Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 79(5): 710-727, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32853546

RESUMEN

The coronavirus disease 2019 (COVID-19) that is wreaking havoc on worldwide public health and economies has heightened awareness about the lack of effective antiviral treatments for human coronaviruses (CoVs). Many current antivirals, notably nucleoside analogs (NAs), exert their effect by incorporation into viral genomes and subsequent disruption of viral replication and fidelity. The development of anti-CoV drugs has long been hindered by the capacity of CoVs to proofread and remove mismatched nucleotides during genome replication and transcription. Here, we review the molecular basis of the CoV proofreading complex and evaluate its potential as a drug target. We also consider existing nucleoside analogs and novel genomic techniques as potential anti-CoV therapeutics that could be used individually or in combination to target the proofreading mechanism.


Asunto(s)
Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/epidemiología , Genoma Viral , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/epidemiología , ARN Viral/genética , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/química , Adenosina Monofosfato/uso terapéutico , Alanina/análogos & derivados , Alanina/química , Alanina/uso terapéutico , Amidas/química , Amidas/uso terapéutico , Antivirales/química , Betacoronavirus/genética , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/virología , Citidina/análogos & derivados , Humanos , Hidroxilaminas , Terapia Molecular Dirigida/métodos , Mutación , Neumonía Viral/virología , Pirazinas/química , Pirazinas/uso terapéutico , ARN Viral/antagonistas & inhibidores , ARN Viral/metabolismo , Ribonucleósidos/química , Ribonucleósidos/uso terapéutico , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Transcripción Genética , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
2.
Trends Biochem Sci ; 46(5): 351-365, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33309323

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently creating a global health emergency. This crisis is driving a worldwide effort to develop effective vaccines, prophylactics, and therapeutics. Nucleic acid (NA)-based treatments hold great potential to combat outbreaks of coronaviruses (CoVs) due to their rapid development, high target specificity, and the capacity to increase druggability. Here, we review key anti-CoV NA-based technologies, including antisense oligonucleotides (ASOs), siRNAs, RNA-targeting clustered regularly interspaced short palindromic repeats-CRISPR-associated protein (CRISPR-Cas), and mRNA vaccines, and discuss improved delivery methods and combination therapies with other antiviral drugs.


Asunto(s)
Vacunas contra la COVID-19 , Sistemas CRISPR-Cas , ARN Mensajero , ARN Viral , SARS-CoV-2 , COVID-19/genética , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/terapia , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/uso terapéutico , Humanos , ARN Mensajero/genética , ARN Mensajero/inmunología , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/inmunología , ARN Viral/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/metabolismo
3.
J Am Chem Soc ; 146(14): 9779-9789, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38561350

RESUMEN

Protein O-linked ß-N-acetylglucosamine modification (O-GlcNAcylation) plays a crucial role in regulating essential cellular processes. The disruption of the homeostasis of O-GlcNAcylation has been linked to various human diseases, including cancer, diabetes, and neurodegeneration. However, there are limited chemical tools for protein- and site-specific O-GlcNAc modification, rendering the precise study of the O-GlcNAcylation challenging. To address this, we have developed heterobifunctional small molecules, named O-GlcNAcylation TArgeting Chimeras (OGTACs), which enable protein-specific O-GlcNAcylation in living cells. OGTACs promote O-GlcNAcylation of proteins such as BRD4, CK2α, and EZH2 in cellulo by recruiting FKBP12F36V-fused O-GlcNAc transferase (OGT), with temporal, magnitude, and reversible control. Overall, the OGTACs represent a promising approach for inducing protein-specific O-GlcNAcylation, thus enabling functional dissection and offering new directions for O-GlcNAc-targeting therapeutic development.


Asunto(s)
Neoplasias , Proteínas Nucleares , Humanos , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Procesamiento Proteico-Postraduccional , N-Acetilglucosaminiltransferasas/metabolismo , Acetilglucosamina/metabolismo , Proteínas que Contienen Bromodominio , Proteínas de Ciclo Celular/metabolismo
5.
J Biol Chem ; 297(2): 100925, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34214498

RESUMEN

Apart from prevention using vaccinations, the management options for COVID-19 remain limited. In retrospective cohort studies, use of famotidine, a specific oral H2 receptor antagonist (antihistamine), has been associated with reduced risk of intubation and death in patients hospitalized with COVID-19. In a case series, nonhospitalized patients with COVID-19 experienced rapid symptom resolution after taking famotidine, but the molecular basis of these observations remains elusive. Here we show using biochemical, cellular, and functional assays that famotidine has no effect on viral replication or viral protease activity. However, famotidine can affect histamine-induced signaling processes in infected Caco2 cells. Specifically, famotidine treatment inhibits histamine-induced expression of Toll-like receptor 3 (TLR3) in SARS-CoV-2 infected cells and can reduce TLR3-dependent signaling processes that culminate in activation of IRF3 and the NF-κB pathway, subsequently controlling antiviral and inflammatory responses. SARS-CoV-2-infected cells treated with famotidine demonstrate reduced expression levels of the inflammatory mediators CCL-2 and IL6, drivers of the cytokine release syndrome that precipitates poor outcome for patients with COVID-19. Given that pharmacokinetic studies indicate that famotidine can reach concentrations in blood that suffice to antagonize histamine H2 receptors expressed in mast cells, neutrophils, and eosinophils, these observations explain how famotidine may contribute to the reduced histamine-induced inflammation and cytokine release, thereby improving the outcome for patients with COVID-19.


Asunto(s)
Famotidina/farmacología , Antagonistas de los Receptores Histamínicos/farmacología , SARS-CoV-2/efectos de los fármacos , Receptor Toll-Like 3/metabolismo , Células A549 , Sitios de Unión , Células CACO-2 , Quimiocina CCL2/metabolismo , Proteasas 3C de Coronavirus/metabolismo , Células HeLa , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interleucina-6/metabolismo , Simulación del Acoplamiento Molecular , FN-kappa B/metabolismo , Unión Proteica , SARS-CoV-2/fisiología , Transducción de Señal , Receptor Toll-Like 3/química , Replicación Viral
6.
ACS Cent Sci ; 7(5): 792-802, 2021 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-34075346

RESUMEN

The outbreak of coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a global threat to human health. Using a multidisciplinary approach, we identified and validated the hepatitis C virus (HCV) protease inhibitor simeprevir as an especially promising repurposable drug for treating COVID-19. Simeprevir potently reduces SARS-CoV-2 viral load by multiple orders of magnitude and synergizes with remdesivir in vitro. Mechanistically, we showed that simeprevir not only inhibits the main protease (Mpro) and unexpectedly the RNA-dependent RNA polymerase (RdRp) but also modulates host immune responses. Our results thus reveal the possible anti-SARS-CoV-2 mechanism of simeprevir and highlight the translational potential of optimizing simeprevir as a therapeutic agent for managing COVID-19 and future outbreaks of CoV.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA