1
|
Simon CS, Garg V, Kuo YY, Niakan KK, Hadjantonakis AK. ETV4 and ETV5 Orchestrate FGF-Mediated Lineage Specification and Epiblast Maturation during Early Mouse Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604964. [PMID: 39091858 PMCID: PMC11291132 DOI: 10.1101/2024.07.24.604964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Cell fate decisions in early mammalian embryos are tightly regulated processes crucial for proper development. While FGF signaling plays key roles in early embryo patterning, its downstream effectors remain poorly understood. Our study demonstrates that the transcription factors Etv4 and Etv5 are critical mediators of FGF signaling in cell lineage specification and maturation in mouse embryos. We show that loss of Etv5 compromises primitive endoderm formation at pre-implantation stages. Furthermore, Etv4/5 deficiency delays naïve pluripotency exit and epiblast maturation, leading to elevated NANOG and reduced OTX2 expression within the blastocyst epiblast. As a consequence of delayed pluripotency progression, Etv4/5 deficient embryos exhibit anterior visceral endoderm migration defects post-implantation, a process essential for coordinated embryonic patterning and gastrulation initiation. Our results demonstrate the successive roles of these FGF signaling effectors in early lineage specification and embryonic body plan establishment, providing new insights into the molecular control of mammalian development.
Collapse
Affiliation(s)
- Claire S. Simon
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- The Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Vidur Garg
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ying-Yi Kuo
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kathy K. Niakan
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- The Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
- Wellcome Trust – Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
2
|
Jin H, Zhang W, Liu H, Bao Y. Genome-wide identification and characteristic analysis of ETS gene family in blood clam Tegillarca granosa. BMC Genomics 2023; 24:700. [PMID: 37990147 PMCID: PMC10664356 DOI: 10.1186/s12864-023-09731-5] [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: 07/18/2023] [Accepted: 10/11/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND ETS transcription factors, known as the E26 transformation-specific factors, assume a critical role in the regulation of various vital biological processes in animals, including cell differentiation, the cell cycle, and cell apoptosis. However, their characterization in mollusks is currently lacking. RESULTS The current study focused on a comprehensive analysis of the ETS genes in blood clam Tegillarca granosa and other mollusk genomes. Our phylogenetic analysis revealed the absence of the SPI and ETV subfamilies in mollusks compared to humans. Additionally, several ETS genes in mollusks were found to lack the PNT domain, potentially resulting in a diminished ability of ETS proteins to bind target genes. Interestingly, the bivalve ETS1 genes exhibited significantly high expression levels during the multicellular proliferation stage and in gill tissues. Furthermore, qRT-PCR results showed that Tg-ETS-14 (ETS1) is upregulated in the high total hemocyte counts (THC) population of T. granosa, suggesting it plays a significant role in stimulating hemocyte proliferation. CONCLUSION Our study significantly contributes to the comprehension of the evolutionary aspects concerning the ETS gene family, while also providing valuable insights into its role in fostering hemocyte proliferation across mollusks.
Collapse
Affiliation(s)
- Hongyu Jin
- School of Marine Sciences, Ningbo University, Ningbo, 315000, China
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Zhejiang, 315100, China
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, Ningbo, 315000, China
| | - Hongxing Liu
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Zhejiang, 315100, China.
| | - Yongbo Bao
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Zhejiang, 315100, China.
| |
Collapse
|
3
|
Rogers MF, Marshall OJ, Secombe J. KDM5-mediated activation of genes required for mitochondrial biology is necessary for viability in Drosophila. Development 2023; 150:dev202024. [PMID: 37800333 PMCID: PMC10651110 DOI: 10.1242/dev.202024] [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: 05/20/2023] [Accepted: 09/29/2023] [Indexed: 10/07/2023]
Abstract
Histone-modifying proteins play important roles in the precise regulation of the transcriptional programs that coordinate development. KDM5 family proteins interact with chromatin through demethylation of H3K4me3 as well as demethylase-independent mechanisms that remain less understood. To gain fundamental insights into the transcriptional activities of KDM5 proteins, we examined the essential roles of the single Drosophila Kdm5 ortholog during development. KDM5 performs crucial functions in the larval neuroendocrine prothoracic gland, providing a model to study its role in regulating key gene expression programs. Integrating genome binding and transcriptomic data, we identify that KDM5 regulates the expression of genes required for the function and maintenance of mitochondria, and we find that loss of KDM5 causes morphological changes to mitochondria. This is key to the developmental functions of KDM5, as expression of the mitochondrial biogenesis transcription factor Ets97D, homolog of GABPα, is able to suppress the altered mitochondrial morphology as well as the lethality of Kdm5 null animals. Together, these data establish KDM5-mediated cellular functions that are important for normal development and could contribute to KDM5-linked disorders when dysregulated.
Collapse
Affiliation(s)
- Michael F. Rogers
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Owen J. Marshall
- Menzies Institute for Medical Research, University of Tasmania, Hobart TAS 7000, Australia
| | - Julie Secombe
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
4
|
Choi Y, Lee Y, Kim JS, Zhang P, Kim J. USP39-Mediated Non-Proteolytic Control of ETS2 Suppresses Nuclear Localization and Activity. Biomolecules 2023; 13:1475. [PMID: 37892157 PMCID: PMC10604658 DOI: 10.3390/biom13101475] [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: 07/01/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
ETS2 is a member of the ETS family of transcription factors and has been implicated in the regulation of cell proliferation, differentiation, apoptosis, and tumorigenesis. The aberrant activation of ETS2 is associated with various human cancers, highlighting its importance as a therapeutic target. Understanding the regulatory mechanisms and interacting partners of ETS2 is crucial for elucidating its precise role in cellular processes and developing novel strategies to modulate its activity. In this study, we conducted binding assays using a human deubiquitinase (DUB) library and identified USP39 as a novel ETS2-binding DUB. USP39 interacts with ETS2 through their respective amino-terminal regions, and the zinc finger and PNT domains are not required for this binding. USP39 deubiquitinates ETS2 without affecting its protein stability. Interestingly, however, USP39 significantly suppresses the transcriptional activity of ETS2. Furthermore, we demonstrated that USP39 leads to a reduction in the nuclear localization of ETS2. Our findings provide valuable insights into the intricate regulatory mechanisms governing ETS2 function. Understanding the interplay between USP39 and ETS2 may have implications for therapeutic interventions targeting ETS2-related diseases, including cancer, where the dysregulation of ETS2 is frequently observed.
Collapse
Affiliation(s)
- Yunsik Choi
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Yuri Lee
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Jin Seo Kim
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| | - Peijing Zhang
- Department of Biological Pharmaceutics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jongchan Kim
- Department of Life Sciences, Sogang University, Seoul 04107, Republic of Korea
| |
Collapse
|
5
|
Zhu M, Dagah OMA, Silaa BB, Lu J. Thioredoxin/Glutaredoxin Systems and Gut Microbiota in NAFLD: Interplay, Mechanism, and Therapeutical Potential. Antioxidants (Basel) 2023; 12:1680. [PMID: 37759983 PMCID: PMC10525532 DOI: 10.3390/antiox12091680] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/20/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common clinical disease, and its pathogenesis is closely linked to oxidative stress and gut microbiota dysbiosis. Recently accumulating evidence indicates that the thioredoxin and glutaredoxin systems, the two thiol-redox dependent antioxidant systems, are the key players in the NAFLD's development and progression. However, the effects of gut microbiota dysbiosis on the liver thiol-redox systems are not well clarified. This review explores the role and mechanisms of oxidative stress induced by bacteria in NAFLD while emphasizing the crucial interplay between gut microbiota dysbiosis and Trx mediated-redox regulation. The paper explores how dysbiosis affects the production of specific gut microbiota metabolites, such as trimethylamine N-oxide (TMAO), lipopolysaccharides (LPS), short-chain fatty acids (SCFAs), amino acids, bile acid, and alcohol. These metabolites, in turn, significantly impact liver inflammation, lipid metabolism, insulin resistance, and cellular damage through thiol-dependent redox signaling. It suggests that comprehensive approaches targeting both gut microbiota dysbiosis and the thiol-redox antioxidant system are essential for effectively preventing and treating NAFLD. Overall, comprehending the intricate relationship between gut microbiota dysbiosis and thiol-redox systems in NAFLD holds significant promise in enhancing patient outcomes and fostering the development of innovative therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - Jun Lu
- Engineering Research Center of Coptis Development and Utilization/Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education (Southwest University), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China; (M.Z.); (O.M.A.D.); (B.B.S.)
| |
Collapse
|
6
|
Sim N, Li Y. NF-κB/p52 augments ETS1 binding genome-wide to promote glioma progression. Commun Biol 2023; 6:445. [PMID: 37087499 PMCID: PMC10122670 DOI: 10.1038/s42003-023-04821-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 04/06/2023] [Indexed: 04/24/2023] Open
Abstract
Gliomas are highly invasive and chemoresistant cancers, making them challenging to treat. Chronic inflammation is a key driver of glioma progression as it promotes aberrant activation of inflammatory pathways such as NF-κB signalling, which drives cancer cell invasion and angiogenesis. NF-κB factors typically dimerise with its own family members, but emerging evidence of their promiscuous interactions with other oncogenic factors has been reported to promote transcription of new target genes and function. Here, we show that non-canonical NF-κB activation directly regulates p52 at the ETS1 promoter, activating its expression. This impacts the genomic and transcriptional landscape of ETS1 in a glioma-specific manner. We further show that enhanced non-canonical NF-κB signalling promotes the co-localisation of p52 and ETS1, resulting in transcriptional activation of non-κB and/or non-ETS glioma-promoting genes. We conclude that p52-induced ETS1 overexpression in glioma cells remodels the genome-wide regulatory network of p52 and ETS1 to transcriptionally drive cancer progression.
Collapse
Affiliation(s)
- Nicholas Sim
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Yinghui Li
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore.
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, 138673, Singapore.
| |
Collapse
|
7
|
Zhang LY, Tan Y, Luo XJ, Wu JF, Ni YR. The roles of ETS transcription factors in liver fibrosis. Hum Cell 2023; 36:528-539. [PMID: 36547849 DOI: 10.1007/s13577-022-00848-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
E26 transformation specific or E twenty-six (ETS) protein family consists of 28 transcription factors, five of which, named ETS1/2, PU.1, ERG and EHF, are known to involve in the development of liver fibrosis, and are expected to become diagnostic markers or therapeutic targets of liver fibrosis. In recent years, some small molecule inhibitors of ETS protein family have been discovered, which might open up a new path for the liver fibrosis therapy targeting ETS. This article reviews the research progress of ETS family members in the development liver fibrosis as well as their prospect of clinical application.
Collapse
Affiliation(s)
- Li-Ye Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- College of Basic Medical Science, China Three Gorges University, Yichang, China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, Yichang, China
| | - Yong Tan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- College of Basic Medical Science, China Three Gorges University, Yichang, China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, Yichang, China
| | - Xiao-Jie Luo
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- College of Basic Medical Science, China Three Gorges University, Yichang, China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, Yichang, China
| | - Jiang-Feng Wu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China.
- College of Basic Medical Science, China Three Gorges University, Yichang, China.
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, Yichang, China.
| | - Yi-Ran Ni
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China.
- College of Basic Medical Science, China Three Gorges University, Yichang, China.
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, Yichang, China.
| |
Collapse
|
8
|
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
|
9
|
Transcriptional Regulation of Siglec-15 by ETS-1 and ETS-2 in Hepatocellular Carcinoma Cells. Int J Mol Sci 2023; 24:ijms24010792. [PMID: 36614238 PMCID: PMC9821606 DOI: 10.3390/ijms24010792] [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: 10/25/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 01/04/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) has been identified as a crucial immune suppressor in human cancers, comparable to programmed cell death 1 ligand (PD-L1). However, the regulatory mechanisms underlying its transcriptional upregulation in human cancers remain largely unknown. Here, we show that the transcription factors ETS-1 and ETS-2 bound to the Siglec-15 promoter to enhance transcription and expression of Siglec-15 in hepatocellular carcinoma (HCC) cells and that transforming growth factor β-1 (TGF-β1) upregulated the expression of ETS-1 and ETS-2 and facilitated the binding of ETS-1 and ETS-2 to the Siglec-15 promoter. We further demonstrate that TGF-β1 activated the Ras/C-Raf/MEK/ERK1/2 signaling pathway, leading to phosphorylation of ETS-1 and ETS-2, which consequently upregulates the transcription and expression of Siglec-15. Our study defines a detailed molecular profile of how Siglec-15 is transcriptionally regulated which may offer significant opportunity for therapeutic intervention on HCC immunotherapy.
Collapse
|
10
|
Li J, Du H, Ji X, Chen Y, Li Y, Heng BC, Xu J. ETV2 promotes osteogenic differentiation of human dental pulp stem cells through the ERK/MAPK and PI3K-Akt signaling pathways. Stem Cell Res Ther 2022; 13:495. [PMID: 36195958 PMCID: PMC9533526 DOI: 10.1186/s13287-022-03052-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 07/18/2022] [Indexed: 11/10/2022] Open
Abstract
Background The repair of cranio-maxillofacial bone defects remains a formidable clinical challenge. The Ets variant 2 (ETV2) transcription factor, which belongs to the E26 transformation-specific (ETS) family, has been reported to play a key role in neovascularization. However, the role of ETV2 in the osteogenesis of human dental pulp stem cells (hDPSCs) remains unexplored. Methods Transgenic overexpression of ETV2 was achieved using a lentiviral vector, based on a Dox-inducible system. The effects of Dox-induced overexpression of ETV2 on the osteogenesis of hDPSCs were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR), western blot, immunofluorescence staining, alkaline phosphatase (ALP) staining, and Alizarin Red S (ARS) staining. Additionally, RNA-sequencing (RNA-Seq) analysis was performed to analyze the underlying mechanisms of ETV2-induced osteogenesis. Additionally, the role of ETV2 overexpression in bone formation in vivo was validated by animal studies with a rat calvarial defect model and a nude mice model. Results Our results demonstrated that ETV2 overexpression significantly upregulated the mRNA and protein expression levels of osteogenic markers, markedly enhanced ALP activity, and promoted matrix mineralization of hDPSCs. Moreover, the results of RNA-Seq analysis and western blot showed that the ERK/MAPK and PI3K-Akt signaling pathways were activated upon transgenic overexpression of ETV2. The enhanced osteogenic differentiation of hDPSCs due to ETV2 overexpression was partially reversed by treatment with inhibitors of ERK/MAPK or PI3K-AKT signaling. Furthermore, the results of in vivo studies demonstrated that ETV2 overexpression improved bone healing in a rat calvarial defect model and increased ectopic bone formation in nude mice. Conclusions Collectively, our results indicated that ETV2 overexpression exerted positive effects on the osteogenesis of hDPSCs, at least partially via the ERK/MAPK and PI3K/AKT signaling pathways. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03052-2.
Collapse
Affiliation(s)
- Jing Li
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, People's Republic of China
| | - Haoran Du
- Key Lab of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, 69 Meishan Road, Hefei, 230032, People's Republic of China
| | - Xin Ji
- Key Lab of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, 69 Meishan Road, Hefei, 230032, People's Republic of China
| | - Yihan Chen
- Key Lab of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, 69 Meishan Road, Hefei, 230032, People's Republic of China
| | - Yishuai Li
- Key Lab of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, 69 Meishan Road, Hefei, 230032, People's Republic of China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, People's Republic of China
| | - Jianguang Xu
- Key Lab of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, 69 Meishan Road, Hefei, 230032, People's Republic of China.
| |
Collapse
|
11
|
Du L, Liu Y, Li C, Deng J, Sang Y. The interaction between ETS transcription factor family members and microRNAs: A novel approach to cancer therapy. Biomed Pharmacother 2022; 150:113069. [PMID: 35658214 DOI: 10.1016/j.biopha.2022.113069] [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: 03/08/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 11/18/2022] Open
Abstract
In cancer biology, ETS transcription factors promote tumorigenesis by mediating transcriptional regulation of numerous genes via the conserved ETS DNA-binding domain. MicroRNAs (miRNAs) act as posttranscriptional regulators to regulate various tumor-promoting or tumor-suppressing factors. Interactions between ETS factors and miRNAs regulate complex tumor-promoting and tumor-suppressing networks. This review discusses the progress of ETS factors and miRNAs in cancer research in detail. We focused on characterizing the interaction of the miRNA/ETS axis with competing endogenous RNAs (ceRNAs) and its regulation in posttranslational modifications (PTMs) and the tumor microenvironment (TME). Finally, we explore the prospect of ETS factors and miRNAs in therapeutic intervention. Generally, interactions between ETS factors and miRNAs provide fresh perspectives into tumorigenesis and development and novel therapeutic approaches for malignant tumors.
Collapse
Affiliation(s)
- Liwei Du
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Department of Center Laboratory, The Third Affiliated Hospital of Nanchang University & The First Hospital of Nanchang, Nanchang 330008, China
| | - Yuchen Liu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Department of Center Laboratory, The Third Affiliated Hospital of Nanchang University & The First Hospital of Nanchang, Nanchang 330008, China; Stomatology College of Nanchang University, Nanchang, China
| | - Chenxi Li
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Department of Center Laboratory, The Third Affiliated Hospital of Nanchang University & The First Hospital of Nanchang, Nanchang 330008, China
| | - Jinkuang Deng
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Department of Center Laboratory, The Third Affiliated Hospital of Nanchang University & The First Hospital of Nanchang, Nanchang 330008, China
| | - Yi Sang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Department of Center Laboratory, The Third Affiliated Hospital of Nanchang University & The First Hospital of Nanchang, Nanchang 330008, China.
| |
Collapse
|
12
|
Huang L, Zhai Y, Fajardo CD, Lang D. YK-4-279 Attenuates Progression of Pre-Existing Pigmented Lesions to Nodular Melanoma in a Mouse Model. Cancers (Basel) 2021; 14:143. [PMID: 35008307 PMCID: PMC8749984 DOI: 10.3390/cancers14010143] [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: 12/06/2021] [Revised: 12/23/2021] [Accepted: 12/26/2021] [Indexed: 11/25/2022] Open
Abstract
More options are needed for the effective treatment of melanoma. In a previous study, we discovered the small molecule drug YK-4-279 almost completely inhibited tumor progression in the BrafCA;Tyr-CreERT2;Ptenflox/flox transgenic mouse model. YK-4-279 had no effect on tumor initiation but blocked progression of invasive melanoma. Our current study was designed as a treatment model, where YK-4-279 was administered during pigmented lesion formation. The study design included the use of three groups: (1) a control group that received only DMSO without a drug (MOCK), (2) mice following our prior studies with YK-4-279 administered at the time of tumor induction (YK-4-279), and (3) mice treated during tumor initiation (YK-4-279 delay). While the MOCK mice had progression of tumors, both YK-4-279 and YK-4-279 delay groups had a significant block or delay of progression. The majority of mice in the YK-4-279 groups had a block of progression, while the YK-4-279 delay group had either a partial block (60% in male mice or 29% in females) or a delay in disease progression in females (28 days in controls to 50 days in YK-4-279 delay group). Here, we demonstrate that YK-4-279 has a significant impact on blocking or delaying tumor progression in a pre-clinical treatment model of melanoma.
Collapse
Affiliation(s)
| | | | | | - Deborah Lang
- Department of Dermatology, Boston University, Boston, MA 02118, USA; (L.H.); (Y.Z.); (C.D.F.)
| |
Collapse
|
13
|
Comprehensive Analysis Identified ETV7 as a Potential Prognostic Biomarker in Bladder Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8530186. [PMID: 34926692 PMCID: PMC8678556 DOI: 10.1155/2021/8530186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/12/2021] [Accepted: 10/27/2021] [Indexed: 11/18/2022]
Abstract
Background The tumor microenvironment (TME) plays a crucial role in the initiation and progression of cancer. Bladder cancer (BLCA) is a malignant tumor of the genitourinary system. Its heterogeneity results in significant differences in the prognosis of patients. To date, this is still a huge challenge for clinical treatment. In recent years, more and more evidence showed that dysregulation of transcription factors (TFs) plays an important role in tumor progression, invasion, and metastasis. Unfortunately, the role of TFs on the tumor microenvironment in bladder cancer is unclear. Methods The original data of BLCA and corresponding adjacent tissues were obtained from The Cancer Genome Atlas (TCGA) database. TFs were downloaded from the Animal Transcription Factor DataBase (Animal TFDB). Intersection analysis was used to obtain TFs that were differentially expressed between tumor and adjacent tissues. Gene Set Cancer Analysis (GSCALite) and CIBERSORT software were used to reveal the key differentially expressed TFs (DE-TFs). Subsequently, UALCAN and Human Protein Atlas (HPA) databases were used to disclose the expression of key DE-TFs in BLCA. The K-M curve divulged the relationship between the key DE-TFs and the patient's overall survival (OS), and the univariate and multivariate Cox regression analyses were conducted to explore independent prognostic factors. The cluster profiler package and Gene Set Enrichment Analysis (GSEA) were used for functional enrichment of genes related to the key DE-TFs. Finally, CIBERSORT software analyzed the immune landscape of BLCA. Results We obtained a total of 117 BLCA-related DE-TFs. Among them, ETV7 was identified as the key DE-TFs due to its association with the autophagy activation pathway and various immune cells in cancer. Online databases of UALCAN and HPA indicated that ETV7 was overexpressed in tumors and negatively correlated with tumor severity. The K-M curve showed that the OS of patients with high expression of ETV7 was poor, which indicated that it was an independent prognostic factor. Functional enrichment of 87 DEGs between ETV7-high and -low expression groups indicated that it was closely related to the immune response and the functions of a variety of immune cells. Finally, CIBERSORT results proved that the high and low expression of ETV7 also caused significant differences in the tumor immune microenvironment of patients. Conclusion Overall, we proved that the transcription factor ETV7 was a novel prognostic factor, which may improve the individualized outcome prediction in BLCA by regulating the tumor immune microenvironment.
Collapse
|
14
|
Harel S, Sanchez V, Moamer A, Sanchez-Galan JE, Abid Hussein MN, Mayaki D, Blanchette M, Hussain SNA. ETS1, ELK1, and ETV4 Transcription Factors Regulate Angiopoietin-1 Signaling and the Angiogenic Response in Endothelial Cells. Front Physiol 2021; 12:683651. [PMID: 34381375 PMCID: PMC8350579 DOI: 10.3389/fphys.2021.683651] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 07/05/2021] [Indexed: 12/03/2022] Open
Abstract
Background Angiopoietin-1 (Ang-1) is the main ligand of Tie-2 receptors. It promotes endothelial cell (EC) survival, migration, and differentiation. Little is known about the transcription factors (TFs) in ECs that are downstream from Tie-2 receptors. Objective The main objective of this study is to identify the roles of the ETS family of TFs in Ang-1 signaling and the angiogenic response. Methods In silico enrichment analyses that were designed to predict TF binding sites of the promotors of eighty-six Ang-1-upregulated genes showed significant enrichment of ETS1, ELK1, and ETV4 binding sites in ECs. Human umbilical vein endothelial cells (HUVECs) were exposed for different time periods to recombinant Ang-1 protein and mRNA levels of ETS1, ELK1, and ETV4 were measured with qPCR and intracellular localization of these transcription factors was assessed with immunofluorescence. Electrophoretic mobility shift assays and reporter assays were used to assess activation of ETS1, ELK1, and ETV4 in response to Ang-1 exposure. The functional roles of these TFs in Ang-1-induced endothelial cell survival, migration, differentiation, and gene regulation were evaluated by using a loss-of-function approach (transfection with siRNA oligos). Results Ang-1 exposure increased ETS1 mRNA levels but had no effect on ELK1 or ETV4 levels. Immunostaining revealed that in control ECs, ETS1 has nuclear localization whereas ELK1 and ETV4 are localized to the nucleus and the cytosol. Ang-1 exposure increased nuclear intensity of ETS1 protein and enhanced nuclear mobilization of ELK1 and ETV4. Selective siRNA knockdown of ETS1, ELK1, and ETV4 showed that these TFs are required for Ang-1-induced EC survival and differentiation of cells, while ETS1 and ETV4 are required for Ang-1-induced EC migration. Moreover, ETS1, ELK1, and ETV4 knockdown inhibited Ang-1-induced upregulation of thirteen, eight, and nine pro-angiogenesis genes, respectively. Conclusion We conclude that ETS1, ELK1, and ETV4 transcription factors play significant angiogenic roles in Ang-1 signaling in ECs.
Collapse
Affiliation(s)
- Sharon Harel
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Critical Care, McGill University Health Centre, Montreal, QC, Canada.,Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Veronica Sanchez
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Critical Care, McGill University Health Centre, Montreal, QC, Canada.,Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Alaa Moamer
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Critical Care, McGill University Health Centre, Montreal, QC, Canada.,Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Javier E Sanchez-Galan
- School of Computer Science, McGill Centre for Bioinformatics, McGill University, Montreal, QC, Canada
| | - Mohammad N Abid Hussein
- School of Engineering and Technology (SET), Aldar University College, Dubai, United Arab Emirates
| | - Dominique Mayaki
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Critical Care, McGill University Health Centre, Montreal, QC, Canada.,Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Mathieu Blanchette
- School of Computer Science, McGill Centre for Bioinformatics, McGill University, Montreal, QC, Canada
| | - Sabah N A Hussain
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Critical Care, McGill University Health Centre, Montreal, QC, Canada.,Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
15
|
Zeng G, Wang T, Zhang J, Kang YJ, Feng L. FLI1 mediates the selective expression of hypoxia-inducible factor 1 target genes in endothelial cells under hypoxic conditions. FEBS Open Bio 2021. [PMID: 34102031 PMCID: PMC8329784 DOI: 10.1002/2211-5463.13220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/13/2021] [Accepted: 06/07/2021] [Indexed: 02/05/2023] Open
Abstract
The selective expression of hypoxia‐inducible factor (HIF) target genes in different physiological and pathological environments forms the basis for cellular adaptation to hypoxia in development and disease. Several E26 transformation‐specific (ETS) transcription factors have been shown to specifically regulate the expression of a subset of HIF‐2 target genes. However, it is unknown whether there are ETS factors that specifically regulate hypoxia‐induced HIF‐1 target genes. The present study was undertaken to explore whether friend leukemia integration 1 (FLI1), an ETS transcription factor, regulates the expression of HIF‐1 target genes. To investigate this possibility, EA.hy926 cells were exposed to 20% O2 (normoxia) or 1% O2 (hypoxia). Western blotting, immunofluorescence staining, and RT‐qPCR revealed that FLI1 mRNA and protein levels increased slightly and that the FLI1 protein co‐localized with HIF‐1α in the nucleus under hypoxic conditions. Further analysis showed that, in the absence of FLI1, the hypoxia‐mediated induction of HIF‐1 target genes was selectively inhibited. The results from immunoprecipitation and luciferase reporter assays indicated that FLI1 cooperates with HIF‐1α and is required for the transcriptional activation of a subset of HIF‐1 target genes with a core promoter region containing FBS in proximity to a functional hypoxia response element (HRE). Furthermore, ChIP analysis further confirmed the direct interaction between FLI1 and the promoter region of FLI1‐dependent HIF‐1 target genes under hypoxia. Together, this study demonstrates that FLI1 is involved in the transactivation of certain HIF‐1 target genes in endothelial cells under hypoxic conditions.
Collapse
Affiliation(s)
- Guodan Zeng
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, China.,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Wang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingyao Zhang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Y James Kang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.,Memphis Institute of Regenerative Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Li Feng
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, China.,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Qu H, Zhao H, Zhang X, Liu Y, Li F, Sun L, Song Z. Integrated Analysis of the ETS Family in Melanoma Reveals a Regulatory Role of ETV7 in the Immune Microenvironment. Front Immunol 2020; 11:612784. [PMID: 33424867 PMCID: PMC7786291 DOI: 10.3389/fimmu.2020.612784] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/19/2020] [Indexed: 12/22/2022] Open
Abstract
The ETS family modulates immune response and drug efficiency to targeted therapies, but their role in melanoma is largely unclear. In this study, the ETS family was systematically analyzed in multiple public data sets. Bioinformatics tools were used to characterize the function of ETV7 in melanoma. A prognostic model was constructed using the LASSO Cox regression method. We found that ETV7 was the only differentially expressed gene with significant prognostic relevance in melanoma. Enrichment analysis of seven independent data sets indicated ETV7 participation in various immune-related pathways. ETV7 particularly showed a strong positive correlation with CD8+ T cell infiltration. The prognostic model based on ETV7 and its hub genes showed a relatively good predictive value in training and testing data sets. Thus, ETV7 can potentially regulate the immune microenvironment in melanoma.
Collapse
Affiliation(s)
- Hui Qu
- Department of Plastic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Hui Zhao
- Department of Urology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xi Zhang
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yang Liu
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Feng Li
- Department of Plastic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liyan Sun
- Department of Plastic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Zewen Song
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
17
|
Ye T, Feng J, Wan X, Xie D, Liu J. Double Agent: SPDEF Gene with Both Oncogenic and Tumor-Suppressor Functions in Breast Cancer. Cancer Manag Res 2020; 12:3891-3902. [PMID: 32547225 PMCID: PMC7259446 DOI: 10.2147/cmar.s243748] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 04/25/2020] [Indexed: 12/16/2022] Open
Abstract
The dichotomy of cancer-regulatory genes into “oncogenes (OCGs)” and “tumor-suppressor genes (TSGs)” has greatly helped us in learning molecular details of tumor biology. SPDEF, known as the prostate-derived ETS factor, is reported to play a pivotal role in normal cell development and survival, which has also been endowed with dual characteristics in cancers. Breast cancer (BC) is a highly heterogeneous disease which becomes the leading reason for cancer-related fatality among women worldwide. The involvement of SPDEF in many aspects of BC has been postulated, whereas the mechanism governing the regulation of the pro- and anti-oncogenic activities of SPDEF in BC state remains poorly defined. In this review, we summarized SPDEF as the double agent involving in expression profiles, the regulatory mechanism in BC progression, as well as the role in diagnosis, treatment and prognosis of BC. The understanding of SPDEF duality has contributed to gain insight into the tumor biology and also add a new dimension to the new therapy targets for BC.
Collapse
Affiliation(s)
- Ting Ye
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| | - Jia Feng
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| | - Xue Wan
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| | - Dan Xie
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| |
Collapse
|
18
|
Garg A, Hannan A, Wang Q, Makrides N, Zhong J, Li H, Yoon S, Mao Y, Zhang X. Etv transcription factors functionally diverge from their upstream FGF signaling in lens development. eLife 2020; 9:e51915. [PMID: 32043969 PMCID: PMC7069720 DOI: 10.7554/elife.51915] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
The signal regulated transcription factors (SRTFs) control the ultimate transcriptional output of signaling pathways. Here, we examined a family of FGF-induced SRTFs - Etv1, Etv 4, and Etv 5 - in murine lens development. Contrary to FGF receptor mutants that displayed loss of ERK signaling and defective cell differentiation, Etv deficiency augmented ERK phosphorylation without disrupting the normal lens fiber gene expression. Instead, the transitional zone for lens differentiation was shifted anteriorly as a result of reduced Jag1-Notch signaling. We also showed that Etv proteins suppresses mTOR activity by promoting Tsc2 expression, which is necessary for the nuclei clearance in mature lens. These results revealed the functional divergence between Etv and FGF in lens development, demonstrating that these SRTFs can operate outside the confine of their upstream signaling.
Collapse
Affiliation(s)
- Ankur Garg
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Abdul Hannan
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Qian Wang
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Neoklis Makrides
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Jian Zhong
- Burke Neurological Institute and Feil Family Brain and Mind Research Institute, Weill Cornell MedicineWhite PlainsUnited States
| | - Hongge Li
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Sungtae Yoon
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Yingyu Mao
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Xin Zhang
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| |
Collapse
|
19
|
Gupta A, Towers C, Willenbrock F, Brant R, Hodgson DR, Sharpe A, Smith P, Cutts A, Schuh A, Asher R, Myers K, Love S, Collins L, Wise A, Middleton MR, Macaulay VM. Dual-specificity protein phosphatase DUSP4 regulates response to MEK inhibition in BRAF wild-type melanoma. Br J Cancer 2019; 122:506-516. [PMID: 31839677 PMCID: PMC7028919 DOI: 10.1038/s41416-019-0673-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/30/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023] Open
Abstract
Background Aiming to improve treatment options for BRAF wild-type melanoma, we previously conducted the DOC-MEK study of docetaxel with MEK inhibitor (MEKi) selumetinib or placebo, revealing trends to prolongation of progression-free survival (hazard ratio 0.75, P = 0.130), and improved response rates (32% vs 14%, P = 0.059) with docetaxel plus selumetinib. NRAS status did not associate with outcome. Here, the aim was to identify novel biomarkers of response to MEKi. Methods A MEK 6 gene signature was quantified using NanoString and correlated with clinical outcomes. Two components of the gene signature were investigated by gene silencing in BRAF/NRAS wild-type melanoma cells. Results In melanomas of patients on the selumetinib but not the placebo arm, two gene signature components, dual-specificity protein phosphatase 4 (DUSP4) and ETS translocation variant 4 (ETV4), were expressed more highly in responders than non-responders. In vitro, ETV4 depletion inhibited cell survival but did not influence sensitivity to MEKi selumetinib or trametinib. In contrast, DUSP4-depleted cells showed enhanced cell survival and increased resistance to both selumetinib and trametinib. Conclusions ETV4 and DUSP4 associated with clinical response to docetaxel plus selumetinib. DUSP4 depletion induced MEKi resistance, suggesting that DUSP4 is not only a biomarker but also a mediator of MEKi sensitivity. Clinical Trial Registration DOC-MEK (EudraCT no: 2009-018153-23).
Collapse
Affiliation(s)
- Avinash Gupta
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK. .,Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford, UK.
| | - Christopher Towers
- Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | - Frances Willenbrock
- Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | - Roz Brant
- Translational Science, Oncology iMED, AstraZeneca, Macclesfield, UK
| | | | | | - Paul Smith
- Cancer BioSciences, AstraZeneca, Cambridge, UK
| | - Anthony Cutts
- Molecular Diagnostics Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Anna Schuh
- Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford, UK.,National Institute for Health Research Biomedical Research Centre, Oxford, UK
| | - Ruth Asher
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Kevin Myers
- Experimental Cancer Medicine Centre, Oxford, UK
| | - Sharon Love
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Linda Collins
- Oncology Clinical Trials Office, University of Oxford, Oxford, UK
| | - Adelyn Wise
- Oncology Clinical Trials Office, University of Oxford, Oxford, UK
| | - Mark Roy Middleton
- Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford, UK.,National Institute for Health Research Biomedical Research Centre, Oxford, UK
| | - Valentine Moya Macaulay
- Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford, UK.,National Institute for Health Research Biomedical Research Centre, Oxford, UK
| |
Collapse
|
20
|
Ma X, Jiang Z, Li N, Jiang W, Gao P, Yang M, Yu X, Wang G, Zhang Y. Ets2 suppresses inflammatory cytokines through MAPK/NF-κB signaling and directly binds to the IL-6 promoter in macrophages. Aging (Albany NY) 2019; 11:10610-10625. [PMID: 31785145 PMCID: PMC6914388 DOI: 10.18632/aging.102480] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022]
Abstract
Proper activation of Toll-like receptor (TLR)-mediated signaling and production of proinflammatory cytokines are critical for the initiation of innate immunity, while the specific mechanism maintaining inflammatory homeostasis remains mostly unknown. Here, we show that Ets2 is upregulated following LPS and VSV stimulation. Ets2 knockdown or knockout leads to increased IL-6, TNF-α, and IFN-β production in macrophages. Consistently, Ets2-deficient mice show exacerbated inflammatory cytokine production and are more susceptible to CLP-induced sepsis. Mechanistically, Ets2 inhibits the LPS- and VSV-induced activation of ERK1/2, JNK, p38, and p65. Ets2 also binds to the promoter of IL-6 to inhibit transcription. Collectively, the results of the present study show the negative regulatory role of Ets2 in LPS- and VSV-induced inflammation through the suppression of MAPK/NF-κB signaling, direct binding to the IL-6 promoter and inhibition of transcription.
Collapse
Affiliation(s)
- Xianwei Ma
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Zhengyu Jiang
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai 200433, China
| | - Na Li
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai 200433, China
| | - Wei Jiang
- Department of Respiration, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Peng Gao
- Cancer Institute, Institute of Translational Medicine, Second Military Medical University/Naval Medical University, Shanghai 200433, China
| | - Mingjin Yang
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University/Naval Medical University, Shanghai 200433, China
| | - Xiya Yu
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai 200433, China
| | - Guifang Wang
- Department of Respiratory Diseases, Huashan Hospital, Fudan University, Shanghai 200433, China
| | - Yan Zhang
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai 200433, China
| |
Collapse
|
21
|
Shi X, Wang X. LINC00473 mediates cyclin D1 expression through a balance between activation and repression signals in breast cancer cells. FEBS Lett 2019; 593:751-759. [PMID: 30848493 DOI: 10.1002/1873-3468.13353] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 12/15/2022]
Abstract
Long noncoding RNAs (lncRNAs) are critical regulators in tumorigenesis. However, their roles in breast cancer remain unclear. Here, we found that lncRNA LINC00473 is significantly upregulated in breast cancer cells. Loss- or gain-of-function experiments show that LINC00473 promotes cell proliferation. Mechanistically, LINC00473 is required for the activation of cyclin D1 (CCND1) expression through recruitment of phosphorylated CREB and histone acetylation to the CCND1 promoter. Interestingly, we found that LINC00473 is also required for maintaining the expression levels of the noncoding RNACCND1 s and recruiting corepressor FUS to the CCND1 promoter. Altogether, the activation effect of LINC00473 on CCND1 is a net effect of two antagonistic regulatory pathways. Our finding provides a novel lncRNA-mediated precise transcriptional control of CCND1.
Collapse
Affiliation(s)
- Xiangmin Shi
- Hefei National Laboratory for Physical Sciences at the Microscale, Hefei, Anhui, China.,Department of Molecular and Cell Biology, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiangting Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, Hefei, Anhui, China.,Department of Molecular and Cell Biology, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
22
|
FGF-induced Pea3 transcription factors program the genetic landscape for cell fate determination. PLoS Genet 2018; 14:e1007660. [PMID: 30188892 PMCID: PMC6143274 DOI: 10.1371/journal.pgen.1007660] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/18/2018] [Accepted: 08/27/2018] [Indexed: 12/01/2022] Open
Abstract
FGF signaling is a potent inducer of lacrimal gland development in the eye, capable of transforming the corneal epithelium into glandular tissues. Here, we show that genetic ablation of the Pea3 family of transcription factors not only disrupted the ductal elongation and branching of the lacrimal gland, but also biased the lacrimal gland epithelium toward an epidermal cell fate. Analysis of high-throughput gene expression and chromatin immunoprecipitation data revealed that the Pea3 genes directly control both the positive and negative feedback loops of FGF signaling. Importantly, Pea3 genes are also required to suppress aberrant Notch signaling which, if gone unchecked, can compromise lacrimal gland development by preventing the expression of both Sox and Six family genes. These results demonstrate that Pea3 genes are key FGF early response transcriptional factors, programing the genetic landscape for cell fate determination. FGF signaling regulates cell fate decision by inducing genome-wide changes in gene expression. We identified Pea3 family transcription factors as the key effectors of FGF signaling in reprograming the epithelia transcriptome. Pea3 factors control both the feedback and feedforward circuities of FGF signaling in lacrimal gland development. They also activate specific expression of Six and Sox family genes and suppress aberrant activation of Notch signaling. In the absence of Pea3 genes, the lacrimal gland progenitors become epidermal-like in their gene expression patterns. The study of Pea3 function resolves the long standing conundrum of how FGF induces the lacrimal gland fate, providing direction for regenerating the lacrimal gland to treat dry eye diseases.
Collapse
|
23
|
Lu Z, Miao Z, Zhu J, Zhu G. ETS-domain containing protein (Elk1) suppression protects cortical neurons against oxygen-glucose deprivation injury. Exp Cell Res 2018; 371:42-49. [PMID: 30053446 DOI: 10.1016/j.yexcr.2018.07.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022]
Abstract
ETS-domain containing protein (Elk1), which is a transcription factor, is reported to be closely related to the apoptosis of primary neurons and could be activated by hypoxia in human microvascular endothelial cells. In this study, we aimed to investigate the role of Elk1 in cortical neurons under oxygen-glucose deprivation (OGD) conditions. The OGD model of cortical neurons was established the anoxia/hypoglycemia-induced injury and the in vivo model was established by middle cerebral artery occlusion (MCAO). Elk1 mRNA and protein expression was significantly up-regulated in neurons exposed to OGD for 24 h, and mRNA expression was also markedly increased in cerebral cortex of rats with MCAO after 10 days. The knockdown of Elk1 in neurons without OGD obviously constrained Fra-1 and promoted Nrf2 expression. Also, Elk1 inhibition suppressed neuronal apoptosis, caspase-3 activity, LDH leakage, and MDA and SOD contents, while it increased cell viability in the neurons with OGD. The overexpression of Fra-1 showed a reverse effect on caspase-3 activity, cell viability and SOD contents in neurons under OGD conditions compared with Elk1 knockdown. Thus, Elk1 inhibition has a protective effect on neurons against OGD-induced injury.
Collapse
Affiliation(s)
- Zhaofeng Lu
- Department of Emergency, First Affiliated Hospital, Henan University of Science and Technology, Luoyang 471000, China
| | - Zhuang Miao
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Jian Zhu
- Department of Neurosurgery, The First People's Hospital of Yancheng( The Fourth Affiliated Hospital of Nantong University), Yancheng 224006, China
| | - Gangyi Zhu
- Department of Emergency, First Affiliated Hospital, Henan University of Science and Technology, Luoyang 471000, China; Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China; Department of Neurosurgery, The First People's Hospital of Yancheng( The Fourth Affiliated Hospital of Nantong University), Yancheng 224006, China
| |
Collapse
|
24
|
Increased expression of EHF contributes to thyroid tumorigenesis through transcriptionally regulating HER2 and HER3. Oncotarget 2018; 7:57978-57990. [PMID: 27517321 PMCID: PMC5295405 DOI: 10.18632/oncotarget.11154] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 07/27/2016] [Indexed: 12/19/2022] Open
Abstract
E26 transformation-specific (ETS) transcription factor EHF plays a tumor suppressor role in prostate cancer and esophageal squamous cell carcinoma (ESCC), whereas it is overexpressed and may act as an oncogene in ovarian and mammary cancers. However, its biological role in thyroid cancer remains totally unknown. The aim of this study was to explore the biological functions of EHF and its potential as a therapeutic target in thyroid cancer. Using quantitative RT-PCR (qRT-PCR) assay, we evaluated mRNA expression of EHF in a cohort of primary papillary thyroid cancers (PTCs) and matched non-cancerous thyroid tissues. The functions of knockdown and ectopic expression of EHF in thyroid cancer cells were determine by a series of in vitro and in vivo experiments. Moreover, dual-luciferase reporter and chromatin immunoprecipitation (ChIP) assays were performed to identify its downstream targets. Our data showed that EHF expression was significantly increased in PTCs compared with matched non-cancerous thyroid tissues. EHF knockdown significantly inhibited thyroid cancer cell proliferation, colony formation, migration, invasion and tumorigenic potential in nude mice and induced cell cycle arrested and apoptosis by modulating the PI3K/Akt and MAPK/Erk signaling pathways. On the other hand, ectopic expression of EHF in thyroid cancer cells notably promoted cell growth and invasiveness. Importantly, EHF was identified as a new transcription factor for HER2 and HER3, contributing to thyroid tumorigenesis. Altogether, our findings suggest that EHF is a novel functional oncogene in thyroid cancer by transcriptionally regulating HER2 and HER3, and may represent a potential therapeutic target for this cancer.
Collapse
|
25
|
Vallarelli AF, Rachakonda PS, André J, Heidenreich B, Riffaud L, Bensussan A, Kumar R, Dumaz N. TERT promoter mutations in melanoma render TERT expression dependent on MAPK pathway activation. Oncotarget 2018; 7:53127-53136. [PMID: 27449293 PMCID: PMC5288173 DOI: 10.18632/oncotarget.10634] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/09/2016] [Indexed: 11/25/2022] Open
Abstract
The mechanism of telomerase re-activation in cancer had remained elusive until the discovery of frequent mutations in the promoter of the TERT gene that encodes the catalytic reverse transcriptase subunit of telomerase. We investigated the regulation of TERT expression in melanoma cell lines and our results show that promoter mutations render TERT expression dependent on MAPK activation due to oncogenic BRAF or NRAS mutations. Mutations in the TERT promoter create binding sites for ETS transcription factors. ETS1, expressed in melanoma cell lines, undergoes activating phosphorylation by ERK at Thr38 residue as a consequence of constitutively activated MAPK pathway. We demonstrate that ETS1 binds on the mutated TERT promoter leading to the re-expression of the gene. The inhibition of ETS1 resulted in reduced TERT expression. We provide evidence that the TERT promoter mutations provide a direct link between TERT expression and MAPK pathway activation due to BRAF or NRAS mutations via the transcription factor ETS1.
Collapse
Affiliation(s)
- Andrelou F Vallarelli
- INSERM, U976, Skin Research Centre, Hôpital Saint-Louis, Paris, F-75010, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS976, Paris, F-75010, France.,Department of Internal Medicine, School of Medicine Sciences, State University of Campinas (UNICAMP), 13083-970, Campinas, SP, Brazil
| | | | - Jocelyne André
- INSERM, U976, Skin Research Centre, Hôpital Saint-Louis, Paris, F-75010, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS976, Paris, F-75010, France
| | - Barbara Heidenreich
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Laurence Riffaud
- INSERM, U976, Skin Research Centre, Hôpital Saint-Louis, Paris, F-75010, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS976, Paris, F-75010, France
| | - Armand Bensussan
- INSERM, U976, Skin Research Centre, Hôpital Saint-Louis, Paris, F-75010, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS976, Paris, F-75010, France
| | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Nicolas Dumaz
- INSERM, U976, Skin Research Centre, Hôpital Saint-Louis, Paris, F-75010, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS976, Paris, F-75010, France
| |
Collapse
|
26
|
Wang V, Davis DA, Yarchoan R. Identification of functional hypoxia inducible factor response elements in the human lysyl oxidase gene promoter. Biochem Biophys Res Commun 2017. [PMID: 28624448 DOI: 10.1016/j.bbrc.2017.06.066] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Human lysyl oxidase (LOX) is a hypoxia-responsive gene whose product catalyzes collagen crosslinking and is thought to be important in cancer metastasis and osteoarthritis. We previously demonstrated that LOX was upregulated by hypoxia inducible factor 2 (HIF-2) more strongly than hypoxia inducible 1 (HIF-1). Here, we further investigated the response of the LOX gene and LOX promoter to HIFs. LOX mRNA, measured by real time reverse transcriptase-PCR, was strongly up-regulated (almost 40-fold), by transfection of HEK-293T cells with a plasmid encoding the HIF-2α subunit of HIF-2, but only three-fold by a plasmid encoding HIF-1α. LOX protein was detectable by Western blot of cells transfected with HIF-2α, but not with HIF-1α. Analysis of a 1487 bp promoter sequence upstream of the human LOX gene revealed 9 potential hypoxia response elements (HREs). Promoter truncation allowed the mapping of two previously unidentified functional HREs, called here HRE8 and HRE7; -455 to -451 and -382 to -386 bp, respectively, upstream of the start codon for LOX. Removal or mutation of these HREs led to a substantial reduction in both HIF-1α and HIF-2α responsiveness. Also, expression of LOX was significantly inhibited by a small molecule specific HIF-2 inhibitor. In conclusion, LOX is highly responsive to HIF-2α and this is largely mediated by two previously unidentified HREs. These observations enhance our understanding of the regulation of this important gene involved in cancer and osteoarthritis, and suggest that these conditions may be targeted by HIF-2 inhibitors.
Collapse
Affiliation(s)
- Victoria Wang
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - David A Davis
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
27
|
Sizemore GM, Pitarresi JR, Balakrishnan S, Ostrowski MC. The ETS family of oncogenic transcription factors in solid tumours. Nat Rev Cancer 2017; 17:337-351. [PMID: 28450705 DOI: 10.1038/nrc.2017.20] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Findings over the past decade have identified aberrant activation of the ETS transcription factor family throughout all stages of tumorigenesis. Specifically in solid tumours, gene rearrangement and amplification, feed-forward growth factor signalling loops, formation of gain-of-function co-regulatory complexes and novel cis-acting mutations in ETS target gene promoters can result in increased ETS activity. In turn, pro-oncogenic ETS signalling enhances tumorigenesis through a broad mechanistic toolbox that includes lineage specification and self-renewal, DNA damage and genome instability, epigenetics and metabolism. This Review discusses these different mechanisms of ETS activation and subsequent oncogenic implications, as well as the clinical utility of ETS factors.
Collapse
Affiliation(s)
- Gina M Sizemore
- The Comprehensive Cancer Center, The Ohio State University
- Department of Cancer Biology and Genetics, The Ohio State University, 598 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, Ohio 43210, USA
| | - Jason R Pitarresi
- The Comprehensive Cancer Center, The Ohio State University
- Department of Cancer Biology and Genetics, The Ohio State University, 598 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, Ohio 43210, USA
| | - Subhasree Balakrishnan
- The Comprehensive Cancer Center, The Ohio State University
- Department of Cancer Biology and Genetics, The Ohio State University, 598 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, Ohio 43210, USA
| | - Michael C Ostrowski
- The Comprehensive Cancer Center, The Ohio State University
- Department of Cancer Biology and Genetics, The Ohio State University, 598 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, Ohio 43210, USA
| |
Collapse
|
28
|
Alterations in leukocyte transcriptional control pathway activity associated with major depressive disorder and antidepressant treatment. Transl Psychiatry 2016; 6:e821. [PMID: 27219347 PMCID: PMC5070063 DOI: 10.1038/tp.2016.79] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 03/23/2016] [Accepted: 03/31/2016] [Indexed: 12/20/2022] Open
Abstract
Major depressive disorder (MDD) is associated with a significantly elevated risk of developing serious medical illnesses such as cardiovascular disease, immune impairments, infection, dementia and premature death. Previous work has demonstrated immune dysregulation in subjects with MDD. Using genome-wide transcriptional profiling and promoter-based bioinformatic strategies, we assessed leukocyte transcription factor (TF) activity in leukocytes from 20 unmedicated MDD subjects versus 20 age-, sex- and ethnicity-matched healthy controls, before initiation of antidepressant therapy, and in 17 of the MDD subjects after 8 weeks of sertraline treatment. In leukocytes from unmedicated MDD subjects, bioinformatic analysis of transcription control pathway activity indicated an increased transcriptional activity of cAMP response element-binding/activating TF (CREB/ATF) and increased activity of TFs associated with cellular responses to oxidative stress (nuclear factor erythroid-derived 2-like 2, NFE2l2 or NRF2). Eight weeks of antidepressant therapy was associated with significant reductions in Hamilton Depression Rating Scale scores and reduced activity of NRF2, but not in CREB/ATF activity. Several other transcriptional regulation pathways, including the glucocorticoid receptor (GR), nuclear factor kappa-B cells (NF-κB), early growth response proteins 1-4 (EGR1-4) and interferon-responsive TFs, showed either no significant differences as a function of disease or treatment, or activities that were opposite to those previously hypothesized to be involved in the etiology of MDD or effective treatment. Our results suggest that CREB/ATF and NRF2 signaling may contribute to MDD by activating immune cell transcriptome dynamics that ultimately influence central nervous system (CNS) motivational and affective processes via circulating mediators.
Collapse
|
29
|
Nagore E, Heidenreich B, Rachakonda S, Garcia-Casado Z, Requena C, Soriano V, Frank C, Traves V, Quecedo E, Sanjuan-Gimenez J, Hemminki K, Landi MT, Kumar R. TERT promoter mutations in melanoma survival. Int J Cancer 2016; 139:75-84. [PMID: 26875008 DOI: 10.1002/ijc.30042] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/03/2016] [Accepted: 02/08/2016] [Indexed: 12/20/2022]
Abstract
Despite advances in targeted therapies, the treatment of advanced melanoma remains an exercise in disease management, hence a need for biomarkers for identification of at-risk primary melanoma patients. In this study, we aimed to assess the prognostic value of TERT promoter mutations in primary melanomas. Tumors from 300 patients with stage I/II melanoma were sequenced for TERT promoter and BRAF/NRAS mutations. Cumulative curves were drawn for patients with and without mutations with progression-free and melanoma-specific survival as outcomes. Cox proportional hazard regression models were used to determine the effect of the mutations on survivals. Individually, presence of TERT promoter and BRAF/NRAS mutations associated with poor disease-free and melanoma-specific survival with modification of the effect by the rs2853669 polymorphism within the TERT promoter. Hazard ratio (HR) for simultaneous occurrence of TERT promoter and BRAF/NRAS mutations for disease-free survival was 2.3 (95% CI 1.2-4.4) and for melanoma-specific survival 5.8 (95% CI 1.9-18.3). The effect of the mutations on melanoma-specific survival in noncarriers of variant allele of the polymorphism was significant (HR 4.5, 95% CI 1.4-15.2) but could not be calculated for the carriers due to low number of events. The variant allele per se showed association with increased survival (HR 0.3, 95% CI 0.1-0.9). The data in this study provide preliminary evidence that TERT promoter mutations in combination with BRAF/NRAS mutations can be used to identify patients at risk of aggressive disease and the possibility of refinement of the classification with inclusion of the rs2853669 polymorphism within TERT promoter.
Collapse
Affiliation(s)
- Eduardo Nagore
- Department of Dermatology, Instituto Valenciano De Oncologia, Valencia, Spain
| | - Barbara Heidenreich
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | | | - Zaida Garcia-Casado
- Laboratory of Molecular Biology, Instituto Valenciano De Oncologia, Valencia, Spain
| | - Celia Requena
- Department of Dermatology, Instituto Valenciano De Oncologia, Valencia, Spain
| | - Virtudes Soriano
- Medical Oncology Department, Instituto Valenciano De Oncologia, Valencia, Spain
| | - Christoph Frank
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Victor Traves
- Department of Pathology, Instituto Valenciano De Oncologia, Valencia, Spain
| | - Esther Quecedo
- Department of Dermatology, Hospital Arnau De Vilanova De Valencia, Valencia, Spain
| | | | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Maria Teresa Landi
- Division of Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, Bethesda, MD
| | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
30
|
Ewing-like sarcoma with CIC-DUX4 gene fusion in a patient with neurofibromatosis type 1. A hitherto unreported association. Pathol Res Pract 2015; 211:877-82. [DOI: 10.1016/j.prp.2015.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 07/18/2015] [Accepted: 08/14/2015] [Indexed: 01/28/2023]
|
31
|
Nabet B, Ó Broin P, Reyes JM, Shieh K, Lin CY, Will CM, Popovic R, Ezponda T, Bradner JE, Golden AA, Licht JD. Deregulation of the Ras-Erk Signaling Axis Modulates the Enhancer Landscape. Cell Rep 2015; 12:1300-13. [PMID: 26279576 DOI: 10.1016/j.celrep.2015.06.078] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 05/11/2015] [Accepted: 06/23/2015] [Indexed: 01/05/2023] Open
Abstract
Unrestrained receptor tyrosine kinase (RTK) signaling and epigenetic deregulation are root causes of tumorigenesis. We establish linkage between these processes by demonstrating that aberrant RTK signaling unleashed by oncogenic HRas(G12V) or loss of negative feedback through Sprouty gene deletion remodels histone modifications associated with active typical and super-enhancers. However, although both lesions disrupt the Ras-Erk axis, the expression programs, enhancer signatures, and transcription factor networks modulated upon HRas(G12V) transformation or Sprouty deletion are largely distinct. Oncogenic HRas(G12V) elevates histone 3 lysine 27 acetylation (H3K27ac) levels at enhancers near the transcription factor Gata4 and the kinase Prkcb, as well as their expression levels. We show that Gata4 is necessary for the aberrant gene expression and H3K27ac marking at enhancers, and Prkcb is required for the oncogenic effects of HRas(G12V)-driven cells. Taken together, our findings demonstrate that dynamic reprogramming of the cellular enhancer landscape is a major effect of oncogenic RTK signaling.
Collapse
Affiliation(s)
- Behnam Nabet
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Pilib Ó Broin
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jaime M Reyes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Kevin Shieh
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Charles Y Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Christine M Will
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Relja Popovic
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Teresa Ezponda
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Aaron A Golden
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Mathematical Sciences, Yeshiva University, New York, NY 10033, USA
| | - Jonathan D Licht
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
32
|
Cheng Z, Guo J, Chen L, Luo N, Yang W, Qu X. Knockdown of EHF inhibited the proliferation, invasion and tumorigenesis of ovarian cancer cells. Mol Carcinog 2015; 55:1048-59. [PMID: 26258986 DOI: 10.1002/mc.22349] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 05/06/2015] [Accepted: 05/15/2015] [Indexed: 12/31/2022]
Abstract
Ovarian cancer is the most lethal gynecologic malignancy worldwide. ETS homologous factor (EHF), a member of E26 transformation specific (ETS) transcription factors, has been reported overexpressed in ovarian cancer. However, the molecular mechanism underlying the biological function of EHF in ovarian cancer is still unclear. Here, we found that EHF was elevated in ovarian cancer tissues compared with non-tumorous tissues. Moreover, high EHF expression level was correlated with short survival time of patients with ovarian cancer. Knockdown of EHF in ovarian cancer cells, SKOV3 and OVCAR3, significantly inhibited cell proliferation and increased cells population in G1 phase. The proteins promoting cell cycles (Cyclin B1, Cyclin D1, and PCNA) were down-regulated and the protein negatively regulating cell cycle progression (P21) was up-regulated after EHF knockdown. Moreover, inhibition of EHF in ovarian cancer cells dramatically induced cell apoptosis, but impaired cell adhesion and cell invasion. Furthermore, phosphorylation levels of ERK and AKT were notably reduced in EHF knockdown cells. Finally, in vivo data showed that knockdown of EHF inhibited tumor growth in nude mice. Our data indicates that EHF could be a potential prognosis marker for ovarian cancer and work as an oncogene by targeting ERK and AKT signaling, which can serve as a new target for ovarian cancer treatment. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zhongping Cheng
- Department of Gynecology & Obstetrics, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Jing Guo
- Department of Gynecology & Obstetrics, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Li Chen
- Department of Gynecology & Obstetrics, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Ning Luo
- Department of Gynecology & Obstetrics, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Weihong Yang
- Department of Gynecology & Obstetrics, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoyan Qu
- Department of Gynecology & Obstetrics, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.,Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
33
|
Cooper CDO, Newman JA, Aitkenhead H, Allerston CK, Gileadi O. Structures of the Ets Protein DNA-binding Domains of Transcription Factors Etv1, Etv4, Etv5, and Fev: DETERMINANTS OF DNA BINDING AND REDOX REGULATION BY DISULFIDE BOND FORMATION. J Biol Chem 2015; 290:13692-709. [PMID: 25866208 PMCID: PMC4447949 DOI: 10.1074/jbc.m115.646737] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Indexed: 12/31/2022] Open
Abstract
Ets transcription factors, which share the conserved Ets DNA-binding domain, number nearly 30 members in humans and are particularly involved in developmental processes. Their deregulation following changes in expression, transcriptional activity, or by chromosomal translocation plays a critical role in carcinogenesis. Ets DNA binding, selectivity, and regulation have been extensively studied; however, questions still arise regarding binding specificity outside the core GGA recognition sequence and the mode of action of Ets post-translational modifications. Here, we report the crystal structures of Etv1, Etv4, Etv5, and Fev, alone and in complex with DNA. We identify previously unrecognized features of the protein-DNA interface. Interactions with the DNA backbone account for most of the binding affinity. We describe a highly coordinated network of water molecules acting in base selection upstream of the GGAA core and the structural features that may account for discrimination against methylated cytidine residues. Unexpectedly, all proteins crystallized as disulfide-linked dimers, exhibiting a novel interface (distant to the DNA recognition helix). Homodimers of Etv1, Etv4, and Etv5 could be reduced to monomers, leading to a 40–200-fold increase in DNA binding affinity. Hence, we present the first indication of a redox-dependent regulatory mechanism that may control the activity of this subset of oncogenic Ets transcription factors.
Collapse
Affiliation(s)
- Christopher D O Cooper
- From the Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Joseph A Newman
- From the Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Hazel Aitkenhead
- From the Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Charles K Allerston
- From the Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Opher Gileadi
- From the Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
34
|
Selvaraj N, Kedage V, Hollenhorst PC. Comparison of MAPK specificity across the ETS transcription factor family identifies a high-affinity ERK interaction required for ERG function in prostate cells. Cell Commun Signal 2015; 13:12. [PMID: 25885538 PMCID: PMC4338625 DOI: 10.1186/s12964-015-0089-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/04/2015] [Indexed: 01/01/2023] Open
Abstract
Background The RAS/MAPK signaling pathway can regulate gene expression by phosphorylating and altering the function of some, but not all, ETS transcription factors. ETS family transcription factors bind similar DNA sequences and can compete for genomic binding sites. However, MAPK regulation varies across the ETS family. Therefore, changing the ETS factor bound to a cis-regulatory element can alter MAPK regulation of gene expression. To understand RAS/MAPK regulated gene expression programs, comprehensive knowledge of the ETS family members that are MAPK targets and relative MAPK targeting efficiency across the family is needed. Results An in vitro kinase assay was used to rank-order 27 human ETS family transcription factors based on phosphorylation by ERK2, JNK1, and p38α. Many novel MAPK targets and specificities were identified within the ETS family, including the identification of the prostate cancer oncoprotein ERG as a specific target of ERK2. ERK2 phosphorylation of ERG S215 required a DEF docking domain and was necessary for ERG to activate transcription of cell migration genes and promote prostate cell migration. The ability of ERK2 to bind ERG with higher affinity than ETS1 provided a potential molecular explanation for why ERG overexpression drives migration of prostate cells with low levels of RAS/ERK signaling, while ETS1 has a similar function only when RAS/ERK signaling is high. Conclusions The rank ordering of ETS transcription factors as MAPK targets provides an important resource for understanding ETS proteins as mediators of MAPK signaling. This is emphasized by the difference in rank order of ERG and ETS1, which allows these factors to have distinct roles based on the level of RAS/ERK signaling present in the cell. Electronic supplementary material The online version of this article (doi:10.1186/s12964-015-0089-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nagarathinam Selvaraj
- Medical Sciences, Indiana University School of Medicine, 1001 E 3rd St, Bloomington, IN, 47405, USA.
| | - Vivekananda Kedage
- Medical Sciences, Indiana University School of Medicine, 1001 E 3rd St, Bloomington, IN, 47405, USA.
| | - Peter C Hollenhorst
- Medical Sciences, Indiana University School of Medicine, 1001 E 3rd St, Bloomington, IN, 47405, USA.
| |
Collapse
|
35
|
Tsoyi K, Geldart AM, Christou H, Liu X, Chung SW, Perrella MA. Elk-3 is a KLF4-regulated gene that modulates the phagocytosis of bacteria by macrophages. J Leukoc Biol 2014; 97:171-80. [PMID: 25351511 DOI: 10.1189/jlb.4a0214-087r] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
ETS family proteins play a role in immune responses. A unique member of this family, Elk-3, is a transcriptional repressor that regulates the expression of HO-1. Elk-3 is very sensitive to the effects of inflammatory mediators and is down-regulated by bacterial endotoxin (LPS). In the present study, exposure of mouse macrophages to Escherichia coli LPS resulted in decreased, full-length, and splice-variant isoforms of Elk-3. We isolated the Elk-3 promoter and demonstrated that LPS also decreased promoter activity. The Elk-3 promoter contains GC-rich regions that are putative binding sites for zinc-finger transcription factors, such as Sp1 and KLFs. Mutation of the GC-rich region from bp -613 to -603 blunted LPS-induced down-regulation of the Elk-3 promoter. Similar to the LPS response, coexpression of KLF4 led to repression of Elk-3 promoter activity, whereas coexpression of Sp1 increased activity. ChIP assays revealed that KLF4 binding to the Elk-3 promoter was increased by LPS exposure, and Sp1 binding was decreased. Thus, down-regulation of Elk-3 by bacterial LPS is regulated, in part, by the transcriptional repressor KLF4. Overexpression of Elk-3, in the presence of E. coli bacteria, resulted in decreased macrophage phagocytosis. To determine whether limited expression of HO-1 may contribute to this response, we exposed HO-1-deficient bone marrow-derived macrophages to E. coli and found a comparable reduction in bacterial phagocytosis. These data suggest that down-regulation of Elk-3 and the subsequent induction of HO-1 are important for macrophage function during the inflammatory response to infection.
Collapse
Affiliation(s)
- Konstantin Tsoyi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Adriana M Geldart
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA; Division of Newborn Medicine, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA; and
| | - Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA; Division of Newborn Medicine, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA; and
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Su Wol Chung
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and School of Biological Sciences, University of Ulsan, South Korea
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
36
|
Plotnik JP, Budka JA, Ferris MW, Hollenhorst PC. ETS1 is a genome-wide effector of RAS/ERK signaling in epithelial cells. Nucleic Acids Res 2014; 42:11928-40. [PMID: 25294825 PMCID: PMC4231772 DOI: 10.1093/nar/gku929] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The RAS/ERK pathway is commonly activated in carcinomas and promotes oncogenesis by altering transcriptional programs. However, the array of cis-regulatory elements and trans-acting factors that mediate these transcriptional changes is still unclear. Our genome-wide analysis determined that a sequence consisting of neighboring ETS and AP-1 transcription factor binding sites is enriched near cell migration genes activated by RAS/ERK signaling in epithelial cells. In vivo screening of candidate ETS proteins revealed that ETS1 is specifically required for migration of RAS/ERK activated cells. Furthermore, both migration and transcriptional activation through ETS/AP-1 required ERK phosphorylation of ETS1. Genome-wide mapping of multiple ETS proteins demonstrated that ETS1 binds specifically to enhancer ETS/AP-1 sequences. ETS1 occupancy, and its role in cell migration, was conserved in epithelial cells derived from multiple tissues, consistent with a chromatin organization common to epithelial cell lines. Genome-wide expression analysis showed that ETS1 was required for activation of RAS-regulated cell migration genes, but also identified a surprising role for ETS1 in the repression of genes such as DUSP4, DUSP6 and SPRY4 that provide negative feedback to the RAS/ERK pathway. Consistently, ETS1 was required for robust RAS/ERK pathway activation. Therefore, ETS1 has dual roles in mediating epithelial-specific RAS/ERK transcriptional functions.
Collapse
Affiliation(s)
- Joshua P Plotnik
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| | - 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
| | - Peter C Hollenhorst
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana, USA
| |
Collapse
|
37
|
Etv1 and Ewsr1 cooperatively regulate limb mesenchymal Fgf10 expression in response to apical ectodermal ridge-derived fibroblast growth factor signal. Dev Biol 2014; 394:181-90. [DOI: 10.1016/j.ydbio.2014.07.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 07/02/2014] [Accepted: 07/30/2014] [Indexed: 11/21/2022]
|
38
|
MAP kinase activity supported by BRAF (V600E) mutation rather than gene amplification is associated with ETV1 expression in melanoma brain metastases. Arch Dermatol Res 2014; 306:873-84. [PMID: 25073704 DOI: 10.1007/s00403-014-1490-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 07/15/2014] [Accepted: 07/21/2014] [Indexed: 12/19/2022]
Abstract
In primary melanoma, ETV1 transcription factor was suggested to be activated mainly by gene amplification and to promote tumor growth in cooperation with BRAF (V600E) . Aim of this study was to investigate ETV1 expression in human melanoma with a focus on brain metastases. We investigated ETV1 in 68 human melanoma brain metastases using FISH for ETV1 gene (located at chromosome 7p21) and centromere chromosome 7 and immunohistochemistry for ETV1, BRAF (V600E) , and ETV1/BRAF associated proteins pMSK1, pRSK1, pp38, pMEK1/2, MAPKAP kinase 2, CIC, HIF-1alpha and Ki-67. We further studied ETV1 copy number variations in 32 melanoma cell lines from primary and metastatic lesions using array CGH. The influence of the MAP kinase pathway activity on ETV1 mRNA and protein expression under BRAF wild-type and BRAF (V600E) conditions were determined in melanoma cell lines using qRT-PCR and Western Blot. No ETV1 high grade amplifications were observed in tissue samples, but low grade ETV1 gene amplifications were found in 7 (10.3 %) melanoma brain metastases. ETV1 protein expression in tissue samples (15 %) correlated with BRAF (V600E) status (p = 0.007) and HIF-1alpha expression (p = 0.049), but not with ETV1 gene dose. Application of the BRAF(V600E)-specific inhibitor vemurafenib and the BRAF(V6ooE/V600K)-inhibitor dabrafenib revealed predominant regulation of ETV-1 mRNA and protein via MAPK-pathway. ETV1 expression is a rare event in human melanoma and seems to be rather based on hyperactivation of MAPK signals, by BRAF (V600E) mutation, than on ETV1 gene amplification. Consequently, therapeutic inhibition of BRAF and the downstream MAPK pathway also down-regulates oncogenic ETV1 expression.
Collapse
|
39
|
Findlay VJ, LaRue AC, Turner DP, Watson PM, Watson DK. Understanding the role of ETS-mediated gene regulation in complex biological processes. Adv Cancer Res 2014; 119:1-61. [PMID: 23870508 DOI: 10.1016/b978-0-12-407190-2.00001-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ets factors are members of one of the largest families of evolutionarily conserved transcription factors, regulating critical functions in normal cell homeostasis, which when perturbed contribute to tumor progression. The well-documented alterations in ETS factor expression and function during cancer progression result in pleiotropic effects manifested by the downstream effect on their target genes. Multiple ETS factors bind to the same regulatory sites present on target genes, suggesting redundant or competitive functions. The anti- and prometastatic signatures obtained by examining specific ETS regulatory networks will significantly improve our ability to accurately predict tumor progression and advance our understanding of gene regulation in cancer. Coordination of multiple ETS gene functions also mediates interactions between tumor and stromal cells and thus contributes to the cancer phenotype. As such, these new insights may provide a novel view of the ETS gene family as well as a focal point for studying the complex biological control involved in tumor progression. One of the goals of molecular biology is to elucidate the mechanisms that contribute to the development and progression of cancer. Such an understanding of the molecular basis of cancer will provide new possibilities for: (1) earlier detection, as well as better diagnosis and staging of disease; (2) detection of minimal residual disease recurrences and evaluation of response to therapy; (3) prevention; and (4) novel treatment strategies. Increased understanding of ETS-regulated biological pathways will directly impact these areas.
Collapse
Affiliation(s)
- Victoria J Findlay
- Department of Pathology and Laboratory Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | |
Collapse
|
40
|
Regulation of the human thioredoxin gene promoter and its key substrates: a study of functional and putative regulatory elements. Biochim Biophys Acta Gen Subj 2013; 1840:303-14. [PMID: 24041992 DOI: 10.1016/j.bbagen.2013.09.013] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Revised: 07/26/2013] [Accepted: 09/06/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND The thioredoxin system maintains redox balance through the action of thioredoxin and thioredoxin reductase. Thioredoxin regulates the activity of various substrates, including those that function to counteract cellular oxidative stress. These include the peroxiredoxins, methionine sulfoxide reductase A and specific transcription factors. Of particular relevance is Redox Factor-1, which in turn activates other redox-regulated transcription factors. SCOPE OF REVIEW Experimentally defined transcription factor binding sites in the human thioredoxin and thioredoxin reductase gene promoters together with promoters of the major thioredoxin system substrates involved in regulating cellular redox status are discussed. An in silico approach was used to identify potential putative binding sites for these transcription factors in all of these promoters. MAJOR CONCLUSIONS Our analysis reveals that many redox gene promoters contain the same transcription factor binding sites. Several of these transcription factors are in turn redox regulated. The ARE is present in several of these promoters and is bound by Nrf2 during various oxidative stress stimuli to upregulate gene expression. Other transcription factors also bind to these promoters during the same oxidative stress stimuli, with this redundancy supporting the importance of the antioxidant response. Putative transcription factor sites were identified in silico, which in combination with specific regulatory knowledge for that gene promoter may inform future experiments. GENERAL SIGNIFICANCE Redox proteins are involved in many cellular signalling pathways and aberrant expression can lead to disease or other pathological conditions. Therefore understanding how their expression is regulated is relevant for developing therapeutic agents that target these pathways.
Collapse
|
41
|
Kumar S, Maiti S. Effect of different arginine methylations on the thermodynamics of Tat peptide binding to HIV-1 TAR RNA. Biochimie 2013; 95:1422-31. [PMID: 23541506 DOI: 10.1016/j.biochi.2013.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 03/18/2013] [Indexed: 11/18/2022]
Abstract
RNA-binding proteins are an important class of mediators that regulate cell function and differentiation. Methylation of arginine, a post-translational modification (PTM) found in these proteins, can modulate their function. Arginine can be monomethylated or dimethylated, depending on the type of methyl transferases involved. This paper describes a comparative study of the thermodynamics of unmodified and modified Tat peptide interaction with TAR RNA, where the peptide is methylated at epsilon (ɛ) and eta (η) nitrogen atoms of guanidinium group of arginine side chain at position 52 or 53. The results indicate that monomethylation of arginine at epsilon (ɛ) nitrogen atom enhances binding affinity, owing to a more favourable enthalpy component which overrides the less favourable entropy change. In contrast, monomethylation of arginine residue at η nitrogen results in reduced binding affinity originating exclusively from a less favourable enthalpy change leaving entropic component unaffected. However, in case of simultaneous methylation at ɛ and η positions, the binding parameters remain almost unaffected, when compared to the unmodified peptide. In case of symmetric dimethylation at η position the observed enthalpy change of the binding was found to be smaller than the values obtained for the unmodified peptide. Asymmetric dimethylation at η position showed the most reduced binding affinities owing to less favourable enthalpy changes. These results provide insights that enable elucidation of the biological outcome of arginine methylation as PTMs that regulate protein function, and will contribute to our understanding of how these PTMs are established in vitro and in vivo.
Collapse
Affiliation(s)
- Santosh Kumar
- Proteomics and Structural Biology Unit, Institute of Genomics and Integrative Biology, CSIR, Mall Road, Delhi 110 007, India
| | | |
Collapse
|
42
|
Sprater F, Hovden AO, Appel S. Expression of ESE-3 isoforms in immunogenic and tolerogenic human monocyte-derived dendritic cells. PLoS One 2012. [PMID: 23185370 PMCID: PMC3501485 DOI: 10.1371/journal.pone.0049577] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Dendritic cells (DC) are the only hematopoietic cells expressing the epithelial specific Ets transcription factor ESE-3. Here we analyzed presence and quantity of isoforms ESE-3a, ESE-3b and ESE-3j in various immunogenic and tolerogenic human monocyte-derived DC (moDC) and blood DC populations using quantitative real time PCR and immunoblot analyses. ESE-3a and ESE-3b were detectable in all moDC populations with ESE-3b being the main transcript. ESE-3b expression was upregulated in immunogenic moDC and downregulated in tolerogenic moDC compared to immature moDC. ESE-3a had similar transcript levels in immature and immunogenic moDC and had very low levels in tolerogenic moDC. In blood DC populations only splice variant ESE-3b was detectable. ESE-3j was not detectable in any of the DC populations. These findings suggest that ESE-3b is the functionally most important ESE-3 isoform in DC.
Collapse
Affiliation(s)
- Florian Sprater
- Broegelmann Research Laboratory, The Gade Institute, University of Bergen, Bergen, Norway
| | - Arnt-Ove Hovden
- Broegelmann Research Laboratory, The Gade Institute, University of Bergen, Bergen, Norway
| | - Silke Appel
- Broegelmann Research Laboratory, The Gade Institute, University of Bergen, Bergen, Norway
- * E-mail:
| |
Collapse
|
43
|
Lelli KM, Slattery M, Mann RS. Disentangling the many layers of eukaryotic transcriptional regulation. Annu Rev Genet 2012; 46:43-68. [PMID: 22934649 DOI: 10.1146/annurev-genet-110711-155437] [Citation(s) in RCA: 165] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Regulation of gene expression in eukaryotes is an extremely complex process. In this review, we break down several critical steps, emphasizing new data and techniques that have expanded current gene regulatory models. We begin at the level of DNA sequence where cis-regulatory modules (CRMs) provide important regulatory information in the form of transcription factor (TF) binding sites. In this respect, CRMs function as instructional platforms for the assembly of gene regulatory complexes. We discuss multiple mechanisms controlling complex assembly, including cooperative DNA binding, combinatorial codes, and CRM architecture. The second section of this review places CRM assembly in the context of nucleosomes and condensed chromatin. We discuss how DNA accessibility and histone modifications contribute to TF function. Lastly, new advances in chromosomal mapping techniques have provided increased understanding of intra- and interchromosomal interactions. We discuss how these topological maps influence gene regulatory models.
Collapse
Affiliation(s)
- Katherine M Lelli
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | |
Collapse
|
44
|
Do PM, Varanasi L, Fan S, Li C, Kubacka I, Newman V, Chauhan K, Daniels SR, Boccetta M, Garrett MR, Li R, Martinez LA. Mutant p53 cooperates with ETS2 to promote etoposide resistance. Genes Dev 2012; 26:830-45. [PMID: 22508727 DOI: 10.1101/gad.181685.111] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mutant p53 (mtp53) promotes chemotherapy resistance through multiple mechanisms, including disabling proapoptotic proteins and regulating gene expression. Comparison of genome wide analysis of mtp53 binding revealed that the ETS-binding site motif (EBS) is prevalent within predicted mtp53-binding sites. We demonstrate that mtp53 regulates gene expression through EBS in promoters and that ETS2 mediates the interaction with this motif. Importantly, we identified TDP2, a 5'-tyrosyl DNA phosphodiesterase involved in the repair of DNA damage caused by etoposide, as a transcriptional target of mtp53. We demonstrate that suppression of TDP2 sensitizes mtp53-expressing cells to etoposide and that mtp53 and TDP2 are frequently overexpressed in human lung cancer; thus, our analysis identifies a potentially "druggable" component of mtp53's gain-of-function activity.
Collapse
Affiliation(s)
- Phi M Do
- Department of Biochemistry, University of Mississippi Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hollenhorst PC. RAS/ERK pathway transcriptional regulation through ETS/AP-1 binding sites. Small GTPases 2012; 3:154-8. [PMID: 22653334 DOI: 10.4161/sgtp.19630] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The RAS/RAF/MEK/ERK signaling pathway is activated by mutation in many cancers. Neighboring ETS and AP-1 DNA binding sequences can act as response elements for transcriptional activation by this pathway. ERK phosphorylation of an ETS transcription factor is one mechanism of activating the RAS/ERK gene expression program that can promote cancer cell phenotypes such as proliferation, invasion, and metastasis. Recent genome-wide mapping of ETS proteins over-expressed by chromosomal rearrangement in prostate cancer reveals a second mechanism for activation of this gene expression program. An oncogenic subset of ETS transcription factors can activate RAS/ERK target genes even in the absence of RAS/ERK pathway activation by binding ETS/AP-1 sequences. Thus, regulation of cancer cell invasion and metastasis via ETS/AP-1 sequence elements depends on which ETS protein is bound, and the status of the RAS/ERK pathway. This commentary will focus on what is known about the selectivity of ETS/AP-1 sequences for different ETS transcription factors and the transcriptional consequences of ETS protein selection.
Collapse
Affiliation(s)
- Peter C Hollenhorst
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN, USA.
| |
Collapse
|
46
|
Fu YP, Kohaar I, Rothman N, Earl J, Figueroa JD, Ye Y, Malats N, Tang W, Liu L, Garcia-Closas M, Muchmore B, Chatterjee N, Tarway M, Kogevinas M, Porter-Gill P, Baris D, Mumy A, Albanes D, Purdue MP, Hutchinson A, Carrato A, Tardón A, Serra C, García-Closas R, Lloreta J, Johnson A, Schwenn M, Karagas MR, Schned A, Diver WR, Gapstur SM, Thun MJ, Virtamo J, Chanock SJ, Fraumeni JF, Silverman DT, Wu X, Real FX, Prokunina-Olsson L. Common genetic variants in the PSCA gene influence gene expression and bladder cancer risk. Proc Natl Acad Sci U S A 2012; 109:4974-9. [PMID: 22416122 PMCID: PMC3324016 DOI: 10.1073/pnas.1202189109] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Genome-wide association studies have identified a SNP, rs2294008, on 8q24.3 within the prostate stem cell antigen (PSCA) gene, as a risk factor for bladder cancer. To fine-map this region, we imputed 642 SNPs within 100 Kb of rs2294008 in addition to 33 markers genotyped in one of the reported genome-wide association study in 8,652 subjects. A multivariable logistic regression model adjusted for rs2294008 revealed a unique signal, rs2978974 (r(2) = 0.02, D' = 0.19 with rs2294008). In the combined analysis of 5,393 cases and 7,324 controls, we detected a per-allele odds ratio (OR) = 1.11 [95% confidence interval (CI) = 1.06-1.17, P = 5.8 × 10(-5)] for rs2294008 and OR = 1.07 (95% CI = 1.02-1.13, P = 9.7 × 10(-3)) for rs2978974. The effect was stronger in carriers of both risk variants (OR = 1.24, 95% CI = 1.08-1.41, P = 1.8 × 10(-3)) and there was a significant multiplicative interaction (P = 0.035) between these two SNPs, which requires replication in future studies. The T risk allele of rs2294008 was associated with increased PSCA mRNA expression in two sets of bladder tumor samples (n = 36, P = 0.0007 and n = 34, P = 0.0054) and in normal bladder samples (n = 35, P = 0.0155), but rs2978974 was not associated with PSCA expression. SNP rs2978974 is located 10 Kb upstream of rs2294008, within an alternative untranslated first exon of PSCA. The non-risk allele G of rs2978974 showed strong interaction with nuclear proteins from five cell lines tested, implying a regulatory function. In conclusion, a joint effect of two PSCA SNPs, rs2294008 and rs2978974, suggests that both variants may be important for bladder cancer susceptibility, possibly through different mechanisms that influence the control of mRNA expression and interaction with regulatory factors.
Collapse
MESH Headings
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Cell Line, Tumor
- DNA, Neoplasm/metabolism
- Electrophoretic Mobility Shift Assay
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Genetic Association Studies
- Genetic Markers
- Genetic Predisposition to Disease
- Humans
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Physical Chromosome Mapping
- Polymorphism, Single Nucleotide/genetics
- Protein Binding
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Recombination, Genetic/genetics
- Risk Factors
- Sequence Analysis, RNA
- Urinary Bladder Neoplasms/genetics
Collapse
Affiliation(s)
- Yi-Ping Fu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, and
| | - Indu Kohaar
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, and
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Julie Earl
- Epithelial Carcinogenesis Group, Molecular Pathology Programme, Centro Nacional de Investigaciones Oncológicas, Madrid 28029, Spain
| | - Jonine D. Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yuanqing Ye
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid 28029, Spain
| | - Wei Tang
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, and
| | - Luyang Liu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, and
| | - Montserrat Garcia-Closas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
- Division of Genetics and Epidemiology, Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Brian Muchmore
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, and
| | - Nilanjan Chatterjee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - McAnthony Tarway
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, and
| | - Manolis Kogevinas
- Centre for Research in Environmental Epidemiology, Barcelona 08003, Spain
- Municipal Institute of Medical Research, Barcelona 08003, Spain
- Centro de Investiación de Biomédica en Red Epidemiología y Salud Pública, Barcelona 08003, Spain
- National School of Public Health, Athens 11521, Greece
| | - Patricia Porter-Gill
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, and
| | - Dalsu Baris
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Adam Mumy
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, and
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Mark P. Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Amy Hutchinson
- Core Genotyping Facility, SAIC-Frederick, Inc., National Cancer Institute at Frederick, Frederick, MD 21702
| | | | - Adonina Tardón
- Municipal Institute of Medical Research, Barcelona 08003, Spain
- Instituto Universitario de Oncología, Universidad de Oviedo, Oviedo 33003, Spain
| | - Consol Serra
- Departament de Ciències Experimentals i de la Salut, and
| | - Reina García-Closas
- Unidad de Investigación, Hospital Universitario de Canarias, La Laguna 38320, Spain
| | - Josep Lloreta
- Hospital del Mar-Institut Municipal d'Investigació Mèdica, Universitat Pompeu Fabra, Barcelona 08003, Spain
| | | | | | | | | | - W. Ryan Diver
- Epidemiology Research Program, American Cancer Society, Atlanta, GA 30303; and
| | - Susan M. Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, GA 30303; and
| | - Michael J. Thun
- Epidemiology Research Program, American Cancer Society, Atlanta, GA 30303; and
| | - Jarmo Virtamo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Stephen J. Chanock
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, and
| | - Joseph F. Fraumeni
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Debra T. Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xifeng Wu
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Francisco X. Real
- Epithelial Carcinogenesis Group, Molecular Pathology Programme, Centro Nacional de Investigaciones Oncológicas, Madrid 28029, Spain
- Departament de Ciències Experimentals i de la Salut, and
| | | |
Collapse
|
47
|
Hollenhorst PC, Ferris MW, Hull MA, Chae H, Kim S, Graves BJ. Oncogenic ETS proteins mimic activated RAS/MAPK signaling in prostate cells. Genes Dev 2011; 25:2147-57. [PMID: 22012618 DOI: 10.1101/gad.17546311] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The aberrant expression of an oncogenic ETS transcription factor is implicated in the progression of the majority of prostate cancers, 40% of melanomas, and most cases of gastrointestinal stromal tumor and Ewing's sarcoma. Chromosomal rearrangements in prostate cancer result in overexpression of any one of four ETS transcription factors. How these four oncogenic ETS genes differ from the numerous other ETS genes expressed in normal prostate and contribute to tumor progression is not understood. We report that these oncogenic ETS proteins, but not other ETS factors, enhance prostate cell migration. Genome-wide binding analysis matched this specific biological function to occupancy of a unique set of genomic sites highlighted by the presence of ETS- and AP-1-binding sequences. ETS/AP-1-binding sequences are prototypical RAS-responsive elements, but oncogenic ETS proteins activated a RAS/MAPK transcriptional program in the absence of MAPK activation. Thus, overexpression of oncogenic ETS proteins can replace RAS/MAPK pathway activation in prostate cells. The genomic description of this ETS/AP-1-regulated, RAS-responsive, gene expression program provides a resource for understanding the role of these ETS factors in both an oncogenic setting and the developmental processes where these genes normally function.
Collapse
|
48
|
Steffan JJ, Koul HK. Prostate derived ETS factor (PDEF): A putative tumor metastasis suppressor. Cancer Lett 2011; 310:109-17. [DOI: 10.1016/j.canlet.2011.06.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 06/03/2011] [Accepted: 06/12/2011] [Indexed: 01/31/2023]
|
49
|
Hollenhorst PC, McIntosh LP, Graves BJ. Genomic and biochemical insights into the specificity of ETS transcription factors. Annu Rev Biochem 2011; 80:437-71. [PMID: 21548782 DOI: 10.1146/annurev.biochem.79.081507.103945] [Citation(s) in RCA: 375] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
ETS proteins are a group of evolutionarily related, DNA-binding transcriptional factors. These proteins direct gene expression in diverse normal and disease states by binding to specific promoters and enhancers and facilitating assembly of other components of the transcriptional machinery. The highly conserved DNA-binding ETS domain defines the family and is responsible for specific recognition of a common sequence motif, 5'-GGA(A/T)-3'. Attaining specificity for biological regulation in such a family is thus a conundrum. We present the current knowledge of routes to functional diversity and DNA binding specificity, including divergent properties of the conserved ETS and PNT domains, the involvement of flanking structured and unstructured regions appended to these dynamic domains, posttranslational modifications, and protein partnerships with other DNA-binding proteins and coregulators. The review emphasizes recent advances from biochemical and biophysical approaches, as well as insights from genomic studies that detect ETS-factor occupancy in living cells.
Collapse
Affiliation(s)
- Peter C Hollenhorst
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana 47405, USA.
| | | | | |
Collapse
|
50
|
Meadows SM, Myers CT, Krieg PA. Regulation of endothelial cell development by ETS transcription factors. Semin Cell Dev Biol 2011; 22:976-84. [PMID: 21945894 DOI: 10.1016/j.semcdb.2011.09.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 09/14/2011] [Indexed: 10/17/2022]
Abstract
The ETS family of transcription factors plays an essential role in controlling endothelial gene expression. Multiple members of the ETS family are expressed in the developing endothelium and evidence suggests that the proteins function, to some extent, redundantly. However, recent studies have demonstrated a crucial non-redundant role for ETV2, as a primary player in specification and differentiation of the endothelial lineage. Here, we review the contribution of ETS factors, and their partner proteins, to the regulation of embryonic vascular development.
Collapse
Affiliation(s)
- Stryder M Meadows
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, United States
| | | | | |
Collapse
|