Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 339
Filter
1.
J Virol ; 95(13): e0013621, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33853958

ABSTRACT

Arboviruses are transmitted by specific vectors, and the reasons for this specificity are not fully understood. One contributing factor is the existence of tissue barriers within the vector such as the midgut escape barrier. We used microRNA (miRNA) targeting of Sindbis virus (SINV) to study how replication in midgut cells contributes to overcoming this barrier in the mosquito Aedes aegypti. SINV constructs were designed to be attenuated specifically in midgut cells by inserting binding sites for midgut-specific miRNAs into either the 3' untranslated region (MRE3'miRT) or the structural open reading frame (MRE-ORFmiRT) of the SINV genome. Both miRNA-targeted viruses replicated less efficiently than control viruses in the presence of these miRNAs. When mosquitoes were given infectious blood meals containing miRNA-targeted viruses, only around 20% (MRE3'miRT) or 40% (MRE-ORFmiRT) of mosquitoes developed disseminated infection. In contrast, dissemination occurred in almost all mosquitoes fed control viruses. Deep sequencing of virus populations from individual mosquitoes ruled out selection for mutations in the inserted target sequences as the cause for dissemination in these mosquitoes. In mosquitoes that became infected with miRNA-targeted viruses, titers were equivalent to those of mosquitoes infected with control virus in both the midgut and the carcass, and there was no evidence of a threshold titer necessary for dissemination. Instead, it appeared that if infection was successfully established in the midgut, replication and dissemination were largely normal. Our results support the hypothesis that replication is an important factor in allowing SINV to overcome the midgut escape barrier but hint that other factors are also likely involved. IMPORTANCE When a mosquito ingests an arbovirus during a blood meal, the arbovirus must escape from the midgut of the vector and infect the salivary glands in order to be transmitted to a new host. We used tissue-specific miRNA targeting to examine the requirement for Sindbis virus (SINV) to replicate in midgut epithelium in order to cause disseminated infection in the mosquito Aedes aegypti. Our results indicate that specifically reducing the ability of SINV to replicate in the mosquito midgut reduces its overall ability to establish infection in the mosquito, but if infection is established, replication and dissemination occur normally. These results are consistent with an importance for replication in the midgut epithelium in aiding arboviruses in crossing the midgut barrier.


Subject(s)
Aedes/virology , Gastrointestinal Tract/virology , MicroRNAs/genetics , Sindbis Virus/growth & development , Virus Replication/genetics , Animals , Cell Line , Cricetinae , Mosquito Vectors/virology , Organ Specificity , Salivary Glands/virology , Sindbis Virus/genetics , Sindbis Virus/metabolism
2.
PLoS Negl Trop Dis ; 13(11): e0007346, 2019 11.
Article in English | MEDLINE | ID: mdl-31693659

ABSTRACT

BACKGROUND: Aedes aegypti is a vector mosquito of major public health importance, transmitting arthropod-borne viruses (arboviruses) such as chikungunya, dengue, yellow fever and Zika viruses. Wild mosquito populations are persistently infected at high prevalence with insect-specific viruses that do not replicate in vertebrate hosts. In experimental settings, acute infections with insect-specific viruses have been shown to modulate arbovirus infection and transmission in Ae. aegypti and other vector mosquitoes. However, the impact of persistent insect-specific virus infections, which arboviruses encounter more commonly in nature, has not been investigated extensively. Cell lines are useful models for studying virus-host interactions, however the available Ae. aegypti cell lines are poorly defined and heterogenous cultures. METHODOLOGY/PRINCIPLE FINDINGS: We generated single cell-derived clonal cell lines from the commonly used Ae. aegypti cell line Aag2. Two of the fourteen Aag2-derived clonal cell lines generated harboured markedly and consistently reduced levels of the insect-specific bunyavirus Phasi Charoen-like virus (PCLV) known to persistently infect Aag2 cells. In contrast to studies with acute insect-specific virus infections in cell culture and in vivo, we found that pre-existing persistent PCLV infection had no major impact on the replication of the flaviviruses dengue virus and Zika virus, the alphavirus Sindbis virus, or the rhabdovirus vesicular stomatitis virus. We also performed a detailed characterisation of the morphology, transfection efficiency and immune status of our Aag2-derived clonal cell lines, and have made a clone that we term Aag2-AF5 available to the research community as a well-defined cell culture model for arbovirus-vector interaction studies. CONCLUSIONS/SIGNIFICANCE: Our findings highlight the need for further in vivo studies that more closely recapitulate natural arbovirus transmission settings in which arboviruses encounter mosquitoes harbouring persistent rather than acute insect-specific virus infections. Furthermore, we provide the well-characterised Aag2-derived clonal cell line as a valuable resource to the arbovirus research community.


Subject(s)
Aedes/virology , Arboviruses/growth & development , Coinfection/virology , Mosquito Vectors/virology , Orthobunyavirus/growth & development , Virus Replication , Alphavirus/growth & development , Animals , Arboviruses/genetics , Base Sequence , Cell Culture Techniques/methods , Cell Line , Dengue Virus/growth & development , Flavivirus/genetics , Flavivirus/growth & development , Genome, Viral , Host-Pathogen Interactions/physiology , Orthobunyavirus/genetics , RNA Viruses/genetics , RNA Viruses/growth & development , Rhabdoviridae/growth & development , Sindbis Virus/growth & development , Transfection , Zika Virus/growth & development
3.
J Virol ; 93(24)2019 12 15.
Article in English | MEDLINE | ID: mdl-31554691

ABSTRACT

Alphaviruses are enveloped, positive-sense RNA viruses that are important causes of viral encephalomyelitis. Sindbis virus (SINV) infects the neurons of rodents and is a model for studying factors that regulate infection of neuronal cells. The outcome of alphavirus infection of the central nervous system is dependent on neuronal maturation status. Differentiated mature neurons survive and control viral replication better than undifferentiated immature neurons. The cellular factors involved in age-dependent susceptibility include higher levels of antiapoptotic and innate immune factors in mature neurons. Because NF-κB pathway activation is required for the initiation of both apoptosis and the host antiviral response, we analyzed the role of NF-κB during SINV infection of differentiated and undifferentiated rat neuronal cells. SINV infection induced canonical NF-κB activation, as evidenced by the degradation of IκBα and the phosphorylation and nuclear translocation of p65. Inhibition or deletion of the upstream IκB kinase substantially reduced SINV replication in differentiated but not in undifferentiated neuronal cells or mouse embryo fibroblasts. NF-κB inhibition did not affect the establishment of infection, replication complex formation, the synthesis of nonstructural proteins, or viral RNA synthesis in differentiated neurons. However, the translation of structural proteins was impaired, phosphorylation of the α subunit of eukaryotic translation initiation factor 2 (eIF2α) was decreased, and host protein synthesis was maintained, suggesting that NF-κB activation was involved in the regulation of translation during infection of mature neurons. Inhibition or deletion of double-stranded RNA-activated protein kinase (PKR) also decreased eIF2α phosphorylation, the translation of viral structural proteins, and virus production. Therefore, canonical NF-κB activation synergizes with PKR to promote SINV replication in differentiated neurons by facilitating viral structural protein translation.IMPORTANCE Mosquito-borne alphaviruses are a significant and growing cause of viral encephalomyelitis worldwide. The outcome of alphaviral neuronal infections is host age dependent and greatly affected by neuronal maturation status, with differentiated, mature neurons being more resistant to infection than undifferentiated, immature neurons. The biological factors that change during neuronal maturation and that influence the outcome of viral infection are currently only partially defined. These studies investigated the role of NF-κB in determining the outcome of alphaviral infection in mature and immature neurons. Inhibition of canonical NF-κB activation decreased alphavirus replication in mature neurons by regulating protein synthesis and limiting the production of the viral structural proteins but had little effect on viral replication in immature neurons or fibroblasts. Therefore, NF-κB is a signaling pathway that influences the maturation-dependent outcome of alphaviral infection in neurons and that highlights the importance of cellular context in determining the effects of signal pathway activation.


Subject(s)
Alphavirus Infections/virology , Alphavirus/drug effects , Alphavirus/growth & development , NF-kappa B/pharmacology , Neurons/virology , Virus Replication/drug effects , Animals , Cell Differentiation , Cell Line , Culicidae/virology , Eukaryotic Initiation Factor-2/metabolism , Gene Knockout Techniques , Mice , NF-kappa B/genetics , Neurogenesis , Phosphorylation , RNA, Viral/metabolism , Rats , Signal Transduction , Sindbis Virus/drug effects , Sindbis Virus/growth & development , Transcriptome , eIF-2 Kinase/metabolism
4.
Mol Cell ; 74(1): 196-211.e11, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30799147

ABSTRACT

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.


Subject(s)
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
5.
J Gen Virol ; 100(1): 46-62, 2019 01.
Article in English | MEDLINE | ID: mdl-30451651

ABSTRACT

Interferon (IFN) regulatory factors (IRFs) are important determinants of the innate response to infection. We evaluated the role(s) of combined and individual IRF deficiencies in the outcome of infection of C57BL/6 mice with Sindbis virus, an alphavirus that infects neurons and causes encephalomyelitis. The brain and spinal cord levels of Irf7, but not Irf3 mRNAs, were increased after infection. IRF3/5/7-/- and IRF3/7-/- mice died within 3-4 days with uncontrolled virus replication, similar to IFNα receptor-deficient mice, while all wild-type (WT) mice recovered. IRF3-/- and IRF7-/- mice had brain levels of IFNα that were lower, but brain and spinal cord levels of IFNß and IFN-stimulated gene mRNAs that were similar to or higher than WT mice without detectable serum IFN or increases in Ifna or Ifnb mRNAs in the lymph nodes, indicating that the differences in outcome were not due to deficiencies in the central nervous system (CNS) type I IFN response. IRF3-/- mice developed persistent neurological deficits and had more spinal cord inflammation and higher CNS levels of Il1b and Ifnγ mRNAs than WT mice, but all mice survived. IRF7-/- mice died 5-8 days after infection with rapidly progressive paralysis and differed from both WT and IRF3-/- mice in the induction of higher CNS levels of IFNß, tumour necrosis factor (TNF) α and Cxcl13 mRNA, delayed virus clearance and more extensive cell death. Therefore, fatal disease in IRF7-/- mice is likely due to immune-mediated neurotoxicity associated with failure to regulate the production of inflammatory cytokines such as TNFα in the CNS.


Subject(s)
Alphavirus Infections/physiopathology , Encephalomyelitis/physiopathology , Host-Pathogen Interactions , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Sindbis Virus/growth & development , Animals , Brain/pathology , Disease Models, Animal , Gene Expression Profiling , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-7/deficiency , Mice, Inbred C57BL , Mice, Knockout , Spinal Cord/pathology , Survival Analysis
6.
Nat Commun ; 9(1): 5326, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30552337

ABSTRACT

Alphaviruses are enveloped RNA viruses that contain several human pathogens. Due to intrinsic heterogeneity of alphavirus particles, a high resolution structure of the virion is currently lacking. Here we provide a 3.5 Å cryo-EM structure of Sindbis virus, using block based reconstruction method that overcomes the heterogeneity problem. Our structural analysis identifies a number of conserved residues that play pivotal roles in the virus life cycle. We identify a hydrophobic pocket in the subdomain D of E2 protein that is stabilized by an unknown pocket factor near the viral membrane. Residues in the pocket are conserved in different alphaviruses. The pocket strengthens the interactions of the E1/E2 heterodimer and may facilitate virus assembly. Our study provides structural insights into alphaviruses that may inform the design of drugs and vaccines.


Subject(s)
Alphavirus/growth & development , Cryoelectron Microscopy/methods , Protein Interaction Domains and Motifs , Virus Assembly , Virus Internalization , Animals , Chlorocebus aethiops , Crystallography, X-Ray , Life Cycle Stages , Membrane Glycoproteins/chemistry , Models, Molecular , Protein Conformation , Sindbis Virus/growth & development , Sindbis Virus/ultrastructure , Vero Cells , Viral Envelope Proteins/chemistry , Virion/growth & development , Virion/ultrastructure
7.
mBio ; 9(6)2018 12 11.
Article in English | MEDLINE | ID: mdl-30538185

ABSTRACT

Alphaviruses are arthropod-borne RNA viruses that are capable of causing severe disease and are a significant burden to public health. Alphaviral replication results in the production of both capped and noncapped viral genomic RNAs (ncgRNAs), which are packaged into virions during infections of vertebrate and invertebrate cells. However, the roles that the ncgRNAs play during alphaviral infection have yet to be exhaustively characterized. Here, the importance of the ncgRNAs to alphaviral infection was assessed by using mutations of the nsP1 protein of Sindbis virus (SINV), which altered the synthesis of the ncgRNAs during infection by modulating the protein's capping efficiency. Specifically, point mutations at residues Y286A and N376A decreased capping efficiency whereas a point mutation at D355A increased the capping efficiency of the SINV genomic RNA during genuine viral infection. Viral growth kinetics levels were significantly reduced for the D355A mutant relative to wild-type infection, whereas the Y286A and N376A mutants showed modest decreases in growth kinetics. Overall genomic translation and nonstructural protein accumulation were found to correlate with increases and decreases in capping efficiency. However, genomic, minus-strand, and subgenomic viral RNA synthesis were largely unaffected by the modulation of alphaviral capping activity. In addition, translation of the subgenomic alphaviral RNA (vRNA) was found not to be impacted by changes in capping efficiency. The mechanism by which the decreased presence of ncgRNAs reduced viral growth kinetics levels operated through the impaired production of viral particles. Collectively, these data illustrate the importance of ncgRNAs to viral infection and suggest that they play an integral role in the production of viral progeny.IMPORTANCE Alphaviruses have been the cause of both localized outbreaks and large epidemics of severe disease. Currently, there are no strategies or vaccines which are either safe or effective for preventing alphaviral infection or treating alphaviral disease. This deficit of viable therapeutics highlights the need to better understand the mechanisms behind alphaviral infection in order to develop novel antiviral strategies for treatment of alphaviral disease. In particular, this report details a previously uncharacterized aspect of the alphaviral life cycle: the importance of noncapped genomic viral RNAs for alphaviral infection. This offers new insights into the mechanisms of alphaviral replication and the impact of the noncapped genomic RNAs on viral packaging.


Subject(s)
Sindbis Virus/enzymology , Sindbis Virus/growth & development , Viral Nonstructural Proteins/metabolism , Amino Acid Substitution , Animals , Cell Line , Cricetinae , Mutant Proteins/genetics , Mutant Proteins/metabolism , Point Mutation , Protein Biosynthesis , RNA, Viral/metabolism , Sindbis Virus/genetics , Viral Nonstructural Proteins/genetics
8.
mBio ; 8(1)2017 02 14.
Article in English | MEDLINE | ID: mdl-28196962

ABSTRACT

Sindbis virus (SINV [genus Alphavirus, family Togaviridae]) is an enveloped, mosquito-borne virus. Alphaviruses cause cytolytic infections in mammalian cells while establishing noncytopathic, persistent infections in mosquito cells. Mosquito vector adaptation of alphaviruses is a major factor in the transmission of epidemic strains of alphaviruses. Though extensive studies have been performed on infected mammalian cells, the morphological and structural elements of alphavirus replication and assembly remain poorly understood in mosquito cells. Here we used high-resolution live-cell imaging coupled with single-particle tracking and electron microscopy analyses to delineate steps in the alphavirus life cycle in both the mammalian host cell and insect vector cells. Use of dually labeled SINV in conjunction with cellular stains enabled us to simultaneously determine the spatial and temporal differences of alphavirus replication complexes (RCs) in mammalian and insect cells. We found that the nonstructural viral proteins and viral RNA in RCs exhibit distinct spatial organization in mosquito cytopathic vacuoles compared to replication organelles from mammalian cells. We show that SINV exploits filopodial extensions for virus dissemination in both cell types. Additionally, we propose a novel mechanism for replication complex formation around glycoprotein-containing vesicles in mosquito cells that produced internally released particles that were seen budding from the vesicles by live imaging. Finally, by characterizing mosquito cell lines that were persistently infected with fluorescent virus, we show that the replication and assembly machinery are highly modified, and this allows continuous production of alphaviruses at reduced levels.IMPORTANCE Reemerging mosquito-borne alphaviruses cause serious human epidemics worldwide. Several structural and imaging studies have helped to define the life cycle of alphaviruses in mammalian cells, but the mode of virus replication and assembly in the invertebrate vector and mechanisms producing two disease outcomes in two types of cells are yet to be identified. Using transmission electron microscopy and live-cell imaging with dual fluorescent protein-tagged SINV, we show that while insect and mammalian cells display similarities in entry and exit, they present distinct spatial and temporal organizations in virus replication and assembly. By characterizing acutely and persistently infected cells, we provide new insights into alphavirus replication and assembly in two distinct hosts, resulting in high-titer virus production in mammalian cells and continuous virus production at reduced levels in mosquito cells-presumably a prerequisite for alphavirus maintenance in nature.


Subject(s)
Aedes/virology , Sindbis Virus/physiology , Spatio-Temporal Analysis , Virus Assembly , Virus Replication , Animals , Cell Line , DNA Replication , Humans , Kinetics , Life Cycle Stages , Luminescent Proteins , Microscopy, Energy-Filtering Transmission Electron , RNA, Viral , Sindbis Virus/growth & development , Red Fluorescent Protein
9.
J Virol ; 91(9)2017 05 01.
Article in English | MEDLINE | ID: mdl-28202764

ABSTRACT

Zinc finger antiviral protein (ZAP) is a host factor that specifically inhibits the replication of certain viruses by binding to viral mRNAs and repressing the translation and/or promoting the degradation of target mRNA. In addition, ZAP regulates the expression of certain cellular genes. Here, we report that tripartite motif-containing protein 25 (TRIM25), a ubiquitin E3 ligase, is required for the antiviral activity of ZAP. Downregulation of endogenous TRIM25 abolished ZAP's antiviral activity. The E3 ligase activity of TRIM25 is required for this regulation. TRIM25 mediated ZAP ubiquitination, but the ubiquitination of ZAP itself did not seem to be required for its antiviral activity. Downregulation of endogenous ubiquitin or overexpression of the deubiquitinase OTUB1 impaired ZAP's activity. We provide evidence indicating that TRIM25 modulates the target RNA binding activity of ZAP. These results uncover a mechanism by which the antiviral activity of ZAP is regulated.IMPORTANCE ZAP is a host antiviral factor that specifically inhibits the replication of certain viruses, including HIV-1, Sindbis virus, and Ebola virus. ZAP binds directly to target mRNA, and it represses the translation and promotes the degradation of target mRNA. While the mechanisms by which ZAP posttranscriptionally inhibits target RNA expression have been extensively studied, how its antiviral activity is regulated is not very clear. Here, we report that TRIM25, a ubiquitin E3 ligase, is required for the antiviral activity of ZAP. Downregulation of endogenous TRIM25 remarkably abolished ZAP's activity. TRIM25 is required for ZAP optimal binding to target mRNA. These results help us to better understand how the antiviral activity of ZAP is regulated.


Subject(s)
Antiviral Agents/metabolism , RNA-Binding Proteins/metabolism , Sindbis Virus/growth & development , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Virus Replication/genetics , Cell Line , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzymes , HEK293 Cells , Humans , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Viral/genetics , Sindbis Virus/genetics , Ubiquitination , Zinc Fingers
10.
PLoS Pathog ; 13(1): e1006145, 2017 01.
Article in English | MEDLINE | ID: mdl-28060952

ABSTRACT

The host factor and interferon (IFN)-stimulated gene (ISG) product, zinc-finger antiviral protein (ZAP), inhibits a number of diverse viruses by usurping and intersecting with multiple cellular pathways. To elucidate its antiviral mechanism, we perform a loss-of-function genome-wide RNAi screen to identify cellular cofactors required for ZAP antiviral activity against the prototype alphavirus, Sindbis virus (SINV). In order to exclude off-target effects, we carry out stringent confirmatory assays to verify the top hits. Important ZAP-liaising partners identified include proteins involved in membrane ion permeability, type I IFN signaling, and post-translational protein modification. The factor contributing most to the antiviral function of ZAP is TRIM25, an E3 ubiquitin and ISG15 ligase. We demonstrate here that TRIM25 interacts with ZAP through the SPRY domain, and TRIM25 mutants lacking the RING or coiled coil domain fail to stimulate ZAP's antiviral activity, suggesting that both TRIM25 ligase activity and its ability to form oligomers are critical for its cofactor function. TRIM25 increases the modification of both the short and long ZAP isoforms by K48- and K63-linked polyubiquitin, although ubiquitination of ZAP does not directly affect its antiviral activity. However, TRIM25 is critical for ZAP's ability to inhibit translation of the incoming SINV genome. Taken together, these data uncover TRIM25 as a bona fide ZAP cofactor that leads to increased ZAP modification enhancing its translational inhibition activity.


Subject(s)
Alphavirus Infections/prevention & control , Antiviral Agents/metabolism , RNA-Binding Proteins/metabolism , Sindbis Virus/growth & development , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , Cricetinae , HEK293 Cells , Humans , Interferon Type I/metabolism , Protein Domains , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
11.
Biochem Pharmacol ; 120: 15-21, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27664855

ABSTRACT

Previously, we reported that salicylate-based analogs of bryostatin protect cells from chikungunya virus (CHIKV)-induced cell death. Interestingly, 'capping' the hydroxyl group at C26 of a lead bryostatin analog, a position known to be crucial for binding to and modulation of protein kinase C (PKC), did not abrogate the anti-CHIKV activity of the scaffold, putatively indicating the involvement of a pathway independent of PKC. The work detailed in this study demonstrates that salicylate-derived analog 1 and two capped analogs (2 and 3) are not merely cytoprotective compounds, but act as selective and specific inhibitors of CHIKV replication. Further, a detailed comparative analysis of the effect of the non-capped versus the two capped analogs revealed that compound 1 acts both at early and late stages in the chikungunya virus replication cycle, while the capped analogs only interfere with a later stage process. Co-dosing with the PKC inhibitors sotrastaurin and Gö6976 counteracts the antiviral activity of compound 1 without affecting that of capped analogs 2 and 3, providing further evidence that the latter elicit their anti-CHIKV activity independently of PKC. Remarkably, treatment of CHIKV-infected cells with a combination of compound 1 and a capped analog resulted in a pronounced synergistic antiviral effect. Thus, these salicylate-based bryostatin analogs can inhibit CHIKV replication through a novel, yet still elusive, non-PKC dependent pathway.


Subject(s)
Antiviral Agents/pharmacology , Bryostatins/pharmacology , Chikungunya virus/drug effects , Drug Design , Protein Kinase C/metabolism , Viral Proteins/metabolism , Acetylation , Animals , Antiviral Agents/agonists , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/chemistry , Bryostatins/agonists , Bryostatins/antagonists & inhibitors , Bryostatins/chemistry , Carbazoles/chemistry , Carbazoles/pharmacology , Cell Line , Chikungunya virus/growth & development , Chikungunya virus/metabolism , Chlorocebus aethiops , Drug Synergism , Gene Expression Regulation, Viral/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Kinetics , Methylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/chemistry , Protein Kinase C/genetics , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacology , Semliki forest virus/drug effects , Semliki forest virus/growth & development , Semliki forest virus/metabolism , Sindbis Virus/drug effects , Sindbis Virus/growth & development , Sindbis Virus/metabolism , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication/drug effects
13.
J Virol Methods ; 223: 19-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26205552

ABSTRACT

Eastern Equine Encephalitis virus (EEEV) is a medically important pathogen that can cause severe encephalitis in humans, with mortality rates ranging from 30 to 80%. Unfortunately there are no antivirals or licensed vaccines available for human use, and laboratory diagnosis is essential to differentiate EEEV infection from other pathogens with similar clinical manifestations. The Arboviral Diseases Branch (ADB) reference laboratory at the CDC Division of Vector-Borne Diseases (DVBD) produces reference antigens used in serological assays such as the EEEV immunoglobulin M antibody-capture enzyme-linked immunosorbent assay (MAC-ELISA). However, EEEV is classified as a HHS select agent and requires biosafety level (BSL) three containment, limiting EEEV antigen production in non-select agent and BSL-2 laboratories. A recombinant Sindbis virus (SINV)/EEEV has been constructed for use under BSL-2 conditions and is not regulated as a select agent. Cell culture production of inactivated EEEV antigen from SINV/EEEV for use in the EEEV MAC-ELISA is reported here. Cell culture conditions and inactivation procedures were analyzed for SINV/EEEV using a recently developed antigen production algorithm, with the MAC-ELISA as the performance indicator.


Subject(s)
Antigens, Viral/genetics , Antigens, Viral/isolation & purification , Encephalitis Virus, Eastern Equine/genetics , Encephalomyelitis, Equine/diagnosis , Sindbis Virus/genetics , Animals , Antibodies, Viral/blood , Antigens, Viral/immunology , Enzyme-Linked Immunosorbent Assay/methods , Horses , Humans , Immunoglobulin M/blood , Sindbis Virus/growth & development , Virus Cultivation/methods
14.
Immunol Res ; 62(3): 255-62, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25948474

ABSTRACT

In the present research, we have studied an influence of enhanced expression TRIM14 on alphavirus Sindbis (SINV, Togaviridae family) infection. In the HEK293 cells transfected with human trim14 gene (HEK-trim14), SINV yield after infection was decreased 1000-10,000 times (3-4 lg of TCD50/ml) at 24 h p.i. and considerably less (1-2 lg of TCD50/ml) at 48 h p.i. Analysis of the expression of 43 genes directly or indirectly involved in innate immune machine in HEK-trim14 non-infected cells comparing with the control (non-transfected) HEK293 cells revealed that stable trim14 transfection in HEK293 cells caused increased transcription of 18 genes (ifna, il6 (ifnß2), isg15, raf-1, NF-kB (nf-kb1, rela, nf-kb2, relb), grb2, grb3-3, traf3ip2, junB, c-myb, pu.1, akt1, tyk2, erk2, mek2) and lowered transcription of 3 genes (ifnγ, gata1, il-17a). The similar patterns of genes expression observe in SINV-infected non-transfected HEK293 cells. However, SINV infection of HEK-trim14 cells caused inhibition of the most interferon cascade genes as well as subunits of transcription factor NF-κB. Thus, stable enhanced expression of trim14 gene in cells activates the transcription of many immunity genes and suppresses the SINV reproduction, but SINV infection of HEK-trim14 cells promotes inhibition of some genes involved in innate immune system.


Subject(s)
Alphavirus Infections/immunology , Carrier Proteins/genetics , Immunity, Innate/genetics , Sindbis Virus/growth & development , Virus Replication/genetics , Alphavirus Infections/virology , Carrier Proteins/biosynthesis , Cell Line , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Intracellular Signaling Peptides and Proteins , Sindbis Virus/immunology , Transfection , Tripartite Motif Proteins
15.
J Virol ; 88(24): 14057-69, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25253354

ABSTRACT

UNLABELLED: Drosophila C virus (DCV) is a positive-sense RNA virus belonging to the Dicistroviridae family. This natural pathogen of the model organism Drosophila melanogaster is commonly used to investigate antiviral host defense in flies, which involves both RNA interference and inducible responses. Although lethality is used routinely as a readout for the efficiency of the antiviral immune response in these studies, virus-induced pathologies in flies still are poorly understood. Here, we characterize the pathogenesis associated with systemic DCV infection. Comparison of the transcriptome of flies infected with DCV or two other positive-sense RNA viruses, Flock House virus and Sindbis virus, reveals that DCV infection, unlike those of the other two viruses, represses the expression of a large number of genes. Several of these genes are expressed specifically in the midgut and also are repressed by starvation. We show that systemic DCV infection triggers a nutritional stress in Drosophila which results from intestinal obstruction with the accumulation of peritrophic matrix at the entry of the midgut and the accumulation of the food ingested in the crop, a blind muscular food storage organ. The related virus cricket paralysis virus (CrPV), which efficiently grows in Drosophila, does not trigger this pathology. We show that DCV, but not CrPV, infects the smooth muscles surrounding the crop, causing extensive cytopathology and strongly reducing the rate of contractions. We conclude that the pathogenesis associated with systemic DCV infection results from the tropism of the virus for an important organ within the foregut of dipteran insects, the crop. IMPORTANCE: DCV is one of the few identified natural viral pathogens affecting the model organism Drosophila melanogaster. As such, it is an important virus for the deciphering of host-virus interactions in insects. We characterize here the pathogenesis associated with DCV infection in flies and show that it results from the tropism of the virus for an essential but poorly characterized organ in the digestive tract, the crop. Our results may have relevance for other members of the Dicistroviridae, some of which are pathogenic to beneficial or pest insect species.


Subject(s)
Dicistroviridae/growth & development , Drosophila melanogaster/virology , Intestinal Obstruction/virology , Animals , Dicistroviridae/physiology , Female , Gastrointestinal Tract/pathology , Gastrointestinal Tract/physiopathology , Gastrointestinal Tract/virology , Gene Expression Profiling , Muscle, Smooth/virology , Nodaviridae/growth & development , Sindbis Virus/growth & development , Viral Tropism
16.
Proc Natl Acad Sci U S A ; 111(19): 7108-13, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24778219

ABSTRACT

Utilization of antiviral small interfering RNAs is thought to be largely restricted to plants, nematodes, and arthropods. In an effort to determine whether a physiological interplay exists between the host small RNA machinery and the cellular response to virus infection in mammals, we evaluated antiviral activity in the presence and absence of Dicer or Drosha, the RNase III nucleases responsible for generating small RNAs. Although loss of Dicer did not compromise the cellular response to virus infection, Drosha deletion resulted in a significant increase in virus levels. Here, we demonstrate that diverse RNA viruses trigger exportin 1 (XPO1/CRM1)-dependent Drosha translocation into the cytoplasm in a manner independent of de novo protein synthesis or the canonical type I IFN system. Additionally, increased virus infection in the absence of Drosha was not due to a loss of viral small RNAs but, instead, correlated with cleavage of viral genomic RNA and modulation of the host transcriptome. Taken together, we propose that Drosha represents a unique and conserved arm of the cellular defenses used to combat virus infection.


Subject(s)
Alphavirus Infections/immunology , Drosophila Proteins/immunology , Drosophila melanogaster/immunology , Drosophila melanogaster/virology , RNA, Viral/metabolism , Ribonuclease III/immunology , Sindbis Virus/immunology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Fibroblasts/cytology , HEK293 Cells , Humans , Interferon Type I/immunology , Karyopherins/metabolism , MicroRNAs/genetics , MicroRNAs/immunology , Protein Transport/immunology , Receptors, Cytoplasmic and Nuclear/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Sindbis Virus/genetics , Sindbis Virus/growth & development , Virus Replication/immunology , Exportin 1 Protein
17.
J Virol ; 87(20): 11231-43, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23926357

ABSTRACT

Rational design and directed evolution are powerful tools to generate and improve protein function; however, their uses are mostly limited to enzyme and antibody engineering. Here we describe a directed-evolution strategy, named the tandem selection and enrichment system (TSES), and its use in generating virus with exclusive specificity for a particular cellular receptor. In TSES, evolving viruses are sequentially and iteratively transferred between two different host cells, one for selection of receptor specificity and the other for enrichment of the fittest virus. By combining rational design and TSES, we generated human epidermal growth factor receptor (EGFR)-specific virus 1 (ESV1). ESV1 has the backbone of Sindbis virus (SINV) and displays an EGF domain engrafted onto structural protein E2 after residue Pro192, together with eight amino acid changes stabilizing the E2-EGF chimera. ESV1 uses EGFR to initiate infection and has lost the capacity to interact with all known SINV receptors. A 12.2-Å cryoelectron microscopic (cryoEM) reconstruction of ESV1 reveals that the E2-EGF fusion adopts a fixed conformation, with EGF sitting at the top of the E2 spike; The EGFR binding interface faces outward, and the EGF domain completely masks SINV receptor binding. The cryoEM structure of ESV1 explains the desirable properties of ESV1 and provides insights for its further modification. TSES expands the scope of directed evolution and can be easily extended to other targeting molecules and viral systems.


Subject(s)
Directed Molecular Evolution/methods , ErbB Receptors/metabolism , Receptors, Virus/metabolism , Sindbis Virus/growth & development , Sindbis Virus/genetics , Virology/methods , Virus Attachment , Cell Line , Cryoelectron Microscopy , ErbB Receptors/genetics , Humans , Receptors, Virus/genetics , Selection, Genetic , Sindbis Virus/physiology , Sindbis Virus/ultrastructure , Virion/ultrastructure
18.
Intervirology ; 56(5): 337-42, 2013.
Article in English | MEDLINE | ID: mdl-23838061

ABSTRACT

Transcription of the subgenomic mRNA of Sindbis virus (SINV) is initiated at a subgenomic promoter (SP). Alignment of SINV sequences identified a 68-nucleotide conserved domain spanning -19 to +49 relative to the subgenomic mRNA start site. Nucleotide T or C is present at -18 or +49 in all known SINVs while a Sindbis-like virus XJ-160 has an A or T at a corresponding position. Our results indicate that deletion or substitution of the T at +49 decreased the activity of SP, while substituting T for A at -18 did not decrease the activity of SP or genetic stability of recombinant SINV.


Subject(s)
Point Mutation , Promoter Regions, Genetic , Sequence Deletion , Sindbis Virus/growth & development , Sindbis Virus/genetics , Animals , Cell Line , Cricetinae , RNA, Messenger/biosynthesis , RNA, Viral/biosynthesis , Transcription, Genetic , Viral Load , Viral Plaque Assay
19.
J Virol ; 87(15): 8317-26, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23740988

ABSTRACT

Due to inactivation of the α1,3-galactosyltransferase gene (GGTA1, or the α1,3GT gene) approximately 28 million years ago, the carbohydrate αGal (Galα1,3Galß1,4GlcNAc) is not expressed on the cells of Old World monkeys and apes (including humans) but is expressed in all other mammals. The proposed selective advantage of this mutation for these primates is the ability to produce anti-Gal antibodies, which may be an effective immune component in neutralizing αGal-expressing pathogens. However, loss of α1,3GT expression may have been advantageous by providing natural resistance against viral pathogens that exploited the α1,3GT pathway or cell surface αGal for infection. Infections of paired cell lines with differential expression of α1,3GT showed that Sindbis viruses (SINV) preferentially replicate in α1,3GT-positive cells, whereas herpes simplex viruses type 1 and type 2 (HSV-1 and HSV-2) preferentially grow in cells lacking α1,3GT. Viral growth and spread correlated with the ability of the different viruses to successfully initiate infection in the presence or absence of α1,3GT expression. GT knockout (KO) suckling mice infected with SINV strains (AR339 and S.A.AR86) experienced significant delay in onset of disease symptoms and mortality compared to wild-type (WT) B6 suckling mice. In contrast, HSV-2-infected GT KO mice had higher viral titers in spleen and liver and exhibited significantly more focal hepatic necrosis than WT B6 mice. This study demonstrates that α1,3GT activity plays a role in the course of infections for certain viruses. Furthermore, this study has implications for the evolution of resistance to viral infections in primates.


Subject(s)
Disease Resistance , Evolution, Molecular , Galactosyltransferases/genetics , Receptors, Virus/genetics , Virus Diseases/immunology , Virus Internalization , Virus Physiological Phenomena , Alphavirus Infections/pathology , Alphavirus Infections/virology , Animals , Cell Line , Cercopithecidae , Disease Models, Animal , Female , Galactosyltransferases/metabolism , Gene Deletion , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/growth & development , Herpesvirus 1, Human/physiology , Herpesvirus 2, Human/growth & development , Herpesvirus 2, Human/physiology , Humans , Liver/pathology , Liver/virology , Male , Mice , Mice, Knockout , Receptors, Virus/metabolism , Selection, Genetic , Sindbis Virus/growth & development , Sindbis Virus/pathogenicity , Sindbis Virus/physiology , Spleen/pathology , Spleen/virology
20.
Antimicrob Agents Chemother ; 57(1): 155-67, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23275491

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-transmitted virus that has reemerged as a significant public health threat in the last decade. Since the 2005-2006 chikungunya fever epidemic in the Indian Ocean island of La Réunion, millions of people in more than 40 countries have been infected. Despite this, there is currently no antiviral treatment for chikungunya infection. In this study, an immunofluorescence-based screening platform was developed to identify potential inhibitors of CHIKV infection. A primary screen was performed using a highly purified natural product compound library, and 44 compounds exhibiting ≥70% inhibition of CHIKV infection were identified as positive hits. Among these, four were selected for dose-dependent inhibition assays to confirm their anti-CHIKV activity. Harringtonine, a cephalotaxine alkaloid, displayed potent inhibition of CHIKV infection (50% effective concentration [EC(50)] = 0.24 µM) with minimal cytotoxicity and was selected for elucidation of its antiviral mechanism. Time-of-addition studies, cotreatment assays, and direct transfection of viral genomic RNA indicated that harringtonine inhibited an early stage of the CHIKV replication cycle which occurred after viral entry into cells. In addition, quantitative reverse transcription-PCR (qRT-PCR) and Western blot analyses indicated that harringtonine affects CHIKV RNA production as well as viral protein expression. Treatment of harringtonine against Sindbis virus, a related alphavirus, suggested that harringtonine could inhibit other alphaviruses. This study suggests for the first time that harringtonine exerts its antiviral effects by inhibiting CHIKV viral protein synthesis.


Subject(s)
Antiviral Agents/pharmacology , Biological Products/pharmacology , Chikungunya virus/drug effects , Harringtonines/pharmacology , Protein Biosynthesis/drug effects , RNA, Viral/antagonists & inhibitors , Virus Replication/drug effects , Aedes , Animals , Antiviral Agents/isolation & purification , Biological Products/isolation & purification , Cell Line , Chikungunya virus/genetics , Chikungunya virus/growth & development , Cricetinae , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Gene Expression/drug effects , Harringtonines/isolation & purification , High-Throughput Screening Assays , Humans , RNA, Viral/genetics , Sindbis Virus/drug effects , Sindbis Virus/genetics , Sindbis Virus/growth & development , Small Molecule Libraries/isolation & purification , Small Molecule Libraries/pharmacology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...