Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 99
Filtrer
1.
bioRxiv ; 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-39026801

RÉSUMÉ

Defining the subset of cellular factors governing SARS-CoV-2 replication can provide critical insights into viral pathogenesis and identify targets for host-directed antiviral therapies. While a number of genetic screens have previously reported SARS-CoV-2 host dependency factors, these approaches relied on utilizing pooled genome-scale CRISPR libraries, which are biased towards the discovery of host proteins impacting early stages of viral replication. To identify host factors involved throughout the SARS-CoV-2 infectious cycle, we conducted an arrayed genome-scale siRNA screen. Resulting data were integrated with published datasets to reveal pathways supported by orthogonal datasets, including transcriptional regulation, epigenetic modifications, and MAPK signalling. The identified proviral host factors were mapped into the SARS-CoV-2 infectious cycle, including 27 proteins that were determined to impact assembly and release. Additionally, a subset of proteins were tested across other coronaviruses revealing 17 potential pan-coronavirus targets. Further studies illuminated a role for the heparan sulfate proteoglycan perlecan in SARS-CoV-2 viral entry, and found that inhibition of the non-canonical NF-kB pathway through targeting of BIRC2 restricts SARS-CoV-2 replication both in vitro and in vivo. These studies provide critical insight into the landscape of virus-host interactions driving SARS-CoV-2 replication as well as valuable targets for host-directed antivirals.

3.
Nat Commun ; 15(1): 1853, 2024 Feb 29.
Article de Anglais | MEDLINE | ID: mdl-38424040

RÉSUMÉ

Many machine learning applications in bioinformatics currently rely on matching gene identities when analyzing input gene signatures and fail to take advantage of preexisting knowledge about gene functions. To further enable comparative analysis of OMICS datasets, including target deconvolution and mechanism of action studies, we develop an approach that represents gene signatures projected onto their biological functions, instead of their identities, similar to how the word2vec technique works in natural language processing. We develop the Functional Representation of Gene Signatures (FRoGS) approach by training a deep learning model and demonstrate that its application to the Broad Institute's L1000 datasets results in more effective compound-target predictions than models based on gene identities alone. By integrating additional pharmacological activity data sources, FRoGS significantly increases the number of high-quality compound-target predictions relative to existing approaches, many of which are supported by in silico and/or experimental evidence. These results underscore the general utility of FRoGS in machine learning-based bioinformatics applications. Prediction networks pre-equipped with the knowledge of gene functions may help uncover new relationships among gene signatures acquired by large-scale OMICs studies on compounds, cell types, disease models, and patient cohorts.


Sujet(s)
Apprentissage profond , Humains , Apprentissage machine , Biologie informatique , Développement de médicament
4.
J Mol Biol ; 436(4): 168409, 2024 02 15.
Article de Anglais | MEDLINE | ID: mdl-38128824

RÉSUMÉ

Human immunodeficiency virus type 1 (HIV-1) stimulates innate immune responses upon infection, including cyclic GMP-AMP synthase (cGAS) signaling that results in type I interferon production. HIV-1-induced activation of cGAS requires the host cell factor polyglutamine binding protein 1 (PQBP1), an intrinsically disordered protein that bridges capsid recognition and cGAS recruitment. However, the molecular details of PQBP1 interactions with the HIV-1 capsid and their functional implications remain poorly understood. Here, we show that PQBP1 binds to HIV-1 capsids through charge complementing contacts between acidic residues in the N-terminal region of PQBP1 and an arginine ring in the central channel of the HIV-1 CA hexamer that makes up the viral capsid. These studies reveal the molecular details of PQBP1's primary interaction with the HIV-1 capsid and suggest that additional elements are likely to contribute to stable capsid binding.


Sujet(s)
Capside , Protéines de liaison à l'ADN , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , Capside/composition chimique , Protéines de capside/composition chimique , Protéines de liaison à l'ADN/composition chimique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/composition chimique , Immunité innée , Nucleotidyltransferases/composition chimique , Liaison aux protéines , Conformation des protéines
5.
Viruses ; 15(12)2023 12 15.
Article de Anglais | MEDLINE | ID: mdl-38140676

RÉSUMÉ

Antiretroviral therapy (ART) has brought the HIV/AIDS epidemic under control, but a curative strategy for viral eradication is still needed. The cessation of ART results in rapid viral rebound from latently infected CD4+ T cells, showing that control of viral replication alone does not fully restore immune function, nor does it eradicate viral reservoirs. With a better understanding of factors and mechanisms that promote viral latency, current approaches are primarily focused on the permanent silencing of latently infected cells ("block and lock") or reactivating HIV-1 gene expression in latently infected cells, in combination with immune restoration strategies to eliminate HIV infected cells from the host ("shock and kill"). In this review, we provide a summary of the current, most promising approaches for HIV-1 cure strategies, including an analysis of both latency-promoting agents (LPA) and latency-reversing agents (LRA) that have shown promise in vitro, ex vivo, and in human clinical trials to reduce the HIV-1 reservoir.


Sujet(s)
Infections à VIH , Séropositivité VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , Latence virale , Réplication virale , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Lymphocytes T CD4+ , Activation virale
6.
Nat Commun ; 14(1): 6030, 2023 09 27.
Article de Anglais | MEDLINE | ID: mdl-37758692

RÉSUMÉ

Influenza A Virus (IAV) is a recurring respiratory virus with limited availability of antiviral therapies. Understanding host proteins essential for IAV infection can identify targets for alternative host-directed therapies (HDTs). Using affinity purification-mass spectrometry and global phosphoproteomic and protein abundance analyses using three IAV strains (pH1N1, H3N2, H5N1) in three human cell types (A549, NHBE, THP-1), we map 332 IAV-human protein-protein interactions and identify 13 IAV-modulated kinases. Whole exome sequencing of patients who experienced severe influenza reveals several genes, including scaffold protein AHNAK, with predicted loss-of-function variants that are also identified in our proteomic analyses. Of our identified host factors, 54 significantly alter IAV infection upon siRNA knockdown, and two factors, AHNAK and coatomer subunit COPB1, are also essential for productive infection by SARS-CoV-2. Finally, 16 compounds targeting our identified host factors suppress IAV replication, with two targeting CDK2 and FLT3 showing pan-antiviral activity across influenza and coronavirus families. This study provides a comprehensive network model of IAV infection in human cells, identifying functional host targets for pan-viral HDT.


Sujet(s)
COVID-19 , Sous-type H5N1 du virus de la grippe A , Virus de la grippe A , Grippe humaine , Humains , Virus de la grippe A/génétique , Grippe humaine/génétique , Sous-type H5N1 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/métabolisme , Protéomique , Réplication virale/génétique , SARS-CoV-2 , Antiviraux/métabolisme , Interactions hôte-pathogène/génétique
7.
Nat Commun ; 14(1): 948, 2023 02 20.
Article de Anglais | MEDLINE | ID: mdl-36804936

RÉSUMÉ

Small molecule inhibitors of glycosylation enzymes are valuable tools for dissecting glycan functions and potential drug candidates. Screening for inhibitors of glycosyltransferases are mainly performed by in vitro enzyme assays with difficulties moving candidates to cells and animals. Here, we circumvent this by employing a cell-based screening assay using glycoengineered cells expressing tailored reporter glycoproteins. We focused on GalNAc-type O-glycosylation and selected the GalNAc-T11 isoenzyme that selectively glycosylates endocytic low-density lipoprotein receptor (LDLR)-related proteins as targets. Our screen of a limited small molecule compound library did not identify selective inhibitors of GalNAc-T11, however, we identify two compounds that broadly inhibited Golgi-localized glycosylation processes. These compounds mediate the reversible fragmentation of the Golgi system without affecting secretion. We demonstrate how these inhibitors can be used to manipulate glycosylation in cells to induce expression of truncated O-glycans and augment binding of cancer-specific Tn-glycoprotein antibodies and to inhibit expression of heparan sulfate and binding and infection of SARS-CoV-2.


Sujet(s)
COVID-19 , SARS-CoV-2 , Animaux , Glycosylation , SARS-CoV-2/métabolisme , Glycoprotéines/métabolisme , Polyosides/métabolisme
8.
Stem Cells Dev ; 32(9-10): 225-236, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36803114

RÉSUMÉ

SARS-CoV-2 infection during pregnancy has been associated with poor maternal and neonatal outcomes and placental defects. The placenta, which acts as a physical and immunological barrier at the maternal-fetal interface, is not established until the end of the first trimester. Therefore, localized viral infection of the trophoblast compartment early in gestation could trigger an inflammatory response resulting in altered placental function and consequent suboptimal conditions for fetal growth and development. In this study, we investigated the effect of SARS-CoV-2 infection in early gestation placentae using placenta-derived human trophoblast stem cells (TSCs), a novel in vitro model, and their extravillous trophoblast (EVT) and syncytiotrophoblast (STB) derivatives. SARS-CoV-2 was able to productively replicate in TSC-derived STB and EVT, but not undifferentiated TSCs, which is consistent with the expression of SARS-CoV-2 entry host factors, ACE2 (angiotensin-converting enzyme 2) and TMPRSS2 (transmembrane cellular serine protease) in these cells. In addition, both TSC-derived EVT and STB infected with SARS-CoV-2 elicited an interferon-mediated innate immune response. Combined, these results suggest that placenta-derived TSCs are a robust in vitro model to investigate the effect of SARS-CoV-2 infection in the trophoblast compartment of the early placenta and that SARS-CoV-2 infection in early gestation activates the innate immune response and inflammation pathways. Therefore, placental development could be adversely affected by early SARS-CoV-2 infection by directly infecting the developing differentiated trophoblast compartment, posing a higher risk for poor pregnancy outcomes.


Sujet(s)
COVID-19 , SARS-CoV-2 , Nouveau-né , Grossesse , Femelle , Humains , COVID-19/métabolisme , Trophoblastes/métabolisme , Interférons , Placenta
9.
PLoS Biol ; 20(11): e3001845, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-36327326

RÉSUMÉ

The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19), which was rapidly declared a pandemic by the World Health Organization (WHO). Early clinical symptomatology focused mainly on respiratory illnesses. However, a variety of neurological manifestations in both adults and newborns are now well-documented. To experimentally determine whether SARS-CoV-2 could replicate in and affect human brain cells, we infected iPSC-derived human brain organoids. Here, we show that SARS-CoV-2 can productively replicate and promote death of neural cells, including cortical neurons. This phenotype was accompanied by loss of excitatory synapses in neurons. Notably, we found that the U.S. Food and Drug Administration (FDA)-approved antiviral Sofosbuvir was able to inhibit SARS-CoV-2 replication and rescued these neuronal alterations in infected brain organoids. Given the urgent need for readily available antivirals, these results provide a cellular basis supporting repurposed antivirals as a strategic treatment to alleviate neurocytological defects that may underlie COVID-19- related neurological symptoms.


Sujet(s)
Traitements médicamenteux de la COVID-19 , SARS-CoV-2 , Nouveau-né , Humains , Sofosbuvir/pharmacologie , Sofosbuvir/usage thérapeutique , Organoïdes , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Encéphale , Mort cellulaire , Synapses
10.
Sci Adv ; 8(40): eabm5859, 2022 10 07.
Article de Anglais | MEDLINE | ID: mdl-36197970

RÉSUMÉ

Molecular responses to influenza A virus (IAV) infections vary between mammalian species. To identify conserved and species-specific molecular responses, we perform a comparative study of transcriptomic data derived from blood cells, primary epithelial cells, and lung tissues collected from IAV-infected humans, ferrets, and mice. The molecular responses in the human host have unique functions such as antigen processing that are not observed in mice or ferrets. Highly conserved gene coexpression modules across the three species are enriched for IAV infection-induced pathways including cell cycle and interferon (IFN) signaling. TDRD7 is predicted as an IFN-inducible host factor that is up-regulated upon IAV infection in the three species. TDRD7 is required for antiviral IFN response, potentially modulating IFN signaling via the JAK/STAT/IRF9 pathway. Identification of the common and species-specific molecular signatures, networks, and regulators of IAV infection provides insights into host-defense mechanisms and will facilitate the development of novel therapeutic interventions against IAV infection.


Sujet(s)
Maladies transmissibles , Virus de la grippe A , Grippe humaine , Infections à Orthomyxoviridae , Animaux , Antiviraux , Furets/métabolisme , Humains , Virus de la grippe A/physiologie , Grippe humaine/génétique , Interférons/métabolisme , Souris , Infections à Orthomyxoviridae/génétique , Ribonucléoprotéines
11.
Mol Cell ; 82(15): 2871-2884.e6, 2022 08 04.
Article de Anglais | MEDLINE | ID: mdl-35809572

RÉSUMÉ

We have previously described polyglutamine-binding protein 1 (PQBP1) as an adapter required for the cyclic GMP-AMP synthase (cGAS)-mediated innate response to the human immunodeficiency virus 1 (HIV-1) and other lentiviruses. Cytoplasmic HIV-1 DNA is a transient and low-abundance pathogen-associated molecular pattern (PAMP), and the mechanism for its detection and verification is not fully understood. Here, we show a two-factor authentication strategy by the innate surveillance machinery to selectively respond to the low concentration of HIV-1 DNA, while distinguishing these species from extranuclear DNA molecules. We find that, upon HIV-1 infection, PQBP1 decorates the intact viral capsid, and this serves as a primary verification step for the viral nucleic acid cargo. As reverse transcription and capsid disassembly initiate, cGAS is recruited to the capsid in a PQBP1-dependent manner. This positions cGAS at the site of PAMP generation and sanctions its response to a low-abundance DNA PAMP.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Capside/métabolisme , ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Humains , Immunité innée , Nucleotidyltransferases/métabolisme , Molécules contenant des motifs associés aux pathogènes/métabolisme
12.
ACS Infect Dis ; 8(7): 1265-1279, 2022 07 08.
Article de Anglais | MEDLINE | ID: mdl-35766385

RÉSUMÉ

There is a pressing need for host-directed therapeutics that elicit broad-spectrum antiviral activities to potentially address current and future viral pandemics. Apratoxin S4 (Apra S4) is a potent Sec61 inhibitor that prevents cotranslational translocation of secretory proteins into the endoplasmic reticulum (ER), leading to anticancer and antiangiogenic activity both in vitro and in vivo. Since Sec61 has been shown to be an essential host factor for viral proteostasis, we tested Apra S4 in cellular models of viral infection, including SARS-CoV-2, influenza A virus, and flaviviruses (Zika, West Nile, and Dengue virus). Apra S4 inhibited viral replication in a concentration-dependent manner and had high potency particularly against SARS-CoV-2 and influenza A virus, with subnanomolar activity in human cells. Characterization studies focused on SARS-CoV-2 revealed that Apra S4 impacted a post-entry stage of the viral life-cycle. Transmission electron microscopy revealed that Apra S4 blocked formation of stacked double-membrane vesicles, the sites of viral replication. Apra S4 reduced dsRNA formation and prevented viral protein production and trafficking of secretory proteins, especially the spike protein. Given the potent and broad-spectrum activity of Apra S4, further preclinical evaluation of Apra S4 and other Sec61 inhibitors as antivirals is warranted.


Sujet(s)
Traitements médicamenteux de la COVID-19 , Virus de la grippe A , Infection par le virus Zika , Virus Zika , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Depsipeptides , Humains , Pandémies , SARS-CoV-2 , Infection par le virus Zika/traitement médicamenteux
13.
iScience ; 25(5): 104311, 2022 May 20.
Article de Anglais | MEDLINE | ID: mdl-35502318

RÉSUMÉ

Novel strategies are needed to identify drug targets and treatments for the COVID-19 pandemic. The altered gene expression of virus-infected host cells provides an opportunity to specifically inhibit viral propagation via targeting the synthetic lethal and synthetic dosage lethal (SL/SDL) partners of such altered host genes. Pursuing this disparate antiviral strategy, here we comprehensively analyzed multiple in vitro and in vivo bulk and single-cell RNA-sequencing datasets of SARS-CoV-2 infection to predict clinically relevant candidate antiviral targets that are SL/SDL with altered host genes. The predicted SL/SDL-based targets are highly enriched for infected cell inhibiting genes reported in four SARS-CoV-2 CRISPR-Cas9 genome-wide genetic screens. We further selected a focused subset of 26 genes that we experimentally tested in a targeted siRNA screen using human Caco-2 cells. Notably, as predicted, knocking down these targets reduced viral replication and cell viability only under the infected condition without harming noninfected healthy cells.

14.
PLoS Pathog ; 17(11): e1009409, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34843601

RÉSUMÉ

The HIV-1 accessory protein Vpu modulates membrane protein trafficking and degradation to provide evasion of immune surveillance. Targets of Vpu include CD4, HLAs, and BST-2. Several cellular pathways co-opted by Vpu have been identified, but the picture of Vpu's itinerary and activities within membrane systems remains incomplete. Here, we used fusion proteins of Vpu and the enzyme ascorbate peroxidase (APEX2) to compare the ultrastructural locations and the proximal proteomes of wild type Vpu and Vpu-mutants. The proximity-omes of the proteins correlated with their ultrastructural locations and placed wild type Vpu near both retromer and ESCRT-0 complexes. Hierarchical clustering of protein abundances across the mutants was essential to interpreting the data and identified Vpu degradation-targets including CD4, HLA-C, and SEC12 as well as Vpu-cofactors including HGS, STAM, clathrin, and PTPN23, an ALIX-like protein. The Vpu-directed degradation of BST-2 was supported by STAM and PTPN23 and to a much lesser extent by the retromer subunits Vps35 and SNX3. PTPN23 also supported the Vpu-directed decrease in CD4 at the cell surface. These data suggest that Vpu directs targets from sorting endosomes to degradation at multi-vesicular bodies via ESCRT-0 and PTPN23.


Sujet(s)
Complexes de tri endosomique requis pour le transport/métabolisme , Infections à VIH/virologie , Protéines du virus de l'immunodéficience humaine/métabolisme , Protein Tyrosine Phosphatases, Non-Receptor/métabolisme , Protéome/métabolisme , Nexines de tri/métabolisme , Protéines du transport vésiculaire/métabolisme , Protéines virales régulatrices ou accessoires/métabolisme , Protéines viroporines/métabolisme , Complexes de tri endosomique requis pour le transport/génétique , Infections à VIH/génétique , Infections à VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Cellules HeLa , Protéines du virus de l'immunodéficience humaine/génétique , Humains , Microscopie électronique , Motifs et domaines d'intéraction protéique , Multimérisation de protéines , Transport des protéines , Protein Tyrosine Phosphatases, Non-Receptor/génétique , Protéome/analyse , Nexines de tri/composition chimique , Nexines de tri/génétique , Protéines du transport vésiculaire/composition chimique , Protéines du transport vésiculaire/génétique , Protéines virales régulatrices ou accessoires/génétique , Protéines viroporines/génétique
15.
Mol Syst Biol ; 17(11): e10260, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34709707

RÉSUMÉ

Tremendous progress has been made to control the COVID-19 pandemic caused by the SARS-CoV-2 virus. However, effective therapeutic options are still rare. Drug repurposing and combination represent practical strategies to address this urgent unmet medical need. Viruses, including coronaviruses, are known to hijack host metabolism to facilitate viral proliferation, making targeting host metabolism a promising antiviral approach. Here, we describe an integrated analysis of 12 published in vitro and human patient gene expression datasets on SARS-CoV-2 infection using genome-scale metabolic modeling (GEM), revealing complicated host metabolism reprogramming during SARS-CoV-2 infection. We next applied the GEM-based metabolic transformation algorithm to predict anti-SARS-CoV-2 targets that counteract the virus-induced metabolic changes. We successfully validated these targets using published drug and genetic screen data and by performing an siRNA assay in Caco-2 cells. Further generating and analyzing RNA-sequencing data of remdesivir-treated Vero E6 cell samples, we predicted metabolic targets acting in combination with remdesivir, an approved anti-SARS-CoV-2 drug. Our study provides clinical data-supported candidate anti-SARS-CoV-2 targets for future evaluation, demonstrating host metabolism targeting as a promising antiviral strategy.


Sujet(s)
AMP/analogues et dérivés , Alanine/analogues et dérivés , Antiviraux/usage thérapeutique , COVID-19/métabolisme , Voies et réseaux métaboliques/génétique , Pandémies , SARS-CoV-2/physiologie , AMP/usage thérapeutique , Alanine/usage thérapeutique , Animaux , COVID-19/virologie , Cellules Caco-2 , Chlorocebus aethiops , Jeux de données comme sujet , Développement de médicament , Repositionnement des médicaments , Interactions hôte-pathogène , Humains , Petit ARN interférent , Analyse de séquence d'ARN , Cellules Vero , Traitements médicamenteux de la COVID-19
16.
Nat Microbiol ; 6(10): 1319-1333, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34556855

RÉSUMÉ

The fate of influenza A virus (IAV) infection in the host cell depends on the balance between cellular defence mechanisms and viral evasion strategies. To illuminate the landscape of IAV cellular restriction, we generated and integrated global genetic loss-of-function screens with transcriptomics and proteomics data. Our multi-omics analysis revealed a subset of both IFN-dependent and independent cellular defence mechanisms that inhibit IAV replication. Amongst these, the autophagy regulator TBC1 domain family member 5 (TBC1D5), which binds Rab7 to enable fusion of autophagosomes and lysosomes, was found to control IAV replication in vitro and in vivo and to promote lysosomal targeting of IAV M2 protein. Notably, IAV M2 was observed to abrogate TBC1D5-Rab7 binding through a physical interaction with TBC1D5 via its cytoplasmic tail. Our results provide evidence for the molecular mechanism utilised by IAV M2 protein to escape lysosomal degradation and traffic to the cell membrane, where it supports IAV budding and growth.


Sujet(s)
Autophagie , Échappement immunitaire , Virus de la grippe A/physiologie , Antiviraux/métabolisme , Protéines d'activation de la GTPase/génétique , Protéines d'activation de la GTPase/métabolisme , Interactions hôte-pathogène , Humains , Virus de la grippe A/pathogénicité , Lysosomes/métabolisme , Liaison aux protéines , Protéines de la matrice virale/métabolisme , Réplication virale , Protéines G rab/métabolisme , Protéines Rab7 liant le GTP
17.
J Virol ; 95(22): e0099621, 2021 10 27.
Article de Anglais | MEDLINE | ID: mdl-34468177

RÉSUMÉ

Zika virus (ZIKV) is a mosquito-borne pathogen classified by the World Health Organization (WHO) as a public health emergency of international concern in 2016, and it is still identified as a priority disease. Although most infected individuals are asymptomatic or show mild symptoms, a risk of neurologic complications is associated with infection in adults. Additionally, infection during pregnancy is directly linked to microcephaly and other congenital malformations. Since there are no currently available vaccines or approved therapeutics for this virus, there is a critical unmet need in developing treatments to prevent future ZIKV outbreaks. Toward this end, we performed a large-scale cell-based high-content screen of 51,520 chemical compounds to identify potential antiviral drug candidates. The compound (2E)-N-benzyl-3-(4-butoxyphenyl)prop-2-enamide (SBI-0090799) was found to inhibit replication of multiple ZIKV strains and in different cell systems. SBI-0090799 did not affect viral entry or RNA translation but suppressed RNA replication by preventing the formation of the membranous replication compartment. Selection of drug-resistant viruses identified single-amino-acid substitutions in the N-terminal region of nonstructural protein NS4A, arguing this is the likely drug target. These resistance mutations rescued viral RNA replication and restored the formation of the membranous replication compartment. This mechanism of action is similar to clinically approved NS5A inhibitors for hepatitis C virus (HCV). Taken together, SBI-0090799 represents a promising lead candidate for the development of an antiviral treatment against ZIKV infection for the mitigation of severe complications and potential resurgent outbreaks of the virus. IMPORTANCE This study describes the elucidation of (2E)-N-benzyl-3-(4-butoxyphenyl)prop-2-enamide (SBI-0090799) as a selective and potent inhibitor of Zika virus (ZIKV) replication using a high-throughput screening approach. Mapping and resistance studies, supported by electron microscopy observations, indicate that the small molecule is functioning through inhibition of NS4A-mediated formation of ZIKV replication compartments in the endoplasmic reticulum (ER). Intriguingly, this defines a novel nonenzymatic target and chemical matter for the development of a new class of ZIKV antivirals. Moreover, chemical modulation affecting this nonstructural protein mirrors the identification and development of hepatitis C virus (HCV) NS5A inhibitor daclatasvir and its derivatives, similarly interfering with the formation of the viral replication compartment and also targeting a protein with no enzymatic activity, which have been part of a curative strategy for HCV.


Sujet(s)
Antiviraux/pharmacologie , Découverte de médicament/méthodes , Évaluation préclinique de médicament/méthodes , Réplication virale/effets des médicaments et des substances chimiques , Infection par le virus Zika/traitement médicamenteux , Virus Zika/effets des médicaments et des substances chimiques , Animaux , Astrocytes , Chlorocebus aethiops , Cellules dendritiques , Cellules HEK293 , Humains , Culture de cellules primaires , Cellules Vero , Compartiments de réplication virale/effets des médicaments et des substances chimiques
18.
bioRxiv ; 2021 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-34545363

RÉSUMÉ

Novel strategies are needed to identify drug targets and treatments for the COVID-19 pandemic. The altered gene expression of virus-infected host cells provides an opportunity to specifically inhibit viral propagation via targeting the synthetic lethal (SL) partners of such altered host genes. Pursuing this antiviral strategy, here we comprehensively analyzed multiple in vitro and in vivo bulk and single-cell RNA-sequencing datasets of SARS-CoV-2 infection to predict clinically relevant candidate antiviral targets that are SL with altered host genes. The predicted SL-based targets are highly enriched for infected cell inhibiting genes reported in four SARS-CoV-2 CRISPR-Cas9 genome-wide genetic screens. Integrating our predictions with the results of these screens, we further selected a focused subset of 26 genes that we experimentally tested in a targeted siRNA screen using human Caco-2 cells. Notably, as predicted, knocking down these targets reduced viral replication and cell viability only under the infected condition without harming non-infected cells. Our results are made publicly available, to facilitate their in vivo testing and further validation.

19.
Mol Cell ; 81(12): 2656-2668.e8, 2021 06 17.
Article de Anglais | MEDLINE | ID: mdl-33930332

RÉSUMÉ

A deficient interferon (IFN) response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been implicated as a determinant of severe coronavirus disease 2019 (COVID-19). To identify the molecular effectors that govern IFN control of SARS-CoV-2 infection, we conducted a large-scale gain-of-function analysis that evaluated the impact of human IFN-stimulated genes (ISGs) on viral replication. A limited subset of ISGs were found to control viral infection, including endosomal factors inhibiting viral entry, RNA binding proteins suppressing viral RNA synthesis, and a highly enriched cluster of endoplasmic reticulum (ER)/Golgi-resident ISGs inhibiting viral assembly/egress. These included broad-acting antiviral ISGs and eight ISGs that specifically inhibited SARS-CoV-2 and SARS-CoV-1 replication. Among the broad-acting ISGs was BST2/tetherin, which impeded viral release and is antagonized by SARS-CoV-2 Orf7a protein. Overall, these data illuminate a set of ISGs that underlie innate immune control of SARS-CoV-2/SARS-CoV-1 infection, which will facilitate the understanding of host determinants that impact disease severity and offer potential therapeutic strategies for COVID-19.


Sujet(s)
Antigènes CD/génétique , Interactions hôte-pathogène/génétique , Facteurs de régulation d'interféron/génétique , Interféron de type I/génétique , SARS-CoV-2/génétique , Protéines virales/génétique , Animaux , Antigènes CD/composition chimique , Antigènes CD/immunologie , Sites de fixation , Lignée cellulaire tumorale , Chlorocebus aethiops , Réticulum endoplasmique/génétique , Réticulum endoplasmique/immunologie , Réticulum endoplasmique/virologie , Protéines liées au GPI/composition chimique , Protéines liées au GPI/génétique , Protéines liées au GPI/immunologie , Régulation de l'expression des gènes , Appareil de Golgi/génétique , Appareil de Golgi/immunologie , Appareil de Golgi/virologie , Cellules HEK293 , Interactions hôte-pathogène/immunologie , Humains , Immunité innée , Facteurs de régulation d'interféron/classification , Facteurs de régulation d'interféron/immunologie , Interféron de type I/immunologie , Simulation de docking moléculaire , Liaison aux protéines , Structure en hélice alpha , Structure en brin bêta , Motifs et domaines d'intéraction protéique , SARS-CoV-2/immunologie , Transduction du signal , Cellules Vero , Protéines virales/composition chimique , Protéines virales/immunologie , Pénétration virale , Libération de particules virales/génétique , Libération de particules virales/immunologie , Réplication virale/génétique , Réplication virale/immunologie
20.
bioRxiv ; 2021 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-33791697

RÉSUMÉ

We identify the prolyl-tRNA synthetase (PRS) inhibitor halofuginone 1 , a compound in clinical trials for anti-fibrotic and anti-inflammatory applications 2 , as a potent inhibitor of SARS-CoV-2 infection and replication. The interaction of SARS-CoV-2 spike protein with cell surface heparan sulfate (HS) promotes viral entry 3 . We find that halofuginone reduces HS biosynthesis, thereby reducing spike protein binding, SARS-CoV-2 pseudotyped virus, and authentic SARS-CoV-2 infection. Halofuginone also potently suppresses SARS-CoV-2 replication post-entry and is 1,000-fold more potent than Remdesivir 4 . Inhibition of HS biosynthesis and SARS-CoV-2 infection depends on specific inhibition of PRS, possibly due to translational suppression of proline-rich proteins. We find that pp1a and pp1ab polyproteins of SARS-CoV-2, as well as several HS proteoglycans, are proline-rich, which may make them particularly vulnerable to halofuginone's translational suppression. Halofuginone is orally bioavailable, has been evaluated in a phase I clinical trial in humans and distributes to SARS-CoV-2 target organs, including the lung, making it a near-term clinical trial candidate for the treatment of COVID-19.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE