Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 93(4)2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30463972

RESUMO

Defective interfering particles (DIPs) replicate at the expense of coinfecting, fully infectious homologous virus. Typically, they contain a highly deleted form of the viral genome. Utilizing single-cell analysis, here we report the discovery of a yet-unknown DIP type, derived from influenza A viruses (IAVs), termed OP7 virus. Instead of deletions, the genomic viral RNA (vRNA) of segment 7 (S7) carried 37 point mutations compared to the reference sequence, affecting promoter regions, encoded proteins, and genome packaging signals. Coinfection experiments demonstrated strong interference of OP7 virus with IAV replication, manifested by a dramatic decrease in the infectivity of released virions. Moreover, an overproportional quantity of S7 in relation to other genome segments was observed, both intracellularly and in the released virus population. Concurrently, OP7 virions lacked a large fraction of other vRNA segments, which appears to constitute its defect in virus replication. OP7 virus might serve as a promising candidate for antiviral therapy. Furthermore, this novel form of DIP may also be present in other IAV preparations.IMPORTANCE Defective interfering particles (DIPs) typically contain a highly deleted form of the viral genome, rendering them defective in virus replication. Yet upon complementation through coinfection with fully infectious standard virus (STV), interference with the viral life cycle can be observed, leading to suppressed STV replication and the release of mainly noninfectious DIPs. Interestingly, recent research indicates that DIPs may serve as an antiviral agent. Here we report the discovery of a yet-unknown type of influenza A virus-derived DIP (termed "OP7" virus) that contains numerous point mutations instead of large deletions in its genome. Furthermore, the underlying principles that render OP7 virions interfering and apparently defective seem to differ from those of conventional DIPs. In conclusion, we believe that OP7 virus might be a promising candidate for antiviral therapy. Moreover, it exerts strong effects, both on virus replication and on the host cell response, and may have been overlooked in other IAV preparations.


Assuntos
Vírus Defeituosos/genética , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Células A549 , Animais , Antivirais , Cães , Genoma Viral/genética , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Nucleotídeos/genética , RNA Viral/genética , Vírion/genética , Replicação Viral/genética
2.
PLoS Comput Biol ; 15(4): e1006944, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30973879

RESUMO

The best measure to limit spread of contagious diseases caused by influenza A viruses (IAVs) is annual vaccination. The growing global demand for low-cost vaccines requires the establishment of high-yield production processes. One possible option to address this challenge is the engineering of novel vaccine producer cell lines by manipulating gene expression of host cell factors relevant for virus replication. To support detailed characterization of engineered cell lines, we fitted an ordinary differential equation (ODE)-based model of intracellular IAV replication previously established by our group to experimental data obtained from infection studies in human A549 cells. Model predictions indicate that steps of viral RNA synthesis, their regulation and particle assembly and virus budding are promising targets for cell line engineering. The importance of these steps was confirmed in four of five single gene overexpression cell lines (SGOs) that showed small, but reproducible changes in early dynamics of RNA synthesis and virus release. Model-based analysis suggests, however, that overexpression of the selected host cell factors negatively influences specific RNA synthesis rates. Still, virus yield was rescued by an increase in the virus release rate. Based on parameter estimations obtained for SGOs, we predicted that there is a potential benefit associated with overexpressing multiple host cell genes in one cell line, which was validated experimentally. Overall, this model-based study on IAV replication in engineered cell lines provides a step forward in the dynamic and quantitative characterization of IAV-host cell interactions. Furthermore, it suggests targets for gene editing and indicates that overexpression of multiple host cell factors may be beneficial for the design of novel producer cell lines.


Assuntos
Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Vírus da Influenza A/fisiologia , Modelos Biológicos , Replicação Viral/fisiologia , Células A549 , Transporte Ativo do Núcleo Celular , Animais , Biologia Computacional , Simulação por Computador , Cães , Engenharia Genética , Genoma Viral , Humanos , Vírus da Influenza A/genética , Vacinas contra Influenza/biossíntese , Cinética , Células Madin Darby de Rim Canino , Replicação Viral/genética
3.
Biotechnol Bioeng ; 116(10): 2503-2513, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31180133

RESUMO

Interleukin-2 (IL-2) is a potent molecule in cancer therapy. Clinical application, however, is limited due to its strong side effects during the treatment. We developed an IL-2 variant (IL-2v) immunocytokine to circumvent the drawbacks of the current IL-2 therapy. During the production of the IL-2v immunocytokine in Chinese hamster ovary (CHO) cells, molecules with fragmented IL-2v and therefore reduced cytokine activity can be observed. To control product fragmentation different production process conditions were investigated. By shifting temperature or pH after the cell growth phase to lower values, fragmented species can be reduced from 10% to 12% to about 4%. However, with the adopted process conditions, the effective titer is decreased concomitantly. Moreover, fermentation length and inoculation cell density are parameters to adjust fragmentation and effective titer. A suitable method for efficient process optimization is the design of experiment approach. With this procedure, novel optimal values for temperature, pH value, harvest day, and inoculation cell densities were proposed and tested subsequently. In comparison to the former process, the improved process reduces fragmentation by 66% while keeping the effective titer comparable. In summary, these findings will help to control fragmentation in CHO production processes of different IL-2v or IL-2 containing therapeutic proteins.


Assuntos
Técnicas de Cultura de Células , Interleucina-2/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Animais , Células CHO , Cricetulus , Humanos , Interleucina-2/genética , Estabilidade Proteica , Proteínas Recombinantes de Fusão/genética
4.
J Gen Virol ; 97(2): 293-298, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26647282

RESUMO

Activation of the innate immune response represents one of the most important cellular mechanisms to limit virus replication and spread in cell culture. Here, we examined the effect of adenoviral gene expression on the antiviral response in adenovirus-transformed cell lines; HEK293, HEK293SF and AGE1.HN. We demonstrate that the expression of the early region protein 1A in these cell lines impairs their ability to activate antiviral genes by the IFN pathway. This property may help in the isolation of newly emerging viruses and the propagation of interferon-sensitive virus strains.


Assuntos
Adenoviridae/imunologia , Adenoviridae/fisiologia , Evasão da Resposta Imune , Imunidade Inata , Replicação Viral , Linhagem Celular Transformada , Humanos , Proteínas Virais/biossíntese , Proteínas Virais/imunologia
5.
Appl Microbiol Biotechnol ; 100(16): 7181-92, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27129532

RESUMO

Influenza viruses are respiratory pathogens and can cause severe disease. The best protection against influenza is provided by annual vaccination. These vaccines are produced in embryonated chicken eggs or using continuous animal cell lines. The latter processes are more flexible and scalable to meet the growing global demand. However, virus production in cell cultures is more expensive. Hence, further research is needed to make these processes more cost-effective and robust. We studied influenza virus replication dynamics to identify factors that limit the virus yield in adherent Madin-Darby canine kidney (MDCK) cells. The cell cycle stage of MDCK cells had no impact during early infection. Yet, our results showed that the influenza virus RNA synthesis levels out already 4 h post infection at a time when viral genome segments are exported from the nucleus. Nevertheless, virus release occurred at a constant rate in the following 16 h. Thereafter, the production of infectious viruses dramatically decreased, but cells continued to produce particles contributing to the hemagglutination (HA) titer. The majority of these particles from the late phase of infection were deformed or broken virus particles as well as large membranous structures decorated with viral surface proteins. These changes in particle characteristics and morphology need to be considered for the optimization of influenza virus production and vaccine purification steps. Moreover, our data suggest that in order to achieve higher cell-specific yields, a prolonged phase of viral RNA synthesis and/or a more efficient release of influenza virus particles is required.


Assuntos
Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vacinas contra Influenza/biossíntese , Influenza Humana/prevenção & controle , RNA Viral/biossíntese , Cultura de Vírus/métodos , Replicação Viral , Animais , Linhagem Celular , Cães , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Células Madin Darby de Rim Canino
6.
PLoS Comput Biol ; 9(11): e1003372, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24278009

RESUMO

Influenza A viruses are respiratory pathogens that cause seasonal epidemics with up to 500,000 deaths each year. Yet there are currently only two classes of antivirals licensed for treatment and drug-resistant strains are on the rise. A major challenge for the discovery of new anti-influenza agents is the identification of drug targets that efficiently interfere with viral replication. To support this step, we developed a multiscale model of influenza A virus infection which comprises both the intracellular level where the virus synthesizes its proteins, replicates its genome, and assembles new virions and the extracellular level where it spreads to new host cells. This integrated modeling approach recapitulates a wide range of experimental data across both scales including the time course of all three viral RNA species inside an infected cell and the infection dynamics in a cell population. It also allowed us to systematically study how interfering with specific steps of the viral life cycle affects virus production. We find that inhibitors of viral transcription, replication, protein synthesis, nuclear export, and assembly/release are most effective in decreasing virus titers whereas targeting virus entry primarily delays infection. In addition, our results suggest that for some antivirals therapy success strongly depends on the lifespan of infected cells and, thus, on the dynamics of virus-induced apoptosis or the host's immune response. Hence, the proposed model provides a systems-level understanding of influenza A virus infection and therapy as well as an ideal platform to include further levels of complexity toward a comprehensive description of infectious diseases.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas/métodos , Vírus da Influenza A/efeitos dos fármacos , Influenza Humana/virologia , Modelos Biológicos , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/química , Morte Celular , Biologia Computacional , Cães , Espaço Extracelular/virologia , Humanos , Vírus da Influenza A/fisiologia , Espaço Intracelular/virologia , Células Madin Darby de Rim Canino , Internalização do Vírus/efeitos dos fármacos
7.
Appl Microbiol Biotechnol ; 98(21): 8999-9008, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25132064

RESUMO

During the replication of influenza viruses, defective interfering particles (DIPs) can be generated. These are noninfectious deletion mutants that require coinfection with a wild-type virus but interfere with its helper virus replication. Consequently, coinfected cells mainly produce DIPs. Little is known about how such noninfectious virus particles affect the virus yield of cell culture-based influenza vaccine production. We compared infections of Madin-Darby canine kidney cells with two seed virus preparations of the influenza virus strain A/Puerto Rico/8/34 that contain different amounts of DIPs. A combination of conventional RT-PCR, RT-qPCR, and flow cytometry revealed that DI genomes indeed strongly accumulate in coinfected cells and impede the viral RNA synthesis. Additionally, cells infected at the higher DIP concentration showed a stronger antiviral response characterized by increased interferon-ß expression and apoptosis induction. Furthermore, in the presence of DIPs, a significant fraction of cells did not show any productive accumulation of viral proteins at all. Together, these effects of DIPs significantly reduce the virus yield. Therefore, the accumulation of DIPs should be avoided during influenza vaccine production which can be achieved by quality controls of working seed viruses based on conventional RT-PCR. The strategy for the depletion of DIPs presented here can help to make cell culture-based vaccine production more reliable and robust.


Assuntos
Vírus Defeituosos/fisiologia , Interações Hospedeiro-Patógeno , Vírus da Influenza A/imunologia , Vírus da Influenza A/fisiologia , Vacinas contra Influenza/isolamento & purificação , Replicação Viral , Animais , Antivirais/metabolismo , Apoptose , Cães , Citometria de Fluxo , Vírus da Influenza A/crescimento & desenvolvimento , Interferons/metabolismo , Células Madin Darby de Rim Canino , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Carga Viral
8.
J Virol ; 86(15): 7806-17, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22593159

RESUMO

Influenza viruses transcribe and replicate their negative-sense RNA genome inside the nucleus of host cells via three viral RNA species. In the course of an infection, these RNAs show distinct dynamics, suggesting that differential regulation takes place. To investigate this regulation in a systematic way, we developed a mathematical model of influenza virus infection at the level of a single mammalian cell. It accounts for key steps of the viral life cycle, from virus entry to progeny virion release, while focusing in particular on the molecular mechanisms that control viral transcription and replication. We therefore explicitly consider the nuclear export of viral genome copies (vRNPs) and a recent hypothesis proposing that replicative intermediates (cRNA) are stabilized by the viral polymerase complex and the nucleoprotein (NP). Together, both mechanisms allow the model to capture a variety of published data sets at an unprecedented level of detail. Our findings provide theoretical support for an early regulation of replication by cRNA stabilization. However, they also suggest that the matrix protein 1 (M1) controls viral RNA levels in the late phase of infection as part of its role during the nuclear export of viral genome copies. Moreover, simulations show an accumulation of viral proteins and RNA toward the end of infection, indicating that transport processes or budding limits virion release. Thus, our mathematical model provides an ideal platform for a systematic and quantitative evaluation of influenza virus replication and its complex regulation.


Assuntos
Genoma Viral/fisiologia , Vírus da Influenza A/fisiologia , Modelos Biológicos , RNA Viral/biossíntese , Replicação Viral/fisiologia , Humanos
9.
Appl Microbiol Biotechnol ; 93(2): 601-11, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21915610

RESUMO

Trypsin is commonly used in Madin-Darby canine kidney (MDCK) cell culture-based influenza vaccine production to facilitate virus infection by proteolytic activation of viral haemagglutinin, which enables multi-cycle replication. In this study, we were able to demonstrate that trypsin also interferes with pathogen defence mechanisms of host cells. In particular, a trypsin concentration of 5 BAEE U/mL (4.5 µg/mL porcine trypsin) used in vaccine manufacturing strongly inhibited interferon (IFN) signalling by proteolytic degradation of secreted IFN. Consequently, absence of trypsin during infection resulted in a considerably stronger induction of IFN signalling and apoptosis, which significantly reduced virus yields. Under this condition, multi-cycle virus replication in MDCK cells was not prevented but clearly delayed. Therefore, incomplete infection can be ruled out as the reason for the lower virus titres. However, suppression of IFN signalling by overexpression of viral IFN antagonists (influenza virus PR8-NS1, rabies virus phosphoprotein) partially rescued virus titres in the absence of trypsin. In addition, virus yields could be almost restored by using the influenza strain A/WSN/33 in combination with fetal calf serum (FCS). For this strain, FCS enabled trypsin-independent fast propagation of virus infection, probably outrunning cellular defence mechanisms and apoptosis induction in the absence of trypsin. Overall, addition of trypsin provided optimal conditions for high yield vaccine production in MDCK cells by two means. On the one hand, proteolytic degradation of IFN keeps cellular defence at a low level. On the other hand, enhanced virus spreading enables viruses to replicate before the cellular response becomes fully activated.


Assuntos
Vacinas contra Influenza/isolamento & purificação , Orthomyxoviridae/crescimento & desenvolvimento , Orthomyxoviridae/isolamento & purificação , Tripsina/metabolismo , Animais , Antivirais/metabolismo , Biotecnologia/métodos , Técnicas de Cultura de Células , Linhagem Celular , Cães , Interferons/metabolismo , Tecnologia Farmacêutica/métodos
10.
J Gen Virol ; 91(Pt 7): 1754-63, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20357039

RESUMO

Because of their high susceptibility to infection with various influenza virus strains, Madin-Darby canine kidney (MDCK) cells have been widely used as a substrate for influenza virus isolation and vaccine production. However, MDCK cells are also interferon (IFN) competent, and the type I IFN response is commonly thought to be a factor strongly inhibiting virus replication. Therefore, the inhibition of influenza virus replication by IFN signalling was analysed for an adherent MDCK cell line used in vaccine manufacturing. Depending on the respective virus strain, different levels of IFN induction and a corresponding upregulation of the IFN-induced myxovirus resistance protein 1 (Mx1) were observed. Suppression of IFN induction by transient expression of the viral non-structural protein 1 protein enhanced replication of an influenza virus lacking NS1, but not wild-type strains. In agreement with this, stimulation of cells with MDCK cell-derived IFN prior to infection resulted only in a decrease in replication rate, and not in a change of final yields for wild-type influenza viruses. This lack of IFN-induced antiviral activity correlated with missing anti-influenza activity of MDCK Mx proteins. No inhibitory effect on viral polymerase activity was found for canine Mx1 (cMx1) and cMx2 in minireplicon assays. In conclusion, in MDCK cells, IFN expression is not a limiting factor for influenza virus replication and this might partially be caused by a lack of anti-influenza activity of canine Mx proteins.


Assuntos
Vírus da Influenza A/fisiologia , Interferons/metabolismo , Replicação Viral/fisiologia , Animais , Linhagem Celular , Cães , Regulação Viral da Expressão Gênica , Humanos , Rim/citologia , Transdução de Sinais , Proteínas não Estruturais Virais
11.
Biochim Biophys Acta ; 1779(2): 139-44, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18082154

RESUMO

The neuregulin-1 (Nrg1) gene encodes for a group of growth factors with multiple functions during mammalian development. Overexpression of Nrg1 is found in many different cancer types and correlates with cancer progression and an aggressive phenotype. In this study we identified the promoter of Nrg1 type I isoforms. Reporter gene assays revealed that 850 bp upstream from the translation initiation codon are necessary for high transcriptional activity in murine Neuro-2A neuroblastoma cells. The core promoter is highly conserved among mammals, has multiple transcription start sites and is located in a CpG island. The conserved promoter region contains GC-and GT-box elements and overexpression of Sp1 increased promoter activity, while ZBP-89 decreased the activity. Overexpression of the NF-kappaB subunit p65 (RelA) led to a strong activation of the promoter mediated through a single NF-kappaB binding site. Reflecting that the transcriptional activity of NF-kappaB and Sp1 are increased upon Nrg-stimulation in breast cancer cells this study suggests a potential mechanism of a positive feedback loop/autoregulation of neuregulin.


Assuntos
NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas , Ativação Transcricional , Animais , Sequência de Bases , Sequência Conservada , Cães , Genes Reporter , Humanos , Camundongos , Dados de Sequência Molecular , NF-kappa B/genética , Neuregulina-1 , Isoformas de Proteínas/genética , Ratos , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Sítio de Iniciação de Transcrição
12.
Virus Res ; 218: 86-95, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27208847

RESUMO

Like many other viral pathogens, influenza A viruses can form defective interfering particles (DIPs). These particles carry a large internal deletion in at least one of their genome segments. Thus, their replication depends on the co-infection of cells by standard viruses (STVs), which supply the viral protein(s) encoded by the defective segment. However, DIPs also interfere with STV replication at the molecular level and, despite considerable research efforts, the mechanism of this interference remains largely elusive. Here, we present a mechanistic mathematical model for the intracellular replication of DIPs. In this model, we account for the common hypothesis that defective interfering RNAs (DI RNAs) possess a replication advantage over full-length (FL) RNAs due to their reduced length. By this means, the model captures experimental data from yield reduction assays and from studies testing different co-infection timings. In addition, our model predicts that one important aspect of interference is the competition for viral proteins, namely the heterotrimeric viral RNA-dependent RNA polymerase (RdRp) and the viral nucleoprotein (NP), which are needed for encapsidation of naked viral RNA. Moreover, we find that there may be an optimum for both the DI RNA synthesis rate and the time point of successive co-infection of a cell by DIPs and STVs. Comparing simulations for the growth of DIPs with a deletion in different genome segments suggests that DI RNAs derived from segments which encode for the polymerase subunits are more competitive than others. Overall, our model, thus, helps to elucidate the interference mechanism of DI RNAs and provides a novel hypothesis why DI RNAs derived from the polymerase-encoding segments are more abundant in DIP preparations.

13.
Virus Res ; 213: 90-99, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26592173

RESUMO

Like many other viral pathogens, influenza A viruses can form defective interfering particles (DIPs). These particles carry a large internal deletion in at least one of their genome segments. Thus, their replication depends on the co-infection of cells by standard viruses (STVs), which supply the viral protein(s) encoded by the defective segment. However, DIPs also interfere with STV replication at the molecular level and, despite considerable research efforts, the mechanism of this interference remains largely elusive. Here, we present a mechanistic mathematical model for the intracellular replication of DIPs. In this model, we account for the common hypothesis that defective interfering RNAs (DI RNAs) possess a replication advantage over full-length (FL) RNAs due to their reduced length. By this means, the model captures experimental data from yield reduction assays and from studies testing different co-infection timings. In addition, our model predicts that one important aspect of interference is the competition for viral proteins, namely the heterotrimeric viral RNA-dependent RNA polymerase (RdRp) and the viral nucleoprotein (NP), which are needed for encapsidation of naked viral RNA. Moreover, we find that there may be an optimum for both the DI RNA synthesis rate and the time point of successive co-infection of a cell by DIPs and STVs. Comparing simulations for the growth of DIPs with a deletion in different genome segments suggests that DI RNAs derived from segments which encode for the polymerase subunits are more competitive than others. Overall, our model, thus, helps to elucidate the interference mechanism of DI RNAs and provides a novel hypothesis why DI RNAs derived from the polymerase-encoding segments are more abundant in DIP preparations.


Assuntos
Vírus Defeituosos/crescimento & desenvolvimento , Vírus da Influenza A/crescimento & desenvolvimento , RNA Viral/genética , RNA Viral/metabolismo , Replicação Viral , Vírus Defeituosos/genética , Vírus da Influenza A/genética , Modelos Teóricos
14.
Biotechnol J ; 10(5): 681-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25728309

RESUMO

Defective interfering particles (DIPs) have been found for many important viral pathogens and it is believed that most viruses generate DIPs. This article reviews the current knowledge of the generation and amplification of DIPs, which possess deletions in the viral genome but retain the ability to replicate in the presence of a complete helper virus. In addition, mechanisms are discussed by which DIPs interfere with the replication of their helper virus leading to the production of mainly progeny DIPs by coinfected cells. Even though DIPs cannot replicate on their own, they are biologically active and it is well known that they have a huge impact on virus replication, evolution, and pathogenesis. Moreover, defective genomes are potent inducers of the innate immune response. Yet, little attention has been paid to DIPs in recent years and their impact on biotechnological products such as vaccines and viral vectors remains elusive in most cases. With a focus on influenza virus, this review demonstrates that DIPs are important for basic research on viruses and for the production of viral vaccines and vectors. Reducing the generation and/or amplification of DIPs ensures reproducible results as well as high yields and consistent product quality in virus production.


Assuntos
Vírus Auxiliares/fisiologia , Vírus Satélites/fisiologia , Animais , Biotecnologia/métodos , Vetores Genéticos/fisiologia , Humanos , Vacinas Virais , Replicação Viral
15.
Nat Commun ; 6: 8938, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26586423

RESUMO

Biochemical reactions are subject to stochastic fluctuations that can give rise to cell-to-cell variability. Yet, how this variability affects viral infections, which themselves involve noisy reactions, remains largely elusive. Here we present single-cell experiments and stochastic simulations that reveal a large heterogeneity between influenza A virus (IAV)-infected cells. In particular, experimental data show that progeny virus titres range from 1 to 970 plaque-forming units and intracellular viral RNA (vRNA) levels span three orders of magnitude. Moreover, the segmentation of IAV genomes seems to increase the susceptibility of their replication to noise, since the level of different genome segments can vary substantially within a cell. In addition, simulations suggest that the abortion of virus entry and random degradation of vRNAs can result in a large fraction of non-productive cells after single-hit infection. These results challenge current beliefs that cell population measurements and deterministic simulations are an accurate representation of viral infections.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/fisiopatologia , Animais , Linhagem Celular , Sobrevivência Celular , Humanos , Vírus da Influenza A/química , Vírus da Influenza A/genética , Influenza Humana/virologia , Cinética , Modelos Teóricos , Análise de Célula Única
16.
PLoS One ; 8(9): e72288, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24039749

RESUMO

Influenza viruses are a major public health burden during seasonal epidemics and a continuous threat due to their potential to cause pandemics. Annual vaccination provides the best protection against the contagious respiratory illness caused by influenza viruses. However, the current production capacities for influenza vaccines are insufficient to meet the increasing demands. We explored the possibility to establish a continuous production process for influenza viruses using the duck-derived suspension cell line AGE1.CR. A two-stage bioreactor setup was designed in which cells were cultivated in a first stirred tank reactor where an almost constant cell concentration was maintained. Cells were then constantly fed to a second bioreactor where virus infection and replication took place. Using this two-stage reactor system, it was possible to continuously produce influenza viruses. Surprisingly, virus titers showed a periodic increase and decrease during the run-time of 17 days. These titer fluctuations were caused by the presence of defective interfering particles (DIPs), which we detected by PCR. Mathematical modeling confirmed this observation showing that constant virus titers can only emerge in the absence of DIPs. Even with very low amounts of DIPs in the seed virus and very low rates for de novo DIP generation, defective viruses rapidly accumulate and, therefore, represent a serious challenge for continuous vaccine production. Yet, the continuous replication of influenza virus using a two-stage bioreactor setup is a novel tool to study aspects of viral evolution and the impact of DIPs.


Assuntos
Vírus Defeituosos/genética , Vírus da Influenza A Subtipo H1N1/genética , Animais , Reatores Biológicos , Linhagem Celular , Vírus Defeituosos/fisiologia , Patos , Genoma Viral , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Vacinas contra Influenza , Modelos Biológicos , Carga Viral , Cultura de Vírus , Replicação Viral
17.
Vaccine ; 29(41): 7125-9, 2011 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-21651940

RESUMO

Influenza B virus infections are mainly restricted to humans, which is partially caused by the inability of influenza B virus NS1 protein to counteract the innate immune response of other species. However, for cell culture-based influenza vaccine production non-human cells, such as Madin-Darby canine kidney (MDCK) cells, are commonly used. Therefore, the impact of cellular pathogen defence mechanisms on influenza B virus propagation in MDCK cells was analysed in this study. Activation of the cellular antiviral defence by interferon stimulation slowed down influenza B virus replication at early time points but after 48h the same virus titres were reached in stimulated and control cells. Furthermore, suppression of the antiviral host defence by transient expression of a viral antagonist, the rabies virus phosphoprotein, could not increase influenza B virus replication. Finally, canine Myxovirus resistance (Mx) proteins showed no antiviral activity in an influenza B virus-specific minireplicon assay in contrast to the murine Mx1 protein. Taken together, these results indicate that an insufficient antiviral defence in MDCK cells promotes efficient influenza B virus replication favouring the use of MDCK cells in influenza vaccine production.


Assuntos
Vírus da Influenza B/crescimento & desenvolvimento , Vírus da Influenza B/imunologia , Interferons/imunologia , Animais , Técnicas de Cultura de Células/métodos , Linhagem Celular , Cães
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA