Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Microbiol Spectr ; 10(1): e0061821, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35019698

RESUMO

The host transmembrane protein MARCH8 is a RING finger E3 ubiquitin ligase that downregulates various host transmembrane proteins, such as MHC-II. We have recently reported that MARCH8 expression in virus-producing cells impairs viral infectivity by reducing virion incorporation of not only HIV-1 envelope glycoprotein but also vesicular stomatitis virus G-glycoprotein through two different pathways. However, the MARCH8 inhibition spectrum remains largely unknown. Here, we show the antiviral spectrum of MARCH8 using viruses pseudotyped with a variety of viral envelope glycoproteins. Infection experiments revealed that viral envelope glycoproteins derived from the rhabdovirus, arenavirus, coronavirus, and togavirus (alphavirus) families were sensitive to MARCH8-mediated inhibition. Lysine mutations at the cytoplasmic tails of rabies virus-G, lymphocytic choriomeningitis virus glycoproteins, SARS-CoV and SARS-CoV-2 spike proteins, and Chikungunya virus and Ross River virus E2 proteins conferred resistance to MARCH8. Immunofluorescence showed impaired downregulation of the mutants of these viral envelope glycoproteins by MARCH8, followed by lysosomal degradation, suggesting that MARCH8-mediated ubiquitination leads to intracellular degradation of these envelopes. Indeed, rabies virus-G and Chikungunya virus E2 proteins proved to be clearly ubiquitinated. We conclude that MARCH8 has inhibitory activity on a variety of viral envelope glycoproteins whose cytoplasmic lysine residues are targeted by this antiviral factor. IMPORTANCE A member of the MARCH E3 ubiquitin ligase family, MARCH8, downregulates many different kinds of host transmembrane proteins, resulting in the regulation of cellular homeostasis. On the other hands, MARCH8 acts as an antiviral factor when it binds to and downregulates HIV-1 envelope glycoprotein and vesicular stomatitis virus G-glycoprotein that are viral transmembrane proteins. This study reveals that, as in the case of cellular membrane proteins, MARCH8 shows broad-spectrum inhibition against various viral envelope glycoproteins by recognizing their cytoplasmic lysine residues, resulting in lysosomal degradation.


Assuntos
Antivirais/farmacologia , Lisina/efeitos dos fármacos , Ubiquitina-Proteína Ligases/farmacologia , Proteínas do Envelope Viral/química , Western Blotting , Regulação para Baixo , Células HEK293 , Células HeLa , Humanos , Imunoprecipitação , Lisina/metabolismo , Ubiquitinação/fisiologia , Proteínas do Envelope Viral/efeitos dos fármacos
2.
Molecules ; 26(22)2021 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-34833913

RESUMO

Dengue fever is a dangerous infectious endemic disease that affects over 100 nations worldwide, from Africa to the Western Pacific, and is caused by the dengue virus, which is transmitted to humans by an insect bite of Aedes aegypti. Millions of citizens have died as a result of dengue fever and dengue hemorrhagic fever across the globe. Envelope (E), serine protease (NS3), RNA-directed RNA polymerase (NS5), and non-structural protein 1 (NS1) are mostly required for cell proliferation and survival. Some of the diterpenoids and their derivatives produced by nature possess anti-dengue viral properties. The goal of the computational study was to scrutinize the effectiveness of diterpenoids and their derivatives against dengue viral proteins through in silico study. Methods: molecular docking was performed to analyze the binding affinity of compounds against four viral proteins: the envelope (E) protein, the NS1 protein, the NS3 protein, and the NS5 protein. Results: among the selected drug candidates, triptolide, stevioside, alepterolic acid, sphaeropsidin A, methyl dodovisate A, andrographolide, caesalacetal, and pyrimethamine have demonstrated moderate to good binding affinities (-8.0 to -9.4 kcal/mol) toward the selected proteins: E protein, NS3, NS5, and NS1 whereas pyrimethamine exerts -7.5, -6.3, -7.8, and -6.6 kcal/mol with viral proteins, respectively. Interestingly, the binding affinities of these lead compounds were better than those of an FDA-approved anti-viral medication (pyrimethamine), which is underused in dengue fever. Conclusion: we can conclude that diterpenoids can be considered as a possible anti-dengue medication option. However, in vivo investigation is recommended to back up the conclusions of this study.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Diterpenos/farmacologia , Antivirais/química , Antivirais/farmacocinética , Sítios de Ligação , Simulação por Computador , Dengue/tratamento farmacológico , Dengue/virologia , Diterpenos/química , Diterpenos/farmacocinética , Desenho de Fármacos , Humanos , Simulação de Acoplamento Molecular , Compostos Fitoquímicos/química , Compostos Fitoquímicos/farmacocinética , Compostos Fitoquímicos/farmacologia , Ligação Proteica , RNA Helicases/química , RNA Helicases/efeitos dos fármacos , RNA Helicases/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/efeitos dos fármacos , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/efeitos dos fármacos , Proteínas não Estruturais Virais/metabolismo
3.
Viruses ; 13(10)2021 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-34696459

RESUMO

Zika virus (ZIKV) is a mosquito-borne flavivirus, and its infection may cause severe neurodegenerative diseases. The outbreak of ZIKV in 2015 in South America has caused severe human congenital and neurologic disorders. Thus, it is vitally important to determine the inner mechanism of ZIKV infection. Here, our data suggested that the ubiquitin-specific peptidase 38 (USP38) played an important role in host resistance to ZIKV infection, during which ZIKV infection did not affect USP38 expression. Mechanistically, USP38 bound to the ZIKV envelope (E) protein through its C-terminal domain and attenuated its K48-linked and K63-linked polyubiquitination, thereby repressed the infection of ZIKV. In addition, we found that the deubiquitinase activity of USP38 was essential to inhibit ZIKV infection, and the mutant that lacked the deubiquitinase activity of USP38 lost the ability to inhibit infection. In conclusion, we found a novel host protein USP38 against ZIKV infection, and this may represent a potential therapeutic target for the treatment and prevention of ZIKV infection.


Assuntos
Proteases Específicas de Ubiquitina/farmacologia , Ubiquitinação , Infecção por Zika virus/tratamento farmacológico , Zika virus/efeitos dos fármacos , Células A549 , Células HeLa , Humanos , Receptor EphB2 , Proteases Específicas de Ubiquitina/metabolismo , Proteínas do Envelope Viral/efeitos dos fármacos , Infecção por Zika virus/virologia
4.
mBio ; 12(1)2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436439

RESUMO

Despite the effectiveness of antiretroviral (ARV) therapy, virological failure can occur in some HIV-1-infected patients in the absence of mutations in drug target genes. We previously reported that, in vitro, the lab-adapted HIV-1 NL4-3 strain can acquire resistance to the integrase inhibitor dolutegravir (DTG) by acquiring mutations in the envelope glycoprotein (Env) that enhance viral cell-cell transmission. In this study, we investigated whether Env-mediated drug resistance extends to ARVs other than DTG and whether it occurs in other HIV-1 isolates. We demonstrate that Env mutations can reduce susceptibility to multiple classes of ARVs and also increase resistance to ARVs when coupled with target-gene mutations. We observe that the NL4-3 Env mutants display a more stable and closed Env conformation and lower rates of gp120 shedding than the WT virus. We also selected for Env mutations in clinically relevant HIV-1 isolates in the presence of ARVs. These Env mutants exhibit reduced susceptibility to DTG, with effects on replication and Env structure that are HIV-1 strain dependent. Finally, to examine a possible in vivo relevance of Env-mediated drug resistance, we performed single-genome sequencing of plasma-derived virus from five patients failing an integrase inhibitor-containing regimen. This analysis revealed the presence of several mutations in the highly conserved gp120-gp41 interface despite low frequency of resistance mutations in integrase. These results suggest that mutations in Env that enhance the ability of HIV-1 to spread via a cell-cell route may increase the opportunity for the virus to acquire high-level drug resistance mutations in ARV target genes.IMPORTANCE Although combination antiretroviral (ARV) therapy is highly effective in controlling the progression of HIV disease, drug resistance can be a major obstacle. Recent findings suggest that resistance can develop without ARV target gene mutations. We previously reported that mutations in the HIV-1 envelope glycoprotein (Env) confer resistance to an integrase inhibitor. Here, we investigated the mechanism of Env-mediated drug resistance and the possible contribution of Env to virological failure in vivo We demonstrate that Env mutations can reduce sensitivity to major classes of ARVs in multiple viral isolates and define the effect of the Env mutations on Env subunit interactions. We observed that many Env mutations accumulated in individuals failing integrase inhibitor therapy despite a low frequency of resistance mutations in integrase. Our findings suggest that broad-based Env-mediated drug resistance may impact therapeutic strategies and provide clues toward understanding how ARV-treated individuals fail therapy without acquiring mutations in drug target genes.


Assuntos
Antirretrovirais/farmacologia , Farmacorresistência Viral/genética , HIV-1/efeitos dos fármacos , HIV-1/genética , Proteínas do Envelope Viral/genética , Linhagem Celular , Células HEK293 , Proteína gp120 do Envelope de HIV/química , Proteína gp120 do Envelope de HIV/genética , Proteína gp41 do Envelope de HIV/química , Proteína gp41 do Envelope de HIV/genética , Infecções por HIV/transmissão , Infecções por HIV/virologia , Inibidores de Integrase de HIV/farmacologia , Células HeLa , Compostos Heterocíclicos com 3 Anéis , Humanos , Mutação/efeitos dos fármacos , Oxazinas , Piperazinas , Piridonas , Linfócitos T , Proteínas do Envelope Viral/efeitos dos fármacos , Produtos do Gene env do Vírus da Imunodeficiência Humana/química , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética
5.
Indian J Pharmacol ; 53(6): 471-479, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975135

RESUMO

OBJECTIVE: The study was performed to evaluate in silico binding ability of lutein and rosmarinic acid (RA) with the envelope domain III (EDIII) proteins of the four serotypes of dengue virus (DENV), enlightening potential antiviral activity of the two compounds. MATERIALS AND METHODS: EDIII protein structures for the four DENV serotypes were retrieved from RCSB Protein data bank (PDB) and used as receptors. Four ligands of lutein and four of RA were selected from the ZINC database and used for computational molecular docking and ligand interaction analysis with the four receptors using bioinformatics tools like AutoDock Vina and Molecular Operating Environment (MOE) software. RESULTS: The EDIII of the four serotypes demonstrated significant interaction with ligands of lutein and RA. RA ligand ZINC899870, particularly presented best-binding energy values of 6.4, -7.0, and 6.9 kcal/mol with EDIII of serotype DENV-1, DENV-2, and DENV-4 respectively. Whereas, lutein ligand, ZINC14879959 presented best-binding energy value of 7.9 kcal/mol for EDIII of serotype DENV-3. From the results predicted by MOE, the hydroxyl (OH) of 3, 4-dihydroxyphenyl group of RA ligand ZINC899870 is actively involved in interaction with all four serotypes. CONCLUSION: RA is a competent candidate for further evaluation of potential in vitro antiviral activity that can be effective in conferring protection against the four serotypes of DENV.


Assuntos
Antivirais/farmacologia , Cinamatos/farmacologia , Vírus da Dengue/efeitos dos fármacos , Dengue/tratamento farmacológico , Depsídeos/farmacologia , Luteína/farmacologia , Dengue/virologia , Humanos , Simulação de Acoplamento Molecular , Fitoterapia , Proteínas do Envelope Viral/efeitos dos fármacos , Ácido Rosmarínico
6.
J Am Chem Soc ; 142(40): 17024-17038, 2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-32926779

RESUMO

Broad-spectrum antivirals are powerful weapons against dangerous viruses where no specific therapy exists, as in the case of the ongoing SARS-CoV-2 pandemic. We discovered that a lysine- and arginine-specific supramolecular ligand (CLR01) destroys enveloped viruses, including HIV, Ebola, and Zika virus, and remodels amyloid fibrils in semen that promote viral infection. Yet, it is unknown how CLR01 exerts these two distinct therapeutic activities. Here, we delineate a novel mechanism of antiviral activity by studying the activity of tweezer variants: the "phosphate tweezer" CLR01, a "carboxylate tweezer" CLR05, and a "phosphate clip" PC. Lysine complexation inside the tweezer cavity is needed to antagonize amyloidogenesis and is only achieved by CLR01. Importantly, CLR01 and CLR05 but not PC form closed inclusion complexes with lipid head groups of viral membranes, thereby altering lipid orientation and increasing surface tension. This process disrupts viral envelopes and diminishes infectivity but leaves cellular membranes intact. Consequently, CLR01 and CLR05 display broad antiviral activity against all enveloped viruses tested, including herpesviruses, Measles virus, influenza, and SARS-CoV-2. Based on our mechanistic insights, we potentiated the antiviral, membrane-disrupting activity of CLR01 by introducing aliphatic ester arms into each phosphate group to act as lipid anchors that promote membrane targeting. The most potent ester modifications harbored unbranched C4 units, which engendered tweezers that were approximately one order of magnitude more effective than CLR01 and nontoxic. Thus, we establish the mechanistic basis of viral envelope disruption by specific tweezers and establish a new class of potential broad-spectrum antivirals with enhanced activity.


Assuntos
Antivirais/química , Antivirais/farmacologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Organofosfatos/farmacologia , Proteínas do Envelope Viral/efeitos dos fármacos , Fosfatase Ácida/química , Fosfatase Ácida/metabolismo , Amiloide/antagonistas & inibidores , Fármacos Anti-HIV/química , Fármacos Anti-HIV/farmacologia , Arginina/química , Betacoronavirus/efeitos dos fármacos , Hidrocarbonetos Aromáticos com Pontes/química , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/virologia , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Humanos , Lipídeos/química , Lisina/química , Espectroscopia de Ressonância Magnética , Organofosfatos/química , SARS-CoV-2 , Proteínas Secretadas pela Vesícula Seminal/química , Proteínas Secretadas pela Vesícula Seminal/metabolismo , Relação Estrutura-Atividade , Proteínas do Envelope Viral/metabolismo , Zika virus/efeitos dos fármacos
7.
Int J Mol Sci ; 21(16)2020 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-32824072

RESUMO

The rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has created a severe global health crisis. In this paper, we used docking and simulation methods to identify potential targets and the mechanism of action of chloroquine (CQ) and hydroxychloroquine (HCQ) against SARS-CoV-2. Our results showed that both CQ and HCQ influenced the functionality of the envelope (E) protein, necessary in the maturation processes of the virus, due to interactions that modify the flexibility of the protein structure. Furthermore, CQ and HCQ also influenced the proofreading and capping of viral RNA in SARS-CoV-2, performed by nsp10/nsp14 and nsp10/nsp16. In particular, HCQ demonstrated a better energy binding with the examined targets compared to CQ, probably due to the hydrogen bonding of the hydroxyl group of HCQ with polar amino acid residues.


Assuntos
Antivirais/farmacologia , Betacoronavirus/efeitos dos fármacos , Cloroquina/farmacologia , Exorribonucleases/metabolismo , Hidroxicloroquina/farmacologia , Metiltransferases/metabolismo , Proteínas não Estruturais Virais/metabolismo , COVID-19 , Proteínas do Envelope de Coronavírus , Infecções por Coronavirus/tratamento farmacológico , Humanos , Simulação de Acoplamento Molecular , Pandemias , Pneumonia Viral/tratamento farmacológico , RNA Viral/efeitos dos fármacos , RNA Viral/genética , SARS-CoV-2 , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Replicação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
8.
J Virol ; 94(19)2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32699087

RESUMO

Chikungunya fever, a mosquito-borne disease manifested by fever, rash, myalgia, and arthralgia, is caused by chikungunya virus (CHIKV), which belongs to the genus Alphavirus of the family Togaviridae Anti-CHIKV IgG from convalescent patients is known to directly neutralize CHIKV, and the state of immunity lasts throughout life. Here, we examined the epitope of a neutralizing mouse monoclonal antibody against CHIKV, CHE19, which inhibits viral fusion and release. In silico docking analysis showed that the epitope of CHE19 was localized in the viral E2 envelope and consisted of two separate segments, an N-linker and a ß-ribbon connector, and that its bound Fab fragment on E2 overlapped the position that the E3 glycoprotein originally occupied. We showed that CHIKV-E2 is lost during the viral internalization and that CHE19 inhibits the elimination of CHIKV-E2. These findings suggested that CHE19 stabilizes the E2-E1 heterodimer instead of E3 and inhibits the protrusion of the E1 fusion loop and subsequent membrane fusion. In addition, the antigen-bound Fab fragment configuration showed that CHE19 connects to the CHIKV spikes existing on the two individual virions, leading us to conclude that the CHE19-CHIKV complex was responsible for the large virus aggregations. In our subsequent filtration experiments, large viral aggregations by CHE19 were trapped by a 0.45-µm filter. This virion-connecting characteristic of CHE19 could explain the inhibition of viral release from infected cells by the tethering effect of the virion itself. These findings provide clues toward the development of effective prophylactic and therapeutic monoclonal antibodies against the Alphavirus infection.IMPORTANCE Recent outbreaks of chikungunya fever have increased its clinical importance. Neither a specific antiviral drug nor a commercial vaccine for CHIKV infection are available. Here, we show a detailed model of the docking between the envelope glycoprotein of CHIKV and our unique anti-CHIKV-neutralizing monoclonal antibody (CHE19), which inhibits CHIKV membrane fusion and virion release from CHIKV-infected cells. Homology modeling of the neutralizing antibody CHE19 and protein-protein docking analysis of the CHIKV envelope glycoprotein and CHE19 suggested that CHE19 inhibits the viral membrane fusion by stabilizing the E2-E1 heterodimer and inhibits virion release by facilitating the formation of virus aggregation due to the connecting virions, and these predictions were confirmed by experiments. Sequence information of CHE19 and the CHIKV envelope glycoprotein and their docking model will contribute to future development of an effective prophylactic and therapeutic agent.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Febre de Chikungunya/imunologia , Vírus Chikungunya/imunologia , Proteínas do Envelope Viral/imunologia , Internalização do Vírus/efeitos dos fármacos , Liberação de Vírus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/química , Anticorpos Antivirais/farmacologia , Vírus Chikungunya/genética , Vírus Chikungunya/fisiologia , Chlorocebus aethiops , Epitopos/imunologia , Feminino , Fusão de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Simulação de Acoplamento Molecular , Domínios e Motivos de Interação entre Proteínas , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/imunologia , Liberação de Vírus/efeitos dos fármacos
9.
Eur J Pharm Sci ; 153: 105465, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32668312

RESUMO

COVID-19, is a disease resulting from the SARS-CoV-2 global pandemic. Due to the current global emergency and the length of time required to develop specific antiviral agent(s) and a vaccine for SARS-CoV-2, the world health organization (WHO) adopted the strategy of repurposing existing medications to treat COVID-19. Iron oxide nanoparticles (IONPs) were previously approved by the US food and drug administration (FDA) for anemia treatment and studies have also demonstrated its antiviral activity in vitro. Therefore, we performed a docking study to explore the interaction of IONPs (Fe2O3 and Fe3O4) with the spike protein receptor binding domain (S1-RBD) of SARS-CoV-2 that is required for virus attachment to the host cell receptors. A similar docking analysis was also performed with hepatitis C virus (HCV) glycoproteins E1 and E2. These studies revealed that both Fe2O3 and Fe3O4 interacted efficiently with the SARS-CoV-2 S1-RBD and to HCV glycoproteins, E1 and E2. Fe3O4 formed a more stable complex with S1-RBD whereas Fe2O3 favored HCV E1 and E2. These interactions of IONPs are expected to be associated with viral proteins conformational changes and hence, viral inactivation. Therefore, we recommend FDA-approved-IONPs to proceed for COVID-19 treatment clinical trials.


Assuntos
Infecções por Coronavirus/tratamento farmacológico , Compostos Férricos/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Simulação de Acoplamento Molecular , Pneumonia Viral/tratamento farmacológico , COVID-19 , Aprovação de Drogas , Reposicionamento de Medicamentos , Humanos , Pandemias , Conformação Proteica , Glicoproteína da Espícula de Coronavírus/efeitos dos fármacos , Estados Unidos , United States Food and Drug Administration , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Tratamento Farmacológico da COVID-19
10.
Emerg Microbes Infect ; 9(1): 390-412, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32397950

RESUMO

Emerging studies have indicated that some penaeidins restrict virus infection; however, the mechanism(s) involved are poorly understood. In the present study, we uncovered that penaeidins are a novel family of antiviral effectors against white spot syndrome virus (WSSV), which antagonize the envelope proteins to block viral entry. We found that the expression levels of four identified penaeidins from Litopenaeus vannamei, including BigPEN, PEN2, PEN3, and PEN4, were significantly induced in hemocytes during the early stage of WSSV infection. Knockdown of each penaeidin in vivo via RNA interference resulted in elevated viral loads and rendered shrimp more susceptible to WSSV, while the survival rate was rescued via the injection of recombinant penaeidins. All penaeidins, except PEN4, were shown to interact with several envelope proteins of WSSV, and all four penaeidins were observed to be located on the outer surface of the WSSV virion. Co-incubation of each recombinant penaeidin with WSSV inhibited virion internalization into hemocytes. More importantly, we found that PEN2 competitively bound to the envelope protein VP24 to release it from polymeric immunoglobulin receptor (pIgR), the cellular receptor required for WSSV infection. Moreover, we also demonstrated that BigPEN was able to bind to VP28 of WSSV, which disrupted the interaction between VP28 and Rab7 - the Rab GTPase that contributes to viral entry by binding with VP28. Taken together, our results demonstrated that penaeidins interact with the envelope proteins of WSSV to block multiple viral infection processes, thereby protecting the host against WSSV.


Assuntos
Antivirais/farmacologia , Penaeidae/virologia , Peptídeos , Vírus da Síndrome da Mancha Branca 1/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/farmacologia , Expressão Gênica , Hemócitos/virologia , Peptídeos/genética , Peptídeos/farmacologia , Proteínas do Envelope Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
11.
Viruses ; 11(11)2019 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31683742

RESUMO

Flaviviruses, such as Zika virus (ZIKV), Japanese encephalitis virus (JEV), Dengue virus (DENV), and West Nile virus (WNV), are important arthropod-borne pathogens that present an immense global health problem. Their unpredictable disease severity, unusual clinical features, and severe neurological manifestations underscore an urgent need for antiviral interventions. Furin, a host proprotein convertase, is a key contender in processing flavivirus prM protein to M protein, turning the inert virus to an infectious particle. For this reason, the current study was planned to evaluate the antiviral activity of decanoyl-Arg-Val-Lys-Arg-chloromethylketone, a specific furin inhibitor, against flaviviruses, including ZIKV and JEV. Analysis of viral proteins revealed a significant increase in the prM/E index of ZIKV or JEV in dec-RVKR-cmk-treated Vero cells compared to DMSO-treated control cells, indicating dec-RVKR-cmk inhibits prM cleavage. Plaque assay, qRT-PCR, and immunofluorescence assay revealed a strong antiviral activity of dec-RVKR-cmk against ZIKV and JEV in terms of the reduction in virus progeny titer and in viral RNA and protein production in both mammalian cells and mosquito cells. Time-of-drug addition assay revealed that the maximum reduction of virus titer was observed in post-infection treatment. Furthermore, our results showed that dec-RVKR-cmk exerts its inhibitory action on the virus release and next round infectivity but not on viral RNA replication. Taken together, our study highlights an interesting antiviral activity of dec-RVKR-cmk against flaviviruses.


Assuntos
Clorometilcetonas de Aminoácidos/farmacologia , Flavivirus/efeitos dos fármacos , Furina/antagonistas & inibidores , Proteínas do Envelope Viral , Animais , Antivirais/farmacologia , Linhagem Celular , Chlorocebus aethiops , Vírus da Encefalite Japonesa (Espécie)/efeitos dos fármacos , Furina/metabolismo , Células Vero , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Zika virus/efeitos dos fármacos
12.
Antiviral Res ; 172: 104636, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31654671

RESUMO

Dengue virus (DENV) is a global health problem that affects approximately 3.9 billion people worldwide. Since safety concerns were raised for the only licensed vaccine, Dengvaxia, and since the present treatment is only supportive care, the development of more effective therapeutic anti-DENV agents is urgently needed. In this report, we identified a potential small-molecule inhibitor, BP34610, via cell-based high-throughput screening (HTS) of 12,000 compounds using DENV-2 reporter viruses. BP34610 reduced the virus yields of type 2 DENV-infected cells with a 50% effective concentration (EC50) and selectivity index value of 0.48 ±â€¯0.06 µM and 197, respectively. Without detectable cytotoxicity, the compound inhibited not only all four serotypes of DENV but also Japanese encephalitis virus (JEV). Time-of-addition experiments suggested that BP34610 may act at an early stage of DENV virus infection. Sequencing analyses of several individual clones derived from BP34610-resistant viruses revealed a consensus amino acid substitution (S397P) in the N-terminal stem region of the E protein. Introduction of S397P into the DENV reporter viruses conferred an over 14.8-fold EC90 shift for BP34610. Importantly, the combination of BP34610 with a viral replication inhibitor, ribavirin, displayed synergistic enhancement of anti-DENV-2 activity. Our results identify an effective small-molecule inhibitor, BP34610, which likely targets the DENV E protein. BP34610 could be developed as an anti-flavivirus agent in the future.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Flavivirus/efeitos dos fármacos , Proteínas do Envelope Viral/efeitos dos fármacos , Animais , Antivirais/toxicidade , Linhagem Celular , Dengue/tratamento farmacológico , Sinergismo Farmacológico , Vírus da Encefalite Japonesa (Espécie)/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Humanos , Ribavirina/farmacologia , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
13.
Biochem Biophys Res Commun ; 515(2): 366-371, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31155294

RESUMO

Flaviviruses are emerging arthropod-borne viruses posing a great threat to human beings worldwide. The E dimer configuration of the flavivirus was prominent during viral assembly, maturation and entry. Neutralization antibodies targeting E dimer played the important role in controlling the flavivirus infection. Previously, the ideal drug target of small molecular inhibitors of JEV was viral proteases and polymerases. The crystal structure of JEV E protein showed a conserved pocket in it is important at membrane fusion step. Recently, a set of anti-virus drugs has been found by virtual screening. Here, we show that the fusion-loop pocket of JEV E protein was a conservative region and an ideal drug target. ChemDiv-3 from virtual screening as the lead compound was found to show a relatively modest inhibition effect for JEV in vitro and in vivo test and could interfere with the formation of JEV sE dimer. ChemDiv-3 interacts with the amino acid residues ASN 313, PRO 314, ALA 315, and VAL 323 in E protein via hydrogen bonds for occupation of the fusion-loop pocket. The key binding sites LYS 312, ALA 513 and THR 317 forming the fusion-loop pocket are the same and other auxiliary sites are similar among the flavivirus. Taken together, the fusion-loop pocket of the flavivirus could be one promising target for drug discovery.


Assuntos
Antivirais/química , Antivirais/farmacologia , Vírus da Encefalite Japonesa (Espécie)/química , Vírus da Encefalite Japonesa (Espécie)/efeitos dos fármacos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Bases de Dados de Produtos Farmacêuticos , Modelos Animais de Doenças , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Vírus da Encefalite Japonesa (Espécie)/genética , Encefalite Japonesa/tratamento farmacológico , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Simulação de Acoplamento Molecular , Multimerização Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína/efeitos dos fármacos , Relação Estrutura-Atividade , Interface Usuário-Computador , Proteínas do Envelope Viral/genética
15.
JCI Insight ; 2(17)2017 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-28878130

RESUMO

Human endogenous retroviruses (HERVs), remnants of ancestral viral genomic insertions, are known to represent 8% of the human genome and are associated with several pathologies. In particular, the envelope protein of HERV-W family (HERV-W-Env) has been involved in multiple sclerosis pathogenesis. Investigations to detect HERV-W-Env in a few other autoimmune diseases were negative, except in type-1 diabetes (T1D). In patients suffering from T1D, HERV-W-Env protein was detected in 70% of sera, and its corresponding RNA was detected in 57% of peripheral blood mononuclear cells. While studies on human Langerhans islets evidenced the inhibition of insulin secretion by HERV-W-Env, this endogenous protein was found to be expressed by acinar cells in 75% of human T1D pancreata. An extensive immunohistological analysis further revealed a significant correlation between HERV-W-Env expression and macrophage infiltrates in the exocrine part of human pancreata. Such findings were corroborated by in vivo studies on transgenic mice expressing HERV-W-env gene, which displayed hyperglycemia and decreased levels of insulin, along with immune cell infiltrates in their pancreas. Altogether, these results strongly suggest an involvement of HERV-W-Env in T1D pathogenesis. They also provide potentially novel therapeutic perspectives, since unveiling a pathogenic target in T1D.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/virologia , Retrovirus Endógenos/efeitos dos fármacos , Proteínas do Envelope Viral/fisiologia , Animais , Antivirais/uso terapêutico , Estudos de Coortes , Diabetes Mellitus Tipo 1/complicações , Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidade , Feminino , Humanos , Hiperglicemia/complicações , Insulina/metabolismo , Antagonistas da Insulina/farmacologia , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , RNA Viral/sangue , Proteínas do Envelope Viral/efeitos dos fármacos
16.
Artigo em Inglês | MEDLINE | ID: mdl-28536674

RESUMO

Dengue virus (DENV) co-circulates as four serotypes (DENV1-4). Primary infection only leads to self-limited dengue fever. But secondary infection with another serotype carries a higher risk of increased disease severity, causing life-threatening dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). Serotype cross-reactive antibodies facilitate DENV infection in Fc-receptor-bearing cells by promoting virus entry via Fcγ receptors (FcγR), a process known as antibody dependent enhancement (ADE). Most studies suggested that enhancing antibodies were mainly specific to the structural premembrane protein (prM) of DENV. However, there is still no effective drugs or vaccines to prevent ADE. In this study, we firstly confirmed that both DENV-2 infected human sera (anti-DENV-2) and DENV-2 prM monoclonal antibody (prM mAb) could significantly enhance DENV-1 infection in K562 cells. Then we developed anti-idiotypic antibodies (prM-AIDs) specific to prM mAb by immunizing of Balb/c mice. Results showed that these polyclonal antibodies can dramatically reduce ADE phenomenon of DENV-1 infection in K562 cells. To further confirm the anti-ADE effect of prM-AIDs in vivo, interferon-α and γ receptor-deficient mice (AG6) were used as the mouse model for DENV infection. We found that administration of DENV-2 prM mAb indeed caused a higher DENV-1 titer as well as interleukin-10 (IL-10) and alaninea minotransferase (ALT) in mice infected with DENV-1, similar to clinical ADE symptoms. But when we supplemented prM-AIDs to DENV-1 challenged AG6 mice, the viral titer, IL-10 and ALT were obviously decreased to the negative control level. Of note, the number of platelets in peripheral blood of prM-AIDs group were significantly increased at day 3 post infection with DENV-1 compared that of prM-mAb group. These results confirmed that our prM-AIDs could prevent ADE not only in vitro but also in vivo, suggested that anti-idiotypic antibodies might be a new choice to be considered to treat DENV infection.


Assuntos
Anticorpos Anti-Idiotípicos/farmacologia , Anticorpos Facilitadores/efeitos dos fármacos , Vírus da Dengue/imunologia , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Anti-Idiotípicos/administração & dosagem , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Anticorpos Facilitadores/imunologia , Reações Cruzadas , Dengue/tratamento farmacológico , Dengue/virologia , Vírus da Dengue/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Interleucina-10/metabolismo , Células K562 , Camundongos , Camundongos Endogâmicos BALB C , Internalização do Vírus , Replicação Viral/efeitos dos fármacos
17.
Sci Rep ; 7(1): 1028, 2017 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-28432348

RESUMO

Several studies have demonstrated that injection of double-stranded RNAs (dsRNA) homologous to mRNA for the white spot syndrome virus (WSSV) viral protein 28 (VP28) can induce protection in shrimp against WSSV through RNA interference (RNAi). In comparison to shrimp injected with either PBS or a green fluorescent protein (GFP) nonspecific dsRNA, we obtained nearly complete protection against WSSV infection in shrimp injected with VP28 dsRNA. Upregulation of host genes associated with small RNA silencing was measured 48 hours post treatment in groups injected with dsRNA, and although the VP28-treated group remained moderately upregulated after challenge with WSSV, many-fold higher induction was observed in both control groups reflecting the ongoing viral infection. RNA sequencing of VP28-treated shrimp demonstrated a siRNA population dominated by high levels of 22 nt long molecules narrowly targeting the VP28 mRNA both before and after challenge with WSSV. Conversely, while no siRNAs targeting WSSV were detected before challenge, a broad response of 22 nt siRNAs mapping across the entire WSSV genome were found in both control groups after challenge. These results give detailed insight to how dsRNA targeting VP28 function to induce protection against WSSV, by generating a highly focused population of 22 nt long siRNA molecules.


Assuntos
Penaeidae/crescimento & desenvolvimento , RNA Interferente Pequeno/farmacologia , Proteínas do Envelope Viral/genética , Vírus da Síndrome da Mancha Branca 1/genética , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Penaeidae/genética , Penaeidae/virologia , Vírus de RNA/genética , Análise de Sequência de RNA , Proteínas do Envelope Viral/efeitos dos fármacos , Vírus da Síndrome da Mancha Branca 1/efeitos dos fármacos
18.
Microb Pathog ; 106: 65-68, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27816678

RESUMO

Tetherin (BST-2) is an important host restriction factor that can inhibit the release of a diverse array of enveloped viruses from infected cells. Conversely, to facilitate their release and spread, many viruses have evolved various strategies to overcome the antiviral effect of tetherin in a species-specific manner. During the development of an attenuated equine infectious anemia virus (EIAV) vaccine in our laboratory, we found that serial passage of a field-isolated virulent EIAV strains in horse and donkey as well as the cultivated donkey cells, produces several typical EIAV strains, including EIAVDV, EIAVDLV, and EIAVFDDV, which exhibit distinct virulence and replication features in vivo and in vitro. However, the role of host restriction factors in EIAV evolution during the serial passage is not well understood. This study aimed to evaluate whether these newly generated strains adapt differently to donkey tetherin (do-tetherin) based on their virulence. We found that do-tetherin exerts an inhibition on the release of the viral particles produced by all three strains, albeit with varying intensity: EIAVDV < EIAVDLV < EIAVFDDV. Additionally, all three EIAV strains could counteract the restriction mediated by do-tetherin via their envelope proteins (Env) with varying strength: EIAVDV > EIAVDLV > EIAVFDDV. These results indicate that donkey tetherin is involved in shaping of EIAV evolution during serial passage.


Assuntos
Antígenos CD/imunologia , Antígenos CD/farmacologia , Proteínas Ligadas por GPI/imunologia , Proteínas Ligadas por GPI/farmacologia , Vírus da Anemia Infecciosa Equina/efeitos dos fármacos , Vírus da Anemia Infecciosa Equina/imunologia , Animais , Evolução Biológica , Células Cultivadas , DNA Viral , Equidae , Células HEK293 , Cavalos , Humanos , Imunidade Inata , Vírus da Anemia Infecciosa Equina/crescimento & desenvolvimento , Mutação , Projetos Piloto , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/genética , Vacinas Virais/imunologia , Vírion/efeitos dos fármacos , Virulência , Replicação Viral/efeitos dos fármacos
19.
Methods Mol Biol ; 1435: 249-91, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27188563

RESUMO

The persistence of West Nile virus (WNV) infections throughout the USA since its inception in 1999 and its continuous spread throughout the globe calls for an urgent need of effective treatments and prevention measures. Although the licensing of several WNV vaccines for veterinary use provides a proof of concept, similar efforts on the development of an effective vaccine for humans remain still unsuccessful. Increased understanding of biology and pathogenesis of WNV together with recent technological advancements have raised hope that an effective WNV vaccine may be available in the near future. In addition, rapid progress in the structural and functional characterization of WNV and other flaviviral proteins have provided a solid base for the design and development of several classes of inhibitors as potential WNV therapeutics. Moreover, the therapeutic monoclonal antibodies demonstrate an excellent efficacy against WNV in animal models and represent a promising class of WNV therapeutics. However, there are some challenges as to the design and development of a safe and efficient WNV vaccine or therapeutic. In this chapter, we discuss the current approaches, progress, and challenges toward the development of WNV vaccines, therapeutic antibodies, and antiviral drugs.


Assuntos
Antivirais/uso terapêutico , Febre do Nilo Ocidental/tratamento farmacológico , Febre do Nilo Ocidental/prevenção & controle , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/farmacologia , Anticorpos Antivirais/uso terapêutico , Antivirais/farmacologia , Ensaios Clínicos como Assunto , Descoberta de Drogas , Humanos , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/imunologia , Febre do Nilo Ocidental/imunologia , Vacinas contra o Vírus do Nilo Ocidental/farmacologia , Vacinas contra o Vírus do Nilo Ocidental/uso terapêutico , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus do Nilo Ocidental/imunologia
20.
Dev Comp Immunol ; 53(1): 47-54, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26123888

RESUMO

Antimicrobial peptides (AMPs) have a great potential to be used as a substitute for antibiotics since AMPs don't lead to bacteria's drug resistance. Anti-lipopolysaccharide factors (ALFs) are one type of AMPs and exist in crustaceans. In the present study, we produced a recombinant protein (rFcALF5) of an ALF isoform (FcALF5) from Chinese shrimp Fenneropenaeus chinensis through a prokaryotic expression system. The rFcALF5 exhibited varied antibacterial activities against different bacteria. Besides its antibacterial activities, it could also inhibit the infection of white spot syndrome virus (WSSV) to shrimp after pre-incubation with this virus. In order to learn the antiviral mechanism on how rFcALF5 influences WSSV infection, the interaction between the total proteins of WSSV and rFcALF5 was analyzed and the data showed that rFcALF5 had direct interaction with the envelope protein VP24 of WSSV. The LPS binding domain (LBD) of FcALF5 also showed direct interaction with VP24 of WSSV. Therefore we inferred that the antiviral activity of FcALF5 might be achieved through the binding of its LBD to VP24 of WSSV. These findings provided more information to develop new strategies for the control of shrimp disease in aquaculture.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Antivirais/farmacologia , Proteínas de Artrópodes/farmacologia , Vírus da Síndrome da Mancha Branca 1/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/genética , Proteínas de Artrópodes/biossíntese , Proteínas de Artrópodes/genética , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Lipopolissacarídeos/imunologia , Testes de Sensibilidade Microbiana , Penaeidae , Isoformas de Proteínas/genética , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA