1
|
Ulloa-Aguilar JM, Herrera Moro Huitron L, Benítez-Zeferino RY, Cerna-Cortes JF, García-Cordero J, León-Reyes G, Guzman-Bautista ER, Farfan-Morales CN, Reyes-Ruiz JM, Miranda-Labra RU, De Jesús-González LA, León-Juárez M. The Nucleolus and Its Interactions with Viral Proteins Required for Successful Infection. Cells 2024; 13:1591. [PMID: 39329772 PMCID: PMC11430610 DOI: 10.3390/cells13181591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Nuclear bodies are structures in eukaryotic cells that lack a plasma membrane and are considered protein condensates, DNA, or RNA molecules. Known nuclear bodies include the nucleolus, Cajal bodies, and promyelocytic leukemia nuclear bodies. These bodies are involved in the concentration, exclusion, sequestration, assembly, modification, and recycling of specific components involved in the regulation of ribosome biogenesis, RNA transcription, and RNA processing. Additionally, nuclear bodies have been shown to participate in cellular processes such as the regulation of transcription of the cell cycle, mitosis, apoptosis, and the cellular stress response. The dynamics and functions of these bodies depend on the state of the cell. It is now known that both DNA and RNA viruses can direct their proteins to nuclear bodies, causing alterations in their composition, dynamics, and functions. Although many of these mechanisms are still under investigation, it is well known that the interaction between viral and nuclear body proteins is necessary for the success of the viral infection cycle. In this review, we concisely describe the interaction between viral and nuclear body proteins. Furthermore, we focus on the role of the nucleolus in RNA virus infections. Finally, we discuss the possible implications of the interaction of viral proteins on cellular transcription and the formation/degradation of non-coding RNAs.
Collapse
Affiliation(s)
- José Manuel Ulloa-Aguilar
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (J.M.U.-A.); (L.H.M.H.); (R.Y.B.-Z.); (E.R.G.-B.)
- Posgrado en Biología Experimental, Departamento de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09310, Mexico
| | - Luis Herrera Moro Huitron
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (J.M.U.-A.); (L.H.M.H.); (R.Y.B.-Z.); (E.R.G.-B.)
- Laboratorio de Microbiología Molecular, Departamento de Microbiología, Escuela Nacional de Ciencias Biologícas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Rocío Yazmin Benítez-Zeferino
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (J.M.U.-A.); (L.H.M.H.); (R.Y.B.-Z.); (E.R.G.-B.)
- Laboratorio de Microbiología Molecular, Departamento de Microbiología, Escuela Nacional de Ciencias Biologícas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Jorge Francisco Cerna-Cortes
- Laboratorio de Microbiología Molecular, Departamento de Microbiología, Escuela Nacional de Ciencias Biologícas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Julio García-Cordero
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico;
| | - Guadalupe León-Reyes
- Laboratorio de Nutrigenética y Nutrigenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Edgar Rodrigo Guzman-Bautista
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (J.M.U.-A.); (L.H.M.H.); (R.Y.B.-Z.); (E.R.G.-B.)
| | - Carlos Noe Farfan-Morales
- Departamento de Ciencias Naturales, Universidad Autonoma Metropolitana (UAM), Unidad Cuajimalpa, Mexico City 05348, Mexico;
| | - José Manuel Reyes-Ruiz
- Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS), Veracruz 91897, Mexico;
| | - Roxana U. Miranda-Labra
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09310, Mexico;
| | | | - Moises León-Juárez
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (J.M.U.-A.); (L.H.M.H.); (R.Y.B.-Z.); (E.R.G.-B.)
| |
Collapse
|
2
|
Itoh Y, Miyamoto Y, Tokunaga M, Suzuki T, Takada A, Ninomiya A, Hishinuma T, Matsuda M, Yoneda Y, Oka M, Suzuki R, Matsuura Y, Okamoto T. Importin-7-dependent nuclear translocation of the Flavivirus core protein is required for infectious virus production. PLoS Pathog 2024; 20:e1012409. [PMID: 39146232 PMCID: PMC11326614 DOI: 10.1371/journal.ppat.1012409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/08/2024] [Indexed: 08/17/2024] Open
Abstract
Flaviviridae is a family of positive-stranded RNA viruses, including human pathogens, such as Japanese encephalitis virus (JEV), dengue virus (DENV), Zika virus (ZIKV), and West Nile virus (WNV). Nuclear localization of the viral core protein is conserved among Flaviviridae, and this feature may be targeted for developing broad-ranging anti-flavivirus drugs. However, the mechanism of core protein translocation to the nucleus and the importance of nuclear translocation in the viral life cycle remain unknown. We aimed to identify the molecular mechanism underlying core protein nuclear translocation. We identified importin-7 (IPO7), an importin-β family protein, as a nuclear carrier for Flaviviridae core proteins. Nuclear import assays revealed that core protein was transported into the nucleus via IPO7, whereas IPO7 deletion by CRISPR/Cas9 impaired their nuclear translocation. To understand the importance of core protein nuclear translocation, we evaluated the production of infectious virus or single-round-infectious-particles in wild-type or IPO7-deficient cells; both processes were significantly impaired in IPO7-deficient cells, whereas intracellular infectious virus levels were equivalent in wild-type and IPO7-deficient cells. These results suggest that IPO7-mediated nuclear translocation of core proteins is involved in the release of infectious virus particles of flaviviruses.
Collapse
Affiliation(s)
- Yumi Itoh
- Department of Microbiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoichi Miyamoto
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Laboratory of Biofunctional Molecular Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Makoto Tokunaga
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tatsuya Suzuki
- Department of Microbiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Akira Takada
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Akinori Ninomiya
- Central Instrumentation Laboratory, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tomomi Hishinuma
- Department of Microbiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Mami Matsuda
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshihiro Yoneda
- The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
| | - Masahiro Oka
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Toru Okamoto
- Department of Microbiology, Juntendo University School of Medicine, Tokyo, Japan
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
3
|
Jing X, Wang P, Liu J, Xiang M, Song X, Wang C, Li P, Li H, Wu Z, Zhang C. A viral protein competitively bound to rice CIPK23 inhibits potassium absorption and facilitates virus systemic infection in rice. PLANT BIOTECHNOLOGY JOURNAL 2024; 22:2348-2363. [PMID: 38578842 PMCID: PMC11258980 DOI: 10.1111/pbi.14350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/02/2024] [Accepted: 03/20/2024] [Indexed: 04/07/2024]
Abstract
Potassium (K+) plays a crucial role as a macronutrient in the growth and development of plants. Studies have definitely determined the vital roles of K+ in response to pathogen invasion. Our previous investigations revealed that rice plants infected with rice grassy stunt virus (RGSV) displayed a reduction in K+ content, but the mechanism by which RGSV infection subverts K+ uptake remains unknown. In this study, we found that overexpression of RGSV P1, a specific viral protein encoded by viral RNA1, results in enhanced sensitivity to low K+ stress and exhibits a significantly lower rate of K+ influx compared to wild-type rice plants. Further investigation revealed that RGSV P1 interacts with OsCIPK23, an upstream regulator of Shaker K+ channel OsAKT1. Moreover, we found that the P1 protein recruits the OsCIPK23 to the Cajal bodies (CBs). In vivo assays demonstrated that the P1 protein competitively binds to OsCIPK23 with both OsCBL1 and OsAKT1. In the nucleus, the P1 protein enhances the binding of OsCIPK23 to OsCoilin, a homologue of the signature protein of CBs in Arabidopsis, and facilitates their trafficking through these CB structures. Genetic analysis indicates that mutant in oscipk23 suppresses RGSV systemic infection. Conversely, osakt1 mutants exhibited increased sensitivity to RGSV infection. These findings suggest that RGSV P1 hinders the absorption of K+ in rice plants by recruiting the OsCIPK23 to the CB structures. This process potentially promotes virus systemic infection but comes at the expense of inhibiting OsAKT1 activity.
Collapse
Affiliation(s)
- Xinxin Jing
- The Engineering Research Center for Plant Health Protection Technology in Henan ProvinceCollege of Plant ProtectionHenan Agricultural UniversityZhengzhouChina
- Fujian Province Key Laboratory of Plant VirologyCollege of Plant ProtectionFujian Agriculture and Forestry UniversityFuzhouChina
| | - Pengyue Wang
- The Engineering Research Center for Plant Health Protection Technology in Henan ProvinceCollege of Plant ProtectionHenan Agricultural UniversityZhengzhouChina
- Fujian Province Key Laboratory of Plant VirologyCollege of Plant ProtectionFujian Agriculture and Forestry UniversityFuzhouChina
| | - Jianjian Liu
- The Engineering Research Center for Plant Health Protection Technology in Henan ProvinceCollege of Plant ProtectionHenan Agricultural UniversityZhengzhouChina
- Hubei Engineering Research Center for Pest Forewarning and ManagementCollege of AgronomyYangtze UniversityJingzhouChina
| | - Meirong Xiang
- The Engineering Research Center for Plant Health Protection Technology in Henan ProvinceCollege of Plant ProtectionHenan Agricultural UniversityZhengzhouChina
| | - Xia Song
- Fujian Province Key Laboratory of Plant VirologyCollege of Plant ProtectionFujian Agriculture and Forestry UniversityFuzhouChina
| | - Chaonan Wang
- The Engineering Research Center for Plant Health Protection Technology in Henan ProvinceCollege of Plant ProtectionHenan Agricultural UniversityZhengzhouChina
| | - Pengbai Li
- The Engineering Research Center for Plant Health Protection Technology in Henan ProvinceCollege of Plant ProtectionHenan Agricultural UniversityZhengzhouChina
| | - Honglian Li
- The Engineering Research Center for Plant Health Protection Technology in Henan ProvinceCollege of Plant ProtectionHenan Agricultural UniversityZhengzhouChina
| | - Zujian Wu
- Fujian Province Key Laboratory of Plant VirologyCollege of Plant ProtectionFujian Agriculture and Forestry UniversityFuzhouChina
| | - Chao Zhang
- The Engineering Research Center for Plant Health Protection Technology in Henan ProvinceCollege of Plant ProtectionHenan Agricultural UniversityZhengzhouChina
| |
Collapse
|
4
|
King MR, Ruff KM, Lin AZ, Pant A, Farag M, Lalmansingh JM, Wu T, Fossat MJ, Ouyang W, Lew MD, Lundberg E, Vahey MD, Pappu RV. Macromolecular condensation organizes nucleolar sub-phases to set up a pH gradient. Cell 2024; 187:1889-1906.e24. [PMID: 38503281 DOI: 10.1016/j.cell.2024.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 01/02/2024] [Accepted: 02/22/2024] [Indexed: 03/21/2024]
Abstract
Nucleoli are multicomponent condensates defined by coexisting sub-phases. We identified distinct intrinsically disordered regions (IDRs), including acidic (D/E) tracts and K-blocks interspersed by E-rich regions, as defining features of nucleolar proteins. We show that the localization preferences of nucleolar proteins are determined by their IDRs and the types of RNA or DNA binding domains they encompass. In vitro reconstitutions and studies in cells showed how condensation, which combines binding and complex coacervation of nucleolar components, contributes to nucleolar organization. D/E tracts of nucleolar proteins contribute to lowering the pH of co-condensates formed with nucleolar RNAs in vitro. In cells, this sets up a pH gradient between nucleoli and the nucleoplasm. By contrast, juxta-nucleolar bodies, which have different macromolecular compositions, featuring protein IDRs with very different charge profiles, have pH values that are equivalent to or higher than the nucleoplasm. Our findings show that distinct compositional specificities generate distinct physicochemical properties for condensates.
Collapse
Affiliation(s)
- Matthew R King
- Department of Biomedical Engineering, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Kiersten M Ruff
- Department of Biomedical Engineering, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Andrew Z Lin
- Department of Biomedical Engineering, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Avnika Pant
- Department of Biomedical Engineering, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Mina Farag
- Department of Biomedical Engineering, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Jared M Lalmansingh
- Department of Biomedical Engineering, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Tingting Wu
- Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Department of Electrical and Systems Engineering, James F. McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Martin J Fossat
- Department of Biomedical Engineering, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Wei Ouyang
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA; Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Matthew D Lew
- Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Department of Electrical and Systems Engineering, James F. McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Emma Lundberg
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA; Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Michael D Vahey
- Department of Biomedical Engineering, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Rohit V Pappu
- Department of Biomedical Engineering, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Biomolecular Condensates, James McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
5
|
Nguyen NLT, Wu W, Panté N. Contribution of the Nuclear Localization Sequences of Influenza A Nucleoprotein to the Nuclear Import of the Influenza Genome in Infected Cells. Viruses 2023; 15:1641. [PMID: 37631984 PMCID: PMC10459959 DOI: 10.3390/v15081641] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Replication of the RNA genome of influenza A virus occurs in the nucleus of infected cells. The influenza nucleoprotein (NP) associated with the viral RNA into ribonucleoprotein complexes (vRNPs) is involved in the nuclear import of the viral genome. NP has two nuclear localization sequences (NLSs), NLS1 and NLS2. Most studies have concentrated on the role of NP's NLSs using in vitro-assembled or purified vRNPs, which may differ from incoming vRNPs released in the cytoplasm during an infection. Here, we study the contribution of the NP's NLSs to the nuclear import of vRNPs in a cell culture model system for influenza infection: human lung carcinoma cells infected with viruses containing NP-carrying mutations in NLS1 or NLS2 (NLS2MT), generated by reverse genetics. We found that cells infected with these mutant viruses were defective in the nuclear import of incoming vRNPs and produced reduced amounts of newly synthesized NP, newly assembled vRNP, and progeny virus. In addition, NLS2MT-infected cells were also defective in the nucleolar accumulation of NP, confirming the nucleolar localization role of NLS2. Our findings indicate that both NLS1 and NLS2 have to be present for successful infection and demonstrate the crucial role of these two NLSs in the infection cycle of the influenza A virus.
Collapse
Affiliation(s)
| | | | - Nelly Panté
- Department of Zoology and Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (N.L.T.N.); (W.W.)
| |
Collapse
|
6
|
Rawlinson SM, Zhao T, Ardipradja K, Zhang Y, Veugelers PF, Harper JA, David CT, Sundaramoorthy V, Moseley GW. Henipaviruses and lyssaviruses target nucleolar treacle protein and regulate ribosomal RNA synthesis. Traffic 2023; 24:146-157. [PMID: 36479968 PMCID: PMC10947316 DOI: 10.1111/tra.12877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022]
Abstract
The nucleolus is a common target of viruses and viral proteins, but for many viruses the functional outcomes and significance of this targeting remains unresolved. Recently, the first intranucleolar function of a protein of a cytoplasmically-replicating negative-sense RNA virus (NSV) was identified, with the finding that the matrix (M) protein of Hendra virus (HeV) (genus Henipavirus, family Paramyxoviridae) interacts with Treacle protein within nucleolar subcompartments and mimics a cellular mechanism of the nucleolar DNA-damage response (DDR) to suppress ribosomal RNA (rRNA) synthesis. Whether other viruses utilise this mechanism has not been examined. We report that sub-nucleolar Treacle targeting and modulation is conserved between M proteins of multiple Henipaviruses, including Nipah virus and other potentially zoonotic viruses. Furthermore, this function is also evident for P3 protein of rabies virus, the prototype virus of a different RNA virus family (Rhabdoviridae), with Treacle depletion in cells also found to impact virus production. These data indicate that unrelated proteins of viruses from different families have independently developed nucleolar/Treacle targeting function, but that modulation of Treacle has distinct effects on infection. Thus, subversion of Treacle may be an important process in infection by diverse NSVs, and so could provide novel targets for antiviral approaches with broad specificity.
Collapse
Affiliation(s)
- Stephen M. Rawlinson
- Department of Microbiology, Biomedicine Discovery InstituteMonash UniversityMelbourneVictoriaAustralia
| | - Tianyue Zhao
- Department of Microbiology, Biomedicine Discovery InstituteMonash UniversityMelbourneVictoriaAustralia
| | - Katie Ardipradja
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)Australian Centre for Disease Preparedness (ACDP)East GeelongVictoriaAustralia
| | - Yilin Zhang
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneMelbourneAustralia
| | - Patrick F. Veugelers
- Department of Microbiology, Biomedicine Discovery InstituteMonash UniversityMelbourneVictoriaAustralia
| | - Jennifer A. Harper
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)Australian Centre for Disease Preparedness (ACDP)East GeelongVictoriaAustralia
| | - Cassandra T. David
- Department of Microbiology, Biomedicine Discovery InstituteMonash UniversityMelbourneVictoriaAustralia
| | - Vinod Sundaramoorthy
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)Australian Centre for Disease Preparedness (ACDP)East GeelongVictoriaAustralia
- School of MedicineDeakin UniversityGeelongVictoriaAustralia
| | - Gregory W. Moseley
- Department of Microbiology, Biomedicine Discovery InstituteMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
7
|
Merino VF, Yan Y, Ordonez AA, Bullen CK, Lee A, Saeki H, Ray K, Huang T, Jain SK, Pomper MG. Nucleolin mediates SARS-CoV-2 replication and viral-induced apoptosis of host cells. Antiviral Res 2023; 211:105550. [PMID: 36740097 PMCID: PMC9896859 DOI: 10.1016/j.antiviral.2023.105550] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023]
Abstract
Host-oriented antiviral therapeutics are promising treatment options to combat COVID-19 and its emerging variants. However, relatively little is known about the cellular proteins hijacked by SARS-CoV-2 for its replication. Here we show that SARS-CoV-2 induces expression and cytoplasmic translocation of the nucleolar protein, nucleolin (NCL). NCL interacts with SARS-CoV-2 viral proteins and co-localizes with N-protein in the nucleolus and in stress granules. Knockdown of NCL decreases the stress granule component G3BP1, viral replication and improved survival of infected host cells. NCL mediates viral-induced apoptosis and stress response via p53. SARS-CoV-2 increases NCL expression and nucleolar size and number in lungs of infected hamsters. Inhibition of NCL with the aptamer AS-1411 decreases viral replication and apoptosis of infected cells. These results suggest nucleolin as a suitable target for anti-COVID therapies.
Collapse
Affiliation(s)
- Vanessa F Merino
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Yu Yan
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Alvaro A Ordonez
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - C Korin Bullen
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Albert Lee
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Harumi Saeki
- Department of Human Pathology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Krishanu Ray
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA; Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sanjay K Jain
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
Sun N, Zhang RR, Song GY, Cai Q, Aliyari SR, Nielsen-Saines K, Jung JU, Yang H, Cheng G, Qin CF. SERTAD3 induces proteasomal degradation of ZIKV capsid protein and represents a therapeutic target. J Med Virol 2023; 95:e28451. [PMID: 36594413 PMCID: PMC9975044 DOI: 10.1002/jmv.28451] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/18/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne RNA virus that belongs to the Flaviviridae family. While flavivirus replication is known to occur in the cytoplasm, a significant portion of the viral capsid protein localizes to the nucleus during infection. However, the role of the nuclear capsid is less clear. Herein, we demonstrated SERTA domain containing 3 (SERTAD3) as an antiviral interferon stimulatory gene product had an antiviral ability to ZIKV but not JEV. Mechanistically, we found that SERTAD3 interacted with the capsid protein of ZIKV in the nucleolus and reduced capsid protein abundance through proteasomal degradation. Furthermore, an eight amino acid peptide of SERTAD3 was identified as the minimum motif that binds with ZIKV capsid protein. Remarkably, the eight amino acids synthetic peptide from SERTAD3 significantly prevented ZIKV infection in culture and pregnant mouse models. Taken together, these findings not only reveal the function of SERTAD3 in promoting proteasomal degradation of a specific viral protein but also provide a promising host-targeted therapeutic strategy against ZIKV infection.
Collapse
Affiliation(s)
- Nina Sun
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, China
| | - Rong-Rong Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Guang-Yuan Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Qiaomei Cai
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, China
| | - Saba R. Aliyari
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Karin Nielsen-Saines
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Jae U. Jung
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Heng Yang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, China
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- School of Basic Medicine, Anhui Medical University, Hefei, China
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Liu Z, Larocque É, Xie Y, Xiao Y, Lemay G, Peloponese JM, Mesnard JM, Rassart É, Lin R, Zhou S, Zeng Y, Gao H, Cen S, Barbeau B. A newly identified interaction between nucleolar NPM1/B23 and the HTLV-I basic leucine zipper factor in HTLV-1 infected cells. Front Microbiol 2022; 13:988944. [PMID: 36532440 PMCID: PMC9753777 DOI: 10.3389/fmicb.2022.988944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/08/2022] [Indexed: 08/05/2023] Open
Abstract
Human T-cell leukemia virus type 1 is the causative agent of HTLV-1-associated myelopathy/tropical spastic paraparesis and adult T-cell leukemia-lymphoma (ATL). The HTLV-1 basic leucine zipper factor (HBZ) has been associated to the cancer-inducing properties of this virus, although the exact mechanism is unknown. In this study, we identified nucleophosmin (NPM1/B23) as a new interaction partner of HBZ. We show that sHBZ and the less abundant uHBZ isoform interact with nucleolar NPM1/B23 in infected cells and HTLV-1 positive patient cells, unlike equivalent antisense proteins of related non-leukemogenic HTLV-2, -3 and-4 viruses. We further demonstrate that sHBZ association to NPM1/B23 is sensitive to RNase. Interestingly, sHBZ was shown to interact with its own RNA. Through siRNA and overexpression experiments, we further provide evidence that NPM1/B23 acts negatively on viral gene expression with potential impact on cell transformation. Our results hence provide a new insight over HBZ-binding partners in relation to cellular localization and potential function on cell proliferation and should lead to a better understanding of the link between HBZ and ATL development.
Collapse
Affiliation(s)
- Zhenlong Liu
- Département de chimie, Université du Québec à Montréal, Montréal, QC, Canada
- Centre d’excellence en recherche sur les maladies orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada
- Lady Davis Institute, Jewish General Hospital & Department of Medicine, McGill University, Montreal, QC, Canada
| | - Émilie Larocque
- Centre d’excellence en recherche sur les maladies orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada
- Département de microbiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Yongli Xie
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, China
| | - Yong Xiao
- Département de chimie, Université du Québec à Montréal, Montréal, QC, Canada
- Centre d’excellence en recherche sur les maladies orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada
| | - Guy Lemay
- Département de microbiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Jean-Marie Peloponese
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université Montpellier, Montpellier, France
| | - Jean-Michel Mesnard
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université Montpellier, Montpellier, France
| | - Éric Rassart
- Centre d’excellence en recherche sur les maladies orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada
- Département des sciences biologiques, Université du Québec à Montréal, Montréal, QC, Canada
| | - Rongtuan Lin
- Lady Davis Institute, Jewish General Hospital & Department of Medicine, McGill University, Montreal, QC, Canada
| | - Shuang Zhou
- Neurosurgery Department, 2nd Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yiming Zeng
- Neurosurgery Department, 2nd Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Hongzhi Gao
- Neurosurgery Department, 2nd Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, China
| | - Benoit Barbeau
- Département de chimie, Université du Québec à Montréal, Montréal, QC, Canada
- Centre d’excellence en recherche sur les maladies orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada
- Département de microbiologie et immunologie, Université de Montréal, Montréal, QC, Canada
- Département des sciences biologiques, Université du Québec à Montréal, Montréal, QC, Canada
| |
Collapse
|
10
|
Wakata A, Katoh H, Kato F, Takeda M. Nucleolar Protein Treacle Is Important for the Efficient Growth of Mumps Virus. J Virol 2022; 96:e0072222. [PMID: 36135364 PMCID: PMC9555161 DOI: 10.1128/jvi.00722-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The nucleolus is the largest structure in the nucleus, and it plays roles in mediating cellular stress responses and regulating cell proliferation, as well as in ribosome biosynthesis. The nucleolus is composed of a variety of nucleolar factors that interact with each other in a complex manner to enable its function. Many viral proteins interact with nucleolar factors as well, affecting cellular morphology and function. Here, to investigate the association between mumps virus (MuV) infection and the nucleolus, we evaluated the necessity of nucleolar factors for MuV proliferation by performing a knockdown of these factors with small interfering (si)RNAs. Our results reveal that suppressing the expression of Treacle, which is required for ribosome biosynthesis, reduced the proliferative potential of MuV. Additionally, the one-step growth kinetics results indicate that Treacle knockdown did not affect the viral RNA and protein synthesis of MuV, but it did impair the production of infectious virus particles. Viral matrix protein (M) was considered a candidate Treacle interaction partner because it functions in the process of particle formation in the viral life cycle and is partially localized in the nucleolus. Our data confirm that MuV M can interact with Treacle and colocalize with it in the nucleolus. Furthermore, we found that viral infection induces relocalization of Treacle in the nucleus. Together, these findings suggest that interaction with Treacle in the nucleolus is important for the M protein to exert its functions late in the MuV life cycle. IMPORTANCE The nucleolus, which is the site of ribosome biosynthesis, is a target organelle for many viruses. It is increasingly evident that viruses can favor their own replication and multiplication by interacting with various nucleolar factors. In this study, we found that the nucleolar protein Treacle, known to function in the transcription and processing of pre-rRNA, is required for the efficient propagation of mumps virus (MuV). Specifically, our data indicate that Treacle is not involved in viral RNA or protein synthesis but is important in the processes leading to viral particle production in MuV infection. Additionally, we determined that MuV matrix protein (M), which functions mainly in viral particle assembly and budding, colocalized and interacted with Treacle. Furthermore, we found that Treacle is distributed throughout the nucleus in MuV-infected cells. Our research shows that the interaction between M and Treacle supports efficient viral growth in the late stage of MuV infection.
Collapse
Affiliation(s)
- Aika Wakata
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hiroshi Katoh
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Fumihiro Kato
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Makoto Takeda
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
11
|
Lytic Reactivation of the Kaposi’s Sarcoma-Associated Herpesvirus (KSHV) Is Accompanied by Major Nucleolar Alterations. Viruses 2022; 14:v14081720. [PMID: 36016343 PMCID: PMC9412354 DOI: 10.3390/v14081720] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
The nucleolus is a subnuclear compartment whose primary function is the biogenesis of ribosomal subunits. Certain viral infections affect the morphology and composition of the nucleolar compartment and influence ribosomal RNA (rRNA) transcription and maturation. However, no description of nucleolar morphology and function during infection with Kaposi’s sarcoma-associated herpesvirus (KSHV) is available to date. Using immunofluorescence microscopy, we documented extensive destruction of the nuclear and nucleolar architecture during the lytic reactivation of KSHV. This was manifested by the redistribution of key nucleolar proteins, including the rRNA transcription factor UBF. Distinct delocalization patterns were evident; certain nucleolar proteins remained together whereas others dissociated, implying that nucleolar proteins undergo nonrandom programmed dispersion. Significantly, the redistribution of UBF was dependent on viral DNA replication or late viral gene expression. No significant changes in pre-rRNA levels and no accumulation of pre-rRNA intermediates were found by RT-qPCR and Northern blot analysis. Furthermore, fluorescent in situ hybridization (FISH), combined with immunofluorescence, revealed an overlap between Fibrillarin and internal transcribed spacer 1 (ITS1), which represents the primary product of the pre-rRNA, suggesting that the processing of rRNA proceeds during lytic reactivation. Finally, small changes in the levels of pseudouridylation (Ψ) and 2′-O-methylation (Nm) were documented across the rRNA; however, none were localized to the functional domain. Taken together, our results suggest that despite dramatic changes in the nucleolar organization, rRNA transcription and processing persist during lytic reactivation of KSHV. Whether the observed nucleolar alterations favor productive infection or signify cellular anti-viral responses remains to be determined.
Collapse
|
12
|
Zhou J, Dai Y, Lin C, Zhang Y, Feng Z, Dong W, Jin Y, Yan Y, Zhou J, Gu J. Nucleolar protein NPM1 is essential for circovirus replication by binding to viral capsid. Virulence 2021; 11:1379-1393. [PMID: 33073687 PMCID: PMC7575006 DOI: 10.1080/21505594.2020.1832366] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Entry of circovirus into the host cell nucleus is essential for viral replication during the early stage of infection. However, the mechanisms by which nucleolar shuttle proteins are used during viral replication is still not well understood. Here, we report a previously unidentified nucleolar localization signal in circovirus capsid protein (Cap), and that circovirus hijacks the nucleolar phosphoprotein nucleophosmin-1 (NPM1) to facilitate its replication. Colocalization analysis showed that NPM1 translocates from the nucleolus to the nucleoplasm and cytoplasm during viral infection. Coimmunoprecipitation and glutathione S-transferase pull-down assays showed that Cap interacts directly with NPM1. Binding domain mapping showed that the arginine-rich N-terminal motif 1MTYPRRRYRRRRHRPRSHLG20 of Cap, and residue serine-48 of the N-terminal oligomerization domain of NPM1, are essential for the interaction. Virus rescue experiments showed that all arginine to alanine substitution in the N-terminal arginine-rich motif of Cap resulted in diminished viral replication. Knockdown of NPM1 and substitution of serine-48 in NPM1 to glutamic acid also decreased viral replication. In addition, binding assays showed that the arginine-rich motif of Cap is a nucleolar localization signal. Taken together, our findings demonstrate that circovirus protein Cap is a nucleolus-located, and regulates viral replication by directly binding to NPM1.
Collapse
Affiliation(s)
- Jianwei Zhou
- MOA Key Laboratory of Animal Virology, Center of Veterinary Sciences, Zhejiang University , Hangzhou, Zhejiang, PR China
| | - Yadong Dai
- MOA Key Laboratory of Animal Virology, Center of Veterinary Sciences, Zhejiang University , Hangzhou, Zhejiang, PR China
| | - Cui Lin
- MOA Key Laboratory of Animal Virology, Center of Veterinary Sciences, Zhejiang University , Hangzhou, Zhejiang, PR China
| | - Ying Zhang
- MOA Key Laboratory of Animal Virology, Center of Veterinary Sciences, Zhejiang University , Hangzhou, Zhejiang, PR China
| | - Zixuan Feng
- MOA Key Laboratory of Animal Virology, Center of Veterinary Sciences, Zhejiang University , Hangzhou, Zhejiang, PR China
| | - Weiren Dong
- MOA Key Laboratory of Animal Virology, Center of Veterinary Sciences, Zhejiang University , Hangzhou, Zhejiang, PR China
| | - Yulan Jin
- MOA Key Laboratory of Animal Virology, Center of Veterinary Sciences, Zhejiang University , Hangzhou, Zhejiang, PR China
| | - Yan Yan
- MOA Key Laboratory of Animal Virology, Center of Veterinary Sciences, Zhejiang University , Hangzhou, Zhejiang, PR China
| | - Jiyong Zhou
- MOA Key Laboratory of Animal Virology, Center of Veterinary Sciences, Zhejiang University , Hangzhou, Zhejiang, PR China.,Collaborative innovation center and State Key laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University , Hangzhou, China
| | - Jinyan Gu
- MOA Key Laboratory of Animal Virology, Center of Veterinary Sciences, Zhejiang University , Hangzhou, Zhejiang, PR China
| |
Collapse
|
13
|
Iarovaia OV, Ioudinkova ES, Velichko AK, Razin SV. Manipulation of Cellular Processes via Nucleolus Hijaking in the Course of Viral Infection in Mammals. Cells 2021; 10:cells10071597. [PMID: 34202380 PMCID: PMC8303250 DOI: 10.3390/cells10071597] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/16/2022] Open
Abstract
Due to their exceptional simplicity of organization, viruses rely on the resources, molecular mechanisms, macromolecular complexes, regulatory pathways, and functional compartments of the host cell for an effective infection process. The nucleolus plays an important role in the process of interaction between the virus and the infected cell. The interactions of viral proteins and nucleic acids with the nucleolus during the infection process are universal phenomena and have been described for almost all taxonomic groups. During infection, proteins of the nucleolus in association with viral components can be directly used for the processes of replication and transcription of viral nucleic acids and the assembly and transport of viral particles. In the course of a viral infection, the usurpation of the nucleolus functions occurs and the usurpation is accompanied by profound changes in ribosome biogenesis. Recent studies have demonstrated that the nucleolus is a multifunctional and dynamic compartment. In addition to the biogenesis of ribosomes, it is involved in regulating the cell cycle and apoptosis, responding to cellular stress, repairing DNA, and transcribing RNA polymerase II-dependent genes. A viral infection can be accompanied by targeted transport of viral proteins to the nucleolus, massive release of resident proteins of the nucleolus into the nucleoplasm and cytoplasm, the movement of non-nucleolar proteins into the nucleolar compartment, and the temporary localization of viral nucleic acids in the nucleolus. The interaction of viral and nucleolar proteins interferes with canonical and non-canonical functions of the nucleolus and results in a change in the physiology of the host cell: cell cycle arrest, intensification or arrest of ribosome biogenesis, induction or inhibition of apoptosis, and the modification of signaling cascades involved in the stress response. The nucleolus is, therefore, an important target during viral infection. In this review, we discuss the functional impact of viral proteins and nucleic acid interaction with the nucleolus during infection.
Collapse
|
14
|
Hidaka C, Soda K, Nomura F, Kashiwabara Y, Ito H, Ito T. The chicken-derived velogenic Newcastle disease virus can acquire high pathogenicity in domestic ducks via serial passaging. Avian Pathol 2021; 50:1-12. [PMID: 33576245 DOI: 10.1080/03079457.2021.1889461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/08/2021] [Indexed: 10/22/2022]
Abstract
Velogenic Newcastle disease virus (NDV) strains, which show high mortality in chickens, generally do not cause severe disease in waterfowl such as ducks. To elucidate the difference in the pathogenic mechanisms of NDV between chickens and ducks, a chicken-derived velogenic strain (9a5b) was passaged in domestic ducks five times in their air sacs, followed by 20 times in their brains. Eventually, 9a5b acquired higher intracerebral and intranasal pathogenicity in ducks. The intracerebral pathogenicity index (ICPI) value increased from 1.10 to 1.88. All one-week-old ducks intranasally inoculated with the passaged virus (d5a20b) died by 5 days post-inoculation, whereas 70% of the ducks inoculated with parental 9a5b survived for 8 days. The d5a20b strain replicated in broader systemic tissues in ducks compared with the 9a5b strain. The velogenic profile of 9a5b in chickens was maintained after passaging in ducks. The d5a20b suppressed IFN-β gene expression in duck embryo fibroblasts and replicated more rapidly than 9a5b. A total of 11 amino acid substitutions were found in the P, V, M, F, HN, and L proteins of d5a20b. These results suggest that chicken-derived velogenic NDVs have the potential to become virulent in both chickens and ducks during circulation in domesticated waterfowl populations. RESEARCH HIGHLIGHTSChicken-derived NDV acquired high pathogenicity in ducks with serial passaging.The passaged NDV showed intracerebral and intranasal pathogenicity in ducks.The passaged NDV efficiently replicated in systemic tissues in ducks.Of 11 amino acid substitutions some or all are likely involved in pathogenicity.
Collapse
Affiliation(s)
- Chiharu Hidaka
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
- The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Kosuke Soda
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
- The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
- Avian Zoonosis Research Center, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Fumie Nomura
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Yukie Kashiwabara
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Hiroshi Ito
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
- The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
- Avian Zoonosis Research Center, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Toshihiro Ito
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
- The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
- Avian Zoonosis Research Center, Faculty of Agriculture, Tottori University, Tottori, Japan
| |
Collapse
|
15
|
Hennessy EJ, FitzGerald GA. Battle for supremacy: nucleic acid interactions between viruses and cells. J Clin Invest 2021; 131:144227. [PMID: 33290272 PMCID: PMC7843224 DOI: 10.1172/jci144227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Since the COVID-19 pandemic swept across the globe, researchers have been trying to understand its origin, life cycle, and pathogenesis. There is a striking variability in the phenotypic response to infection with SARS-CoV-2 that may reflect differences in host genetics and/or immune response. It is known that the human epigenome is influenced by ethnicity, age, lifestyle, and environmental factors, including previous viral infections. This Review examines the influence of viruses on the host epigenome. We describe general lessons and methodologies that can be used to understand how the virus evades the host immune response. We consider how variation in the epigenome may contribute to heterogeneity in the response to SARS-CoV-2 and may identify a precision medicine approach to treatment.
Collapse
|
16
|
Ujike M, Kawachi Y, Matsunaga Y, Etho Y, Asanuma H, Kamitani W, Taguchi F. Characterization of Localization and Export Signals of Bovine Torovirus Nucleocapsid Protein Responsible for Extensive Nuclear and Nucleolar Accumulation and Their Importance for Virus Growth. J Virol 2021; 95:e02111-20. [PMID: 33177195 PMCID: PMC7925113 DOI: 10.1128/jvi.02111-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 11/20/2022] Open
Abstract
Torovirus (ToV) has recently been classified into the new family Tobaniviridae, although historically, it belonged to the Coronavirus (CoV) family. The nucleocapsid (N) proteins of CoVs are predominantly localized in the cytoplasm, where the viruses replicate, but in some cases the proteins are partially located in the nucleolus. Many studies have investigated the subcellular localization and nucleocytoplasmic trafficking signals of the CoV N proteins, but little is known about ToV N proteins. Here, we studied the subcellular localization of the bovine ToV (BToV) N protein (BToN) and characterized its nucleocytoplasmic trafficking signals. Unlike other CoVs, BToN in infected cells was transported mainly to the nucleolus during early infection but was distributed predominantly in the nucleoplasm rather than in the nucleolus during late infection. Interestingly, a small quantity of BToN was detected in the cytoplasm during infection. Examination of a comprehensive set of substitution or deletion mutants of BToN fused with enhanced green fluorescent protein (EGFP) revealed that clusters of arginine (R) residues comprise nuclear/nucleolar localization signals (NLS/NoLS), and the C-terminal region served as a chromosomal maintenance 1 (CRM1)-independent nuclear export signal (NES). Moreover, recombinant viruses with mutations in the NLS/NoLS, but retaining nuclear accumulation, were successfully rescued and showed slightly reduced growth ability, while the virus that lost the NLS/NoLS-mediated nuclear accumulation of BToN was not rescued. These results indicate that BToN uniquely accumulates mainly in nuclear compartments during infection, regulated by an R-rich NLS/NoLS and a CRM1-independent NES, and that the BToN accumulation in the nuclear compartment driven by NLS/NoLS is important for virus growth.IMPORTANCE ToVs are diarrhea-causing pathogens detected in many species, including humans. BToV has spread worldwide, leading to economic loss, and there is currently no treatment or vaccine available. Positive-stranded RNA viruses, including ToVs, replicate in the cytoplasm, and their structural proteins generally accumulate in the cytoplasm. Interestingly, BToN accumulated predominantly in the nucleus/nucleolus during all infectious processes, with only a small fraction accumulating in the cytoplasm despite being a major structural protein. Furthermore, we identified unique nucleocytoplasmic trafficking signals and demonstrated the importance of NLS/NoLS for virus growth. This study is the first to undertake an in-depth investigation of the subcellular localization and intracellular trafficking signals of BToN. Our findings additionally suggest that the NLS/NoLS-mediated nuclear accumulation of BToN is important for virus replication. An understanding of the unique features of BToV may provide novel insights into the assembly mechanisms of not only ToVs but also other positive-stranded RNA viruses.
Collapse
Affiliation(s)
- Makoto Ujike
- Laboratory of Veterinary Infectious Diseases, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Tokyo, Japan
- Research Center for Animal Life Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Yukako Kawachi
- Laboratory of Veterinary Infectious Diseases, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Yui Matsunaga
- Laboratory of Veterinary Infectious Diseases, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Yuka Etho
- Laboratory of Veterinary Infectious Diseases, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Hideki Asanuma
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Wataru Kamitani
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Fumihiro Taguchi
- Laboratory of Veterinary Infectious Diseases, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Tokyo, Japan
| |
Collapse
|
17
|
Kaliatsi EG, Giarimoglou N, Stathopoulos C, Stamatopoulou V. Non-Coding RNA-Driven Regulation of rRNA Biogenesis. Int J Mol Sci 2020; 21:E9738. [PMID: 33419375 PMCID: PMC7766524 DOI: 10.3390/ijms21249738] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/30/2022] Open
Abstract
Ribosomal RNA (rRNA) biogenesis takes place in the nucleolus, the most prominent condensate of the eukaryotic nucleus. The proper assembly and integrity of the nucleolus reflects the accurate synthesis and processing of rRNAs which in turn, as major components of ribosomes, ensure the uninterrupted flow of the genetic information during translation. Therefore, the abundant production of rRNAs in a precisely functional nucleolus is of outmost importance for the cell viability and requires the concerted action of essential enzymes, associated factors and epigenetic marks. The coordination and regulation of such an elaborate process depends on not only protein factors, but also on numerous regulatory non-coding RNAs (ncRNAs). Herein, we focus on RNA-mediated mechanisms that control the synthesis, processing and modification of rRNAs in mammals. We highlight the significance of regulatory ncRNAs in rRNA biogenesis and the maintenance of the nucleolar morphology, as well as their role in human diseases and as novel druggable molecular targets.
Collapse
Affiliation(s)
| | | | - Constantinos Stathopoulos
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece; (E.G.K.); (N.G.)
| | - Vassiliki Stamatopoulou
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece; (E.G.K.); (N.G.)
| |
Collapse
|
18
|
Razin SV, Gavrilov AA, Iarovaia OV. Modification of Nuclear Compartments and the 3D Genome in the Course of a Viral Infection. Acta Naturae 2020; 12:34-46. [PMID: 33456976 PMCID: PMC7800604 DOI: 10.32607/actanaturae.11041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
The review addresses the question of how the structural and functional compartmentalization of the cell nucleus and the 3D organization of the cellular genome are modified during the infection of cells with various viruses. Particular attention is paid to the role of the introduced changes in the implementation of the viral strategy to evade the antiviral defense systems and provide conditions for viral replication. The discussion focuses on viruses replicating in the cell nucleus. Cytoplasmic viruses are mentioned in cases when a significant reorganization of the nuclear compartments or the 3D genome structure occurs during an infection with these viruses.
Collapse
Affiliation(s)
- S. V. Razin
- Institute of Gene Biology Russian Academy of Sciences
| | | | | |
Collapse
|
19
|
Labrecque M, Marchand C, Archambault D. Characterization of Signal Sequences Determining the Nuclear/Nucleolar Import and Nuclear Export of the Caprine Arthritis-Encephalitis Virus Rev Protein. Viruses 2020; 12:v12080900. [PMID: 32824614 PMCID: PMC7471974 DOI: 10.3390/v12080900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/08/2020] [Accepted: 08/12/2020] [Indexed: 11/16/2022] Open
Abstract
Caprine arthritis-encephalitis virus (CAEV), a lentivirus, relies on the action of the Rev protein for its replication. The CAEV Rev fulfills its function by allowing the nuclear exportation of partially spliced or unspliced viral mRNAs. In this study, we characterized the nuclear and nucleolar localization signals (NLS and NoLS, respectively) and the nuclear export signal (NES) of the CAEV Rev protein. These signals are key actors in the nucleocytoplasmic shuttling of a lentiviral Rev protein. Several deletion and alanine substitution mutants were generated from a plasmid encoding the CAEV Rev wild-type protein that was fused to the enhanced green fluorescent protein (EGFP). Following cell transfection, images were captured by confocal microscopy and the fluorescence was quantified in the different cell compartments. The results showed that the NLS region is localized between amino acids (aa) 59 to 75, has a monopartite-like structure and is exclusively composed of arginine residues. The NoLS was found to be partially associated with the NLS. Finally, the CAEV Rev protein’s NES mapped between aa 89 to 101, with an aa spacing between the hydrophobic residues that was found to be unconventional as compared to that of other retroviral Rev/Rev-like proteins.
Collapse
Affiliation(s)
- Marlène Labrecque
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada
| | - Claude Marchand
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Denis Archambault
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, Montréal, QC H3C 3J7, Canada
| |
Collapse
|
20
|
Expression of Rta in B Lymphocytes during Epstein-Barr Virus Latency. J Mol Biol 2020; 432:5227-5243. [PMID: 32710985 DOI: 10.1016/j.jmb.2020.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/29/2020] [Accepted: 07/20/2020] [Indexed: 11/21/2022]
Abstract
Rta of Epstein-Barr virus (EBV) is thought to be expressed only during the lytic cycle to promote the transcription of lytic genes. However, we found that Rta is expressed in EBV-infected B cells during viral latency, at levels detectable by immunoblot analysis. Latent Rta expression cannot be attributed to spontaneous lytic activation, as we observed that more than 90% of Akata, P3HR1, and 721 cells latently infected by EBV express Rta. We further found that Rta is sequestered in the nucleolus during EBV latency through its interaction with MCRS2, a nucleolar protein. When Rta is sequestered in the nucleolus, it no longer activates RNA polymerase II-driven transcription, thus explaining why Rta expression during latency does not transactivate EBV lytic genes. Additional experiments showed that Rta can bind to 18S rRNA and become incorporated into ribosomes, and a transient transfection experiment showed that Rta promotes translation from an mRNA reporter. These findings reveal that Rta has novel functions beyond transcriptional activation during EBV latency and may have interesting implications for the concept of EBV latency.
Collapse
|
21
|
Wu K, Zou J, Chang HY. RNA-GPS Predicts SARS-CoV-2 RNA Localization to Host Mitochondria and Nucleolus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.04.28.065201. [PMID: 32511373 PMCID: PMC7263502 DOI: 10.1101/2020.04.28.065201] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The SARS-CoV-2 coronavirus is driving a global pandemic, but its biological mechanisms are less well understood. SARS-CoV-2 is an RNA virus whose multiple genomic and subgenomic RNA (sgRNA) transcripts hijack the host cell's machinery, located across distinct cytotopic locations. Subcellular localization of its viral RNA could play important roles in viral replication and host antiviral immune response. Here we perform computational modeling of SARS-CoV-2 viral RNA localization across eight subcellular neighborhoods. We compare hundreds of SARS-CoV-2 genomes to the human transcriptome and other coronaviruses and perform systematic sub-sequence analyses to identify the responsible signals. Using state-of-the-art machine learning models, we predict that the SARS-CoV-2 RNA genome and all sgRNAs are enriched in the host mitochondrial matrix and nucleolus. The 5' and 3' viral untranslated regions possess the strongest and most distinct localization signals. We discuss the mitochondrial localization signal in relation to the formation of double-membrane vesicles, a critical stage in the coronavirus life cycle. Our computational analysis serves as a hypothesis generation tool to suggest models for SARS-CoV-2 biology and inform experimental efforts to combat the virus.
Collapse
Affiliation(s)
- Kevin Wu
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305, USA
- Center for Personal and Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - James Zou
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Howard Y Chang
- Center for Personal and Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
22
|
DEAD-Box Helicases: Sensors, Regulators, and Effectors for Antiviral Defense. Viruses 2020; 12:v12020181. [PMID: 32033386 PMCID: PMC7077277 DOI: 10.3390/v12020181] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 12/13/2022] Open
Abstract
DEAD-box helicases are a large family of conserved RNA-binding proteins that belong to the broader group of cellular DExD/H helicases. Members of the DEAD-box helicase family have roles throughout cellular RNA metabolism from biogenesis to decay. Moreover, there is emerging evidence that cellular RNA helicases, including DEAD-box helicases, play roles in the recognition of foreign nucleic acids and the modulation of viral infection. As intracellular parasites, viruses must evade detection by innate immune sensing mechanisms and degradation by cellular machinery while also manipulating host cell processes to facilitate replication. The ability of DEAD-box helicases to recognize RNA in a sequence-independent manner, as well as the breadth of cellular functions carried out by members of this family, lead them to influence innate recognition and viral infections in multiple ways. Indeed, DEAD-box helicases have been shown to contribute to intracellular immune sensing, act as antiviral effectors, and even to be coopted by viruses to promote their replication. However, our understanding of the mechanisms underlying these interactions, as well as the cellular roles of DEAD-box helicases themselves, is limited in many cases. We will discuss the diverse roles that members of the DEAD-box helicase family play during viral infections.
Collapse
|
23
|
Tokunaga M, Miyamoto Y, Suzuki T, Otani M, Inuki S, Esaki T, Nagao C, Mizuguchi K, Ohno H, Yoneda Y, Okamoto T, Oka M, Matsuura Y. Novel anti-flavivirus drugs targeting the nucleolar distribution of core protein. Virology 2019; 541:41-51. [PMID: 31826845 DOI: 10.1016/j.virol.2019.11.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 01/04/2023]
Abstract
The risk of infectious diseases caused by Flavivirus is increasing globally. Here, we developed a novel high-throughput screening (HTS) system to evaluate the inhibitory effects of compounds targeting the nuclear localization of the flavivirus core protein. We screened 4000 compounds based on their ability to inhibit the nuclear localization of the core protein, and identified over 20 compounds including inhibitors for cyclin dependent kinase and glycogen synthase kinase. The efficacy of the identified compounds to suppress viral growth was validated in a cell-based infection system. Remarkably, the nucleolus morphology was affected by the treatment with the compounds, suggesting that the nucleolus function is critical for viral propagation. The present HTS system provides a useful strategy for the identification of antivirals against flavivirus by targeting the nucleolar localization of the core protein.
Collapse
Affiliation(s)
- Makoto Tokunaga
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoichi Miyamoto
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Tatsuya Suzuki
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Mayumi Otani
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Esaki
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institute of Biomedical Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Center for Data Science Education and Research, Shiga University, Shiga, Japan
| | - Chioko Nagao
- Laboratory of In-silico Drug Design, Center for Drug Design Research, National Institute of Biomedical Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institute of Biomedical Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Laboratory of In-silico Drug Design, Center for Drug Design Research, National Institute of Biomedical Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Institute for Protein Research, Osaka University, Osaka, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Yoshihiro Yoneda
- National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Toru Okamoto
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
| | - Masahiro Oka
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.
| | - Yoshiharu Matsuura
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
24
|
Acar DD, Stroobants VJE, Favoreel H, Saelens X, Nauwynck HJ. Identification of peptide domains involved in the subcellular localization of the feline coronavirus 3b protein. J Gen Virol 2019; 100:1417-1430. [PMID: 31483243 PMCID: PMC7079696 DOI: 10.1099/jgv.0.001321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Feline coronavirus (FCoV) has been identified as the aetiological agent of feline infectious peritonitis (FIP), a highly fatal systemic disease in cats. FCoV open reading frame 3 (ORF3) encodes accessory proteins 3a, 3b and 3 c. The FCoV 3b accessory protein consists of 72 amino acid residues and localizes to nucleoli and mitochondria. The present work focused on peptide domains within FCoV 3b that drive its intracellular trafficking. Transfection of different cell types with FCoV 3b fused to enhanced green fluorescent protein (EGFP) or 3×FLAG confirmed localization of FCoV 3b in the mitochondria and nucleoli. Using serial truncated mutants, we showed that nucleolar accumulation is controlled by a joint nucleolar and nuclear localization signal (NoLS/NLS) in which the identified overlapping pat4 motifs (residues 53–57) play a critical role. Mutational analysis also revealed that mitochondrial translocation is mediated by N-terminal residues 10–35, in which a Tom20 recognition motif (residues 13–17) and two other overlapping hexamers (residues 24–30) associated with mitochondrial targeting were identified. In addition, a second Tom20 recognition motif was identified further downstream (residues 61–65), although the mitochondrial translocation evoked by these residues seemed less efficient as a diffuse cytoplasmic distribution was also observed. Assessing the spatiotemporal distribution of FCoV 3b did not provide convincing evidence of dynamic shuttling behaviour between the nucleoli and the mitochondria.
Collapse
Affiliation(s)
- Delphine D. Acar
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Veerle J. E. Stroobants
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Herman Favoreel
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Hans J. Nauwynck
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- *Correspondence: Hans J. Nauwynck,
| |
Collapse
|
25
|
The Jembrana disease virus Rev protein: Identification of nuclear and novel lentiviral nucleolar localization and nuclear export signals. PLoS One 2019; 14:e0221505. [PMID: 31437223 PMCID: PMC6706053 DOI: 10.1371/journal.pone.0221505] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/08/2019] [Indexed: 11/19/2022] Open
Abstract
The lentiviral Rev protein, which is a regulatory protein essential for virus replication, has been first studied in the human immunodeficiency virus type 1 (HIV-1). The main function of Rev is to mediate the nuclear exportation of viral RNAs. To fulfill its function, Rev shuttles between the cytoplasm and the nucleus. The Jembrana disease virus (JDV), a lentivirus, is the etiologic agent of the Jembrana disease which was first described in Bali cattle in Indonesia in 1964. Despite the high mortality rate associated with JDV, this virus remains poorly studied. Herein the subcellular distribution of JDV Rev, the nuclear and nucleolar localization signals (NLS and NoLS, respectively) and the nuclear export signal (NES) of the protein were examined. JDV Rev fused to the enhanced green fluorescent protein (EGFP) predominantly localized to the cytoplasm and nucleolus of transfected cells, as determined by fluorescence microscopy analyses. Through transfection of a series of deletion mutants of JDV Rev, it was possible to localize the NLS/NoLS region between amino acids (aa) 74 to 105. By substituting basic residues with alanine within this sequence, we demonstrated that the JDV Rev NLS encompasses aa 76 to 86, and is exclusively composed of arginine residues, whereas a bipartite NoLS was observed for the first time in any retroviral Rev/Rev-like proteins. Finally, a NES was identified downstream of the NLS/NoLS and encompasses aa 116 to 128 of the JDV Rev protein. The JDV Rev NES was found to be of the protein kinase A inhibitor (PKI) class instead of the HIV-1 Rev class. It also corresponds to the most optimal consensus sequence of PKI NES and, as such, is novel among lentiviral Rev NES.
Collapse
|
26
|
Matos-Perdomo E, Machín F. Nucleolar and Ribosomal DNA Structure under Stress: Yeast Lessons for Aging and Cancer. Cells 2019; 8:cells8080779. [PMID: 31357498 PMCID: PMC6721496 DOI: 10.3390/cells8080779] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/15/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023] Open
Abstract
Once thought a mere ribosome factory, the nucleolus has been viewed in recent years as an extremely sensitive gauge of diverse cellular stresses. Emerging concepts in nucleolar biology include the nucleolar stress response (NSR), whereby a series of cell insults have a special impact on the nucleolus. These insults include, among others, ultra-violet radiation (UV), nutrient deprivation, hypoxia and thermal stress. While these stresses might influence nucleolar biology directly or indirectly, other perturbances whose origin resides in the nucleolar biology also trigger nucleolar and systemic stress responses. Among the latter, we find mutations in nucleolar and ribosomal proteins, ribosomal RNA (rRNA) processing inhibitors and ribosomal DNA (rDNA) transcription inhibition. The p53 protein also mediates NSR, leading ultimately to cell cycle arrest, apoptosis, senescence or differentiation. Hence, NSR is gaining importance in cancer biology. The nucleolar size and ribosome biogenesis, and how they connect with the Target of Rapamycin (TOR) signalling pathway, are also becoming important in the biology of aging and cancer. Simple model organisms like the budding yeast Saccharomyces cerevisiae, easy to manipulate genetically, are useful in order to study nucleolar and rDNA structure and their relationship with stress. In this review, we summarize the most important findings related to this topic.
Collapse
Affiliation(s)
- Emiliano Matos-Perdomo
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
- Escuela de Doctorado y Estudios de Postgrado, Universidad de La Laguna, 38200 Tenerife, Spain
| | - Félix Machín
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain.
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, 38200 Tenerife, Spain.
- Facultad de Ciencias de la Salud, Universidad Fernando Pessoa Canarias, 35450 Santa María de Guía, Gran Canaria, Spain.
| |
Collapse
|
27
|
Duan Z, Deng S, Ji X, Zhao J, Yuan C, Gao H. Nuclear localization of Newcastle disease virus matrix protein promotes virus replication by affecting viral RNA synthesis and transcription and inhibiting host cell transcription. Vet Res 2019; 50:22. [PMID: 30894203 PMCID: PMC6425612 DOI: 10.1186/s13567-019-0640-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/11/2019] [Indexed: 01/24/2023] Open
Abstract
Nuclear localization of paramyxovirus proteins is crucial for virus life cycle, including the regulation of viral replication and the evasion of host immunity. We previously showed that a recombinant Newcastle disease virus (NDV) with nuclear localization signal mutation in the matrix (M) protein results in a pathotype change and attenuates viral pathogenicity in chickens. However, little is known about the nuclear localization functions of NDV M protein. In this study, the potential functions of the M protein in the nucleus were investigated. We first demonstrate that nuclear localization of the M protein could not only promote the cytopathogenicity of NDV but also increase viral RNA synthesis and transcription efficiency in DF-1 cells. Using microarray analysis, we found that nuclear localization of the M protein might inhibit host cell transcription, represented by numerous up-regulating genes associated with transcriptional repressor activity and down-regulating genes associated with transcriptional activator activity. The role of representative up-regulated gene prospero homeobox 1 (PROX1) and down-regulated gene aryl hydrocarbon receptor (AHR) in the replication of NDV was then evaluated. The results show that siRNA-mediated knockdown of PROX1 or AHR significantly reduced or increased the viral RNA synthesis and viral replication, respectively, demonstrating the important roles of the expression changes of these genes in NDV replication. Together, our findings demonstrate for the first time that nuclear localization of NDV M protein promotes virus replication by affecting viral RNA synthesis and transcription and inhibiting host cell transcription, improving our understanding of the molecular mechanism of NDV replication and pathogenesis.
Collapse
Affiliation(s)
- Zhiqiang Duan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China. .,College of Animal Science, Guizhou University, Guiyang, China.
| | - Shanshan Deng
- College of Animal Science, Guizhou University, Guiyang, China
| | - Xinqin Ji
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China.,College of Animal Science, Guizhou University, Guiyang, China
| | - Jiafu Zhao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China.,College of Animal Science, Guizhou University, Guiyang, China
| | - Chao Yuan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China.,College of Animal Science, Guizhou University, Guiyang, China
| | - Hongbo Gao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China.,College of Animal Science, Guizhou University, Guiyang, China
| |
Collapse
|
28
|
Stamm S, Lodmell JS. C/D box snoRNAs in viral infections: RNA viruses use old dogs for new tricks. Noncoding RNA Res 2019; 4:46-53. [PMID: 31193534 PMCID: PMC6533054 DOI: 10.1016/j.ncrna.2019.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/27/2019] [Accepted: 02/13/2019] [Indexed: 12/17/2022] Open
Abstract
C/D box snoRNAs (SNORDs) are a highly expressed class of non-coding RNAs. Besides their well-established role in rRNA modification, C/D box snoRNAs form protein complexes devoid of fibrillarin and regulate pre-mRNA splicing and polyadenylation of numerous genes. There is an emerging body of evidence for functional interactions between RNA viruses and C/D box snoRNAs. The infectivity of some RNA viruses depends on enzymatically active fibrillarin, and many RNA viral proteins associate with nucleolin or nucleophosmin, suggesting that viruses benefit from their cytosolic accumulation. These interactions are likely reflected by morphological changes in the nucleolus, often leading to relocalization of nucleolar proteins and ncRNAs to the cytosol that are a characteristic feature of viral infections. Knock-down studies have also shown that RNA viruses need specific C/D box snoRNAs for optimal replication, suggesting that RNA viruses benefit from gene expression programs regulated by SNORDs, or that viruses have evolved “new” uses for these humble ncRNAs to advance their prospects during infection.
Collapse
Affiliation(s)
- Stefan Stamm
- University of Kentucky, Molecular and Cellular Biochemistry, 741 South Limestone, Lexington, KY 40536, USA
| | - J Stephen Lodmell
- Division of Biological Sciences and Center for Biomolecular Structure and Dynamics, The University of Montana, Missoula, MT, USA
| |
Collapse
|
29
|
Wang W, Cheng X, Buske PJ, Suzich JA, Jin H. Attenuate Newcastle disease virus by codon modification of the glycoproteins and phosphoprotein genes. Virology 2019; 528:144-151. [PMID: 30616204 DOI: 10.1016/j.virol.2018.12.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/10/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022]
Abstract
A codon modification strategy was used to attenuate the avian pathogenicity of an oncolytic mesogenic Newcastle disease virus (NDV) by targeting the three major virulence factors: the fusion (F) protein, hemagglutinin neuraminidase (HN) and phosphoprotein (P). Recoding the F and HN genes with rare codons greatly reduced expression of both F and HN proteins and resulted in their low incorporation into virions. The F and HN recoded virus was partially attenuated in chickens even when the F protein cleavage site was modified. Full attenuation was achieved when the 5' portion of the P gene was recoded. The recoded P, F and HN triple gene mutant exhibited delayed cell death in human cancer cells with prolonged expression of a GFP transgene. While this engineered attenuated NDV strain has lower oncolytic potency, its capacity for prolonged transgene expression may allow its use as a vaccine or gene delivery vector.
Collapse
Affiliation(s)
- Weijia Wang
- MedImmune LLC, 121 Oyster Point Boulevard, South San Francisco, CA, USA
| | - Xing Cheng
- MedImmune LLC, 121 Oyster Point Boulevard, South San Francisco, CA, USA
| | - Paul J Buske
- MedImmune LLC, 121 Oyster Point Boulevard, South San Francisco, CA, USA
| | | | - Hong Jin
- MedImmune LLC, 121 Oyster Point Boulevard, South San Francisco, CA, USA.
| |
Collapse
|
30
|
Lobaina Y, Perera Y. Implication of B23/NPM1 in Viral Infections, Potential Uses of B23/NPM1 Inhibitors as Antiviral Therapy. Infect Disord Drug Targets 2019; 19:2-16. [PMID: 29589547 DOI: 10.2174/1871526518666180327124412] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/08/2018] [Accepted: 02/12/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND B23/nucleophosmin (B23/NPM1) is an abundant multifunctional protein mainly located in the nucleolus but constantly shuttling between the nucleus and cytosol. As a consequence of its constitutive expression, intracellular dynamics and binding capacities, B23/NPM1 interacts with multiple cellular factors in different cellular compartments, but also with viral proteins from both DNA and RNA viruses. B23/NPM1 influences overall viral replication of viruses like HIV, HBV, HCV, HDV and HPV by playing functional roles in different stages of viral replication including nuclear import, viral genome transcription and assembly, as well as final particle formation. Of note, some virus modify the subcellular localization, stability and/or increases B23/NPM1 expression levels on target cells, probably to foster B23/NPM1 functions in their own replicative cycle. RESULTS This review summarizes current knowledge concerning the interaction of B23/NPM1 with several viral proteins during relevant human infections. The opportunities and challenges of targeting this well-conserved host protein as a potentially new broad antiviral treatment are discussed in detail. Importantly, although initially conceived to treat cancer, a handful of B23/NPM1 inhibitors are currently available to test on viral infection models. CONCLUSION As B23/NPM1 partakes in key steps of viral replication and some viral infections remain as unsolved medical needs, an appealing idea may be the expedite evaluation of B23/NPM1 inhibitors in viral infections. Furthermore, worth to be addressed is if the up-regulation of B23/NPM1 protein levels that follows persistent viral infections may be instrumental to the malignant transformation induced by virus like HBV and HCV.
Collapse
Affiliation(s)
- Yadira Lobaina
- Therapeutic Hepatitis B Vaccine Group, Vaccine Division, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Havana, CP 10600, Cuba
| | - Yasser Perera
- Molecular Oncology Group, Pharmaceuticals Division, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Havana, CP 10600, Cuba
| |
Collapse
|
31
|
Lemay G. Synthesis and Translation of Viral mRNA in Reovirus-Infected Cells: Progress and Remaining Questions. Viruses 2018; 10:E671. [PMID: 30486370 PMCID: PMC6315682 DOI: 10.3390/v10120671] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 11/23/2018] [Accepted: 11/25/2018] [Indexed: 12/11/2022] Open
Abstract
At the end of my doctoral studies, in 1988, I published a review article on the major steps of transcription and translation during the mammalian reovirus multiplication cycle, a topic that still fascinates me 30 years later. It is in the nature of scientific research to generate further questioning as new knowledge emerges. Our understanding of these fascinating viruses thus remains incomplete but it seemed appropriate at this moment to look back and reflect on our progress and most important questions that still puzzle us. It is also essential of being careful about concepts that seem so well established, but could still be better validated using new approaches. I hope that the few reflections presented here will stimulate discussions and maybe attract new investigators into the field of reovirus research. Many other aspects of the viral multiplication cycle would merit our attention. However, I will essentially limit my discussion to these central aspects of the viral cycle that are transcription of viral genes and their phenotypic expression through the host cell translational machinery. The objective here is not to review every aspect but to put more emphasis on important progress and challenges in the field.
Collapse
Affiliation(s)
- Guy Lemay
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC H3C 3J7, Canada.
| |
Collapse
|
32
|
Abstract
Zika virus (ZIKV) is a significant global health threat, as infection has been linked to serious neurological complications, including microcephaly. Using a human stem cell-derived neural progenitor model system, we find that a critical cellular quality control process called the nonsense-mediated mRNA decay (NMD) pathway is disrupted during ZIKV infection. Importantly, disruption of the NMD pathway is a known cause of microcephaly and other neurological disorders. We further identify an interaction between the capsid protein of ZIKV and up-frameshift protein 1 (UPF1), the master regulator of NMD, and show that ZIKV capsid targets UPF1 for degradation. Together, these results offer a new mechanism for how ZIKV infection can cause neuropathology in the developing brain. Zika virus (ZIKV) infection of neural progenitor cells (NPCs) in utero is associated with neurological disorders, such as microcephaly, but a detailed molecular understanding of ZIKV-induced pathogenesis is lacking. Here we show that in vitro ZIKV infection of human cells, including NPCs, causes disruption of the nonsense-mediated mRNA decay (NMD) pathway. NMD is a cellular mRNA surveillance mechanism that is required for normal brain size in mice. Using affinity purification-mass spectrometry, we identified multiple cellular NMD factors that bind to the viral capsid protein, including the central NMD regulator up-frameshift protein 1 (UPF1). Endogenous UPF1 interacted with the ZIKV capsid protein in coimmunoprecipitation experiments, and capsid expression posttranscriptionally downregulated UPF1 protein levels, a process that we confirmed occurs during ZIKV infection. Cellular fractionation studies show that the ZIKV capsid protein specifically targets nuclear UPF1 for degradation via the proteasome. A further decrease in UPF1 levels by RNAi significantly enhanced ZIKV infection in NPC cultures, consistent with a model in which NMD restricts ZIKV infection in the fetal brain. We propose that ZIKV, via the capsid protein, has evolved a strategy to lower UPF1 levels and dampen antiviral activities of NMD, which in turn contributes to neuropathology in vivo.
Collapse
|
33
|
Duan Z, Xu H, Ji X, Zhao J, Xu H, Hu Y, Deng S, Hu S, Liu X. Importin α5 negatively regulates importin β1-mediated nuclear import of Newcastle disease virus matrix protein and viral replication and pathogenicity in chicken fibroblasts. Virulence 2018. [PMID: 29532715 PMCID: PMC5955436 DOI: 10.1080/21505594.2018.1449507] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The matrix (M) protein of Newcastle disease virus (NDV) is demonstrated to localize in the nucleus via intrinsic nuclear localization signal (NLS), but cellular proteins involved in the nuclear import of NDV M protein and the role of M's nuclear localization in the replication and pathogenicity of NDV remain unclear. In this study, importin β1 was screened to interact with NDV M protein by yeast two-hybrid screening. This interaction was subsequently confirmed by co-immunoprecipitation and pull-down assays. In vitro binding studies indicated that the NLS region of M protein and the amino acids 336–433 of importin β1 that belonged to the RanGTP binding region were important for binding. Importantly, a recombinant virus with M/NLS mutation resulted in a pathotype change of NDV and attenuated viral replication and pathogenicity in chicken fibroblasts and SPF chickens. In agreement with the binding data, nuclear import of NDV M protein in digitonin-permeabilized HeLa cells required both importin β1 and RanGTP. Interestingly, importin α5 was verified to interact with M protein through binding importin β1. However, importin β1 or importin α5 depletion by siRNA resulted in different results, which showed the obviously cytoplasmic or nuclear accumulation of M protein and the remarkably decreased or increased replication ability and pathogenicity of NDV in chicken fibroblasts, respectively. Our findings therefore demonstrate for the first time the nuclear import mechanism of NDV M protein and the negative regulation role of importin α5 in importin β1-mediated nuclear import of M protein and the replication and pathogenicity of a paramyxovirus.
Collapse
Affiliation(s)
- Zhiqiang Duan
- a Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education , Guizhou University , Guiyang , China.,b College of Animal Science , Guizhou University , Guiyang , China
| | - Haixu Xu
- c Key Laboratory of Animal Infectious Diseases of Ministry of Agriculture , Yangzhou University , Yangzhou , China
| | - Xinqin Ji
- a Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education , Guizhou University , Guiyang , China.,b College of Animal Science , Guizhou University , Guiyang , China
| | - Jiafu Zhao
- a Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education , Guizhou University , Guiyang , China.,b College of Animal Science , Guizhou University , Guiyang , China
| | - Houqiang Xu
- a Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education , Guizhou University , Guiyang , China.,b College of Animal Science , Guizhou University , Guiyang , China
| | - Yan Hu
- b College of Animal Science , Guizhou University , Guiyang , China
| | - Shanshan Deng
- b College of Animal Science , Guizhou University , Guiyang , China
| | - Shunlin Hu
- c Key Laboratory of Animal Infectious Diseases of Ministry of Agriculture , Yangzhou University , Yangzhou , China
| | - Xiufan Liu
- c Key Laboratory of Animal Infectious Diseases of Ministry of Agriculture , Yangzhou University , Yangzhou , China
| |
Collapse
|
34
|
Strand-Specific Dual RNA Sequencing of Bronchial Epithelial Cells Infected with Influenza A/H3N2 Viruses Reveals Splicing of Gene Segment 6 and Novel Host-Virus Interactions. J Virol 2018; 92:JVI.00518-18. [PMID: 29976658 DOI: 10.1128/jvi.00518-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/24/2018] [Indexed: 02/08/2023] Open
Abstract
Host-influenza virus interplay at the transcript level has been extensively characterized in epithelial cells. Yet, there are no studies that simultaneously characterize human host and influenza A virus (IAV) genomes. We infected human bronchial epithelial BEAS-2B cells with two seasonal IAV/H3N2 strains, Brisbane/10/07 and Perth/16/09 (reference strains for past vaccine seasons) and the well-characterized laboratory strain Udorn/307/72. Strand-specific RNA sequencing (RNA-seq) of the infected BEAS-2B cells allowed for simultaneous analysis of host and viral transcriptomes, in addition to pathogen genomes, to reveal changes in mRNA expression and alternative splicing (AS). In general, patterns of global and immune gene expression induced by the three IAVs were mostly shared. However, AS of host transcripts and small nuclear RNAs differed between the seasonal and laboratory strains. Analysis of viral transcriptomes showed deletions of the polymerase components (defective interfering-like RNAs) within the genome. Surprisingly, we found that the neuraminidase gene undergoes AS and that the splicing event differs between seasonal and laboratory strains. Our findings reveal novel elements of the host-virus interaction and highlight the importance of RNA-seq in identifying molecular changes at the genome level that may contribute to shaping RNA-based innate immunity.IMPORTANCE The use of massively parallel RNA sequencing (RNA-seq) has revealed insights into human and pathogen genomes and their evolution. Dual RNA-seq allows simultaneous dissection of host and pathogen genomes and strand-specific RNA-seq provides information about the polarity of the RNA. This is important in the case of negative-strand RNA viruses like influenza virus, which generate positive (complementary and mRNA) and negative-strand RNAs (genome) that differ in their potential to trigger innate immunity. Here, we characterize interactions between human bronchial epithelial cells and three influenza A/H3N2 strains using strand-specific dual RNA-seq. We focused on this subtype because of its epidemiological importance in causing significant morbidity and mortality during influenza epidemics. We report novel elements that differ between seasonal and laboratory strains highlighting the complexity of the host-virus interplay at the RNA level.
Collapse
|
35
|
Rawlinson SM, Zhao T, Rozario AM, Rootes CL, McMillan PJ, Purcell AW, Woon A, Marsh GA, Lieu KG, Wang LF, Netter HJ, Bell TDM, Stewart CR, Moseley GW. Viral regulation of host cell biology by hijacking of the nucleolar DNA-damage response. Nat Commun 2018; 9:3057. [PMID: 30076298 PMCID: PMC6076271 DOI: 10.1038/s41467-018-05354-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 06/28/2018] [Indexed: 12/11/2022] Open
Abstract
Recent studies indicate that nucleoli play critical roles in the DNA-damage response (DDR) via interaction of DDR machinery including NBS1 with nucleolar Treacle protein, a key mediator of ribosomal RNA (rRNA) transcription and processing. Here, using proteomics, confocal and single molecule super-resolution imaging, and infection under biosafety level-4 containment, we show that this nucleolar DDR pathway is targeted by infectious pathogens. We find that the matrix proteins of Hendra virus and Nipah virus, highly pathogenic viruses of the Henipavirus genus in the order Mononegavirales, interact with Treacle and inhibit its function, thereby silencing rRNA biogenesis, consistent with mimicking NBS1–Treacle interaction during a DDR. Furthermore, inhibition of Treacle expression/function enhances henipavirus production. These data identify a mechanism for viral modulation of host cells by appropriating the nucleolar DDR and represent, to our knowledge, the first direct intranucleolar function for proteins of any mononegavirus. Many RNA viruses that replicate in the cytoplasm express proteins that localize to nucleoli, but the nucleolar functions remain largely unknown. Here, the authors show that the Henipavirus matrix protein mimics an endogenous Treacle partner of the DNA-damage response, resulting in suppression of rRNA biogenesis.
Collapse
Affiliation(s)
- Stephen M Rawlinson
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.,Department of Biochemistry and Molecular Biology, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Tianyue Zhao
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.,Department of Biochemistry and Molecular Biology, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Ashley M Rozario
- School of Chemistry, Monash University, Clayton, Victoria, 3800, Australia
| | - Christina L Rootes
- CSIRO Health & Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, 3220, Australia
| | - Paul J McMillan
- Department of Biochemistry and Molecular Biology, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, 3010, Australia.,Biological Optical Microscopy Platform, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Anthony W Purcell
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Amanda Woon
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Glenn A Marsh
- CSIRO Health & Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, 3220, Australia
| | - Kim G Lieu
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Hans J Netter
- Victorian Infectious Diseases Reference Laboratory, Melbourne Health, The Peter Doherty Institute, Victoria, 3000, Australia
| | - Toby D M Bell
- School of Chemistry, Monash University, Clayton, Victoria, 3800, Australia
| | - Cameron R Stewart
- CSIRO Health & Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, 3220, Australia
| | - Gregory W Moseley
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia. .,Department of Biochemistry and Molecular Biology, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, 3010, Australia.
| |
Collapse
|
36
|
Tarakhovsky A, Prinjha RK. Drawing on disorder: How viruses use histone mimicry to their advantage. J Exp Med 2018; 215:1777-1787. [PMID: 29934321 PMCID: PMC6028506 DOI: 10.1084/jem.20180099] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/24/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022] Open
Abstract
Humans carry trillions of viruses that thrive because of their ability to exploit the host. In this exploitation, viruses promote their own replication by suppressing the host antiviral response and by inducing changes in host biosynthetic processes, often with extremely small genomes of their own. In the review, we discuss the phenomenon of histone mimicry by viral proteins and how this mimicry allows the virus to dial in to the cell's transcriptional processes and establish a cell state that promotes infection. We suggest that histone mimicry is part of a broader viral strategy to use intrinsic protein disorder as a means to overcome the size limitations of its own genome and to maximize its impact on host protein networks. In particular, we discuss how intrinsic protein disorder may enable viral proteins to interfere with phase-separated host protein condensates, including those that contribute to chromatin-mediated control of gene expression.
Collapse
Affiliation(s)
- Alexander Tarakhovsky
- Laboratory of the Immune Cell Epigenetics and Signaling, The Rockefeller University, New York, NY
| | - Rab K Prinjha
- Epigenetics DPU, Oncology and Immuno-inflammation TA Units, GlaxoSmithKline Medicines Research Centre, Stevenage, England, UK
| |
Collapse
|
37
|
Strange DP, Green R, Siemann DN, Gale M, Verma S. Immunoprofiles of human Sertoli cells infected with Zika virus reveals unique insights into host-pathogen crosstalk. Sci Rep 2018; 8:8702. [PMID: 29880853 PMCID: PMC5992156 DOI: 10.1038/s41598-018-27027-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/16/2018] [Indexed: 02/07/2023] Open
Abstract
Confirmed reports of Zika virus (ZIKV) in seminal fluid months after clearance of viremia suggests that ZIKV can establish persistent infection in the seminiferous tubules, an immune privileged site of the testis. The seminiferous tubule epithelium is mainly composed of Sertoli cells that function to nourish and protect developing germ cells. We recently demonstrated that primary human Sertoli cells (hSeC) were highly susceptible to ZIKV as compared to dengue virus without causing cell death and thus may act as a reservoir for ZIKV in the testes. However, the cellular and immune responses of hSeC to infection with ZIKV or any other virus are not yet characterized. Using genome-wide RNA-seq to compare immunoprofiles of hSeC, we show that the most prominent response to ZIKV at early stage of infection was suppression of cell growth and proliferation functional pathways. Peak virus replication was associated with induction of multiple antiviral defense pathways. Unique ZIKV-associated signatures included dysregulation of germ cell-Sertoli cell junction signaling. This study demonstrates that hSeC are capable of signaling through canonical pro-inflammatory pathways and provides insights into unique cell-type-specific response induced by ZIKV in association with viral persistence in the testes.
Collapse
Affiliation(s)
- Daniel P Strange
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Richard Green
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, Washington, USA
| | - David N Siemann
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Michael Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, Washington, USA.
| | - Saguna Verma
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA.
| |
Collapse
|
38
|
Li Z, Zhang Y, Jiang Z, Jin X, Zhang K, Wang X, Han C, Yu J, Li D. Hijacking of the nucleolar protein fibrillarin by TGB1 is required for cell-to-cell movement of Barley stripe mosaic virus. MOLECULAR PLANT PATHOLOGY 2018; 19:1222-1237. [PMID: 28872759 PMCID: PMC6638131 DOI: 10.1111/mpp.12612] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/31/2017] [Accepted: 09/04/2017] [Indexed: 05/18/2023]
Abstract
Barley stripe mosaic virus (BSMV) Triple Gene Block1 (TGB1) is a multifunctional movement protein with RNA-binding, ATPase and helicase activities which mainly localizes to the plasmodesmata (PD) in infected cells. Here, we show that TGB1 localizes to the nucleus and the nucleolus, as well as the cytoplasm, and that TGB1 nuclear-cytoplasmic trafficking is required for BSMV cell-to-cell movement. Prediction analyses and laser scanning confocal microscopy (LSCM) experiments verified that TGB1 possesses a nucleolar localization signal (NoLS) (amino acids 95-104) and a nuclear localization signal (NLS) (amino acids 227-238). NoLS mutations reduced BSMV cell-to-cell movement significantly, whereas NLS mutations almost completely abolished movement. Furthermore, neither the NoLS nor NLS mutant viruses could infect Nicotiana benthamiana systemically, although the NoLS mutant virus was able to establish systemic infections of barley. Protein interaction experiments demonstrated that TGB1 interacts directly with the glycine-arginine-rich (GAR) domain of the nucleolar protein fibrillarin (Fib2). Moreover, in BSMV-infected cells, Fib2 accumulation increased by about 60%-70% and co-localized with TGB1 in the plasmodesmata. In addition, BSMV cell-to-cell movement in fib2 knockdown transgenic plants was reduced to less than one-third of that of non-transgenic plants. Fib2 also co-localized with both TGB1 and BSMV RNA, which are the main components of the ribonucleoprotein (RNP) movement complex. Collectively, these results show that TGB1-Fib2 interactions play a direct role in cell-to-cell movement, and we propose that Fib2 is hijacked by BSMV TGB1 to form a BSMV RNP which functions in cell-to-cell movement.
Collapse
Affiliation(s)
- Zhenggang Li
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological SciencesChina Agricultural UniversityBeijing 100193China
| | - Yongliang Zhang
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological SciencesChina Agricultural UniversityBeijing 100193China
| | - Zhihao Jiang
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological SciencesChina Agricultural UniversityBeijing 100193China
| | - Xuejiao Jin
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological SciencesChina Agricultural UniversityBeijing 100193China
| | - Kun Zhang
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological SciencesChina Agricultural UniversityBeijing 100193China
| | - Xianbing Wang
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological SciencesChina Agricultural UniversityBeijing 100193China
| | - Chenggui Han
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological SciencesChina Agricultural UniversityBeijing 100193China
| | - Jialin Yu
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological SciencesChina Agricultural UniversityBeijing 100193China
| | - Dawei Li
- State Key Laboratory of Agro‐Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological SciencesChina Agricultural UniversityBeijing 100193China
| |
Collapse
|
39
|
Passos-Castilho AM, Marchand C, Archambault D. B23/nucleophosmin interacts with bovine immunodeficiency virus Rev protein and facilitates viral replication. Virology 2018; 515:158-164. [DOI: 10.1016/j.virol.2017.12.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022]
|
40
|
Slomnicki LP, Chung DH, Parker A, Hermann T, Boyd NL, Hetman M. Ribosomal stress and Tp53-mediated neuronal apoptosis in response to capsid protein of the Zika virus. Sci Rep 2017; 7:16652. [PMID: 29192272 PMCID: PMC5709411 DOI: 10.1038/s41598-017-16952-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 11/20/2017] [Indexed: 02/07/2023] Open
Abstract
We report here that in rat and human neuroprogenitor cells as well as rat embryonic cortical neurons Zika virus (ZIKV) infection leads to ribosomal stress that is characterized by structural disruption of the nucleolus. The anti-nucleolar effects were most pronounced in postmitotic neurons. Moreover, in the latter system, nucleolar presence of ZIKV capsid protein (ZIKV-C) was associated with ribosomal stress and apoptosis. Deletion of 22 C-terminal residues of ZIKV-C prevented nucleolar localization, ribosomal stress and apoptosis. Consistent with a casual relationship between ZIKV-C-induced ribosomal stress and apoptosis, ZIKV-C-overexpressing neurons were protected by loss-of-function manipulations targeting the ribosomal stress effector Tp53 or knockdown of the ribosomal stress mediator RPL11. Finally, capsid protein of Dengue virus, but not West Nile virus, induced ribosomal stress and apoptosis. Thus, anti-nucleolar and pro-apoptotic effects of protein C are flavivirus-species specific. In the case of ZIKV, capsid protein-mediated ribosomal stress may contribute to neuronal death, neurodevelopmental disruption and microcephaly.
Collapse
Affiliation(s)
- Lukasz P Slomnicki
- Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, 40292, USA
| | - Dong-Hoon Chung
- Center of Predictive Medicine and the Department of Microbiology & Immunology, University of Louisville, Louisville, Kentucky, 40292, USA
| | - Austin Parker
- Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, 40292, USA
| | - Taylor Hermann
- Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, 40292, USA
| | - Nolan L Boyd
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky, 40292, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center and the Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, 40292, USA.
- Pharmacology & Toxicology, University of Louisville, Louisville, Kentucky, 40292, USA.
| |
Collapse
|
41
|
Tripp RA, Tompkins SM, Foo CH, Bean AGD, Wang LF. A Functional Genomics Approach to Henipavirus Research: The Role of Nuclear Proteins, MicroRNAs and Immune Regulators in Infection and Disease. Curr Top Microbiol Immunol 2017; 419:191-213. [PMID: 28674944 PMCID: PMC7122743 DOI: 10.1007/82_2017_28] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hendra and Nipah viruses (family Paramyxoviridae, genus Henipavirus) are zoonotic RNA viruses that cause lethal disease in humans and are designated as Biosafety Level 4 (BSL4) agents. Moreover, henipaviruses belong to the same group of viruses that cause disease more commonly in humans such as measles, mumps and respiratory syncytial virus. Due to the relatively recent emergence of the henipaviruses and the practical constraints of performing functional genomics studies at high levels of containment, our understanding of the henipavirus infection cycle is incomplete. In this chapter we describe recent loss-of-function (i.e. RNAi) functional genomics screens that shed light on the henipavirus-host interface at a genome-wide level. Further to this, we cross-reference RNAi results with studies probing host proteins targeted by henipavirus proteins, such as nuclear proteins and immune modulators. These functional genomics studies join a growing body of evidence demonstrating that nuclear and nucleolar host proteins play a crucial role in henipavirus infection. Furthermore these studies will underpin future efforts to define the role of nucleolar host-virus interactions in infection and disease.
Collapse
Affiliation(s)
- Ralph A. Tripp
- grid.213876.90000 0004 1936 738XDepartment Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA USA
| | - S. Mark Tompkins
- grid.213876.90000 0004 1936 738XCenter for Vaccines and Immunology, University of Georgia, Athens, GA USA
| | - Chwan Hong Foo
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Andrew G D Bean
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, 169857, Singapore
| |
Collapse
|
42
|
Nonstructural Protein NSs of Schmallenberg Virus Is Targeted to the Nucleolus and Induces Nucleolar Disorganization. J Virol 2016; 91:JVI.01263-16. [PMID: 27795408 PMCID: PMC5165206 DOI: 10.1128/jvi.01263-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/04/2016] [Indexed: 01/04/2023] Open
Abstract
Schmallenberg virus (SBV) was discovered in Germany in late 2011 and then spread rapidly to many European countries. SBV is an orthobunyavirus that causes abortion and congenital abnormalities in ruminants. A virus-encoded nonstructural protein, termed NSs, is a major virulence factor of SBV, and it is known to promote the degradation of Rpb1, a subunit of the RNA polymerase II (Pol II) complex, and therefore hampers global cellular transcription. In this study, we found that NSs is mainly localized in the nucleus of infected cells and specifically appears to target the nucleolus through a nucleolar localization signal (NoLS) localized between residues 33 and 51 of the protein. NSs colocalizes with nucleolar markers such as B23 (nucleophosmin) and fibrillarin. We observed that in SBV-infected cells, B23 undergoes a nucleolus-to-nucleoplasm redistribution, evocative of virus-induced nucleolar disruption. In contrast, the nucleolar pattern of B23 was unchanged upon infection with an SBV recombinant mutant with NSs lacking the NoLS motif (SBVΔNoLS). Interestingly, unlike wild-type SBV, the inhibitory activity of SBVΔNoLS toward RNA Pol II transcription is impaired. Overall, our results suggest that a putative link exists between NSs-induced nucleolar disruption and its inhibitory function on cellular transcription, which consequently precludes the cellular antiviral response and/or induces cell death. IMPORTANCE Schmallenberg virus (SBV) is an emerging arbovirus of ruminants that spread in Europe between 2011 and 2013. SBV induces fetal abnormalities during gestation, with the central nervous system being one of the most affected organs. The virus-encoded NSs protein acts as a virulence factor by impairing host cell transcription. Here, we show that NSs contains a nucleolar localization signal (NoLS) and induces disorganization of the nucleolus. The NoLS motif in the SBV NSs is absolutely necessary for virus-induced inhibition of cellular transcription. To our knowledge, this is the first report of nucleolar functions for NSs within the Bunyaviridae family.
Collapse
|
43
|
Audsley MD, Jans DA, Moseley GW. Roles of nuclear trafficking in infection by cytoplasmic negative-strand RNA viruses: paramyxoviruses and beyond. J Gen Virol 2016; 97:2463-2481. [PMID: 27498841 DOI: 10.1099/jgv.0.000575] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Genome replication and virion production by most negative-sense RNA viruses (NSVs) occurs exclusively in the cytoplasm, but many NSV-expressed proteins undergo active nucleocytoplasmic trafficking via signals that exploit cellular nuclear transport pathways. Nuclear trafficking has been reported both for NSV accessory proteins (including isoforms of the rabies virus phosphoprotein, and V, W and C proteins of paramyxoviruses) and for structural proteins. Trafficking of the former is thought to enable accessory functions in viral modulation of antiviral responses including the type I IFN system, but the intranuclear roles of structural proteins such as nucleocapsid and matrix proteins, which have critical roles in extranuclear replication and viral assembly, are less clear. Nevertheless, nuclear trafficking of matrix protein has been reported to be critical for efficient production of Nipah virus and Respiratory syncytial virus, and nuclear localization of nucleocapsid protein of several morbilliviruses has been linked to mechanisms of immune evasion. Together, these data point to the nucleus as a significant host interface for viral proteins during infection by NSVs with otherwise cytoplasmic life cycles. Importantly, several lines of evidence now suggest that nuclear trafficking of these proteins may be critical to pathogenesis and thus could provide new targets for vaccine development and antiviral therapies.
Collapse
Affiliation(s)
- Michelle D Audsley
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - David A Jans
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Gregory W Moseley
- Department of Biochemistry and Molecular Biology, BIO21 Molecular Science and Biotechnology Institute, University of Melbourne, VIC 3000, Australia
| |
Collapse
|
44
|
Terrier O, Carron C, De Chassey B, Dubois J, Traversier A, Julien T, Cartet G, Proust A, Hacot S, Ressnikoff D, Lotteau V, Lina B, Diaz JJ, Moules V, Rosa-Calatrava M. Nucleolin interacts with influenza A nucleoprotein and contributes to viral ribonucleoprotein complexes nuclear trafficking and efficient influenza viral replication. Sci Rep 2016; 6:29006. [PMID: 27373907 PMCID: PMC4931502 DOI: 10.1038/srep29006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/09/2016] [Indexed: 01/18/2023] Open
Abstract
Influenza viruses replicate their single-stranded RNA genomes in the nucleus of infected cells and these replicated genomes (vRNPs) are then exported from the nucleus to the cytoplasm and plasma membrane before budding. To achieve this export, influenza viruses hijack the host cell export machinery. However, the complete mechanisms underlying this hijacking remain not fully understood. We have previously shown that influenza viruses induce a marked alteration of the nucleus during the time-course of infection and notably in the nucleolar compartment. In this study, we discovered that a major nucleolar component, called nucleolin, is required for an efficient export of vRNPs and viral replication. We have notably shown that nucleolin interacts with the viral nucleoprotein (NP) that mainly constitutes vRNPs. Our results suggest that this interaction could allow vRNPs to "catch" the host cell export machinery, a necessary step for viral replication.
Collapse
Affiliation(s)
- Olivier Terrier
- Virologie et Pathologie Humaine - Team VirPath - Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Lyon, France
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Coralie Carron
- Virologie et Pathologie Humaine - Team VirPath - Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Lyon, France
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Benoît De Chassey
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Julia Dubois
- Virologie et Pathologie Humaine - Team VirPath - Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Lyon, France
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Aurélien Traversier
- Virologie et Pathologie Humaine - Team VirPath - Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Lyon, France
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Thomas Julien
- Virologie et Pathologie Humaine - Team VirPath - Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Lyon, France
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Lyon 1, Lyon, France
| | - Gaëlle Cartet
- Virologie et Pathologie Humaine - Team VirPath - Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Lyon, France
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Anaïs Proust
- Virologie et Pathologie Humaine - Team VirPath - Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Lyon, France
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Lyon 1, Lyon, France
| | - Sabine Hacot
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052 CNRS 5286, Centre Léon Bérard, Lyon, France and Université de Lyon, Lyon, France
| | - Denis Ressnikoff
- CIQLE, Centre d’imagerie quantitative Lyon-Est, Université Claude Bernard Lyon 1, Lyon, France
| | - Vincent Lotteau
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Bruno Lina
- Virologie et Pathologie Humaine - Team VirPath - Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Lyon, France
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Hospices Civils de Lyon, Laboratory of Virology, Lyon, France
| | - Jean-Jacques Diaz
- Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052 CNRS 5286, Centre Léon Bérard, Lyon, France and Université de Lyon, Lyon, France
| | - Vincent Moules
- Virologie et Pathologie Humaine - Team VirPath - Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Lyon, France
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Lyon 1, Lyon, France
| | - Manuel Rosa-Calatrava
- Virologie et Pathologie Humaine - Team VirPath - Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, Lyon, France
- CIRI, International Center for Infectiology Research, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Lyon 1, Lyon, France
| |
Collapse
|
45
|
Dynamic Nucleolar Targeting of Dengue Virus Polymerase NS5 in Response to Extracellular pH. J Virol 2016; 90:5797-5807. [PMID: 27076639 DOI: 10.1128/jvi.02727-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/29/2016] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED The nucleolar subcompartment of the nucleus is increasingly recognized as an important target of RNA viruses. Here we document for the first time the ability of dengue virus (DENV) polymerase, nonstructural protein 5 (NS5), to accumulate within the nucleolus of infected cells and to target green fluorescent protein (GFP) to the nucleolus of live transfected cells. Intriguingly, NS5 exchange between the nucleus and nucleolus is dynamically modulated by extracellular pH, responding rapidly and reversibly to pH change, in contrast to GFP alone or other nucleolar and non-nucleolar targeted protein controls. The minimal pH-sensitive nucleolar targeting region (pHNTR), sufficient to target GFP to the nucleolus in a pH-sensitive fashion, was mapped to NS5 residues 1 to 244, with mutation of key hydrophobic residues, Leu-165, Leu-167, and Val-168, abolishing pHNTR function in NS5-transfected cells, and severely attenuating DENV growth in infected cells. This is the first report of a viral protein whose nucleolar targeting ability is rapidly modulated by extracellular stimuli, suggesting that DENV has the ability to detect and respond dynamically to the extracellular environment. IMPORTANCE Infections by dengue virus (DENV) threaten 40% of the world's population yet there is no approved vaccine or antiviral therapeutic to treat infections. Understanding the molecular details that govern effective viral replication is key for the development of novel antiviral strategies. Here, we describe for the first time dynamic trafficking of DENV nonstructural protein 5 (NS5) to the subnuclear compartment, the nucleolus. We demonstrate that NS5's targeting to the nucleolus occurs in response to acidic pH, identify the key amino acid residues within NS5 that are responsible, and demonstrate that their mutation severely impairs production of infectious DENV. Overall, this study identifies a unique subcellular trafficking event and suggests that DENV is able to detect and respond dynamically to environmental changes.
Collapse
|
46
|
Ito N, Moseley GW, Sugiyama M. The importance of immune evasion in the pathogenesis of rabies virus. J Vet Med Sci 2016; 78:1089-98. [PMID: 27041139 PMCID: PMC4976263 DOI: 10.1292/jvms.16-0092] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Rabies is a zoonotic disease caused by the Lyssavirus rabies virus
(RABV) that can infect most mammals, including humans, where it has a case-fatality rate
of almost 100%. Although preventable by vaccination, rabies causes c. 59,000 human
fatalities every year worldwide. Thus, there exists an urgent need to establish an
effective therapy and/or improve dissemination of vaccines for humans and animals. These
outcomes require greater understanding of the mechanisms of RABV pathogenesis to identify
new molecular targets for the development of therapeutics and/or live vaccines with high
levels of safety. Importantly, a number of studies in recent years have indicated that
RABV specifically suppresses host immunity through diverse mechanisms and that this is a
key process in pathogenicity. Here, we review current understanding of immune modulation
by RABV, with an emphasis on its significance to pathogenicity and the potential
exploitation of this knowledge to develop new vaccines and antivirals.
Collapse
Affiliation(s)
- Naoto Ito
- Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, Gifu 501-1193, Japan
| | | | | |
Collapse
|
47
|
Ma TH, Lee LW, Lee CC, Yi YH, Chan SP, Tan BCM, Lo SJ. Genetic control of nucleolar size: An evolutionary perspective. Nucleus 2016; 7:112-20. [PMID: 27003693 DOI: 10.1080/19491034.2016.1166322] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Exploiting a C. elegans mutant (ncl-1) exhibiting nucleolar abnormalities, we recently identified the let-7/ncl-1/fib-1 genetic cascade underlying proper rRNA abundance and nucleolar size. These 3 factors, let-7 (a miRNA), NCL-1 (a member of the TRIM-NHL family), and fibrillarin (a nucleolar methyltransferase), are evolutionarily conserved across metazoans. In this article, we provide several lines of bioinformatic evidence showing that human and Drosophila homologues of C. elegans NCL-1, TRIM-71 and Brat, respectively, likely act as translational suppressors of fibrillarin. Moreover, since their 3'-UTRs contain putative target sites, they may also be under the control of the let-7 miRNA. We hypothesize that let-7, TRIM and fibrillarin contribute activities in concert, and constitute a conserved network controlling nucleolar size in eukaryotes. We provide an in-depth literature review of various molecular pathways, including the let-7/ncl-1/fib-1 genetic cascade, implicated in the regulation of nucleolar size.
Collapse
Affiliation(s)
- Tian-Hsiang Ma
- a Department of Biomedical Sciences, College of Medicine , Chang Gung University , TaoYuan , Taiwan.,b Graduate Institute of Biomedical Sciences , College of Medicine , Chang Gung University , TaoYuan , Taiwan
| | - Li-Wei Lee
- a Department of Biomedical Sciences, College of Medicine , Chang Gung University , TaoYuan , Taiwan.,c Molecular Medicine Research Center , College of Medicine , Chang Gung University , TaoYuan , Taiwan
| | - Chi-Chang Lee
- d Institute of Chemistry , Academia Sinica , Taipei , Taiwan
| | - Yung-Hsiang Yi
- a Department of Biomedical Sciences, College of Medicine , Chang Gung University , TaoYuan , Taiwan.,c Molecular Medicine Research Center , College of Medicine , Chang Gung University , TaoYuan , Taiwan
| | - Shih-Peng Chan
- e Graduate Institute of Microbiology , College of Medicine , National Taiwan University , Taipei , Taiwan
| | - Bertrand Chin-Ming Tan
- a Department of Biomedical Sciences, College of Medicine , Chang Gung University , TaoYuan , Taiwan.,b Graduate Institute of Biomedical Sciences , College of Medicine , Chang Gung University , TaoYuan , Taiwan.,c Molecular Medicine Research Center , College of Medicine , Chang Gung University , TaoYuan , Taiwan
| | - Szecheng J Lo
- a Department of Biomedical Sciences, College of Medicine , Chang Gung University , TaoYuan , Taiwan.,b Graduate Institute of Biomedical Sciences , College of Medicine , Chang Gung University , TaoYuan , Taiwan.,c Molecular Medicine Research Center , College of Medicine , Chang Gung University , TaoYuan , Taiwan
| |
Collapse
|
48
|
Musinova YR, Sheval EV, Dib C, Germini D, Vassetzky YS. Functional roles of HIV-1 Tat protein in the nucleus. Cell Mol Life Sci 2016; 73:589-601. [PMID: 26507246 PMCID: PMC11108392 DOI: 10.1007/s00018-015-2077-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/01/2015] [Accepted: 10/16/2015] [Indexed: 02/06/2023]
Abstract
Human immunodeficiency virus-1 (HIV-1) Tat protein is one of the most important regulatory proteins for viral gene expression in the host cell and can modulate different cellular processes. In addition, Tat is secreted by the infected cell and can be internalized by neighboring cells; therefore, it affects both infected and uninfected cells. Tat can modulate cellular processes by interacting with different cellular structures and signaling pathways. In the nucleus, Tat might be localized either in the nucleoplasm or the nucleolus depending on its concentration. Here we review the distinct functions of Tat in the nucleoplasm and the nucleolus in connection with viral infection and HIV-induced oncogenesis.
Collapse
Affiliation(s)
- Yana R Musinova
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, 119991, Moscow, Russia
- LIA 1066 French-Russian Joint Cancer Research Laboratory, 94805, Villejuif, France
| | - Eugene V Sheval
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, 119991, Moscow, Russia
- LIA 1066 French-Russian Joint Cancer Research Laboratory, 94805, Villejuif, France
| | - Carla Dib
- LIA 1066 French-Russian Joint Cancer Research Laboratory, 94805, Villejuif, France
- UMR8126, Université Paris-Sud, CNRS, Institut de cancérologie Gustave Roussy, 94805, Villejuif, France
| | - Diego Germini
- LIA 1066 French-Russian Joint Cancer Research Laboratory, 94805, Villejuif, France
- UMR8126, Université Paris-Sud, CNRS, Institut de cancérologie Gustave Roussy, 94805, Villejuif, France
| | - Yegor S Vassetzky
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, 119991, Moscow, Russia.
- LIA 1066 French-Russian Joint Cancer Research Laboratory, 94805, Villejuif, France.
- UMR8126, Université Paris-Sud, CNRS, Institut de cancérologie Gustave Roussy, 94805, Villejuif, France.
| |
Collapse
|
49
|
Stewart M, Hardy A, Barry G, Pinto RM, Caporale M, Melzi E, Hughes J, Taggart A, Janowicz A, Varela M, Ratinier M, Palmarini M. Characterization of a second open reading frame in genome segment 10 of bluetongue virus. J Gen Virol 2015; 96:3280-3293. [PMID: 26290332 PMCID: PMC4806581 DOI: 10.1099/jgv.0.000267] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Viruses have often evolved overlapping reading frames in order to maximize their coding capacity. Until recently, the segmented dsRNA genome of viruses of the Orbivirus genus was thought to be monocistronic, but the identification of the bluetongue virus (BTV) NS4 protein changed this assumption. A small ORF in segment 10, overlapping the NS3 ORF in the +1 position, is maintained in more than 300 strains of the 27 different BTV serotypes and in more than 200 strains of the phylogenetically related African horse sickness virus (AHSV). In BTV, this ORF (named S10-ORF2 in this study) encodes a putative protein 50–59 residues in length and appears to be under strong positive selection. HA- or GFP-tagged versions of S10-ORF2 expressed from transfected plasmids localized within the nucleoli of transfected cells, unless a putative nucleolar localization signal was mutated. S10-ORF2 inhibited gene expression, but not RNA translation, in transient transfection reporter assays. In both mammalian and insect cells, BTV S10-ORF2 deletion mutants (BTV8ΔS10-ORF2) displayed similar replication kinetics to wt virus. In vivo, S10-ORF2 deletion mutants were pathogenic in mouse models of disease. Although further evidence is required for S10-ORF2 expression during infection, the data presented provide an initial characterization of this ORF.
Collapse
Affiliation(s)
- Meredith Stewart
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Alexandra Hardy
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Gerald Barry
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Rute Maria Pinto
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Marco Caporale
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.,Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise 'G. Caporale', Teramo, Italy
| | - Eleonora Melzi
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Aislynn Taggart
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Anna Janowicz
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Mariana Varela
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Maxime Ratinier
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | |
Collapse
|