Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 96(14): e0066522, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35762760

RESUMO

Human astrovirus VA1 has been associated with neurological disease in immunocompromised patients, and its recent propagation in cell culture has opened the possibility to study its biology. Unlike classical human astroviruses, VA1 growth was found to be independent of trypsin during virus replication in vitro. In this work, we show that despite its independence on trypsin activation for cell infection, the VA1 capsid precursor protein, of 86 kDa (VP86), is processed intracellularly, and this proteolytic processing is important for astrovirus VA1 infectivity. Antibodies raised against different regions of the capsid precursor showed that the polyprotein can be processed starting at either its amino- or carboxy-terminal end, and they allowed us to identify those proteins of about 33 (VP33) and 38 (VP38) kDa constitute the core and the spike proteins of the mature infectious virus particles, respectively. The amino-terminal end of the spike protein was found to be Thr-348. Whether the protease involved in intracellular cleavage of the capsid precursor is of viral or cellular origin remains to be determined, but the cleavage is independent of caspases. Also, trypsin is able to degrade the capsid precursor but has no effect on VP33 and VP38 proteins when assembled into virus particles. These studies provide the basis for advancement of the knowledge of astrovirus VA1 cell entry and replication. IMPORTANCE Human astrovirus VA1 has been associated with neurological disease in immunocompromised patients. Its recent propagation in cell culture has facilitated the study of its biology. In this work, we show that despite the ability of this virus to grow in the absence of trypsin, a marked feature of human classical astroviruses, the capsid precursor protein of astrovirus VA1 is cleaved intracellularly to yield the mature infectious particles, formed by two polypeptides, VP33 that constitutes the core domain of the virus particle, and VP38 that forms the spike of the virus. These studies provide a platform to advance our knowledge on astrovirus VA1 cell entry and replication.


Assuntos
Infecções por Astroviridae , Proteínas do Capsídeo , Mamastrovirus , Precursores de Proteínas , Infecções por Astroviridae/virologia , Células CACO-2 , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Humanos , Espaço Intracelular/virologia , Mamastrovirus/fisiologia , Precursores de Proteínas/metabolismo , Tripsina/metabolismo
2.
Sci Rep ; 9(1): 8682, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31213630

RESUMO

Dengue fever is one of the most important mosquito-borne viral infections in large parts of tropical and subtropical countries and is a significant public health concern and socioeconomic burden. There is an urgent need to develop antivirals that can effectively reduce dengue virus (DENV) replication and decrease viral load. Niclosamide, an antiparasitic drug approved for human use, has been recently identified as an effective antiviral agent against a number of pH-dependent viruses, including flaviviruses. Here, we reveal that neutralization of low-pH intracellular compartments by niclosamide affects multiple steps of the DENV infectious cycle. Specifically, niclosamide-induced endosomal neutralization not only prevents viral RNA replication but also affects the maturation of DENV particles, rendering them non-infectious. We found that niclosamide-induced endosomal neutralization prevented E glycoprotein conformational changes on the virion surface of flaviviruses, resulting in the release of non-infectious immature virus particles with uncleaved pr peptide from host cells. Collectively, our findings support the potential application of niclosamide as an antiviral agent against flavivirus infection and highlight a previously uncharacterized mechanism of action of the drug.


Assuntos
Vesículas Citoplasmáticas/efeitos dos fármacos , Vírus da Dengue/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Espaço Intracelular/efeitos dos fármacos , Niclosamida/farmacologia , Animais , Antivirais/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Vesículas Citoplasmáticas/química , Vesículas Citoplasmáticas/virologia , Vírus da Dengue/genética , Vírus da Dengue/crescimento & desenvolvimento , Endossomos/química , Endossomos/virologia , Humanos , Concentração de Íons de Hidrogênio , Espaço Intracelular/química , Espaço Intracelular/virologia , Estágios do Ciclo de Vida/efeitos dos fármacos , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Vírion/efeitos dos fármacos , Vírion/genética , Vírion/crescimento & desenvolvimento , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
3.
Biomed Pharmacother ; 116: 108976, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31103827

RESUMO

With the development of more effective direct-acting antivirals (DAAs), dual- or triple-therapy regimens represent the major strategy used to cure chronic hepatitis C virus (HCV) infection. Thus, shorter treatment duration regimens with low burden, few adverse effects and good patient adherence are urgently needed. This study theoretically demonstrates a proof-of-concept approach for shortening therapy duration by examining HCV-infected Huh7.5 cells after treatment with a high or low fixed dose of three DAAs (simeprevir + daclatasvir + sofosbuvir) for 6-15 days. The results demonstrated that HCV-infected Huh7.5 cells achieved an ultrarapid virologic response with undetectable HCV RNA and protein and were cured after treatment with the triple-therapy regimen for 15 days. When the treatment duration was shortened, virologic relapse might occur after treatment with a low fixed dose of the three DAAs for 9 days and did occur after treatment with a low fixed dose for 6 days, although HCV was below detectable levels at the end of treatment. However, virologic relapse could be avoided with treatment of a high fixed dose of the three DAAs for 9 or 6 days. Although a virologic breakthrough occurred after an intermittent treatment regimen at the low fixed dose, the high fixed dose cured HCV-positive Huh7.5 cells with intermittent treatment. In conclusion, HCV is persistently present below detectable levels in HCV-infected Huh7.5 cells for a long time after treatment, and a shortened therapy duration is associated with an increased risk of virologic relapse, but virologic relapse or breakthrough might be avoided by treatment with a combination of more highly effective DAAs.


Assuntos
Antivirais/uso terapêutico , Hepacivirus/fisiologia , Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/virologia , Antivirais/farmacologia , Carbamatos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Quimioterapia Combinada , Hepacivirus/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Espaço Intracelular/virologia , Pirrolidinas , Recidiva , Simeprevir/farmacologia , Simeprevir/uso terapêutico , Sofosbuvir/farmacologia , Sofosbuvir/uso terapêutico , Valina/análogos & derivados , Replicação Viral/efeitos dos fármacos
4.
Clin Chim Acta ; 495: 100-108, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30959045

RESUMO

Inflammasomes play a very important role in the host defense against multiple pathogenic microbes, including bacteria and viruses. Inflammasomes are multiprotein complex platforms that mediate the processing of the two most important inflammatory cytokines, pro-IL-1ß and pro-IL-18, to their active forms. The inflammasome is formed by the apoptosis-associated speck-like protein containing a CARD (ASC), procaspase-1 and a sensor protein, either a NOD-like receptor (NLR) or an absent in melanoma 2 (AIM2)-like receptor. The sensor molecule determines inflammasome specificity by detecting specific and conserved microbial products or cell stress signals. Compared with the other inflammasomes, there is much more unknown about the activation or regulation mechanisms of the AIM2 inflammasome. In this review, we will discuss these mechanisms and the specific roles of the AIM2 inflammasome in response to diverse pathogens.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Infecções/microbiologia , Infecções/virologia , Inflamassomos/metabolismo , Animais , Caspases/metabolismo , Humanos , Infecções/metabolismo , Infecções/patologia , Espaço Intracelular/metabolismo , Espaço Intracelular/microbiologia , Espaço Intracelular/virologia
5.
J Theor Biol ; 473: 20-27, 2019 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-31004612

RESUMO

Even in the presence of a successful combination therapy stalling the progress of AIDS, developing a cure for this disease is still an open question. One of the major steps towards a cure would be to be able to eradicate latent HIV reservoirs present in patients. During the last decade, multiple findings point to the dominant role of the viral protein Tat in the establishment of latency. Here we present a mathematical study to understand the potential role of Tat inhibitors as virus-suppressing agents. For this aim, we implemented a computational model that reproduces intracellular dynamics. Simulating an HIV-infected cell and its intracellular feedback we observed that removing Tat protein from the system via inhibitors resulted in a temporary and reversible viral suppression. In contrast, we observed that compounds that interact with Tat protein and disrupt the integrated viral genome produced a more permanent viral suppression.


Assuntos
Antivirais/farmacologia , HIV/fisiologia , Modelos Biológicos , Latência Viral/efeitos dos fármacos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Simulação por Computador , Regulação Viral da Expressão Gênica/efeitos dos fármacos , HIV/efeitos dos fármacos , HIV/genética , Espaço Intracelular/virologia , Fatores de Tempo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
6.
ACS Infect Dis ; 4(2): 88-92, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29298032

RESUMO

There is a large, global unmet need for the development of countermeasures to combat intracellular pathogens. The development of novel antimicrobials is expensive and slow and typically focuses on selective inhibition of proteins encoded by a single pathogen, thereby providing a narrow spectrum of coverage. The repurposing of approved drugs targeting host functions required for microbial infections represents a promising alternative. This review summarizes progress and challenges in the repurposing of approved drugs as host-targeted broad-spectrum agents for the treatment of intracellular pathogens. These strategies include targeting both cellular factors required for infection by various viruses, intracellular bacteria, and/or protozoa as well as factors that modulate the host immune response to these microbial infections. The repurposed approach offers complementary means to develop therapeutics against existing and emerging intracellular microbial threats.


Assuntos
Anti-Infecciosos/farmacologia , Reposicionamento de Medicamentos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/microbiologia , Animais , Antineoplásicos/farmacologia , Humanos , Espaço Intracelular/virologia , Terapia de Alvo Molecular
7.
PLoS One ; 11(12): e0168939, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28033419

RESUMO

The budded phenotype (BV) of the baculovirus AcMNPV has been demonstrated to have strong immunostimulatory properties that are relevant for the development of vaccines and antiviral therapies. Although the occluded phenotype (ODV) shares the main structural proteins and its genome with BV, it has been poorly studied in mammals. In this study, we assessed the capacity of ODV to induce immune responses in mice. In contrast to BVs, ODVs failed to promote the secretion of IFN-gamma, IL-6 and Il-12 and to induce antiviral activity against VSV in the short term. Furthermore, ODVs were unable to induce cellular immunity against a coadministered antigen 7 days after inoculation. By analyzing the interaction of ODVs with BMDCs, we observed that although ODVs entered the cells reaching late and acidic endosomes, they did not induce their maturation. Finally, we also analyzed if BVs and ODVs followed different routes in the cell during the infection. BVs, but not ODVs, colocalized with the protein ovalbumin in compartments with the presence of proteases. The results suggest that structural differences could be responsible for their different destinies in the dendritic cell and this could lead to a different impact on the immune response.


Assuntos
Espaço Intracelular/virologia , Nucleopoliedrovírus/fisiologia , Fenótipo , Animais , Células da Medula Óssea/citologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Endossomos/virologia , Feminino , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Endogâmicos C57BL , Células Sf9 , Spodoptera
8.
Proc Natl Acad Sci U S A ; 112(29): 8887-92, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26195777

RESUMO

In the present article we examine clonality in virus evolution. Most viruses retain an active recombination machinery as a potential means to initiate new levels of genetic exploration that go beyond those attainable solely by point mutations. However, despite abundant recombination that may be linked to molecular events essential for genome replication, herein we provide evidence that generation of recombinants with altered biological properties is not essential for the completion of the replication cycles of viruses, and that viral lineages (near-clades) can be defined. We distinguish mechanistically active but inconsequential recombination from evolutionarily relevant recombination, illustrated by episodes in the field and during experimental evolution. In the field, recombination has been at the origin of new viral pathogens, and has conferred fitness advantages to some viruses once the parental viruses have attained a sufficient degree of diversification by point mutations. In the laboratory, recombination mediated a salient genome segmentation of foot-and-mouth disease virus, an important animal pathogen whose genome in nature has always been characterized as unsegmented. We propose a model of continuous mutation and recombination, with punctuated, biologically relevant recombination events for the survival of viruses, both as disease agents and as promoters of cellular evolution. Thus, clonality is the standard evolutionary mode for viruses because recombination is largely inconsequential, since the decisive events for virus replication and survival are not dependent on the exchange of genetic material and formation of recombinant (mosaic) genomes.


Assuntos
Evolução Biológica , Espaço Intracelular/virologia , Poliploidia , Vírus/genética , Animais , Células Clonais , Vírus da Febre Aftosa/genética , Modelos Biológicos , Recombinação Genética/genética , Terminologia como Assunto
9.
J Theor Biol ; 366: 91-102, 2015 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-25476731

RESUMO

UNLABELLED: The progression of acute hepatitis B virus (HBV) to chronic infection or clearance is highly dependent on the host immune response composed of cytolytic (CTL) and non-cytolytic (non-CTL) effects. Cytolytic processes induce hepatocyte killing while non-CTL processes inhibit intracellular replication. Both effects are widely recognized and accepted. However, there are uncertainties about the assistance provided by either the loss of covalently circular closed DNA (cccDNA) during cell proliferation or the emergence of refractory cells to immune mediated clearance. We developed an agent-based mathematical model and tested the relative roles of different mechanisms of the immune system in the clearance of acute HBV infection. HBV viremia clearance time and hepatocyte turnover (HT) were used as the two major criteria in determining reasonable outcomes. Modelling results in 90% of cells containing between 1 and 17 cccDNA copies and normally distributed at the peak of infection. Variations in p36 levels, responsible for determining export of virions or recirculation to amplify cccDNA numbers, have a much greater impact on mean cccDNA level/cell at peak viremia than virus infectivity and cccDNA half-life. A strong CTL effect alone failed to clear infection with HT ≈ 10. Acute infection clearance was possible with combined CTL and non-CTL effects along with complete loss of intracellular viral components during cell proliferation resulting in the desired range of HT (0.7-1). The emergence of cells refractory to infection can reduce HT by up to 90%. However their impact was less effective than complete loss of intracellular viral components during cell proliferation. CONCLUSION: the existence of refractory cells is not necessary when there is complete loss of intracellular quantities during cell proliferation but is essential with only partial clearance.


Assuntos
Simulação por Computador , Vírus da Hepatite B/fisiologia , Hepatite B/imunologia , Hepatite B/virologia , Doença Aguda , Imunidade Adaptativa , Animais , Antivirais/uso terapêutico , Proliferação de Células , Citotoxicidade Imunológica , DNA Circular/genética , DNA Viral/genética , Hepatite B/patologia , Vírus da Hepatite B/patogenicidade , Hepatite B Crônica/tratamento farmacológico , Hepatite B Crônica/virologia , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Espaço Intracelular/virologia , Viremia/imunologia , Viremia/virologia , Vírion/patogenicidade , Replicação Viral
10.
Biomed Res Int ; 2013: 965853, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24078930

RESUMO

It has been reported that diabetes mellitus (DM) was an epidemiologically identified risk factor for development of dengue hemorrhagic fever (DHF)/severe dengue in dengue virus (DENV) affected patients, and T helper 2 (Th2) cytokines such as interleukin-4 (IL-4) and IL-10 each plays an important role in the immunopathogenesis of DHF in studies involving general population. To better understand the relationship between these epidemiological and immunological findings, we performed an in vitro study evaluating the sequential immunological reactions and viral load in the DENV infected mononuclear cells of adults with type 2 DM (T2DM group, n = 33) and normal adults (control group, n = 29). We found in the T2DM group significantly higher IL-4 level on the first (P = 0.049) and the third (P = 0.022) postinfection days, while higher IL-10 (P = 0.042) and granulocyte-macrophage colony-stimulating factor (GM-CSF) (P = 0.009) were detected on the third postinfection day. No significant difference in DENV viral load between the cultured mononuclear cells from both groups was found on the first and third post-infection days. These data immunologically suggest that patients with T2DM are at higher risk for development of DHF/severe dengue and strengthen the previously epidemiologically identified role of DM being a predictive risk factor for progressing into DHF/severe dengue in DENV-affected patients.


Assuntos
Vírus da Dengue/fisiologia , Diabetes Mellitus Tipo 2/virologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Interleucina-10/biossíntese , Interleucina-4/biossíntese , Leucócitos Mononucleares/virologia , Replicação Viral/fisiologia , Estudos de Casos e Controles , Quimiocina CCL2/metabolismo , Demografia , Dengue/complicações , Dengue/virologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Espaço Intracelular/virologia , Masculino , Pessoa de Meia-Idade , Fator de Necrose Tumoral alfa/metabolismo , Carga Viral
11.
PLoS One ; 8(8): e71316, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951130

RESUMO

Human Rhinovirus (HRV) infection results in shut down of essential cellular processes, in part through disruption of nucleocytoplasmic transport by cleavage of the nucleoporin proteins (Nups) that make up the host cell nuclear pore. Although the HRV genome encodes two proteases (2A and 3C) able to cleave host proteins such as Nup62, little is known regarding the specific contribution of each. Here we use transfected as well as HRV-infected cells to establish for the first time that 3C protease is most likely the mediator of cleavage of Nup153 during HRV infection, while Nup62 and Nup98 are likely to be targets of HRV2A protease. HRV16 3C protease was also able to elicit changes in the appearance and distribution of the nuclear speckle protein SC35 in transfected cells, implicating it as a key mediator of the mislocalisation of SC35 in HRV16-infected cells. In addition, 3C protease activity led to the redistribution of the nucleolin protein out of the nucleolus, but did not affect nuclear localisation of hnRNP proteins, implying that complete disruption of nucleocytoplasmic transport leading to relocalisation of hnRNP proteins from the nucleus to the cytoplasm in HRV-infected cells almost certainly requires 2A in addition to 3C protease. Thus, a specific role for HRV 3C protease in cleavage and mislocalisation of host cell nuclear proteins, in concert with 2A, is implicated for the first time in HRV pathogenesis.


Assuntos
Cisteína Endopeptidases/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Nucleares/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Virais/metabolismo , Proteases Virais 3C , Transporte Ativo do Núcleo Celular , Animais , Western Blotting , Células COS , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Chlorocebus aethiops , Cisteína Endopeptidases/genética , Citoplasma/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Espaço Intracelular/metabolismo , Espaço Intracelular/virologia , Microscopia Confocal , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteólise , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Rhinovirus/genética , Rhinovirus/metabolismo , Rhinovirus/fisiologia , Ribonucleoproteínas/genética , Fatores de Processamento de Serina-Arginina , Transfecção , Proteínas Virais/genética , Nucleolina
12.
Biomaterials ; 34(13): 3511-22, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23415044

RESUMO

Recently, peptide drugs have evolved into mainstream therapeutics, representing a significant portion of the pharmaceutical market. However, their bioavailability remains to be improved compared with that of chemical drugs. Here, we screened and identified a new peptide, Ctry2459, from a scorpion venom peptide library that was proven to inhibit hepatitis C virus (HCV) infection via inactivating infectious viral particles. However, Ctry2459 cannot suppress established infection of HCV because of the poor cellular uptake and restriction of endosomes. Based on the molecular template of the Ctry2459 peptide, we designed two histidine-rich peptides (Ctry2459-H2 and Ctry2459-H3) with significantly enhanced cellular uptake and improved intracellular distribution. Moreover, the two mutated peptides, as well as the wild-type peptide Ctry2459, exhibited virucidal activities against HCV. In distinct contrast to the Ctry2459 peptide, the mutated peptides significantly suppressed the established HCV infection at the cellular level but demonstrated lower cytotoxic and hemolytic activities. Our work presents an effective design strategy for optimizing natural antiviral peptides and opens a new avenue for enhancing the bioavailability of peptide drugs.


Assuntos
Antivirais/farmacologia , Antivirais/farmacocinética , Hepacivirus/efeitos dos fármacos , Peptídeos/farmacologia , Peptídeos/farmacocinética , Proteínas/farmacologia , Ácidos/metabolismo , Sequência de Aminoácidos , Antivirais/análise , Antivirais/química , Disponibilidade Biológica , Linhagem Celular Tumoral , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/virologia , Biblioteca Gênica , Hemólise/efeitos dos fármacos , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/virologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Peptídeos/análise , Peptídeos/química , Transporte Proteico/efeitos dos fármacos , Proteínas/química , ATPases Translocadoras de Prótons/metabolismo , Venenos de Escorpião/química , Venenos de Escorpião/farmacocinética , Venenos de Escorpião/farmacologia , Fatores de Tempo , Inativação de Vírus/efeitos dos fármacos
13.
Int J Mol Med ; 31(4): 867-73, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23403777

RESUMO

Clemastanin B, 7S,8R,8'R-(-)-lariciresinol-4,4'-bis-O-ß-D-glucopyranoside, is one of the major lignans extracted from Isatis indigotica root (IIR). In this study, the anti-influenza activities of clemastanin B were evaluated in vitro. Clemastanin B was found to inhibit different subtypes of human (H1N1, including swine-origin H1N1; H3N2 and influenza B) and avian influenza viruses (H6N2, H7N3, H9N2) at different magnitudes of activity (IC50 0.087-0.72 mg/ml) while this compound was inactive against respiratory syncytial virus (RSV), adenovirus 3 (ADV3), parainfluenza virus 3 (PIV3), enterovirus 71 (EV71) and human rhinovirus (HRV). An apparent virus titer reduction was detected when MDCK cells were treated with clemastanin B after viral infection, particularly at the early stage, and the ribonucleoprotein (RNP) of the influenza virus was retained in the nucleus after treatment with clemastanin B. These results demonstrated that clemastanin B targets viral endocytosis, uncoating or RNP export from the nucleus. Furthermore, treatment with clemastanin B did not easily result in the emergence of viral drug resistance. The effects of clemastanin B demonstrated in this study may promote the antiviral study of IIR, but additional studies are required to define the anti-influenza mechanism(s).


Assuntos
Antivirais/farmacologia , Furanos/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Isatis/química , Extratos Vegetais/farmacologia , Adsorção , Análise de Variância , Animais , Antivirais/química , Linhagem Celular , Núcleo Celular , Cães , Farmacorresistência Viral/efeitos dos fármacos , Furanos/química , Humanos , Espaço Intracelular/virologia , Células Madin Darby de Rim Canino , Extratos Vegetais/química , Raízes de Plantas/química , Ensaio de Placa Viral
14.
Microbes Infect ; 15(1): 18-27, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23108316

RESUMO

Coxsackievirus B4 (CV-B4), in presence of antibodies and through a specific viral receptor CAR and Fcγ receptors II and III, can infect monocytes which results in interferon-α synthesis. The antibody-dependent enhancement of CV-B4 infection in the human monocytic-like THP-1 cell line has been investigated. The preincubation of CV-B4 with human plasma or human polyvalent immunoglobulins enhanced the infection of phorbol-myristate-acetate (PMA)-activated THP-1 cell cultures. CV-B4 replicated in these cells as demonstrated by the intracellular detection of infectious particles, viral protein VP1 (immunofluorescence), positive and negative viral RNA (RT-PCR). The viability of infected and control cell cultures was not different up to 20 days post-infection. Activated cell cultures inoculated with CV-B4 harbored intracellular RNA up to 14 days post-infection and produced IFNα that was detected by intracellular immunofluorescence staining as soon as 4 h post-infection with a maximum at 48 h post-infection and by RT-PCR all along the experiment. Together, these data demonstrate that PMA-activated THP-1 cells can be infected with CV-B4, can produce IFNα as a result of interactions between the virus, antibodies and specific receptors. This cellular model can be used to investigate further the mechanism and the result of the antibody-dependent enhancement of CV-B4 infection.


Assuntos
Enterovirus Humano B/imunologia , Imunoglobulinas/farmacologia , Monócitos/imunologia , Monócitos/virologia , Células Cultivadas , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/imunologia , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/virologia , Enterovirus Humano B/patogenicidade , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Interferon-alfa/biossíntese , Interferon-alfa/imunologia , Espaço Intracelular/metabolismo , Espaço Intracelular/virologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Receptores de IgG/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia
15.
PLoS One ; 7(11): e48721, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23173037

RESUMO

Cationic cell-penetrating peptides (CPPs) and their lipid domain-conjugates (CatLip) are agents for the delivery of (uncharged) biologically active molecules into the cell. Using infection and transfection assays we surprisingly discovered that CatLip peptides were able to inhibit replication of Duck Hepatitis B Virus (DHBV), a reference model for human HBV. Amongst twelve CatLip peptides we identified Deca-(Arg)8 having a particularly potent antiviral activity, leading to a drastic inhibition of viral particle secretion without detectable toxicity. Inhibition of virion secretion was correlated with a dose-dependent increase in intracellular viral DNA. Deca-(Arg)8 peptide did neither interfere with DHBV entry, nor with formation of mature nucleocapsids nor with their travelling to the nucleus. Instead, Deca-(Arg)8 caused envelope protein accumulation in large clusters as revealed by confocal laser scanning microscopy indicating severe structural changes of preS/S. Sucrose gradient analysis of supernatants from Deca-(Arg)8-treated cells showed unaffected naked viral nucleocapsids release, which was concomitant with a complete arrest of virion and surface protein-containing subviral particle secretion. This is the first report showing that a CPP is able to drastically block hepadnaviral release from infected cells by altering late stages of viral morphogenesis via interference with enveloped particle formation, without affecting naked nucleocapsid egress, thus giving a view inside the mode of inhibition. Deca-(Arg)8 may be a useful tool for elucidating the hepadnaviral secretory pathway, which is not yet fully understood. Moreover we provide the first evidence that a modified CPP displays a novel antiviral mechanism targeting another step of viral life cycle compared to what has been so far described for other enveloped viruses.


Assuntos
Peptídeos Penetradores de Células/metabolismo , Peptídeos Penetradores de Células/farmacologia , Vírus da Hepatite B do Pato/efeitos dos fármacos , Vírus da Hepatite B do Pato/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Capsídeo/efeitos dos fármacos , Capsídeo/metabolismo , Linhagem Celular , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Vírus da Hepatite B do Pato/genética , Vírus da Hepatite B do Pato/metabolismo , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/virologia , Metabolismo dos Lipídeos , Transporte Proteico , Fatores de Tempo , Proteínas Estruturais Virais/metabolismo
16.
PLoS One ; 7(9): e44965, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028707

RESUMO

The unique properties of the hepatitis C virus (HCV) JFH1 isolate have made it possible to produce and study HCV in an infectious cell culture system. However, relatively low virus titers restrict some of the uses of this system and preparing infectious chimeric reporter viruses have been difficult. In this study, we report cell culture-adapted mutations in wild-type JFH1 yielding higher titers of infectious particles of both JFH1 and chimeric JFH1 viruses carrying reporter genes. Sequencing analyses determined that ten of the sixteen nonsynonymous mutations were in the NS5A region. Individual viruses harboring specific adaptive mutations were prepared and studied. The mutations in the NS5A region, which included all three domains, were most effective in increasing infectious virus production. Insertion of two reporter genes in JFH1 without the adaptive mutations ablated the production of infectious HCV particles. However, the introduction of specific adaptive mutations in the NS5A region permitted reporter genes, Renilla luciferase (Rluc) and EGFP, to be introduced into JHF1 to produce chimeric HCV-NS5A-EGFP and HCV-NS5A-Rluc reporter viruses at relatively high titers of infectious virus. The quantity of hyperphosphorylated NS5A (p58) was decreased in the adapted JFH1 compared wild type JFH1 and is likely be involved in increased production of infectious virus based on previous studies of p58. The JFH1-derived mutant viruses and chimeric reporter viruses described here provide new tools for studying HCV biology, identifying HCV antivirals, and enable new ways of engineering additional infectious chimeric viruses.


Assuntos
Adaptação Fisiológica , Genes Reporter/genética , Engenharia Genética/métodos , Hepacivirus/fisiologia , Mutação , Carga Viral , Replicação Viral , Sequência de Bases , Linhagem Celular Tumoral , DNA Recombinante/genética , Proteínas de Fluorescência Verde/genética , Hepacivirus/genética , Hepacivirus/patogenicidade , Humanos , Espaço Intracelular/metabolismo , Espaço Intracelular/virologia , Luciferases de Renilla/genética , Fosforilação , Estrutura Terciária de Proteína , Transporte Proteico , Inoculações Seriadas , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
17.
Hum Gene Ther ; 23(10): 1090-100, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22779406

RESUMO

Adoptive immunotherapy with genetically modified natural killer (NK) cells is a promising approach for cancer treatment. Yet, optimization of highly efficient and clinically applicable gene transfer protocols for NK cells still presents a challenge. In this study, we aimed at identifying conditions under which optimum lentiviral gene transfer to NK cells can be achieved. Our results demonstrate that stimulation of NK cells with interleukin (IL)-2 and IL-21 supports efficient transduction using a VSV-G pseudotyped lentiviral vector. Moreover, we have identified that inhibition of innate immune receptor signaling greatly enhances transduction efficiency. We were able to boost the efficiency of lentiviral genetic modification on average 3.8-fold using BX795, an inhibitor of the TBK1/IKKɛ complex acting downstream of RIG-I, MDA-5, and TLR3. We have also observed that the use of BX795 enhances lentiviral transduction efficiency in a number of human and mouse cell lines, indicating a broadly applicable, practical, and safe approach that has the potential of being applicable to various gene therapy protocols.


Assuntos
Antivirais/farmacologia , Terapia Genética , Espaço Intracelular/virologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Lentivirus/efeitos dos fármacos , Transdução Genética , Animais , Linhagem Celular , Citocinas/farmacologia , Humanos , Imunidade Inata/efeitos dos fármacos , Espaço Intracelular/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Lentivirus/genética , Camundongos , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Fatores de Tempo
18.
PLoS One ; 7(5): e36571, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22693552

RESUMO

Chronic HIV infection leads to the development of cognitive impairments, designated as HIV-associated neurocognitive disorders (HAND). The secretion of soluble neurotoxic factors by HIV-infected macrophages plays a central role in the neuronal dysfunction and cell death associated with HAND. One potentially neurotoxic protein secreted by HIV-1 infected macrophages is cathepsin B. To explore the potential role of cathepsin B in neuronal cell death after HIV infection, we cultured HIV-1(ADA) infected human monocyte-derived macrophages (MDM) and assayed them for expression and activity of cathepsin B and its inhibitors, cystatins B and C. The neurotoxic activity of the secreted cathepsin B was determined by incubating cells from the neuronal cell line SK-N-SH with MDM conditioned media (MCM) from HIV-1 infected cultures. We found that HIV-1 infected MDM secreted significantly higher levels of cathepsin B than did uninfected cells. Moreover, the activity of secreted cathepsin B was significantly increased in HIV-infected MDM at the peak of viral production. Incubation of neuronal cells with supernatants from HIV-infected MDM resulted in a significant increase in the numbers of apoptotic neurons, and this increase was reversed by the addition of either the cathepsin B inhibitor CA-074 or a monoclonal antibody to cathepsin B. In situ proximity ligation assays indicated that the increased neurotoxic activity of the cathepsin B secreted by HIV-infected MDM resulted from decreased interactions between the enzyme and its inhibitors, cystatins B and C. Furthermore, preliminary in vivo studies of human post-mortem brain tissue suggested an upregulation of cathepsin B immunoreactivity in the hippocampus and basal ganglia in individuals with HAND. Our results demonstrate that HIV-1 infection upregulates cathepsin B in macrophages, increases cathepsin B activity, and reduces cystatin-cathepsin interactions, contributing to neuronal apoptosis. These findings provide new evidence for the role of cathepsin B in neuronal cell death induced by HIV-infected macrophages.


Assuntos
Apoptose , Catepsina B/metabolismo , HIV-1/fisiologia , Macrófagos/metabolismo , Neurônios/citologia , Neurônios/virologia , Gânglios da Base/metabolismo , Gânglios da Base/patologia , Gânglios da Base/fisiopatologia , Gânglios da Base/virologia , Catepsina B/antagonistas & inibidores , Catepsina B/genética , Linhagem Celular Tumoral , Cognição , Cistatina B/genética , Cistatina B/metabolismo , Cistatina C/genética , Cistatina C/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Hipocampo/virologia , Humanos , Espaço Intracelular/metabolismo , Espaço Intracelular/virologia , Lisossomos/metabolismo , Lisossomos/virologia , Macrófagos/virologia , Monócitos/citologia
19.
Plant Biotechnol J ; 10(4): 410-21, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22260326

RESUMO

Human papillomavirus 8 (HPV-8), one of the high-risk cutaneous papillomaviruses (cHPVs), is associated with epidermodysplasia verruciformis and nonmelanoma skin cancer in immuno-compromised individuals. Currently, no vaccines against cHPVs have been reported; however, recent studies on cross-neutralizing properties of their capsid proteins (CP) have fostered an interest in vaccine production against these viruses. We examined the potential of producing HPV-8 major CP L1 in Nicotiana benthamiana by agroinfiltration of different transient expression vectors: (i) the binary vector pBIN19 with or without silencing suppressor constructs, (ii) the nonreplicating Cowpea mosaic virus-derived expression vector pEAQ-HT and (iii) a replicating Tobacco mosaic virus (TMV)-based vector alone or with signal peptides. Although HPV-8 L1 was successfully expressed using pEAQ-HT and TMV, a 15-fold increase was obtained with pEAQ-HT. In contrast, no L1 protein could be immune detected using pBIN19 irrespective of whether silencing suppressors were coexpressed, although such constructs were required for identifying L1-specific transcripts. A fourfold yield increase in L1 expression was obtained when 22 C-terminal amino acids were deleted (L1ΔC22), possibly eliminating a nuclear localization signal. Electron microscopy showed that plant-made HPV-8 L1 proteins assembled in appropriate virus-like particles (VLPs) of T = 1 or T = 7 symmetry. Ultrathin sections of L1ΔC22-expressing cells revealed their accumulation in the cytoplasm in the form of VLPs or paracrystalline arrays. These results show for the first time the production and localization of HPV-8 L1 protein in planta and its assembly into VLPs representing promising candidate for potential vaccine production.


Assuntos
Proteínas do Capsídeo/biossíntese , Proteínas do Capsídeo/isolamento & purificação , Expressão Gênica , Técnicas Genéticas , Nicotiana/metabolismo , Proteínas Oncogênicas Virais/biossíntese , Proteínas Oncogênicas Virais/isolamento & purificação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Northern Blotting , Proteínas do Capsídeo/ultraestrutura , DNA Bacteriano/genética , Vetores Genéticos/genética , Humanos , Immunoblotting , Espaço Intracelular/metabolismo , Espaço Intracelular/virologia , Proteínas Mutantes/metabolismo , Proteínas Oncogênicas Virais/ultraestrutura , Exsudatos de Plantas/metabolismo , Proteínas Recombinantes/ultraestrutura , Frações Subcelulares/virologia , Vírion/metabolismo , Vírion/ultraestrutura
20.
Virology ; 415(2): 69-82, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21524776

RESUMO

Severe acute respiratory syndrome (SARS) coronavirus (CoV) envelope (E) protein is a transmembrane protein. Several subcellular locations and topological conformations of E protein have been proposed. To identify the correct ones, polyclonal and monoclonal antibodies specific for the amino or the carboxy terminus of E protein, respectively, were generated. E protein was mainly found in the endoplasmic reticulum-Golgi intermediate compartment (ERGIC) of cells transfected with a plasmid encoding E protein or infected with SARS-CoV. No evidence of E protein presence in the plasma membrane was found by using immunofluorescence, immunoelectron microscopy and cell surface protein labeling. In addition, measurement of plasma membrane voltage gated ion channel activity by whole-cell patch clamp suggested that E protein was not present in the plasma membrane. A topological conformation in which SARS-CoV E protein amino terminus is oriented towards the lumen of intracellular membranes and carboxy terminus faces cell cytoplasm is proposed.


Assuntos
Espaço Intracelular/virologia , Síndrome Respiratória Aguda Grave/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Chlorocebus aethiops , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Complexo de Golgi/metabolismo , Complexo de Golgi/virologia , Humanos , Espaço Intracelular/metabolismo , Dados de Sequência Molecular , Transporte Proteico , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Síndrome Respiratória Aguda Grave/metabolismo , Células Vero , Proteínas do Envelope Viral/genética , Proteínas Viroporinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA