Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 211
1.
mBio ; 11(6)2020 12 01.
Article En | MEDLINE | ID: mdl-33262258

Alphaviruses are positive-sense RNA viruses that utilize a 5' cap structure to facilitate translation of viral proteins and to protect the viral RNA genome. Nonetheless, significant quantities of viral genomic RNAs that lack a canonical 5' cap structure are produced during alphaviral replication and packaged into viral particles. However, the role/impact of the noncapped genomic RNA (ncgRNA) during alphaviral infection in vivo has yet to be characterized. To determine the importance of the ncgRNA in vivo, the previously described D355A and N376A nsP1 mutations, which increase or decrease nsP1 capping activity, respectively, were incorporated into the neurovirulent AR86 strain of Sindbis virus to enable characterization of the impact of altered capping efficiency in a murine model of infection. Mice infected with the N376A nsP1 mutant exhibited slightly decreased rates of mortality and delayed weight loss and neurological symptoms, although levels of inflammation in the brain were similar to those of wild-type infection. Although the D355A mutation resulted in decreased antiviral gene expression and increased resistance to interferon in vitro, mice infected with the D355A mutant showed significantly reduced mortality and morbidity compared to mice infected with wild-type virus. Interestingly, expression of proinflammatory cytokines was found to be significantly decreased in mice infected with the D355A mutant, suggesting that capping efficiency and the production of ncgRNA are vital to eliciting pathogenic levels of inflammation. Collectively, these data indicate that the ncgRNA have important roles during alphaviral infection and suggest a novel mechanism by which noncapped viral RNAs aid in viral pathogenesis.IMPORTANCE Mosquito-transmitted alphaviruses have been the cause of widespread outbreaks of disease that can range from mild illness to lethal encephalitis or severe polyarthritis. There are currently no safe and effective vaccines or therapeutics with which to prevent or treat alphaviral disease, highlighting the need to better understand alphaviral pathogenesis to develop novel antiviral strategies. This report reveals production of noncapped genomic RNAs (ncgRNAs) to be a novel determinant of alphaviral virulence and offers insight into the importance of inflammation to pathogenesis. Taken together, the findings reported here suggest that the ncgRNAs contribute to alphaviral pathogenesis through the sensing of the ncgRNAs during alphaviral infection and are necessary for the development of severe disease.


Alphavirus Infections/virology , Gene Expression Regulation, Viral , Genome, Viral , RNA, Viral , Sindbis Virus/genetics , Alphavirus Infections/genetics , Alphavirus Infections/metabolism , Animals , Brain/metabolism , Brain/virology , Cell Line , Cell Survival , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Inflammation Mediators , Interferon Type I/metabolism , Mice , Neurons/virology , RNA Caps , Sindbis Virus/pathogenicity , Virulence , Virus Replication
2.
PLoS One ; 15(11): e0241592, 2020.
Article En | MEDLINE | ID: mdl-33180795

Superinfection exclusion (SIE) is a process by which a virally infected cell is protected from subsequent infection by the same or a closely related virus. By preventing cell coinfection, SIE favors preservation of genome integrity of a viral strain and limits its recombination potential with other viral genomes, thereby impacting viral evolution. Although described in virtually all viral families, the precise step(s) impacted by SIE during the viral life cycle have not been systematically explored. Here, we describe for the first time SIE triggered by chikungunya virus (CHIKV), an alphavirus of public health importance. Using single-cell technologies, we demonstrate that CHIKV excludes subsequent infection with: CHIKV; Sindbis virus, a related alphavirus; and influenza A, an unrelated RNA virus. We further demonstrate that SIE does not depend on the action of type I interferon, nor does it rely on host cell transcription. Moreover, exclusion is not mediated by the action of a single CHIKV protein; in particular, we observed no role for non-structural protein 2 (nsP2), making CHIKV unique among characterized alphaviruses. By stepping through the viral life cycle, we show that CHIKV exclusion occurs at the level of replication, but does not directly influence virus binding, nor viral structural protein translation. In sum, we characterized co-infection during CHIKV replication, which likely influences the rate of viral diversification and evolution.


Chikungunya Fever/virology , Chikungunya virus/physiology , Superinfection/virology , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Cells, Cultured , Chikungunya virus/genetics , Chikungunya virus/pathogenicity , Chlorocebus aethiops , Genome, Viral , Influenza A virus/pathogenicity , Mice , Sindbis Virus/pathogenicity , Vero Cells , Viral Nonstructural Proteins/genetics
3.
PLoS One ; 15(8): e0238254, 2020.
Article En | MEDLINE | ID: mdl-32841293

The identification of host / pathogen interactions is essential to both understanding the molecular biology of infection and developing rational intervention strategies to overcome disease. Alphaviruses, such as Sindbis virus, Chikungunya virus, and Venezuelan Equine Encephalitis virus are medically relevant positive-sense RNA viruses. As such, they must interface with the host machinery to complete their infectious lifecycles. Nonetheless, exhaustive RNA:Protein interaction discovery approaches have not been reported for any alphavirus species. Thus, the breadth and evolutionary conservation of host interactions on alphaviral RNA function remains a critical gap in the field. Herein we describe the application of the Cross-Link Assisted mRNP Purification (CLAMP) strategy to identify conserved alphaviral interactions. Through comparative analyses, conserved alphaviral host / pathogen interactions were identified. Approximately 100 unique host proteins were identified as a result of these analyses. Ontological assessments reveal enriched Molecular Functions and Biological Processes relevant to alphaviral infection. Specifically, as anticipated, Poly(A) RNA Binding proteins are significantly enriched in virus specific CLAMP data sets. Moreover, host proteins involved in the regulation of mRNA stability, proteasome mediated degradation, and a number of 14-3-3 proteins were identified. Importantly, these data expand the understanding of alphaviral host / pathogen interactions by identifying conserved interactants.


Alphavirus/genetics , Alphavirus/pathogenicity , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Alphavirus/physiology , Animals , Cell Line , Chikungunya virus/genetics , Chikungunya virus/pathogenicity , Chikungunya virus/physiology , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalitis Virus, Venezuelan Equine/physiology , Evolution, Molecular , HEK293 Cells , Humans , Protein Interaction Maps , Ribonucleoproteins/genetics , Ribonucleoproteins/isolation & purification , Ribonucleoproteins/metabolism , Sindbis Virus/genetics , Sindbis Virus/pathogenicity , Sindbis Virus/physiology , Species Specificity
4.
mBio ; 11(1)2020 02 11.
Article En | MEDLINE | ID: mdl-32047134

Macrodomain (MD), a highly conserved protein fold present in a subset of plus-strand RNA viruses, binds to and hydrolyzes ADP-ribose (ADPr) from ADP-ribosylated proteins. ADPr-binding by the alphavirus nonstructural protein 3 (nsP3) MD is necessary for the initiation of virus replication in neural cells, whereas hydrolase activity facilitates replication complex amplification. To determine the importance of these activities for pathogenesis of alphavirus encephalomyelitis, mutations were introduced into the nsP3 MD of Sindbis virus (SINV), and the effects on ADPr binding and hydrolase activities, virus replication, immune responses, and disease were assessed. Elimination of ADPr-binding and hydrolase activities (G32E) severely impaired in vitro replication of SINV in neural cells and in vivo replication in the central nervous systems of 2-week-old mice with reversion to wild type (WT) (G) or selection of a less compromising change (S) during replication. SINVs with decreased binding and hydrolase activities (G32S and G32A) or with hydrolase deficiency combined with better ADPr-binding (Y114A) were less virulent than WT virus. Compared to the WT, the G32S virus replicated less well in both the brain and spinal cord, induced similar innate responses, and caused less severe disease with full recovery of survivors, whereas the Y114A virus replicated well, induced higher expression of interferon-stimulated and NF-κB-induced genes, and was cleared more slowly from the spinal cord with persistent paralysis in survivors. Therefore, MD function was important for neural cell replication both in vitro and in vivo and determined the outcome from alphavirus encephalomyelitis in mice.IMPORTANCE Viral encephalomyelitis is an important cause of long-term disability, as well as acute fatal disease. Identifying viral determinants of outcome helps in assessing disease severity and developing new treatments. Mosquito-borne alphaviruses infect neurons and cause fatal disease in mice. The highly conserved macrodomain of nonstructural protein 3 binds and can remove ADP-ribose (ADPr) from ADP-ribosylated proteins. To determine the importance of these functions for virulence, recombinant mutant viruses were produced. If macrodomain mutations eliminated ADPr-binding or hydrolase activity, viruses did not grow. If the binding and hydrolase activities were impaired, the viruses grew less well than the wild-type virus, induced similar innate responses, and caused less severe disease, and most of the infected mice recovered. If binding was improved, but hydrolase activity was decreased, the virus replicated well and induced greater innate responses than did the WT, but clearance from the nervous system was impaired, and mice remained paralyzed. Therefore, macrodomain function determined the outcome of alphavirus encephalomyelitis.


Adenosine Diphosphate Ribose/metabolism , Hydrolases/metabolism , Sindbis Virus/genetics , Sindbis Virus/pathogenicity , Viral Nonstructural Proteins/metabolism , Adenosine Diphosphate Ribose/genetics , Animals , Brain/virology , Cell Line , Encephalomyelitis/virology , Female , Hydrolases/genetics , Mice , Mutation , Neurons/virology , Signal Transduction , Viral Nonstructural Proteins/genetics , Virulence , Virus Replication/genetics
5.
PLoS Pathog ; 15(5): e1007798, 2019 05.
Article En | MEDLINE | ID: mdl-31116799

Cellular antiviral programs encode molecules capable of targeting multiple steps in the virus lifecycle. Zinc-finger antiviral protein (ZAP) is a central and general regulator of antiviral activity that targets pathogen mRNA stability and translation. ZAP is diffusely cytoplasmic, but upon infection ZAP is targeted to particular cytoplasmic structures, termed stress granules (SGs). However, it remains unclear if ZAP's antiviral activity correlates with SG localization, and what molecular cues are required to induce this localization event. Here, we use Sindbis virus (SINV) as a model infection and find that ZAP's localization to SGs can be transient. Sometimes no apparent viral infection follows ZAP SG localization but ZAP SG localization always precedes accumulation of SINV non-structural protein, suggesting virus replication processes trigger SG formation and ZAP recruitment. Data from single-molecule RNA FISH corroborates this finding as the majority of cells with ZAP localization in SGs contain low levels of viral RNA. Furthermore, ZAP recruitment to SGs occurred in ZAP-expressing cells when co-cultured with cells replicating full-length SINV, but not when co-cultured with cells replicating a SINV replicon. ZAP recruitment to SGs is functionally important as a panel of alanine ZAP mutants indicate that the anti-SINV activity is correlated with ZAP's ability to localize to SGs. As ZAP is a central component of the cellular antiviral programs, these data provide further evidence that SGs are an important cytoplasmic antiviral hub. These findings provide insight into how antiviral components are regulated upon virus infection to inhibit virus spread.


Alphavirus Infections/prevention & control , Antiviral Agents/pharmacology , Cytoplasmic Granules/metabolism , RNA-Binding Proteins/pharmacology , Sindbis Virus/pathogenicity , Stress, Physiological , Virus Replication/drug effects , Alphavirus Infections/metabolism , Alphavirus Infections/virology , Antiviral Agents/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/virology , Humans , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/virology , Protein Transport , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Tumor Cells, Cultured
6.
Mol Cell ; 74(1): 196-211.e11, 2019 04 04.
Article En | MEDLINE | ID: mdl-30799147

The compendium of RNA-binding proteins (RBPs) has been greatly expanded by the development of RNA-interactome capture (RIC). However, it remained unknown if the complement of RBPs changes in response to environmental perturbations and whether these rearrangements are important. To answer these questions, we developed "comparative RIC" and applied it to cells challenged with an RNA virus called sindbis (SINV). Over 200 RBPs display differential interaction with RNA upon SINV infection. These alterations are mainly driven by the loss of cellular mRNAs and the emergence of viral RNA. RBPs stimulated by the infection redistribute to viral replication factories and regulate the capacity of the virus to infect. For example, ablation of XRN1 causes cells to be refractory to SINV, while GEMIN5 moonlights as a regulator of SINV gene expression. In summary, RNA availability controls RBP localization and function in SINV-infected cells.


Epithelial Cells/virology , Gene Expression Profiling/methods , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Sindbis Virus/genetics , Transcriptome , Uterine Cervical Neoplasms/virology , 5' Untranslated Regions , Binding Sites , Epithelial Cells/metabolism , Exoribonucleases/genetics , Exoribonucleases/metabolism , Female , Gene Expression Regulation, Viral , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Protein Binding , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins , Sindbis Virus/growth & development , Sindbis Virus/metabolism , Sindbis Virus/pathogenicity , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Virus Replication
7.
mBio ; 9(4)2018 08 21.
Article En | MEDLINE | ID: mdl-30131356

Infection with Ross River virus (RRV) causes debilitating polyarthritis and arthralgia in individuals. Alphaviruses are highly sensitive to type I interferon (IFN). Mutations at the conserved P3 position of the cleavage site between nonstructural protein 1 (nsP1) and nsP2 (1/2 site) modulate type I IFN induction for both RRV and Sindbis virus (SINV). We constructed and characterized RRV-T48A534V, a mutant harboring an A534V substitution in the P1 position of the 1/2 site, and compared it to parental RRV-T48 and to RRV-T48A532V, SINVI538 and SINVT538 harboring different substitutions in the same region. A534V substitution resulted in impaired processing of RRV nonstructural polyprotein and in elevated production of replicase-generated pathogen-associated molecular pattern (PAMP) RNAs that induce expression of type I IFN. Both A532V and A534V substitutions affected synthesis of viral RNAs, though the effects of these closely located mutations were drastically different affecting mostly either the viral negative-strand RNA or genomic and subgenomic RNA levels, respectively. Synthesis of PAMP RNAs was also observed for SINV replicase, and it was increased by I538T substitution. In comparison to RRV-T48, RRV-T48A534V was attenuated in vitro and in vivo Interestingly, when type I IFN-deficient cells and type I IFN receptor-deficient mice were infected with RRV-T48 or RRV-T48A534V, differences between these viruses were no longer apparent. Compared to RRV-T48, RRV-T48A534V infection was associated with increased upregulation of type I IFN signaling proteins. We demonstrate novel mechanisms by which the A534V mutation affect viral nonstructural polyprotein processing that can impact PAMP RNA production, type I IFN induction/sensitivity, and disease.IMPORTANCE This study gives further insight into mechanisms of type I IFN modulation by the medically important alphaviruses Ross River virus (RRV) and Sindbis virus (SINV). By characterizing attenuated RRV mutants, the crucial role of amino acid residues in P1 and P3 positions (the first and third amino acid residues preceding the scissile bond) of the cleavage site between nsP1 and nsP2 regions was highlighted. The study uncovers a unique relationship between alphavirus nonstructural polyprotein processing, RNA replication, production of different types of pathogen-associated molecular pattern (PAMP) RNAs, type I IFN induction, and disease pathogenesis. This study also highlights the importance of the host innate immune response in RRV infections. The viral determinants of type I IFN modulation provide potential drug targets for clinical treatment of alphaviral disease and offer new approaches for rational attenuation of alphaviruses for construction of vaccine candidates.


Interferons/metabolism , Mutant Proteins/immunology , Mutation, Missense , Polyproteins/metabolism , RNA, Viral/immunology , Ross River virus/pathogenicity , Viral Nonstructural Proteins/metabolism , Alphavirus Infections/pathology , Alphavirus Infections/virology , Animals , Antiviral Agents/metabolism , Disease Models, Animal , Mice , Mutant Proteins/genetics , Polyproteins/genetics , RNA, Viral/metabolism , Ross River virus/genetics , Ross River virus/immunology , Sindbis Virus/genetics , Sindbis Virus/immunology , Sindbis Virus/pathogenicity , Viral Nonstructural Proteins/genetics , Virulence
8.
J Virol ; 92(7)2018 04 01.
Article En | MEDLINE | ID: mdl-29321325

Arthropod-borne viruses, such as the members of the genus Alphavirus, are a significant concern to global public health. As obligate intracellular pathogens, RNA viruses must interact with the host cell machinery to establish and complete their life cycles. Despite considerable efforts to define the host-pathogen interactions essential for alphaviral replication, an unbiased and inclusive assessment of alphaviral RNA-protein interactions has not been undertaken. Moreover, the biological and molecular importance of these interactions, in the full context of their molecular function as RNA-binding proteins, has not been fully realized. The data presented here introduce a robust viral RNA-protein discovery method to elucidate the Sindbis virus (SINV) RNA-protein host interface. Cross-link-assisted mRNP purification (CLAMP) assessment revealed an extensive array of host-pathogen interactions centered on the viral RNAs (vRNAs). After prioritization of the host proteins associated with the vRNAs, we identified the site of protein-vRNA interaction by a UV cross-linking and immunoprecipitation sequencing (CLIP-seq) approach and assessed the consequences of the RNA-protein binding event of hnRNP K, hnRNP I, and hnRNP M in regard to viral infection. Here, we demonstrate that mutation of the prioritized hnRNP-vRNA interaction sites effectively disrupts hnRNP-vRNA interaction. Correlating with disrupted hnRNP-vRNA binding, SINV growth kinetics were reduced relative to wild-type parental viral infections in vertebrate and invertebrate tissue culture models of infection. The molecular mechanism leading to reduced viral growth kinetics was found to be dysregulated structural-gene expression. Collectively, this study further defines the scope and importance of the alphavirus host-pathogen vRNA-protein interactions.IMPORTANCE Members of the genus Alphavirus are widely recognized for their potential to cause severe disease. Despite this recognition, there are no antiviral therapeutics, or safe and effective vaccines, currently available to treat alphaviral infection. Alphaviruses utilize the host cell machinery to efficiently establish and complete their life cycle. However, the extent and importance of host-pathogen RNA-protein interactions are woefully undercharacterized. The efforts detailed in this study fill this critical gap, and the significance of this research is 3-fold. First, the data presented here fundamentally expand the scope and understanding of alphavirus host-pathogen interactions. Second, this study identifies the sites of interaction for several prioritized interactions and defines the contribution of the RNA-protein interaction at the molecular level. Finally, these studies build a strategy by which the importance of the given host-pathogen interactions may be assessed in the future, using a mouse model of infection.


Alphavirus Infections/virology , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Host-Pathogen Interactions , RNA, Viral/metabolism , Sindbis Virus/pathogenicity , Virus Replication , Alphavirus Infections/metabolism , Cells, Cultured , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , RNA, Viral/genetics , Sindbis Virus/genetics , Virus Assembly
9.
Nucleic Acids Res ; 46(7): 3657-3670, 2018 04 20.
Article En | MEDLINE | ID: mdl-29361131

Alphaviruses are mosquito-borne pathogens that cause human diseases ranging from debilitating arthritis to lethal encephalitis. Studies with Sindbis virus (SINV), which causes fever, rash, and arthralgia in humans, and Venezuelan equine encephalitis virus (VEEV), which causes encephalitis, have identified RNA structural elements that play key roles in replication and pathogenesis. However, a complete genomic structural profile has not been established for these viruses. We used the structural probing technique SHAPE-MaP to identify structured elements within the SINV and VEEV genomes. Our SHAPE-directed structural models recapitulate known RNA structures, while also identifying novel structural elements, including a new functional element in the nsP1 region of SINV whose disruption causes a defect in infectivity. Although RNA structural elements are important for multiple aspects of alphavirus biology, we found the majority of RNA structures were not conserved between SINV and VEEV. Our data suggest that alphavirus RNA genomes are highly divergent structurally despite similar genomic architecture and sequence conservation; still, RNA structural elements are critical to the viral life cycle. These findings reframe traditional assumptions about RNA structure and evolution: rather than structures being conserved, alphaviruses frequently evolve new structures that may shape interactions with host immune systems or co-evolve with viral proteins.


Encephalitis Virus, Venezuelan Equine/genetics , RNA/genetics , Sindbis Virus/genetics , Virus Replication/genetics , Alphavirus/chemistry , Alphavirus/genetics , Alphavirus/pathogenicity , Animals , Encephalitis/genetics , Encephalitis/virology , Encephalitis Virus, Venezuelan Equine/chemistry , Encephalitis Virus, Venezuelan Equine/pathogenicity , Genome, Viral/genetics , Horses/virology , Humans , Nucleic Acid Conformation , RNA/chemistry , Sindbis Virus/chemistry , Sindbis Virus/pathogenicity
10.
J Virol ; 92(6)2018 03 15.
Article En | MEDLINE | ID: mdl-29263262

Alphaviruses are an important cause of mosquito-borne outbreaks of arthritis, rash, and encephalomyelitis. Previous studies in mice with a virulent strain (neuroadapted SINV [NSV]) of the alphavirus Sindbis virus (SINV) identified a role for Th17 cells and regulation by interleukin-10 (IL-10) in the pathogenesis of fatal encephalomyelitis (K. A. Kulcsar, V. K. Baxter, I. P. Greene, and D. E. Griffin, Proc Natl Acad Sci U S A 111:16053-16058, 2014, https://doi.org/10.1073/pnas.1418966111). To determine the role of virus virulence in generation of immune responses, we analyzed the modulatory effects of IL-10 on disease severity, virus clearance, and the CD4+ T cell response to infection with a recombinant strain of SINV of intermediate virulence (TE12). The absence of IL-10 during TE12 infection led to longer morbidity, more weight loss, higher mortality, and slower viral clearance than in wild-type mice. More severe disease and impaired virus clearance in IL-10-/- mice were associated with more Th1 cells, fewer Th2 cells, innate lymphoid type 2 cells, regulatory cells, and B cells, and delayed production of antiviral antibody in the central nervous system (CNS) without an effect on Th17 cells. Therefore, IL-10 deficiency led to more severe disease in TE12-infected mice by increasing Th1 cells and by hampering development of the local B cell responses necessary for rapid production of antiviral antibody and virus clearance from the CNS. In addition, the shift from Th17 to Th1 responses with decreased virus virulence indicates that the effects of IL-10 deficiency on immunopathologic responses in the CNS during alphavirus infection are influenced by virus strain.IMPORTANCE Alphaviruses cause mosquito-borne outbreaks of encephalomyelitis, but determinants of outcome are incompletely understood. We analyzed the effects of the anti-inflammatory cytokine IL-10 on disease severity and virus clearance after infection with an alphavirus strain of intermediate virulence. The absence of IL-10 led to longer illness, more weight loss, more death, and slower viral clearance than in mice that produced IL-10. IL-10 influenced development of disease-causing T cells and entry into the brain of B cells producing antiviral antibody. The Th1 pathogenic cell subtype that developed in IL-10-deficient mice infected with a less virulent virus was distinct from the Th17 subtype that developed in response to a more virulent virus, indicating a role for virus strain in determining the immune response. Slow production of antibody in the nervous system led to delayed virus clearance. Therefore, both the virus strain and the host response to infection are important determinants of outcome.


Alphavirus Infections/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis/immunology , Interleukin-10/immunology , Sindbis Virus/immunology , Alphavirus Infections/genetics , Alphavirus Infections/pathology , Animals , Antibodies, Viral/immunology , Antibody Formation/genetics , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/pathology , Cell Line , Cricetinae , Encephalomyelitis/genetics , Encephalomyelitis/pathology , Interleukin-10/genetics , Mice , Mice, Knockout , Sindbis Virus/genetics , Sindbis Virus/pathogenicity
11.
Sci Rep ; 7(1): 6448, 2017 07 25.
Article En | MEDLINE | ID: mdl-28743868

Recent high annual losses of honey bee colonies are associated with many factors, including RNA virus infections. Honey bee antiviral responses include RNA interference and immune pathway activation, but their relative roles in antiviral defense are not well understood. To better characterize the mechanism(s) of honey bee antiviral defense, bees were infected with a model virus in the presence or absence of dsRNA, a virus associated molecular pattern. Regardless of sequence specificity, dsRNA reduced virus abundance. We utilized next generation sequencing to examine transcriptional responses triggered by virus and dsRNA at three time-points post-infection. Hundreds of genes exhibited differential expression in response to co-treatment of dsRNA and virus. Virus-infected bees had greater expression of genes involved in RNAi, Toll, Imd, and JAK-STAT pathways, but the majority of differentially expressed genes are not well characterized. To confirm the virus limiting role of two genes, including the well-characterized gene, dicer, and a probable uncharacterized cyclin dependent kinase in honey bees, we utilized RNAi to reduce their expression in vivo and determined that virus abundance increased, supporting their involvement in antiviral defense. Together, these results further our understanding of honey bee antiviral defense, particularly the role of a non-sequence specific dsRNA-mediated antiviral pathway.


Bees/physiology , Bees/virology , Host-Pathogen Interactions/physiology , Insect Proteins/genetics , RNA, Double-Stranded/genetics , Animals , Endocytosis/genetics , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Polymerase Chain Reaction/methods , RNA Interference , Reproducibility of Results , Signal Transduction/genetics , Sindbis Virus/pathogenicity , Transcription, Genetic
12.
Cell Host Microbe ; 22(1): 48-60.e5, 2017 Jul 12.
Article En | MEDLINE | ID: mdl-28704652

Asymptomatic infections often proceed undetected, yet can still prime the host to be sensitive to secondary environmental stress. While the mechanisms underlying disease caused by asymptomatic infections are unknown, it is believed that productive pathogen replication is required. We report that the environmental stress of carbon dioxide (CO2) anesthesia converts an asymptomatic rhabdovirus infection in Drosophila to one that is lethal. This lethality results from a pool of infectious virus in glial cells and is regulated by the antiviral RNAi pathway of the host. CO2 sensitivity is caused by the fusogenic activity of the viral glycoprotein, which results in fusion of neurons and glia. Expression of the viral glycoprotein, but not a fusion defective mutant, is sufficient to cause CO2 sensitivity, which can occur even in the absence of productive viral replication. These findings highlight how viral proteins, independent of pathogen replication, may predispose hosts to life-threatening environmental stress.


Carbon Dioxide/pharmacology , Environment , Neurons/virology , Stress, Physiological , Vesicular stomatitis Indiana virus/drug effects , Vesicular stomatitis Indiana virus/pathogenicity , Virus Diseases , Animals , Animals, Genetically Modified , Anopheles/immunology , Anopheles/virology , Antiviral Agents/pharmacology , Behavior, Animal , Drosophila melanogaster/immunology , Drosophila melanogaster/virology , Glycoproteins/metabolism , Hydrogen-Ion Concentration , Immunity, Innate , Neuroglia/immunology , Neuroglia/virology , Neurons/immunology , RNA Interference , Sindbis Virus/immunology , Sindbis Virus/pathogenicity , Viral Plaque Assay , Viral Proteins/metabolism , Virus Diseases/immunology , Virus Diseases/virology , Virus Replication/drug effects
13.
PLoS Pathog ; 13(6): e1006473, 2017 Jun.
Article En | MEDLINE | ID: mdl-28662211

Alphaviruses are arthropod-borne viruses that represent a significant threat to public health at a global level. While the formation of alphaviral nucleocapsid cores, consisting of cargo nucleic acid and the viral capsid protein, is an essential molecular process of infection, the precise interactions between the two partners are ill-defined. A CLIP-seq approach was used to screen for candidate sites of interaction between the viral Capsid protein and genomic RNA of Sindbis virus (SINV), a model alphavirus. The data presented in this report indicates that the SINV capsid protein binds to specific viral RNA sequences in the cytoplasm of infected cells, but its interaction with genomic RNA in mature extracellular viral particles is largely non-specific in terms of nucleotide sequence. Mutational analyses of the cytoplasmic viral RNA-capsid interaction sites revealed a functional role for capsid binding early in infection. Interaction site mutants exhibited decreased viral growth kinetics; however, this defect was not a function of decreased particle production. Rather mutation of the cytoplasmic capsid-RNA interaction sites negatively affected the functional capacity of the incoming viral genomic RNAs leading to decreased infectivity. Furthermore, cytoplasmic capsid interaction site mutants are attenuated in a murine model of neurotropic alphavirus infection. Collectively, the findings of this study indicate that the identified cytoplasmic interactions of the viral capsid protein and genomic RNA, while not essential for particle formation, are necessary for genomic RNA function early during infection. This previously unappreciated role of capsid protein during the alphaviral replication cycle also constitutes a novel virulence determinant.


Capsid Proteins/metabolism , RNA, Viral/metabolism , Sindbis Virus/metabolism , Animals , Capsid/metabolism , Cytoplasm/metabolism , Genome, Viral/genetics , Sindbis Virus/genetics , Sindbis Virus/pathogenicity , Viral Envelope Proteins/metabolism , Virion/metabolism , Virulence/physiology , Virus Assembly/physiology
14.
Proc Natl Acad Sci U S A ; 114(10): 2681-2686, 2017 03 07.
Article En | MEDLINE | ID: mdl-28213497

The innate immune system senses RNA viruses by pattern recognition receptors (PRRs) and protects the host from virus infection. PRRs mediate the production of immune modulatory factors and direct the elimination of RNA viruses. Here, we show a unique PRR that mediates antiviral response. Tetrachlorodibenzo-p-dioxin (TCDD)-inducible poly(ADP ribose) polymerase (TIPARP), a Cysteine3 Histidine (CCCH)-type zinc finger-containing protein, binds to Sindbis virus (SINV) RNA via its zinc finger domain and recruits an exosome to induce viral RNA degradation. TIPARP typically localizes in the nucleus, but it accumulates in the cytoplasm after SINV infection, allowing targeting of cytoplasmic SINV RNA. Redistribution of TIPARP is induced by reactive oxygen species (ROS)-dependent oxidization of the nuclear pore that affects cytoplasmic-nuclear transport. BCL2-associated X protein (BAX) and BCL2 antagonist/killer 1 (BAK1), B-cell leukemia/lymphoma 2 (BCL2) family members, mediate mitochondrial damage to generate ROS after SINV infection. Thus, TIPARP is a viral RNA-sensing PRR that mediates antiviral responses triggered by BAX- and BAK1-dependent mitochondrial damage.


Immunity, Innate/genetics , Poly(ADP-ribose) Polymerases/genetics , RNA Viruses/genetics , Receptors, Pattern Recognition/genetics , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/immunology , Cytoplasm/genetics , Cytoplasm/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Mitochondria/genetics , Mitochondria/pathology , Mitochondria/virology , Nucleoside Transport Proteins , Poly(ADP-ribose) Polymerases/immunology , RNA Viruses/immunology , Reactive Oxygen Species/metabolism , Receptors, Pattern Recognition/immunology , Sindbis Virus/genetics , Sindbis Virus/immunology , Sindbis Virus/pathogenicity , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/immunology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/immunology
15.
Neurotherapeutics ; 13(3): 455-60, 2016 07.
Article En | MEDLINE | ID: mdl-27114366

Mosquito-borne viruses are important causes of death and long-term neurologic disability due to encephalomyelitis. Studies of mice infected with the alphavirus Sindbis virus have shown that outcome is dependent on the age and genetic background of the mouse and virulence of the infecting virus. Age-dependent susceptibility reflects the acquisition by neurons of resistance to virus replication and virus-induced cell death with maturation. In mature mice, the populations of neurons most susceptible to infection are in the hippocampus and anterior horn of the spinal cord. Hippocampal infection leads to long-term memory deficits in mice that survive, while motor neuron infection can lead to paralysis and death. Neuronal death is immune-mediated, rather than a direct consequence of virus infection, and associated with entry and differentiation of pathogenic T helper 17 cells in the nervous system. To modulate glutamate excitotoxicity, mice were treated with an N-methyl-D-aspartate receptor antagonist, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor antagonists or a glutamine antagonist. The N-methyl-D-aspartate receptor antagonist MK-801 protected hippocampal neurons but not motor neurons, and mice still became paralyzed and died. α-Amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor antagonists GYKI-52466 and talampanel protected both hippocampal and motor neurons and prevented paralysis and death. Glutamine antagonist 6-diazo-5-l-norleucine protected hippocampal neurons and improved memory generation in mice surviving infection with an avirulent virus. Surprisingly, in all cases protection was associated with inhibition of the antiviral immune response, reduced entry of inflammatory cells into the central nervous system, and delayed virus clearance, emphasizing the importance of treatment approaches that include prevention of immunopathologic damage.


Alphavirus Infections/drug therapy , Encephalomyelitis/drug therapy , Encephalomyelitis/virology , Neurons/virology , Sindbis Virus/physiology , Alphavirus Infections/immunology , Alphavirus Infections/prevention & control , Alphavirus Infections/virology , Animals , Benzodiazepines/administration & dosage , Cell Death , Diazooxonorleucine/administration & dosage , Dizocilpine Maleate/administration & dosage , Encephalomyelitis/immunology , Encephalomyelitis/prevention & control , Hippocampus/drug effects , Hippocampus/virology , Humans , Inflammation/metabolism , Memory/drug effects , Mice , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Sindbis Virus/pathogenicity , Survival Analysis , Th17 Cells/metabolism
16.
Rev Med Virol ; 26(4): 221-41, 2016 07.
Article En | MEDLINE | ID: mdl-26990827

Sindbis virus (SINV; family Togaviridae, genus Alphavirus) is an enveloped RNA virus widely distributed in Eurasia, Africa, Oceania and Australia. SINV is transmitted among its natural bird hosts via mosquitoes. Human disease caused by SINV infection has been reported mainly in South Africa and in Northern Europe. Vector mosquito abundance affects the annual incidence of SINV infections with occasional outbreaks of up to 1500 patients. Symptoms include fever, malaise, rash and musculoskeletal pain. In a significant portion of patients the debilitating musculoskeletal symptoms persist for years. Chronic disease after SINV infection shares many features with autoimmune diseases. Currently there is no specific treatment available. Recently SINV infections have been detected outside the previously known distribution range. In this article we will summarize the current knowledge on epidemiology, clinical disease and pathogenesis of SINV infection in man. Copyright © 2016 John Wiley & Sons, Ltd.


Alphavirus Infections/epidemiology , Alphavirus Infections/pathology , Sindbis Virus/pathogenicity , Zoonoses/epidemiology , Zoonoses/virology , Alphavirus Infections/veterinary , Alphavirus Infections/virology , Animals , Bird Diseases/epidemiology , Bird Diseases/virology , Birds , Disease Outbreaks , Global Health , Humans , Incidence , Topography, Medical
17.
Emerg Infect Dis ; 21(12): 2225-9, 2015 Dec.
Article En | MEDLINE | ID: mdl-26583836

Old World alphaviruses were identified in 52 of 623 horses with febrile or neurologic disease in South Africa. Five of 8 Sindbis virus infections were mild; 2 of 3 fatal cases involved co-infections. Of 44 Middelburg virus infections, 28 caused neurologic disease; 12 were fatal. Middelburg virus likely has zoonotic potential.


Horses/virology , Sindbis Virus/pathogenicity , Zoonoses/epidemiology , Alphavirus/genetics , Animals , Horses/genetics , Humans , Phylogeny , Sindbis Virus/genetics , South Africa/epidemiology
18.
Biochem Biophys Res Commun ; 462(4): 426-32, 2015 Jul 10.
Article En | MEDLINE | ID: mdl-25976675

We examined the impact of Sindbis virus (SINV) infection on cell cycle progression in a cancer cell line, HeLa, and a non-cancerous cell line, Vero. Cell cycle analyses showed that SINV infection is able to alter the cell cycle progression in both HeLa and Vero cells, but differently, especially during the early stage of infection. SINV infection affected the expression of several cell cycle regulators (CDK4, CDK6, cyclin E, p21, cyclin A and cyclin B) in HeLa cells and caused HeLa cells to accumulate in S phase during the early stage of infection. Monitoring SINV replication in HeLa and Vero cells expressing cell cycle indicators revealed that SINV which infected HeLa cells during G1 phase preferred to proliferate during S/G2 phase, and the average time interval for viral replication was significantly shorter in both HeLa and Vero cells infected during G1 phase than in cells infected during S/G2 phase.


Cell Cycle , Sindbis Virus/pathogenicity , HeLa Cells , Humans , Sindbis Virus/physiology , Virus Replication
19.
Virus Res ; 196: 1-4, 2015 Jan 22.
Article En | MEDLINE | ID: mdl-25449361

Sindbis virus (SV) can be rendered neurovirulent for adult mice by a double substitution within the E2 glycoprotein, including replacing Gln at position 55 of E2 with a His (E2-55: Gln-His) and E2-70: Glu to Lys. However, the mutant Sindbis-like virus XJ-160 with the double substitution (BR-E5570) does not show neurovirulence for adult mice, although the mutant apparently reduced the average survival time of neonatal mice. To produce an XJ-160 virus neurovirulent for adult mice, the BR-E5570 virus containing the double substitution was provided with another substitution in the nsP1 region (nsP1-173: Thr-Ile), which could enhance viral infectivity and neurovirulence for neonatal mice. The mutant containing these three substitutions was accordingly designated as BR-5570-ns173. Like the BR-XJ160 virus derived from the wild-type clone, BR-E5570 and BR-E5570-ns173 were both virulent for newborn mice, between which BR-E5570-ns173 virus showed the greatest neurovirulence. Furthermore, only BR-E5570-ns173 virus was fully neurovirulent for 14-day-old mice, and this fatal adult mouse-virulence was dependent on the E2 double substitutions at positions 55 and 70. Compared with BR-XJ160, both the mutants showed a higher capacity for propagation both in cultured cells and in the mouse brain. In particular, BR-E5570-ns173 virus showed a more persistent existence and higher titer in the brains of 7-day-old mice. These findings indicate that the substitution at nsP1-173 combination with a double substitution in the E2 region renders the XJ-160 virus fully neurovirulent for adult mice, and this neurovirulence may be related to the increased efficiency and persistence of propagation of this virus.


Alphavirus Infections/virology , Nervous System Diseases/virology , Sindbis Virus/physiology , Viral Envelope Proteins/genetics , Viral Nonstructural Proteins/genetics , Alphavirus Infections/mortality , Amino Acid Substitution , Animals , Cell Line , Mice , Nervous System Diseases/mortality , Sindbis Virus/pathogenicity , Viral Envelope Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Virulence/genetics
20.
Cell Rep ; 5(6): 1737-48, 2013 Dec 26.
Article En | MEDLINE | ID: mdl-24332855

Alphaviruses are a large class of insect-borne human pathogens and little is known about the host-factor requirements for infection. To identify such factors, we performed a genome-wide RNAi screen using model Drosophila cells and validated 94 genes that impacted infection of Sindbis virus (SINV), the prototypical alphavirus. We identified a conserved role for SEC61A and valosin-containing protein (VCP) in facilitating SINV entry in insects and mammals. SEC61A and VCP selectively regulate trafficking of the entry receptor NRAMP2, and loss or pharmacological inhibition of these proteins leads to altered NRAMP2 trafficking to lysosomal compartments and proteolytic digestion within lysosomes. NRAMP2 is the major iron transporter in cells, and loss of NRAMP2 attenuates intracellular iron transport. Thus, this study reveals genes and pathways involved in both infection and iron homeostasis that may serve as targets for antiviral therapeutics or for iron-imbalance disorders.


Adenosine Triphosphatases/metabolism , Drosophila Proteins/metabolism , Genome, Insect , Membrane Proteins/metabolism , Sindbis Virus/pathogenicity , Virus Internalization , Adenosine Triphosphatases/genetics , Aedes/genetics , Aedes/metabolism , Aedes/virology , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Drosophila/genetics , Drosophila/metabolism , Drosophila/virology , Drosophila Proteins/genetics , Humans , Iron/metabolism , Membrane Proteins/genetics , Protein Transport , RNA Interference , RNA, Small Interfering/genetics , SEC Translocation Channels , Valosin Containing Protein
...