Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 573
Filtrar
1.
Biomed Pharmacother ; 174: 116561, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38593705

RESUMEN

Pectin and its derivatives have been shown to modulate immune signaling as well as gut microbiota in preclinical studies, which may constitute the mechanisms by which supplementation of specific pectic polysaccharides confers protection against viral respiratory infections. In a double-blind, placebo-controlled rhinovirus (RV16) challenge study, healthy volunteers were randomized to consume placebo (0.0 g/day) (N = 46), low-dose (0.3 g/day) (N = 49) or high-dose (1.5 g/day) (N = 51) of carrot derived rhamnogalacturonan-I (cRG-I) for eight weeks and they were subsequently challenged with RV-16. Here, the effect of 8-week cRG-I supplementation on the gut microbiota was studied. While the overall gut microbiota composition in the population was generally unaltered by this very low dose of fibre, the relative abundance of Bifidobacterium spp. (mainly B. adolescentis and B. longum) was significantly increased by both doses of cRG-1. Moreover, daily supplementation of cRG-I led to a dose-dependent reduction in inter- and intra-individual microbiota heterogeneity, suggesting a stabilizing effect on the gut microbiota. The severity of respiratory symptoms did not directly correlate with the cRG-I-induced microbial changes, but several dominant groups of the Ruminococcaceae family and microbiota richness were positively associated with a reduced and hence desired post-infection response. Thus, the present results on the modulation of the gut microbiota composition support the previously demonstrated immunomodulatory and protective effect of cRG-I during a common cold infection.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal , Voluntarios Sanos , Pectinas , Humanos , Pectinas/administración & dosificación , Pectinas/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Adulto , Método Doble Ciego , Femenino , Adulto Joven , Rhinovirus/efectos de los fármacos , Persona de Mediana Edad , Heces/microbiología , Bifidobacterium/efectos de los fármacos
2.
Antiviral Res ; 226: 105897, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38685531

RESUMEN

Human respiratory viruses have an enormous impact on national health systems, societies, and economy due to the rapid airborne transmission and epidemic spread of such pathogens, while effective specific antiviral drugs to counteract infections are still lacking. Here, we identified two Keggin-type polyoxometalates (POMs), [TiW11CoO40]8- (TiW11Co) and [Ti2PW10O40]7- (Ti2PW10), endowed with broad-spectrum activity against enveloped and non-enveloped human respiratory viruses, i.e., coronavirus (HCoV-OC43), rhinovirus (HRV-A1), respiratory syncytial virus (RSV-A2), and adenovirus (AdV-5). Ti2PW10 showed highly favorable selectivity indexes against all tested viruses (SIs >700), and its antiviral potential was further investigated against human coronaviruses and rhinoviruses. This POM was found to inhibit replication of multiple HCoV and HRV strains, in different cell systems. Ti2PW10 did not affect virus binding or intracellular viral replication, but selectively inhibited the viral entry. Serial passaging of virus in presence of the POM revealed a high barrier to development of Ti2PW10-resistant variants of HRV-A1 or HCoV-OC43. Moreover, Ti2PW10 was able to inhibit HRV-A1 production in a 3D model of the human nasal epithelium and, importantly, the antiviral treatment did not determine cytotoxicity or tissue damage. A mucoadhesive thermosensitive in situ hydrogel formulation for nasal delivery was also developed for Ti2PW10. Overall, good biocompatibility on cell lines and human nasal epithelia, broad-spectrum activity, and absence of antiviral resistance development reveal the potential of Ti2PW10 as an antiviral candidate for the development of a treatment of acute respiratory viral diseases, warranting further studies to identify the specific target/s of the polyanion and assess its clinical potential.


Asunto(s)
Antivirales , Compuestos de Tungsteno , Internalización del Virus , Replicación Viral , Humanos , Internalización del Virus/efectos de los fármacos , Antivirales/farmacología , Replicación Viral/efectos de los fármacos , Compuestos de Tungsteno/farmacología , Rhinovirus/efectos de los fármacos , Rhinovirus/fisiología , Línea Celular , Infecciones del Sistema Respiratorio/virología , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Coronavirus Humano OC43/efectos de los fármacos , Coronavirus Humano OC43/fisiología , Animales
3.
J Virol ; 96(2): e0106021, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34705560

RESUMEN

Rhinoviruses (RVs) cause recurrent infections of the nasal and pulmonary tracts, life-threatening conditions in chronic respiratory illness patients, predisposition of children to asthmatic exacerbation, and large economic cost. RVs are difficult to treat. They rapidly evolve resistance and are genetically diverse. Here, we provide insight into RV drug resistance mechanisms against chemical compounds neutralizing low pH in endolysosomes. Serial passaging of RV-A16 in the presence of the vacuolar proton ATPase inhibitor bafilomycin A1 (BafA1) or the endolysosomotropic agent ammonium chloride (NH4Cl) promoted the emergence of resistant virus populations. We found two reproducible point mutations in viral proteins 1 and 3 (VP1 and VP3), A2526G (serine 66 to asparagine [S66N]), and G2274U (cysteine 220 to phenylalanine [C220F]), respectively. Both mutations conferred cross-resistance to BafA1, NH4Cl, and the protonophore niclosamide, as identified by massive parallel sequencing and reverse genetics, but not the double mutation, which we could not rescue. Both VP1-S66 and VP3-C220 locate at the interprotomeric face, and their mutations increase the sensitivity of virions to low pH, elevated temperature, and soluble intercellular adhesion molecule 1 receptor. These results indicate that the ability of RV to uncoat at low endosomal pH confers virion resistance to extracellular stress. The data endorse endosomal acidification inhibitors as a viable strategy against RVs, especially if inhibitors are directly applied to the airways. IMPORTANCE Rhinoviruses (RVs) are the predominant agents causing the common cold. Anti-RV drugs and vaccines are not available, largely due to rapid evolutionary adaptation of RVs giving rise to resistant mutants and an immense diversity of antigens in more than 160 different RV types. In this study, we obtained insight into the cell biology of RVs by harnessing the ability of RVs to evolve resistance against host-targeting small chemical compounds neutralizing endosomal pH, an important cue for uncoating of normal RVs. We show that RVs grown in cells treated with inhibitors of endolysosomal acidification evolved capsid mutations yielding reduced virion stability against elevated temperature, low pH, and incubation with recombinant soluble receptor fragments. This fitness cost makes it unlikely that RV mutants adapted to neutral pH become prevalent in nature. The data support the concept of host-directed drug development against respiratory viruses in general, notably at low risk of gain-of-function mutations.


Asunto(s)
Cápside/química , Mutación/efectos de los fármacos , Rhinovirus/fisiología , Desencapsidación Viral/fisiología , Antivirales/farmacología , Cápside/efectos de los fármacos , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Endosomas/química , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Molécula 1 de Adhesión Intercelular/metabolismo , Conformación Proteica , Rhinovirus/química , Rhinovirus/efectos de los fármacos , Rhinovirus/genética , Virión/química , Virión/genética , Virión/metabolismo , Internalización del Virus/efectos de los fármacos , Desencapsidación Viral/efectos de los fármacos , Desencapsidación Viral/genética
4.
Viruses ; 13(12)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34960671

RESUMEN

Rhinoviruses (RV), like many other viruses, modulate programmed cell death to their own advantage. The viral protease, 3C has an integral role in the modulation, and we have shown that RVA-16 3C protease cleaves Receptor-interacting protein kinase-1 (RIPK1), a key host factor that modulates various cell death and cell survival pathways. In the current study, we have investigated whether this cleavage is conserved across selected RV strains. RIPK1 was cleaved in cells infected with strains representing diversity across phylogenetic groups (A and B) and receptor usage (major and minor groups). The cleavage was abrogated in the presence of the specific 3C protease inhibitor, Rupintrivir. Interestingly, there appears to be involvement of another protease (maybe 2A protease) in RIPK1 cleavage in strains belonging to genotype B. Our data show that 3C protease from diverse RV strains cleaves RIPK1, highlighting the importance of the cleavage to the RV lifecycle.


Asunto(s)
Proteasas Virales 3C/metabolismo , Infecciones por Picornaviridae/enzimología , Rhinovirus/enzimología , Proteasas Virales 3C/genética , Antivirales/química , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Isoxazoles/química , Isoxazoles/farmacología , Fenilalanina/análogos & derivados , Fenilalanina/química , Fenilalanina/farmacología , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/virología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Pirrolidinonas/química , Pirrolidinonas/farmacología , Rhinovirus/química , Rhinovirus/efectos de los fármacos , Rhinovirus/genética , Valina/análogos & derivados , Valina/química , Valina/farmacología
5.
Int J Mol Sci ; 22(20)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34681727

RESUMEN

The ongoing COVID-19 pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) became a globally leading public health concern over the past two years. Despite the development and administration of multiple vaccines, the mutation of newer strains and challenges to universal immunity has shifted the focus to the lack of efficacious drugs for therapeutic intervention for the disease. As with SARS-CoV, MERS-CoV, and other non-respiratory viruses, flavonoids present themselves as a promising therapeutic intervention given their success in silico, in vitro, in vivo, and more recently, in clinical studies. This review focuses on data from in vitro studies analyzing the effects of flavonoids on various key SARS-CoV-2 targets and presents an analysis of the structure-activity relationships for the same. From 27 primary papers, over 69 flavonoids were investigated for their activities against various SARS-CoV-2 targets, ranging from the promising 3C-like protease (3CLpro) to the less explored nucleocapsid (N) protein; the most promising were quercetin and myricetin derivatives, baicalein, baicalin, EGCG, and tannic acid. We further review promising in silico studies featuring activities of flavonoids against SARS-CoV-2 and list ongoing clinical studies involving the therapeutic potential of flavonoid-rich extracts in combination with synthetic drugs or other polyphenols and suggest prospects for the future of flavonoids against SARS-CoV-2.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Flavonoides/uso terapéutico , Antivirales/química , Antivirales/farmacología , COVID-19/virología , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/metabolismo , Proteínas de la Nucleocápside de Coronavirus/antagonistas & inhibidores , Proteínas de la Nucleocápside de Coronavirus/metabolismo , Flavonoides/química , Flavonoides/farmacología , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Rhinovirus/efectos de los fármacos , Rhinovirus/fisiología , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/metabolismo , Internalización del Virus/efectos de los fármacos
6.
Viruses ; 13(9)2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34578365

RESUMEN

Rhinoviruses (RVs) are the main cause of recurrent infections with rather mild symptoms characteristic of the common cold. Nevertheless, RVs give rise to enormous numbers of absences from work and school and may become life-threatening in particular settings. Vaccination is jeopardised by the large number of serotypes eliciting only poorly cross-neutralising antibodies. Conversely, antivirals developed over the years failed FDA approval because of a low efficacy and/or side effects. RV species A, B, and C are now included in the fifteen species of the genus Enteroviruses based upon the high similarity of their genome sequences. As a result of their comparably low pathogenicity, RVs have become a handy model for other, more dangerous members of this genus, e.g., poliovirus and enterovirus 71. We provide a short overview of viral proteins that are considered potential drug targets and their corresponding drug candidates. We briefly mention more recently identified cellular enzymes whose inhibition impacts on RVs and comment novel approaches to interfere with infection via aggregation, virus trapping, or preventing viral access to the cell receptor. Finally, we devote a large part of this article to adding the viral RNA genome to the list of potential drug targets by dwelling on its structure, folding, and the still debated way of its exit from the capsid. Finally, we discuss the recent finding that G-quadruplex stabilising compounds impact on RNA egress possibly via obfuscating the unravelling of stable secondary structural elements.


Asunto(s)
Antivirales/farmacología , ARN Viral/efectos de los fármacos , Rhinovirus/efectos de los fármacos , Aminoquinolinas/farmacología , Animales , Cápside/metabolismo , Proteínas de la Cápside/genética , Enterovirus/genética , Infecciones por Enterovirus/virología , Genoma Viral/efectos de los fármacos , Humanos , Ácidos Picolínicos/farmacología , Poliovirus/genética , Proteínas no Estructurales Virales/efectos de los fármacos , Proteínas Virales/genética
7.
SLAS Discov ; 26(8): 974-983, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34151629

RESUMEN

Affinity selection mass spectrometry (ASMS) has emerged as a powerful high-throughput screening tool used in drug discovery to identify novel ligands against therapeutic targets. This report describes the first high-throughput screen using a novel self-assembled monolayer desorption ionization (SAMDI)-ASMS methodology to reveal ligands for the human rhinovirus 3C (HRV3C) protease. The approach combines self-assembled monolayers of alkanethiolates on gold with matrix-assisted laser desorption ionization time-of-flight (MALDI TOF) mass spectrometry (MS), a technique termed SAMDI-ASMS. The primary screen of more than 100,000 compounds in pools of 8 compounds per well was completed in less than 8 h, and informs on the binding potential and selectivity of each compound. Initial hits were confirmed in follow-up SAMDI-ASMS experiments in single-concentration and dose-response curves. The ligands identified by SAMDI-ASMS were further validated using differential scanning fluorimetry (DSF) and in functional protease assays against HRV3C and the related SARS-CoV-2 3CLpro enzyme. SAMDI-ASMS offers key benefits for drug discovery over traditional ASMS approaches, including the high-throughput workflow and readout, minimizing compound misbehavior by using smaller compound pools, and up to a 50-fold reduction in reagent consumption. The flexibility of this novel technology opens avenues for high-throughput ASMS assays of any target, thereby accelerating drug discovery for diverse diseases.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Ensayos Analíticos de Alto Rendimiento , Rhinovirus/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Proteasas Virales 3C/química , COVID-19/virología , Descubrimiento de Drogas , Humanos , Ligandos , Espectrometría de Masas , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , Bibliotecas de Moléculas Pequeñas/aislamiento & purificación , Bibliotecas de Moléculas Pequeñas/uso terapéutico
8.
Comput Biol Chem ; 92: 107499, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33932782

RESUMEN

Rhinoviruses (RV), especially Human rhinovirus (HRVs) have been accepted as the most common cause for upper respiratory tract infections (URTIs). Pleconaril, a broad spectrum anti-rhinoviral compound, has been used as a drug of choice for URTIs for over a decade. Unfortunately, for various complications associated with this drug, it was rejected, and a replacement is highly desirable. In silico screening and prediction methods such as sub-structure search and molecular docking have been widely used to identify alternative compounds. In our study, we have utilised sub-structure search to narrow down our quest in finding relevant chemical compounds. Molecular docking studies were then used to study their binding interaction at the molecular level. Interestingly, we have identified 3 residues that is worth further investigation in upcoming molecular dynamics simulation systems of their contribution in stable interaction.


Asunto(s)
Antivirales/química , Simulación del Acoplamiento Molecular , Antivirales/farmacología , Evaluación Preclínica de Medicamentos , Humanos , Enlace de Hidrógeno , Estructura Molecular , Rhinovirus/efectos de los fármacos
9.
J Gen Virol ; 102(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33956593

RESUMEN

Host cell lipids play a pivotal role in the pathogenesis of respiratory virus infection. However, a direct comparison of the lipidomic profile of influenza virus and rhinovirus infections is lacking. In this study, we first compared the lipid profile of influenza virus and rhinovirus infection in a bronchial epithelial cell line. Most lipid features were downregulated for both influenza virus and rhinovirus, especially for the sphingomyelin features. Pathway analysis showed that sphingolipid metabolism was the most perturbed pathway. Functional study showed that bacterial sphingomyelinase suppressed influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication, but promoted rhinovirus replication. These findings suggest that sphingomyelin pathway can be a potential target for antiviral therapy, but should be carefully evaluated as it has opposite effects on different respiratory viruses. Furthermore, the differential effect of sphingomyelinase on rhinovirus and influenza virus may explain the interference between rhinovirus and influenza virus infection.


Asunto(s)
Orthomyxoviridae/efectos de los fármacos , Rhinovirus/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Esfingomielinas/farmacología , Animales , Enfermedades Bronquiales/virología , Línea Celular , Perros , Células Epiteliales/virología , Humanos , Gripe Humana , Lipidómica , Células de Riñón Canino Madin Darby , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Esfingomielina Fosfodiesterasa , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
10.
Antiviral Res ; 187: 105020, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33515606

RESUMEN

The 3-chymotrypsin-like cysteine protease (3CLpro) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is considered a major target for the discovery of direct antiviral agents. We previously reported the evaluation of SARS-CoV-2 3CLpro inhibitors in a novel self-assembled monolayer desorption ionization mass spectrometry (SAMDI-MS) enzymatic assay (Gurard-Levin et al., 2020). The assay was further improved by adding the rhinovirus HRV3C protease to the same well as the SARS-CoV-2 3CLpro enzyme. High substrate specificity for each enzyme allowed the proteases to be combined in a single assay reaction without interfering with their individual activities. This novel duplex assay was used to profile a diverse set of reference protease inhibitors. The protease inhibitors were grouped into three categories based on their relative potency against 3CLpro and HRV3C including those that are: equipotent against 3CLpro and HRV3C (GC376 and calpain inhibitor II), selective for 3CLpro (PF-00835231, calpain inhibitor XII, boceprevir), and selective for HRV3C (rupintrivir). Structural analysis showed that the combination of minimal interactions, conformational flexibility, and limited bulk allows GC376 and calpain inhibitor II to potently inhibit both enzymes. In contrast, bulkier compounds interacting more tightly with pockets P2, P3, and P4 due to optimization for a specific target display a more selective inhibition profile. Consistently, the most selective viral protease inhibitors were relatively weak inhibitors of human cathepsin L. Taken together, these results can guide the design of cysteine protease inhibitors that are either virus-specific or retain a broad antiviral spectrum against coronaviruses and rhinoviruses.


Asunto(s)
Antivirales/farmacología , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Rhinovirus/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Antivirales/química , Sitios de Unión , Catepsina L/metabolismo , Descubrimiento de Drogas , Glicoproteínas/farmacología , Humanos , Cinética , Modelos Moleculares , Inhibidores de Proteasas/química , Pirrolidinas/farmacología , Ácidos Sulfónicos
11.
Eur J Pharmacol ; 893: 173839, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33359650

RESUMEN

Inhaled corticosteroids (ICS) are recommended treatments for all degrees of asthma severity and in combination with bronchodilators are indicated for COPD patients with a history of frequent exacerbations. However, the long-term side effects of glucocorticoids (GCs) may include increased risk of respiratory infections, including viral triggered exacerbations. Rhinovirus (RV) infection is the main trigger of asthma and COPD exacerbations. Thus, we sought to explore the influence of GCs on viral replication. We demonstrate the ICS fluticasone propionate (FP) and two selective non-steroidal (GRT7) and steroidal (GRT10) glucocorticoid receptor (GR) agonists significantly suppress pro-inflammatory (IL-6 and IL-8) and antiviral (IFN-λ1) cytokine production and the expression of the interferon-stimulated genes (ISGs) OAS and viperin in RV-infected bronchial epithelial cells, with a consequent increase of viral replication. We also show that FP, GRT7 and GRT10 inhibit STAT1 Y701 and/or STAT2 Y690 phosphorylation and ISG mRNA induction following cell stimulation with recombinant IFN-ß. In addition, we investigated the effects of the ICS budesonide (BD) and the long-acting ß2 agonist (LABA) formoterol, alone or as an ICS/LABA combination, on RV-induced ISG expression and viral replication. Combination of BD/formoterol increases the suppression of OAS and viperin mRNA observed with both BD and formoterol alone, but an increase in viral RNA was only observed with BD treatment and not with formoterol. Overall, we provide evidence of an impairment of the innate antiviral immune response by GC therapy and the potential for GCs to enhance viral replication. These findings could have important clinical implications.


Asunto(s)
Bronquios/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Glucocorticoides/toxicidad , Mediadores de Inflamación/metabolismo , Interferón Tipo I/metabolismo , Rhinovirus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/toxicidad , Bronquios/inmunología , Bronquios/metabolismo , Bronquios/virología , Quimioterapia Combinada , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Fumarato de Formoterol/toxicidad , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata/efectos de los fármacos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Proteínas/genética , Proteínas/metabolismo , Rhinovirus/crecimiento & desarrollo , Rhinovirus/inmunología , Transducción de Señal
12.
BMC Complement Med Ther ; 20(1): 380, 2020 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-33357221

RESUMEN

BACKGROUND: Rhinoviruses and influenza viruses cause millions of acute respiratory infections annually. Symptoms of mild acute respiratory infections are commonly treated with over-the-counter products like ambroxol, bromhexine, and N-acetyl cysteine, as well as of thyme and pelargonium extracts today. Because the direct antiviral activity of these over-the-counter products has not been studied in a systematic way, the current study aimed to compare their inhibitory effect against rhinovirus and influenza virus replication in an in vitro setting. METHODS: The cytotoxicity of ambroxol, bromhexine, and N-acetyl cysteine, as well as of thyme and pelargonium extracts was analyzed in Madin Darby canine kidney (MDCK) and HeLa Ohio cells. The antiviral effect of these over-the-counter products was compared by analyzing the dose-dependent inhibition (i) of rhinovirus A2- and B14-induced cytopathic effect in HeLa Ohio cells and (ii) of influenza virus A/Hong Kong/68 (subtype H3N2)- and A/Jena/8178/09 (subtype H1N1, pandemic)-induced cytopathic effect in MDCK cells at non-cytotoxic concentrations. To get insights into the mechanism of action of pelargonium extract against influenza virus, we performed time-of-addition assays as well as hemagglutination and neuraminidase inhibition assays. RESULTS: N-acetyl cysteine, thyme and pelargonium extract showed no or only marginal cytotoxicity in MDCK and HeLa Ohio cells in the tested concentration range. The 50% cytotoxic concentration of ambroxol and bromhexine was 51.85 and 61.24 µM, respectively. No anti-rhinoviral activity was detected at non-cytotoxic concentrations in this in vitro study setting. Ambroxol, bromhexine, and N-acetyl cysteine inhibited the influenza virus-induced cytopathic effect in MDCK cells no or less than 50%. In contrast, a dose-dependent anti-influenza virus activity of thyme and pelargonium extracts was demonstrated. The time-of addition assays revealed an inhibition of early and late steps of influenza virus replication by pelargonium extract whereas zanamivir acted on late steps only. The proven block of viral neuraminidase activity might explain the inhibition of influenza virus replication when added after viral adsorption. CONCLUSION: The study results indicate a distinct inhibition of influenza A virus replication by thyme and pelargonium extract which might contribute to the beneficial effects of these plant extracts on acute respiratory infections symptoms.


Asunto(s)
Virus de la Influenza A/efectos de los fármacos , Pelargonium , Extractos Vegetales/uso terapéutico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Rhinovirus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Acetilcisteína , Ambroxol , Animales , Bromhexina , Perros , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Pruebas de Sensibilidad Microbiana , Fitoterapia , Extractos Vegetales/farmacología , Thymus (Planta) , Pruebas de Toxicidad
13.
Nat Rev Immunol ; 20(11): 709-713, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33024281

RESUMEN

Immunity is a multifaceted phenomenon. For T cell-mediated memory responses to SARS-CoV-2, it is relevant to consider their impact both on COVID-19 disease severity and on viral spread in a population. Here, we reflect on the immunological and epidemiological aspects and implications of pre-existing cross-reactive immune memory to SARS-CoV-2, which largely originates from previous exposure to circulating common cold coronaviruses. We propose four immunological scenarios for the impact of cross-reactive CD4+ memory T cells on COVID-19 severity and viral transmission. For each scenario, we discuss its implications for the dynamics of herd immunity and on projections of the global impact of SARS-CoV-2 on the human population, and assess its plausibility. In sum, we argue that key potential impacts of cross-reactive T cell memory are already incorporated into epidemiological models based on data of transmission dynamics, particularly with regard to their implications for herd immunity. The implications of immunological processes on other aspects of SARS-CoV-2 epidemiology are worthy of future study.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Betacoronavirus/inmunología , Infecciones por Coronaviridae/prevención & control , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/prevención & control , Vacunas Virales/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Betacoronavirus/efectos de los fármacos , Betacoronavirus/patogenicidad , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , COVID-19 , Vacunas contra la COVID-19 , Coronaviridae/efectos de los fármacos , Coronaviridae/inmunología , Infecciones por Coronaviridae/epidemiología , Infecciones por Coronaviridae/inmunología , Infecciones por Coronaviridae/virología , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Reacciones Cruzadas , Humanos , Inmunidad Colectiva/efectos de los fármacos , Memoria Inmunológica , Neumonía Viral/epidemiología , Neumonía Viral/inmunología , Neumonía Viral/virología , Rhinovirus/efectos de los fármacos , Rhinovirus/inmunología , SARS-CoV-2 , Vacunas Virales/administración & dosificación , Vacunas Virales/biosíntesis
14.
Antiviral Res ; 183: 104933, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32949635

RESUMEN

Stimulator of interferon genes (STING), as a signaling hub in innate immunity, plays a central role for the effective initiation of host defense mechanisms against microbial infections. Upon binding of its ligand cyclic dinucleotides (CDNs) produced by the cyclic GMP-AMP synthase (cGAS) or invading bacteria, STING is activated, leading to the induction of both type I interferon responses and autophagy, which are critical for the control of certain microbial infections. RNA viruses, such as Parainfluenza virus (PIV) and Rhinovirus (HRV), are among the leading causes of respiratory infections that affect human health without effective treatments. Activation of STING pathway may provide a new therapeutic approach fighting against these viruses. However, the role of STING in the control of RNA virus infection remains largely unexplored. In this study, using dimeric amidobenzimidazole (diABZI), a newly discovered synthetic small molecule STING receptor agonist with much higher potency than CDNs, we found that activation of STING elicits potent antiviral effects against parainfluenza virus type 3 (PIV3) and human rhinovirus 16 (HRV16), two representative respiratory viral pathogens. Notably, while anti-PIV3 activity was depend on the induction of type I interferon responses through TANK-binding kinase 1 (TBK1), anti-HRV16 activity required the induction of autophagy-related gene 5 (ATG5)-dependent autophagy, indicating that two distinct antiviral mechanisms are engaged upon STING activation. Antiviral activity and individual specific pathway was further confirmed in infected primary bronchial epithelial cells. Our findings thus demonstrate the distinct antiviral mechanisms triggered by STING agonist and uncover the potential of therapeutic effect against different viruses.


Asunto(s)
Antivirales/farmacología , Proteínas de la Membrana/agonistas , Virus de la Parainfluenza 3 Humana/efectos de los fármacos , Rhinovirus/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/síntesis química , Autofagia , Línea Celular , Células Cultivadas , Células HeLa , Humanos , Inmunidad Innata , Ratones , Virus de la Parainfluenza 3 Humana/fisiología , Células RAW 264.7 , Rhinovirus/fisiología , Transducción de Señal/inmunología , Células THP-1
15.
Nat Commun ; 11(1): 4252, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32843628

RESUMEN

The 2019 novel respiratory virus (SARS-CoV-2) causes COVID-19 with rapid global socioeconomic disruptions and disease burden to healthcare. The COVID-19 and previous emerging virus outbreaks highlight the urgent need for broad-spectrum antivirals. Here, we show that a defensin-like peptide P9R exhibited potent antiviral activity against pH-dependent viruses that require endosomal acidification for virus infection, including the enveloped pandemic A(H1N1)pdm09 virus, avian influenza A(H7N9) virus, coronaviruses (SARS-CoV-2, MERS-CoV and SARS-CoV), and the non-enveloped rhinovirus. P9R can significantly protect mice from lethal challenge by A(H1N1)pdm09 virus and shows low possibility to cause drug-resistant virus. Mechanistic studies indicate that the antiviral activity of P9R depends on the direct binding to viruses and the inhibition of virus-host endosomal acidification, which provides a proof of concept that virus-binding alkaline peptides can broadly inhibit pH-dependent viruses. These results suggest that the dual-functional virus- and host-targeting P9R can be a promising candidate for combating pH-dependent respiratory viruses.


Asunto(s)
Antivirales/farmacología , Coronavirus/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Antivirales/química , Antivirales/metabolismo , Antivirales/uso terapéutico , Línea Celular , Endosomas/química , Endosomas/efectos de los fármacos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Virus de la Influenza A/metabolismo , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/metabolismo , Péptidos/química , Péptidos/metabolismo , Péptidos/uso terapéutico , Unión Proteica , Conformación Proteica , Rhinovirus/efectos de los fármacos , Rhinovirus/metabolismo , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
16.
Viruses ; 12(7)2020 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-32635420

RESUMEN

The neutrophil extracellular trap (ET) is a eukaryotic host defense machinery that operates by capturing and concentrating pathogens in a filamentous network manufactured by neutrophils and made of DNA, histones, and many other components. Respiratory virus-induced ETs are involved in tissue damage and impairment of the alveolar-capillary barrier, but they also aid in fending off infection. We found that the small organic compound pyridostatin (PDS) forms somewhat similar fibrillary structures in Tris buffer in a concentration-dependent manner. Common cold viruses promote this process and become entrapped in the network, decreasing their infectivity by about 70% in tissue culture. We propose studying this novel mechanism of virus inhibition for its utility in preventing viral infection.


Asunto(s)
Aminoquinolinas/farmacología , Antivirales/farmacología , Ácidos Picolínicos/farmacología , Rhinovirus/efectos de los fármacos , Trometamina/química , Células Cultivadas , Resfriado Común/prevención & control , Resfriado Común/virología , Trampas Extracelulares/fisiología , Células HeLa , Humanos , Microscopía Electrónica de Transmisión , Neutrófilos , Rhinovirus/ultraestructura
17.
Org Biomol Chem ; 18(31): 6155-6161, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32716466

RESUMEN

The aminated mimetics of 2-keto-3-deoxy-sugar acids such as the anti-influenza clinical drugs oseltamivir (Tamiflu) and zanamivir (Relenza) are important bioactive molecules. Development of synthetic methodologies for accessing such compound collections is highly desirable. Herein, we describe a simple, catalyst-free glycal diazidation protocol enabled by visible light-driven conditions. This new method requires neither acid promoters nor transition-metal catalysts and takes place at ambient temperature within 1-2 hours. Notably, the desired transformations could be promoted by thermal conditions as well, albeit with lower efficacy compared to the light-induced conditions. Different sugar acid-derived glycal templates have been converted into a range of 2,3-diazido carbohydrate analogs by harnessing this mild and scalable approach, leading to the discovery of new antiviral agents.


Asunto(s)
Antivirales/farmacología , Azidas/farmacología , Carbohidratos/farmacología , Calor , Luz , Rhinovirus/efectos de los fármacos , Azúcares Ácidos/farmacología , Virus Zika/efectos de los fármacos , Antivirales/síntesis química , Antivirales/química , Azidas/síntesis química , Azidas/química , Conformación de Carbohidratos , Carbohidratos/síntesis química , Carbohidratos/química , Pruebas de Sensibilidad Microbiana , Azúcares Ácidos/química
18.
SLAS Discov ; 25(6): 634-645, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32189556

RESUMEN

Human rhinovirus (RV) is the most common cause of acute upper respiratory tract infections and has recently been shown to play a significant role in exacerbations of asthma and chronic obstructive pulmonary disease (COPD). There is a significant unmet medical need for agents for the prevention and/or treatment of exacerbations triggered by human RV infection. Phenotypic drug discovery programs using different perturbation modalities, for example, siRNA, small-molecule compounds, and CRISPR, hold significant value for identifying novel drug targets. We have previously reported the identification of lanosterol synthase as a novel regulator of RV2 replication through a phenotypic screen of a library of siRNAs against druggable genes in normal human bronchial epithelial (NHBE) cells. Here, we describe a follow-up phenotypic screen of small-molecule compounds that are annotated to be pharmacological regulators of target genes that were identified to significantly affect RV2 replication in the siRNA primary screen of 10,500 druggable genes. Two hundred seventy small-molecule compounds selected for interacting with 122 target gene hits were screened in the primary RV2 assay in NHBE cells by quantifying viral replication via in situ hybridization followed by secondary quantitative PCR-based assays for RV2, RV14, and RV16. The described follow-up phenotypic screening allowed us to identify Fms-related tyrosine kinase 4 (FLT4) as a novel target regulating RV replication. We demonstrate that a combination of siRNA and small-molecule compound screening models is a useful phenotypic drug discovery approach for the identification of novel drug targets.


Asunto(s)
Transferasas Intramoleculares/genética , Rhinovirus/efectos de los fármacos , Virosis/tratamiento farmacológico , Replicación Viral/efectos de los fármacos , Bronquios/efectos de los fármacos , Bronquios/virología , Sistemas CRISPR-Cas/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , ARN Interferente Pequeño/genética , Rhinovirus/patogenicidad , Bibliotecas de Moléculas Pequeñas/farmacología , Virosis/genética , Virosis/virología
19.
Arch Pharm Res ; 43(5): 526-539, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-28861755

RESUMEN

Rhinoviral infection is associated with an increased risk of asthma attacks. The macrolide clarithromycin decreases cytokine production in nasopharyngeal aspirates from patients with wheezing, but the effects of macrolides on cytokine production in nasal epithelial cells obtained from asthmatic subjects remain unclear. Here, human nasal epithelial cells were infected with type-14 rhinovirus (RV14), a major RV group. Titers and RNA of RV14 and cytokine concentrations, including IL-1ß and IL-6, were higher in the supernatants of the cells obtained from subjects with bronchial asthma (asthmatic group) than in those from the non-asthmatic group. Pretreatment with clarithromycin decreased RV14 titers, viral RNA and cytokine concentrations, and susceptibility to RV14 infection. Pretreatment with clarithromycin also decreased IL-33 production, which was detected after infection. Pretreatment with clarithromycin decreased the expression of intercellular adhesion molecule-1, the receptor for RV14, after infection, the number and fluorescence intensity of the acidic endosomes through which RV RNA enters the cytoplasm, and the activation of nuclear factor kappa-B proteins in nuclear extracts. These findings suggested that RV replication and cytokine production may be enhanced in nasal epithelial cells obtained from subjects with bronchial asthma and may be modulated by clarithromycin.


Asunto(s)
Antivirales/farmacología , Asma/tratamiento farmacológico , Claritromicina/farmacología , Citocinas/biosíntesis , Células Epiteliales/efectos de los fármacos , Rhinovirus/efectos de los fármacos , Asma/metabolismo , Células Cultivadas , Células Epiteliales/metabolismo , Femenino , Humanos , Interleucina-33/antagonistas & inhibidores , Interleucina-33/biosíntesis , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Interleucina-8/antagonistas & inhibidores , Interleucina-8/biosíntesis , Masculino , Persona de Mediana Edad , Replicación Viral/efectos de los fármacos
20.
Proc Natl Acad Sci U S A ; 116(38): 19109-19115, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31462495

RESUMEN

Viral inhibitors, such as pleconaril and vapendavir, target conserved regions in the capsids of rhinoviruses (RVs) and enteroviruses (EVs) by binding to a hydrophobic pocket in viral capsid protein 1 (VP1). In resistant RVs and EVs, bulky residues in this pocket prevent their binding. However, recently developed pyrazolopyrimidines inhibit pleconaril-resistant RVs and EVs, and computational modeling has suggested that they also bind to the hydrophobic pocket in VP1. We studied the mechanism of inhibition of pleconaril-resistant RVs using RV-B5 (1 of the 7 naturally pleconaril-resistant rhinoviruses) and OBR-5-340, a bioavailable pyrazolopyrimidine with proven in vivo activity, and determined the 3D-structure of the protein-ligand complex to 3.6 Å with cryoelectron microscopy. Our data indicate that, similar to other capsid binders, OBR-5-340 induces thermostability and inhibits viral adsorption and uncoating. However, we found that OBR-5-340 attaches closer to the entrance of the pocket than most other capsid binders, whose viral complexes have been studied so far, showing only marginal overlaps of the attachment sites. Comparing the experimentally determined 3D structure with the control, RV-B5 incubated with solvent only and determined to 3.2 Å, revealed no gross conformational changes upon OBR-5-340 binding. The pocket of the naturally OBR-5-340-resistant RV-A89 likewise incubated with OBR-5-340 and solved to 2.9 Å was empty. Pyrazolopyrimidines have a rigid molecular scaffold and may thus be less affected by a loss of entropy upon binding. They interact with less-conserved regions than known capsid binders. Overall, pyrazolopyrimidines could be more suitable for the development of new, broadly active inhibitors.


Asunto(s)
Antivirales/metabolismo , Cápside/metabolismo , Microscopía por Crioelectrón/métodos , Farmacorresistencia Viral , Oxadiazoles/farmacología , Rhinovirus/metabolismo , Proteínas Virales/química , Antivirales/farmacología , Sitios de Unión , Cápside/efectos de los fármacos , Cápside/ultraestructura , Células HeLa , Humanos , Modelos Moleculares , Estructura Molecular , Oxazoles , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/virología , Unión Proteica , Conformación Proteica , Rhinovirus/efectos de los fármacos , Rhinovirus/ultraestructura , Relación Estructura-Actividad , Proteínas Virales/genética , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA