FOXF1 Defines the Core-Regulatory Circuitry in Gastrointestinal Stromal Tumor

Total Page:16

File Type:pdf, Size:1020Kb

FOXF1 Defines the Core-Regulatory Circuitry in Gastrointestinal Stromal Tumor Published OnlineFirst November 21, 2017; DOI: 10.1158/2159-8290.CD-17-0468 RESEARCH ARTICLE FOXF1 Defi nes the Core-Regulatory Circuitry in Gastrointestinal Stromal Tumor Leili Ran 1 , Yuedan Chen 1 , 2 , Jessica Sher 1 , Elissa W.P. Wong 1 , Devan Murphy 1 , Jenny Q. Zhang 1 , Dan Li1 , Kemal Deniz 3 , Inna Sirota 4 , Zhen Cao 1 , 2 , Shangqian Wang 1 , Youxin Guan 1 , Shipra Shukla 1 , Katie Yang Li5 , Alan Chramiec 5 , 6 , Yuanyuan Xie 1 , Deyou Zheng 7 , 8 , 9 , Richard P. Koche 5 , Cristina R. Antonescu 10 , Yu Chen 1 , 2 , 11 , 12 , and Ping Chi 1 , 2 , 11 , 12 ABSTRACT The cellular context that integrates upstream signaling and downstream nuclear response dictates the oncogenic behavior and shapes treatment responses in distinct cancer types. Here, we uncover that in gastrointestinal stromal tumor (GIST), the forkhead family member FOXF1 directly controls the transcription of two master regulators, KIT and ETV1 , both required for GIST precursor-interstitial cells of Cajal lineage specifi cation and GIST tumorigenesis. Further, FOXF1 colocalizes with ETV1 at enhancers and functions as a pioneer factor that regulates the ETV1-dependent GIST lineage-specifi c transcriptome through modulation of the local chromatin con- text, including chromatin accessibility, enhancer maintenance, and ETV1 binding. Functionally, FOXF1 is required for human GIST cell growth in vitro and murine GIST tumor growth and maintenance in vivo . The simultaneous control of the upstream signaling and nuclear response sets up a unique regulatory paradigm and highlights the critical role of FOXF1 in enforcing the GIST cellular context for highly lineage-restricted clinical behavior and treatment response. SIGNIFICANCE: We uncover that FOXF1 defi nes the core-regulatory circuitry in GIST through both direct transcriptional regulation and pioneer factor function. The unique and simultaneous control of signaling and transcriptional circuitry by FOXF1 sets up an enforced transcriptional addiction to FOXF1 in GIST, which can be exploited diagnostically and therapeutically. Cancer Discov; 8(2); 234–51. ©2017 AACR. See related commentary by Lee and Duensing, p. 146. 1 Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Memorial Sloan Kettering Cancer Center, New York, New York. 12 Depart- Cancer Center, New York, New York . 2 Weill Cornell Graduate School of ment of Medicine, Weill Cornell Medical College, New York, New York. 3 Medical Sciences, Cornell University, New York, New York. Department of Note: Supplementary data for this article are available at Cancer Discovery 4 Pathology, Erciyes University, Kayseri, Turkey. Department of Molecular Online (http://cancerdiscovery.aacrjournals.org/). Biology and Genetics, Cornell University, Ithaca, New York. 5 Center of Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, L. Ran and Y. Chen are the co–fi rst authors of this article. New York. 6 Biomedical Engineering, Columbia University, New York, New Corresponding Authors: P. Chi , Memorial Sloan Kettering Cancer Center, York. 7 Department of Genetics, Albert Einstein College of Medicine, Bronx, 1275 York Avenue, New York, NY 10065. Phone: 646-888-3338; Fax: New York. 8 Department of Neurology, Albert Einstein College of Medicine, 646-888-3406; E-mail: [email protected] ; and Y. Chen, [email protected] 9 Bronx, New York. Department of Neuroscience, Albert Einstein College of doi: 10.1158/2159-8290.CD-17-0468 Medicine, Bronx, New York. 10 Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York. 11 Department of Medicine, © 2017 American Association for Cancer Research. 234 | CANCER DISCOVERY FEBRUARY 2018 www.aacrjournals.org Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 21, 2017; DOI: 10.1158/2159-8290.CD-17-0468 INTRODUCTION and melanoma, where it regulates distinct transcriptional programs (1, 9–11). The enhancer landscape of accessible Gastrointestinal stromal tumor (GIST) is one of the most chromatin defines cellular lineage and the distinct cis- common subtypes of human soft-tissue sarcoma. GIST arises trome and transcriptional output of individual transcrip- from the interstitial cells of Cajal (ICC), a cell lineage that tion factors in different cell types. We thus speculate that requires KIT, the principal signaling regulator, and ETV1, a additional master regulator(s) may function as “pioneer lineage-specific master transcription factor, for lineage speci- factor(s)” that modulate chromatin accessibility and help fication and survival (1–3). Physiologically, normal levels of define and maintain the cistrome of ETV1, analogous to KIT activation by the KIT ligand stabilize the ETV1 protein the pioneer function of FOXA1 to androgen receptor (AR) through active downstream MAPK signaling, and result in in prostate cancer and estrogen receptor-α in breast can- physiologic transcriptional output critical for ICC lineage cer (12–17). Here, we describe the discovery of FOXF1, as specification and development. GIST is characterized by fre- a novel ICC/GIST lineage-specific master regulator that quent activating mutations in KIT. Mutant KIT aberrantly directly regulates KIT, ETV1 expression, and the ICC/GIST activates downstream MAPK signaling, which stabilizes the lineage-specific transcriptome. Moreover, FOXF1 functions ETV1 protein, and stabilized ETV1 in turn enhances mutant as a pioneer factor required to maintain open chromatin and KIT expression. Therefore, mutant KIT and ETV1 form a ETV1 binding at many lineage-specific ETV1-binding sites. positive feedback loop and cooperate in GIST oncogenesis We further demonstrate that FOXF1 functionally is required (4). The lineage-specific expression ofKIT and ETV1 and their for GIST cell growth and survival in vitro and GIST tumor interplay in GIST underline the exquisite therapeutic sen- growth and maintenance in genetically engineered mouse sitivity and clinical success of targeting the lineage depend- models. Overall, our data demonstrate a unique regulatory ence on KIT and ETV1 (5–8). However, how KIT and ETV1 hierarchy of FOXF1 that distinguishes itself from other pio- are regulated and what defines the cellular context in GIST neer factors, e.g., FOXA1, in that beyond chromatin context remain unclear. modulation and active recruitment of ETV1, it also directly In addition to GIST, ETV1 is involved in the tumori- controls the expression of ETV1 and the cooperative signaling genesis of multiple cancer types, including prostate cancer factor KIT. FEBRUARY 2018 CANCER DISCOVERY | 235 Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst November 21, 2017; DOI: 10.1158/2159-8290.CD-17-0468 RESEARCH ARTICLE Ran et al. RESULTS prostate cancer and breast cancer (Fig. 1D). However, FOXA1 expression is low in GIST tumors as well as cell lines (Fig. 1D; FOXF1 Is Nearly Universally and Uniquely Supplementary Fig. S1A and S1B). Expressed in Human GISTs We thus speculate that a different FOX family transcription To identify critical factor(s) that regulate the lineage-spe- factor is involved in the modulation of the ETV1 cistrome cific cellular context for oncogenic transformation, we focused in GIST. We examined the expression of all FOX factors our initial analyses on ETV1, a transcription factor that drives and uncovered that FOXF1 is the highest in both absolute tumorigenesis in two distinct cancer types: prostate cancer expression and significance of differential expression in GIST and GIST (1, 9, 10). We generated genome-wide localization of compared with other cancer types (Fig. 1E; Supplementary ETV1 by chromatin immunoprecipitation sequencing (ChIP- Fig. S1A and Supplementary Table S5). We further examined seq) in two human GIST cell lines (GIST-T1 and GIST48) and RNA-seq profiles of GIST48 and GIST882 cells and observed two prostate cancer cell lines that harbor aberrant expression that FOXF1 was the highest-expressing FOX family member of full-length ETV1 due to translocation of its entire cod- (Supplementary Fig. S1B). We confirmed the presence of ing locus (LNCaP and MDA-PCa2b; refs. 1, 9, 10, 18–20). FOXF1 protein in all three human GIST cell lines, but not in ETV1 cistrome analyses demonstrated that the majority of the negative control melanoma cell lines (OMIM1.3, A375, the ETV1 promoter binding sites (TSS ± 1 kb) were shared and A2058; Fig. 1F). Furthermore, we examined FOXF1 pro- between prostate cancer and GIST, whereas the majority of tein expression in tissue microarrays (TMA) of GIST and nonpromoter (referred as “enhancer” hereafter) binding sites several other sarcoma subtypes from MSK-archived tumor were distinct between the two cancer types (Fig. 1A and B). specimens by immunohistochemistry (IHC). Independent Unsupervised k-means clustering divided enhancer ETV1- review by two sarcoma pathologists confirmed positive FOXF1 binding sites into three distinct clusters of GIST-specific (C1), staining in >98% of all human GIST samples regardless of KIT/ prostate-specific (C2), and shared (C3) sites. This is consistent PDGFRA mutational status, but rarely in other sarcoma sub- with previous observation that enhancer landscape is more types, including myxofibrosarcoma, myxoid liposarcoma, and lineage-specific than promoter (12, 14, 15, 17, 21–24). The synovial sarcoma (Fig. 1G and H). These data demonstrate that observation that ETV1 binds to distinct
Recommended publications
  • Chromatin State Barriers Enforce an Irreversible Mammalian Cell Fate Decision
    bioRxiv preprint doi: https://doi.org/10.1101/2021.05.12.443709; this version posted May 14, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Chromatin state barriers… Blanco et al. 2021 Chromatin state barriers enforce an irreversible mammalian cell fate decision M. Andrés Blanco1,19,*,†,, David B. Sykes6,8,19, Lei Gu2,15,17,18,19, Mengjun Wu2,4,15, Ricardo Petroni1, Rahul Karnik7,8,9, Mathias Wawer10, Joshua Rico1, Haitao Li1, William D. Jacobus2,12,15, Ashwini Jambhekar2,15,11, Sihem Cheloufi5, Alexander Meissner7,8,9,13, Konrad Hochedlinger6,7,8,14, David T. Scadden6,8,9,*, and Yang Shi2,3,* 1 Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA 2 Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA 3 Ludwig Institute for Cancer Research, Oxford Branch, Oxford University, UK 4 Current address: The Bioinformatics Centre, Department of Biology and Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark 5 Department of Biochemistry, Stem Cell Center, University of California, Riverside, Riverside, CA 92521, USA. 6 Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA. 7 Broad Institute of MIT and Harvard, Cambridge, MA, USA 8 Harvard Stem Cell Institute, Cambridge, Massachusetts,
    [Show full text]
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Figure S1. Representative Report Generated by the Ion Torrent System Server for Each of the KCC71 Panel Analysis and Pcafusion Analysis
    Figure S1. Representative report generated by the Ion Torrent system server for each of the KCC71 panel analysis and PCaFusion analysis. (A) Details of the run summary report followed by the alignment summary report for the KCC71 panel analysis sequencing. (B) Details of the run summary report for the PCaFusion panel analysis. A Figure S1. Continued. Representative report generated by the Ion Torrent system server for each of the KCC71 panel analysis and PCaFusion analysis. (A) Details of the run summary report followed by the alignment summary report for the KCC71 panel analysis sequencing. (B) Details of the run summary report for the PCaFusion panel analysis. B Figure S2. Comparative analysis of the variant frequency found by the KCC71 panel and calculated from publicly available cBioPortal datasets. For each of the 71 genes in the KCC71 panel, the frequency of variants was calculated as the variant number found in the examined cases. Datasets marked with different colors and sample numbers of prostate cancer are presented in the upper right. *Significantly high in the present study. Figure S3. Seven subnetworks extracted from each of seven public prostate cancer gene networks in TCNG (Table SVI). Blue dots represent genes that include initial seed genes (parent nodes), and parent‑child and child‑grandchild genes in the network. Graphical representation of node‑to‑node associations and subnetwork structures that differed among and were unique to each of the seven subnetworks. TCNG, The Cancer Network Galaxy. Figure S4. REVIGO tree map showing the predicted biological processes of prostate cancer in the Japanese. Each rectangle represents a biological function in terms of a Gene Ontology (GO) term, with the size adjusted to represent the P‑value of the GO term in the underlying GO term database.
    [Show full text]
  • A Dissertation Entitled the Androgen Receptor
    A Dissertation entitled The Androgen Receptor as a Transcriptional Co-activator: Implications in the Growth and Progression of Prostate Cancer By Mesfin Gonit Submitted to the Graduate Faculty as partial fulfillment of the requirements for the PhD Degree in Biomedical science Dr. Manohar Ratnam, Committee Chair Dr. Lirim Shemshedini, Committee Member Dr. Robert Trumbly, Committee Member Dr. Edwin Sanchez, Committee Member Dr. Beata Lecka -Czernik, Committee Member Dr. Patricia R. Komuniecki, Dean College of Graduate Studies The University of Toledo August 2011 Copyright 2011, Mesfin Gonit This document is copyrighted material. Under copyright law, no parts of this document may be reproduced without the expressed permission of the author. An Abstract of The Androgen Receptor as a Transcriptional Co-activator: Implications in the Growth and Progression of Prostate Cancer By Mesfin Gonit As partial fulfillment of the requirements for the PhD Degree in Biomedical science The University of Toledo August 2011 Prostate cancer depends on the androgen receptor (AR) for growth and survival even in the absence of androgen. In the classical models of gene activation by AR, ligand activated AR signals through binding to the androgen response elements (AREs) in the target gene promoter/enhancer. In the present study the role of AREs in the androgen- independent transcriptional signaling was investigated using LP50 cells, derived from parental LNCaP cells through extended passage in vitro. LP50 cells reflected the signature gene overexpression profile of advanced clinical prostate tumors. The growth of LP50 cells was profoundly dependent on nuclear localized AR but was independent of androgen. Nevertheless, in these cells AR was unable to bind to AREs in the absence of androgen.
    [Show full text]
  • Integrative Epigenomic and Genomic Analysis of Malignant Pheochromocytoma
    EXPERIMENTAL and MOLECULAR MEDICINE, Vol. 42, No. 7, 484-502, July 2010 Integrative epigenomic and genomic analysis of malignant pheochromocytoma Johanna Sandgren1,2* Robin Andersson3*, pression examination in a malignant pheochromocy- Alvaro Rada-Iglesias3, Stefan Enroth3, toma sample. The integrated analysis of the tumor ex- Goran̈ Akerstro̊ m̈ 1, Jan P. Dumanski2, pression levels, in relation to normal adrenal medulla, Jan Komorowski3,4, Gunnar Westin1 and indicated that either histone modifications or chromo- somal alterations, or both, have great impact on the ex- Claes Wadelius2,5 pression of a substantial fraction of the genes in the in- vestigated sample. Candidate tumor suppressor 1Department of Surgical Sciences genes identified with decreased expression, a Uppsala University, Uppsala University Hospital H3K27me3 mark and/or in regions of deletion were for SE-75185 Uppsala, Sweden 2 instance TGIF1, DSC3, TNFRSF10B, RASSF2, HOXA9, Department of Genetics and Pathology Rudbeck Laboratory, Uppsala University PTPRE and CDH11. More genes were found with in- SE-75185 Uppsala, Sweden creased expression, a H3K4me3 mark, and/or in re- 3The Linnaeus Centre for Bioinformatics gions of gain. Potential oncogenes detected among Uppsala University those were GNAS, INSM1, DOK5, ETV1, RET, NTRK1, SE-751 24 Uppsala, Sweden IGF2, and the H3K27 trimethylase gene EZH2. Our ap- 4Interdisciplinary Centre for Mathematical and proach to associate histone methylations and DNA Computational Modelling copy number changes to gene expression revealed ap- Warsaw University parent impact on global gene transcription, and en- PL-02-106 Warszawa, Poland abled the identification of candidate tumor genes for 5Corresponding author: Tel, 46-18-471-40-76; further exploration.
    [Show full text]
  • Saethre–Chotzen Syndrome Caused by TWIST 1 Gene Mutations: Functional Differentiation from Muenke Coronal Synostosis Syndrome
    European Journal of Human Genetics (2006) 14, 39–48 & 2006 Nature Publishing Group All rights reserved 1018-4813/06 $30.00 www.nature.com/ejhg ARTICLE Saethre–Chotzen syndrome caused by TWIST 1 gene mutations: functional differentiation from Muenke coronal synostosis syndrome Wolfram Kress*,1, Christian Schropp2, Gabriele Lieb2, Birgit Petersen2, Maria Bu¨sse-Ratzka2, Ju¨rgen Kunz3, Edeltraut Reinhart4, Wolf-Dieter Scha¨fer5, Johanna Sold5, Florian Hoppe6, Jan Pahnke6, Andreas Trusen7, Niels So¨rensen8,Ju¨rgen Krauss8 and Hartmut Collmann8 1Institute of Human Genetics, University of Wu¨rzburg, Wu¨rzburg, Germany; 2Department of Pediatrics, University of Wu¨rzburg, Wu¨rzburg, Germany; 3Institute of Human Genetics, University of Marburg, Marburg, Germany; 4Department of Maxillo-facial Surgery, University of Wu¨rzburg, Wu¨rzburg, Germany; 5Department of Ophthalmology, University of Wu¨rzburg, Wu¨rzburg, Germany; 6Department of Otorhinolaryngology, University of Wu¨rzburg, Wu¨rzburg, Germany; 7Department of Diagnostic Radiology, University of Wu¨rzburg, Wu¨rzburg, Germany; 8Sect. Pediatric Neurosurgery, University of Wu¨rzburg, Wu¨rzburg, Germany The Saethre–Chotzen syndrome (SCS) is an autosomal dominant craniosynostosis syndrome with uni- or bilateral coronal synostosis and mild limb deformities. It is caused by loss-of-function mutations of the TWIST 1 gene. In an attempt to delineate functional features separating SCS from Muenke’s syndrome, we screened patients presenting with coronal suture synostosis for mutations in the TWIST 1 gene, and for the Pro250Arg mutation in FGFR3. Within a total of 124 independent pedigrees, 39 (71 patients) were identified to carry 25 different mutations of TWIST 1 including 14 novel mutations, to which six whole gene deletions were added.
    [Show full text]
  • Engineered Type 1 Regulatory T Cells Designed for Clinical Use Kill Primary
    ARTICLE Acute Myeloid Leukemia Engineered type 1 regulatory T cells designed Ferrata Storti Foundation for clinical use kill primary pediatric acute myeloid leukemia cells Brandon Cieniewicz,1* Molly Javier Uyeda,1,2* Ping (Pauline) Chen,1 Ece Canan Sayitoglu,1 Jeffrey Mao-Hwa Liu,1 Grazia Andolfi,3 Katharine Greenthal,1 Alice Bertaina,1,4 Silvia Gregori,3 Rosa Bacchetta,1,4 Norman James Lacayo,1 Alma-Martina Cepika1,4# and Maria Grazia Roncarolo1,2,4# Haematologica 2021 Volume 106(10):2588-2597 1Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA; 2Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA; 3San Raffaele Telethon Institute for Gene Therapy, Milan, Italy and 4Center for Definitive and Curative Medicine, Stanford School of Medicine, Stanford, CA, USA *BC and MJU contributed equally as co-first authors #AMC and MGR contributed equally as co-senior authors ABSTRACT ype 1 regulatory (Tr1) T cells induced by enforced expression of interleukin-10 (LV-10) are being developed as a novel treatment for Tchemotherapy-resistant myeloid leukemias. In vivo, LV-10 cells do not cause graft-versus-host disease while mediating graft-versus-leukemia effect against adult acute myeloid leukemia (AML). Since pediatric AML (pAML) and adult AML are different on a genetic and epigenetic level, we investigate herein whether LV-10 cells also efficiently kill pAML cells. We show that the majority of primary pAML are killed by LV-10 cells, with different levels of sensitivity to killing. Transcriptionally, pAML sensitive to LV-10 killing expressed a myeloid maturation signature.
    [Show full text]
  • A Small Molecule Inhibitor of ETV1, YK-4- 279, Prevents Prostate
    RESEARCH ARTICLE A Small Molecule Inhibitor of ETV1, YK-4- OPEN ACCESS 279, Prevents Prostate Cancer Growth and Citation: Rahim S, Minas T, Hong S-H, Justvig S, C¸ elik H, et al. (2014) A Small Molecule Inhibitor of ETV1, YK-4-279, Prevents Prostate Cancer Metastasis in a Mouse Xenograft Model Growth and Metastasis in a Mouse Xenograft Model. PLoS ONE 9(12): e114260. doi:10.1371/ Said Rahim1, Tsion Minas1, Sung-Hyeok Hong1, Sarah Justvig1, Haydar C¸ elik1, journal.pone.0114260 Yasemin Saygideger Kont1, Jenny Han1, Abraham T. Kallarakal1, Yali Kong1, 2 1 1 1 Editor: Irina U Agoulnik, Florida International Michelle A. Rudek , Milton L. Brown , Bhaskar Kallakury , Jeffrey A. Toretsky , University, United States of America Aykut U¨ ren1* Received: May 21, 2014 1. Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United Accepted: November 5, 2014 States of America, 2. The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States of America Published: December 5, 2014 *[email protected] Copyright: ß 2014 Rahim et al. This is an open- access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and repro- duction in any medium, provided the original author and source are credited. Abstract Data Availability: The authors confirm that all data Background: The erythroblastosis virus E26 transforming sequences (ETS) family underlying the findings are fully available without restriction. All relevant data are within the paper of transcription factors consists of a highly conserved group of genes that play and its Supporting Information files.
    [Show full text]
  • Overview of Research on Fusion Genes in Prostate Cancer
    2011 Review Article Overview of research on fusion genes in prostate cancer Chunjiao Song1,2, Huan Chen3 1Medical Research Center, Shaoxing People’s Hospital, Shaoxing University School of Medicine, Shaoxing 312000, China; 2Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing 312000, China; 3Key Laboratory of Microorganism Technology and Bioinformatics Research of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou 310000, China Contributions: (I) Conception and design: C Song; (II) Administrative support: Shaoxing Municipal Health and Family Planning Science and Technology Innovation Project (2017CX004) and Shaoxing Public Welfare Applied Research Project (2018C30058); (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: C Song; (V) Data analysis and interpretation: H Chen; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors. Correspondence to: Chunjiao Song. No. 568 Zhongxing Bei Road, Shaoxing 312000, China. Email: [email protected]. Abstract: Fusion genes are known to drive and promote carcinogenesis and cancer progression. In recent years, the rapid development of biotechnologies has led to the discovery of a large number of fusion genes in prostate cancer specimens. To further investigate them, we summarized the fusion genes. We searched related articles in PubMed, CNKI (Chinese National Knowledge Infrastructure) and other databases, and the data of 92 literatures were summarized after preliminary screening. In this review, we summarized approximated 400 fusion genes since the first specific fusion TMPRSS2-ERG was discovered in prostate cancer in 2005. Some of these are prostate cancer specific, some are high-frequency in the prostate cancer of a certain ethnic group. This is a summary of scientific research in related fields and suggests that some fusion genes may become biomarkers or the targets for individualized therapies.
    [Show full text]
  • Antagonism of Forkhead Box Subclass O Transcription Factors Elicits Loss of Soluble Guanylyl Cyclase Expression S
    Supplemental material to this article can be found at: http://molpharm.aspetjournals.org/content/suppl/2019/04/15/mol.118.115386.DC1 1521-0111/95/6/629–637$35.00 https://doi.org/10.1124/mol.118.115386 MOLECULAR PHARMACOLOGY Mol Pharmacol 95:629–637, June 2019 Copyright ª 2019 by The Author(s) This is an open access article distributed under the CC BY-NC Attribution 4.0 International license. Antagonism of Forkhead Box Subclass O Transcription Factors Elicits Loss of Soluble Guanylyl Cyclase Expression s Joseph C. Galley, Brittany G. Durgin, Megan P. Miller, Scott A. Hahn, Shuai Yuan, Katherine C. Wood, and Adam C. Straub Heart, Lung, Blood and Vascular Medicine Institute (J.C.G., B.G.D., M.P.M., S.A.H., S.Y., K.C.W., A.C.S.) and Department of Pharmacology and Chemical Biology (J.C.G., A.C.S.), University of Pittsburgh, Pittsburgh, Pennsylvania Received November 29, 2018; accepted March 31, 2019 Downloaded from ABSTRACT Nitric oxide (NO) stimulates soluble guanylyl cyclase (sGC) protein expression showed a concentration-dependent down- activity, leading to elevated intracellular cyclic guano- regulation. Consistent with the loss of sGC a and b mRNA and sine 39,59-monophosphate (cGMP) and subsequent vascular protein expression, pretreatment of vascular smooth muscle smooth muscle relaxation. It is known that downregulation of cells with the FoxO inhibitor decreased sGC activity mea- sGC expression attenuates vascular dilation and contributes to sured by cGMP production following stimulation with an NO molpharm.aspetjournals.org the pathogenesis of cardiovascular disease. However, it is not donor.
    [Show full text]
  • Xo PANEL DNA GENE LIST
    xO PANEL DNA GENE LIST ~1700 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes (at 400 -500x average coverage on tumor) Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4 HOXA11
    [Show full text]
  • Single Cell Transcriptomic Analysis of Human Pluripotent Stem Cell Chondrogenesis
    ARTICLE https://doi.org/10.1038/s41467-020-20598-y OPEN Single cell transcriptomic analysis of human pluripotent stem cell chondrogenesis Chia-Lung Wu1,2,5,6, Amanda Dicks1,2,3,6, Nancy Steward1,2, Ruhang Tang1,2, Dakota B. Katz1,2,3, ✉ Yun-Rak Choi1,2,4 & Farshid Guilak 1,2,3 The therapeutic application of human induced pluripotent stem cells (hiPSCs) for cartilage regeneration is largely hindered by the low yield of chondrocytes accompanied by unpre- 1234567890():,; dictable and heterogeneous off-target differentiation of cells during chondrogenesis. Here, we combine bulk RNA sequencing, single cell RNA sequencing, and bioinformatic analyses, including weighted gene co-expression analysis (WGCNA), to investigate the gene reg- ulatory networks regulating hiPSC differentiation under chondrogenic conditions. We identify specific WNTs and MITF as hub genes governing the generation of off-target differentiation into neural cells and melanocytes during hiPSC chondrogenesis. With heterocellular signaling models, we further show that WNT signaling produced by off-target cells is responsible for inducing chondrocyte hypertrophy. By targeting WNTs and MITF, we eliminate these cell lineages, significantly enhancing the yield and homogeneity of hiPSC-derived chondrocytes. Collectively, our findings identify the trajectories and molecular mechanisms governing cell fate decision in hiPSC chondrogenesis, as well as dynamic transcriptome profiles orches- trating chondrocyte proliferation and differentiation. 1 Dept. of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO 63110, USA. 2 Shriners Hospitals for Children—St. Louis, St. Louis, MO 63110, USA. 3 Dept. of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO 63110, USA. 4 Dept. of Orthopaedic Surgery, Yonsei University, Seoul, South Korea.
    [Show full text]