Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
PLoS Pathog ; 15(6): e1007827, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31181119

RESUMO

P10 is a small, abundant baculovirus protein that accumulates to high levels in the very late stages of the infection cycle. It is associated with a number of intracellular structures and implicated in diverse processes from occlusion body maturation to nuclear stability and lysis. However, studies have also shown that it is non-essential for virus replication, at least in cell culture. Here, we describe the use of serial block-face scanning electron microscopy to achieve high-resolution 3D characterisation of P10 structures within Trichoplusia ni TN-368 cells infected with Autographa californica multiple nucleopolyhedrovirus. This has enabled unparalleled visualisation of P10 and determined the independent formation of dynamic perinuclear and nuclear vermiform fibrous structures. Our 3D data confirm the sequence of ultrastructural changes that create a perinuclear cage from thin angular fibrils within the cytoplasm. Over the course of infection in cultured cells, the cage remodels to form a large polarised P10 mass and we suggest that these changes are critical for nuclear lysis to release occlusion bodies. In contrast, nuclear P10 forms a discrete vermiform structure that was observed in close spatial association with both electron dense spacers and occlusion bodies; supporting a previously suggested role for P10 and electron dense spacers in the maturation of occlusion bodies. We also demonstrate that P10 hyper-expression is critical for function. Decreasing levels of p10 expression, achieved by manipulation of promoter length, correlated with reduced P10 production, a lack of formation of P10 structures and a concomitant decrease in nuclear lysis.


Assuntos
Núcleo Celular/metabolismo , Regulação Viral da Expressão Gênica/fisiologia , Nucleopoliedrovírus/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Mariposas , Nucleopoliedrovírus/química , Nucleopoliedrovírus/genética , Domínios Proteicos , Proteínas Virais/química , Proteínas Virais/genética
2.
Curr Issues Mol Biol ; 34: 215-230, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31167962

RESUMO

Over 35 years since it was established to make recombinant proteins, the baculovirus expression vector system continues to develop and improve. Early systems for recombinant virus selection were laborious, but better methods were rapidly devised that enabled non-virologists to use baculovirus vectors successfully in a wide range of applications. These applications include multiple gene expression for complex molecules, production of adeno-associated virus-like particles for gene therapy, the use of baculovirus budded virus for the same purpose, numerous potential human and animal vaccines, and for other therapeutic proteins. A number of products for human and veterinary use are now on the market, which attests to the utility of the systems. Despite these successes, baculovirus vectors essentially remain in a relatively primitive state of development. Many proteins, particularly membrane-bound or secreted products, continue to be difficult to produce. Various research groups are working to identify potential areas of improvement, which if combined into an ideal vector might offer considerable advances to the system. This chapter will review some of the most recent reports and highlight those that might have generic application for recombinant protein synthesis in insect cells. We also summarize parallel developments in host cells used for baculovirus expression and how culture conditions can influence protein production.


Assuntos
Baculoviridae/genética , Expressão Gênica , Engenharia Genética , Vetores Genéticos/genética , Animais , Engenharia Genética/métodos , Humanos , Engenharia de Proteínas , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
3.
J Virol ; 91(13)2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28424279

RESUMO

Baculoviruses encode a variety of auxiliary proteins that are not essential for viral replication but provide them with a selective advantage in nature. P10 is a 10-kDa auxiliary protein produced in the very late phase of gene transcription by Autographa californica multiple nucleopolyhedrovirus (AcMNPV). The P10 protein forms cytoskeleton-like structures in the host cell that associate with microtubules varying from filamentous forms in the cytoplasm to aggregated perinuclear tubules that form a cage-like structure around the nucleus. These P10 structures may have a role in the release of occlusion bodies (OBs) and thus mediate the horizontal transmission of the virus between insect hosts. Here, using mass spectrometric analysis, it is demonstrated that the C terminus of P10 is phosphorylated during virus infection of cells in culture. Analysis of P10 mutants encoded by recombinant baculoviruses in which putative phosphorylation residues were mutated to alanine showed that serine 93 is a site of phosphorylation. Confocal microscopy examination of the serine 93 mutant structures revealed aberrant formation of the perinuclear tubules. Thus, the phosphorylation of serine 93 may induce the aggregation of filaments to form tubules. Together, these data suggest that the phosphorylation of serine 93 affects the structural conformation of P10.IMPORTANCE The baculovirus P10 protein has been researched intensively since it was first observed in 1969, but its role during viral infection remains unclear. It is conserved in the alphabaculoviruses and expressed at high levels during virus infection. Producing large amounts of a protein is wasteful for the virus unless it is advantageous for the survival of its progeny, and therefore, P10 presents an enigma. As P10 polymerizes to form organized cytoskeletal structures that colocalize with host cell microtubules, the structural relationship of the protein with the host cell may present a key to help understand the function and importance of this protein. This study addresses the importance of the structural changes in P10 during infection and how they may be governed by phosphorylation. The P10 structures affected by phosphorylation are closely associated with the viral progeny and thus may potentially be responsible for its dissemination and survival.


Assuntos
Processamento de Proteína Pós-Traducional , Proteínas Virais/química , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Análise Mutacional de DNA , Insetos , Espectrometria de Massas , Fosforilação , Conformação Proteica , Multimerização Proteica , Proteínas Virais/genética
4.
J Virol ; 88(6): 3548-56, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24403587

RESUMO

UNLABELLED: Superinfection exclusion is the ability of an established virus to interfere with a second virus infection. This effect was studied in vitro during lepidopteran-specific nucleopolyhedrovirus (genus Alphabaculovirus, family Baculoviridae) infection. Homologous interference was detected in Sf9 cells sequentially infected with two genotypes of Autographa californica multiple nucleopolyhedrovirus (AcMNPV), each one expressing a different fluorescent protein. This was a progressive process in which a sharp decrease in the signs of infection caused by the second virus was observed, affecting not only the number of coinfected cells observed, but also the level of protein expression due to the second virus infection. Superinfection exclusion was concurrent with reorganization of cytoplasmic actin to F-actin in the nucleus, followed by budded virus production (16 to 20 h postinfection). Disruption of actin filaments by cell treatment with cytochalasin D resulted in a successful second infection. Protection against heterologous nucleopolyhedrovirus infection was also demonstrated, as productive infection of Sf9 cells by Spodoptera frugiperda nucleopolyhedrovirus (SfMNPV) was inhibited by prior infection with AcMNPV, and vice versa. Finally, coinfected cells were observed following inoculation with mixtures of these two phylogenetically distant nucleopolyhedroviruses--AcMNPV and SfMNPV--but at a frequency lower than predicted, suggesting interspecific virus interference during infection or replication. The temporal window of infection is likely necessary to maintain genotypic diversity that favors virus survival but also permits dual infection by heterospecific alphabaculoviruses. IMPORTANCE: Infection of a cell by more than one virus particle implies sharing of cell resources. We show that multiple infection, by closely related or distantly related baculoviruses, is possible only during a brief window of time that allows additional virus particles to enter an infected cell over a period of ca. 16 h but then blocks multiple infections as newly generated virus particles begin to leave the infected cell. This temporal window has two important consequences. First, it allows multiple genotypes to almost simultaneously infect cells within the host, thus generating genetically diverse virus particles for transmission. Second, it provides a mechanism by which different viruses replicating in the same cell nucleus can exchange genetic material, so that the progeny viruses may be a mosaic of genes from each of the parental viruses. This opens a completely new avenue of research into the evolution of these insect pathogens.


Assuntos
Actinas/metabolismo , Coinfecção/veterinária , Nucleopoliedrovírus/fisiologia , Spodoptera/virologia , Superinfecção/veterinária , Animais , Núcleo Celular/metabolismo , Coinfecção/metabolismo , Coinfecção/virologia , Citoplasma/metabolismo , Proteínas de Insetos/metabolismo , Nucleopoliedrovírus/genética , Células Sf9 , Spodoptera/metabolismo , Superinfecção/metabolismo , Superinfecção/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
5.
J Extracell Biol ; 3(7): e163, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38947876

RESUMO

Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is an enveloped DNA virus of the Baculoviridae family. This baculovirus is widely exploited for the biological control of insect pest species and as an expression platform to produce recombinant proteins in insect cells. Extracellular vesicles (EVs) are secreted by all cells and are involved in key roles in many biological processes through their cargo consisting of proteins, RNA or DNA. In viral infections, EVs have been found to transfer both viral and cellular cargo that can elicit either a pro- or antiviral response in recipient cells. Here, small EVs (sEVs) released by Spodoptera frugiperda (Sf) insect cells were characterised for the first time. Using S. frugiperda (SfC1B5) cells stably expressing the baculovirus gp64, the viral envelope protein GP64 was shown to be incorporated into sEVs. Sf9 cells were also transfected with a bacmid AcMNPV genome lacking p6.9 (AcΔP6.9) to prevent budded virus production. The protein content of sEVs from both mock- and AcΔP6.9-transfected cells were analysed by mass spectrometry. In addition to GP64, viral proteins Ac-F, ME-53 and viral ubiquitin were identified, as well as many host proteins including TSG101-which may be useful as a protein marker for sEVs.

6.
J Immunol Methods ; 512: 113405, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36496007

RESUMO

Humanised antibodies targeting Crimean-Congo Haemorrhagic virus (CCHFV) are needed for the development and standardisation of serological assays. These assays are needed to address a shortfall in available tests that meet regulatory diagnostic standards and to aid surveillance activities to extend knowledge on the distribution of CCHFV. To generate a humanised monoclonal antibody against CCHFV, we have compared two methods: the traditional mouse hybridoma approach with subsequent sequencing and humanisation of antibodies versus a non-animal alternative using a human combinatorial antibody library (HuCAL). Our results demonstrated that the mouse hybridoma followed by humanisation protocol gave higher affinity antibodies. Whilst not yet able to demonstrate the generation of equivalent humanised antibodies without the use of animals, sequencing data enables the subsequent production of recombinant antibodies, thus providing a reduction in future animal usage for this application. Ultimately, our report provides information on development of a humanised standardised control, which can form an important positive control component of serological assays against CCHFV.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Humanos , Animais , Camundongos , Febre Hemorrágica da Crimeia/diagnóstico , Febre Hemorrágica da Crimeia/epidemiologia , Hibridomas , Anticorpos Antivirais , Imunoglobulina G
7.
Methods ; 55(1): 52-7, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21945427

RESUMO

Baculoviruses have a unique bi-phasic life cycle and powerful promoters, which greatly facilitates their use for recombinant protein expression in insect cells. We have developed an expression system that utilizes homologous recombination in insect cells between a transfer plasmid containing a gene to be expressed and a replication-deficient virus (bacmid). Only recombinant virus can replicate facilitating the rapid production of multiple recombinant viruses using robotic liquid handlers. The bacmid has also been genetically optimized for improved protein expression and stability. We describe the application of this system for high level production of recombinant proteins.


Assuntos
Baculoviridae/genética , Expressão Gênica , Vetores Genéticos/genética , Ensaios de Triagem em Larga Escala , Plasmídeos/genética , Spodoptera/metabolismo , Animais , Automação Laboratorial , Linhagem Celular , Vetores Genéticos/química , Recombinação Homóloga , Plasmídeos/química , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Spodoptera/citologia , Spodoptera/virologia , Replicação Viral/genética
8.
J Gen Virol ; 92(Pt 5): 1061-1070, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21228125

RESUMO

A laboratory culture of Spodoptera exigua was examined to assess covert (latent or persistent) baculovirus infections and spontaneous disease outbreaks. Two nucleopolyhedrovirus (NPV) species were found to be reactivated from a covert state in a laboratory culture of S. exigua to fully lethal forms. These were identified as S. exigua multinucleopolyhedrovirus (SeMNPV) and Mamestra brassicae NPV (MbNPV) using restriction enzyme analysis of purified viral DNA. Sequence data derived from both overtly and covertly virus-infected insects revealed highly conserved sequences for lef-8, lef-9 and polyhedrin gene sequence (98-100 % nucleotide identity to SeMNPV published sequence). By monitoring spontaneous overt infections and quantifying viral DNA (by quantitative-PCR) in asymptomatic individuals over two generations we identified fluctuating trends in viral DNA levels from covert SeMNPV and MbNPV within an S. exigua host population. Virus levels per insect life stage ranged from 3.51±0.101×10(5) to 0.29±0.036 pg (detection limit at 0.06 pg). Bioassays performed with this culture of larvae showed a differential susceptibility to SeMNPV-like or MbNPV-like viruses, with SeMNPV superinfections being extremely virulent. The data presented has broad implications relating to our understanding of transmission patterns of baculovirus in the environment and the role of covert infections in host-pathogen interaction dynamics.


Assuntos
Baculoviridae/isolamento & purificação , Lepidópteros/virologia , Animais , Baculoviridae/genética , Baculoviridae/patogenicidade , Linhagem Celular , Sequência Conservada , Dados de Sequência Molecular , Análise de Sequência de DNA , Homologia de Sequência do Ácido Nucleico , Análise de Sobrevida , Proteínas Virais/genética , Virulência , Ativação Viral , Latência Viral
9.
Appl Environ Microbiol ; 76(3): 803-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20008167

RESUMO

The stabilities of the Spodoptera frugiperda multiple nucleopolyhedrovirus (SfMNPV) complete genome bacmid (Sfbac) and a deletion recombinant (Sf29null) in which the Sf29 gene was replaced by a kanamycin resistance cassette were determined during sequential rounds of per os infection in insect larvae. The Sf29 gene is a viral factor that determines the number of virions in occlusion bodies (OBs). The Sf29null bacmid virus was able to recover the Sf29 gene during passage. After the third passage (P3) of Sf29null bacmid OBs, the population was observed to reach an equilibrium involving a mixture of those with a kanamycin resistance cassette and those with the Sf29 gene. The biological activity of Sf29null bacmid OBs at P3 was similar to that of Sfbac OBs. The recovered gene in the Sf29null virus was 98 to 100% homologous to the Sf29 genes of different SfMNPV genotypes. Reverse transcription-PCR analysis of uninoculated S. frugiperda larvae confirmed the expression of the SfMNPV ie-0 and Sf29 genes, indicating that the insect colony harbors a covert SfMNPV infection. Additionally, the nonessential bacterial artificial chromosome vector was spontaneously deleted from both viral genomes upon passage in insects.


Assuntos
Deleção de Genes , Nucleopoliedrovírus/genética , Spodoptera/virologia , Animais , Cromossomos Artificiais Bacterianos/metabolismo , Clonagem Molecular , DNA Viral/genética , DNA Viral/metabolismo , Regulação Viral da Expressão Gênica , Frequência do Gene , Variação Genética , Vetores Genéticos , Genoma Viral , Genótipo , Larva/genética , Larva/metabolismo , Larva/virologia , Nucleopoliedrovírus/metabolismo , Controle Biológico de Vetores , Filogenia , Análise de Sequência de DNA , Inoculações Seriadas , Spodoptera/genética , Spodoptera/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vírion/genética , Vírion/metabolismo , Replicação Viral/genética
10.
Cell Biol Toxicol ; 26(1): 57-68, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19655260

RESUMO

Generating large amounts of recombinant protein in transgenic animals is often challenging and has a number of drawbacks compared to cell culture systems. The baculovirus expression vector system (BEVS) uses virus-infected insect cells to produce recombinant proteins to high levels, and these are usually processed in a similar way to the native protein. Interestingly, since the development of the BEVS, the virus most often used (Autographa californica multi-nucleopolyhedovirus; AcMNPV) has been little altered genetically from its wild-type parental virus. In this study, we modified the AcMNPV genome in an attempt to improve recombinant protein yield, by deleting genes that are non-essential in cell culture. We deleted the p26, p10 and p74 genes from the virus genome, replacing them with an antibiotic selection cassette, allowing us to isolate recombinants. We screened and identified recombinant viruses by restriction enzyme analysis, PCR and Western blot. Cell viability analysis showed that the deletions did not improve the viability of infected cells, compared to non-deletion viruses. However, expression studies showed that recombinant protein levels for the deletion viruses were significantly higher than the expression levels of non-deletion viruses. These results confirm that there is still great potential for improving the BEVS, further increasing recombinant protein expression yields and stability in insect cells.


Assuntos
Baculoviridae/genética , Deleção de Genes , Genes Virais , Engenharia Genética/métodos , Vetores Genéticos , Proteínas Recombinantes/biossíntese , Animais , Linhagem Celular , Expressão Gênica , Insetos , Regulação para Cima
11.
Biotechnol Appl Biochem ; 56(3): 85-93, 2010 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-20441568

RESUMO

Secretory and membrane-bound proteins are generally produced in lower amounts in insect cells compared with cytoplasmic and nuclear proteins. There may be many reasons for this, including degradation of recombinant proteins by proteases, competition for cellular resources between native and recombinant proteins, and physical blockage of the secretory pathways. In the present study, we describe the construction of a baculovirus in which chiA (chitinase) and cath (cathepsin) genes have been deleted and show improved recombinant protein expression using this vector. We confirmed the complete removal of both genes by PCR, restriction enzyme analysis and enzyme assays, and the modified virus DNA was shown to be stable in bacterial cells over multiple passages. A selection of recombinant genes were inserted into the double-deletion virus and their expression levels compared with recombinant viruses that had single or no gene deletions. In all instances, the double-deletion viruses showed greatly enhanced levels of protein production for both secreted and nuclear/cytoplasmic proteins. In summary, we have conclusively demonstrated the importance of this deletion vector for the high-level production of recombinant proteins.


Assuntos
Baculoviridae/genética , Proteínas de Membrana/biossíntese , Engenharia de Proteínas/métodos , Proteínas Recombinantes/biossíntese , Animais , Baculoviridae/enzimologia , Catepsinas/genética , Células Cultivadas , Quitinases/genética , Deleção de Genes , Expressão Gênica , Humanos , Insetos/citologia , Proteínas de Membrana/genética
12.
Viruses ; 12(1)2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31906433

RESUMO

Many steps in the baculovirus life cycle, from initial ingestion to the subsequent infection of all larval cells, remain largely unknown; primarily because it has hitherto not been possible to follow individual genomes and their lineages. Use of ANCHORTM technology allows a high intensity fluorescent labelling of DNA. When applied to a virus genome, it is possible to follow individual particles, and the overall course of infection. This technology has been adapted to enable labelling of the baculovirus Autographa californica Multiple NucleoPolyhedroVirus genome, as a first step to its application to other baculoviruses. AcMNPV was modified by inserting the two components of ANCHORTM: a specific DNA-binding protein fused to a fluorescent reporter, and the corresponding DNA recognition sequence. The resulting modified virus was stable, infectious, and replicated correctly in Spodoptera frugiperda 9 (Sf9) cells and in vivo. Both budded viruses and occlusion bodies were clearly distinguishable, and infecting cells or larvae allowed the infection process to be monitored in living cells or tissues. The level of fluorescence in the culture medium of infected cells in vitro showed a good correlation with the number of infectious budded viruses. A cassette that can be used in other baculoviruses has been designed. Altogether our results introduce for the first time the generation of autofluorescent baculovirus and their application to follow infection dynamics directly in living cells or tissues.


Assuntos
DNA Viral/metabolismo , Nucleopoliedrovírus/fisiologia , Replicação Viral , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fluorometria , Genoma Viral/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Larva/virologia , Microscopia de Fluorescência , Células Sf9 , Spodoptera
13.
J Virol ; 82(16): 7897-904, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550678

RESUMO

The genome of Spodoptera frugiperda multiple nucleopolyhedrovirus (NPV) was inserted into a bacmid (Sfbac) and used to produce a mutant lacking open reading frame 29 (Sf29null). Sf29null bacmid DNA was able to generate an infection in S. frugiperda. Approximately six times less DNA was present in occlusion bodies (OBs) produced by the Sf29null bacmid in comparison to viruses containing this gene. This reduction in DNA content was consistent with fewer virus particles being packaged within Sf29null bacmid OBs, as determined by fractionation of dissolved polyhedra and comparison of occlusion-derived virus (ODV) infectivity in cell culture. DNA from Sfbac, Sf29null, or Sf29null-repair, in which the gene deletion had been repaired, were equally infectious when used to transfect S. frugiperda. All three viruses produced similar numbers of OBs, although those from Sf29null were 10-fold less infectious than viruses with the gene. Insects infected with Sf29null bacmid died approximately 24 h later than positive controls, consistent with the reduced virus particle content of Sf29null OBs. Transcripts from Sf29 were detected in infected insects 12 h prior to those from the polyhedrin gene. Homologs to Sf29 were present in other group II NPVs, and similar sequences were present in entomopoxviruses. Analysis of the Sf29 predicted protein sequence revealed signal peptide and transmembrane domains, but the presence of 12 potential N-glycosylation sites suggest that it is not an ODV envelope protein. Other motifs, including zinc-binding and threonine-rich regions, suggest degradation and adhesion functions. We conclude that Sf29 is a viral factor that determines the number of ODVs occluded in each OB.


Assuntos
Regulação Viral da Expressão Gênica , Nucleocapsídeo/genética , Nucleopoliedrovírus/metabolismo , Vírion , Animais , Células Cultivadas , DNA Viral/metabolismo , Deleção de Genes , Genes Virais , Glicosilação , Humanos , Insetos , Nucleocapsídeo/metabolismo , Fases de Leitura Aberta , Sinais Direcionadores de Proteínas , Vírion/química
14.
Virus Res ; 136(1-2): 202-5, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18538883

RESUMO

Homologs to Autographa californica multiple nucleopolyhedrovirus (AcMNPV) open reading frame (ORF) 136 or Acp26 are present within almost all nucleopolyhedroviruses (NPVs). Two copies of the gene are found in some members of group II NPVs, suggesting that it may play an important role in transmission or replication. Phylogenetic analysis revealed that the predicted protein has some similarity with Camelpox virus v-slfn protein, which reduces the virulence of orthopoxviruses in vivo. To investigate the influence of ACP26 on the infectivity and virulence of AcMNPV a bacmid system was used to delete the Acp26 ORF. The Acp26null bacmid was able to generate a transmissible infection in cell culture and larvae, indicating that Acp26 is not essential for propagation of viral infection in vitro or in vivo. Deletion of Acp26 from the AcMNPV genome had no apparent effect on timing or production of infectious BV in cell culture or in insect larvae. Additional comparisons of AcMNPV and Acp26null bacmid viruses showed that deletion of Acp26 did not significantly influence the infectivity and virulence of AcMNPV occlusion bodies or the number produced. The Acp26 may be an auxiliary gene with subtle effects on virus replication and transmission.


Assuntos
Nucleopoliedrovírus/fisiologia , Nucleopoliedrovírus/patogenicidade , Proteínas Virais/fisiologia , Fatores de Virulência/fisiologia , Animais , Linhagem Celular , Deleção de Genes , Larva/virologia , Nucleopoliedrovírus/genética , Spodoptera , Proteínas Virais/genética , Virulência , Fatores de Virulência/genética
15.
Virus Res ; 133(2): 157-66, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18261819

RESUMO

An Autographa californica nucleopolyhedrovirus (AcMNPV) mutant (AcdefrT) isolated from virus-infected Trichoplusia ni (TN-368) cells produced plasma membrane blebbing and caspase-3-like activity late in infection. It also synthesized less polyhedra, but displayed enhanced budded virus formation in TN-368 cells. This phenotype resulted from dual mutations in p35 and FP-25. In this study we showed that enhanced budded virus production occurs because the hourly rate of release of virus from AcdefrT-infected cells is higher than that for AcMNPV and it continues for longer. This may be the trigger for the induction of apoptosis late in AcdefrT-infected TN-368 cells. However, laddering of host DNA was absent in TN-368 cells infected with AcdefrT, but was observed in Spodoptera frugiperda cells. Very late polyhedrin protein production and occlusion body formation was reduced in AcdefrT-infected TN-368 cells, but chitinase and capsid late gene expression remained unchanged. The AcdefrT was rescued with a copy of a baculovirus iap3, to replace the absent p35. This modification abolished most plasma membrane blebbing in AcdefrT-infected TN-368 cells, but did not affect enhanced budded virus production. These data suggest that inhibitors of apoptosis are required in T. ni cells, particularly when the production of budded virus is enhanced.


Assuntos
Apoptose , Mariposas/virologia , Mutação , Proteínas do Nucleocapsídeo/genética , Nucleopoliedrovírus/metabolismo , Proteínas Virais/genética , Animais , Células Cultivadas , Nucleopoliedrovírus/genética , Spodoptera/virologia
16.
Virus Res ; 135(1): 42-7, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18405997

RESUMO

A reovirus was isolated from Operophtera brumata (ObRV) and its parasitoid wasp Phobocampe tempestiva. Each of the 10 dsRNA genome segments of ObRV was sequenced and shown to contain a single open reading frame (ORF). Conserved motifs ([+ve] 5'-AAATAAA ...(G)/(T)AGGTT-3') were found at the termini of each segment, with the exception of Seg-6 and Seg-8, where the 5' termini were 5'-AACAAA...-3'. The putative proteins encoded by each segment were compared with those of other members of the family Reoviridae. Phylogenetic comparisons to published sequences for the RNA-dependent RNA polymerase genes from other reoviruses indicated that ObRV is most closely related to members of the genus Cypovirus. However, unlike the cypoviruses, ObRV has a double-layered capsid structure. When the protein encoded by ObRV Seg-10 was expressed (by inserting the open reading frame into a baculovirus expression vector) no 'occlusion bodies' were observed in the recombinant baculovirus infected insect cell cultures. This suggests that unlike the cypoviruses, Seg-10 of ObRV does not contain a polyhedrin gene. Further phylogenetic comparisons also identified relationships between Seg-2 and Seg-10 of ObRV, and genes of Diadromus pulchellus Idnoreovirus 1 (DpIRV1), suggesting that ObRV represents a new species from the genus Idnoreovirus.


Assuntos
Vírus de Insetos/genética , Vírus de Insetos/isolamento & purificação , Mariposas/virologia , Reoviridae/genética , Reoviridae/isolamento & purificação , Vespas/virologia , Animais , Expressão Gênica , Genoma Viral , Vírus de Insetos/classificação , Vírus de Insetos/metabolismo , Dados de Sequência Molecular , Fases de Leitura Aberta , Filogenia , Reoviridae/classificação , Reoviridae/metabolismo , Análise de Sequência , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
17.
J Med Virol ; 80(12): 2127-33, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19040289

RESUMO

Junín arenavirus is the etiologic agent of Argentine hemorrhagic fever. Due to its morbidity and high mortality rate in untreated cases, this endemic disease is of mandatory report in Argentina. Secure and accurate diagnostic methods are needed for the epidemiological surveillance of the disease. Current assays rely on antigens prepared from lysates of virus infected mammalian cells. The bio-safety issue related to the manipulation of large quantities of virus restricts such antigen production to laboratories with the appropriate containment facilities. In this report, we describe the development of an enzyme linked immunosorbent assay for the etiologic confirmation of the disease, based on recombinant antigens expressed in insect cells. Eight different variables of the assay were optimized with the Taguchi approach for experimental design (L18 design, seven three-level factors and one two-level factor). The area under the receiver operating characteristics (ROC) curve was 0.966, showing the high accuracy of the test discriminating positive from negative samples. Taking into account the biosafety benefits, the high yields of antigen in cell culture, and the general performance of the assay, it is expected that it will be a useful alternative to the current ELISA for the detection of antibodies in sera from convalescent patients.


Assuntos
Testes Diagnósticos de Rotina/métodos , Febre Hemorrágica Americana/diagnóstico , Vírus Junin/genética , Proteínas Recombinantes , Proteínas Virais , Animais , Linhagem Celular , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Insetos , Curva ROC
18.
Biotechnol Bioeng ; 101(6): 1115-22, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18781697

RESUMO

The baculovirus expression system is one of the most popular methods used for the production of recombinant proteins but has several complex steps which have proved inherently difficult to adapt to a multi-parallel process. We have developed a bacmid vector that does not require any form of selection pressure to separate recombinant virus from non-recombinant parental virus. The method relies on homologous recombination in insect cells between a transfer vector containing a gene to be expressed and a replication-deficient bacmid. The target gene replaces a bacterial replicon at the polyhedrin loci, simultaneously restoring a virus gene essential for replication. Therefore, only recombinant virus can replicate facilitating the rapid production of multiple recombinant viruses on automated platforms in a one-step procedure. Using this vector allowed us to automate the generation of multiple recombinant viruses with a robotic liquid handler and then rapidly screen infected insect cell supernatant for the presence of secreted proteins.


Assuntos
Baculoviridae/genética , Biotecnologia/métodos , Vetores Genéticos , Proteínas Recombinantes/genética , Animais , Linhagem Celular , Clonagem Molecular , Expressão Gênica , Insetos , Biologia Molecular/métodos , Recombinação Genética , Replicação Viral
19.
Curr Protoc Protein Sci ; 91: 5.4.1-5.4.6, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29516481

RESUMO

This unit provides information on the replication cycle of insect baculovirus to provide an understanding of how this virus has been adapted for use as an expression vector for recombinant proteins in insect cells. We provide an overview of the virus structure and its unique bi-phasic replication cycle, which has been exploited in developing the virus as an expression vector. We also review the development of the baculovirus expression vector system (BEVS), from the mid-1980s to the present day in which the BEVS is now an established tool for the production of a range of recombinant proteins and multi-protein complexes including virus-like particles. We describe advances made to the BEVS to allow the rapid and easy production of recombinant viruses and developments to improve protein yield. We finish by describing the application of recombinant BacMam as vectors for the delivery of genes into mammalian and human cells. © 2018 by John Wiley & Sons, Inc.


Assuntos
Baculoviridae/genética , Baculoviridae/metabolismo , Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Proteínas Recombinantes/biossíntese , Animais , Humanos , Proteínas Recombinantes/genética
20.
Curr Protoc Protein Sci ; 91: 5.5.1-5.5.22, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29516484

RESUMO

Baculovirus expression systems are well established as an easy and reliable way to produce high quality recombinant proteins. Baculoviruses can also be used to transduce mammalian cells, termed 'BacMam', with considerable potential in biomedical applications. This chapter explains the process of making a recombinant baculovirus, encompassing production of a recombinant virus by homologous recombination in insect cells, followed by amplification and titration of the virus-all steps needed before commencing gene expression and protein production. We also cover the use of small-scale test expression to provide an initial indication of quality and protein yield. Whereas proteins expressed at high levels can be directly scaled up, more challenging proteins may require optimization of cell lines, growth conditions, or harvest times. Scale-up and purification approaches are discussed, focusing on working with large shake cultures and use of the Wave bioreactor. © 2018 by John Wiley & Sons, Inc.


Assuntos
Baculoviridae/genética , Baculoviridae/metabolismo , Reatores Biológicos , Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Proteínas Recombinantes/biossíntese , Animais , Humanos , Proteínas Recombinantes/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa