Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 15(12)2023 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-38140557

RESUMO

BACKGROUND: As long as COVID-19 endures, viral surface proteins will keep changing and new viral strains will emerge, rendering prior vaccines and treatments decreasingly effective. To provide durable targets for preventive and therapeutic agents, there is increasing interest in slowly mutating viral proteins, including non-surface proteins like RdRp. METHODS: A scoping review of studies was conducted describing RdRp in the context of COVID-19 through MEDLINE/PubMed and EMBASE. An iterative approach was used with input from content experts and three independent reviewers, focused on studies related to either RdRp activity inhibition or RdRp mechanisms against SARS-CoV-2. RESULTS: Of the 205 records screened, 43 studies were included in the review. Twenty-five evaluated RdRp activity inhibition, and eighteen described RdRp mechanisms of existing drugs or compounds against SARS-CoV-2. In silico experiments suggested that RdRp inhibitors developed for other RNA viruses may be effective in disrupting SARS-CoV-2 replication, indicating a possible reduction of disease progression from current and future variants. In vitro, in vivo, and human clinical trial studies were largely consistent with these findings. CONCLUSIONS: Future risk mitigation and treatment strategies against forthcoming SARS-CoV-2 variants should consider targeting RdRp proteins instead of surface proteins.


Assuntos
Antivirais , COVID-19 , RNA Polimerase Dependente de RNA , Humanos , Antivirais/farmacologia , Antivirais/uso terapêutico , Antivirais/metabolismo , COVID-19/genética , COVID-19/metabolismo , Proteínas de Membrana , Simulação de Acoplamento Molecular , Pandemias , RNA Polimerase Dependente de RNA/efeitos dos fármacos , RNA Polimerase Dependente de RNA/genética , SARS-CoV-2/metabolismo , Tratamento Farmacológico da COVID-19/métodos
2.
Molecules ; 27(3)2022 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-35164069

RESUMO

The human population is still facing appalling conditions due to several outbreaks of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) virus. The absence of specific drugs, appropriate vaccines for mutants, and knowledge of potential therapeutic agents makes this situation more difficult. Several 1, 2, 4-triazolo [1, 5-a] pyrimidine (TP)-derivative compounds were comprehensively studied for antiviral activities against RNA polymerase of HIV, HCV, and influenza viruses, and showed immense pharmacological interest. Therefore, TP-derivative compounds can be repurposed against the RNA-dependent RNA polymerase (RdRp) protein of SARS-CoV-2. In this study, a meta-analysis was performed to ensure the genomic variability and stability of the SARS-CoV-2 RdRp protein. The molecular docking of natural and synthetic TP compounds to RdRp and molecular dynamic (MD) simulations were performed to analyse the dynamic behaviour of TP compounds at the active site of the RdRp protein. TP compounds were also docked against other non-structural proteins (NSP1, NSP2, NSP3, NSP5, NSP8, NSP13, and NSP15) of SARS-CoV-2. Furthermore, the inhibition potential of TP compounds was compared with Remdesivir and Favipiravir drugs as a positive control. Additionally, TP compounds were analysed for inhibitory activity against SARS-CoV RdRp protein. This study demonstrates that TP analogues (monomethylated triazolopyrimidine and essramycin) represent potential lead molecules for designing an effective inhibitor to control viral replication. Furthermore, in vitro and in vivo studies will strengthen the use of these inhibitors as suitable drug candidates against SARS-CoV-2.


Assuntos
RNA-Polimerase RNA-Dependente de Coronavírus/efeitos dos fármacos , RNA-Polimerase RNA-Dependente de Coronavírus/metabolismo , Pirimidinas/farmacologia , Triazóis/farmacologia , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Alanina/análogos & derivados , Alanina/farmacologia , Amidas/farmacologia , COVID-19/metabolismo , Domínio Catalítico/efeitos dos fármacos , Biologia Computacional/métodos , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Pirazinas/farmacologia , Pirimidinas/química , RNA Viral/efeitos dos fármacos , RNA Polimerase Dependente de RNA/efeitos dos fármacos , RNA Polimerase Dependente de RNA/metabolismo , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/metabolismo , Triazóis/química , Replicação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
3.
Antiviral Res ; 196: 105209, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34801588

RESUMO

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative agent of Coronavirus Disease 2019 (COVID-19) pandemic. Despite intensive and global efforts to discover and develop novel antiviral therapies, only Remdesivir has been approved as a treatment for COVID-19. Therefore, effective antiviral therapeutics are still urgently needed to combat and halt the pandemic. Viral RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 demonstrates high potential as a reliable target for the development of antivirals. We previously developed a cell-based assay to assess the efficiency of compounds that target SARS-CoV-2 RdRp, as well as their tolerance to viral exoribonuclease-mediated proof-reading. In our previous study, we discovered that 2-((1H-indol-3-yl)thio)-N-phenyl-acetamides specifically targets the RdRp of both respiratory syncytial virus (RSV) and influenza A virus. Thus, we hypothesize that 2-((1H-indol-3-yl)thio)-N-phenyl-acetamides may also have the ability to inhibit SARS-CoV-2 replication by targeting its RdRp activity. In this research, we test a compound library containing 103 of 2-((1H-indol-3-yl)thio)-N-phenyl-acetamides against SARS-CoV-2 RdRp, using our cell-based assay. Among these compounds, the top five candidates strongly inhibit SARS-CoV-2 RdRp activity while exhibiting low cytotoxicity and resistance to viral exoribonuclease. Compound 6-72-2a is the most promising candidate with the lowest EC50 value of 1.41 µM and highest selectivity index (CC50/EC50) (above 70.92). Furthermore, our data suggests that 4-46b and 6-72-2a also inhibit the replication of HCoV-OC43 and HCoV-NL63 virus in a dose-dependent manner. Compounds 4-46b and 6-72-2a exhibit EC50 values of 1.13 µM and 0.94 µM, respectively, on HCoV-OC43 viral replication. However, higher concentrations of these compounds are needed to effectively block HCoV-NL63 replication. Together, our findings successfully identified 4-46b and 6-72-2a as promising inhibitors against SARS-CoV-2 RdRp.


Assuntos
Acetamidas/farmacologia , Tratamento Farmacológico da COVID-19 , RNA Polimerase Dependente de RNA , Antivirais/farmacologia , Sistemas de Liberação de Medicamentos , Humanos , RNA Viral/biossíntese , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
4.
Cell Rep ; 37(4): 109882, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34653416

RESUMO

Remdesivir (RDV), a nucleotide analog with broad-spectrum features, has exhibited effectiveness in COVID-19 treatment. However, the precise working mechanism of RDV when targeting the viral RNA-dependent RNA polymerase (RdRP) has not been fully elucidated. Here, we solve a 3.0-Å structure of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RdRP elongation complex (EC) and assess RDV intervention in polymerase elongation phase. Although RDV could induce an "i+3" delayed termination in meta-stable complexes, only pausing and subsequent elongation are observed in the EC. A comparative investigation using an enterovirus RdRP further confirms similar delayed intervention and demonstrates that steric hindrance of the RDV-characteristic 1'-cyano at the -4 position is responsible for the "i+3" intervention, although two representative Flaviviridae RdRPs do not exhibit similar behavior. A comparison of representative viral RdRP catalytic complex structures indicates that the product RNA backbone encounters highly conserved structural elements, highlighting the broad-spectrum intervention potential of 1'-modified nucleotide analogs in anti-RNA virus drug development.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Antivirais/farmacologia , RNA Polimerase Dependente de RNA/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Proteínas Virais/efeitos dos fármacos , Monofosfato de Adenosina/farmacologia , Alanina/farmacologia , Microscopia Crioeletrônica , Humanos , RNA Viral/química , RNA Viral/efeitos dos fármacos , RNA Polimerase Dependente de RNA/química , SARS-CoV-2/química , Proteínas Virais/química , Replicação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
5.
Microbiol Spectr ; 9(1): e0047221, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34378968

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a causative agent of the coronavirus disease 2019 (COVID-19) pandemic, and the development of therapeutic interventions is urgently needed. So far, monoclonal antibodies and drug repositioning are the main methods for drug development, and this effort was partially successful. Since the beginning of the COVID-19 pandemic, the emergence of SARS-CoV-2 variants has been reported in many parts of the world, and the main concern is whether the current vaccines and therapeutics are still effective against these variant viruses. Viral entry and viral RNA-dependent RNA polymerase (RdRp) are the main targets of current drug development; therefore, the inhibitory effects of transmembrane serine protease 2 (TMPRSS2) and RdRp inhibitors were compared among the early SARS-CoV-2 isolate (lineage A) and the two recent variants (lineage B.1.1.7 and lineage B.1.351) identified in the United Kingdom and South Africa, respectively. Our in vitro analysis of viral replication showed that the drugs targeting TMPRSS2 and RdRp are equally effective against the two variants of concern. IMPORTANCE The COVID-19 pandemic is causing unprecedented global problems in both public health and human society. While some vaccines and monoclonal antibodies were successfully developed very quickly and are currently being used, numerous variants of the causative SARS-CoV-2 are emerging and threatening the efficacy of vaccines and monoclonal antibodies. In order to respond to this challenge, we assessed antiviral efficacy of small-molecule inhibitors that are being developed for treatment of COVID-19 and found that they are still very effective against the SARS-CoV-2 variants. Since most small-molecule inhibitors target viral or host factors other than the mutated sequence of the viral spike protein, they are expected to be potent control measures against the COVID-19 pandemic.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , RNA Polimerase Dependente de RNA/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Serina Endopeptidases/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Chlorocebus aethiops , Humanos , África do Sul , Reino Unido , Células Vero , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
6.
Nat Commun ; 12(1): 279, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436624

RESUMO

Remdesivir is the only FDA-approved drug for the treatment of COVID-19 patients. The active form of remdesivir acts as a nucleoside analog and inhibits the RNA-dependent RNA polymerase (RdRp) of coronaviruses including SARS-CoV-2. Remdesivir is incorporated by the RdRp into the growing RNA product and allows for addition of three more nucleotides before RNA synthesis stalls. Here we use synthetic RNA chemistry, biochemistry and cryo-electron microscopy to establish the molecular mechanism of remdesivir-induced RdRp stalling. We show that addition of the fourth nucleotide following remdesivir incorporation into the RNA product is impaired by a barrier to further RNA translocation. This translocation barrier causes retention of the RNA 3'-nucleotide in the substrate-binding site of the RdRp and interferes with entry of the next nucleoside triphosphate, thereby stalling RdRp. In the structure of the remdesivir-stalled state, the 3'-nucleotide of the RNA product is matched and located with the template base in the active center, and this may impair proofreading by the viral 3'-exonuclease. These mechanistic insights should facilitate the quest for improved antivirals that target coronavirus replication.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Alanina/análogos & derivados , Alanina/farmacologia , RNA Polimerase Dependente de RNA/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Antivirais/farmacologia , Aptâmeros de Nucleotídeos , RNA-Polimerase RNA-Dependente de Coronavírus/efeitos dos fármacos , Nucleotídeos , RNA Viral , RNA Polimerase Dependente de RNA/genética , SARS-CoV-2/enzimologia , Replicação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
7.
Artigo em Inglês | MEDLINE | ID: mdl-32122918

RESUMO

The influenza virus RNA-dependent RNA polymerase is highly conserved among influenza A, B, C, and D viruses. It comprises three subunits: polymerase basic protein 1 (PB1), polymerase basic protein 2 (PB2), and polymerase acidic protein (PA) in influenza A and B viruses or polymerase 3 protein (P3) in influenza C and D viruses. Because this polymerase is essential for influenza virus replication, it has been considered as a target for antiviral agents. Recently, several polymerase inhibitors that target each subunit have been developed. This review discusses the mechanism of action, antiviral activity, and emergence of resistance to three inhibitors approved for the treatment of influenza or in late-phase clinical trials: the PB1 inhibitor favipiravir, the PB2 inhibitor pimodivir, and the PA inhibitor baloxavir marboxil.


Assuntos
Farmacorresistência Viral , Influenza Humana/genética , Influenza Humana/virologia , Orthomyxoviridae/efeitos dos fármacos , RNA Polimerase Dependente de RNA , Amidas , Antivirais/uso terapêutico , Dibenzotiepinas , Humanos , Morfolinas , Pirazinas , Piridinas , Piridonas , Pirimidinas , Pirróis , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Triazinas , Replicação Viral/efeitos dos fármacos
8.
J Virol ; 94(24)2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-32907977

RESUMO

Dengue virus (DENV) NS5 RNA-dependent RNA polymerase (RdRp), an important drug target, synthesizes viral RNA and is essential for viral replication. While a number of allosteric inhibitors have been reported for hepatitis C virus RdRp, few have been described for DENV RdRp. Following a diverse compound screening campaign and a rigorous hit-to-lead flowchart combining biochemical and biophysical approaches, two DENV RdRp nonnucleoside inhibitors were identified and characterized. These inhibitors show low- to high-micromolar inhibition in DENV RNA polymerization and cell-based assays. X-ray crystallography reveals that they bind in the enzyme RNA template tunnel. One compound (NITD-434) induced an allosteric pocket at the junction of the fingers and palm subdomains by displacing residue V603 in motif B. Binding of another compound (NITD-640) ordered the fingers loop preceding the F motif, close to the RNA template entrance. Most of the amino acid residues that interacted with these compounds are highly conserved in flaviviruses. Both sites are important for polymerase de novo initiation and elongation activities and essential for viral replication. This work provides evidence that the RNA tunnel in DENV RdRp offers interesting target sites for inhibition.IMPORTANCE Dengue virus (DENV), an important arthropod-transmitted human pathogen that causes a spectrum of diseases, has spread dramatically worldwide in recent years. Despite extensive efforts, the only commercial vaccine does not provide adequate protection to naive individuals. DENV NS5 polymerase is a promising drug target, as exemplified by the development of successful commercial drugs against hepatitis C virus (HCV) polymerase and HIV-1 reverse transcriptase. High-throughput screening of compound libraries against this enzyme enabled the discovery of inhibitors that induced binding sites in the RNA template channel. Characterizations by biochemical, biophysical, and reverse genetics approaches provide a better understanding of the biological relevance of these allosteric sites and the way forward to design more-potent inhibitors.


Assuntos
Vírus da Dengue/genética , Vírus da Dengue/metabolismo , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Sítio Alostérico , Antivirais/farmacologia , Sítios de Ligação , Cristalografia por Raios X , Dengue/virologia , Transcriptase Reversa do HIV , Ensaios de Triagem em Larga Escala , Humanos , Modelos Moleculares , RNA Polimerase Dependente de RNA/efeitos dos fármacos , RNA Polimerase Dependente de RNA/genética , Replicon , Alinhamento de Sequência , Análise de Sequência de Proteína , Proteínas não Estruturais Virais/efeitos dos fármacos , Proteínas não Estruturais Virais/genética , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia
9.
Comput Biol Med ; 124: 103936, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32738628

RESUMO

Virtual screening of phytochemicals was performed through molecular docking, simulations, in silico ADMET and drug-likeness prediction to identify the potential hits that can inhibit the effects of SARS-CoV-2. Considering the published literature on medicinal importance, 154 phytochemicals with analogous structure from limonoids and triterpenoids were selected to search potential inhibitors for the five therapeutic protein targets of SARS-CoV-2, i.e., 3CLpro (main protease), PLpro (papain-like protease), SGp-RBD (spike glycoprotein-receptor binding domain), RdRp (RNA dependent RNA polymerase) and ACE2 (angiotensin-converting enzyme 2). The in silico computational results revealed that the phytochemicals such as glycyrrhizic acid, limonin, 7-deacetyl-7-benzoylgedunin, maslinic acid, corosolic acid, obacunone and ursolic acid were found to be effective against the target proteins of SARS-CoV-2. The protein-ligand interaction study revealed that these phytochemicals bind with the amino acid residues at the active site of the target proteins. Therefore, the core structure of these potential hits can be used for further lead optimization to design drugs for SARS-CoV-2. Also, the medicinal plants containing these phytochemicals like licorice, neem, tulsi, citrus and olives can be used to formulate suitable therapeutic approaches in traditional medicines.


Assuntos
Antivirais/farmacologia , Betacoronavirus/química , Betacoronavirus/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/virologia , Limoninas/farmacologia , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/virologia , Triterpenos/farmacologia , Enzima de Conversão de Angiotensina 2 , Antivirais/química , Antivirais/farmacocinética , Sítios de Ligação , COVID-19 , Biologia Computacional , Simulação por Computador , RNA-Polimerase RNA-Dependente de Coronavírus , Avaliação Pré-Clínica de Medicamentos , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Humanos , Limoninas/química , Limoninas/farmacocinética , Simulação de Acoplamento Molecular , Pandemias , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/efeitos dos fármacos , Compostos Fitoquímicos/química , Compostos Fitoquímicos/farmacocinética , Compostos Fitoquímicos/farmacologia , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/efeitos dos fármacos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/efeitos dos fármacos , Triterpenos/química , Triterpenos/farmacocinética , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/efeitos dos fármacos , Proteínas Virais/química , Proteínas Virais/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
10.
J Med Microbiol ; 69(6): 864-873, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32469301

RESUMO

Introduction. The emergence of SARS-CoV-2 has taken humanity off guard. Following an outbreak of SARS-CoV in 2002, and MERS-CoV about 10 years later, SARS-CoV-2 is the third coronavirus in less than 20 years to cross the species barrier and start spreading by human-to-human transmission. It is the most infectious of the three, currently causing the COVID-19 pandemic. No treatment has been approved for COVID-19. We previously proposed targets that can serve as binding sites for antiviral drugs for multiple coronaviruses, and here we set out to find current drugs that can be repurposed as COVID-19 therapeutics.Aim. To identify drugs against COVID-19, we performed an in silico virtual screen with the US Food and Drug Administration (FDA)-approved drugs targeting the RNA-dependent RNA polymerase (RdRP), a critical enzyme for coronavirus replication.Methodology. Initially, no RdRP structure of SARS-CoV-2 was available. We performed basic sequence and structural analysis to determine if RdRP from SARS-CoV was a suitable replacement. We performed molecular dynamics simulations to generate multiple starting conformations that were used for the in silico virtual screen. During this work, a structure of RdRP from SARS-CoV-2 became available and was also included in the in silico virtual screen.Results. The virtual screen identified several drugs predicted to bind in the conserved RNA tunnel of RdRP, where many of the proposed targets were located. Among these candidates, quinupristin is particularly interesting because it is expected to bind across the RNA tunnel, blocking access from both sides and suggesting that it has the potential to arrest viral replication by preventing viral RNA synthesis. Quinupristin is an antibiotic that has been in clinical use for two decades and is known to cause relatively minor side effects.Conclusion. Quinupristin represents a potential anti-SARS-CoV-2 therapeutic. At present, we have no evidence that this drug is effective against SARS-CoV-2 but expect that the biomedical community will expeditiously follow up on our in silico findings.


Assuntos
Antivirais/farmacologia , Betacoronavirus/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Pneumonia Viral/tratamento farmacológico , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Animais , Antivirais/uso terapêutico , Betacoronavirus/enzimologia , Betacoronavirus/genética , Betacoronavirus/fisiologia , COVID-19 , Infecções por Coronavirus/virologia , Avaliação Pré-Clínica de Medicamentos/métodos , Sinergismo Farmacológico , Humanos , Conformação Molecular , Pandemias , Filogenia , Pneumonia Viral/virologia , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Rifampina/farmacologia , SARS-CoV-2 , Alinhamento de Sequência , Análise de Sequência de Proteína , Virginiamicina/análogos & derivados , Virginiamicina/farmacologia , Replicação Viral/efeitos dos fármacos
11.
Antiviral Res ; 165: 55-64, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30885750

RESUMO

Influenza viruses are major respiratory pathogens responsible for both seasonal epidemics and occasional pandemics worldwide. The current available treatment options have limited efficacy and thus the development of new antivirals is highly needed. We previously reported the identification of a series of cycloheptathiophene-3-carboxamide compounds as influenza A virus inhibitors that act by targeting the protein-protein interactions between the PA-PB1 subunits of the viral polymerase. In this study, we characterized the antiviral properties of the most promising compounds as well as investigated their propensity to induce drug resistance. Our results show that some of the selected compounds possess potent, broad-spectrum anti-influenza activity as they efficiently inhibited the replication of several strains of influenza A and B viruses, including an oseltamivir-resistant clinical isolate, with nanomolar or low-micromolar potency. The most promising compounds specifically inhibited the PA-PB1 binding in vitro and interfered with the influenza A virus polymerase activity in a cellular context, without showing cytotoxicity. The most active PA-PB1 inhibitors showed to possess a drug resistance barrier higher than that of oseltamivir. Indeed, no viral variants with reduced susceptibility to the selected compounds emerged after serial passages of influenza A virus under drug selective pressure. Overall, our studies identified potent PA-PB1 inhibitors as promising candidates for the development of new anti-influenza drugs.


Assuntos
Antivirais/farmacologia , Orthomyxoviridae/efeitos dos fármacos , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Animais , Farmacorresistência Viral , Humanos , Vírus da Influenza A/metabolismo , Vírus da Influenza B/metabolismo , Oseltamivir/farmacologia , RNA Polimerase Dependente de RNA/biossíntese , Proteínas Virais/genética , Replicação Viral/efeitos dos fármacos
12.
Appl Biochem Biotechnol ; 185(3): 799-814, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29349531

RESUMO

A precipitous increase in the number of flaviviral infections has been noted over the last 5 years. Despite these outbreaks, treatment protocols for infected individuals remain ambiguous. Numerous studies have identified NITD008 as a potent flavivirus inhibitor; however, clinical testing was dismissed due to undesirable toxic effects. The binding landscape of NITD008 in complex with five detrimental flaviviruses at the RNA-dependent RNA polymerase active sites was explored. An "all-in-one" pharmacophore model was created for the design of small molecules that may inhibit a broad spectrum of flaviviruses. This pharmacophore model approach serves as a robust cornerstone, thus assisting medicinal experts in the composition of multifunctional inhibitors that will eliminate cross-resistance and toxicity and enhance patient adherence.


Assuntos
Adenosina/análogos & derivados , Antivirais/farmacologia , Descoberta de Drogas/métodos , Flaviviridae/efeitos dos fármacos , Modelos Químicos , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Adenosina/química , Adenosina/metabolismo , Adenosina/farmacologia , Domínio Catalítico , Flaviviridae/enzimologia , Flaviviridae/fisiologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , RNA Polimerase Dependente de RNA/metabolismo , Termodinâmica , Replicação Viral/efeitos dos fármacos
13.
Antiviral Res ; 143: 38-47, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28390873

RESUMO

Chikungunya virus (CHIKV) is an important arboviral infectious agent in tropical and subtropical regions, often causing persistent and debilitating disease. The viral enzyme non-structural protein 4 (nsP4), as RNA-dependent RNA polymerase (RdRP), catalyzes the formation of negative-sense, genomic and subgenomic viral RNAs. Here we report a truncated nsP4 construct that is soluble, stable and purified recombinantly from Escherichia coli. Sequence analyses and homology modelling indicate that all necessary RdRP elements are included. Hydrogen/deuterium exchange with mass spectrometry was used to analyze solvent accessibility and flexibility of subdomains. Fluorophore-conjugated RNA ligands were designed and screened by using fluorescence anisotropy to select a suitable substrate for RdRP assays. Assay trials revealed that nsP4 core domain is conditionally active upon choice of detergent species, and carries out both primed extension and terminal adenylyltransferase activities. The polymerization assay can be further developed to screen for antiviral compounds in vitro.


Assuntos
Vírus Chikungunya/enzimologia , Detergentes/farmacologia , RNA Polimerase Dependente de RNA/efeitos dos fármacos , RNA Polimerase Dependente de RNA/metabolismo , Proteínas não Estruturais Virais/química , Antivirais , Domínio Catalítico , Febre de Chikungunya/virologia , Vírus Chikungunya/genética , Clonagem Molecular , Detergentes/química , Ensaios Enzimáticos , Escherichia coli/genética , Polarização de Fluorescência , Cinética , Nucleotidiltransferases/genética , Proteínas com Motivo de Reconhecimento de RNA/química , RNA Viral/genética , RNA Polimerase Dependente de RNA/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Alinhamento de Sequência , Análise de Sequência , Homologia Estrutural de Proteína , Proteínas não Estruturais Virais/genética
14.
PLoS One ; 12(4): e0174523, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28380007

RESUMO

BACKGROUND: Influenza virus infection is a major threat to human health. Small interfering RNA (siRNA) is a promising approach for the prevention and treatment of viral infections. In this study, we constructed a series of DNA vector-based short hairpin RNAs (shRNAs) that target various genes of the influenza A virus using the polymerase III U6-RNA promoter to prevent influenza virus infection in vitro and in a mouse model. RESULTS: Three sets of DNA vector-based shRNA, two targeting genes encoding the polymerase acidic protein (PA) and one targeting polymerase basic protein 2 (PB2), efficiently inhibited the replication of influenza virus A/WSN/33(H1N1) in vitro. We also successfully prevented influenza virus A/WSN/33(H1N1) infection in a C57BL/6 mouse model by intratracheal delivery of anti-PB2 shRNA. CONCLUSIONS: Our findings suggest that the PB2-targeting shRNA plasmid showed potential for use as an RNAi-based therapeutic for influenza virus infection.


Assuntos
Vírus da Influenza A/efeitos dos fármacos , Infecções por Orthomyxoviridae/prevenção & controle , Interferência de RNA , RNA Interferente Pequeno/farmacologia , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Terapêutica com RNAi/métodos , Proteínas Virais/efeitos dos fármacos , Animais , Feminino , Vírus da Influenza A/fisiologia , Intubação Intratraqueal , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Replicação Viral/efeitos dos fármacos
15.
Antiviral Res ; 134: 226-235, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27649989

RESUMO

RNA dependent RNA polymerases (RdRp) are essential enzymes for flavivirus replication. Starting from an in silico docking analysis we identified a pyridobenzothiazole compound, HeE1-2Tyr, able to inhibit West Nile and Dengue RdRps activity in vitro, which proved effective against different flaviviruses in cell culture. Crystallographic data show that HeE1-2Tyr binds between the fingers domain and the priming loop of Dengue virus RdRp (Site 1). Conversely, enzyme kinetics, binding studies and mutational analyses suggest that, during the catalytic cycle and assembly of the RdRp-RNA complex, HeE1-2Tyr might be hosted in a distinct binding site (Site 2). RdRp mutational studies, driven by in silico docking analysis, allowed us to locate the inhibition Site 2 in the thumb domain. Taken together, our results provide innovative concepts for optimization of a new class of anti-flavivirus compounds.


Assuntos
Benzotiazóis/farmacologia , Flavivirus/enzimologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Antivirais/farmacologia , Benzotiazóis/química , Sítios de Ligação , Domínio Catalítico , Cristalização , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/enzimologia , Descoberta de Drogas , Flavivirus/efeitos dos fármacos , Cinética , Modelos Moleculares , Simulação de Acoplamento Molecular , Mutação , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus do Nilo Ocidental/enzimologia
16.
Antiviral Res ; 134: 63-76, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27575793

RESUMO

Worldwide, respiratory syncytial virus (RSV) causes severe disease in infants, the elderly, and immunocompromised people. No vaccine or effective antiviral treatment is available. RSV is a member of the non-segmented, negative-strand (NNS) group of RNA viruses and relies on its RNA-dependent RNA polymerase to transcribe and replicate its genome. Because of its essential nature and unique properties, the RSV polymerase has proven to be a good target for antiviral drugs, with one compound, ALS-8176, having already achieved clinical proof-of-concept efficacy in a human challenge study. In this article, we first provide an overview of the role of the RSV polymerase in viral mRNA transcription and genome replication. We then review past and current approaches to inhibiting the RSV polymerase, including use of nucleoside analogs and non-nucleoside inhibitors. Finally, we consider polymerase inhibitors that hold promise for treating infections with other NNS RNA viruses, including measles and Ebola.


Assuntos
Antivirais/uso terapêutico , RNA Polimerases Dirigidas por DNA/antagonistas & inibidores , Mononegavirais/efeitos dos fármacos , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Antivirais/farmacologia , Ensaios Clínicos como Assunto , RNA Polimerases Dirigidas por DNA/efeitos dos fármacos , RNA Polimerases Dirigidas por DNA/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Doença pelo Vírus Ebola/tratamento farmacológico , Humanos , Sarampo/tratamento farmacológico , Mononegavirais/enzimologia , Mononegavirais/genética , Nucleosídeos/agonistas , RNA Mensageiro , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sincicial Respiratório Humano/enzimologia , Vírus Sincicial Respiratório Humano/genética , Transcrição Gênica , Replicação Viral/efeitos dos fármacos
17.
Eur J Med Chem ; 117: 321-34, 2016 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-27161176

RESUMO

In this work, we present and discuss a comprehensive set of both newly and previously synthesized compounds belonging to 5 distinct molecular classes of linear aromatic N-polycyclic systems that efficiently inhibits bovine viral diarrhea virus (BVDV) infection. A coupled in silico/in vitro investigation was employed to formulate a molecular rationale explaining the notable affinity of all molecules to BVDV RNA dependent RNA polymerase (RdRp) NS5B. We initially developed a three-dimensional common-feature pharmacophore model according to which two hydrogen bond acceptors and one hydrophobic aromatic feature are shared by all molecular series in binding the viral polymerase. The pharmacophoric information was used to retrieve a putative binding site on the surface of the BVDV RdRp and to guide compound docking within the protein binding site. The affinity of all compounds towards the enzyme was scored via molecular dynamics-based simulations, showing high correlation with in vitro EC50 data. The determination of the interaction spectra of the protein residues involved in inhibitor binding highlighted amino acids R295 and Y674 as the two fundamental H-bond donors, while two hydrophobic cavities HC1 (residues A221, I261, I287, and Y289) and HC2 (residues V216, Y303, V306, K307, P408, and A412) fulfill the third pharmacophoric requirement. Three RdRp (K263, R295 and Y674) residues critical for drug binding were selected and mutagenized, both in silico and in vitro, into alanine, and the affinity of a set of selected compounds towards the mutant RdRp isoforms was determined accordingly. The agreement between predicted and experimental data confirmed the proposed common molecular rationale shared by molecules characterized by different chemical scaffolds in binding to the BVDV RdRp, ultimately yielding compound 6b (EC50 = 0.3 µM; IC50 = 0.48 µM) as a new, potent inhibitor of this Pestivirus.


Assuntos
Vírus da Diarreia Viral Bovina/efeitos dos fármacos , Hidrocarbonetos Policíclicos Aromáticos/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Animais , Antivirais/química , Antivirais/farmacologia , Sítios de Ligação/genética , Doença das Mucosas por Vírus da Diarreia Viral Bovina/tratamento farmacológico , Doença das Mucosas por Vírus da Diarreia Viral Bovina/prevenção & controle , Bovinos , Vírus da Diarreia Viral Bovina/enzimologia , Ligação de Hidrogênio , Modelos Moleculares , Hidrocarbonetos Policíclicos Aromáticos/farmacologia , RNA Polimerase Dependente de RNA/efeitos dos fármacos
18.
J Biomed Sci ; 21: 14, 2014 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-24521134

RESUMO

Enterovirus 71 (EV-71) is the main etiological agent of hand, foot and mouth disease (HFMD). Recent EV-71 outbreaks in Asia-Pacific were not limited to mild HFMD, but were associated with severe neurological complications such as aseptic meningitis and brainstem encephalitis, which may lead to cardiopulmonary failure and death. The absence of licensed therapeutics for clinical use has intensified research into anti-EV-71 development. This review highlights the potential antiviral agents targeting EV-71 attachment, entry, uncoating, translation, polyprotein processing, virus-induced formation of membranous RNA replication complexes, and RNA-dependent RNA polymerase. The strategies for antiviral development include target-based synthetic compounds, anti-rhinovirus and poliovirus libraries screening, and natural compound libraries screening. Growing knowledge of the EV-71 life cycle will lead to successful development of antivirals. The continued effort to develop antiviral agents for treatment is crucial in the absence of a vaccine. The coupling of antivirals with an effective vaccine will accelerate eradication of the disease.


Assuntos
Antivirais/uso terapêutico , Enterovirus Humano A/efeitos dos fármacos , Enterovirus Humano A/genética , Infecções por Enterovirus/tratamento farmacológico , Antivirais/química , Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/genética , Infecções por Enterovirus/patologia , Humanos , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
19.
Curr Drug Targets ; 15(1): 103-13, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24102407

RESUMO

Australia is facing a major national medical challenge with the emergence of the Hendra virus (HeV) as a medically and economically important pathogen of humans and animals. Clinical symptoms of human HeV infection can include fever, hypotension, dizziness, encephalitis, respiratory haemorrhage and edema. The window of opportunity for successful patient treatment remains unknown, but is likely to be very narrow. Currently, very few effective therapeutic options are available for the case management of severe HeV infections or the rapid silencing of local outbreaks. This underscores the need for more activity in the drug discovery arena to develop much needed therapeutics that specifically targets this deadly disease. The structural analysis of HeV is very much in its infancy, which leaves many gaps in our understanding of the biology of HeV and makes structure-guided drug design difficult. Structural studies of the viral RNAdependent- RNA polymerase (RdRp), which is the heart of the viral replication machinery, will set the stage for rational drug design and fill a major gap in our understanding of the HeV replication machinery. This review examines the current knowledge based on the multi-domain architecture of the Hendra RdRp and highlights which essential domain functions represent tangible targets for drug development against this deadly disease.


Assuntos
Antivirais/farmacologia , Vírus Hendra/efeitos dos fármacos , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Animais , Vírus Hendra/enzimologia , Humanos , RNA Polimerase Dependente de RNA/metabolismo
20.
Antiviral Res ; 78(3): 194-201, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18258312

RESUMO

RNA polymerase of influenza virus is a specific enzyme necessary for the viral replication. A siRNA against the RNA polymerase and the RNA polymerase inhibitor L-742,001 reduced accumulation of viral RNAs in the infected cells. L-742,001 strongly inhibited virus re-growth after removal of the agent from the culture, whereas the neuraminidase inhibitor zanamivir did not. L-742,001-resistant mutants showed a Thr-20 to Ala substitution in the PA subunit of RNA polymerase. The drug-resistant virus showed a slight reduction in the susceptibility to L-742,001 in both the plaque assay (threefold reduction) and enzyme assay (two- to three-fold reduction). The resistance levels were lower than those of zanamivir-resistant mutants in the plaque assay. Against zanamivir-resistant mutants, L-742,001 retained the same antiviral activity as against the wild-type strain. These results indicate that L-742,001 is most likely to act at the PA subunit, and possesses a unique profile. It is suggested that PA subunit of RNA polymerase is a promising target for anti-influenza virus agents.


Assuntos
Antivirais/farmacologia , Desenho de Fármacos , Hidroxibutiratos/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Piperidinas/farmacologia , RNA Interferente Pequeno/metabolismo , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Proteínas Virais/efeitos dos fármacos , Animais , Linhagem Celular , Farmacorresistência Viral , Inibidores Enzimáticos/farmacologia , Humanos , Vírus da Influenza A/enzimologia , Vírus da Influenza A/genética , Testes de Sensibilidade Microbiana , Mutação , RNA Interferente Pequeno/genética , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/genética , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/química , Proteínas Virais/genética , Zanamivir/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...