Protein Arginine Methyltransferases: Promising Targets for Cancer Therapy Jee Won Hwang1,Yenacho1,Gyu-Unbae1,Su-Namkim2 and Yong Kee Kim 1

Total Page:16

File Type:pdf, Size:1020Kb

Protein Arginine Methyltransferases: Promising Targets for Cancer Therapy Jee Won Hwang1,Yenacho1,Gyu-Unbae1,Su-Namkim2 and Yong Kee Kim 1 Hwang et al. Experimental & Molecular Medicine (2021) 53:788–808 https://doi.org/10.1038/s12276-021-00613-y Experimental & Molecular Medicine REVIEW ARTICLE Open Access Protein arginine methyltransferases: promising targets for cancer therapy Jee Won Hwang1,YenaCho1,Gyu-UnBae1,Su-NamKim2 and Yong Kee Kim 1 Abstract Protein methylation, a post-translational modification (PTM), is observed in a wide variety of cell types from prokaryotes to eukaryotes. With recent and rapid advancements in epigenetic research, the importance of protein methylation has been highlighted. The methylation of histone proteins that contributes to the epigenetic histone code is not only dynamic but is also finely controlled by histone methyltransferases and demethylases, which are essential for the transcriptional regulation of genes. In addition, many nonhistone proteins are methylated, and these modifications govern a variety of cellular functions, including RNA processing, translation, signal transduction, DNA damage response, and the cell cycle. Recently, the importance of protein arginine methylation, especially in cell cycle regulation and DNA repair processes, has been noted. Since the dysregulation of protein arginine methylation is closely associated with cancer development, protein arginine methyltransferases (PRMTs) have garnered significant interest as novel targets for anticancer drug development. Indeed, several PRMT inhibitors are in phase 1/2 clinical trials. In this review, we discuss the biological functions of PRMTs in cancer and the current development status of PRMT inhibitors in cancer therapy. 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; G G Introduction ω-N ,N -asymmetric dimethyl arginine (ADMA), and G G Since the discovery of arginine residue methylation on ω-N ,N’ -symmetric dimethyl arginine (SDMA)8. PRMTs histone proteins1, protein arginine methylation has been are classified into three subgroups based on the type of emphasized as an indispensable post-translational mod- methyl arginine they produce: Type I PRMTs (PRMT1, 2, ification (PTM) and an epigenetic regulation mechan- 3, 4, 6, and 8) generate MMA and ADMA, Type II PRMTs ism2,3. Arginine methylation is catalyzed by a family of (PRMT5 and 9) produce MMA and SDMA, and Type III enzymes called protein arginine methyltransferases PRMT (PRMT7) produces only MMA7,9. (PRMTs), and nine PRMTs have been identified in The arginine residue consists of a guanidino group on mammals to date (Fig. 1a)2,4,5. All PRMTs share four its side chain, which is protonated and positively charged conserved sequence motifs (I, post-I, II, and III) and one at physiological pH3,5. The guanidino group forms mul- THW loop, which compose the S-adenosyl-L-methionine tiple hydrogen bonds that bind with other interacting (AdoMet) binding pocket in the tertiary structure6,7. proteins or cofactors2,5. Although the methylated arginine PRMTs transfer a methyl group from the AdoMet residue retains its positive charge, the ability to form molecule to the guanidino group of the arginine residue in hydrogen bonds is reduced, probably affecting the substrate proteins8. There are three types of methyl protein-protein interaction. In addition, arginine methy- G arginine (Fig. 1b): ω-N -monomethyl arginine (MMA), lation is very stable compared to that of other PTMs, and hence, its kinetics are less dynamic2,5. PRMTs are asso- ciated with many essential cellular processes, including Correspondence: Yong Kee Kim ([email protected]) transcription, splicing, translation, signal transduction, 1Research Institute of Pharmaceutical Sciences, College of Pharmacy, ’ DNA damage and repair, and cell cycle regulation Sookmyung Women s University, Seoul 04310, Republic of Korea 2–4 2Natural Product Research Institute, Korea Institute of Science and Technology, (Fig. 2) , and the knockout phenotypes of some PRMTs Gangneung 25451, Republic of Korea © The Author(s) 2021 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a linktotheCreativeCommons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/. Official journal of the Korean Society for Biochemistry and Molecular Biology Hwang et al. Experimental & Molecular Medicine (2021) 53:788–808 789 (a) (b) Name Domain structureClassificaon Cellular localizaon arginine 1ab c d e 371 NH + NH PRMT1 type I cytoplasm/nucleus 2 2 50 361 C 1ab c d e 433 PRMT2 SH3 type I cytoplasm/nucleus NH 30 89 99 432 (CH2)3 1ab c d e 531 PRMT3 Zn-F type I cytoplasm Protein 48 71 271 531 1 608 ab c d e type I, II, and III CARM1 TAD type I cytoplasm/nucleus 146 453 499 608 1ab c d e 637 asymmetric di-methyl mono-methyl symmetric di-methyl PRMT5 TIM-barrel type II cytoplasm/nucleus arginine (ADMA) arginine (MMA) arginine (SDMA) 13 292 308 615 1ab c d e 375 PRMT6 type I nucleus CH CH CH 3 3 3 CH3 44 374 + + + NH2 N NH2 NH NH NH 1 692 ab c d e ab c d CH PRMT7 type III cytoplasm/nucleus C 3 C C 14 345 358 684 NH type I NH type II NH SH3 binding mof 1 394 ab c d e (CH ) (CH ) (CH ) PRMT8 myr type I plasma membrane 2 3 2 3 2 3 73 394 TPR Protein Protein Protein 1ab c d e ab c d e 845 PRMT9 type II cytoplasm 137 466 530 845 Fig. 1 Protein arginine methylation and responsible enzymes. a The mammalian PRMT family. Nine PRMTs were identified, and these have unique signatures (dark blue lines) with high sequence similarity (a, Motif I: VLD/EVGXGXG; b, Post-I: V/IXG/AXD/E; c, Motif II: F/I/VDI/L/K; d, Motif III: LR/KXXG; e, THW loop). Their enzymatic types and cellular localization are shown. b Types of arginine methylation. The arginine residue has two equivalent nitrogen atoms in its guanidino group. Types I, II, and III PRMTs generate monomethyl arginine (MMA) marks. The subsequent generation of asymmetric dimethyl arginine (ADMA) is catalyzed by type I enzymes (PRMT1, PRMT2, PRMT3, CARM1, PRMT6, and PRMT8), and symmetric dimethyl arginine (SDMA) is produced by type II enzymes (PRMT5 and PRMT9). PRMT7, a type III enzyme, generates only MMA. – show embryonic or perinatal lethality2,10 12, indicating the arginine residues not only serve as key epigenetic marks significance of PRMTs in maintaining functional home- but also engage in crosstalk with other epigenetic ostasis in biological systems. Tissue-specific deletion marks21,22. These orchestrated epigenetic modifications studies of PRMTs strongly support the supposition that are recognized by epigenetic reader proteins, leading to they are involved in cancer and metabolic, immune, the recruitment of activating/repressing transcriptional neurodegenerative, and muscular disorders4,13,14. Since machinery. The histone modifications generated by the dysregulation of PRMTs has been closely associated PRMTs and their roles are summarized in Table 1. The with cancer development2,15,16, the use of PRMTs as novel methylation status of an arginine residue in histones can targets for anticancer drug development is rapidly determine whether the transcription process is activated increasing. Recent studies have revealed considerable or suppressed. For example, H4R3me2a, a modification advances in the identification of clinically relevant PRMT generated by PRMT1/PRMT3, acts as a mark of activated inhibitors17,18. Here, we focus on the biological functions transcription, whereas H4R3me2s, generated by PRMT5, of PRMTs in cancer and the therapeutic potential of functions as a repression mark, implying that there is a PRMT inhibitors. sophisticated and competitive mechanism between PRMTs for regulating the transcription process. In addi- Biological functions of protein arginine tion to histone proteins, various proteins involved in methylation transcription, such as transcription factors, coactivators, As histone proteins tightly regulate gene transcription and corepressors, are also methylated by PRMTs (Table 1 through various PTMs, including acetylation, lysine and Fig. 2)23. Hence, PRMTs also contribute to the precise methylation, phosphorylation, ubiquitination, and regulation of the transcription process. A number of SUMOylation19,20, early studies of PRMTs have also RNA-binding proteins (RBPs) have RG/RGG-rich motifs focused on their epigenetic functions. PRMTs synthesize that have been established as representative consensus methyl arginine on nucleosomes after being recruited into sequences of PRMTs24,25. Indeed, theoretical insights and chromatin remodeling complexes, and these methylated proteomic analysis revealed that several RBPs are Official journal of the Korean Society for Biochemistry and Molecular Biology Hwang et al. Experimental & Molecular Medicine (2021) 53:788–808 790 Genec Mutaons Diet Aging Hormones PRMTs Environments MMA, ADMA, SDMA Histones Non-histones H3: R2me2a(s), R8me2a(s), R17me2a, R26me2a, C/EBPα, FOXO1, GLI1/2, Sm proteins, hnRNPs, R42me2a EGFR, CRAF, p65/RelA, Smad6/7, INCENP, CDK4, H4: R3me2a(s) H2A: R29me2a p53, RUVBL1, 53BP1, BRCA1,
Recommended publications
  • Behavioural Brain Research 217 (2011) 271–281
    Behavioural Brain Research 217 (2011) 271–281 Contents lists available at ScienceDirect Behavioural Brain Research journal homepage: www.elsevier.com/locate/bbr Research report The telomeric part of the human chromosome 21 from Cstb to Prmt2 is not necessary for the locomotor and short-term memory deficits observed in the Tc1 mouse model of Down syndrome Arnaud Duchon a, Stéphanie Pothion b, Véronique Brault a, Andrew J. Sharp c, Victor L.J. Tybulewicz d, Elizabeth M.C. Fisher e, Yann Herault a,b,f,∗ a Institut de Génétique Biologie Moléculaire et Cellulaire, Translational Medicine and Neuroscience Program, IGBMC, CNRS, INSERM, Université de Strasbourg, UMR7104, UMR964, 1 rue Laurent Fries, 67404 Illkirch, France b Transgenese et Archivage Animaux Modèles, TAAM, CNRS, UPS44, 3B rue de la Férollerie 45071 Orléans, France c Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, 1425 Madison Avenue, Room 14-75B, Box 1498, New York, NY 10029, USA d MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK e UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK f Institut Clinique de la Souris, ICS, 1 rue Laurent Fries, 67404 Illkirch, France article info abstract Article history: Trisomy 21 or Down syndrome (DS) is the most common form of human aneuploid disorder. Increase Received 5 September 2010 in the copy number of human chromosome 21 genes leads to several alterations including mental retar- Received in revised form 6 October 2010 dation, heart and skeletal dysmorphologies with additional physiological defects. To better understand Accepted 17 October 2010 the genotype and phenotype relationships, several mouse models have been developed, including the Available online 31 October 2010 transchromosomic Tc1 mouse, which carries an almost complete human chromosome 21, that displays several locomotor and cognitive alterations related to DS.
    [Show full text]
  • An Animal Model with a Cardiomyocyte-Specific Deletion of Estrogen Receptor Alpha: Functional, Metabolic, and Differential Netwo
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2014 An animal model with a cardiomyocyte-specific deletion of estrogen receptor alpha: Functional, metabolic, and differential network analysis Sriram Devanathan Washington University School of Medicine in St. Louis Timothy Whitehead Washington University School of Medicine in St. Louis George G. Schweitzer Washington University School of Medicine in St. Louis Nicole Fettig Washington University School of Medicine in St. Louis Attila Kovacs Washington University School of Medicine in St. Louis See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Devanathan, Sriram; Whitehead, Timothy; Schweitzer, George G.; Fettig, Nicole; Kovacs, Attila; Korach, Kenneth S.; Finck, Brian N.; and Shoghi, Kooresh I., ,"An animal model with a cardiomyocyte-specific deletion of estrogen receptor alpha: Functional, metabolic, and differential network analysis." PLoS One.9,7. e101900. (2014). https://digitalcommons.wustl.edu/open_access_pubs/3326 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. Authors Sriram Devanathan, Timothy Whitehead, George G. Schweitzer, Nicole Fettig, Attila Kovacs, Kenneth S. Korach, Brian N. Finck, and Kooresh I. Shoghi This open access publication is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/open_access_pubs/3326 An Animal Model with a Cardiomyocyte-Specific Deletion of Estrogen Receptor Alpha: Functional, Metabolic, and Differential Network Analysis Sriram Devanathan1, Timothy Whitehead1, George G. Schweitzer2, Nicole Fettig1, Attila Kovacs3, Kenneth S.
    [Show full text]
  • A Molecular and Genetic Analysis of Otosclerosis
    A molecular and genetic analysis of otosclerosis Joanna Lauren Ziff Submitted for the degree of PhD University College London January 2014 1 Declaration I, Joanna Ziff, confirm that the work presented in this thesis is my own. Where information has been derived from other sources, I confirm that this has been indicated in the thesis. Where work has been conducted by other members of our laboratory, this has been indicated by an appropriate reference. 2 Abstract Otosclerosis is a common form of conductive hearing loss. It is characterised by abnormal bone remodelling within the otic capsule, leading to formation of sclerotic lesions of the temporal bone. Encroachment of these lesions on to the footplate of the stapes in the middle ear leads to stapes fixation and subsequent conductive hearing loss. The hereditary nature of otosclerosis has long been recognised due to its recurrence within families, but its genetic aetiology is yet to be characterised. Although many familial linkage studies and candidate gene association studies to investigate the genetic nature of otosclerosis have been performed in recent years, progress in identifying disease causing genes has been slow. This is largely due to the highly heterogeneous nature of this condition. The research presented in this thesis examines the molecular and genetic basis of otosclerosis using two next generation sequencing technologies; RNA-sequencing and Whole Exome Sequencing. RNA–sequencing has provided human stapes transcriptomes for healthy and diseased stapes, and in combination with pathway analysis has helped identify genes and molecular processes dysregulated in otosclerotic tissue. Whole Exome Sequencing has been employed to investigate rare variants that segregate with otosclerosis in affected families, and has been followed by a variant filtering strategy, which has prioritised genes found to be dysregulated during RNA-sequencing.
    [Show full text]
  • Protein Identities in Evs Isolated from U87-MG GBM Cells As Determined by NG LC-MS/MS
    Protein identities in EVs isolated from U87-MG GBM cells as determined by NG LC-MS/MS. No. Accession Description Σ Coverage Σ# Proteins Σ# Unique Peptides Σ# Peptides Σ# PSMs # AAs MW [kDa] calc. pI 1 A8MS94 Putative golgin subfamily A member 2-like protein 5 OS=Homo sapiens PE=5 SV=2 - [GG2L5_HUMAN] 100 1 1 7 88 110 12,03704523 5,681152344 2 P60660 Myosin light polypeptide 6 OS=Homo sapiens GN=MYL6 PE=1 SV=2 - [MYL6_HUMAN] 100 3 5 17 173 151 16,91913397 4,652832031 3 Q6ZYL4 General transcription factor IIH subunit 5 OS=Homo sapiens GN=GTF2H5 PE=1 SV=1 - [TF2H5_HUMAN] 98,59 1 1 4 13 71 8,048185945 4,652832031 4 P60709 Actin, cytoplasmic 1 OS=Homo sapiens GN=ACTB PE=1 SV=1 - [ACTB_HUMAN] 97,6 5 5 35 917 375 41,70973209 5,478027344 5 P13489 Ribonuclease inhibitor OS=Homo sapiens GN=RNH1 PE=1 SV=2 - [RINI_HUMAN] 96,75 1 12 37 173 461 49,94108966 4,817871094 6 P09382 Galectin-1 OS=Homo sapiens GN=LGALS1 PE=1 SV=2 - [LEG1_HUMAN] 96,3 1 7 14 283 135 14,70620005 5,503417969 7 P60174 Triosephosphate isomerase OS=Homo sapiens GN=TPI1 PE=1 SV=3 - [TPIS_HUMAN] 95,1 3 16 25 375 286 30,77169764 5,922363281 8 P04406 Glyceraldehyde-3-phosphate dehydrogenase OS=Homo sapiens GN=GAPDH PE=1 SV=3 - [G3P_HUMAN] 94,63 2 13 31 509 335 36,03039959 8,455566406 9 Q15185 Prostaglandin E synthase 3 OS=Homo sapiens GN=PTGES3 PE=1 SV=1 - [TEBP_HUMAN] 93,13 1 5 12 74 160 18,68541938 4,538574219 10 P09417 Dihydropteridine reductase OS=Homo sapiens GN=QDPR PE=1 SV=2 - [DHPR_HUMAN] 93,03 1 1 17 69 244 25,77302971 7,371582031 11 P01911 HLA class II histocompatibility antigen,
    [Show full text]
  • BMP Signaling Orchestrates a Transcriptional Network to Control the Fate of Mesenchymal Stem Cells (Mscs)
    bioRxiv preprint doi: https://doi.org/10.1101/104927; this version posted February 1, 2017. 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 4.0 International license. BMP signaling orchestrates a transcriptional network to control the fate of mesenchymal stem cells (MSCs) Jifan Feng1, Junjun Jing1,2, Jingyuan Li1, Hu Zhao1, Vasu Punj3, Tingwei Zhang1,2, Jian Xu1 and Yang Chai1,* 1Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA 2State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China 3Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA *Corresponding author: Yang Chai 2250 Alcazar Street – CSA 103 Center for Craniofacial Molecular Biology University of Southern California Los Angeles, CA Phone number: 323-442-3480 [email protected] Running Title: BMP regulates cell fate of MSCs Key Words: BMP, mesenchymal stem cells (MSCs), odontogenesis, Gli1 Summary Statement: BMP signaling activity is required for the lineage commitment of MSCs and transcription factors downstream of BMP signaling may determine distinct cellular identities within the dental mesenchyme. 1 bioRxiv preprint doi: https://doi.org/10.1101/104927; this version posted February 1, 2017. 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 4.0 International license.
    [Show full text]
  • Mir-96 Regulates the Progression of Differentiation in Mammalian Cochlear Inner and Outer Hair Cells
    miR-96 regulates the progression of differentiation in mammalian cochlear inner and outer hair cells Stephanie Kuhna,1, Stuart L. Johnsona,1, David N. Furnessb, Jing Chenc, Neil Inghamc, Jennifer M. Hiltonc, Georg Steffesc, Morag A. Lewisc, Valeria Zampinia,d, Carole M. Hackneya, Sergio Masettod, Matthew C. Holleya, Karen P. Steelc, and Walter Marcottia,2 aDepartment of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom; bInstitute for Science and Technology in Medicine, Keele University, Keele ST5 5BG, United Kingdom; cWellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom; and dDepartment of Physiology, University of Pavia, 27100 Pavia, Italy Edited by Mary-Claire King, University of Washington, Seattle, WA, and approved December 27, 2010 (received for review November 8, 2010) MicroRNAs (miRNAs) are small noncoding RNAs able to regulate cells up to at least P5 (23) and in the spiral ganglion up to P14 a broad range of protein-coding genes involved in many biological (19). Mutations in miR-96 have been associated with non- processes. miR-96 is a sensory organ-specific miRNA expressed in syndromic progressive hearing loss in humans (15) and mice (23). the mammalian cochlea during development. Mutations in miR-96 We found that hair cell development in diminuendo mice with cause nonsyndromic progressive hearing loss in humans and mice. a Mir96 mutation is arrested at around the day of birth, a time The mouse mutant diminuendo has a single base change in the when IHCs and OHCs still exhibit qualitatively similar bio- seed region of the Mir96 gene leading to widespread changes in physical properties.
    [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]
  • PRMT8 Antibody (C-Term) Purified Rabbit Polyclonal Antibody (Pab) Catalog # Ap1219b
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 PRMT8 Antibody (C-term) Purified Rabbit Polyclonal Antibody (Pab) Catalog # AP1219b Specification PRMT8 Antibody (C-term) - Product Information Application WB,E Primary Accession Q9NR22 Other Accession Q6PAK3 Reactivity Human Predicted Mouse Host Rabbit Clonality Polyclonal Isotype Rabbit Ig Calculated MW 45291 Antigen Region 344-373 PRMT8 Antibody (C-term) - Additional Information Gene ID 56341 Western blot analysis of lysates from human Other Names brain, mouse brain tissue lysate (from left to Protein arginine N-methyltransferase 8, right), using PRMT8 Antibody (C-term)(Cat. 211-, Heterogeneous nuclear #AP1219b). AP1219b was diluted at 1:1000 ribonucleoprotein methyltransferase-like at each lane. A goat anti-rabbit IgG H&L(HRP) protein 4, PRMT8, HRMT1L3, HRMT1L4 at 1:10000 dilution was used as the Target/Specificity secondary antibody. Lysates at 35ug per This PRMT8 antibody is generated from lane. rabbits immunized with a KLH conjugated synthetic peptide between 344-373 amino acids from the C-terminal region of human PRMT8. Dilution WB~~1:1000 Format Purified polyclonal antibody supplied in PBS with 0.09% (W/V) sodium azide. This antibody is prepared by Saturated Ammonium Sulfate (SAS) precipitation followed by dialysis against PBS. Storage Western blot analysis of PRMT8 Antibody Maintain refrigerated at 2-8°C for up to 2 (C-term) (Cat.#AP1219b) in K562 cell line weeks. For long term storage store at -20°C lysates (35ug/lane). PRMT8 (arrow) was in small aliquots to prevent freeze-thaw detected using the purified Pab. cycles.
    [Show full text]
  • The Endogenous Transposable Element Tgm9 Is Suitable for Generating Knockout Mutants for Functional Analyses of Soybean Genes and Genetic Improvement in Soybean
    RESEARCH ARTICLE The endogenous transposable element Tgm9 is suitable for generating knockout mutants for functional analyses of soybean genes and genetic improvement in soybean Devinder Sandhu1*, Jayadri Ghosh2, Callie Johnson3, Jordan Baumbach2, Eric Baumert3, Tyler Cina3, David Grant2,4, Reid G. Palmer2,4², Madan K. Bhattacharyya2* a1111111111 1 USDA-ARS, US Salinity Laboratory, Riverside, CA, United States of America, 2 Department of Agronomy, a1111111111 Iowa State University, Ames, IA, United States of America, 3 Department of Biology, University of Wisconsin- a1111111111 Stevens Point, Stevens Point, WI, United States of America, 4 USDA-ARS Corn Insects and Crop Genomics a1111111111 Research Unit, Ames, IA, United States of America a1111111111 ² Deceased. * [email protected] (DS); [email protected] (MKB) OPEN ACCESS Abstract Citation: Sandhu D, Ghosh J, Johnson C, In soybean, variegated flowers can be caused by somatic excision of the CACTA-type trans- Baumbach J, Baumert E, Cina T, et al. (2017) The posable element Tgm9 from Intron 2 of the DFR2 gene encoding dihydroflavonol-4-reduc- endogenous transposable element Tgm9 is suitable for generating knockout mutants for tase of the anthocyanin pigment biosynthetic pathway. DFR2 was mapped to the W4 locus, functional analyses of soybean genes and genetic where the allele containing Tgm9 was termed w4-m. In this study we have demonstrated improvement in soybean. PLoS ONE 12(8): that previously identified morphological mutants (three chlorophyll deficient mutants, one e0180732. https://doi.org/10.1371/journal. male sterile-female fertile mutant, and three partial female sterile mutants) were caused by pone.0180732 insertion of Tgm9 following its excision from DFR2.
    [Show full text]
  • NIH Public Access Author Manuscript ACS Chem Biol
    NIH Public Access Author Manuscript ACS Chem Biol. Author manuscript; available in PMC 2013 July 20. NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: ACS Chem Biol. 2012 July 20; 7(7): 1198–1204. doi:10.1021/cb300024c. Novel Inhibitors for PRMT1 Discovered by High–Throughput Screening Using Activity–Based Fluorescence Polarization Myles B. C. Dillon1, Daniel A. Bachovchin1, Steven J. Brown1, M.G. Finn2, Hugh Rosen1, Benjamin F. Cravatt1, and Kerri A. Mowen*,1 1Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 2Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 Abstract Protein Arginine Methyltransferases (PRMTs) catalyze the posttranslational methylation of arginine using S–adenosyl–methionine (SAM) as a methyl–donor. The PRMT family is widely expressed and has been implicated in biological functions such as RNA splicing, transcriptional control, signal transduction, and DNA repair. Therefore, specific inhibitors of individual PRMTs have potentially significant research and therapeutic value. In particular, PRMT1 is responsible for >85% of arginine methyltransferase activity, but currently available inhibitors of PRMT1 lack specificity, efficacy, and bioavailability. To address this limitation, we developed a high– throughput screening assay for PRMT1 that utilizes a hyper–reactive cysteine within the active– site, which is lacking in almost all other PRMTs. This assay, which monitors the kinetics of the fluorescence polarization signal increase upon PRMT1 labeling by a rhodamine–containing cysteine–reactive probe, successfully identified two novel inhibitors selective for PRMT1 over other SAM–dependent methyltransferases.
    [Show full text]
  • PRMT1 and PRMT8 Regulate Retinoic Acid Dependent Neuronal
    EMBRYONIC STEM CELLS/INDUCED PLURIPOTENT STEM CELLS 1Department of Biochemistry and Molecular PRMT1 and PRMT8 Regulate Retinoic Acid De‐ Biology, University of Debrecen, Debrecen, Hungary, H‐4012; 2Department of Physiolo‐ pendent Neuronal Differentiation with Impli‐ gy, University of Debrecen, Debrecen, Hun‐ gary, H‐4012; 3Department of Dermatology, cations to Neuropathology University of Debrecen, Debrecen, Hungary, 4 H‐4012; Department of Surgery and Opera‐ 1 1 1 2 tive Techniques, Faculty of Dentistry, Uni‐ Zoltan Simandi , Erik Czipa , Attila Horvath , Aron Koszeghy , versity of Debrecen, Debrecen, Hungary, H‐ 2 1 3,4 5 Csilla Bordas , Szilárd Póliska , István Juhász , László 4012; Department of Biophysics and Cell 5 5 6 1 biology, University of Debrecen, Debrecen, Imre , Gábor Szabó , Balazs Dezso , Endre Barta , Sa‐ Hungary, H‐4012; 6Department of Patholo‐ scha Sauer7, Katalin Karolyi8, Ilona Kovacs8, Gábor gy, University of Debrecen, Debrecen, Hun‐ 9 9 9 gary, H‐4012; 7Otto Warburg Laboratory, Hutóczky , László Bognár , Álmos Klekner , Peter 2,10 1 1,11,12# Max Planck Institute for Molecular Genetics, Szucs , Bálint L.Bálint and Laszlo Nagy Berlin, Germany; 8Department of Pathology, Kenézy Hospital and Outpatient Clinic, Bar‐ tok Bela 2‐26, Debrecen, Hungary, H‐4010; Key words. PRMT1 • PRMT8 • retinoid signaling • neural differentiation • 9Department of Neurosurgery, University of glioblastoma Debrecen, Debrecen, Hungary, H‐4012; 10MTA‐DE‐NAP B‐Pain Control Group, Uni‐ versity of Debrecen, Debrecen, Hungary, H‐ ABSTRACT 4012; 11MTA‐DE “Lendulet” Immunogenom‐ ics Research Group, University of Debrecen, Retinoids are morphogens and have been implicated in cell fate commit‐ Debrecen, Hungary, H‐4012; 12Sanford‐ Burnham Medical Research Institute at Lake ment of embryonic stem cells (ESCs) to neurons.
    [Show full text]
  • CNOT4 Antibody (Pab)
    21.10.2014CNOT4 antibody (pAb) Rabbit Anti-Human/Mouse/Rat CCR4-NOT Transcription Complex Subunit 4 (NOT4, NOT4H) Instruction Manual Catalog Number PK-AB718-4813 Synonyms CNOT4 Antibody: CCR4-NOT transcription complex subunit 4, NOT4, NOT4H Description CNOT4 is a component of the CCR4-NOT transcription complex, a complex that is implicated in the repression of RNA polymerase II transcription. In the CCR4-NOT complex, CNOT4 acts as an E3 ubiquitin-protein ligase and interacts with a subset of E2 ubiquitin-conjugating enzymes through a unique C4C4 RING domain. This E3 ligase activity was shown to be dependent on the selective and specific interaction with the ubiquitin conjugating enzyme UbcH5B. In yeast, mutations in CNOT4 that prevented its interaction with the UbcH5B homolog UBC4 caused increased sensitivity to hydroxyurea, heat shock, and hygromycin B, suggesting that CNOT4 and UbcH5B are involved in stress response in vivo. Multiple isoforms of CNOT4 are known to exist. Quantity 100 µg Source / Host Rabbit Immunogen CNOT4 antibody was raised against a 19 amino acid peptide near the amino terminus of the human CNOT4. Purification Method Affinity chromatography purified via peptide column. Clone / IgG Subtype Polyclonal antibody Species Reactivity Human, Mouse, Rat Specificity Formulation Antibody is supplied in PBS containing 0.02% sodium azide. Reconstitution During shipment, small volumes of antibody will occasionally become entrapped in the seal of the product vial. For products with volumes of 200 μl or less, we recommend gently tapping the vial on a hard surface or briefly centrifuging the vial in a tabletop centrifuge to dislodge any liquid in the container’s cap.
    [Show full text]