Emerin Suppresses Notch Signaling by Restricting the Notch Intracellular Domain to the Nuclear Membrane

Total Page:16

File Type:pdf, Size:1020Kb

Emerin Suppresses Notch Signaling by Restricting the Notch Intracellular Domain to the Nuclear Membrane Emerin Suppresses Notch Signaling by Restricting the Notch Intracellular Domain to the Nuclear Membrane Byongsun Lee a, Tae-Hee Lee b and Jaekyung Shim a,b* a Department of Molecular Biology, Sejong University, Seoul 05006, Republic of Korea b Laboratory for Cancer & Stem Cell Biology, Plant Engineering Institute, Sejong University, Seoul 05006, Korea Contact Information: *All correspondence should be addressed to Jaekyung Shim , Ph.D. Department of Molecular Biology, Sejong University, 98, Kunja-Dong, Kwangjin-Gu, Seoul 05006, Korea Phone: 82-2-3408-3944, Fax: 82-2-3408-4336, E-mail: [email protected] Running Title: Inhibition of Notch signaling by emerin Keywords: Emerin, Notch signaling, nuclear membrane Abbreviations: CSL, CBF1/suppressor hairless/Lag-1; NEXT, Notch1 extracellular truncation; NICD, Notch intracellular domain; ADAM, A disintegrin and metalloproteinase; CoA, Coactivators; CoR, Corepressors; PM, Plasma membrane; NE, Nuclear envelope; NGPS, Néstor- Guillermo progeria syndrome; LEM, LAP2-emerin-MAN1; TM, Transmembrane ABSTRACT Emerin is an inner nuclear membrane protein that is involved in maintaining the mechanical integrity of the nuclear membrane. Increasing evidence supports the involvement of emerin in the regulation of gene expression; however, its precise function remains to be elucidated. Here, we show that emerin downregulated genes downstream of Notch signaling, which are activated exclusively by the Notch intracellular domain (NICD). Deletion mutant experiments revealed that the transmembrane domain of emerin is important for the inhibition of Notch signaling. Emerin interacted directly and colocalized with the NICD at the nuclear membrane. Emerin knockdown induced the phosphorylation of ERK and AKT, increased endogenous Notch signaling, and inhibited hydrogen peroxide-induced apoptosis in HeLa cells. Notably, the downregulation of barrier-to-autointegration factor (BAF) or lamin A/C increased Notch signaling by inducing the release of emerin into the cytosol, implying that nuclear membrane-bound emerin acts as an endogenous inhibitor of Notch signaling. Taken together, our results indicate that emerin negatively regulates Notch signaling by promoting the retention of the NICD at the nuclear membrane. This mechanism could constitute a new therapeutic target for the treatment of emerin-related diseases. 1. Introduction The nuclear lamina establishes the mechanical support for the nucleus and provides a platform for protein interactions that contribute to gene regulation, DNA replication, and genome stability [ 1- 4]. Nuclear proteins that bind to the lamina include emerin, MAN1, lamin-associated polypeptide 2 (LAP2), and LEMD, which scaffold potentially hundreds of proteins [5, 6]. Multiple human diseases are caused by loss of individual nuclear lamina proteins, highlighting the importance of this network [2, 7]. Emerin was originally identified as a 35 kDa protein encoded by the EMD gene, which is located on the human X-chromosome. Emery-Dreifuss muscular dystrophy (EDMD) consists of X- linked EDMD (X-EDMD) and autosomal dominant EDMD (AD-EDMD) [4]. X-EDMD is caused by mutations in EMD (encoding Emerin) located on chromosome Xq28 and AD-EDMD is caused by mutations in LMNA (encoding Lamin A) located on chromosome 1q11-q23 [8-10]. EDMD is characterized by skeletal muscle wasting and cardiac defects, and mutations in the emerin gene that cause X-EDMD result in the loss of the emerin protein [11, 12]; however, the role of emerin loss in this disease has not been precisely elucidated. In addition to its known function in supporting the mechanical integrity of the nuclear membrane, emerin plays a role in the regulation of gene expression. Emerin interacts with many proteins, including nuclear lamins, germ cell-less (GCL), nesprin-1α, and BAF [13-16]. In particular, GCL proteins localize to the nuclear envelope and bind directly to emerin with high affinity [14, 17, 18]. The emerin bound GCL can also interact with the DP3 subunit of E2F-DP heterodimers and represses E2F-DP-dependent gene expression [17, 18]. However, emerin downregulation leads to the mislocalization of GCL from the nuclear envelope and the concomitant increase in E2F-DP-dependent gene expression, suggesting that emerin plays an important role in transcriptional repression by GCL. Similarly, the transcriptional repressor Btf (Bcl- 2-associated transcription factor), which is highly expressed in skeletal muscle [19, 20], interacts with emerin to exert its inhibitory role on transcription [21], suggesting that Btf may be relevant to EDMD. These findings indicate that emerin can negatively modulate gene expression through the recruitment of transcriptional repressors. Emerin also functions as a negative regulator of gene expression by trapping transcriptional activators at the nuclear membrane. For example, the direct interaction of emerin with β-catenin causes the downregulation of Wnt signaling [22]. In the absence of emerin, the levels of nuclear β- catenin increase, resulting in the upregulation of target genes [22, 23]. Emerin induces the nuclear envelope localization of LIM Domain Only 7 (LMO 7), a transcriptional activator for myogenic differentiation, and suppresses its transcriptional function [24]. The mislocalization of transcriptional activators could represent a new addition to the list of already well-known regulatory mechanisms of gene expression such as transcription, translation, and post-translational modification; however, few studies have explored this regulatory mechanism. In the present study, we examined the role of emerin as a transcriptional regulator and assessed its effect on gene expression using a transcription factor profiling assay covering 84 genes. We identified Notch signaling as a potential target pathway regulated by emerin. In Notch signaling, a Notch receptor interacts extracellularly with its canonical ligand on a contacting cell and is cleaved proteolytically by metalloproteinases and secretases. The NICD released from the receptor translocates to the nucleus, where it interacts with a CBF1/suppressor hairless/Lag-1 (CSL) family DNA-binding protein, resulting in the initiation of transcription of Notch target genes involved in several biological functions, such as development, growth, differentiation, and survival [25]. Here, we show that emerin can modulate Notch signaling by inducing the retention of the NICD at the nuclear membrane; thus, emerin may participate in some genetic laminopathy disorders, at least in part through the modulation of Notch signaling. 2. Materials and methods 2.1. Cell culture and transfection HeLa cells (American Type Culture Collection, Manassas, VA) were cultured in DMEM (Welgene, South Korea) supplemented with 10% FBS and 1% penicillin-streptomycin (Welgene, South Korea). Transfection was performed using the Lipofectamine 2000 reagent (Invitrogen, Grand Island, NY) according to the manufacturer's instruction. The transfected cells were cultured for 24–72 h, washed with DPBS, and harvested with lysis buffer (#FNN0011; Life technology, Grand Island, NY). 2.2. Antibodies For immunoblotting, primary antibodies specific for emerin (1:5000, #sc-15378,), BAF (1:100, #sc- 166324), lamin A/C (1:5000, #sc-20681), ERK1 (1:3000. #sc-93), pERK (1:2500, #sc-7383), pAKT/Thr 308 (1:3000, #sc-16646), AKT (1:3000, #sc-8312), pSTAT3 (1:2500, #sc-8059), and STAT3 (1:3000, #sc-482) were purchased from Santa Cruz Biotechnology (Santa Cruz Biotechnology, Santa Cruz, CA). NICD antibody (1:3000, #4147S) was obtained from Cell Signaling Technology (Cell Signaling Technology, Beverly, MA). Primary antibodies specific for HA (1:5000, #G036), Flag (1:1000, #G191), α-tubulin (1:5000, #G094), and β-actin (1:5000, #G043) were purchased from Applied Biological Materials (Applied Biological Materials, Richmond, BC, Canada). Primary antibodies specific for GAPDH (1:5000, #csb-ma000071m0m) and GST (1:3000, #csb- ma000031m0m) were purchased from Cusabio (Cusabio, Wuhan, China). HES1 (1:1000, #ab 5702) and HES5 (1:1000, #ab 5708) antibodies were purchased from Millipore (Millipore, Darmstadt, Germany). FITC (1:500, # 209-095-082) and TRITC (1:500, # 209-025-082) antibodies were purchased from Jackson Immuno Research Laboratories (Jackson Immuno Research Laboratories, West Grove, PA). 2.3. Plasmid constructs Human emerin and NICD cDNA were provided by 21C Frontier Human Gene Bank (South Korea). The emerin cDNA was amplified by PCR and inserted into the restriction enzyme sites of HA- pcDNA3 for biochemical studies. The amplified full-length NICD cDNA was inserted into the restriction enzyme sites of pEGFP-C1 (Clontech, Mountain View, CA) for immunocytochemistry. Alternatively, both amplified genes were inserted into the restriction enzyme sites of pcDNA3 for biological assays. To generate the GST-NICD or GST-YTHDC1 fusion protein, the coding region of each cDNA was amplified by PCR and inserted into the restriction enzyme sites of pGEX-4T-1 (GE Healthcare, Marlborough, MA). For construction of emerin deletion mutants or NICD deletion mutants, the corresponding regions were amplified by PCR and inserted into the restriction enzyme sites of HA-pcDNA3. 2.4. Transcription factor profiling assay Total RNA was isolated using Trizol reagent (Life Technologies), and 1 μg of total RNA was used for cDNA synthesis. The human transcription factor profiling PCR array was performed according to the manufacturer’s protocol (#PAHS-075ZC-2; Qiagen, Valencia, CA). Data were obtained using the manufacturer’s software. 2.5.
Recommended publications
  • SRC Antibody - N-Terminal Region (ARP32476 P050) Data Sheet
    SRC antibody - N-terminal region (ARP32476_P050) Data Sheet Product Number ARP32476_P050 Product Name SRC antibody - N-terminal region (ARP32476_P050) Size 50ug Gene Symbol SRC Alias Symbols ASV; SRC1; c-SRC; p60-Src Nucleotide Accession# NM_005417 Protein Size (# AA) 536 amino acids Molecular Weight 60kDa Product Format Lyophilized powder NCBI Gene Id 6714 Host Rabbit Clonality Polyclonal Official Gene Full Name V-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog (avian) Gene Family SH2D This is a rabbit polyclonal antibody against SRC. It was validated on Western Blot by Aviva Systems Biology. At Aviva Systems Biology we manufacture rabbit polyclonal antibodies on a large scale (200-1000 Description products/month) of high throughput manner. Our antibodies are peptide based and protein family oriented. We usually provide antibodies covering each member of a whole protein family of your interest. We also use our best efforts to provide you antibodies recognize various epitopes of a target protein. For availability of antibody needed for your experiment, please inquire (). Peptide Sequence Synthetic peptide located within the following region: QTPSKPASADGHRGPSAAFAPAAAEPKLFGGFNSSDTVTSPQRAGPLAGG This gene is highly similar to the v-src gene of Rous sarcoma virus. This proto-oncogene may play a role in the Description of Target regulation of embryonic development and cell growth. SRC protein is a tyrosine-protein kinase whose activity can be inhibited by phosphorylation by c-SRC kinase. Mutations in this gene could be involved in the
    [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]
  • Systematic Description of the Expression and Prognostic Value of M6A Regulators in Human Ovarian Cancer
    Systematic Description of the Expression and Prognostic Value of M6A Regulators in Human Ovarian Cancer Yuwei Chen Fujian Medical University Fujian Cancer Hospital Yang Sun ( [email protected] ) Fujian Cancer Hospital https://orcid.org/0000-0003-4224-8832 Xinbei Chen Fujian Medical University Fujian Cancer Hospital Siming Li Fujian Medical University Fujian Cancer Hospital Hongmei Xia Fujian Cancer Hospital Research Keywords: m6A regulators, m6A, ovarian cancer, Prognostic values, bioinformatics analysis. Posted Date: December 22nd, 2020 DOI: https://doi.org/10.21203/rs.3.rs-131682/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Systematic description of the expression and prognostic value of m6A regulators in human ovarian cancer 1 Yuwei Chen1, Yang Sun1*, Xinbei Chen1, Siming Li1, Hongmei Xia1 2 * Correspondence: 3 Yang Sun 4 [email protected] 5 Total words:5253 6 Total Figures and Tables: 11 Figures and 3 Tables 7 Keywords: m6A regulators, m6A, ovarian cancer, Prognostic values, 8 bioinformatics analysis. 9 Abstract 10 Background: N6-methyladenine (m6A) methylation, known as a kind of RNA 11 methylation regulator, has become a hotspot for research in the life sciences in recent 12 years. The existing studies revealed that m6A regulators helped to regulate the 13 progression of several malignant tumors. However, the expression mode and 14 prognostic value of m6A regulators in ovarian cancer have not been fully elucidated. 15 Methods: ONCOMINE, GEPIA, Human protein atlas, Kaplan-meier, UALCAN, 16 cBioPortal, GeneMANIA, DAVID, Sring, and Metascape were utilized in this study. 17 Results: In this study, through a comprehensive use of multiple database systems, 18 m6A regulators-induced mRNA and protein expression levels and their prognostic 19 value in the epithelial ovarian cancer of various histological types were analyzed.
    [Show full text]
  • A Novel Hypoxic Long Noncoding RNA KB-1980E6.3 Maintains Breast Cancer Stem Cell Stemness Via Interacting with IGF2BP1 to Facilitate C-Myc Mrna Stability
    Oncogene (2021) 40:1609–1627 https://doi.org/10.1038/s41388-020-01638-9 ARTICLE A novel hypoxic long noncoding RNA KB-1980E6.3 maintains breast cancer stem cell stemness via interacting with IGF2BP1 to facilitate c-Myc mRNA stability 1 2 3 4 1 1 1 1 1 Pengpeng Zhu ● Fang He ● Yixuan Hou ● Gang Tu ● Qiao Li ● Ting Jin ● Huan Zeng ● Yilu Qin ● Xueying Wan ● 1 1 5 1 Yina Qiao ● Yuxiang Qiu ● Yong Teng ● Manran Liu Received: 15 June 2020 / Revised: 13 November 2020 / Accepted: 18 December 2020 / Published online: 19 January 2021 © The Author(s), under exclusive licence to Springer Nature Limited 2021. This article is published with open access Abstract The hostile hypoxic microenvironment takes primary responsibility for the rapid expansion of breast cancer tumors. However, the underlying mechanism is not fully understood. Here, using RNA sequencing (RNA-seq) analysis, we identified a hypoxia-induced long noncoding RNA (lncRNA) KB-1980E6.3, which is aberrantly upregulated in clinical breast cancer tissues and closely correlated with poor prognosis of breast cancer patients. The enhanced lncRNA KB- 1980E6.3 facilitates breast cancer stem cells (BCSCs) self-renewal and tumorigenesis under hypoxic microenvironment both 1234567890();,: 1234567890();,: in vitro and in vivo. Mechanistically, lncRNA KB-1980E6.3 recruited insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) to form a lncRNA KB-1980E6.3/IGF2BP1/c-Myc signaling axis that retained the stability of c-Myc mRNA through increasing binding of IGF2BP1 with m6A-modified c-Myc coding region instability determinant (CRD) mRNA. In conclusion, we confirm that lncRNA KB-1980E6.3 maintains the stemness of BCSCs through lncRNA KB-1980E6.3/ IGF2BP1/c-Myc axis and suggest that disrupting this axis might provide a new therapeutic target for refractory hypoxic tumors.
    [Show full text]
  • The VE-Cadherin/Amotl2 Mechanosensory Pathway Suppresses Aortic InAmmation and the Formation of Abdominal Aortic Aneurysms
    The VE-cadherin/AmotL2 mechanosensory pathway suppresses aortic inammation and the formation of abdominal aortic aneurysms Yuanyuan Zhang Karolinska Institute Evelyn Hutterer Karolinska Institute Sara Hultin Karolinska Institute Otto Bergman Karolinska Institute Maria Forteza Karolinska Institute Zorana Andonovic Karolinska Institute Daniel Ketelhuth Karolinska University Hospital, Stockholm, Sweden Joy Roy Karolinska Institute Per Eriksson Karolinska Institute Lars Holmgren ( [email protected] ) Karolinska Institute Article Keywords: arterial endothelial cells (ECs), vascular disease, abdominal aortic aneurysms Posted Date: June 15th, 2021 DOI: https://doi.org/10.21203/rs.3.rs-600069/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License The VE-cadherin/AmotL2 mechanosensory pathway suppresses aortic inflammation and the formation of abdominal aortic aneurysms Yuanyuan Zhang1, Evelyn Hutterer1, Sara Hultin1, Otto Bergman2, Maria J. Forteza2, Zorana Andonovic1, Daniel F.J. Ketelhuth2,3, Joy Roy4, Per Eriksson2 and Lars Holmgren1*. 1Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden. 2Department of Medicine Solna, BioClinicum, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden. 3Department of Cardiovascular and Renal Research, Institutet of Molecular Medicine, Univ. of Southern Denmark, Odense, Denmark 4Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm,
    [Show full text]
  • Figure S1. Reverse Transcription‑Quantitative PCR Analysis of ETV5 Mrna Expression Levels in Parental and ETV5 Stable Transfectants
    Figure S1. Reverse transcription‑quantitative PCR analysis of ETV5 mRNA expression levels in parental and ETV5 stable transfectants. (A) Hec1a and Hec1a‑ETV5 EC cell lines; (B) Ishikawa and Ishikawa‑ETV5 EC cell lines. **P<0.005, unpaired Student's t‑test. EC, endometrial cancer; ETV5, ETS variant transcription factor 5. Figure S2. Survival analysis of sample clusters 1‑4. Kaplan Meier graphs for (A) recurrence‑free and (B) overall survival. Survival curves were constructed using the Kaplan‑Meier method, and differences between sample cluster curves were analyzed by log‑rank test. Figure S3. ROC analysis of hub genes. For each gene, ROC curve (left) and mRNA expression levels (right) in control (n=35) and tumor (n=545) samples from The Cancer Genome Atlas Uterine Corpus Endometrioid Cancer cohort are shown. mRNA levels are expressed as Log2(x+1), where ‘x’ is the RSEM normalized expression value. ROC, receiver operating characteristic. Table SI. Clinicopathological characteristics of the GSE17025 dataset. Characteristic n % Atrophic endometrium 12 (postmenopausal) (Control group) Tumor stage I 91 100 Histology Endometrioid adenocarcinoma 79 86.81 Papillary serous 12 13.19 Histological grade Grade 1 30 32.97 Grade 2 36 39.56 Grade 3 25 27.47 Myometrial invasiona Superficial (<50%) 67 74.44 Deep (>50%) 23 25.56 aMyometrial invasion information was available for 90 of 91 tumor samples. Table SII. Clinicopathological characteristics of The Cancer Genome Atlas Uterine Corpus Endometrioid Cancer dataset. Characteristic n % Solid tissue normal 16 Tumor samples Stagea I 226 68.278 II 19 5.740 III 70 21.148 IV 16 4.834 Histology Endometrioid 271 81.381 Mixed 10 3.003 Serous 52 15.616 Histological grade Grade 1 78 23.423 Grade 2 91 27.327 Grade 3 164 49.249 Molecular subtypeb POLE 17 7.328 MSI 65 28.017 CN Low 90 38.793 CN High 60 25.862 CN, copy number; MSI, microsatellite instability; POLE, DNA polymerase ε.
    [Show full text]
  • Migration of Thymic Pre-T Cells Regulates Development
    Alternative Splicing Controlled by Heterogeneous Nuclear Ribonucleoprotein L Regulates Development, Proliferation, and Migration of Thymic Pre-T Cells This information is current as of October 2, 2021. Marie-Claude Gaudreau, Florian Heyd, Rachel Bastien, Brian Wilhelm and Tarik Möröy J Immunol published online 20 April 2012 http://www.jimmunol.org/content/early/2012/04/20/jimmun ol.1103142 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2012/04/20/jimmunol.110314 Material 2.DC1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication by guest on October 2, 2021 *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2012 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published April 20, 2012, doi:10.4049/jimmunol.1103142 The Journal of Immunology Alternative Splicing Controlled by Heterogeneous Nuclear Ribonucleoprotein L Regulates Development, Proliferation, and Migration of Thymic Pre-T Cells Marie-Claude Gaudreau,*,† Florian Heyd,‡ Rachel Bastien,* Brian Wilhelm,x and Tarik Mo¨ro¨y*,† The regulation of posttranscriptional modifications of pre-mRNA by alternative splicing is important for cellular function, devel- opment, and immunity.
    [Show full text]
  • A High-Throughput Approach to Uncover Novel Roles of APOBEC2, a Functional Orphan of the AID/APOBEC Family
    Rockefeller University Digital Commons @ RU Student Theses and Dissertations 2018 A High-Throughput Approach to Uncover Novel Roles of APOBEC2, a Functional Orphan of the AID/APOBEC Family Linda Molla Follow this and additional works at: https://digitalcommons.rockefeller.edu/ student_theses_and_dissertations Part of the Life Sciences Commons A HIGH-THROUGHPUT APPROACH TO UNCOVER NOVEL ROLES OF APOBEC2, A FUNCTIONAL ORPHAN OF THE AID/APOBEC FAMILY A Thesis Presented to the Faculty of The Rockefeller University in Partial Fulfillment of the Requirements for the degree of Doctor of Philosophy by Linda Molla June 2018 © Copyright by Linda Molla 2018 A HIGH-THROUGHPUT APPROACH TO UNCOVER NOVEL ROLES OF APOBEC2, A FUNCTIONAL ORPHAN OF THE AID/APOBEC FAMILY Linda Molla, Ph.D. The Rockefeller University 2018 APOBEC2 is a member of the AID/APOBEC cytidine deaminase family of proteins. Unlike most of AID/APOBEC, however, APOBEC2’s function remains elusive. Previous research has implicated APOBEC2 in diverse organisms and cellular processes such as muscle biology (in Mus musculus), regeneration (in Danio rerio), and development (in Xenopus laevis). APOBEC2 has also been implicated in cancer. However the enzymatic activity, substrate or physiological target(s) of APOBEC2 are unknown. For this thesis, I have combined Next Generation Sequencing (NGS) techniques with state-of-the-art molecular biology to determine the physiological targets of APOBEC2. Using a cell culture muscle differentiation system, and RNA sequencing (RNA-Seq) by polyA capture, I demonstrated that unlike the AID/APOBEC family member APOBEC1, APOBEC2 is not an RNA editor. Using the same system combined with enhanced Reduced Representation Bisulfite Sequencing (eRRBS) analyses I showed that, unlike the AID/APOBEC family member AID, APOBEC2 does not act as a 5-methyl-C deaminase.
    [Show full text]
  • RNA M6 a Modification Enzymes Shape Innate Responses to DNA by Regulating Interferon Β
    Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press RNA m6A modification enzymes shape innate responses to DNA by regulating interferon β Rosa M. Rubio,1 Daniel P. Depledge,1 Christopher Bianco,1 Letitia Thompson,1 and Ian Mohr1,2 1Department of Microbiology, 2Laura and Isaac Perlmutter Cancer Institute, New York University School of Medicine, New York, New York 10016, USA Modification of mRNA by N6-adenosine methylation (m6A) on internal bases influences gene expression in eukaryotes. How the dynamic genome-wide landscape of m6A-modified mRNAs impacts virus infection and host immune responses remains poorly understood. Here, we show that type I interferon (IFN) production triggered by dsDNA or human cytomegalovirus (HCMV) is controlled by the cellular m6A methyltrasferase subunit METTL14 and ALKBH5 demethylase. While METTL14 depletion reduced virus reproduction and stimulated dsDNA- or HCMV-induced IFNB1 mRNA accumulation, ALKBH5 depletion had the opposite effect. Depleting METTL14 increased both nascent IFNB1 mRNA production and stability in response to dsDNA. In contrast, ALKBH5 deple- tion reduced nascent IFNB1 mRNA production without detectably influencing IFN1B mRNA decay. Genome-wide transcriptome profiling following ALKBH5 depletion identified differentially expressed genes regulating antiviral immune responses, while METTL14 depletion altered pathways impacting metabolic reprogramming, stress responses, and aging. Finally, we determined that IFNB1 mRNA was m6A-modified within both the coding sequence and the 3′ untranslated region (UTR). This establishes that the host m6A modification machinery controls IFNβ production triggered by HCMV or dsDNA. Moreover, it demonstrates that responses to nonmicrobial dsDNA in uninfected cells, which shape host immunity and contribute to autoimmune disease, are regulated by enzymes controlling m6A epitranscriptomic changes.
    [Show full text]
  • M6a)-Dependent Fate Decision for the Lncrna THOR Hongmei Liu1,Yuxinxu1,Bingyao1, Tingting Sui1,Liangxuelai1,2,3,4 and Zhanjun Li1
    Liu et al. Cell Death and Disease (2020) 11:613 https://doi.org/10.1038/s41419-020-02833-y Cell Death & Disease ARTICLE Open Access A novel N6-methyladenosine (m6A)-dependent fate decision for the lncRNA THOR Hongmei Liu1,YuxinXu1,BingYao1, Tingting Sui1,LiangxueLai1,2,3,4 and Zhanjun Li1 Abstract Previous studies have revealed the critical roles of the N6-methyladenosine (m6A) modification of long non-coding RNAs (lncRNAs) in cancers, but the relationship between the oncogenic role of the lncRNA THOR (a representative of cancer/testis lncRNAs) and m6A modification remains unclear. Here, we show that the internal m6A modification of the lncRNA THOR via an m6A-reader-dependent modality regulates the proliferation of cancer cells. Our findings demonstrated that the loss of the lncRNA THOR inhibits the proliferation, migration, and invasion of cancer cells in vitro and in vivo. In addition, m6A is highly enriched on lncRNA THOR transcripts, which contain GA (m6A) CA, GG (m6A) CU, and UG (m6A) CU sequence motifs. RIP-qRT-PCR and RNA pull-down assay results revealed that the specific m6A readers YTHDF1 and YTHDF2 can read the m6A motifs and regulate the stability of the lncRNA THOR (stabilization and decay). These m6A-dependent RNA-protein interactions can maintain the oncogenic role of the lncRNA THOR. Collectively, these findings highlight the critical role of the m6A modification in oncogenic lncRNA THOR and reveal a novel long non-coding RNA regulatory mechanism, providing a new way to explore RNA epigenetic regulatory patterns in the future. 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; Introduction Dysregulated expression of lncRNA THOR has been Long non-coding RNA (lncRNA), which is an abundant reported to modulate the progression of various types of and functionally diverse species of non-coding RNA cancers, such as melanoma, non-small cell lung cancer, – (ncRNA), is more than 200 nt transcripts long with lim- osteosarcoma and renal cell carcinoma10 12.
    [Show full text]
  • Whole Genome Sequencing Identifies Homozygous BRCA2 Deletion Guiding Treatment in De-Differentiated Prostate Cancer
    Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Whole Genome Sequencing identifies homozygous BRCA2 deletion guiding treatment in de-differentiated prostate cancer Karin Purshouse1~, Anna Schuh2~, Benjamin P Fairfax1, 5, Sam Knight3, Pavlos Antoniou2, Helene Dreau2, Niko Popitsch3, Kevin Gatter4, Ian Roberts5, Lisa Browning6, Zoe Traill7, David Kerr1, Clare Verrill6, Mark Tuthill1, Jenny C Taylor2,3*, Andrew Protheroe1* ~ joint first authors * Joint senior authors 1 - Oxford Cancer and Haematology Centre, Churchill Hospital, Headington, Oxford, OX3 7LE, UK. 2- Oxford National Institute for Health Research, Biomedical Research Centre/NHS Translational Diagnostics Centre, The Joint Research Office, Block 60, The Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK 3- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK 4- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK. 5- Molecular Oncology and Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, OX3 9DS, UK 6- Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 9DU, UK. 7- Department of Radiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 9DU, UK. Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Abstract Whole genome sequencing (WGS) has transformed the understanding of the genetic drivers of cancer and is increasingly being used in cancer medicine to identify personalised therapies. Here we describe a case where the application of WGS identified a tumoural BRCA2 deletion in a patient with aggressive dedifferentiated prostate cancer that was repeat biopsied after disease progression.
    [Show full text]
  • A Heroin Addiction Severity-Associated Intronic Single
    11048 • The Journal of Neuroscience, August 13, 2014 • 34(33):11048–11066 Cellular/Molecular A Heroin Addiction Severity-Associated Intronic Single Nucleotide Polymorphism Modulates Alternative Pre-mRNA Splicing of the ␮ Opioid Receptor Gene OPRM1 via hnRNPH Interactions Jin Xu,1* Zhigang Lu,1* Mingming Xu,1 Ling Pan,1 Yi Deng,1 Xiaohu Xie,3 Huifen Liu,3 Shixiong Ding,4 Yasmin L. Hurd,5 X Gavril W. Pasternak,1 X Robert J. Klein,2 Luca Cartegni,6 X Wenhua Zhou,3† and Ying-Xian Pan1† 1Department of Neurology and the Molecular Pharmacology & Chemistry Program, and 2Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, 3Laboratory of Behavioral Neuroscience, Ningbo Addiction Research and Treatment Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, People’s Republic of China, 4Laboratory of Clinical Biochemistry, Ningbo No. 2 Hospital, Zhejiang 315100, People’s Republic of China, 5Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, and 6Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 Single nucleotide polymorphisms (SNPs) in the OPRM1 gene have been associated with vulnerability to opioid dependence. The current study identifies an association of an intronic SNP (rs9479757) with the severity of heroin addiction among Han-Chinese male heroin addicts. Individual SNP analysis and haplotype-based analysis with additional SNPs in the OPRM1 locus showed that mild heroin addiction was associated with the AG genotype, whereas severe heroin addiction was associated with the GG genotype.
    [Show full text]