Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.336
Filtrar
1.
J Med Chem ; 67(19): 16951-16966, 2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39303015

RESUMO

The global pharmaceutical market has been profoundly impacted by the coronavirus pandemic, leading to an increased demand for specific drugs. Consequently, drug resistance has prompted continuous innovation in drug design strategies to effectively combat resistant pathogens or disease variants. Protein dimers play crucial roles in vivo, including catalytic reactions, signal transduction, and structural stability. The site of action for protein dimerization modulators typically does not reside within the active site of the protein, thereby potentially impeding resistance development. Therefore, harnessing viral protein dimerization modulators could represent a promising avenue for combating viral infections. In this Perspective, we provide a detailed introduction to the design principles and applications of dimerization modulators in antiviral research. Furthermore, we analyze various representative examples to elucidate their modes of action while presenting our perspective on dimerization modulators along with the opportunities and challenges associated with this groundbreaking area of investigation.


Assuntos
Antivirais , Desenho de Fármacos , Multimerização Proteica , Antivirais/farmacologia , Antivirais/química , Multimerização Proteica/efeitos dos fármacos , Humanos , Proteínas Virais/química , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , SARS-CoV-2/efeitos dos fármacos , Controle de Qualidade , Tratamento Farmacológico da COVID-19
2.
J Med Virol ; 96(9): e29926, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39295251

RESUMO

H5N1, a highly pathogenic avian influenza virus, poses an ongoing and significant threat to global public health, primarily due to its potential to cause severe respiratory illness and high mortality rates in humans. Despite extensive efforts in vaccination and antiviral therapy, H5N1 continues to exhibit high mutation rates, resulting in recurrent outbreaks and the emergence of drug-resistant strains. Traditional antiviral therapies, such as neuraminidase inhibitors and M2 ion channel blockers, have demonstrated limited efficacy, necessitating the exploration of innovative therapeutic strategies. Proteolysis-targeting chimeras (PROTACs) emerge as a novel and promising approach, leveraging the ubiquitin-proteasome system to selectively degrade pathogenic proteins. Unlike conventional inhibitors that only block protein function, PROTACs eliminate the target protein, providing a sustained therapeutic effect and potentially reducing the development of resistance. This paper offers a comprehensive examination of the current landscape of H5N1 infections, detailing the pathogenesis and challenges associated with existing treatments. It further explores the mechanism of action, design, and therapeutic potential of PROTACs in inhibiting H5N1. By targeting essential viral proteins, such as hemagglutinin and the RNA-dependent RNA polymerase complex, PROTACs hold the potential to revolutionize the treatment of H5N1 infections, offering a new frontier in antiviral therapy.


Assuntos
Antivirais , Virus da Influenza A Subtipo H5N1 , Influenza Humana , Proteólise , Humanos , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Antivirais/farmacologia , Antivirais/uso terapêutico , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Proteólise/efeitos dos fármacos , Animais , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Proteínas Virais/genética , Quimera de Direcionamento de Proteólise
3.
J Mol Graph Model ; 132: 108837, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39098150

RESUMO

Monkeypox is an infectious disease caused by the monkeypox virus (MPXV), a member of the Orthopoxvirus genus closely related to smallpox. The structure of the A42R profilin-like protein is the first and only available structure among MPXV proteins. Biochemical studies of A42R were conducted in the 1990s and later work also analyzed the protein's function in viral replication in cells. This study aims to screen tripeptides for their potential inhibition of the A42R profilin-like protein using computational methods, with implications for MPXV therapy. A total of 8000 tripeptides underwent molecular docking simulations, resulting in the identification of 20 compounds exhibiting strong binding affinity to A42R. To validate the docking results, molecular dynamics simulations and free energy perturbation calculations were performed. These analyses revealed two tripeptides with sequences TRP-THR-TRP and TRP-TRP-TRP, which displayed robust binding affinity to A42R. Markedly, electrostatic interactions predominated over van der Waals interactions in the binding process between tripeptides and A42R. Three A42R residues, namely Glu9, Ser12, and Arg38, appear to be pivotal in mediating the interaction between A42R and the tripeptide ligands. Notably, tripeptides containing two or three tryptophan residues demonstrate a pronounced binding affinity, with the tripeptide comprising three tryptophan amino acids showing the highest level of affinity. These findings offer valuable insights for the selection of compounds sharing a similar structure and possessing a high affinity for A42R, potentially capable of inhibiting its enzyme activity. The study highlights a structural advantage and paves the way for the development of targeted therapies against MPXV infections.


Assuntos
Antivirais , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Monkeypox virus , Proteínas Virais , Antivirais/química , Antivirais/farmacologia , Proteínas Virais/química , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Monkeypox virus/química , Monkeypox virus/efeitos dos fármacos , Ligação Proteica , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Sítios de Ligação , Termodinâmica , Descoberta de Drogas
4.
J Mol Graph Model ; 132: 108846, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39151375

RESUMO

African swine fever (ASF) causes high mortality in pigs and threatens global swine production. There is still a lack of therapeutics available, with two vaccines under scrutiny and no approved small-molecule drugs. Eleven (11) viral proteins were used to identify potential antivirals in in silico screening of secondary metabolites (127) from Chlorella spp. The metabolites were screened for affinity and binding selectivity. High-scoring compounds were assessed through in silico ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) predictions, compared to structurally similar drugs, and checked for off-target docking with prepared swine receptors. Molecular dynamics (MD) simulations determined binding stability while binding energy was measured in Molecular Mechanics - Generalized Born Surface Area (MMGBSA) or Poisson-Boltzmann Surface Area (MMPBSA). Only six (6) compounds passed until MD analyses, of which five (5) were stable after 100 ns of MD runs. Of these five compounds, only three had binding affinities that were comparable to or stronger than controls. Specifically, phytosterols 24,25-dihydrolanosterol and CID 4206521 that interact with the RNA capping enzyme (pNP868R), and ergosterol which bound to the Erv-like thioreductase (pB119L). The compounds identified in this study can be used as a theoretical basis for in vitro screening to develop potent antiviral drugs against ASFV.


Assuntos
Vírus da Febre Suína Africana , Antivirais , Chlorella , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Vírus da Febre Suína Africana/efeitos dos fármacos , Vírus da Febre Suína Africana/química , Antivirais/farmacologia , Antivirais/química , Animais , Chlorella/química , Suínos , Proteínas Virais/química , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Avaliação Pré-Clínica de Medicamentos
5.
Antiviral Res ; 230: 105980, 2024 10.
Artigo em Inglês | MEDLINE | ID: mdl-39117284

RESUMO

In search of novel therapeutic options to treat influenza virus (IV) infections, we previously identified a series of inhibitors that act by disrupting the interactions between the PA and PB1 subunits of the viral RNA polymerase. These compounds showed broad-spectrum antiviral activity against human influenza A and B viruses and a high barrier to the induction of drug resistance in vitro. In this short communication, we investigated the effects of combinations of the PA-PB1 interaction inhibitor 54 with oseltamivir carboxylate (OSC), zanamivir (ZA), favipiravir (FPV), and baloxavir marboxil (BXM) on the inhibition of influenza A and B virus replication in vitro. We observed a synergistic effect of the 54/OSC and 54/ZA combinations and an antagonistic effect when 54 was combined with either FPV or BXM. Moreover, we demonstrated the efficacy of 54 against highly pathogenic avian influenza viruses (HPAIVs) both in cell culture and in the embryonated chicken eggs model. Finally, we observed that 54 enhances OSC protective effect against HPAIV replication in the embryonated eggs model. Our findings represent an advance in the development of alternative therapeutic strategies against both human and avian IV infections.


Assuntos
Antivirais , Sinergismo Farmacológico , Vírus da Influenza A , Oseltamivir , Pirazinas , Proteínas Virais , Replicação Viral , Oseltamivir/farmacologia , Oseltamivir/análogos & derivados , Animais , Antivirais/farmacologia , Humanos , Replicação Viral/efeitos dos fármacos , Pirazinas/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Embrião de Galinha , Proteínas Virais/metabolismo , Proteínas Virais/antagonistas & inibidores , Amidas/farmacologia , Dibenzotiepinas/farmacologia , Vírus da Influenza B/efeitos dos fármacos , Vírus da Influenza B/fisiologia , Zanamivir/farmacologia , Triazinas/farmacologia , Piridonas/farmacologia , Influenza Aviária/tratamento farmacológico , Influenza Aviária/virologia , Morfolinas/farmacologia , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Cães , RNA Polimerases Dirigidas por DNA/antagonistas & inibidores , RNA Polimerases Dirigidas por DNA/metabolismo , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/metabolismo , Linhagem Celular , Células Madin Darby de Rim Canino
6.
Eur J Med Chem ; 277: 116737, 2024 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-39153334

RESUMO

Influenza viruses (IV) are single-stranded RNA viruses with a negative-sense genome and have the potential to cause pandemics. While vaccines exist for influenza, their protection is only partial. Additionally, there is only a limited number of approved anti-IV drugs, which are associated to emergence of drug resistance. To address these issues, for years we have focused on the development of small-molecules that can interfere with the heterodimerization of PA and PB1 subunits of the IV RNA-dependent RNA polymerase (RdRP). In this study, starting from a cycloheptathiophene-3-carboxamide compound that we recently identified, we performed iterative cycles of medicinal chemistry optimization that led to the identification of compounds 43 and 45 with activity in the nanomolar range against circulating A and B strains of IV. Mechanistic studies demonstrated the ability of 43 and 45 to interfere with viral RdRP activity by disrupting PA-PB1 subunits heterodimerization and to bind to the PA C-terminal domain through biophysical assays. Most important, ADME studies of 45 also showed an improvement in the pharmacokinetic profile with respect to the starting hit.


Assuntos
Antivirais , RNA Polimerase Dependente de RNA , Antivirais/farmacologia , Antivirais/química , Antivirais/síntese química , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/metabolismo , Humanos , Animais , Relação Estrutura-Atividade , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Proteínas Virais/química , Estrutura Molecular , Multimerização Proteica/efeitos dos fármacos , Relação Dose-Resposta a Droga , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/enzimologia , Testes de Sensibilidade Microbiana , Cães
7.
Antiviral Res ; 230: 105976, 2024 10.
Artigo em Inglês | MEDLINE | ID: mdl-39117283

RESUMO

Coronaviruses are highly transmissible respiratory viruses that cause symptoms ranging from mild congestion to severe respiratory distress. The recent outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the need for new antivirals with broad-acting mechanisms to combat increasing emergence of new variants. Currently, there are only a few antivirals approved for treatment of SARS-CoV-2. Previously, the rocaglate natural product silvestrol and synthetic rocaglates such as CR-1-31b were shown to have antiviral effects by inhibiting eukaryotic translation initiation factor 4A1 (eIF4A) function and virus protein synthesis. In this study, we evaluated amidino-rocaglates (ADRs), a class of synthetic rocaglates with the most potent eIF4A-inhibitory activity to-date, for inhibition of SARS-CoV-2 infection. This class of compounds showed low nanomolar potency against multiple SARS-CoV-2 variants and in multiple cell types, including human lung-derived cells, with strong inhibition of virus over host protein synthesis and low cytotoxicity. The most potent ADRs were also shown to be active against two highly pathogenic and distantly related coronaviruses, SARS-CoV and MERS-CoV. Mechanistically, cells with mutations of eIF4A1, which are known to reduce rocaglate interaction displayed reduced ADR-associated loss of cellular function, consistent with targeting of protein synthesis. Overall, ADRs and derivatives may offer new potential treatments for SARS-CoV-2 with the goal of developing a broad-acting anti-coronavirus agent.


Assuntos
Antivirais , Biossíntese de Proteínas , SARS-CoV-2 , Replicação Viral , SARS-CoV-2/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Antivirais/síntese química , Antivirais/química , Humanos , Chlorocebus aethiops , Animais , Biossíntese de Proteínas/efeitos dos fármacos , Células Vero , Tratamento Farmacológico da COVID-19 , Benzofuranos/farmacologia , Benzofuranos/síntese química , Benzofuranos/química , COVID-19/virologia , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Proteínas Virais/genética , Fator de Iniciação 4A em Eucariotos/antagonistas & inibidores , Fator de Iniciação 4A em Eucariotos/metabolismo
8.
Antimicrob Agents Chemother ; 68(10): e0080024, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39162479

RESUMO

Small-molecule antivirals can be used as chemical probes to stabilize transitory conformational stages of viral target proteins, facilitating structural analyses. Here, we evaluate allosteric pneumo- and paramyxovirus polymerase inhibitors that have the potential to serve as chemical probes and aid the structural characterization of short-lived intermediate conformations of the polymerase complex. Of multiple inhibitor classes evaluated, we discuss in-depth distinct scaffolds that were selected based on well-understood structure-activity relationships, insight into resistance profiles, biochemical characterization of the mechanism of action, and photoaffinity-based target mapping. Each class is thought to block structural rearrangements of polymerase domains albeit target sites and docking poses are distinct. This review highlights validated druggable targets in the paramyxo- and pneumovirus polymerase proteins and discusses discrete structural stages of the polymerase complexes required for bioactivity.


Assuntos
Antivirais , Pneumovirus , Antivirais/farmacologia , Antivirais/química , Relação Estrutura-Atividade , Pneumovirus/efeitos dos fármacos , Humanos , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/química , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Proteínas Virais/química
9.
Virol Sin ; 39(4): 685-693, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39025463

RESUMO

Myocarditis is an inflammatory disease of the cardiac muscle and one of the primary causes of dilated cardiomyopathy. Group B coxsackievirus (CVB) is one of the leading causative pathogens of viral myocarditis, which primarily affects children and young adults. Due to the lack of vaccines, the development of antiviral medicines is crucial to controlling CVB infection and the progression of myocarditis. In this study, we investigated the antiviral effect of baicalein, a flavonoid extracted from Scutellaria baicaleinsis. Our results demonstrated that baicalein treatment significantly reduced cytopathic effect and increased cell viability in CVB3-infected cells. In addition, significant reductions in viral protein 3D, viral RNA, and viral particles were observed in CVB3-infected cells treated with baicalein. We found that baicalein exerted its inhibitory effect in the early stages of CVB3 infection. Baicalein also suppressed viral replication in the myocardium and effectively alleviated myocarditis induced by CVB3 infection. Our study revealed that baicalein exerts its antiviral effect by inhibiting the activity of caspase-1 and viral protease 2A. Taken together, our findings demonstrate that baicalein has antiviral activity against CVB3 infection and may serve as a potential therapeutic option for the myocarditis caused by enterovirus infection.


Assuntos
Antivirais , Caspase 1 , Enterovirus Humano B , Flavanonas , Miocardite , Replicação Viral , Flavanonas/farmacologia , Replicação Viral/efeitos dos fármacos , Enterovirus Humano B/efeitos dos fármacos , Enterovirus Humano B/fisiologia , Antivirais/farmacologia , Animais , Miocardite/tratamento farmacológico , Miocardite/virologia , Humanos , Caspase 1/metabolismo , Proteínas Virais/metabolismo , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/genética , Infecções por Coxsackievirus/tratamento farmacológico , Infecções por Coxsackievirus/virologia , Camundongos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cisteína Endopeptidases/metabolismo , Camundongos Endogâmicos BALB C , Masculino , Scutellaria baicalensis/química , Efeito Citopatogênico Viral/efeitos dos fármacos
10.
Antiviral Res ; 229: 105959, 2024 09.
Artigo em Inglês | MEDLINE | ID: mdl-38986873

RESUMO

Avian influenza outbreaks, including ones caused by highly pathogenic A(H5N1) clade 2.3.4.4b viruses, have devastated animal populations and remain a threat to humans. Risk elements assessed for emerging influenza viruses include their susceptibility to approved antivirals. Here, we screened >20,000 neuraminidase (NA) or polymerase acidic (PA) protein sequences of potentially pandemic A(H5Nx), A(H7Nx), and A(H9N2) viruses that circulated globally in 2010-2023. The frequencies of NA or PA substitutions associated with reduced inhibition (RI) or highly reduced inhibition (HRI) by NA inhibitors (NAIs) (oseltamivir, zanamivir) or a cap-dependent endonuclease inhibitor (baloxavir) were low: 0.60% (137/22,713) and 0.62% (126/20,347), respectively. All tested subtypes were susceptible to NAIs and baloxavir at sub-nanomolar concentrations. A(H9N2) viruses were the most susceptible to oseltamivir, with IC50s 3- to 4-fold lower than for other subtypes (median IC50: 0.18 nM; n = 22). NA-I222M conferred RI of A(H5N1) viruses by oseltamivir (with a 26-fold IC50 increase), but NA-S246N did not reduce inhibition. PA-E23G, PA-K34R, PA-I38M/T, and the previously unreported PA-A36T caused RI by baloxavir in all subtypes tested. Avian A(H9N2) viruses endemic in Egyptian poultry predominantly acquired PA-I38V, which causes only a <3-fold decrease in the baloxavir EC50 and fails to meet the RI criteria. PA-E199A/D in A(H7Nx) and A(H9N2) viruses caused a 2- to 4-fold decrease in EC50 (close to the borderline for RI) and should be closely monitored. Our data indicate antiviral susceptibility is high among avian influenza A viruses with pandemic potential and present novel markers of resistance to existing antiviral interventions.


Assuntos
Antivirais , Aves , Dibenzotiepinas , Farmacorresistência Viral , Inibidores Enzimáticos , Genótipo , Vírus da Influenza A , Influenza Aviária , Neuraminidase , Oseltamivir , Piridonas , Triazinas , Neuraminidase/antagonistas & inibidores , Neuraminidase/genética , Antivirais/farmacologia , Influenza Aviária/virologia , Animais , Inibidores Enzimáticos/farmacologia , Dibenzotiepinas/farmacologia , Farmacorresistência Viral/genética , Piridonas/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/genética , Vírus da Influenza A/enzimologia , Triazinas/farmacologia , Oseltamivir/farmacologia , Aves/virologia , Morfolinas/farmacologia , Endonucleases/antagonistas & inibidores , Endonucleases/genética , Endonucleases/metabolismo , Vírus da Influenza A Subtipo H9N2/efeitos dos fármacos , Vírus da Influenza A Subtipo H9N2/genética , Proteínas Virais/genética , Proteínas Virais/antagonistas & inibidores , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/enzimologia , Zanamivir/farmacologia , Fenótipo , Humanos , Concentração Inibidora 50
11.
Antiviral Res ; 228: 105923, 2024 08.
Artigo em Inglês | MEDLINE | ID: mdl-38844175

RESUMO

There are no approved vaccines or therapeutics for Lassa virus (LASV) infections. To identify compounds with anti-LASV activity, we conducted a cell-based screening campaign at biosafety level 4 and tested almost 60,000 compounds for activity against an infectious reporter LASV. Hits from this screen included several structurally related macrocycles. The most potent, Mac128, had a sub-micromolar EC50 against the reporter virus, inhibited wild-type clade IV LASV, and reduced viral titers by 4 orders of magnitude. Mechanistic studies suggested that Mac128 inhibited viral replication at the level of the polymerase.


Assuntos
Antivirais , Vírus Lassa , Compostos Macrocíclicos , Replicação Viral , Vírus Lassa/efeitos dos fármacos , Antivirais/farmacologia , Antivirais/química , Replicação Viral/efeitos dos fármacos , Compostos Macrocíclicos/farmacologia , Compostos Macrocíclicos/química , Humanos , Animais , Chlorocebus aethiops , Células Vero , Febre Lassa/virologia , Febre Lassa/tratamento farmacológico , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Proteínas Virais/genética
12.
Antiviral Res ; 227: 105907, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38772503

RESUMO

Respiratory syncytial virus (RSV) can cause pulmonary complications in infants, elderly and immunocompromised patients. While two vaccines and two prophylactic monoclonal antibodies are now available, treatment options are still needed. JNJ-7184 is a non-nucleoside inhibitor of the RSV-Large (L) polymerase, displaying potent inhibition of both RSV-A and -B strains. Resistance selection and hydrogen-deuterium exchange experiments suggest JNJ-7184 binds RSV-L in the connector domain. JNJ-7184 prevents RSV replication and transcription by inhibiting initiation or early elongation. JNJ-7184 is effective in air-liquid interface cultures and therapeutically in neonatal lambs, acting to drastically reverse the appearance of lung pathology.


Assuntos
Antivirais , Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Replicação Viral , Antivirais/farmacologia , Antivirais/química , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/virologia , Animais , Humanos , Replicação Viral/efeitos dos fármacos , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Ovinos , Farmacorresistência Viral , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Proteínas Virais/genética , Pulmão/virologia
13.
Curr Top Med Chem ; 24(15): 1343-1358, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38698747

RESUMO

BACKGROUND: Human rhinovirus 3C protease (HRV-3Cpro) plays a crucial role in viral proliferation, establishing it as a prime target for antiviral therapy. However, research on identifying HRV-3Cpro inhibitors is still limited. OBJECTIVE: This study had two primary objectives: first, to validate the efficacy of an end-point colorimetric assay, previously developed by our team, for identifying potential inhibitors of HRV-3Cpro; and second, to discover phytochemicals in medicinal plants that inhibit the enzyme's activity. METHODS: Rupintrivir, a well-known inhibitor of HRV-3Cpro, was used to validate the colorimetric assay. Following this, we conducted a two-step in silico screening of 2532 phytochemicals, which led to the identification of eight active compounds: apigenin, carnosol, chlorogenic acid, kaempferol, luteolin, quercetin, rosmarinic acid, and rutin. We subsequently evaluated these candidates in vitro. To further investigate the inhibitory potential of the most promising candidates, namely, carnosol and rosmarinic acid, molecular docking studies were performed to analyze their binding interactions with HRV-3Cpro. RESULTS: The colorimetric assay we previously developed is effective in identifying compounds that selectively inhibit HRV-3Cpro. Carnosol and rosmarinic acid emerged as potent inhibitors, inhibiting HRV-3Cpro activity in vitro by over 55%. Our analysis indicated that carnosol and rosmarinic acid exert their inhibitory effects through a competitive mechanism. Molecular docking confirmed their competitive binding to the enzyme's active site. CONCLUSION: Carnosol and rosmarinic acid warrant additional investigation for their potential in the development of common cold treatment. By highlighting these compounds as effective HRV-3Cpro inhibitors, our study presents a promising approach for discovering phytochemical inhibitors against proteases from similar pathogens.


Assuntos
Proteases Virais 3C , Antivirais , Resfriado Comum , Simulação de Acoplamento Molecular , Compostos Fitoquímicos , Rhinovirus , Compostos Fitoquímicos/farmacologia , Compostos Fitoquímicos/química , Compostos Fitoquímicos/isolamento & purificação , Proteases Virais 3C/antagonistas & inibidores , Rhinovirus/enzimologia , Rhinovirus/efeitos dos fármacos , Humanos , Antivirais/farmacologia , Antivirais/química , Resfriado Comum/tratamento farmacológico , Resfriado Comum/virologia , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Estrutura Molecular , Relação Estrutura-Atividade , Inibidores de Proteases/farmacologia , Inibidores de Proteases/química
14.
J Med Chem ; 67(11): 8791-8816, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38775356

RESUMO

The spread of the influenza virus has caused devastating pandemics and huge economic losses worldwide. Antiviral drugs with diverse action modes are urgently required to overcome the challenges of viral mutation and drug resistance, and targeted protein degradation strategies constitute excellent candidates for this purpose. Herein, the first degradation of the influenza virus polymerase acidic (PA) protein using small-molecule degraders developed by hydrophobic tagging (HyT) technology to effectively combat the influenza virus was reported. The SAR results revealed that compound 19b with Boc2-(L)-Lys demonstrated excellent inhibitory activity against A/WSN/33/H1N1 (EC50 = 0.015 µM) and amantadine-resistant strain (A/PR/8/H1N1), low cytotoxicity, high selectivity, substantial degradation ability, and good drug-like properties. Mechanistic studies demonstrated that the proteasome system and autophagic lysosome pathway were the potential drivers of these HyT degraders. Thus, this study provides a powerful tool for investigating the targeted degradation of influenza virus proteins and for antiviral drug development.


Assuntos
Antivirais , Interações Hidrofóbicas e Hidrofílicas , Tioureia , Antivirais/farmacologia , Antivirais/química , Antivirais/síntese química , Humanos , Cães , Animais , Tioureia/farmacologia , Tioureia/análogos & derivados , Tioureia/química , Relação Estrutura-Atividade , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Células Madin Darby de Rim Canino , Proteólise/efeitos dos fármacos , Proteínas Virais/metabolismo , Proteínas Virais/química , Proteínas Virais/antagonistas & inibidores , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/metabolismo , Farmacorresistência Viral/efeitos dos fármacos
15.
IUCrJ ; 11(Pt 3): 374-383, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38656310

RESUMO

The large Bunyavirales order includes several families of viruses with a segmented ambisense (-) RNA genome and a cytoplasmic life cycle that starts by synthesizing viral mRNA. The initiation of transcription, which is common to all members, relies on an endonuclease activity that is responsible for cap-snatching. In La Crosse virus, an orthobunyavirus, it has previously been shown that the cap-snatching endonuclease resides in the N-terminal domain of the L protein. Orthobunyaviruses are transmitted by arthropods and cause diseases in cattle. However, California encephalitis virus, La Crosse virus and Jamestown Canyon virus are North American species that can cause encephalitis in humans. No vaccines or antiviral drugs are available. In this study, three known Influenza virus endonuclease inhibitors (DPBA, L-742,001 and baloxavir) were repurposed on the La Crosse virus endonuclease. Their inhibition was evaluated by fluorescence resonance energy transfer and their mode of binding was then assessed by differential scanning fluorimetry and microscale thermophoresis. Finally, two crystallographic structures were obtained in complex with L-742,001 and baloxavir, providing access to the structural determinants of inhibition and offering key information for the further development of Bunyavirales endonuclease inhibitors.


Assuntos
Antivirais , Endonucleases , Vírus La Crosse , Triazinas , Vírus La Crosse/efeitos dos fármacos , Vírus La Crosse/enzimologia , Antivirais/farmacologia , Antivirais/química , Endonucleases/antagonistas & inibidores , Endonucleases/metabolismo , Endonucleases/química , Dibenzotiepinas , Morfolinas/farmacologia , Morfolinas/química , Piridonas/farmacologia , Piridonas/química , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/química , Transferência Ressonante de Energia de Fluorescência , Humanos , Animais , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/química , Proteínas Virais/metabolismo
16.
J Virol Methods ; 323: 114838, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37914041

RESUMO

In influenza A virus-infected cells, newly synthesized viral neuraminidases (NAs) transiently localize at the host cell Golgi due to glycosylation, before their expression on the cell surface. It remains unproven whether Golgi-localized intracellular NAs exhibit sialidase activity. We have developed a sialidase imaging probe, [2-(benzothiazol-2-yl)-5-(non-1-yn-1-yl) phenyl]-α-D-N-acetylneuraminic acid (BTP9-Neu5Ac). This probe is designed to be cleaved by sialidase activity, resulting in the release of a hydrophobic fluorescent compound, 2-(benzothiazol-2-yl)-5-(non-1-yn-1-yl) phenol (BTP9). BTP9-Neu5Ac makes the location of sialidase activity visually detectable by the BTP9 fluorescence that results from the action of sialidase activity. In this study, we established a protocol to visualize the sialidase activity of intracellular NA at the Golgi of influenza A virus-infected cells using BTP9-Neu5Ac. Furthermore, we employed this fluorescence imaging protocol to elucidate the intracellular inhibition of laninamivir octanoate, an anti-influenza drug. At approximately 7 h after infection, newly synthesized viral NAs localized at the Golgi. Using our developed protocol, we successfully histochemically stained the sialidase activity of intracellular viral NAs localized at the Golgi. Importantly, we observed that laninamivir octanoate effectively inhibited the intracellular viral NA, in contrast to drugs like zanamivir or laninamivir. Our study establishes a visualization protocol for intracellular viral NA sialidase activity and visualizes the inhibitory effect of laninamivir octanoate on Golgi-localized intracellular viral NA in infected cells.


Assuntos
Antivirais , Inibidores Enzimáticos , Vírus da Influenza A , Neuraminidase , Proteínas Virais , Humanos , Antivirais/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/enzimologia , Neuraminidase/análise , Neuraminidase/antagonistas & inibidores , Imagem Óptica/métodos , Zanamivir/farmacologia , Proteínas Virais/análise , Proteínas Virais/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia
17.
J Virol ; 97(10): e0078623, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37796126

RESUMO

IMPORTANCE: EV71 poses a significant health threat to children aged 5 and below. The process of EV71 infection and replication is predominantly influenced by ubiquitination modifications. Our previous findings indicate that EV71 prompts the activation of host deubiquitinating enzymes, thereby impeding the host interferon signaling pathway as a means of evading the immune response. Nevertheless, the precise mechanisms by which the host employs ubiquitination modifications to hinder EV71 infection remain unclear. The present study demonstrated that the nonstructural protein 2Apro, which is encoded by EV71, exhibits ubiquitination and degradation mediated by the host E3 ubiquitin ligase SPOP. In addition, it is the first report, to our knowledge, that SPOP is involved in the host antiviral response.


Assuntos
Cisteína Endopeptidases , Enterovirus Humano A , Infecções por Enterovirus , Interações entre Hospedeiro e Microrganismos , Ubiquitina-Proteína Ligases , Ubiquitina , Ubiquitinação , Proteínas Virais , Criança , Humanos , Enterovirus Humano A/enzimologia , Enterovirus Humano A/fisiologia , Infecções por Enterovirus/metabolismo , Infecções por Enterovirus/virologia , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Cisteína Endopeptidases/metabolismo
18.
J Virol ; 97(10): e0060223, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37754760

RESUMO

IMPORTANCE: Influenza A viruses (IAVs) contain hemagglutinin (HA) proteins involved in sialoglycan receptor binding and neuraminidase (NA) proteins that cleave sialic acids. While the importance of the NA protein in virion egress is well established, its role in virus entry remains to be fully elucidated. NA activity is needed for the release of virions from mucus decoy receptors, but conflicting results have been reported on the importance of NA activity in virus entry in the absence of decoy receptors. We now show that inhibition of NA activity affects virus entry depending on the receptor-binding properties of HA and the receptor repertoire present on cells. Inhibition of entry by the presence of mucus correlated with the importance of NA activity for virus entry, with the strongest inhibition being observed when mucus and OsC were combined. These results shed light on the importance in virus entry of the NA protein, an important antiviral drug target.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vírus da Influenza A , Neuraminidase , Receptores Virais , Proteínas Virais , Internalização do Vírus , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Vírus da Influenza A/enzimologia , Vírus da Influenza A/metabolismo , Influenza Humana/enzimologia , Influenza Humana/metabolismo , Neuraminidase/antagonistas & inibidores , Neuraminidase/metabolismo , Ligação Proteica , Receptores Virais/metabolismo , Especificidade por Substrato , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Linhagem Celular , Muco
19.
Carbohydr Res ; 532: 108918, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37586142

RESUMO

A triazolylsialoside-human serum albumin conjugate was prepared as a multivalent hemagglutinin and neuraminidase inhibitor using a di-(N-succinimidyl) adipate strategy. Matrix-Assisted Laser Desorption/Ionization-Time of Flight-Mass Spectrometry (MALDI-TOF-MS) indicated that five tetravalent sialyl galactosides were grafted onto the protein backbone resulting in an eicosavalent triazolylsialoside-protein complex. Compared with monomeric sialic acid, molecular interaction studies showed that the synthetic pseudo-glycoprotein bound tightly not only to hemagglutinin (HA)/neuraminidase (NA) but also to mutated drug-resistant NA on the surface of the influenza virus with a dissociation constant (KD) in the 1 µM range, attributed to the cluster effect. Moreover, this glycoconjugate exhibited potent antiviral activity against a broad spectrum of virus strains and showed no cytotoxicity towards Human Umbilical Vein Endothelial Cells (HUVECs) and Madin-Darby canine kidney (MDCK) cells at high concentrations. Further mechanistic studies demonstrated this multivalent sialyl conjugate showed strong capture and trapping of influenza virions, thus disrupting the ability of the influenza virus to infect host cells. This research lays the experimental foundation for the development of new antiviral agents based on multivalent sialic acid-protein conjugates.


Assuntos
Influenza Humana , Animais , Cães , Humanos , Antivirais/química , Células Endoteliais/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Hemaglutininas/metabolismo , Células Madin Darby de Rim Canino , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/metabolismo , Albumina Sérica Humana , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/química , Proteínas Virais/metabolismo , Vírion/metabolismo
20.
Antiviral Res ; 217: 105673, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37478917

RESUMO

Human cytomegalovirus (HCMV) can cause serious diseases in immunocompromised patients. Use of current antivirals is limited by their adverse effects and emergence of drug resistance mutations. Thus, new drugs are an urgent need. The terminase complex (pUL56-pUL89-pUL51) represents a target of choice for new antivirals development. pUL51 was shown to be crucial for the cleavage of concatemeric HCMV DNA and viral replication. Its C-terminal part plays a critical role for the terminase complex assembly. However, no interaction domain is clearly identified. Sequence comparison of herpesvirus homologs and protein modelling were performed on pUL51. Importance of a putative interaction domain is validated by the generation of recombinant viruses with specific alanine substitutions of amino acids implicated in the domain. We identified a Leucine-Zipper (LZ) domain involving the leucine residues L126-X6-L133-X6-L140-X6-L147 in C-terminal part of pUL51. These leucines are crucial for viral replication, suggesting the significance for pUL51 structure and function. A mimetic-peptide approach has been used and tested in antiviral assays to validate the interaction domain as a new therapeutic target. Cytotoxicity was evaluated by LDH release measurement. The peptide TAT-HK29, homologous to the pUL51-LZ domain, inhibits HCMV replication by 27% ± 9% at 1.25 µM concentration without cytotoxicity. Our results highlight the importance of a leucine zipper domain in the C-terminal part of pUL51 involving leucines L126, L133, L140 and L147. We also confirm the potential of mimetic peptides to inhibit HCMV replication and the importance to target interaction domains to develop antiviral agents.


Assuntos
Antivirais , Materiais Biomiméticos , Citomegalovirus , Endodesoxirribonucleases , Zíper de Leucina , Proteínas Virais , Replicação Viral , Replicação Viral/efeitos dos fármacos , Citomegalovirus/efeitos dos fármacos , Antivirais/química , Antivirais/farmacologia , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/química , Proteínas Virais/genética , Desenvolvimento de Medicamentos , Endodesoxirribonucleases/antagonistas & inibidores , Endodesoxirribonucleases/química , Humanos , Peptídeos/química , Peptídeos/farmacologia , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...