RESUMEN
During infection, the hepatitis C virus (HCV) can evade immune response and cause chronic disease. Formation of effective T-cell response is important for the control of HCV infection. Dendritic cells derived from peripheral blood monocytes activated by immunodominant epitopes of the pathogen can effectively stimulate T-lymphocytes. Previously, we obtained recombinant proteins containing cytotoxic T-lymphocyte epitopes of NS3 and NS4ab proteins of HCV, the T-helper epitope PADRE, and self-assembling peptides that cause the formation of nanoparticles. Here, we studied the activation of human dendritic cells isolated from peripheral blood monocytes and from monocytes derived from induced pluripotent stem cells. Both types of dendritic cells effectively respond to activation by recombinant HCV proteins and stimulated lymphocytes along the Th1 pathway. Recombinant nanoparticles induced more efficient responses. These results open prospects for immunotherapy of patients with chronic hepatitis C using activated dendritic cells derived from their induced pluripotent stem cells.
Asunto(s)
Células Dendríticas , Hepacivirus , Células Madre Pluripotentes Inducidas , Activación de Linfocitos , Células Dendríticas/inmunología , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/citología , Hepacivirus/inmunología , Activación de Linfocitos/inmunología , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/genética , Monocitos/inmunología , Epítopos de Linfocito T/inmunología , Proteínas Recombinantes/inmunología , Linfocitos T/inmunología , Diferenciación Celular/inmunología , Antígenos de la Hepatitis C/inmunología , Células TH1/inmunología , Hepatitis C Crónica/inmunología , Hepatitis C Crónica/virología , Proteasas Virales , Serina Endopeptidasas , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
The tick-borne encephalitis virus (TBEV) strain C11-13 (GenBank acc. no. OQ565596) of the Siberian genotype was previously isolated from the brain of a deceased person. TBEV C11-13 variants obtained at passages 3 and 8 in SPEV cells were inoculated into the brains of white mice for subsequent passages. Full genome sequences of all virus variants were analyzed by high-throughput sequencing. A total of 41 single nucleotide substitutions were found to occur mainly in the genes for the nonstructural proteins NS3 and NS5 (GenBank MF043953, OP902894, and OP902895), and 12 amino acid substitutions were identified in the deduced protein sequences. Reverse nucleotide and amino acid substitutions were detected after three passages through mouse brains. The substitutions restored the primary structures that were characteristic of the isolate C11-13 from a human patient and changed during the eight subsequent passages in SPEV cells. In addition, the 3'-untranslated region (3'-UTR) of the viral genome increased by 306 nt. The Y3 and Y2 3'-UTR elements were found to contain imperfect L and R repeats, which were probably associated with inhibition of cellular XRN1 RNase and thus involved in the formation of subgenomic flaviviral RNAs (sfRNAs). All TBEV variants showed high-level reproduction in both cell cultures and mouse brains. The genomic changes that occurred during successive passages of TBEV are most likely due to its significant genetic variability, which ensures its efficient reproduction in various hosts and its broad distribution in various climatic zones.
Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Genoma Viral , Proteínas no Estructurales Virales , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Animales , Ratones , Humanos , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Regiones no Traducidas 3'/genética , Encefalitis Transmitida por Garrapatas/virología , Encefalitis Transmitida por Garrapatas/genética , Sustitución de Aminoácidos , Cultivo de Virus/métodos , Encéfalo/virología , Encéfalo/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Línea Celular , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
It is believed that DNA double-strand breaks induced by Zika virus (ZIKV) infection in pregnant women is a main reason of brain damage (e.g. microcephaly, severe brain malformation, and neuropathy) in newborn babies [1,2], but its underlying mechanism is poorly understood. In this study, we report that the depletion of ERp57, a member of the protein disulphide isomerase (PDI) family, leads to the limited production of ZIKV in nerve cells. ERp57 knockout not only suppresses viral induced reactive oxygen species (ROS) mediated host DNA damage, but also decreases apoptosis. Strikingly, DNA damage depends on ERp57-bridged complex formation of viral protein NS2B/NS3. LOC14, an ERp57 inhibitor, restricts ZIKV infection and virus-induced DNA damage. Our work reveals an important role of ERp57 in both ZIKV propagation and virus-induced DNA damage, suggesting a potential target against ZIKV infection.
Asunto(s)
Daño del ADN , Proteína Disulfuro Isomerasas , Proteínas no Estructurales Virales , Infección por el Virus Zika , Virus Zika , Virus Zika/genética , Virus Zika/fisiología , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Humanos , Proteína Disulfuro Isomerasas/metabolismo , Proteína Disulfuro Isomerasas/genética , Infección por el Virus Zika/virología , Infección por el Virus Zika/metabolismo , Animales , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Apoptosis , Especies Reactivas de Oxígeno/metabolismo , Células Vero , Chlorocebus aethiops , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
The emergence of drug-resistance-inducing mutations in Hepatitis C virus (HCV) coupled with genotypic heterogeneity has made targeting NS3/4A serine protease difficult. In this work, we investigated the mutagenic variations in the binding pocket of Genotype 3 (G3) HCV NS3/4A and evaluated ligands for efficacious inhibition. We report mutations at 14 positions within the ligand-binding residues of HCV NS3/4A, including H57R and S139P within the catalytic triad. We then modelled each mutational variant for pharmacophore-based virtual screening (PBVS) followed by covalent docking towards identifying a potential covalent inhibitor, i.e., cpd-217. The binding stability of cpd-217 was then supported by molecular dynamic simulation followed by MM/GBSA binding free energy calculation. The free energy decomposition analysis indicated that the resistant mutants alter the HCV NS3/4A-ligand interaction, resulting in unbalanced energy distribution within the binding site, leading to drug resistance. Cpd-217 was identified as interacting with all NS3/4A G3 variants with significant covalent docking scores. In conclusion, cpd-217 emerges as a potential inhibitor of HCV NS3/4A G3 variants that warrants further in vitro and in vivo studies. This study provides a theoretical foundation for drug design and development targeting HCV G3 NS3/4A.
Asunto(s)
Antivirales , Farmacorresistencia Viral , Genotipo , Hepacivirus , Simulación del Acoplamiento Molecular , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Hepacivirus/genética , Hepacivirus/enzimología , Hepacivirus/efectos de los fármacos , Farmacorresistencia Viral/genética , Antivirales/farmacología , Antivirales/química , Humanos , Mutación , Simulación de Dinámica Molecular , Hepatitis C/virología , Hepatitis C/tratamiento farmacológico , Sitios de Unión , Unión Proteica , Farmacóforo , Serina Proteasas , Proteasas Virales , ARN Helicasas DEAD-box , Nucleósido-Trifosfatasa , Serina EndopeptidasasRESUMEN
West Nile virus (WNV) nonstructural protein 5 (NS5) possesses multiple enzymatic domains essential for viral RNA replication. During infection, NS5 predominantly localizes to unique replication organelles (ROs) at the rough endoplasmic reticulum (RER), known as vesicle packets (VPs) and convoluted membranes (CMs), with a portion of NS5 accumulating in the nucleus. NS5 is a soluble protein that must be in the VP, where its enzymatic activities are required for viral RNA synthesis. However, the mechanistic processes behind the recruitment of NS5 from the cytoplasm to the RER membrane remain unclear. Here, we utilize high-resolution confocal microscopy and sucrose density gradient ultracentrifugation to investigate whether the association of NS5 with other NS proteins contributes to its membrane recruitment and retention. We demonstrate that NS1 or NS3 partially influences the NS5 association with the membrane. We further demonstrate that processed NS5 is predominantly in the cytoplasm and nucleus, indicating that the processing of NS5 from the viral polyprotein does not contribute to its membrane localization. These observations suggest that other host or viral factors, such as the enwrapment of NS5 by the RO, may also be necessary for the complete membrane retention of NS5. Therefore, studies on the inhibitors that disrupt the membrane localization of WNV NS5 are warranted for antiviral drug development.
Asunto(s)
Proteínas no Estructurales Virales , Virus del Nilo Occidental , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Virus del Nilo Occidental/enzimología , Virus del Nilo Occidental/fisiología , Humanos , Animales , Replicación Viral , ARN Helicasas/metabolismo , ARN Helicasas/genética , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Chlorocebus aethiops , Citoplasma/metabolismo , Células Vero , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Fiebre del Nilo Occidental/virología , Línea Celular , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
Microcephaly, Guillain-Barré syndrome, and potential sexual transmission stand as prominent complications associated with Zika virus (ZIKV) infection. The absence of FDA-approved drugs or vaccines presents a substantial obstacle in combatting the virus. Furthermore, the inclusion of pregnancy in the pharmacological screening process complicates and extends the endeavor to ensure molecular safety and minimal toxicity. Given its pivotal role in viral assembly and maturation, the NS2B-NS3 viral protease emerges as a promising therapeutic target against ZIKV. In this context, a dipeptide inhibitor was specifically chosen as a control against 200 compounds for docking analysis. Subsequent molecular dynamics simulations extending over 200 ns were conducted to ascertain the stability of the docked complex and confirm the binding of the inhibitor at the protein's active site. The simulation outcomes exhibited conformity to acceptable thresholds, encompassing parameters such as root mean square deviation (RMSD), root mean square fluctuation (RMSF), ligand-protein interaction analysis, ligand characterization, and surface area analysis. Notably, analysis of ligand angles bolstered the identification of prospective ligands capable of inhibiting viral protein activity and impeding virus dissemination. In this study, the integration of molecular docking and dynamics simulations has pinpointed the dipeptide inhibitor as a potential candidate ligand against ZIKV protease, thereby offering promise for therapeutic intervention against the virus.
Asunto(s)
Dipéptidos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Inhibidores de Proteasas , Proteínas no Estructurales Virales , Virus Zika , Virus Zika/enzimología , Virus Zika/efectos de los fármacos , Dipéptidos/química , Dipéptidos/farmacología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Antivirales/farmacología , Antivirales/química , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Humanos , Unión Proteica , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
Zika virus (ZIKV) infection may lead to severe neurological consequences, including seizures, and early infancy death. However, the involved mechanisms are still largely unknown. TRPC channels play an important role in regulating nervous system excitability and are implicated in seizure development. We investigated whether TRPCs might be involved in the pathogenesis of ZIKV infection. We found that ZIKV infection increases TRPC4 expression in host cells via the interaction between the ZIKV-NS3 protein and CaMKII, enhancing TRPC4-mediated calcium influx. Pharmacological inhibition of CaMKII decreased both pCREB and TRPC4 protein levels, whereas the suppression of either TRPC4 or CaMKII improved the survival rate of ZIKV-infected cells and reduced viral protein production, likely by impeding the replication phase of the viral life cycle. TRPC4 or CaMKII inhibitors also reduced seizures and increased the survival of ZIKV-infected neonatal mice and blocked the spread of ZIKV in brain organoids derived from human-induced pluripotent stem cells. These findings suggest that targeting CaMKII or TRPC4 may offer a promising approach for developing novel anti-ZIKV therapies, capable of preventing ZIKV-associated seizures and death.
Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Canales Catiónicos TRPC , Infección por el Virus Zika , Virus Zika , Infección por el Virus Zika/virología , Infección por el Virus Zika/metabolismo , Animales , Humanos , Virus Zika/fisiología , Virus Zika/efectos de los fármacos , Ratones , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Células HEK293 , Proteínas Virales/metabolismo , Convulsiones/virología , Convulsiones/metabolismo , Convulsiones/tratamiento farmacológico , Proteasas Virales , Serina Endopeptidasas , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
For the rational design of epitope-specific vaccines, identifying epitopes that can be processed and presented is essential. As algorithm-based epitope prediction is frequently discordant with actually recognized CD8+ T-cell epitopes, we developed an in vitro CD8 T-cell priming protocol to enable the identification of truly and functionally expressed HLA class I epitopes. The assay was established and validated to identify epitopes presented by hepatitis C virus (HCV)-infected cells. In vitro priming of naïve CD8 T cells was achieved by culturing unfractionated PBMCs in the presence of a specific cocktail of growth factors and cytokines, and next exposing the cells to hepatic cells expressing the NS3 protein of HCV. After a 10-day co-culture, HCV-specific T-cell responses were identified based on IFN-γ ELISpot analysis. For this, the T cells were restimulated with long synthetic peptides (SLPs) spanning the whole NS3 protein sequence allowing the identification of HCV-specificity. We demonstrated that this protocol resulted in the in vitro priming of naïve precursors to antigen-experienced T-cells specific for 11 out of 98 SLPs tested. These 11 SLPs contain 12 different HLA-A*02:01-restricted epitopes, as predicted by a combination of three epitope prediction algorithms. Furthermore, we identified responses against 3 peptides that were not predicted to contain any immunogenic HLA class I epitopes, yet showed HCV-specific responses in vitro. Separation of CD8+ and CD8- T cells from PBMCs primed in vitro showed responses only upon restimulation with short peptides. We established an in vitro method that enables the identification of HLA class I epitopes resulting from cross-presented antigens and that can cross-prime T cells and allows the effective selection of functional immunogenic epitopes, but also less immunogenic ones, for the design of tailored therapeutic vaccines against persistent viral infections and tumor antigens.
Asunto(s)
Linfocitos T CD8-positivos , Epítopos de Linfocito T , Hepacivirus , Vacunas contra Hepatitis Viral , Proteínas no Estructurales Virales , Linfocitos T CD8-positivos/inmunología , Humanos , Epítopos de Linfocito T/inmunología , Hepacivirus/inmunología , Hepacivirus/genética , Vacunas contra Hepatitis Viral/inmunología , Proteínas no Estructurales Virales/inmunología , Hepatitis C/inmunología , Hepatitis C/prevención & control , Ensayo de Immunospot Ligado a Enzimas/métodos , Antígeno HLA-A2/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Proteasas Virales , Serina Endopeptidasas , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
BACKGROUND: Peptide drugs are advantageous because they are subject to rational design and exhibit highly diverse structures and broad biological activities. The NS2B-NS3 protein is a particularly promising flavivirus therapeutic target, with extensive research on the development of inhibitors as therapeutic candidates, and was used as a model in this work to determine the mechanism by which GA-Hecate inhibits ZIKV replication. OBJECTIVE: The present study aimed to evaluate the potential of GA-Hecate, a new antiviral developed by our group, against the Brazilian Zika virus and to evaluate the mechanism of action of this compound on the flavivirus NS2B-NS3 protein. METHODS: Solid-phase peptide Synthesis, High-Performance Liquid Chromatography, and Mass Spectrometry were used to obtain, purify, and characterize the synthesized compound. Real-time and enzymatic assays were used to determine the antiviral potential of GA-Hecate against ZIKV. RESULTS: The RT-qPCR results showed that GA-Hecate decreased the number of ZIKV RNA copies in the virucidal, pre-treatment, and post-entry assays, with 5- to 6-fold fewer RNA copies at the higher nontoxic concentration in Vero cells (HNTC: 10 µM) than in the control cells. Enzymatic and kinetic assays indicated that GA-Hecate acts as a competitive ZIKV NS2B-NS3 protease inhibitor with an IC50 of 32 nM and has activity against the yellow fever virus protease. CONCLUSION: The results highlight the antiviral potential of the GA-Hecate bioconjugate and open the door for the development of new antivirals.
Asunto(s)
Antivirales , Proteínas no Estructurales Virales , Replicación Viral , Virus Zika , Virus Zika/efectos de los fármacos , Antivirales/farmacología , Antivirales/química , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Chlorocebus aethiops , Células Vero , Replicación Viral/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Péptidos/farmacología , Péptidos/química , ARN Helicasas/metabolismo , ARN Helicasas/antagonistas & inhibidores , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/virología , Humanos , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
Dengue fever represents a significant medical and socio-economic burden in (sub)tropical regions, yet antivirals for treatment or prophylaxis are lacking. JNJ-A07 was described as highly active against the different genotypes within each serotype of the disease-causing dengue virus (DENV). Based on clustering of resistance mutations it has been assumed to target DENV non-structural protein 4B (NS4B). Using a photoaffinity labeling compound with high structural similarity to JNJ-A07, here we demonstrate binding to NS4B and its precursor NS4A-2K-NS4B. Consistently, we report recruitment of the compound to intracellular sites enriched for these proteins. We further specify the mechanism-of-action of JNJ-A07, which has virtually no effect on viral polyprotein cleavage, but targets the interaction between the NS2B/NS3 protease/helicase complex and the NS4A-2K-NS4B cleavage intermediate. This interaction is functionally linked to de novo formation of vesicle packets (VPs), the sites of DENV RNA replication. JNJ-A07 blocks VPs biogenesis with little effect on established ones. A similar mechanism-of-action was found for another NS4B inhibitor, NITD-688. In summary, we unravel the antiviral mechanism of these NS4B-targeting molecules and show how DENV employs a short-lived cleavage intermediate to carry out an early step of the viral life cycle.
Asunto(s)
Antivirales , Virus del Dengue , Dengue , Proteínas no Estructurales Virales , Replicación Viral , Virus del Dengue/efectos de los fármacos , Virus del Dengue/genética , Virus del Dengue/fisiología , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Antivirales/farmacología , Humanos , Dengue/virología , Dengue/tratamiento farmacológico , Serogrupo , ARN Helicasas/metabolismo , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/genética , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Unión Proteica , Animales , Orgánulos/metabolismo , Orgánulos/efectos de los fármacos , Proteasas Virales , Aminofenoles , Proteínas de la Membrana , Indoles , ARN Helicasas DEAD-box , Nucleósido-Trifosfatasa , ButiratosRESUMEN
Viruses have developed sophisticated strategies to control metabolic activity of infected cells in order to supply replication machinery with energy and metabolites. Dengue virus (DENV), a mosquito-borne flavivirus responsible for dengue fever, is no exception. Previous reports have documented DENV interactions with metabolic pathways and shown in particular that glycolysis is increased in DENV-infected cells. However, underlying molecular mechanisms are still poorly characterized and dependence of DENV on this pathway has not been investigated in details yet. Here, we identified an interaction between the non-structural protein 3 (NS3) of DENV and glucokinase regulator protein (GCKR), a host protein that inhibits the liver-specific hexokinase GCK. NS3 expression was found to increase glucose consumption and lactate secretion in hepatic cell line expressing GCK. Interestingly, we observed that GCKR interaction with GCK decreases DENV replication, indicating the dependence of DENV to GCK activity and supporting the role of NS3 as an inhibitor of GCKR function. Accordingly, in the same cells, DENV replication both induces and depends on glycolysis. By targeting NAD(H) biosynthesis with the antimetabolite 6-Amino-Nicotinamide (6-AN), we decreased cellular glycolytic activity and inhibited DENV replication in hepatic cells. Infection of primary organotypic liver cultures (OLiC) from hamsters was also inhibited by 6-AN. Altogether, our results show that DENV has evolved strategies to control glycolysis in the liver, which could account for hepatic dysfunctions associated to infection. Besides, our findings suggest that lowering intracellular availability of NAD(H) could be a valuable therapeutic strategy to control glycolysis and inhibit DENV replication in the liver.
Asunto(s)
Virus del Dengue , Dengue , Glucoquinasa , Glucólisis , NAD , Proteínas no Estructurales Virales , Replicación Viral , Glucólisis/efectos de los fármacos , Virus del Dengue/efectos de los fármacos , Glucoquinasa/metabolismo , Glucoquinasa/antagonistas & inhibidores , Humanos , Replicación Viral/efectos de los fármacos , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Animales , Dengue/tratamiento farmacológico , Dengue/virología , Dengue/metabolismo , NAD/metabolismo , NAD/biosíntesis , Línea Celular , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Glucosa/metabolismo , Hígado/virología , Hígado/metabolismo , Antivirales/farmacología , Proteasas Virales , Serina Endopeptidasas , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
The Orthoflavivirus NS3 helicase (NS3h) is crucial in virus replication, representing a potential drug target for pathogenesis. NS3h utilizes nucleotide triphosphate (ATP) for hydrolysis energy to translocate on single-stranded nucleic acids, which is an important step in the unwinding of double-stranded nucleic acids. Intermediate states along the ATP hydrolysis cycle and conformational changes between these states, represent important yet difficult-to-identify targets for potential inhibitors. Extensive molecular dynamics simulations of West Nile virus NS3h+ssRNA in the apo, ATP, ADP+Pi and ADP bound states were used to model the conformational ensembles along this cycle. Energetic and structural clustering analyses depict a clear trend of differential enthalpic affinity of NS3h with ADP, demonstrating a probable mechanism of hydrolysis turnover regulated by the motif-VI loop (MVIL). Based on these results, MVIL mutants (D471L, D471N and D471E) were found to have a substantial reduction in ATPase activity and RNA replication compared to the wild-type. Simulations of the mutants in the apo state indicate a shift in MVIL populations favoring either a closed or open 'valve' conformation, affecting ATP entry or stabilization, respectively. Combining our molecular modeling with experimental evidence highlights a conformation-dependent role for MVIL as a 'valve' for the ATP-pocket, presenting a promising target for antiviral development.
Asunto(s)
Adenosina Trifosfato , Simulación de Dinámica Molecular , ARN Helicasas , Proteínas no Estructurales Virales , Virus del Nilo Occidental , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Virus del Nilo Occidental/enzimología , Virus del Nilo Occidental/genética , ARN Helicasas/metabolismo , ARN Helicasas/química , ARN Helicasas/genética , Adenosina Trifosfato/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Difosfato/química , Secuencias de Aminoácidos , Mutación , Nucleótidos/metabolismo , Nucleótidos/química , Hidrólisis , Replicación Viral/genética , Conformación Proteica , Proteasas Virales , Serina Endopeptidasas , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
Zika virus (ZIKV) continues to pose a significant global public health threat, with recurring regional outbreaks and potential for pandemic spread. Despite often being asymptomatic, ZIKV infections can have severe consequences, including neurological disorders and congenital abnormalities. Unfortunately, there are currently no approved vaccines or antiviral drugs for the prevention or treatment of ZIKV. One promising target for drug development is the ZIKV NS2B-NS3 protease due to its crucial role in the virus life cycle. In this study, we established a cell-based ZIKV protease inhibition assay designed for high-throughput screening (HTS). Our assay relies on the ZIKV protease's ability to cleave a cyclised firefly luciferase fused to a natural cleavage sequence between NS2B and NS3 protease within living cells. We evaluated the performance of our assay in HTS setting using the pharmacologic controls (JNJ-40418677 and MK-591) and by screening a Library of Pharmacologically Active Compounds (LOPAC). The results confirmed the feasibility of our assay for compound library screening to identify potential ZIKV protease inhibitors.
Asunto(s)
Antivirales , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Inhibidores de Proteasas , Infección por el Virus Zika , Virus Zika , Virus Zika/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Inhibidores de Proteasas/farmacología , Humanos , Antivirales/farmacología , Descubrimiento de Drogas/métodos , Infección por el Virus Zika/virología , Infección por el Virus Zika/tratamiento farmacológico , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Animales , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
Three new series of macrocyclic active site-directed inhibitors of the Zika virus (ZIKV) NS2B-NS3 protease were synthesized. First, attempts were made to replace the basic P3 lysine residue of our previously described inhibitors with uncharged and more hydrophobic residues. This provided numerous compounds with inhibition constants between 30 and 50 nM. A stronger reduction of the inhibitory potency was observed when the P2 lysine was replaced by neutral residues, all of these inhibitors possess Ki values >1 µM. However, it is possible to replace the P2 lysine with the less basic 3-aminomethylphenylalanine, which provides a similarly potent inhibitor of the ZIKV protease (Ki = 2.69 nM). Crystal structure investigations showed that the P2 benzylamine structure forms comparable interactions with the protease as lysine. Twelve additional structures of these inhibitors in complex with the protease were determined, which explain many, but not all, SAR data obtained in this study. All individual modifications in the P2 or P3 position resulted in inhibitors with low antiviral efficacy in cell culture. Therefore, a third inhibitor series with combined modifications was synthesized; all of them contain a more hydrophobic d-cyclohexylalanine in the linker segment. At a concentration of 40 µM, two of these compounds possess similar antiviral potency as ribavirin at 100 µM. Due to their reliable crystallization in complex with the ZIKV protease, these cyclic compounds are very well suited for a rational structure-based development of improved inhibitors.
Asunto(s)
Antivirales , Compuestos Macrocíclicos , Virus Zika , Virus Zika/enzimología , Virus Zika/efectos de los fármacos , Relación Estructura-Actividad , Compuestos Macrocíclicos/farmacología , Compuestos Macrocíclicos/síntesis química , Compuestos Macrocíclicos/química , Antivirales/farmacología , Antivirales/síntesis química , Antivirales/química , Estructura Molecular , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Relación Dosis-Respuesta a Droga , Serina Endopeptidasas/metabolismo , Humanos , Inhibidores de Proteasa Viral/farmacología , Inhibidores de Proteasa Viral/síntesis química , Inhibidores de Proteasa Viral/química , Cristalografía por Rayos X , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
Flaviviruses can cause severe illness in humans. Effective and safe vaccines are available for some species; however, for many flaviviruses disease prevention or specific treatments remain unavailable. The viral replication cycle depends on the proteolytic activity of the NS2B-NS3 protease, which releases functional viral proteins from a non-functional polyprotein precursor, rendering the protease a promising drug target. In this study, we characterised recombinant NS2B-NS3 proteases from ten flaviviruses including three unreported proteases from the Usutu, Kyasanur forest disease and Powassan viruses. All protease constructs comprise a covalent Gly4-Ser-Gly4 linker connecting the NS3 serine protease domain with its cofactor NS2B. We conducted a comprehensive cleavage site analysis revealing areas of high conversion. While all proteases were active in enzymatic assays, we noted a 1000-fold difference in catalytic efficiency across proteases from different flaviviruses. Two bicyclic peptide inhibitors displayed anti-pan-flaviviral protease activity with inhibition constants ranging from 10 to 1000 nM.
Asunto(s)
Antivirales , Flavivirus , Serina Endopeptidasas , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Flavivirus/efectos de los fármacos , Flavivirus/enzimología , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/química , Antivirales/farmacología , Antivirales/química , Humanos , ARN Helicasas/metabolismo , ARN Helicasas/química , ARN Helicasas/genética , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/química , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-boxRESUMEN
In the global pandemic scenario, dengue and zika viruses (DENV and ZIKV, respectively), both mosquito-borne members of the flaviviridae family, represent a serious health problem, and considering the absence of specific antiviral drugs and available vaccines, there is a dire need to identify new targets to treat these types of viral infections. Within this drug discovery process, the protease NS2B/NS3 is considered the primary target for the development of novel anti-flavivirus drugs. The NS2B/NS3 is a serine protease that has a dual function both in the viral replication process and in the elusion of the innate immunity. To date, two main classes of NS2B/NS3 of DENV and ZIKV protease inhibitors have been discovered: those that bind to the orthosteric site and those that act at the allosteric site. Therefore, this perspective article aims to discuss the main features of the use of the most potent NS2B/NS3 inhibitors and their impact at the social level.
Asunto(s)
Antivirales , Dengue , Inhibidores de Proteasas , Infección por el Virus Zika , Animales , Humanos , Antivirales/uso terapéutico , Antivirales/farmacología , ARN Helicasas DEAD-box , Dengue/tratamiento farmacológico , Dengue/virología , Virus del Dengue/efectos de los fármacos , Nucleósido-Trifosfatasa , Inhibidores de Proteasas/uso terapéutico , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/química , Proteasas Virales , Virus Zika/efectos de los fármacos , Virus Zika/enzimología , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/virologíaRESUMEN
Hepatitis C virus (HCV) is the most common infection worldwide. The correlation between HCV and renal cell carcinoma (RCC) is still mysterious. Therefore, the relationship between HCV and RCC was investigated. The study included 100 patients with RCC; 32 with HCV infection, and 68 without HCV infection. Expressions of viral proteins (NS3 and NS5A) were tested using an immune electron-microscope (IEM) and immunohistochemistry (IHC). IHC and quantitative real time-PCR investigated the presentation of human proteins TP53 and p21 genes. Transmission electron (TEM) detected viral-like particles in infected RCC tissues. The gene and protein expression of P53 was higher in HCV positive versus HCV negative patients and p21 was lower in HCV positive versus HCV negative in both tumor and normal tissue samples. Viral like particles were observed by TEM in the infected tumor and normal portion of the RCC tissues and the plasma samples. The IEM showed the depositions of NS3 and NS5A in infected renal tissues, while in noninfected samples, were not observed. The study hypothesizes that a correlation between HCV and RCC could exist through successfully detecting HCV-like particles, HCV proteins, and (p53 and p21) in RCC-infected patients.
Asunto(s)
Carcinoma de Células Renales , Genotipo , Hepacivirus , Neoplasias Renales , Proteína p53 Supresora de Tumor , Proteínas no Estructurales Virales , Humanos , Carcinoma de Células Renales/virología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Hepacivirus/genética , Proteínas no Estructurales Virales/genética , Neoplasias Renales/virología , Neoplasias Renales/patología , Neoplasias Renales/genética , Masculino , Proteína p53 Supresora de Tumor/genética , Femenino , Persona de Mediana Edad , Hepatitis C/virología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Anciano , Adulto , Inmunohistoquímica , Proteasas Virales , ARN Polimerasa Dependiente del ARN , ARN Helicasas DEAD-box , Nucleósido-Trifosfatasa , Serina EndopeptidasasRESUMEN
Current management of HCV infection is based on Direct-Acting Antiviral Drugs (DAAs). However, resistance-associated mutations, especially in the NS3 and NS5B regions are gradually decreasing the efficacy of DAAs. Among the most effective HCV NS3/4A protease drugs, Sofosbuvir also develops resistance due to mutations in the NS3 and NS5B regions. Four mutations at positions A156Y, L36P, Q41H, and Q80K are classified as high-level resistance mutations. The resistance mechanism of HCV NS3/4A protease toward Sofosbuvir caused by these mutations is still unclear, as there is less information available regarding the structural and functional effects of the mutations against Sofosbuvir. In this work, we combined molecular dynamics simulation, molecular mechanics/Generalized-Born surface area calculation, principal component analysis, and free energy landscape analysis to explore the resistance mechanism of HCV NS3/4A protease due to these mutations, as well as compare interaction changes in wild-type. Subsequently, we identified that the mutant form of HCV NS3/4A protease affects the activity of Sofosbuvir. In this study, the resistance mechanism of Sofosbuvir at the atomic level is proposed. The proposed drug-resistance mechanism will provide valuable guidance for the design of HCV drugs.
Asunto(s)
Antivirales , Farmacorresistencia Viral , Hepacivirus , Simulación de Dinámica Molecular , Mutación , Sofosbuvir , Proteínas no Estructurales Virales , Antivirales/farmacología , Antivirales/química , ARN Helicasas DEAD-box , Farmacorresistencia Viral/genética , Hepacivirus/efectos de los fármacos , Hepacivirus/genética , Hepacivirus/enzimología , Nucleósido-Trifosfatasa , Serina Endopeptidasas , Serina Proteasas , Sofosbuvir/farmacología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Proteasas ViralesRESUMEN
AlphaFold protein structure database (AlphaFold DB) archives a vast number of predicted models. We conducted systematic data mining against AlphaFold DB and discovered an uncharacterized P-loop NTPase family. The structure of the protein family was surprisingly novel, showing an atypical topology for P-loop NTPases, noticeable twofold symmetry, and two pairs of independent putative active sites. Our findings show that structural data mining is a powerful approach to identifying undiscovered protein families.
Asunto(s)
Nucleósido-Trifosfatasa , Proteínas , Nucleósido-Trifosfatasa/química , Nucleósido-Trifosfatasa/metabolismo , Proteínas/química , Dominio Catalítico , Proteínas AAA/metabolismoRESUMEN
Cytosolic long dsRNA, among the most potent proinflammatory signals, is recognized by melanoma differentiation-associated protein 5 (MDA5). MDA5 binds dsRNA cooperatively forming helical filaments. ATP hydrolysis by MDA5 fulfills a proofreading function by promoting dissociation of shorter endogenous dsRNs from MDA5 while allowing longer viral dsRNAs to remain bound leading to activation of interferon-ß responses. Here, we show that adjacent MDA5 subunits in MDA5-dsRNA filaments hydrolyze ATP cooperatively, inducing cooperative filament disassembly. Consecutive rounds of ATP hydrolysis amplify the filament footprint, displacing tightly bound proteins from dsRNA. Our electron microscopy and biochemical assays show that LGP2 binds to dsRNA at internal binding sites through noncooperative ATP hydrolysis. Unlike MDA5, LGP2 has low nucleic acid selectivity and can hydrolyze GTP and CTP as well as ATP. Binding of LGP2 to dsRNA promotes nucleation of MDA5 filament assembly resulting in shorter filaments. Molecular modeling identifies an internally bound MDA5-LGP2-RNA complex, with the LGP2 C-terminal tail forming the key contacts with MDA5. These contacts are specifically required for NTP-dependent internal RNA binding. We conclude that NTPase-dependent binding of LGP2 to internal dsRNA sites complements NTPase-independent binding to dsRNA ends, via distinct binding modes, to increase the number and signaling output of MDA5-dsRNA complexes.