Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Virol ; 97(11): e0087823, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37905840

RESUMEN

IMPORTANCE: Remodeling of the cellular endomembrane system by viruses allows for efficient and coordinated replication of the viral genome in distinct subcellular compartments termed replication organelles. As a critical step in the viral life cycle, replication organelle formation is an attractive target for therapeutic intervention, but factors central to this process are only partially understood. In this study, we corroborate that two viral proteins, nsp3 and nsp4, are the major drivers of membrane remodeling in SARS-CoV-2 infection. We further report a number of host cell factors interacting with these viral proteins and supporting the viral replication cycle, some of them by contributing to the formation of the SARS-CoV-2 replication organelle.


Asunto(s)
COVID-19 , SARS-CoV-2 , Proteínas no Estructurales Virales , Replicación Viral , Humanos , Orgánulos/metabolismo , Proteómica , SARS-CoV-2/fisiología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
2.
Mol Cell ; 83(14): 2559-2577.e8, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37421942

RESUMEN

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) remodels the endoplasmic reticulum (ER) to form replication organelles, leading to ER stress and unfolded protein response (UPR). However, the role of specific UPR pathways in infection remains unclear. Here, we found that SARS-CoV-2 infection causes marginal activation of signaling sensor IRE1α leading to its phosphorylation, clustering in the form of dense ER-membrane rearrangements with embedded membrane openings, and XBP1 splicing. By investigating the factors regulated by IRE1α-XBP1 during SARS-CoV-2 infection, we identified stress-activated kinase NUAK2 as a novel host-dependency factor for SARS-CoV-2, HCoV-229E, and MERS-CoV entry. Reducing NUAK2 abundance or kinase activity impaired SARS-CoV-2 particle binding and internalization by decreasing cell surface levels of viral receptors and viral trafficking likely by modulating the actin cytoskeleton. IRE1α-dependent NUAK2 levels were elevated in SARS-CoV-2-infected and bystander non-infected cells, promoting viral spread by maintaining ACE2 cell surface levels and facilitating virion binding to bystander cells.


Asunto(s)
Proteínas Serina-Treonina Quinasas , SARS-CoV-2 , Internalización del Virus , Humanos , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/metabolismo , COVID-19/metabolismo , COVID-19/patología , COVID-19/virología , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , SARS-CoV-2/fisiología , Respuesta de Proteína Desplegada
3.
Antimicrob Agents Chemother ; 67(2): e0133122, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36700643

RESUMEN

Dengue virus (DENV) is a Flavivirus that causes the most prevalent arthropod-borne viral disease. Clinical manifestation of DENV infection ranges from asymptomatic to severe symptoms that can lead to death. Unfortunately, no antiviral treatments against DENV are currently available. In order to identify novel DENV inhibitors, we screened a library of 1,604 chemically diversified fragment-based compounds using DENV reporter viruses that allowed quantification of viral replication in infected cells. Following a validation screening, the two best inhibitor candidates were N-phenylpyridine-3-carboxamide (NPP3C) and 6-acetyl-1H-indazole (6A1HI). The half maximal effective concentration of NPP3C and 6A1H1 against DENV were 7.1 µM and 6.5 µM, respectively. 6A1H1 decreased infectious DENV particle production up to 1,000-fold without any cytotoxicity at the used concentrations. While 6A1HI was DENV-specific, NPP3C also inhibited the replication of other flaviviruses such as West Nile virus and Zika virus. Structure-activity relationship (SAR) studies with 151 analogues revealed key structural elements of NPP3C and 6A1HI required for their antiviral activity. Time-of-drug-addition experiments identified a postentry step as a target of these compounds. Consistently, using a DENV subgenomic replicon, we demonstrated that these compounds specifically impede the viral RNA replication step and exhibit a high genetic barrier-to-resistance. In contrast, viral RNA translation and the de novo biogenesis of DENV replication organelles were not affected. Overall, our data unveil NPP3C and 6A1H1 as novel DENV inhibitors. The information revealed by our SAR studies will help chemically optimize NPP3C and 6A1H1 in order to improve their anti-flaviviral potency and to challenge them in in vivo models.


Asunto(s)
Virus del Dengue , Dengue , Flavivirus , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Antivirales/farmacología , Antivirales/uso terapéutico , Dengue/tratamiento farmacológico , Virus del Dengue/genética , Estadios del Ciclo de Vida , Replicación de ARN , ARN Viral/genética , Replicación Viral , Virus Zika/genética , ARN Subgenómico/genética
4.
Commun Biol ; 5(1): 45, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-35022513

RESUMEN

SARS-CoV-2 is a novel virus that has rapidly spread, causing a global pandemic. In the majority of infected patients, SARS-CoV-2 leads to mild disease; however, in a significant proportion of infections, individuals develop severe symptoms that can lead to long-lasting lung damage or death. These severe cases are often associated with high levels of pro-inflammatory cytokines and low antiviral responses, which can cause systemic complications. Here, we have evaluated transcriptional and cytokine secretion profiles and detected a distinct upregulation of inflammatory cytokines in infected cell cultures and samples taken from infected patients. Building on these observations, we found a specific activation of NF-κB and a block of IRF3 nuclear translocation in SARS-CoV-2 infected cells. This NF-κB response was mediated by cGAS-STING activation and could be attenuated through several STING-targeting drugs. Our results show that SARS-CoV-2 directs a cGAS-STING mediated, NF-κB-driven inflammatory immune response in human epithelial cells that likely contributes to inflammatory responses seen in patients and could be therapeutically targeted to suppress severe disease symptoms.


Asunto(s)
COVID-19/metabolismo , Síndrome de Liberación de Citoquinas , Mediadores de Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Nucleotidiltransferasas/metabolismo , COVID-19/virología , Humanos , SARS-CoV-2/aislamiento & purificación , Transducción de Señal
5.
Infection ; 50(2): 395-406, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34383260

RESUMEN

PURPOSE: Rapid antigen-detecting tests (Ag-RDTs) for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can transform pandemic control. Thus far, sensitivity (≤ 85%) of lateral-flow assays has limited scale-up. Conceivably, microfluidic immunofluorescence Ag-RDTs could increase sensitivity for SARS-CoV-2 detection. METHODS: This multi-centre diagnostic accuracy study investigated performance of the microfluidic immunofluorescence LumiraDx™ assay, enrolling symptomatic and asymptomatic participants with suspected SARS-CoV-2 infection. Participants collected a supervised nasal mid-turbinate (NMT) self-swab for Ag-RDT testing, in addition to a professionally collected nasopharyngeal (NP) swab for routine testing with reverse transcriptase polymerase chain reaction (RT-PCR). Results were compared to calculate sensitivity and specificity. Sub-analyses investigated the results by viral load, symptom presence and duration. An analytical study assessed exclusivity and limit-of-detection (LOD). In addition, we evaluated ease-of-use. RESULTS: The study was conducted between November 2nd 2020 and 4th of December 2020. 761 participants were enrolled, with 486 participants reporting symptoms on testing day. 120 out of 146 RT-PCR positive cases were detected positive by LumiraDx™, resulting in a sensitivity of 82.2% (95% CI 75.2-87.5%). Specificity was 99.3% (CI 98.3-99.7%). Sensitivity was increased in individuals with viral load ≥ 7 log10 SARS-CoV2 RNA copies/ml (93.8%; CI 86.2-97.3%). Testing against common respiratory commensals and pathogens showed no cross-reactivity and LOD was estimated to be 2-56 PFU/mL. The ease-of-use-assessment was favourable for lower throughput settings. CONCLUSION: The LumiraDx™ assay showed excellent analytical sensitivity, exclusivity and clinical specificity with good clinical sensitivity using supervised NMT self-sampling. TRIAL REGISTRATION NUMBER AND REGISTRATION DATE: DRKS00021220 and 01.04.2020.


Asunto(s)
COVID-19 , SARS-CoV-2 , COVID-19/diagnóstico , Humanos , Pandemias , Sistemas de Atención de Punto , ARN Viral , Sensibilidad y Especificidad
6.
Nat Commun ; 12(1): 7276, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34907161

RESUMEN

Double membrane vesicles (DMVs) serve as replication organelles of plus-strand RNA viruses such as hepatitis C virus (HCV) and SARS-CoV-2. Viral DMVs are morphologically analogous to DMVs formed during autophagy, but lipids driving their biogenesis are largely unknown. Here we show that production of the lipid phosphatidic acid (PA) by acylglycerolphosphate acyltransferase (AGPAT) 1 and 2 in the ER is important for DMV biogenesis in viral replication and autophagy. Using DMVs in HCV-replicating cells as model, we found that AGPATs are recruited to and critically contribute to HCV and SARS-CoV-2 replication and proper DMV formation. An intracellular PA sensor accumulated at viral DMV formation sites, consistent with elevated levels of PA in fractions of purified DMVs analyzed by lipidomics. Apart from AGPATs, PA is generated by alternative pathways and their pharmacological inhibition also impaired HCV and SARS-CoV-2 replication as well as formation of autophagosome-like DMVs. These data identify PA as host cell lipid involved in proper replication organelle formation by HCV and SARS-CoV-2, two phylogenetically disparate viruses causing very different diseases, i.e. chronic liver disease and COVID-19, respectively. Host-targeting therapy aiming at PA synthesis pathways might be suitable to attenuate replication of these viruses.


Asunto(s)
Hepacivirus/genética , Ácidos Fosfatidicos/metabolismo , SARS-CoV-2/genética , Replicación Viral/fisiología , 1-Acilglicerol-3-Fosfato O-Aciltransferasa , Aciltransferasas , Autofagosomas/metabolismo , Autofagia , COVID-19/virología , Línea Celular , Supervivencia Celular , Virus del Dengue , Células HEK293 , Humanos , Proteínas de la Membrana , Glicoproteína de la Espiga del Coronavirus , Proteínas no Estructurales Virales , Proteínas Virales , Virus Zika
7.
Cell Rep ; 37(8): 110049, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34788596

RESUMEN

Positive-strand RNA viruses replicate in close association with rearranged intracellular membranes. For hepatitis C virus (HCV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), these rearrangements comprise endoplasmic reticulum (ER)-derived double membrane vesicles (DMVs) serving as RNA replication sites. Cellular factors involved in DMV biogenesis are poorly defined. Here, we show that despite structural similarity of viral DMVs with autophagosomes, conventional macroautophagy is dispensable for HCV and SARS-CoV-2 replication. However, both viruses exploit factors involved in autophagosome formation, most notably class III phosphatidylinositol 3-kinase (PI3K). As revealed with a biosensor, PI3K is activated in cells infected with either virus to produce phosphatidylinositol 3-phosphate (PI3P) while kinase complex inhibition or depletion profoundly reduces replication and viral DMV formation. The PI3P-binding protein DFCP1, recruited to omegasomes in early steps of autophagosome formation, participates in replication and DMV formation of both viruses. These results indicate that phylogenetically unrelated HCV and SARS-CoV-2 exploit similar components of the autophagy machinery to create their replication organelles.


Asunto(s)
Autofagia/fisiología , Hepacivirus/fisiología , SARS-CoV-2/fisiología , Compartimentos de Replicación Viral/metabolismo , Autofagosomas/metabolismo , Proteínas Portadoras/metabolismo , Fosfatidilinositol 3-Quinasas Clase III/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Humanos , Fosfatos de Fosfatidilinositol/metabolismo , ARN Viral/biosíntesis , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
8.
Viruses ; 13(10)2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34696522

RESUMEN

The dengue virus (DENV) causes the most prevalent arthropod-borne viral disease worldwide. While its incidence is increasing in many countries, there is no approved antiviral therapy currently available. In infected cells, the DENV induces extensive morphological alterations of the endoplasmic reticulum (ER) to generate viral replication organelles (vRO), which include convoluted membranes (CM) and vesicle packets (VP) hosting viral RNA replication. The viral non-structural protein NS4B localizes to vROs and is absolutely required for viral replication through poorly defined mechanisms, which might involve cellular protein partners. Previous interactomic studies identified the ATPase valosin-containing protein (VCP) as a DENV NS4B-interacting host factor in infected cells. Using both pharmacological and dominant-negative inhibition approaches, we show, in this study, that VCP ATPase activity is required for efficient DENV replication. VCP associates with NS4B when expressed in the absence of other viral proteins while in infected cells, both proteins colocalize within large DENV-induced cytoplasmic structures previously demonstrated to be CMs. Consistently, VCP inhibition dramatically reduces the abundance of DENV CMs in infected cells. Most importantly, using a recently reported replication-independent plasmid-based vRO induction system, we show that de novo VP biogenesis is dependent on VCP ATPase activity. Overall, our data demonstrate that VCP ATPase activity is required for vRO morphogenesis and/or stability. Considering that VCP was shown to be required for the replication of other flaviviruses, our results argue that VCP is a pan-flaviviral host dependency factor. Given that new generation VCP-targeting drugs are currently evaluated in clinical trials for cancer treatment, VCP may constitute an attractive broad-spectrum antiviral target in drug repurposing approaches.


Asunto(s)
Virus del Dengue/metabolismo , Proteína que Contiene Valosina/metabolismo , Compartimentos de Replicación Viral/fisiología , Adenosina Trifosfatasas/genética , Línea Celular , Dengue/virología , Virus del Dengue/genética , Virus del Dengue/patogenicidad , Retículo Endoplásmico/virología , Humanos , ARN Viral/genética , Proteína que Contiene Valosina/genética , Proteínas no Estructurales Virales/genética , Replicación Viral/fisiología
9.
J Virol ; 95(24): e0059621, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34586867

RESUMEN

Cellular factors have important roles in all facets of the flavivirus replication cycle. Deciphering viral-host protein interactions is essential for understanding the flavivirus life cycle as well as development of effective antiviral strategies. To uncover novel host factors that are co-opted by multiple flaviviruses, a CRISPR/Cas9 genome wide knockout (KO) screen was employed to identify genes required for replication of Zika virus (ZIKV). Receptor for Activated Protein C Kinase 1 (RACK1) was identified as a novel host factor required for ZIKV replication, which was confirmed via complementary experiments. Depletion of RACK1 via siRNA demonstrated that RACK1 is important for replication of a wide range of mosquito- and tick-borne flaviviruses, including West Nile Virus (WNV), Dengue Virus (DENV), Powassan Virus (POWV) and Langat Virus (LGTV) as well as the coronavirus SARS-CoV-2, but not for YFV, EBOV, VSV or HSV. Notably, flavivirus replication was only abrogated when RACK1 expression was dampened prior to infection. Utilising a non-replicative flavivirus model, we show altered morphology of viral replication factories and reduced formation of vesicle packets (VPs) in cells lacking RACK1 expression. In addition, RACK1 interacted with NS1 protein from multiple flaviviruses; a key protein for replication complex formation. Overall, these findings reveal RACK1's crucial role to the biogenesis of pan-flavivirus replication organelles. IMPORTANCE Cellular factors are critical in all facets of viral lifecycles, where overlapping interactions between the virus and host can be exploited as possible avenues for the development of antiviral therapeutics. Using a genome-wide CRISPR knockout screening approach to identify novel cellular factors important for flavivirus replication we identified RACK1 as a pro-viral host factor for both mosquito- and tick-borne flaviviruses in addition to SARS-CoV-2. Using an innovative flavivirus protein expression system, we demonstrate for the first time the impact of the loss of RACK1 on the formation of viral replication factories known as 'vesicle packets' (VPs). In addition, we show that RACK1 can interact with numerous flavivirus NS1 proteins as a potential mechanism by which VP formation can be induced by the former.


Asunto(s)
Sistemas CRISPR-Cas , Flavivirus/genética , Proteínas de Neoplasias/genética , Receptores de Cinasa C Activada/genética , Replicación Viral , Células A549 , Aedes , Animales , COVID-19 , Chlorocebus aethiops , Culicidae , Virus del Dengue/genética , Estudio de Asociación del Genoma Completo , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , ARN Interferente Pequeño/metabolismo , ARN Viral/metabolismo , SARS-CoV-2 , Células Vero , Virus del Nilo Occidental/genética , Virus Zika/genética , Infección por el Virus Zika/virología
10.
J Virol ; 95(21): e0131021, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34379504

RESUMEN

Dengue virus (DENV) constitutes one of the most important arboviral pathogens affecting humans. The high prevalence of DENV infections, which cause more than 20,000 deaths annually, and the lack of effective vaccines or direct-acting antiviral drugs make it a global health concern. DENV genome replication occurs in close association with the host endomembrane system, which is remodeled to form the viral replication organelle that originates from endoplasmic reticulum (ER) membranes. To date, the viral and cellular determinants responsible for the biogenesis of DENV replication organelles are still poorly defined. The viral nonstructural protein 4A (NS4A) can remodel membranes and has been shown to associate with numerous host factors in DENV-replicating cells. In the present study, we used reverse and forward genetic screens and identified sites within NS4A required for DENV replication. We also mapped the determinants in NS4A required for interactions with other viral proteins. Moreover, taking advantage of our recently developed polyprotein expression system, we evaluated the role of NS4A in the formation of DENV replication organelles. Together, we report a detailed map of determinants within NS4A required for RNA replication, interaction with other viral proteins, and replication organelle formation. Our results suggest that NS4A might be an attractive target for antiviral therapy. IMPORTANCE DENV is the most prevalent mosquito-borne virus, causing around 390 million infections each year. There are no approved therapies to treat DENV infection, and the only available vaccine shows limited efficacy. The viral nonstructural proteins have emerged as attractive drug targets due to their pivotal role in RNA replication and establishment of virus-induced membranous compartments, designated replication organelles (ROs). The transmembrane protein NS4A, generated by cleavage of the NS4A-2K-4B precursor, contributes to DENV replication by unknown mechanisms. Here, we report a detailed genetic interaction map of NS4A and identify residues required for RNA replication and interaction between NS4A-2K-4B and NS2B-3 as well as NS1. Importantly, by means of an expression-based system, we demonstrate the essential role of NS4A in RO biogenesis and identify determinants in NS4A required for this process. Our data suggest that NS4A is an attractive target for antiviral therapy.


Asunto(s)
Virus del Dengue/fisiología , Dengue/virología , Biogénesis de Organelos , Orgánulos/virología , Proteínas no Estructurales Virales/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Chlorocebus aethiops , Virus del Dengue/ultraestructura , Interacciones Microbiota-Huesped , Humanos , Proteínas Mutantes/fisiología , Mutación , Orgánulos/ultraestructura , Unión Proteica , ARN/metabolismo , ARN Viral , Genética Inversa/métodos , Células Vero , Replicación Viral
11.
Mol Cell ; 81(13): 2851-2867.e7, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34118193

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19). SARS-CoV-2 relies on cellular RNA-binding proteins (RBPs) to replicate and spread, although which RBPs control its life cycle remains largely unknown. Here, we employ a multi-omic approach to identify systematically and comprehensively the cellular and viral RBPs that are involved in SARS-CoV-2 infection. We reveal that SARS-CoV-2 infection profoundly remodels the cellular RNA-bound proteome, which includes wide-ranging effects on RNA metabolic pathways, non-canonical RBPs, and antiviral factors. Moreover, we apply a new method to identify the proteins that directly interact with viral RNA, uncovering dozens of cellular RBPs and six viral proteins. Among them are several components of the tRNA ligase complex, which we show regulate SARS-CoV-2 infection. Furthermore, we discover that available drugs targeting host RBPs that interact with SARS-CoV-2 RNA inhibit infection. Collectively, our results uncover a new universe of host-virus interactions with potential for new antiviral therapies against COVID-19.


Asunto(s)
COVID-19/metabolismo , Proteoma/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , SARS-CoV-2/fisiología , Proteínas Virales/metabolismo , Replicación Viral/fisiología , Células A549 , COVID-19/genética , Humanos , Proteoma/genética , ARN Viral/genética , Proteínas de Unión al ARN/genética , Proteínas Virales/genética
12.
ACS Infect Dis ; 7(6): 1457-1468, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33570381

RESUMEN

Two proteases produced by the SARS-CoV-2 virus, the main protease and papain-like protease, are essential for viral replication and have become the focus of drug development programs for treatment of COVID-19. We screened a highly focused library of compounds containing covalent warheads designed to target cysteine proteases to identify new lead scaffolds for both Mpro and PLpro proteases. These efforts identified a small number of hits for the Mpro protease and no viable hits for the PLpro protease. Of the Mpro hits identified as inhibitors of the purified recombinant protease, only two compounds inhibited viral infectivity in cellular infection assays. However, we observed a substantial drop in antiviral potency upon expression of TMPRSS2, a transmembrane serine protease that acts in an alternative viral entry pathway to the lysosomal cathepsins. This loss of potency is explained by the fact that our lead Mpro inhibitors are also potent inhibitors of host cell cysteine cathepsins. To determine if this is a general property of Mpro inhibitors, we evaluated several recently reported compounds and found that they are also effective inhibitors of purified human cathepsins L and B and showed similar loss in activity in cells expressing TMPRSS2. Our results highlight the challenges of targeting Mpro and PLpro proteases and demonstrate the need to carefully assess selectivity of SARS-CoV-2 protease inhibitors to prevent clinical advancement of compounds that function through inhibition of a redundant viral entry pathway.


Asunto(s)
COVID-19 , SARS-CoV-2 , Antivirales/farmacología , Antivirales/uso terapéutico , Humanos , Péptido Hidrolasas , Inhibidores de Proteasas
13.
J Virol ; 95(4)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33257477

RESUMEN

Positive-strand RNA viruses have been the etiological agents in several major disease outbreaks over the last few decades. Examples of this include flaviviruses, such as dengue virus and Zika virus, which cause millions of yearly infections around the globe, and coronaviruses, such as SARS-CoV-2, the source of the current pandemic. The severity of outbreaks caused by these viruses stresses the importance of research aimed at determining methods to limit virus spread and to curb disease severity. Such studies require molecular tools to decipher virus-host interactions and to develop effective treatments. Here, we describe the generation and characterization of a reporter system that can be used to visualize and identify cells infected with dengue virus or SARS-CoV-2. This system is based on viral protease activity that mediates cleavage and nuclear translocation of an engineered fluorescent protein stably expressed in cells. We show the suitability of this system for live cell imaging, for visualization of single infected cells, and for screening and testing of antiviral compounds. With the integrated modular building blocks, this system is easy to manipulate and can be adapted to any virus encoding a protease, thus offering a high degree of flexibility.IMPORTANCE Reporter systems are useful tools for fast and quantitative visualization of virus-infected cells within a host cell population. Here, we describe a reporter system that takes advantage of virus-encoded proteases expressed in infected cells to cleave an ER-anchored fluorescent protein fused to a nuclear localization sequence. Upon cleavage, the GFP moiety translocates to the nucleus, allowing for rapid detection of the infected cells. Using this system, we demonstrate reliable reporting activity for two major human pathogens from the Flaviviridae and the Coronaviridae families: dengue virus and SARS-CoV-2. We apply this reporter system to live cell imaging and use it for proof-of-concept to validate antiviral activity of a nucleoside analogue. This reporter system is not only an invaluable tool for the characterization of viral replication, but also for the discovery and development of antivirals that are urgently needed to halt the spread of these viruses.


Asunto(s)
COVID-19/virología , Virus del Dengue/aislamiento & purificación , Dengue/virología , SARS-CoV-2/aislamiento & purificación , Células A549 , Animales , COVID-19/diagnóstico , COVID-19/metabolismo , COVID-19/patología , Línea Celular , Chlorocebus aethiops , Dengue/diagnóstico , Dengue/metabolismo , Dengue/patología , Virus del Dengue/genética , Virus del Dengue/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Señales de Localización Nuclear/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Células Vero , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
14.
Bioessays ; 43(3): e2000257, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33377226

RESUMEN

Emergence of the novel pathogenic coronavirus SARS-CoV-2 and its rapid pandemic spread presents challenges that demand immediate attention. Here, we describe the development of a semi-quantitative high-content microscopy-based assay for detection of three major classes (IgG, IgA, and IgM) of SARS-CoV-2 specific antibodies in human samples. The possibility to detect antibodies against the entire viral proteome together with a robust semi-automated image analysis workflow resulted in specific, sensitive and unbiased assay that complements the portfolio of SARS-CoV-2 serological assays. Sensitive, specific and quantitative serological assays are urgently needed for a better understanding of humoral immune response against the virus as a basis for developing public health strategies to control viral spread. The procedure described here has been used for clinical studies and provides a general framework for the application of quantitative high-throughput microscopy to rapidly develop serological assays for emerging virus infections.


Asunto(s)
Anticuerpos Antivirales/sangre , COVID-19/diagnóstico , Inmunoensayo , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Microscopía/métodos , SARS-CoV-2/inmunología , COVID-19/inmunología , COVID-19/virología , Prueba de COVID-19/métodos , Técnica del Anticuerpo Fluorescente , Ensayos Analíticos de Alto Rendimiento , Humanos , Procesamiento de Imagen Asistido por Computador/estadística & datos numéricos , Sueros Inmunes/química , Aprendizaje Automático , Sensibilidad y Especificidad
15.
STAR Protoc ; 1(3): 100173, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33377067

RESUMEN

Positive-strand RNA viruses replicate in distinct membranous structures called replication organelles (ROs). Mechanistic studies of RO formation have been difficult because perturbations affecting viral replication have an impact on viral protein amounts, thus affecting RO biogenesis. Here, we present a detailed guide on how to use a replication-independent expression system, designated pIRO (plasmid-induced replication organelle formation), inducing bona fide flavivirus ROs in transfected cells. This will be useful for mechanistic studies of viral and cellular factors driving flavivirus RO biogenesis. For complete details on the use and execution of this protocol, please refer to Cerikan et al. (2020).


Asunto(s)
Técnicas Citológicas/métodos , Flavivirus/fisiología , Orgánulos/ultraestructura , Replicación Viral/fisiología , Línea Celular Tumoral , Flavivirus/ultraestructura , Humanos , Manejo de Especímenes
16.
Nat Commun ; 11(1): 5885, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33208793

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of the COVID19 pandemic, is a highly pathogenic ß-coronavirus. As other coronaviruses, SARS-CoV-2 is enveloped, replicates in the cytoplasm and assembles at intracellular membranes. Here, we structurally characterize the viral replication compartment and report critical insights into the budding mechanism of the virus, and the structure of extracellular virions close to their native state by in situ cryo-electron tomography and subtomogram averaging. We directly visualize RNA filaments inside the double membrane vesicles, compartments associated with viral replication. The RNA filaments show a diameter consistent with double-stranded RNA and frequent branching likely representing RNA secondary structures. We report that assembled S trimers in lumenal cisternae do not alone induce membrane bending but laterally reorganize on the envelope during virion assembly. The viral ribonucleoprotein complexes (vRNPs) are accumulated at the curved membrane characteristic for budding sites suggesting that vRNP recruitment is enhanced by membrane curvature. Subtomogram averaging shows that vRNPs are distinct cylindrical assemblies. We propose that the genome is packaged around multiple separate vRNP complexes, thereby allowing incorporation of the unusually large coronavirus genome into the virion while maintaining high steric flexibility between the vRNPs.


Asunto(s)
Betacoronavirus/química , Betacoronavirus/fisiología , Replicación Viral , Células A549 , Animales , COVID-19 , Línea Celular , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Microscopía por Crioelectrón , Vesículas Citoplasmáticas/virología , Tomografía con Microscopio Electrónico , Retículo Endoplásmico/virología , Humanos , Pandemias , Neumonía Viral/virología , ARN Viral/química , ARN Viral/metabolismo , SARS-CoV-2 , Células Vero , Virión/química , Virión/metabolismo , Ensamble de Virus
17.
Cell Host Microbe ; 28(6): 853-866.e5, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33245857

RESUMEN

Pathogenesis induced by SARS-CoV-2 is thought to result from both an inflammation-dominated cytokine response and virus-induced cell perturbation causing cell death. Here, we employ an integrative imaging analysis to determine morphological organelle alterations induced in SARS-CoV-2-infected human lung epithelial cells. We report 3D electron microscopy reconstructions of whole cells and subcellular compartments, revealing extensive fragmentation of the Golgi apparatus, alteration of the mitochondrial network and recruitment of peroxisomes to viral replication organelles formed by clusters of double-membrane vesicles (DMVs). These are tethered to the endoplasmic reticulum, providing insights into DMV biogenesis and spatial coordination of SARS-CoV-2 replication. Live cell imaging combined with an infection sensor reveals profound remodeling of cytoskeleton elements. Pharmacological inhibition of their dynamics suppresses SARS-CoV-2 replication. We thus report insights into virus-induced cytopathic effects and provide alongside a comprehensive publicly available repository of 3D datasets of SARS-CoV-2-infected cells for download and smooth online visualization.


Asunto(s)
COVID-19/genética , Retículo Endoplásmico/ultraestructura , SARS-CoV-2/ultraestructura , Compartimentos de Replicación Viral/ultraestructura , COVID-19/diagnóstico por imagen , COVID-19/patología , COVID-19/virología , Muerte Celular/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/virología , Humanos , Microscopía Electrónica , Pandemias , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Compartimentos de Replicación Viral/metabolismo , Replicación Viral/genética
18.
Nature ; 588(7838): 498-502, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32805734

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virions are surrounded by a lipid bilayer from which spike (S) protein trimers protrude1. Heavily glycosylated S trimers bind to the angiotensin-converting enzyme 2 receptor and mediate entry of virions into target cells2-6. S exhibits extensive conformational flexibility: it modulates exposure of its receptor-binding site and subsequently undergoes complete structural rearrangement to drive fusion of viral and cellular membranes2,7,8. The structures and conformations of soluble, overexpressed, purified S proteins have been studied in detail using cryo-electron microscopy2,7,9-12, but the structure and distribution of S on the virion surface remain unknown. Here we applied cryo-electron microscopy and tomography to image intact SARS-CoV-2 virions and determine the high-resolution structure, conformational flexibility and distribution of S trimers in situ on the virion surface. These results reveal the conformations of S on the virion, and provide a basis from which to understand interactions between S and neutralizing antibodies during infection or vaccination.


Asunto(s)
Microscopía por Crioelectrón , SARS-CoV-2/metabolismo , SARS-CoV-2/ultraestructura , Glicoproteína de la Espiga del Coronavirus/análisis , Glicoproteína de la Espiga del Coronavirus/ultraestructura , Virión/química , Virión/ultraestructura , Anticuerpos Neutralizantes/inmunología , COVID-19/inmunología , Vacunas contra la COVID-19/inmunología , Línea Celular Tumoral , Humanos , Modelos Moleculares , Docilidad , Conformación Proteica , Multimerización de Proteína , SARS-CoV-2/química , SARS-CoV-2/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/aislamiento & purificación , Virión/aislamiento & purificación , Virión/metabolismo
19.
Cell Rep ; 32(1): 107859, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32640225

RESUMEN

Dengue virus (DENV) and Zika virus (ZIKV), members of the Flavivirus genus, rearrange endoplasmic reticulum membranes to induce invaginations known as vesicle packets (VPs), which are the assumed sites for viral RNA replication. Mechanistic information on VP biogenesis has so far been difficult to attain due to the necessity of studying their formation under conditions of viral replication, where perturbations reducing replication will inevitably impact VP formation. Here, we report a replication-independent expression system, designated pIRO (plasmid-induced replication organelle formation) that induces bona fide DENV and ZIKV VPs that are morphologically indistinguishable from those in infected cells. Using this system, we demonstrate that sequences in the 3' terminal RNA region of the DENV, but not the ZIKV genome, contribute to VP formation in a non-replicative manner. These results validate the pIRO system that opens avenues for mechanistically dissecting virus replication from membrane reorganization.


Asunto(s)
Virus del Dengue/genética , Virus del Dengue/fisiología , Genoma Viral , Orgánulos/metabolismo , Replicación Viral/genética , Regiones no Traducidas 3'/genética , Regiones no Traducidas 5'/genética , Línea Celular , ADN Polimerasa Dirigida por ADN/metabolismo , Dengue/virología , Virus del Dengue/enzimología , Virus del Dengue/ultraestructura , Humanos , Membranas , Conformación de Ácido Nucleico , Orgánulos/ultraestructura , Plásmidos/genética , Poliproteínas/metabolismo , ARN Viral/genética , Virus Zika/genética
20.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-152587

RESUMEN

Emergence of the novel pathogenic coronavirus SARS-CoV-2 and its rapid pandemic spread presents numerous questions and challenges that demand immediate attention. Among these is the urgent need for a better understanding of humoral immune response against the virus as a basis for developing public health strategies to control viral spread. For this, sensitive, specific and quantitative serological assays are required. Here we describe the development of a semi-quantitative high-content microscopy-based assay for detection of three major classes (IgG, IgA and IgM) of SARS-CoV-2 specific antibodies in human samples. The possibility to detect antibodies against the entire viral proteome together with a robust semi-automated image analysis workflow resulted in specific, sensitive and unbiased assay which complements the portfolio of SARS-CoV-2 serological assays. The procedure described here has been used for clinical studies and provides a general framework for the application of quantitative high-throughput microscopy to rapidly develop serological assays for emerging virus infections.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...