On Mechanisms of Zika Virus-Induced Microcephaly and Host Invasion [Version 2; Peer Review: 2 Approved, 1 Approved with Reservations, 1 Not Approved]

Total Page:16

File Type:pdf, Size:1020Kb

On Mechanisms of Zika Virus-Induced Microcephaly and Host Invasion [Version 2; Peer Review: 2 Approved, 1 Approved with Reservations, 1 Not Approved] F1000Research 2017, 5:1919 Last updated: 30 JUL 2021 RESEARCH NOTE Predicted protein interactions of IFITMs may shed light on mechanisms of Zika virus-induced microcephaly and host invasion [version 2; peer review: 2 approved, 1 approved with reservations, 1 not approved] Previously titled: Predicted protein interactions of IFITMs which inhibit Zika virus infection Madhavi K. Ganapathiraju1,2, Kalyani B. Karunakaran3, Josefina Correa-Menéndez2 1Intelligent Systems Program, University of Pittsburgh, Pittsburgh, PA, USA 2Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA 3Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore, India v2 First published: 05 Aug 2016, 5:1919 Open Peer Review https://doi.org/10.12688/f1000research.9364.1 Latest published: 21 Nov 2017, 5:1919 https://doi.org/10.12688/f1000research.9364.2 Reviewer Status Invited Reviewers Abstract After the first reported case of Zika virus (ZIKV) in Brazil, in 2015, a 1 2 3 4 significant increase in the reported cases of microcephaly was observed. Microcephaly is a neurological condition in which the version 2 infant’s head is significantly smaller with complications in brain (revision) report report report development. Recently, two small membrane-associated interferon- 21 Nov 2017 inducible transmembrane proteins (IFITM1 and IFITM3) have been shown to repress members of the flaviviridae family which includes version 1 ZIKV. However, the exact mechanisms leading to the inhibition of the 05 Aug 2016 report report report virus are yet unknown. Here, we assembled an interactome of IFITM1 and IFITM3 with known protein-protein interactions (PPIs) collected from publicly available databases and novel PPIs predicted using the 1. Judith Klein-Seetharaman, University of High-confidence Protein-Protein Interaction Prediction (HiPPIP) Warwick, Warwick, UK model. We analyzed the functional and pathway associations of the interacting proteins, and found that there are several immunity 2. Nicholas Eyre, University of Adelaide, pathways (toll-like receptor signaling, cd28 signaling in T-helper cells, Adelaide, Australia crosstalk between dendritic cells and natural killer cells), neuronal pathways (axonal guidance signaling, neural tube closure and actin 3. Sandeep Chakraborty , University of cytoskeleton signaling) and developmental pathways (neural tube California, Davis, USA closure, embryonic skeletal system development) that are associated with these interactors. Our novel PPIs associate cilia dysfunction in 4. Rama Rao Damerla , Memorial Sloan ependymal cells to microcephaly, and may also shed light on potential Kettering Cancer Center, New York, USA targets of ZIKV for host invasion by immunosuppression and cytoskeletal rearrangements. These results could help direct future Any reports and responses or comments on the research in elucidating the mechanisms underlying host defense to ZIKV and other flaviviruses. Page 1 of 20 F1000Research 2017, 5:1919 Last updated: 30 JUL 2021 Keywords article can be found at the end of the article. Zika, , virus infection, protein interaction, interferon-inducible transmembrane proteins This article is included in the Disease Outbreaks gateway. This article is included in the Zika & Arbovirus Outbreaks collection. Corresponding author: Madhavi K. Ganapathiraju ([email protected]) Author roles: Ganapathiraju MK: Conceptualization, Data Curation, Formal Analysis, Funding Acquisition, Investigation, Methodology, Project Administration, Supervision, Visualization, Writing – Original Draft Preparation, Writing – Review & Editing; Karunakaran KB: Investigation, Writing – Original Draft Preparation; Correa-Menéndez J: Writing – Original Draft Preparation Competing interests: No competing interests were disclosed. Grant information: MKG’s work is funded by Biobehavioral Research Awards for Innovative New Scientists (BRAINS) grant (R01MH094564) from National Institute of Mental Health of National Institutes of Health of USA. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Copyright: © 2017 Ganapathiraju MK et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. How to cite this article: Ganapathiraju MK, Karunakaran KB and Correa-Menéndez J. Predicted protein interactions of IFITMs may shed light on mechanisms of Zika virus-induced microcephaly and host invasion [version 2; peer review: 2 approved, 1 approved with reservations, 1 not approved] F1000Research 2017, 5:1919 https://doi.org/10.12688/f1000research.9364.2 First published: 05 Aug 2016, 5:1919 https://doi.org/10.12688/f1000research.9364.1 Page 2 of 20 F1000Research 2017, 5:1919 Last updated: 30 JUL 2021 Recently, it was discovered that two small membrane-associated REVISE D Amendments from Version 1 interferon inducible transmembrane proteins (IFITMs) IFITM1 In this revision, we have added additional details to the and IFITM3 play a protective role against ZIKV infection by Introduction and Methods sections, and added a substantial inhibiting replication of the virus and preventing cell death7. The Discussion section that presents 3 hypotheses about mechanisms IFITM protein family belongs to a group of small (10 – 15kDa)20 of Zika-virus induced microcephaly and host-invasion by the interferon stimulated genes (ISGs), which are in turn produced by virus, based on the functional significance of predicted interactors the interferon system21. The interferon system is the host’s primary of IFITMs. We have added 2 new authors, Kalyani B. Karunakaran and Josefina Correa-Menéndez who contributed to this revision. response against infection which restricts entry and fusion from late endosomes22, with some ISGs being involved in suppressing We have added a brief description about HiPPIP, the 21,23,24 computational method that was employed to predict protein- the early stages of viral replication . IFITMs are located at protein interactions. This method has been described in detail in the cell plasma and endosomal membranes, which are the means our prior publication (npj Schizophrenia 2, Article number: 16012, of entry of many viruses25. This protein family protects the host 2016, doi:10.1038/npjschz.2016.12). against viral infection by directly restricting entry and fusion from See referee reports late endosomes26. Flaviviruses depend on the intracellular membrane of their host throughout their lifecycle27–29. Mice that are lacking immune Introduction sensors, signaling pathways and effector molecules were found Zika virus (ZIKV) is a flavivirus that was initially isolated from to be susceptible to flavivirus infection highlighting the impor- rhesus monkeys in 1947 and was first reported in humans in tance of the host’s innate immune response to these viruses5,30,31. 19521. The flavivirus genus contains around 70 viruses belong- Recent literature has demonstrated that during ZIKV infection, the ing to the family Flaviviridiae, a family of positive sense, single- production of several types of interferon and ISGs is increased, stranded, enveloped RNA viruses2, that also include Dengue virus, and that IFITM1 and IFITM3 are involved in inhibiting ZIKV West nile virus (WNV), tick-borne encephalitis virus, Japanese infection during the early stages of pathogenesis7,30–33. Enveloped encephalitis virus, yellow fever virus, and Hepatitis C virus3. Until viruses, such as ZIKV, have a membrane of host cell lipids that recently, ZIKV reports had been limited to Africa and Asia4 but contains viral fusion proteins; these proteins mediate fusion it became a wide-spread ZIKV epidemic5. The virus spread rap- between the virus and the endosomal membrane of the host cell, idly across the Americas and was declared a ‘global emergency’ a necessary step for initiating infection21,34. The precise mecha- by the World Health Organization6. It is mostly transmitted by nisms by which IFITMs inhibit infection are yet to be described. mosquitoes and clinical manifestations include rash, mild fever, However, IFITM3 has been shown to impact the function of arthralgia, conjunctivitis, myalgia, and headaches. In addition, vesicle-associated membrane protein (VAMP)-associated the virus can be transmitted sexually, with the risk of infection protein A (VAPA) and oxysterol-binding protein (OSBP) by persisting for several months after initial contact7. While the directly interacting with VAPA. This leads to the accumula- symptoms of ZIKV can be mild1, the virus has been linked to tion of cholesterol in multivesicular bodies and late endosomes, two more serious afflictions: Guillen-Barré syndrome (GBS)8,9 preventing the fusion of the intraluminal virion-containing and microcephaly10–14, both of which are serious neurological vesicles with endosomal membranes35. Additionally, IFITM1 has conditions. Microcephaly results in reduced head circumference been shown to suppress cell to cell fusion (syncytia formation) by measurement in infants, exhibiting complications in brain devel- localizing to the plasma membrane after interferon induction21. opment. Of particular concern is the attribution of microcephaly to infection with ZIKV occurring between the first two trimesters There are additional reasons to explore the role of IFITMs in of pregnancy13,14. Evidence linking ZIKV to microcephaly includes ZIKV restriction. For
Recommended publications
  • 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]
  • Opposing Activities of IFITM Proteins in SARS-Cov-2 Infection
    bioRxiv preprint doi: https://doi.org/10.1101/2020.08.11.246678; this version posted August 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. Opposing activities of IFITM proteins in SARS-CoV-2 infection Guoli Shi1*, Adam D. Kenney2,3*, Elena Kudryashova3,4, Lizhi Zhang2,3, Luanne Hall-Stoodley2, Richard T. Robinson2, Dmitri S. Kudryashov3,4, Alex A. Compton1,#, and Jacob S. Yount2,3,# 1HIV Dynamics and Replication Program, National Cancer Institute, Frederick, MD, USA. 2Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH, USA 3Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, OH, USA 4Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA *These authors contributed equally to this work #Address correspondence to Alex A. Compton, [email protected], and Jacob S. Yount, [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.08.11.246678; this version posted August 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. Abstract Interferon-induced transmembrane proteins (IFITMs) restrict infections by many viruses, but a subset of IFITMs enhance infections by specific coronaviruses through currently unknown mechanisms. Here we show that SARS-CoV-2 Spike-pseudotyped virus and genuine SARS- CoV-2 infections are generally restricted by expression of human IFITM1, IFITM2, and IFITM3, using both gain- and loss-of-function approaches.
    [Show full text]
  • Virus-Host Interaction: the Multifaceted Roles of Ifitms And
    Virus-Host Interaction: The Multifaceted Roles of IFITMs and LY6E in HIV Infection DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Jingyou Yu Graduate Program in Comparative and Veterinary Medicine The Ohio State University 2018 Dissertation Committee: Shan-Lu Liu, MD, PhD, Advisor Patrick L. Green, PhD Jianrong Li, DVM., PhD Jesse J. Kwiek, PhD Copyrighted by Jingyou Yu 2018 Abstract With over 1.8 million newly infected people each year, the worldwide HIV-1 epidemic remains an imperative challenge for public health. Recent work has demonstrated that type I interferons (IFNs) efficiently suppress HIV infection through induction of hundreds of interferon stimulated genes (ISGs). These ISGs target distinct infection stages of invading pathogens and shape innate immunity. Among these, interferon induced transmembrane proteins (IFITMs) and lymphocyte antigen 6 complex, locus E (LY6E) have been shown to differentially modulate viral infections. However, their effects on HIV are not fully understood. In my thesis work, I provided evidence in Chapter 2 showing that IFITM proteins, particularly IFITM2 and IFITM3, specifically antagonize the HIV-1 envelope glycoprotein (Env), thereby inhibiting viral infection. IFITM proteins interacted with HIV-1 Env in viral producer cells, leading to impaired Env processing and virion incorporation. Notably, the level of IFITM incorporation into HIV-1 virions did not strictly correlate with the extent of inhibition. Prolonged passage of HIV-1 in IFITM-expressing T lymphocytes led to emergence of Env mutants that overcome IFITM restriction. The ability of IFITMs to inhibit cell-to-cell infection can be extended to HIV-1 primary isolates, HIV-2 and SIVs; however, the extent of inhibition appeared to be virus- strain dependent.
    [Show full text]
  • Prognostic Significance of IFITM1 Expression and Correlation With
    Pathology - Research and Practice 215 (2019) 152444 Contents lists available at ScienceDirect Pathology - Research and Practice journal homepage: www.elsevier.com/locate/prp Prognostic significance of IFITM1 expression and correlation with microvessel density and epithelial–mesenchymal transition signature in lung T adenocarcinoma ⁎ Young Wha Koha, , Jae-Ho Hana, Dongjun Jeongb, Chang-Jin Kimb a Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea b Department of Pathology, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea ARTICLE INFO ABSTRACT Keywords: We evaluated the relationship between interferon-induced transmembrane protein 1 (IFITM1) expression, epi- Lung adenocarcinoma thelial–mesenchymal transition (EMT) signature and angiogenesis in lung adenocarcinoma. Additionally, we Interferon-induced transmembrane protein 1 examined prognostic significance of IFITM1 according to pTNM stage to confirm that IFITM1 can serve as a Microvessel complement to the pTNM stage. A total of 141 lung adenocarcinoma specimens were evaluated retrospectively Density by immunohistochemical staining for IFITM1, EMT markers (e-cadherin, β-catenin, and vimentin), and CD31 to Epithelial–mesenchymal transition measure microvessel density. IFITM1was expressed in 46.8% of the specimens. IFITM1 expression was sig- Prognosis nificantly correlated with increased microvessel density (P = 0.048). However, IFITM1 expression was not as- sociated with three EMT markers. In a multivariate analysis, IFITM1 was an independent prognostic factor for overall survival in a multivariate analysis (hazard ratio: 2.59, P = 0.01). Online database with data from 720 lung adenocarcinoma patients also revealed a negative prognostic significance of IFITM1 (P < 0.001). Furthermore, high IFITM1 expression was significantly correlated with decreased OS rates in each pTNM stage.
    [Show full text]
  • Human Macrophage Activation Programs Induced by Bacterial Pathogens
    Human macrophage activation programs induced by bacterial pathogens Gerard J. Nau*†‡, Joan F. L. Richmond*‡, Ann Schlesinger*‡, Ezra G. Jennings*§, Eric S. Lander*§, and Richard A. Young*§¶ *Whitehead Institute, 9 Cambridge Center, Cambridge, MA 02142; †Infectious Disease Unit, Massachusetts General Hospital, Boston, MA 02114; and §Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 Communicated by Gerald R. Fink, Whitehead Institute for Biomedical Research, Cambridge, MA, December 5, 2001 (received for review October 2, 2001) Understanding the response of innate immune cells to pathogens bia, MD). Monocytes were cultured at a density of 2 ϫ 107 may provide insights to host defenses and the tactics used by cells͞10 ml of DMEM (Invitrogen) with 20% FCS (Intergen, pathogens to circumvent these defenses. We used DNA microar- Purchase, NY), 10% human serum (Nabi, Boca Raton, FL), and rays to explore the responses of human macrophages to a variety 50 ␮g͞ml gentamicin (Invitrogen) in Primaria T-25 flasks (Bec- of bacteria. Macrophages responded to a broad range of bacteria ton Dickinson) for 5 days at 37°C, 5% CO2. On days 5 and 7, half with a robust, shared pattern of gene expression. The shared of the media was removed and replaced with media lacking FCS. response includes genes encoding receptors, signal transduction Media on the cultured macrophages was changed to 5 ml of molecules, and transcription factors. This shared activation pro- DMEM with 1% human serum on day 9, 1 hour before exper- gram transforms the macrophage into a cell primed to interact with iments were begun. its environment and to mount an immune response.
    [Show full text]
  • A Membrane Topology Model for Human Interferon Inducible Transmembrane Protein 1
    A Membrane Topology Model for Human Interferon Inducible Transmembrane Protein 1 Stuart Weston1*, Stephanie Czieso1, Ian J. White1, Sarah E. Smith2, Paul Kellam2,3, Mark Marsh1,3* 1 MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom, 2 Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom, 3 MRC Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, London, United Kingdom Abstract InterFeron Inducible TransMembrane proteins 1–3 (IFITM1, IFITM2 and IFITM3) are a family of proteins capable of inhibiting the cellular entry of numerous human and animal viruses. IFITM1-3 are unique amongst the currently described viral restriction factors in their apparent ability to block viral entry. This restrictive property is dependant on the localisation of the proteins to plasma and endosomal membranes, which constitute the main portals of viral entry into cells. The topology of the IFITM proteins within cell membranes is an unresolved aspect of their biology. Here we present data from immunofluorescence microscopy, protease cleavage, biotin-labelling and immuno-electron microscopy assays, showing that human IFITM1 has a membrane topology in which the N-terminal domain resides in the cytoplasm, and the C-terminal domain is extracellular. Furthermore, we provide evidence that this topology is conserved for all of the human interferon- induced IFITM proteins. This model is consistent with that recently proposed for murine IFITM3, but differs from that proposed for murine IFITM1. Citation: Weston S, Czieso S, White IJ, Smith SE, Kellam P, et al. (2014) A Membrane Topology Model for Human Interferon Inducible Transmembrane Protein 1.
    [Show full text]
  • Cx3cr1 Mediates the Development of Monocyte-Derived Dendritic Cells During Hepatic Inflammation
    CX3CR1 MEDIATES THE DEVELOPMENT OF MONOCYTE-DERIVED DENDRITIC CELLS DURING HEPATIC INFLAMMATION. Supplementary material Supplementary Figure 1: Liver CD45+ myeloid cells were pre-gated for Ly6G negative cells for excluding granulocytes and HDCs subsequently analyzed among the cells that were CD11c+ and had high expression of MHCII. Supplementary Table 1 low/- high + Changes in gene expression between CX3CR1 and CX3CR1 CD11b myeloid hepatic dendritic cells (HDCs) from CCl4-treated mice high Genes up-regulated in CX3CR1 HDCs Gene Fold changes P value Full name App 4,01702 5,89E-05 amyloid beta (A4) precursor protein C1qa 9,75881 1,69E-22 complement component 1, q subcomponent, alpha polypeptide C1qb 9,19882 3,62E-20 complement component 1, q subcomponent, beta polypeptide Ccl12 2,51899 0,011769 chemokine (C-C motif) ligand 12 Ccl2 6,53486 6,37E-11 chemokine (C-C motif) ligand 2 Ccl3 4,99649 5,84E-07 chemokine (C-C motif) ligand 3 Ccl4 4,42552 9,62E-06 chemokine (C-C motif) ligand 4 Ccl6 3,9311 8,46E-05 chemokine (C-C motif) ligand 6 Ccl7 2,60184 0,009272 chemokine (C-C motif) ligand 7 Ccl9 4,17294 3,01E-05 chemokine (C-C motif) ligand 9 Ccr2 3,35195 0,000802 chemokine (C-C motif) receptor 2 Ccr5 3,23358 0,001222 chemokine (C-C motif) receptor 5 Cd14 6,13325 8,61E-10 CD14 antigen Cd36 2,94367 0,003243 CD36 antigen Cd44 4,89958 9,60E-07 CD44 antigen Cd81 6,49623 8,24E-11 CD81 antigen Cd9 3,06253 0,002195 CD9 antigen Cdkn1a 4,65279 3,27E-06 cyclin-dependent kinase inhibitor 1A (P21) Cebpb 6,6083 3,89E-11 CCAAT/enhancer binding protein (C/EBP),
    [Show full text]
  • HIV-1 Mutates to Evade IFITM1 Restriction
    Virology 454-455 (2014) 11–24 Contents lists available at ScienceDirect Virology journal homepage: www.elsevier.com/locate/yviro HIV-1 mutates to evade IFITM1 restriction Shilei Ding a,b, Qinghua Pan a, Shan-Lu Liu d,nn, Chen Liang a,b,c,n a Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada H3T 1E2 b Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada H3A 2B4 c Department of Medicine, McGill University, Montreal, QC, Canada H3A 2B4 d Department of Molecular Microbiology & Immunology, School of Medicine, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310, USA article info abstract Article history: Interferon-induced transmembrane (IFITM) proteins inhibit the infection of a wide range of viruses Received 7 November 2013 including human immunodeficiency virus type 1 (HIV-1). At present, little is known about how viruses Returned to author for revisions overcome IFITM restriction. In this study, we have utilized HIV-1 as a model and selected IFITM1- 25 November 2013 resistant viruses after multiple passages of HIV-1 in IFITM1-expressing SupT1 cells. Sequencing the entire Accepted 25 January 2014 viral genome revealed several mutations in the vpu and envelope genes, among which mutations Vpu34 Available online 20 February 2014 and EnvG367E together enable efficient HIV-1 replication in IFITM1-expressing cells. Vpu34 introduces a Key words: stop codon at amino acid position 35 of Vpu, whereas EnvG367E changes the G367 residue at the CD4- HIV-1 binding site of gp120. These two mutations do not appear to overcome the downregulation of viral p24 IFITM1 expression caused by IFITM1, but rather enhance HIV-1 replication by promoting cell-to-cell virus Cell-to-cell transmission transmission.
    [Show full text]
  • IFITM3 Inhibits Virus-Triggered Induction of Type I Interferon by Mediating Autophagosome-Dependent Degradation of IRF3
    Cellular & Molecular Immunology (2018) 15, 858–867 & 2017 CSI and USTC All rights reserved 2042-0226/17 www.nature.com/cmi RESEARCH ARTICLE IFITM3 inhibits virus-triggered induction of type I interferon by mediating autophagosome-dependent degradation of IRF3 Li-Qun Jiang1, Tian Xia1, Yun-Hong Hu1, Ming-Shun Sun1, Shuang Yan1, Cao-Qi Lei1, Hong-Bing Shu1, Ji-Hua Guo2 and Yu Liu1 Interferon-induced transmembrane protein 3 (IFITM3) is a restriction factor that can be induced by viral infection and interferons (IFNs). It inhibits the entry and replication of many viruses, which are independent of receptor usage but dependent on processes that occur in endosomes. In this study, we demonstrate that IFITM3 plays important roles in regulating the RNA-virus-triggered production of IFN-β in a negative-feedback manner. Overexpression of IFITM3 inhibited Sendai virus-triggered induction of IFN-β, whereas knockdown of IFITM3 had the opposite effect. We also showed that IFITM3 was constitutively associated with IRF3 and regulated the homeostasis of IRF3 by mediating the autophagic degradation of IRF3. These findings suggest a novel inhibitory function of IFITM3 on the RNA-virus-triggered production of type I IFNs and cellular antiviral responses. Cellular & Molecular Immunology advance online publication, 24 April 2017; doi:10.1038/cmi.2017.15 Keywords: autophagy; IFITM3; IFNs; IRF3 INTRODUCTION TLRs and RIG-I-like receptors (RLRs) detect RNA virus As the first line of host defense, the innate immune system infection. For example, TLR3 recognizes viral double-stranded counters viral infection by expressing a number of intrinsic RNA (dsRNA) released by infected cells and triggers TIR- antiviral proteins, triggering the production of interferons domain- containing adapter-inducing interferon-β-mediated (IFNs) and facilitating the activation of adaptive immunity.
    [Show full text]
  • The IFITM1 Model Carlos E Parra-Herran, Liping Yuan, Marisa R Nucci and Bradley J Quade
    Modern Pathology (2014) 27, 569–579 & 2014 USCAP, Inc. All rights reserved 0893-3952/14 $32.00 569 Targeted development of specific biomarkers of endometrial stromal cell differentiation using bioinformatics: the IFITM1 model Carlos E Parra-Herran, Liping Yuan, Marisa R Nucci and Bradley J Quade Department of Pathology, Women’s and Perinatal Pathology Division, Brigham and Women’s Hospital, Boston, MA, USA When classifying cellular uterine mesenchymal neoplasms, histological distinction of endometrial stromal from smooth muscle neoplasms can be difficult. The only widely established marker of endometrial stromal differentiation, CD10, has marginal specificity. We took a bioinformatics approach to identify more specific markers of endometrial stromal differentiation by searching the Human Protein Atlas, a public database of protein expression profiles. After screening the database using different methods, interferon-induced transmembrane protein 1 (IFITM1) was selected for further analysis. Immunohistochemistry for IFITM1 was performed using tissue sections from the selected cases of proliferative endometrium (22), secretory endometrium (6), inactive endometrium (19), adenomyosis (10), conventional leiomyoma (11), cellular leiomyoma (16), endometrial stromal nodule (2), low-grade endometrial stromal sarcoma (16), high-grade endometrial stromal sarcoma (2) and undifferentiated uterine sarcoma (2). Stained slides were scored in terms of intensity and distribution. Normal endometrial samples uniformly showed diffuse and strong IFITM1 staining.
    [Show full text]
  • Comparative Transcriptome Analyses Reveal Genes Associated with SARS-Cov-2 Infection of Human Lung Epithelial Cells
    bioRxiv preprint doi: https://doi.org/10.1101/2020.06.24.169268; this version posted June 30, 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. Comparative transcriptome analyses reveal genes associated with SARS-CoV-2 infection of human lung epithelial cells Darshan S. Chandrashekar1, *, Upender Manne1,2,#, Sooryanarayana Varambally1,2,3,#* 1Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 2O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 3Informatics Institute, University of Alabama at Birmingham, Birmingham, AL *Correspondence to: Sooryanarayana Varambally, Ph.D., Molecular and Cellular Pathology, Department of Pathology, Wallace Tumor Institute, 4th floor, 20B, University of Alabama at Birmingham, Birmingham, AL 35233, USA Phone: (205) 996-1654 Email: [email protected] And Darshan S. Chandrashekar Ph.D., Department of Pathology, University of Alabama at Birmingham, Birmingham, AL Email: [email protected] # Share Senior Authorship (UM Email: [email protected]) Running Title: SARS-CoV-2 gene signature in infected lung epithelial cells Disclosure of Potential Conflicts of Interest: No potential conflicts of interest were disclosed. Page | 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.06.24.169268; this version posted June 30, 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: Understanding the molecular mechanism of SARS-CoV-2 infection (the cause of COVID-19) is a scientific priority for 2020. Various research groups are working toward development of vaccines and drugs, and many have published genomic and transcriptomic data related to this viral infection.
    [Show full text]
  • Interferon-Induced Transmembrane Protein 1 (IFITM1) Overexpression
    Ogony et al. Breast Cancer Research (2016) 18:25 DOI 10.1186/s13058-016-0683-7 RESEARCHARTICLE Open Access Interferon-induced transmembrane protein 1(IFITM1) overexpression enhances the aggressive phenotype of SUM149 inflammatory breast cancer cells in a signal transducer and activator of transcription 2 (STAT2)-dependent manner Joshua Ogony1, Hye Joung Choi1, Asona Lui1,2, Massimo Cristofanilli3 and Joan Lewis-Wambi1* Abstract Background: Inflammatory breast cancer (IBC) is a very aggressive and lethal subtype of breast cancer that accounts for about 4 % of all breast cancers diagnosed in the United States. Despite the efforts of several investigators to identify the molecular factors driving the aggressive phenotype of IBC, a great deal is still unknown about the molecular underpinnings of the disease. In the present study, we investigated the role of interferon-induced transmembrane protein 1 (IFITM1), a well-known interferon-stimulated gene (ISG), in promoting the aggressiveness of SUM149 IBC cells. Methods: Western blot and real-time polymerase chain reaction analyses were performed to assess the protein and messenger RNA (mRNA) levels of IFITM1 and other ISGs in three IBC cell lines: SUM149, MDA-IBC-3, and SUM190. IFITM1 expression and cellular localization were assessed by using immunofluorescence, while the tumorigenic potential was assessed by performing cell migration, invasion, and colony formation assays. Small interfering RNA and short hairpin RNA knockdowns, enzyme-linked immunosorbent assays, and luciferase assays were performed to determine the functional significance of IFITM1 and signal transducers and activators of transcription 1 and 2 (STAT1/2) in SUM149 cells. Results: We found that IFITM1 was constitutively overexpressed at the mRNA and protein levels in triple-negative SUM149 IBC cells, but that it was not expressed in SUM190 and MDA-IBC-3 IBC cells, and that suppression of IFITM1 or blockade of the IFNα signaling pathway significantly reduced the aggressive phenotype of SUM149 cells.
    [Show full text]