Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Microbiol Spectr ; 12(8): e0365123, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-38916347

RESUMO

The rapidly developing field of oncolytic virus (OV) therapy necessitates the development of new and improved analytical approaches for the characterization of the virus during production and development. Accurate monitoring and absolute quantification of viral proteins are crucial for OV product characterization and can facilitate the understanding of infection, immunogenicity, and development stages of viral replication. Targeted mass spectrometry methods like multiple reaction monitoring (MRM) offer a robust way to directly detect and quantify specific targeted proteins represented by surrogate peptides. We have leveraged the power of MRM by combining ultra-high performance liquid chromatography (UPLC) with a Sciex 6500 triple-stage quadrupole mass spectrometer to develop an assay that accurately and absolutely quantifies the structural proteins of a pseudotyped vesicular stomatitis virus (VSV) intended for use as a new biotherapeutic (designated hereafter as VSV-GP to differentiate it from native VSV). The new UPLC-MRM method provides absolute quantification with the use of heavy-labeled reference standard surrogate peptides. When added in known exact amounts to standards and samples, the reference standards normalize and account for any small perturbations during sample preparation and/or instrument performance, resulting in accurate and precise quantification. Because of the multiplexed nature of MRM, all targeted proteins are quantified at the same time. The optimized assay has been enhanced to quantify the ratios of the processed GP1 and GP2 proteins while simultaneously measuring any remaining or unprocessed form of the envelope protein GP complex (GPC; full-length GPC). IMPORTANCE: The development of oncolytic viral therapy has gained considerable momentum in recent years. Vesicular stomatitis virus glycoprotein (VSV-GP) is a new biotherapeutic emerging in the oncolytic viral therapy platform. Novel analytical assays that can accurately and precisely quantify the viral proteins are a necessity for the successful development of viral vector as a biotherapeutic. We developed an ultra-high performance liquid chromatography multiple reaction monitoring-based assay to quantify the absolute concentrations of the different structural proteins of VSV-GP. The complete processing of GP complex (GPC) is a prerequisite for the infectivity of the virus. The assay extends the potential for quantifying full-length GPC, which provides an understanding of the processing of GPC (along with the quantification of GP1 and GP2 separately). We used this assay in tracking GPC processing in HEK-293-F production cell lines infected with VSV-GP.


Assuntos
Vesiculovirus , Proteínas Virais , Humanos , Cromatografia Líquida de Alta Pressão/métodos , Espectrometria de Massas/métodos , Vírus Oncolíticos/genética , Vírus da Estomatite Vesicular Indiana/genética , Vesiculovirus/genética , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
2.
Mol Ther Methods Clin Dev ; 28: 190-207, 2023 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-36700123

RESUMO

Viral replication places oncolytic viruses (OVs) in a unique niche in the field of drug pharmacokinetics (PK) as their self-amplification obscures exposure-response relationships. Moreover, standard bioanalytical techniques are unable to distinguish the input from replicated drug products. Here, we combine two novel approaches to characterize PK and biodistribution (BD) after systemic administration of vesicular stomatitis virus pseudotyped with lymphocytic choriomeningitis virus glycoprotein (VSV-GP) in healthy mice. First: to decouple input drug PK/BD versus replication PK/BD, we developed and fully characterized a replication-incompetent tool virus that retained all other critical attributes of the drug. We used this approach to quantify replication in blood and tissues and to determine its impact on PK and BD. Second: to discriminate the genomic and antigenomic viral RNA strands contributing to replication dynamics in tissues, we developed an in situ hybridization method using strand-specific probes and assessed their spatiotemporal distribution in tissues. This latter approach demonstrated that distribution, transcription, and replication localized to tissue-resident macrophages, indicating their role in PK and BD. Ultimately, our study results in a refined PK/BD profile for a replicating OV, new proposed PK parameters, and deeper understanding of OV PK/BD using unique approaches that could be applied to other replicating vectors.

3.
J Virol ; 93(1)2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30305352

RESUMO

The incoming influenza A virus (IAV) genome must pass through two distinct barriers in order to establish infection in the cell: the plasma membrane and the nuclear membrane. A precise understanding of the challenges imposed by the nuclear barrier remains outstanding. Passage across is mediated by host karyopherins (KPNAs), which bind to the viral nucleoprotein (NP) via its N-terminal nuclear localization sequence (NLS). The binding affinity between the two molecules is low, but NP is present in a high copy number, which suggests that binding avidity plays a compensatory role during import. Using nanobody-based technology, we demonstrate that a high binding avidity is required for infection, though the absolute value differs between cell types and correlates with their relative susceptibility to infection. In addition, we demonstrate that increasing the affinity level caused a decrease in avidity requirements for some cell types but blocked infection in others. Finally, we show that genomes that become frustrated by low avidity and remain cytoplasmic trigger the type I interferon response. Based on these results, we conclude that IAV balances affinity and avidity considerations in order to overcome the nuclear barrier across a broad range of cell types. Furthermore, these results provide evidence to support the long-standing hypothesis that IAV's strategy of import and replication in the nucleus facilitates immune evasion.IMPORTANCE We used intracellular nanobodies to block influenza virus infection at the step prior to nuclear import of its ribonucleoproteins. By doing so, we were able to answer an important but outstanding question that could not be addressed with conventional tools: how many of the ∼500 available NLS motifs are needed to establish infection? Furthermore, by controlling the subcellular localization of the incoming viral ribonucleoproteins and measuring the cell's antiviral response, we were able to provide direct evidence for the long-standing hypothesis that influenza virus exploits nuclear localization to delay activation of the innate immune response.


Assuntos
Vírus da Influenza A/patogenicidade , Interferon Tipo I/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Anticorpos de Domínio Único/metabolismo , Proteínas do Core Viral/química , Proteínas do Core Viral/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Cricetinae , Cães , Células HEK293 , Células HeLa , Humanos , Evasão da Resposta Imune , Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , Influenza Humana , Carioferinas , Células Madin Darby de Rim Canino , Proteínas do Nucleocapsídeo , Infecções por Orthomyxoviridae
4.
Cell Host Microbe ; 23(6): 819-831.e5, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29902443

RESUMO

Ubiquitylation is one of the most versatile protein post-translational modifications and is frequently altered during virus infections. Here we employed a functional proteomics screen to identify host proteins that are differentially ubiquitylated upon dengue virus (DENV) infection. Among the several differentially modified proteins identified in infected cells was AUP1, a lipid droplet-localized type-III membrane protein, which exists predominantly in the mono-ubiquitylated form. AUP1 associated with DENV NS4A and relocalized from lipid droplets to autophagosomes upon infection. Virus production was abolished in cells deleted for AUP1 or expressing an AUP1 acyltransferase domain mutant. Ubiquitylation disrupted the AUP1-NS4A interaction, resulting in inhibited acyltransferase activity, defective lipophagy, and attenuated virus production. Our results show that DENV-NS4A exploits the acyltransferase activity of AUP1 to trigger lipophagy, a process regulated by ubiquitylation. This mechanism appears to be a general phenomenon in biogenesis of flaviviruses and underscores the critical role of post-translational modifications in virus infections.


Assuntos
Autofagia/fisiologia , Proteínas de Transporte/metabolismo , Flavivirus/metabolismo , Flavivirus/patogenicidade , Domínios e Motivos de Interação entre Proteínas/fisiologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Células A549 , Aciltransferases/metabolismo , Autofagossomos/virologia , Proteínas de Transporte/genética , Dengue/imunologia , Dengue/metabolismo , Vírus da Dengue/patogenicidade , Técnicas de Inativação de Genes , Células HeLa , Células Hep G2 , Humanos , Gotículas Lipídicas , Proteínas de Membrana/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Proteico , Proteômica , Ubiquitinação
5.
J Virol ; 89(5): 2792-800, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25540369

RESUMO

UNLABELLED: Perturbation of protein-protein interactions relies mostly on genetic approaches or on chemical inhibition. Small RNA viruses, such as influenza A virus, do not easily lend themselves to the former approach, while chemical inhibition requires that the target protein be druggable. A lack of tools thus constrains the functional analysis of influenza virus-encoded proteins. We generated a panel of camelid-derived single-domain antibody fragments (VHHs) against influenza virus nucleoprotein (NP), a viral protein essential for nuclear trafficking and packaging of the influenza virus genome. We show that these VHHs can target NP in living cells and perturb NP's function during infection. Cytosolic expression of NP-specific VHHs (αNP-VHHs) disrupts virus replication at an early stage of the life cycle. Based on their specificity, these VHHs fall into two distinct groups. Both prevent nuclear import of the viral ribonucleoprotein (vRNP) complex without disrupting nuclear import of NP alone. Different stages of the virus life cycle thus rely on distinct nuclear localization motifs of NP. Their molecular characterization may afford new means of intervention in the virus life cycle. IMPORTANCE: Many proteins encoded by RNA viruses are refractory to manipulation due to their essential role in replication. Thus, studying their function and determining how to disrupt said function through pharmaceutical intervention are difficult. We present a novel method based on single-domain-antibody technology that permits specific targeting and disruption of an essential influenza virus protein in the absence of genetic manipulation of influenza virus itself. Characterization of such interactions may help identify new targets for pharmaceutical intervention. This approach can be extended to study proteins encoded by other viral pathogens.


Assuntos
Núcleo Celular/química , Núcleo Celular/virologia , Vírus da Influenza A/fisiologia , Proteínas de Ligação a RNA/análise , Anticorpos de Domínio Único/metabolismo , Proteínas do Core Viral/análise , Virologia/métodos , Replicação Viral , Animais , Linhagem Celular , Cães , Expressão Gênica , Biologia Molecular/métodos , Proteínas do Nucleocapsídeo , Anticorpos de Domínio Único/genética
6.
J Biol Chem ; 288(12): 8061-8073, 2013 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-23362274

RESUMO

Influenza is a severe disease in humans and animals with few effective therapies available. All strains of influenza virus are prone to developing drug resistance due to the high mutation rate in the viral genome. A therapeutic agent that targets a highly conserved region of the virus could bypass resistance and also be effective against multiple strains of influenza. Influenza uses many individually weak ligand binding interactions for a high avidity multivalent attachment to sialic acid-bearing cells. Polymerized sialic acid analogs can form multivalent interactions with influenza but are not ideal therapeutics due to solubility and toxicity issues. We used liposomes as a novel means for delivery of the glycan sialylneolacto-N-tetraose c (LSTc). LSTc-bearing decoy liposomes form multivalent, polymer-like interactions with influenza virus. Decoy liposomes competitively bind influenza virus in hemagglutination inhibition assays and inhibit infection of target cells in a dose-dependent manner. Inhibition is specific for influenza virus, as inhibition of Sendai virus and respiratory syncytial virus is not observed. In contrast, monovalent LSTc does not bind influenza virus or inhibit infectivity. LSTc decoy liposomes prevent the spread of influenza virus during multiple rounds of replication in vitro and extend survival of mice challenged with a lethal dose of virus. LSTc decoy liposomes co-localize with fluorescently tagged influenza virus, whereas control liposomes do not. Considering the conservation of the hemagglutinin binding pocket and the ability of decoy liposomes to form high avidity interactions with influenza hemagglutinin, our decoy liposomes have potential as a new therapeutic agent against emerging influenza strains.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Polissacarídeos/farmacologia , Ácidos Siálicos/farmacologia , Animais , Antivirais/administração & dosagem , Linhagem Celular , Chlorocebus aethiops , Cães , Avaliação Pré-Clínica de Medicamentos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Feminino , Hemaglutinação/efeitos dos fármacos , Humanos , Vírus da Influenza A/fisiologia , Lipossomos , Camundongos , Camundongos Endogâmicos C57BL , Polissacarídeos/administração & dosagem , Vírus do Sarcoma de Rous/efeitos dos fármacos , Vírus Sendai/efeitos dos fármacos , Ácidos Siálicos/administração & dosagem , Células Vero , Replicação Viral/efeitos dos fármacos
7.
J Virol ; 83(11): 5408-18, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19279106

RESUMO

The mammalian interferon (IFN) signaling pathway is a primary component of the innate antiviral response. As such, viral pathogens have devised multiple mechanisms to antagonize this pathway and thus facilitate infection. Dengue virus (DENV) encodes several proteins (NS2a, NS4a, and NS4b) that have been shown individually to inhibit the IFN response. In addition, DENV infection results in reduced levels of expression of STAT2, which is required for IFN signaling (M. Jones, A. Davidson, L. Hibbert, P. Gruenwald, J. Schlaak, S. Ball, G. R. Foster, and M. Jacobs, J. Virol. 79:5414-5420, 2005). Translation of the DENV genome results in a single polypeptide, which is processed by viral and host proteases into at least 10 separate proteins. To date, no single DENV protein has been implicated in the targeting of STAT2 for decreased levels of expression. We demonstrate here that the polymerase of the virus, NS5, binds to STAT2 and is necessary and sufficient for its reduced level of expression. The decrease in protein level observed requires ubiquitination and proteasome activity, strongly suggesting an active degradation process. Furthermore, we show that the degradation of but not binding to STAT2 is dependent on the expression of the polymerase in the context of a polyprotein that undergoes proteolytic processing for NS5 maturation. Thus, the mature form of NS5, when not expressed as a precursor, was able to bind to STAT2 but was unable to target it for degradation, establishing a unique role for viral polyprotein processing in providing an additional function to a viral polypeptide. Therefore, we have identified both a novel mechanism by which DENV evades the innate immune response and a potential target for antiviral therapeutics.


Assuntos
Vírus da Dengue/metabolismo , Fator de Transcrição STAT2/metabolismo , Proteínas não Estruturais Virais/metabolismo , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Vírus da Dengue/genética , Regulação da Expressão Gênica , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Fator de Transcrição STAT2/genética , Ubiquitina/metabolismo , Proteínas não Estruturais Virais/genética
8.
J Exp Med ; 205(8): 1929-38, 2008 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-18663127

RESUMO

Tumor suppressor p53 is activated by several stimuli, including DNA damage and oncogenic stress. Previous studies (Takaoka, A., S. Hayakawa, H. Yanai, D. Stoiber, H. Negishi, H. Kikuchi, S. Sasaki, K. Imai, T. Shibue, K. Honda, and T. Taniguchi. 2003. Nature. 424:516-523) have shown that p53 is also induced in response to viral infections as a downstream transcriptional target of type I interferon (IFN) signaling. Moreover, many viruses, including SV40, human papillomavirus, Kaposi's sarcoma herpesvirus, adenoviruses, and even RNA viruses such as polioviruses, have evolved mechanisms designated to abrogate p53 responses. We describe a novel p53 function in the activation of the IFN pathway. We observed that infected mouse and human cells with functional p53 exhibited markedly decreased viral replication early after infection. This early inhibition of viral replication was mediated both in vitro and in vivo by a p53-dependent enhancement of IFN signaling, specifically the induction of genes containing IFN-stimulated response elements. Of note, p53 also contributed to an increase in IFN release from infected cells. We established that this p53-dependent enhancement of IFN signaling is dependent to a great extent on the ability of p53 to activate the transcription of IFN regulatory factor 9, a central component of the IFN-stimulated gene factor 3 complex. Our results demonstrate that p53 contributes to innate immunity by enhancing IFN-dependent antiviral activity independent of its functions as a proapoptotic and tumor suppressor gene.


Assuntos
Genes p53 , Imunidade Inata/genética , Interferons/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , Apoptose , Linhagem Celular Tumoral , Células Cultivadas , Marcação de Genes , Proteínas de Fluorescência Verde/genética , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Camundongos , Camundongos Knockout , Transdução de Sinais , Transcrição Gênica , Vírus da Estomatite Vesicular Indiana/patogenicidade , Vírus da Estomatite Vesicular Indiana/fisiologia , Replicação Viral/genética , Replicação Viral/imunologia
9.
J Virol ; 79(13): 8004-13, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15956546

RESUMO

Flaviviruses are insect-borne, positive-strand RNA viruses that have been disseminated worldwide. Their genome is translated into a polyprotein, which is subsequently cleaved by a combination of viral and host proteases to produce three structural proteins and seven nonstructural proteins. The nonstructural protein NS4B of dengue 2 virus partially blocks activation of STAT1 and interferon-stimulated response element (ISRE) promoters in cells stimulated with interferon (IFN). We have found that this function of NS4B is conserved in West Nile and yellow fever viruses. Deletion analysis shows that that the first 125 amino acids of dengue virus NS4B are sufficient for inhibition of alpha/beta IFN (IFN-alpha/beta) signaling. The cleavable signal peptide at the N terminus of NS4B, a peptide with a molecular weight of 2,000, is required for IFN antagonism but can be replaced by an unrelated signal peptide. Coexpression of dengue virus NS4A and NS4B together results in enhanced inhibition of ISRE promoter activation in response to IFN-alpha/beta. In contrast, expression of the precursor NS4A/B fusion protein does not cause an inhibition of IFN signaling unless this product is cleaved by the viral peptidase NS2B/NS3, indicating that proper viral polyprotein processing is required for anti-interferon function.


Assuntos
Flavivirus/fisiologia , Interferon-alfa/fisiologia , Interferon beta/fisiologia , Sinais Direcionadores de Proteínas/fisiologia , Proteínas não Estruturais Virais/farmacologia , Vírus do Nilo Ocidental/fisiologia , Animais , Sequência de Bases , Clonagem Molecular , Primers do DNA , Interferon-alfa/efeitos dos fármacos , Interferon beta/efeitos dos fármacos , Mamíferos , Reação em Cadeia da Polimerase , Sinais Direcionadores de Proteínas/efeitos dos fármacos , Mapeamento por Restrição , Proteínas não Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA