Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 269
Filtrar
1.
Virus Res ; 349: 199448, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39127240

RESUMEN

Adenoviral infections, particularly in children, remain a significant public health issue with no approved targeted treatments. Artemisinin and its derivatives, well-known for their use in malaria treatment, have shown antiviral activities in recent studies. However, their efficacy against human adenovirus (HAdV) remains unexplored. This study aimed to assess the activity of artemisinin and its derivatives against HAdV infection in vitro using cell lines and primary cells. Our data revealed that artemisinin exhibited dose-dependent anti-HAdV activity with no apparent cytotoxicity over a wide concentration range. Mechanistically, artemisinin did not affect viral attachment or entry into target cells, nor the viral genome entry into cell nucleus. Instead, it inhibited HAdV through suppression of viral DNA replication. Comparative analysis with its derivatives, artesunate and artemisone, showed distinct cytotoxicity and anti-adenoviral profiles, with artemisone showing superior efficacy and lower toxicity. Further validation using a primary airway epithelial cell model confirmed the anti-adenoviral activity of both artemisinin and artemisone against different virus strains. Together, our findings suggest that artemisinin and its derivatives may be promising candidates for anti-HAdV treatment.


Asunto(s)
Adenovirus Humanos , Antivirales , Artemisininas , Replicación Viral , Artemisininas/farmacología , Humanos , Antivirales/farmacología , Replicación Viral/efectos de los fármacos , Adenovirus Humanos/efectos de los fármacos , Adenovirus Humanos/fisiología , Línea Celular , Artesunato/farmacología , Células Epiteliales/virología , Células Epiteliales/efectos de los fármacos , Infecciones por Adenovirus Humanos/virología , Infecciones por Adenovirus Humanos/tratamiento farmacológico
2.
J Water Health ; 22(6): 1102-1110, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38935460

RESUMEN

Ferrate (Fe(VI): HFeO4- /FeO42-), a potent oxidant, has been investigated as an alternative chemical disinfectant in water treatment due to its reduced production of disinfection by-products. In this study, we assessed the disinfecting ability of potassium ferrate against a variety of microorganisms, including waterborne pathogens, under varying pH and water temperature conditions. We presented CT values, a metric of ferrate concentrations (C) and contact time (T), to quantify microbial inactivation rates. Among the tested microorganisms, human adenovirus was the least resistant to ferrate, followed by waterborne bacteria such as Escherichia coli and Vibrio cholerae, and finally, the protozoan parasite Giardia duodenalis. We further investigated the impact of two pH values (7 and 8) and two temperatures (5 and 25 °C) on microbial inactivation rates, observing that inactivation rates increased with lower pH and higher temperature. In addition to showcasing ferrate's capacity to effectively inactivate a range of the tested microorganisms, we offer a ferrate CT table to facilitate the comparison of the effectiveness of various disinfection methods.


Asunto(s)
Desinfectantes , Giardia lamblia , Temperatura , Concentración de Iones de Hidrógeno , Desinfectantes/farmacología , Giardia lamblia/efectos de los fármacos , Adenovirus Humanos/efectos de los fármacos , Compuestos de Potasio/farmacología , Compuestos de Potasio/química , Microbiología del Agua , Desinfección/métodos , Purificación del Agua/métodos , Compuestos de Hierro/farmacología , Compuestos de Hierro/química , Humanos , Escherichia coli/efectos de los fármacos
3.
Antimicrob Agents Chemother ; 68(7): e0048924, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38775484

RESUMEN

Human adenoviruses can cause serious, disseminated infections in immunocompromised patients. For pediatric allogeneic stem cell transplant patients, the case fatality rate can reach 80%. Still, there is no available antiviral drug that is specifically approved by the Food and Drug Administration for the treatment of adenovirus infections. To fill this pressing medical need, we have developed NPP-669, a prodrug of cidofovir with broad activity against double-stranded DNA viruses, including adenoviruses. Here, we report on the in vivo anti-adenoviral efficacy of NPP-669. Using the immunosuppressed Syrian hamster as the model, we show that NPP-669 is highly efficacious when dosed orally at 1 mg/kg and 3 mg/kg. In a delayed administration experiment, NPP-669 was more effective than brincidofovir, a similar compound that reached Phase III clinical trials. Furthermore, parenteral administration of NPP-669 increased its efficacy approximately 10-fold compared to oral dosing without apparent toxicity, suggesting that this route may be preferable in a hospital setting. Based on these findings, we believe that NPP-669 is a promising new compound that needs to be further investigated.


Asunto(s)
Antivirales , Cidofovir , Citosina , Mesocricetus , Organofosfonatos , Profármacos , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Profármacos/farmacología , Profármacos/uso terapéutico , Humanos , Cidofovir/farmacología , Cidofovir/uso terapéutico , Organofosfonatos/farmacología , Organofosfonatos/uso terapéutico , Citosina/análogos & derivados , Citosina/farmacología , Citosina/uso terapéutico , Adenovirus Humanos/efectos de los fármacos , Infecciones por Adenovirus Humanos/tratamiento farmacológico , Infecciones por Adenovirus Humanos/virología , Modelos Animales de Enfermedad , Cricetinae , Administración Oral
4.
Biomed Pharmacother ; 174: 116558, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38603887

RESUMEN

Human adenovirus (HAdV) infection is a major cause of respiratory disease, yet no antiviral drugs have been approved for its treatment. Herein, we evaluated the antiviral and anti-inflammatory effects of cyclin-dependent protein kinase (CDK) inhibitor indirubin-3'-monoxime (IM) against HAdV infection in cells and a transgenic mouse model. After evaluating its cytotoxicity, cytopathic effect reduction, antiviral replication kinetics, and viral yield reduction assays were performed to assess the anti-HAdV activity of IM. Quantitative real-time polymerase chain reaction (qPCR), quantitative reverse transcription PCR (qRT-PCR), and western blotting were used to assess the effects of IM on HAdV DNA replication, transcription, and protein expression, respectively. IM significantly inhibited HAdV DNA replication as well as E1A and Hexon transcription, in addition to significantly suppressing the phosphorylation of the RNA polymerase II C-terminal domain (CTD). IM mitigated body weight loss, reduced viral burden, and lung injury, decreasing cytokine and chemokine secretion to a greater extent than cidofovir. Altogether, IM inhibits HAdV replication by downregulating CTD phosphorylation to suppress viral infection and corresponding innate immune reactions as a promising therapeutic agent.


Asunto(s)
Adenovirus Humanos , Antiinflamatorios , Antivirales , Indoles , Oximas , Replicación Viral , Indoles/farmacología , Animales , Oximas/farmacología , Humanos , Antivirales/farmacología , Adenovirus Humanos/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Antiinflamatorios/farmacología , Ratones , Ratones Transgénicos , Infecciones por Adenovirus Humanos/tratamiento farmacológico , Infecciones por Adenovirus Humanos/virología , Células A549 , Citocinas/metabolismo , Fosforilación/efectos de los fármacos
5.
Viruses ; 16(4)2024 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-38675973

RESUMEN

Differentiated HepaRG cells are popular in vitro cell models for hepatotoxicity studies. Their differentiation is usually supported by the addition of dimethyl sulfoxide (DMSO), an amphipathic solvent widely used in biomedicine, for example, in potential novel therapeutic drugs and cryopreservation of oocytes. Recent studies have demonstrated drastic effects, especially on epigenetics and extracellular matrix composition, induced by DMSO, making its postulated inert character doubtful. In this work, the influence of DMSO and DMSO-mediated modulation of differentiation on human adenovirus (HAdV) infection of HepaRG cells was investigated. We observed an increase in infectivity of HepaRG cells by HAdVs in the presence of 1% DMSO. However, this effect was dependent on the type of medium used for cell cultivation, as cells in William's E medium showed significantly stronger effects compared with those cultivated in DMEM. Using different DMSO concentrations, we proved that the impact of DMSO on infectability was dose-dependent. Infection of cells with a replication-deficient HAdV type demonstrated that the mode of action of DMSO was based on viral entry rather than on viral replication. Taken together, these results highlight the strong influence of the used cell-culture medium on the performed experiments as well as the impact of DMSO on infectivity of HepaRG cells by HAdVs. As this solvent is widely used in cell culture, those effects must be considered, especially in screening of new antiviral compounds.


Asunto(s)
Adenovirus Humanos , Diferenciación Celular , Dimetilsulfóxido , Replicación Viral , Dimetilsulfóxido/farmacología , Humanos , Adenovirus Humanos/efectos de los fármacos , Adenovirus Humanos/fisiología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Replicación Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Hepatocitos/virología , Hepatocitos/efectos de los fármacos , Infecciones por Adenovirus Humanos/virología , Medios de Cultivo/química
6.
Food Environ Virol ; 16(2): 143-158, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38308001

RESUMEN

Removal of pathogenic viruses from water resources is critically important for sanitation and public health. Nanotechnology is a promising technology for virus inactivation. In this paper, the effects of titanium dioxide (TiO2) anatase nanoparticles (NPs) on human adenovirus type 35 (HAdV-35) removal under static and dynamic (with agitation) batch conditions were comprehensively studied. Batch experiments were performed at room temperature (25 °C) with and without ambient light using three different initial virus concentrations. The virus inactivation experimental data were satisfactorily fitted with a pseudo-first-order expression with a time-dependent rate coefficient. The experimental results demonstrated that HAdV-35 sorption onto TiO2 NPs was favored with agitation under both ambient light and dark conditions. However, no distinct relationships between virus initial concentration and removal efficiency could be established from the experimental data.


Asunto(s)
Adenovirus Humanos , Nanopartículas , Titanio , Inactivación de Virus , Titanio/química , Titanio/farmacología , Adenovirus Humanos/efectos de los fármacos , Adenovirus Humanos/química , Adenovirus Humanos/fisiología , Adenovirus Humanos/genética , Adsorción , Humanos , Inactivación de Virus/efectos de los fármacos , Nanopartículas/química , Purificación del Agua/métodos , Purificación del Agua/instrumentación , Cinética
7.
Int J Antimicrob Agents ; 63(5): 107116, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38401774

RESUMEN

Human adenovirus (HAdV) and cytomegalovirus (HCMV) cause high morbidity and mortality in patients undergoing solid organ transplantation (SOT) and haematopoietic stem cell transplantation (HSCT). Immunosuppressors are used universally to prevent graft-vs-host disease in HSCT and graft rejection in SOT. The long-term use of these drugs is associated with a high risk of infection, but there is also evidence of their specific interference with viral infection. This study evaluated the antiviral activity of immunosuppressors commonly used in clinical practice in SOT and HSCT recipients in vitro to determine whether their use could be associated with reduced risk of HAdV and HCMV infection. Cyclophosphamide, tacrolimus, cyclosporine, mycophenolic acid, methotrexate, everolimus and sirolimus presented antiviral activity, with 50% inhibitory concentration (IC50) values at low micromolar and sub-micromolar concentrations. Mycophenolic acid and methotrexate showed the greatest antiviral effects against HAdV (IC50=0.05 µM and 0.3 µM, respectively) and HCMV (IC50=10.8 µM and 0.02 µM, respectively). The combination of tacrolimus and mycophenolic acid showed strong synergistic antiviral activity against both viruses, with combinatory indexes (CI50) of 0.02 and 0.25, respectively. Additionally, mycophenolic acid plus cyclosporine, and mycophenolic acid plus everolimus/sirolimus showed synergistic antiviral activity against HAdV (CI50=0.05 and 0.09, respectively), while methotrexate plus cyclosporine showed synergistic antiviral activity against HCMV (CI50=0.29). These results, showing antiviral activity in vitro against both HAdV and HCMV, at concentrations below the human Cmax values, may be relevant for the selection of specific immunosuppressant therapies in patients at risk of HAdV and HCMV infections.


Asunto(s)
Adenovirus Humanos , Antivirales , Citomegalovirus , Inmunosupresores , Humanos , Inmunosupresores/farmacología , Antivirales/farmacología , Adenovirus Humanos/efectos de los fármacos , Citomegalovirus/efectos de los fármacos , Sinergismo Farmacológico , Concentración 50 Inhibidora , Ácido Micofenólico/farmacología , Tacrolimus/farmacología , Ciclosporina/farmacología , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/virología , Infecciones por Citomegalovirus/prevención & control
8.
Viruses ; 13(10)2021 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-34696411

RESUMEN

Viral proteases are indispensable for successful virion maturation, thus making them a prominent drug target. Their enzyme activity is tightly spatiotemporally regulated by expression in the precursor form with little or no activity, followed by activation via autoprocessing. These cleavage events are frequently triggered upon transportation to a specific compartment inside the host cell. Typically, precursor oligomerization or the presence of a co-factor is needed for activation. A detailed understanding of these mechanisms will allow ligands with non-canonical mechanisms of action to be designed, which would specifically modulate the initial irreversible steps of viral protease autoactivation. Binding sites exclusive to the precursor, including binding sites beyond the protease domain, can be exploited. Both inhibition and up-regulation of the proteolytic activity of viral proteases can be detrimental for the virus. All these possibilities are discussed using examples of medically relevant viruses including herpesviruses, adenoviruses, retroviruses, picornaviruses, caliciviruses, togaviruses, flaviviruses, and coronaviruses.


Asunto(s)
Antivirales/farmacología , Inhibidores de Proteasa Viral/farmacología , Proteasas Virales/metabolismo , Virosis/tratamiento farmacológico , Adenovirus Humanos/efectos de los fármacos , Adenovirus Humanos/metabolismo , Flavivirus/efectos de los fármacos , Flavivirus/metabolismo , VIH-1/efectos de los fármacos , Herpesviridae/efectos de los fármacos , Herpesviridae/metabolismo , Humanos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , Proteasas Virales/biosíntesis
9.
Int J Mol Sci ; 22(18)2021 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-34576237

RESUMEN

Previous studies reported on the broad-spectrum antiviral function of heparin. Here we investigated the antiviral function of magnesium-modified heparin and found that modified heparin displayed a significantly enhanced antiviral function against human adenovirus (HAdV) in immortalized and primary cells. Nuclear magnetic resonance analyses revealed a conformational change of heparin when complexed with magnesium. To broadly explore this discovery, we tested the antiviral function of modified heparin against herpes simplex virus type 1 (HSV-1) and found that the replication of HSV-1 was even further decreased compared to aciclovir. Moreover, we investigated the antiviral effect against the new severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and measured a 55-fold decreased viral load in the supernatant of infected cells associated with a 38-fold decrease in virus growth. The advantage of our modified heparin is an increased antiviral effect compared to regular heparin.


Asunto(s)
Antivirales/farmacología , Heparina/farmacología , Cloruro de Magnesio/farmacología , Aciclovir/farmacología , Adenovirus Humanos/efectos de los fármacos , Adenovirus Humanos/fisiología , Animales , Antivirales/química , Células CHO , Línea Celular Tumoral , Chlorocebus aethiops , Cricetulus , Evaluación Preclínica de Medicamentos , Fibroblastos , Heparina/química , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/fisiología , Humanos , Cloruro de Magnesio/química , Espectroscopía de Resonancia Magnética , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Cultivo Primario de Células , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Relación Estructura-Actividad , Células Vero , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
10.
Viruses ; 13(8)2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34452467

RESUMEN

Type III interferons (lambda IFNs) are a quite new, small family of three closely related cytokines with interferon-like activity. Attention to IFN-λ is mainly focused on direct antiviral activity in which, as with IFN-α, viral genome replication is inhibited without the participation of immune system cells. The heterodimeric receptor for lambda interferons is exposed mainly on epithelial cells, which limits its possible action on other cells, thus reducing the likelihood of developing undesirable side effects compared to type I IFN. In this study, we examined the antiviral potential of exogenous human IFN-λ1 in cellular models of viral infection. To study the protective effects of IFN-λ1, three administration schemes were used: 'preventive' (pretreatment); 'preventive/therapeutic' (pre/post); and 'therapeutic' (post). Three IFN-λ1 concentrations (from 10 to 500 ng/mL) were used. We have shown that human IFN-λ1 restricts SARS-CoV-2 replication in Vero cells with all three treatment schemes. In addition, we have shown a decrease in the viral loads of CHIKV and IVA with the 'preventive' and 'preventive/therapeutic' regimes. No significant antiviral effect of IFN-λ1 against AdV was detected. Our study highlights the potential for using IFN-λ as a broad-spectrum therapeutic agent against respiratory RNA viruses.


Asunto(s)
Adenovirus Humanos/efectos de los fármacos , Virus Chikungunya/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Interferones/farmacología , SARS-CoV-2/efectos de los fármacos , Células A549 , Adenovirus Humanos/fisiología , Animales , Virus Chikungunya/fisiología , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Humanos , Virus de la Influenza A/fisiología , Interferones/uso terapéutico , Interleucinas , Infecciones por Virus ARN/tratamiento farmacológico , Infecciones por Virus ARN/prevención & control , Proteínas Recombinantes/farmacología , SARS-CoV-2/fisiología , Células Vero , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Interferón lambda
11.
Molecules ; 26(11)2021 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-34198721

RESUMEN

There is no approved antiviral therapy for adenovirus (HAdV) ocular infections. Astodrimer sodium (SPL7013) is a polyanionic dendrimer with antiviral activity. The current study evaluated the ocular tolerability and anti-adenoviral efficacy of topical SPL7013 in rabbit ocular models. In a tolerability study, rabbits were treated with 3% SPL7013, vehicle, or 0.5% cidofovir. Their eyes were graded using the Draize scale. In antiviral efficacy studies, HAdV5 inoculated eyes were treated with 3% SPL7013, vehicle, or 0.5% cidofovir. Eyes were cultured for the virus on days 0, 1, 3, 4, 5, 7, 9, 11, and 14. Viral titers were determined. There were no differences in Draize scores between 3% SPL7013 and vehicle on any day. Cidofovir produced significantly higher Draize scores on day 12 than SPL7013 and vehicle. The 3% SPL7013 and 0.5% cidofovir significantly reduced daily viral titers and positive cultures per total compared with vehicle on several different days. The 3% SPL7013 and 0.5% cidofovir significantly reduced the duration of HAdV5 shedding compared to vehicle. The 3% SPL7013 demonstrated significantly more antiviral activity compared with vehicle in the Ad5/NZW rabbit ocular model. The 3% SPL7013 induced "minimal" to "practically non-irritating" Draize scores in the ocular tolerability study. Further development of astodrimer sodium as a topical antiviral therapy for adenoviral ocular infections is indicated.


Asunto(s)
Infecciones por Adenoviridae/tratamiento farmacológico , Cidofovir/administración & dosificación , Dendrímeros/administración & dosificación , Infecciones Virales del Ojo/tratamiento farmacológico , Polilisina/administración & dosificación , Células A549 , Adenovirus Humanos/efectos de los fármacos , Adenovirus Humanos/fisiología , Administración Tópica , Animales , Cidofovir/farmacología , Dendrímeros/farmacología , Modelos Animales de Enfermedad , Femenino , Humanos , Polilisina/farmacología , Conejos , Resultado del Tratamiento , Carga Viral/efectos de los fármacos
12.
Antiviral Res ; 188: 105034, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33577808

RESUMEN

Human adenoviruses (HAdV) are ubiquitous human pathogens that cause a significant burden of respiratory, ocular, and gastrointestinal illnesses. Although HAdV infections are generally self-limiting, pediatric and immunocompromised individuals are at particular risk for developing severe disease. Currently, no approved antiviral therapies specific to HAdV exist. Recent outbreaks underscore the need for effective antiviral agents to treat life-threatening infections. In this review we will focus on recent developments in search of potential therapeutic agents for controlling HAdV infections, with a focus on those targeting post-entry stages of the virus replicative cycle.


Asunto(s)
Infecciones por Adenovirus Humanos/tratamiento farmacológico , Adenovirus Humanos/efectos de los fármacos , Antivirales/uso terapéutico , Transporte Activo de Núcleo Celular/efectos de los fármacos , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/genética , Adenovirus Humanos/fisiología , Antivirales/farmacología , Replicación del ADN/efectos de los fármacos , Reposicionamiento de Medicamentos , Quimioterapia Combinada , Epigénesis Genética/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Inmunoterapia Adoptiva , Linfocitos T/inmunología , Replicación Viral/efectos de los fármacos
13.
Int J Mol Sci ; 22(4)2021 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-33562748

RESUMEN

Human adenoviruses (HAdVs) display a wide range of tissue tropism and can cause an array of symptoms from mild respiratory illnesses to disseminated and life-threatening infections in immunocompromised individuals. However, no antiviral drug has been approved specifically for the treatment of HAdV infections. Herein, we report our continued efforts to optimize salicylamide derivatives and discover compound 16 (JMX0493) as a potent inhibitor of HAdV infection. Compound 16 displays submicromolar IC50 values, a higher selectivity index (SI > 100) and 2.5-fold virus yield reduction compared to our hit compound niclosamide. Moreover, unlike niclosamide, our mechanistic studies suggest that the antiviral activity of compound 16 against HAdV is achieved through the inhibition of viral particle escape from the endosome, which bars subsequent uncoating and the presentation of lytic protein VI.


Asunto(s)
Adenovirus Humanos/fisiología , Antivirales/farmacología , Endosomas/virología , Niclosamida/farmacología , Salicilamidas/farmacología , Células A549 , Adenovirus Humanos/efectos de los fármacos , Descubrimiento de Drogas , Endosomas/efectos de los fármacos , Células HEK293 , Humanos , Concentración 50 Inhibidora , Niclosamida/química , Salicilamidas/química , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Tropismo Viral , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
14.
Virology ; 555: 102-110, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33032802

RESUMEN

Human adenovirus (HAdV) can cause severe disease in certain at-risk populations such as newborns, young children, the elderly and individuals with a compromised immune system. Unfortunately, no FDA-approved antiviraldrug is currently available for the treatment of HAdV infections. Within the nucleus of infected cells, the HAdV genome associates with histones and forms a chromatin-like structure during early infection, and viral gene expression appears to be regulated by cellular epigenetic processes. Thus, one potential therapeutic strategy to combat HAdV disease may be to target the cellular proteins involved in modifying the viral nucleoprotein structure and facilitating HAdV gene expression and replication. We have screened a panel of small molecules that modulate the activity of epigenetic regulatory proteins for compounds affecting HAdV gene expression. Several of the compounds, specifically chaetocin, gemcitabine and lestaurtinib, reduced HAdV recovery by 100- to 1000-fold, while showing limited effects on cell health, suggesting that these compounds may indeed be promising as anti-HAdV therapeutics.


Asunto(s)
Infecciones por Adenovirus Humanos/tratamiento farmacológico , Adenovirus Humanos/efectos de los fármacos , Antivirales/farmacología , Replicación Viral/efectos de los fármacos , Carbazoles/farmacología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Furanos/farmacología , Humanos , Piperazinas/farmacología , Gemcitabina
15.
Eur J Ophthalmol ; 31(2): 379-384, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31813297

RESUMEN

PURPOSE: The aim of this study was to test the antiviral effectivity of potassium peroxymonosulfate (RUBYSTA®, KYORIN) against five epidemic keratoconjunctivitis-related types of Human adenovirus D in vitro. METHODS: Five types of Human adenovirus D (8, 37, 53, 54 and 56) were incubated with 1% potassium peroxymonosulfate, 0.1% sodium hypochlorite (NaClO) or alcohol-based disinfectant for 30 s or 1 min. These solutions were subjected to measurements of viral titres by infection assays in A549 cells. At day 6 post-infection, both, supernatants and cells, were collected and the viral genome was assessed by real-time polymerase chain reaction analysis. RESULTS: Treatments with 1% potassium peroxymonosulfate led to significant reduction in all tested Human adenovirus D types comparable to disinfecting effects by 0.1% NaClO. Overall, potassium peroxymonosulfate demonstrated sufficient inactivation of the major epidemic keratoconjunctivitis-causing Human adenovirus D to be considered for disinfection and prevention purposes in ophthalmological clinics and hospitals. CONCLUSION: This study demonstrated that potassium peroxymonosulfate is a promising disinfectant for the prevention of epidemic keratoconjunctivitis nosocomial infections in ophthalmological clinics.


Asunto(s)
Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/efectos de los fármacos , Desinfectantes/farmacología , Queratoconjuntivitis/virología , Oxidantes/farmacología , Peróxidos/farmacología , Células A549 , Infección Hospitalaria/prevención & control , Epidemias , Humanos , Replicación Viral/efectos de los fármacos
16.
J Med Chem ; 63(21): 12830-12852, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33112138

RESUMEN

An effective therapy for human adenovirus (HAdV) infections in immunocompromised patients and healthy individuals with community-acquired pneumonia remains an unmet medical need. We herein reported a series of novel substituted N-(4-amino-2-chlorophenyl)-5-chloro-2-hydroxybenzamide analogues as potent HAdV inhibitors. Compounds 6, 15, 29, 40, 43, 46, 47, and 54 exhibited increased selectivity indexes (SI > 100) compared to the lead compound niclosamide, while maintaining sub-micromolar to low micromolar potency against HAdV. The preliminary mechanistic studies indicated that compounds 6 and 43 possibly target the HAdV DNA replication process, while compounds 46 and 47 suppress later steps of HAdV life cycle. Notably, among these derivatives, compound 15 showed improved anti-HAdV activity (IC50 = 0.27 µM), significantly decreased cytotoxicity (CC50 = 156.8 µM), and low in vivo toxicity (maximum tolerated dose = 150 mg/kg in hamster) as compared with niclosamide, supporting its further in vivo efficacy studies for the treatment of HAdV infections.


Asunto(s)
Adenovirus Humanos/fisiología , Antivirales/química , Benzamidas/química , Adenovirus Humanos/efectos de los fármacos , Animales , Antivirales/síntesis química , Antivirales/farmacología , Benzamidas/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cricetinae , Evaluación Preclínica de Medicamentos , Humanos , Dosificación Letal Mediana , Relación Estructura-Actividad , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
17.
ACS Chem Biol ; 15(10): 2683-2691, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-32845119

RESUMEN

Coxsackievirus A24 variant (CVA24v) and human adenovirus 37 (HAdV-37) are leading causative agents of the severe and highly contagious ocular infections acute hemorrhagic conjunctivitis and epidemic keratoconjunctivitis, respectively. Currently, neither vaccines nor antiviral agents are available for treating these diseases, which affect millions of individuals worldwide. CVA24v and HAdV-37 utilize sialic acid as attachment receptors facilitating entry into host cells. Previously, we and others have shown that derivatives based on sialic acid are effective in preventing HAdV-37 binding and infection of cells. Here, we designed and synthesized novel pentavalent sialic acid conjugates and studied their inhibitory effect against CVA24v and HAdV-37 binding and infection of human corneal epithelial cells. The pentavalent conjugates are the first reported inhibitors of CVA24v infection and proved efficient in blocking HAdV-37 binding. Taken together, the pentavalent conjugates presented here form a basis for the development of general inhibitors of these highly contagious ocular pathogens.


Asunto(s)
Adenovirus Humanos/efectos de los fármacos , Antivirales/farmacología , Enterovirus Humano C/efectos de los fármacos , Ácidos Siálicos/farmacología , Adenovirus Humanos/química , Sitios de Unión , Enterovirus Humano C/química , Humanos , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos
18.
Antiviral Res ; 182: 104872, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32768412

RESUMEN

OBJECTIVES: Human adenovirus (HAdV) infections are associated with a high morbidity and mortality in transplant patients requiring the use of antiviral treatments. Brincidofovir (BCV), a cytidine analog, inhibits HAdV replication through viral DNA elongation termination and likely through other mechanisms. To elucidate if BCV regulates cellular antiviral pathways, we analyzed its impact on HAdV-infected and non-HAdV-infected lung epithelial cells. METHODS: We assessed the cellular and viral transcriptome of A549 cells infected and non-infected with HAdV C5 and treated or non-treated with BCV by RNAseq after 72 h. RESULTS: BCV treatment of HAdV infected cells resulted in a profound decrease of viral transcription associated with a relative overexpression of the early genes E1A and E4 and of the late gene L1. BCV had also a profound impact on A549 cells' transcriptome. Ontologic analysis revealed an effect of BCV on several pathways known to interact with adenovirus replication as mTor signalling and Wnt pathways. A549 cells treated with BCV demonstrated a significant inhibition of the biological function of "viral replication" including 25 dysregulated genes involved in inflammation pathways. CONCLUSION: We demonstrated that BCV alters viral gene expression and promotes the expression of antiviral cellular pathways in A549 cells. These results provide new insights how to interfere with cellular pathways to control HAdV infections.


Asunto(s)
Adenovirus Humanos/efectos de los fármacos , Antivirales/farmacología , Citosina/análogos & derivados , Organofosfonatos/farmacología , Transcriptoma , Células A549 , Citosina/farmacología , Interacciones Microbiota-Huesped , Humanos , Replicación Viral/efectos de los fármacos
19.
Sci Data ; 7(1): 265, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32788590

RESUMEN

Human adenoviruses (HAdVs) are fatal to immuno-suppressed individuals, but no effective anti-HAdV therapy is available. Here, we present a novel image-based high-throughput screening (HTS) platform, which scores the full viral replication cycle from virus entry to dissemination of progeny and second-round infections. We analysed 1,280 small molecular weight compounds of the Prestwick Chemical Library (PCL) for interference with HAdV-C2 infection in a quadruplicate, blinded format, and performed robust image analyses and hit filtering. We present the entire set of the screening data including all images, image analyses and data processing pipelines. The data are made available at the Image Data Resource (IDR, idr0081). Our screen identified Nelfinavir mesylate as an inhibitor of HAdV-C2 multi-round plaque formation, but not single round infection. Nelfinavir has been FDA-approved for anti-retroviral therapy in humans. Our results underscore the power of image-based full cycle infection assays in identifying viral inhibitors with clinical potential.


Asunto(s)
Adenovirus Humanos/efectos de los fármacos , Antivirales/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Adenovirus Humanos/fisiología , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Nelfinavir/farmacología , Replicación Viral/efectos de los fármacos
20.
J Virol ; 94(18)2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32641484

RESUMEN

Human adenoviruses (HAdV) are ubiquitous within the human population and comprise a significant burden of respiratory illnesses worldwide. Pediatric and immunocompromised individuals are at particular risk for developing severe disease; however, no approved antiviral therapies specific to HAdV exist. Ivermectin is an FDA-approved broad-spectrum antiparasitic drug that also exhibits antiviral properties against a diverse range of viruses. Its proposed function is inhibiting the classical protein nuclear import pathway mediated by importin-α (Imp-α) and -ß1 (Imp-ß1). Many viruses, including HAdV, rely on this host pathway for transport of viral proteins across the nuclear envelope. In this study, we show that ivermectin inhibits HAdV-C5 early gene transcription, early and late protein expression, genome replication, and production of infectious viral progeny. Similarly, ivermectin inhibits genome replication of HAdV-B3, a clinically important pathogen responsible for numerous recent outbreaks. Mechanistically, we show that ivermectin disrupts binding of the viral E1A protein to Imp-α without affecting the interaction between Imp-α and Imp-ß1. Our results further extend ivermectin's broad antiviral activity and provide a mechanistic underpinning for its mode of action as an inhibitor of cellular Imp-α/ß1-mediated nuclear import.IMPORTANCE Human adenoviruses (HAdVs) represent a ubiquitous and clinically important pathogen without an effective antiviral treatment. HAdV infections typically cause mild symptoms; however, individuals such as children, those with underlying conditions, and those with compromised immune systems can develop severe disseminated disease. Our results demonstrate that ivermectin, an FDA-approved antiparasitic agent, is effective at inhibiting replication of several HAdV types in vitro This is in agreement with the growing body of literature suggesting ivermectin has broad antiviral activity. This study expands our mechanistic knowledge of ivermectin by showing that ivermectin targets the ability of importin-α (Imp-α) to recognize nuclear localization sequences, without effecting the Imp-α/ß1 interaction. These data also exemplify the applicability of targeting host factors upon which viruses rely as a viable antiviral strategy.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Adenovirus Humanos/efectos de los fármacos , Antiparasitarios/farmacología , Ivermectina/farmacología , Replicación Viral/efectos de los fármacos , alfa Carioferinas/genética , beta Carioferinas/genética , Células A549 , Transporte Activo de Núcleo Celular/genética , Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Adenovirus Humanos/patogenicidad , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/virología , Citosol/efectos de los fármacos , Citosol/metabolismo , Citosol/virología , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Regulación de la Expresión Génica , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Transducción de Señal , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Proteínas Virales/metabolismo , alfa Carioferinas/antagonistas & inhibidores , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA