• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 141
  • 28
  • 13
  • 11
  • 7
  • 7
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 257
  • 257
  • 47
  • 38
  • 32
  • 28
  • 26
  • 26
  • 25
  • 23
  • 22
  • 20
  • 18
  • 17
  • 17
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
91

Innate immune responses and signaling pathways in influenza A (H5N1) infected human primary macrophages

Hui, Pui-yan., 許珮茵. January 2008 (has links)
published_or_final_version / Microbiology / Doctoral / Doctor of Philosophy
92

The role of the interaction of the influenza B virus NS1 protein with the cellular Brd2 protein

Park, Jang Won 22 October 2009 (has links)
Influenza B virus is a major human pathogen causing highly contagious respiratory disease. It accounts for approximately ~30% of influenza virus infection per year. The effector domain of the NS1 protein of influenza B virus (NS1B protein), encompassing the carboxy terminal two thirds of the protein, suppresses interferon-β (IFN-β) synthesis in virus-infected cells by unknown mechanism(s). The induced IFN-β mediates innate immunity. To elucidate the mechanism by which the NS1B effector domain suppresses the production of IFN-β, we identified cellular proteins that interact with the NS1B effector domain. Two approaches were used. The approach that succeeded employed the transfection into cells of plasmids expressing the NS1B effector domain containing two affinity tags. After double affinity purification, co-purified cellular proteins were identified by mass spectrometry. We identified Brd2 as a cellular protein that interacts with the NS1B protein. We established that Brd2 specifically binds to the NS1B effector domain in vitro, in vivo, and in virus-infected cells. Serial mutagenesis experiments showed the phenylalanine at position 171 (F171) of the NS1B protein is essential for Brd2 binding. To determine the function of the interaction of Brd2 with the NS1B protein, we generated a recombinant virus encoding an NS1B protein in which F at position 171 was replaced by an alanine. The F171A mutant virus was attenuated, and unlike the wild-type virus, induced the synthesis of IFN-β mRNA. IRF3, a key transcription factor for transcription of the IFN-β gene, was activated in mutant virusinfected cells, but not in wild-type virus-infected cells. Transfection assays implicated the activation of the TBK1 kinase as the step in IRF3 activation that is induced in mutant virus-infected cells. We interpreted these results as showing that Brd2 binding to the NS1B protein is required for suppressing IRF3 activation and IFN-β induction. Attempts at further confirmation by depletion of endogenous Brd2 using RNA interference were not successful because of inefficient knock-down efficiency and nonspecific IFN-β induction. A further complication is that another bromodomain protein, Brd4, interacts with the NS1B protein and could compensate for depletion of Brd2. / text
93

Effects of PB1-F2 and PA-X on the pathogenicity of H1N1 influenza virus

Lee, Jinhwa January 1900 (has links)
Doctor of Philosophy / Department of Diagnostic Medicine/Pathobiology / Wenjun Ma / Influenza A virus (IAV) is a negative sense, single-stranded, segmented RNA virus with eight gene segments. It is an important respiratory pathogen which causes annual epidemics and occasional pandemics worldwide in humans and leads to considerable economic problems for the livestock industry. To control and prevent this significant disease, understanding the pathogenesis of IAVs is critical. Although some molecular mechanisms regarding virulence have been determined, IAV pathogenesis is not completely understood and is difficult to predict. The eight viral gene segments of IAV were thought to encode for 10 viral proteins. Since 2001, eight additional viral proteins have been identified, including PB1-F2, PB1-N40, PA-X, NS3, PA-N155, PA-N182, M42, and PB2-S1. However, the functions of these novel proteins in influenza virus replication as well as pathogenesis have not been fully elucidated. Although PB1-F2 protein is an important virulence factor of IAV, the effects of this protein on viral pathogenicity of swine influenza virus (SIV) remain unclear. In Chapter 2, we investigated the contribution of the PB1-F2 protein to viral pathogenicity of a virulent triple-reassortant (TR) H1N1 SIV in different hosts, pigs and mice. Our data indicate that PB1-F2 expression in virulent TR H1N1 SIV modulates virus replication and pathogenicity in the natural host, pigs, but not in mice. In addition, single amino acid (aa) substitution at position 66 (N/S) in the PB1-F2 has a critical role in virulence in mice but no effect was found in pigs. A novel IAV protein, PA-X consists of the N-terminal 191aa of PA protein and a unique C-terminal 41 (truncated form) or 61 (full-length form) aa residues encoded by +1 ribosomal frameshifting. Although several studies have demonstrated the PA-X protein as an important immune modulator and virulence factor, the impact of different expressions of PA-X protein including full-length, truncated or PA-X deficient forms on viral pathogenicity and host response remains unclear. In Chapter 3, we showed that expression of either truncated or full-length PA-X protein in 2009 human pandemic H1N1 (pH1N1) viruses suppresses host antiviral response by host shutoff activity which promotes viral growth and virulence in mice when compared to loss of PA-X expression. Furthermore, full-length PA-X expression displayed stronger impact on viral pathogenicity and host immune response compared to truncated PA-X expression. Taken together, our results provide new insights into the impact of PB1-F2 and PA-X proteins on virus replication, pathogenicity and modulation of host immune responses. This knowledge is important for better understanding of IAV pathogenesis.
94

Die Rolle des Transkriptionsfaktors NF-KappaB in Influenza-A-Virus infizierten Zellen / The role of the transcriptionfactor NF-kB in influenza-A-virus infected cells

Wurzer, Walter January 2003 (has links) (PDF)
Die Aktivierung von Transkriptionsfaktors NF-kB ist ein Charakteristikum viraler Infektionen, einschließlich der Infektion durch Influenza-A-Viren (Hiscott J. et al., 2001). Da die Expression vieler proinflammatorischer und antiviraler Zytokine, wie IFNb oder TNF-a durch NF-kB kontrolliert wird, hat sich ein Konzept entwickelt, welches besagt, dass NF-kB und sein übergeordneter Aktivator IKK wichtige Bestandteile der angeborenen, antiviralen Immunität im Kontext einer Infektion mit RNA-Viren sind (Chu WM. Et al., 1999). Im Gegensatz zu dieser weithin akzeptierten Ansicht, wurde in der hier vorliegenden Arbeit gezeigt, dass die Aktivierung von NF-kB für eine effiziente Influenzareplikation von großer Wichtigkeit ist. Auf einer molekularen Ebene wurde dies durch die NF-kB-abhängige virale Aktivierung des proapoptotischen Faktors TRAIL gezeigt, welcher die Virusvermehrung sowohl auto- als auch parakrin erhöht. Somit kann man sagen, dass NF-kB im Kontext einer Influenza-A-Virusinfektion sowohl proapoptotisch als auch proviral wirkt. Die Induktion der Apoptose ist ein weiteres, charakteristisches Merkmal, das man im Zusammenhang mit Virusinfektionen beobachten kann. Da die Rolle der Apoptose während einer Influenza-A-Virusinfektion noch unklar war, wurde diese Frage adressiert. Dabei wurde versucht mit einem wichtigen, virus-induzierten Apoptose-Effektor, nämlich Kaspase-3 zu interferieren. Überraschenderweise wurde die Influenzavermehrung in Anwesenheit eines Kaspase-3-Inhibitors stark negativ beeinflusst. Im Einklang mit diesem Befund konnte gezeigt werden, dass die Virustiter in Zellen, in denen XIAP überexprimiert wurde, rückläufig waren. Gegengleich führte Überexpression von Prokaspase-3 zu einem Titeranstieg. Mechanistisch scheint der Blockade der Virusvermehrung eine Retention der viralen RNP-Komplexe im Zellkern zu Grunde zu liegen, die die Bildung von reifen Viruspartikel verhindert. Die Erklärung dürfte in der Aktivität von Kaspase-3 zu finden sein, die an dem Abbau von Kernporenkomplexproteinen in apoptotischen Zellen beteiligt ist und was in Folge die freie Diffusion viraler RNPs ermöglichen dürfte. Abschließend entwickelte sich aufgrund der vorliegenden Arbeit eine neue Hypothese über die Rolle des IKK-NF-kB-Signalweges, seinen Einfluss auf die Apoptoseregulation in Influenza-infizierten Zellen und der Auswirkung auf das Virus. / Activation of the transcription factor NF-kB is a hallmark of infections by viral pathogens including influenza-A-viruses (Hiscott J. et al., 2001). Since gene expression of many proinflammatory and antiviral cytokines, such as IFNb of TNF-a is controlled by NF-kB the concept emerged that NF-kB and its upstream regulator IKK are essential components of the innate antiviral immune response to infections with RNA viruses (Chu WM. et al., 1999). In contrast to this common view this work presents data that for efficient influenza virus production NF-kB activity is required. On a molecular level this is due to NF-kB-dependent viral induction of the proapoptotic factor TRAIL which enhances virus propagation in an auto- and parakrine fashion. Thus, NF-kB acts both proapoptotic and proviral in the context of an influenza virus infection. Induction of apoptosis is a hallmark observed upon infection with many viral pathogens, including influenza-A-virus. Since the consequences of apoptosis induction for the outcome of an influenza virus infection are not clear, we have addressed this issue by interfering with the function of a major virus-induced apoptosis effector, caspase-3. Surprisingly, influenza virus propagation was strongly impaired in the presence of a caspase-3 inhibitor in cultures cells. Consistent with these findings, virus yields are reduced in cells expressing XIAP and enhanced when procaspase-3 is overexpressed. Mechanistically, the block in virus propagation appears to be due to retention of the viral RNP complexes in the nucleus preventing formation of progeny virus particles. An explanation might given by an effect of caspase-3, which is involved in cleavage of protein of the nuclear pore complex in cells undergoing apoptosis, which results in fusion of the pores and might thus allow free diffusion of viral RNA complexes. Finally, this work presented here led to a new hypothesis concerning the role of the IKK-NF-kB-pathway, its influence on the regulation of apoptosis in influenza infected cells and the outcome for the virus.
95

The role of the NS segment of Influenza A virus in setting host range and pathogenicity

Turnbull, Matthew Luke January 2017 (has links)
Influenza A virus (IAV) circulates in waterfowl, causing mostly asymptomatic infections. IAV can undergo host adaptation and evolve to cause significant disease and mortality in domestic poultry and mammals, applying an enormous socio-economic burden on society. Sporadically, IAV causes global pandemics in man due to its zoonotic nature, and this can result in millions of deaths worldwide during a single outbreak. Host adaptation of IAV is an incompletely understood phenomenon, but is known to involve both host and viral determinants. It is essential to improve the understanding of the factors governing host range and pathogenicity of avian IAV, especially given the absence of a universal influenza vaccine and a limited weaponry of effective antiviral compounds. This study set out to improve the understanding of host adaptation of avian IAV, focussing on segment 8 (NS segment) of the virus genome. The NS segment of non-chiropteran IAV circulates as two phylogenetically distinct clades – the ‘A-’ and ‘B-alleles’. The A-allele is found in avian and mammalian viruses, but the B-allele is considered to be almost exclusively avian. This might result from one or both of the major NS gene products (NS1 and NEP) being non-functional in mammalian host cells, or from an inability of segment 8 RNA to package into mammalian-adapted strains. To investigate this, the NS segments from a panel of avian A- and B-allele strains were introduced into human H1N1 and H3N2 viruses by reverse genetics. A- and B-allele reassortant viruses replicated equally well in a variety of mammalian cell types in vitro. Surprisingly, the consensus B-allele segment 8 out-competed an A-allele counterpart when reassortant H1N1 viruses were co-infected, with the parental WT segment 8 being most fit in this system. A- and B-allele NS1 proteins were equally efficient at blocking the mammalian IFN response both in the context of viral infection and in transfection-based reporter assays. Consensus A- and B-allele H1N1 viruses also caused disease in mice and replicated to high virus titre in the lung. Interestingly, the B-allele virus induced more weight-loss than the A-allele, although the parental WT virus was most pathogenic in vivo. To re-address the hypothesis that B-allele NS genes really are avian-restricted, the relative rates of independent Aves to Mammalia incursion events of A- and B-allele lineage IAV strains was estimated and compared using phylogenetic analyses of all publically available segment 8 sequences. 32 A-allele introduction events were estimated compared to 6 B-allele incursions, however the total number of avian Aallele sequences outnumbered B-allele sequences by over 3.5 to 1, and the relative rates of introduction were not significantly different across the two lineages suggesting no bias against avian B-allele NS segments entering mammalian hosts in nature. Therefore, this study provides evidence that avian B-allele NS genes are not attenuating in mammalian hosts and are able to cause severe disease. Thus, this lineage of IAV genes, previously assumed to be avian-restricted, should be considered when assessing zoonotic potential and pandemic risk of circulating avian IAVs.
96

Targeting cellular nuclear export to inhibit influenza A virus replication

Dewar, Rebecca Amy January 2018 (has links)
Influenza A virus (IAV) is a global health threat, causing seasonal epidemics and potential pandemics leading to morbidity, death and economic losses. Currently, there are two main classes of licensed antivirals against IAV available in the US and Europe; adamantanes and neuraminidase inhibitors, both of which are hindered by the generation of resistant virus variants. The viral polymerase has a high error rate leading to mutations that allow the virus to overcome selection pressures directed at its own genome from conventional antivirals. The prospect of inhibiting host proteins that the virus exploits to facilitate its replication is of increasing interest as an antiviral strategy as the emergence of resistance has been predicted to be slower when targeting a host cellular factor. IAV utilizes the host nuclear export protein CRM1 to transport viral ribonucleoproteins (vRNPs) from the nucleus to the cytoplasm of an infected cell, a critical late stage of the influenza lifecycle. Leptomycin B (LMB), a Streptomyces metabolite, has been previously shown to target this pathway, resulting in reduced viral propagation; however, LMB's potent cytotoxicity has limited its use as a therapeutic agent. This thesis examined two novel selective inhibitors of nuclear export (SINE), KPT-335 and KPT-185, with less cytotoxicity. In vitro, KPT-335 inhibited replication of human and animal IAV strains in a dose-dependent manner with minimal cytotoxicity. To assess the resistance potential of KPT-335, IAV viruses were serially passaged in the presence of a sub-optimal concentration of the compound and assayed for the development of resistance. Resistance to KPT-335 became evident at 8-10 rounds of passage. Sequencing analysis of independently derived resistant virus clones identified 4 single amino acid changes on a surface exposed patch of the viral nucleoprotein (NP). Introduction of these amino acid changes, into otherwise wild type viruses by reverse genetics, confirmed that changes Q311R and N309T conferred a drug-resistant phenotype. However, these substitutions came at a fitness cost to virus replication. The molecular basis for resistance was unclear but Q311R and N309T NP-mutant viruses produced increased levels of M1 during infection as well as producing virus particles with increased M1:NP ratios. Furthermore, the KPT-335-resistance mutations were surprisingly similar to NP sequence polymorphisms previously associated with susceptibility to the innate defence protein MxA. Consistent with this, viruses harbouring the Q311R mutation displayed increased susceptibility to MxA inhibition compared to wild-type virus. Altogether this study confirms that SINEs have the potential to be successful therapeutic agents against IAV replication and that although resistance could be generated, it may be difficult for the virus to overcome both drug selection pressures and the human innate immune response restrictions by escape mutations.
97

JMJD6 dioxygenase regulates macrophage host responses and is a proviral host factor for vaccinia and influenza A virus growth

Kwok, Chi Ting Janice January 2018 (has links)
Jumonji C (JmjC) domain containing proteins comprise a large family of enzymes that catalyse oxidative reactions. The jumonji domain containing protein 6 (JMJD6) has pleiotropic functions as a lysyl hydroxylase and arginyl demethylase. Previous studies have shown that Jmjd6 is involved in histone modification, mRNA splicing and regulation of polymerase II pause release. A constitutive knockout of Jmjd6 in mice is neonatal lethal and shows defects in macrophage host responses. Recently, JMJD6 was shown to support Foot-and-mouth disease virus replication through interactions with the dead-box RNA helicase Dhx9. This PhD thesis aims to further explore functions of Jmjd6 in macrophages and its roles during viral infections. The hypothesis is that through interactions with RNA helicases, Jmjd6 regulates host responses to foreign nucleic acids and/or has functions as a host factor for replication of DNA and/or RNA viruses. Testing of this hypothesis required the generation of Jmjd6-deficient cell recourses. A new conditional Jmjd6 mouse allele was characterised and a method optimised to knockout the gene in bone marrow-derived macrophages (BMDM) using TAT-Cre recombinase. To study vaccinia (VACV) and influenza A virus (IAV) infections in human cell lines, JMJD6 was depleted using RNA interference or CRISPR-Cas9 gene editing. In BMDM, JMJD6 expression was up-regulated in the late phase of lipopolysaccharide stimulation. The nuclear expression pattern of Jmjd6 in BMDM overlapped with that of DDX41 but not with DHX9, two RNA helicases that have been implicated in sensing of viral DNA and RNA, respectively. Deletion of Jmjd6 in BMDM reduced induction of type I interferon response genes after stimulation with synthetic analogs of viral RNA. To characterise the role of JMJD6 during infection with a DNA virus, Jmjd6-deficient cells were infected with VACV. Knockout of Jmjd6 reduced VACV growth in macrophages but not in HeLa cells. In contrast to HeLa cells, Jmjd6-deficient macrophages displayed abnormal localisations of viral factories and increased cell death, showing that Jmjd6 is specifically required for productive VACV infection in macrophages. To further analyse whether Jmjd6 has pro- or anti-viral functions during RNA virus infection, JMJD6 depleted A549 cells were infected with IAV. JMJD6 depletion in A549 drastically reduced IAV growth from an early stage of infection. Preliminary data indicate that this phenotype is related to a defect in nuclear import of IAV ribonucleoprotein complexes. In summary, this work has identified JMJD6 as a novel pro-viral host factor for VACV and IAV infection and has underpinned its importance for macrophage functions.
98

Untersuchung Influenza Virus-induzierter Signalprozesse und deren Bedeutung in der Wirtszell-Abwehr / Investigation of Influenza virus induced signal-transduction processes and their role in host defense

Ehrhardt, Christina January 2002 (has links) (PDF)
Eine Influenza A Virus Infektion induziert die Expression zahlreicher Gene, einschließlich der TypI Interferone, die eine erste Abwehrlinie gegen virale Infektionen bilden. Hierbei ist IFNb das wichtigste Zytokin. IFNb wird durch einen multimeren Komplex, das Enhanceosom kontrolliert, das Bindungsstellen für die Transkriptionsfaktoren AP-1, NF-kB und IRF-3 in seiner Promotorsequenz besitzt. In früheren Arbeiten konnten wir zeigen, dass die Influenza Virus-induzierte AP-1 abhängige Genexpression über den JNK/SAPK-Signalweg erfolgt (Ehrhardt, 1999; Ludwig et al., 2001). Unter den, an DNA Elemente bindenden AP-1 Faktoren waren solche, die aufgrund von Phosphorylierung durch die JNKs reguliert werden, wie beispielsweise ATF-2. Weiterhin korrelierte die Induktion der AP-1 abhängigen Genexpression mit der starken Aktivierung von JNK und seiner upstream Regulatoren in permissiven Zellen. Die Virusmengen transfizierter und infizierter Zellen, in denen JNK inhibiert wurde, waren höher im Vergleich zu Virusmengen der Kontrollzellen. Demzufolge kann die Virus-induzierte Aktivierung von JNK und AP-1 nicht der Virusreplikation dienen, sondern gehört vielmehr zu einer antiviralen Immunantwort. Daten aus einem Virus-freien, auf Plasmiden basierenden vRNA Replikations-System deuten darauf hin, dass die JNK Aktivierung aus der Akkumulation viraler RNA resultiert. Entsprechend bewirkte die Infektion von Zellen mit einem Virus, dem das virale NS1 Protein fehlt, welches RNA binden und somit "wegfangen" kann, eine gesteigerte JNK Aktivität im Vergleich zu den Kontroll-Infektionen. Damit konnte das NS1 Protein als erstes virales Protein identifiziert werden, das der Virus- und dsRNA-induzierten Aktivierung des JNK/SAPK-Signalweges entgegen wirkt. Der Transkriptionsfaktor IRF-3 wird spezifisch infolge einer viralen Infektion aktiviert und ist daher ein potenter Kandidat, die schnelle und starke antivirale Genexpression zu regulieren. Infolge einer Influenza Virus Infektion wird IRF-3 phosphoryliert, wandert in den Kern und bindet dort an Promotoren, die die antivirale Genexpression steuern. Bislang sind die IRF-3 Kinase und zelluläre Signalwege, die eine IRF-3 Phosphorylierunge induzieren, unbekannt. Um in unserem Labor Signalmediatoren, die upstream von IRF-3 liegen, zu suchen, wurde ein IRF-3 responsives Promotor-Reportergen-Plasmid, aus dem IFNb Promotor stammend, konstruiert. Die kleine Rho-GTPase Rac1 wurde als erster nicht an RNA bindender, zellulärer Mediator identifiziert, der in die Influenza Virus-induzierte IRF-3 abhängige Genexpression involviert ist. Die Inaktivierung der Rho-GTPasen durch das spezifische Inhibitor Toxin B oder dominant negatives Rac1 resultierten in der Inhibierung der Virus- und dsRNA-induzierten IRF-3 Phosphorylierung und DNA Bindung, sowie der IRF-3 abhängigen Promotoraktivität, beispielsweise des IFNb Promotors. Damit konnten zwei wichtige Komponenten der Virus-induzierten Immunantwort identifiziert und charakterisiert werden. / Infection of cells with Influenza A virus induces the expression of a variety of genes, including the type I interferons which are a first line of defense against viral infections. IFNb, the most important cytokine, is controlled by a higher order complex, the enhanceosom, which contains binding sites for the transcription factors AP-1, NF-kB and IRF-3. We could show that the Influenza Virus induced AP-1 dependent gene expression occurs via the JNK/SAPK pathway (Ehrhardt, 1999; Ludwig et al., 2001). Among the AP-1 factors which were identified to bind their cognate DNA element during viral infection are those, that are regulated via phosphorylation by JNKs, such as ATF-2. Accordingly, the induction of AP-1 dependent gene expression correlates with a strong activation of JNK and its upstream activators MKK4 and 7 in permissive cells. Virus yields from transfected and infected cells in which JNK signaling was inhibited by different approaches were higher compared to the levels from control cells. Therefore we conclude that virus-induced activation of JNK and AP-1 is not exploited by the virus to support its replication but rather is required for the innate antiviral immune response. Data obtained with a virus-free plasmid-based vRNA replication system indicated that JNK activation is a cause of viral RNA accumulation during infection. This was supported by the observation, that infection of cells with a virus lacking viral NS1 protein, which is known to bind and to sequester RNA from cellular signaling intermediates, caused a strongly enhanced JNK activity compared to control infections. Furthermore, the NS1 protein was identified as the first viral protein that antagonizes virus- and dsRNA-induced activation of the stress response signaling pathway mediated through Jun N-terminal kinase. IRF-3 is specificially activated in response to viral infection and is therefore the most potent candidate to regulate the fast and strong antiviral gene expression. After an Influenza virus infection IRF-3 becomes phosphorylated and migrates to the nucleus where it binds to antiviral gene promoters. However, the IRF-3 kinase and the cellular signaling pathways leading to IRF-3 phosphorylation are unknown. To investigate signaling mediators upstream of IRF-3, we have constructed an IRF-3 responsive promoter-reporter gene plasmid derived from the IFNb promoter. The small Rho-GTPase Rac1 was identified as the first non-RNA binding cellular mediator involved in the Influenza virus-induced IRF-3 dependent gene expression. Inactivation of these Rho GTPases by the specific inhibitor toxin B or dominant negative Rac1 resulted in the inhibition of virus- and dsRNA-induced IRF-3 phosphorylation and DNA binding as well as of IRF-3 dependent promoter activity, e.g. of the IFNb promoter. Thus two important components of virus-mediated immune response were identified and characterised.
99

Alternative Approaches In The Preparation And Growth Of Influenza B Vaccine Viruses

Audsley, Jennifer M, jennifer.audsley@med.monash.edu.au January 2008 (has links)
Influenza B viruses are a significant cause of disease and influenza B antigens are present in all human vaccines. Achieving suitable yields of seed viruses is often difficult for vaccine manufacturers. With influenza A viruses increases in yields have been achieved by the preparation of reassortants between a high-yielding donor strain and an epidemic strain. However, reassortment of influenza B viruses for the preparation of seeds has not been usually undertaken due to the lack suitable donor strains. Such an approach, which formed the basis of this thesis, could improve vaccine yields, lower costs and introduce a further element of predictability to vaccine manufacture. Potential donor strains were prepared from B/Lee/40 (B/Lee) by two approaches involving the selection of stable cold- and high- temperature mutants. Initial passaging was undertaken in specific-pathogen-free (SPF) chicken embryo kidney (CEK) cultures and later passage in SPF embryonated chicken eggs. Both approaches were successful, although a smaller number of viable progeny could be isolated from plaques obtained at 38„aC. Potential donor strains, isolated by selection at either 25 or 38„aC and plaque-purified in SPF CEK cultures, were tested for haemagglutinin and infectious titre, in comparison with the original parental strain by three methods, and for differences in antigenicity by cross-haemagglutination-inhibition tests. Potential donor strains selected at temperatures of 25„aC (C25) and 38„aC (H38) produced haemagglutination titres of 320 units/50ƒÝL and infectivities of 8.57 and 8.39 50% egg infectious doses, respectively, when grown in eggs at the permissive temperature (34„aC). Reassorting experiments using the B/Lee-derived potential donor strains C25 and H38 and the epidemic strain, B/Johannesburg/5/99 (B/Johannesburg), showed that the preparation of reassortant progeny with both epidemic strain HA and NA was difficult. Only 1/24 of the resulting reassortants possessed both the HA and NA of the epidemic strain. None of the reassortant progeny produced in reassorting experiments using C25 and H38 and the epidemic strain B/Panama/45/90 (B/Panama) possessed the desired 6:2 gene constellation (i.e. genes for the two surface antigens of the epidemic strain and the remainder from the donor strain). The infectious titre of selected progeny from the reassortment experiments were determined by three methods and compared with their respective epidemic parents. Yields of several influenza B epidemic strains and potential donor strains were measured after growth in Madin-Darby canine kidney (MDCK) cells prepared in serum-containing (SC) and animal- and human-derived protein-free (AHPF) media. Optimal multiplicities of infection were determined for B/Panama, B/Johannesburg and C25 in MDCK cultures grown in SC medium. A series of experiments were then undertaken to determine the maximum virus yields in MDCK cells grown in SC medium, followed by a further experiment using C25, B/Panama, B/Johannesburg, and selected reassortants after preparation in AHPF medium. Cell culture yields from 5/6 viruses grown in MDCK cells prepared in AHPF medium were higher than in cells prepared in SC medium and approached those obtained in eggs.
100

Viral determinants of influenza A (H5N1) associated TNF-a hyper-induction in human primary monocyte-derived macrophages

Wong, Hing-ki, Charmaine. January 2006 (has links)
Thesis (M. Phil.)--University of Hong Kong, 2006. / Title proper from title frame. Also available in printed format.

Page generated in 0.0454 seconds