Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Metab ; 4(5): 534-546, 2022 05.
Article in English | MEDLINE | ID: mdl-35655026

ABSTRACT

Although the immunomodulatory and cytoprotective properties of itaconate have been studied extensively, it is not known whether its naturally occurring isomers mesaconate and citraconate have similar properties. Here, we show that itaconate is partially converted to mesaconate intracellularly and that mesaconate accumulation in macrophage activation depends on prior itaconate synthesis. When added to human cells in supraphysiological concentrations, all three isomers reduce lactate levels, whereas itaconate is the strongest succinate dehydrogenase (SDH) inhibitor. In cells infected with influenza A virus (IAV), all three isomers profoundly alter amino acid metabolism, modulate cytokine/chemokine release and reduce interferon signalling, oxidative stress and the release of viral particles. Of the three isomers, citraconate is the strongest electrophile and nuclear factor-erythroid 2-related factor 2 (NRF2) agonist. Only citraconate inhibits catalysis of itaconate by cis-aconitate decarboxylase (ACOD1), probably by competitive binding to the substrate-binding site. These results reveal mesaconate and citraconate as immunomodulatory, anti-oxidative and antiviral compounds, and citraconate as the first naturally occurring ACOD1 inhibitor.


Subject(s)
Fumarates/pharmacology , Interferons , Macrophages , Maleates/pharmacology , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Carboxy-Lyases , Catalysis , Humans , Inflammation/metabolism , Macrophages/metabolism , Oxidative Stress
2.
Infect Genet Evol ; 40: 80-90, 2016 06.
Article in English | MEDLINE | ID: mdl-26917362

ABSTRACT

It is almost a decade since the highly pathogenic H5N1 avian influenza virus (A/H5N1) of clade 2.2.1 was introduced to Egypt in 2005, most likely, via wild birds; marking the longest endemic status of influenza viruses in poultry outside Asia. The endemic A/H5N1 in Egypt still compromises the poultry industry, poses serious hazards to public health and threatens to become potentially pandemic. The control strategies adopted for A/H5N1 in Egyptian poultry using diverse vaccines in commercialized poultry neither eliminated the virus nor did they decrease its evolutionary rate. Several virus clades have evolved, a few of them disappeared and others prevailed. Disparate evolutionary traits in both birds and humans were manifested by accumulation of clade-specific mutations across viral genomes driven by a variety of selection pressures. Viruses in vaccinated poultry populations displayed higher mutation rates at the immunogenic epitopes, promoting viral escape and reducing vaccine efficiency. On the other hand, viruses isolated from humans displayed changes in the receptor binding domain, which increased the viral affinity to bind to human-type glycan receptors. Moreover, viral pathogenicity exhibited several patterns in different hosts. This review aims to provide an overview of the viral evolution, pathogenicity and vaccine efficacy of A/H5N1 in Egypt during the last ten years.


Subject(s)
Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/epidemiology , Influenza, Human/epidemiology , Mutation Rate , Poultry Diseases/virology , Animals , Egypt/epidemiology , Evolution, Molecular , Humans , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/pathogenicity , Poultry/virology , Poultry Diseases/epidemiology , Virulence , Virulence Factors/genetics
3.
J Immunol ; 167(9): 5209-16, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11673534

ABSTRACT

Infection of cells by vesicular stomatitis virus (VSV) results in the inhibition of host transcription. We show in this study that infection of HeLa cells with VSV leads to a strongly diminished activation of STAT3 and STAT1 by the inflammatory cytokine IL-6. This effect was mimicked by forced expression of a single viral protein, the matrix (M)-protein of VSV, which blocked STAT activation via chimeric receptors containing the cytoplasmic domain of the IL-6 signal transducer gp130. Western blot analysis revealed that VSV M-protein did not inhibit the nuclear translocation of activated STAT3 but did inhibit its tyrosine phosphorylation. Inhibition of STAT activation was not dependent on tyrosine 759 of the IL-6 signal transducer gp130, suggesting that the inhibitory action of VSV M-protein is not mediated by the induction of the suppressor of cytokine signaling 3. VSV M-protein inhibited gene transcription from cotransfected alpha(2)-macroglobulin or antichymotrypsin promoter/luciferase reporter constructs which contain STAT3-binding sites. However, transcription from a STAT5-dependent construct was not negatively affected. In conclusion, our data suggest that infection by VSV and specifically overexpression of the viral M-protein interferes with an important signaling pathway necessary for triggering antiviral and inflammatory responses.


Subject(s)
Antigens, CD/physiology , DNA-Binding Proteins/physiology , JNK Mitogen-Activated Protein Kinases , Membrane Glycoproteins/physiology , Trans-Activators/physiology , Vesicular stomatitis Indiana virus/pathogenicity , Viral Matrix Proteins/toxicity , Cytokine Receptor gp130 , HeLa Cells , Humans , Interleukin-6/pharmacology , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases/physiology , Phosphorylation , STAT3 Transcription Factor , Transcription, Genetic , Tyrosine/metabolism
4.
J Biol Chem ; 276(24): 10990-8, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11441823

ABSTRACT

Influenza A virus infection of cells results in the induction of a variety of antiviral cytokines, including those that are regulated by transcription factors of the activating protein-1 (AP-1) family. Here we show that influenza virus infection induces AP-1-dependent gene expression in productively infected cells but not in cells that do not support viral replication. Among the AP-1 factors identified to bind to their cognate DNA element during viral infections of Madin-Darby canine kidney and U937 cells are those that are regulated via phosphorylation by JNKs. Accordingly, we observed that induction of AP-1-dependent gene expression correlates with a strong activation of JNK in permissive cells, which appears to be caused by viral RNA accumulation during replication. Blockade of JNK signaling at several levels of the cascade by transient expression of dominant negative kinase mutants and inhibitory proteins resulted in inhibition of virus-induced JNK activation, reduced AP-1 activity, and impaired transactivation of the IFN-beta promoter. Virus yields from transfected and infected cells in which JNK signaling was inhibited were higher compared with the levels from control cells. Therefore, we conclude that virus-induced activation of JNK and AP-1 is part of the innate antiviral response of the cell.


Subject(s)
Gene Expression Regulation, Viral , Influenza A virus/physiology , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 4 , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Transcription Factor AP-1/physiology , Activating Transcription Factor 2 , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , DNA/metabolism , DNA-Directed DNA Polymerase/metabolism , Enzyme Activation , HeLa Cells , Humans , Interferon-beta/genetics , MAP Kinase Kinase 7 , Phosphorylation , Promoter Regions, Genetic , Protein Binding , RNA, Viral/metabolism , Species Specificity , Transcription Factors/metabolism , U937 Cells , Virus Replication
5.
J Virol ; 75(10): 4871-7, 2001 May.
Article in English | MEDLINE | ID: mdl-11312358

ABSTRACT

Borna disease virus (BDV) is a highly neurotropic virus that causes Borna disease, a virus-induced immune-mediated encephalomyelitis, in a variety of warm-blooded animals. Recent studies reported that BDV can be detected in patients with psychiatric disorders. BDV is noncytopathic, replicates in the nucleus of infected cells, and spreads intraaxonally in vivo. Upon infection of susceptible cultured cells, virus can be detected in foci. Little is known about the cellular components required for BDV replication. Here, we show that the cellular Raf/MEK/ERK signaling cascade is activated upon infection with BDV. In the presence of the MEK-specific inhibitor U0126, cells get infected with BDV; however, there is a block in virus spread to neighboring cells. The effect of the inhibitor on virus spread was still observed when the compound was added 2 h postinfection but not if treatment was initiated as late as 4 h after infection. Our results provide new insights into the BDV-host cell interaction and show that virus infection can be controlled with drugs interfering with a cellular signaling pathway. Since concentrations of the MEK inhibitor required to block BDV focus formation are not toxic for the host cells, our finding may be important with respect to antiviral drug development.


Subject(s)
Borna disease virus/drug effects , Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Nitriles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Borna disease virus/growth & development , Borna disease virus/physiology , Cell Line , Cells, Cultured , Enzyme Activation , Guinea Pigs , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism
6.
Nat Cell Biol ; 3(3): 301-5, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231581

ABSTRACT

Influenza A viruses are important worldwide pathogens in humans and different animal species. The functions of most of the ten different viral proteins of this negative-strand RNA virus have been well elucidated. However, little is known about the virus-induced intracellular signalling events that support viral replication. The Raf/MEK/ERK cascade is the prototype of mitogen-activated protein (MAP) kinase cascades and has an important role in cell growth, differentiation and survival. Investigation of the function of this pathway has been facilitated by the identification of specific inhibitors such as U0126, which blocks the cascade at the level of MAPK/ERK kinase (MEK). Here we show that infection of cells with influenza A virus leads to biphasic activation of the Raf/MEK/ERK cascade. Inhibition of Raf signalling results in nuclear retention of viral ribonucleoprotein complexes (RNPs), impaired function of the nuclear-export protein (NEP/NS2) and concomitant inhibition of virus production. Thus, signalling through the mitogenic cascade seems to be essential for virus production and RNP export from the nucleus during the viral life cycle.


Subject(s)
Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Influenza A virus/physiology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Nitriles/pharmacology , Active Transport, Cell Nucleus , Animals , Blotting, Western , Cell Line , Genes, Reporter , Humans , Immunohistochemistry , Influenza A virus/genetics , MAP Kinase Signaling System/drug effects , Microscopy, Confocal , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/metabolism , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins/metabolism , Transfection , Viral Proteins/metabolism , Virus Replication
7.
J Biol Chem ; 276(14): 10990-8, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11150300

ABSTRACT

Influenza A virus infection of cells results in the induction of a variety of antiviral cytokines, including those that are regulated by transcription factors of the activating protein-1 (AP-1) family. Here we show that influenza virus infection induces AP-1-dependent gene expression in productively infected cells but not in cells that do not support viral replication. Among the AP-1 factors identified to bind to their cognate DNA element during viral infections of Madin-Darby canine kidney and U937 cells are those that are regulated via phosphorylation by JNKs. Accordingly, we observed that induction of AP-1-dependent gene expression correlates with a strong activation of JNK in permissive cells, which appears to be caused by viral RNA accumulation during replication. Blockade of JNK signaling at several levels of the cascade by transient expression of dominant negative kinase mutants and inhibitory proteins resulted in inhibition of virus-induced JNK activation, reduced AP-1 activity, and impaired transactivation of the IFN-beta promoter. Virus yields from transfected and infected cells in which JNK signaling was inhibited were higher compared with the levels from control cells. Therefore, we conclude that virus-induced activation of JNK and AP-1 is part of the innate antiviral response of the cell.


Subject(s)
Influenza, Human/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/metabolism , Orthomyxoviridae/physiology , Signal Transduction , Transcription Factor AP-1/metabolism , Animals , Cell Line , Gene Expression Regulation, Viral , Humans , Influenza, Human/genetics , Influenza, Human/virology , MAP Kinase Kinase 4 , Virus Replication
8.
J Virol ; 74(14): 6316-23, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10864641

ABSTRACT

The hemagglutinin (HA) of fowl plague virus A/FPV/Rostock/34 (H7N1) carries two N-linked oligosaccharides attached to Asn123 and Asn149 in close vicinity to the receptor-binding pocket. In previous studies in which HA mutants lacking either one (mutants G1 and G2) or both (mutant G1,2) glycosylation sites had been expressed from a simian virus 40 vector, we showed that these glycans regulate receptor binding affinity (M. Ohuchi, R. Ohuchi, A. Feldmann, and H. D. Klenk, J. Virol. 71:8377-8384, 1997). We have now investigated the effect of these mutations on virus growth using recombinant viruses generated by an RNA polymerase I-based reverse genetics system. Two reassortants of influenza virus strain A/WSN/33 were used as helper viruses to obtain two series of HA mutant viruses differing only in the neuraminidase (NA). Studies using N1 NA viruses revealed that loss of the oligosaccharide from Asn149 (mutant G2) or loss of both oligosaccharides (mutant G1,2) has a pronounced effect on virus growth in MDCK cells. Growth of virus lacking both oligosaccharides from infected cells was retarded, and virus yields in the medium were decreased about 20-fold. Likewise, there was a reduction in plaque size that was distinct with G1,2 and less pronounced with G2. These effects could be attributed to a highly impaired release of mutant progeny viruses from host cells. In contrast, with recombinant viruses containing N2 NA, these restrictions were much less apparent. N1 recombinants showed lower neuraminidase activity than N2 recombinants, indicating that N2 NA is able to partly overrule the high-affinity binding of mutant HA to the receptor. These results demonstrate that N-glycans flanking the receptor-binding site of the HA molecule are potent regulators of influenza virus growth, with the glycan at Asn149 being dominant and that at Asn123 being less effective. In addition, we show here that HA and NA activities need to be highly balanced in order to allow productive influenza virus infection.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A virus/growth & development , Neuraminidase/genetics , Animals , Cattle , Cell Line , Chickens , Erythrocytes/virology , Glycosylation , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , In Vitro Techniques , Influenza A virus/genetics , Influenza A virus/metabolism , Mutagenesis, Site-Directed , Neuraminidase/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Recombination, Genetic , Viral Plaque Assay
9.
Viral Immunol ; 12(3): 175-96, 1999.
Article in English | MEDLINE | ID: mdl-10532647

ABSTRACT

Influenza A viruses are important worldwide pathogens for humans and different animal species. The infectious agent is the prototype of the orthomyxoviridae which are characterized by a segmented negative strand RNA genome that is replicated in the nucleus of the infected cell. The genome has a combined coding capacity of about 13 kb and contains the genetic information for ten viral proteins. Despite this relatively small coding capacity--large DNA viruses like herpes or poxviruses express about 150-200 gene products--influenza A viruses are able to successfully infect and multiply in a wide range of mammalian and avian species. It is therefore not surprising that influenza A viruses extensively use and manipulate host cell functions. This includes multiple interactions of viral proteins with cellular proteins. In recent years an increasing amount of information about the identity of the cellular factors that are involved in viral transcription and replication, intracellular trafficking of viral components and assembly of the virus particle has accumulated. This article aims to review recent developments in this field with a focus on cellular factors and processes which are activated by the virus to either support viral replication or to counteract host-cell defense mechanisms.


Subject(s)
Orthomyxoviridae/physiology , Animals , Apoptosis , Cell Nucleus/virology , Gene Expression , Humans , Orthomyxoviridae/immunology , Orthomyxoviridae/pathogenicity , Protein Kinases/physiology , Viral Envelope Proteins/biosynthesis , Virus Replication
10.
Proc Natl Acad Sci U S A ; 93(21): 11354-8, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8876139

ABSTRACT

The negative-strand RNA viruses are a broad group of animal viruses that comprise several important human pathogens, including influenza, measles, mumps, rabies, respiratory syncytial, Ebola, and hantaviruses. The development of new strategies to genetically manipulate the genomes of negative-strand RNA viruses has provided us with new tools to study the structure-function relationships of the viral components and their contributions to the pathogenicity of these viruses. It is also now possible to envision rational approaches--based on genetic engineering techniques--to design live attenuated vaccines against some of these viral agents. In addition, the use of different negative-strand RNA viruses as vectors to efficiently express foreign polypeptides has also become feasible, and these novel vectors have potential applications in disease prevention as well as in gene therapy.


Subject(s)
Genetic Engineering , RNA Viruses/genetics , Vaccines, Synthetic , Viral Vaccines , Virus Diseases/virology , Animals , Chimera , Drug Design , Genome, Viral , Humans , Influenza Vaccines , Orthomyxoviridae/genetics , Orthomyxoviridae/immunology , RNA Viruses/pathogenicity , Virulence , Virus Diseases/immunology , Virus Diseases/prevention & control
11.
J Virol ; 70(6): 4188-92, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8648766

ABSTRACT

A reverse genetics system for negative-strand RNA viruses was first successfully developed for influenza viruses. This technology involved the transfection of in vitro-reconstituted ribonucleoprotein (RNP) complexes into influenza virus-infected cells. We have now developed a method that allows intracellular reconstitution of RNP complexes from plasmid-based expression vectors. Expression of a viral RNA-like transcript is achieved from a plasmid containing a truncated human polymerase I (polI) promoter and a ribozyme sequence that generates the desired 3' end by autocatalytic cleavage. The polI-driven plasmid is cotransfected into human 293 cells with polII-responsive plasmids that express the viral PB1, PB2, PA, and NP proteins. This exclusively plasmid-driven system results in the efficient transcription and replication of the viral RNA-like reporter and allows the study of cis- and trans-acting signals involved in the transcription and replication of influenza virus RNAs. Using this system, we have also been able to rescue a synthetic neuraminidase gene into a recombinant influenza virus. This method represents a convenient alternative to the previously established RNP transfection system.


Subject(s)
Influenza A virus/genetics , Plasmids , RNA, Viral/genetics , Ribonucleoproteins/genetics , Base Sequence , Humans , Molecular Sequence Data , RNA, Viral/biosynthesis , Transcription, Genetic , Transfection
12.
J Gen Virol ; 76 ( Pt 10): 2529-37, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7595356

ABSTRACT

Influenza C virus is able to inactivate its own cellular receptors by virtue of a sialate 9-O-acetylesterase that releases the acetyl residue at position C-9 of N-acetyl-9-O-acetylneuraminic acid (Neu5,9Ac2). The receptor-destroying enzyme activity is a function of the surface glycoprotein HEF and this esterase belongs to the class of serine hydrolases. In their active site, these enzymes contain a catalytic triad made up of a serine, a histidine and an aspartic acid residue. Sequence comparison with other serine esterases has indicated that, in addition to serine-71 (S71), the amino acids histidine-368 or -369 (H368/369) and aspartic acid 261 (D261) are the most likely candidates to form the catalytic triad of the influenza C virus glycoprotein. By site-directed mutagenesis, mutants were generated in which alanine substituted for either of these amino acids. Using a phagemid expression vector, pSP1D-HEF the HEF gene was expressed in both COS 7 and MDCK I cells. The glycoprotein was obtained in a functional form only in the latter cells, as indicated by its transport to the cell surface and measurable enzyme activity. The low level of expression could be increased by stimulating the NF-KB-binding activity of the cytomegalovirus immediate-early promoter/enhancer element of the vector. The esterase activity of the mutant proteins was compared with that of the wild-type glycoprotein. With Neu5,9Ac2 as the substrate, the esterase specific activities of the S71/A mutant and the H368,369/A mutant were reduced by more than 90%. In the case of the D261/A mutant the specific activity was reduced by 64%. From this data we conclude that S71, H368/369 and D261 are likely to represent the catalytic triad of the influenza C virus glycoprotein HEF. In addition, N280 is proposed to stabilize the oxyanion of the presumptive transition state intermediate formed by the enzyme-substrate complex.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Gammainfluenzavirus/enzymology , Hemagglutinins, Viral/metabolism , Viral Fusion Proteins , Acetylesterase , Amino Acid Sequence , Animals , Aspartic Acid/physiology , Binding Sites , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/biosynthesis , Carboxylic Ester Hydrolases/genetics , Cell Line , Dogs , Enzyme Inhibitors/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Histidine/physiology , Isoflurophate/pharmacology , Molecular Sequence Data , Mutagenesis, Site-Directed , NF-kappa B/metabolism , Recombinant Fusion Proteins/biosynthesis , Serine/physiology , Serine Endopeptidases/genetics , Sialic Acids/metabolism , Viral Proteins/genetics
13.
J Gen Virol ; 75 ( Pt 5): 1023-30, 1994 May.
Article in English | MEDLINE | ID: mdl-8176364

ABSTRACT

The post-translational processing of the influenza C virus glycoprotein HEF was analysed. In cells infected with influenza C virus, HEF protein is synthesized as a glycosylated 80K polypeptide. A post-translational conformational rearrangement involving the formation of intramolecular disulphide bonds results in a decrease in its electrophoretic mobility. Therefore, SDS-PAGE under non-reducing conditions suggests an Mr of about 100K, whereas under reducing conditions an 80K protein is observed which is in accordance with the sequence data. The 100K form was detected 10 min after synthesis of HEF, and transport to the cell surface took about 60 min. This result indicates that the conformational change presumably occurs in the endoplasmic reticulum. A difference in post-translational processing was observed when the HEF gene was expressed in the absence of other influenza C virus genes. In cells infected with recombinant simian virus 40, the 80K precursor was synthesized, but this protein was neither converted to the 100K form nor transported to the cell surface. Deletion of the short cytoplasmic tail of HEF (Arg-Thr-Lys) or replacement of the two basic amino acids by hydrophobic (Ile) or acidic residues (Glu) resulted in HEF protein which was partially converted to the 100K form. Influenza C virus glycoprotein obtained after transient expression of the HEF gene using the vaccinia virus system was completely converted to the 100K form. However, in neither expression system was HEF transported to the cell surface. The possibility is discussed that the interaction of HEF with another viral protein is required for the post-translational folding and transport of this glycoprotein. The M protein of influenza C virus is suggested as a candidate for the chaperone which might interact with the cytoplasmic tail of HEF.


Subject(s)
Gammainfluenzavirus/metabolism , Hemagglutinins, Viral/metabolism , Protein Folding , Protein Processing, Post-Translational , Amino Acid Sequence , Base Sequence , Biological Transport , Cloning, Molecular , DNA Mutational Analysis , Genes, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Hemagglutinins, Viral/genetics , Gammainfluenzavirus/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Recombinant Proteins/metabolism , Simian virus 40/genetics , Simian virus 40/growth & development , Transfection , Vaccinia virus/genetics , Vaccinia virus/growth & development
14.
Virus Res ; 30(1): 105-10, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8266718

ABSTRACT

The influenza C glycoprotein HEF was analyzed for acetylesterase activity after SDS-polyacrylamide gel electrophoresis and transfer to nitrocellulose membranes. Using a histological esterase assay, the glycoprotein was detected as a colored band indicating that it is enzymatically active. The enzyme activity was not affected by low pH, but was abolished after denaturation by SDS as well as after breaking the disulfide bonds by reducing agents. Glycoprotein inactivated by SDS regained its enzyme activity if the ionic detergent was displaced by either bovine serum albumin or a nonionic detergent. The stability of the enzyme combined with the color assay provides a convenient tool to study the acetylesterase activity of the influenza C virus glycoprotein.


Subject(s)
Acetylesterase/analysis , Gammainfluenzavirus/enzymology , Viral Envelope Proteins/chemistry , Acetylesterase/drug effects , Acetylesterase/metabolism , Collodion , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Hydrogen-Ion Concentration , Protein Denaturation , Serum Albumin, Bovine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...