Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 195
Filtrar
1.
J Virol ; 98(3): e0187423, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38329336

RESUMO

Subacute sclerosing panencephalitis (SSPE) is a rare but fatal late neurological complication of measles, caused by persistent measles virus (MeV) infection of the central nervous system. There are no drugs approved for the treatment of SSPE. Here, we followed the clinical progression of a 5-year-old SSPE patient after treatment with the nucleoside analog remdesivir, conducted a post-mortem evaluation of the patient's brain, and characterized the MeV detected in the brain. The quality of life of the patient transiently improved after the first two courses of remdesivir, but a third course had no further clinical effect, and the patient eventually succumbed to his condition. Post-mortem evaluation of the brain displayed histopathological changes including loss of neurons and demyelination paired with abundant presence of MeV RNA-positive cells throughout the brain. Next-generation sequencing of RNA isolated from the brain revealed a complete MeV genome with mutations that are typically detected in SSPE, characterized by a hypermutated M gene. Additional mutations were detected in the polymerase (L) gene, which were not associated with resistance to remdesivir. Functional characterization showed that mutations in the F gene led to a hyperfusogenic phenotype predominantly mediated by N465I. Additionally, recombinant wild-type-based MeV with the SSPE-F gene or the F gene with the N465I mutation was no longer lymphotropic but instead efficiently disseminated in neural cultures. Altogether, this case encourages further investigation of remdesivir as a potential treatment of SSPE and highlights the necessity to functionally understand SSPE-causing MeV.IMPORTANCEMeasles virus (MeV) causes acute, systemic disease and remains an important cause of morbidity and mortality in humans. Despite the lack of known entry receptors in the brain, MeV can persistently infect the brain causing the rare but fatal neurological disorder subacute sclerosing panencephalitis (SSPE). SSPE-causing MeVs are characterized by a hypermutated genome and a hyperfusogenic F protein that facilitates the rapid spread of MeV throughout the brain. No treatment against SSPE is available, but the nucleoside analog remdesivir was recently demonstrated to be effective against MeV in vitro. We show that treatment of an SSPE patient with remdesivir led to transient clinical improvement and did not induce viral escape mutants, encouraging the future use of remdesivir in SSPE patients. Functional characterization of the viral proteins sheds light on the shared properties of SSPE-causing MeVs and further contributes to understanding how those viruses cause disease.


Assuntos
Monofosfato de Adenosina , Alanina , Vírus do Sarampo , Sarampo , Panencefalite Esclerosante Subaguda , Proteínas Virais , Pré-Escolar , Humanos , Monofosfato de Adenosina/administração & dosagem , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/uso terapêutico , Alanina/administração & dosagem , Alanina/análogos & derivados , Alanina/uso terapêutico , Autopsia , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Progressão da Doença , Evolução Fatal , Genoma Viral/genética , Sequenciamento de Nucleotídeos em Larga Escala , Sarampo/complicações , Sarampo/tratamento farmacológico , Sarampo/virologia , Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/genética , Vírus do Sarampo/metabolismo , Proteínas Mutantes/análise , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Qualidade de Vida , RNA Viral/análise , RNA Viral/genética , Panencefalite Esclerosante Subaguda/tratamento farmacológico , Panencefalite Esclerosante Subaguda/etiologia , Panencefalite Esclerosante Subaguda/virologia , Proteínas Virais/análise , Proteínas Virais/genética , Proteínas Virais/metabolismo
2.
Biomed Res Int ; 2021: 9998420, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34527748

RESUMO

The global burden of viral infection, especially the current pandemics of SARS-CoV-2, HIV/AIDS, and hepatitis, is a very risky one. Additionally, HCV expresses the necessity for antiviral therapeutic elements. Venoms are known to contain an array of bioactive peptides that are commonly used in the treatment of various medical issues. Several peptides isolated from scorpion venom have recently been proven to possess an antiviral activity against several viral families. The aim of this review is to provide an up-to-date overview of scorpion antiviral peptides and to discuss their modes of action and potential biomedical application against different viruses.


Assuntos
Antivirais/química , Antivirais/farmacologia , Peptídeos/farmacologia , Venenos de Escorpião/química , Viroses/tratamento farmacológico , Animais , Coronavirus/efeitos dos fármacos , HIV-1/efeitos dos fármacos , Vírus de Hepatite/efeitos dos fármacos , Herpesvirus Humano 1/efeitos dos fármacos , Humanos , Vírus do Sarampo/efeitos dos fármacos , Peptídeos/química , Peptídeos/isolamento & purificação , Viroses/virologia
3.
Nat Commun ; 12(1): 5233, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34475387

RESUMO

Measles virus (MeV) is a highly contagious pathogen that enters the human host via the respiratory route. Besides acute pathologies including fever, cough and the characteristic measles rash, the infection of lymphocytes leads to substantial immunosuppression that can exacerbate the outcome of infections with additional pathogens. Despite the availability of effective vaccine prophylaxis, measles outbreaks continue to occur worldwide. We demonstrate that prophylactic and post-exposure therapeutic treatment with an orally bioavailable small-molecule polymerase inhibitor, ERDRP-0519, prevents measles disease in squirrel monkeys (Saimiri sciureus). Treatment initiation at the onset of clinical signs reduced virus shedding, which may support outbreak control. Results show that this clinical candidate has the potential to alleviate clinical measles and augment measles virus eradication.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Sarampo/prevenção & controle , Morfolinas/uso terapêutico , Piperidinas/uso terapêutico , Pirazóis/uso terapêutico , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Animais , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacocinética , Tolerância Imunológica/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Vírus do Sarampo/efeitos dos fármacos , Morfolinas/farmacocinética , Piperidinas/farmacocinética , Pirazóis/farmacocinética , Saimiri , Replicação Viral/efeitos dos fármacos , Eliminação de Partículas Virais/efeitos dos fármacos
4.
PLoS Pathog ; 17(2): e1009371, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33621266

RESUMO

Morbilliviruses, such as measles virus (MeV) and canine distemper virus (CDV), are highly infectious members of the paramyxovirus family. MeV is responsible for major morbidity and mortality in non-vaccinated populations. ERDRP-0519, a pan-morbillivirus small molecule inhibitor for the treatment of measles, targets the morbillivirus RNA-dependent RNA-polymerase (RdRP) complex and displayed unparalleled oral efficacy against lethal infection of ferrets with CDV, an established surrogate model for human measles. Resistance profiling identified the L subunit of the RdRP, which harbors all enzymatic activity of the polymerase complex, as the molecular target of inhibition. Here, we examined binding characteristics, physical docking site, and the molecular mechanism of action of ERDRP-0519 through label-free biolayer interferometry, photoaffinity cross-linking, and in vitro RdRP assays using purified MeV RdRP complexes and synthetic templates. Results demonstrate that unlike all other mononegavirus small molecule inhibitors identified to date, ERDRP-0519 inhibits all phosphodiester bond formation in both de novo initiation of RNA synthesis at the promoter and RNA elongation by a committed polymerase complex. Photocrosslinking and resistance profiling-informed ligand docking revealed that this unprecedented mechanism of action of ERDRP-0519 is due to simultaneous engagement of the L protein polyribonucleotidyl transferase (PRNTase)-like domain and the flexible intrusion loop by the compound, pharmacologically locking the polymerase in pre-initiation conformation. This study informs selection of ERDRP-0519 as clinical candidate for measles therapy and identifies a previously unrecognized druggable site in mononegavirus L polymerase proteins that can silence all synthesis of viral RNA.


Assuntos
Antivirais/farmacologia , Inibidores Enzimáticos/farmacologia , Vírus do Sarampo/efeitos dos fármacos , Sarampo/tratamento farmacológico , Morfolinas/farmacologia , Piperidinas/farmacologia , Pirazóis/farmacologia , RNA Viral/biossíntese , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Chlorocebus aethiops , Sarampo/metabolismo , Sarampo/virologia , Vírus do Sarampo/enzimologia , Mutação , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Células Vero
5.
Jpn J Infect Dis ; 74(2): 154-156, 2021 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-32863356

RESUMO

Subacute sclerosing panencephalitis (SSPE) is a late-onset, intractable, and fatal viral disease caused by persistent infection of the central nervous system with a measles virus mutant (SSPE virus). In Japan, interferon-α and ribavirin are administered intracerebroventricularly to patients with SSPE. However, as the therapeutic effect is insufficient, more effective drugs are needed. Favipiravir, which is clinically used as an anti-influenza drug, demonstrates anti-viral effects against RNA viruses. In this study, the antiviral effect of favipiravir against measles virus (Edmonston strain) and SSPE virus (Yamagata-1 strain) was examined in vitro. The 50% effective concentration (EC50) of favipiravir (inhibiting viral plaque formation by 50%) against Edmonston and Yamagata-1 strains were 108.7 ± 2.0 µM (17.1 ± 0.3 µg/mL) and 38.6 ± 6.0 µM (6.1 ± 0.9 µg/mL), respectively, which were similar to those of ribavirin. The antiviral activity of favipiravir against the SSPE virus was demonstrated for the first time in this study.


Assuntos
Amidas/farmacologia , Antivirais/farmacologia , Sarampo/tratamento farmacológico , Pirazinas/farmacologia , Panencefalite Esclerosante Subaguda/tratamento farmacológico , Animais , Chlorocebus aethiops , Humanos , Interferon-alfa/farmacologia , Japão , Sarampo/patologia , Vírus do Sarampo/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Ribavirina/farmacologia , Vírus SSPE/efeitos dos fármacos , Panencefalite Esclerosante Subaguda/patologia , Células Vero
6.
Curr Opin Virol ; 41: 8-17, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32247280

RESUMO

Measles caused an estimated minimum of one million fatalities annually before vaccination. Outstanding progress towards controlling the virus has been made since the measles vaccine was introduced, but reduction of measles case-fatalities has stalled at around 100,000 annually for the last decade and a 2019 resurgence in several geographical regions threatens some of these past accomplishments. Whereas measles eradication through vaccination is feasible, a potentially open-ended endgame of elimination may loom. Other than doubling-down on existing approaches, is it worthwhile to augment vaccination efforts with antiviral therapeutics to solve the conundrum? This question is hypothetical at present, since no drugs have yet been approved specifically for the treatment of measles, or infection by any other pathogen of the paramyxovirus family. This article will consider obstacles that have hampered anti-measles and anti-paramyxovirus drug development, discuss MeV-specific challenges of clinical testing, and define drug properties suitable to address some of these problems.


Assuntos
Desenvolvimento de Medicamentos , Sarampo/prevenção & controle , Animais , Antivirais/farmacologia , Saúde Global , Humanos , Sarampo/epidemiologia , Sarampo/virologia , Vacina contra Sarampo/administração & dosagem , Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/genética , Vírus do Sarampo/imunologia
7.
Molecules ; 25(7)2020 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-32260270

RESUMO

Different parts of Nuphar lutea L. (yellow water lily) have been used to treat several inflammatory and pathogen-related diseases. It has shown that Nuphar lutea extracts (NUP) are active against various pathogens including bacteria, fungi, and leishmanial parasites. In an effort to detect novel therapeutic agents against negative-stranded RNA (- RNA) viruses, we have tested the effect of a partially-purified alkaloid mixture of Nuphar lutea leaves on the measles virus (MV). The MV vaccine's Edmonston strain was used to acutely or persistently infect cells. The levels of several MV proteins were detected by a Western blot and immunocytochemistry. Viral RNAs were quantitated by qRT-PCR. Virus infectivity was monitored by infecting African green monkey kidney VERO cells' monolayers. We showed that NUP protected cells from acute infection. Decreases in the MV P-, N-, and V-proteins were observed in persistently infected cells and the amount of infective virus released was reduced as compared to untreated cells. By examining viral RNAs, we suggest that NUP acts at the post-transcriptional level. We conclude, as a proof of concept, that NUP has anti-viral therapeutic activity against the MV. Future studies will determine the mechanism of action and the effect of NUP on other related viruses.


Assuntos
Alcaloides/farmacologia , Antivirais/farmacologia , Vírus do Sarampo/crescimento & desenvolvimento , Nuphar/química , Alcaloides/química , Animais , Antivirais/química , Chlorocebus aethiops , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/genética , Extratos Vegetais/química , Estudo de Prova de Conceito , RNA Viral/efeitos dos fármacos , Células Vero , Proteínas Virais/efeitos dos fármacos , Proteínas Virais/metabolismo
8.
Antiviral Res ; 178: 104750, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32205137

RESUMO

Picornaviridae are positive-sense single stranded RNA viruses with a similar genomic structure lacking a cap at the 5' end, but with a highly structured 5'-untranslated region (UTR) containing an internal ribosome entry site (IRES). IRES allows ribosomes to be recruited by the viral RNA and initiate translation in a cap-independent manner. Coxsackie virus type B (CV-B) belong to Picornaviridae and are widespread in human population. They usually cause subclinical infections but, occasionally, also severe diseases with various clinical manifestations. CV-B have no specific therapy. DEAD-box polypeptide 3 (DDX3) is a member of the Asp-Glu-Ala-Asp (DEAD)-box family with an ATP-dependent RNA unwinding helicase activity. Recently, several positive-sense single strand RNA viruses have been shown to need DDX3 for their translation. Here, we show that several DDX3 inhibitors reduced CV-B replication and production of viral protein, particularly when added within 12 h of infection. Based on in vitro and in silico data, we hypothesized that DDX3 inhibitors hamper interaction between DDX3 and viral IRES in a stereodynamic fashion. Accordingly, the DDX3 inhibitors tested have no activity against the Vesicular Stomatitis virus and Measles virus, which are negative-sense single stranded RNA viruses and use cap-dependent translation. This study suggests that DDX3 is required by RNA viruses lacking a cap and show that this enzyme is a valuable target to design antiviral molecules against CV-B. Thus, DDX3 is dispensable for cap-dependent translation, but required for translation of transcripts containing secondary structure in their UTRs.


Assuntos
Antivirais/farmacologia , RNA Helicases DEAD-box/antagonistas & inibidores , Enterovirus Humano B/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Antivirais/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , RNA Helicases DEAD-box/metabolismo , Enterovirus Humano B/classificação , Enterovirus Humano B/fisiologia , Inibidores Enzimáticos/química , Humanos , Concentração Inibidora 50 , Sítios Internos de Entrada Ribossomal , Células KB , Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/fisiologia , Vírus de RNA de Sentido Negativo/efeitos dos fármacos , Vírus de RNA de Sentido Negativo/fisiologia , Conformação de Ácido Nucleico , Vírus de RNA de Cadeia Positiva/efeitos dos fármacos , Vírus de RNA de Cadeia Positiva/fisiologia , RNA Viral/química , RNA Viral/genética , RNA Viral/metabolismo , Ribavirina/farmacologia , Sorogrupo , Vesiculovirus/efeitos dos fármacos , Vesiculovirus/fisiologia , Ensaio de Placa Viral , Proteínas Virais/biossíntese , Replicação Viral/efeitos dos fármacos
9.
Vaccine ; 38(3): 460-469, 2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31732326

RESUMO

BACKGROUND: In settings where measles has been eliminated, vaccine-derived immunity may in theory wane more rapidly due to a lack of immune boosting by circulating measles virus. We aimed to assess whether measles vaccine effectiveness (VE) waned over time, and if so, whether differentially in measles-eliminated and measles-endemic settings. METHODS: We performed a systematic literature review of studies that reported VE and time since vaccination with measles-containing vaccine (MCV). We extracted information on case definition (clinical symptoms and/or laboratory diagnosis), method of vaccination status ascertainment (medical record or vaccine registry), as well as any biases which may have arisen from cold chain issues and a lack of an age at first dose of MCV. We then used linear regression to evaluate VE as a function of age at first dose of MCV and time since MCV. RESULTS: After screening 14,782 citations, we identified three full-text articles from measles-eliminated settings and 33 articles from measles-endemic settings. In elimination settings, two-dose VE estimates increased as age at first dose of MCV increased and decreased as time since MCV increased; however, the small number of studies available limited interpretation. In measles-endemic settings, one-dose VE increased by 1.5% (95% CI 0.5, 2.5) for every month increase in age at first dose of MCV. We found no evidence of waning VE in endemic settings. CONCLUSIONS: The paucity of data from measles-eliminated settings indicates that additional studies and approaches (such as studies using proxies including laboratory correlates of protection) are needed to answer the question of whether VE in measles-eliminated settings wanes. Age at first dose of MCV was the most important factor in determining VE. More VE studies need to be conducted in elimination settings, and standards should be developed for information collected and reported in such studies.


Assuntos
Esquemas de Imunização , Vacina contra Sarampo/administração & dosagem , Sarampo/prevenção & controle , Vacinação/tendências , Fatores Etários , Humanos , Lactente , Sarampo/epidemiologia , Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/fisiologia , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos , Resultado do Tratamento
10.
Viruses ; 11(12)2019 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-31801280

RESUMO

Measles virus (MeV) is a paramyxovirus that infects humans, principally children. Despite the existence of an effective and safe vaccine, the number of cases of measles has increased due to lack of vaccination coverage. The World Health Organization (WHO) reports that the number of cases worldwide multiplied fourfold between January and March 2019, to 112,000. Today, there is no treatment available for MeV. In recent years, it has been demonstrated that natural extracts (herbal or algal) with antiviral activity can also work as reducing agents that, in combination with nanotechnology, offer an innovative option to counteract viral infections. Here, we synthetized and evaluated the antiviral activity of gold nanoparticles using garlic extract (Allium sativa) as a reducing agent (AuNPs-As). These nanoparticles actively inhibited MeV replication in Vero cells at a 50% effective concentration (EC50) of 8.829 µg/mL, and the selectivity index (SI) obtained was 16.05. AuNPs-As likely inhibit viral infection by blocking viral particles directly, showing a potent virucidal effect. Gold nanoparticles may be useful as a promising strategy for treating and controlling the infection of MeV and other related enveloped viruses.


Assuntos
Antivirais/farmacologia , Alho/química , Ouro/farmacologia , Vírus do Sarampo/efeitos dos fármacos , Sarampo/tratamento farmacológico , Extratos Vegetais/farmacologia , Animais , Antivirais/química , Chlorocebus aethiops , Ouro/química , Humanos , Sarampo/virologia , Vírus do Sarampo/ultraestrutura , Extratos Vegetais/química , Células Vero
11.
Eur J Epidemiol ; 34(10): 897-915, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31624970

RESUMO

Measles vaccination schedules and targets of herd immunity have been designed according to the paradigm that the vaccine is as protective as natural infection, and the virus has remained of a single serotype over many decades. As a result, ongoing measles resurgence is mostly attributed to gaps in immunization. Using official data, we investigated the correlation between the rate of vaccine coverage reported and aggregated at the national level, and the incidence of cases. We discussed the limits of this indicator considered in isolation. We provide a literature overview of measles vaccine efficacy and failures. We questioned whether measles strains could escape the vaccine. Immunization tools and strategies for measles control deserve to be optimized in the current context.


Assuntos
Surtos de Doenças/prevenção & controle , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/imunologia , Vírus do Sarampo/efeitos dos fármacos , Sarampo/prevenção & controle , Genótipo , Humanos , Vírus do Sarampo/genética , Vacinação
12.
Antiviral Res ; 161: 144-153, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30500403

RESUMO

Secretory IgA (SIgA) antibody is unique for its capability to transit through epithelial cells by transcytosis and thus has opportunities and probabilities to interact with all viral components during viral replication which may result in the inhibition of viral replication intracellularly. Here, we report a novel IgA mAb 1D11-IgA against phosphoprotein (P) of measles virus (MV), which is able to interact specifically with P in MV infected Vero-pIgR cells grown in a two-chamber transwell system. The binding epitope of 1D11-IgA involves a key residue proline 23 in P protein, which is among the α-molecular recognition element (α-MoRE) of P and critical for N0-P complex. The antibody appears to block P to interact with N in P-N complex and thus may inhibit the function of viral RdRp complex, which results in decreased synthesis of viral genome RNA and mRNA. Our data together demonstrate that IgA is able to interact with viral phosphoprotein intraepithelial cells and neutralize viral replication by interrupting formation of P-N complex and function of RdRp. The findings highlight that IgA has a unique anti-viral activity by targeting viral conserved components critical for viral replication, which serves as a proof-of-concept assessment of the druggability of mononegavirales P-N interfaces.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Antivirais/farmacologia , Imunoglobulina A/farmacologia , Vírus do Sarampo/efeitos dos fármacos , Fosfoproteínas/imunologia , Proteínas Virais/imunologia , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Citoplasma/virologia , Genoma Viral , Masculino , Vírus do Sarampo/fisiologia , Camundongos , Testes de Neutralização , Transcitose
13.
ACS Nano ; 12(10): 9855-9865, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30230818

RESUMO

Measles remains one of the leading causes of child mortality worldwide and is re-emerging in some countries due to poor vaccine coverage, concomitant with importation of measles virus (MV) from endemic areas. The lack of specific chemotherapy contributes to negative outcomes, especially in infants or immunodeficient individuals. Fusion inhibitor peptides derived from the MV Fusion protein C-terminal Heptad Repeat (HRC) targeting MV envelope fusion glycoproteins block infection at the stage of entry into host cells, thus preventing viral multiplication. To improve efficacy of such entry inhibitors, we have modified a HRC peptide inhibitor by introducing properties of self-assembly into nanoparticles (NP) and higher affinity for both viral and cell membranes. Modification of the peptide consisted of covalent grafting with tocopherol to increase amphipathicity and lipophilicity (HRC5). One additional peptide inhibitor consisting of a peptide dimer grafted to tocopherol was also used (HRC6). Spectroscopic, imaging, and simulation techniques were used to characterize the NP and explore the molecular basis for their antiviral efficacy. HRC5 forms micellar stable NP while HRC6 aggregates into amorphous, loose, unstable NP. Interpeptide cluster bridging governs NP assembly into dynamic metastable states. The results are consistent with the conclusion that the improved efficacy of HRC6 relative to HRC5 can be attributed to NP instability, which leads to more extensive partition to target membranes and binding to viral target proteins.


Assuntos
Antivirais/farmacologia , Vírus do Sarampo/efeitos dos fármacos , Nanopartículas/química , Peptídeos/farmacologia , Tocoferóis/farmacologia , Antivirais/química , Testes de Sensibilidade Microbiana , Peptídeos/química , Tocoferóis/química , Proteínas Virais de Fusão/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos
14.
Viruses ; 10(9)2018 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-30200234

RESUMO

Although preventable by vaccination, Measles still causes thousands of deaths among young children worldwide. The discovery of new antivirals is a good approach to control new outbreaks that cause such death. In this study, we tested the antiviral activity against Measles virus (MeV) of Polyphenol-rich extracts (PPs) coming from five seaweeds collected and cultivated in Mexico. An MTT assay was performed to determine cytotoxicity effect, and antiviral activity was measured by syncytia reduction assay and confirmed by qPCR. PPs from Ecklonia arborea (formerly Eisenia arborea, Phaeophyceae) and Solieria filiformis (Rhodophyta) showed the highest Selectivity Index (SI), >3750 and >576.9 respectively. Both PPs extracts were selected to the subsequent experiments owing to their high efficacy and low cytotoxicity compared with ribavirin (SI of 11.57). The combinational effect of PPs with sulphated polysaccharides (SPs) and ribavirin were calculated by using Compusyn software. Synergistic activity was observed by combining both PPs with low concentrations of Solieria filiformis SPs (0.01 µg/mL). The antiviral activity of the best combinations was confirmed by qPCR. Virucidal assay, time of addition, and viral penetration evaluations suggested that PPs act mainly by inactivating the viral particle. To our knowledge, this is the first report of the virucidal effect of Polyphenol-rich extracts of seaweeds.


Assuntos
Antivirais/farmacologia , Sinergismo Farmacológico , Vírus do Sarampo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Polifenóis/farmacologia , Alga Marinha/química , Animais , Antivirais/isolamento & purificação , Antivirais/toxicidade , Chlorocebus aethiops , México , Testes de Sensibilidade Microbiana , Viabilidade Microbiana , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/toxicidade , Polifenóis/isolamento & purificação , Polifenóis/toxicidade , Polissacarídeos/farmacologia , Ribavirina/farmacologia , Células Vero
15.
J Virol ; 92(17)2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29925665

RESUMO

We found earlier that ectopic expression of the cytidine deaminase APOBEC3G (A3G) in Vero cells inhibits measles virus (MV), respiratory syncytial virus, and mumps virus, while the mechanism of inhibition remained unclear. A microarray analysis revealed that in A3G-transduced Vero cells, several cellular transcripts were differentially expressed, suggesting that A3G regulates the expression of host factors. One of the most upregulated host cell factors, REDD1 (regulated in development and DNA damage response-1, also called DDIT4), reduced MV replication ∼10-fold upon overexpression in Vero cells. REDD1 is an endogenous inhibitor of mTORC1 (mammalian target of rapamycin complex-1), the central regulator of cellular metabolism. Interestingly, rapamycin reduced the MV replication similarly to REDD1 overexpression, while the combination of both did not lead to further inhibition, suggesting that the same pathway is affected. REDD1 silencing in A3G-expressing Vero cells abolished the inhibitory effect of A3G. In addition, silencing of A3G led to reduced REDD1 expression, confirming that its expression is regulated by A3G. In primary human peripheral blood lymphocytes (PBL), expression of A3G and REDD1 was found to be stimulated by phytohemagglutinin (PHA) and interleukin-2. Small interfering RNA (siRNA)-mediated depletion of A3G in PHA-stimulated PBL reduced REDD1 expression and increased viral titers, which corroborates our findings in Vero cells. Silencing of REDD1 also increased viral titers, confirming the antiviral role of REDD1. Finally, pharmacological inhibition of mTORC1 by rapamycin in PHA-stimulated PBL reduced viral replication to the level found in unstimulated lymphocytes, indicating that mTORC1 activity supports MV replication as a proviral host factor.IMPORTANCE Knowledge about host factors supporting or restricting virus replication is required for a deeper understanding of virus-cell interactions and may eventually provide the basis for therapeutic intervention. This work was undertaken predominantly to explain the mechanism of A3G-mediated inhibition of MV, a negative-strand RNA virus that is not affected by the deaminase activity of A3G acting on single-stranded DNA. We found that A3G regulates the expression of several cellular proteins, which influences the capacity of the host cell to replicate MV. One of these, REDD1, which modulates the cellular metabolism in a central position by regulating the kinase complex mTORC1, was identified as the major cellular factor impairing MV replication. These findings show interesting aspects of the function of A3G and the dependence of the MV replication on the metabolic state of the cell. Interestingly, pharmacological inhibition of mTORC1 can be utilized to inhibit MV replication in Vero cells and primary human peripheral blood lymphocytes.


Assuntos
Desaminase APOBEC-3G/genética , Vírus do Sarampo/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Replicação Viral/genética , Desaminase APOBEC-3G/metabolismo , Animais , Antivirais/farmacologia , Linhagem Celular , Chlorocebus aethiops , Replicação do DNA , Interações Hospedeiro-Patógeno/genética , Humanos , Interleucina-2/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/virologia , Linfócitos/efeitos dos fármacos , Linfócitos/virologia , Vírus do Sarampo/efeitos dos fármacos , Alvo Mecanístico do Complexo 1 de Rapamicina/efeitos dos fármacos , Fito-Hemaglutininas/farmacologia , RNA Interferente Pequeno , Sirolimo/farmacologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/efeitos dos fármacos , Células Vero , Replicação Viral/efeitos dos fármacos
17.
J Virol ; 91(23)2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28904193

RESUMO

The inhibitors carbobenzoxy (Z)-d-Phe-l-Phe-Gly (fusion inhibitor peptide [FIP]) and 4-nitro-2-phenylacetyl amino-benzamide (AS-48) have similar efficacies in blocking membrane fusion and syncytium formation mediated by measles virus (MeV). Other homologues, such as Z-d-Phe, are less effective but may act through the same mechanism. In an attempt to map the site of action of these inhibitors, we generated mutant viruses that were resistant to the inhibitory effects of Z-d-Phe-l-Phe-Gly. These 10 mutations were localized to the heptad repeat B (HRB) region of the fusion protein, and no changes were observed in the viral hemagglutinin, which is the receptor attachment protein. Mutations were validated in a luciferase-based membrane fusion assay, using transfected fusion and hemagglutinin expression plasmids or with syncytium-based assays in Vero, Vero-SLAM, and Vero-Nectin 4 cell lines. The changes I452T, D458N, D458G/V459A, N462K, N462H, G464E, and I483R conferred resistance to both FIP and AS-48 without compromising membrane fusion. The inhibitors did not block hemagglutinin protein-mediated binding to the target cell. Edmonston vaccine/laboratory and IC323 wild-type strains were equally affected by the inhibitors. Escape mutations were mapped upon a three-dimensional (3D) structure modeled from the published crystal structure of parainfluenzavirus 5 fusion protein. The most effective mutations were situated in a region located near the base of the globular head and its junction with the alpha-helical stalk of the prefusion protein. We hypothesize that the fusion inhibitors could interfere with the structural changes that occur between the prefusion and postfusion conformations of the fusion protein.IMPORTANCE Due to lapses in vaccination worldwide that have caused localized outbreaks, measles virus (MeV) has regained importance as a pathogen. Antiviral agents against measles virus are not commercially available but could be useful in conjunction with MeV eradication vaccine programs and as a safeguard in oncolytic viral therapy. Three decades ago, the small hydrophobic peptide Z-d-Phe-l-Phe-Gly (FIP) was shown to block MeV infections and syncytium formation in monkey kidney cell lines. The exact mechanism of its action has yet to be determined, but it does appear to have properties similar to those of another chemical inhibitor, AS-48, which appears to interfere with the conformational change in the viral F protein that is required to elicit membrane fusion. Escape mutations were used to map the site of action for FIP. Knowledge gained from these studies could help in the design of new inhibitors against morbilliviruses and provide additional knowledge concerning the mechanism of virus-mediated membrane fusion.


Assuntos
Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/genética , Mutação , Oligopeptídeos/farmacologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Animais , Antivirais/farmacologia , Benzamidas/farmacologia , Chlorocebus aethiops , Hemaglutininas Virais/genética , Hemaglutininas Virais/metabolismo , Fusão de Membrana/efeitos dos fármacos , Modelos Moleculares , Ligação Proteica , Células Vero , Proteínas Virais de Fusão/química , Internalização do Vírus/efeitos dos fármacos
18.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28250131

RESUMO

Measles virus (MeV) is a member of the family Paramixoviridae that causes a highly contagious respiratory disease but has emerged as a promising oncolytic platform. Previous studies of MeV entry focused on the identification of cellular receptors. However, the endocytic and trafficking pathways utilized during MeV entry remain poorly described. The contribution of each endocytic pathway has been examined in cells that express the MeV receptors SLAM (signaling lymphocyte-activating molecule) and PVRL4 (poliovirus receptor-like 4) (nectin-4). Recombinant MeVs expressing either firefly luciferase or green fluorescent protein together with a variety of inhibitors were used. The results showed that MeV uptake was dynamin independent in the Vero.hPVRL4, Vero.hSLAM, and PVRL4-positive MCF7 breast cancer cell lines. However, MeV infection was blocked by 5-(N-ethyl-N-propyl)amiloride (EIPA), the hallmark inhibitor of macropinocytosis, as well as inhibitors of actin polymerization. By using phalloidin staining, MeV entry was shown to induce actin rearrangements and the formation of membrane ruffles accompanied by transient elevated fluid uptake. Small interfering RNA (siRNA) knockdown of p21-activated kinase 1 (PAK1) demonstrated that MeV enters both Vero.hPVRL4 and Vero.hSLAM cells in a PAK1-independent manner using a macropinocytosis-like pathway. In contrast, MeV entry into MCF7 human breast cancer cells relied upon Rac1 and its effector PAK1 through a PVRL4-mediated macropinocytosis pathway. MeV entry into DLD-1 colon and HTB-20 breast cancer cells also appeared to use the same pathway. Overall, these findings provide new insight into the life cycle of MeV, which could lead to therapies that block virus entry or methods that improve the uptake of MeV by cancer cells during oncolytic therapy.IMPORTANCE In the past decades, measles virus (MeV) has emerged as a promising oncolytic platform. Previous studies concerning MeV entry focused mainly on the identification of putative receptors for MeV. Nectin-4 (PVRL4) was recently identified as the epithelial cell receptor for MeV. However, the specific endocytic and trafficking pathways utilized during MeV infections are poorly documented. In this study, we demonstrated that MeV enters host cells via a dynamin-independent and actin-dependent endocytic pathway. Moreover, we show that MeV gains entry into MCF7, DLD-1, and HTB-20 cancer cells through a PVRL4-mediated macropinocytosis pathway and identified the typical cellular GTPase and kinase involved. Our findings provide new insight into the life cycle of MeV, which may lead to the development of therapies that block the entry of the virus into the host cell or alternatively promote the uptake of oncolytic MeV into cancer cells.


Assuntos
Moléculas de Adesão Celular/metabolismo , Vírus do Sarampo/fisiologia , Pinocitose , Internalização do Vírus , Actinas/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Neoplasias da Mama , Linhagem Celular , Chlorocebus aethiops , Neoplasias do Colo , Células Epiteliais/virologia , Feminino , Humanos , Células MCF-7 , Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/genética , Vírus Oncolíticos/fisiologia , Pinocitose/efeitos dos fármacos , RNA Interferente Pequeno/genética , Células Vero , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
19.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27733647

RESUMO

Measles virus (MV) infection is undergoing resurgence and remains one of the leading causes of death among young children worldwide despite the availability of an effective measles vaccine. MV infects its target cells by coordinated action of the MV hemagglutinin (H) and fusion (F) envelope glycoproteins; upon receptor engagement by H, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. By interfering with this structural transition of F, peptides derived from the heptad repeat (HR) regions of F can inhibit MV infection at the entry stage. In previous work, we have generated potent MV fusion inhibitors by dimerizing the F-derived peptides and conjugating them to cholesterol. We have shown that prophylactic intranasal administration of our lead fusion inhibitor efficiently protects from MV infection in vivo We show here that peptides tagged with lipophilic moieties self-assemble into nanoparticles until they reach the target cells, where they are integrated into cell membranes. The self-assembly feature enhances biodistribution and the half-life of the peptides, while integration into the target cell membrane increases fusion inhibitor potency. These factors together modulate in vivo efficacy. The results suggest a new framework for developing effective fusion inhibitory peptides. IMPORTANCE: Measles virus (MV) infection causes an acute illness that may be associated with infection of the central nervous system (CNS) and severe neurological disease. No specific treatment is available. We have shown that fusion-inhibitory peptides delivered intranasally provide effective prophylaxis against MV infection. We show here that specific biophysical properties regulate the in vivo efficacy of MV F-derived peptides.


Assuntos
Hemaglutininas Virais/imunologia , Vacina contra Sarampo/administração & dosagem , Vírus do Sarampo/efeitos dos fármacos , Sarampo/prevenção & controle , Nanopartículas/administração & dosagem , Peptídeos/imunologia , Proteínas Virais de Fusão/imunologia , Administração Intranasal , Sequência de Aminoácidos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Colesterol/química , Feminino , Meia-Vida , Hemaglutininas Virais/química , Humanos , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Masculino , Sarampo/imunologia , Sarampo/mortalidade , Sarampo/virologia , Vacina contra Sarampo/síntese química , Vírus do Sarampo/química , Vírus do Sarampo/imunologia , Nanopartículas/química , Peptídeos/síntese química , Sigmodontinae , Análise de Sobrevida , Proteínas Virais de Fusão/química , Internalização do Vírus/efeitos dos fármacos
20.
PLoS One ; 11(9): e0162823, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27612283

RESUMO

Subacute sclerosing panencephalitis (SSPE) is a persistent, progressive, and fatal degenerative disease resulting from persistent measles virus (MV) infection of the central nervous system. Most drugs used to treat SSPE have been reported to have limited effects. Therefore, novel therapeutic strategies are urgently required. The SSPE virus, a variant MV strain, differs virologically from wild-type MV strain. One characteristic of the SSPE virus is its defective production of cell-free virus, which leaves cell-to-cell infection as the major mechanism of viral dissemination. The fusion protein plays an essential role in this cell-to-cell spread. It contains two critical heptad repeat regions that form a six-helix bundle in the trimer similar to most viral fusion proteins. In the case of human immunodeficiency virus type-1 (HIV-1), a synthetic peptide derived from the heptad repeat region of the fusion protein enfuvirtide inhibits viral replication and is clinically approved as an anti-HIV-1 agent. The heptad repeat regions of HIV-1 are structurally and functionally similar to those of the MV fusion protein. We therefore designed novel peptides derived from the fusion protein heptad repeat region of the MV and examined their effects on the measles and SSPE virus replication in vitro and in vivo. Some of these synthetic novel peptides demonstrated high antiviral activity against both the measles (Edmonston strain) and SSPE (Yamagata-1 strain) viruses at nanomolar concentrations with no cytotoxicity in vitro. In particular, intracranial administration of one of the synthetic peptides increased the survival rate from 0% to 67% in an SSPE virus-infected nude mouse model.


Assuntos
Vírus do Sarampo/fisiologia , Peptídeos/farmacologia , Sequências Repetitivas de Aminoácidos , Proteínas Virais de Fusão/química , Replicação Viral/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Antivirais/farmacologia , Chlorocebus aethiops , Modelos Animais de Doenças , HIV-1/metabolismo , Humanos , Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Peptídeos/química , Peptídeos/metabolismo , Panencefalite Esclerosante Subaguda/patologia , Panencefalite Esclerosante Subaguda/virologia , Fatores de Tempo , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...