Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 65
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Pathogens ; 13(7)2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-39057782

RÉSUMÉ

Zika virus (ZIKV), a mosquito-borne flavivirus, is prominently associated with microcephaly in babies born to infected mothers as well as Guillain-Barré Syndrome in adults. Each cell type infected by ZIKV-neuronal cells (radial glial cells, neuronal progenitor cells, astrocytes, microglia cells, and glioblastoma stem cells) and non-neuronal cells (primary fibroblasts, epidermal keratinocytes, dendritic cells, monocytes, macrophages, and Sertoli cells)-displays its own characteristic changes to their cell physiology and has various impacts on disease. Here, we provide an in-depth review of the ZIKV life cycle and its cellular targets, and discuss the current knowledge of how infections cause neuropathologies, as well as what approaches researchers are currently taking to further advance such knowledge. A key aspect of ZIKV neuropathogenesis is virus-induced neuronal apoptosis via numerous mechanisms including cell cycle dysregulation, mitochondrial fragmentation, ER stress, and the unfolded protein response. These, in turn, result in the activation of p53-mediated intrinsic cell death pathways. A full spectrum of infection models including stem cells and co-cultures, transwells to simulate blood-tissue barriers, brain-region-specific organoids, and animal models have been developed for ZIKV research.

2.
Article de Anglais | MEDLINE | ID: mdl-36561642

RÉSUMÉ

Background: Recently, the in vitro blood brain barrier (BBB) models derived from human pluripotent stem cells have been given extensive attention in therapeutics due to the implications it has with the health of the central nervous system. It is essential to create an accurate BBB model in vitro in order to better understand the properties of the BBB and how it can respond to inflammatory stimulation and be passed by targeted or non-targeted cell therapeutics, more specifically extracellular vesicles. Methods: Brain-specific pericytes (iPCs) were differentiated from iPSK3 cells using dual SMAD signaling inhibitors and Wnt activation plus fibroblast growth factor 2 (FGF-2). The derived cells were characterized by immunostaining, flow cytometry and RT-PCR. In parallel, blood vessels organoids were derived using Wnt activation, BMP4, FGF2, VEGF and SB431542. The organoids were replated and treated with retinoic acid to enhance the blood brain barrier (BBB) features in the differentiated brain endothelial cells (iECs). Co-culture was performed for the iPCs and iECs in transwell system and 3-D microfluidics channels. Results: The derived iPCs expressed common markers PDGFRb and NG2, as well as brain-specific genes FOXF2, ABCC9, KCNJ8, and ZIC1. The derived iECs expressed common endothelial cell markers CD31, VE-cadherin, as well as BBB-associated genes BRCP, GLUT-1, PGP, ABCC1, OCLN, SLC2A1. The co-culture of the two cell types responded to the stimulation of amyloid ß42 oligomers by the upregulation of expression of TNFa, IL6, NFKB, Casp3, SOD2 and TP53. The co-culture also showed the property of trans-endothelial electrical resistance. The proof-of-concept vascularization strategy was demonstrated in a 3-D microfluidics-based device. Conclusion: The derived iPCs and iECs have brain-specific properties and the co-culture of iPCs and iECs provides an in vitro BBB model that show inflammatory response. This study has significance in establishing micro-physiological systems for neurological disease modeling and drug screening.

3.
Cell Rep ; 39(9): 110885, 2022 05 31.
Article de Anglais | MEDLINE | ID: mdl-35649379

RÉSUMÉ

Physiological blood-tissue barriers play a critical role in separating the circulation from immune-privileged sites and denying access to blood-borne viruses. The mechanism of virus restriction by these barriers is poorly understood. We utilize induced pluripotent stem cell (iPSC)-derived human brain microvascular endothelial cells (iBMECs) to study virus-blood-brain barrier (BBB) interactions. These iPSC-derived cells faithfully recapitulate a striking difference in in vivo neuroinvasion by two alphavirus isolates and are selectively permissive to neurotropic flaviviruses. A model of cocultured iBMECs and astrocytes exhibits high transendothelial electrical resistance and blocks non-neurotropic flaviviruses from getting across the barrier. We find that iBMECs constitutively express an interferon-induced gene, IFITM1, which preferentially restricts the replication of non-neurotropic flaviviruses. Barrier cells from blood-testis and blood-retinal barriers also constitutively express IFITMs that contribute to the viral resistance. Our application of a renewable human iPSC-based model for studying virus-BBB interactions reveals that intrinsic immunity at the barriers contributes to virus exclusion.


Sujet(s)
Barrière hémato-encéphalique , Cellules souches pluripotentes induites , Antiviraux , Encéphale/physiologie , Cellules endothéliales/physiologie , Humains , Cellules souches pluripotentes induites/physiologie , Mâle
4.
Methods Mol Biol ; 2429: 201-213, 2022.
Article de Anglais | MEDLINE | ID: mdl-35507163

RÉSUMÉ

The blood-brain barrier is a tissue structure that modulates the selective entry of molecules into the brain compartment. This barrier offers protection to the brain microenvironment from toxins or any fluctuations in the composition of the blood plasma via a layer of endothelial cells connected by tight junctions and supported by pericytes and astrocytes. Disruption of the barrier can be either a cause or a consequence of central nervous system pathogenesis. Therefore, research based on understanding the structure, function, and the mechanisms of breaching the blood-brain barrier is of primary interest for diverse disciplines including drug discovery, brain pathology, and infectious disease. The following protocol describes a detailed differentiation method that uses defined serum components during stem cell culture to deliver cellular cues in order to drive the cells towards brain endothelial cell lineage. This method can be used to obtain reproducible and scalable cultures of brain microvascular endothelial cells with barrier characteristics and functionality. These endothelial cells can also be stored long term or shipped frozen.


Sujet(s)
Barrière hémato-encéphalique , Cellules endothéliales , Astrocytes , Encéphale , Différenciation cellulaire/physiologie , Péricytes , Jonctions serrées
6.
Cells ; 10(12)2021 11 27.
Article de Anglais | MEDLINE | ID: mdl-34943843

RÉSUMÉ

Zika virus (ZIKV) became a global health concern in 2016 due to its links to congenital microcephaly and other birth defects. Flaviviruses, including ZIKV, reorganize the endoplasmic reticulum (ER) to form a viroplasm, a compartment where virus particles are assembled. Microtubules (MTs) and microtubule-organizing centers (MTOCs) coordinate structural and trafficking functions in the cell, and MTs also support replication of flaviviruses. Here we investigated the roles of MTs and the cell's MTOCs on ZIKV viroplasm organization and virus production. We show that a toroidal-shaped viroplasm forms upon ZIKV infection, and MTs are organized at the viroplasm core and surrounding the viroplasm. We show that MTs are necessary for viroplasm organization and impact infectious virus production. In addition, the centrosome and the Golgi MTOC are closely associated with the viroplasm, and the centrosome coordinates the organization of the ZIKV viroplasm toroidal structure. Surprisingly, viroplasm formation and virus production are not significantly impaired when infected cells have no centrosomes and impaired Golgi MTOC, and we show that MTs are anchored to the viroplasm surface in these cells. We propose that the viroplasm is a site of MT organization, and the MTs organized at the viroplasm are sufficient for efficient virus production.


Sujet(s)
Centre organisateur de microtubules/métabolisme , Microtubules/métabolisme , Compartiments de réplication virale/physiologie , Infection par le virus Zika/virologie , Lignée cellulaire , Centrosome/métabolisme , Réticulum endoplasmique/métabolisme , Appareil de Golgi/métabolisme , Humains , Virion/métabolisme
7.
Viruses ; 13(11)2021 10 20.
Article de Anglais | MEDLINE | ID: mdl-34834920

RÉSUMÉ

Zika virus (ZIKV) is a re-emerging flavivirus that has caused large-scale epidemics. Infection during pregnancy can lead to neurologic developmental abnormalities in children. There is no approved vaccine or therapy for ZIKV. To uncover cellular pathways required for ZIKV that can be therapeutically targeted, we transcriptionally upregulated all known human coding genes with an engineered CRISPR-Cas9 activation complex in human fibroblasts deficient in interferon (IFN) signaling. We identified Ras homolog family member V (RhoV) and WW domain-containing transcription regulator 1 (WWTR1) as proviral factors, and found them to play important roles during early ZIKV infection in A549 cells. We then focused on RhoV, a Rho GTPase with atypical terminal sequences and membrane association, and validated its proviral effects on ZIKV infection and virion production in SNB-19 cells. We found that RhoV promotes infection of some flaviviruses and acts at the step of viral entry. Furthermore, RhoV proviral effects depend on the complete GTPase cycle. By depleting Rho GTPases and related proteins, we identified RhoB and Pak1 as additional proviral factors. Taken together, these results highlight the positive role of RhoV in ZIKV infection and confirm CRISPR activation as a relevant method to identify novel host-pathogen interactions.


Sujet(s)
Protéines G/métabolisme , Protéines tumorales/métabolisme , Infection par le virus Zika/enzymologie , Virus Zika/physiologie , Protéine G RhoB/métabolisme , Cellules A549 , Systèmes CRISPR-Cas , Protéines G/génétique , Humains , Protéines tumorales/génétique , Transcriptional coactivator with PDZ-binding motif proteins/génétique , Transcriptional coactivator with PDZ-binding motif proteins/métabolisme , Pénétration virale , Réplication virale , Virus Zika/génétique , Infection par le virus Zika/génétique , Infection par le virus Zika/virologie , p21-Activated Kinases/génétique , p21-Activated Kinases/métabolisme , Protéine G RhoB/génétique
8.
Bioorg Med Chem Lett ; 40: 127906, 2021 05 15.
Article de Anglais | MEDLINE | ID: mdl-33689873

RÉSUMÉ

Zika virus has emerged as a potential threat to human health globally. A previous drug repurposing screen identified the approved anthelminthic drug niclosamide as a small molecule inhibitor of Zika virus infection. However, as antihelminthic drugs are generally designed to have low absorption when dosed orally, the very limited bioavailability of niclosamide will likely hinder its potential direct repurposing as an antiviral medication. Here, we conducted SAR studies focusing on the anilide and salicylic acid regions of niclosamide to improve physicochemical properties such as microsomal metabolic stability, permeability and solubility. We found that the 5-bromo substitution in the salicylic acid region retains potency while providing better drug-like properties. Other modifications in the anilide region with 2'-OMe and 2'-H substitutions were also advantageous. We found that the 4'-NO2 substituent can be replaced with a 4'-CN or 4'-CF3 substituents. Together, these modifications provide a basis for optimizing the structure of niclosamide to improve systemic exposure for application of niclosamide analogs as drug lead candidates for treating Zika and other viral infections. Indeed, key analogs were also able to rescue cells from the cytopathic effect of SARS-CoV-2 infection, indicating relevance for therapeutic strategies targeting the COVID-19 pandemic.


Sujet(s)
Antiviraux/pharmacologie , Niclosamide/analogues et dérivés , Niclosamide/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Virus Zika/effets des médicaments et des substances chimiques , Animaux , Antiviraux/synthèse chimique , Antiviraux/métabolisme , Sites de fixation , Chlorocebus aethiops , Stabilité de médicament , Humains , Tests de sensibilité microbienne , Microsomes du foie/métabolisme , Simulation de docking moléculaire , Structure moléculaire , Niclosamide/métabolisme , Liaison aux protéines , Rats , Serine endopeptidases/composition chimique , Serine endopeptidases/métabolisme , Relation structure-activité , Cellules Vero , Protéines virales non structurales/composition chimique , Protéines virales non structurales/métabolisme , Protéines virales/composition chimique , Protéines virales/métabolisme
9.
Genomics Proteomics Bioinformatics ; 19(1): 108-122, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33610792

RÉSUMÉ

The Zika virus (ZIKV) and dengue virus (DENV) flaviviruses exhibit similar replicative processes but have distinct clinical outcomes. A systematic understanding of virus-host protein-protein interaction networks can reveal cellular pathways critical to viral replication and disease pathogenesis. Here we employed three independent systems biology approaches toward this goal. First, protein array analysis of direct interactions between individual ZIKV/DENV viral proteins and 20,240 human proteins revealed multiple conserved cellular pathways and protein complexes, including proteasome complexes. Second, an RNAi screen of 10,415 druggable genes identified the host proteins required for ZIKV infection and uncovered that proteasome proteins were crucial in this process. Third, high-throughput screening of 6016 bioactive compounds for ZIKV inhibition yielded 134 effective compounds, including six proteasome inhibitors that suppress both ZIKV and DENV replication. Integrative analyses of these orthogonal datasets pinpoint proteasomes as critical host machinery for ZIKV/DENV replication. Our study provides multi-omics datasets for further studies of flavivirus-host interactions, disease pathogenesis, and new drug targets.


Sujet(s)
Virus de la dengue , Dengue/génétique , Proteasome endopeptidase complex , Infection par le virus Zika , Virus Zika , Virus de la dengue/génétique , Virus de la dengue/physiologie , Humains , Proteasome endopeptidase complex/génétique , Biologie des systèmes , Réplication virale , Virus Zika/génétique , Virus Zika/physiologie , Infection par le virus Zika/génétique
10.
Nat Biotechnol ; 39(6): 747-753, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33623157

RÉSUMÉ

Computational approaches for drug discovery, such as quantitative structure-activity relationship, rely on structural similarities of small molecules to infer biological activity but are often limited to identifying new drug candidates in the chemical spaces close to known ligands. Here we report a biological activity-based modeling (BABM) approach, in which compound activity profiles established across multiple assays are used as signatures to predict compound activity in other assays or against a new target. This approach was validated by identifying candidate antivirals for Zika and Ebola viruses based on high-throughput screening data. BABM models were then applied to predict 311 compounds with potential activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Of the predicted compounds, 32% had antiviral activity in a cell culture live virus assay, the most potent compounds showing a half-maximal inhibitory concentration in the nanomolar range. Most of the confirmed anti-SARS-CoV-2 compounds were found to be viral entry inhibitors and/or autophagy modulators. The confirmed compounds have the potential to be further developed into anti-SARS-CoV-2 therapies.


Sujet(s)
Antiviraux/pharmacologie , Traitements médicamenteux de la COVID-19 , Tests de criblage à haut débit/méthodes , SARS-CoV-2/effets des médicaments et des substances chimiques , COVID-19/génétique , COVID-19/virologie , Découverte de médicament/méthodes , Évaluation préclinique de médicament/méthodes , Humains , SARS-CoV-2/pathogénicité
11.
Cell Stem Cell ; 27(4): 503-504, 2020 10 01.
Article de Anglais | MEDLINE | ID: mdl-33007231

RÉSUMÉ

Among the common flaviviruses that infect humans, Zika virus, and the contemporary Asian strain in particular, is distinctively associated with microcephaly. Zeng et al. (2020) report in this issue of Cell Stem Cell that strong binding and inhibition of human Dicer enzyme by the capsid protein is a potential mechanism for this unique pathogenic potential.


Sujet(s)
microARN , Microcéphalie , Infection par le virus Zika , Virus Zika , Capside , Protéines de capside , Humains , Ribonuclease III/génétique
12.
Antiviral Res ; 183: 104935, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32949636

RÉSUMÉ

Zika virus (ZIKV) outbreaks have been reported worldwide, including a recent occurrence in Brazil where it spread rapidly, and an association with increased cases of microcephaly was observed in addition to neurological issues such as GBS that were reported during previous outbreaks. Following infection of neuronal tissues, ZIKV can cause inflammation, which may lead to neuronal abnormalities, including seizures and paralysis. Therefore, a drug containing both anti-viral and immunosuppressive properties would be of great importance in combating ZIKV related neurological abnormalities. Castanospermine (CST) is potentially a right candidate drug as it reduced viral load and brain inflammation with the resulting appearance of delayed neuronal disorders, including seizures and paralysis in an Ifnar1-/- mouse.


Sujet(s)
Antiviraux/usage thérapeutique , Indolizine/usage thérapeutique , Inflammation/traitement médicamenteux , Crises épileptiques/traitement médicamenteux , Crises épileptiques/virologie , Infection par le virus Zika/complications , Infection par le virus Zika/traitement médicamenteux , Animaux , Lignée cellulaire tumorale , Chlorocebus aethiops , Modèles animaux de maladie humaine , Femelle , Glioblastome , Humains , Immunosuppresseurs/usage thérapeutique , Inflammation/virologie , Mâle , Souris , Crises épileptiques/immunologie , Cellules Vero , Charge virale/effets des médicaments et des substances chimiques , Virus Zika/effets des médicaments et des substances chimiques
13.
Bioorg Chem ; 104: 104205, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32916389

RÉSUMÉ

Zika virus (ZIKV) infection represents a significant threat to the global health system, and the search for efficient antivirals to ZIKV remains necessary and urgent. In this study, we extended the exploration of our previously discovered scaffold of 1H-pyrrolo[1,2-c]imidazol-1-one and revealed that two trans isomers of compounds 2 and 7 and one mixture with major trans isomer of compound 3 as novel tetrahydroquinoline-fused imidazolone derivatives are active against ZIKV infection but they are not virucidal. Western Blot and ELISA analyses of ZIKV NS5 and NS1 further demonstrate that compounds of (±)-2, (±)-3 and (±)-7 act as effective agents against ZIKV infection. We show that the N10's basicity is not the basic requirement for these compounds' antiviral activity in the current work. Importantly, tuning of some pharmacophores including substituents at arene can generate promising candidates for anti-ZIKV agents.


Sujet(s)
Antiviraux/pharmacologie , Infection par le virus Zika/traitement médicamenteux , Virus Zika/effets des médicaments et des substances chimiques , Animaux , Antiviraux/synthèse chimique , Antiviraux/composition chimique , Cellules cultivées , Chlorocebus aethiops , Relation dose-effet des médicaments , Humains , Tests de sensibilité microbienne , Modèles moléculaires , Structure moléculaire , Relation structure-activité , Cellules Vero , Protéines virales non structurales/analyse , Protéines virales non structurales/antagonistes et inhibiteurs , Protéines virales non structurales/métabolisme , Infection par le virus Zika/métabolisme
14.
bioRxiv ; 2020 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-32766591

RÉSUMÉ

The recent global pandemic caused by the new coronavirus SARS-CoV-2 presents an urgent need for new therapeutic candidates. While the importance of traditional in silico approaches such as QSAR in such efforts in unquestionable, these models fundamentally rely on structural similarity to infer biological activity and are thus prone to becoming trapped in the very nearby chemical spaces of already known ligands. For novel and unprecedented threats such as COVID-19 much faster and efficient paradigms must be devised to accelerate the identification of new chemical classes for rapid drug development. Here we report the development of a new biological activity-based modeling (BABM) approach that builds on the hypothesis that compounds with similar activity patterns tend to share similar targets or mechanisms of action. In BABM, compound activity profiles established on massive scale across multiple assays are used as signatures to predict compound activity in a new assay or against a new target. We first trained and validated this approach by identifying new antiviral lead candidates for Zika and Ebola based on data from ~0.5 million compounds screened against ~2,000 assays. BABM models were then applied to predict ~300 compounds not previously reported to have activity for SARS-CoV-2, which were then tested in a live virus assay with high (>30%) hit rates. The most potent compounds showed antiviral activities in the nanomolar range. These potent confirmed compounds have the potential to be further developed in novel chemical space into new anti-SARS-CoV-2 therapies. These results demonstrate unprecedented ability using BABM to predict novel structures as chemical leads significantly beyond traditional methods, and its application in rapid drug discovery response in a global public health crisis.

15.
Front Microbiol ; 11: 598203, 2020.
Article de Anglais | MEDLINE | ID: mdl-33424801

RÉSUMÉ

The 2015 to 2016 outbreak of Zika virus (ZIKV) infections in the Americas coincided with a dramatic increase in neurodevelopmental abnormalities, including fetal microcephaly, in newborns born to infected women. In this study, we observed mitochondrial fragmentation and disrupted mitochondrial membrane potential after 24 h of ZIKV infection in human neural stem cells and the SNB-19 glioblastoma cell line. The severity of these changes correlated with the amount of ZIKV proteins expressed in infected cells. ZIKV infection also decreased the levels of mitofusin 2, which modulates mitochondria fusion. Mitochondrial division inhibitor 1 (Mdivi-1), a small molecule inhibiting mitochondria fission, ameliorated mitochondria disruptions and reduced cell death in ZIKV-infected cells. Collectively, this study suggests that abnormal mitochondrial fragmentation contributes to ZIKV-induced neuronal cell death; rebalancing mitochondrial dynamics of fission-fusion could be a therapeutic strategy for drug development to treat ZIKV-mediated neuronal apoptosis.

16.
Eur J Med Chem ; 187: 111925, 2020 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-31838328

RÉSUMÉ

The Zika endemic established by imported and local transmission is of significant concern and effective anti-ZIKV drugs remain an urgent unmet need. As andrographolide was identified to be an inhibitor of DENV and CHIKV and the importance of quinoline structure against infectious diseases was considered, we are interested in studying its andrographolide derivatives with quinoline moiety against Zika virus infection. In addition to screening eight in-house derivatives of andrographolide, sixteen new derivatives were designed, synthesized and tested against Zika virus infection. Among these compounds, two most potent anti-Zika compounds of 19-acetylated 14α-(5',7'-dichloro-8'-quinolyloxy) derivative 17b and 14ß-(8'-quinolyloxy)-3,19- diol derivative 3 with the highest selectivity were discovered. The SAR analysis indicates that rational and optimal combined modification/s at 3-, 14-, or 19-positions can make derivatives less toxic and more potent against Zika infection, and both of 3 and 17b are suitable as leads for designing new generation of andrographolide derivatives with quinoline or its structure- and property-related moieties against Zika virus and other arboviruses.


Sujet(s)
Antiviraux/pharmacologie , Diterpènes/synthèse chimique , Diterpènes/pharmacologie , Conception de médicament , Découverte de médicament , Infection par le virus Zika/traitement médicamenteux , Virus Zika/effets des médicaments et des substances chimiques , Antiviraux/synthèse chimique , Antiviraux/composition chimique , Diterpènes/composition chimique , Relation dose-effet des médicaments , Humains , Tests de sensibilité microbienne , Structure moléculaire , Relation structure-activité , Cellules cancéreuses en culture
17.
J Virol ; 93(20)2019 10 15.
Article de Anglais | MEDLINE | ID: mdl-31375586

RÉSUMÉ

Zika virus (ZIKV) infection attenuates the growth of human neural progenitor cells (hNPCs). As these hNPCs generate the cortical neurons during early brain development, the ZIKV-mediated growth retardation potentially contributes to the neurodevelopmental defects of the congenital Zika syndrome. Here, we investigate the mechanism by which ZIKV manipulates the cell cycle in hNPCs and the functional consequence of cell cycle perturbation on the replication of ZIKV and related flaviviruses. We demonstrate that ZIKV, but not dengue virus (DENV), induces DNA double-strand breaks (DSBs), triggering the DNA damage response through the ATM/Chk2 signaling pathway while suppressing the ATR/Chk1 signaling pathway. Furthermore, ZIKV infection impedes the progression of cells through S phase, thereby preventing the completion of host DNA replication. Recapitulation of the S-phase arrest state with inhibitors led to an increase in ZIKV replication, but not of West Nile virus or DENV. Our data identify ZIKV's ability to induce DSBs and suppress host DNA replication, which results in a cellular environment favorable for its replication.IMPORTANCE Clinically, Zika virus (ZIKV) infection can lead to developmental defects in the cortex of the fetal brain. How ZIKV triggers this event in developing neural cells is not well understood at a molecular level and likely requires many contributing factors. ZIKV efficiently infects human neural progenitor cells (hNPCs) and leads to growth arrest of these cells, which are critical for brain development. Here, we demonstrate that infection with ZIKV, but not dengue virus, disrupts the cell cycle of hNPCs by halting DNA replication during S phase and inducing DNA damage. We further show that ZIKV infection activates the ATM/Chk2 checkpoint but prevents the activation of another checkpoint, the ATR/Chk1 pathway. These results unravel an intriguing mechanism by which an RNA virus interrupts host DNA replication. Finally, by mimicking virus-induced S-phase arrest, we show that ZIKV manipulates the cell cycle to benefit viral replication.


Sujet(s)
Altération de l'ADN , Cellules souches neurales/métabolisme , Cellules souches neurales/virologie , Réplication virale , Infection par le virus Zika/génétique , Infection par le virus Zika/virologie , Virus Zika/physiologie , Marqueurs biologiques , Cycle cellulaire , Lignée cellulaire , Interactions hôte-pathogène/génétique , Humains , Modèles biologiques
18.
Assay Drug Dev Technol ; 17(3): 128-139, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30958701

RÉSUMÉ

Zika virus has recently emerged as a worldwide pathogen and public health burden due to its rapid spread and identification as a causative agent for multiple neurological defects, including congenital microcephaly. While there has been a flurry of recent research to address this emerging pathogen, there are currently no approved drug treatments for ZIKV infection. The gold standard for testing antiviral activity is to quantify infectious virion production. However, current infectious viral production assays, such as the plaque-forming or focus-forming unit assay, are tedious and labor intensive with a low-screening throughput. To facilitate drug development, we developed a Zika viral titration assay using an automated imaging system and an image analysis algorithm for viral colony quantification. This assay retained the principle of the classical virus titer assay, while improving workflow and offering higher screening throughput. In addition, this assay can be broadly adapted to quantification of other viruses.


Sujet(s)
Antiviraux/pharmacologie , Évaluation préclinique de médicament , Tests de criblage à haut débit , Charge virale/effets des médicaments et des substances chimiques , Virus Zika/effets des médicaments et des substances chimiques , Algorithmes , Antiviraux/composition chimique , Automatisation , Humains , Tests de sensibilité microbienne , Imagerie optique , Cellules cancéreuses en culture
19.
Stem Cell Reports ; 11(2): 348-362, 2018 08 14.
Article de Anglais | MEDLINE | ID: mdl-29983385

RÉSUMÉ

Zika virus (ZIKV) and dengue virus (DENV) are two closely related flaviviruses that lead to different clinical outcomes. The mechanism for the distinct pathogenesis of ZIKV and DENV is poorly understood. Here, we investigate ZIKV and DENV infection of macrophages using a human pluripotent stem cell (hPSC)-derived macrophage model and discover key virus-specific responses. ZIKV and DENV productively infect hPSC-derived macrophages. DENV, but not ZIKV, infection of macrophages strongly activates macrophage migration inhibitory factor (MIF) secretion and decreases macrophage migration. Neutralization of MIF leads to improved migratory ability of DENV-infected macrophages. In contrast, ZIKV-infected macrophages exhibit prolonged migration and express low levels of pro-inflammatory cytokines and chemokines. Mechanistically, ZIKV disrupts the nuclear factor κB (NF-κB)-MIF positive feedback loop by inhibiting the NF-κB signaling pathway. Our results demonstrate the utility of hPSC-derived macrophages in infectious disease modeling and suggest that the distinct impact of ZIKV and DENV on macrophage immune response may underlie different pathogenesis of Zika and dengue diseases.


Sujet(s)
Différenciation cellulaire , Virus de la dengue/immunologie , Dengue/immunologie , Macrophages/cytologie , Macrophages/immunologie , Cellules souches pluripotentes/cytologie , Infection par le virus Zika/immunologie , Virus Zika/immunologie , Marqueurs biologiques , Mouvement cellulaire/immunologie , Cellules cultivées , Cytokines/génétique , Interactions hôte-pathogène/immunologie , Humains , Immunophénotypage , Macrophages/métabolisme , Macrophages/virologie , Cellules souches pluripotentes/métabolisme , Réplication virale/immunologie
20.
Cell Discov ; 4: 31, 2018.
Article de Anglais | MEDLINE | ID: mdl-29872540

RÉSUMÉ

The re-emergence of Zika virus (ZIKV) and Ebola virus (EBOV) poses serious and continued threats to the global public health. Effective therapeutics for these maladies is an unmet need. Here, we show that emetine, an anti-protozoal agent, potently inhibits ZIKV and EBOV infection with a low nanomolar half maximal inhibitory concentration (IC50) in vitro and potent activity in vivo. Two mechanisms of action for emetine are identified: the inhibition of ZIKV NS5 polymerase activity and disruption of lysosomal function. Emetine also inhibits EBOV entry. Cephaeline, a desmethyl analog of emetine, which may be better tolerated in patients than emetine, exhibits a similar efficacy against both ZIKV and EBOV infections. Hence, emetine and cephaeline offer pharmaceutical therapies against both ZIKV and EBOV infection.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE