Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.916
Filter
Add more filters

Publication year range
1.
Cell ; 186(10): 2208-2218.e15, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37098345

ABSTRACT

Semliki Forest virus (SFV) is an alphavirus that uses the very-low-density lipoprotein receptor (VLDLR) as a receptor during infection of its vertebrate hosts and insect vectors. Herein, we used cryoelectron microscopy to study the structure of SFV in complex with VLDLR. We found that VLDLR binds multiple E1-DIII sites of SFV through its membrane-distal LDLR class A (LA) repeats. Among the LA repeats of the VLDLR, LA3 has the best binding affinity to SFV. The high-resolution structure shows that LA3 binds SFV E1-DIII through a small surface area of 378 Å2, with the main interactions at the interface involving salt bridges. Compared with the binding of single LA3s, consecutive LA repeats around LA3 promote synergistic binding to SFV, during which the LAs undergo a rotation, allowing simultaneous key interactions at multiple E1-DIII sites on the virion and enabling the binding of VLDLRs from divergent host species to SFV.


Subject(s)
Receptors, LDL , Semliki forest virus , Alphavirus/metabolism , Cryoelectron Microscopy , Semliki forest virus/metabolism , Semliki forest virus/ultrastructure , Receptors, LDL/metabolism , Receptors, LDL/ultrastructure , Receptors, Virus/metabolism , Receptors, Virus/ultrastructure
2.
Annu Rev Biochem ; 89: 21-43, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569520

ABSTRACT

My coworkers and I have used animal viruses and their interaction with host cells to investigate cellular processes difficult to study by other means. This approach has allowed us to branch out in many directions, including membrane protein characterization, endocytosis, secretion, protein folding, quality control, and glycobiology. At the same time, our aim has been to employ cell biological approaches to expand the fundamental understanding of animal viruses and their pathogenic lifestyles. We have studied mechanisms of host cell entry and the uncoating of incoming viruses as well as the synthesis, folding, maturation, and intracellular movement of viral proteins and molecular assemblies. I have had the privilege to work in institutions in four different countries. The early years in Finland (the University of Helsinki) were followed by 6 years in Germany (European Molecular Biology Laboratory), 16 years in the United States (Yale School of Medicine), and 16 years in Switzerland (ETH Zurich).


Subject(s)
Calnexin/genetics , Calreticulin/genetics , Host-Pathogen Interactions/genetics , Influenza A virus/genetics , Picornaviridae/genetics , Viral Proteins/genetics , Virology/history , Animals , Calnexin/chemistry , Calnexin/metabolism , Calreticulin/chemistry , Calreticulin/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Endosomes/metabolism , Endosomes/virology , Gene Expression Regulation , History, 20th Century , History, 21st Century , Humans , Influenza A virus/metabolism , Picornaviridae/metabolism , Protein Folding , Semliki forest virus/genetics , Semliki forest virus/metabolism , Vesiculovirus/genetics , Vesiculovirus/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Internalization
3.
Nature ; 602(7897): 475-480, 2022 02.
Article in English | MEDLINE | ID: mdl-34929721

ABSTRACT

Alphaviruses, like many other arthropod-borne viruses, infect vertebrate species and insect vectors separated by hundreds of millions of years of evolutionary history. Entry into evolutionarily divergent host cells can be accomplished by recognition of different cellular receptors in different species, or by binding to receptors that are highly conserved across species. Although multiple alphavirus receptors have been described1-3, most are not shared among vertebrate and invertebrate hosts. Here we identify the very low-density lipoprotein receptor (VLDLR) as a receptor for the prototypic alphavirus Semliki forest virus. We show that the E2 and E1 glycoproteins (E2-E1) of Semliki forest virus, eastern equine encephalitis virus and Sindbis virus interact with the ligand-binding domains (LBDs) of VLDLR and apolipoprotein E receptor 2 (ApoER2), two closely related receptors. Ectopic expression of either protein facilitates cellular attachment, and internalization of virus-like particles, a VLDLR LBD-Fc fusion protein or a ligand-binding antagonist block Semliki forest virus E2-E1-mediated infection of human and mouse neurons in culture. The administration of a VLDLR LBD-Fc fusion protein has protective activity against rapidly fatal Semliki forest virus infection in mouse neonates. We further show that invertebrate receptor orthologues from mosquitoes and worms can serve as functional alphavirus receptors. We propose that the ability of some alphaviruses to infect a wide range of hosts is a result of their engagement of evolutionarily conserved lipoprotein receptors and contributes to their pathogenesis.


Subject(s)
Mosquito Vectors , Semliki forest virus , Animals , LDL-Receptor Related Proteins , Ligands , Mice , Receptors, LDL , Semliki forest virus/metabolism , Sindbis Virus/physiology
4.
Immunity ; 44(6): 1455-69, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27332734

ABSTRACT

Aedes aegypti mosquitoes are responsible for transmitting many medically important viruses such as those that cause Zika and dengue. The inoculation of viruses into mosquito bite sites is an important and common stage of all mosquito-borne virus infections. We show, using Semliki Forest virus and Bunyamwera virus, that these viruses use this inflammatory niche to aid their replication and dissemination in vivo. Mosquito bites were characterized by an edema that retained virus at the inoculation site and an inflammatory influx of neutrophils that coordinated a localized innate immune program that inadvertently facilitated virus infection by encouraging the entry and infection of virus-permissive myeloid cells. Neutrophil depletion and therapeutic blockade of inflammasome activity suppressed inflammation and abrogated the ability of the bite to promote infection. This study identifies facets of mosquito bite inflammation that are important determinants of the subsequent systemic course and clinical outcome of virus infection.


Subject(s)
Arbovirus Infections/immunology , Bunyamwera virus/physiology , Inflammation/immunology , Insect Bites and Stings/immunology , Neutrophils/immunology , Semliki forest virus/physiology , Virus Replication , Animals , Cell Movement , Cells, Cultured , Culicidae/immunology , Humans , Immunity, Innate , Inflammasomes/metabolism , Inflammation/virology , Insect Bites and Stings/virology , Mice , Neutrophils/virology
5.
PLoS Pathog ; 18(1): e1010202, 2022 01.
Article in English | MEDLINE | ID: mdl-34990484

ABSTRACT

The exogenous small interfering RNA (exo-siRNA) pathway is a key antiviral mechanism in the Aedes aegypti mosquito, a widely distributed vector of human-pathogenic arboviruses. This pathway is induced by virus-derived double-stranded RNAs (dsRNA) that are cleaved by the ribonuclease Dicer 2 (Dcr2) into predominantly 21 nucleotide (nt) virus-derived small interfering RNAs (vsiRNAs). These vsiRNAs are used by the effector protein Argonaute 2 within the RNA-induced silencing complex to cleave target viral RNA. Dcr2 contains several domains crucial for its activities, including helicase and RNase III domains. In Drosophila melanogaster Dcr2, the helicase domain has been associated with binding to dsRNA with blunt-ended termini and a processive siRNA production mechanism, while the platform-PAZ domains bind dsRNA with 3' overhangs and subsequent distributive siRNA production. Here we analyzed the contributions of the helicase and RNase III domains in Ae. aegypti Dcr2 to antiviral activity and to the exo-siRNA pathway. Conserved amino acids in the helicase and RNase III domains were identified to investigate Dcr2 antiviral activity in an Ae. aegypti-derived Dcr2 knockout cell line by reporter assays and infection with mosquito-borne Semliki Forest virus (Togaviridae, Alphavirus). Functionally relevant amino acids were found to be conserved in haplotype Dcr2 sequences from field-derived Ae. aegypti across different continents. The helicase and RNase III domains were critical for silencing activity and 21 nt vsiRNA production, with RNase III domain activity alone determined to be insufficient for antiviral activity. Analysis of 21 nt vsiRNA sequences (produced by functional Dcr2) to assess the distribution and phasing along the viral genome revealed diverse yet highly consistent vsiRNA pools, with predominantly short or long sequence overlaps including 19 nt overlaps (the latter representing most likely true Dcr2 cleavage products). Combined with the importance of the Dcr2 helicase domain, this suggests that the majority of 21 nt vsiRNAs originate by processive cleavage. This study sheds new light on Ae. aegypti Dcr2 functions and properties in this important arbovirus vector species.


Subject(s)
Aedes/immunology , Aedes/virology , Alphavirus Infections/immunology , Ribonuclease III/immunology , Aedes/genetics , Animals , DNA Mutational Analysis , Mosquito Vectors/virology , RNA, Small Interfering/immunology , RNA, Viral/immunology , Ribonuclease III/genetics , Semliki forest virus
6.
J Med Virol ; 96(1): e29376, 2024 01.
Article in English | MEDLINE | ID: mdl-38235850

ABSTRACT

Semliki Forest virus (SFV) viral replicon particles (VRPs) have been frequently used in various animal models and clinical trials. Chimeric replicon particles offer different advantages because of their unique biological properties. We here constructed a novel three-plasmid packaging system for chimeric SFV/SIN VRPs. The capsid and envelope of SIN structural proteins were generated using two-helper plasmids separately, and the SFV replicon contained the SFV replicase gene, packaging signal of SIN, subgenomic promoter followed by the exogenous gene, and 3' UTR of SIN. The chimeric VRPs carried luciferase or eGFP as reporter genes. The fluorescence and electron microscopy results revealed that chimeric VRPs were successfully packaged. The yield of the purified chimeric VRPs was approximately 2.5 times that of the SFV VRPs (1.38 × 107 TU/ml vs. 5.41 × 106 TU/ml) (p < 0.01). Furthermore, chimeric VRPs could be stored stably at 4°C for at least 60 days. Animal experiments revealed that mice immunized with chimeric VRPs (luciferase) had stronger luciferase expression than those immunized with equivalent amount of SFV VRPs (luciferase) (p < 0.01), and successfully expressed luciferase for approximately 12 days. Additionally, the chimeric VRPs expressed the RBD of SARS-CoV-2 efficiently and induced robust RBD-specific antibody responses in mice. In conclusion, the chimeric VRPs constructed here met the requirements of a gene delivery tool for vaccine development and cancer therapy.


Subject(s)
Semliki forest virus , Sindbis Virus , Mice , Animals , Semliki forest virus/genetics , Sindbis Virus/genetics , Plasmids/genetics , Replicon , Luciferases/genetics , Genetic Vectors
7.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063100

ABSTRACT

The Semliki Forest virus capsid protein (C) is an RNA binding protein which exhibits both specific and unspecific affinities to single-strand nucleic acids. The putative use of the self-amplifying RNAs (saRNAs) of alphaviruses for biotechnological purpose is one of the main studied strategies concerning RNA-based therapies or immunization. In this work, a recombinant C protein from SFV was expressed and purified from bacteria and used to associate in vitro with a saRNA derived from SFV. Results showed that the purified form of C protein can associate with the saRNA even after high temperature treatment. The C protein was associated with a modified saRNA coding for the green fluorescent protein (GFP) and delivered to murine macrophage cells which expressed the GFP, showing that the saRNA was functional after being associated with the recombinant purified C protein.


Subject(s)
Capsid Proteins , Macrophages , RNA, Viral , Recombinant Proteins , Semliki forest virus , Semliki forest virus/genetics , Animals , Capsid Proteins/genetics , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Mice , Macrophages/metabolism , Macrophages/virology , Recombinant Proteins/genetics , RNA, Viral/genetics , Cell Line , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism
8.
Immunol Cell Biol ; 101(4): 333-344, 2023 04.
Article in English | MEDLINE | ID: mdl-36702633

ABSTRACT

Central nervous system virus infections are a major cause of morbidity and mortality worldwide and a significant global public health concern. As in many tissues, inflammation and immune responses in the brain, despite their protective roles, can also be harmful. Control of brain inflammation is important in many neurological diseases from encephalitis to multiple sclerosis and neurogenerative disease. The suppressors of cytokine signaling (SOCS) proteins are a key mechanism controlling inflammatory and immune responses across all tissues including the brain. Using a mouse model system, we demonstrate that lack of SOCS4 results in changes in the pathogenesis and clinical outcome of a neurotropic virus infection. Relative to wild-type mice, SOCS4-deficient mice showed accelerated clearance of virus from the brain, lower levels of persisting viral RNA in the brain, increased neuroinflammation and more severe neuropathology. We conclude that, in the mouse brain, SOCS4 is a vital regulator of antiviral immunity that mediates the critical balance between immunopathology and virus persistence.


Subject(s)
Cytokines , Encephalitis , Suppressor of Cytokine Signaling Proteins , Animals , Mice , Cytokines/immunology , Encephalitis/immunology , Encephalitis/virology , Immunity , Semliki forest virus , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
9.
PLoS Pathog ; 17(5): e1009603, 2021 05.
Article in English | MEDLINE | ID: mdl-34019569

ABSTRACT

The positive-sense, single-stranded RNA alphaviruses pose a potential epidemic threat. Understanding the complex interactions between the viral and the host cell proteins is crucial for elucidating the mechanisms underlying successful virus replication strategies and for developing specific antiviral interventions. Here we present the first comprehensive protein-protein interaction map between the proteins of Semliki Forest Virus (SFV), a mosquito-borne member of the alphaviruses, and host cell proteins. Among the many identified cellular interactors of SFV proteins, the enrichment of factors involved in translation and nonsense-mediated mRNA decay (NMD) was striking, reflecting the virus' hijacking of the translation machinery and indicating viral countermeasures for escaping NMD by inhibiting NMD at later time points during the infectious cycle. In addition to observing a general inhibition of NMD about 4 hours post infection, we also demonstrate that transient expression of the SFV capsid protein is sufficient to inhibit NMD in cells, suggesting that the massive production of capsid protein during the SFV reproduction cycle is responsible for NMD inhibition.


Subject(s)
Alphavirus Infections/virology , Capsid Proteins/metabolism , Host-Pathogen Interactions , Nonsense Mediated mRNA Decay/genetics , Semliki forest virus/physiology , Capsid Proteins/genetics , HeLa Cells , Humans , Semliki forest virus/genetics , Virus Replication
10.
PLoS Pathog ; 17(5): e1009549, 2021 05.
Article in English | MEDLINE | ID: mdl-33984068

ABSTRACT

The antiviral innate immune response mainly involves type I interferon (IFN) in mammalian cells. The contribution of the RNA silencing machinery remains to be established, but several recent studies indicate that the ribonuclease DICER can generate viral siRNAs in specific conditions. It has also been proposed that type I IFN and RNA silencing could be mutually exclusive antiviral responses. In order to decipher the implication of DICER during infection of human cells with alphaviruses such as the Sindbis virus and Semliki forest virus, we determined its interactome by proteomics analysis. We show that DICER specifically interacts with several double-stranded RNA binding proteins and RNA helicases during viral infection. In particular, proteins such as DHX9, ADAR-1 and the protein kinase RNA-activated (PKR) are enriched with DICER in virus-infected cells. We demonstrate that the helicase domain of DICER is essential for this interaction and that its deletion confers antiviral properties to this protein in an RNAi-independent, PKR-dependent, manner.


Subject(s)
Alphavirus Infections/drug therapy , Antiviral Agents/pharmacology , DEAD-box RNA Helicases/metabolism , Protein Interaction Domains and Motifs/drug effects , Ribonuclease III/metabolism , Semliki forest virus/drug effects , Virus Replication , eIF-2 Kinase/metabolism , Alphavirus Infections/metabolism , Alphavirus Infections/pathology , DEAD-box RNA Helicases/genetics , HEK293 Cells , Humans , Interferon Type I/pharmacology , Ribonuclease III/genetics , eIF-2 Kinase/genetics
11.
Int J Mol Sci ; 24(11)2023 May 26.
Article in English | MEDLINE | ID: mdl-37298266

ABSTRACT

African swine fever virus (ASFV) causes a devastating viral hemorrhagic disease in domestic pigs and Eurasian wild boars, posing a foremost threat to the swine industry and pig farming. The development of an effective vaccine is urgently needed, but has been hampered by the lack of an in-depth, mechanistic understanding of the host immune response to ASFV infection and the induction of protective immunity. In this study, we report that immunization of pigs with Semliki Forest Virus (SFV) replicon-based vaccine candidates expressing ASFV p30, p54, and CD2v, as well as their ubiquitin-fused derivatives, elicits T cell differentiation and expansion, promoting specific T cell and humoral immunity. Due to significant variations in the individual non-inbred pigs in response to the vaccination, a personalized analysis was conducted. Using integrated analysis of differentially expressed genes (DEGs), Venn, KEGG and WGCNA, Toll-like receptor, C-type lectin receptor, IL17 receptor, NOD-like receptor and nucleic acid sensor-mediated signaling pathways were demonstrated to be positively correlated to the antigen-stimulated antibody production and inversely correlated to the IFN-γ secreting cell counts in peripheral blood mononuclear cells (PBMCs). An up-regulation of CIQA, CIQB, CIQC, C4BPA, SOSC3, S100A8 and S100A9, and down-regulation of CTLA4, CXCL2, CXCL8, FOS, RGS1, EGR1 and SNAI1 are general in the innate immune response post-the second boost. This study reveals that pattern recognition receptors TLR4, DHX58/DDX58 and ZBP1, and chemokines CXCL2, CXCL8 and CXCL10 may play important roles in regulating this vaccination-stimulated adaptive immune response.


Subject(s)
African Swine Fever Virus , Swine , Animals , African Swine Fever Virus/genetics , Semliki forest virus , Immunity, Humoral , Leukocytes, Mononuclear , Sus scrofa
12.
J Virol ; 95(14): e0043321, 2021 06 24.
Article in English | MEDLINE | ID: mdl-33952645

ABSTRACT

Negeviruses are a group of insect-specific viruses (ISVs) that have been found in many arthropods. Their presence in important vector species led us to examine their interactions with arboviruses during coinfections. Wild-type negeviruses reduced the replication of several alphaviruses during coinfections in mosquito cells. Negev virus (NEGV) isolates were also used to express green fluorescent protein (GFP) and anti-chikungunya virus (CHIKV) antibody fragments during coinfections with CHIKV. NEGV expressing anti-CHIKV antibody fragments was able to further reduce replication of CHIKV during coinfections, while reductions of CHIKV with NEGV expressing GFP were similar to titers with wild-type NEGV alone. These results are the first to show that negeviruses induce superinfection exclusion of arboviruses and to demonstrate a novel approach to deliver antiviral antibody fragments with paratransgenic ISVs. The ability to inhibit arbovirus replication and express exogenous proteins in mosquito cells makes negeviruses a promising platform for control of arthropod-borne pathogens. IMPORTANCE Negeviruses are a group of insect-specific viruses (ISVs), viruses known to infect only insects. They have been discovered over a wide geographical and species range. Their ability to infect mosquito species that transmit dangerous arboviruses makes negeviruses a candidate for a pathogen control platform. Coinfections of mosquito cells with a negevirus and an alphavirus demonstrated that negeviruses can inhibit the replication of alphaviruses. Additionally, modifying Negev virus (NEGV) to express a fragment of an anti-CHIKV antibody further reduced the replication of CHIKV in coinfected cells. This is the first evidence to demonstrate that negeviruses can inhibit the replication of important arboviruses in mosquito cells. The ability of a modified NEGV to drive the expression of antiviral proteins also highlights a method for negeviruses to target specific pathogens and limit the incidence of vector-borne diseases.


Subject(s)
Alphavirus/physiology , Insect Viruses/physiology , Virus Replication , Aedes/virology , Animals , Cells, Cultured , Chikungunya virus/physiology , Chlorocebus aethiops , Culex/virology , O'nyong-nyong Virus/physiology , Semliki forest virus/physiology , Vero Cells
13.
J Virol ; 95(20): e0097321, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34319778

ABSTRACT

Alphaviruses (family Togaviridae) include both human pathogens such as chikungunya virus (CHIKV) and Sindbis virus (SINV) and model viruses such as Semliki Forest virus (SFV). The alphavirus positive-strand RNA genome is translated into nonstructural (ns) polyprotein(s) that are precursors for four nonstructural proteins (nsPs). The three-dimensional structures of nsP2 and the N-terminal 2/3 of nsP3 reveal that these proteins consist of several domains. Cleavage of the ns-polyprotein is performed by the strictly regulated protease activity of the nsP2 region. Processing results in the formation of a replicase complex that can be considered a network of functional modules. These modules work cooperatively and should perform the same task for each alphavirus. To investigate functional interactions between replicase components, we generated chimeras using the SFV genome as a backbone. The functional modules corresponding to different parts of nsP2 and nsP3 were swapped with their counterparts from CHIKV and SINV. Although some chimeras were nonfunctional, viruses harboring the CHIKV N-terminal domain of nsP2 or any domain of nsP3 were viable. Viruses harboring the protease part of nsP2, the full-length nsP2 of CHIKV, or the nsP3 macrodomain of SINV required adaptive mutations for functionality. Seven mutations that considerably improved the infectivity of the corresponding chimeric genomes affected functionally important hot spots recurrently highlighted in previous alphavirus studies. These data indicate that alphaviruses utilize a rather limited set of strategies to survive and adapt. Furthermore, functional analysis revealed that the disturbance of processing was the main defect resulting from chimeric alterations within the ns-polyprotein. IMPORTANCE Alphaviruses cause debilitating symptoms and have caused massive outbreaks. There are currently no approved antivirals or vaccines for treating these infections. Understanding the functions of alphavirus replicase proteins (nsPs) provides valuable information for both antiviral drug and vaccine development. The nsPs of all alphaviruses consist of similar functional modules; however, to what extent these are independent in functionality and thus interchangeable among homologous viruses is largely unknown. Homologous domain swapping was used to study the functioning of modules from nsP2 and nsP3 of other alphaviruses in the context of Semliki Forest virus. Most of the introduced substitutions resulted in defects in the processing of replicase precursors that were typically compensated by adaptive mutations that mapped to determinants of polyprotein processing. Understanding the principles of virus survival strategies and identifying hot spot mutations that permit virus adaptation highlight a route to the rapid development of attenuated viruses as potential live vaccine candidates.


Subject(s)
Adaptation, Biological/genetics , Alphavirus/genetics , Semliki forest virus/genetics , Cell Line , Chikungunya virus/genetics , Chimera/genetics , Chimera/metabolism , DNA Viruses/genetics , Humans , Mutation/genetics , Polyproteins/metabolism , RNA, Viral/metabolism , Sindbis Virus/genetics , Viral Nonstructural Proteins/genetics , Viral Replication Compartments/metabolism , Virus Replication/genetics
14.
J Virol ; 95(20): e0079021, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34346765

ABSTRACT

Rabies is a fatal zoonosis that causes encephalitis in mammals, and vaccination is the most effective method to control and eliminate rabies. Virus-like vesicles (VLVs), which are characterized as infectious, self-propagating membrane-enveloped particles composed of only Semliki Forest virus (SFV) replicase and vesicular stomatitis virus glycoprotein (VSV-G), have been proven safe and efficient as vaccine candidates. However, previous studies showed that VLVs containing rabies virus glycoprotein (RABV-G) grew at relatively low titers in cells, impeding their potential use as a rabies vaccine. In this study, we constructed novel VLVs by transfection of a mutant SFV RNA replicon encoding RABV-G. We found that these VLVs could self-propagate efficiently in cell culture and could evolve to high titers (approximately 108 focus-forming units [FFU]/ml) by extensive passaging 25 times in BHK-21 cells. Furthermore, we found that the evolved amino acid changes in SFV nonstructural protein 1 (nsP1) at positions 470 and 482 was critical for this high-titer phenotype. Remarkably, VLVs could induce robust type I interferon (IFN) expression in BV2 cells and were highly sensitive to IFN-α. We found that direct inoculation of VLVs into the mouse brain caused reduced body weight loss, mortality, and neuroinflammation compared with the RABV vaccine strain. Finally, it could induce increased generation of germinal center (GC) B cells, plasma cells (PCs), and virus-neutralizing antibodies (VNAs), as well as provide protection against virulent RABV challenge in immunized mice. This study demonstrated that VLVs containing RABV-G could proliferate in cells and were highly evolvable, revealing the feasibility of developing an economic, safe, and efficacious rabies vaccine. IMPORTANCE VLVs have been shown to represent a more versatile and superior vaccine platform. In previous studies, VLVs containing the Semliki Forest virus replicase (SFV nsP1 to nsP4) and rabies virus glycoprotein (RABV-G) grew to relatively low titers in cells. In our study, we not only succeeded in generating VLVs that proliferate in cells and stably express RABV-G, but the VLVs that evolved grew to higher titers, reaching 108 FFU/ml. We also found that nucleic acid changes at positions 470 and 482 in nsP1 were vital for this high-titer phenotype. Moreover, the VLVs that evolved in our studies were highly attenuated in mice, induced potent immunity, and protected mice from lethal RABV infection. Collectively, our study showed that high titers of VLVs containing RABV-G were achieved, demonstrating that these VLVs could be an economical, safe, and efficacious rabies vaccine candidate.


Subject(s)
Rabies Vaccines/immunology , Rabies/immunology , Vaccination/methods , Animals , Antibodies, Viral/blood , B-Lymphocytes/immunology , Disease Models, Animal , Female , Genetic Engineering/methods , Germinal Center/immunology , Glycoproteins/genetics , Immunization/methods , Male , Mice , Mice, Inbred ICR , Rabies/metabolism , Rabies Vaccines/metabolism , Rabies Vaccines/pharmacology , Rabies virus/immunology , Semliki forest virus/immunology , Vesiculovirus/genetics , Viral Proteins/genetics
15.
Mol Ther ; 29(2): 611-625, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33160073

ABSTRACT

A first-in-human phase I trial of Vvax001, an alphavirus-based therapeutic cancer vaccine against human papillomavirus (HPV)-induced cancers was performed assessing immunological activity, safety, and tolerability. Vvax001 consists of replication-incompetent Semliki Forest virus replicon particles encoding HPV16-derived antigens E6 and E7. Twelve participants with a history of cervical intraepithelial neoplasia were included. Four cohorts of three participants were treated per dose level, ranging from 5 × 105 to 2.5 × 108 infectious particles per immunization. The participants received three immunizations with a 3-week interval. For immune monitoring, blood was drawn before immunization and 1 week after the second and third immunization. Immunization with Vvax001 was safe and well tolerated, with only mild injection site reactions, and resulted in both CD4+ and CD8+ T cell responses against E6 and E7 antigens. Even the lowest dose of 5 × 105 infectious particles elicited E6/E7-specific interferon (IFN)-γ responses in all three participants in this cohort. Overall, immunization resulted in positive vaccine-induced immune responses in 12 of 12 participants in one or more assays performed. In conclusion, Vvax001 was safe and induced immune responses in all participants. These data strongly support further clinical evaluation of Vvax001 as a therapeutic vaccine in patients with HPV-related malignancies.


Subject(s)
Cancer Vaccines/immunology , Genetic Vectors/genetics , Neoplasms/etiology , Neoplasms/therapy , Papillomavirus Infections/complications , Papillomavirus Vaccines/immunology , Semliki forest virus/genetics , Alphapapillomavirus/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Genetic Vectors/administration & dosage , Humans , Immunization , Neoplasms/prevention & control , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/genetics , Repressor Proteins/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome , Vaccination
16.
J Virol ; 94(3)2020 01 17.
Article in English | MEDLINE | ID: mdl-31694940

ABSTRACT

RNA interference (RNAi) is a conserved antiviral immune defense in eukaryotes, and numerous viruses have been found to encode viral suppressors of RNAi (VSRs) to counteract antiviral RNAi. Alphaviruses are a large group of positive-stranded RNA viruses that maintain their transmission and life cycles in both mosquitoes and mammals. However, there is little knowledge about how alphaviruses antagonize RNAi in both host organisms. In this study, we identified that Semliki Forest virus (SFV) capsid protein can efficiently suppress RNAi in both insect and mammalian cells by sequestrating double-stranded RNA and small interfering RNA. More importantly, when the VSR activity of SFV capsid was inactivated by reverse genetics, the resulting VSR-deficient SFV mutant showed severe replication defects in mammalian cells, which could be rescued by blocking the RNAi pathway. Besides, capsid protein of Sindbis virus also inhibited RNAi in cells. Together, our findings show that SFV uses capsid protein as VSR to antagonize RNAi in infected mammalian cells, and this mechanism is probably used by other alphaviruses, which shed new light on the knowledge of SFV and alphavirus.IMPORTANCE Alphaviruses are a genus of positive-stranded RNA viruses and include numerous important human pathogens, such as Chikungunya virus, Ross River virus, Western equine encephalitis virus, etc., which create the emerging and reemerging public health threat worldwide. RNA interference (RNAi) is one of the most important antiviral mechanisms in plants and insects. Accumulating evidence has provided strong support for the existence of antiviral RNAi in mammals. In response to antiviral RNAi, viruses have evolved to encode viral suppressors of RNAi (VSRs) to antagonize the RNAi pathway. It is unclear whether alphaviruses encode VSRs that can suppress antiviral RNAi during their infection in mammals. In this study, we first uncovered that capsid protein encoded by Semliki Forest virus (SFV), a prototypic alphavirus, had a potent VSR activity that can antagonize antiviral RNAi in the context of SFV infection in mammalian cells, and this mechanism is probably used by other alphaviruses.


Subject(s)
Capsid Proteins/genetics , Capsid Proteins/metabolism , RNA Interference/physiology , Semliki forest virus/genetics , Semliki forest virus/metabolism , Animals , Capsid , Cell Line , Chikungunya virus/physiology , Drosophila , Encephalitis Virus, Western Equine/physiology , HEK293 Cells , Humans , RNA, Small Interfering , RNA, Viral , Sindbis Virus/physiology , Virion , Virus Replication
17.
PLoS Pathog ; 15(6): e1007842, 2019 06.
Article in English | MEDLINE | ID: mdl-31199850

ABSTRACT

G3BP-1 and -2 (hereafter referred to as G3BP) are multifunctional RNA-binding proteins involved in stress granule (SG) assembly. Viruses from diverse families target G3BP for recruitment to replication or transcription complexes in order to block SG assembly but also to acquire pro-viral effects via other unknown functions of G3BP. The Old World alphaviruses, including Semliki Forest virus (SFV) and chikungunya virus (CHIKV) recruit G3BP into viral replication complexes, via an interaction between FGDF motifs in the C-terminus of the viral non-structural protein 3 (nsP3) and the NTF2-like domain of G3BP. To study potential proviral roles of G3BP, we used human osteosarcoma (U2OS) cell lines lacking endogenous G3BP generated using CRISPR-Cas9 and reconstituted with a panel of G3BP1 mutants and truncation variants. While SFV replicated with varying efficiency in all cell lines, CHIKV could only replicate in cells expressing G3BP1 variants containing both the NTF2-like and the RGG domains. The ability of SFV to replicate in the absence of G3BP allowed us to study effects of different domains of the protein. We used immunoprecipitation to demonstrate that that both NTF2-like and RGG domains are necessary for the formation a complex between nsP3, G3BP1 and the 40S ribosomal subunit. Electron microscopy of SFV-infected cells revealed that formation of nsP3:G3BP1 complexes via the NTF2-like domain was necessary for clustering of cytopathic vacuoles (CPVs) and that the presence of the RGG domain was necessary for accumulation of electron dense material containing G3BP1 and nsP3 surrounding the CPV clusters. Clustered CPVs also exhibited localised high levels of translation of viral mRNAs as detected by ribopuromycylation staining. These data confirm that G3BP is a ribosomal binding protein and reveal that alphaviral nsP3 uses G3BP to concentrate viral replication complexes and to recruit the translation initiation machinery, promoting the efficient translation of viral mRNAs.


Subject(s)
Carrier Proteins/metabolism , Chikungunya Fever/metabolism , Chikungunya virus/physiology , DNA Helicases/metabolism , Peptide Chain Initiation, Translational , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Semliki forest virus/physiology , Virus Replication , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Chikungunya Fever/genetics , Chikungunya Fever/pathology , Cricetinae , DNA Helicases/genetics , Humans , Poly-ADP-Ribose Binding Proteins/genetics , Protein Domains , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins , Ribosome Subunits, Small, Eukaryotic/genetics , Ribosome Subunits, Small, Eukaryotic/metabolism
18.
Arch Virol ; 166(4): 1015-1033, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33582855

ABSTRACT

Multiple sclerosis (MS) is a common inflammatory demyelinating disease of the central nervous system. Although the etiology of MS is unknown, genetics and environmental factors, such as infections, play a role. Viral infections of mice have been used as model systems to study this demyelinating disease of humans. Three viruses that have long been studied in this capacity are Theiler's murine encephalomyelitis virus, mouse hepatitis virus, and Semliki Forest virus. This review describes the viruses themselves, the infection process, the disease caused by infection and its accompanying pathology, and the model systems and their usefulness in studying MS.


Subject(s)
Disease Models, Animal , Multiple Sclerosis/pathology , Multiple Sclerosis/virology , RNA Virus Infections/pathology , RNA Virus Infections/virology , Animals , Central Nervous System/pathology , Central Nervous System/physiology , Central Nervous System/virology , Humans , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Murine hepatitis virus/pathogenicity , Murine hepatitis virus/physiology , RNA Virus Infections/immunology , RNA Virus Infections/physiopathology , Semliki forest virus/pathogenicity , Semliki forest virus/physiology , Theilovirus/pathogenicity , Theilovirus/physiology
19.
Mol Ther ; 28(1): 119-128, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31624015

ABSTRACT

Here, we present a potent RNA vaccine approach based on a novel bipartite vector system using trans-amplifying RNA (taRNA). The vector cassette encoding the vaccine antigen originates from an alphaviral self-amplifying RNA (saRNA), from which the replicase was deleted to form a transreplicon. Replicase activity is provided in trans by a second molecule, either by a standard saRNA or an optimized non-replicating mRNA (nrRNA). The latter delivered 10- to 100-fold higher transreplicon expression than the former. Moreover, expression driven by the nrRNA-encoded replicase in the taRNA system was as efficient as in a conventional monopartite saRNA system. We show that the superiority of nrRNA- over saRNA-encoded replicase to drive expression of the transreplicon is most likely attributable to its higher translational efficiency and lack of interference with cellular translation. Testing the novel taRNA system in mice, we observed that doses of influenza hemagglutinin antigen-encoding RNA as low as 50 ng were sufficient to induce neutralizing antibodies and mount a protective immune response against live virus challenge. These findings, together with a favorable safety profile, a simpler production process, and the universal applicability associated with this bipartite vector system, warrant further exploration of taRNA.


Subject(s)
Immunogenicity, Vaccine , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/metabolism , Orthomyxoviridae Infections/prevention & control , RNA, Viral/genetics , Semliki forest virus/genetics , Vaccination , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cricetinae , Dogs , Female , Genetic Vectors , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza Vaccines/immunology , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology , Viral Replicase Complex Proteins/genetics
20.
Article in English | MEDLINE | ID: mdl-31964798

ABSTRACT

Alphaviruses are arthropod-borne, positive-stranded RNA viruses capable of causing severe disease with high morbidity. Chikungunya virus (CHIKV) is an alphavirus that causes a febrile illness which can progress into chronic arthralgia. The current lack of vaccines and specific treatment for CHIKV infection underscores the need to develop new therapeutic interventions. To discover new antiviral agents, we performed a compound screen in cell culture-based infection models and identified two carbocyclic adenosine analogues, 6'-ß-fluoro-homoaristeromycin (FHA) and 6'-fluoro-homoneplanocin A (FHNA), that displayed potent activity against CHIKV and Semliki Forest virus (SFV) with 50% effective concentrations in the nanomolar range at nontoxic concentrations. The compounds, designed as inhibitors of the host enzyme S-adenosylhomocysteine (SAH) hydrolase, impeded postentry steps in CHIKV and SFV replication. Selection of FHNA-resistant mutants and reverse genetics studies demonstrated that the combination of mutations G230R and K299E in CHIKV nonstructural protein 1 (nsP1) conferred resistance to the compounds. Enzymatic assays with purified wild-type (wt) SFV nsP1 suggested that an oxidized (3'-keto) form, rather than FHNA itself, directly inhibited the MTase activity, while a mutant protein with the K231R and K299E substitutions was insensitive to the compound. Both wt nsP1 and the resistant mutant were equally sensitive to the inhibitory effect of SAH. Our combined data suggest that FHA and FHNA inhibit CHIKV and SFV replication by directly targeting the MTase activity of nsP1, rather than through an indirect effect on host SAH hydrolase. The high potency and selectivity of these novel alphavirus mRNA capping inhibitors warrant further preclinical investigation of these compounds.


Subject(s)
Adenosine/analogs & derivatives , Antiviral Agents/pharmacology , Chikungunya virus/drug effects , Chikungunya virus/physiology , Adenosine/pharmacology , Animals , Chikungunya virus/pathogenicity , Chlorocebus aethiops , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Guanosine Monophosphate/metabolism , Mutation , Phosphorus Radioisotopes , Semliki forest virus/drug effects , Vero Cells , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL