The 8P11-P12 Amplicon Oncogenes ASH2L and NSD3 Regulate Cell

Total Page:16

File Type:pdf, Size:1020Kb

The 8P11-P12 Amplicon Oncogenes ASH2L and NSD3 Regulate Cell Medical University of South Carolina MEDICA MUSC Theses and Dissertations 2017 The 8p11-P12 Amplicon Oncogenes ASH2L and NSD3 Regulate Cell Cycle Progression via Epigenetic Alterations and Result in Overexpression and Estrogen-Independent Activation of ERα in Breast Cancer Jamie Nicole Mills Medical University of South Carolina Follow this and additional works at: https://medica-musc.researchcommons.org/theses Recommended Citation Mills, Jamie Nicole, "The 8p11-P12 Amplicon Oncogenes ASH2L and NSD3 Regulate Cell Cycle Progression via Epigenetic Alterations and Result in Overexpression and Estrogen-Independent Activation of ERα in Breast Cancer" (2017). MUSC Theses and Dissertations. 335. https://medica-musc.researchcommons.org/theses/335 This Dissertation is brought to you for free and open access by MEDICA. It has been accepted for inclusion in MUSC Theses and Dissertations by an authorized administrator of MEDICA. For more information, please contact [email protected]. TABLE OF CONTENTS Acknowledgements…………………………………………………………….…...... ii List of Figures…………………………………………………….………………....... iii List of Tables.…………………………………………………………………….…… iv List of Appendices……………………………………………………………...…….. v List of Abbreviations………………………………………………………………….. vi ABSTRACT.…………………………………………………………….….…………. viii CHAPTER 1: Introduction and Review of Literature…………….…………..……. 2 CHAPTER 2: NSD3 amplification and overexpression results in overexpression and estrogen-independent activation of the estrogen receptor in human breast cancer……………………………………………………………… 55 CHAPTER 3: ASH2L regulates gene expression via H3K4me3 in promoters and knockdown of ASH2L reduces expression of NSD3, ERα, and other genes important in cell proliferation processes………………….......................... 70 CHAPTER 4: Discussion and Future Directions….………….…….…………..…. 94 CHAPTER 5: Materials and Methods…………………………………………….... 124 REFERENCES CITED……………………………………………………………….. 136 Appendix Materials………………………………………………………………….... 162 Page | i ACKNOWLEDGEMENTS First and foremost, I would like to thank Steve Ethier for his mentorship. I appreciate all he taught me, the time and energy he invested in me, and the guidance he gave while also allowing me the freedom to explore scientific thought. Steve always challenged me and I will always be grateful to have had him as my mentor. I would also like to thank the members of the Ethier lab for support and advice through this process. It has been a pleasure working with Brittany Turner-Ivey, Christy Kappler, and Ericka Smith. Brittany has been a wonderful mentor and friend and I am thankful for all she taught me. Thank you to Jon Irish, Steve Guest, Ginny Davis, members of adjacent labs, and the Department of Pathology and Laboratory Medicine as a whole for your friendship and for acting as invaluable resources in my training process. I would like to thank my committee members, Melissa Cunningham, John Wrangle, Victoria Findlay, and Amanda LaRue for taking time from their busy schedules to invest in my scientific development. Perry Halushka, Amy Connolly, and my colleagues from the MSTP at MUSC have been an amazing source of support, as have the MUSC core facilities and personnel. Thank you to my clinical mentors and the faculty at MUSC as a whole for participating in my training. Finally, I would like to thank my friends and family. My husband, Ian Mills, always supported me and I could not have accomplished this without him. Thank you to my extended family, especially my mom, Diane Lingenfelter, and grandma, Vel Helton. I am grateful for support from my friends that have become family, especially Gerry Barnard, Renee Vick, Dawn Simmons, and the members of Redeemer Presbyterian Church. Last but not least, I have to thank my daughters, Gwendolyn and Eleanor. More than anyone, they have challenged me and shaped me in ways I never thought possible and I hope they continue to do so at every stage of life and career. Page | ii LIST OF FIGURES Figure 1.1. Schematic representation of ERα structure. Figure 1.2. Breast cancer intrinsic subtype proportions and characteristics of luminal tumors. Figure 1.3. Schematic representation of the 8p11-p12 amplicon. Figure 1.4. Frequencies of ASH2L and NSD3 alterations in primary tumors (TCGA). Figure 1.5. Illustration of major histone modifications and effects on chromatin. Figure 1.6. Hierarchical representation of histone lysine methyltransferase enzyme relationships. Figure 1.7. Representation of the three isoforms of NSD3. Figure 2.1. NSD3 is overexpressed in 8p11-p12 amplicon-bearing cell lines and is a verified oncogene. Figure 2.2. NSD3 knockdown results in knockdown of estrogen receptor alpha (ERα). Figure 2.3. NSD3 overexpression results in overexpression of estrogen receptor alpha (ERα). Figure 2.4. SUM-44 cells are estrogen-independent despite high-level ERα expression. Figure 2.5. SUM-44 cells are ERα-dependent. Figure 3.1. ASH2L is overexpressed in ER+, 8p11-p12 amplicon-bearing breast cancer. Figure 3.2. Overall H3K4 tri-methylation patterns in SUM-44 cells are similar between control and ASH2L knockdown on a global scale. Figure 3.3. ASH2L is responsible for H3K4me3 in the promoters of genes involved in breast cancer tumorigenesis. Figure 3.4. ASH2L knockdown reduces expression of genes with decreased promoter H3K4me3 levels. Figure 3.5. ASH2L, NSD3, and ERα influence the expression of similar suites of genes related to cell cycle progression. Figure 3.6. Knockdown of ASH2L results in decreased proliferation and clonogenicity in luminal B breast cancer. Figure 3.7. Knockdown of ASH2L alters response to palbociclib. Figure 4.1. Summary model of NSD3 functions. Figure 4.2. Summary model of ASH2L functions. Figure 4.3. Summary model. Page | iii LIST OF TABLES Table 2.1. Genes with reduced transcript expression following knockdown of NSD3-S in SUM-44 cells. Table 2.2. Biological processes to which genes with altered expression following knockdown of NSD3-S and ESR1 were annotated. Table 3.1. Genes with promoter H3K4me3 ChIP-seq peaks called in control but not ASH2L knockdown datasets. Table 3.2. Genes with reduced transcript expression following knockdown of ASH2L in SUM-44 cells. Table 3.3. Summary of genes identified by ASH2L knockdown H3K4me3 ChIP-seq, ASH2L knockdown RNA-seq, NSD3-S knockdown array, or ESR1 knockdown array. Table 3.4. Genes annotated to palbociclib response in ToppFun. Page | iv LIST OF APPENDICES Appendix A. NSD3-S and ESR1 knockdown microarray results. Appendix B. H3K4me3 ChIP-seq quality control and enrichment reports. Appendix C. H3K4me3 ChIP-seq peak datasets: LacZ control and ASH2L knockdown. Appendix D. Gene lists: overall H3K4me3 ChIP-seq peaks. Appendix E. Gene lists and ToppFun analysis: promoter only H3K4me3 ChIP-seq peaks. Appendix F. ASH2L knockdown RNA-seq results. Appendix G. Gene lists and ToppFun analysis: ASH2L knockdown RNA-seq downregulated genes. Appendix H. Gene lists and ToppFun analysis: ASH2L knockdown H3K4me3 ChIP-seq and RNA-seq. Appendix I. Gene lists and ToppFun analysis: ASH2L, NSD3-S, and ESR1 knockdown expression analysis and ASH2L knockdown H3K4me3 ChIP-seq overlap. Page | v LIST OF ABBREVIATIONS 4OH-tamoxifen – hydroxytamoxifen HDAC(i) – histone deacetylase (inhibitor) AF – activating function (ERα domain) HER2 – human epidermal growth factor receptor 2 AI – aromatase inhibitor HOX – homeobox gene family AML – acute myeloid leukemia HMT – histone methyltransferase ASH2L – Absent, Small, or Homeotic disc 2-Like HMTi – histone methyltransferase inhibitors/inhibition BET – bromodomain and extraterminal domain hPTM – histone post-translational modification CCND1 – cyclin D1 gene IGV – integrated genome viewer CDK – cyclin-dependent kinase IHC - immunohistochemistry CHD – chromodomain IP - immunoprecipitation ChIP – chromatin immunoprecipitation IPTG – isopropyl β-D-1- – copy number alteration CNA thiogalactopytanoside CTD – comparative toxicogenomics K – lysine database LBD – ligand binding domain DBD – DNA binding domain LSCC – lung squamous cell carcinoma DNMT – DNA methyltransferase MLL – mixed lineage leukemia EGFR – epidermal growth factor receptor NLS – nuclear localization signal ER/ERα – estrogen receptor (alpha) NS – non-significant ERE – estrogen response element NSD – Nuclear-receptor binding SET ESR1 – estrogen receptor alpha gene Domain-containing FDR – false discovery rate NSD3-L – long isoform of NSD3 GISTIC – genomic identification of NSD3-S – short isoform of NSD3 significant targets in cancer NSD3-T – total NSD3 (both isoforms) GO – gene ontology PBS – phosphate buffered saline GRH – gonadotrophin releasing hormone PCA – principal component analysis H – histone PcG – polycomb group HAT – histone acetyltransferase PHD – plant homeodomain HBSS – Hank’s buffered saline solution Page | vi PR – progesterone receptor PRC – polycomb repressive complex PWWP – proline-tryptophan-tryptophan- proline R – arginine RT – room temperature RT-PCR – reverse transcription polymerase chain reaction SAC – SET-associated cysteine-rich SERD – selective estrogen receptor degrader SERM – selective estrogen receptor modulator SET – Suppressor of variegation 3-9, Enhancer of zeste and Trithorax -seq – high throughput sequencing shRNA – short hairpin RNA TAD – transcription activating domain TBST – tris-buffered saline + Tween 20 TCGA – the cancer genome atlas TNBC – triple negative breast cancer trxG – trithorax group WAR – WDR5-ASH2L-RbBP5 sub- complex WRAD – WDR5-RbBP5-ASH2L-DPY-30 sub-complex WHSC1L1 – Wolf-Hirschhorn Syndrome Candidate 1-Like 1 Page | vii ABSTRACT
Recommended publications
  • Severe Infantile Neuropathy with Diaphragmatic Weakness and Its Relationship to SMARD1
    DOI: 10.1093/brain/awg278 Advanced Access publication September 23, 2003 Brain (2003), 126, 2682±2692 Severe infantile neuropathy with diaphragmatic weakness and its relationship to SMARD1 Matthew Pitt,1 Henry Houlden,2 Jean Jacobs,2 Quen Mok,1 Brian Harding,1 Mary Reilly2 and Robert Surtees1 1Great Ormond Street Hospital for Children and Correspondence to: Dr Matthew Pitt, Department of 2The National Hospital for Neurology and Neurosurgery, Clinical Neurophysiology, Great Ormond Street Hospital London, UK for Children NHS Trust, Great Ormond Street, London WC1N 3JH, UK E-mail [email protected] Downloaded from Summary A group of 13 patients with early onset diaphragmatic cord in one patient failed to ®nd any evidence of an palsy in association with a progressive neuropathy is SMA. Four out of the ®ve not tested genetically were presented. All eight of those tested were found to have positive for all diagnostic criteria. None of the cases of mutations in the same gene encoding the immunoglobu- early onset neuropathies or spinal muscular atrophies http://brain.oxfordjournals.org/ lin mu-binding protein 2 (IGHMBP2) in patients with with early respiratory failure reviewed in the literature spinal muscular atrophy (SMA) with respiratory dis- shares the exact characteristics, but many do have very tress type 1. Six out of these eight patients had either close similarities. Their classi®cation varies, but the dis- homozygous or compound heterozygous mutations, and covery of mutations in IGHMBP2 in cases that are vari- two had only a single heterozygous mutation. Detailed ously classi®ed as SMA plus or severe infantile analysis of the clinical picture and the neurophysiologi- neuropathy with respiratory distress points to a need cal and histopathological ®ndings indicated that these for the search for this genetic defect to be widened to patients shared similar characteristics, which were fur- include both groups.
    [Show full text]
  • An Interactive Web Application to Explore Regeneration-Associated Gene Expression and Chromatin Accessibility
    Supplementary Materials Regeneration Rosetta: An interactive web application to explore regeneration-associated gene expression and chromatin accessibility Andrea Rau, Sumona P. Dhara, Ava J. Udvadia, Paul L. Auer 1. Table S1. List of cholesterol metabolic genes from MGI database 2. Table S2. List of differentially expressed transcripts during optic nerve regeneration in zebrafish using the MGI cholesterol metabolic gene queries in the Regeneration Rosetta app 3. Table S3. List of transcription factor encoding genes from brain cell bodies following spinal cord injury in lamprey over a course of 12 weeKs 4. Table S4. List of transcription factor encoding genes from spinal cell bodies following spinal cord injury in lamprey over a course of 12 weeks Ensembl ID MGI Gene ID Symbol Name ENSMUSG00000015243 MGI:99607 Abca1 ATP-binding cassette, sub-family A (ABC1), member 1 ENSMUSG00000026944 MGI:99606 Abca2 ATP-binding cassette, sub-family A (ABC1), member 2 ENSMUSG00000024030 MGI:107704 Abcg1 ATP binding cassette subfamily G member 1 ENSMUSG00000026003 MGI:87866 Acadl acyl-Coenzyme A dehydrogenase, long-chain ENSMUSG00000018574 MGI:895149 Acadvl acyl-Coenzyme A dehydrogenase, very long chain ENSMUSG00000038641 MGI:2384785 Akr1d1 aldo-keto reductase family 1, member D1 ENSMUSG00000028553 MGI:1353627 Angptl3 angiopoietin-like 3 ENSMUSG00000031996 MGI:88047 Aplp2 amyloid beta (A4) precursor-like protein 2 ENSMUSG00000032083 MGI:88049 Apoa1 apolipoprotein A-I ENSMUSG00000005681 MGI:88050 Apoa2 apolipoprotein A-II ENSMUSG00000032080 MGI:88051 Apoa4
    [Show full text]
  • Proteomic Identification of the Transcription Factors Ikaros And
    European School of Molecular Medicine (SEMM) University of Milan and University of Naples “Federico II” PhD degree in Systems Medicine (curriculum in Molecular Oncology) Settore disciplinare: BIO/11 Proteomic identification of the transcription factors Ikaros and Aiolos as new Myc interactors on chromatin Chiara Veronica Locarno Matricola: R10755 Center for Genomic Science IIT@SEMM, Milan Supervisor: Bruno Amati, PhD IEO, Milan Added Supervisor: Arianna Sabò, PhD IEO, Milan Academic year 2017-2018 Table of contents List of abbreviations ........................................................................................................... 4 List of figures ....................................................................................................................... 8 List of tables ....................................................................................................................... 11 Abstract .............................................................................................................................. 12 1. INTRODUCTION ......................................................................................................... 13 1.1 Myc ........................................................................................................................................ 13 1.1.1 Myc discovery and structure ........................................................................................... 13 1.1.2. Role of Myc in physiological and pathological conditions ...........................................
    [Show full text]
  • Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin
    cells Review Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin Agnieszka Bochy ´nska,Juliane Lüscher-Firzlaff and Bernhard Lüscher * ID Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany; [email protected] (A.B.); jluescher-fi[email protected] (J.L.-F.) * Correspondence: [email protected]; Tel.: +49-241-8088850; Fax: +49-241-8082427 Received: 18 January 2018; Accepted: 27 February 2018; Published: 2 March 2018 Abstract: Regulation of gene expression is achieved by sequence-specific transcriptional regulators, which convey the information that is contained in the sequence of DNA into RNA polymerase activity. This is achieved by the recruitment of transcriptional co-factors. One of the consequences of co-factor recruitment is the control of specific properties of nucleosomes, the basic units of chromatin, and their protein components, the core histones. The main principles are to regulate the position and the characteristics of nucleosomes. The latter includes modulating the composition of core histones and their variants that are integrated into nucleosomes, and the post-translational modification of these histones referred to as histone marks. One of these marks is the methylation of lysine 4 of the core histone H3 (H3K4). While mono-methylation of H3K4 (H3K4me1) is located preferentially at active enhancers, tri-methylation (H3K4me3) is a mark found at open and potentially active promoters. Thus, H3K4 methylation is typically associated with gene transcription. The class 2 lysine methyltransferases (KMTs) are the main enzymes that methylate H3K4. KMT2 enzymes function in complexes that contain a necessary core complex composed of WDR5, RBBP5, ASH2L, and DPY30, the so-called WRAD complex.
    [Show full text]
  • TASOR Is a Pseudo-PARP That Directs HUSH Complex Assembly and Epigenetic Transposon Control
    bioRxiv preprint doi: https://doi.org/10.1101/2020.03.09.974832; this version posted March 11, 2020. 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. TASOR is a pseudo-PARP that directs HUSH complex assembly and epigenetic transposon control Christopher H. Douse1,‡, Iva A. Tchasovnikarova2,‡, Richard T. Timms2,‡, Anna V. Protasio2, Marta Seczynska2, Daniil M. Prigozhin1, Anna Albecka1,2, Jane Wagstaff3, James C. Williamson2, Stefan M.V. Freund3, Paul J. Lehner2*, Yorgo Modis1,2* 1 Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK 2 Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK 3 Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK Current addresses: Department of Experimental Medical Science, Lund University, Sölvegatan 19, Lund, Sweden (C.H.D.); The Gurdon Institute, Tennis Court Road, Cambridge, UK (I.A.T.); Department of Pathology, Tennis Court Road, Cambridge, UK (A.V.P.); Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA (D.M.P.) ‡These authors contributed equally *co-corresponding authors Keywords: Transcriptional repression; epigenetic silencing; antiretroviral response; genome stability; histone H3 lysine 9 methylation (H3K9me3); transposable element (TE); long interspersed nuclear element-1 (LINE-1); poly-ADP ribose polymerase (PARP); RNA-binding protein; RNA-induced transcriptional silencing; CUT&RUN; CUT&Tag; genome profiling 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.03.09.974832; this version posted March 11, 2020.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Download Download
    Supplementary Figure S1. Results of flow cytometry analysis, performed to estimate CD34 positivity, after immunomagnetic separation in two different experiments. As monoclonal antibody for labeling the sample, the fluorescein isothiocyanate (FITC)- conjugated mouse anti-human CD34 MoAb (Mylteni) was used. Briefly, cell samples were incubated in the presence of the indicated MoAbs, at the proper dilution, in PBS containing 5% FCS and 1% Fc receptor (FcR) blocking reagent (Miltenyi) for 30 min at 4 C. Cells were then washed twice, resuspended with PBS and analyzed by a Coulter Epics XL (Coulter Electronics Inc., Hialeah, FL, USA) flow cytometer. only use Non-commercial 1 Supplementary Table S1. Complete list of the datasets used in this study and their sources. GEO Total samples Geo selected GEO accession of used Platform Reference series in series samples samples GSM142565 GSM142566 GSM142567 GSM142568 GSE6146 HG-U133A 14 8 - GSM142569 GSM142571 GSM142572 GSM142574 GSM51391 GSM51392 GSE2666 HG-U133A 36 4 1 GSM51393 GSM51394 only GSM321583 GSE12803 HG-U133A 20 3 GSM321584 2 GSM321585 use Promyelocytes_1 Promyelocytes_2 Promyelocytes_3 Promyelocytes_4 HG-U133A 8 8 3 GSE64282 Promyelocytes_5 Promyelocytes_6 Promyelocytes_7 Promyelocytes_8 Non-commercial 2 Supplementary Table S2. Chromosomal regions up-regulated in CD34+ samples as identified by the LAP procedure with the two-class statistics coded in the PREDA R package and an FDR threshold of 0.5. Functional enrichment analysis has been performed using DAVID (http://david.abcc.ncifcrf.gov/)
    [Show full text]
  • Investigation of Candidate Genes and Mechanisms Underlying Obesity
    Prashanth et al. BMC Endocrine Disorders (2021) 21:80 https://doi.org/10.1186/s12902-021-00718-5 RESEARCH ARTICLE Open Access Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules G. Prashanth1 , Basavaraj Vastrad2 , Anandkumar Tengli3 , Chanabasayya Vastrad4* and Iranna Kotturshetti5 Abstract Background: Obesity associated type 2 diabetes mellitus is a metabolic disorder ; however, the etiology of obesity associated type 2 diabetes mellitus remains largely unknown. There is an urgent need to further broaden the understanding of the molecular mechanism associated in obesity associated type 2 diabetes mellitus. Methods: To screen the differentially expressed genes (DEGs) that might play essential roles in obesity associated type 2 diabetes mellitus, the publicly available expression profiling by high throughput sequencing data (GSE143319) was downloaded and screened for DEGs. Then, Gene Ontology (GO) and REACTOME pathway enrichment analysis were performed. The protein - protein interaction network, miRNA - target genes regulatory network and TF-target gene regulatory network were constructed and analyzed for identification of hub and target genes. The hub genes were validated by receiver operating characteristic (ROC) curve analysis and RT- PCR analysis. Finally, a molecular docking study was performed on over expressed proteins to predict the target small drug molecules. Results: A total of 820 DEGs were identified between
    [Show full text]
  • Aberrant Activity of Histone–Lysine N-Methyltransferase 2 (KMT2) Complexes in Oncogenesis
    International Journal of Molecular Sciences Review Aberrant Activity of Histone–Lysine N-Methyltransferase 2 (KMT2) Complexes in Oncogenesis Elzbieta Poreba 1,* , Krzysztof Lesniewicz 2 and Julia Durzynska 1,* 1 Institute of Experimental Biology, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Pozna´nskiego6, 61-614 Pozna´n,Poland 2 Department of Molecular and Cellular Biology, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Pozna´nskiego6, 61-614 Pozna´n,Poland; [email protected] * Correspondence: [email protected] (E.P.); [email protected] (J.D.); Tel.: +48-61-829-5857 (E.P.) Received: 19 November 2020; Accepted: 6 December 2020; Published: 8 December 2020 Abstract: KMT2 (histone-lysine N-methyltransferase subclass 2) complexes methylate lysine 4 on the histone H3 tail at gene promoters and gene enhancers and, thus, control the process of gene transcription. These complexes not only play an essential role in normal development but have also been described as involved in the aberrant growth of tissues. KMT2 mutations resulting from the rearrangements of the KMT2A (MLL1) gene at 11q23 are associated with pediatric mixed-lineage leukemias, and recent studies demonstrate that KMT2 genes are frequently mutated in many types of human cancers. Moreover, other components of the KMT2 complexes have been reported to contribute to oncogenesis. This review summarizes the recent advances in our knowledge of the role of KMT2 complexes in cell transformation. In addition, it discusses the therapeutic targeting of different components of the KMT2 complexes. Keywords: histone–lysine N-methyltransferase 2; COMPASS; COMPASS-like; H3K4 methylation; oncogenesis; cancer; epigenetics; chromatin 1.
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. 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. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. 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 The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • PRODUCT SPECIFICATION Anti-C22orf23 Product Datasheet
    Anti-C22orf23 Product Datasheet Polyclonal Antibody PRODUCT SPECIFICATION Product Name Anti-C22orf23 Product Number HPA000650 Gene Description chromosome 22 open reading frame 23 Clonality Polyclonal Isotype IgG Host Rabbit Antigen Sequence Recombinant Protein Epitope Signature Tag (PrEST) antigen sequence: LQCSPTSSQRVLPSKQIASPIYLPPILAARPHLRPANMCQANGAYSREQF KPQATRDLEKEKQRLQNIFATGKDMEERKRKAPPARQKAPAPELDRFEEL VKEIQERKEFLADMEALGQGKQYRGIILAEISQKLREMEDIDHRRSEE Purification Method Affinity purified using the PrEST antigen as affinity ligand Verified Species Human Reactivity Recommended ICC-IF (Immunofluorescence) Applications - Fixation/Permeabilization: PFA/Triton X-100 - Working concentration: 0.25-2 µg/ml Characterization Data Available at atlasantibodies.com/products/HPA000650 Buffer 40% glycerol and PBS (pH 7.2). 0.02% sodium azide is added as preservative. Concentration Lot dependent Storage Store at +4°C for short term storage. Long time storage is recommended at -20°C. Notes Gently mix before use. Optimal concentrations and conditions for each application should be determined by the user. For protocols, additional product information, such as images and references, see atlasantibodies.com. Product of Sweden. For research use only. Not intended for pharmaceutical development, diagnostic, therapeutic or any in vivo use. No products from Atlas Antibodies may be resold, modified for resale or used to manufacture commercial products without prior written approval from Atlas Antibodies AB. Warranty: The products supplied by Atlas Antibodies are warranted to meet stated product specifications and to conform to label descriptions when used and stored properly. Unless otherwise stated, this warranty is limited to one year from date of sales for products used, handled and stored according to Atlas Antibodies AB's instructions. Atlas Antibodies AB's sole liability is limited to replacement of the product or refund of the purchase price.
    [Show full text]