Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 51
1.
Free Radic Biol Med ; 213: 266-273, 2024 03.
Article En | MEDLINE | ID: mdl-38278309

Yellow fever (YF) presents a wide spectrum of severity, with clinical manifestations in humans ranging from febrile and self-limited to fatal cases. Although YF is an old disease for which an effective and safe vaccine exists, little is known about the viral- and host-specific mechanisms that contribute to liver pathology. Several studies have demonstrated that oxidative stress triggered by viral infections contributes to pathogenesis. We evaluated whether yellow fever virus (YFV), when infecting human hepatocytes cells, could trigger an imbalance in redox homeostasis, culminating in oxidative stress. YFV infection resulted in a significant increase in reactive oxygen species (ROS) levels from 2 to 4 days post infection (dpi). When measuring oxidative parameters at 4 dpi, YFV infection caused oxidative damage to lipids, proteins, and DNA, evidenced by an increase in lipid peroxidation/8-isoprostane, carbonyl protein, and 8-hydroxy-2'-deoxyguanosine, respectively. Furthermore, there was a significant reduction in the activity of the antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GPx), in addition to a reduction in the ratio of reduced to oxidized glutathione (GSH/GSSG), indicating a pro-oxidant environment. However, no changes were observed in the enzymatic activity of the enzyme catalase (CAT) or in the gene expression of SOD isoforms (1/2/3), CAT, or GPx. Therefore, our results show that YFV infection generates an imbalance in redox homeostasis, with the overproduction of ROS and depletion of antioxidant enzymes, which induces oxidative damage to cellular constituents. Moreover, as it has been demonstrated that oxidative stress is a conspicuous event in YFV infection, therapeutic strategies based on antioxidant biopharmaceuticals may be new targets for the treatment of YF.


Antioxidants , Yellow Fever , Humans , Antioxidants/metabolism , Reactive Oxygen Species/metabolism , Yellow fever virus/metabolism , Glutathione/metabolism , Oxidative Stress , Oxidation-Reduction , Catalase/genetics , Catalase/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Glutathione Disulfide/metabolism , Hepatocytes/metabolism , Lipid Peroxidation , Glutathione Peroxidase/metabolism , 8-Hydroxy-2'-Deoxyguanosine/metabolism
2.
Viruses ; 15(7)2023 06 23.
Article En | MEDLINE | ID: mdl-37515107

As obligate intracellular parasites, viruses rely heavily on host cells for replication, and therefore dysregulate several cellular processes for their benefit. In return, host cells activate multiple signaling pathways to limit viral replication and eradicate viruses. The present study explores the complex interplay between viruses and host cells through next generation RNA sequencing as well as mass spectrometry (SILAC). Both the coding transcriptome and the proteome of human brain-derived U87 cells infected with Kunjin virus, Zika virus, or Yellow Fever virus were compared to the transcriptome and the proteome of mock-infected cells. Changes in the abundance of several hundred mRNAs and proteins were found in each infection. Moreover, the alternative splicing of hundreds of mRNAs was found to be modulated upon viral infection. Interestingly, a significant disconnect between the changes in the transcriptome and those in the proteome of infected cells was observed. These findings provide a global view of the coding transcriptome and the proteome of Flavivirus-infected cells, leading to a better comprehension of Flavivirus-host interactions.


Flavivirus , West Nile virus , Yellow Fever , Zika Virus Infection , Zika Virus , Humans , Zika Virus/genetics , Zika Virus/metabolism , West Nile virus/genetics , Yellow fever virus/genetics , Yellow fever virus/metabolism , Proteome/genetics , Transcriptome , Flavivirus/genetics , Virus Replication , Brain/metabolism
3.
J Virol ; 97(4): e0194922, 2023 04 27.
Article En | MEDLINE | ID: mdl-37017533

Genome cyclization is essential for viral RNA (vRNA) replication of the vertebrate-infecting flaviviruses, and yet its regulatory mechanisms are not fully understood. Yellow fever virus (YFV) is a notorious pathogenic flavivirus. Here, we demonstrated that a group of cis-acting RNA elements in YFV balance genome cyclization to govern efficient vRNA replication. It was shown that the downstream of the 5'-cyclization sequence hairpin (DCS-HP) is conserved in the YFV clade and is important for efficient YFV propagation. By using two different replicon systems, we found that the function of the DCS-HP is determined primarily by its secondary structure and, to a lesser extent, by its base-pair composition. By combining in vitro RNA binding and chemical probing assays, we found that the DCS-HP orchestrates the balance of genome cyclization through two different mechanisms, as follows: the DCS-HP assists the correct folding of the 5' end in a linear vRNA to promote genome cyclization, and it also limits the overstabilization of the circular form through a potential crowding effect, which is influenced by the size and shape of the DCS-HP structure. We also provided evidence that an A-rich sequence downstream of the DCS-HP enhances vRNA replication and contributes to the regulation of genome cyclization. Interestingly, diversified regulatory mechanisms of genome cyclization, involving both the downstream of the 5'-cyclization sequence (CS) and the upstream of the 3'-CS elements, were identified among different subgroups of the mosquito-borne flaviviruses. In summary, our work highlighted how YFV precisely controls the balance of genome cyclization to ensure viral replication. IMPORTANCE Yellow fever virus (YFV), the prototype of the Flavivirus genus, can cause devastating yellow fever disease. Although it is preventable by vaccination, there are still tens of thousands of yellow fever cases per year, and no approved antiviral medicine is available. However, the understandings about the regulatory mechanisms of YFV replication are obscure. In this study, by a combination of bioinformatics, reverse genetics, and biochemical approaches, it was shown that the downstream of the 5'-cyclization sequence hairpin (DCS-HP) promotes efficient YFV replication by modulating the conformational balance of viral RNA. Interestingly, we found specialized combinations for the downstream of the 5'-cyclization sequence (CS) and upstream of the 3'-CS elements in different groups of the mosquito-borne flaviviruses. Moreover, possible evolutionary relationships among the various downstream of the 5'-CS elements were implied. This work highlighted the complexity of RNA-based regulatory mechanisms in the flaviviruses and will facilitate the design of RNA structure-targeted antiviral therapies.


Virus Replication , Yellow fever virus , Animals , Humans , Cyclization , RNA, Viral/metabolism , Virus Replication/genetics , Yellow Fever/virology , Yellow fever virus/metabolism , Genome, Viral/genetics , Cell Line , Cricetinae , Mesocricetus , A549 Cells
4.
Emerg Microbes Infect ; 10(1): 1739-1750, 2021 Dec.
Article En | MEDLINE | ID: mdl-34379047

Yellow fever virus (YFV) is a re-emerging flavivirus, which can lead to severe clinical manifestations and high mortality, with no specific antiviral therapies available. The live-attenuated yellow fever vaccine 17D (YF17D) has been widely used for over eighty years. However, the emergence of yellow fever vaccine-associated viscerotropic disease (YFL-AVD) and yellow fever vaccine-associated neurotropic disease (YFL-AND) raised non-negligible concerns. Additionally, the attenuation mechanism of YF17D is still unclear. Thus, the development of convenient models is crucial to understand the mechanisms behind YF17D attenuation and its adverse effects. In this work, we generated a reporter YF17D expressing nano-luciferase (NLuc). In vitro and in vivo characterization demonstrated that the NLuc-YF17D shared similar biological properties with its parental strain and the NLuc activity can reflect viral infectivity reliably. Combined with in vivo bioluminescence imaging, a series of mice models of YF17D infection was established, which will be useful for the evaluation of antiviral medicines and novel vaccine candidates. Especially, we demonstrated that intraperitoneally (i.p.) infection of NLuc-YF17D in type I interferon receptor-deficient mice A129 resulted in outcomes resembling YEL-AVD and YEL-AND, evidenced by viral replication in multiple organs and invasion of the central neuronal system. Finally, in vitro and in vivo assays based on this reporter virus were established to evaluate the antiviral activities of validated antiviral agents. In conclusion, the bioluminescent reporter virus described herein provides a powerful platform to study YF17D attenuation and vaccine-associated diseases as well as to develop novel countermeasures against YFV.


Luminescent Measurements/methods , Yellow Fever/virology , Yellow fever virus/metabolism , Animals , Cell Line , Imaging, Three-Dimensional/methods , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Virus Replication , Yellow fever virus/genetics
5.
Sci Rep ; 11(1): 6081, 2021 03 16.
Article En | MEDLINE | ID: mdl-33727688

Although there are many studies on the control of mosquito vectors of the yellow fever virus (YFV) in tropical forests, there are still few ecological studies regarding abiotic factors effect on these mosquitoes. Here we characterize these effects on oviposition behavior, abundance, and diversity of mosquito vectors of YFV. The study was conducted in Córrego da Luz Municipal Park, in Casimiro de Abreu, Rio de Janeiro state, Brazil, from July 2018 to December 2019. Ovitraps were placed at ground level and 3 m high. The data were tested for normality using the Shapiro-Wilk test, followed by an independent sample analysis, the Mann-Whitney test. The Shannon Diversity Index was used to evaluate the abundance of mosquitos' eggs collected at both ground level and 3 m high. We highlight the presence of Haemagogus janthinomys and Hg. leucocelaenus, primary YFV vectors in forest areas. The abundance of Hg. leucocelaenus (63%), Hg. janthinomys (75%), and Aedes terrens (58%) was higher at the height of 3 m, while Ae. albopictus (52%) was higher at ground level. Aedes albopictus was positively correlated with temperature. Culicidae monitoring is essential for assessing the YFV transmission cycle in Atlantic forest fragments.


Culicidae/physiology , Mosquito Vectors/physiology , Oviposition , Seasons , Aedes/anatomy & histology , Aedes/physiology , Aedes/virology , Animals , Brazil/epidemiology , Culicidae/anatomy & histology , Culicidae/virology , Female , Mosquito Vectors/anatomy & histology , Mosquito Vectors/virology , Yellow Fever/epidemiology , Yellow Fever/transmission , Yellow fever virus/metabolism
6.
Virus Res ; 294: 198291, 2021 03.
Article En | MEDLINE | ID: mdl-33388393

Yellow fever virus, the prototype in the genus Flavivirus, was used to develop viruses in which the nonstructural protein NS1 is genetically fused to GFP in the context of viruses capable of autonomous replication. The GFP-tagging of NS1 at the amino-terminus appeared possible despite the presence of a small and functionally important domain at the NS1's amino-terminus which can be distorted by such fusing. GFP-tagged NS1 viruses were rescued from DNA-launched molecular clones. The initially produced GFP-tagged NS1 virus was capable of only poor replication. Sequential passages of the virus in cell cultures resulted in the appearance of mutations in GFP, NS4A, NS4B and NS5. The mutations which change amino acid sequences of GFP, NS4A and NS5 have the adaptive effect on the replication of GFP-tagged NS1 viruses. The pattern of GFP-fluorescence indicates that the GFP-NS1 fusion protein is produced into the endoplasmic reticulum. The intracellular GFP-NS1 fusion protein colocalizes with dsRNA. The discovered forms of extracellular GFP-NS1 possibly include tetramers and hexamers.


Flavivirus , Yellow fever virus , Amino Acid Sequence , Flavivirus/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication , Yellow fever virus/genetics , Yellow fever virus/metabolism
7.
Viruses ; 12(11)2020 11 12.
Article En | MEDLINE | ID: mdl-33198381

Yellow fever virus (YFV) is a mosquito-borne member of the genus flavivirus, including other important human-pathogenic viruses, such as dengue, Japanese encephalitis, and Zika. Herein, we report identifying 129 YFV Class II epitopes in donors vaccinated with the live attenuated YFV vaccine (YFV-17D). A total of 1156 peptides predicted to bind 17 different common HLA-DRB1 allelic variants were tested using IFNγ ELISPOT assays in vitro re-stimulated peripheral blood mononuclear cells from twenty-six vaccinees. Overall, we detected responses against 215 YFV epitopes. We found that the capsid and envelope proteins, as well as the non-structural (NS) proteins NS3 and NS5, were the most targeted proteins by CD4+ T cells from YF-VAX vaccinated donors. In addition, we designed and validated by flow cytometry a CD4+ mega pool (MP) composed of structural and non-structural epitopes in an independent cohort of vaccinated donors. Overall, this study provides a comprehensive prediction and validation of YFV epitopes in a cohort of YF-17D vaccinated individuals. With the design of a CD4 epitope MP, we further provide a useful tool to detect ex vivo responses of YFV-specific CD4 T cells in small sample volumes.


Epitopes/immunology , Histocompatibility Antigens Class II/immunology , Yellow Fever/immunology , Yellow Fever/virology , Yellow fever virus/immunology , Alleles , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Epitopes/chemistry , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Humans , Immunophenotyping , Peptides , Protein Binding , T-Cell Antigen Receptor Specificity , Vaccination , Viral Proteins/immunology , Yellow Fever Vaccine/immunology , Yellow fever virus/metabolism
8.
J Virol ; 94(22)2020 10 27.
Article En | MEDLINE | ID: mdl-32878892

Yellow fever virus (YFV) is an RNA virus primarily targeting the liver. Severe YF cases are responsible for hemorrhagic fever, plausibly precipitated by excessive proinflammatory cytokine response. Pathogen recognition receptors (PRRs), such as the cytoplasmic retinoic acid inducible gene I (RIG-I)-like receptors (RLRs), and the viral RNA sensor protein kinase R (PKR), are known to initiate a proinflammatory response upon recognition of viral genomes. Here, we sought to reveal the main determinants responsible for the acute cytokine expression occurring in human hepatocytes following YFV infection. Using a RIG-I-defective human hepatoma cell line, we found that RIG-I largely contributes to cytokine secretion upon YFV infection. In infected RIG-I-proficient hepatoma cells, RIG-I was localized in stress granules. These granules are large aggregates of stalled translation preinitiation complexes known to concentrate RLRs and PKR and are so far recognized as hubs orchestrating RNA virus sensing. Stable knockdown of PKR in hepatoma cells revealed that PKR contributes to both stress granule formation and cytokine induction upon YFV infection. However, stress granule disruption did not affect the cytokine response to YFV infection, as assessed by small interfering RNA (siRNA)-knockdown-mediated inhibition of stress granule assembly. Finally, no viral RNA was detected in stress granules using a fluorescence in situ hybridization approach coupled with immunofluorescence. Our findings suggest that both RIG-I and PKR mediate proinflammatory cytokine induction in YFV-infected hepatocytes, in a stress granule-independent manner. Therefore, by showing the uncoupling of the cytokine response from the stress granule formation, our model challenges the current view in which stress granules are required for the mounting of the acute antiviral response.IMPORTANCE Yellow fever is a mosquito-borne acute hemorrhagic disease caused by yellow fever virus (YFV). The mechanisms responsible for its pathogenesis remain largely unknown, although increased inflammation has been linked to worsened outcome. YFV targets the liver, where it primarily infects hepatocytes. We found that two RNA-sensing proteins, RIG-I and PKR, participate in the induction of proinflammatory mediators in human hepatocytes infected with YFV. We show that YFV infection promotes the formation of cytoplasmic structures, termed stress granules, in a PKR- but not RIG-I-dependent manner. While stress granules were previously postulated to be essential platforms for immune activation, we found that they are not required for the production of proinflammatory mediators upon YFV infection. Collectively, our work uncovered molecular events triggered by the replication of YFV, which could prove instrumental in clarifying the pathogenesis of the disease, with possible repercussions for disease management.


DEAD Box Protein 58/metabolism , Yellow fever virus/metabolism , eIF-2 Kinase/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinoma, Hepatocellular , Cell Line , Cell Line, Tumor , Cytokines/metabolism , DEAD Box Protein 58/deficiency , DEAD Box Protein 58/genetics , DNA Helicases/genetics , Gene Knockdown Techniques , Haplorhini , Hepatocytes/virology , Humans , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , RNA, Small Interfering , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Receptors, Immunologic , T-Cell Intracellular Antigen-1/genetics , Transcriptome , eIF-2 Kinase/genetics
9.
Talanta ; 208: 120338, 2020 Feb 01.
Article En | MEDLINE | ID: mdl-31816752

Arboviruses have been emerging as a significant global health problem due to the recurrent epidemics. Arboviruses require the development of new diagnostic devices due to the nonspecific clinical manifestations. Herein, we report a biosensor based on cysteine (Cys), zinc oxide nanoparticles (ZnONp), and Concanavalin A (ConA) lectin to differentiate between arboviruses infections. ConA is capable of interacting with the saccharide components of the viral capsid. In this study, we evaluated the reproducibility, sensitivity, and specificity of the sensor for the virus of Dengue type 2 (DENV2), Zika (ZIKV), Chikungunya (CHIKV), and Yellow fever (YFV). Atomic force microscopy measurements confirmed the electrode surface modification and revealed a heterogeneous topography during the biorecognition process. Cyclic voltammetry (CV) and impedance spectroscopy (EIS) were used to characterize the biosensor. The blockage of the oxidation-reduction process is related to the formation of Cys-ZnONp-ConA system on the electroactive area and its subsequent interaction with viral glycoproteins. The sensor exhibited a linear response to different concentrations of the studied arboviruses. Our study demonstrates that ConA lectin recognizes the structural glycoproteins of the DENV2, ZIKV, CHIKV, and YFV. DENV2 is the most structurally similar to ZIKV. Our results have shown that the impedimetric response correlates with the structural glycoproteins, as follow: DENV2 (18.6 kΩ) > ZIKV (14.6 kΩ) > CHIKV (6.86 kΩ) > YFV (5.98 kΩ). The homologous structural regions contribute to ConA-arboviruses recognition. Our results demonstrate the use of the proposed system for the development of biosensors for arboviruses infections.


Arbovirus Infections/diagnosis , Arboviruses/metabolism , Biosensing Techniques/methods , Concanavalin A/chemistry , Electrochemistry/methods , Electrodes , Metal Nanoparticles/chemistry , Arbovirus Infections/blood , Arbovirus Infections/virology , Arboviruses/isolation & purification , Chikungunya Fever/blood , Chikungunya Fever/diagnosis , Chikungunya Fever/virology , Chikungunya virus/isolation & purification , Chikungunya virus/metabolism , Cysteine/chemistry , Dengue/blood , Dengue/diagnosis , Dengue/virology , Dengue Virus/isolation & purification , Dengue Virus/metabolism , Diagnosis, Differential , Glucose/analysis , Humans , Mannose/analysis , Yellow Fever/blood , Yellow Fever/diagnosis , Yellow Fever/virology , Yellow fever virus/isolation & purification , Yellow fever virus/metabolism , Zika Virus/isolation & purification , Zika Virus/metabolism , Zika Virus Infection/blood , Zika Virus Infection/diagnosis , Zika Virus Infection/virology , Zinc Oxide/chemistry
10.
Antiviral Res ; 169: 104536, 2019 09.
Article En | MEDLINE | ID: mdl-31202975

Yellow fever virus (YFV) is responsible for devastating outbreaks of Yellow fever (YF) in humans and is associated with high mortality rates. Recent large epidemics and epizootics and exponential increases in the numbers of YF cases in humans and non-human primates highlight the increasing threat YFV poses, despite the availability of an effective YFV vaccine. YFV is the first human virus discovered, but the structures of several of the viral proteins remain poorly understood. Here we report the structure of the full-length NS5 protein, a key enzyme for the replication of flaviviruses that contains both a methyltransferase domain and an RNA dependent RNA polymerase domain, at 3.1 Šresolution. The viral polymerase adopts right-hand fold, demonstrating the similarities of the Yellow fever, Dengue and Zika polymerases. Together this data suggests NS5 as a prime and ideal target for the design of pan-flavivirus inhibitors.


Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/drug effects , Yellow Fever/virology , Yellow fever virus/drug effects , Yellow fever virus/metabolism , Animals , Binding Sites , Dengue , Disease Outbreaks , Drug Design , Methyltransferases/chemistry , Models, Molecular , Primates , Protein Conformation , Protein Domains , RNA-Dependent RNA Polymerase/chemistry , Viral Nonstructural Proteins/genetics , Virus Replication/drug effects , Yellow Fever Vaccine , Yellow fever virus/genetics , Zika Virus Infection , Zinc
11.
Cell Rep ; 26(6): 1598-1613.e8, 2019 02 05.
Article En | MEDLINE | ID: mdl-30726741

Flaviviruses cause systemic or neurotropic-encephalitic pathology in humans. The flavivirus nonstructural protein 1 (NS1) is a secreted glycoprotein involved in viral replication, immune evasion, and vascular leakage during dengue virus infection. However, the contribution of secreted NS1 from related flaviviruses to viral pathogenesis remains unknown. Here, we demonstrate that NS1 from dengue, Zika, West Nile, Japanese encephalitis, and yellow fever viruses selectively binds to and alters permeability of human endothelial cells from lung, dermis, umbilical vein, brain, and liver in vitro and causes tissue-specific vascular leakage in mice, reflecting the pathophysiology of each flavivirus. Mechanistically, each flavivirus NS1 leads to differential disruption of endothelial glycocalyx components, resulting in endothelial hyperpermeability. Our findings reveal the capacity of a secreted viral protein to modulate endothelial barrier function in a tissue-specific manner both in vitro and in vivo, potentially influencing virus dissemination and pathogenesis and providing targets for antiviral therapies and vaccine development.


Dengue Virus/genetics , Endothelial Cells/virology , Glycocalyx/virology , Viral Nonstructural Proteins/genetics , Animals , Brain/pathology , Brain/virology , Cell Line , Cell Membrane Permeability , Dengue/genetics , Dengue/metabolism , Dengue/pathology , Dengue Virus/metabolism , Dengue Virus/pathogenicity , Dermis/pathology , Dermis/virology , Encephalitis Virus, Japanese/genetics , Encephalitis Virus, Japanese/metabolism , Encephalitis Virus, Japanese/pathogenicity , Endothelial Cells/pathology , Gene Expression , Glycocalyx/chemistry , Humans , Liver/pathology , Liver/virology , Lung/pathology , Lung/virology , Male , Mice , Organ Specificity , Primary Cell Culture , Umbilical Veins/pathology , Umbilical Veins/virology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Virus Replication , West Nile virus/genetics , West Nile virus/metabolism , West Nile virus/pathogenicity , Yellow fever virus/genetics , Yellow fever virus/metabolism , Yellow fever virus/pathogenicity , Zika Virus/genetics , Zika Virus/metabolism , Zika Virus/pathogenicity
12.
Cell Host Microbe ; 16(3): 269-71, 2014 Sep 10.
Article En | MEDLINE | ID: mdl-25211068

Given the potency of interferon-α/ß, viral evasion of this pathway is crucial for infection. In this issue of Cell Host & Microbe, Laurent-Rolle et al. (2014) report that during yellow fever virus infection, interferon-α/ß stimulates the polyubiquitination of viral NS5, which binds to STAT2 and inhibits transcription of interferon-stimulated genes.


Interferon-beta/metabolism , Viral Nonstructural Proteins/metabolism , Yellow Fever/metabolism , Yellow fever virus/metabolism , Animals , Humans
13.
Cell Host Microbe ; 16(3): 314-327, 2014 Sep 10.
Article En | MEDLINE | ID: mdl-25211074

To successfully establish infection, flaviviruses have to overcome the antiviral state induced by type I interferon (IFN-I). The nonstructural NS5 proteins of several flaviviruses antagonize IFN-I signaling. Here we show that yellow fever virus (YFV) inhibits IFN-I signaling through a unique mechanism that involves binding of YFV NS5 to the IFN-activated transcription factor STAT2 only in cells that have been stimulated with IFN-I. This NS5-STAT2 interaction requires IFN-I-induced tyrosine phosphorylation of STAT1 and the K63-linked polyubiquitination at a lysine in the N-terminal region of YFV NS5. We identified TRIM23 as the E3 ligase that interacts with and polyubiquitinates YFV NS5 to promote its binding to STAT2 and trigger IFN-I signaling inhibition. Our results demonstrate the importance of YFV NS5 in overcoming the antiviral action of IFN-I and offer a unique example of a viral protein that is activated by the same host pathway that it inhibits.


Interferon-beta/metabolism , Viral Nonstructural Proteins/metabolism , Yellow Fever/metabolism , Yellow fever virus/metabolism , Amino Acid Motifs , Animals , Cell Line , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Host-Pathogen Interactions , Humans , Phosphorylation , Protein Binding , STAT1 Transcription Factor/chemistry , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Signal Transduction , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Yellow Fever/genetics , Yellow Fever/virology , Yellow fever virus/genetics
14.
J Biomol Screen ; 19(8): 1147-53, 2014 Sep.
Article En | MEDLINE | ID: mdl-24793430

RNA-protein interactions are vital to the replication of the flaviviral genome. Discovery focused on small molecules that disrupt these interactions represent a viable path for identification of new inhibitors. The viral RNA (vRNA) cap methyltransferase (MTase) of the flaviviruses has been validated as a suitable drug target. Here we report the development of a high-throughput screen for the discovery of compounds that target the RNA binding site of flaviviral protein NS5A. The assay described here is based on displacement of an MT-bound polynucleotide aptamer, decathymidylate derivatized at its 5' end with fluorescein (FL-dT10). Based on the measurement of fluorescence polarization, FL-dT10 bound to yellow fever virus (YFV) MTase in a saturable manner with a Kd= 231 nM. The binding was reversed by a 250-nucleotide YFV messenger RNA (mRNA) transcript and by the triphenylmethane dye aurintricarboxylic acid (ATA). The EC50for ATA displacement was 1.54 µM. The MTase cofactors guanosine-5'-triphosphate and S-adenosyl-methionine failed to displace FL-dT10. Analysis by electrophoretic mobility shift assay (EMSA) suggests that ATA binds YFV MTase so as to displace the vRNA. The assay was determined to have a Z' of 0.83 and was successfully used to screen a library of known bioactives.


Aptamers, Nucleotide , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Flavivirus/enzymology , Methyltransferases/antagonists & inhibitors , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Aurintricarboxylic Acid/pharmacology , Binding Sites , Enzyme Inhibitors/chemistry , Fluorescence Polarization , Guanosine Triphosphate/metabolism , High-Throughput Screening Assays/methods , Molecular Targeted Therapy/methods , Oligodeoxyribonucleotides/metabolism , RNA Caps , RNA, Messenger , Small Molecule Libraries/pharmacology , Viral Nonstructural Proteins/metabolism , Yellow fever virus/genetics , Yellow fever virus/metabolism
15.
Biosci Rep ; 33(1): 113-24, 2012 Dec 20.
Article En | MEDLINE | ID: mdl-23137297

The human RPSA [ribosomal protein SA; also known as LamR1(laminin receptor 1)] belongs to the ribosome but is also a membrane receptor for laminin, growth factors, prion, pathogens and the anticarcinogen EGCG (epigallocatechin-gallate). It contributes to the crossing of the blood-brain barrier by neurotropic viruses and bacteria, and is a biomarker of metastasis. RPSA includes an N-terminal domain, which is folded and homologous to the prokaryotic RPS2, and a C-terminal extension, which is intrinsically disordered and conserved in vertebrates. We used recombinant derivatives of RPSA and its N- and C-domains to quantify its interactions with ligands by in-vitro immunochemical and spectrofluorimetric methods. Both N- and C-domains bound laminin with K(D) (dissociation constants) of 300 nM. Heparin bound only to the N-domain and competed for binding to laminin with the negatively charged C-domain, which therefore mimicked heparin. EGCG bound only to the N-domain with a K(D) of 100 nM. Domain 3 of the envelope protein from yellow fever virus and serotypes-1 and -2 of dengue virus bound preferentially to the C-domain whereas that from West Nile virus bound only to the N-domain. Our quantitative in-vitro approach should help clarify the mechanisms of action of RPSA, and ultimately fight against cancer and infectious agents.


Cell Membrane/metabolism , Protein Folding , Protein Interaction Mapping/methods , Receptors, Laminin/metabolism , Ribosomal Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Escherichia coli/metabolism , Heparin/metabolism , Humans , Immunochemistry , Laminin/metabolism , Ligands , Protein Binding , Protein Structure, Tertiary , Receptors, Laminin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribosomal Proteins/genetics , Spectrometry, Fluorescence/methods , Tryptophan/metabolism , Viral Envelope Proteins/metabolism , West Nile virus/metabolism , Yellow fever virus/metabolism
16.
PLoS One ; 6(12): e27753, 2011.
Article En | MEDLINE | ID: mdl-22163273

BACKGROUND: Yellow fever vaccination (YF-17D) can cause serious adverse events (SAEs). The mechanism of these SAEs is poorly understood. Older age has been identified as a risk factor. We tested the hypothesis that the humoral immune response to yellow fever vaccine develops more slowly in elderly than in younger subjects. METHOD: We vaccinated young volunteers (18-28 yrs, N = 30) and elderly travelers (60-81 yrs, N = 28) with YF-17D and measured their neutralizing antibody titers and plasma YF-17D RNA copy numbers before vaccination and 3, 5, 10, 14 and 28 days after vaccination. RESULTS: Ten days after vaccination seroprotection was attained by 77% (23/30) of the young participants and by 50% (14/28) of the elderly participants (p = 0.03). Accordingly, the Geometric Mean Titer of younger participants was higher than the GMT of the elderly participants. At day 10 the difference was +2.9 IU/ml (95% CI 1.8-4.7, p = 0.00004) and at day 14 +1.8 IU/ml (95% CI 1.1-2.9, p = 0.02, using a mixed linear model. Viraemia was more common in the elderly (86%, 24/28) than in the younger participants (60%, 14/30) (p = 0.03) with higher YF-17D RNA copy numbers in the elderly participants. CONCLUSIONS: We found that elderly subjects had a delayed antibody response and higher viraemia levels after yellow fever primovaccination. We postulate that with older age, a weaker immune response to yellow fever vaccine allows the attenuated virus to cause higher viraemia levels which may increase the risk of developing SAEs. This may be one piece in the puzzle of the pathophysiology of YEL-AVD. TRIAL REGISTRATION: Trialregitser.nl NTR1040.


Yellow Fever Vaccine/adverse effects , Yellow Fever/prevention & control , Yellow fever virus/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Antibodies, Neutralizing/chemistry , Cohort Studies , Humans , Neutralization Tests , Risk , Risk Factors , Time Factors , Vaccination , Viremia/blood , Young Adult
18.
Interdiscip Sci ; 3(1): 64-77, 2011 Mar.
Article En | MEDLINE | ID: mdl-21369890

Yellow fever is among one of the most lethal viral diseases for which approved antiviral therapies were yet to be discovered. Herein, functional assignment of complete YFV proteome was done through support vector machine. Major envelope (E) protein that mediates entry of YFV into host cell was selected as a potent molecular target. Three dimensional structure of the molecular target was predicted using Modeller9v7. The model was optimized in Maestro9.0 applying OPLS AA force field and was evaluated using PROCHECK, ProSA, ProQ and Profile 3D. The BOG pocket residues Val48, Glu197, Thr200, Ile204, Thr265, Thr268 and Gly278 were located in YFV E protein using SiteMap2.3. More than one million compounds of Ligandinfo Meta database were explored using a computational virtual screening protocol targeting BOG pocket of the E protein. Finally, ten top ranked lead molecules with strong binding affinity to BOG pocket of YFV E protein were identified based on XP Gscore. Drug likeliness and comparative bioactivity analysis for these leads using QikProp3.2 had shown that these molecules would have the potential to act as better drug. Thus, the 10 lead molecules suggested in the present study would be of interest as promising starting point for designing antiviral compound against yellow fever.


Antiviral Agents/chemistry , Viral Envelope Proteins/chemistry , Yellow fever virus/metabolism , Amino Acid Sequence , Binding Sites , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Proteome , Small Molecule Libraries , Software , Support Vector Machine , Viral Envelope Proteins/metabolism
19.
Arch Virol ; 156(6): 931-8, 2011 Jun.
Article En | MEDLINE | ID: mdl-21298455

The flavivirus NS5 protein is one of the most important proteins of the replication complex, and cellular proteins can interact with it. This study shows for the first time that the yellow fever virus (YFV) NS5 protein is able to interact with U1A, a protein involved in splicing and polyadenylation. We confirmed this interaction by GST-pulldown assay and by co-immunoprecipitation in YFV-infected cells. A region between amino acids 368 and 448 was identified as the site of interaction of the NS5 protein with U1A. This region was conserved among some flaviviruses of medical importance. The implications of this interaction for flavivirus replication are discussed.


Protein Interaction Domains and Motifs , Ribonucleoprotein, U1 Small Nuclear/metabolism , Viral Nonstructural Proteins/metabolism , Yellow fever virus , Amino Acid Sequence , Animals , Binding Sites , Chlorocebus aethiops , Conserved Sequence , HeLa Cells , Humans , Immunoprecipitation , Polymerase Chain Reaction , Protein Binding , RNA, Viral , Ribonucleoprotein, U1 Small Nuclear/chemistry , Two-Hybrid System Techniques , Vero Cells , Viral Nonstructural Proteins/chemistry , Yellow fever virus/genetics , Yellow fever virus/metabolism
20.
Microbes Infect ; 13(1): 85-95, 2011 Jan.
Article En | MEDLINE | ID: mdl-21044891

The ESCRT (endosomal sorting complex required for transport) machinery normally executes cargo sorting and internalization during multivesicular body biogenesis, but is also utilized by several enveloped viruses to facilitate their budding from cellular membranes. Although the mechanisms of flavivirus infectious particle assembly and release are poorly understood, the nonstructural protein NS3 has been reported to have an essential role via an undescribed mechanism. Here, we shed light on the role of NS3 by connecting it to the host factor Alix, a protein intimately connected with the ESCRT machinery. We demonstrate that NS3 and Alix interact and show that dominant negative versions of Alix inhibit YFV release. Furthermore, we show that NS3 supplied in trans rescues this effect. We propose that the interaction between NS3 and Alix contributes to YFV release.


Endosomal Sorting Complexes Required for Transport/metabolism , Viral Nonstructural Proteins/metabolism , Virus Release/physiology , Yellow fever virus/metabolism , Amino Acid Sequence , Animals , Chlorocebus aethiops , Endosomal Sorting Complexes Required for Transport/chemistry , Molecular Sequence Data , Peptide Fragments/metabolism , Rabbits , Sequence Alignment , Sequence Homology, Amino Acid , Vero Cells , Viral Nonstructural Proteins/chemistry
...