Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.012
Filter
1.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273370

ABSTRACT

Zika virus (ZIKV; family, Flaviviridae), which causes congenital Zika syndrome, Guillain-Barré Syndrome, and other severe diseases, is transmitted mainly by mosquitoes; however, the virus can be transmitted through other routes. Among the three structural and seven nonstructural proteins, the surface envelope (E) protein of ZIKV plays a critical role in viral entry and pathogenesis, making it a key target for the development of effective entry inhibitors. This review article describes the life cycle, genome, and encoded proteins of ZIKV, illustrates the structure and function of the ZIKV E protein, summarizes E protein-targeting entry inhibitors (with a focus on those based on natural products and small molecules), and highlights challenges that may potentially hinder the development of effective inhibitors of ZIKV infection. Overall, the article will provide useful guidance for further development of safe and potent ZIKV entry inhibitors targeting the viral E protein.


Subject(s)
Antiviral Agents , Viral Envelope Proteins , Virus Internalization , Zika Virus Infection , Zika Virus , Zika Virus/drug effects , Zika Virus/physiology , Virus Internalization/drug effects , Humans , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/antagonists & inhibitors , Zika Virus Infection/virology , Zika Virus Infection/drug therapy , Antiviral Agents/pharmacology , Animals
2.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273400

ABSTRACT

Zika virus (ZIKV), transmitted by Aedes mosquitoes, has been a global health concern since 2007. It primarily causes fetal microcephaly and neuronal defects through maternal transmission and induces neurological complications in adults. Recent studies report elevated proinflammatory cytokines and persistent neurological alterations post recovery, but the in vivo mechanisms remain unclear. In our study, viral RNA loads in the brains of mice infected with ZIKV peaked at 7 days post infection and returned to baseline by day 21, indicating recovery. RNA sequencing of the cerebral cortex at 7 and 21 days revealed upregulated genes related to neuroinflammation and microglial activation. Histological analyses indicated neuronal cell death and altered neurite morphology owing to severe neuroinflammation. Additionally, sustained microglial activation was associated with increased phospho-Tau levels, constituting a marker of neurodegeneration. These findings highlight how persistent microglial activation leads to neuronal dysfunction post ZIKV recovery, providing insights into the molecular pathogenesis of ZIKV-induced brain abnormalities.


Subject(s)
Microglia , Neurons , Zika Virus Infection , Zika Virus , Animals , Zika Virus Infection/virology , Zika Virus Infection/pathology , Zika Virus Infection/metabolism , Microglia/virology , Microglia/metabolism , Microglia/pathology , Mice , Zika Virus/physiology , Zika Virus/pathogenicity , Neurons/virology , Neurons/metabolism , Neurons/pathology , Synapses/pathology , Synapses/metabolism , Brain/virology , Brain/pathology , Brain/metabolism , Disease Models, Animal , Female , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/virology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Viral Load
3.
Viruses ; 16(9)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39339867

ABSTRACT

The establishment of effective antiviral responses within host cells is intricately related to their metabolic status, shedding light on immunometabolism. In this study, we investigated the hypothesis that cellular reliance on glutamine metabolism contributes to the development of a potent antiviral response. We evaluated the antiviral response in the presence or absence of L-glutamine in the culture medium, revealing a bivalent response hinging on cellular metabolism. While certain interferon-stimulated genes (ISGs) exhibited higher expression in an oxidative phosphorylation (OXPHOS)-dependent manner, others were surprisingly upregulated in a glycolytic-dependent manner. This metabolic dichotomy was influenced in part by variations in interferon-ß (IFN-ß) expression. We initially demonstrated that the presence of L-glutamine induced an enhancement of OXPHOS in A549 cells. Furthermore, in cells either stimulated by poly:IC or infected with dengue virus and Zika virus, a marked increase in ISGs expression was observed in a dose-dependent manner with L-glutamine supplementation. Interestingly, our findings unveiled a metabolic dependency in the expression of specific ISGs. In particular, genes such as ISG54, ISG12 and ISG15 exhibited heightened expression in cells cultured with L-glutamine, corresponding to higher OXPHOS rates and IFN-ß signaling. Conversely, the expression of viperin and 2'-5'-oligoadenylate synthetase 1 was inversely related to L-glutamine concentration, suggesting a glycolysis-dependent regulation, confirmed by inhibition experiments. This study highlights the intricate interplay between cellular metabolism, especially glutaminergic and glycolytic, and the establishment of the canonical antiviral response characterized by the expression of antiviral effectors, potentially paving the way for novel strategies to modulate antiviral responses through metabolic interventions.


Subject(s)
Glutamine , Interferon-beta , Oxidative Phosphorylation , Poly I-C , Zika Virus , Humans , Glutamine/metabolism , A549 Cells , Poly I-C/pharmacology , Interferon-beta/metabolism , Oxidative Phosphorylation/drug effects , Zika Virus/drug effects , Zika Virus/physiology , Antiviral Agents/pharmacology , Glycolysis/drug effects , Dengue Virus/drug effects , Dengue Virus/physiology , Virus Replication/drug effects , Host-Pathogen Interactions , Viperin Protein
4.
Nat Commun ; 15(1): 8221, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300135

ABSTRACT

The main vectors of Zika virus (ZIKV) and dengue virus (DENV) are Aedes aegypti and Ae. albopictus, with Ae. aegypti being more competent. However, the underlying mechanisms remain unclear. Here, we find Ae. albopictus shows comparable vector competence to ZIKV/DENV with Ae. aegypti by blood-feeding after antibiotic treatment or intrathoracic injection. This suggests that midgut microbiota can influence vector competence. Enterobacter hormaechei_B17 (Eh_B17) is isolated from field-collected Ae. albopictus and conferred resistance to ZIKV/DENV infection in Ae. aegypti after gut-transplantation. Sphingosine, a metabolite secreted by Eh_B17, effectively suppresses ZIKV infection in both Ae. aegypti and cell cultures by blocking viral entry during the fusion step, with an IC50 of approximately 10 µM. A field survey reveals that Eh_B17 preferentially colonizes Ae. albopictus compared to Ae. aegypti. And field Ae. albopictus positive for Eh_B17 are more resistant to ZIKV infection. These findings underscore the potential of gut symbiotic bacteria, such as Eh_B17, to modulate the arbovirus vector competence of Aedes mosquitoes. As a natural antiviral agent, Eh_B17 holds promise as a potential candidate for blocking ZIKV/DENV transmission.


Subject(s)
Aedes , Dengue Virus , Enterobacter , Gastrointestinal Microbiome , Mosquito Vectors , Sphingosine , Symbiosis , Zika Virus , Aedes/virology , Aedes/microbiology , Aedes/drug effects , Animals , Mosquito Vectors/microbiology , Mosquito Vectors/virology , Mosquito Vectors/drug effects , Zika Virus/physiology , Zika Virus/drug effects , Dengue Virus/drug effects , Dengue Virus/physiology , Gastrointestinal Microbiome/drug effects , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/pharmacology , Enterobacter/drug effects , Enterobacter/physiology , Zika Virus Infection/transmission , Zika Virus Infection/virology , Dengue/transmission , Dengue/virology , Dengue/prevention & control , Female , Virus Internalization/drug effects , Humans
5.
Commun Biol ; 7(1): 1089, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237833

ABSTRACT

Viruses depend on host metabolic pathways and flaviviruses are specifically linked to lipid metabolism. During dengue virus infection lipid droplets are degraded to fuel replication and Zika virus (ZIKV) infection depends on triglyceride biosynthesis. Here, we systematically investigated the neutral lipid-synthesizing enzymes diacylglycerol O-acyltransferases (DGAT) and the sterol O-acyltransferase (SOAT) 1 in orthoflavivirus infection. Downregulation of DGAT1 and SOAT1 compromises ZIKV infection in hepatoma cells but only SOAT1 and not DGAT inhibitor treatment reduces ZIKV infection. DGAT1 interacts with the ZIKV capsid protein, indicating that protein interaction might be required for ZIKV replication. Importantly, inhibition of SOAT1 severely impairs ZIKV infection in neural cell culture models and cerebral organoids. SOAT1 inhibitor treatment decreases extracellular viral RNA and E protein level and lowers the specific infectivity of virions, indicating that ZIKV morphogenesis is compromised, likely due to accumulation of free cholesterol. Our findings provide insights into the importance of cholesterol and cholesterol ester balance for efficient ZIKV replication and implicate SOAT1 as an antiviral target.


Subject(s)
Organoids , Sterol O-Acyltransferase , Virus Replication , Zika Virus Infection , Zika Virus , Humans , Zika Virus Infection/virology , Zika Virus Infection/metabolism , Zika Virus/physiology , Organoids/virology , Organoids/metabolism , Virus Replication/drug effects , Sterol O-Acyltransferase/metabolism , Sterol O-Acyltransferase/antagonists & inhibitors , Animals , Antiviral Agents/pharmacology
6.
Cell Rep ; 43(9): 114694, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39196777

ABSTRACT

Subgenomic flavivirus RNAs (sfRNAs) are structured RNAs encoded by flaviviruses that promote viral infection by inhibiting cellular RNA decay machinery. Herein, we analyze sfRNA production and localization using single-molecule RNA fluorescence in situ hybridization (smRNA-FISH) throughout West Nile virus, Zika virus, or dengue virus serotype 2 infection. We observe that sfRNAs are generated during the RNA replication phase of viral infection in the cytosol and accumulate in processing bodies (P-bodies), which contain RNA decay machinery such as XRN1 and Dcp1b. However, upon activation of the host antiviral endoribonuclease, ribonuclease L (RNase L), sfRNAs re-localize to ribonucleoprotein complexes known as RNase L-induced bodies (RLBs). RLB-mediated sequestration of sfRNAs reduces sfRNA association with RNA decay machinery in P-bodies, which coincides with increased viral RNA decay. These findings establish a functional role for RLBs in enhancing the cell-mediated decay of viral RNA by sequestering functional viral RNA decay products.


Subject(s)
Endoribonucleases , Flavivirus , RNA Stability , RNA, Viral , RNA, Viral/metabolism , RNA, Viral/genetics , Endoribonucleases/metabolism , Humans , Flavivirus/metabolism , Zika Virus/metabolism , Zika Virus/physiology , Zika Virus/genetics , Animals , West Nile virus/physiology , Virus Replication
7.
Virus Res ; 348: 199447, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39117146

ABSTRACT

One third of all emerging infectious diseases are vector-borne, with no licensed antiviral therapies available against any vector-borne viruses. Zika virus and Usutu virus are two emerging flaviviruses transmitted primarily by mosquitoes. These viruses modulate different host pathways, including the PI3K/AKT/mTOR pathway. Here, we report the effect on ZIKV and USUV replication of two AKT inhibitors, Miransertib (ARQ-092, allosteric inhibitor) and Capivasertib (AZD5363, competitive inhibitor) in different mammalian and mosquito cell lines. Miransertib showed a stronger inhibitory effect against ZIKV and USUV than Capivasertib in mammalian cells, while Capivasertib showed a stronger effect in mosquito cells. These findings indicate that AKT plays a conserved role in flavivirus infection, in both the vertebrate host and invertebrate vector. Nevertheless, the specific function of AKT may vary depending on the host species. These findings indicate that AKT may be playing a conserved role in flavivirus infection in both, the vertebrate host and the invertebrate vector. However, the specific function of AKT may vary depending on the host species. A better understanding of virus-host interactions is therefore required to develop new treatments to prevent human disease and new approaches to control transmission by insect vectors.


Subject(s)
Flavivirus Infections , Flavivirus , Proto-Oncogene Proteins c-akt , Virus Replication , Zika Virus , Animals , Flavivirus/physiology , Flavivirus/drug effects , Flavivirus/genetics , Humans , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Cell Line , Zika Virus/physiology , Zika Virus/drug effects , Flavivirus Infections/virology , Flavivirus Infections/transmission , Vertebrates/virology , Antiviral Agents/pharmacology , Mosquito Vectors/virology , Chlorocebus aethiops , Culicidae/virology , Host-Pathogen Interactions
8.
PLoS Negl Trop Dis ; 18(8): e0012146, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39178324

ABSTRACT

Mosquito-borne Zika virus (ZIKV) from sub-Saharan Africa has recently gained attention due to its epidemic potential and its capacity to be highly teratogenic. To improve our knowledge on currently circulating strains of African ZIKV, we conducted protein sequence alignment and identified contemporary West Africa NS1 (NS1CWA) protein as a highly conserved viral protein. Comparison of NS1CWA with the NS1 of the historical African ZIKV strain MR766 (NS1MR766), revealed seven amino acid substitutions. The effects of NS1 mutations on protein expression, virus replication, and innate immune activation were assessed in human cells using recombinant NS1 proteins and a chimeric viral clone MR766 with NS1CWA replacing NS1MR766. Our data indicated higher secretion efficiency of NS1CWA compared to NS1MR766 associated with a change in subcellular distribution. A chimeric MR766 virus with NS1CWA instead of authentic protein displayed a greater viral replication efficiency, leading to more pronounced cell death compared to parental virus. Enhanced viral growth was associated with reduced activation of innate immunity. Our data raise questions of the importance of NS1 protein in the pathogenicity of contemporary ZIKV from sub-Saharan Africa and point to differences within viral strains of African lineage.


Subject(s)
Immunity, Innate , Viral Nonstructural Proteins , Virus Replication , Zika Virus , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/immunology , Zika Virus/genetics , Zika Virus/immunology , Zika Virus/physiology , Humans , Zika Virus Infection/virology , Zika Virus Infection/immunology , Animals , Chlorocebus aethiops , Vero Cells , Africa, Western , Amino Acid Substitution , Cell Line
9.
Sci Rep ; 14(1): 18470, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39122799

ABSTRACT

The microbial communities residing in the mosquito midgut play a key role in determining the outcome of mosquito pathogen infection. Elizabethkingia anophelis, originally isolated from the midgut of Anopheles gambiae possess a broad-spectrum antiviral phenotype, yet a gap in knowledge regarding the mechanistic basis of its interaction with viruses exists. The current study aims to identify pathways and genetic factors linked to E. anophelis antiviral activity. The understanding of E. anophelis antiviral mechanism could lead to novel transmission barrier tools to prevent arboviral outbreaks. We utilized a non-targeted multi-omics approach, analyzing extracellular lipids, proteins, metabolites of culture supernatants coinfected with ZIKV and E. anophelis. We observed a significant decrease in arginine and phenylalanine levels, metabolites that are essential for viral replication and progression of viral infection. This study provides insights into the molecular basis of E. anophelis antiviral phenotype. The findings lay a foundation for in-depth mechanistic studies.


Subject(s)
Flavobacteriaceae , Zika Virus , Zika Virus/physiology , Animals , Flavobacteriaceae/metabolism , Flavobacteriaceae/genetics , Anopheles/virology , Anopheles/microbiology , Zika Virus Infection/virology , Antiviral Agents/pharmacology , Antiviral Agents/metabolism , Virus Replication , Phenylalanine/metabolism , Arginine/metabolism , Multiomics
10.
Proc Natl Acad Sci U S A ; 121(34): e2403235121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39145933

ABSTRACT

The ZIKA virus (ZIKV) evades the host immune response by degrading STAT2 through its NS5 protein, thereby inhibiting type I interferon (IFN)-mediated antiviral immunity. However, the molecular mechanism underlying this process has remained elusive. In this study, we performed a genome-wide CRISPR/Cas9 screen, revealing that ZSWIM8 as the substrate receptor of Cullin3-RING E3 ligase is required for NS5-mediated STAT2 degradation. Genetic depletion of ZSWIM8 and CUL3 substantially impeded NS5-mediated STAT2 degradation. Biochemical analysis illuminated that NS5 enhances the interaction between STAT2 and the ZSWIM8-CUL3 E3 ligase complex, thereby facilitating STAT2 ubiquitination. Moreover, ZSWIM8 knockout endowed A549 and Huh7 cells with partial resistance to ZIKV infection and protected cells from the cytopathic effects induced by ZIKV, which was attributed to the restoration of STAT2 levels and the activation of IFN signaling. Subsequent studies in a physiologically relevant model, utilizing human neural progenitor cells, demonstrated that ZSWIM8 depletion reduced ZIKV infection, resulting from enhanced IFN signaling attributed to the sustained levels of STAT2. Our findings shed light on the role of ZIKV NS5, serving as the scaffold protein, reprograms the ZSWIM8-CUL3 E3 ligase complex to orchestrate STAT2 proteasome-dependent degradation, thereby facilitating evasion of IFN antiviral signaling. Our study provides unique insights into ZIKV-host interactions and holds promise for the development of antivirals and prophylactic vaccines.


Subject(s)
Cullin Proteins , Interferon Type I , Proteolysis , STAT2 Transcription Factor , Signal Transduction , Ubiquitin-Protein Ligases , Ubiquitination , Viral Nonstructural Proteins , Zika Virus Infection , Zika Virus , Humans , STAT2 Transcription Factor/metabolism , Zika Virus/immunology , Zika Virus/physiology , Zika Virus/metabolism , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Interferon Type I/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Zika Virus Infection/metabolism , Zika Virus Infection/immunology , Zika Virus Infection/virology , Cullin Proteins/metabolism , A549 Cells , HEK293 Cells , CRISPR-Cas Systems
11.
mSphere ; 9(8): e0040124, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39092912

ABSTRACT

As climate change alters Earth's biomes, it is expected the transmission dynamics of mosquito-borne viruses will change. While the effects of temperature changes on mosquito-virus interactions and the spread of the pathogens have been elucidated over the last decade, the impact of relative humidity changes is still relatively unknown. To overcome this knowledge gap, we exposed Aedes aegypti females to various humidity conditions. We measured different components of vectorial capacity such as survival, blood-feeding rates, and changes in infection and dissemination of Zika virus. Survival decreased as the humidity level decreased, while infection rates increased as the humidity level decreased. Alternatively, blood feeding rates and disseminated infection rates peaked at the intermediate 50% relative humidity treatment but were the same in the 30% and 80% relative humidity treatments. These results provide empirical evidence that Ae. aegypti exposure to low humidity can enhance Zika virus infection in the mosquito, which has important implications in predicting how climate change will impact mosquito-borne viruses.IMPORTANCEViruses transmitted by mosquitoes to humans are a major public health burden and are expected to increase under climate change. While we know that temperature is an important driver of variation in arbovirus replication in the mosquito, very little is known about how other relevant climate variables such as humidity will influence the interaction between mosquitoes and the viruses they transmit. Given the variability in humidity across environments, and the predicted changes in humidity under climate change, it is imperative that we also study the impact that it has on mosquito infection and transmission of arboviruses.


Subject(s)
Aedes , Climate Change , Humidity , Mosquito Vectors , Zika Virus Infection , Zika Virus , Aedes/virology , Aedes/physiology , Animals , Zika Virus Infection/transmission , Zika Virus Infection/virology , Mosquito Vectors/virology , Zika Virus/physiology , Female , Temperature , Feeding Behavior
12.
Viruses ; 16(8)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39205304

ABSTRACT

Zika virus (ZIKV) impacts the developing brain. Here, a technique was applied to define, in 3D, developmental changes in the brains of ZIKV-infected mice. Postnatal day 1 mice were uninfected or ZIKV-infected, then analysed by iodine staining and micro-CT scanning (diffusible iodine contrast-enhanced micro-CT; diceCT) at 3-, 6-, and 10-days post-infection (dpi). Multiple brain regions were visualised using diceCT: the olfactory bulb, cerebrum, hippocampus, midbrain, interbrain, and cerebellum, along with the lens and retina of the eye. Brain regions were computationally segmented and quantitated, with increased brain volumes and developmental time in uninfected mice. Conversely, in ZIKV-infected mice, no quantitative differences were seen at 3 or 6 dpi when there were no clinical signs, but qualitatively, diverse visual defects were identified at 6-10 dpi. By 10 dpi, ZIKV-infected mice had significantly lower body weight and reduced volume of brain regions compared to 10 dpi-uninfected or 6 dpi ZIKV-infected mice. Nissl and immunofluorescent Iba1 staining on post-diceCT tissue were successful, but RNA extraction was not. Thus, diceCT shows utility for detecting both 3D qualitative and quantitative changes in the developing brain of ZIKV-infected mice, with the benefit, post-diceCT, of retaining the ability to apply traditional histology and immunofluorescent analysis to tissue.


Subject(s)
Brain , Disease Models, Animal , Zika Virus Infection , Zika Virus , Animals , Zika Virus Infection/virology , Zika Virus Infection/pathology , Brain/virology , Brain/pathology , Brain/diagnostic imaging , Mice , Zika Virus/physiology , X-Ray Microtomography , Female
13.
Sci Rep ; 14(1): 20095, 2024 08 29.
Article in English | MEDLINE | ID: mdl-39209987

ABSTRACT

Usutu (USUV), West Nile (WNV), and Zika virus (ZIKV) are neurotropic arthropod-borne viruses (arboviruses) that cause severe neurological disease in humans. However, USUV-associated neurological disease is rare, suggesting a block in entry to or infection of the brain. We determined the replication, cell tropism and neurovirulence of these arboviruses in human brain tissue using a well-characterized human fetal organotypic brain slice culture model. Furthermore, we assessed the efficacy of interferon-ß and 2'C-methyl-cytidine, a synthetic nucleoside analogue, in restricting viral replication. All three arboviruses replicated within the brain slices, with WNV reaching the highest titers, and all primarily infected neuronal cells. USUV- and WNV-infected cells exhibited a shrunken morphology, not associated with detectable cell death. Pre-treatment with interferon-ß inhibited replication of all arboviruses, while 2'C-methyl-cytidine reduced only USUV and ZIKV titers. Collectively, USUV can infect human brain tissue, showing similarities in tropism and neurovirulence as WNV and ZIKV. These data suggest that a blockade to infection of the human brain may not be the explanation for the low clinical incidence of USUV-associated neurological disease. However, USUV replicated more slowly and to lower titers than WNV, which could help to explain the reduced severity of neurological disease resulting from USUV infection.


Subject(s)
Brain , Flavivirus , Virus Replication , West Nile virus , Zika Virus , Humans , West Nile virus/pathogenicity , West Nile virus/physiology , Zika Virus/pathogenicity , Zika Virus/physiology , Brain/virology , Virus Replication/drug effects , Flavivirus/pathogenicity , Flavivirus/physiology , Flavivirus/drug effects , Fetus/virology , Interferon-beta/pharmacology , Animals , Virulence , Organ Culture Techniques , Viral Tropism , Neurons/virology , Flavivirus Infections/virology , Zika Virus Infection/virology , Chlorocebus aethiops , Vero Cells
14.
RNA Biol ; 21(1): 1-10, 2024 Jan.
Article in English | MEDLINE | ID: mdl-39183472

ABSTRACT

One of the most recent advances in the analysis of viral RNA-cellular protein interactions is the Comprehensive Identification of RNA-binding Proteins by Mass Spectrometry (ChIRP-MS). Here, we used ChIRP-MS in mock-infected and Zika-infected wild-type cells and cells knockout for the zinc finger CCCH-type antiviral protein 1 (ZAP). We characterized 'ZAP-independent' and 'ZAP-dependent' cellular protein interactomes associated with flavivirus RNA and found that ZAP affects cellular proteins associated with Zika virus RNA. The ZAP-dependent interactome identified with ChIRP-MS provides potential ZAP co-factors for antiviral activity against Zika virus and possibly other viruses. Identifying the full spectrum of ZAP co-factors and mechanisms of how they act will be critical to understanding the ZAP antiviral system and may contribute to the development of antivirals.


Subject(s)
RNA, Viral , RNA-Binding Proteins , Zika Virus Infection , Zika Virus , Zika Virus/genetics , Zika Virus/physiology , Zika Virus/metabolism , Humans , RNA, Viral/metabolism , RNA, Viral/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Zika Virus Infection/virology , Zika Virus Infection/metabolism , Protein Binding , Host-Pathogen Interactions/genetics , Mass Spectrometry , HEK293 Cells
15.
Sci Immunol ; 9(98): eadk9872, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39121194

ABSTRACT

The Aedes aegypti mosquito is a vector of many infectious agents, including flaviviruses such as Zika virus. Components of mosquito saliva have pleomorphic effects on the vertebrate host to enhance blood feeding, and these changes also create a favorable niche for pathogen replication and dissemination. Here, we demonstrate that human CD47, which is known to be involved in various immune processes, interacts with a 34-kilodalton mosquito salivary protein named Nest1. Nest1 is up-regulated in blood-fed female A. aegypti and facilitates Zika virus dissemination in human skin explants. Nest1 has a stronger affinity for CD47 than its natural ligand, signal regulatory protein α, competing for binding at the same interface. The interaction between Nest1 with CD47 suppresses phagocytosis by human macrophages and inhibits proinflammatory responses by white blood cells, thereby suppressing antiviral responses in the skin. This interaction elucidates how an arthropod protein alters the human response to promote arbovirus infectivity.


Subject(s)
Aedes , Skin , Zika Virus , Aedes/immunology , Aedes/virology , Animals , Humans , Skin/immunology , Skin/virology , Zika Virus/immunology , Zika Virus/physiology , Female , Insect Proteins/immunology , Zika Virus Infection/immunology , Salivary Proteins and Peptides/immunology , Mosquito Vectors/immunology , Mosquito Vectors/virology , CD47 Antigen
16.
Sci Rep ; 14(1): 18112, 2024 08 05.
Article in English | MEDLINE | ID: mdl-39103482

ABSTRACT

This study presents a computational investigation of a stochastic Zika virus along with optimal control model using the Legendre spectral collocation method (LSCM). By accumulation of stochasticity into the model through the proposed stochastic differential equations, we appropriating the random fluctuations essential in the progression and disease transmission. The stability, convergence and accuracy properties of the LSCM are conscientiously analyzed and also demonstrating its strength for solving the complex epidemiological models. Moreover, the study evaluates the various control strategies, such as treatment, prevention and treatment pesticide control, and identifies optimal combinations that the intervention costs and also minimize the proposed infection rates. The basic properties of the given model, such as the reproduction number, were determined with and without the presence of the control strategies. For R 0 < 0 , the model satisfies the disease-free equilibrium, in this case the disease die out after some time, while for R 0 > 1 , then endemic equilibrium is satisfied, in this case the disease spread in the population at higher scale. The fundamental findings acknowledge the significant impact of stochastic phonemes on the robustness and effectiveness of control strategies that accelerating the need for cost-effective and multi-faceted approaches. In last the results provide the valuable insights for public health department to enabling more impressive mitigation of Zika virus outbreaks and management in real-world scenarios.


Subject(s)
Stochastic Processes , Zika Virus Infection , Zika Virus , Zika Virus Infection/epidemiology , Zika Virus Infection/prevention & control , Zika Virus Infection/transmission , Humans , Zika Virus/physiology , Computer Simulation , Epidemiological Models
17.
EMBO Mol Med ; 16(8): 1817-1839, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39009885

ABSTRACT

Zika virus (ZIKV) infection may lead to severe neurological consequences, including seizures, and early infancy death. However, the involved mechanisms are still largely unknown. TRPC channels play an important role in regulating nervous system excitability and are implicated in seizure development. We investigated whether TRPCs might be involved in the pathogenesis of ZIKV infection. We found that ZIKV infection increases TRPC4 expression in host cells via the interaction between the ZIKV-NS3 protein and CaMKII, enhancing TRPC4-mediated calcium influx. Pharmacological inhibition of CaMKII decreased both pCREB and TRPC4 protein levels, whereas the suppression of either TRPC4 or CaMKII improved the survival rate of ZIKV-infected cells and reduced viral protein production, likely by impeding the replication phase of the viral life cycle. TRPC4 or CaMKII inhibitors also reduced seizures and increased the survival of ZIKV-infected neonatal mice and blocked the spread of ZIKV in brain organoids derived from human-induced pluripotent stem cells. These findings suggest that targeting CaMKII or TRPC4 may offer a promising approach for developing novel anti-ZIKV therapies, capable of preventing ZIKV-associated seizures and death.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , TRPC Cation Channels , Zika Virus Infection , Zika Virus , Zika Virus Infection/virology , Zika Virus Infection/metabolism , Animals , Humans , Zika Virus/physiology , Zika Virus/drug effects , Mice , TRPC Cation Channels/metabolism , TRPC Cation Channels/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Virus Replication/drug effects , HEK293 Cells , Viral Proteins/metabolism , Seizures/virology , Seizures/metabolism , Seizures/drug therapy , Viral Proteases , Serine Endopeptidases , Nucleoside-Triphosphatase , DEAD-box RNA Helicases
18.
J Exp Med ; 221(9)2024 09 02.
Article in English | MEDLINE | ID: mdl-39042188

ABSTRACT

The contribution of placental immune responses to congenital Zika virus (ZIKV) syndrome remains poorly understood. Here, we leveraged a mouse model of ZIKV infection to identify mechanisms of innate immune restriction exclusively in the fetal compartment of the placenta. ZIKV principally infected mononuclear trophoblasts in the junctional zone, which was limited by mitochondrial antiviral-signaling protein (MAVS) and type I interferon (IFN) signaling mechanisms. Single nuclear RNA sequencing revealed MAVS-dependent expression of IFN-stimulated genes (ISGs) in spongiotrophoblasts but not in other placental cells that use alternate pathways to induce ISGs. ZIKV infection of Ifnar1-/- or Mavs-/- placentas was associated with greater infection of the adjacent immunocompetent decidua, and heterozygous Mavs+/- or Ifnar1+/- dams carrying immunodeficient fetuses sustained greater maternal viremia and tissue infection than dams carrying wild-type fetuses. Thus, MAVS-IFN signaling in the fetus restricts ZIKV infection in junctional zone trophoblasts, which modulates dissemination and outcome for both the fetus and the pregnant mother.


Subject(s)
Adaptor Proteins, Signal Transducing , Decidua , Fetus , Interferon Type I , Placenta , Receptor, Interferon alpha-beta , Signal Transduction , Trophoblasts , Zika Virus Infection , Zika Virus , Female , Animals , Pregnancy , Interferon Type I/metabolism , Interferon Type I/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Placenta/immunology , Placenta/virology , Placenta/metabolism , Zika Virus Infection/immunology , Zika Virus Infection/virology , Zika Virus/immunology , Zika Virus/physiology , Mice , Decidua/immunology , Decidua/virology , Decidua/metabolism , Fetus/immunology , Fetus/virology , Trophoblasts/immunology , Trophoblasts/virology , Trophoblasts/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , Immunity, Innate , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Disease Models, Animal
19.
PLoS Negl Trop Dis ; 18(7): e0012066, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38968296

ABSTRACT

Zika virus (ZIKV) has become a global health problem over the past decade due to the extension of the geographic distribution of the Asian/American genotype. Recent epidemics of Asian/American ZIKV have been associated with developmental disorders in humans. There is mounting evidence that African ZIKV may be associated with increased fetal pathogenicity necessitating to pay a greater attention towards currently circulating viral strains in sub-Saharan Africa. Here, we generated an infectious molecular clone GUINEA-18 of a recently transmitted human ZIKV isolate from West Africa, ZIKV-15555. The available infectious molecular clone MR766MC of historical African ZIKV strain MR766-NIID was used for a molecular clone-based comparative study. Viral clones GUINEA-18 and MR766MC were compared for their ability to replicate in VeroE6, A549 and HCM3 cell lines. There was a lower replication rate for GUINEA-18 associated with weaker cytotoxicity and reduced innate immune system activation compared with MR766MC. Analysis of chimeric viruses between viral clones stressed the importance of NS1 to NS4B proteins, with a particular focus of NS4B on GUINEA-18 replicative properties. ZIKV has developed strategies to prevent cytoplasmic stress granule formation which occurs in response to virus infection. GUINEA-18 was greatly efficient in inhibiting stress granule assembly in A549 cells subjected to a physiological stressor, with NS1 to NS4B proteins also being critical in this process. The impact of these GUINEA-18 proteins on viral replicative abilities and host-cell responses to viral infection raises the question of the role of nonstructural proteins in the pathogenicity of currently circulating ZIKV in sub-Saharan Africa.


Subject(s)
Virus Replication , Zika Virus Infection , Zika Virus , Zika Virus/genetics , Zika Virus/physiology , Humans , Africa, Western/epidemiology , Zika Virus Infection/virology , Animals , Chlorocebus aethiops , Cell Line , Vero Cells , A549 Cells
20.
FASEB J ; 38(14): e23764, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39042395

ABSTRACT

The mosquito, Aedes aegypti, is the principal vector for several arboviruses. The mosquito midgut is the initial tissue that gets infected with an arbovirus acquired along with a blood meal from a vertebrate host. Blood meal ingestion leads to midgut tissue distention thereby increasing the pore size of the surrounding basal lamina. This allows newly synthesized virions to exit the midgut by traversing the distended basal lamina to infect secondary tissues of the mosquito. We conducted a quantitative label-free proteomic time course analysis with saline meal-fed Ae. aegypti females to identify host factors involved in midgut tissue distention. Around 2000 proteins were detected during each of the seven sampling time points and 164 of those were uniquely expressed. Forty-five of 97 differentially expressed proteins were upregulated during the 96-h time course and most of those were involved in cytoskeleton modulation, metabolic activity, and vesicle/vacuole formation. The F-actin-modulating Ae. aegypti (Aa)-gelsolin was selected for further functional studies. Stable knockout of Aa-gelsolin resulted in a mosquito line, which showed distorted actin filaments in midgut-associated tissues likely due to diminished F-actin processing by gelsolin. Zika virus dissemination from the midgut of these mosquitoes was diminished and delayed. The loss of Aa-gelsolin function was associated with an increased induction of apoptosis in midgut tissue indicating an involvement of Aa-gelsolin in apoptotic signaling in mosquitoes. Here, we used proteomics to discover a novel host factor, Aa-gelsolin, which affects the midgut escape barrier for arboviruses in mosquitoes and apoptotic signaling in the midgut.


Subject(s)
Aedes , Arboviruses , Gelsolin , Insect Proteins , Animals , Aedes/virology , Aedes/metabolism , Gelsolin/metabolism , Gelsolin/genetics , Insect Proteins/metabolism , Insect Proteins/genetics , Arboviruses/physiology , Cytoskeleton/metabolism , Female , Mosquito Vectors/virology , Mosquito Vectors/metabolism , Proteomics/methods , Zika Virus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL