1
|
Semizhon PA, Scheslenok EP, Dubkov NA, Sukhotskaya EA, Stolbunova KA, Popov IV, Popov IV, Alekseev AY, Kabwe E, Davidyuk YN. [Identification of orthohantaviruses detected for the first time in the Republic of Belarus]. Vopr Virusol 2025; 70:87-98. [PMID: 40233340 DOI: 10.36233/0507-4088-292] [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: 02/13/2025] [Indexed: 04/17/2025]
Abstract
INTRODUCTION Monitoring of hemorrhagic fever with renal syndrome (HFRS) pathogens in the Republic of Belarus is necessary and relevant, since the number of HFRS cases in the population has increased in recent years, and genetic characteristics of the pathogens remain unidentified. AIM OF THE STUDY Identification of orthohantaviruses circulating in the territory of the Republic of Belarus and defining of their genetic characteristics. MATERIALS AND METHODS Screening of 613 samples from small mammals caught in the territory of the Republic of Belarus was carried out by the real time PCR method using the test system «Belar-GLPS-PCR/RV». Positive samples were sequenced by the Sanger method. Comparative and phylogenetic analysis was carried out using the MegAlign programs from the Lasergene package (DNASTAR, USA) and MEGA 11. RESULTS The primary screening yielded 32 PCR-positive samples (5.2%), of which 24 belonged to Puumala virus (PUUV) and 8 to Dobrava-Belgrade virus (DOBV). Three nucleotide sequences of the M-segment region of PUUV, two sequences of the 291-base pair (bp) M-segment region and one sequence of the 348-bp S-segment region of DOBV were sequenced. Comparative and phylogenetic analysis showed that the identified PUUV sequences belong to the Russian genetic lineage, to the same sublineage as the strains common in the Moscow and Kursk regions. The identified DOBV ssequences demonstrated the closest relationship to the strains from the central region of the European part of Russia. CONCLUSION The results of molecular biological analysis showed that PUUV circulates in the territory of the Republic of Belarus and is widespread. At the same time, DOBV was detected in four regions of the republic, which indicates an expansion of the range of this HFRS pathogen. In the Republic of Belarus, nucleotide sequences of orthohantaviruses were obtained for the first time and their molecular genetic analysis was carried out.
Collapse
Affiliation(s)
- P A Semizhon
- Republican Center for Hygiene, Epidemiology and Public Health of the Ministry of Health of the Republic of Belarus
| | - E P Scheslenok
- Republican Center for Hygiene, Epidemiology and Public Health of the Ministry of Health of the Republic of Belarus
| | - N A Dubkov
- Republican Center for Hygiene, Epidemiology and Public Health of the Ministry of Health of the Republic of Belarus
| | - E A Sukhotskaya
- Republican Center for Hygiene, Epidemiology and Public Health of the Ministry of Health of the Republic of Belarus
| | - K A Stolbunova
- Research Institute of Virology, Federal Research Center for Fundamental and Translational Medicine
| | | | | | - A Y Alekseev
- Research Institute of Virology, Federal Research Center for Fundamental and Translational Medicine
| | - E Kabwe
- Institute of Fundamental Medicine and Biology, Kazan Federal University
| | - Y N Davidyuk
- Institute of Fundamental Medicine and Biology, Kazan Federal University
| |
Collapse
|
2
|
Shkair L, Sharma D, Hamza S, Garanina E, Shakirova V, Khaertynova I, Markelova M, Pavelkina V, Rizvanov A, Khaiboullina S, Baranwal M, Martynova E. Cross-reactivity of hantavirus antibodies after immunization with PUUV antigens. Biotechnol Appl Biochem 2024; 71:1139-1153. [PMID: 38779849 DOI: 10.1002/bab.2604] [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: 11/29/2023] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
Nephropathia epidemica (NE), caused by Puumala (PUUV) orthohantavirus, is endemic in the Republic of Tatarstan (RT). There are limited options for NE prevention in RT. Currently, available vaccines are made using Haantan (HNTV) orthohantavirus antigens. In this study, the efficacy of microvesicles (MVs) loaded with PUUV antigens to induce the humoral immune response in small mammals was analyzed. Additionally, the cross-reactivity of serum from immunized small mammals and NE patients with HNTV, Dobrava, and Andes orthohantaviruses was investigated using nucleocapsid (N) protein peptide libraries. Finally, the selected peptides were analyzed for allergenicity, their ability to induce an autoimmune response, and their interaction with Class II HLA. Several N protein peptides were found to be cross-reactive with serum from MVs immunized small mammals. These cross-reactive epitopes were located in oligomerization perinuclear targeting and Daxx-interacting domains. Most cross-reactive peptides lack allergenic and autoimmune reactivity. Molecular docking revealed two cross-reacting peptides, N6 and N19, to have good binding with three Class II HLA alleles. These peptides could be candidates for developing vaccines and therapeutics for NE.
Collapse
Affiliation(s)
- Layaly Shkair
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Diksha Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Shaimaa Hamza
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Ekaterina Garanina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Venara Shakirova
- Department of Infectious Diseases, Kazan State Medical Academy, Kazan, Russia
| | - Ilsiyar Khaertynova
- Department of Infectious Diseases, Kazan State Medical Academy, Kazan, Russia
| | - Maria Markelova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Vera Pavelkina
- Infectious Diseases Department, National Research Ogarev Mordovia State University, Saransk, Russia
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Svetlana Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Manoj Baranwal
- Infectious Diseases Department, National Research Ogarev Mordovia State University, Saransk, Russia
| | - Ekaterina Martynova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
3
|
Chang M, Min YQ, Xu Z, Deng F, Wang H, Ning YJ. Host factor MxA restricts Dabie bandavirus infection by targeting the viral NP protein to inhibit NP-RdRp interaction and ribonucleoprotein activity. J Virol 2024; 98:e0156823. [PMID: 38054738 PMCID: PMC10805036 DOI: 10.1128/jvi.01568-23] [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: 10/08/2023] [Accepted: 11/20/2023] [Indexed: 12/07/2023] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease with high case mortality rates, which is caused by Dabie bandavirus (DBV), a novel pathogen also termed as SFTS virus (SFTSV). Currently, no specific therapeutic drugs or vaccines are available for SFTS. Myxovirus resistance protein A (MxA) has been shown to inhibit multiple viral pathogens; however, the role of MxA in DBV infection is unknown. Here, we demonstrated that DBV stimulates MxA expression which, in turn, restricts DBV infection. Mechanistic target analysis revealed that MxA specifically interacts with the viral nucleocapsid protein (NP) in a manner independent of RNA. Minigenome reporter assay showed that in agreement with its targeting of NP, MxA inhibits DBV ribonucleoprotein (RNP) activity. In detail, MxA interacts with the NP N-terminal and disrupts the interaction of NP with the viral RNA-dependent RNA polymerase (RdRp) but not NP multimerization, the critical activities of NP for RNP formation and function. Furthermore, MxA N-terminal domain was identified as the functional domain inhibiting DBV infection, and, consistently, then was shown to interact with NP and obstruct the NP-RdRp interaction. Additionally, threonine 103 within the N-terminal domain is important for MxA inhibition to DBV, and its mutation (T103A) attenuates MxA binding to NP and obstruction of the NP-RdRp interaction. This study uncovers MxA inhibition of DBV with a series of functional and mechanistical analyses, providing insights into the virus-host interactions and probably helping inform the development of antiviral agents in the future.IMPORTANCEDBV/SFTSV is an emerging high-pathogenic virus. Since its first identification in China in 2009, cases of DBV infection have been reported in many other countries, posing a significant threat to public health. Uncovering the mechanisms of DBV-host interactions is necessary to understand the viral pathogenesis and host response and may advance the development of antiviral therapeutics. Here, we found that host factor MxA whose expression is induced by DBV restricts the virus infection. Mechanistically, MxA specifically interacts with the viral NP and blocks the NP-RdRp interaction, inhibiting the viral RNP activity. Further studies identified the key domain and amino acid residue required for MxA inhibition to DBV. Consistently, they were then shown to be important for MxA targeting of NP and obstruction of the NP-RdRp association. These findings unravel the restrictive role of MxA in DBV infection and the underlying mechanism, expanding our knowledge of the virus-host interactions.
Collapse
Affiliation(s)
- Meng Chang
- Key Laboratory of Virology and Biosafety and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuan-Qin Min
- Key Laboratory of Virology and Biosafety and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Virology and Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Zhao Xu
- Key Laboratory of Virology and Biosafety and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fei Deng
- Key Laboratory of Virology and Biosafety and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Virology and Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Hualin Wang
- Key Laboratory of Virology and Biosafety and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Virology and Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yun-Jia Ning
- Key Laboratory of Virology and Biosafety and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Virology and Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| |
Collapse
|
4
|
Fan Y, Li X, Zhang L, Zong Z, Wang F, Huang J, Zeng L, Zhang C, Yan H, Zhang L, Zhou F. SUMOylation in Viral Replication and Antiviral Defense. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104126. [PMID: 35060688 PMCID: PMC8895153 DOI: 10.1002/advs.202104126] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/07/2021] [Indexed: 05/22/2023]
Abstract
SUMOylation is a ubiquitination-like post-translational modification that plays an essential role in the regulation of protein function. Recent studies have shown that proteins from both RNA and DNA virus families can be modified by SUMO conjugation, which facilitates viral replication. Viruses can manipulate the entire process of SUMOylation through interplay with the SUMO pathway. By contrast, SUMOylation can eliminate viral infection by regulating host antiviral immune components. A deeper understanding of how SUMOylation regulates viral proteins and cellular antiviral components is necessary for the development of effective antiviral therapies. In the present review, the regulatory mechanism of SUMOylation in viral replication and infection and the antiviral immune response, and the consequences of this regulation for viral replication and engagement with antiviral innate immunity are summarized. The potential therapeutic applications of SUMOylation in diseases caused by viruses are also discussed.
Collapse
Affiliation(s)
- Yao Fan
- Department of PharmacologyZhejiang University City College School of MedicineHangzhouZhejiang310015China
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123China
| | - Xiang Li
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Lei Zhang
- Department of Orthopaedic SurgeryThe Third Affiliated Hospital of Wenzhou Medical UniversityRui'an325200China
| | - Zhi Zong
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Fangwei Wang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Jun Huang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Linghui Zeng
- Department of PharmacologyZhejiang University City College School of MedicineHangzhouZhejiang310015China
| | - Chong Zhang
- Department of PharmacologyZhejiang University City College School of MedicineHangzhouZhejiang310015China
| | - Haiyan Yan
- Department of PharmacologyZhejiang University City College School of MedicineHangzhouZhejiang310015China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Fangfang Zhou
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123China
| |
Collapse
|
5
|
Welke RW, Sperber HS, Bergmann R, Koikkarah A, Menke L, Sieben C, Krüger DH, Chiantia S, Herrmann A, Schwarzer R. Characterization of Hantavirus N Protein Intracellular Dynamics and Localization. Viruses 2022; 14:v14030457. [PMID: 35336863 PMCID: PMC8954124 DOI: 10.3390/v14030457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/02/2022] [Accepted: 02/18/2022] [Indexed: 02/07/2023] Open
Abstract
Hantaviruses are enveloped viruses that possess a tri-segmented, negative-sense RNA genome. The viral S-segment encodes the multifunctional nucleocapsid protein (N), which is involved in genome packaging, intracellular protein transport, immunoregulation, and several other crucial processes during hantavirus infection. In this study, we generated fluorescently tagged N protein constructs derived from Puumalavirus (PUUV), the dominant hantavirus species in Central, Northern, and Eastern Europe. We comprehensively characterized this protein in the rodent cell line CHO-K1, monitoring the dynamics of N protein complex formation and investigating co-localization with host proteins as well as the viral glycoproteins Gc and Gn. We observed formation of large, fibrillar PUUV N protein aggregates, rapidly coalescing from early punctate and spike-like assemblies. Moreover, we found significant spatial correlation of N with vimentin, actin, and P-bodies but not with microtubules. N constructs also co-localized with Gn and Gc albeit not as strongly as the glycoproteins associated with each other. Finally, we assessed oligomerization of N constructs, observing efficient and concentration-dependent multimerization, with complexes comprising more than 10 individual proteins.
Collapse
Affiliation(s)
- Robert-William Welke
- Department of Molecular Biophysics, Humboldt University, 10115 Berlin, Germany; (R.-W.W.); (R.B.); (A.H.)
| | - Hannah Sabeth Sperber
- Institute for Translational HIV Research, University Hospital Essen, 45147 Essen, Germany;
| | - Ronny Bergmann
- Department of Molecular Biophysics, Humboldt University, 10115 Berlin, Germany; (R.-W.W.); (R.B.); (A.H.)
| | - Amit Koikkarah
- Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany; (A.K.); (S.C.)
| | - Laura Menke
- Nanoscale Infection Biology Group, Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (L.M.); (C.S.)
| | - Christian Sieben
- Nanoscale Infection Biology Group, Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (L.M.); (C.S.)
- Institute for Genetics, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Detlev H. Krüger
- Institut für Virologie, Charité–Universitätsmedizin Berlin, Gliedkörperschaft der Freien Universität Berlin und der Humboldt-Universität zu Berlin, 10117 Berlin, Germany;
| | - Salvatore Chiantia
- Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany; (A.K.); (S.C.)
| | - Andreas Herrmann
- Department of Molecular Biophysics, Humboldt University, 10115 Berlin, Germany; (R.-W.W.); (R.B.); (A.H.)
- Biophysikalische Chemie, Freie Universität, 14195 Berlin, Germany
| | - Roland Schwarzer
- Institute for Translational HIV Research, University Hospital Essen, 45147 Essen, Germany;
- Correspondence:
| |
Collapse
|
6
|
Imbert F, Langford D. Viruses, SUMO, and immunity: the interplay between viruses and the host SUMOylation system. J Neurovirol 2021; 27:531-541. [PMID: 34342851 PMCID: PMC8330205 DOI: 10.1007/s13365-021-00995-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/19/2021] [Accepted: 06/29/2021] [Indexed: 12/14/2022]
Abstract
The conjugation of small ubiquitin-like modifier (SUMO) proteins to substrates is a well-described post-translational modification that regulates protein activity, subcellular localization, and protein-protein interactions for a variety of downstream cellular activities. Several studies describe SUMOylation as an essential post-translational modification for successful viral infection across a broad range of viruses, including RNA and DNA viruses, both enveloped and un-enveloped. These viruses include but are not limited to herpes viruses, human immunodeficiency virus-1, and coronaviruses. In addition to the SUMOylation of viral proteins during infection, evidence shows that viruses manipulate the SUMO pathway for host protein SUMOylation. SUMOylation of host and viral proteins greatly impacts host innate immunity through viral manipulation of the host SUMOylation machinery to promote viral replication and pathogenesis. Other post-translational modifications like phosphorylation can also modulate SUMO function. For example, phosphorylation of COUP-TF interacting protein 2 (CTIP2) leads to its SUMOylation and subsequent proteasomal degradation. The SUMOylation of CTIP2 and subsequent degradation prevents CTIP2-mediated recruitment of a multi-enzymatic complex to the HIV-1 promoter that usually prevents the transcription of integrated viral DNA. Thus, the "SUMO switch" could have implications for CTIP2-mediated transcriptional repression of HIV-1 in latency and viral persistence. In this review, we describe the consequences of SUMO in innate immunity and then focus on the various ways that viral pathogens have evolved to hijack the conserved SUMO machinery. Increased understanding of the many roles of SUMOylation in viral infections can lead to novel insight into the regulation of viral pathogenesis with the potential to uncover new targets for antiviral therapies.
Collapse
Affiliation(s)
- Fergan Imbert
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, PA, 19140, Philadelphia, USA
| | - Dianne Langford
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, PA, 19140, Philadelphia, USA.
| |
Collapse
|
7
|
Abstract
Over the past 20 years, humankind has encountered three severe coronavirus outbreaks. Currently ongoing, COVID-19 (coronavirus disease 2019) was declared a pandemic due to its massive impact on global health and the economy. Numerous scientists are working to identify efficacious therapeutic agents for COVID-19, although treatment ability has yet to be demonstrated. The SUMO (small ubiquitin-like modifier) system has diverse roles in viral manipulation, but the function of SUMO in coronaviruses is still unknown. The objective of this review article is to present recently published data suggesting contributions of the host SUMO system to coronavirus infection. These findings underscore the potential of SUMO as a novel target for anti-coronavirus therapy, and the need for a deeper understanding of coronavirus pathology to prepare and prevail against the current and emerging coronavirus outbreaks.
Collapse
Affiliation(s)
- Hong-Yeoul Ryu
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of National Sciences, Kyungpook National University, Daegu, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
8
|
Arragain B, Reguera J, Desfosses A, Gutsche I, Schoehn G, Malet H. High resolution cryo-EM structure of the helical RNA-bound Hantaan virus nucleocapsid reveals its assembly mechanisms. eLife 2019; 8:43075. [PMID: 30638449 PMCID: PMC6365055 DOI: 10.7554/elife.43075] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/10/2019] [Indexed: 12/21/2022] Open
Abstract
Negative-strand RNA viruses condense their genome into helical nucleocapsids that constitute essential templates for viral replication and transcription. The intrinsic flexibility of nucleocapsids usually prevents their full-length structural characterisation at high resolution. Here, we describe purification of full-length recombinant metastable helical nucleocapsid of Hantaan virus (Hantaviridae family, Bunyavirales order) and determine its structure at 3.3 Å resolution by cryo-electron microscopy. The structure reveals the mechanisms of helical multimerisation via sub-domain exchanges between protomers and highlights nucleotide positions in a continuous positively charged groove compatible with viral genome binding. It uncovers key sites for future structure-based design of antivirals that are currently lacking to counteract life-threatening hantavirus infections. The structure also suggests a model of nucleoprotein-polymerase interaction that would enable replication and transcription solely upon local disruption of the nucleocapsid.
Collapse
Affiliation(s)
- Benoît Arragain
- Electron Microscopy and Methods Group, Université Grenoble Alpes, CNRS, CEA, Institute for Structural Biology, Grenoble, France
| | - Juan Reguera
- Complexes Macromoléculaires Viraux, Aix-Marseille Université, CNRS, INSERM, AFMB UMR 7257, Marseille, France
| | - Ambroise Desfosses
- Electron Microscopy and Methods Group, Université Grenoble Alpes, CNRS, CEA, Institute for Structural Biology, Grenoble, France
| | - Irina Gutsche
- Electron Microscopy and Methods Group, Université Grenoble Alpes, CNRS, CEA, Institute for Structural Biology, Grenoble, France
| | - Guy Schoehn
- Electron Microscopy and Methods Group, Université Grenoble Alpes, CNRS, CEA, Institute for Structural Biology, Grenoble, France
| | - Hélène Malet
- Electron Microscopy and Methods Group, Université Grenoble Alpes, CNRS, CEA, Institute for Structural Biology, Grenoble, France
| |
Collapse
|
9
|
Mutational analysis of Rift Valley fever phlebovirus nucleocapsid protein indicates novel conserved, functional amino acids. PLoS Negl Trop Dis 2017; 11:e0006155. [PMID: 29267287 PMCID: PMC5764413 DOI: 10.1371/journal.pntd.0006155] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 01/11/2018] [Accepted: 12/07/2017] [Indexed: 11/19/2022] Open
Abstract
Rift Valley fever phlebovirus (RVFV; Phenuiviridae, Phlebovirus) is an important mosquito-borne pathogen of both humans and ruminants. The RVFV genome is composed of tripartite, single stranded, negative or ambisense RNAs. The small (S) segment encodes both the nucleocapsid protein (N) and the non-structural protein (NSs). The N protein is responsible for the formation of the viral ribonucleoprotein (RNP) complexes, which are essential in the virus life cycle and for the transcription and replication of the viral genome. There is currently limited knowledge surrounding the roles of the RVFV nucleocapsid protein in viral infection other than its key functions: N protein multimerisation, encapsidation of the RNA genome and interactions with the RNA-dependent RNA polymerase, L. By bioinformatic comparison of the N sequences of fourteen phleboviruses, mutational analysis, minigenome assays and packaging assays, we have further characterised the RVFV N protein. Amino acids P11 and F149 in RVFV N play an essential role in the function of RNPs and are neither associated with N protein multimerisation nor known nucleocapsid protein functions and may have additional roles in the virus life cycle. Amino acid Y30 exhibited increased minigenome activity despite reduced RNA binding capacity. Additionally, we have determined that the N-terminal arm of N protein is not involved in N-L interactions. Elucidating the fundamental processes that involve the nucleocapsid protein will add to our understanding of this important viral protein and may influence future studies in the development of novel antiviral strategies.
Collapse
|
10
|
Reuter M, Krüger DH. The nucleocapsid protein of hantaviruses: much more than a genome-wrapping protein. Virus Genes 2017; 54:5-16. [PMID: 29159494 DOI: 10.1007/s11262-017-1522-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/11/2017] [Indexed: 12/11/2022]
Abstract
The nucleocapsid (N) protein of hantaviruses represents an impressive example of a viral multifunctional protein. It encompasses properties as diverse as genome packaging, RNA chaperoning, intracellular protein transport, DNA degradation, intervention in host translation, and restricting host immune responses. These functions all rely on the capability of N to interact with RNA and other viral and cellular proteins. We have compiled data on the N protein of different hantavirus species together with information of the recently published three-dimensional structural data of the protein. The array of diverse functional activities accommodated in the hantaviral N protein goes far beyond to be a static structural protein and makes it an interesting target in the development of antiviral therapeutics.
Collapse
Affiliation(s)
- Monika Reuter
- Institute of Virology, Helmut-Ruska-Haus, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| | - Detlev H Krüger
- Institute of Virology, Helmut-Ruska-Haus, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
11
|
Moreli ML, Novaes DPDS, Flor EC, Saivish MV, Costa VGD. Seropositivity diagnosis for hantavirus in Jataí, Goiás State, Brazil. Rev Soc Bras Med Trop 2017; 50:530-534. [DOI: 10.1590/0037-8682-0035-2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/17/2017] [Indexed: 11/22/2022] Open
|
12
|
Abstract
Hantaviruses are emerging zoonotic pathogens that belong to the Bunyaviridae family. They have been classified as category A pathogens by CDC (centers for disease control and prevention). Hantaviruses pose a serious threat to human health because their infection causes two highly fatal diseases, hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS). These pathogens are transmitted to humans through aerosolized excreta of their infected rodent hosts. Hantaviruses have a tripartite-segmented negative-sense RNA genome. The three genomic RNA segments, S, M, and L, encode a nucleocapsid protein (N), a precursor glycoprotein that is processed into two envelope glycoproteins (Gn and Gc) and the viral RNA-dependent RNA polymerase (RdRp), respectively. N protein is the major structural component of the virus, its main function is to protect and encapsidate the three genomic RNAs forming three viral ribonucleocapsids. Recent studies have proposed that N in conjunction with RdRp plays important roles in the transcription and replication of viral genome. In addition, N preferentially facilitates the translation of viral mRNA in cells. Glycoproteins, Gn and Gc, play major roles in viral attachment and entry to the host cells, virulence, and assembly and packaging of new virions in infected cells. RdRp functions as RNA replicase and transcriptase to replicate and transcribe the viral RNA and is also thought to have endonuclease activity. Currently, no antiviral therapy or vaccine is available for the treatment of hantavirus-associated diseases. Understanding the molecular details of hantavirus life cycle will help in the identification of targets for antiviral therapeutics and in the design of potential antiviral drug for the treatment of HFRS and HCPS. Due to the alarming fatality of hantavirus diseases, development of an effective vaccine against hantaviruses is a necessity.
Collapse
|
13
|
Olal D, Daumke O. Structure of the Hantavirus Nucleoprotein Provides Insights into the Mechanism of RNA Encapsidation. Cell Rep 2016; 14:2092-2099. [PMID: 26923588 DOI: 10.1016/j.celrep.2016.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/07/2015] [Accepted: 01/25/2016] [Indexed: 11/19/2022] Open
Abstract
Hantaviruses are etiological agents of life-threatening hemorrhagic fever with renal syndrome and hantavirus cardiopulmonary syndrome. The nucleoprotein (N) of hantavirus is essential for viral transcription and replication, thus representing an attractive target for therapeutic intervention. We have determined the crystal structure of hantavirus N to 3.2 Å resolution. The structure reveals a two-lobed, mostly α-helical structure that is distantly related to that of orthobunyavirus Ns. A basic RNA binding pocket is located at the intersection between the two lobes. We provide evidence that oligomerization is mediated by amino- and C-terminal arms that bind to the adjacent monomers. Based on these findings, we suggest a model for the oligomeric ribonucleoprotein (RNP) complex. Our structure provides mechanistic insights into RNA encapsidation in the genus Hantavirus and constitutes a template for drug discovery efforts aimed at combating hantavirus infections.
Collapse
Affiliation(s)
- Daniel Olal
- Crystallography, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125 Berlin, Germany.
| | - Oliver Daumke
- Crystallography, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125 Berlin, Germany; Biochemie, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany.
| |
Collapse
|
14
|
Phosphorylation of the nucleocapsid protein of Hantaan virus by casein kinase II. J Microbiol 2015; 53:343-7. [PMID: 25935306 DOI: 10.1007/s12275-015-5095-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/03/2015] [Accepted: 04/10/2015] [Indexed: 10/23/2022]
Abstract
Hantaanvirus (HTNV) is the prototype of the genus Hantavirus, which belongs to the family Bunyaviridae. Hantaviruses are carried and transmitted by rodents and are known to cause two serious disease syndromes in humans i.e., hemorrhagic fever with renal syndrome (HFRS) and the hantavirus pulmonary syndrome (HPS). HTNV is an enveloped virus that contains a tripartite genome consisting of three negative-sense RNA segments (L, M, S), and the S and M segment of HTNV, respectively, encode the viral nucleocapsid protein (NP) and envelope glycoproteins. Possible phosphorylation motifs of casein kinase II (CKII) and protein kinase C (PKC) were identified in HTNV NP through bioinformatics searches. Sucrose gradient SDS-PAGE analysis indicated that dephosphorylated HTNV NP migrated faster than non-dephosphorylated NP, suggesting that HTNV NP is phosphorylated in infected Vero E6 cells. Immunoblot anaylsis of HTNV particles with anti-phosphoserine antibody and anti-phosphothreonine antibody after immunoprecipitation showed that viral particles are readily phosphorylated at threonine residues. In vitro kinase assay further showed that HTNV NP is phosphorylated by CK II, but not by PKC. Full length or truncated HTNV NPs expressed in E. coli were phosphorylated in vitro by CKII suggesting that phosphorylation may occur in vivo at multiple sites. Site specific mutagenesis studies suggest that HTNV NP phosphorylation might occur at unknown sites excluding the site-directly mutagenized locations. Taken together, HTNV NP can be phosphorylated mainly at threonine residues in vivo by CK II treatment.
Collapse
|
15
|
Ribosomal protein S19-binding domain provides insights into hantavirus nucleocapsid protein-mediated translation initiation mechanism. Biochem J 2015; 464:109-21. [PMID: 25062117 DOI: 10.1042/bj20140449] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The hantaviral zoonotic diseases pose a significant threat to human health due to the lack of potential antiviral therapeutics or a vaccine against hantaviruses. N (Sin Nombre hantavirus nucleocapsid protein) augments mRNA translation. N binds to both the mRNA 5' cap and 40S ribosomal subunit via RPS19 (ribosomal protein S19). N with the assistance of the viral mRNA 5'-UTR preferentially favours the translation of a downstream ORF. We identified and characterized the RPS19-binding domain at the N-terminus of N. Its deletion did not influence the secondary structure, but affected the conformation of trimeric N molecules. The N variant lacking the RPS19-binding region was able to bind both the mRNA 5' cap and panhandle-like structure, formed by the termini of viral genomic RNA. In addition, the N variant formed stable trimers similar to wild-type N. Use of this variant in multiple experiments provided insights into the mechanism of ribosome loading during N-mediated translation strategy. The present study suggests that N molecules individually associated with the mRNA 5' cap and RPS19 of the 40S ribosomal subunit undergo N-N interaction to facilitate the engagement of N-associated ribosomes at the mRNA 5' cap. This has revealed new targets for therapeutic intervention of hantavirus infection.
Collapse
|
16
|
Ali HS, Drewes S, Weber de Melo V, Schlegel M, Freise J, Groschup MH, Heckel G, Ulrich RG. Complete genome of a Puumala virus strain from Central Europe. Virus Genes 2014; 50:292-8. [PMID: 25543297 DOI: 10.1007/s11262-014-1157-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 12/12/2014] [Indexed: 12/30/2022]
Abstract
Puumala virus (PUUV) is one of the predominant hantavirus species in Europe causing mild to moderate cases of haemorrhagic fever with renal syndrome. Parts of Lower Saxony in north-western Germany are endemic for PUUV infections. In this study, the complete PUUV genome sequence of a bank vole-derived tissue sample from the 2007 outbreak was determined by a combined primer-walking and RNA ligation strategy. The S, M and L genome segments were 1,828, 3,680 and 6,550 nucleotides in length, respectively. Sliding-window analyses of the nucleotide sequences of all available complete PUUV genomes indicated a non-homogenous distribution of variability with hypervariable regions located at the 3'-ends of the S and M segments. The overall similarity of the coding genome regions to the other PUUV strains ranged between 80.1 and 84.7 % at the level of the nucleotide sequence and between 89.5 and 98.1 % for the deduced amino acid sequences. In comparison to the phylogenetic trees of the complete coding sequences, trees based on partial segments revealed a general drop in phylogenetic support and a lower resolution. The Astrup strain S and M segment sequences showed the highest similarity to sequences of strains from geographically close sites in the Osnabrück Hills region. In conclusion, a primer-walking-mediated strategy resulted in the determination of the first complete nucleotide sequence of a PUUV strain from Central Europe. Different levels of variability along the genome provide the opportunity to choose regions for analyses according to the particular research question, e.g., large-scale phylogenetics or within-host evolution.
Collapse
Affiliation(s)
- Hanan Sheikh Ali
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, OIE Collaborating Centre for Zoonoses in Europe, Institute for Novel and Emerging Infectious Diseases, Südufer 10, 17493, Greifswald - Insel Riems, Germany
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Antigenic properties of N protein of hantavirus. Viruses 2014; 6:3097-109. [PMID: 25123683 PMCID: PMC4147688 DOI: 10.3390/v6083097] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/21/2014] [Accepted: 07/21/2014] [Indexed: 01/08/2023] Open
Abstract
Hantavirus causes two important rodent-borne viral zoonoses, hemorrhagic fever with renal syndrome (HFRS) in Eurasia and hantavirus pulmonary syndrome (HPS) in North and South America. Twenty-four species that represent sero- and genotypes have been registered within the genus Hantavirus by the International Committee on Taxonomy of Viruses (ICTV). Among the viral proteins, nucleocapsid (N) protein possesses an immunodominant antigen. The antigenicitiy of N protein is conserved compared with that of envelope glycoproteins. Therefore, N protein has been used for serological diagnoses and seroepidemiological studies. An understanding of the antigenic properties of N protein is important for the interpretation of results from serological tests using N antigen. N protein consists of about 430 amino acids and possesses various epitopes. The N-terminal quarter of N protein bears linear and immunodominant epitopes. However, a serotype-specific and multimerization-dependent antigenic site was found in the C-terminal half of N protein. In this paper, the structure, function, and antigenicity of N protein are reviewed.
Collapse
|
18
|
Varadaraj A, Mattoscio D, Chiocca S. SUMO Ubc9 enzyme as a viral target. IUBMB Life 2014; 66:27-33. [PMID: 24395713 DOI: 10.1002/iub.1240] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 12/22/2013] [Indexed: 01/20/2023]
Abstract
Viruses alter specific host cell targets to counteract possible defense mechanisms aimed at eliminating infectivity and viral propagation. The SUMO conjugating enzyme Ubc9 functions as a hub for protein sumoylation, whilst also providing an interactive surface for sumoylated proteins through noncovalent interactions. The targeting of Ubc9 by viruses and viral proteins is thus highly beneficial for the disruption of both protein modification and protein-protein interaction mechanisms with which proteins increase their functional repertoire in cells. This review explores some of the clever mechanisms adopted by viruses to deregulate Ubc9, influence effector pathways and positively impact viral persistence consequently.
Collapse
Affiliation(s)
- Archana Varadaraj
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | | | | |
Collapse
|
19
|
Abstract
ABSTRACT: Hantaviruses productively infect endothelial cells in their rodent reservoirs and humans, but the infection only causes disease in humans – hantavirus pulmonary syndrome and hemorrhagic fever with renal syndrome. Despite the enormous progress that has been made in understanding the pathogenesis and immune responses of hantavirus infection, there is a large gap in our molecular-based knowledge of hantaviral proteins in their structures, functions and the mechanisms that facilitate their entry, replication and assembly. Importantly, we know little about the specific viral determinants and viral protein–host interactions that drive differences noted in immune responses between the reservoir and humans. This review discusses our current understanding and future work needed for unraveling the biology of these viruses in their reservoirs and in humans.
Collapse
Affiliation(s)
- Ryan C McAllister
- Department of Pharmacology & Toxicology, University of Louisville, KY 40202, USA
- Center for Predictive Medicine for Biodefense & Emerging Infectious Diseases, KY, USA
| | - Colleen B Jonsson
- Department of Pharmacology & Toxicology, University of Louisville, KY 40202, USA
- Center for Predictive Medicine for Biodefense & Emerging Infectious Diseases, KY, USA
- Department of Microbiology and Immunology, University of Louisville, KY 40202, USA
- Departments of Microbiology & Immunology & Pharmacology & Toxicology, Center for Predictive Medicine for Biodefense & Emerging Infectious Diseases, University of Louisville, Clinical & Translational Research Building, 505 South Hancock Avenue, Louisville, KY 40202, USA
| |
Collapse
|
20
|
Hoenen A, Gillespie L, Morgan G, van der Heide P, Khromykh A, Mackenzie J. The West Nile virus assembly process evades the conserved antiviral mechanism of the interferon-induced MxA protein. Virology 2014; 448:104-16. [DOI: 10.1016/j.virol.2013.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 06/28/2013] [Accepted: 10/03/2013] [Indexed: 02/04/2023]
|
21
|
Niu F, Shaw N, Wang YE, Jiao L, Ding W, Li X, Zhu P, Upur H, Ouyang S, Cheng G, Liu ZJ. Structure of the Leanyer orthobunyavirus nucleoprotein-RNA complex reveals unique architecture for RNA encapsidation. Proc Natl Acad Sci U S A 2013; 110:9054-9. [PMID: 23569220 PMCID: PMC3670306 DOI: 10.1073/pnas.1300035110] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Negative-stranded RNA viruses cover their genome with nucleoprotein (N) to protect it from the human innate immune system. Abrogation of the function of N offers a unique opportunity to combat the spread of the viruses. Here, we describe a unique fold of N from Leanyer virus (LEAV, Orthobunyavirus genus, Bunyaviridae family) in complex with single-stranded RNA refined to 2.78 Å resolution as well as a 2.68 Å resolution structure of LEAV N-ssDNA complex. LEAV N is made up of an N- and a C-terminal lobe, with the RNA binding site located at the junction of these lobes. The LEAV N tetramer binds a 44-nucleotide-long single-stranded RNA chain. Hence, oligomerization of N is essential for encapsidation of the entire genome and is accomplished by using extensions at the N and C terminus. Molecular details of the oligomerization of N are illustrated in the structure where a circular ring-like tertiary assembly of a tetramer of LEAV N is observed tethering the RNA in a positively charged cavity running along the inner edge. Hydrogen bonds between N and the C2 hydroxyl group of ribose sugar explain the specificity of LEAV N for RNA over DNA. In addition, base-specific hydrogen bonds suggest that some regions of RNA bind N more tightly than others. Hinge movements around F20 and V125 assist in the reversal of capsidation during transcription and replication of the virus. Electron microscopic images of the ribonucleoprotein complexes of LEAV N reveal a filamentous assembly similar to those found in phleboviruses.
Collapse
Affiliation(s)
- Fengfeng Niu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Neil Shaw
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| | - Yao E. Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095; and
| | - Lianying Jiao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Ding
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaomin Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ping Zhu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Halmurat Upur
- Department of Drug Analysis, Xinjiang Medical University, Urumqi 830011, Xinjiang, China
| | - Songying Ouyang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095; and
| | - Zhi-Jie Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
22
|
Barletta J, Bartolome A. Immuno-polymerase chain reaction as a unique molecular tool for detection of infectious agents. ACTA ACUST UNITED AC 2013; 1:267-88. [PMID: 23489312 DOI: 10.1517/17530059.1.2.267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Theoretically, the immuno-polymerase chain reaction (IPCR) method is the most sensitive technique for the detection of proteins and gains its uniqueness through the exponential amplification of a signal-generating nucleic acid intermediate attached to a protein target. This method is similar to PCR for the detection of nucleic acid targets, and has now been shown to offer the ability to detect infectious agents where nucleic acids are not present. Although the technical development of IPCR has taken a torturous path down a winding avenue of encouraging advances, the method remains rarely utilized by the scientific community and completely unused as a clinical diagnostic test approved by a national accrediting agency. Although the use of real-time instrumentation has enhanced the performance of IPCR to higher levels of statistical accuracy and reproducibility, as compared with the conventional method, its application remains limited by the high standards required for clinical diagnoses of infectious diseases. This review summarizes experimental data published to date describing the utilization of the IPCR method as it relates to the detection and diagnosis of human infectious disease, and examines the progressive development of this method, as well as the factors impeding its universal application as a clinical diagnostic tool. With further standardization and validation, the IPCR method has the potential to become the most analytically sensitive method available for the detection of target proteins of infectious diseases.
Collapse
Affiliation(s)
- Janet Barletta
- University of Maryland Baltimore, Department of Pathology, 725 W. Lombard St., Baltimore, MD 21201, USA +1 410 706 2215 ; +1 410 706 0155 ;
| | | |
Collapse
|
23
|
Cheng E, Mir MA. Signatures of host mRNA 5' terminus for efficient hantavirus cap snatching. J Virol 2012; 86:10173-85. [PMID: 22787213 PMCID: PMC3446632 DOI: 10.1128/jvi.05560-11] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 07/04/2012] [Indexed: 02/06/2023] Open
Abstract
Hantaviruses, similarly to other negative-strand segmented RNA viruses, initiate the synthesis of translation-competent capped mRNAs by a unique cap-snatching mechanism. Hantavirus nucleocapsid protein (N) binds to host mRNA caps and requires four nucleotides adjacent to the 5' cap for high-affinity binding. N protects the 5' caps of cellular transcripts from degradation by the cellular decapping machinery. The rescued 5' capped mRNA fragments are stored in cellular P bodies by N, which are later efficiently used as primers by the hantaviral RNA-dependent RNA polymerase (RdRp) for transcription initiation. We showed that N also protects the host mRNA caps in P-body-deficient cells. However, the rescued caps were not effectively used by the hantavirus RdRp during transcription initiation, suggesting that caps stored in cellular P bodies by N are preferred for cap snatching. We examined the characteristics of the 5' terminus of a capped test mRNA to delineate the minimum requirements for a capped transcript to serve as an efficient cap donor during hantavirus cap snatching. We showed that hantavirus RdRp preferentially snatches caps from the nonsense mRNAs compared to mRNAs engaged in translation. Hantavirus RdRp preferentially cleaves the cap donor mRNA at a G residue located 14 nucleotides downstream of the 5' cap. The sequence complementarity between the 3' terminus of viral genomic RNA and the nucleotides located in the vicinity of the cleavage site of the cap donor mRNA favors cap snatching. Our results show that hantavirus RdRp snatches caps from viral mRNAs. However, the negligible cap-donating efficiency of wild-type mRNAs in comparison to nonsense mRNAs suggests that viral mRNAs will not be efficiently used for cap snatching during viral infection due to their continuous engagement in protein synthesis. Our results suggest that efficiency of an mRNA to donate caps for viral mRNA synthesis is primarily regulated at the translational level.
Collapse
Affiliation(s)
- Erdong Cheng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | |
Collapse
|
24
|
The Role of the Endothelium in HPS Pathogenesis and Potential Therapeutic Approaches. Adv Virol 2012; 2012:467059. [PMID: 22811711 PMCID: PMC3395186 DOI: 10.1155/2012/467059] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 05/16/2012] [Accepted: 05/18/2012] [Indexed: 02/07/2023] Open
Abstract
American hantaviruses cause a highly lethal acute pulmonary edema termed hantavirus pulmonary syndrome (HPS). Hantaviruses nonlytically infect endothelial cells and cause dramatic changes in barrier functions of the endothelium without disrupting the endothelium. Instead hantaviruses cause changes in the function of infected endothelial cells that normally regulate fluid barrier functions of capillaries. The endothelium of arteries, veins, and lymphatic vessels is unique and central to the function of vast pulmonary capillary beds, which regulate pulmonary fluid accumulation. The endothelium maintains vascular barrier functions through a complex series of redundant receptors and signaling pathways that serve to both permit fluid and immune cell efflux into tissues and restrict tissue edema. Infection of the endothelium provides several mechanisms for hantaviruses to alter capillary permeability but also defines potential therapeutic targets for regulating acute pulmonary edema and HPS disease. Here we discuss interactions of HPS causing hantaviruses with the endothelium, potential endothelial cell-directed permeability mechanisms, and therapeutic targeting of the endothelium as a means of reducing the severity of HPS disease.
Collapse
|
25
|
Hepojoki J, Strandin T, Lankinen H, Vaheri A. Hantavirus structure--molecular interactions behind the scene. J Gen Virol 2012; 93:1631-1644. [PMID: 22622328 DOI: 10.1099/vir.0.042218-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Viruses of the genus Hantavirus, carried and transmitted by rodents and insectivores, are the exception in the vector-borne virus family Bunyaviridae, since viruses of the other genera are transmitted via arthropods. The single-stranded, negative-sense, RNA genome of hantaviruses is trisegmented into small, medium and large (S, M and L) segments. The segments, respectively, encode three structural proteins: nucleocapsid (N) protein, two glycoproteins Gn and Gc and an RNA-dependent RNA-polymerase. The genome segments, encapsidated by the N protein to form ribonucleoproteins, are enclosed inside a lipid envelope that is decorated by spikes composed of Gn and Gc. The virion displays round or pleomorphic morphology with a diameter of roughly 120-160 nm depending on the detection method. This review focuses on the structural components of hantaviruses, their interactions, the mechanisms behind virion assembly and the interactions that maintain virion integrity. We attempt to summarize recent results on the virion structure and to suggest mechanisms on how the assembly is driven. We also compare hantaviruses to other bunyaviruses with known structure.
Collapse
Affiliation(s)
- Jussi Hepojoki
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| | - Tomas Strandin
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| | - Hilkka Lankinen
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| |
Collapse
|
26
|
Abstract
The Bunyaviridae family is comprised of a large number of negative-sense, single-stranded RNA viruses that infect animals, insects, and plants. The tripartite genome of bunyaviruses, encapsidated in the form of individual ribonucleoprotein complexes, encodes four structural proteins, the glycoproteins Gc and Gn, the nucleoprotein N, and the viral polymerase L. Some bunyaviruses also use an ambi-sense strategy to encode the nonstructural proteins NSs and NSm. While some bunyaviruses have a T = 12 icosahedral symmetry, others only have locally ordered capsids, or capsids with no detectable symmetry. Bunyaviruses enter cells through clathrin-mediated endocytosis or phagocytosis. In endosome, viral glycoproteins facilitate membrane fusion at acidic pH, thus allowing bunyaviruses to uncoat and deliver their genomic RNA into host cytoplasm. Bunyaviruses replicate in cytoplasm where the viral polymerase L catalyzes both transcription and replication of the viral genome. While transcription requires a cap primer for initiation and ends at specific termination signals before the 3' end of the template is reached, replication copies the entire template and does not depend on any primer for initiation. This review will discuss some of the most interesting aspects of bunyavirus replication, including L protein/N protein-mediated cap snatching, prime-and-realign for transcription and replication initiation, translation-coupled transcription, sequence/secondary structure-dependent transcription termination, ribonucleoprotein encapsidation, and N protein-mediated initiation of viral protein translation. Recent developments on the structure and functional characterization of the bunyavirus capsid and the RNA synthesis machineries (including both protein L and N) will also be discussed.
Collapse
|
27
|
Estrada DF, Conner M, Jeor SC, Guzman RND. The Structure of the Hantavirus Zinc Finger Domain is Conserved and Represents the Only Natively Folded Region of the Gn Cytoplasmic Tail. Front Microbiol 2011; 2:251. [PMID: 22203819 PMCID: PMC3243910 DOI: 10.3389/fmicb.2011.00251] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 11/27/2011] [Indexed: 11/25/2022] Open
Abstract
Hantaviruses, of the family Bunyaviridae, are present throughout the world and cause a variety of infections ranging from the asymptomatic to mild and severe hemorrhagic fevers. Hantaviruses are enveloped anti-sense RNA viruses that contain three genomic segments that encode for a nucleocapsid protein, two membrane glycoproteins (Gn and Gc), and an RNA polymerase. Recently, the pathogenicity of hantaviruses has been mapped to the carboxyl end of the 150 residue Gn cytoplasmic tail. The Gn tail has also been shown to play a role in binding the ribonucleoprotein (RNP), a step critical for virus assembly. In this study, we use NMR spectroscopy to compare the structure of a Gn tail zinc finger domain of both a pathogenic (Andes) and a non-pathogenic (Prospect Hill) hantavirus. We demonstrate that despite a stark difference in the virulence of both of these viruses, the structure of the Gn core zinc finger domain is largely conserved in both strains. We also use NMR backbone relaxation studies to demonstrate that the regions of the Andes virus Gn tail immediately outside the zinc finger domain, sites known to bind the RNP, are disordered and flexible, thus intimating that the zinc finger domain is the only structured region of the Gn tail. These structural observations provide further insight into the role of the Gn tail during viral assembly as well as its role in pathogenesis.
Collapse
Affiliation(s)
- D Fernando Estrada
- Department of Molecular Biosciences, University of Kansas Lawrence, KS, USA
| | | | | | | |
Collapse
|
28
|
Walter CT, Barr JN. Recent advances in the molecular and cellular biology of bunyaviruses. J Gen Virol 2011; 92:2467-2484. [PMID: 21865443 DOI: 10.1099/vir.0.035105-0] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The family Bunyaviridae of segmented, negative-stranded RNA viruses includes over 350 members that infect a bewildering variety of animals and plants. Many of these bunyaviruses are the causative agents of serious disease in their respective hosts, and are classified as emerging viruses because of their increased incidence in new populations and geographical locations throughout the world. Emerging bunyaviruses, such as Crimean-Congo hemorrhagic fever virus, tomato spotted wilt virus and Rift Valley fever virus, are currently attracting great interest due to migration of their arthropod vectors, a situation possibly linked to climate change. These and other examples of continued emergence suggest that bunyaviruses will probably continue to pose a sustained global threat to agricultural productivity, animal welfare and human health. The threat of emergence is particularly acute in light of the lack of effective preventative or therapeutic treatments for any of these viruses, making their study an important priority. This review presents recent advances in the understanding of the bunyavirus life cycle, including aspects of their molecular, cellular and structural biology. Whilst special emphasis is placed upon the emerging bunyaviruses, we also describe the extensive body of work involving model bunyaviruses, which have been the subject of major contributions to our overall understanding of this important group of viruses.
Collapse
Affiliation(s)
- Cheryl T Walter
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK
| | - John N Barr
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT, UK
| |
Collapse
|
29
|
Strandin T, Hepojoki J, Wang H, Vaheri A, Lankinen H. The cytoplasmic tail of hantavirus Gn glycoprotein interacts with RNA. Virology 2011; 418:12-20. [PMID: 21807393 PMCID: PMC7172371 DOI: 10.1016/j.virol.2011.06.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 06/06/2011] [Accepted: 06/16/2011] [Indexed: 11/15/2022]
Abstract
We recently characterized the interaction between the intraviral domains of envelope glycoproteins (Gn and Gc) and ribonucleoprotein (RNP) of Puumala and Tula hantaviruses (genus Hantavirus, family Bunyaviridae). Herein we report a direct interaction between spike-forming glycoprotein and nucleic acid. We show that the envelope glycoprotein Gn of hantaviruses binds genomic RNA through its cytoplasmic tail (CT). The nucleic acid binding of Gn-CT is unspecific, as demonstrated by interactions with unrelated RNA and with single-stranded DNA. Peptide scan and protein deletions of Gn-CT mapped the nucleic acid binding to regions that overlap with the previously characterized N protein binding sites and demonstrated the carboxyl-terminal part of Gn-CT to be the most potent nucleic acid-binding site. We conclude that recognition of the RNP complex by the Gn-CT could be mediated by interactions with both genomic RNA and the N protein. This would provide the required selectivity for the genome packaging of hantaviruses.
Collapse
Affiliation(s)
- Tomas Strandin
- Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, PO Box 21, FI-00014, University of Helsinki, Finland.
| | | | | | | | | |
Collapse
|
30
|
Cheng E, Haque A, Rimmer MA, Hussein ITM, Sheema S, Little A, Mir MA. Characterization of the Interaction between hantavirus nucleocapsid protein (N) and ribosomal protein S19 (RPS19). J Biol Chem 2011; 286:11814-24. [PMID: 21296889 DOI: 10.1074/jbc.m110.210179] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Hantaviruses, members of the Bunyaviridae family, are negative-stranded emerging RNA viruses and category A pathogens that cause serious illness when transmitted to humans through aerosolized excreta of infected rodent hosts. Hantaviruses have evolved a novel translation initiation mechanism, operated by nucleocapsid protein (N), which preferentially facilitates the translation of viral mRNAs. N binds to the ribosomal protein S19 (RPS19), a structural component of the 40 S ribosomal subunit. In addition, N also binds to both the viral mRNA 5' cap and a highly conserved triplet repeat sequence of the viral mRNA 5' UTR. The simultaneous binding of N at both the terminal cap and the 5' UTR favors ribosome loading on viral transcripts during translation initiation. We characterized the binding between N and RPS19 and demonstrate the role of the N-RPS19 interaction in N-mediated translation initiation mechanism. We show that N specifically binds to RPS19 with high affinity and a binding stoichiometry of 1:1. The N-RPS19 interaction is an enthalpy-driven process. RPS19 undergoes a conformational change after binding to N. Using T7 RNA polymerase, we synthesized the hantavirus S segment mRNA, which matches the transcript generated by the viral RNA-dependent RNA polymerase in cells. We show that the N-RPS19 interaction plays a critical role in the translation of this mRNA both in cells and rabbit reticulocyte lysates. Our results demonstrate that the N-mediated translation initiation mechanism, which lures the host translation machinery for the preferential translation of viral transcripts, primarily depends on the N-RPS19 interaction. We suggest that the N-RPS19 interaction is a novel target to shut down the N-mediated translation strategy and hence virus replication in cells.
Collapse
Affiliation(s)
- Erdong Cheng
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas 66103, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Tacaribe virus (TCRV) belongs to the Arenaviridae family. Its bisegmented negative-stranded RNA genome encodes the nucleoprotein (N), the precursor of the envelope glycoproteins, the polymerase (L), and a RING finger matrix (Z) protein. The 570-amino-acid N protein binds to viral RNA, forming nucleocapsids, which are the template for transcription and replication by the viral polymerase. We have previously shown that the interaction between N and Z is required for assembly of infectious virus-like particles (VLPs) (J. C. Casabona et al., J. Virol. 83:7029-7039, 2009). Here, we examine the functional organization of TCRV N protein. A series of deletions and point mutations were introduced into the N-coding sequence, and the ability of the mutants to sustain heterotypic (N-Z) or homotypic (N-N) interactions was analyzed. We found that N protein displays two functional domains. By using coimmunoprecipitation studies, VLP incorporation assays, and double immunofluorescence staining, the carboxy-terminal region of N was found to be required for N-Z interaction and also necessary for incorporation of N protein into VLPs. Moreover, further analysis of this region showed that the integrity of a putative zinc-finger motif, as well as its amino-flanking sequence (residues 461 to 489), are critical for Z binding and N incorporation into VLPs. In addition, we provide evidence of an essential role of the amino-terminal region of N protein for N-N interaction. In this regard, using reciprocal coimmunoprecipitation analysis, we identified a 28-residue region predicted to form a coiled-coil domain (residues 92 to 119) as a newly recognized molecular determinant of N homotypic interactions.
Collapse
|
32
|
Li J, Zhang Q, Wang T, Li C, Liang M, Li D. Tracking hantavirus nucleocapsid protein using intracellular antibodies. Virol J 2010; 7:339. [PMID: 21092325 PMCID: PMC3002308 DOI: 10.1186/1743-422x-7-339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 11/24/2010] [Indexed: 11/30/2022] Open
Abstract
Background Hantavirus nucleocapsid (N) protein is a multifunctional viral macromolecule involved in multiple stages of the viral replication cycle. The intracellular trafficking of N protein during virus assembly remains unclear. Methods We used N protein-specific intracellular expressed antibodies to track the localization and distribution of Hantaan virus and Seoul virus N protein. The N protein-specific antibody single-chain variable antibody fragments (scFvs), which bind an N-terminal linear epitope (L13F3) and C-terminal conformational domain (H34), were intracellularly expressed in the endoplasmic reticulum (ER) by fusion of the SEKDEL retention signal peptide at the carboxyl terminus, and in the cytoplasm (Cyto) by deletion of the ER membrane target signal peptide. Stable Vero-E6 cell lines expressing intracellular scFvs were either infected with hantavirus or transfected with an N protein expression plasmid; virus replication and N protein intracellular localization were determined. Result N protein co-localized with scFvs in the ER and cytoplasm with or without viral membrane glycoproteins. Hantavirus replication was inhibited in both the scFvs-ER- and scFvs-Cyto-expressing stable cell lines. Conclusion N protein may be expressed in the ER retention signal peptide of KDEL circulating region (ER/cis-Golgi) without the assistance of G protein, and so expression of N protein in both the cytoplasm and within the ER/cis-Golgi plays an important role in virus replication.
Collapse
Affiliation(s)
- Jiandong Li
- State Key Laboratory, Molecular Virology and Genetic Engineering, Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Changping District, Beijing 102206, PR China
| | | | | | | | | | | |
Collapse
|
33
|
Brown BA, Panganiban AT. Identification of a region of hantavirus nucleocapsid protein required for RNA chaperone activity. RNA Biol 2010; 7:830-7. [PMID: 21378500 DOI: 10.4161/rna.7.6.13862] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Sin Nombre hantavirus (SNV) is a New World hantavirus and causes hantavirus cardiopulmonary syndrome. The viral nucleocapsid protein (N) is an RNA chaperone and has multiple functions important in virus replication. The three negative sense RNA segments of hantaviruses form panhandle structures through imperfect hydrogen bonding of the 5' and 3' termini, and the chaperone activity of N can mediate correct panhandle formation. N also functions during transcription and translation initiation and the chaperone activity of N is likely to be involved in aspects of these processes. Using a series of mutations in the N gene we identified a region of N required for chaperone activity. The N-terminal 100 amino acids of N contain a domain that is both necessary and sufficient for RNA chaperone activity. We propose that this region of N may reside in one of two potential states. First, the region may be highly disordered and function in N-mediated RNA chaperone activity. Alternatively, in trimeric form, the region likely becomes ordered and serves in high affinity vRNA panhandle recognition.
Collapse
Affiliation(s)
- Bradley A Brown
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | | |
Collapse
|
34
|
Abstract
Hantaviruses, members of the Bunyaviridae family, are emerging category A pathogens that initiate the translation of their capped mRNAs by a novel mechanism mediated by viral nucleocapsid protein (N). N specifically binds to the mRNA 5' m7G cap and 40S ribosomal subunit, a complex of 18S rRNA and multiple ribosomal proteins. Here, we show that N specifically interacts with the ribosomal protein S19 (RPS19), located at the head region of the 40S subunit. We suggest that this N-RPS19 interaction facilitates ribosome loading on capped mRNAs during N-mediated translation initiation.
Collapse
|
35
|
Katz A, Freiberg AN, Backström V, Schulz AR, Mateos A, Holm L, Pettersson RF, Vaheri A, Flick R, Plyusnin A. Oligomerization of Uukuniemi virus nucleocapsid protein. Virol J 2010; 7:187. [PMID: 20698970 PMCID: PMC2925374 DOI: 10.1186/1743-422x-7-187] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 08/10/2010] [Indexed: 11/26/2022] Open
Abstract
Background Uukuniemi virus (UUKV) belongs to the Phlebovirus genus in the family Bunyaviridae. As a non-pathogenic virus for humans UUKV has served as a safe model bunyavirus in a number of studies addressing fundamental questions such as organization and regulation of viral genes, genome replication, structure and assembly. The present study is focused on the oligomerization of the UUKV nucleocapsid (N) protein, which plays an important role in several steps of virus replication. The aim was to locate the domains involved in the N protein oligomerization and study the process in detail. Results A set of experiments concentrating on the N- and C-termini of the protein was performed, first by completely or partially deleting putative N-N-interaction domains and then by introducing point mutations of amino acid residues. Mutagenesis strategy was based on the computer modeling of secondary and tertiary structure of the N protein. The N protein mutants were studied in chemical cross-linking, immunofluorescence, mammalian two-hybrid, minigenome, and virus-like particle-forming assays. The data showed that the oligomerization ability of UUKV-N protein depends on the presence of intact α-helices on both termini of the N protein molecule and that a specific structure in the N-terminal region plays a crucial role in the N-N interaction(s). This structure is formed by two α-helices, rich in amino acid residues with aromatic (W7, F10, W19, F27, F31) or long aliphatic (I14, I24) side chains. Furthermore, some of the N-terminal mutations (e.g. I14A, I24A, F31A) affected the N protein functionality both in mammalian two-hybrid and minigenome assays. Conclusions UUKV-N protein has ability to form oligomers in chemical cross-linking and mammalian two-hybrid assays. In mutational analysis, some of the introduced single-point mutations abolished the N protein functionality both in mammalian two-hybrid and minigenome assays, suggesting that especially the N-terminal region of the UUKV-N protein is essential for the N-N interaction.
Collapse
Affiliation(s)
- Anna Katz
- Department of Virology, Infection Biology Research Program, Haartman Institute, PO Box 21, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bouloy M, Weber F. Molecular biology of rift valley Fever virus. Open Virol J 2010; 4:8-14. [PMID: 20517489 PMCID: PMC2878978 DOI: 10.2174/1874357901004020008] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 11/25/2009] [Accepted: 11/25/2009] [Indexed: 12/25/2022] Open
Abstract
Rift Valley fever virus (RVFV) causes large outbreaks of acute febrile and often fatal illness among humans and domesticated animals in sub-saharan Africa and the Arabian peninsula. RVFV is a member of the family Bunyaviridae, genus Phlebovirus. Like all members of this large virus family, it contains a three-segmented genome of negative/ambisense strand RNA, packaged into viral nucleocapsid protein, and enveloped by a lipid bilayer containing two viral glycoproteins. During the past years, there was an increased interest in RVFV epidemiology, molecular biology, and virulence mechanisms. Here, we will try to provide an overview over the basic features of this significant pathogen, and review the latest developments in this highly active research field.
Collapse
Affiliation(s)
- Michele Bouloy
- Unite de Genetique Moleculaire des Bunyavirus, Institut Pasteur, Paris, France
| | - Friedemann Weber
- Department of Virology, University of Freiburg, D-79008 Freiburg, Germany
| |
Collapse
|
37
|
Truncated hantavirus nucleocapsid proteins for serotyping Sin Nombre, Andes, and Laguna Negra hantavirus infections in humans and rodents. J Clin Microbiol 2010; 48:1635-42. [PMID: 20335425 DOI: 10.1128/jcm.00072-10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Sin Nombre virus (SNV), Andes virus (ANDV), and Laguna Negra virus (LANV) have been known as the dominant causative agents of hantavirus pulmonary syndrome (HPS). ANDV and LANV, with different patterns of pathogenicity, exist in a sympatric relationship. Moreover, there is documented evidence of person-to-person transmission of ANDV. Therefore, it is important in clinical medicine and epidemiology to know the serotype of a hantavirus causing infection. Truncated SNV, ANDV, and LANV recombinant nucleocapsid proteins (trNs) missing 99 N-terminal amino acids (trN100) were expressed using a baculovirus system, and their applicability for serotyping SNV, ANDV, and LANV infection by the use of enzyme-linked immunosorbent assays (ELISA) was examined. HPS patient sera and natural-reservoir rodent sera infected with SNV, ANDV, and LANV showed the highest optical density (OD) values for homologous trN100 antigens. Since even patient sera with lower IgM and IgG antibody titers were serotyped, the trN100s are therefore considered useful for serotyping with early-acute-phase sera. In contrast, assays testing whole recombinant nucleocapsid protein antigens of SNV, ANDV, and LANV expressed in Escherichia coli detected homologous and heterologous antibodies equally. These results indicated that a screening ELISA using an E. coli-expressed antigen followed by a serotyping ELISA using trN100s is useful for epidemiological surveillance in regions where two or more hantavirus species cocirculate.
Collapse
|
38
|
Ontiveros SJ, Li Q, Jonsson CB. Modulation of apoptosis and immune signaling pathways by the Hantaan virus nucleocapsid protein. Virology 2010; 401:165-78. [PMID: 20227103 DOI: 10.1016/j.virol.2010.02.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 12/12/2009] [Accepted: 02/10/2010] [Indexed: 01/23/2023]
Abstract
Herein, we show a direct relationship between the Hantaan virus (HTNV) nucleocapsid (N) protein and the modulation of apoptosis. We observed an increase in caspase-7 and -8, but not -9 in cells expressing HTNV N protein mutants lacking amino acids 270-330. Similar results were observed for the New World hantavirus, Andes virus. Nuclear factor kappa B (NF-kappaB) was sequestered in the cytoplasm after tumor necrosis factor receptor (TNFR) stimulation in cells expressing HTNV N protein. Further, TNFR stimulated cells expressing HTNV N protein inhibited caspase activation. In contrast, cells expressing N protein truncations lacking the region from amino acids 270-330 were unable to inhibit nuclear import of NF-kappaB and the mutants also triggered caspase activity. These results suggest that the HTNV circumvents host antiviral signaling and apoptotic response mediated by the TNFR pathway through host interactions with the N protein.
Collapse
Affiliation(s)
- Steven J Ontiveros
- Graduate Program in Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
39
|
Mir MA, Sheema S, Haseeb A, Haque A. Hantavirus nucleocapsid protein has distinct m7G cap- and RNA-binding sites. J Biol Chem 2010; 285:11357-68. [PMID: 20164193 DOI: 10.1074/jbc.m110.102459] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Hantaviruses, members of the Bunyaviridae family, are emerging category A pathogens that carry three negative stranded RNA molecules as their genome. Hantavirus nucleocapsid protein (N) is encoded by the smallest S segment genomic RNA (viral RNA). N specifically binds mRNA caps and requires four nucleotides adjacent to the cap for high affinity binding. We show that the N peptide has distinct cap- and RNA-binding sites that independently interact with mRNA cap and viral genomic RNA, respectively. In addition, N can simultaneously bind with both mRNA cap and vRNA. N undergoes distinct conformational changes after binding with either mRNA cap or vRNA or both mRNA cap and vRNA simultaneously. Hantavirus RNA-dependent RNA polymerase (RdRp) uses a capped RNA primer for transcription initiation. The capped RNA primer is generated from host cell mRNA by the cap-snatching mechanism and is supposed to anneal with the 3' terminus of vRNA template during transcription initiation by single G-C base pairing. We show that the capped RNA primer binds at the cap-binding site and induces a conformational change in N. The conformationally altered N with a capped primer loaded at the cap-binding site specifically binds the conserved 3' nine nucleotides of vRNA and assists the bound primer to anneal at the 3' terminus. We suggest that the cap-binding site of N, in conjunction with RdRp, plays a key role during the transcription and replication initiation of vRNA genome.
Collapse
Affiliation(s)
- Mohammad A Mir
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas 66103, USA.
| | | | | | | |
Collapse
|
40
|
Ribeiro D, Borst JW, Goldbach R, Kormelink R. Tomato spotted wilt virus nucleocapsid protein interacts with both viral glycoproteins Gn and Gc in planta. Virology 2008; 383:121-30. [PMID: 18973913 DOI: 10.1016/j.virol.2008.09.028] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 08/29/2008] [Accepted: 09/18/2008] [Indexed: 10/21/2022]
Abstract
Recently, the Tomato Spotted Wilt Virus (TSWV) Gn and Gc glycoproteins were shown to induce the formation of (pseudo-) circular and pleomorphic membrane structures upon transient expression in plant cells. Furthermore, when singly expressed, Gc retains in the ER, while Gn is able to further migrate to the Golgi. Upon co-expression, Gn rescues Gc and co-migrates to the Golgi complex. Here, we have studied the behavior of the glycoproteins in the presence of the viral nucleocapsid (N) protein and in vivo analyzed the occurrence of protein-protein interactions by fluorescence life time imaging microscopy (FLIM). The analysis demonstrated that N co-localizes and interacts with both glycoproteins, with a preference for Gn. Additionally, it is shown that N causes a dramatic change in the distribution of Gc within the ER, from reticular to punctate spots. The observations are discussed in the context of the virus particle formation during the infection process.
Collapse
Affiliation(s)
- Daniela Ribeiro
- Wageningen University, Laboratory of Virology, Binnenhaven 11, 6709 PD Wageningen, The Netherlands
| | | | | | | |
Collapse
|
41
|
Alminaite A, Backström V, Vaheri A, Plyusnin A. Oligomerization of hantaviral nucleocapsid protein: charged residues in the N-terminal coiled-coil domain contribute to intermolecular interactions. J Gen Virol 2008; 89:2167-2174. [PMID: 18753226 DOI: 10.1099/vir.0.2008/004044-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The nucleocapsid (N) protein of hantaviruses (family Bunyaviridae) is the most abundant component of the virion; it encapsidates genomic RNA segments and participates in viral genome transcription and replication, as well as in virus assembly. During RNA encapsidation, the N protein forms intermediate trimers and then oligomers via 'head-to-head, tail-to-tail' interactions. In previous work, using Tula hantavirus (TULV) N protein as a model, it was demonstrated that an intact coiled-coil structure of the N terminus is crucial for the oligomerization capacity of the N protein and that the hydrophobic 'a' residues from the second alpha-helix are especially important. Here, the importance of charged amino acid residues located within the coiled-coil for trimer formation and oligomerization was analysed. To predict the interacting surfaces of the monomers, the previous in silico model of TULV coiled-coils was first upgraded, taking advantage of the recently published crystal structure of the N-terminal coiled-coil of the Sin Nombre virus N protein. The results obtained using a mammalian two-hybrid assay suggested that conserved, charged amino acid residues within the coiled-coil make a substantial contribution to N protein oligomerization. This contribution probably involves (i) the formation of interacting surfaces of the N monomers (residues D35 and D38, located at the tip of the coiled-coil loop, and R63 appear particularly important) and (ii) stabilization of the coiled-coil via intramolecular ionic bridging (with E55 as a key player). It is hypothesized that the tips of the coiled-coils are the first to come into direct contact and thus to initiate tight packing of the three structures.
Collapse
Affiliation(s)
- Agne Alminaite
- Department of Virology, Haartman Institute, PO Box 21, FIN-00014 University of Helsinki, Finland
| | - Vera Backström
- Department of Biochemistry and Pharmacy, Åbo Akademi University, Turku, Finland
| | - Antti Vaheri
- Department of Virology, Haartman Institute, PO Box 21, FIN-00014 University of Helsinki, Finland
| | - Alexander Plyusnin
- Department of Virology, Haartman Institute, PO Box 21, FIN-00014 University of Helsinki, Finland
| |
Collapse
|
42
|
Wang Y, Boudreaux DM, Estrada DF, Egan CW, St Jeor SC, De Guzman RN. NMR structure of the N-terminal coiled coil domain of the Andes hantavirus nucleocapsid protein. J Biol Chem 2008; 283:28297-304. [PMID: 18687679 DOI: 10.1074/jbc.m804869200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The hantaviruses are emerging infectious viruses that in humans can cause a cardiopulmonary syndrome or a hemorrhagic fever with renal syndrome. The nucleocapsid (N) is the most abundant viral protein, and during viral assembly, the N protein forms trimers and packages the viral RNA genome. Here, we report the NMR structure of the N-terminal domain (residues 1-74, called N1-74) of the Andes hantavirus N protein. N1-74 forms two long helices (alpha1 and alpha2) that intertwine into a coiled coil domain. The conserved hydrophobic residues at the helix alpha1-alpha2 interface stabilize the coiled coil; however, there are many conserved surface residues whose function is not known. Site-directed mutagenesis, CD spectroscopy, and immunocytochemistry reveal that a point mutation in the conserved basic surface formed by Arg22 or Lys26 lead to antibody recognition based on the subcellular localization of the N protein. Thus, Arg22 and Lys26 are likely involved in a conformational change or molecular recognition when the N protein is trafficked from the cytoplasm to the Golgi, the site of viral assembly and maturation.
Collapse
Affiliation(s)
- Yu Wang
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045, USA
| | | | | | | | | | | |
Collapse
|
43
|
Tischler ND, Rosemblatt M, Valenzuela PDT. Characterization of cross-reactive and serotype-specific epitopes on the nucleocapsid proteins of hantaviruses. Virus Res 2008; 135:1-9. [PMID: 18342973 DOI: 10.1016/j.virusres.2008.01.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Revised: 01/15/2008] [Accepted: 01/21/2008] [Indexed: 11/19/2022]
Abstract
The hantavirus nucleocapsid (N) protein fulfills several key roles in virus replication and assembly and is the major antigen in humoral immune responses in humans and mice. Here we report on epitopes involved in serotype-specific and cross-reactive recognition of the N proteins of hantaviruses using monoclonal antibodies (mAbs) against the N proteins of Andes virus (ANDV) and Sin Nombre virus (SNV). The mAbs define at least twelve different epitopic patterns which span eight sequences, including amino acids 17-59, 66-78, 79-91, 157-169, 222-234, 244-263, 274-286 and 326-338 on the SNV and ANDV N proteins. Studies on the cross-reactivity of these mAbs with different hantavirus N proteins indicated that epitopes located within amino acids 244-286 are related to serotype specificity. We analyzed further the location of epitopes with available three-dimensional structure information including the N-terminal coiled-coil and derived exposed and hidden residues of these epitopes. The generated recombinant N proteins and the characterized mAbs are functional tools being now available for hantavirus diagnostics and replication studies.
Collapse
|
44
|
Barklis E, Still A, Sabri MI, Hirsch AJ, Nikolich-Zugich J, Brien J, Dhenub TC, Scholz I, Alfadhli A. Sultam thiourea inhibition of West Nile virus. Antimicrob Agents Chemother 2007; 51:2642-5. [PMID: 17452483 PMCID: PMC1913232 DOI: 10.1128/aac.00007-07] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2007] [Revised: 02/26/2007] [Accepted: 04/11/2007] [Indexed: 12/30/2022] Open
Abstract
We have identified sultam thioureas as novel inhibitors of West Nile virus (WNV) replication. One such compound inhibited WNV, with a 50% effective concentration of 0.7 microM, and reduced reporter expression from cells that harbored a WNV-based replicon. Our results demonstrate that sultam thioureas can block a postentry, preassembly step of WNV replication.
Collapse
Affiliation(s)
- Eric Barklis
- Vollum Institute and Department of Molecular Microbiology and Immunology, Oregon Health & Sciences University, Mail Code L220, 3181 SW Sam Jackson Park Road, Portland, OR 97201-3098, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ramanathan HN, Chung DH, Plane SJ, Sztul E, Chu YK, Guttieri MC, McDowell M, Ali G, Jonsson CB. Dynein-dependent transport of the hantaan virus nucleocapsid protein to the endoplasmic reticulum-Golgi intermediate compartment. J Virol 2007; 81:8634-47. [PMID: 17537852 PMCID: PMC1951367 DOI: 10.1128/jvi.00418-07] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In contrast to most negative-stranded RNA viruses, hantaviruses and other viruses in the family Bunyaviridae mature intracellularly, deriving the virion envelope from the endoplasmic reticulum (ER) or Golgi compartment. While it is generally accepted that Old World hantaviruses assemble and bud into the Golgi compartment, some studies with New World hantaviruses have raised the possibility of maturation at the plasma membrane as well. Overall, the steps leading to virion assembly remain largely undetermined for hantaviruses. Because hantaviruses do not have matrix proteins, the nucleocapsid protein (N) has been proposed to play a key role in assembly. Herein, we examine the intracellular trafficking and morphogenesis of the prototype Old World hantavirus, Hantaan virus (HTNV). Using confocal microscopy, we show that N colocalized with the ER-Golgi intermediate compartment (ERGIC) in HTNV-infected Vero E6 cells, not with the ER, Golgi compartment, or early endosomes. Brefeldin A, which effectively disperses the ER, the ERGIC, and Golgi membranes, redistributed N with the ERGIC, implicating membrane association; however, subcellular fractionation experiments showed the majority of N in particulate fractions. Confocal microscopy revealed that N was juxtaposed to and distributed along microtubules and, over time, became surrounded by vimentin cages. To probe cytoskeletal association further, we probed trafficking of N in cells treated with nocodazole and cytochalasin D, which depolymerize microtubules and actin, respectively. We show that nocodazole, but not cytochalasin D, affected the distribution of N and reduced levels of intracellular viral RNA. These results suggested the involvement of microtubules in trafficking of N, whose movement could occur via molecular motors such as dynein. Overexpression of dynamitin, which is associated with dynein-mediated transport, creates a dominant-negative phenotype blocking transport on microtubules. Overexpression of dynamitin reduced N accumulation in the perinuclear region, which further supports microtubule components in N trafficking. The combined results of these experiments support targeting of N to the ERGIC prior to its movement to the Golgi compartment and the requirement of an intact ERGIC for viral replication and, thus, the possibility of virus factories in this region.
Collapse
Affiliation(s)
- Harish N Ramanathan
- Graduate Program in Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Velappan N, Martinez JS, Valero R, Chasteen L, Ponce L, Bondu-Hawkins V, Kelly C, Pavlik P, Hjelle B, Bradbury ARM. Selection and characterization of scFv antibodies against the Sin Nombre hantavirus nucleocapsid protein. J Immunol Methods 2007; 321:60-9. [PMID: 17336997 DOI: 10.1016/j.jim.2007.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Revised: 12/02/2006] [Accepted: 01/07/2007] [Indexed: 11/16/2022]
Abstract
Rodent-borne hantaviruses cause hemorrhagic fever with renal syndrome (HFRS) in the old world and hantavirus cardio-pulmonary syndrome (HCPS) in the new. Most cases of HCPS in North America are caused by Sin Nombre Virus (SNV). Current viral detection technologies depend upon the identification of anti-viral antibodies in patient serum. Detection of viral antigen may facilitate earlier detection of the pathogen. We describe here the characterization of two single-chain Fv antibodies (scFvs), selected from a large naïve phage antibody library, which are capable of identifying the Sin Nombre Virus nucleocapsid protein (SNV-N), with no cross reactivity with the nucleocapsid protein from other hantaviruses. The utility of such selected scFvs was increased by the creation of an scFv-alkaline phosphatase fusion protein which was able to directly detect virally produced material without the need for additional reagents.
Collapse
Affiliation(s)
- Nileena Velappan
- Los Alamos National Laboratory, TA-43, HRL-1, MS M888, Los Alamos NM 87545, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Boudko SP, Kuhn RJ, Rossmann MG. The coiled-coil domain structure of the Sin Nombre virus nucleocapsid protein. J Mol Biol 2006; 366:1538-44. [PMID: 17222867 PMCID: PMC1820746 DOI: 10.1016/j.jmb.2006.12.046] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2006] [Revised: 10/09/2006] [Accepted: 12/15/2006] [Indexed: 11/17/2022]
Abstract
Hantaviruses can cause hemorrhagic fever with a renal syndrome and hantavirus pulmonary syndrome when transmitted to humans. The nucleocapsid protein of hantaviruses encapsidates viral genomic RNA and associates with transcription and replication complexes. Both the amino and carboxy termini of the nucleocapsid protein had been predicted to form trimers prior to the formation of the ribonucleoprotein. Crystal structures of amino-terminal fragments of the nucleocapsid protein showed the formation of intramolecular antiparallel coiled coils, but not intermolecular trimers. Thus, the amino-terminal part of the nucleocapsid protein is probably insufficient to initiate the trimerization of the full-length molecule.
Collapse
Affiliation(s)
| | | | - Michael G. Rossmann
- *Corresponding author: Telephone, 765-494-4911; fax, 765-496-1189; e-mail address,
| |
Collapse
|
48
|
Lindgren L, Lindkvist M, Overby A, Ahlm C, Bucht G, Holmström A. Regions of importance for interaction of puumala virus nucleocapsid subunits. Virus Genes 2006; 33:169-74. [PMID: 16972031 DOI: 10.1007/s11262-005-0045-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Accepted: 11/25/2005] [Indexed: 10/24/2022]
Abstract
Puumala virus (PUUV) is a hantavirus that causes a mild form of hemorrhagic fever with renal syndrome in northern and central Europe, and in large parts of Russia. The nucleocapsid (N) protein encoded by hantaviruses plays an important role in the life-cycle of these viruses, and one important function for the N-protein is to oligomerize, surround and protect the viral RNAs. We have identified amino- and carboxy-terminal regions involved in PUUV N-N interactions, which comprise amino acids 100-120 and 330-405. Our findings strengthen the hypothesis that the amino-terminus of the N-protein of hantaviruses holds a more regulatory function regarding N-N interactions, while conserved residues in the carboxy-terminal region, F335 together with F336 and W392, in concert with Y388 and/or F400 seems to play a more critical role in the PUUV N-N formation. This study provides evidence that the amino-terminal regions involved in the N-N interaction of Puumala virus are similar to those reported for Seoul virus (SEOV) and to some extent Hantaan virus (HTNV), even though the identity between PUUV N and SEOV/HTNV N is markedly lower than between PUUV N and Tula virus (TULV) N or Sin Nombre virus (SNV) N.
Collapse
Affiliation(s)
- Lena Lindgren
- Department of Medical Countermeasures, Division of NBC Defence, Swedish Defence Research Agency, SE-901 82 Umeå, Sweden
| | | | | | | | | | | |
Collapse
|
49
|
Alminaite A, Halttunen V, Kumar V, Vaheri A, Holm L, Plyusnin A. Oligomerization of hantavirus nucleocapsid protein: analysis of the N-terminal coiled-coil domain. J Virol 2006; 80:9073-81. [PMID: 16940519 PMCID: PMC1563903 DOI: 10.1128/jvi.00515-06] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hantaviruses constitute a genus in the family Bunyaviridae. They are enveloped negative-strand RNA viruses with a tripartite genome encoding the nucleocapsid (N) protein, the two surface glycoproteins Gn and Gc, and an RNA-dependent RNA polymerase. The N protein is the most abundant component of the virion; it encapsidates genomic RNA segments forming ribonucleoproteins and participates in genome transcription and replication as well as virus assembly. In the course of RNA encapsidation, N protein forms intermediate trimers via head-to-head and tail-to-tail interactions. We analyzed the amino-terminal trimerization domain (amino acid residues 1 to 77) of Tula hantavirus using computer modeling, mammalian two-hybrid assay, and immunofluorescence assay. The results obtained were consistent with the existence of an antiparallel coiled-coil stabilized by interactions between hydrophobic residues. Residues L44, V51, and L58 were important for the N-N interaction; other residues, e.g., L25 and V32, also made a contribution, albeit a modest one. Our alignments of the N-terminal domain of the hantaviral N proteins suggest the coiled-coil structure, and hence the mode of N-protein oligomerization, is conserved among hantaviruses.
Collapse
Affiliation(s)
- Agne Alminaite
- Department of Virology, Haartman Institute, P.O. Box 21, FIN-00014 University of Helsinki, Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
50
|
Mir MA, Brown B, Hjelle B, Duran WA, Panganiban AT. Hantavirus N protein exhibits genus-specific recognition of the viral RNA panhandle. J Virol 2006; 80:11283-92. [PMID: 16971445 PMCID: PMC1642145 DOI: 10.1128/jvi.00820-06] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
A key genomic characteristic that helps define Hantavirus as a genus of the family Bunyaviridae is the presence of distinctive terminal complementary nucleotides that promote the folding of the viral genomic segments into "panhandle" hairpin structures. The hantavirus nucleocapsid protein (N protein), which is encoded by the smallest of the three negative-sense genomic RNA segments, undergoes in vivo and in vitro trimerization. Trimeric hantavirus N protein specifically recognizes the panhandle structure formed by complementary base sequence of 5' and 3' ends of viral genomic RNA. N protein trimers from the Andes, Puumala, Prospect Hill, Seoul, and Sin Nombre viruses recognize their individual homologous panhandles as well as other hantavirus panhandles with high affinity. In contrast, these hantavirus N proteins bind with markedly reduced affinity to the panhandles from the genera Bunyavirus, Tospovirus, and Phlebovirus or Nairovirus. Interactions between most hantavirus N and heterologous hantavirus viral RNA panhandles are mediated by the nine terminal conserved nucleotides of the panhandle, whereas Sin Nombre virus N requires the first 23 nucleotides for high-affinity binding. Trimeric hantavirus N complexes undergo a prominent conformational change while interacting with panhandles from members of the genus Hantavirus but not while interacting with panhandles from viruses of other genera of the family Bunyaviridae. These data indicate that high-affinity interactions between trimeric N and hantavirus panhandles are conserved within the genus Hantavirus.
Collapse
Affiliation(s)
- M A Mir
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | | | | | |
Collapse
|