Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Elife ; 122024 May 24.
Article in English | MEDLINE | ID: mdl-38787378

ABSTRACT

Severe dengue infections are characterized by endothelial dysfunction shown to be associated with the secreted nonstructural protein 1 (sNS1), making it an attractive vaccine antigen and biotherapeutic target. To uncover the biologically relevant structure of sNS1, we obtained infection-derived sNS1 (isNS1) from dengue virus (DENV)-infected Vero cells through immunoaffinity purification instead of recombinant sNS1 (rsNS1) overexpressed in insect or mammalian cell lines. We found that isNS1 appeared as an approximately 250 kDa complex of NS1 and ApoA1 and further determined the cryoEM structures of isNS1 and its complex with a monoclonal antibody/Fab. Indeed, we found that the major species of isNS1 is a complex of the NS1 dimer partially embedded in a high-density lipoprotein (HDL) particle. Crosslinking mass spectrometry studies confirmed that the isNS1 interacts with the major HDL component ApoA1 through interactions that map to the NS1 wing and hydrophobic domains. Furthermore, our studies demonstrated that the sNS1 in sera from DENV-infected mice and a human patient form a similar complex as isNS1. Our results report the molecular architecture of a biological form of sNS1, which may have implications for the molecular pathogenesis of dengue.


Subject(s)
Dengue Virus , Dengue , Lipoproteins, HDL , Viral Nonstructural Proteins , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Animals , Dengue Virus/genetics , Dengue Virus/metabolism , Chlorocebus aethiops , Mice , Humans , Lipoproteins, HDL/metabolism , Vero Cells , Dengue/virology , Dengue/metabolism , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/chemistry , Protein Multimerization , Cryoelectron Microscopy
2.
EBioMedicine ; 91: 104570, 2023 May.
Article in English | MEDLINE | ID: mdl-37068347

ABSTRACT

BACKGROUND: The Asian lineage Zika virus (ZIKV) emerged as a public health emergency in 2016 causing severe neurological pathologies with no apparent historical correlate to the mild, disease-causing innocuous member of the mosquito-borne flavivirus genus that was discovered in Africa in 1947. Replication error rate of RNA viruses combined with viral protein/RNA structural plasticity can lead to evolution of virus-induced pathogenicity that is critical to identify and validate. METHODS: Infection studies in cells and A129 interferon alpha/beta receptor deficient mice with ZIKV French Polynesian H/PF/2013 clinical isolate, plaque-purified isogenic clone derivatives as well as infectious cDNA clone derived wild-type and site-specific mutant viruses, were employed together with Next-Generation Sequencing (NGS) to pin-point the contributions of specific viral variants in neurovirulence recapitulated in our ZIKV mouse model. FINDINGS: NGS analysis of the low-passage inoculum virus as well as mouse serum, brain and testis derived virus, revealed specific enrichment in the mouse brain that were not found in the other tissues. Specifically, non-structural (NS) protein 2A variant at position 117 along with changes in NS1 and NS4B were uniquely associated with the mouse brain isolate. Mutational analysis of these variants in cDNA infectious clones identified the NS2A A117V as the lethal pathogenic determinant with potential epistatic contribution of NS1 and NS4B variants in ZIKV brain penetrance. INTERPRETATION: Our findings confirm that viral subpopulations drive ZIKV neuropathogenicity and identify specific sequence variants that expand in the mouse brain that associates with this phenotype which can serve as predictors of severe epidemics. FUNDING: Duke-NUS Khoo Post-doctoral Fellowship Award 2020 (KWKC) and National Medical Research Council of Singapore grants MOH-000524 (OFIRG) (SW) and MOH-OFIRG20nov-0002 (SGV).


Subject(s)
Zika Virus Infection , Zika Virus , Male , Chlorocebus aethiops , Animals , Mice , Zika Virus/genetics , Vero Cells , DNA, Complementary/genetics , DNA, Complementary/metabolism , Virus Replication , RNA, Viral/genetics , RNA, Viral/metabolism
3.
Virology ; 583: 1-13, 2023 06.
Article in English | MEDLINE | ID: mdl-37060797

ABSTRACT

Type I interferon (IFN-I) evasion by Dengue virus (DENV) is key in DENV pathogenesis. The non-structural protein 5 (NS5) antagonizes IFN-I response through the degradation of the signal transducer and activator of transcription 2 (STAT2). We developed a K562 cell-based platform, for high throughput screening of compounds potentially counteracting the NS5-mediated antagonism of IFN-I signaling. Upon a screening with a library of 1220 approved drugs, 3 compounds previously linked to DENV inhibition (Apigenin, Chrysin, and Luteolin) were identified. Luteolin and Apigenin determined a significant inhibition of DENV2 replication in Huh7 cells and the restoration of STAT2 phosphorylation in both cell systems. Apigenin and Luteolin were able to stimulate STAT2 even in the absence of infection. Despite the "promiscuous" and "pan-assay-interfering" nature of Luteolin, Apigenin promotes STAT2 Tyr 689 phosphorylation and activation, highlighting the importance of screening for compounds able to interact with host factors, to counteract viral proteins capable of dampening innate immune responses.


Subject(s)
Dengue Virus , Apigenin/pharmacology , Dengue Virus/physiology , Luteolin/pharmacology , Signal Transduction , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Humans
4.
Eur J Med Chem ; 252: 115283, 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-36965228

ABSTRACT

Dengue virus (DENV), a mosquito-borne flavivirus, continues to be a major public health threat in many countries and no approved antiviral therapeutics are available yet. In this work, we designed and synthesized a series of sulfonyl anthranilic acid (SAA) derivatives using a ligand-based scaffold morphing approach of the 2,1-benzothiazine 2,2-dioxide core, previously used by us to develop DENV polymerase inhibitors resulting devoid of any cell-based antiviral activity. Several derivatives based on the new SAA chemotype exhibited potent inhibition against DENV infection in the cell-based assay but did not inhibit DENV NS5 polymerase activity in the in vitro de novo initiation and elongation assays. Notably, best compounds 26 and 39 showed EC50 values in the range of 0.54-1.36 µM against cells infected with the four dengue serotypes (DENV-1-4). Time-of-drug-addition assay revealed that analogue 26 is a post-entry replication inhibitor that appears to be specific for cells of primate origin, implicating a host target with a high barrier to resistance. In conclusion, SAA derivatives offer a valuable starting point for developing effective Dengue antiviral therapeutics.


Subject(s)
Dengue Virus , Dengue , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Dengue/drug therapy , Serogroup , Virus Replication
5.
Antiviral Res ; 210: 105517, 2023 02.
Article in English | MEDLINE | ID: mdl-36592668

ABSTRACT

Flaviviruses are vector-borne pathogens capable of causing devastating human diseases. The re-emergence of Zika in 2016 notoriously led to a widescale epidemic in the Americas. New daunting evidence suggests that a single mutation in Zika virus genome may increase transmission and pathogenesis, further highlighting the need to be prepared for flavivirus outbreaks. Dengue, in particular infects about 400 million people each year, leading to reoccurring local outbreaks. Public health efforts to mitigate flavivirus transmission is largely dependent on vector control strategies, as only a limited number of flavivirus vaccines have been developed thus far. There are currently no commercially available antivirals for flaviviruses, leaving supportive care as the primary treatment option. In this review, we will briefly paint a broad picture of the flavivirus landscape in terms of therapeutics, with particular focus on viral targets, promising novel compounds entering the drug discovery pipeline, as well as model systems for evaluating drug efficacy.


Subject(s)
Flavivirus Infections , Flavivirus , Viral Vaccines , Zika Virus Infection , Zika Virus , Humans , Flavivirus/genetics , Zika Virus/genetics , Zika Virus Infection/drug therapy , Zika Virus Infection/prevention & control
6.
EBioMedicine ; 77: 103930, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35290828

ABSTRACT

BACKGROUND: Congenital disorders associated with prenatal vertical transmission of Zika virus (ZIKV) is well established since the 2016 outbreak in the Americas. However, despite clinical reports of similar mode of transmission for other flaviviruses such as dengue virus (DENV), the phenomenon has not been experimentally explored. METHODS: Pregnant AG129 mice were infected with DENV1 in the presence or absence of enhancing antibodies at different gestational time points. ZIKV was used for comparison. We quantified viral load in fetus and placentas and performed comprehensive gene expression profiling in the maternal (decidua) and fetal portion of placenta separately. FINDINGS: We demonstrate in a laboratory experimental setting that DENV can be transmitted vertically in a gestation stage-dependent manner similar to ZIKV, and this incidence drastically increases in the presence of enhancing antibodies. Interestingly, a high rate of DENV fetal infection occurs even though the placental viral load is significantly lower than that found in ZIKV-infected dams. Comprehensive gene expression profiling revealed DENV infection modulates a variety of inflammation-associated genes comparable to ZIKV in decidua and fetal placenta in early pregnancy. INTERPRETATION: Our findings suggest that the virus-induced modulation of host gene expression may facilitate DENV to cross the placental barrier in spite of lower viral burden compared to ZIKV. This mouse model may serve to identify the host determinants required for the vertical transmission of flaviviruses and develop appropriate countermeasures. FUNDING: National Medical Research Council/Open Fund Individual Research Grant MOH-000524 (SW), MOH-000086 and OFIRG20nov-0017 (SGV).


Subject(s)
Dengue Virus , Zika Virus Infection , Zika Virus , Animals , Antibodies, Viral , Female , Humans , Mice , Placenta , Pregnancy , Zika Virus/genetics
7.
RNA ; 28(2): 177-193, 2022 02.
Article in English | MEDLINE | ID: mdl-34759006

ABSTRACT

The commitment to replicate the RNA genome of flaviviruses without a primer involves RNA-protein interactions that have been shown to include the recognition of the stem-loop A (SLA) in the 5' untranslated region (UTR) by the nonstructural protein NS5. We show that DENV2 NS5 arginine 888, located within the carboxy-terminal 18 residues, is completely conserved in all flaviviruses and interacts specifically with the top-loop of 3'SL in the 3'UTR which contains the pentanucleotide 5'-CACAG-3' previously shown to be critical for flavivirus RNA replication. We present virological and biochemical data showing the importance of this Arg 888 in virus viability and de novo initiation of RNA polymerase activity in vitro. Based on our binding studies, we hypothesize that ternary complex formation of NS5 with 3'SL, followed by dimerization, leads to the formation of the de novo initiation complex that could be regulated by the reversible zipping and unzipping of cis-acting RNA elements.


Subject(s)
Dengue Virus/physiology , RNA/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication , 3' Untranslated Regions , Animals , Arginine/chemistry , Cell Line , Conserved Sequence , Cricetinae , Cricetulus , DNA-Directed RNA Polymerases/metabolism , Dengue Virus/genetics , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
8.
Antiviral Res ; 195: 105194, 2021 11.
Article in English | MEDLINE | ID: mdl-34699863

ABSTRACT

The flavivirus NS5 protein contains an N-terminal methyl-transferase (MTase) connected through a flexible linker with a C-terminal RNA-dependent RNA-polymerase (RdRp) domain, that work cooperatively to replicate and methylate the viral genome. In this study we probed the importance of an evolutionary-conserved hydrophobic residue (Val266) located at the start of the ten-residue interdomain linker of Zika virus (ZIKV) NS5. In flavivirus NS5 crystal structures, the start of the linker forms a 310 helix when NS5 adopts a compact conformation, but becomes disordered or extended in open conformations. Using reverse genetics system, we either introduced rigidity in the linker through mutation to a proline or flexibility through a glycine mutation at position 266. ZIKV NS5 Val 266 to Pro mutation was lethal for viral RNA replication while the Gly mutation was severely attenuated. Serial passaging of cell culture supernatant derived from C6/36 mosquito cells transfected with mutant ZIKV RNA showed that the attenuation can be rescued. Next generation deep sequencing revealed four single nucleotide polymorphisms that occur with an allele frequency >98%. The single non-synonymous NS5 mutation Glu419 to Lys is adjacent to RdRp motif G at the tip of the fingers subdomain, while the remaining three are synonymous variants at nucleotide positions 1403, 4403 and 6653 in the genome. Reverse engineering the changes into the ZIKV NS5/Val266Gly background followed by serial passaging revealed that residue 266 is under strong positive selection to revert back to Val. The interaction of the specific conformation of the NS5 linker with Val at position 266 and the RNA binding motif G region may present a potential strategy for allosteric antiviral drug development.


Subject(s)
Antiviral Agents/chemistry , Methyltransferases/chemistry , Viral Nonstructural Proteins/chemistry , Virus Replication/drug effects , Zika Virus/enzymology , Allosteric Site , Animals , Cell Line , Cricetinae , Crystallography, X-Ray , Drug Design , Methyltransferases/biosynthesis , Models, Molecular , Protein Binding , RNA-Dependent RNA Polymerase , Viral Nonstructural Proteins/biosynthesis , Zika Virus Infection
9.
Eur J Med Chem ; 224: 113695, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34298282

ABSTRACT

The flavivirus genus of the Flaviviridae family comprises Dengue, Zika and West-Nile viruses which constitute unmet medical needs as neither appropriate antivirals nor safe vaccines are available. The dengue NS2BNS3 protease is one of the most promising validated targets for developing a dengue treatment however reported protease inhibitors suffer from toxicity and cellular inefficacy. Here we report SAR on our previously reported Zika-active carbazole scaffold, culminating prodrug compound SP-471P (EC50 1.10 µM, CC50 > 100 µM) that generates SP-471; one of the most potent, non-cytotoxic and cell-active protease inhibitors described in the dengue literature. In cell-based assays, SP-471P leads to inhibition of viral RNA replication and complete abolishment of infective viral particle production even when administered 6 h post-infection. Mechanistically, SP-471 appears to inhibit both normal intermolecular protease processes and intramolecular cleavage events at the NS2BNS3 junction, as well as at NS3 internal sites, all critical for virus replication. These render SP-471 a unique to date multimodal inhibitor of the dengue protease.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Oximes/pharmacology , Peptide Hydrolases/metabolism , Prodrugs/pharmacology , Protease Inhibitors/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dengue Virus/enzymology , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Oximes/chemical synthesis , Oximes/chemistry , Prodrugs/chemical synthesis , Prodrugs/chemistry , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Structure-Activity Relationship
10.
Antiviral Res ; 185: 104991, 2021 01.
Article in English | MEDLINE | ID: mdl-33279522

ABSTRACT

In mouse models of dengue virus (DENV) infection, 18F-FDG PET is able to sensitively detect tissue-specific sites of inflammation and disease activity, as well as track therapeutic response to anti- DENV agents. However, the use of 18F-FDG PET to study the pathogenesis of inflammation and disease activity in DENV infection in humans, has not been clinically validated. Here we report the 18F-FDG PET imaging results of two patients during the febrile phase of acute DENV infection, paired with serial serum viral load, NS1 and proinflammatory cytokine measurements. Our findings demonstrate that 18F-FDG PET is able to sensitively detect and quantify organ-specific inflammation in the lymph nodes and spleen, in classic acute dengue fever. This raises the potential for 18F-FDG PET to be used as a research tool that may provide further insights into disease pathogenesis.


Subject(s)
Dengue/blood , Dengue/physiopathology , Fluorodeoxyglucose F18/metabolism , Inflammation/physiopathology , Positron Emission Tomography Computed Tomography/methods , Positron-Emission Tomography/methods , Acute Disease , Adult , Convalescence , Cytokines/analysis , Dengue/virology , Female , Humans , Inflammation/immunology , Lymph Nodes/pathology , Male , Middle Aged , Spleen/pathology , Viral Load
11.
J Gen Virol ; 101(9): 941-953, 2020 09.
Article in English | MEDLINE | ID: mdl-32589122

ABSTRACT

The dengue virus (DENV) replication complex is made up of its non-structural (NS) proteins and yet-to-be identified host proteins, but the molecular interactions between these proteins are not fully elucidated. In this work, we sought to uncover the interactions between DENV NS1 and its fellow NS proteins using a yeast two-hybrid (Y2H) approach, and found that domain II of NS1 binds to an N-terminal cytoplasmic fragment of NS4A. Mutations in amino acid residues 41 and 43 in this cytoplasmic region of NS4A disrupted the interaction between NS1 and the NS4A-2K-4B precursor protein. When the NS4A Y41F mutation was introduced into the context of the virus via a DENV2 infectious clone, this mutant virus exhibited impaired viral fitness and decreased infectious virus production. The NS4A Y41F mutant virus triggered a significantly muted transcriptional activation of interferon-stimulated genes compared to wild-type virus that is independent of NS4A's ability to antagonize type I interferon signalling. Taken together, we have identified a link between DENV NS1 and the cytoplasmic domain in NS4A that is important for its cellular and viral functions.


Subject(s)
Dengue Virus/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Dengue Virus/physiology , Genetic Fitness , Humans , Interferon Type I/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Two-Hybrid System Techniques , Viral Nonstructural Proteins/chemistry , Virion/metabolism , Virus Replication
12.
Cells ; 8(12)2019 11 26.
Article in English | MEDLINE | ID: mdl-31779251

ABSTRACT

The Zika virus (ZIKV) non-structural protein 5 (NS5) plays multiple viral and cellular roles during infection, with its primary role in virus RNA replication taking place in the cytoplasm. However, immunofluorescence assay studies have detected the presence of ZIKV NS5 in unique spherical shell-like structures in the nuclei of infected cells, suggesting potentially important cellular roles of ZIKV NS5 in the nucleus. Hence ZIKV NS5's subcellular distribution and localization must be tightly regulated during ZIKV infection. Both ZIKV NS5 expression or ZIKV infection antagonizes type I interferon signaling, and induces a pro-inflammatory transcriptional response in a cell type-specific manner, but the mechanisms involved and the role of nuclear ZIKV NS5 in these cellular functions has not been elucidated. Intriguingly, these cells originate from the brain and placenta, which are also organs that exhibit a pro-inflammatory signature and are known sites of pathogenesis during ZIKV infection in animal models and humans. Here, we discuss the regulation of the subcellular localization of the ZIKV NS5 protein, and its putative role in the induction of an inflammatory response and the occurrence of pathology in specific organs during ZIKV infection.


Subject(s)
Host-Pathogen Interactions/immunology , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/metabolism , Zika Virus Infection/immunology , Zika Virus Infection/virology , Zika Virus/physiology , Amino Acid Sequence , Cell Nucleus/metabolism , Humans , Immunity, Innate , Intracellular Space/metabolism , Protein Conformation , Protein Transport , Structure-Activity Relationship , Viral Nonstructural Proteins/chemistry
13.
Sci Transl Med ; 11(498)2019 06 26.
Article in English | MEDLINE | ID: mdl-31243154

ABSTRACT

Dengue viruses cause severe and sudden human epidemics worldwide. The secreted form of the nonstructural protein 1 (sNS1) of dengue virus causes vascular leakage, a hallmark of severe dengue disease. Here, we reverse engineered the T164S mutation of NS1, associated with the severity of dengue epidemics in the Americas, into a dengue virus serotype 2 mildly infectious strain. The T164S mutant virus decreased infectious virus production and increased sNS1 production in mammalian cell lines and human peripheral blood mononuclear cells (PBMCs) without affecting viral RNA replication. Gene expression profiling of 268 inflammation-associated human genes revealed up-regulation of genes induced in response to vascular leakage. Infection of the mosquito vector Aedes aegypti with the T164S mutant virus resulted in increased viral load in the mosquito midgut and higher sNS1 production compared to wild-type virus infection. Infection of type 1 and 2 interferon receptor-deficient AG129 mice with the T164S mutant virus resulted in severe disease coupled with increased complement activation, tissue inflammation, and more rapid mortality compared to AG129 mice infected with wild-type virus. Molecular dynamics simulations predicted that mutant sNS1 formed stable dimers similar to the wild-type protein, whereas the hexameric mutant sNS1 was predicted to be unstable. Immunoaffinity-purified sNS1 from T164S mutant virus-infected mammalian cells was associated with different lipid classes compared to wild-type sNS1. Treatment of human PBMCs with sNS1 purified from T164S mutant virus resulted in a twofold higher production of proinflammatory cytokines, suggesting a mechanism for how mutant sNS1 may cause more severe dengue disease.


Subject(s)
Dengue Virus/genetics , Dengue/pathology , Dengue/virology , Mutation/genetics , Severity of Illness Index , Viral Nonstructural Proteins/genetics , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , Culicidae/virology , Gene Expression Regulation , Inflammation/genetics , Kinetics , Leukocytes, Mononuclear/virology , Mice , Models, Molecular , Mutant Proteins/chemistry , Phylogeny , Protein Multimerization , Protein Stability , Viral Nonstructural Proteins/chemistry , Virus Replication
14.
FEBS Lett ; 593(12): 1272-1291, 2019 06.
Article in English | MEDLINE | ID: mdl-31090058

ABSTRACT

Zika virus (ZIKV) relies on its nonstructural protein 5 (NS5) for capping and synthesis of the viral RNA. Recent small-angle X-ray scattering (SAXS) data of recombinant ZIKV NS5 protein showed that it is dimeric in solution. Here, we present insights into the critical residues responsible for its dimer formation. SAXS studies of the engineered ZIKV NS5 mutants revealed that R681A mutation on NS5 (NS5R681A ) disrupts the dimer formation and affects its RNA-dependent RNA polymerase activity as well as the subcellular localization of NS5R681A in mammalian cells. The critical residues involved in the dimer arrangement of ZIKV NS5 are discussed, and the data provide further insights into the diversity of flaviviral NS5 proteins in terms of their propensity for oligomerization.


Subject(s)
Viral Nonstructural Proteins/metabolism , Zika Virus/metabolism , Amino Acid Sequence , Cell Line, Tumor , Dimerization , Humans , Mutation , Protein Conformation , Scattering, Small Angle , Sequence Homology, Amino Acid , Subcellular Fractions/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , X-Ray Diffraction
15.
Antiviral Res ; 167: 104-109, 2019 07.
Article in English | MEDLINE | ID: mdl-31051186

ABSTRACT

Zika virus (ZIKV) infection during pregnancy has been associated with adverse outcomes and birth defects such as microcephaly in newborn children. Congenital malformations associated with ZIKV are believed to occur via direct infection of the fetus. Unfortunately, there are no licensed therapeutic or preventative tools to block maternal-fetal transmission of ZIKV. In this study, we developed a mouse model of ZIKV infection that specifically establishes vertical maternal-fetal transmission of ZIKV in 40-60% of fetuses when the dams acquire ZIKV infection during pregnancy. This mouse model somewhat mirrors the experience in humans at the peak of the epidemic in the Americas. Using this model, we demonstrate that a well-documented directly acting antiviral (DAA) compound that targets flaviviral RNA synthesis can completely prevent fetal infection when the treatment is started at the time of infection. Notably, we show that the treatment commenced at the time of peak viremia is still able to reduce the risk of fetal infection concomitant with significant reduction in placental viral load. Our results show for the first time the potential for clinical development of antiviral drugs for preventing vertical maternal-fetal transmission of ZIKV.


Subject(s)
Adenosine/analogs & derivatives , Antiviral Agents/therapeutic use , Infectious Disease Transmission, Vertical , Nervous System Malformations/virology , Zika Virus Infection , Adenosine/therapeutic use , Animals , Disease Models, Animal , Female , Fetus/abnormalities , Fetus/virology , Humans , Mice , Microcephaly/virology , Nervous System Malformations/drug therapy , Nervous System Malformations/prevention & control , Pregnancy , Pregnancy Complications, Infectious , Viral Load/drug effects , Zika Virus/isolation & purification , Zika Virus Infection/drug therapy , Zika Virus Infection/transmission
16.
ACS Infect Dis ; 5(6): 932-948, 2019 06 14.
Article in English | MEDLINE | ID: mdl-30848123

ABSTRACT

The Zika virus (ZIKV) epidemic in the Americas was alarming because of its link with microcephaly in neonates and Guillain-Barré syndrome in adults. The unusual pathologies induced by ZIKV infection and the knowledge that the flaviviral nonstructural protein 5 (NS5), the most conserved protein in the flavivirus proteome, can modulate the host immune response during ZIKV infection prompted us to investigate the subcellular localization of NS5 during ZIKV infection and explore its functional significance. A monopartite nuclear localization signal (NLS) sequence within ZIKV NS5 was predicted by the cNLS Mapper program, and we observed localization of ZIKV NS5 in the nucleus of infected cells by immunostaining with specific antibodies. Strikingly, ZIKV NS5 forms spherical shell-like nuclear bodies that exclude DNA. The putative monopartite NLS 390KRPR393 is necessary to direct FLAG-tagged NS5 to the nucleus as the NS5 390ARPA393 mutant protein accumulates in the cytoplasm. Furthermore, coimmunostaining experiments reveal that NS5 localizes with and sequesters importin-α, but not importin-ß, in the observed nuclear bodies during virus infection. Structural and biochemical data demonstrate binding of ZIKV NS5 with importin-α and reveal important binding determinants required for their interaction and formation of complexes that give rise to the supramolecular nuclear bodies. Significantly, we demonstrate a neuronal-specific activation of the host immune response to ZIKV infection and a possible role of ZIKV NS5's nuclear localization toward this activation. This suggests that ZIKV pathogenesis may arise from a tissue-specific host response to ZIKV infection.


Subject(s)
Host Microbial Interactions/immunology , Neurons/immunology , Neurons/virology , Viral Nonstructural Proteins/metabolism , Zika Virus/immunology , alpha Karyopherins/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/virology , HEK293 Cells , Humans , Inflammation/genetics , Male , Mice , Protein Binding , Viral Nonstructural Proteins/genetics , Virus Replication , Zika Virus/genetics , Zika Virus/physiology
17.
J Infect Dis ; 219(2): 223-233, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30085051

ABSTRACT

Preexisting immunity to Zika virus (ZIKV) or dengue virus (DENV) may alter the course of their infection, and here we use robust mouse models to examine pathological outcomes following passive immunization, sequential cross-infection, or vaccination with inactivated virus. DENV infection was enhanced (through antibody-dependent enhancement [ADE]) or was suppressed by both DENV and ZIKV immunity. Notably, inactivated ZIKV vaccination enhanced dengue disease severity, although it was highly protective against ZIKV infection. On the other hand, ADE was not observed upon ZIKV infection in mice that were passively immunized or preinfected with DENV. Surprisingly, however, we found that vaccination with inactivated DENV enhanced ZIKV infection, mainly in the mesenteric lymph node, indicating the potential for DENV immunity to cause ADE in vivo. Collectively, our data call for greater attention to detail in the design of ZIKV or DENV vaccines.


Subject(s)
Cross Reactions/immunology , Dengue Virus/immunology , Dengue Virus/pathogenicity , Dengue/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Zika Virus/pathogenicity , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibody-Dependent Enhancement/immunology , Cell Line , Dengue/blood , Dengue/pathology , Dengue Vaccines , Dengue Virus/genetics , Disease Models, Animal , Genome, Viral , Humans , Immunity , Immunization , Lymph Nodes , Mice , THP-1 Cells , Vaccination , Virus Inactivation , Zika Virus/genetics , Zika Virus Infection/blood , Zika Virus Infection/pathology
18.
Antiviral Res ; 154: 87-96, 2018 06.
Article in English | MEDLINE | ID: mdl-29665375

ABSTRACT

We previously showed that luteolin, a well-known plant-derived component found in the "heat clearing" class of Traditional Chinese Medicine (TCM) herbs, is an uncompetitive inhibitor (Ki 58.6 µM) of the host proprotein convertase furin, an endoprotease that is required for maturation of flaviviruses in the trans-Golgi compartment. Luteolin also weakly inhibited recombinant dengue virus NS2B/NS3 protease (Ki 140.36 µM) non-competitively. In order to further explore the mechanism of inhibition we isolated resistant mutants by continuous passaging of DENV2 in the presence of increasing concentrations of luteolin. Nucleotide sequence analysis of the luteolin-resistant escape mutants revealed nucleotide changes that lead to amino acid substitutions in the prM (T79R) and NS2B (I114M) genes. These mutations were introduced into a DENV2 infectious clone and tested for replication in Huh-7 cells. Interestingly we found that the replication kinetics of prM T19R-NS2B I114M double-mutant (DM) was similar to wild-type virus (WT). On the other hand the prM T79R single mutant (SM1) was attenuated and the NS2B I114M single mutant (SM2) showed enhanced replication. Time of drug addition assay with luteolin showed that the mutant viruses were able to produce more mature virions than WT in the order DM > SM2>SM1>WT. Exogenous addition of furin to purified immature WT or mutant viruses revealed that luteolin blocked the prM cleavage of WT and SM2 at a similar level. On the other hand the SM1 immature virus showed some cleavage while the DM immature virus revealed efficient furin cleavage of prM even in the presence of 50 µM luteolin. Our findings suggest that luteolin inhibition of furin may occur at host/pathogen interface that permits the virus to escape the suppression by mutating key residue that may lead to an altered interface.


Subject(s)
Dengue Virus/drug effects , Dengue Virus/genetics , Luteolin/pharmacology , Viral Envelope Proteins/genetics , Viral Nonstructural Proteins/genetics , Virus Assembly/drug effects , Amino Acid Substitution , Cell Line, Tumor , Dengue/virology , Drug Resistance, Viral , Furin/pharmacology , Humans , Mutation , Nucleotides/genetics
19.
J Virol ; 92(7)2018 04 01.
Article in English | MEDLINE | ID: mdl-29321322

ABSTRACT

A primary question in dengue virus (DENV) biology is the molecular strategy for recruitment of host cell protein synthesis machinery. Here, we combined cell fractionation, ribosome profiling, and transcriptome sequencing (RNA-seq) to investigate the subcellular organization of viral genome translation and replication as well as host cell translation and its response to DENV infection. We report that throughout the viral life cycle, DENV plus- and minus-strand RNAs were highly partitioned to the endoplasmic reticulum (ER), identifying the ER as the primary site of DENV translation. DENV infection was accompanied by an ER compartment-specific remodeling of translation, where ER translation capacity was subverted from host transcripts to DENV plus-strand RNA, particularly at late stages of infection. Remarkably, translation levels and patterns in the cytosol compartment were only modestly affected throughout the experimental time course of infection. Comparisons of ribosome footprinting densities of the DENV plus-strand RNA and host mRNAs indicated that DENV plus-strand RNA was only sparsely loaded with ribosomes. Combined, these observations suggest a mechanism where ER-localized translation and translational control mechanisms, likely cis encoded, are used to repurpose the ER for DENV virion production. Consistent with this view, we found ER-linked cellular stress response pathways commonly associated with viral infection, namely, the interferon response and unfolded protein response, to be only modestly activated during DENV infection. These data support a model where DENV reprograms the ER protein synthesis and processing environment to promote viral survival and replication while minimizing the activation of antiviral and proteostatic stress response pathways.IMPORTANCE DENV, a prominent human health threat with no broadly effective or specific treatment, depends on host cell translation machinery for viral replication, immune evasion, and virion biogenesis. The molecular mechanism by which DENV commandeers the host cell protein synthesis machinery and the subcellular organization of DENV replication and viral protein synthesis is poorly understood. Here, we report that DENV has an almost exclusively ER-localized life cycle, with viral replication and translation largely restricted to the ER. Surprisingly, DENV infection largely affects only ER-associated translation, with relatively modest effects on host cell translation in the cytosol. DENV RNA translation is very inefficient, likely representing a strategy to minimize disruption of ER proteostasis. Overall these findings demonstrate that DENV has evolved an ER-compartmentalized life cycle; thus, targeting the molecular signatures and regulation of the DENV-ER interaction landscape may reveal strategies for therapeutic intervention.


Subject(s)
Dengue Virus/physiology , Dengue/immunology , Endoplasmic Reticulum/immunology , Immune Evasion , Protein Biosynthesis/immunology , RNA, Messenger/immunology , RNA, Viral/immunology , Virus Replication/immunology , Cell Line, Tumor , Dengue/pathology , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/virology , Humans , Interferons/immunology , Unfolded Protein Response/immunology
20.
Antiviral Res ; 143: 176-185, 2017 07.
Article in English | MEDLINE | ID: mdl-28389141

ABSTRACT

In many countries afflicted with dengue fever, traditional medicines are widely used as panaceas for illness, and here we describe the systematic evaluation of a widely known natural product, luteolin, originating from the "heat clearing" class of herbs. We show that luteolin inhibits the replication of all four serotypes of dengue virus, but the selectivity of the inhibition was weak. In addition, ADE-mediated dengue virus infection of human cell lines and primary PBMCs was inhibited. In a time-of-drug-addition study, luteolin was found to reduce infectious virus particle formation, but not viral RNA synthesis, in Huh-7 cells. During the virus life cycle, the host protease furin cleaves the pr moiety from prM protein of immature virus particles in the trans-Golgi network to produce mature virions. Analysis of virus particles from luteolin-treated cells revealed that prM was not cleaved efficiently. Biochemical interrogation of human furin showed that luteolin inhibited the enzyme activity in an uncompetitive manner, with Ki value of 58.6 µM, suggesting that treatment may restrict the virion maturation process. Luteolin also exhibited in vivo antiviral activity in mice infected with DENV, causing reduced viremia. Given the mode of action of luteolin and its widespread source, it is possible that it can be tested in combination with other dengue virus inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Furin/metabolism , Luteolin/antagonists & inhibitors , Proprotein Convertases/drug effects , Virus Replication/drug effects , A549 Cells , Animals , Antiviral Agents/chemistry , Cell Line , Cell Survival/drug effects , Cricetinae , DNA Replication/drug effects , Dengue/drug therapy , Dengue/virology , Dengue Virus/classification , Dengue Virus/genetics , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Enzyme Activation/drug effects , HEK293 Cells , Humans , Kinetics , Luteolin/administration & dosage , Luteolin/chemistry , Male , Mice , Proprotein Convertases/metabolism , RNA, Viral/drug effects , Viremia/drug therapy , Virion/drug effects , trans-Golgi Network/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...