Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 218
Filtrar
2.
ACS Appl Mater Interfaces ; 16(23): 29621-29633, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38806169

RESUMEN

The ongoing challenge of viral transmission, exemplified by the Covid pandemic and recurrent viral outbreaks, necessitates the exploration of sustainable antiviral solutions. This study investigates the underexplored antiviral potential of wooden surfaces. We evaluated the antiviral efficacy of various wood types, including coniferous and deciduous trees, against enveloped coronaviruses and nonenveloped enteroviruses like coxsackie virus A9. Our findings revealed excellent antiviral activity manifesting already within 10 to 15 min in Scots pine and Norway spruce, particularly against enveloped viruses. In contrast, other hardwoods displayed varied efficacy, with oak showing effectiveness against the enterovirus. This antiviral activity was consistently observed across a spectrum of humidity levels (20 to 90 RH%), while the antiviral efficacy manifested itself more rapidly at 37 °C vs 21 °C. Key to our findings is the chemical composition of these woods. Resin acids and terpenes were prevalent in pine and spruce, correlating with their antiviral performance, while oak's high phenolic content mirrored its efficacy against enterovirus. The pine surface absorbed a higher fraction of the coronavirus in contrast to oak, whereas enteroviruses were not absorbed on those surfaces. Thermal treatment of wood or mixing wood with plastic, such as in wood-plastic composites, strongly compromised the antiviral functionality of wood materials. This study highlights the role of bioactive chemicals in the antiviral action of wood and opens new avenues for employing wood surfaces as a natural and sustainable barrier against viral transmissions.


Asunto(s)
Antivirales , Enterovirus , Madera , Madera/química , Antivirales/química , Antivirales/farmacología , Enterovirus/efectos de los fármacos , Coronavirus/efectos de los fármacos , Inactivación de Virus/efectos de los fármacos , Propiedades de Superficie , Quercus/química , Humanos , Pinus/química , Picea/química , Árboles/virología
3.
J Agric Food Chem ; 72(18): 10640-10654, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38661066

RESUMEN

Coronaviruses have consistently posed a major global concern in the field of livestock industry and public health. However, there is currently a lack of efficient drugs with broad-spectrum antiviral activity to address the challenges presented by emerging mutated strains or drug resistance. Additionally, the method for identifying multitarget drugs is also insufficient. Aminopeptidase N (APN) and 3C-like proteinase (3CLpro) represent promising targets for host-directed and virus-directed strategies, respectively, in the development of effective drugs against various coronaviruses. In this study, maduramycin ammonium demonstrated a broad-spectrum antiviral effect by targeting both of the proteins. The binding domains 4 Å from the ligand of both target proteins shared a structural similarity, suggesting that screening and designing drugs based on these domains might exhibit broad-spectrum and highly effective antiviral activity. Furthermore, it was identified that the polyether ionophores' ability to carry zinc ion might be one of the reasons why they were able to target APN and exhibit antiviral effect. The findings of this experiment provide novel perspectives for future drug screening and design, while also offering valuable references for the utilization of polyether ionophores in the management of livestock health.


Asunto(s)
Antivirales , Antígenos CD13 , Ionóforos , Ganado , Animales , Antivirales/farmacología , Antivirales/química , Ionóforos/farmacología , Ionóforos/química , Antígenos CD13/metabolismo , Antígenos CD13/química , Proteasas 3C de Coronavirus/química , Proteasas 3C de Coronavirus/metabolismo , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Drogas Veterinarias/farmacología , Drogas Veterinarias/química , Coronavirus/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/química , Policétidos Poliéteres
4.
J Virol ; 98(4): e0017124, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38488361

RESUMEN

The global impact of emerging viral infections emphasizes the urgent need for effective broad-spectrum antivirals. The cellular organelle, lipid droplet (LD), is utilized by many types of viruses for replication, but its reduction does not affect cell survival. Therefore, LD is a potential target for developing broad-spectrum antivirals. In this study, we found that 2-bromopalmitate (2 BP), a previously defined palmitoylation inhibitor, depletes LD across all studied cell lines and exerts remarkable antiviral effects on different coronaviruses. We comprehensively utilized 2 BP, alongside other palmitoylation inhibitors such as cerulenin and 2-fluoro palmitic acid (2-FPA), as well as the enhancer palmostatin B and evaluated their impact on LD and the replication of human coronaviruses (hCoV-229E, hCoV-Oc43) and murine hepatitis virus (MHV-A59) at non-cytotoxic concentrations. While cerulenin and 2-FPA exhibited moderate inhibition of viral replication, 2 BP exhibited a much stronger suppressive effect on MHV-A59 replication, although they share similar inhibitory effects on palmitoylation. As expected, palmostatin B significantly enhanced viral replication, it failed to rescue the inhibitory effects of 2 BP, whereas it effectively counteracted the effects of cerulenin and 2-FPA. This suggests that the mechanism that 2 BP used to inhibit viral replication is beyond palmitoylation inhibition. Further investigations unveil that 2 BP uniquely depletes LDs, a phenomenon not exhibited by 2-FPA and cerulenin. Importantly, the depletion of LDs was closely associated with the inhibition of viral replication because the addition of oleic acid to 2 BP significantly rescued LD depletion and its inhibitory effects on MHV-A59. Our findings indicate that the inhibitory effects of 2 BP on viral replication primarily stem from LD disruption rather than palmitoylation inhibition. Intriguingly, fatty acid (FA) assays demonstrated that 2 BP reduces the FA level in mitochondria while concurrently increasing FA levels in the cytoplasm. These results highlight the crucial role of LDs in viral replication and uncover a novel biological activity of 2 BP. These insights contribute to the development of broad-spectrum antiviral strategies. IMPORTANCE: In our study, we conducted a comparative investigation into the antiviral effects of palmitoylation inhibitors including 2-bromopalmitate (2-BP), 2-fluoro palmitic acid (2-FPA), and cerulenin. Surprisingly, we discovered that 2-BP has superior inhibitory effects on viral replication compared to 2-FPA and cerulenin. However, their inhibitory effects on palmitoylation were the same. Intrigued by this finding, we delved deeper into the underlying mechanism of 2-BP's potent antiviral activity, and we unveiled a novel biological activity of 2-BP: depletion of lipid droplets (LDs). Importantly, we also highlighted the crucial role of LDs in viral replication. Our insights shed new light on the antiviral mechanism of LD depletion paving the way for the development of broad-spectrum antiviral strategies by targeting LDs.


Asunto(s)
Antivirales , Coronavirus , Virus de la Hepatitis Murina , Palmitatos , Animales , Humanos , Ratones , Antivirales/farmacología , Antivirales/metabolismo , Cerulenina/metabolismo , Cerulenina/farmacología , Coronavirus/efectos de los fármacos , Coronavirus/fisiología , Gotas Lipídicas/efectos de los fármacos , Palmitatos/farmacología , Ácido Palmítico/farmacología , Ácido Palmítico/metabolismo , Propiolactona/análogos & derivados , Replicación Viral/efectos de los fármacos , Virus de la Hepatitis Murina/efectos de los fármacos , Virus de la Hepatitis Murina/fisiología
5.
J Mol Biol ; 436(6): 168474, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38311236

RESUMEN

The main protease (Mpro) of coronaviruses participates in viral replication, serving as a hot target for drug design. GC376 is able to effectively inhibit the activity of Mpro, which is due to nucleophilic addition of GC376 by binding covalently with Cys145 in Mpro active site. Here, we used fluorescence resonance energy transfer (FRET) assay to analyze the IC50 values of GC376 against Mpros from six different coronaviruses (SARS-CoV-2, HCoV-229E, HCoV-HUK1, MERS-CoV, SARS-CoV, HCoV-NL63) and five Mpro mutants (G15S, M49I, K90R, P132H, S46F) from SARS-CoV-2 variants. The results showed that GC376 displays effective inhibition to various coronaviral Mpros and SARS-CoV-2 Mpro mutants. In addition, the crystal structures of SARS-CoV-2 Mpro (wide type)-GC376, SARS-CoV Mpro-GC376, MERS-CoV Mpro-GC376, and SARS-CoV-2 Mpro mutants (G15S, M49I, S46F, K90R, and P132H)-GC376 complexes were solved. We found that GC376 is able to fit into the active site of Mpros from different coronaviruses and different SARS-CoV-2 variants properly. Detailed structural analysis revealed key molecular determinants necessary for inhibition and illustrated the binding patterns of GC376 to these different Mpros. In conclusion, we not only proved the inhibitory activity of GC376 against different Mpros including SARS-CoV-2 Mpro mutants, but also revealed the molecular mechanism of inhibition by GC376, which will provide scientific guidance for the development of broad-spectrum drugs against SARS-CoV-2 as well as other coronaviruses.


Asunto(s)
Antivirales , Proteasas 3C de Coronavirus , Coronavirus , Lactamas , Leucina , Ácidos Sulfónicos , Humanos , Antivirales/química , Antivirales/farmacología , Coronavirus/efectos de los fármacos , Coronavirus/enzimología , Lactamas/farmacología , Leucina/análogos & derivados , SARS-CoV-2/enzimología , Ácidos Sulfónicos/farmacología , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química
6.
J Virol ; 97(10): e0039623, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37706687

RESUMEN

IMPORTANCE: This study highlights the crucial role RNA processing plays in regulating viral gene expression and replication. By targeting SR kinases, we identified harmine as a potent inhibitor of HIV-1 as well as coronavirus (HCoV-229E and multiple SARS-CoV-2 variants) replication. Harmine inhibits HIV-1 protein expression and reduces accumulation of HIV-1 RNAs in both cell lines and primary CD4+ T cells. Harmine also suppresses coronavirus replication post-viral entry by preferentially reducing coronavirus sub-genomic RNA accumulation. By focusing on host factors rather than viral targets, our study offers a novel approach to combating viral infections that is effective against a range of unrelated viruses. Moreover, at doses required to inhibit virus replication, harmine had limited toxicity and minimal effect on the host transcriptome. These findings support the viability of targeting host cellular processes as a means of developing broad-spectrum anti-virals.


Asunto(s)
Antivirales , Coronavirus , VIH-1 , Harmina , Humanos , Antivirales/farmacología , Antivirales/uso terapéutico , Coronavirus/efectos de los fármacos , Coronavirus/fisiología , Infecciones por Coronavirus/tratamiento farmacológico , Harmina/farmacología , Harmina/uso terapéutico , VIH-1/efectos de los fármacos , VIH-1/fisiología , Replicación Viral/efectos de los fármacos
7.
Phytochemistry ; 212: 113713, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37169138

RESUMEN

The potential antiviral effects of indole-3-carbinol (I3C), a phytochemical found in Cruciferous vegetables, were investigated. Fibroblasts and epithelial cells were co-cultured on Alvetex® scaffolds, to obtain ad hoc 3D in vitro platforms able to mimic the trachea and intestinal mucosae, which represent the primary structures involved in the coronavirus pathogenesis. The two barriers generated in vitro were treated with various concentrations of I3C for different incubation periods. A protective effect of I3C on both intestinal and trachea models was demonstrated. A significant reduction in the transcription of the two main genes belonging to the Homologous to E6AP C-terminus (HECT)-E3 ligase family members, namely NEDD4 E3 Ubiquitin Protein Ligase (NEDD4) and WW Domain Containing E3 Ubiquitin Protein Ligase 1 (WWP1), which promote virus matrix protein ubiquitination and inhibit viral egression, were detected. These findings indicate I3C potential effect in preventing coronavirus cell egression processes that inhibit viral production. Although further studies are needed to clarify the molecular mechanisms whereby HECT family members control virus life cycle, this work paves the way to the possible therapeutic use of new natural compounds that may reduce the clinical severity of future pandemics.


Asunto(s)
Antivirales , Brassicaceae , Coronavirus , Intestinos , Modelos Biológicos , Fitoquímicos , Tráquea , Verduras , Antivirales/farmacología , Brassicaceae/química , Coronavirus/efectos de los fármacos , Coronavirus/metabolismo , Técnicas In Vitro , Intestinos/efectos de los fármacos , Intestinos/metabolismo , Intestinos/virología , Fitoquímicos/farmacología , Tráquea/efectos de los fármacos , Tráquea/metabolismo , Tráquea/virología , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Verduras/química , Proteínas de la Matriz Viral/metabolismo , Reproducibilidad de los Resultados , Porcinos , Animales , Humanos , Técnicas de Cultivo Tridimensional de Células
8.
Braz. J. Pharm. Sci. (Online) ; 59: e21067, 2023. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1429947

RESUMEN

Abstract We critically analyzed clinical trials performed with chloroquine (CQ) and hydroxychloroquine (HCQ) with or without macrolides during the first wave of COVID-19 and discussed the design and limitations of peer-reviewed studies from January to July 2020. Seventeen studies were eligible for the discussion. CQ and HCQ did not demonstrate clinical advantages that justified their inclusion in therapeutic regimens of free prescription for treatment or prophylactic purposes, as suggested by health authorities, including in Brazil, during the first wave. Around August 2020, robust data had already indicated that pharmacological effects of CQ, HCQ and macrolides as anti-SARS-CoV-2 molecules were limited to in vitro conditions and largely based on retrospective trials with low quality and weak internal validity, which made evidence superficial for decision-making. Up to that point, most randomized and nonrandomized clinical trials did not reveal beneficial effects of CQ or HCQ with or without macrolides to reduce lethality, rate of intubation, days of hospitalization, respiratory support/mechanical ventilation requirements, duration, type and number of symptoms, and death and were unsuccessful in increasing virus elimination and/or days alive in hospitalized or ambulatory patients with COVID-19. In addition, many studies have demonstrated that side effects are more common in CQ-or HCQ-treated patients.


Asunto(s)
Macrólidos/análisis , Pandemias/clasificación , COVID-19/patología , Antimaláricos/análisis , Comorbilidad , Ensayos Clínicos como Asunto/instrumentación , Coronavirus/efectos de los fármacos , Aminoquinolinas/agonistas , Hospitalización
9.
Viruses ; 14(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35215770

RESUMEN

Recurrent outbreaks of novel zoonotic coronavirus (CoV) diseases in recent years have highlighted the importance of developing therapeutics with broad-spectrum activity against CoVs. Because all CoVs use -1 programmed ribosomal frameshifting (-1 PRF) to control expression of key viral proteins, the frameshift signal in viral mRNA that stimulates -1 PRF provides a promising potential target for such therapeutics. To test the viability of this strategy, we explored whether small-molecule inhibitors of -1 PRF in SARS-CoV-2 also inhibited -1 PRF in a range of bat CoVs-the most likely source of future zoonoses. Six inhibitors identified in new and previous screens against SARS-CoV-2 were evaluated against the frameshift signals from a panel of representative bat CoVs as well as MERS-CoV. Some drugs had strong activity against subsets of these CoV-derived frameshift signals, while having limited to no effect on -1 PRF caused by frameshift signals from other viruses used as negative controls. Notably, the serine protease inhibitor nafamostat suppressed -1 PRF significantly for multiple CoV-derived frameshift signals. These results suggest it is possible to find small-molecule ligands that inhibit -1 PRF specifically in a broad spectrum of CoVs, establishing frameshift signals as a viable target for developing pan-coronaviral therapeutics.


Asunto(s)
Antivirales/farmacología , Coronavirus/efectos de los fármacos , Coronavirus/genética , Mutación del Sistema de Lectura , Sistema de Lectura Ribosómico/efectos de los fármacos , Proteínas Virales/antagonistas & inhibidores , Animales , Antivirales/uso terapéutico , Quirópteros/virología , Coronavirus/clasificación , Infecciones por Coronavirus/tratamiento farmacológico , Conformación de Ácido Nucleico , ARN Mensajero/genética , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/genética , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos
10.
Molecules ; 27(3)2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35163977

RESUMEN

COVID-19 has spread around the world and caused serious public health and social problems. Although several vaccines have been authorized for emergency use, new effective antiviral drugs are still needed. Some repurposed drugs including Chloroquine, Hydroxychloroquine and Remdesivir were immediately used to treat COVID-19 after the pandemic. However, the therapeutic effects of these drugs have not been fully demonstrated in clinical studies. In this paper, we found an antimalarial drug, Naphthoquine, showed good broad-spectrum anti-coronavirus activity. Naphthoquineinhibited HCoV-229E, HCoV-OC43 and SARS-CoV-2 replication in vitro, with IC50 = 2.05 ± 1.44 µM, 5.83 ± 0.74 µM, and 2.01 ± 0.38 µM, respectively. Time-of-addition assay was also performed to explore at which stage Naphthoquine functions during SARS-CoV-2 replication. The results suggested that Naphthoquine may influence virus entry and post-entry replication. Considering the safety of Naphthoquine was even better than that of Chloroquine, we think Naphthoquine has the potential to be used as a broad-spectrum drug for coronavirus infection.


Asunto(s)
1-Naftilamina/análogos & derivados , Aminoquinolinas/farmacología , Antivirales/farmacología , Coronavirus/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , 1-Naftilamina/farmacología , Animales , Línea Celular , Chlorocebus aethiops , Coronavirus Humano 229E/efectos de los fármacos , Coronavirus Humano NL63/efectos de los fármacos , Coronavirus Humano OC43/efectos de los fármacos , Humanos , Técnicas In Vitro , Células Vero , Replicación Viral/efectos de los fármacos
11.
Viruses ; 14(1)2022 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-35062314

RESUMEN

Photodynamic inactivation (PDI) employs a photosensitizer, light, and oxygen to create a local burst of reactive oxygen species (ROS) that can inactivate microorganisms. The botanical extract PhytoQuinTM is a powerful photosensitizer with antimicrobial properties. We previously demonstrated that photoactivated PhytoQuin also has antiviral properties against herpes simplex viruses and adenoviruses in a dose-dependent manner across a broad range of sub-cytotoxic concentrations. Here, we report that human coronaviruses (HCoVs) are also susceptible to photodynamic inactivation. Photoactivated-PhytoQuin inhibited the replication of the alphacoronavirus HCoV-229E and the betacoronavirus HCoV-OC43 in cultured cells across a range of sub-cytotoxic doses. This antiviral effect was light-dependent, as we observed minimal antiviral effect of PhytoQuin in the absence of photoactivation. Using RNase protection assays, we observed that PDI disrupted HCoV particle integrity allowing for the digestion of viral RNA by exogenous ribonucleases. Using lentiviruses pseudotyped with the SARS-CoV-2 Spike (S) protein, we once again observed a strong, light-dependent antiviral effect of PhytoQuin, which prevented S-mediated entry into human cells. We also observed that PhytoQuin PDI altered S protein electrophoretic mobility. The PhytoQuin constituent emodin displayed equivalent light-dependent antiviral activity to PhytoQuin in matched-dose experiments, indicating that it plays a central role in PhytoQuin PDI against CoVs. Together, these findings demonstrate that HCoV lipid envelopes and proteins are damaged by PhytoQuin PDI and expands the list of susceptible viruses.


Asunto(s)
Antivirales/farmacología , Coronavirus/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Inactivación de Virus/efectos de los fármacos , Animales , Antivirales/efectos de la radiación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cricetinae , Emodina/farmacología , Emodina/efectos de la radiación , Humanos , Luz , Fármacos Fotosensibilizantes/efectos de la radiación , Extractos Vegetales/farmacología , Extractos Vegetales/efectos de la radiación , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/efectos de los fármacos , Virión/efectos de los fármacos
12.
Med Sci Monit ; 28: e934102, 2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35075100

RESUMEN

BACKGROUND Heat-clearing and detoxifying herbs (HDHs) play an important role in the prevention and treatment of coronavirus infection. However, their mechanism of action needs further study. This study aimed to explore the anti-coronavirus basis and mechanism of HDHs. MATERIAL AND METHODS Database mining was performed on 7 HDHs. Core ingredients and targets were screened according to ADME rules combined with Neighborhood, Co-occurrence, Co-expression, and other algorithms. GO enrichment and KEGG pathway analyses were performed using the R language. Finally, high-throughput molecular docking was used for verification. RESULTS HDHs mainly acts on NOS3, EGFR, IL-6, MAPK8, PTGS2, MAPK14, NFKB1, and CASP3 through quercetin, luteolin, wogonin, indirubin alkaloids, ß-sitosterol, and isolariciresinol. These targets are mainly involved in the regulation of biological processes such as inflammation, activation of MAPK activity, and positive regulation of NF-kappaB transcription factor activity. Pathway analysis further revealed that the pathways regulated by these targets mainly include: signaling pathways related to viral and bacterial infections such as tuberculosis, influenza A, Ras signaling pathways; inflammation-related pathways such as the TLR, TNF, MAPK, and HIF-1 signaling pathways; and immune-related pathways such as NOD receptor signaling pathways. These pathways play a synergistic role in inhibiting lung inflammation and regulating immunity and antiviral activity. CONCLUSIONS HDHs play a role in the treatment of coronavirus infection by regulating the body's immunity, fighting inflammation, and antiviral activities, suggesting a molecular basis and new strategies for the treatment of COVID-19 and a foundation for the screening of new antiviral drugs.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Coronavirus/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , SARS-CoV-2/efectos de los fármacos , Alcaloides/química , Alcaloides/farmacología , Caspasa 3/efectos de los fármacos , Caspasa 3/genética , Coronavirus/metabolismo , Infecciones por Coronavirus/tratamiento farmacológico , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/genética , Bases de Datos Farmacéuticas , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/uso terapéutico , Flavanonas/química , Flavanonas/farmacología , Humanos , Indoles/química , Indoles/farmacología , Interleucina-6/genética , Lignina/química , Lignina/farmacología , Luteolina/química , Luteolina/farmacología , Proteína Quinasa 14 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 8 Activada por Mitógenos/genética , Simulación del Acoplamiento Molecular , Subunidad p50 de NF-kappa B/efectos de los fármacos , Subunidad p50 de NF-kappa B/genética , Naftoles/química , Naftoles/farmacología , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/genética , Mapas de Interacción de Proteínas , Quercetina/química , Quercetina/farmacología , SARS-CoV-2/metabolismo , Transducción de Señal , Sitoesteroles/química , Sitoesteroles/farmacología , Transcriptoma/efectos de los fármacos , Transcriptoma/genética
13.
Nucleic Acids Res ; 50(2): 635-650, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35018474

RESUMEN

Coronaviral methyltransferases (MTases), nsp10/16 and nsp14, catalyze the last two steps of viral RNA-cap creation that takes place in cytoplasm. This cap is essential for the stability of viral RNA and, most importantly, for the evasion of innate immune system. Non-capped RNA is recognized by innate immunity which leads to its degradation and the activation of antiviral immunity. As a result, both coronaviral MTases are in the center of scientific scrutiny. Recently, X-ray and cryo-EM structures of both enzymes were solved even in complex with other parts of the viral replication complex. High-throughput screening as well as structure-guided inhibitor design have led to the discovery of their potent inhibitors. Here, we critically summarize the tremendous advancement of the coronaviral MTase field since the beginning of COVID pandemic.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Coronavirus/efectos de los fármacos , Coronavirus/enzimología , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/química , Metiltransferasas/metabolismo , Secuencia de Aminoácidos , Aminoácidos/química , Sitios de Unión , Coronavirus/genética , Descubrimiento de Drogas , Humanos , Metilación , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Unión Proteica , ARN Viral/química , ARN Viral/genética , ARN Viral/metabolismo , Relación Estructura-Actividad
14.
Sci Rep ; 11(1): 23315, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857794

RESUMEN

The COVID-19 pandemic has highlighted the urgent need for the identification of new antiviral drug therapies for a variety of diseases. COVID-19 is caused by infection with the human coronavirus SARS-CoV-2, while other related human coronaviruses cause diseases ranging from severe respiratory infections to the common cold. We developed a computational approach to identify new antiviral drug targets and repurpose clinically-relevant drug compounds for the treatment of a range of human coronavirus diseases. Our approach is based on graph convolutional networks (GCN) and involves multiscale host-virus interactome analysis coupled to off-target drug predictions. Cell-based experimental assessment reveals several clinically-relevant drug repurposing candidates predicted by the in silico analyses to have antiviral activity against human coronavirus infection. In particular, we identify the MET inhibitor capmatinib as having potent and broad antiviral activity against several coronaviruses in a MET-independent manner, as well as novel roles for host cell proteins such as IRAK1/4 in supporting human coronavirus infection, which can inform further drug discovery studies.


Asunto(s)
Antivirales/farmacología , Coronavirus/efectos de los fármacos , Coronavirus/metabolismo , Desarrollo de Medicamentos/métodos , Reposicionamiento de Medicamentos/métodos , Benzamidas/farmacología , Línea Celular , Simulación por Computador , Coronavirus/química , Bases de Datos Farmacéuticas , Descubrimiento de Drogas/métodos , Interacciones Huésped-Patógeno , Humanos , Imidazoles/farmacología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , SARS-CoV-2/química , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Triazinas/farmacología , Tratamiento Farmacológico de COVID-19
15.
mBio ; 12(6): e0334721, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34933447

RESUMEN

The world was unprepared for coronavirus disease 2019 (COVID-19) and remains ill-equipped for future pandemics. While unprecedented strides have been made developing vaccines and treatments for COVID-19, there remains a need for highly effective and widely available regimens for ambulatory use for novel coronaviruses and other viral pathogens. We posit that a priority is to develop pan-family drug cocktails to enhance potency, limit toxicity, and avoid drug resistance. We urge cocktail development for all viruses with pandemic potential both in the short term (<1 to 2 years) and longer term with pairs of drugs in advanced clinical testing or repurposed agents approved for other indications. While significant efforts were launched against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in vitro and in the clinic, many studies employed solo drugs and had disappointing results. Here, we review drug combination studies against SARS-CoV-2 and other viruses and introduce a model-driven approach to assess drug pairs with the highest likelihood of clinical efficacy. Where component agents lack sufficient potency, we advocate for synergistic combinations to achieve therapeutic levels. We also discuss issues that stymied therapeutic progress against COVID-19, including testing of agents with low likelihood of efficacy late in clinical disease and lack of focus on developing virologic surrogate endpoints. There is a need to expedite efficient clinical trials testing drug combinations that could be taken at home by recently infected individuals and exposed contacts as early as possible during the next pandemic, whether caused by a coronavirus or another viral pathogen. The approach herein represents a proactive plan for global viral pandemic preparedness.


Asunto(s)
Antivirales/farmacología , Coronavirus/efectos de los fármacos , Combinación de Medicamentos , Animales , Coronavirus/clasificación , Coronavirus/patogenicidad , Infecciones por Coronavirus/tratamiento farmacológico , Humanos , Ratones , Pandemias/prevención & control , SARS-CoV-2/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
16.
Sci Rep ; 11(1): 24442, 2021 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-34952911

RESUMEN

Therapeutic interventions targeting viral infections remain a significant challenge for both the medical and scientific communities. While specific antiviral agents have shown success as therapeutics, viral resistance inevitably develops, making many of these approaches ineffective. This inescapable obstacle warrants alternative approaches, such as the targeting of host cellular factors. Respiratory syncytial virus (RSV), the major respiratory pathogen of infants and children worldwide, causes respiratory tract infection ranging from mild upper respiratory tract symptoms to severe life-threatening lower respiratory tract disease. Despite the fact that the molecular biology of the virus, which was originally discovered in 1956, is well described, there is no vaccine or effective antiviral treatment against RSV infection. Here, we demonstrate that targeting host factors, specifically, mTOR signaling, reduces RSV protein production and generation of infectious progeny virus. Further, we show that this approach can be generalizable as inhibition of mTOR kinases reduces coronavirus gene expression, mRNA transcription and protein production. Overall, defining virus replication-dependent host functions may be an effective means to combat viral infections, particularly in the absence of antiviral drugs.


Asunto(s)
Coronavirus/metabolismo , Virus Sincitial Respiratorio Humano/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Virales/metabolismo , Células A549 , Coronavirus/efectos de los fármacos , Coronavirus/genética , Regulación Viral de la Expresión Génica/efectos de los fármacos , Humanos , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/antagonistas & inhibidores , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Proteína Reguladora Asociada a mTOR/antagonistas & inhibidores , Proteína Reguladora Asociada a mTOR/genética , Proteína Reguladora Asociada a mTOR/metabolismo , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitial Respiratorio Humano/aislamiento & purificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Proteínas Virales/genética
17.
Science ; 374(6575): 1586-1593, 2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-34726479

RESUMEN

The worldwide outbreak of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic. Alongside vaccines, antiviral therapeutics are an important part of the healthcare response to countering the ongoing threat presented by COVID-19. Here, we report the discovery and characterization of PF-07321332, an orally bioavailable SARS-CoV-2 main protease inhibitor with in vitro pan-human coronavirus antiviral activity and excellent off-target selectivity and in vivo safety profiles. PF-07321332 has demonstrated oral activity in a mouse-adapted SARS-CoV-2 model and has achieved oral plasma concentrations exceeding the in vitro antiviral cell potency in a phase 1 clinical trial in healthy human participants.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Lactamas/farmacología , Lactamas/uso terapéutico , Leucina/farmacología , Leucina/uso terapéutico , Nitrilos/farmacología , Nitrilos/uso terapéutico , Prolina/farmacología , Prolina/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Inhibidores de Proteasa Viral/farmacología , Inhibidores de Proteasa Viral/uso terapéutico , Administración Oral , Animales , COVID-19/virología , Ensayos Clínicos Fase I como Asunto , Coronavirus/efectos de los fármacos , Modelos Animales de Enfermedad , Quimioterapia Combinada , Humanos , Lactamas/administración & dosificación , Lactamas/farmacocinética , Leucina/administración & dosificación , Leucina/farmacocinética , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Nitrilos/administración & dosificación , Nitrilos/farmacocinética , Prolina/administración & dosificación , Prolina/farmacocinética , Ensayos Clínicos Controlados Aleatorios como Asunto , Ritonavir/administración & dosificación , Ritonavir/uso terapéutico , SARS-CoV-2/fisiología , Inhibidores de Proteasa Viral/administración & dosificación , Inhibidores de Proteasa Viral/farmacocinética , Replicación Viral/efectos de los fármacos
18.
ACS Appl Mater Interfaces ; 13(41): 48469-48477, 2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34623127

RESUMEN

The COVID-19 pandemic highlighted the importance of developing surfaces and coatings with antiviral activity. Here, we present, for the first time, peptide-based assemblies that can kill viruses. The minimal inhibitory concentration (MIC) of the assemblies is in the range tens of micrograms per milliliter. This value is 2 orders of magnitude smaller than the MIC of metal nanoparticles. When applied on a surface, by drop casting, the peptide spherical assemblies adhere to the surface and form an antiviral coating against both RNA- and DNA-based viruses including coronavirus. Our results show that the coating reduced the number of T4 bacteriophages (DNA-based virus) by 3 log, compared with an untreated surface and 6 log, when compared with a stock solution. Importantly, we showed that this coating completely inactivated canine coronavirus (RNA-based virus). This peptide-based coating can be useful wherever sterile surfaces are needed to reduce the risk of viral transmission.


Asunto(s)
Antivirales/química , Péptidos/química , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Bacteriófagos/efectos de los fármacos , COVID-19/virología , Coronavirus/efectos de los fármacos , Coronavirus/aislamiento & purificación , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Dihidroxifenilalanina/química , Enfermedades de los Perros/tratamiento farmacológico , Enfermedades de los Perros/virología , Perros , Humanos , Nanopartículas del Metal/química , Péptidos/farmacología , Péptidos/uso terapéutico , SARS-CoV-2/aislamiento & purificación , Inactivación de Virus/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
19.
Best Pract Res Clin Anaesthesiol ; 35(3): 269-292, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34511219

RESUMEN

Coronaviruses belong to the family Coronaviridae order Nidovirales and are known causes of respiratory and intestinal disease in various mammalian and avian species. Species of coronaviruses known to infect humans are referred to as human coronaviruses (HCoVs). While traditionally, HCoVs have been a significant cause of the common cold, more recently, emergent viruses, including severe acute respiratory syndrome coronavirus (SARS-CoV-2) has caused a global pandemic. Here, we discuss coronavirus disease (COVID-19) biology, pathology, epidemiology, signs and symptoms, diagnosis, treatment, and recent clinical trials involving promising treatments.


Asunto(s)
Antivirales/administración & dosificación , COVID-19/epidemiología , COVID-19/terapia , SARS-CoV-2 , Adenosina Monofosfato/administración & dosificación , Adenosina Monofosfato/análogos & derivados , Corticoesteroides/administración & dosificación , Alanina/administración & dosificación , Alanina/análogos & derivados , Animales , COVID-19/diagnóstico , COVID-19/inmunología , Coronavirus/efectos de los fármacos , Coronavirus/inmunología , Tos/epidemiología , Tos/terapia , Diabetes Mellitus/epidemiología , Diabetes Mellitus/terapia , Fatiga/epidemiología , Fatiga/terapia , Fiebre , Cardiopatías/epidemiología , Cardiopatías/terapia , Humanos , Respiración con Presión Positiva/métodos , Pronóstico , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Enfermedad Pulmonar Obstructiva Crónica/terapia , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Resultado del Tratamiento
20.
Food Funct ; 12(20): 9607-9619, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34549212

RESUMEN

At the end of 2019, the COVID-19 virus spread worldwide, infecting millions of people. Infectious diseases induced by pathogenic microorganisms such as the influenza virus, hepatitis virus, and Mycobacterium tuberculosis are also a major threat to public health. The high mortality caused by infectious pathogenic microorganisms is due to their strong virulence, which leads to the excessive counterattack by the host immune system and severe inflammatory damage of the immune system. This paper reviews the efficacy, mechanism and related immune regulation of epigallocatechin-3-gallate (EGCG) as an anti-pathogenic microorganism drug. EGCG mainly shows both direct and indirect anti-infection effects. EGCG directly inhibits early infection by interfering with the adsorption on host cells, inhibiting virus replication and reducing bacterial biofilm formation and toxin release; EGCG indirectly inhibits infection by regulating immune inflammation and antioxidation. At the same time, we reviewed the bioavailability and safety of EGCG in vivo. At present, the bioavailability of EGCG can be improved to some extent using nanostructured drug delivery systems and molecular modification technology in combination with other drugs. This study provides a theoretical basis for the development of EGCG as an adjuvant drug for anti-pathogenic microorganisms.


Asunto(s)
Antiinfecciosos/farmacología , Catequina/análogos & derivados , Catequina/farmacología , Factores Inmunológicos/farmacología , Animales , Antioxidantes/farmacología , Coronavirus/efectos de los fármacos , Virus de Hepatitis/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Mycobacterium tuberculosis/efectos de los fármacos , Orthomyxoviridae/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA