors (ELF3, ELF5, EHF and SPDEF) are defined by their highly conserved ETS DNA binding domain and predominant epithelial-specific expression profile. ESE transcription factors maintain normal cell homeostasis and differentiation of a number of epithelial tissues, and their genetic alteration and deregulated expression has been linked to the progression of several epithelial cancers. Herein we review the normal function of the ESE transcription factors, the mechanisms by which they are dysregulated in cancers, and the current evidence for their role in cancer progression. Finally, we discuss potential therapeutic strategies for targeting or reactivating these factors as a novel means of cancer treatment.
Collapse
Affiliation(s)
- Ian Y Luk
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia.
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria 3086, Australia.
| | - Camilla M Reehorst
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia.
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria 3086, Australia.
| | - John M Mariadason
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia.
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria 3086, Australia.
| |
Collapse
|
40
|
Liu S, Wang X, Qin W, Genchev GZ, Lu H. Transcription Factors Contribute to Differential Expression in Cellular Pathways in Lung Adenocarcinoma and Lung Squamous Cell Carcinoma. Interdiscip Sci 2018; 10:836-847. [PMID: 30039492 DOI: 10.1007/s12539-018-0300-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 06/08/2018] [Accepted: 06/13/2018] [Indexed: 12/25/2022]
Abstract
Lung cancers are broadly classified into small cell lung cancers and non-small cell lung cancers (NSCLC). Lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) are two common subtypes of NSCLC, and despite the fact that both occur in lung tissues, these two subtypes show a number of different pathological characteristics. To investigate the differences and seek potential therapy targets, we used bioinformatics methods to analyze RNA-Seq data from different aspects. The previous studies and comparative pathway enrichment analysis on publicly available data showed that expressed or inhibited genes are different in two cancer subtypes through important pathways. Some of these genes could not only affect cell function through expression, but also could regulate other genes' expression by binding to a specific DNA sequence. This kind of genes is called transcription factor (TF) or sequence-specific DNA-binding factor. Transcription factors play important roles in controlling gene expression in carcinoma pathways. Our results revealed transcription factors that may cause differential expression of genes in cellular pathways of LUAD and LUSC, which provide new clues for study and treatment. Once such TF is NFE2l2 which may regulate genes in the Wnt signaling pathway, and the MAPK signaling pathway, thus leading to an increase the cell growth, cell division, and gene transcription. Another TF-XBP1 has high correlation with genes related to cell adhesion molecules and cytokine-cytokine receptor interaction pathways that may further affect the immune system. Moreover, the two TF and high correlated genes also show similar patterns in an independent GEO data set.
Collapse
Affiliation(s)
- Shiyi Liu
- Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai, China
| | - Xujun Wang
- Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai, China.,SJTU-Yale Joint Center for Biostatistics, Shanghai Jiaotong University, Shanghai, China
| | - Wenyi Qin
- SJTU-Yale Joint Center for Biostatistics, Shanghai Jiaotong University, Shanghai, China.,Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan, Rm 218, Chicago, IL, 60607, USA
| | - Georgi Z Genchev
- SJTU-Yale Joint Center for Biostatistics, Shanghai Jiaotong University, Shanghai, China
| | - Hui Lu
- Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai, China. .,SJTU-Yale Joint Center for Biostatistics, Shanghai Jiaotong University, Shanghai, China. .,Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan, Rm 218, Chicago, IL, 60607, USA.
| |
Collapse
|
41
|
Dual tumor suppressing and promoting function of Notch1 signaling in human prostate cancer. Oncotarget 2018; 7:48011-48026. [PMID: 27384993 PMCID: PMC5216996 DOI: 10.18632/oncotarget.10333] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/12/2016] [Indexed: 12/22/2022] Open
Abstract
Adenocarcinomas of the prostate arise as multifocal heterogeneous lesions as the likely result of genetic and epigenetic alterations and deranged cell-cell communication. Notch signaling is an important form of intercellular communication with a role in growth/differentiation control and tumorigenesis. Contrasting reports exist in the literature on the role of this pathway in prostate cancer (PCa) development. We show here that i) compared to normal prostate tissue, Notch1 expression is significantly reduced in a substantial fraction of human PCas while it is unaffected or even increased in others; ii) acute Notch activation both inhibits and induces process networks associated with prostatic neoplasms; iii) down-modulation of Notch1 expression and activity in immortalized normal prostate epithelial cells increases their proliferation potential, while increased Notch1 activity in PCa cells suppresses growth and tumorigenicity through a Smad3-dependent mechanism involving p21WAF1/CIP1; iv) prostate cancer cells resistant to Notch growth inhibitory effects retain Notch1-induced upregulation of pro-oncogenic genes, like EPAS1 and CXCL6, also overexpressed in human PCas with high Notch1 levels. Taken together, these results reconcile conflicting data on the role of Notch1 in prostate cancer.
Collapse
|
42
|
Tamura RE, Paccez JD, Duncan KC, Morale MG, Simabuco FM, Dillon S, Correa RG, Gu X, Libermann TA, Zerbini LF. GADD45α and γ interaction with CDK11p58 regulates SPDEF protein stability and SPDEF-mediated effects on cancer cell migration. Oncotarget 2017; 7:13865-79. [PMID: 26885618 PMCID: PMC4924684 DOI: 10.18632/oncotarget.7355] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/28/2016] [Indexed: 01/02/2023] Open
Abstract
The epithelium-specific Ets transcription factor, SPDEF, plays a critical role in metastasis of prostate and breast cancer cells. While enhanced SPDEF expression blocks migration and invasion, knockdown of SPDEF expression enhances migration, invasion, and metastasis of cancer cells. SPDEF expression and activation is tightly regulated in cancer cells; however, the precise mechanism of SPDEF regulation has not been explored in detail. In this study we provide evidence that the cell cycle kinase CDK11p58, a protein involved in G2/M transition and degradation of several transcription factors, directly interacts with and phosphorylates SPDEF on serine residues, leading to subsequent ubiquitination and degradation of SPDEF through the proteasome pathway. As a consequence of CDK11p58 mediated degradation of SPDEF, this loss of SPDEF protein results in increased prostate cancer cell migration and invasion. In contrast, knockdown of CDK11p58 protein expression by interfering RNA or SPDEF overexpression inhibit migration and invasion of cancer cells. We demonstrate that CDK11p58 mediated degradation of SPDEF is attenuated by Growth Arrest and DNA damage-inducible 45 (GADD45) α and, two proteins inducing G2/M cell cycle arrest. We show that GADD45 α and γ, directly interact with CDK11p58 and thereby inhibit CDK11p58 activity, and consequentially SPDEF phosphorylation and degradation, ultimately reducing prostate cancer cell migration and invasion. Our findings provide new mechanistic insights into the complex regulation of SPDEF activity linked to cancer metastasis and characterize a previously unidentified SPDEF/CDK11p58/GADD45α/γ pathway that controls SPDEF protein stability and SPDEF-mediated effects on cancer cell migration and invasion.
Collapse
Affiliation(s)
- Rodrigo E Tamura
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Medical Biochemistry Division, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Juliano D Paccez
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Medical Biochemistry Division, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Kristal C Duncan
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Medical Biochemistry Division, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mirian G Morale
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Medical Biochemistry Division, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Fernando M Simabuco
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Simon Dillon
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Ricardo G Correa
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Xuesong Gu
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Towia A Libermann
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Luiz F Zerbini
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Medical Biochemistry Division, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
43
|
Angulo JC, López JI, Ropero S. DNA Methylation and Urological Cancer, a Step Towards Personalized Medicine: Current and Future Prospects. Mol Diagn Ther 2017; 20:531-549. [PMID: 27501813 DOI: 10.1007/s40291-016-0231-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Urologic malignancies are some of the commonest tumors often curable when diagnosed at early stage. However, accurate diagnostic markers and faithful predictors of prognosis are needed to avoid over-diagnosis leading to overtreatment. Many promising exploratory studies have identified epigenetic markers in urinary malignancies based on DNA methylation, histone modification and non-coding ribonucleic acid (ncRNA) expression that epigenetically regulate gene expression. We review and discuss the current state of development and the future potential of epigenetic biomarkers for more accurate and less invasive detection of urological cancer, tumor recurrence and progression of disease serving to establish diagnosis and monitor treatment efficacies. The specific clinical implications of such methylation tests on therapeutic decisions and patient outcome and current limitations are also discussed.
Collapse
Affiliation(s)
- Javier C Angulo
- Servicio de Urología, Hospital Universitario de Getafe, Departamento Clínico, Facultad de Ciencias Biomédicas, Universidad Europea de Madrid, Laureate Universities, Hospital Universitario de Getafe, Carretera de Toledo Km 12.5, Getafe, 28905, Madrid, Spain.
| | - Jose I López
- Servicio de Anatomía Patológica, Hospital Universitario de Cruces, Instituto BioCruces,Universidad del País Vasco (UPV-EHU), Bilbao, Spain
| | - Santiago Ropero
- Departamento de Biología de Sistemas, Unidad Docente de Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
44
|
O'Sullivan AG, Eivers SB, Mulvaney EP, Kinsella BT. Regulated expression of the TPβ isoform of the human T prostanoid receptor by the tumour suppressors FOXP1 and NKX3.1: Implications for the role of thromboxane in prostate cancer. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3153-3169. [PMID: 28890397 DOI: 10.1016/j.bbadis.2017.09.005] [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: 06/02/2017] [Revised: 08/16/2017] [Accepted: 09/07/2017] [Indexed: 12/13/2022]
Abstract
The prostanoid thromboxane (TX)A2 signals through the TPα and TPβ isoforms of T Prostanoid receptor (TP) that are transcriptionally regulated by distinct promoters termed Prm1 and Prm3, respectively, within the TBXA2R gene. We recently demonstrated that expression of TPα and TPβ is increased in PCa, differentially correlating with Gleason grade and with altered CpG methylation of the individual Prm1/Prm3 regions within the TBXA2R. The current study sought to localise the sites of CpG methylation within Prm1 and Prm3, and to identify the main transcription factors regulating TPβ expression through Prm3 in the prostate adenocarcinoma PC-3 and LNCaP cell lines. Bisulfite sequencing revealed extensive differences in the pattern and status of CpG methylation of the individual Prm1 and Prm3 regions that regulate TPα and TPβ expression, respectively, within the TBXA2R. More specifically, Prm1 is predominantly hypomethylated while Prm3 is hypermethylated across its entire sequence in PC-3 and LNCaP cells. Furthermore, the tumour suppressors FOXP1 and NKX3.1, strongly implicated in PCa development, were identified as key transcription factors regulating TPβ expression through Prm3 in both PCa cell lines. Specific siRNA-disruption of FOXP1 and NKX3.1 each coincided with up-regulated TPβ protein and mRNA expression, while genetic-reporter and chromatin immunoprecipitation (ChIP) analyses confirmed that both FOXP1 and NKX3.1 bind to cis‑elements within Prm3 to transcriptionally repress TPβ in the PCa lines. Collectively these data identify Prm3/TPβ as a bona fide target of FOXP1 and NKX3.1 regulation, providing a mechanistic basis, at least in part, for the highly significant upregulation of TPβ expression in PCa.
Collapse
Affiliation(s)
- Aine G O'Sullivan
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sarah B Eivers
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eamon P Mulvaney
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - B Therese Kinsella
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
45
|
Polycomb Repressor Complex 2 in Genomic Instability and Cancer. Int J Mol Sci 2017; 18:ijms18081657. [PMID: 28758948 PMCID: PMC5578047 DOI: 10.3390/ijms18081657] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 07/19/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023] Open
Abstract
Polycomb repressor complexes PRC1 and PRC2 regulate chromatin compaction and gene expression, and are widely recognized for their fundamental contributions to developmental processes. Herein, we summarize the existing evidence and molecular mechanisms linking PRC-mediated epigenetic aberrations to genomic instability and malignancy, with a particular focus on the role of deregulated PRC2 in tumor suppressor gene expression, the DNA damage response, and the fidelity of DNA replication. We also discuss some of the recent advances in the development of pharmacological and dietary interventions affecting PRC2, which point to promising applications for the prevention and management of human malignancies.
Collapse
|
46
|
Fossum SL, Mutolo MJ, Tugores A, Ghosh S, Randell SH, Jones LC, Leir SH, Harris A. Ets homologous factor (EHF) has critical roles in epithelial dysfunction in airway disease. J Biol Chem 2017; 292:10938-10949. [PMID: 28461336 DOI: 10.1074/jbc.m117.775304] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/27/2017] [Indexed: 12/16/2022] Open
Abstract
The airway epithelium forms a barrier between the internal and external environments. Epithelial dysfunction is critical in the pathology of many respiratory diseases, including cystic fibrosis. Ets homologous factor (EHF) is a key member of the transcription factor network that regulates gene expression in the airway epithelium in response to endogenous and exogenous stimuli. EHF, which has altered expression in inflammatory states, maps to the 5' end of an intergenic region on Chr11p13 that is implicated as a modifier of cystic fibrosis airway disease. Here we determine the functions of EHF in primary human bronchial epithelial (HBE) cells and relevant airway cell lines. Using EHF ChIP followed by deep sequencing (ChIP-seq) and RNA sequencing after EHF depletion, we show that EHF targets in HBE cells are enriched for genes involved in inflammation and wound repair. Furthermore, changes in gene expression impact cell phenotype because EHF depletion alters epithelial secretion of a neutrophil chemokine and slows wound closure in HBE cells. EHF activates expression of the SAM pointed domain-containing ETS transcription factor, which contributes to goblet cell hyperplasia. Our data reveal a critical role for EHF in regulating epithelial function in lung disease.
Collapse
Affiliation(s)
- Sara L Fossum
- From the Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, Illinois 60614.,the Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Michael J Mutolo
- From the Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, Illinois 60614.,the Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Antonio Tugores
- the Unidad de Investigación, Complejo Hospitalario Universitario Insular Materno Infantil, 35016 Las Palmas de Gran Canaria, Spain
| | - Sujana Ghosh
- From the Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, Illinois 60614.,the Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Scott H Randell
- the Marsico Lung Institute, University of North Carolina Cystic Fibrosis Center, University of North Carolina, Chapel Hill, North Carolina 27599, and
| | - Lisa C Jones
- the Marsico Lung Institute, University of North Carolina Cystic Fibrosis Center, University of North Carolina, Chapel Hill, North Carolina 27599, and
| | - Shih-Hsing Leir
- From the Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, Illinois 60614.,the Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611.,the Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio 44016
| | - Ann Harris
- From the Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, Illinois 60614, .,the Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611.,the Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio 44016
| |
Collapse
|
47
|
Kumar B, Lupold SE. MicroRNA expression and function in prostate cancer: a review of current knowledge and opportunities for discovery. Asian J Androl 2017; 18:559-67. [PMID: 27056344 PMCID: PMC4955179 DOI: 10.4103/1008-682x.177839] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are well-conserved noncoding RNAs that broadly regulate gene expression through posttranscriptional silencing of coding genes. Dysregulated miRNA expression in prostate and other cancers implicates their role in cancer biology. Moreover, functional studies provide support for the contribution of miRNAs to several key pathways in cancer initiation and progression. Comparative analyses of miRNA gene expression between malignant and nonmalignant prostate tissues, healthy controls and prostate cancer (PCa) patients, as well as less aggressive versus more aggressive disease indicate that miRNAs may be future diagnostic or prognostic biomarkers in tumor tissue, blood, or urine. Further, miRNAs may be future therapeutics or therapeutic targets. In this review, we examine the miRNAs most commonly observed to be de-regulated in PCa gene expression analyses and review the potential contribution of these miRNAs to important pathways in PCa initiation and progression.
Collapse
Affiliation(s)
- Binod Kumar
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Shawn E Lupold
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
48
|
Archer LK, Frame FM, Maitland NJ. Stem cells and the role of ETS transcription factors in the differentiation hierarchy of normal and malignant prostate epithelium. J Steroid Biochem Mol Biol 2017; 166:68-83. [PMID: 27185499 DOI: 10.1016/j.jsbmb.2016.05.006] [Citation(s) in RCA: 11] [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: 02/19/2016] [Revised: 04/25/2016] [Accepted: 05/07/2016] [Indexed: 12/18/2022]
Abstract
Prostate cancer is the most common cancer of men in the UK and accounts for a quarter of all new cases. Although treatment of localised cancer can be successful, there is no cure for patients presenting with invasive prostate cancer and there are less treatment options. They are generally treated with androgen-ablation therapies but eventually the tumours become hormone resistant and patients develop castration-resistant prostate cancer (CRPC) for which there are no further successful or curative treatments. This highlights the need for new treatment strategies. In order to prevent prostate cancer recurrence and treatment resistance, all the cell populations in a heterogeneous prostate tumour must be targeted, including the rare cancer stem cell (CSC) population. The ETS transcription factor family members are now recognised as a common feature in multiple cancers including prostate cancer; with aberrant expression, loss of tumour suppressor function, inactivating mutations and the formation of fusion genes observed. Most notably, the TMPRSS2-ERG gene fusion is present in approximately 50% of prostate cancers and in prostate CSCs. However, the role of other ETS transcription factors in prostate cancer is less well understood. This review will describe the prostate epithelial cell hierarchy and discuss the evidence behind prostate CSCs and their inherent resistance to conventional cancer therapies. The known and proposed roles of the ETS family of transcription factors in prostate epithelial cell differentiation and regulation of the CSC phenotype will be discussed, as well as how they might be targeted for therapy.
Collapse
Affiliation(s)
- Leanne K Archer
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, United Kingdom
| | - Fiona M Frame
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, United Kingdom
| | - Norman J Maitland
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, United Kingdom.
| |
Collapse
|
49
|
Lu H, Li G, Zhou C, Jin W, Qian X, Wang Z, Pan H, Jin H, Wang X. Regulation and role of post-translational modifications of enhancer of zeste homologue 2 in cancer development. Am J Cancer Res 2016; 6:2737-2754. [PMID: 28042497 PMCID: PMC5199751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 08/09/2016] [Indexed: 06/06/2023] Open
Abstract
Post-translational modifications (PTMs) are critical molecular events which alter protein conformation after their synthesis and diversity protein properties by modulating their stability, localization, interacting partners or the activity of their substrates, consequently exerting pivotal roles in regulating the functions of many important eukaryotic proteins. It has been well acknowledged that PTMs are of great importance in a broad range of biological processes such as gene regulation, cell proliferation, differentiation and apoptosis, tissue development, diseases, tumor progression and drug resistance. As the core and contributing catalytic subunit of Polycomb repressive complex 2(PRC2), Enhancer of zeste homolog 2 (EZH2) is a master epigenetic regulator, often serving as a highly conserved histone methyltransferase (HMTase) to induce histone H3 lysine 27 trimethylation (H3K27me3) and repress gene transcription and expression. Dysregulated EZH2 expression is frequently associated with cancer development and poor prognosis in a wide variety of cancers. Considered its essential role in carcinogenesis, EZH2 is a potential candidate for cancer targeted therapy. Remarkably, mounting evidence highlights that EZH2 expression, activity and stability can be regulated by PTMs including phosphorylation, acetylation, ubiquitination, sumoylation and GlcNAcylation aside from its well-validated modifications in transcriptional and post-transcriptional levels. However, the precise regulatory mechanisms underlying EZH2 PTMs and whether other types of PTMs orchestrate in EZH2 remain largely unclear. In this review, we summarize current advances in the understanding of EZH2 regulation by PTMs and their associated biological functions during tumorigenesis.
Collapse
Affiliation(s)
- Haiqi Lu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
- Laboratory of Cancer Biology, Provincial Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Guangliang Li
- Department of Medical Oncology, Zhejiang Cancer HospitalHangzhou, Zhejiang, China
| | - Chenyi Zhou
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Wei Jin
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Xiaoling Qian
- Laboratory of Cancer Biology, Provincial Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Zhuo Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
- Laboratory of Cancer Biology, Provincial Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Provincial Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| |
Collapse
|
50
|
Zhang YY, Huang SH, Zhou HR, Chen CJ, Tian LH, Shen JZ. Role of HOTAIR in the diagnosis and prognosis of acute leukemia. Oncol Rep 2016; 36:3113-3122. [PMID: 27748863 PMCID: PMC5112607 DOI: 10.3892/or.2016.5147] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/16/2016] [Indexed: 12/22/2022] Open
Abstract
HOX antisense intergenic RNA (HOTAIR), a long non-coding RNA, plays an important role in the development of many types of cancers. Its function in acute leukemia (AL), however, has not been examined. The present study investigated the role of HOTAIR and its downstream genes in AL, and determined whether it could act as a molecular marker for prediction of leukemia development and prognosis. Real-time quantitative PCR was used to examine the expression of each gene in the HOTAIR signaling pathway in AL patients. The relationship between expression of HOTAIR and downstream genes and AL prognosis was analyzed. Expression of HOTAIR in patients with acute monocytic leukemia (M5) was increased as compared to controls (P<0.05). Compared to patients with low HOTAIR expression, overall survival and event-free survival of patients with high HOTAIR expression was significantly reduced. In addition, the expression of downstream genes in the HOTAIR signaling pathway including EZH2, LSD1, DNMT3A and DNMT3B was significantly increased in AL patients, and showed a significant positive correlation with high expression of HOTAIR (P<0.05). In conclusion, HOTAIR was closely related with a poor prognosis in AL patients. It may be involved in the development of leukemia by mediating methylation of DNA and histones.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- DNA Methyltransferase 3A
- Female
- Gene Expression
- Humans
- Kaplan-Meier Estimate
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Male
- Middle Aged
- Multivariate Analysis
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality
- Prognosis
- Proportional Hazards Models
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Signal Transduction
- Tumor Cells, Cultured
- Young Adult
- DNA Methyltransferase 3B
Collapse
Affiliation(s)
- Yuan-Yuan Zhang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Si-Han Huang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Hua-Rong Zhou
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Cong-Jie Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Li-Hong Tian
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jian-Zhen Shen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|