Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Water Res ; 88: 346-357, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26512813

ABSTRACT

In this study, laboratory scale digesters were operated to simulate potential shocks to the Anaerobic Digestion (AD) process at a 350 ML/day wastewater treatment plant. The shocks included high (42 °C) and low (32 °C) temperature (either side of mesophilic 37 °C) and a 20% loading of fats, oil and grease (FOG; 20% w:v). These variables were explored at two sludge retention times (12 and 20 days) and two organic loading rates (2.0 and 2.5 kgTS/m(3)day OLR). Metagenomic and metabolomic approaches were then used to characterise the impact of operational shocks in regard to temperature and FOG addition, as determined through monitoring of biogas production, the microbial profile and their metabolism. Results showed that AD performance was not greatly affected by temperature shocks, with the biggest impact being a reduction in biogas production at 42 °C that persisted for 32 ± 1 days. The average biogas production across all digesters at the completion of the experiment was 264.1 ± 76.5 mL/day, with FOG addition observed to significantly promote biogas production (+87.8 mL/day). Metagenomic and metabolomic analyses of the digesters indicated that methanogens and methane oxidising bacteria (MOB) were low in relative abundance, and that the ratio of oxidising bacteria (methane, sulphide and sulphate) with respect to sulphate reducing bacteria (SRB) had a noticeable influence on biogas production. Furthermore, increased biogas production correlated with an increase in short chain fatty acids, a product of the addition of 20% FOG. This work demonstrates the application of metagenomics and metabolomics to characterise the microbiota and their metabolism in AD digesters, providing insight to the resilience of crucial microbial populations when exposed to operational shocks.


Subject(s)
Bioreactors/microbiology , Metabolomics/methods , Metagenomics/methods , Microbial Consortia/physiology , Waste Disposal, Fluid/methods , Anaerobiosis , Archaea/genetics , Archaea/metabolism , Bacteria/genetics , Bacteria/metabolism , Biofuels , High-Throughput Nucleotide Sequencing , Methane/metabolism , Sulfates/metabolism , Waste Disposal, Fluid/instrumentation
2.
Vaccine ; 32(17): 1897-900, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24560674

ABSTRACT

UNLABELLED: We investigated the cross-neutralising potential of serum and nasal wash samples from volunteers who were intranasally immunised once with a monovalent replication-deficient delNS1-H1N1 influenza virus vaccine (7.7log10TCID50/volunteer). Eight out of twelve (8/12) vaccinees responded to vaccination with a significant increase of antibody levels in serum IgG ELISA, mucosal IgA ELISA, MNA or HAI. Four responders showed delNS1-specific ELISA IgA increases and revealed excellent homosubtypic neutralising activity in serum and mucosal washings (4/4). However, 0/4 of the sera but 3/4 of the nasal washings neutralised also heterosubtypic H3N2 and H5N1 influenza viruses. Depletion experiments proved that IgA but not IgG is responsible for the cross-neutralising activity of the nasal wash sample. Our findings indicate that the induction of virus-neutralising IgA may represent a valuable correlate of cross-protection of intranasal influenza vaccines and that the delNS1 concept constitutes a promising approach to protect humans from seasonal and pandemic influenza threats. CLINICAL TRIAL REGISTRATION: NCT00724997.


Subject(s)
Immunity, Mucosal , Immunoglobulin A/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Administration, Intranasal , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Cross Protection , Double-Blind Method , Hemagglutination Inhibition Tests , Humans , Immunoglobulin G/blood , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza A Virus, H5N1 Subtype , Neutralization Tests , Vaccines, Inactivated/immunology
3.
Chemosphere ; 91(5): 676-84, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23453605

ABSTRACT

Precipitation in the Mg-Ca-NH3-PO4 system has been explored to improve understanding of likely phases recoverable from complex wastewaters. Over a range of Mg/Ca combinations (0-100%) and pH 5-11, at least seven identifiable crystalline phases could be precipitated from artificial wastewater including: struvite, hydroxylapatite, newberyite, brushite, merrilite/whitlockite, octocalcium phosphate, and monetite. This experimental study has outlined the physicochemical conditions required to produce various phosphate products from synthetic wastewater, and found that large differences exist between experimentally formed phases and thermodynamical predictions. Struvite formation is the most desirable precipitate for the recovery of phosphate based upon purity, growth characteristics, dewatering properties, phosphate removal efficiency, and its ability to simultaneously remove ammonia. This study has also demonstrated that in specific cases the preliminary precipitation of brushite is a possible means of decreasing calcium content such that subsequent struvite formation could achieve higher-purity. Utilising experimental results and information on current commodity prices, discussion on the choice of Mg and Ca sources for phosphorus recovery provides guidance on appropriate means to optimise the formation and yield of high quality cost-optimised products.


Subject(s)
Phosphorus/analysis , Recycling/methods , Waste Disposal, Fluid/methods , Wastewater/chemistry , Water Pollutants, Chemical/analysis
4.
J Clin Virol ; 56(4): 323-30, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23375739

ABSTRACT

BACKGROUND: The hemagglutination inhibition assay (HAI) is universally regarded as the gold standard in influenza virus serology. Nevertheless, difficulties in titre readouts are common and interlaboratory variations are frequently reported. OBJECTIVE: We developed and validated the modified HAI to facilitate reliable, accurate and reproducible analysis of sera derived from influenza vaccination studies. STUDY DESIGN: Clinical and preclinical serum samples, NIBSC reference sera and seasonal influenza virus type A (H1N1 and H3N2) and type B antigens were employed to validate the mHAI. Moreover, pandemic virus strains (H5N1 and H1N1pdm09) were used to prove assay robustness. RESULTS: Utilisation of a 0.08% solution of stabilised human erythrocytes, assay buffer containing bovine serum albumin and microscopical plate readout are the major differences between the modified and standard HAI assay protocols. Validation experiments revealed that the mHAI is linear, specific and up to eightfold more sensitive than the standard HAI. In 95.6% of all measurements mHAI titres were precisely measured irrespective of the assay day, run or operator. Moreover, 96.4% (H1N1) or 95.2% (H3N2 and B), respectively, of all serum samples were determined within one dilution step of the nominal values for spiked samples. Finally, the mHAI results remained unaffected by variations in virus antigens, erythrocytes, reagents, laboratory location, sample storage conditions or matrix components. CONCLUSION: The modified HAI is easy to analyse, requires only a single source of erythrocytes and allows utilisation of numerous influenza virus antigens, also including virus strains which are difficult to handle by the standard HAI (e.g. H3N2, H5N1 and H1N1pdm09).


Subject(s)
Antibodies, Viral/blood , Hemagglutination Inhibition Tests/methods , Orthomyxoviridae Infections/immunology , Animals , Erythrocytes/immunology , Ferrets/immunology , Ferrets/virology , Hemagglutinins, Viral/analysis , Humans , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Reproducibility of Results , Sensitivity and Specificity , Serum Albumin, Bovine , Viral Load
5.
Vopr Virusol ; 56(6): 19-22, 2011.
Article in Russian | MEDLINE | ID: mdl-22359944

ABSTRACT

The paper gives the results of evaluating the efficiency of deINS1 pandemic H5N1 vaccine candidate VN1203delNS1 which was constructed by reverse genetics on the basis of influenza virus strain A/Vietnam/1203/04. The safety, immunogenicity and cross-protection of the vaccine strain against different H5N1 virus clades were demonstrated in mouse and macaque models. The results showed the possibility of designing a new-generation replication-deficient intranasal influenza vaccine, by applying an approach to deleting the NS1 pathogenicity factor, an antagonist of the interferon system.


Subject(s)
Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Orthomyxoviridae Infections/prevention & control , Vaccines, Attenuated/therapeutic use , Viral Nonstructural Proteins/genetics , Administration, Intranasal , Animals , Chlorocebus aethiops , Cross Protection/immunology , Drug Evaluation, Preclinical , Humans , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/genetics , Influenza, Human/immunology , Interferons/metabolism , Macaca fascicularis , Mice , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Reverse Genetics/methods , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vero Cells , Viral Nonstructural Proteins/immunology
6.
Cancer Res ; 61(22): 8188-93, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11719449

ABSTRACT

The NS1 protein of influenza virus is a virulence factor that counteracts the PKR-mediated antiviral response by the host. As a consequence, influenza NS1 gene knockout virus delNS1 (an influenza A virus lacking the NS1 open reading frame) fails to replicate in normal cells but produces infectious particles in PKR-deficient cells. Because it is known that oncogenic ras induces an inhibitor of PKR, we addressed the question of whether the delNS1 virus selectively replicates in cells expressing oncogenic ras. We show that upon transfection and expression of oncogenic N-ras, cells become permissive for productive delNS1 virus replication, suggesting that the delNS1 virus has specific oncolytic properties. Viral growth in the oncogenic ras-transfected cells is associated with a reduction of PKR activation during infection. Moreover, treatment of s.c. established N-ras-expressing melanomas in severe combined immunodeficiency mice with the delNS1 virus revealed that this virus has tumor-ablative potentials. The delNS1 virus does not replicate in nonmalignant cell lines such as melanocytes, keratinocytes, or endothelial cells. The apathogenic nature of the delNS1 virus combined with the selective replication properties of this virus in oncogenic ras-expressing cells renders this virus an attractive candidate for the therapy of tumors with an activated ras-signaling pathway.


Subject(s)
Genes, ras/physiology , Influenza A virus/physiology , Animals , Cell Transformation, Viral/genetics , Cell Transformation, Viral/physiology , Chlorocebus aethiops , Enzyme Activation , Genes, ras/genetics , Humans , Influenza A virus/genetics , Influenza A virus/metabolism , Male , Melanoma/therapy , Melanoma/virology , Mice , Mice, SCID , Transfection , Tumor Cells, Cultured , Vero Cells , Viral Nonstructural Proteins/genetics , Virus Replication , Xenograft Model Antitumor Assays , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
7.
Melanoma Res ; 11(4): 379-83, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11479426

ABSTRACT

The pro-apoptotic prostate apoptosis response-4 gene product Par-4 sensitizes prostate cells to the induction of programmed cell death. In this study we examined Par-4 expression in human melanoma cell lines and melanoma metastases. The heterogeneous expression detected prompted us to investigate the biological relevance of Par-4 in a human melanoma xenotransplantation model. Overexpression of Par-4 by transfection decreased tumour development in xenotransplanted A375-C6 melanoma cells in SCID mice and correlated to an increase in tumour cell apoptosis. These data suggest that high expression of the pro-apoptotic protein Par-4 could qualify as a prognostic marker in human melanoma.


Subject(s)
Apoptosis/genetics , Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins , Melanoma/genetics , Melanoma/pathology , Animals , Apoptosis Regulatory Proteins , Biomarkers, Tumor/genetics , Blotting, Western , Female , Mice , Mice, SCID , Neoplasm Transplantation , Prognosis , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
8.
J Virol ; 74(24): 11566-73, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090154

ABSTRACT

The alpha/beta interferon (IFN-alpha/beta) system represents one of the first lines of defense against virus infections. As a result, most viruses encode IFN antagonistic factors which enhance viral replication in their hosts. We have previously shown that a recombinant influenza A virus lacking the NS1 gene (delNS1) only replicates efficiently in IFN-alpha/beta-deficient systems. Consistent with this observation, we found that infection of tissue culture cells with delNS1 virus, but not with wild-type influenza A virus, induced high levels of mRNA synthesis from IFN-alpha/beta genes, including IFN-beta. It is known that transactivation of the IFN-beta promoter depends on NF-kappaB and several other transcription factors. Interestingly, cells infected with delNS1 virus showed high levels of NF-kappaB activation compared with those infected with wild-type virus. Expression of dominant-negative inhibitors of the NF-kappaB pathway during delNS1 virus infection prevented the transactivation of the IFN-beta promoter, demonstrating a functional link between NF-kappaB activation and IFN-alpha/beta synthesis in delNS1 virus-infected cells. Moreover, expression of the NS1 protein prevented virus- and/or double-stranded RNA (dsRNA)-mediated activation of the NF-kappaB pathway and of IFN-beta synthesis. This inhibitory property of the NS1 protein of influenza A virus was dependent on its ability to bind dsRNA, supporting a model in which binding of NS1 to dsRNA generated during influenza virus infection prevents the activation of the IFN system. NS1-mediated inhibition of the NF-kappaB pathway may thus play a key role in the pathogenesis of influenza A virus.


Subject(s)
Influenza A virus , Interferon-alpha/metabolism , Interferon-beta/metabolism , NF-kappa B/metabolism , Orthomyxoviridae Infections/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Gene Expression Regulation, Viral , Interferon-alpha/genetics , Interferon-beta/genetics , Mice , NF-kappa B/genetics , Orthomyxoviridae Infections/genetics , Viral Nonstructural Proteins/genetics
9.
J Virol ; 74(17): 7989-96, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10933707

ABSTRACT

We present a novel mechanism by which viruses may inhibit the alpha/beta interferon (IFN-alpha/beta) cascade. The double-stranded RNA (dsRNA) binding protein NS1 of influenza virus is shown to prevent the potent antiviral interferon response by inhibiting the activation of interferon regulatory factor 3 (IRF-3), a key regulator of IFN-alpha/beta gene expression. IRF-3 activation and, as a consequence, IFN-beta mRNA induction are inhibited in wild-type (PR8) influenza virus-infected cells but not in cells infected with an isogenic virus lacking the NS1 gene (delNS1 virus). Furthermore, NS1 is shown to be a general inhibitor of the interferon signaling pathway. Inhibition of IRF-3 activation can be achieved by the expression of wild-type NS1 in trans, not only in delNS1 virus-infected cells but also in cells infected with a heterologous RNA virus (Newcastle disease virus). We propose that inhibition of IRF-3 activation by a dsRNA binding protein significantly contributes to the virulence of influenza A viruses and possibly to that of other viruses.


Subject(s)
DNA-Binding Proteins/metabolism , Influenza A virus/physiology , Transcription Factors/metabolism , Transcriptional Activation , Viral Nonstructural Proteins/metabolism , Animals , Blotting, Western , Cell Line , Chick Embryo , DNA-Binding Proteins/antagonists & inhibitors , Humans , Interferon Regulatory Factor-3 , Interferon-beta/metabolism , Mutation , Newcastle disease virus/physiology , RNA, Messenger/metabolism , Respirovirus/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/antagonists & inhibitors , Transfection , Viral Nonstructural Proteins/genetics
10.
J Virol ; 74(13): 6203-6, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10846107

ABSTRACT

The availability of an influenza virus NS1 gene knockout virus (delNS1 virus) allowed us to establish the significance of the biological relationship between the influenza virus NS1 protein and double-stranded-RNA-activated protein kinase (PKR) in the life cycle and pathogenicity of influenza virus. Our results show that the lack of functional PKR permits the delNS1 virus to replicate in otherwise nonpermissive hosts, suggesting that the major function of the influenza virus NS1 protein is to counteract or prevent the PKR-mediated antiviral response.


Subject(s)
Influenza A virus/immunology , Influenza A virus/pathogenicity , Influenza, Human/immunology , Viral Nonstructural Proteins/immunology , Virus Replication , eIF-2 Kinase/immunology , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Disease Models, Animal , Humans , Influenza A virus/growth & development , Influenza, Human/genetics , Influenza, Human/virology , Interferons/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT1 Transcription Factor , Trans-Activators/genetics , Trans-Activators/immunology , Viral Nonstructural Proteins/genetics , eIF-2 Kinase/genetics
11.
Proc Natl Acad Sci U S A ; 97(8): 4309-14, 2000 Apr 11.
Article in English | MEDLINE | ID: mdl-10725408

ABSTRACT

We propose a rational approach to the generation of live viral vaccines: alteration of virally encoded type I IFN antagonists to attenuate virulence while retaining immunogenicity. We have explored this concept by using the influenza virus. Previously we have shown that the NS1 protein of influenza A virus possesses anti-IFN activity. We now present evidence that influenza A and B viruses encoding altered viral NS1 proteins are highly attenuated in the mouse host, yet provide protection from challenge with wild-type viruses.


Subject(s)
Influenza A virus/genetics , Influenza B virus/genetics , Influenza Vaccines/genetics , Viral Nonstructural Proteins/genetics , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , DNA Primers , Dogs , Enzyme-Linked Immunosorbent Assay , Female , Influenza A virus/immunology , Influenza B virus/immunology , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Vero Cells , Viral Nonstructural Proteins/immunology
12.
Arch Virol Suppl ; 15: 131-8, 1999.
Article in English | MEDLINE | ID: mdl-10470274

ABSTRACT

Concerted efforts to study the molecular biology of influenza viruses and the ability to genetically engineer them have dramatically advanced our understanding of the functions of influenza viral genes and gene products. The only nonstructural protein (NS1) coded for by the influenza virus was shown to possess interferon antagonist activity and thus to play an important role in countering the interferon (antiviral) response of the host following infection. Influenza A and B virus mutants with "weak" anti-interferon activity are highly attenuated because the host is able to mount an effective interferon response. It is suggested that these NS1-modified attenuated influenza viruses can induce a protective immune response and that they are ideal live virus vaccine candidates against influenza.


Subject(s)
Influenza A virus/chemistry , Influenza A virus/immunology , Influenza B virus/immunology , Influenza Vaccines/immunology , Viral Nonstructural Proteins/physiology , Animals , Genes, Viral , Humans , Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza B virus/chemistry , Influenza B virus/genetics , Influenza B virus/pathogenicity , Influenza, Human/prevention & control , Mice , Vaccines, Attenuated , Vaccines, Inactivated , Viral Nonstructural Proteins/genetics
13.
J Infect Dis ; 178(5): 1359-68, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9780256

ABSTRACT

Previously, a mucosal model of immunization against human immunodeficiency virus type 1 (HIV-1) was established by using influenza virus as a vector for the neutralizing gp41 epitope ELDKWA. Whether replication of this chimeric influenza virus in the upper respiratory tract of mice is sufficient for inducing mucosal immune responses in the genital tract was investigated. An immunization strategy was established that permits the virus to replicate in the murine upper respiratory tracts but not in the lungs. Intranasal application of the chimeric virus induced HIV-1-specific antibodies in sera and genital tract. In addition, chimeric virus-specific antibody-secreting cells were detected in lymphocyte populations obtained from lungs, spleens, and urogenital tracts. These results indicate that replication of the chimeric influenza/ELDKWA virus in the upper respiratory tract is sufficient to induce systemic immune responses as well as local immune responses in the genital tract.


Subject(s)
HIV Antibodies/biosynthesis , HIV-1/immunology , Influenza A virus/immunology , Lung/immunology , Nasal Mucosa/immunology , Reassortant Viruses/immunology , Vagina/immunology , AIDS Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Monoclonal/metabolism , Chimera , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors , HIV Antibodies/immunology , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/immunology , HIV-1/genetics , Humans , Immunity, Mucosal , Influenza A virus/genetics , Influenza A virus/physiology , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Lung/virology , Mice , Mice, Inbred BALB C , Mucous Membrane/immunology , Mucous Membrane/virology , Nasal Mucosa/virology , Peptide Fragments/immunology , Reassortant Viruses/genetics , Vagina/metabolism , Vagina/virology , Virus Replication
14.
J Virol ; 72(8): 6437-41, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9658085

ABSTRACT

We established a reverse genetics system for the nonstructural (NS) gene segment of influenza A virus. This system is based on the use of the temperature-sensitive (ts) reassortant virus 25A-1. The 25A-1 virus contains the NS gene from influenza A/Leningrad/134/57 virus and the remaining gene segments from A/Puerto Rico (PR)/8/34 virus. This particular gene constellation was found to be responsible for the ts phenotype. For reverse genetics of the NS gene, a plasmid-derived NS gene from influenza A/PR/8/34 virus was ribonucleoprotein transfected into cells that were previously infected with the 25A-1 virus. Two subsequent passages of the transfection supernatant at 40 degreesC selected viruses containing the transfected NS gene derived from A/PR/8/34 virus. The high efficiency of the selection process permitted the rescue of transfectant viruses with large deletions of the C-terminal part of the NS1 protein. Viable transfectant viruses containing the N-terminal 124, 80, or 38 amino acids of the NS1 protein were obtained. Whereas all deletion mutants grew to high titers in Vero cells, growth on Madin-Darby canine kidney (MDCK) cells and replication in mice decreased with increasing length of the deletions. In Vero cells expression levels of viral proteins of the deletion mutants were similar to those of the wild type. In contrast, in MDCK cells the level of the M1 protein was significantly reduced for the deletion mutants.


Subject(s)
Influenza A virus/growth & development , Viral Nonstructural Proteins/physiology , Animals , Cell Line , Chick Embryo , Chlorocebus aethiops , Dogs , Humans , Influenza A virus/genetics , Influenza A virus/metabolism , Influenza A virus/physiology , Mice , Mice, Inbred BALB C , Nucleocapsid/biosynthesis , Peptide Biosynthesis , Sequence Deletion , Transfection , Vero Cells , Viral Matrix Proteins/biosynthesis , Viral Nonstructural Proteins/genetics , Virus Replication
15.
J Gen Virol ; 79 ( Pt 6): 1405-9, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9634082

ABSTRACT

During the 1996 influenza epidemic in Vienna we obtained influenza A virus specimens (Vienna/47/96, Vienna/81/96) which grow efficiently in African green monkey kidney (Vero) cells but not in embryonated chicken eggs. Amplification of the specimens in Vero cells resulted in progeny that agglutinated human but not chicken erythrocytes. Reassortment analysis suggested that the haemagglutinin (HA) might be responsible for the host restriction. Vero cells were infected with the Vienna/47/96 virus and then transfected with reconstituted ribonucleoprotein complexes containing HA genes from egg-adapted strains. Subsequent selective passages in embryonated chicken eggs resulted in selection of transfectant viruses, growing in eggs and containing the transfected HAs. The results demonstrate that host restriction of the Vero-adapted Vienna/47/96 virus is due to its HA. Moreover, the experiments showed that the Vienna/47/96 strain can be used as helper virus for reverse genetics experiments.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A virus/genetics , Transfection , Animals , Base Sequence , Cell Line , Chick Embryo , Chlorocebus aethiops , DNA, Viral , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/physiology , Humans , Influenza A virus/pathogenicity , Molecular Sequence Data , Vero Cells
16.
Virology ; 252(2): 324-30, 1998 Dec 20.
Article in English | MEDLINE | ID: mdl-9878611

ABSTRACT

The NS1 protein is the only nonstructural protein encoded by influenza A virus. It has been proposed that the NS1 performs several regulatory functions during the viral replication cycle, including the regulation of synthesis, transport, splicing, and translation of mRNAs. Through the use of reverse genetics, a viable transfectant influenza A virus (delNS1) which lacks the NS1 gene has been generated. Our results indicate that the NS1 of influenza A virus is an auxiliary (virulence) factor which plays a crucial role in inhibiting interferon-mediated antiviral responses of the host.


Subject(s)
Gene Deletion , Influenza A virus/genetics , Orthomyxoviridae Infections/physiopathology , Viral Nonstructural Proteins/genetics , Virus Replication , Animals , Cell Line , Chick Embryo , Chlorocebus aethiops , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Dogs , Influenza A virus/physiology , Kidney , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/genetics , STAT1 Transcription Factor , Signal Transduction , Trans-Activators/genetics , Trans-Activators/physiology , Transfection , Vero Cells , Viral Nonstructural Proteins/metabolism
17.
J Infect Dis ; 176 Suppl 1: S45-9, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9240694

ABSTRACT

Approaches to improve the efficacy of the current (killed) influenza virus vaccines include the generation of cold-adapted and genetically engineered influenza viruses containing specific attenuating mutations. It is hoped that these genetically altered viruses, in which the hemagglutinin and neuraminidase genes from circulating strains have been incorporated by reassortment, can be used as safe live influenza virus vaccines to induce a long-lasting protective immune response in humans. In addition, genetically engineered influenza viruses may provide a means for expressing foreign antigens. Immunization of mice with recombinant influenza and vaccinia viruses expressing specific antigens of Plasmodium yoelii resulted in a dramatic protective immune response against malaria in this model. Mice immunized with recombinant influenza viruses expressing human immunodeficiency virus (HIV) epitopes generated long-lasting HIV-specific serum antibodies and secretory IgA in the secretory nasal, vaginal, and intestinal mucosa. These results suggest that genetically engineered influenza viruses may be developed for use as live virus vaccines against influenza as well as other diseases.


Subject(s)
Influenza Vaccines/immunology , Vaccines, Synthetic/immunology , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors , Humans , Immunity, Mucosal , Mice , Vaccines, Attenuated/immunology
18.
Virus Res ; 44(1): 23-31, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8873410

ABSTRACT

Influenza A virus replication and packaging is mediated by cis-acting signals, which are located at the 3' and the 5' end of the viral segments. The terminal residues can be divided into conserved and nonconserved residues. We have constructed a mutant influenza A/WSN/33 virus, which contains multiple mutations in the nonconserved residues of the neuraminidase (NA) segment. This virus shows a segment-specific reduction of the genomic RNA content in the infected cell and in the progeny virus. Further mutants and revertant viruses revealed that it was not possible to define specific residues, which were responsible for the reduction of the NA-specific RNA. Thus, it appears that an efficient vRNA formation is dependent on the synergistic effect of the terminal sequences.


Subject(s)
Influenza A virus/enzymology , Neuraminidase/genetics , RNA, Viral , Viral Proteins/genetics , Animals , Cattle , Cell Line , Cloning, Molecular , Humans , Influenza A virus/genetics , Influenza A virus/growth & development , Mutagenesis
19.
AIDS ; 10(6): 587-93, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8780812

ABSTRACT

OBJECTIVE: To investigate whether variations of the conserved gp41 amino-acid sequence ELDKWA affect its binding or neutralization by monoclonal antibody (MAb) 2F5. DESIGN AND METHODS: Neutralization assays were performed with primary isolates from different HIV-1 subtypes and the sequences corresponding to the 2F5 epitope region were analysed. Studies of MAb 2F5 peptide reactivity were performed by spot analysis, using peptides immobilized on cellulose. The frequency of emergence of neutralization-resistant virus variants was determined by immune selection experiments in the presence of MAb 2F5. RESULTS: Primary isolates from clades A, B and E were neutralized by MAb 2F5. Neutralization sensitivity correlated with the presence of the LDKW motif. A K-to-N change in the core sequence was identified in a neutralization-resistant patient isolate. Neutralization resistant virus variants that were selected in the presence of MAb 2F5 were found to contain D-to-N, D-to-E, or K-to-N changes within the LDKW sequence. Neither in natural isolates nor in variants obtained under immune selection conditions in the laboratory were changes in the L and W positions observed. Studies of MAb 2F5 binding to variations of the ELDKWA peptide confirmed that the changes at the first and last positions did not significantly reduce binding capacity, whereas amino-acid changes from D to N, D to E, and K to N almost completely abrogated binding of MAb 2F5. CONCLUSION: Sequence analysis of a variety of primary isolates suggests that the major determinant of MAb 2F5 binding corresponds to the amino-acid sequence LDKW. Naturally occurring and in vitro selected neutralization-resistant viruses contained changes in the D and K positions of the ELDKWA motif.


Subject(s)
Antigenic Variation , HIV Envelope Protein gp41/immunology , HIV-1/immunology , Antibodies, Monoclonal/immunology , Humans , Sequence Analysis
20.
J Virol ; 70(2): 1100-8, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8551569

ABSTRACT

We have isolated and characterized human monoclonal antibody 2G12 to the gp120 surface glycoprotein of human immunodeficiency virus type 1 (HIV-1). This antibody potently and broadly neutralizes primary and T-cell line-adapted clade B strains of HIV-1 in a peripheral blood mononuclear cell-based assay and inhibits syncytium formation in the AA-2 cell line. Furthermore, 2G12 possesses neutralizing activity against strains from clade A but not from clade E. Complement- and antibody-dependent cellular cytotoxicity-activating functions of 2G12 were also defined. The gp120 epitope recognized by 2G12 was found to be distinctive; binding of 2G12 to LAI recombinant gp120 was abolished by amino acid substitutions removing N-linked carbohydrates in the C2, C3, V4, and C4 regions of gp120. This gp120 mutant recognition pattern has not previously been observed, indicating that the 2G12 epitope is unusual. consistent with this, antibodies able to block 2G12 binding to recombinant gp120 were not detected in significant quantities in 16 HIV-positive human serum samples.


Subject(s)
Antibodies, Monoclonal/immunology , Epitope Mapping , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Amino Acid Sequence , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line , Complement C3/immunology , Giant Cells/immunology , Giant Cells/virology , Glycosylation , HIV-1/isolation & purification , Humans , Molecular Sequence Data , Mutation , Neutralization Tests , Polysaccharides/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...