Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
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 Virol ; 86(2): 844-53, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22072745

RESUMO

Autographa californica multiple nucleopolyhedrovirus (AcMNPV) replicates in the nucleus of insect cells to produce nucleocapsids, which are transported from the nucleus to the plasma membrane for budding through GP64-enriched areas to form budded viruses. However, little is known about the anterograde trafficking of baculovirus nucleocapsids in insect cells. Preliminary confocal scanning laser microscopy studies showed that enhanced green fluorescent protein (EGFP)-tagged nucleocapsids and capsid proteins aligned and colocalized with the peripheral microtubules of virus-infected insect cells. A colchicine inhibition assay of virus-infected insect cells showed a significant reduction in budded virus production, providing further evidence for the involvement of microtubules and suggesting a possible role of kinesin in baculovirus anterograde trafficking. We investigated the interaction between AcMNPV nucleocapsids and kinesin-1 with fluorescence resonance energy transfer-fluorescence lifetime imaging microscopy (FRET-FLIM) and show for the first time that AcMNPV capsid proteins VP39 and EXON0, but not Orf1629, interact with the tetratricopeptide repeat (TPR) domain of kinesin. The excited-state fluorescence lifetime of EGFP fused to VP39 or EXON0 was quenched from 2.4 ± 1 ns to 2.1 ± 1 ns by monomeric fluorescent protein (mDsRed) fused to TPR (mDsRed-TPR). However, the excited-state fluorescence lifetime of an EGFP fusion of Orf1629 remained unquenched by mDsRed-TPR. These data indicate that kinesin-1 plays an important role in the anterograde trafficking of baculovirus in insect cells.


Assuntos
Proteínas do Capsídeo/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas de Insetos/metabolismo , Cinesinas/metabolismo , Mariposas/metabolismo , Nucleopoliedrovírus/metabolismo , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Linhagem Celular , Cinesinas/química , Mariposas/virologia , Nucleopoliedrovírus/química , Nucleopoliedrovírus/genética
7.
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
8.
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
9.
J Pain Symptom Manage ; 63(6): e601-e610, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35595373

RESUMO

BACKGROUND: Each year, approximately one million older adults die in American intensive care units (ICUs) or survive with significant functional impairment. Inadequate symptom management, surrogates' psychological distress and inappropriate healthcare use are major concerns. Pioneering work by Dr. J. Randall Curtis paved the way for integrating palliative care (PC) specialists to address these needs, but convincing proof of efficacy has not yet been demonstrated. DESIGN: We will conduct a multicenter patient-randomized efficacy trial of integrated specialty PC (SPC) vs. usual care for 500 high-risk ICU patients over age 60 and their surrogate decision-makers from five hospitals in Pennsylvania. INTERVENTION: The intervention will follow recommended best practices for inpatient PC consultation. Patients will receive care from a multidisciplinary SPC team within 24 hours of enrollment that continues until hospital discharge or death. SPC clinicians will meet with patients, families, and the ICU team every weekday. SPC and ICU clinicians will jointly participate in proactive family meetings according to a predefined schedule. Patients in the control arm will receive routine ICU care. OUTCOMES: Our primary outcome is patient-centeredness of care, measured using the modified Patient Perceived Patient-Centeredness of Care scale. Secondary outcomes include surrogates' psychological symptom burden and health resource utilization. Other outcomes include patient survival, as well as interprofessional collaboration. We will also conduct prespecified subgroup analyses using variables such as PC needs, measured by the Needs of Social Nature, Existential Concerns, Symptoms, and Therapeutic Interaction scale. CONCLUSIONS: This trial will provide robust evidence about the impact of integrating SPC with critical care on patient, family, and health system outcomes.


Assuntos
Estado Terminal , Enfermagem de Cuidados Paliativos na Terminalidade da Vida , Idoso , Cuidados Críticos , Estado Terminal/terapia , Humanos , Unidades de Terapia Intensiva , Pessoa de Meia-Idade , Estudos Multicêntricos como Assunto , Cuidados Paliativos/métodos , Ensaios Clínicos Controlados Aleatórios como Assunto
10.
J Virol ; 84(24): 12886-94, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20943972

RESUMO

Using two-photon-induced fluorescence lifetime imaging microscopy, we corroborate an interaction (previously demonstrated by yeast two-hybrid domain analysis) of full-length vaccinia virus (VACV; an orthopoxvirus) A36 protein with the cellular microtubule motor protein kinesin. Quenching of enhanced green fluorescent protein (EGFP), fused to the C terminus of VACV A36, by monomeric red fluorescent protein (mDsRed), fused to the tetratricopeptide repeat (TPR) domain of kinesin, was observed in live chicken embryo fibroblasts infected with either modified vaccinia virus Ankara (MVA) or wild-type fowlpox virus (FWPV; an avipoxvirus), and the excited-state fluorescence lifetime of EGFP was reduced from 2.5 ± 0.1 ns to 2.1 ± 0.1 ns due to resonance energy transfer to mDsRed. FWPV does not encode an equivalent of intracellular enveloped virion surface protein A36, yet it is likely that this virus too must interact with kinesin to facilitate intracellular virion transport. To investigate possible interactions between innate FWPV proteins and kinesin, recombinant FWPVs expressing EGFP fused to the N termini of FWPV structural proteins Fpv140, Fpv168, Fpv191, and Fpv198 (equivalent to VACV H3, A4, p4c, and A34, respectively) were generated. EGFP fusions of intracellular mature virion (IMV) surface protein Fpv140 and type II membrane protein Fpv198 were quenched by mDsRed-TPR in recombinant FWPV-infected cells, indicating that these virion proteins are found within 10 nm of mDsRed-TPR. In contrast, and as expected, EGFP fusions of the IMV core protein Fpv168 did not show any quenching. Interestingly, the p4c-like protein Fpv191, which demonstrates late association with preassembled IMV, also did not show any quenching.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Infecções por Poxviridae/metabolismo , Infecções por Poxviridae/virologia , Proteínas Estruturais Virais/metabolismo , Vírion/metabolismo , Animais , Embrião de Galinha , Fibroblastos/virologia , Transferência Ressonante de Energia de Fluorescência , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Cinesinas , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/genética , Fótons , Plasmídeos , Poxviridae/patogenicidade , Infecções por Poxviridae/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/genética , Proteína Vermelha Fluorescente
11.
Transpl Int ; 24(8): 820-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21623952

RESUMO

Concerns over the safety of conventional viral vectors have limited the translation of gene transfer from an exciting experimental procedure to a successful clinical therapy in transplantation. Baculoviruses are insect viruses, but have the ability to enter mammalian cells and deliver potential therapeutic molecules with no evidence of viral replication. This study provides evidence of the ability of recombinant baculovirus to enter mammalian kidneys and livers during cold preservation. Six kidneys and six liver lobules retrieved from large pigs were perfused with University of Wisconsin (UW) solution containing a baculovirus tagged with green fluorescent protein and preserved for 8 h. In addition, six kidneys were perfused with UW containing a baculovirus expressing red fluorescent protein and preserved for 24 h. Green fluorescent virus particles were detected within transduced kidneys and livers after 8 h standard cold storage and red fluorescent protein mRNA was detected in kidneys after 24 h of cold preservation. There were no significant differences in tissue architecture, cell morphology or ATP content between experimental organs and their controls. Ex vivo transduction of organs with recombinant baculovirus during conventional cold preservation was demonstrated with no evidence of additional injury or reduction in cell viability.


Assuntos
Baculoviridae/genética , Soluções para Preservação de Órgãos/metabolismo , Preservação de Órgãos/métodos , Adenosina/farmacologia , Trifosfato de Adenosina/metabolismo , Alopurinol/farmacologia , Animais , Sobrevivência Celular , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Genômica , Glutationa/farmacologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Hipotermia Induzida , Insulina/farmacologia , Proteínas Luminescentes/metabolismo , Microscopia Confocal/métodos , Soluções para Preservação de Órgãos/farmacologia , Proteômica/métodos , RNA Mensageiro/metabolismo , Rafinose/farmacologia , Suínos , Fatores de Tempo , Proteína Vermelha Fluorescente
12.
J Invertebr Pathol ; 107 Suppl: S49-58, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21784231

RESUMO

The baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is able to transduce a wide range of mammalian cells and shows preferential uptake in some, particularly liver and kidney cells. This suggests that the virus may be useful for delivery of protective genes for ameliorating the effects of ischaemia reperfusion injury (IRI) in solid organs during transplantation procedures. In this chapter we discuss the advantages of the baculovirus over other virus vectors for gene delivery in organ transplantation and describe some of the protective genes which may be used to ameliorate the effects of IRI. We then describe a method for concentrating baculovirus for use in an ex vivo transduction model. Data are also provided for the effects of virus transduction in vitro on the innate and adaptive immune response. We conclude with a discussion on the future considerations for using baculovirus for delivery and expression of protective genes in organ transplantation.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Nucleopoliedrovírus/genética , Transplante de Órgãos , Células Cultivadas , Regulação Viral da Expressão Gênica , Técnicas de Transferência de Genes , Humanos , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/terapia
13.
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
14.
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
15.
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
16.
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
17.
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
18.
Cancer Biother Radiopharm ; 23(1): 108-13, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18298334

RESUMO

UNLABELLED: The cytotoxic ability of T-cells against tumor cells may be increased by interleukin (IL)-2. The infiltration of tumors by these cytotoxic T-cells may be enhanced by low-dose cyclophosphamide, which may also serve to deplete regulatory T-cells. Famotidine may increase IL-2 internalization by the IL-2 receptor on T-lymphocytes. We have treated 14 patients with either metastatic melanoma or kidney cancer, using CY 350 mg/M(2) intravenously (i.v.) over 1 hour followed by a continuous infusion IL-2 9 MIU/M(2)/24 hours for 72 hours and famotidine 20 mg i.v. twice per day. All patients had received prior systemic therapy. Cycles were repeated every 4 weeks until disease progression. PATIENT CHARACTERISTICS: 8 with melanoma, 8 males, median age, 64, median Eastern Cooperative Oncology Group performance status, 1; most common metastatic sites: lungs, lymph nodes, bone, and liver. Median number of cycles received=2 (range, 2-4). Most common toxicities were fever, nausea/emesis, hypomagnesemia, hypophosphatemia, hyponatremia, and rigors. All patients were treated on an oncology inpatient unit. One (1) patient with kidney cancer has had a partial response in lung and lymph nodes for 5 months, while 1 patient with melanoma had a partial response in pulmonary metastases. Cyclophosphamide and IL-2 with famotidine has evidence of antitumor activity in previously treated kidney cancer and melanoma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Melanoma/tratamento farmacológico , Idoso , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/secundário , Ciclofosfamida/administração & dosagem , Famotidina/administração & dosagem , Feminino , Humanos , Infusões Intravenosas , Interleucina-2/administração & dosagem , Neoplasias Renais/patologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Linfonodos/patologia , Metástase Linfática , Masculino , Melanoma/secundário , Pessoa de Meia-Idade , Invasividade Neoplásica , Resultado do Tratamento
19.
Vaccine ; 36(46): 7003-7010, 2018 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-30309744

RESUMO

African horse sickness is a severe, often fatal, arboviral disease of equids. The control of African horse sickness virus (AHSV) in endemic countries is based currently on the use of live attenuated vaccines despite some biosafety concerns derived from its biological properties. Thus, experimental vaccination platforms have been developed over the years in order to avoid the biosafety concerns associated with the use of attenuated vaccines. Various studies showed that baculovirus-expressed AHSV-VP2 or modified Vaccinia Ankara virus expressing AHSV-VP2 (MVA-VP2) induced virus neutralising antibodies and protective immunity in small animals and horses. AHSV is an antigenically diverse pathogen and immunity against AHS is serotype-specific. Therefore, AHS vaccines for use in endemic countries need to induce an immune response capable of protecting against all existing serotypes. For this reason, current live attenuated vaccines are administered as polyvalent preparations comprising combinations of AHSV attenuated strains of different serotypes. Previous studies have shown that it is possible to induce cross-reactive virus neutralising antibodies against different serotypes of AHSV by using polyvalent vaccines comprising combinations of either different serotype-specific VP2 proteins, or MVA-VP2 viruses. However, these strategies could be difficult to implement if induction of protective immunity is highly dependent on using a two-dose vaccination regime for each serotype the vaccine intends to protect against. In our study, we have tested the protective capacity of MVA-VP2 and baculovirus-expressed VP2 vaccines when a single dose was used. Groups of interferon alpha receptor knock-out mice were inoculated with either MVA-VP2 or baculovirus-expressed VP2 vaccines using one dose or the standard two-dose vaccination regime. After vaccination, all four vaccinated groups were challenged with AHSV and clinical responses, lethality and viraemia compared between the groups. Our results show that complete clinical protection was achieved after a single vaccination with either MVA-VP2 or baculovirus sub-unit VP2 vaccines.


Assuntos
Doença Equina Africana/prevenção & controle , Proteínas do Capsídeo/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Animais , Baculoviridae/genética , Modelos Animais de Doenças , Portadores de Fármacos , Feminino , Vetores Genéticos , Camundongos , Análise de Sobrevida , Resultado do Tratamento , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vaccinia virus/genética , Viremia/prevenção & controle
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA