(12) Patent Application Publication (10) Pub. No.: US 2002/014 1975 A1 Olmsted Et Al

Total Page:16

File Type:pdf, Size:1020Kb

(12) Patent Application Publication (10) Pub. No.: US 2002/014 1975 A1 Olmsted Et Al US 2002O141975A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2002/014 1975 A1 Olmsted et al. (43) Pub. Date: Oct. 3, 2002 (54) ALPHAVIRUS VECTORS AND WIROSOMES Publication Classification WITH MODIFIED HIV GENES FOR USE IN WACCINES (51) Int. Cl." ......................... A61K 48/00; A61K 39/21; A61K 39/12; CO7H 21/04; (75) Inventors: Robert Olmsted, Chapel Hill, NC AO1N 63/00; CO7K 1/00; (US); Paula Keith, Holly Springs, NC C07K 14/00; CO7K 17/00 (US); Sergey Dryga, Chapel Hill, NC (52) U.S. Cl. ................. 424/93.2; 424/188.1; 424/186.1; (US); Ian Caley, Durham, NC (US); 530/350; 530/826; 536/23.72 Maureen Maughan, Durham, NC (US); Robert Johnston, Chapel Hill, NC (US); Nancy Davis, Chapel Hill, (57) ABSTRACT NC (US); Ronald Swanstrom, Chapel The present invention provides methods and compositions Hill, NC (US) comprising a population of alphavirus replicon particles Correspondence Address: comprising two or more isolated nucleic acids Selected from NEEDLE & ROSENBERG PC 1) an isolated nucleic acid encoding an env gene product or 127 PEACHTREE STREET N E an immunogenic fragment thereof of a human immunode ATLANTA, GA 30303-1811 (US) ficiency virus, 2) an isolated nucleic acid encoding a gag gene product or an immunogenic fragment thereof of a (73) ASSignee: Alpha Vax, Inc. human immunodeficiency virus, wherein the gag gene prod (21) Appl. No.: 09/991,258 uct or immunogenic fragment thereof is modified to inhibit formation of Virus-like particles containing the gag gene (22) Filed: Nov. 16, 2001 product or the immunogenic fragment thereof and their release from a cell, and 3) an isolated nucleic acid encoding Related U.S. Application Data a pol gene product or an immunogenic fragment thereof of (63) Continuation-in-part of application No. 09/902,537, a human immunodeficiency virus, wherein the poll gene filed on Jul. 9, 2001, now abandoned. product or immunogenic fragment thereof is modified to inhibit protease, integrase, RNase H and/or reverse tran (60) Provisional application No. 60/216,995, filed on Jul. Scriptase activity, and wherein the nucleic acids are each 7, 2000. contained within a separate alphavirus replicon particle. Aba I (12497) EcoRI (12354), KN(R) COLE1 OR EcoRI (2137) nSP2 gag 26S promoter M nsP3 Apa I (7506) nsP4 Patent Application Publication Oct. 3, 2002. Sheet 1 of 15 US 2002/0141975 A1 Xba I (12497) EcoRI (12354). T7P KN(R) COLE1 OR EcoRI (2137) nSP2 of I (9232) gag 26S promoter es ' ' ' ...? nsP3 Apa I (7506) f nsP4 FIG. 1 Patent Application Publication Oct. 3, 2002. Sheet 2 of 15 US 2002/0141975 A1 Xba I (5047) TTP EcoRI (4904) , KN(R) ^ 26S promoter COLE1 ORI Not I (1782) FIG. 2 Patent Application Publication Oct. 3, 2002. Sheet 3 of 15 US 2002/0141975 A1 Xba I (6960) EcoRI (6817) T7P KNR) \ M 26S promoter E2 COLE1 ORI Xba I (1930 Not I (3695) FIG. 3 Patent Application Publication Oct. 3, 2002. Sheet 4 of 15 US 2002/0141975 A1 COLE1 OR nSP2 Not I (9085) p51 26S promoter - InsP3 FIG. 4 Patent Application Publication Oct. 3, 2002. Sheet 5 of 15 US 2002/0141975 A1 Xba I (13549) EcoRI (13406), KNR) lalla nsP coLE1 OR - EcoRI (2131) nsP2 pERK-DU151env Noti (10284) 13584 bp DU15 env V nsP3 Xbai (8642) 26S promoter / Apa I (7500) nsP4 FIG. 5 Patent Application Publication Oct. 3, 2002. Sheet 6 of 15 US 2002/0141975 A1 . * * * . x' \* * :::::::3% . ; ; , s: ' ' ' . 8.3: vs. x - 8: ... ::::::. ::::::: ... : : FIG. 6 Patent Application Publication Oct. 3, 2002. Sheet 8 of 15 US 2002/0141975 A1 Patent Application Publication Oct. 3, 2002 Sheet 9 of 15 US 2002/0141975 A1 1 OO 75 2 > 50 - S. | 25 O 1 OO 50 25 12 6 3 E/T Ratio FIG. 9A 1 OO 50 25 12 6 3 E/T Ratio FIG. 9B 1 OO 75 92 >9. 50 SS 25 O 1 OO 50 25 12 6 3 E IT Ratio FIG. 9C Patent Application Publication Oct. 3, 2002. Sheet 10 of 15 US 2002/0141975 A1 Patent Application Publication Oct. 3, 2002 Sheet 14 of 15 US 2002/0141975A1 Patent Application Publication Oct. 3, 2002 Sheet 15 of 15 US 2002/0141975 A1 US 2002/014 1975 A1 Oct. 3, 2002 ALPHAVIRUS VECTORS AND WIROSOMES WITH Immediately following infection, the nsPs are produced by MODIFIED HIV GENES FOR USE IN WACCNES translation of parental genomes and catalyze the Synthesis of 0001. This application is a continuation-in-part of and a full-length negative-Sense copy of the genome. This Serves claims priority to, U.S. application Ser. No. 09/902,537, as a template for the Synthesis of progeny plus-Stranded filed Jul. 9, 2001 (abandoned), which claims priority to genomeS. provisional application Serial No. 60/216,995, filed Jul. 7, 0009. The negative-sense copy of the genome also serves 2000, which applications are incorporated by reference as the template for the synthesis of subgenomic mRNA at herein in their entirety. approximately 10-fold molar exceSS relative to genomic RNA in infected cells (Schlesinger and Schlesinger, 1990). BACKGROUND OF THE INVENTION Synthesis of Subgenomic 26S mRNA is initiated from the highly active internal 26S mRNA promoter, which is func 0002) 1. Field of the Invention tional only on the negative-Sense RNA. The Subgenomic 0003. The present invention relates to vaccines using mRNA corresponds to the 3' one-third of the genome and Viral antigens, and in particular, to vaccines for the treatment encodes the alphavirus Structural proteins. and prevention of human immunodeficiency virus (HIV) 0010 Full-length, infectious cDNA clones of the RNA infection. The vaccines of this invention comprise alphavi genome of VEE Davis et al., 1989) have been constructed, ruS RNA replicon Systems which contain nucleic acid a panel of mutations which Strongly attenuate the virus have Sequence encoding antigens for eliciting an immune been identified (Johnston and Smith, 1988; Davis et al., response to HIV. 1990), and various constellations of these attenuating muta 0004 2. Background tions have been inserted into the clones to generate Several live attenuated VEE vaccine candidates (Davis et al., 1991; 0005. The successful control of the AIDS epidemic will 1995b, Grieder et al., 1995). The resulting vaccine candi require an effective vaccine for human immunodeficiency dates are avirulent and provide complete protection against virus type 1 (HIV) that significantly reduces or prevents the lethal virus challenge in rodents, horses and nonhuman Spread of infection. Currently, Several viral vector Systems primates. as well as naked DNA are at various Stages of pre-clinical and clinical evaluation as candidate HIV Vaccines. Recom 0011. The alphavirus VRPs are propagation defective, binant poxyiruses are the most widely Studied virus vectors Single cycle vectors that contain a Self-amplifying alphavirus and are furthest along in clinical development (e.g., RNA (replicon RNA) in which the structural protein genes ALVAC). of the virus are replaced by a heterologous antigen gene to be expressed. Alphavirus VRPs are typically made in cul 0006 The alphavirus-based replicon particle systems, tured cells, referred to as packaging cells. Following intro Such as the ones described in U.S. Pat. No. 5,792,462 and duction into mammalian cells, the replicon RNA ig pack herein referred to as “VRPs,” have multiple distinct prop aged into VRP by Supplying the Structural proteins in erties that make them attractive as an HIV vaccine delivery “trans, i.e., the cells are co-transfected with both replicon technology. These properties include: natural targeting to RNA and one or more separate helper RNAS which together and expression in lymphoid tissues (an optimal site for encode the full complement of alphavirus Structural pro induction of an immune response); high antigen expression teins. Importantly, only the replicon RNA is packaged into levels, e.g., up to 20% of total cell protein; induction of VRP, as the helper RNA(s) lack the cis-acting packaging balanced humoral, cellular, and mucosal immune responses, Sequence required for encapsidation. Thus, the VRPs are Sustained efficacy over multiple Simultaneous or Sequential defective, in that they can only infect target cells in culture inoculations of the vector; and a high margin of Safety. or in Vivo, where they express the heterologous antigen gene 0007 Venezuelan equine encephalitis virus (VEE) is a to high level, but they lack critical portions of the VEE member of the Alphaviruses group, which also includes the genome (i.e., the VEE Structural protein genes) necessary to prototype Sindbis virus (SIN) and Semliki Forest virus produce virus particles which could spread to other cells. (SFV), and is comprised of enveloped viruses containing 0012 Delivery of the replicon RNA into target cells (for plus-Stranded RNA genomes within icosahedral capsids vaccination) is facilitated by the VRP following infection of (Strauss, 1994). Alphavirus genomes are: approximately the target cells. In the cytoplasm of the target cell, the 11.5 kb long, capped, polyadenylated, and infectious under replicon RNA is first translated to produce the viral replicase appropriate transfection conditions. The nucleocapsid is proteins necessary to initiate Self-amplification and expres composed of 240 molecules of the capsid protein arranged Sion. The heterologous antigen gene is encoded by a Sub as a T=4 icosahedron, and is Surrounded by a lipoprotein genomic mRNA, abundantly transcribed from the replicon envelope (Paredes et al., 1993). Protruding from the virion RNA, leading to high level expression of the heterologous Surface are 80 glycoprotein Spikes, each of which is a trimer antigen gene product. Since the VEE Structural protein genes of Virally encoded E1 and E2 glycoprotein heterodimers. are not encoded by the replicon RNA delivered to the target The Virions contain no host proteins.
Recommended publications
  • Interactions De La Protéine Nsp1 Du Virus Chikungunya Avec Les Membranes De L’Hôte Et Conséquences Fonctionnelles William Bakhache
    Interactions de la protéine nsP1 du virus Chikungunya avec les membranes de l’hôte et conséquences fonctionnelles William Bakhache To cite this version: William Bakhache. Interactions de la protéine nsP1 du virus Chikungunya avec les membranes de l’hôte et conséquences fonctionnelles. Médecine humaine et pathologie. Université Montpellier, 2020. Français. NNT : 2020MONTT008. tel-03132645 HAL Id: tel-03132645 https://tel.archives-ouvertes.fr/tel-03132645 Submitted on 5 Feb 2021 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. THÈSE POUR OBTENIR LE GRADE DE DOCTEUR DE L’UNIVERSITÉ DE M ONTPELLIER En Biologie Santé École Doctorale Sciences Chimiques et Biologiques pour la Santé UMR 9004 - Institut de Recherche en Infectiologie de Montpellier Interactions de la protéine nsP1 du virus Chikungunya avec les membranes de l’hôte et conséquences fonctionnelles Présentée par William Bakhache Le 27 Mars 2020 Sous la direction de Laurence Briant Devant le jury composé de Jean-Luc Battini, Directeur de Recherche, IRIM, Montpellier Président de jury Ali Amara, Directeur de recherche, U944 INSERM, Paris Rapporteur Juan Reguera, Chargé de recherche, AFMB, Marseille Rapporteur Raphaël Gaudin, Chargé de recherche, IRIM, Montpellier Examinateur Yves Rouillé, Directeur de Recherche, CIIL, Lille Examinateur Bruno Coutard, Professeur des universités, UVE, Marseille Co-encadrant de thèse Laurence Briant, Directeur de recherche, IRIM, Montpellier Directeur de thèse 1 Acknowledgments I want to start by thanking the members of my thesis jury.
    [Show full text]
  • Lessons Learned from Ebola Virus
    membranes Review SARS-CoV-2 Cellular Infection and Therapeutic Opportunities: Lessons Learned from Ebola Virus Jordana Muñoz-Basagoiti 1,†, Daniel Perez-Zsolt 1,†, Jorge Carrillo 1 , Julià Blanco 1,2 , Bonaventura Clotet 1,2,3 and Nuria Izquierdo-Useros 1,* 1 IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Spain; [email protected] (J.M.-B.); [email protected] (D.P.-Z.); [email protected] (J.C.); [email protected] (J.B.); [email protected] (B.C.) 2 Infectious Diseases and Immunity Department, Faculty of Medicine, University of Vic (UVic-UCC), 08500 Vic, Spain 3 Infectious Diseases Department, Germans Trias i Pujol Hospital, 08916 Badalona, Spain * Correspondence: [email protected] † These authors contribution is equally to this work. Abstract: Viruses rely on the cellular machinery to replicate and propagate within newly infected individuals. Thus, viral entry into the host cell sets up the stage for productive infection and disease progression. Different viruses exploit distinct cellular receptors for viral entry; however, numerous viral internalization mechanisms are shared by very diverse viral families. Such is the case of Ebola virus (EBOV), which belongs to the filoviridae family, and the recently emerged coronavirus SARS- CoV-2. These two highly pathogenic viruses can exploit very similar endocytic routes to productively infect target cells. This convergence has sped up the experimental assessment of clinical therapies against SARS-CoV-2 previously found to be effective for EBOV, and facilitated their expedited clinical testing. Here we review how the viral entry processes and subsequent replication and egress strategies of EBOV and SARS-CoV-2 can overlap, and how our previous knowledge on antivirals, Citation: Muñoz-Basagoiti, J.; antibodies, and vaccines against EBOV has boosted the search for effective countermeasures against Perez-Zsolt, D.; Carrillo, J.; Blanco, J.; the new coronavirus.
    [Show full text]
  • A SARS-Cov-2-Human Protein-Protein Interaction Map Reveals Drug Targets and Potential Drug-Repurposing
    A SARS-CoV-2-Human Protein-Protein Interaction Map Reveals Drug Targets and Potential Drug-Repurposing Supplementary Information Supplementary Discussion All SARS-CoV-2 protein and gene functions described in the subnetwork appendices, including the text below and the text found in the individual bait subnetworks, are based on the functions of homologous genes from other coronavirus species. These are mainly from SARS-CoV and MERS-CoV, but when available and applicable other related viruses were used to provide insight into function. The SARS-CoV-2 proteins and genes listed here were designed and researched based on the gene alignments provided by Chan et. al. 1 2020 .​ Though we are reasonably sure the genes here are well annotated, we want to note that not every ​ protein has been verified to be expressed or functional during SARS-CoV-2 infections, either in vitro or in vivo. ​ ​ In an effort to be as comprehensive and transparent as possible, we are reporting the sub-networks of these functionally unverified proteins along with the other SARS-CoV-2 proteins. In such cases, we have made notes within the text below, and on the corresponding subnetwork figures, and would advise that more caution be taken when examining these proteins and their molecular interactions. Due to practical limits in our sample preparation and data collection process, we were unable to generate data for proteins corresponding to Nsp3, Orf7b, and Nsp16. Therefore these three genes have been left out of the following literature review of the SARS-CoV-2 proteins and the protein-protein interactions (PPIs) identified in this study.
    [Show full text]
  • The Role of F-Box Proteins During Viral Infection
    Int. J. Mol. Sci. 2013, 14, 4030-4049; doi:10.3390/ijms14024030 OPEN ACCESS International Journal of Molecular Sciences ISSN 1422-0067 www.mdpi.com/journal/ijms Review The Role of F-Box Proteins during Viral Infection Régis Lopes Correa 1, Fernanda Prieto Bruckner 2, Renan de Souza Cascardo 1,2 and Poliane Alfenas-Zerbini 2,* 1 Department of Genetics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-970, Brazil; E-Mails: [email protected] (R.L.C.); [email protected] (R.S.C.) 2 Department of Microbiology/BIOAGRO, Federal University of Viçosa, Viçosa, MG 36570-000, Brazil; E-Mail: [email protected] * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +55-31-3899-2955; Fax: +55-31-3899-2864. Received: 23 October 2012; in revised form: 14 December 2012 / Accepted: 17 January 2013 / Published: 18 February 2013 Abstract: The F-box domain is a protein structural motif of about 50 amino acids that mediates protein–protein interactions. The F-box protein is one of the four components of the SCF (SKp1, Cullin, F-box protein) complex, which mediates ubiquitination of proteins targeted for degradation by the proteasome, playing an essential role in many cellular processes. Several discoveries have been made on the use of the ubiquitin–proteasome system by viruses of several families to complete their infection cycle. On the other hand, F-box proteins can be used in the defense response by the host. This review describes the role of F-box proteins and the use of the ubiquitin–proteasome system in virus–host interactions.
    [Show full text]
  • Annual Conference 2016
    Annual Conference 2016 POSTER ABSTRACT BOOK 21-24 MARCH 2016 ACC, LIVERPOOL, UK ANNUAL CONFERENCE 2016 SESSION 1 – MEMBRANE TRANSPORTERS S1/P1 the pump in this complex and it is conserved between bacterial species, with an average of 78.5% identity between the DNA Novel tripartite tricarboxylate transporters sequences and approximately 80% similarity between the amino acid sequences amongst Enterobacteriaceae. This pump acts as from Rhodopseudomonas palustris a drug-proton antiporter, four residues have been previously Leonardo Talachia Rosa, John Rafferty, reported as essential for proton translocation in Escherichia coli AcrB: D407, D408, K940 and T978. AcrB of E. coli has an identity David Kelly of 86% and a 94% similarity to that of S. Typhimurium. Based on The University of Sheffield, Sheffield, UK these data, we constructed an AcrB D408A chromosomal mutant in S. Typhimurium SL1344. Western blotting confirmed that the Rhodopseudomonas palustris is a soil non-sulfur purple mutant had the same level of expression of AcrB as the parental bacterium, with ability to degrade lignin-derived compounds and wild type strain. The mutant had no growth deficiencies either in also to generate high yields of hydrogen gas, what raises several LB or MOPS minimal media. However, compared with wild type biotechnological interests in this bacterium. Degradation SL1344, the mutant had decreased efflux activity and was pathways, though, must begin with substrate uptake. In this multi-drug hyper-susceptible. Interestingly, the phenotype of the context, Soluble Binding Proteins (SBP`s) dependant AcrB D408A mutant was almost identical to that of an ΔacrB transporters are responsible for high-affinity and specificity mutant.
    [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]
  • In Vitro Evolution of High-Titer, Virus-Like Vesicles Containing a Single Structural Protein
    In vitro evolution of high-titer, virus-like vesicles containing a single structural protein Nina F. Rosea, Linda Buonocorea, John B. Schella, Anasuya Chattopadhyaya, Kapil Bahla, Xinran Liub, and John K. Rosea,1 aDepartment of Pathology, Yale University School of Medicine, New Haven, CT 06510; and bCenter for Cellular and Molecular Imaging, Yale University School of Medicine, New Haven, CT 06510 Edited* by Robert A. Lamb, Northwestern University, Evanston, IL, and approved October 22, 2014 (received for review August 6, 2014) Self-propagating, infectious, virus-like vesicles (VLVs) are gener- RNA. These proteins form a complex that directs replication of ated when an alphavirus RNA replicon expresses the vesicular the genomic RNA to form antigenomic RNA, which is then stomatitis virus glycoprotein (VSV G) as the only structural protein. copied to form full-length positive strand RNA and a subgenomic The mechanism that generates these VLVs lacking a capsid protein mRNA that encodes the structural proteins. The capsid protein has remained a mystery for over 20 years. We present evidence encases the genomic RNA in the cytoplasm and then buds from that VLVs arise from membrane-enveloped RNA replication facto- the cell surface in a membrane containing the SFV glycoproteins. ries (spherules) containing VSV G protein that are largely trapped Alphavirus RNA replication occurs inside light-bulb shaped, on the cell surface. After extensive passaging, VLVs evolve to membrane-bound compartments called spherules that initially grow to high titers through acquisition of multiple point muta- form on the cell surface and are then endocytosed to form cyto- tions in their nonstructural replicase proteins.
    [Show full text]
  • The SARS-Coronavirus Infection Cycle: a Survey of Viral Membrane Proteins, Their Functional Interactions and Pathogenesis
    International Journal of Molecular Sciences Review The SARS-Coronavirus Infection Cycle: A Survey of Viral Membrane Proteins, Their Functional Interactions and Pathogenesis Nicholas A. Wong * and Milton H. Saier, Jr. * Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0116, USA * Correspondence: [email protected] (N.A.W.); [email protected] (M.H.S.J.); Tel.: +1-650-763-6784 (N.A.W.); +1-858-534-4084 (M.H.S.J.) Abstract: Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is a novel epidemic strain of Betacoronavirus that is responsible for the current viral pandemic, coronavirus disease 2019 (COVID- 19), a global health crisis. Other epidemic Betacoronaviruses include the 2003 SARS-CoV-1 and the 2009 Middle East Respiratory Syndrome Coronavirus (MERS-CoV), the genomes of which, particularly that of SARS-CoV-1, are similar to that of the 2019 SARS-CoV-2. In this extensive review, we document the most recent information on Coronavirus proteins, with emphasis on the membrane proteins in the Coronaviridae family. We include information on their structures, functions, and participation in pathogenesis. While the shared proteins among the different coronaviruses may vary in structure and function, they all seem to be multifunctional, a common theme interconnecting these viruses. Many transmembrane proteins encoded within the SARS-CoV-2 genome play important roles in the infection cycle while others have functions yet to be understood. We compare the various structural and nonstructural proteins within the Coronaviridae family to elucidate potential overlaps Citation: Wong, N.A.; Saier, M.H., Jr.
    [Show full text]
  • Unconventional Viral Gene Expression Mechanisms As Therapeutic Targets
    Review Unconventional viral gene expression mechanisms as therapeutic targets https://doi.org/10.1038/s41586-021-03511-5 Jessica Sook Yuin Ho1,3, Zeyu Zhu1,3 & Ivan Marazzi1,2 ✉ Received: 8 June 2020 Accepted: 22 March 2021 Unlike the human genome that comprises mostly noncoding and regulatory sequences, Published online: 19 May 2021 viruses have evolved under the constraints of maintaining a small genome size while expanding the efciency of their coding and regulatory sequences. As a result, viruses Check for updates use strategies of transcription and translation in which one or more of the steps in the conventional gene–protein production line are altered. These alternative strategies of viral gene expression (also known as gene recoding) can be uniquely brought about by dedicated viral enzymes or by co-opting host factors (known as host dependencies). Targeting these unique enzymatic activities and host factors exposes vulnerabilities of a virus and provides a paradigm for the design of novel antiviral therapies. In this Review, we describe the types and mechanisms of unconventional gene and protein expression in viruses, and provide a perspective on how future basic mechanistic work could inform translational eforts that are aimed at viral eradication. Expression of a gene in the human genome is a multistep and heavily (for example, alternative splicing) or use unique strategies. Here we regulated process that resembles a production line. Protein-coding describe the diverse ways by which viral genomes give rise to genes and genes are transcribed almost exclusively by RNA polymerase II (RNAPII). proteins that deviate from the canonical framework of human genes, During transcription, quality-control checkpoints are implemented to restricting our analyses to eukaryotes and their viruses.
    [Show full text]
  • A SARS-Cov-2 Protein Interaction Map Reveals Targets for Drug Repurposing
    Article A SARS-CoV-2 protein interaction map reveals targets for drug repurposing https://doi.org/10.1038/s41586-020-2286-9 A list of authors and affiliations appears at the end of the paper Received: 23 March 2020 Accepted: 22 April 2020 A newly described coronavirus named severe acute respiratory syndrome Published online: 30 April 2020 coronavirus 2 (SARS-CoV-2), which is the causative agent of coronavirus disease 2019 (COVID-19), has infected over 2.3 million people, led to the death of more than Check for updates 160,000 individuals and caused worldwide social and economic disruption1,2. There are no antiviral drugs with proven clinical efcacy for the treatment of COVID-19, nor are there any vaccines that prevent infection with SARS-CoV-2, and eforts to develop drugs and vaccines are hampered by the limited knowledge of the molecular details of how SARS-CoV-2 infects cells. Here we cloned, tagged and expressed 26 of the 29 SARS-CoV-2 proteins in human cells and identifed the human proteins that physically associated with each of the SARS-CoV-2 proteins using afnity-purifcation mass spectrometry, identifying 332 high-confdence protein–protein interactions between SARS-CoV-2 and human proteins. Among these, we identify 66 druggable human proteins or host factors targeted by 69 compounds (of which, 29 drugs are approved by the US Food and Drug Administration, 12 are in clinical trials and 28 are preclinical compounds). We screened a subset of these in multiple viral assays and found two sets of pharmacological agents that displayed antiviral activity: inhibitors of mRNA translation and predicted regulators of the sigma-1 and sigma-2 receptors.
    [Show full text]
  • Alphavirus-Induced Membrane Rearrangements During Replication, Assembly, and Budding
    pathogens Review Alphavirus-Induced Membrane Rearrangements during Replication, Assembly, and Budding Zeinab Elmasri 1,2, Benjamin L. Nasal 2 and Joyce Jose 1,2,* 1 Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA; [email protected] 2 Department of Biochemistry & Molecular Biology, Eberly College of Science, The Pennsylvania State University, University Park, PA 16802, USA; [email protected] * Correspondence: [email protected]; Tel.: +1-814-863-8806 Abstract: Alphaviruses are arthropod-borne viruses mainly transmitted by hematophagous insects that cause moderate to fatal disease in humans and other animals. Currently, there are no approved vaccines or antivirals to mitigate alphavirus infections. In this review, we summarize the current knowledge of alphavirus-induced structures and their functions in infected cells. Throughout their lifecycle, alphaviruses induce several structural modifications, including replication spherules, type I and type II cytopathic vacuoles, and filopodial extensions. Type I cytopathic vacuoles are replication-induced structures containing replication spherules that are sites of RNA replication on the endosomal and lysosomal limiting membrane. Type II cytopathic vacuoles are assembly induced structures that originate from the Golgi apparatus. Filopodial extensions are induced at the plasma membrane and are involved in budding and cell-to-cell transport of virions. This review provides an overview of the viral and host factors involved in the biogenesis and function of these virus-induced structures. Understanding virus–host interactions in infected cells will lead to the identification of Citation: Elmasri, Z.; Nasal, B.L.; new targets for antiviral discovery. Jose, J. Alphavirus-Induced Membrane Rearrangements during Keywords: Togaviridae; alphavirus; spherule; replication; cytopathic vacuole; nucleocapsid core; Replication, Assembly, and Budding.
    [Show full text]
  • Medical Countermeasures Analysis of 2019-Ncov and Vaccine Risks for Antibody- Dependent Enhancement (ADE)
    Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1 Medical Countermeasures Analysis of 2019-nCoV and Vaccine Risks for Antibody- dependent Enhancement (ADE) Darrell O. Ricke, PhD1 & Robert W. Malone2, MD 1Biological and Chemical Technologies Massachusetts Institute of Technology Lincoln Laboratory Lexington, MA USA 2Chief Medical Officer, Alchem Laboratories 13305 Rachel Boulevard, Alachua FL 32615 USA [email protected] ORCID: 0000-0003-0340-7490 Corresponding author: Darrell O. Ricke, Ph.D. Email: [email protected] ORCID for Darrell Ricke is 0000-0002-2842-2809 Summary Background In 80% of patients, COVID-19 presents as mild disease1,2. 20% of cases develop severe (13%) or critical (6%) illness. More severe forms of COVID-19 present as clinical severe acute respiratory syndrome, but include a T-predominant lymphopenia3, high circulating levels of proinflammatory cytokines and chemokines, accumulation of neutrophils and macrophages in lungs, and immune dysregulation including immunosuppression4. Methods All major SARS-CoV-2 proteins were characterized using an amino acid residue variation analysis method. Results predict that most SARS-CoV-2 proteins are evolutionary constrained, with the exception of the spike (S) protein extended outer surface. Results were interpreted based on known SARS-like coronavirus virology and pathophysiology, with a focus on medical countermeasure development implications. Findings Non-neutralizing antibodies to variable S domains may enable an alternative infection pathway via Fc receptor-mediated uptake. This may be a gating event for the immune response dysregulation observed in more severe COVID-19 disease. Prior studies involving vaccine candidates for FCoV5,6 SARS-CoV-17-10 and Middle East Respiratory Syndrome coronavirus (MERS-CoV) 11 demonstrate vaccination-induced antibody-dependent enhancement of disease (ADE), including infection of phagocytic antigen presenting cells (APC).
    [Show full text]