Heterozygous Deletion of Chromosome 17P Renders Prostate Cancer Vulnerable to Inhibition of RNA Polymerase II

Total Page:16

File Type:pdf, Size:1020Kb

Heterozygous Deletion of Chromosome 17P Renders Prostate Cancer Vulnerable to Inhibition of RNA Polymerase II ARTICLE DOI: 10.1038/s41467-018-06811-z OPEN Heterozygous deletion of chromosome 17p renders prostate cancer vulnerable to inhibition of RNA polymerase II Yujing Li1,2,3, Yunhua Liu 2,3,4, Hanchen Xu1,2,3, Guanglong Jiang3, Kevin Van der Jeught 2,3, Yuanzhang Fang2,3, Zhuolong Zhou3, Lu Zhang2,3, Michael Frieden3, Lifei Wang3, Zhenhua Luo5, Milan Radovich4,6, Bryan P. Schneider7, Yibin Deng8, Yunlong Liu3, Kun Huang7, Bin He9, Jin Wang 10, Xiaoming He 11,12, Xinna Zhang3,4, Guang Ji 1 & Xiongbin Lu2,3,4 1234567890():,; Heterozygous deletion of chromosome 17p (17p) is one of the most frequent genomic events in human cancers. Beyond the tumor suppressor TP53, the POLR2A gene encoding the cat- alytic subunit of RNA polymerase II (RNAP2) is also included in a ~20-megabase deletion region of 17p in 63% of metastatic castration-resistant prostate cancer (CRPC). Using a focused CRISPR-Cas9 screen, we discovered that heterozygous loss of 17p confers a selective dependence of CRPC cells on the ubiquitin E3 ligase Ring-Box 1 (RBX1). RBX1 activates POLR2A by the K63-linked ubiquitination and thus elevates the RNAP2-mediated mRNA synthesis. Combined inhibition of RNAP2 and RBX1 profoundly suppress the growth of CRPC in a synergistic manner, which potentiates the therapeutic effectivity of the RNAP2 inhibitor, α-amanitin-based antibody drug conjugate (ADC). Given the limited therapeutic options for CRPC, our findings identify RBX1 as a potentially therapeutic target for treating human CRPC harboring heterozygous deletion of 17p. 1 Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, China. 2 Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. 3 Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA. 4 Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA. 5 The Liver Care Center and Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA. 6 Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA. 7 Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA. 8 Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN 55912, USA. 9 Biomarker Research Program Center, Houston Methodist Research Institute, Houston, TX 77030, USA. 10 Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA. 11 Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA. 12 Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA. These authors contributed equally: Yujing Li, Yunhua Liu, Hanchen Xu. Correspondence and requests for materials should be addressed to X.Z. (email: [email protected]) or to G.J. (email: [email protected]) or to X.L. (email: [email protected]) NATURE COMMUNICATIONS | (2018) 9:4394 | DOI: 10.1038/s41467-018-06811-z | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-06811-z rostate cancer is among the most common male malig- human 17p13.1) cooperates with Trp53 (mouse orthologue of nancies and one major leading cause of cancer mortality in TP53) deletion to produce more aggressive disease in lymphoma P 1,2 18 fi men . Since the discovery of androgen dependence in and leukemia . In this study, we nd that 63% of metastatic prostate cancer, the backbone therapy for prostate cancer has prostate cancer harbors heterozygous deletion of a region that been to lower androgen levels by surgical castration or androgen- spans up to 20 megabases of DNA at 17p. We demonstrate that deprivation therapy3. However, although many patients initially POLR2A is included in the 17p deletion region along with TP53 respond to androgen deprivation therapy, nearly all the patients in a majority of prostate cancers. POLR2A is the catalytic subunit relapse and eventually develop castration-resistant prostate can- of the RNAP2 complex that is solely in charge of mRNA synthesis cer (CRPC)4. Over the past decade, it has become clear that the in cells. POLR2A and RNAP2 activity is specifically inhibited by androgen receptor (AR) plays a pivotal role in the development of α-amanitin, a small molecule peptide produced by the death cap resistance to hormone therapies in both primary and recurrent mushroom (Amanita phalloides)19,20. Inhibition of POLR2A with prostate cancer. New therapeutic approaches in advanced pros- α-amanitin-based ADC selectively suppresses the proliferation, tate cancer have focused on the AR protein, which led to the survival and tumor growth of CRPC cells harboring this genomic development of AR-targeting agents, abiraterone acetate and event. Using a CRISPR-Cas9 screen, we uncover that hetero- enzalutamide5. zygous deletion of 17p confers a selective dependence on RBX1, Despite the success of androgen deprivation and AR-blocking inhibition of which had a synergistic and robust suppression in therapies, the newly developed therapies also suffer a short the growth of CRPC along with the treatment of α-amanitin- therapeutic durability due to acquired resistance4,6. Thus, conjugated anti-EpCAM antibodies. researchers are now searching for more therapeutic targets, one of fi which is prostate-speci c membrane antigen (PSMA). PSMA is Results highly expressed on the surface of nearly all prostate cancer cells Heterozygous deletion of 17p is frequent in prostate cancer. but is present on only a few normal tissues, making it an excellent TP53, a well-documented tumor suppressor gene, is frequently target for drugs that selectively attack tumors7. Radioactive ele- inactivated by mutation or deletion in a majority of human ment lutetium-177-labeled PSMA antibody has shown promise in – tumors21. However, the mutation and homozygous deletion of treating patients who are resistant to other drug therapies8 10. TP53 are relatively infrequent events in prostate cancer, While it is hard to directly target variant forms of AR or altera- accounting for only 12% (59 out of 492) and 8% (37 out of 492) tions in the AR gene that promote castration resistance, a small of total cases, respectively. In contrast, approximately 26% (126 molecular inhibitor against ROR-γ, an upstream regulator of AR, out of 492) of all prostate tumor samples harbor heterozygous was proven to effectively shut down AR signaling11. In mouse loss of TP53 (Fig. 1a). Our recent studies have shown that het- CRPC models, treatment with ROR-γ inhibitors led to substantial erozygous deletion of TP53 in human colorectal cancer fre- and prolonged regression of tumors, and restored their sensitivity quently encompasses a neighboring essential gene POLR2A, to the treatment of enzalutamide. In-depth understanding of rendering cancer cells with heterozygous loss of TP53 susceptible prostate cancer invasion, metastasis and drug resistance will help to further inhibition of POLR2A22,23. The comprehensive ana- identify more therapeutic targets and lead to new treatment lysis of prostate cancer genomes revealed that the TP53 deletion options. is often included in a large fragment deletion that spans over The whole-genome sequencing of cancer genomes and other almost the whole short arm of chromosome 17 (17p) (Fig. 1b). associated omics efforts have empowered our knowledge of The loss of 17p is frequently detected at relatively late stages of human cancer biology and pathogenesis. These efforts have colorectal cancer development24,25, which may contribute to or facilitated personalized medicine to find new genetic alterations even determine the transition from the early-stage to advanced- in the context of a specific cancer. Comprehensive analyses of key stage cancer. Consistent with the findings in colorectal cancer, genomic changes in prostate cancer will accelerate our progress in analysis of The Cancer Genome Atlas (TCGA) prostate dataset developing more effective ways to diagnose, treat and prevent this revealed that 17p loss is readily detectable in low-grade T1 disease. Recent studies have identified recurrent somatic muta- tumors, but was significantly increased in the advanced T2-T4 tions, copy number variations, and chromosomal rearrangements tumors (Fig. 1c, d). Moreover, the loss of 17p rises up to 63% (95 in prostate cancer12,13. A number of frequent genomic changes out of 150) in metastatic prostate cancer, whereas TP53 mutation are shared by primary and metastatic prostate cancer, including and homozygous deletion account for only 39 and 6% respec- E26 transformation-specific (ETS) fusions, point mutations in tively (Fig. 1e, f). Collectively, these results indicate the 17p loss SPOP, FOXA1, and TP53, and copy number alterations involving may play an important role in prostate cancer progression and MYC, RB1, and PTEN, although these alterations are differentially metastasis. enriched at different stages of prostate cancer12,13. Interestingly, aberrations of BRCA2, BRCA1, and ATM were observed at a much higher frequency in CRPC, indicating the potential appli- RBX1 is an essential gene for 17ploss CRPC cells. In the 17p cation of PARP inhibitors in treating this subset of cancers. In a deletion region, there are as many as 200 protein-coding genes phase II study of olaparib14, a PARP inhibitor, it was proven to and noncoding RNA genes,
Recommended publications
  • Supplementary Materials
    DEPs in osteosarcoma cells comparing to osteoblastic cells Biological Process Protein Percentage of Hits metabolic process (GO:0008152) 29.3 29.3% cellular process (GO:0009987) 20.2 20.2% localization (GO:0051179) 9.4 9.4% biological regulation (GO:0065007) 8 8.0% developmental process (GO:0032502) 7.8 7.8% response to stimulus (GO:0050896) 5.6 5.6% cellular component organization (GO:0071840) 5.6 5.6% multicellular organismal process (GO:0032501) 4.4 4.4% immune system process (GO:0002376) 4.2 4.2% biological adhesion (GO:0022610) 2.7 2.7% apoptotic process (GO:0006915) 1.6 1.6% reproduction (GO:0000003) 0.8 0.8% locomotion (GO:0040011) 0.4 0.4% cell killing (GO:0001906) 0.1 0.1% 100.1% Genes 2179Hits 3870 biological adhesion apoptotic process … reproduction (GO:0000003) , 0.8% (GO:0022610) , 2.7% locomotion (GO:0040011) ,… immune system process cell killing (GO:0001906) , 0.1% (GO:0002376) , 4.2% multicellular organismal process (GO:0032501) , metabolic process 4.4% (GO:0008152) , 29.3% cellular component organization (GO:0071840) , 5.6% response to stimulus (GO:0050896), 5.6% developmental process (GO:0032502) , 7.8% biological regulation (GO:0065007) , 8.0% cellular process (GO:0009987) , 20.2% localization (GO:0051179) , 9.
    [Show full text]
  • A Network Propagation Approach to Prioritize Long Tail Genes in Cancer
    bioRxiv preprint doi: https://doi.org/10.1101/2021.02.05.429983; this version posted February 8, 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. A Network Propagation Approach to Prioritize Long Tail Genes in Cancer Hussein Mohsen1,*, Vignesh Gunasekharan2, Tao Qing2, Sahand Negahban3, Zoltan Szallasi4, Lajos Pusztai2,*, Mark B. Gerstein1,5,6,3,* 1 Computational Biology & Bioinformatics Program, Yale University, New Haven, CT 06511, USA 2 Breast Medical Oncology, Yale School of Medicine, New Haven, CT 06511, USA 3 Department of Statistics & Data Science, Yale University, New Haven, CT 06511, USA 4 Children’s Hospital Informatics Program, Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA 5 Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA 6 Department of Computer Science, Yale University, New Haven, CT 06511, USA * Corresponding author Abstract Introduction. The diversity of genomic alterations in cancer pose challenges to fully understanding the etiologies of the disease. Recent interest in infrequent mutations, in genes that reside in the “long tail” of the mutational distribution, uncovered new genes with significant implication in cancer development. The study of these genes often requires integrative approaches with multiple types of biological data. Network propagation methods have demonstrated high efficacy in uncovering genomic patterns underlying cancer using biological interaction networks. Yet, the majority of these analyses have focused their assessment on detecting known cancer genes or identifying altered subnetworks.
    [Show full text]
  • The Middle Temporal Gyrus Is Transcriptionally Altered in Patients with Alzheimer’S Disease
    1 The middle temporal gyrus is transcriptionally altered in patients with Alzheimer’s Disease. 2 1 3 Shahan Mamoor 1Thomas Jefferson School of Law 4 East Islip, NY 11730 [email protected] 5 6 We sought to understand, at the systems level and in an unbiased fashion, how gene 7 expression was most different in the brains of patients with Alzheimer’s Disease (AD) by mining published microarray datasets (1, 2). Comparing global gene expression profiles between 8 patient and control revealed that a set of 84 genes were expressed at significantly different levels in the middle temporal gyrus (MTG) of patients with Alzheimer’s Disease (1, 2). We used 9 computational analyses to classify these genes into known pathways and existing gene sets, 10 and to describe the major differences in the epigenetic marks at the genomic loci of these genes. While a portion of these genes is computationally cognizable as part of a set of genes 11 up-regulated in the brains of patients with AD (3), many other genes in the gene set identified here have not previously been studied in association with AD. Transcriptional repression, both 12 pre- and post-transcription appears to be affected; nearly 40% of these genes are transcriptional 13 targets of MicroRNA-19A/B (miR-19A/B), the zinc finger protein 10 (ZNF10), or of the AP-1 repressor jun dimerization protein 2 (JDP2). 14 15 16 17 18 19 20 21 22 23 24 25 26 Keywords: Alzheimer’s Disease, systems biology of Alzheimer’s Disease, differential gene 27 expression, middle temporal gyrus.
    [Show full text]
  • Identification of the Key Genes and Pathways in Prostate Cancer
    ONCOLOGY LETTERS 16: 6663-6669, 2018 Identification of the key genes and pathways in prostate cancer SHUTONG FAN1*, ZUMU LIANG1*, ZHIQIN GAO1, ZHIWEI PAN2, SHAOJIE HAN3, XIAOYING LIU1, CHUNLING ZHAO1, WEIWEI YANG1, ZHIFANG PAN1 and WEIGUO FENG1 1College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053; 2Department of Internal Medicine, Laizhou Development Zone Hospital, Yantai, Shandong 261400; 3Animal Epidemic Prevention and Epidemic Control Center, Changle County Bureau of Animal Health and Production, Weifang, Shandong 262400, P.R. China Received March 5, 2018; Accepted September 17, 2018 DOI: 10.3892/ol.2018.9491 Abstract. Prostate cancer (PCa) is one of the most common Introduction malignancies in men globally. The aim of the present study was to identify the key genes and pathways involved in the Prostate cancer (PCa) is one of the most common malignancies occurrence of PCa. Gene expression profile (GSE55945) in men globally and the second leading cause of cancer was downloaded from Gene Expression Omnibus, and associated mortality in developed countries (1,2). Like other the differentially expressed genes (DEGs) were identified. cancers, PCa is considered to be a disease which caused by Subsequently, Gene ontology analysis, KEGG pathway age, diet and gene aberrations (3). Accumulating evidences analysis and protein-protein interaction (PPI) analysis of have demonstrated that a series of genes and pathways involved DEGs were performed. Finally, the identified key genes were in the occurrence, progression and metastasis of PCa (4). At confirmed by immunohistochemistry. The GO analysis results present, the underlying mechanism of PCa occurrence is still showed that the DEGs were mainly participated in cell cycle, unclear, which limits the diagnosis and therapy.
    [Show full text]
  • Protein Purification Protein Localization in Vivo Fluorescent Imaging Protein Arrays Real Time Imaging Protein Interactions Protein Trafficking Protein Turnover
    Overcoming Challenges of Protein Analysis in Mammalian Systems Danette L. Daniels, Ph.D. Current Technologies for Protein Analysis Biochemical/ In Vivo Proteomic Cell Based Animal Analysis Analysis Models Fluorescent proteins Affinity tags Antibodies How about a system applicable to the all approaches that also addresses limitations of current methods? • Minimal interference with protein of interest • Efficient capture/isolation • Detection/real-time imaging • Differential labeling • High Signal/background HaloTag Platform Biochemical/ In Vivo Proteomic Cell Based Animal Analysis Analysis Models Protein purification Protein localization In vivo fluorescent imaging Protein arrays Real time imaging Protein interactions Protein trafficking Protein turnover HaloTag® HaloCHIP™ HaloLink™ HaloTag® Fluorescent Purification Protein:DNA Protein Arrays Pull-Down Ligands HaloTag is a Genetically Engineered Protein Fusion Tag O Functional Protein of Cl O Interest HT + group Protein of Functional HT O O Interest group . A monomeric , 34 kDa, modified bacterial dehalogenase genetically engineered to covalently bind specific, synthetic HaloTag® ligands . Irreversible, covalent attachment of chemical functionalities . Suitable as either N- or C- terminal fusion Mutagenized HaloTag® Protein Enables Covalent HaloTag®-Ligand Complex Hydrolase (DhaA) HaloTag® Catalytic process Facilitated bond formation T r p 1 0 7 T r p 1 0 7 HaloTag®: • 34kDa protein • Monomeric N N H N 4 1 H N A s n - H H • Single change: C l 4 1 A s n C l 2 1 His272Phe for covalent O R O O - C C bond. 3 R O 1 0 6 A s p 1 0 6 A s p O H O H Covalent bond: H H O O • Stable after N C G l u 1 3 0 C G l u 1 3 0 N - O denaturation.
    [Show full text]
  • Novel Driver Strength Index Highlights Important Cancer Genes in TCGA Pancanatlas Patients
    medRxiv preprint doi: https://doi.org/10.1101/2021.08.01.21261447; this version posted August 5, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY-NC-ND 4.0 International license . Novel Driver Strength Index highlights important cancer genes in TCGA PanCanAtlas patients Aleksey V. Belikov*, Danila V. Otnyukov, Alexey D. Vyatkin and Sergey V. Leonov Laboratory of Innovative Medicine, School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Moscow Region, Russia *Corresponding author: [email protected] NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice. 1 medRxiv preprint doi: https://doi.org/10.1101/2021.08.01.21261447; this version posted August 5, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY-NC-ND 4.0 International license . Abstract Elucidating crucial driver genes is paramount for understanding the cancer origins and mechanisms of progression, as well as selecting targets for molecular therapy. Cancer genes are usually ranked by the frequency of mutation, which, however, does not necessarily reflect their driver strength. Here we hypothesize that driver strength is higher for genes that are preferentially mutated in patients with few driver mutations overall, because these few mutations should be strong enough to initiate cancer.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Characterizing Novel Interactions of Transcriptional Repressor Proteins BCL6 & BCL6B
    Characterizing Novel Interactions of Transcriptional Repressor Proteins BCL6 & BCL6B by Geoffrey Graham Lundell-Smith A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Biochemistry University of Toronto © Copyright by Geoffrey Lundell-Smith, 2017 Characterizing Novel Interactions of Transcriptional Repression Proteins BCL6 and BCL6B Geoffrey Graham Lundell-Smith Masters of Science Department of Biochemistry University of Toronto 2016 Abstract B-cell Lymphoma 6 (BCL6) and its close homolog BCL6B encode proteins that are members of the BTB-Zinc Finger family of transcription factors. BCL6 plays an important role in regulating the differentiation and proliferation of B-cells during the adaptive immune response, and is also involved in T cell development and inflammation. BCL6 acts by repressing genes involved in DNA damage response during the affinity maturation of immunoglobulins, and the mis- expression of BCL6 can lead to diffuse large B-cell lymphoma. Although BCL6B shares high sequence similarity with BCL6, the functions of BCL6B are not well-characterized. I used BioID, an in vivo proximity-dependent labeling method, to identify novel BCL6 and BCL6B protein interactors and validated a number of these interactions with co-purification experiments. I also examined the evolutionary relationship between BCL6 and BCL6B and identified conserved residues in an important interaction interface that mediates corepressor binding and gene repression. ii Acknowledgments Thank you to my supervisor, Gil Privé for his mentorship, guidance, and advice, and for giving me the opportunity to work in his lab. Thanks to my committee members, Dr. John Rubinstein and Dr. Jeff Lee for their ideas, thoughts, and feedback during my Masters.
    [Show full text]
  • Attachment PDF Icon
    Spectrum Name of Protein Count of Peptides Ratio (POL2RA/IgG control) POLR2A_228kdBand POLR2A DNA-directed RNA polymerase II subunit RPB1 197 NOT IN CONTROL IP POLR2A_228kdBand POLR2B DNA-directed RNA polymerase II subunit RPB2 146 NOT IN CONTROL IP POLR2A_228kdBand RPAP2 Isoform 1 of RNA polymerase II-associated protein 2 24 NOT IN CONTROL IP POLR2A_228kdBand POLR2G DNA-directed RNA polymerase II subunit RPB7 23 NOT IN CONTROL IP POLR2A_228kdBand POLR2H DNA-directed RNA polymerases I, II, and III subunit RPABC3 19 NOT IN CONTROL IP POLR2A_228kdBand POLR2C DNA-directed RNA polymerase II subunit RPB3 17 NOT IN CONTROL IP POLR2A_228kdBand POLR2J RPB11a protein 7 NOT IN CONTROL IP POLR2A_228kdBand POLR2E DNA-directed RNA polymerases I, II, and III subunit RPABC1 8 NOT IN CONTROL IP POLR2A_228kdBand POLR2I DNA-directed RNA polymerase II subunit RPB9 9 NOT IN CONTROL IP POLR2A_228kdBand ALMS1 ALMS1 3 NOT IN CONTROL IP POLR2A_228kdBand POLR2D DNA-directed RNA polymerase II subunit RPB4 6 NOT IN CONTROL IP POLR2A_228kdBand GRINL1A;Gcom1 Isoform 12 of Protein GRINL1A 6 NOT IN CONTROL IP POLR2A_228kdBand RECQL5 Isoform Beta of ATP-dependent DNA helicase Q5 3 NOT IN CONTROL IP POLR2A_228kdBand POLR2L DNA-directed RNA polymerases I, II, and III subunit RPABC5 5 NOT IN CONTROL IP POLR2A_228kdBand KRT6A Keratin, type II cytoskeletal 6A 3 NOT IN CONTROL IP POLR2A_228kdBand POLR2K DNA-directed RNA polymerases I, II, and III subunit RPABC4 2 NOT IN CONTROL IP POLR2A_228kdBand RFC4 Replication factor C subunit 4 1 NOT IN CONTROL IP POLR2A_228kdBand RFC2
    [Show full text]
  • Extrachromosomal Circular DNA, Microdna, Without Canonical Promoters Produce Short Regulatory Rnas That Suppress Gene Expression
    bioRxiv preprint doi: https://doi.org/10.1101/535831; this version posted January 31, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Extrachromosomal circular DNA, microDNA, without canonical promoters produce short regulatory RNAs that suppress gene expression Teressa Paulsen, Yoshiyuki Shibata, Pankaj Kumar, Laura Dillon, Anindya Dutta* Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA To whom correspondence should be addressed. Tel: 434-924-2466 Email: [email protected] Present Address: Dr. Anindya Dutta, Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA 1 bioRxiv preprint doi: https://doi.org/10.1101/535831; this version posted January 31, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. ABSTRACT Interest in extrachromosomal circular DNA (eccDNA) molecules has increased recently because of their widespread presence in normal cells across every species ranging from yeast to humans, their increased levels in cancer cells, and their overlap with oncogenic and drug- resistant genes. However, the majority of eccDNA (microDNA) are too small to carry protein coding genes. We have tested functional capabilities of microDNA, by creating artificial microDNA molecules mimicking known microDNA sequences and have discovered that they express functional small regulatory RNA including microRNA and novel si-like RNA. MicroDNA is transcribed in vitro and in vivo independent of a canonical promoter sequence.
    [Show full text]
  • The Role of GRHL2 and Epigenetic Remodeling in Epithelial–Mesenchymal Plasticity in Ovarian Cancer Cells
    ARTICLE https://doi.org/10.1038/s42003-019-0506-3 OPEN The role of GRHL2 and epigenetic remodeling in epithelial–mesenchymal plasticity in ovarian cancer cells Vin Yee Chung 1, Tuan Zea Tan 1, Jieru Ye1, Rui-Lan Huang2, Hung-Cheng Lai2, Dennis Kappei 1,3, 1234567890():,; Heike Wollmann4, Ernesto Guccione4 & Ruby Yun-Ju Huang1,5 Cancer cells exhibit phenotypic plasticity during epithelial–mesenchymal transition (EMT) and mesenchymal–epithelial transition (MET) involving intermediate states. To study genome-wide epigenetic remodeling associated with EMT plasticity, we integrate the ana- lyses of DNA methylation, ChIP-sequencing of five histone marks (H3K4me1, H3K4me3, H3K27Ac, H3K27me3 and H3K9me3) and transcriptome profiling performed on ovarian cancer cells with different epithelial/mesenchymal states and on a knockdown model of EMT suppressor Grainyhead-like 2 (GRHL2). We have identified differentially methylated CpG sites associated with EMT, found at promoters of epithelial genes and GRHL2 binding sites. GRHL2 knockdown results in CpG methylation gain and nucleosomal remodeling (reduction in permissive marks H3K4me3 and H3K27ac; elevated repressive mark H3K27me3), resembling the changes observed across progressive EMT states. Epigenetic-modifying agents such as 5-azacitidine, GSK126 and mocetinostat further reveal cell state-dependent plasticity upon GRHL2 overexpression. Overall, we demonstrate that epithelial genes are subject to epigenetic control during intermediate phases of EMT/MET involving GRHL2. 1 Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore. 2 Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, 11031 Taipei, Taiwan. 3 Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore.
    [Show full text]
  • Gene Expression Differences Associated with Human Papillomavirus Status in Head and Neck Squamous Cell Carcinoma Robbertj.C
    Human Cancer Biology Gene Expression Differences Associated with Human Papillomavirus Status in Head and Neck Squamous Cell Carcinoma RobbertJ.C. Slebos,1, 2 Yajun Yi, 7 Kim Ely,3 Jesse Carter,8 Amy Evjen,1Xueqiong Zhang,4 Yu Shyr,4 Barbara M. Murphy,8 Anthony J. Cmelak,5 Brian B. Burkey,2 James L. Netterville,2 Shawn Levy,6 Wendell G. Yarbrough,1, 2 and Christine H. Chung8 Abstract Human papillomavirus (HPV) is associated with a subset of head and neck squamous cell carcinoma (HNSCC). Between 15% and 35% of HNSCCs harbor HPV DNA. Demographic and exposure differences between HPV-positive (HPV+) and negative (HPVÀ) HNSCCs suggest that HPV + tumors may constitute a subclass with different biology, whereas clinical differences have also been observed. Gene expression profiles of HPV+ and HPVÀ tumors were compared with further exploration of the biological effect of HPV in HNSCC. Thirty-six HNSCC tumors were analyzed using Affymetrix Human 133U Plus 2.0 GeneChip and for HPV by PCR and real-time PCR. Eight of 36 (22%) tumors were positive for HPV subtype 16. Statistical analysis using Significance Analysis of Microarrays based on HPV status as a supervising variable resulted in a list of 91genes that were differentially expressed with statistical significance. Results for a subset of these genes were verified by real-time PCR. Genes highly expressed in HPV+ samples included cell cycle regulators (p16INK4A, p18, and CDC7) and transcription factors (TAF7L, RFC4, RPA2, andTFDP2).The microarray data were also investigated by mapping genes by chromosomal loca- tion (DIGMAP).A large number of genes on chromosome 3q24-qter had high levels of expression in HPV+ tumors.
    [Show full text]