Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Virology ; 571: 21-33, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35439707

RESUMO

Since December 2019, the deadly novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the current COVID-19 pandemic. To date, vaccines are available in the developed countries to prevent the infection of this virus; however, medicines are necessary to help control COVID-19. Human coronavirus 229E (HCoV-229E) causes the common cold. The main protease (Mpro) is an essential enzyme required for the multiplication of these two viruses in the host cells, and thus is an appropriate candidate to screen potential medicinal compounds. Flavonols and dihydroflavonols are two groups of plant flavonoids. In this study, we report docking simulation with two Mpro enzymes and five flavonols and three dihydroflavonols, in vitro inhibition of the SARS-CoV-2 Mpro, and in vitro inhibition of the HCoV 229E replication. The docking simulation results predicted that (+)-dihydrokaempferol, (+)- dihydroquercetin, (+)-dihydromyricetin, kaempferol, quercetin, myricentin, isoquercitrin, and rutin could bind to at least two subsites (S1, S1', S2, and S4) in the binding pocket and inhibit the activity of SARS-CoV-2 Mpro. Their affinity scores ranged from -8.8 to -7.4 (kcal/mol). Likewise, these compounds were predicted to bind and inhibit the HCoV-229E Mpro activity with affinity scores ranging from -7.1 to -7.8 (kcal/mol). In vitro inhibition assays showed that seven available compounds effectively inhibited the SARS-CoV-2 Mpro activity and their IC50 values ranged from 0.125 to 12.9 µM. Five compounds inhibited the replication of HCoV-229E in Huh-7 cells. These findings indicate that these antioxidative flavonols and dihydroflavonols are promising candidates for curbing the two viruses.


Assuntos
Coronavirus Humano 229E , Proteases 3C de Coronavírus , Flavonóis , SARS-CoV-2 , COVID-19 , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano 229E/fisiologia , Proteases 3C de Coronavírus/antagonistas & inibidores , Flavonóis/farmacologia , Humanos , Simulação de Acoplamento Molecular , SARS-CoV-2/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
2.
Molecules ; 27(3)2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35163977

RESUMO

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


Assuntos
1-Naftilamina/análogos & derivados , Aminoquinolinas/farmacologia , Antivirais/farmacologia , Coronavirus/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , 1-Naftilamina/farmacologia , Animais , Linhagem Celular , Chlorocebus aethiops , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano NL63/efeitos dos fármacos , Coronavirus Humano OC43/efeitos dos fármacos , Humanos , Técnicas In Vitro , Células Vero , Replicação Viral/efeitos dos fármacos
3.
J Ethnopharmacol ; 287: 114965, 2022 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-34990767

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Coronavirus and influenza virus infection seriously threaten human health. Cangma Huadu Granules (CMHD) is an in-hospital preparation composed of eight traditional Chinese medicines (TCM), which has been clinically used against COVID-19 in China and may be a promising candidate for the treatment of influenza. However, the role of its treatment urgently needs to be studied. AIM OF THE STUDY: To evaluate the therapeutic effects of CMHD on pneumonia induced by coronavirus (HCoV-229E) and influenza A virus (H1N1/FM1) in mice and explore its mechanism of anti-infection. MATERIALS AND METHODS: Mice were infected with HCoV-229E or H1N1/FM1 virus through the nasal cavity. CMHD (12.1, 6.05 and 3.03 g/kg/d) or the positive control drugs were administered intragastrically. The lung index and histopathological changes were used to evaluate the therapeutic effect of CMHD. The expression of TNF-α, IL-1ß, IL-6 and IL-4 in Serum and the proportion of CD4+ and CD8+ T lymphocytes in peripheral blood were detected to evaluate the anti-inflammatory and immune regulation effects of CMHD, respectively. Furthermore, the levels of p-NF-κBp65/ NF-κB p65, which was the key targets of the NF-κB pathway was analyzed. RESULTS: In HCoV-229E-induced pneumonia, the lung index was markedly reduced, and lung pathology was improved in mice that treated with CMHD (12.1, 6.05 g/kg/d). Meanwhile, the expression of TNF-α, IL-6 were obviously inhibited, but the expression of IL-4 was significantly increased in CMHD groups. Compared with the model group, CMHD could also markedly upregulate the level of CD4+ and CD8+. Furthermore, CMHD has a markedly effect on inhibit the expression of p-NF-κB p65/NF-κB p65 in the lung. In H1N1-induced pneumonia, the lung index of mice in the CMHD (12.1 g/kg/d) treatment group was lower than that in the model group, and less inflammatory infiltration could be seen in the lung pathological. Moreover, CMHD could also obviously decrease the expression of TNF-α, IL-1ß, IL-6, but significantly increase the expression of IL-4. Except for that, CMHD could also markedly downregulate the level of CD4+ and upregulate the level of CD8+ compared with the model group. In addition, CMHD has a markedly effect on inhibit the expression of p-NF-κB p65/NF-κB p65 in the lung. CONCLUSION: CMHD can significantly combats viral infections caused by HCoV-229E and H1N1, and the mechanism may be related to its multiple functions of anti-inflammatory, immunity regulating and inhibiting NF-κB signal transduction pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Infecções por Coronavirus/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Medicina Tradicional Chinesa/métodos , Infecções por Orthomyxoviridae/tratamento farmacológico , Animais , Anti-Inflamatórios/uso terapêutico , Coronavirus Humano 229E/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/uso terapêutico , Feminino , Imunidade/efeitos dos fármacos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Pneumonia/tratamento farmacológico , Pneumonia/patologia , Linfócitos T/metabolismo , Fator de Transcrição RelA/metabolismo
4.
Phytomedicine ; 95: 153874, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34923232

RESUMO

BACKGROUND: Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human coronavirus 229E (HCoV-229E) pose a huge threat to human public health, no specific treatment is available. Jinzhen granule (JZ) is a traditional eight ingredients-Chinese medicine with prominent efficacy for treating viral-induced diseases. However, little is known about the antiviral effect and mechanism of JZ against SARS-CoV-2 and HCoV-229E. PURPOSE: This study aimed to reveal the antiviral effects of JZ against SARS-CoV-2 and HCoV-229E, and to further explore the underlying mechanisms regulating the host immune response. METHODS: The chromatographic separation of JZ was performed using a Shimadzu analytical high-performance liquid chromatograph with UV detection and Alltech ELSD 2000ES. We conducted cytopathic effect (CPE) and plaque reduction assays to evaluate the antiviral effect of JZ. A lethal human angiotensin converting enzyme 2 (hACE2) transgenic mouse model of SARS-CoV-2 was established to determine the protective effect of JZ on mortality and lung virus titers. Real-time quantitative PCR assays were used to analyze the expression of proinflammatory cytokines in vitro and in vivo. Western blotting was further performed to determine the activities on regulating the nuclear factor kappa B (NF-κB)/MAPK pathway. Finally, mitochondrial membrane potential assays, flow cytometry analysis and western blotting were used to assess the anti-apoptotic potency toward HCoV-229E infection. RESULTS: The results showed that 13 chemical components were identified and five peaks were determined and quantitated (gallic acid 1.97 mg/g, baicalin 20.69 mg/g, glycyrrhizic acid 4.92 mg/g, hyodeoxycholic acid 4.86 mg/g, cholic acid 4.07 mg/g). We found that JZ exerted inhibitory potency against SARS-CoV-2 and HCoV-229E in vitro by using CPE and plaque reduction assays, and it was further found that JZ protected mice infected by SARS-CoV-2 from death and inhibited lung virus titers. JZ also significantly decreased the induction of inflammatory cytokines (IL-1α, IL-6, CCL-5 and MIP-1ß), similar to the observed in vitro effect. Moreover, JZ suppressed the release of inflammatory cytokines in vitro and it decreased the protein expression of p-p38 MAPK, p-JNK, p-NF-κB p65 and p-IκBα induced by HCoV-229E and increased the expression of IκBα. Notably, JZ significantly protected HCoV-229E-infected Huh-7 cells from mitochondrial damage and decreased apoptotic cells. The activation of the mitochondria-mediated apoptotic pathway was inhibited by JZ, as shown by the reduced expression of cleaved caspase-9, caspase-3 and p-PARP. CONCLUSIONS: In conclusion, JZ (gallic acid 1.97 mg/g, baicalin 20.69 mg/g, glycyrrhizic acid 4.92 mg/g, hyodeoxycholic acid 4.86 mg/g, cholic acid 4.07 mg/g) exhibited antiviral activities against SARS-CoV-2 and HCoV-229E by regulating the NF-κB/MAPK pathway and the mitochondria-mediated apoptotic pathway. These findings demonstrated the efficacy of JZ against CoVs and suggested JZ treatment as a novel clinical therapeutic strategy for COVID-19.


Assuntos
Antivirais , Coronavirus Humano 229E , Medicamentos de Ervas Chinesas/farmacologia , SARS-CoV-2/efeitos dos fármacos , Animais , Antivirais/farmacologia , COVID-19 , Coronavirus Humano 229E/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , NF-kappa B
5.
Int J Mol Sci ; 22(21)2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34769299

RESUMO

The COVID-19 pandemic caused by SARS-CoV-2 infection poses a serious threat to global public health and the economy. The enzymatic product of cholesterol 25-hydroxylase (CH25H), 25-Hydroxycholesterol (25-HC), was reported to have potent anti-SARS-CoV-2 activity. Here, we found that the combination of 25-HC with EK1 peptide, a pan-coronavirus (CoV) fusion inhibitor, showed a synergistic antiviral activity. We then used the method of 25-HC modification to design and synthesize a series of 25-HC-modified peptides and found that a 25-HC-modified EK1 peptide (EK1P4HC) was highly effective against infections caused by SARS-CoV-2, its variants of concern (VOCs), and other human CoVs, such as HCoV-OC43 and HCoV-229E. EK1P4HC could protect newborn mice from lethal HCoV-OC43 infection, suggesting that conjugation of 25-HC with a peptide-based viral inhibitor was a feasible and universal strategy to improve its antiviral activity.


Assuntos
Antivirais/farmacologia , Hidroxicolesteróis/química , Lipopeptídeos/química , SARS-CoV-2/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Antivirais/química , Antivirais/uso terapêutico , Peso Corporal/efeitos dos fármacos , COVID-19/virologia , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano 229E/patogenicidade , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/mortalidade , Infecções por Coronavirus/virologia , Coronavirus Humano OC43/efeitos dos fármacos , Coronavirus Humano OC43/patogenicidade , Modelos Animais de Doenças , Sinergismo Farmacológico , Humanos , Hidroxicolesteróis/farmacologia , Hidroxicolesteróis/uso terapêutico , Lipopeptídeos/farmacologia , Lipopeptídeos/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Polietilenoglicóis/química , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/fisiologia , Taxa de Sobrevida , Internalização do Vírus/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
6.
Cells ; 10(11)2021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34831176

RESUMO

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.


Assuntos
Coronavirus Humano 229E/fisiologia , Coronavirus Humano OC43/fisiologia , Proteína com Valosina/metabolismo , Replicação Viral/fisiologia , Antivirais/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano OC43/efeitos dos fármacos , Efeito Citopatogênico Viral/efeitos dos fármacos , Humanos , Proteoma/efeitos dos fármacos , Proteoma/metabolismo , RNA Interferente Pequeno/genética , RNA Viral/biossíntese , Proteína com Valosina/antagonistas & inibidores , Proteína com Valosina/genética , Replicação Viral/efeitos dos fármacos , Desenvelopamento do Vírus/efeitos dos fármacos
7.
Viruses ; 13(11)2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34835033

RESUMO

The COVID-19 pandemic has deeply influenced sanitization procedures, and high-level disinfection has been massively used to prevent SARS-CoV-2 spread, with potential negative impact on the environment and on the threat of antimicrobial resistance (AMR). Aiming to overcome these concerns, yet preserving the effectiveness of sanitization against enveloped viruses, we assessed the antiviral properties of the Probiotic Cleaning Hygiene System (PCHS), an eco-sustainable probiotic-based detergent previously proven to stably abate pathogen contamination and AMR. PCHS (diluted 1:10, 1:50 and 1:100) was tested in comparison to common disinfectants (70% ethanol and 0.5% sodium hypochlorite), in suspension and carrier tests, according with the European UNI EN 14476:2019 and UNI EN 16777:2019 standards. Human alpha- and beta-coronaviruses hCoV-229E and SARS-CoV-2, human herpesvirus type 1, human and animal influenza viruses, and vaccinia virus were included in the study. The results showed that PCHS was able to inactivate 99.99% of all tested viruses within 1-2 h of contact, both in suspension and on surface. Notably, while control disinfectants became inactive within 2 h after application, the PCHS antiviral action persisted up to 24 h post-application, suggesting that its use may effectively allow a continuous prevention of virus spread via contaminated environment, without worsening environmental pollution and AMR concern.


Assuntos
Desinfecção/métodos , Probióticos/farmacologia , Saneamento/métodos , Viroses/prevenção & controle , Vírus/efeitos dos fármacos , Animais , Antivirais/farmacologia , COVID-19/prevenção & controle , COVID-19/virologia , Coronavirus Humano 229E/efeitos dos fármacos , Desinfetantes/farmacologia , Microbiologia Ambiental , Herpesvirus Humano 1/efeitos dos fármacos , Humanos , Orthomyxoviridae/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Vírus Vaccinia/efeitos dos fármacos , Viroses/virologia
8.
Virology ; 564: 33-38, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34619630

RESUMO

Endemic seasonal coronaviruses cause morbidity and mortality in a subset of patients, but no specific treatment is available. Molnupiravir is a promising pipeline antiviral drug for treating SARS-CoV-2 infection potentially by targeting RNA-dependent RNA polymerase (RdRp). This study aims to evaluate the potential of repurposing molnupiravir for treating seasonal human coronavirus (HCoV) infections. Molecular docking revealed that the active form of molnupiravir, ß-D-N4-hydroxycytidine (NHC), has similar binding affinity to RdRp of SARS-CoV-2 and seasonal HCoV-NL63, HCoV-OC43 and HCoV-229E. In cell culture models, treatment of molnupiravir effectively inhibited viral replication and production of infectious viruses of the three seasonal coronaviruses. A time-of-drug-addition experiment indicates the specificity of molnupiravir in inhibiting viral components. Furthermore, combining molnupiravir with the protease inhibitor GC376 resulted in enhanced antiviral activity. Our findings highlight that the great potential of repurposing molnupiravir for treating seasonal coronavirus infected patients.


Assuntos
Coronavirus Humano 229E/genética , Infecções por Coronavirus/tratamento farmacológico , Coronavirus Humano NL63/genética , Coronavirus Humano OC43/genética , Citidina/análogos & derivados , Hidroxilaminas/farmacologia , Antivirais/química , Antivirais/metabolismo , Antivirais/farmacologia , Resfriado Comum/tratamento farmacológico , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano 229E/fisiologia , Coronavirus Humano NL63/efeitos dos fármacos , Coronavirus Humano NL63/fisiologia , Coronavirus Humano OC43/efeitos dos fármacos , Coronavirus Humano OC43/fisiologia , Citidina/farmacologia , Humanos , Simulação de Acoplamento Molecular , Ligação Proteica/efeitos dos fármacos , Pirrolidinas/farmacologia , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Estações do Ano , Ácidos Sulfônicos/farmacologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
9.
Nat Commun ; 12(1): 6055, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34663813

RESUMO

COVID-19 caused by the SARS-CoV-2 virus has become a global pandemic. 3CL protease is a virally encoded protein that is essential across a broad spectrum of coronaviruses with no close human analogs. PF-00835231, a 3CL protease inhibitor, has exhibited potent in vitro antiviral activity against SARS-CoV-2 as a single agent. Here we report, the design and characterization of a phosphate prodrug PF-07304814 to enable the delivery and projected sustained systemic exposure in human of PF-00835231 to inhibit coronavirus family 3CL protease activity with selectivity over human host protease targets. Furthermore, we show that PF-00835231 has additive/synergistic activity in combination with remdesivir. We present the ADME, safety, in vitro, and in vivo antiviral activity data that supports the clinical evaluation of PF-07304814 as a potential COVID-19 treatment.


Assuntos
Tratamento Farmacológico da COVID-19 , Proteases 3C de Coronavírus/antagonistas & inibidores , Inibidores de Protease de Coronavírus/administração & dosagem , Indóis/administração & dosagem , Leucina/administração & dosagem , Pirrolidinonas/administração & dosagem , Monofosfato de Adenosina/administração & dosagem , Monofosfato de Adenosina/efeitos adversos , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacocinética , Alanina/administração & dosagem , Alanina/efeitos adversos , Alanina/análogos & derivados , Alanina/farmacocinética , Animais , COVID-19/virologia , Chlorocebus aethiops , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano 229E/enzimologia , Inibidores de Protease de Coronavírus/efeitos adversos , Inibidores de Protease de Coronavírus/farmacocinética , Modelos Animais de Doenças , Desenho de Fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Células HeLa , Humanos , Indóis/efeitos adversos , Indóis/farmacocinética , Infusões Intravenosas , Leucina/efeitos adversos , Leucina/farmacocinética , Camundongos , Pirrolidinonas/efeitos adversos , Pirrolidinonas/farmacocinética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/enzimologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/enzimologia , Células Vero
10.
Biosensors (Basel) ; 11(8)2021 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-34436052

RESUMO

Cell-based assays are a valuable tool for examination of virus-host cell interactions and drug discovery processes, allowing for a more physiological setting compared to biochemical assays. Despite the fact that cell-based SPR assays are label-free and thus provide all the associated benefits, they have never been used to study viral growth kinetics and to predict drug antiviral response in cells. In this study, we prove the concept that the cell-based SPR assay can be applied in the kinetic analysis of the early stages of viral infection of cells and the antiviral drug activity in the infected cells. For this purpose, cells immobilized on the SPR slides were infected with human coronavirus HCov-229E and treated with hydroxychloroquine. The SPR response was measured at different time intervals within the early stages of infection. Methyl Thiazolyl Tetrazolium (MTT) assay was used to provide the reference data. We found that the results of the SPR and MTT assays were consistent, and SPR is a reliable tool in investigating virus-host cell interaction and the mechanism of action of viral inhibitors. SPR assay was more sensitive and accurate in the first hours of infection within the first replication cycle, whereas the MTT assay was not so effective. After the second replication cycle, noise was generated by the destruction of the cell layer and by the remnants of dead cells, and masks useful SPR signals.


Assuntos
Antivirais/uso terapêutico , Coronavirus Humano 229E/fisiologia , Infecções por Coronavirus/tratamento farmacológico , Hidroxicloroquina/uso terapêutico , Ressonância de Plasmônio de Superfície/métodos , Animais , Antivirais/farmacologia , Chlorocebus aethiops , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano 229E/isolamento & purificação , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Humanos , Hidroxicloroquina/farmacologia , Cinética , Índice de Gravidade de Doença , Células Vero
11.
Int J Mol Sci ; 22(15)2021 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-34360797

RESUMO

A novel series of N-substituted cis- and trans-3-aryl-4-(diethoxyphosphoryl)azetidin-2-ones were synthesized by the Kinugasa reaction of N-methyl- or N-benzyl-(diethyoxyphosphoryl)nitrone and selected aryl alkynes. Stereochemistry of diastereoisomeric adducts was established based on vicinal H3-H4 coupling constants in azetidin-2-one ring. All the obtained azetidin-2-ones were evaluated for the antiviral activity against a broad range of DNA and RNA viruses. Azetidin-2-one trans-11f showed moderate inhibitory activity against human coronavirus (229E) with EC50 = 45 µM. The other isomer cis-11f was active against influenza A virus H1N1 subtype (EC50 = 12 µM by visual CPE score; EC50 = 8.3 µM by TMS score; MCC > 100 µM, CC50 = 39.9 µM). Several azetidin-2-ones 10 and 11 were tested for their cytostatic activity toward nine cancerous cell lines and several of them appeared slightly active for Capan-1, Hap1 and HCT-116 cells values of IC50 in the range 14.5-97.9 µM. Compound trans-11f was identified as adjuvant of oxacillin with significant ability to enhance the efficacy of this antibiotic toward the highly resistant S. aureus strain HEMSA 5. Docking and molecular dynamics simulations showed that enantiomer (3R,4S)-11f can be responsible for the promising activity due to the potency in displacing oxacillin at ß-lactamase, thus protecting the antibiotic from undesirable biotransformation.


Assuntos
Adjuvantes Farmacêuticos/química , Adjuvantes Farmacêuticos/farmacologia , Antivirais/química , Antivirais/farmacologia , Azetidinas/farmacologia , Infecções/tratamento farmacológico , Antibacterianos/química , Antibacterianos/farmacologia , Azetidinas/química , Proteínas de Bactérias/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Coronavirus Humano 229E/efeitos dos fármacos , Citostáticos/química , Citostáticos/farmacologia , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Simulação de Dinâmica Molecular , Oxacilina/química , Proteínas de Ligação às Penicilinas/química , Staphylococcus aureus/efeitos dos fármacos , Estereoisomerismo , beta-Lactamases/química
12.
Viruses ; 13(8)2021 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-34452529

RESUMO

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.


Assuntos
Antivirais/farmacologia , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano OC43/efeitos dos fármacos , Indóis/farmacologia , SARS-CoV-2/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Animais , Antivirais/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Coronavirus Humano 229E/fisiologia , Coronavirus Humano OC43/fisiologia , Efeito Citopatogênico Viral/efeitos dos fármacos , Humanos , Indóis/administração & dosagem , Testes de Sensibilidade Microbiana , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , SARS-CoV-2/fisiologia , Células Vero , Carga Viral/efeitos dos fármacos , Ensaio de Placa Viral , Replicação Viral/efeitos dos fármacos
13.
Antiviral Res ; 193: 105127, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34217752

RESUMO

In this study, a series of 10 quinoline analogues was evaluated for their in vitro antiviral activity against a panel of alpha- and beta-coronaviruses, including the severe acute respiratory syndrome coronaviruses 1 and 2 (SARS-CoV-1 and SARS-CoV-2), as well as the human coronaviruses (HCoV) 229E and OC43. Chloroquine and hydroxychloroquine were the most potent with antiviral EC50 values in the range of 0.12-12 µM. Chloroquine displayed the most favorable selectivity index (i.e. ratio cytotoxic versus antiviral concentration), being 165 for HCoV-OC43 in HEL cells. Potent anti-coronavirus activity was also observed with amodiaquine, ferroquine and mefloquine, although this was associated with substantial cytotoxicity for mefloquine. Primaquine, quinidine, quinine and tafenoquine only blocked coronavirus replication at higher concentrations, while piperaquine completely lacked antiviral and cytotoxic effects. A time-of-addition experiment in HCoV-229E-infected HEL cells revealed that chloroquine interferes with viral entry at a post-attachment stage. Using confocal microscopy, no viral RNA synthesis could be detected upon treatment of SARS-CoV-2-infected cells with chloroquine. The inhibition of SARS-CoV-2 replication by chloroquine and hydroxychloroquine coincided with an inhibitory effect on the autophagy pathway as visualized by a dose-dependent increase in LC3-positive puncta. The latter effect was less pronounced or even absent with the other quinolines. In summary, we showed that several quinoline analogues, including chloroquine, hydroxychloroquine, amodiaquine, ferroquine and mefloquine, exhibit broad anti-coronavirus activity in vitro.


Assuntos
Antimaláricos/farmacologia , Antivirais/farmacologia , Infecções por Coronavirus/tratamento farmacológico , Coronavirus/efeitos dos fármacos , Quinolinas/farmacologia , Animais , Chlorocebus aethiops , Cloroquina/farmacologia , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano OC43/efeitos dos fármacos , Humanos , Hidroxicloroquina/farmacologia , SARS-CoV-2/efeitos dos fármacos , Células Vero , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
14.
Appl Environ Microbiol ; 87(19): e0109821, 2021 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34288707

RESUMO

A novel and robust approach to evaluate the antiviral activity of coatings was developed, assessing three commercially available leave-on surface coating products for efficacy against human coronaviruses (HCoVs) HCoV-229E and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The assessment is based on three criteria that reflect real-life settings, namely, (i) immediate antiviral effect, (ii) effect after repeated cleaning of the coated surface, and (iii) antiviral activity in the presence of organic material. The results showed that only a copper compound-based coating successfully met all three criteria. A quaternary ammonium compound-based coating did not meet the second criterion, and a coating based on reactive oxygen species showed no antiviral effect. Moreover, the study demonstrated that HCoV-229E is a relevant SARS-CoV-2 surrogate for such experiments. This new approach allows benchmarking of currently available antiviral coatings and future coating developments to avoid unjustified claims. The deployment of efficient antiviral coatings can offer an additional measure to mitigate the risk of transmission of respiratory viruses like SARS-CoV-2 or influenza viruses from high-touch surfaces. IMPORTANCE SARS-CoV-2, the virus responsible for the coronavirus disease 2019 (COVID-19) pandemic, is transmitted mainly person-to-person through respiratory droplets, while the contribution of fomite transmission is less important than suspected at the beginning of the pandemic. Nevertheless, antiviral-coating solutions can offer an additional measure to mitigate the risk of SARS-CoV-2 transmission from high-touch surfaces. The deployment of antiviral coatings is not new, but what is currently lacking is solid scientific evidence of the efficacy of commercially available self-disinfecting surfaces under real-life conditions. Therefore, we developed a novel, robust approach to evaluate the antiviral activity of such coatings, applying strict quality criteria to three commercially available products to test their efficacies against SARS-CoV-2. We also showed that HCoV-229E is a relevant surrogate for such experiments. Our approach will also bring significant benefit to the evaluation of the effects of coatings on the survival of nonenveloped viruses, which are known to be more tolerant to desiccation and disinfectants and for which high-touch surfaces play an important role.


Assuntos
Antivirais/farmacologia , Coronavirus Humano 229E/efeitos dos fármacos , Desinfetantes/farmacologia , SARS-CoV-2/efeitos dos fármacos
15.
PLoS Pathog ; 17(5): e1009599, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34043740

RESUMO

Antiviral therapeutics are a front-line defense against virally induced diseases. Because viruses frequently mutate to escape direct inhibition of viral proteins, there is interest in targeting the host proteins that the virus must co-opt to complete its replication cycle. However, a detailed understanding of the interactions between the virus and the host cell is necessary in order to facilitate development of host-directed therapeutics. As a first step, we performed a genome-wide loss of function screen using the alphacoronavirus HCoV-229E to better define the interactions between coronaviruses and host factors. We report the identification and validation of an ER-resident host protein, TMEM41B, as an essential host factor for not only HCoV-229E but also genetically distinct coronaviruses including the pandemic betacoronavirus SARS-CoV-2. We show that the protein is required at an early, but post-receptor engagement, stage of the viral lifecycle. Further, mechanistic studies revealed that although the protein was not enriched at replication complexes, it likely contributes to viral replication complex formation via mobilization of cholesterol and other lipids to facilitate host membrane expansion and curvature. Continued study of TMEM41B and the development of approaches to prevent its function may lead to broad spectrum anti-coronavirus therapeutics.


Assuntos
Coronavirus Humano 229E/efeitos dos fármacos , Interações entre Hospedeiro e Microrganismos/fisiologia , Proteínas de Membrana/metabolismo , Animais , Antivirais/farmacologia , COVID-19/metabolismo , Linhagem Celular , Chlorocebus aethiops , Coronavirus Humano 229E/fisiologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Proteínas de Membrana/fisiologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Células Vero , Replicação Viral/efeitos dos fármacos
16.
mSphere ; 6(2)2021 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-33910996

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) persists on stainless steel and plastic for up to 7 days, suggesting that coronavirus disease 2019 (COVID-19) could be spread by fomite transmission. There is limited research on the stability of SARS-CoV-2 on textiles, with the risk of textiles acting as fomites not being well understood. To date, there does not appear to be any published research on the stability of coronaviruses during laundering, which is required to determine the efficacy of current laundering policies in the decontamination of health care textiles. The aim of this study was to investigate the environmental stability of human coronaviruses HCoV-OC43 and HCoV-229E on different textile fiber types and the persistence of HCoV-OC43 on textiles during domestic and industrial laundering. This study demonstrated that human coronaviruses (5 log10 50% tissue culture infective doses [TCID50]) remain infectious on polyester for ≥72 h, cotton for ≥24 h, and polycotton for ≥6 h; HCoV-OC43 was also able to transfer from polyester to PVC or polyester after 72 h. Under clean conditions, HCoV-OC43 was not detectable on cotton swatches laundered with industrial and domestic wash cycles without temperature and detergent (≥4.57-log10-TCID50 reduction), suggesting that the dilution and agitation of wash cycles are sufficient to remove human coronaviruses from textiles. In the presence of interfering substances (artificial saliva), ≤1.78 log10 TCID50 HCoV-OC43 was detected after washing domestically without temperature and detergent, unlike industrial laundering, where the virus was completely removed. However, no infectious HCoV-OC43 was detected when washed domestically with detergent.IMPORTANCE Synthetic textiles such as polyester could potentially act as fomites of human coronaviruses, indicating the importance of infection control procedures during handling of contaminated textiles prior to laundering. This study provides novel evidence that human coronaviruses can persist on textiles for up to 3 days and are readily transferred from polyester textile to other surfaces after 72 h of incubation. This is of particular importance for the domestic laundering of contaminated textiles such as health care uniforms in the United Kingdom and United States, where there may be a risk of cross-contaminating the domestic environment. It was demonstrated that human coronaviruses are removed from contaminated textiles by typical domestic and commercial wash cycles, even at low temperatures without detergent, indicating that current health care laundering policies are likely sufficient in the decontamination of SARS-CoV-2 from textiles.


Assuntos
COVID-19/transmissão , Resfriado Comum/transmissão , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano OC43/efeitos dos fármacos , Detergentes/farmacologia , Têxteis/virologia , Linhagem Celular , Fibra de Algodão/virologia , Fômites/virologia , Humanos , Lavanderia , Poliésteres , SARS-CoV-2/efeitos dos fármacos
17.
Viruses ; 13(3)2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33807081

RESUMO

The emergent human coronavirus SARS-CoV-2 and its high infectivity rate has highlighted the strong need for new disinfection systems. Evidence has proven that airborne transmission is an important route of spreading for this virus. Therefore, this short communication introduces CLODOS Technology®, a novel strategy to disinfect contaminated surfaces. It is a product based on stable and 99% pure chlorine dioxide, already certified as a bactericide, fungicide and virucide against different pathogens. In this study, CLODOS Technology®, by direct contact or thermonebulization, showed virucidal activity against the human coronavirus HCoV-229E at non-cytotoxic doses. Different conditions such as nebulization, exposure time and product concentration have been tested to standardize and optimize this new feasible method for disinfection.


Assuntos
Coronavirus Humano 229E/efeitos dos fármacos , Desinfetantes/farmacologia , Desinfecção/métodos , Linhagem Celular , Compostos Clorados/análise , Compostos Clorados/farmacologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Desinfetantes/análise , Desinfecção/instrumentação , Humanos , Nebulizadores e Vaporizadores , Óxidos/análise , Óxidos/farmacologia
18.
J Med Chem ; 64(9): 5632-5644, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33877845

RESUMO

To develop antiviral therapeutics against human coronavirus (HCoV) infections, suitable coronavirus drug targets and corresponding lead molecules must be urgently identified. Here, we describe the discovery of a class of HCoV inhibitors acting on nsp15, a hexameric protein component of the viral replication-transcription complexes, endowed with immune evasion-associated endoribonuclease activity. Structure-activity relationship exploration of these 1,2,3-triazolo-fused betulonic acid derivatives yielded lead molecule 5h as a strong inhibitor (antiviral EC50: 0.6 µM) of HCoV-229E replication. An nsp15 endoribonuclease active site mutant virus was markedly less sensitive to 5h, and selected resistance to the compound mapped to mutations in the N-terminal part of HCoV-229E nsp15, at an interface between two nsp15 monomers. The biological findings were substantiated by the nsp15 binding mode for 5h, predicted by docking. Hence, besides delivering a distinct class of inhibitors, our study revealed a druggable pocket in the nsp15 hexamer with relevance for anti-coronavirus drug development.


Assuntos
Antivirais/farmacologia , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano 229E/enzimologia , Endorribonucleases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Ácido Oleanólico/análogos & derivados , Proteínas não Estruturais Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Antivirais/síntese química , Antivirais/química , Linhagem Celular , Relação Dose-Resposta a Droga , Endorribonucleases/metabolismo , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Ácido Oleanólico/síntese química , Ácido Oleanólico/química , Ácido Oleanólico/farmacologia , Proteínas não Estruturais Virais/metabolismo
19.
Viruses ; 13(2)2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33672333

RESUMO

A novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in China at the end of 2019 causing a large global outbreak. As treatments are of the utmost importance, drug repurposing embodies a rich and rapid drug discovery landscape, where candidate drug compounds could be identified and optimized. To this end, we tested seven compounds for their ability to reduce replication of human coronavirus (HCoV)-229E, another member of the coronavirus family. Among these seven drugs tested, four of them, namely rapamycin, disulfiram, loperamide and valproic acid, were highly cytotoxic and did not warrant further testing. In contrast, we observed a reduction of the viral titer by 80% with resveratrol (50% effective concentration (EC50) = 4.6 µM) and lopinavir/ritonavir (EC50 = 8.8 µM) and by 60% with chloroquine (EC50 = 5 µM) with very limited cytotoxicity. Among these three drugs, resveratrol was less cytotoxic (cytotoxic concentration 50 (CC50) = 210 µM) than lopinavir/ritonavir (CC50 = 102 µM) and chloroquine (CC50 = 67 µM). Thus, among the seven drugs tested against HCoV-229E, resveratrol demonstrated the optimal antiviral response with low cytotoxicity with a selectivity index (SI) of 45.65. Similarly, among the three drugs with an anti-HCoV-229E activity, namely lopinavir/ritonavir, chloroquine and resveratrol, only the latter showed a reduction of the viral titer on SARS-CoV-2 with reduced cytotoxicity. This opens the door to further evaluation to fight Covid-19.


Assuntos
Antivirais/farmacologia , Coronavirus Humano 229E/efeitos dos fármacos , Resveratrol/farmacologia , Ritonavir/farmacologia , SARS-CoV-2/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Cloroquina/farmacologia , Coronavirus Humano 229E/fisiologia , Reposicionamento de Medicamentos , Humanos , Lopinavir/farmacologia , Masculino , SARS-CoV-2/fisiologia , Carga Viral
20.
ACS Infect Dis ; 7(3): 586-597, 2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33645977

RESUMO

As the COVID-19 pandemic continues to unfold, the morbidity and mortality are increasing daily. Effective treatment for SARS-CoV-2 is urgently needed. We recently discovered four SARS-CoV-2 main protease (Mpro) inhibitors including boceprevir, calpain inhibitors II and XII, and GC-376 with potent antiviral activity against infectious SARS-CoV-2 in cell culture. In this study, we further characterized the mechanism of action of these four compounds using the SARS-CoV-2 pseudovirus neutralization assay. It was found that GC-376 and calpain inhibitors II and XII have a dual mechanism of action by inhibiting both viral Mpro and host cathepsin L in Vero cells. To rule out the cell-type dependent effect, the antiviral activity of these four compounds against SARS-CoV-2 was also confirmed in type 2 transmembrane serine protease-expressing Caco-2 cells using the viral yield reduction assay. In addition, we found that these four compounds have broad-spectrum antiviral activity in inhibiting not only SARS-CoV-2 but also SARS-CoV, and MERS-CoV, as well as human coronaviruses (CoVs) 229E, OC43, and NL63. The mechanism of action is through targeting the viral Mpro, which was supported by the thermal shift-binding assay and enzymatic fluorescence resonance energy transfer assay. We further showed that these four compounds have additive antiviral effect when combined with remdesivir. Altogether, these results suggest that boceprevir, calpain inhibitors II and XII, and GC-376 might be promising starting points for further development against existing human coronaviruses as well as future emerging CoVs.


Assuntos
Antivirais/farmacologia , Carbonatos/farmacologia , Glicoproteínas/farmacologia , Leucina/farmacologia , Oligopeptídeos/farmacologia , Prolina/análogos & derivados , SARS-CoV-2/efeitos dos fármacos , Ácidos Sulfônicos/farmacologia , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Alanina/análogos & derivados , Alanina/farmacologia , Animais , Células CACO-2 , Catepsina L/antagonistas & inibidores , Linhagem Celular , Chlorocebus aethiops , Coronavirus Humano 229E/efeitos dos fármacos , Proteases 3C de Coronavírus/antagonistas & inibidores , Coronavirus Humano NL63/efeitos dos fármacos , Coronavirus Humano OC43/efeitos dos fármacos , Combinação de Medicamentos , Células HEK293 , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Prolina/farmacologia , Serina Endopeptidases/metabolismo , Células Vero , Tratamento Farmacológico da COVID-19
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...