1
|
Goode EC, Fachal L, Panousis N, Moutsianas L, McIntyre RE, Bai BYH, Kawasaki N, Wittmann A, Raine T, Rushbrook SM, Anderson CA. Fine-mapping and molecular characterisation of primary sclerosing cholangitis genetic risk loci. Nat Commun 2024; 15:9594. [PMID: 39505854 PMCID: PMC11541731 DOI: 10.1038/s41467-024-53602-w] [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: 08/22/2023] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
Genome-wide association studies of primary sclerosing cholangitis have identified 23 susceptibility loci. The majority of these loci reside in non-coding regions of the genome and are thought to exert their effect by perturbing the regulation of nearby genes. Here, we aim to identify these genes to improve the biological understanding of primary sclerosing cholangitis, and nominate potential drug targets. We first build an eQTL map for six primary sclerosing cholangitis-relevant T-cell subsets obtained from the peripheral blood of primary sclerosing cholangitis and ulcerative colitis patients. These maps identify 10,459 unique eGenes, 87% of which are shared across all six primary sclerosing cholangitis T-cell types. We then search for colocalisations between primary sclerosing cholangitis loci and eQTLs and undertake Bayesian fine-mapping to identify disease-causing variants. In this work, colocalisation analyses nominate likely primary sclerosing cholangitis effector genes and biological mechanisms at five non-coding (UBASH3A, PRKD2, ETS2 and AP003774.1/CCDC88B) and one coding (SH2B3) primary sclerosing cholangitis loci. Through fine-mapping we identify likely causal variants for a third of all primary sclerosing cholangitis-associated loci, including two to single variant resolution.
Collapse
Affiliation(s)
- Elizabeth C Goode
- Wellcome Sanger Institute, Hinxton, Cambridge, UK
- University of Cambridge, Cambridge, UK
- Norfolk and Norwich University Hospital, Norwich, UK
| | - Laura Fachal
- Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | | | | | | | - Benjamin Yu Hang Bai
- Wellcome Sanger Institute, Hinxton, Cambridge, UK
- University of Cambridge, Cambridge, UK
| | | | | | - Tim Raine
- University of Cambridge, Cambridge, UK
| | - Simon M Rushbrook
- Norfolk and Norwich University Hospital, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | |
Collapse
|
2
|
Hurtado-Lorenzo A, Swantek JL. The landscape of new therapeutic opportunities for IBD. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:1-83. [PMID: 39521596 DOI: 10.1016/bs.apha.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
This chapter presents an overview of the emerging strategies to address the unmet needs in the management of inflammatory bowel diseases (IBD). IBD poses significant challenges, as over half of patients experience disease progression despite interventions, leading to irreversible complications, and a substantial proportion do not respond to existing therapies, such as biologics. To overcome these limitations, we describe a diverse array of novel therapeutic approaches. In the area of immune homeostasis restoration, the focus is on targeting cytokine networks, leukocyte trafficking, novel immune pathways, and cell therapies involving regulatory T cells and mesenchymal stem cells (MSC). Recognizing the critical role of impaired intestinal barrier integrity in IBD, we highlight therapies aimed at restoring barrier function and promoting mucosal healing, such as those targeting cell proliferation, tight junctions, and lipid mediators. Addressing the challenges posed by fibrosis and fistulas, we describe emerging targets for reversing fibrosis like kinase and cytokine inhibitors and nuclear receptor agonists, as well as the potential of MSC for fistulas. The restoration of a healthy gut microbiome, through strategies like fecal microbiota transplantation, rationally defined bacterial consortia, and targeted antimicrobials, is also highlighted. We also describe innovative approaches to gut-targeted drug delivery to enhance efficacy and minimize side effects. Reinforcing these advancements is the critical role of precision medicine, which emphasizes the use of multiomics analysis for the discovery of biomarkers to enable personalized IBD care. Overall, the emerging landscape of therapeutic opportunities for IBD holds great potential to surpass the therapeutic ceiling of current treatments.
Collapse
Affiliation(s)
- Andrés Hurtado-Lorenzo
- Translational Research & IBD Ventures, Research Department, Crohn's & Colitis Foundation, New York, NY, United States.
| | - Jennifer L Swantek
- Translational Research & IBD Ventures, Research Department, Crohn's & Colitis Foundation, New York, NY, United States
| |
Collapse
|
3
|
Chen HB, Li W, Yang Z, Liu KL, Lu BS, Wang ZY. ERK/MAPK Signalling Pathway Regulates MMP2 through ETS1 in Renal Clear Cell Carcinoma. Curr Mol Med 2024; 24:780-789. [PMID: 37254537 DOI: 10.2174/1566524023666230529143837] [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: 12/21/2022] [Revised: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND The c-ETS-1 (ETS1) expression is high in clear cell renal cell carcinoma (ccRCC) tissues; however, how it impacts ccRCC is currently unknown. METHODS The online STRING web source was used to construct a protein network interacting with ETS1. The Cell Counting Kit-8 was used to detect the cell viability. A clonogenic assay, a wound-healing assay, and a Transwell assay were used to detect cell proliferation, invasion and migration abilities. Western blot was used to detect the expression of proteins. RESULTS The data showed the expression of ETS1 in ccRCC tissues to be significantly increased compared to adjacent tissues (p<0.05). The positive expression of ETS1 in ccRCC patients aged 20-100 was statistically significant compared to adjacent normal tissues (p<0.05). The grade of ETS1 positive expression (1-4) and lymph node metastasis (N1) in ccRCC were significantly higher than those in adjacent normal tissues (p<0.05). The tumour stage (stages 1-4) in ccRCC patients with positive ETS1 expression was significantly higher than that in adjacent normal tissues (p<0.05). Knockdown of ETS1 and PERK inhibitors significantly inhibited the proliferation, migration and invasion of ccRCC cells. Knockdown of ETS1 inhibited MMP-2 expression, and an extracellular signal-related kinase (ERK) inhibitor inhibited both ETS1 and MMP-2 expression. CONCLUSION A high expression of ETS1 is associated with the progression of ccRCC. This study suggests that ETS1 promotes proliferation by increasing MMP2 expression in ccRCC, and combined knockdown of ETS1 and inhibition of ERK can significantly inhibit the proliferation, migration and invasion of ccRCC. ETS1 may be a therapeutic and prognostic target for renal cell carcinoma.
Collapse
Affiliation(s)
- Hai-Bin Chen
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, 050061, China
- Department of Urology, Affiliated Hospital of Hebei Engineering University, Handan, 056000, China
| | - Wei Li
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, 050061, China
| | - Zhan Yang
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, 050061, China
| | - Kai-Long Liu
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, 050061, China
| | - Bao-Sai Lu
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, 050061, China
| | - Zi-Yi Wang
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, 050061, China
| |
Collapse
|
4
|
Wang Y, Huang Z, Sun M, Huang W, Xia L. ETS transcription factors: Multifaceted players from cancer progression to tumor immunity. Biochim Biophys Acta Rev Cancer 2023; 1878:188872. [PMID: 36841365 DOI: 10.1016/j.bbcan.2023.188872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/18/2023] [Accepted: 01/28/2023] [Indexed: 02/26/2023]
Abstract
The E26 transformation specific (ETS) family comprises 28 transcription factors, the majority of which are involved in tumor initiation and development. Serving as a group of functionally heterogeneous gene regulators, ETS factors possess a structurally conserved DNA-binding domain. As one of the most prominent families of transcription factors that control diverse cellular functions, ETS activation is modulated by multiple intracellular signaling pathways and post-translational modifications. Disturbances in ETS activity often lead to abnormal changes in oncogenicity, including cancer cell survival, growth, proliferation, metastasis, genetic instability, cell metabolism, and tumor immunity. This review systematically addresses the basics and advances in studying ETS factors, from their tumor relevance to clinical translational utility, with a particular focus on elucidating the role of ETS family in tumor immunity, aiming to decipher the vital role and clinical potential of regulation of ETS factors in the cancer field.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zhao Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| |
Collapse
|
5
|
Li Y, Wu T, Peng Z, Tian X, Dai Q, Chen M, Zhu J, Xia S, Sun A, Yang W, Lin Q. ETS1 is a prognostic biomarker of triple-negative breast cancer and promotes the triple-negative breast cancer progression through the YAP signaling. Am J Cancer Res 2022; 12:5074-5084. [PMID: 36504910 PMCID: PMC9729895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 10/26/2022] [Indexed: 12/15/2022] Open
Abstract
E26 transcription factor-1 (ETS1) is involved in extracellular matrix remodeling, migratory infiltration and angiogenesis in tumors and known to play an important role in tumor progression. However, the mechanism by which ETS1 promotes tumor progression remains elusive. In this report, we show that ETS1 is highly expressed in breast tumor tissues and specifically associated with the tumor metastasis and poor survival in triple negative breast cancer (TNBC) tumors, upon analysis by immunohistochemical (IHC) staining of tumor samples from 240 breast cancer cases. Depletion of ETS1 in TNBC cells by shETS1 significantly inhibited the cell proliferation and migration. Mechanistically, knockdown of ETS1 in TNBC cells dramatically reduced expression of YAP and the YAP target genes, and overexpression of YAP in the ETS1 knockdown cells restored the cell proliferation and migration. These data indicate that YAP is a downstream effector mediating the ETS1-promoted TNBC cell proliferation and migration. Taken together, our results suggest that ETS1 promotes TNBC progression through the YAP signaling.
Collapse
Affiliation(s)
- Yanlin Li
- School of Medicine, Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Tiantian Wu
- School of Medicine, Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Ziluo Peng
- School of Medicine, Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Xianyan Tian
- School of Medicine, Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Qian Dai
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
| | - Miao Chen
- School of Medicine, Jiangsu UniversityZhenjiang, Jiangsu, China,The First People’s Hospital of ZhenjiangZhenjiang, Jiangsu, China
| | - Jun Zhu
- School of Medicine, Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Song Xia
- School of Medicine, Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Aiqin Sun
- School of Medicine, Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Wannian Yang
- School of Medicine, Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Qiong Lin
- School of Medicine, Jiangsu UniversityZhenjiang, Jiangsu, China
| |
Collapse
|
6
|
Galasso M, Dalla Pozza E, Chignola R, Gambino S, Cavallini C, Quaglia FM, Lovato O, Dando I, Malpeli G, Krampera M, Donadelli M, Romanelli MG, Scupoli MT. The rs1001179 SNP and CpG methylation regulate catalase expression in chronic lymphocytic leukemia. Cell Mol Life Sci 2022; 79:521. [PMID: 36112236 PMCID: PMC9481481 DOI: 10.1007/s00018-022-04540-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/23/2022] [Accepted: 08/28/2022] [Indexed: 11/26/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is an incurable disease characterized by an extremely variable clinical course. We have recently shown that high catalase (CAT) expression identifies patients with an aggressive clinical course. Elucidating mechanisms regulating CAT expression in CLL is preeminent to understand disease mechanisms and develop strategies for improving its clinical management. In this study, we investigated the role of the CAT promoter rs1001179 single nucleotide polymorphism (SNP) and of the CpG Island II methylation encompassing this SNP in the regulation of CAT expression in CLL. Leukemic cells harboring the rs1001179 SNP T allele exhibited a significantly higher CAT expression compared with cells bearing the CC genotype. CAT promoter harboring the T -but not C- allele was accessible to ETS-1 and GR-β transcription factors. Moreover, CLL cells exhibited lower methylation levels than normal B cells, in line with the higher CAT mRNA and protein expressed by CLL in comparison with normal B cells. Methylation levels at specific CpG sites negatively correlated with CAT levels in CLL cells. Inhibition of methyltransferase activity induced a significant increase in CAT levels, thus functionally validating the role of CpG methylation in regulating CAT expression in CLL. Finally, the CT/TT genotypes were associated with lower methylation and higher CAT levels, suggesting that the rs1001179 T allele and CpG methylation may interact in regulating CAT expression in CLL. This study identifies genetic and epigenetic mechanisms underlying differential expression of CAT, which could be of crucial relevance for the development of therapies targeting redox regulatory pathways in CLL.
Collapse
Affiliation(s)
- Marilisa Galasso
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy
- Section of Hematology, Department of Medicine, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Elisa Dalla Pozza
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy
| | - Roberto Chignola
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134, Verona, Italy
| | - Simona Gambino
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy
| | - Chiara Cavallini
- Research Center LURM, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Francesca Maria Quaglia
- Section of Hematology, Department of Medicine, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Ornella Lovato
- Research Center LURM, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Ilaria Dando
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy
| | - Giorgio Malpeli
- Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Mauro Krampera
- Section of Hematology, Department of Medicine, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy
| | - Massimo Donadelli
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy
| | - Maria G Romanelli
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy.
| | - Maria T Scupoli
- Biology and Genetics Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy.
- Research Center LURM, University of Verona, Policlinico G.B. Rossi, P. L.A. Scuro 10, 37134, Verona, Italy.
| |
Collapse
|
7
|
Vishnoi K, Ke R, Viswakarma N, Srivastava P, Kumar S, Das S, Singh SK, Principe DR, Rana A, Rana B. Ets1 mediates sorafenib resistance by regulating mitochondrial ROS pathway in hepatocellular carcinoma. Cell Death Dis 2022; 13:581. [PMID: 35789155 PMCID: PMC9253325 DOI: 10.1038/s41419-022-05022-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 01/21/2023]
Abstract
The incidence and mortality of hepatocellular carcinoma (HCC) are on a rise in the Western countries including US, attributed mostly to late detection. Sorafenib has been the first-line FDA-approved drug for advanced unresectable HCC for almost a decade, but with limited efficacy due to the development of resistance. More recently, several other multi-kinase inhibitors (lenvatinib, cabozantinib, regorafenib), human monoclonal antibody (ramucirumab), and immune checkpoint inhibitors (nivolumab, pembrolizumab) have been approved as systemic therapies. Despite this, the median survival of patients is not significantly increased. Understanding of the molecular mechanism(s) that govern HCC resistance is critically needed to increase efficacy of current drugs and to develop more efficacious ones in the future. Our studies with sorafenib-resistant (soraR) HCC cells using transcription factor RT2 Profiler PCR Arrays revealed an increase in E26 transformation-specific-1 (Ets-1) transcription factor in all soraR cells. HCC TMA studies showed an increase in Ets-1 expression in advanced HCC compared to the normal livers. Overexpression or knocking down Ets-1 modulated sorafenib resistance-related epithelial-mesenchymal transition (EMT), migration, and cell survival. In addition, the soraR cells showed a significant reduction of mitochondrial damage and mitochondrial reactive oxygen species (mROS) generation, which were antagonized by knocking down Ets-1 expression. More in-depth analysis identified GPX-2 as a downstream mediator of Ets-1-induced sorafenib resistance, which was down-regulated by Ets-1 knockdown while other antioxidant pathway genes were not affected. Interestingly, knocking down GPX2 expression significantly increased sorafenib sensitivity in the soraR cells. Our studies indicate the activation of a novel Ets-1-GPX2 signaling axis in soraR cells, targeting which might successfully antagonize resistance and increase efficacy.
Collapse
Affiliation(s)
- Kanchan Vishnoi
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Rong Ke
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Navin Viswakarma
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Piush Srivastava
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Sandeep Kumar
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Subhasis Das
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.185648.60000 0001 2175 0319University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Sunil Kumar Singh
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Daniel R. Principe
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Ajay Rana
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.185648.60000 0001 2175 0319University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.280892.90000 0004 0419 4711Jesse Brown VA Medical Center, Chicago, IL 60612 USA
| | - Basabi Rana
- grid.185648.60000 0001 2175 0319Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.185648.60000 0001 2175 0319University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612 USA ,grid.280892.90000 0004 0419 4711Jesse Brown VA Medical Center, Chicago, IL 60612 USA
| |
Collapse
|
8
|
Ning XH, Li NY, Qi YY, Li SC, Jia ZK, Yang JJ. Identification of a Hypoxia-Related Gene Model for Predicting the Prognosis and Formulating the Treatment Strategies in Kidney Renal Clear Cell Carcinoma. Front Oncol 2022; 11:806264. [PMID: 35141153 PMCID: PMC8818738 DOI: 10.3389/fonc.2021.806264] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/23/2021] [Indexed: 12/18/2022] Open
Abstract
Purpose The present study aimed to establish a hypoxia related genes model to predict the prognosis of kidney clear cell carcinoma (KIRC) patients using data accessed from The Cancer Genome Atlas (TCGA) database and International Cancer Genome Consortium (ICGC) database. Methods Patients’ data were downloaded from the TCGA and ICGC databases, and hypoxia related genes were accessed from the Molecular Signatures Database. The differentially expressed genes were evaluated and then the differential expressions hypoxia genes were screened. The TCGA cohort was randomly divided into a discovery TCGA cohort and a validation TCGA cohort. The discovery TCGA cohort was used for constructing the hypoxia genes risk model through Lasso regression, univariate and multivariate Cox regression analysis. Receiver operating characteristic (ROC) curves were used to assess the reliability and sensitivity of our model. Then, we established a nomogram to predict the probable one-, three-, and five-year overall survival rates. Lastly, the Tumor Immune Dysfunction and Exclusion (TIDE) score of patients was calculated. Results We established a six hypoxia-related gene prognostic model of KIRC patients in the TCGA database and validated in the ICGC database. The patients with high riskscore present poorer prognosis than those with low riskscore in the three TCGA cohorts and ICGC cohort. ROC curves show our six-gene model with a robust predictive capability in these four cohorts. In addition, we constructed a nomogram for KIRC patients in the TCGA database. Finally, the high risk-group had a high TIDE score than the patients with low riskscore. Conclusions We established a six hypoxia-related gene risk model for independent prediction of the prognosis of KIRC patients was established and constructed a robust nomogram. The different riskscores might be a biomarker for immunotherapy strategy.
Collapse
Affiliation(s)
- Xiang-hui Ning
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Xiang-hui Ning, ; Jin-jian Yang,
| | - Ning-yang Li
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuan-yuan Qi
- Department of Nephrology, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Song-chao Li
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhan-kui Jia
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin-jian Yang
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Xiang-hui Ning, ; Jin-jian Yang,
| |
Collapse
|
9
|
Ma P, Jin X, Fan Z, Wang Z, Yue S, Wu C, Chen S, Wu Y, Chen M, Gu D, Zhang S, Mao R, Fan Y. Super-enhancer receives signals from the extracellular matrix to induce PD-L1-mediated immune evasion via integrin/BRAF/TAK1/ERK/ETV4 signaling. Cancer Biol Med 2021; 19:j.issn.2095-3941.2021.0137. [PMID: 34623791 PMCID: PMC9196059 DOI: 10.20892/j.issn.2095-3941.2021.0137] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE PD-L1 and PD-L2 expression levels determine immune evasion and the therapeutic efficacy of immune checkpoint blockade. The factors that drive inducible PD-L1 expression have been extensively studied, but mechanisms that result in constitutive PD-L1 expression in cancer cells are largely unknown. METHODS DNA elements were deleted in cells by CRISPR/Cas9-mediated knockout. Protein function was inhibited by chemical inhibitors. Protein levels were examined by Western blot, mRNA levels were examined by real-time RT-PCR, and surface protein expression was determined by cellular immunofluorescence and flow cytometry. Immune evasion was examined by in vitro T cell-mediated killing. RESULTS We determined the core regions (chr9: 5, 496, 378-5, 499, 663) of a previously identified PD-L1L2-super-enhancer (SE). Through systematic analysis, we found that the E26 transformation-specific (ETS) variant transcription factor (ETV4) bound to this core DNA region but not to DNA surrounding PD-L1L2SE. Genetic knockout of ETV4 dramatically reduced the expressions of both PD-L1 and PD-L2. ETV4 transcription was dependent on ERK activation, and BRAF/TAK1-induced ERK activation was dependent on extracellular signaling from αvβ3 integrin, which profoundly affected ETV4 transcription and PD-L1/L2 expression. Genetic silencing or pharmacological inhibition of components of the PD-L1L2-SE-associated pathway rendered cancer cells susceptible to T cell-mediated killing. CONCLUSIONS We identified a pathway originating from the extracellular matrix that signaled via integrin/BRAF/TAK1/ERK/ETV4 to PD-L1L2-SE to induce PD-L1-mediated immune evasion. These results provided new insights into PD-L1L2-SE activation and pathways associated with immune checkpoint regulation in cancer.
Collapse
Affiliation(s)
- Panpan Ma
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong 226001, China
- Department of Clinical Laboratory, Yancheng No. 1 People's Hospital, Yancheng 224005, China
| | - Xinxin Jin
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Zhiwei Fan
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Zhou Wang
- School of Life Sciences, Nantong University, Nantong 226001, China
| | - Suhui Yue
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Changyue Wu
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Shiyin Chen
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Yuanyuan Wu
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Miaomiao Chen
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Donghua Gu
- The Department of Urology, the Second Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Siliang Zhang
- The Department of Radiotherapy Oncology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong 226001, China
| | - Yihui Fan
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong 226001, China
| |
Collapse
|
10
|
Chen K, Gorgen A, Ding A, Du L, Jiang K, Ding Y, Sapisochin G, Ghanekar A. Dual-Specificity Phosphatase 9 Regulates Cellular Proliferation and Predicts Recurrence After Surgery in Hepatocellular Carcinoma. Hepatol Commun 2021; 5:1310-1328. [PMID: 34278178 PMCID: PMC8279460 DOI: 10.1002/hep4.1701] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/19/2021] [Accepted: 02/07/2021] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (CC) is a common and deadly cancer with complex molecular pathogenesis. Little is known about dual-specificity phosphatases (DUSPs) in HCC. We investigated DUSP9 expression in human HCC, associations between DUSP9 and patient outcomes, and effects of altered DUSP9 expression on HCC biology. We studied public data sets as well as 196 patients at our institution who had HCC resections. Quantitative real-time reverse transcription polymerase chain reaction and western blot demonstrated that DUSP9 expression was increased >10-fold in HCC compared to adjacent liver and healthy controls (P = 0.005). Kaplan-Meier and multivariable regression analyses revealed that higher DUSP9 expression was associated with shorter disease-free survival (high DUSP9, 1.6; 95% confidence interval, 0.9-2.3 vs. low DUSP9, 3.4; 95% confidence interval, 1.8-5.0 years; P = 0.04) and increased risk of recurrence (hazard ratio 1.55; 95% confidence interval, 1.01-2.67; P = 0.05) after resection. DUSP9 complementary DNA (cDNA) was cloned using rapid amplification of cDNA ends, revealing two DUSP9 isoforms in human HCC cells. Studies of transcriptional regulation using promoter-luciferase reporter constructs suggested that DUSP9 transcription is regulated by E26 transformation-specific transcription factors. Proliferation of hepatic cells in vitro was enhanced by lentiviral-mediated overexpression of DUSP9. In contrast, DUSP9 knockout HCC cells generated using clustered regularly interspaced short palindromic repeats (CRISPR) demonstrated decreased HCC proliferation and doxorubicin resistance in vitro and impaired xenograft growth in vivo. RNA sequencing, gene set enrichment, and network/pathway analysis revealed that DUSP9 knockout is associated with activation of protein kinase activity and apoptosis. Conclusion: DUSP9 regulates cell proliferation and predicts recurrence after surgery in HCC. DUSP9 may represent a novel prognostic candidate and therapeutic target. Additional studies are warranted to further explore the role and regulation of DUSP9 in HCC.
Collapse
Affiliation(s)
- Kui Chen
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
| | - Andre Gorgen
- Division of General SurgeryUniversity Health NetworkTorontoONCanada
- Department of SurgeryUniversity of TorontoTorontoONCanada
| | - Avrilynn Ding
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
| | - Lulu Du
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
| | - Keruo Jiang
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
| | - Yu Ding
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
| | - Gonzalo Sapisochin
- Division of General SurgeryUniversity Health NetworkTorontoONCanada
- Department of SurgeryUniversity of TorontoTorontoONCanada
| | - Anand Ghanekar
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoONCanada
- Division of General SurgeryUniversity Health NetworkTorontoONCanada
- Department of SurgeryUniversity of TorontoTorontoONCanada
| |
Collapse
|
11
|
Sivasudhan E, Blake N, Lu ZL, Meng J, Rong R. Dynamics of m6A RNA Methylome on the Hallmarks of Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:642443. [PMID: 33869193 PMCID: PMC8047153 DOI: 10.3389/fcell.2021.642443] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
Epidemiological data consistently rank hepatocellular carcinoma (HCC) as one of the leading causes of cancer-related deaths worldwide, often posing severe economic burden on health care. While the molecular etiopathogenesis associated with genetic and epigenetic modifications has been extensively explored, the biological influence of the emerging field of epitranscriptomics and its associated aberrant RNA modifications on tumorigenesis is a largely unexplored territory with immense potential for discovering new therapeutic approaches. In particular, the underlying cellular mechanisms of different hallmarks of hepatocarcinogenesis that are governed by the complex dynamics of m6A RNA methylation demand further investigation. In this review, we reveal the up-to-date knowledge on the mechanistic and functional link between m6A RNA methylation and pathogenesis of HCC.
Collapse
Affiliation(s)
- Enakshi Sivasudhan
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Neil Blake
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Zhi-Liang Lu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Jia Meng
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Rong Rong
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China.,Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
12
|
Dinhof C, Pirker C, Kroiss P, Kirchhofer D, Gabler L, Gojo J, Lötsch-Gojo D, Stojanovic M, Timelthaler G, Ferk F, Knasmüller S, Reisecker J, Spiegl-Kreinecker S, Birner P, Preusser M, Berger W. p53 Loss Mediates Hypersensitivity to ETS Transcription Factor Inhibition Based on PARylation-Mediated Cell Death Induction. Cancers (Basel) 2020; 12:cancers12113205. [PMID: 33143299 PMCID: PMC7693367 DOI: 10.3390/cancers12113205] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/13/2020] [Accepted: 10/26/2020] [Indexed: 01/31/2023] Open
Abstract
Simple Summary ETS transcription factors are potent oncogenic drivers in several cancer types and represent promising therapeutic targets. However, molecular factors influencing response to ETS factor inhibition are widely unknown so far. Here, we uncover that sensitivity of cancer cells against ETS factor blockade by the small molecule inhibitor YK-4-279 is strongly promoted by p53 loss in a MAPK-driven background. Induction of a parthanatos-like cell death based on a deregulated MAPK/ETS1/p53/PARP1 signal axis is identified as underlying molecular mechanism. Hence, this study suggests a novel and biomarker-driven therapeutic strategy for p53-deleted tumours, generally known for their profound therapy resistance. Abstract The small-molecule E26 transformation-specific (ETS) factor inhibitor YK-4-279 was developed for therapy of ETS/EWS fusion-driven Ewing’s sarcoma. Here we aimed to identify molecular factors underlying YK-4-279 responsiveness in ETS fusion-negative cancers. Cell viability screenings that deletion of P53 induced hypersensitization against YK-4-279 especially in the BRAFV600E-mutated colon cancer model RKO. This effect was comparably minor in the BRAF wild-type HCT116 colon cancer model. Out of all ETS transcription factor family members, especially ETS1 overexpression at mRNA and protein level was induced by deletion of P53 specifically under BRAF-mutated conditions. Exposure to YK-4-279 reverted ETS1 upregulation induced by P53 knock-out in RKO cells. Despite upregulation of p53 by YK-4-279 itself in RKOp53 wild-type cells, YK-4-279-mediated hyperphosphorylation of histone histone H2A.x was distinctly more pronounced in the P53 knock-out background. YK-4-279-induced cell death in RKOp53-knock-out cells involved hyperPARylation of PARP1, translocation of the apoptosis-inducible factor AIF into nuclei, and induction of mitochondrial membrane depolarization, all hallmarks of parthanatos. Accordingly, pharmacological PARP as well as BRAFV600E inhibition showed antagonistic activity with YK-4-279 especially in the P53 knock-out background. Taken together, we identified ETS factor inhibition as a promising strategy for the treatment of notoriously therapy-resistant p53-null solid tumours with activating MAPK mutations.
Collapse
Affiliation(s)
- Carina Dinhof
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria;
| | - Christine Pirker
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria;
| | - Philipp Kroiss
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria;
| | - Dominik Kirchhofer
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria;
| | - Lisa Gabler
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria;
| | - Johannes Gojo
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria;
- Department of Pediatrics and Adolescent Medicine and Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniela Lötsch-Gojo
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria;
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Mirjana Stojanovic
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria;
| | - Gerald Timelthaler
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
| | - Franziska Ferk
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
| | - Siegfried Knasmüller
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
| | - Johannes Reisecker
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
| | - Sabine Spiegl-Kreinecker
- Department of Neurosurgery, Neuromed Campus, Kepler University Hospital GmbH, Johannes Kepler University, 4040 Linz, Austria;
| | - Peter Birner
- Clinical Institute of Pathology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.D.); (C.P.); (P.K.); (D.K.); (L.G.); (D.L.-G.); (M.S.); (G.T.); (F.F.); (S.K.); (J.R.)
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria;
- Correspondence: ; Tel.: +43-(0)1-40160-57555
| |
Collapse
|
13
|
Fish JE, Flores Suarez CP, Boudreau E, Herman AM, Gutierrez MC, Gustafson D, DiStefano PV, Cui M, Chen Z, De Ruiz KB, Schexnayder TS, Ward CS, Radovanovic I, Wythe JD. Somatic Gain of KRAS Function in the Endothelium Is Sufficient to Cause Vascular Malformations That Require MEK but Not PI3K Signaling. Circ Res 2020; 127:727-743. [PMID: 32552404 PMCID: PMC7447191 DOI: 10.1161/circresaha.119.316500] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: We previously identified somatic activating mutations in the KRAS (Kirsten rat sarcoma viral oncogene homologue) gene in the endothelium of the majority of human sporadic brain arteriovenous malformations; a disorder characterized by direct connections between arteries and veins. However, whether this genetic abnormality alone is sufficient for lesion formation, as well as how active KRAS signaling contributes to arteriovenous malformations, remains unknown. Objective: To establish the first in vivo models of somatic KRAS gain of function in the endothelium in both mice and zebrafish to directly observe the phenotypic consequences of constitutive KRAS activity at a cellular level in vivo, and to test potential therapeutic interventions for arteriovenous malformations. Methods and Results: Using both postnatal and adult mice, as well as embryonic zebrafish, we demonstrate that endothelial-specific gain of function mutations in Kras (G12D or G12V) are sufficient to induce brain arteriovenous malformations. Active KRAS signaling leads to altered endothelial cell morphogenesis and increased cell size, ectopic sprouting, expanded vessel lumen diameter, and direct connections between arteries and veins. Furthermore, we show that these lesions are not associated with altered endothelial growth dynamics or a lack of proper arteriovenous identity but instead seem to feature exuberant angiogenic signaling. Finally, we demonstrate that KRAS-dependent arteriovenous malformations in zebrafish are refractory to inhibition of the downstream effector PI3K but instead require active MEK (mitogen-activated protein kinase kinase 1) signaling. Conclusions: We demonstrate that active KRAS expression in the endothelium is sufficient for brain arteriovenous malformations, even in the setting of uninjured adult vasculature. Furthermore, the finding that KRAS-dependent lesions are reversible in zebrafish suggests that MEK inhibition may represent a promising therapeutic treatment for arteriovenous malformation patients.
Collapse
Affiliation(s)
- Jason E Fish
- From the Toronto General Hospital Research Institute (J.E.F., E.B., D.G., P.V.D., Z.C.), University Health Network, Canada.,Peter Munk Cardiac Centre (J.E.F.), University Health Network, Canada.,Department of Laboratory Medicine and Pathobiology (J.E.F., D.G.), University of Toronto, Canada
| | - Carlos Perfecto Flores Suarez
- Cardiovascular Research Institute (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., J.D.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology and Biophysics (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., T.S.S., C.S.W., J.D.W.), Baylor College of Medicine, Houston, TX
| | - Emilie Boudreau
- From the Toronto General Hospital Research Institute (J.E.F., E.B., D.G., P.V.D., Z.C.), University Health Network, Canada
| | - Alexander M Herman
- Cardiovascular Research Institute (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., J.D.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology and Biophysics (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., T.S.S., C.S.W., J.D.W.), Baylor College of Medicine, Houston, TX
| | - Manuel Cantu Gutierrez
- Cardiovascular Research Institute (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., J.D.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology and Biophysics (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., T.S.S., C.S.W., J.D.W.), Baylor College of Medicine, Houston, TX.,Graduate Program in Developmental Biology (M.C.G., J.D.W.), Baylor College of Medicine, Houston, TX
| | - Dakota Gustafson
- From the Toronto General Hospital Research Institute (J.E.F., E.B., D.G., P.V.D., Z.C.), University Health Network, Canada.,Department of Laboratory Medicine and Pathobiology (J.E.F., D.G.), University of Toronto, Canada
| | - Peter V DiStefano
- From the Toronto General Hospital Research Institute (J.E.F., E.B., D.G., P.V.D., Z.C.), University Health Network, Canada
| | - Meng Cui
- Cardiovascular Research Institute (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., J.D.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology and Biophysics (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., T.S.S., C.S.W., J.D.W.), Baylor College of Medicine, Houston, TX
| | - Zhiqi Chen
- From the Toronto General Hospital Research Institute (J.E.F., E.B., D.G., P.V.D., Z.C.), University Health Network, Canada
| | - Karen Berman De Ruiz
- Cardiovascular Research Institute (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., J.D.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology and Biophysics (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., T.S.S., C.S.W., J.D.W.), Baylor College of Medicine, Houston, TX
| | - Taylor S Schexnayder
- Department of Molecular Physiology and Biophysics (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., T.S.S., C.S.W., J.D.W.), Baylor College of Medicine, Houston, TX.,and Advanced Technology Cores (T.S.S., C.S.W.), Baylor College of Medicine, Houston, TX
| | - Christopher S Ward
- Department of Molecular Physiology and Biophysics (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., T.S.S., C.S.W., J.D.W.), Baylor College of Medicine, Houston, TX.,and Advanced Technology Cores (T.S.S., C.S.W.), Baylor College of Medicine, Houston, TX
| | - Ivan Radovanovic
- Krembil Research Institute (I.R.), University Health Network, Canada.,Division of Neurosurgery, Sprott Department of Surgery (I.R.), University Health Network, Canada.,Department of Surgery (I.R.), University of Toronto, Canada
| | - Joshua D Wythe
- Cardiovascular Research Institute (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., J.D.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology and Biophysics (C.P.F.S., A.M.H., M.C.G., M.C., K.B.D.R., T.S.S., C.S.W., J.D.W.), Baylor College of Medicine, Houston, TX.,Graduate Program in Developmental Biology (M.C.G., J.D.W.), Baylor College of Medicine, Houston, TX
| |
Collapse
|
14
|
Kim GC, Lee CG, Verma R, Rudra D, Kim T, Kang K, Nam JH, Kim Y, Im SH, Kwon HK. ETS1 Suppresses Tumorigenesis of Human Breast Cancer via Trans-Activation of Canonical Tumor Suppressor Genes. Front Oncol 2020; 10:642. [PMID: 32477936 PMCID: PMC7239993 DOI: 10.3389/fonc.2020.00642] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 04/06/2020] [Indexed: 01/18/2023] Open
Abstract
ETS1 has shown dichotomous roles as an oncogene and a tumor suppressor gene in diverse cancers, but its functionality in breast cancer tumorigenesis still remains unclear. We utilized the Cancer Genome Atlas (TCGA) database to analyze comprehensive functions of ETS1 in human breast cancer (BRCA) patients by investigating its expression patterns and methylation status in relation to clinical prognosis. ETS1 expression was significantly diminished by hyper-methylation of the ETS1 promoter region in specimens from BRCA patients compared to a healthy control group. Moreover, ETS1high BRCA patients showed better prognosis and longer survival compared to ETS1low BRCA patients. Consistent with clinical evidence, comparative transcriptome analysis combined with CRISPR/Cas9 or shRNA based perturbation of ETS1 expression revealed direct as well as indirect mechanisms of ETS1 that hinder tumorigenesis of BRCA cells. Taken together, our study enlightens a novel function of ETS1 as a tumor suppressor in breast cancer cells.
Collapse
Affiliation(s)
- Gi-Cheon Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Choong-Gu Lee
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung Institute of Natural Products, Gangneung, South Korea
| | - Ravi Verma
- Academy of Immunology and Microbiology (AIM), Institute for Basic Science (IBS), Pohang, South Korea
| | - Dipayan Rudra
- Academy of Immunology and Microbiology (AIM), Institute for Basic Science (IBS), Pohang, South Korea
| | - Taemook Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Keunsoo Kang
- Department of Microbiology, College of Natural Sciences, Dankook University, Cheonan, South Korea
| | - Jong Hee Nam
- Department of Pathology, Chonnam National University Medical School, Gwangju, South Korea
| | - Young Kim
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Sin-Hyeog Im
- Academy of Immunology and Microbiology (AIM), Institute for Basic Science (IBS), Pohang, South Korea.,Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
15
|
Merrill NM, Lachacz EJ, Vandecan NM, Ulintz PJ, Bao L, Lloyd JP, Yates JA, Morikawa A, Merajver SD, Soellner MB. Molecular determinants of drug response in TNBC cell lines. Breast Cancer Res Treat 2020; 179:337-347. [PMID: 31655920 PMCID: PMC7323911 DOI: 10.1007/s10549-019-05473-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/10/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE There is a need for biomarkers of drug efficacy for targeted therapies in triple-negative breast cancer (TNBC). As a step toward this, we identify multi-omic molecular determinants of anti-TNBC efficacy in cell lines for a panel of oncology drugs. METHODS Using 23 TNBC cell lines, drug sensitivity scores (DSS3) were determined using a panel of investigational drugs and drugs approved for other indications. Molecular readouts were generated for each cell line using RNA sequencing, RNA targeted panels, DNA sequencing, and functional proteomics. DSS3 values were correlated with molecular readouts using a FDR-corrected significance cutoff of p* < 0.05 and yielded molecular determinant panels that predict anti-TNBC efficacy. RESULTS Six molecular determinant panels were obtained from 12 drugs we prioritized based on their efficacy. Determinant panels were largely devoid of DNA mutations of the targeted pathway. Molecular determinants were obtained by correlating DSS3 with molecular readouts. We found that co-inhibiting molecular correlate pathways leads to robust synergy across many cell lines. CONCLUSIONS These findings demonstrate an integrated method to identify biomarkers of drug efficacy in TNBC where DNA predictions correlate poorly with drug response. Our work outlines a framework for the identification of novel molecular determinants and optimal companion drugs for combination therapy based on these correlates.
Collapse
Affiliation(s)
- Nathan M Merrill
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Eric J Lachacz
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Nathalie M Vandecan
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Peter J Ulintz
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Liwei Bao
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - John P Lloyd
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Joel A Yates
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Aki Morikawa
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Sofia D Merajver
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA.
| | - Matthew B Soellner
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
16
|
Abstract
Mutated or dysregulated transcription factors represent a unique class of drug targets that mediate aberrant gene expression, including blockade of differentiation and cell death gene expression programmes, hallmark properties of cancers. Transcription factor activity is altered in numerous cancer types via various direct mechanisms including chromosomal translocations, gene amplification or deletion, point mutations and alteration of expression, as well as indirectly through non-coding DNA mutations that affect transcription factor binding. Multiple approaches to target transcription factor activity have been demonstrated, preclinically and, in some cases, clinically, including inhibition of transcription factor-cofactor protein-protein interactions, inhibition of transcription factor-DNA binding and modulation of levels of transcription factor activity by altering levels of ubiquitylation and subsequent proteasome degradation or by inhibition of regulators of transcription factor expression. In addition, several new approaches to targeting transcription factors have recently emerged including modulation of auto-inhibition, proteolysis targeting chimaeras (PROTACs), use of cysteine reactive inhibitors, targeting intrinsically disordered regions of transcription factors and combinations of transcription factor inhibitors with kinase inhibitors to block the development of resistance. These innovations in drug development hold great promise to yield agents with unique properties that are likely to impact future cancer treatment.
Collapse
Affiliation(s)
- John H Bushweller
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA.
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
17
|
Wang Y, Ding X, Liu B, Li M, Chang Y, Shen H, Xie SM, Xing L, Li Y. ETV4 overexpression promotes progression of non-small cell lung cancer by upregulating PXN and MMP1 transcriptionally. Mol Carcinog 2019; 59:73-86. [PMID: 31670855 DOI: 10.1002/mc.23130] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/09/2019] [Accepted: 10/17/2019] [Indexed: 12/29/2022]
Abstract
ETS variant 4 (ETV4), together with ETV1 and ETV5, constitute the PEA3 subfamily of ETS transcription factors, which are implicated in the progression of many cancers. However, the clinicopathologic significance and molecular events regulated by ETV4 in lung cancer are still poorly understood, especially in squamous cell carcinoma of the lung. Here, we aimed to identify functional targets involved in ETV4-driven lung tumorigenesis. Microarray analysis and validation data revealed that ETV4 was the most preponderant PEA3 factor, which was significantly related to the advanced stage, lymph node metastasis, and poor prognosis of non-small cell lung cancers (NSCLCs; all P < .001). Reduced ETV4 expression suppressed the growth and metastasis of NSCLC both in vivo and in vitro. Microarray, gain, or loss of function and luciferase report assays revealed the direct regulatory effect of ETV4 on the expression of focal adhesion gene PXN and matrix metalloproteinase 1 (MMP1), and PXN and/or MMP1 inhibition partially abolished cell proliferation and migration induced by ETV4. Kaplan-Meier analysis indicated that ETV4 and PXN or MMP1 co-overexpression is associated with poor prognosis in human NSCLCs. In conclusion, the ETV4-PXN and ETV4-MMP1 axes are useful biomarkers of tumor progression and worse outcomes in NSCLCs.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaosong Ding
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Bei Liu
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Minglei Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ying Chang
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haitao Shen
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China.,Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shelly M Xie
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lingxiao Xing
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China.,Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuehong Li
- Department of Pathology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
18
|
Cao Q, Yang S, Lv Q, Liu Y, Li L, Wu X, Qu G, He X, Zhang X, Sun S, Li B, An J, Hu T, Xue J. Five ETS family members, ELF-1, ETV-4, ETV-3L, ETS-1, and ETS-2 upregulate human leukocyte-associated immunoglobulin-like receptor-1 gene basic promoter activity. Aging (Albany NY) 2019; 10:1390-1401. [PMID: 29915163 PMCID: PMC6046229 DOI: 10.18632/aging.101475] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 06/10/2018] [Indexed: 12/14/2022]
Abstract
Human leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1), an immunoinhibitory receptor, is expressed on most types of hematopoietic cells and some tumor cells. LAIR-1 plays an inhibitory role in immune cell maturation, differentiation, and activation. LAIR-1 is also involved in some autoimmune diseases and tumors. However, the mechanism controlling the regulation of the LAIR-1 gene is still unknown. In order to elucidate the molecular mechanisms involved in LAIR-1 regulation, in the present study, we cloned and characterized the promoter region of LAIR-1 gene using a series of truncated promoter plasmids in luciferase reporter assays. Our results show that the basic core promoter of LAIR-1 is located within the region -256/-8 relative to the translational start site. Our further studies indicate that five ETS transcription factors: ELF-1, ETV-4, ETV-3L, ETS-1 and ETS-2, can up-regulate the LAIR-1 basic promoter activity. Of these, ETS-2 is the most effective transcription factor. Moreover, ETS-2 was confirmed to interact directly with the basic promoter of LAIR-1. This study presents the first description of regions/factors capable of up-regulation the promoter activity of LAIR-1. This new knowledge contributes to understanding of the molecular mechanisms involved in LAIR-1 associated immune regulation and diseases.
Collapse
Affiliation(s)
- Qizhi Cao
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Shandong 264003, China.,Anti-aging Research Institution, Binzhou Medical University, Shandong 264003, China
| | - Shude Yang
- School of Agriculture, Ludong University, Shandong 264003, China
| | - Qing Lv
- Anti-aging Research Institution, Binzhou Medical University, Shandong 264003, China.,School of Gerontology, Binzhou Medical University, Shandong 264003, China
| | - Yan Liu
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Shandong 264003, China
| | - Li Li
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaojie Wu
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Shandong 264003, China
| | - Guiwu Qu
- Anti-aging Research Institution, Binzhou Medical University, Shandong 264003, China.,School of Gerontology, Binzhou Medical University, Shandong 264003, China
| | - Xiaoli He
- The People's Liberation Army 107 Hospital, Affiliated Hospital of Bin Zhou Medical University, Yantai 264002, China
| | - Xiaoshu Zhang
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Shandong 264003, China
| | - Shuqin Sun
- School of Gerontology, Binzhou Medical University, Shandong 264003, China
| | - Boqing Li
- Department of Microbiology, School of Basic Medical Sciences, Binzhou Medical University, Shandong 264003, China
| | - Jing An
- School of Medicine, University of California - San Diego, La Jolla, CA 92037, USA
| | - Tao Hu
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Shandong 264003, China
| | - Jiangnan Xue
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Shandong 264003, China
| |
Collapse
|
19
|
Lai X, Eberhardt M, Schmitz U, Vera J. Systems biology-based investigation of cooperating microRNAs as monotherapy or adjuvant therapy in cancer. Nucleic Acids Res 2019; 47:7753-7766. [PMID: 31340025 PMCID: PMC6735922 DOI: 10.1093/nar/gkz638] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/05/2019] [Accepted: 07/13/2019] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs) are short, noncoding RNAs that regulate gene expression by suppressing mRNA translation and reducing mRNA stability. A miRNA can potentially bind many mRNAs, thereby affecting the expression of oncogenes and tumor suppressor genes as well as the activity of whole pathways. The promise of miRNA therapeutics in cancer is to harness this evolutionarily conserved mechanism for the coordinated regulation of gene expression, and thus restoring a normal cell phenotype. However, the promiscuous binding of miRNAs can provoke unwanted off-target effects, which are usually caused by high-dose single-miRNA treatments. Thus, it is desirable to develop miRNA therapeutics with increased specificity and efficacy. To achieve that, we propose the concept of miRNA cooperativity in order to exert synergistic repression on target genes, thus lowering the required total amount of miRNAs. We first review miRNA therapies in clinical application. Next, we summarize the knowledge on the molecular mechanism and biological function of miRNA cooperativity and discuss its application in cancer therapies. We then propose and discuss a systems biology approach to investigate miRNA cooperativity for the clinical setting. Altogether, we point out the potential of miRNA cooperativity to reduce off-target effects and to complement conventional, targeted, or immune-based therapies for cancer.
Collapse
Affiliation(s)
- Xin Lai
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen, 91052 Erlangen, Germany
- Faculty of Medicine, Friedrich-Alexander University Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Martin Eberhardt
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen, 91052 Erlangen, Germany
- Faculty of Medicine, Friedrich-Alexander University Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Ulf Schmitz
- Computational BioMedicine Laboratory Centenary Institute, The University of Sydney, 2006 Camperdown, Australia
- Gene & Stem Cell Therapy Program Centenary Institute, The University of Sydney, 2006 Camperdown, Australia
- Sydney Medical School, The University of Sydney, 2006 Camperdown, Australia
| | - Julio Vera
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen, 91052 Erlangen, Germany
- Faculty of Medicine, Friedrich-Alexander University Erlangen-Nürnberg, 91052 Erlangen, Germany
| |
Collapse
|
20
|
Gabler L, Lötsch D, Kirchhofer D, van Schoonhoven S, Schmidt HM, Mayr L, Pirker C, Neumayer K, Dinhof C, Kastler L, Azizi AA, Dorfer C, Czech T, Haberler C, Peyrl A, Kumar R, Slavc I, Spiegl-Kreinecker S, Gojo J, Berger W. TERT expression is susceptible to BRAF and ETS-factor inhibition in BRAF V600E/TERT promoter double-mutated glioma. Acta Neuropathol Commun 2019; 7:128. [PMID: 31391125 PMCID: PMC6685154 DOI: 10.1186/s40478-019-0775-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 07/18/2019] [Indexed: 12/20/2022] Open
Abstract
The BRAF gene and the TERT promoter are among the most frequently altered genomic loci in low-grade (LGG) and high-grade-glioma (HGG), respectively. The coexistence of BRAF and TERT promoter aberrations characterizes a subset of aggressive glioma. Therefore, we investigated interactions between those alterations in malignant glioma. We analyzed co-occurrence of BRAFV600E and TERT promoter mutations in our clinical data (n = 8) in addition to published datasets (n = 103) and established a BRAFV600E-positive glioma cell panel (n = 9) for in vitro analyses. We investigated altered gene expression, signaling events and TERT promoter activity upon BRAF- and E-twenty-six (ETS)-factor inhibition by qRT-PCR, chromatin immunoprecipitation (ChIP), Western blots and luciferase reporter assays. TERT promoter mutations were significantly enriched in BRAFV600E-mutated HGG as compared to BRAFV600E-mutated LGG. In vitro, BRAFV600E/TERT promoter double-mutant glioma cells showed exceptional sensitivity towards BRAF-targeting agents. Remarkably, BRAF-inhibition attenuated TERT expression and TERT promoter activity exclusively in double-mutant models, while TERT expression was undetectable in BRAFV600E-only cells. Various ETS-factors were broadly expressed, however, only ETS1 expression and phosphorylation were consistently downregulated following BRAF-inhibition. Knock-down experiments and ChIP corroborated the notion of a functional role for ETS1 and, accordingly, all double-mutant tumor cells were highly sensitive towards the ETS-factor inhibitor YK-4-279. In conclusion, our data suggest that concomitant BRAFV600E and TERT promoter mutations synergistically support cancer cell proliferation and immortalization. ETS1 links these two driver alterations functionally and may represent a promising therapeutic target in this aggressive glioma subgroup.
Collapse
Affiliation(s)
- Lisa Gabler
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
| | - Daniela Lötsch
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Dominik Kirchhofer
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Sushilla van Schoonhoven
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
| | - Hannah M. Schmidt
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Lisa Mayr
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
| | - Katharina Neumayer
- Department of Neurosurgery, Kepler University Hospital, Johannes Kepler University, Neuromed Campus, Wagner-Jauregg-Weg 15, 4020 Linz, Austria
| | - Carina Dinhof
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
| | - Lucia Kastler
- Department of Neurosurgery, Kepler University Hospital, Johannes Kepler University, Neuromed Campus, Wagner-Jauregg-Weg 15, 4020 Linz, Austria
| | - Amedeo A. Azizi
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Christian Dorfer
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Thomas Czech
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Christine Haberler
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Institute of Neurology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Andreas Peyrl
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Irene Slavc
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Sabine Spiegl-Kreinecker
- Department of Neurosurgery, Kepler University Hospital, Johannes Kepler University, Neuromed Campus, Wagner-Jauregg-Weg 15, 4020 Linz, Austria
| | - Johannes Gojo
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Walter Berger
- Comprehensive Cancer Center-Central Nervous System Tumors Unit, Medical University of Vienna, Spitalgasse 23, BT86/E 01, 1090 Vienna, Austria
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
| |
Collapse
|
21
|
Roberts SA, Brown AJ, Wyrick JJ. Recurrent Noncoding Mutations in Skin Cancers: UV Damage Susceptibility or Repair Inhibition as Primary Driver? Bioessays 2019; 41:e1800152. [PMID: 30801747 PMCID: PMC6571124 DOI: 10.1002/bies.201800152] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/05/2018] [Indexed: 12/14/2022]
Abstract
Somatic mutations arising in human skin cancers are heterogeneously distributed across the genome, meaning that certain genomic regions (e.g., heterochromatin or transcription factor binding sites) have much higher mutation densities than others. Regional variations in mutation rates are typically not a consequence of selection, as the vast majority of somatic mutations in skin cancers are passenger mutations that do not promote cell growth or transformation. Instead, variations in DNA repair activity, due to chromatin organization and transcription factor binding, have been proposed to be a primary driver of mutational heterogeneity in melanoma. However, as discussed in this review here, recent studies indicate that chromatin organization and transcription factor binding also significantly modulate the rate at which UV lesions form in DNA. The authors propose that local variations in lesion susceptibility may be an important driver of mutational hotspots in melanoma and other skin cancers, particularly at binding sites for ETS transcription factors.
Collapse
Affiliation(s)
- Steven A. Roberts
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA 99164
| | - Alexander J. Brown
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA 99164
| | - John J. Wyrick
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA 99164
| |
Collapse
|
22
|
Russo G, Pennisi M, Boscarino R, Pappalardo F. Continuous Petri Nets and microRNA Analysis in Melanoma. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:1492-1499. [PMID: 28767374 DOI: 10.1109/tcbb.2017.2733529] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Personalized target therapies represent one of the possible treatment strategies to fight the ongoing battle against cancer. New treatment interventions are still needed for an effective and successful cancer therapy. In this scenario, we simulated and analyzed the dynamics of BRAF V600E melanoma patients treated with BRAF inhibitors in order to find potentially interesting targets that may make standard treatments more effective in particularly aggressive tumors that may not respond to selective inhibitor drugs. To this aim, we developed a continuous Petri Net model that simulates fundamental signalling cascades involved in melanoma development, such as MAPK and PI3K/AKT, in order to deeply analyze these complex kinase cascades and predict new crucial nodes involved in melanomagenesis. The model pointed out that some microRNAs, like hsa-mir-132, downregulates expression levels of p120RasGAP: under high concentrations of p120RasGAP, MAPK pathway activation is significantly decreased and consequently also PI3K/PDK1/AKT activation. Furthermore, our analysis carried out through the Genomic Data Commons (GDC) Data Portal shows the evidence that hsa-mir-132 is significantly associated with clinical outcome in melanoma cancer genomic data sets of BRAF-mutated patients. In conclusion, targeting miRNAs through antisense oligonucleotides technology may suggest the way to enhance the action of BRAF-inhibitors.
Collapse
|
23
|
High ETS2 expression predicts poor prognosis in acute myeloid leukemia patients undergoing allogeneic hematopoietic stem cell transplantation. Ann Hematol 2018; 98:519-525. [PMID: 30022221 DOI: 10.1007/s00277-018-3440-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/10/2018] [Indexed: 10/28/2022]
|
24
|
Hung YF, Chen CY, Shih YC, Liu HY, Huang CM, Hsueh YP. Endosomal TLR3, TLR7, and TLR8 control neuronal morphology through different transcriptional programs. J Cell Biol 2018; 217:2727-2742. [PMID: 29777026 PMCID: PMC6080926 DOI: 10.1083/jcb.201712113] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/09/2018] [Accepted: 05/03/2018] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation is associated with diverse neurological disorders. Endosomal Toll-like receptors (TLRs) including TLR3, TLR7, and TLR8 cell-autonomously regulate neuronal differentiation. However, the mechanisms by which these three TLRs affect neuronal morphology are unclear. In this study, we compare these TLRs in mouse neurons. By combining in vitro neuronal cultures, in utero electroporation, and transcriptomic profiling, we show that TLR8, TLR7, and TLR3 promote dendritic pruning via MYD88 signaling. However, they induce different transcriptomic profiles related to innate immunity, signaling, and neuronal development. The temporal expression patterns and the effects on neuronal morphology are not identical upon activation of these endosomal TLRs. Pathway analyses and in vitro studies specifically implicate mitogen-activated protein kinase signaling in TLR8-mediated dendritic pruning. We further show that TLR8 is more critical for dendritic arborization at a late development stage in vivo. The activation of TLR8, TLR7, or TLR3 results in dendritic shortening, and TLR7 and TLR3 but not TLR8 also control axonal growth. In-depth transcriptomic analyses show that TLRs use different downstream pathways to control neuronal morphology, which may contribute to neuronal development and pathological responses.
Collapse
Affiliation(s)
- Yun-Fen Hung
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan.,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chiung-Ya Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Chun Shih
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Hsin-Yu Liu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | | | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
25
|
Fry EA, Inoue K. Aberrant expression of ETS1 and ETS2 proteins in cancer. CANCER REPORTS AND REVIEWS 2018; 2:10.15761/CRR.1000151. [PMID: 29974077 PMCID: PMC6027756 DOI: 10.15761/crr.1000151] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The ETS transcription factors regulate expression of genes involved in normal cell development, proliferation, differentiation, angiogenesis, and apoptosis, consisting of 28 family members in humans. Dysregulation of these transcription factors facilitates cell proliferation in cancers, and several members participate in invasion and metastasis by activating gene transcription. ETS1 and ETS2 are the founding members of the ETS family and regulate transcription by binding to ETS sequences. They are both involved in oncogenesis and tumor suppression depending on the biological situations used. The essential roles of ETS proteins in human telomere maintenance have been suggested, which have been linked to creation of new Ets binding sites. Recently, preferential binding of ETS2 to gain-of-function mutant p53 and ETS1 to wild type p53 (WTp53) has been suggested, raising the tumor promoting role for the former and tumor suppressive role for the latter. The oncogenic and tumor suppressive functions of ETS1 and 2 proteins have been discussed.
Collapse
Affiliation(s)
- Elizabeth A. Fry
- The Dept. of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| | - Kazushi Inoue
- The Dept. of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| |
Collapse
|