Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.142
Filtrar
1.
Virol J ; 21(1): 95, 2024 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664855

RESUMEN

BACKGROUND: African swine fever virus (ASFV) is a major threat to pig production and the lack of effective vaccines underscores the need to develop robust antiviral countermeasures. Pathologically, a significant elevation in pro-inflammatory cytokine production is associated with ASFV infection in pigs and there is high interest in identifying dual-acting natural compounds that exhibit antiviral and anti-inflammatory activities. METHODS: Using the laboratory-adapted ASFV BA71V strain, we screened a library of 297 natural, anti-inflammatory compounds to identify promising candidates that protected Vero cells against virus-induced cytopathic effect (CPE). Virus yield reduction, virucidal, and cell cytotoxicity experiments were performed on positive hits and two lead compounds were further characterized in dose-dependent assays along with time-of-addition, time-of-removal, virus entry, and viral protein synthesis assays. The antiviral effects of the two lead compounds on mitigating virulent ASFV infection in porcine macrophages (PAMs) were also tested using similar methods, and the ability to inhibit pro-inflammatory cytokine production during virulent ASFV infection was assessed by enzyme-linked immunosorbent assay (ELISA). RESULTS: The screen identified five compounds that inhibited ASFV-induced CPE by greater than 50% and virus yield reduction experiments showed that two of these compounds, tetrandrine and berbamine, exhibited particularly high levels of anti-ASFV activity. Mechanistic analysis confirmed that both compounds potently inhibited early stages of ASFV infection and that the compounds also inhibited infection of PAMs by the virulent ASFV Arm/07 isolate. Importantly, during ASFV infection in PAM cells, both compounds markedly reduced the production of pro-inflammatory cytokines involved in disease pathogenesis while tetrandrine had a greater and more sustained anti-inflammatory effect than berbamine. CONCLUSIONS: Together, these findings support that dual-acting natural compounds with antiviral and anti-inflammatory properties hold promise as preventative and therapeutic agents to combat ASFV infection by simultaneously inhibiting viral replication and reducing virus-induced cytokine production.


Asunto(s)
Virus de la Fiebre Porcina Africana , Antiinflamatorios , Antivirales , Animales , Virus de la Fiebre Porcina Africana/efectos de los fármacos , Virus de la Fiebre Porcina Africana/fisiología , Antivirales/farmacología , Porcinos , Antiinflamatorios/farmacología , Chlorocebus aethiops , Células Vero , Macrófagos/efectos de los fármacos , Macrófagos/virología , Macrófagos/inmunología , Fiebre Porcina Africana/virología , Replicación Viral/efectos de los fármacos , Productos Biológicos/farmacología , Evaluación Preclínica de Medicamentos , Efecto Citopatogénico Viral/efectos de los fármacos , Citocinas/metabolismo , Internalización del Virus/efectos de los fármacos
2.
IUBMB Life ; 74(1): 93-100, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34390301

RESUMEN

Unfolded protein response (UPR) and endoplasmic reticulum (ER) stress are aspects of SARS-CoV-2-host cell interaction with proposed role in the cytopathic and inflammatory pathogenesis of this viral infection. The role of the NF-kB pathway in these cellular processes remains poorly characterized. When investigated in VERO-E6 cells, SARS-CoV-2 infection was found to markedly stimulate NF-kB protein expression and activity. NF-kB activation occurs early in the infection process (6 hpi) and it is associated with increased MAPK signaling and expression of the UPR inducer IRE-1α. These signal transduction processes characterize the cellular stress response to the virus promoting a pro-inflammatory environment and caspase activation in the host cell. Inhibition of viral replication by the viral protease inhibitor Nelfinavir reverts all these molecular changes also stimulating c-Jun expression, a key component of the JNK/AP-1 pathway with important role in the IRE-1α-mediated transcriptional regulation of stress response genes with anti-inflammatory and cytoprotection function. The present study demonstrates that UPR signaling and its interaction with cellular MAPKs and the NF-kB activity are important aspects of SARS-CoV-2-host cell interaction that deserve further investigation to identify more efficient therapies for this viral infection.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , COVID-19/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , FN-kappa B/metabolismo , SARS-CoV-2 , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Animales , COVID-19/virología , Caspasa 9/metabolismo , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Modelos Biológicos , Nelfinavir/farmacología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , Respuesta de Proteína Desplegada/efectos de los fármacos , Células Vero
3.
Cells ; 10(11)2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34831176

RESUMEN

Human coronavirus (HCoV) similar to other viruses rely on host cell machinery for both replication and to spread. The p97/VCP ATPase is associated with diverse pathways that may favor HCoV replication. In this study, we assessed the role of p97 and associated host responses in human lung cell line H1299 after HCoV-229E or HCoV-OC43 infection. Inhibition of p97 function by small molecule inhibitors shows antiviral activity, particularly at early stages of the virus life cycle, during virus uncoating and viral RNA replication. Importantly, p97 activity inhibition protects human cells against HCoV-induced cytopathic effects. The p97 knockdown also inhibits viral production in infected cells. Unbiased quantitative proteomics analyses reveal that HCoV-OC43 infection resulted in proteome changes enriched in cellular senescence and DNA repair during virus replication. Further analysis of protein changes between infected cells with control and p97 shRNA identifies cell cycle pathways for both HCoV-229E and HCoV-OC43 infection. Together, our data indicate a role for the essential host protein p97 in supporting HCoV replication, suggesting that p97 is a therapeutic target to treat HCoV infection.


Asunto(s)
Coronavirus Humano 229E/fisiología , Coronavirus Humano OC43/fisiología , Proteína que Contiene Valosina/metabolismo , Replicación Viral/fisiología , Antivirales/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular , Coronavirus Humano 229E/efectos de los fármacos , Coronavirus Humano OC43/efectos de los fármacos , Efecto Citopatogénico Viral/efectos de los fármacos , Humanos , Proteoma/efectos de los fármacos , Proteoma/metabolismo , ARN Interferente Pequeño/genética , ARN Viral/biosíntesis , Proteína que Contiene Valosina/antagonistas & inhibidores , Proteína que Contiene Valosina/genética , Replicación Viral/efectos de los fármacos , Desencapsidación Viral/efectos de los fármacos
4.
Mol Immunol ; 140: 106-119, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34678620

RESUMEN

The recent advances in our understanding of the host factors in orchestrating qualitatively different immune responses against influenza Type A virus (IAV) have changed the perception of conventional approaches for controlling avian influenza virus (AIV) infection in chickens. Given that infection-induced pathogenicity and replication of influenza virus largely rely on regulating host immune responses, immunoregulatory cytokine profiles often determine the disease outcomes. However, in contrast to the function of other inflammatory cytokines, interleukin-17A (IL-17A) has been described as a 'double-edged sword', indicating that in addition to antiviral host responses, IL-17A has a distinct role in promoting viral infection. Therefore, in the present study, we investigated the chicken IL-17A mediated antiviral immune effects on IAVs infection in primary chicken embryo fibroblasts cells (CEFs). To this end, we first bioengineered a food-grade Lactic Acid Producing Bacteria (LAB), Lactococcus lactis (L. lactis), secreting bioactive recombinant chicken IL-17A (sChIL-17A). Next, the functionality of sChIL-17A was confirmed by transcriptional upregulation of several genes associated with antiviral host responses, including granulocyte-monocyte colony-stimulating factor (GM-CSF) (CSF3 in the chickens), interleukin-6 (IL-6), interferon-α (IFN-α), -ß and -γ genes in primary CEFs cells. Consistent with our hypothesis that such a pro-inflammatory state may translate to immunoprotection against IAVs infection, we observed that sChIL-17A pre-treatment could significantly limit the viral replication and protect the primary CEFs cells against two heterotypic IAVs such as A/turkey/Wisconsin/1/1966(H9N2) and A/PR/8/1934(H1N1). Together, the data presented in this work suggest that exogenous application of sChIL-17A secreted by modified LAB vector may represent an alternative strategy for improving antiviral immunity against avian influenza virus infection in chickens.


Asunto(s)
Bioingeniería , Citoprotección , Fibroblastos/virología , Vectores Genéticos/metabolismo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H9N2 del Virus de la Influenza A/fisiología , Interleucina-17/farmacología , Lactobacillales/genética , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Pollos/virología , Efecto Citopatogénico Viral/efectos de los fármacos , Citoprotección/efectos de los fármacos , Perros , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Perfilación de la Expresión Génica , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H9N2 del Virus de la Influenza A/genética , Gripe Aviar/inmunología , Gripe Aviar/virología , Interleucina-17/genética , Células de Riñón Canino Madin Darby , Nisina/farmacología , Fenotipo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/farmacología , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
5.
Cell Mol Life Sci ; 78(23): 7777-7794, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34714362

RESUMEN

The COVID-19 pandemic caused by SARS-CoV-2 requires new treatments both to alleviate the symptoms and to prevent the spread of this disease. Previous studies demonstrated good antiviral and virucidal activity of phospholipase A2s (PLA2s) from snake venoms against viruses from different families but there was no data for coronaviruses. Here we show that PLA2s from snake venoms protect Vero E6 cells against SARS-CoV-2 cytopathic effects. PLA2s showed low cytotoxicity to Vero E6 cells with some activity at micromolar concentrations, but strong antiviral activity at nanomolar concentrations. Dimeric PLA2 from the viper Vipera nikolskii and its subunits manifested especially potent virucidal effects, which were related to their phospholipolytic activity, and inhibited cell-cell fusion mediated by the SARS-CoV-2 spike glycoprotein. Moreover, PLA2s interfered with binding both of an antibody against ACE2 and of the receptor-binding domain of the glycoprotein S to 293T/ACE2 cells. This is the first demonstration of a detrimental effect of PLA2s on ß-coronaviruses. Thus, snake PLA2s are promising for the development of antiviral drugs that target the viral envelope, and could also prove to be useful tools to study the interaction of viruses with host cells.


Asunto(s)
Fosfolipasas A2/farmacología , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/metabolismo , Venenos de Víboras/farmacología , Acoplamiento Viral/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Afinidad de Anticuerpos/efectos de los fármacos , Antivirales/farmacología , Fusión Celular , Línea Celular , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Células HEK293 , Humanos , Modelos Moleculares , Dominios Proteicos/efectos de los fármacos , Resonancia por Plasmón de Superficie , Células Vero , Venenos de Víboras/enzimología , Tratamiento Farmacológico de COVID-19
6.
Viruses ; 13(9)2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34578432

RESUMEN

Human noroviruses (HuNoVs) are the most common cause of viral gastroenteritis resulting annually in ~219,000 deaths and a societal cost of ~USD 60 billion, and no antivirals or vaccines are available. Here, we assess the anti-norovirus activity of new peptidomimetic aldehydes related to the protease inhibitor rupintrivir. The early hit compound 4 inhibited the replication of murine norovirus (MNV) and the HuNoV GI.1 replicon in vitro (EC50 ~1 µM) and swiftly cleared the HuNoV GI.1 replicon from the cells. Compound 4 still inhibits the proteolytic activity. We selected a resistant GI.1 replicon, with a mutation (I109V) in a highly conserved region of the viral protease, conferring a low yield of resistance against compound 4 and rupintrivir. After testing new derivatives, compound 10d was the most potent (EC50 nanomolar range). Molecular docking indicated that the aldehyde group of compounds 4 and 10d bind with Cys139 in the HuNoV 3CL protease by a covalent linkage. Finally, compound 10d inhibited the replication of HuNoV GII.4 in infected zebrafish larvae, and PK studies in mice showed an adequate profile.


Asunto(s)
Infecciones por Caliciviridae/tratamiento farmacológico , Norovirus/efectos de los fármacos , Inhibidores de Proteasa Viral/farmacología , Animales , Infecciones por Caliciviridae/virología , Línea Celular , Efecto Citopatogénico Viral/efectos de los fármacos , Farmacorresistencia Viral , Isoxazoles/farmacología , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Mutación , Norovirus/enzimología , Norovirus/genética , Norovirus/fisiología , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Pirrolidinonas/farmacología , ARN Viral/metabolismo , Replicón , Bibliotecas de Moléculas Pequeñas , Valina/análogos & derivados , Valina/farmacología , Inhibidores de Proteasa Viral/química , Inhibidores de Proteasa Viral/farmacocinética , Replicación Viral/efectos de los fármacos , Pez Cebra/virología
7.
J Med Chem ; 64(19): 14465-14476, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34549580

RESUMEN

In this work, a series of novel substituted polycyclic pyridone derivatives were designed and synthesized as potent anti-influenza agents. The cytopathic effect (CPE) assay and cytotoxicity assay indicated that all of the compounds possessed potent anti-influenza virus activity and relatively low cytotoxicity; some of them inhibited the replication of influenza A virus (IAV) at picomolar concentrations. Further studies revealed that, at a concentration of 3 nM, three compounds (10a, 10d, and 10g) could significantly reduce the M2 RNA amounts and M2 protein expression of IAV and inhibit the activity of RNA-dependent RNA polymerase (RdRp). Among them, (R)-12-(5H-dibenzo[a,d][7]annulen-5-yl)-7-hydroxy-3,4,12,12a-tetrahydro-1H-[1,4]oxazino[3,4-c]pyrido[2,1-f][1,2,4]triazine-6,8-dione (10a) was found to be a promising anti-influenza drug candidate with good human liver microsomal stability, as well as with better selectivity index and oral bioavailability than Baloxavir.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Dibenzotiepinas/química , Virus de la Influenza A/efectos de los fármacos , Morfolinas/química , Piridonas/síntesis química , Piridonas/farmacología , Triazinas/química , Animales , Supervivencia Celular/efectos de los fármacos , Efecto Citopatogénico Viral/efectos de los fármacos , Perros , Humanos , Células de Riñón Canino Madin Darby , Masculino , Piridonas/química , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Viruses ; 13(8)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34452529

RESUMEN

An escalating pandemic of the novel SARS-CoV-2 virus is impacting global health, and effective antivirals are needed. Umifenovir (Arbidol) is an indole-derivative molecule, licensed in Russia and China for prophylaxis and treatment of influenza and other respiratory viral infections. It has been shown that umifenovir has broad spectrum activity against different viruses. We evaluated the sensitivity of different coronaviruses, including the novel SARS-CoV-2 virus, to umifenovir using in vitro assays. Using a plaque assay, we revealed an antiviral effect of umifenovir against seasonal HCoV-229E and HCoV-OC43 coronaviruses in Vero E6 cells, with estimated 50% effective concentrations (EC50) of 10.0 ± 0.5 µM and 9.0 ± 0.4 µM, respectively. Umifenovir at 90 µM significantly suppressed plaque formation in CMK-AH-1 cells infected with SARS-CoV. Umifenovir also inhibited the replication of SARS-CoV-2 virus, with EC50 values ranging from 15.37 ± 3.6 to 28.0 ± 1.0 µM. In addition, 21-36 µM of umifenovir significantly suppressed SARS-CoV-2 virus titers (≥2 log TCID50/mL) in the first 24 h after infection. Repurposing of antiviral drugs is very helpful in fighting COVID-19. A safe, pan-antiviral drug such as umifenovir could be extremely beneficial in combating the early stages of a viral pandemic.


Asunto(s)
Antivirales/farmacología , Coronavirus Humano 229E/efectos de los fármacos , Coronavirus Humano OC43/efectos de los fármacos , Indoles/farmacología , SARS-CoV-2/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Animales , Antivirales/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Coronavirus Humano 229E/fisiología , Coronavirus Humano OC43/fisiología , Efecto Citopatogénico Viral/efectos de los fármacos , Humanos , Indoles/administración & dosificación , Pruebas de Sensibilidad Microbiana , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , SARS-CoV-2/fisiología , Células Vero , Carga Viral/efectos de los fármacos , Ensayo de Placa Viral , Replicación Viral/efectos de los fármacos
9.
Dev Comp Immunol ; 124: 104200, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34237380

RESUMEN

Frog virus 3 (FV3) causes mortality in a range of amphibian species. Despite the importance of the skin epithelium as a first line of defence against FV3, the interaction between amphibian skin epithelial cells and FV3 remains largely uncharacterized. Here, we used newly established Xenopus laevis skin epithelial-like cell lines, Xela DS2 and Xela VS2, to study the susceptibility and permissiveness of frog skin epithelial cells to FV3, and the innate immune antiviral and proinflammatory gene regulatory responses of these cells to FV3. Both cell lines are susceptible and permissive to FV3, yet do not exhibit appreciable transcript levels of scavenger receptors thought to be used by FV3 for cellular entry. Xela DS2 and Xela VS2 upregulate antiviral and proinflammatory cytokine transcripts in response to poly(I:C) but not to FV3 or UV-inactivated FV3. Poly(I:C) pretreatment limits FV3 replication and FV3-induced cytopathic effects in both cell lines. Thus, Xela DS2 and Xela VS2 can support FV3 replication, represent in vitro systems to investigate antiviral responses of frog skin epithelial cells, and can serve as novel tools for screening compounds that initiate effective antiviral programs to limit FV3 replication.


Asunto(s)
Factores de Restricción Antivirales/inmunología , Células Epiteliales/virología , Ranavirus/fisiología , Piel/citología , Replicación Viral/inmunología , Animales , Línea Celular , Citocinas/inmunología , Efecto Citopatogénico Viral/efectos de los fármacos , Efecto Citopatogénico Viral/inmunología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Inmunidad Innata , Poli I-C/farmacología , Replicación Viral/efectos de los fármacos , Xenopus laevis
10.
Emerg Microbes Infect ; 10(1): 1378-1389, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34162308

RESUMEN

The vector-borne flaviviruses (VBFVs) are well known for causing great misery and death in humans worldwide. The VBFVs include those transmitted by mosquitos, such as Zika virus (ZIKV), dengue virus; and those transmitted by ticks including the tick-borne flavivirus serocomplex and Powassan virus (POWV). Two of our recent reports showed that intracranial POWV infection in the reservoir host, Peromyscus leucopus, was restricted and caused no overt clinical disease. Several modes of analyses suggested activation of the LXR pathway. Activation of the LXR pathway leads to increased efflux of cholesterol from cells and consequent disturbances in membrane biogenesis. Because VBFV replication is dependent on membrane biogenesis, we evaluated the effect of an LXR agonist (LXR623) on POWV and ZIKV infection and observed that the compound impaired permissive replication of both viruses in a human neuroblastoma SK-N-SH cell line. The LXR agonist resulted in failure of the viruses to induce ER expansion and elaborate vesicle formation, suggesting that the efflux of cholesterol was part of the antiviral mechanism. We also observed that the LXR agonist contributed to the mechanism of virus suppression by increased expression of mRNAs encoding for the antiviral cytokines CXCL10, RANTES and IFN1ß. In sharp contrast, a LXR antagonist (GSK2033) had no significant effect on VBFV replication. We conclude that LXR623 impairs flavivirus replication by stimulating cellular antiviral factors.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas/efectos de los fármacos , Indazoles/farmacología , Receptores X del Hígado/agonistas , Virus Zika/efectos de los fármacos , Antivirales/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Efecto Citopatogénico Viral/efectos de los fármacos , Vesículas Citoplasmáticas/efectos de los fármacos , Vesículas Citoplasmáticas/metabolismo , Virus de la Encefalitis Transmitidos por Garrapatas/fisiología , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Humanos , Receptores X del Hígado/metabolismo , Replicación Viral/efectos de los fármacos , Virus Zika/fisiología
11.
Molecules ; 26(10)2021 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-34069874

RESUMEN

Since herpes simplex virus type 1 (HSV-1) infection is so widespread, several antiviral drugs have been developed to treat it, among which are uracil nucleosides. However, there are major problems with the current medications such as severe side-effects and drug resistance. Here we present some newly synthesized cyclic and acyclic uracil nucleosides that showed very promising activity against HSV-1 compared to acyclovir.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Uridina/síntesis química , Uridina/farmacología , Aciclovir/farmacología , Animales , Antivirales/química , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Estándares de Referencia , Relación Estructura-Actividad , Uridina/química , Células Vero
12.
Virol J ; 18(1): 123, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34107996

RESUMEN

BACKGROUND: The international SARS-CoV-2 pandemic has resulted in an urgent need to identify new anti-viral drugs for treatment of COVID-19. The initial step to identifying potential candidates usually involves in vitro screening that includes standard cytotoxicity controls. Under-appreciated is that viable, but stressed or otherwise compromised cells, can also have a reduced capacity to replicate virus. A refinement proposed herein for in vitro drug screening thus includes a simple growth assay to identify drug concentrations that cause cellular stress or "cytomorbidity", as distinct from cytotoxicity or loss of viability. METHODS: A simple rapid bioassay is presented for antiviral drug screening using Vero E6 cells and inhibition of SARS-CoV-2 induced cytopathic effects (CPE) measured using crystal violet staining. We use high cell density for cytotoxicity assays, and low cell density for cytomorbidity assays. RESULTS: The assay clearly illustrated the anti-viral activity of remdesivir, a drug known to inhibit SARS-CoV-2 replication. In contrast, nitazoxanide, oleuropein, cyclosporine A and ribavirin all showed no ability to inhibit SARS-CoV-2 CPE. Hydroxychloroquine, cyclohexamide, didemnin B, γ-mangostin and linoleic acid were all able to inhibit viral CPE at concentrations that did not induce cytotoxicity. However, these drugs inhibited CPE at concentrations that induced cytomorbidity, indicating non-specific anti-viral activity. CONCLUSIONS: We describe the methodology for a simple in vitro drug screening assay that identifies potential anti-viral drugs via their ability to inhibit SARS-CoV-2-induced CPE. The additional growth assay illustrated how several drugs display anti-viral activity at concentrations that induce cytomorbidity. For instance, hydroxychloroquine showed anti-viral activity at concentrations that slow cell growth, arguing that its purported in vitro anti-viral activity arises from non-specific impairment of cellular activities. The cytomorbidity assay can therefore rapidly exclude potential false positives.


Asunto(s)
Antivirales/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/química , Bioensayo , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Concentración 50 Inhibidora , Células Vero , Replicación Viral/efectos de los fármacos
13.
Viruses ; 13(4)2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33810356

RESUMEN

The 2019 coronavirus infectious disease (COVID-19) is caused by infection with the new severe acute respiratory syndrome coronavirus (SARS-CoV-2). Currently, the treatment options for COVID-19 are limited. The purpose of the experiments presented here was to investigate the effectiveness of ketotifen, naproxen and indomethacin, alone or in combination, in reducing SARS-CoV-2 replication. In addition, the cytotoxicity of the drugs was evaluated. The findings showed that the combination of ketotifen with indomethacin (SJP-002C) or naproxen both reduce viral yield. Compared to ketotifen alone (60% inhibition at EC50), an increase in percentage inhibition of SARS-CoV-2 to 79%, 83% and 93% was found when co-administered with 25, 50 and 100 µM indomethacin, respectively. Compared to ketotifen alone, an increase in percentage inhibition of SARS-CoV-2 to 68%, 68% and 92% was found when co-administered with 25, 50 and 100 µM naproxen, respectively. For both drug combinations the observations suggest an additive or synergistic effect, compared to administering the drugs alone. No cytotoxic effects were observed for the administered dosages of ketotifen, naproxen, and indomethacin. Further research is warranted to investigate the efficacy of the combination of ketotifen with indomethacin (SJP-002C) or naproxen in the treatment of SARS-CoV-2 infection in humans.


Asunto(s)
Antivirales/farmacología , COVID-19/virología , Indometacina/farmacología , Cetotifen/farmacología , Naproxeno/farmacología , SARS-CoV-2/efectos de los fármacos , Efecto Citopatogénico Viral/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Tratamiento Farmacológico de COVID-19
14.
Arch Virol ; 166(4): 1203-1211, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33606112

RESUMEN

Lactoferrin is part of the innate immune system, with antiviral activity against numerous DNA and RNA viruses. Rhinoviruses, the leading cause of the common cold, are associated with exacerbation of respiratory illnesses such as asthma. Here, we explored the effect of bovine lactoferrin (BLf) on RV-B14 infectivity. Using different assays, we show that the effect of BLf is strongest during adhesion of the virus to the cell and entry. Tracking the internalisation of BLf and virus revealed a degree of colocalisation, although their interaction was only confirmed in vitro using empty viral particles, indicating a possible additional influence of BLf on other infection steps.


Asunto(s)
Antivirales/farmacología , Enterovirus/efectos de los fármacos , Lactoferrina/farmacología , Internalización del Virus/efectos de los fármacos , Antivirales/metabolismo , Supervivencia Celular/efectos de los fármacos , Efecto Citopatogénico Viral/efectos de los fármacos , Enterovirus/fisiología , Infecciones por Enterovirus/virología , Células HeLa , Humanos , Lactoferrina/metabolismo , Acoplamiento Viral/efectos de los fármacos
15.
J Gen Virol ; 102(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33416468

RESUMEN

Chios mastic gum (CMG), a resin of the mastic tree (Pistacia lentiscus var. chia), has been used to treat multiple disorders caused by gastrointestinal malfunctions and bacterial infections for more than 2500 years. However, little is known about CMG's antiviral activity. CMG is known to influence multiple cellular processes such as cell proliferation, differentiation and apoptosis. As virus replication is largely dependent on the host cellular metabolism, it is conceivable that CMG regulates virus infectivity. Therefore, in this study, we evaluated CMG's potential as an antiviral drug to treat influenza A virus (IAV) infection. CMG treatment dramatically reduced the cytopathogenic effect and production of RNAs, proteins and infectious particles of IAV. Interestingly, CMG interfered with the early stage of the virus life cycle after viral attachment. Importantly, the administration of CMG greatly ameliorated morbidity and mortality in IAV-infected mice. The results suggest that CMG displays a potent anti-IAV activity by blocking the early stage of viral replication. Thus, mastic gum could be exploited as a novel therapeutic agent against IAV infection.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Resina Mástique/farmacología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Efecto Citopatogénico Viral/efectos de los fármacos , Perros , Células HEK293 , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Células de Riñón Canino Madin Darby , Resina Mástique/uso terapéutico , Infecciones por Orthomyxoviridae/virología , Virulencia/efectos de los fármacos , Acoplamiento Viral , Replicación Viral/efectos de los fármacos
16.
Nat Prod Res ; 35(6): 1014-1018, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31496276

RESUMEN

The antiviral effect against RVA in cell culture was evaluated by using an aqueous extract of Patallus mollis sea cucumber, applying the titration methodology. This technique is used to measures the ability of the extract dilutions to inhibit the cytopathic effect (CPE) of the virus, expressed as percentage of inhibition (IP). The mean extract cytotoxic concentration (CC50) used in the antiviral assay was 27,042.10 µg/mL and the PI of the antiviral activity extract was greater than 99.9% for each concentration. To determine the viral action mode, the cells were previously treated with the extracts in different stages during the viral infection cycle. The result analysis suggests that the extract inhibits 99% of the virus during the absorption and viral inactivation phase. These results show the P. mollis extract has a remarkable antiviral effect against the RVA in cell culture. So that, it is crucial to investigate its action mechanisms.


Asunto(s)
Antivirales/farmacología , Rotavirus/efectos de los fármacos , Pepinos de Mar/anatomía & histología , Animales , Efecto Citopatogénico Viral/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Rotavirus/fisiología , Metabolismo Secundario/efectos de los fármacos , Replicación Viral/efectos de los fármacos
17.
J Antimicrob Chemother ; 76(2): 413-417, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33164048

RESUMEN

BACKGROUND: As the causative agent of COVID-19, SARS-CoV-2 is a pathogen of immense importance to global public health. Development of innovative direct-acting antiviral agents is sorely needed to address this virus. Peptide-conjugated morpholino oligomers (PPMO) are antisense compounds composed of a phosphorodiamidate morpholino oligomer covalently conjugated to a cell-penetrating peptide. PPMO require no delivery assistance to enter cells and are able to reduce expression of targeted RNA through sequence-specific steric blocking. METHODS: Five PPMO designed against sequences of genomic RNA in the SARS-CoV-2 5'-untranslated region and a negative control PPMO of random sequence were synthesized. Each PPMO was evaluated for its effect on the viability of uninfected cells and its inhibitory effect on the replication of SARS-CoV-2 in Vero-E6 cell cultures. Cell viability was evaluated with an ATP-based method using a 48 h PPMO treatment time. Viral growth was measured with quantitative RT-PCR and TCID50 infectivity assays from experiments where cells received a 5 h PPMO treatment time. RESULTS: PPMO designed to base-pair with sequence in the 5' terminal region or the leader transcription regulatory sequence region of SARS-CoV-2 genomic RNA were highly efficacious, reducing viral titres by up to 4-6 log10 in cell cultures at 48-72 h post-infection, in a non-toxic and dose-responsive manner. CONCLUSIONS: The data indicate that PPMO have the ability to potently and specifically suppress SARS-CoV-2 growth and are promising candidates for further preclinical development.


Asunto(s)
Antivirales/farmacología , COVID-19/virología , Péptidos de Penetración Celular/farmacología , Morfolinos/farmacología , SARS-CoV-2/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/química , Supervivencia Celular/efectos de los fármacos , Péptidos de Penetración Celular/química , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Morfolinos/química , SARS-CoV-2/genética , Células Vero
18.
Virus Res ; 292: 198249, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33253717

RESUMEN

The channel catfish virus (CCV) can cause lethal hemorrhagic infection in channel catfish, resulting in significant economic losses in the fish industry. Effective drugs for the virus are still lacking. Acyclovir is known as a potent antiviral agent against human herpes viruses and some animal DNA viruses. The present study was undertaken to explore the antiviral response and mechanism of acyclovir against CCV in channel catfish ovary (CCO) cells. Acyclovir was able to significantly inhibit the expression of viral genes related to CCV viral DNA synthesis and suppress viral replication at a safe concentration. Furthermore, acyclovir blocked the cytopathic effects and apoptosis induced by CCV, thereby maintaining the normal cellular morphological structure, as shown by the protection of CCO cells from the formation of apoptotic bodies or nuclear fragmentation. Moreover, reverse transcript quantitative polymerase chain reaction (RT-qPCR) demonstrated that acyclovir suppressed the expression of caspase 3, caspase 8 and caspase 9, while there was no significant impact on the expression of the apoptosis-inhibiting gene bcl-2 in CCV-infected cells. In addition, acyclovir did not promote the expression of immune-related genes such as MyD88, Mx1, IRF3, IRF7, IFN-I, NF-kB and IL-1ß, suggesting that the antiviral activity of acyclovir to CCV infection is not achieved by facilitating the expression of immune-related genes in CCO cells. Taken together, the results from this study suggest that acyclovir could effectively regulate CCV-induced infection, and thus is a promising therapeutic agent against CCV. Our results will aid our understanding of the pharmacological mechanisms of antiviral agents.


Asunto(s)
Aciclovir/administración & dosificación , Antivirales/administración & dosificación , Bagres/virología , Enfermedades de los Peces/tratamiento farmacológico , Ictalurivirus/efectos de los fármacos , Ovario/citología , Replicación Viral/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Efecto Citopatogénico Viral/efectos de los fármacos , Femenino , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/fisiopatología , Enfermedades de los Peces/virología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Ictalurivirus/genética , Ictalurivirus/fisiología , Ovario/virología , Proteínas Virales/genética , Proteínas Virales/metabolismo
19.
PLoS Pathog ; 16(9): e1008765, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32970777

RESUMEN

Tilapia is one of the most important economic and fastest-growing species in aquaculture worldwide. In 2015, an epidemic associated with severe mortality occurred in adult tilapia in Hubei, China. The causative pathogen was identified as Tilapia parvovirus (TiPV) by virus isolation, electron microscopy, experimental challenge, In situ hybridization (ISH), indirect immunofluorescence (IFA), and viral gene sequencing. Electron microscopy revealed large numbers of parvovirus particles in the organs of diseased fish, including kidney, spleen, liver, heart, brain, gill, intestine, etc. The virions were spherical in shape, non-enveloped and approximately 30nm in diameter. The TiPV was isolated and propagated in tilapia brain cells (TiB) and induced a typical cytopathic effect (CPE) after 3 days post-infection (dpi). This virus was used to experimentally infect adult tilapia and clinical disease symptoms similar to those observed naturally were replicated. Additionally, the results of ISH and IFA showed positive signals in kidney and spleen tissues from TiPV-infected fish. To identify TiPV-specific sequences, the near complete genome of TiPV was obtained and determined to be 4269 bp in size. Phylogenetic analysis of the NS1 sequence revealed that TiPV is a novel parvovirus, forms a separate branch in proposed genus Chapparvovirus of Parvoviridae. Results presented here confirm that TiPV is a novel parvovirus pathogen that can cause massive mortality in adult tilapia. This provides a basis for the further studies to define the epidemiology, pathology, diagnosis, prevention and treatment of this emerging viral disease.


Asunto(s)
Enfermedades de los Peces/virología , Infecciones por Parvoviridae/virología , Parvovirus/patogenicidad , Tilapia/virología , Animales , China , Efecto Citopatogénico Viral/efectos de los fármacos , Bazo/virología
20.
Sci Rep ; 10(1): 15247, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943689

RESUMEN

Microbicides play critical roles in infection prevention and control of Ebola virus by decontaminating high-touch environmental surfaces (HITES), interrupting the virus-HITES-hands nexus. We evaluated the efficacy of formulations containing different microbicidal actives for inactivating Ebola virus-Makona strain (EBOV/Mak) on stainless-steel carriers per ASTM E2197-11. Formulations of sodium hypochlorite (NaOCl) (0.05-1%), ethanol (70%), chloroxylenol (PCMX) (0.12-0.48% by weight) in hard water, and a ready-to-use disinfectant spray with 58% ethanol (EDS), were tested at contact times of 0, or 0.5 to 10 min at ambient temperature. EBOV/Mak was inactivated (> 6 log10) by 70% ethanol after contact times ≥ 2.5 min, by 0.5% and 1% NaOCl or EDS (> 4 log10) at contact times ≥ 5 min, and by 0.12-0.48% PCMX (> 4.2 log10) at contact times ≥ 5 min. Residual infectious virus in neutralized samples was assessed by passage on cells and evaluation for viral cytopathic effect. No infectious virus was detected in cells inoculated with EBOV/Mak exposed to NaOCl (0.5% or 1%), PCMX (0.12% to 0.48%), or EDS for ≥ 5 min. These results demonstrate ≥ 6 log10 inactivation of EBOV/Mak dried on prototypic surfaces by EDS or formulations of NaOCl (≥ 0.5%), PCMX (≥ 0.12%), or 70% ethanol at contact times ≥ 5 min.


Asunto(s)
Antiinfecciosos/farmacología , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/prevención & control , Inactivación de Virus/efectos de los fármacos , Animales , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Desinfectantes/farmacología , Ebolavirus/patogenicidad , Microbiología Ambiental , Etanol/farmacología , Fiebre Hemorrágica Ebola/transmisión , Fiebre Hemorrágica Ebola/virología , Humanos , Técnicas In Vitro , Porosidad , Hipoclorito de Sodio/farmacología , Propiedades de Superficie , Células Vero , Xilenos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...