Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Virol ; 92(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29321321

RESUMO

In the last few decades, dengue virus, an arbovirus, has spread to over 120 countries. Although a vaccine has been approved in some countries, limitations on its effectiveness and a lack of effective antiviral treatments reinforce the need for additional research. The functions of several viral nonstructural proteins are essentially unknown. To better understand the functions of these proteins and thus dengue virus pathogenesis, we embarked on a genomewide transposon mutagenesis screen with next-generation sequencing to determine sites in the viral genome that tolerate 15-nucleotide insertions. Using this approach, we generated support for several published predicted transmembrane and enzymatic domains. Next, we created 7 mutants containing the 15-nucleotide insertion from the original selection and found 6 of them were capable of replication in both mammalian and mosquito tissue culture cells. Interestingly, one mutation had a significant impairment of viral assembly, and this mutation may lead to a better understanding of viral assembly and release. In addition, we created a fully infectious virus expressing a functionally tagged NS4B protein, which will provide a much-needed tool to elucidate the role of NS4B in viral pathogenesis.IMPORTANCE Dengue virus is a mosquito-borne virus distributed in tropical and subtropical regions globally that can result in hospitalization and even death in some cases. Although a vaccine exists, its limitations and a lack of approved antiviral treatments highlight our limited understanding of dengue virus pathogenesis and host immunity. The functions of many viral proteins are poorly understood. We used a previously published approach using transposon mutagenesis to develop tools to study these proteins' functions by adding insertions randomly throughout the viral genomes. These genomes were transferred into cells, and infectious progeny were recovered to determine sites that tolerated insertions, as only the genomes that tolerated insertions would be able to propagate. Using these results, we created viruses with epitope tags, one in the viral structural protein Capsid and one in the viral nonstructural protein NS4B. Further investigation of these mutants may elucidate the roles of Capsid and NS4B during dengue virus infections.


Assuntos
Vírus da Dengue/genética , Genoma Viral , Sequenciamento de Nucleotídeos em Larga Escala , Mutagênese Insercional , Proteínas não Estruturais Virais/genética , Animais , Chlorocebus aethiops , Humanos , Células Vero
2.
Gastroenterology ; 146(5): 1373-85.e1-11, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24512803

RESUMO

BACKGROUND & AIMS: Positive-sense RNA viruses remodel intracellular membranes to generate specialized membrane compartments for viral replication. Several RNA viruses, including poliovirus and hepatitis C virus (HCV), require phosphatidylinositol (PI) 4-kinases for their replication. However, it is not known how PI 4-kinases and their product, PI(4)P, facilitate host membrane reorganization and viral replication. In addition, although the HCV replication compartment, known as the membranous web, is believed to be cholesterol enriched, the mechanisms by which this occurs have not been elucidated. We aimed to identify and characterize a PI 4-kinase effector in HCV replication. METHODS: We used a combination of microscopic and biochemical methods to study HCV replication, web morphology, the distribution of intracellular protein and PI(4)P, along with cholesterol trafficking in HCV-infected cells. PI 4-kinase and oxysterol-binding protein (OSBP) were inhibited using RNA interference or small molecules in cells expressing a full-length genotype 1b replicon or infected with the JFH-1 strain of HCV. RESULTS: OSBP was required for HCV replication and membranous web integrity. OSBP was recruited to membranous webs in a PI 4-kinase-dependent manner, and both these factors were found to regulate cholesterol trafficking to the web. We also found OSBP to be required for poliovirus infection but dispensable for dengue virus. CONCLUSIONS: OSBP is a PI 4-kinase effector in HCV infection, and contributes to the integrity and cholesterol enrichment of the membranous web. OSBP might also be a PI 4-kinase effector in poliovirus infection and could be involved in replication of other viruses that require PI 4-kinases.


Assuntos
1-Fosfatidilinositol 4-Quinase/metabolismo , Membrana Celular/enzimologia , Colesterol/metabolismo , Hepacivirus/enzimologia , Receptores de Esteroides/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , 1-Fosfatidilinositol 4-Quinase/antagonistas & inibidores , Transporte Biológico , Membrana Celular/efeitos dos fármacos , Genótipo , Células HEK293 , Células HeLa , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Fenótipo , Fosfatos de Fosfatidilinositol/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , RNA Viral/biossíntese , Receptores de Esteroides/genética , Transfecção , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/genética , Replicação Viral/efeitos dos fármacos
3.
PLoS Pathog ; 8(7): e1002783, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22792064

RESUMO

Ubiquitin (Ub) is a vital regulatory component in various cellular processes, including cellular responses to viral infection. As obligate intracellular pathogens, viruses have the capacity to manipulate the ubiquitin (Ub) cycle to their advantage by encoding Ub-modifying proteins including deubiquitinases (DUBs). However, how cellular DUBs modulate specific viral infections, such as norovirus, is poorly understood. To examine the role of DUBs during norovirus infection, we used WP1130, a small molecule inhibitor of a subset of cellular DUBs. Replication of murine norovirus in murine macrophages and the human norovirus Norwalk virus in a replicon system were significantly inhibited by WP1130. Chemical proteomics identified the cellular DUB USP14 as a target of WP1130 in murine macrophages, and pharmacologic inhibition or siRNA-mediated knockdown of USP14 inhibited murine norovirus infection. USP14 is a proteasome-associated DUB that also binds to inositol-requiring enzyme 1 (IRE1), a critical mediator of the unfolded protein response (UPR). WP1130 treatment of murine macrophages did not alter proteasome activity but activated the X-box binding protein-1 (XBP-1) through an IRE1-dependent mechanism. In addition, WP1130 treatment or induction of the UPR also reduced infection of other RNA viruses including encephalomyocarditis virus, Sindbis virus, and La Crosse virus but not vesicular stomatitis virus. Pharmacologic inhibition of the IRE1 endonuclease activity partially rescued the antiviral effect of WP1130. Taken together, our studies support a model whereby induction of the UPR through cellular DUB inhibition blocks specific viral infections, and suggest that cellular DUBs and the UPR represent novel targets for future development of broad spectrum antiviral therapies.


Assuntos
Antivirais/farmacologia , Nitrilas/farmacologia , Norovirus/efeitos dos fármacos , Piridinas/farmacologia , Ubiquitina Tiolesterase/antagonistas & inibidores , Ubiquitina Tiolesterase/metabolismo , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Animais , Infecções por Caliciviridae/tratamento farmacológico , Infecções por Caliciviridae/virologia , Linhagem Celular , Linhagem Celular Tumoral , Cianoacrilatos , Proteínas de Ligação a DNA/metabolismo , Vírus da Encefalomiocardite/efeitos dos fármacos , Vírus da Encefalomiocardite/patogenicidade , Inibidores Enzimáticos/farmacologia , Humanos , Vírus La Crosse/efeitos dos fármacos , Vírus La Crosse/patogenicidade , Macrófagos/virologia , Proteínas de Membrana/metabolismo , Camundongos , Norovirus/fisiologia , Vírus Norwalk/efeitos dos fármacos , Vírus Norwalk/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Fatores de Transcrição de Fator Regulador X , Sindbis virus/efeitos dos fármacos , Sindbis virus/patogenicidade , Fatores de Transcrição/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterase/genética , Replicação Viral/efeitos dos fármacos , Proteína 1 de Ligação a X-Box
4.
J Virol ; 86(10): 5584-93, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22438544

RESUMO

Human norovirus infections are the most common cause of acute nonbacterial gastroenteritis in humans worldwide, and glycan binding plays an important role in the susceptibility to these infections. However, due to the lack of an efficient cell culture system or small animal model for human noroviruses, little is known about the biological role of glycan binding during infection. Murine noroviruses (MNV) are also enteric viruses that bind to cell surface glycans, but in contrast to their human counterparts, they can be grown in tissue culture and a small animal host. In this study, we determined glycan-binding specificities of the MNV strains MNV-1 and CR3 in vitro, identified molecular determinants of glycan binding, and analyzed infection in vivo. We showed that unlike MNV-1, CR3 binding to murine macrophages was resistant to neuraminidase treatment and glycosphingolipid depletion. Both strains depended on N-linked glycoproteins for binding, while only MNV-1 attachment to macrophages was sensitive to O-linked glycoprotein depletion. In vivo, CR3 showed differences in tissue tropism compared to MNV-1 by replicating in the large intestine. Mapping of a glycan-binding site in the MNV-1 capsid by reverse genetics identified a region topologically similar to the histo-blood group antigen (HBGA)-binding sites of the human norovirus strain VA387. The recombinant virus showed distinct changes in tissue tropism compared to wild-type virus. Taken together, our data demonstrate that MNV strains evolved multiple strategies to bind different glycan receptors on the surface of murine macrophages and that glycan binding contributes to tissue tropism in vivo.


Assuntos
Infecções por Caliciviridae/metabolismo , Glicolipídeos/metabolismo , Glicoproteínas/metabolismo , Norovirus/fisiologia , Receptores Virais/metabolismo , Animais , Infecções por Caliciviridae/virologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Norovirus/genética , Polissacarídeos/metabolismo , Ligação Proteica , Especificidade da Espécie , Ligação Viral
5.
Mol Microbiol ; 81(1): 113-28, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21542860

RESUMO

ToxR of Vibrio cholerae directly activates the ompU promoter, but requires a second activator, TcpP to activate the toxT promoter. ompU encodes a porin, while toxT encodes the transcription factor, ToxT, which activates V. cholerae virulence genes including cholera toxin and the toxin co-regulated pilus. Using an ompU-sacB transcriptional fusion, toxR mutant alleles were identified that encode ToxR molecules defective for ompU promoter activation. Many toxR mutants defective for ompU activation affected residues involved in DNA binding. Mutants defective for ompU activation were also tested for activation of the toxT promoter. ToxR-F69A and ToxR-V71A, both in the α-loop of ToxR, were preferentially defective for ompU activation, with ToxR-V71A nearly completely defective. Six mutants from the ompU-sacB selection showed more dramatic defects in toxT activation than ompU activation. All but one of the affected residues map to the wing domain of the winged helix-turn-helix of ToxR. Some ToxR mutants preferentially affecting toxT activation had partial DNA-binding defects, and one mutant, ToxR-P101L, had altered interactions with TcpP. These data suggest that while certain residues in the α-loop of ToxR are utilized to activate the ompU promoter, the wing domain of ToxR contributes to both promoter binding and ToxR/TcpP interaction facilitating toxT activation.


Assuntos
Adesinas Bacterianas/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica , Fatores de Transcrição/metabolismo , Vibrio cholerae/fisiologia , Adesinas Bacterianas/genética , Proteínas de Bactérias/genética , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Sequências Hélice-Volta-Hélice , Modelos Biológicos , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Conformação Proteica , Fatores de Transcrição/genética , Vibrio cholerae/genética
6.
Infect Immun ; 79(12): 4850-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21911458

RESUMO

Macrophages are key mediators of antimicrobial defense and innate immunity. Innate intracellular defense mechanisms can be rapidly regulated at the posttranslational level by the coordinated addition and removal of ubiquitin by ubiquitin ligases and deubiquitinases (DUBs). While ubiquitin ligases have been extensively studied, the contribution of DUBs to macrophage innate immune function is incompletely defined. We therefore employed a small molecule DUB inhibitor, WP1130, to probe the role of DUBs in the macrophage response to bacterial infection. Treatment of activated bone marrow-derived macrophages (BMM) with WP1130 significantly augmented killing of the intracellular bacterial pathogen Listeria monocytogenes. WP1130 also induced killing of phagosome-restricted bacteria, implicating a bactericidal mechanism associated with the phagosome, such as the inducible nitric oxide synthase (iNOS). WP1130 had a minimal antimicrobial effect in macrophages lacking iNOS, indicating that iNOS is an effector mechanism for WP1130-mediated bacterial killing. Although overall iNOS levels were not notably different, we found that WP1130 significantly increased colocalization of iNOS with the Listeria-containing phagosome during infection. Taken together, our data indicate that the deubiquitinase inhibitor WP1130 increases bacterial killing in macrophages by enhancing iNOS localization to the phagosome and suggest a potential role for ubiquitin regulation in iNOS trafficking.


Assuntos
Macrófagos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Nitrilas/farmacologia , Fagossomos/enzimologia , Inibidores de Proteases/farmacologia , Piridinas/farmacologia , Animais , Linhagem Celular , Cianoacrilatos , Endopeptidases/metabolismo , Escherichia coli/fisiologia , Listeria monocytogenes/fisiologia , Macrófagos/enzimologia , Camundongos , Óxido Nítrico Sintase Tipo II/genética , Transporte Proteico/efeitos dos fármacos , Ubiquitina/metabolismo
7.
J Virol ; 84(12): 6163-76, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20375172

RESUMO

Although noroviruses cause the vast majority of nonbacterial gastroenteritis in humans, little is known about their life cycle, including viral entry. Murine norovirus (MNV) is the only norovirus to date that efficiently infects cells in culture. To elucidate the productive route of infection for MNV-1 into murine macrophages, we used a neutral red (NR) infectious center assay and pharmacological inhibitors in combination with dominant-negative (DN) and small interfering RNA (siRNA) constructs to show that clathrin- and caveolin-mediated endocytosis did not play a role in entry. In addition, we showed that phagocytosis or macropinocytosis, flotillin-1, and GRAF1 are not required for the major route of MNV-1 uptake. However, MNV-1 genome release occurred within 1 h, and endocytosis was significantly inhibited by the cholesterol-sequestering drugs nystatin and methyl-beta-cyclodextrin, the dynamin-specific inhibitor dynasore, and the dominant-negative dynamin II mutant K44A. Therefore, we conclude that the productive route of MNV-1 entry into murine macrophages is rapid and requires host cholesterol and dynamin II.


Assuntos
Infecções por Caliciviridae/metabolismo , Colesterol/metabolismo , Dinamina II/metabolismo , Endocitose , Macrófagos/virologia , Norovirus/fisiologia , Animais , Infecções por Caliciviridae/virologia , Linhagem Celular Tumoral , Macrófagos/metabolismo , Camundongos
8.
J Virol ; 83(9): 4092-101, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19244326

RESUMO

Noroviruses are the major cause of nonbacterial gastroenteritis in humans. However, little is known regarding the norovirus life cycle, including cell binding and entry. In contrast to human noroviruses, the recently discovered murine norovirus 1 (MNV-1) readily infects murine macrophages and dendritic cells in culture. Many viruses, including the related feline calicivirus, use terminal sialic acids (SA) as receptors for infection. Therefore, we tested whether SA moieties play a role during MNV-1 infection of murine macrophages. Competition with SA-binding lectins and neuraminidase treatment led to a reduction in MNV-1 binding and infection in cultured and primary murine macrophages, suggesting a role for SA during the initial steps of the MNV-1 life cycle. Because SA moieties can be attached to glycolipids (i.e., gangliosides), we next determined whether MNV-1 uses gangliosides during infection. The gangliosides GD1a, GM1, and asialo-GM1 (GA1) are natural components of murine macrophages. MNV-1 bound to ganglioside GD1a, which is characterized by an SA on the terminal galactose, but not to GM1 or asialo-GM1 in an enzyme-linked immunosorbent assay. The depletion of gangliosides using an inhibitor of glycosylceramide synthase (d-threo-P4) led to a reduction of MNV-1 binding and infection in cultured and primary murine macrophages. This defect was specifically rescued by the addition of GD1a. A similar phenotype was observed for MNV field strains WU11 (GV/WU11/2005/USA) and S99 (GV/Berlin/2006/DE). In conclusion, our data indicate that MNV can use terminal SA on gangliosides as attachment receptors during binding to murine macrophages.


Assuntos
Gangliosídeos/metabolismo , Macrófagos/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Norovirus/metabolismo , Receptores Virais/metabolismo , Internalização do Vírus , Animais , Anticorpos/imunologia , Linhagem Celular , Endotoxinas/metabolismo , Lectinas/metabolismo , Camundongos , Neuraminidase/metabolismo , Norovirus/classificação , Norovirus/genética , Especificidade por Substrato
9.
Virus Res ; 143(1): 125-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19463729

RESUMO

Murine norovirus (MNV) is a recently discovered mouse pathogen. Unlike the fastidious human noroviruses that cause the overwhelming majority of non-bacterial gastroenteritis worldwide, MNV readily infects cells in culture. Its replication in primary murine macrophages and dendritic cells and their derived cell lines allows the study of norovirus cell entry for the first time. In this study we determined the role of pH during MNV-1 infection since the low pH environment of endosomes often triggers uncoating of viruses. We demonstrated that MNV-1 viral titers by plaque assay and expression of the non-structural protein VPg by immunofluorescence were not affected by pH in cultured and primary macrophages and dendritic cells in the presence of two known endosome acidification inhibitors, bafilomycin A1 and chloroquine. These data indicate that MNV-1 enters permissive cells in a pH-independent manner.


Assuntos
Infecções por Caliciviridae/tratamento farmacológico , Células Dendríticas/virologia , Macrófagos/virologia , Norovirus/fisiologia , Internalização do Vírus/efeitos dos fármacos , Animais , Antirreumáticos/farmacologia , Infecções por Caliciviridae/metabolismo , Infecções por Caliciviridae/virologia , Técnicas de Cultura de Células , Cloroquina/farmacologia , Células Dendríticas/química , Células Dendríticas/efeitos dos fármacos , Endossomos/química , Endossomos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Macrolídeos/farmacologia , Macrófagos/química , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Norovirus/efeitos dos fármacos , Norovirus/metabolismo , Proteínas não Estruturais Virais/biossíntese
10.
Bio Protoc ; 8(16)2018 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-30214914

RESUMO

Protein tagging is a powerful method of investigating protein function. However, modifying positive-strand RNA virus proteins in the context of viral infection can be particularly difficult as their compact genomes and multifunctional proteins mean even small changes can inactivate or attenuate the virus. Although targeted approaches to functionally tag viral proteins have been successful, these approaches are time consuming and inefficient. A strategy that has been successfully applied to several RNA viruses is whole-genome transposon insertional mutagenesis. A library of viral genomes, each containing a single randomly placed small insertion, is selected by passaging in cell culture and the insertion sites can be identified using Next Generation Sequencing (NGS). Here we describe a protocol for transposon mutagenesis of the 16681 strain of dengue virus, serotype 2. Mutant dengue virus libraries containing short randomly placed insertions are passaged through mammalian cells and insertions are mapped by NGS of the viable progeny. The protocol is divided into four stages: transposon mutagenesis of a dengue cDNA clone, viral genome transfection into permissive cells, isolation of viral progeny genomes, and sequencing library preparation.

11.
mBio ; 4(4)2013 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-23860770

RESUMO

UNLABELLED: Human noroviruses (HuNoVs) cause significant morbidity and mortality worldwide. However, despite substantial efforts, a small-animal model for HuNoV has not been described to date. Since "humanized" mice have been successfully used to study human-tropic pathogens in the past, we challenged BALB/c mice deficient in recombination activation gene (Rag) 1 or 2 and common gamma chain (γc) (Rag-γc) engrafted with human CD34+ hematopoietic stem cells, nonengrafted siblings, and immunocompetent wild-type controls with pooled stool isolates from patients positive for HuNoV. Surprisingly, both humanized and nonhumanized BALB/c Rag-γc-deficient mice supported replication of a GII.4 strain of HuNoV, as indicated by increased viral loads over input. In contrast, immunocompetent wild-type BALB/c mice were not infected. An intraperitoneal route of infection and the BALB/c genetic background were important for facilitating a subclinical HuNoV infection of Rag-γc-deficient mice. Expression of structural and nonstructural proteins was detected in cells with macrophage-like morphology in the spleens and livers of BALB/c Rag-γc-deficient mice, confirming the ability of HuNoV to replicate in a mouse model. In summary, HuNoV replication in BALB/c Rag-γc-deficient mice is dependent on the immune-deficient status of the host but not on the presence of human immune cells and provides the first genetically manipulable small-animal model for studying HuNoV infection. IMPORTANCE: Human noroviruses are a significant cause of viral gastroenteritis worldwide, resulting in significant morbidity and mortality. Antivirals and vaccines are currently not available, in part due to the inability to study these viruses in a genetically manipulable, small-animal model. Herein, we report the first mouse model for human noroviruses. This model will accelerate our understanding of human norovirus biology and provide a useful resource for evaluating antiviral therapies.


Assuntos
Infecções por Caliciviridae/patologia , Infecções por Caliciviridae/virologia , Modelos Animais de Doenças , Norovirus/patogenicidade , Animais , Fezes/virologia , Genoma Viral , Humanos , Fígado/virologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Dados de Sequência Molecular , Norovirus/genética , Norovirus/crescimento & desenvolvimento , Norovirus/isolamento & purificação , RNA Viral/genética , Análise de Sequência de DNA , Baço/virologia , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA