Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Proteome Res ; 12(8): 3707-20, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23848607

ABSTRACT

Hemagglutinin (HA) is the major antigen in influenza vaccines, and glycosylation is known to influence its antigenicity. Embryonated hen eggs are traditionally used for influenza vaccine production, but vaccines produced in mammalian and insect cells were recently licensed. This raises the concern that vaccines produced with different cell systems might not be equivalent due to differences in their glycosylation patterns. Thus, we developed an analytical method to monitor vaccine glycosylation through a combination of nanoLC/MS(E) and quantitative MALDI-TOF MS permethylation profiling. We then used this method to examine glycosylation of HAs from two different influenza H5N1 strains produced in five different platforms, including hen eggs, three different insect cell lines (High Five, expresSF+ and glycoengineered expresSF+), and a human cell line (HEK293). Our results demonstrated that (1) sequon utilization is not necessarily equivalent in different cell types, (2) there are quantitative and qualitative differences in the overall N-glycosylation patterns and structures produced by different cell types, (3) ∼20% of the N-glycans on the HAs produced by High Five cells are core α1,3-fucosylated structures, which may be allergenic in humans, and (4) our method can be used to monitor differences in glycosylation during the cellular glycoengineering stages of vaccine development.


Subject(s)
Glycomics , Hemagglutinins, Viral/chemistry , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/chemistry , Polysaccharides/analysis , Amino Acid Sequence , Animals , Carbohydrate Sequence , Chick Embryo , Chickens , Glycosylation , HEK293 Cells , Hemagglutinins, Viral/metabolism , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/biosynthesis , Influenza, Human/immunology , Influenza, Human/prevention & control , Molecular Sequence Data , Polysaccharides/chemistry , Sf9 Cells , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spodoptera , Zygote/virology
2.
Glycobiology ; 22(3): 417-28, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22042767

ABSTRACT

The inability to produce recombinant glycoproteins with authentic N-glycans is a limitation of many heterologous protein expression systems. In the baculovirus-insect cell system, this limitation has been addressed by glycoengineering insect cell lines with mammalian genes encoding protein N-glycosylation functions ("glycogenes") under the transcriptional control of constitutive promoters. However, a potential problem with this approach is that the metabolic load imposed by the expression of multiple transgenes could adversely impact the growth and/or stability of glycoengineered insect cell lines. Thus, we created a new transgenic insect cell line (SfSWT-5) with an inducibly mammalianized protein N-glycosylation pathway. Expression of all six glycogenes was induced when uninfected SfSWT-5 cells were cultured in growth medium containing doxycycline. Higher levels of expression and induction were observed when SfSWT-5 cells were cultured with doxycycline and infected with a baculovirus. Interestingly, there were no major differences in the short-term growth properties of SfSWT-5 cells cultured with or without doxycycline. Furthermore, there were no major differences in the phenotypic stability of these cells after continuous culture for over 300 passages with or without doxycycline. Baculovirus-infected Sf9 and SfSWT-5 cells produced about the same amounts of a model recombinant glycoprotein, but only the latter sialylated this product and sialylation was more pronounced when the cells were treated with doxycycline. In summary, this is the first report of a lower eukaryotic system with an inducibly mammalianized protein N-glycosylation pathway and the first to examine how the presumed metabolic load imposed by multiple transgene expression impacts insect cell growth and stability.


Subject(s)
Cell Line/metabolism , Genetic Engineering , Glycosyltransferases/biosynthesis , Oxo-Acid-Lyases/biosynthesis , Spodoptera/cytology , Animals , Baculoviridae/genetics , Cattle , Cell Line/enzymology , Cell Proliferation , Cloning, Molecular , Gene Expression Regulation , Genetic Vectors , Glycoproteins/biosynthesis , Glycoproteins/metabolism , Glycosylation , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Humans , Metabolic Networks and Pathways , Mice , Oxo-Acid-Lyases/genetics , Oxo-Acid-Lyases/metabolism , Phenotype , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
3.
Protein Expr Purif ; 80(2): 274-82, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21864686

ABSTRACT

In an effort to produce processed, soluble Western equine encephalitis virus (WEEV) glycoproteins for subunit therapeutic vaccine studies, we isolated twelve recombinant baculoviruses designed to express four different WEEV glycoprotein constructs under the transcriptional control of three temporally distinct baculovirus promoters. The WEEV glycoprotein constructs encoded full-length E1, the E1 ectodomain, an E26KE1 polyprotein precursor, and an artificial, secretable E2E1 chimera. The three different promoters induced gene expression during the immediate early (ie1), late (p6.9), and very late (polh) phases of baculovirus infection. Protein expression studies showed that the nature of the WEEV construct and the timing of expression both influenced the quantity and quality of recombinant glycoprotein produced. The full-length E1 product was insoluble, irrespective of the timing of expression. Each of the other three constructs yielded soluble products and, in these cases, the timing of expression was important, as higher protein processing efficiencies were generally obtained at earlier times of infection. However, immediate early expression did not yield detectable levels of every WEEV product, and expression during the late (p6.9) or very late (polh) phases of infection provided equal or higher amounts of processed, soluble product. Thus, while earlier foreign gene expression can provide higher recombinant glycoprotein processing efficiencies in the baculovirus system, in the case of the WEEV glycoproteins, earlier expression did not provide larger amounts of high quality, soluble recombinant glycoprotein product.


Subject(s)
Baculoviridae/isolation & purification , Encephalitis Virus, Western Equine/chemistry , Glycoproteins/isolation & purification , Viral Envelope Proteins/isolation & purification , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Baculoviridae/pathogenicity , Blotting, Western , Cell Line , Cloning, Molecular , Encephalitis Virus, Western Equine/genetics , Gene Expression Regulation, Viral , Genes, Viral , Genetic Vectors/genetics , Genetic Vectors/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Solubility , Spodoptera , Time Factors , Transcription, Genetic , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
4.
J Biol Chem ; 283(17): 11330-9, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18303021

ABSTRACT

Manalpha6(Manalpha3)Manbeta4GlcNAcbeta4GlcNAc-R is the core structure of the major processed protein N-glycans produced by insect cells. Ultimately, this paucimannose type structure is produced by an unusual beta-N-acetylglucosaminidase, which removes the terminal N-acetylglucosamine residue from the upstream intermediate, Manalpha6(GlcNAcbeta2Manalpha3)Manbeta4GlcNAcbeta4GlcNAc-R. Because the N-glycan processing pathways leading to the production of this intermediate are probably identical in insects and higher eukaryotes, the presence or absence of this specific, processing beta-N-acetylglucosaminidase is a key factor distinguishing the processing pathways in these two different types of organisms. Recent studies have shown that the fused lobes (fdl) gene encodes the specific, processing beta-N-acetylglucosaminidase of Drosophila melanogaster. However, there are conflicting reports on the identity of the gene encoding this enzyme in the lepidopteran insect, Spodoptera frugiperda. One has suggested that a gene alternatively designated SfGlcNAcase-3 or SfHex encodes this function, whereas another has suggested that this gene encodes a broad-spectrum beta-N-acetylglucosaminidase that functions in glycan and chitin degradation. In this study we resolved this conflict by molecularly cloning an S. frugiperda fdl ortholog (Sf-fdl) and demonstrating that it encodes a product with the substrate specificity expected of the processing beta-N-acetylglucosaminidase. Moreover, we showed that the endogenous levels of specific, processing beta-N-acetylglucosaminidase activity were significantly reduced in S. frugiperda cells engineered to express a double-stranded RNA derived from the Sf-fdl gene. These results indicate that Sf-fdl encodes the specific, processing beta-N-acetylglucosaminidase of S. frugiperda and validate our previous suggestion that the broad-spectrum beta-N-acetylglucosaminidase encoded by the SfGlcNAcase-3/SfHex gene is more likely to be involved in N-glycan and/or chitin degradation.


Subject(s)
Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Drosophila Proteins/genetics , Gene Expression Regulation , Spodoptera/genetics , Acetylglucosaminidase/biosynthesis , Acetylglucosaminidase/chemistry , Amino Acid Sequence , Animals , Cell Line , Chitin/metabolism , Drosophila Proteins/chemistry , Microsomes/metabolism , Models, Biological , Molecular Sequence Data , Phylogeny , Polysaccharides/chemistry , Polysaccharides/metabolism , RNA, Double-Stranded/metabolism , Sequence Homology, Amino Acid , Spodoptera/metabolism , Substrate Specificity
5.
Biotechnol Bioeng ; 95(1): 37-47, 2006 Sep 05.
Article in English | MEDLINE | ID: mdl-16607656

ABSTRACT

The inability to sialylate recombinant glycoproteins is a critical limitation of the baculovirus-insect cell expression system. This limitation is due, at least in part, to the absence of detectable sialyltransferase activities and CMP-sialic acids in the insect cell lines routinely used as hosts in this system. SfSWT-1 is a transgenic insect cell line encoding five mammalian glycosyltransferases, including sialyltransferases, which can contribute to sialylation of recombinant glycoproteins expressed by baculovirus vectors. However, sialylation of recombinant glycoproteins requires culturing SfSWT-1 cells in the presence of fetal bovine serum or another exogenous source of sialic acid. To eliminate this requirement and extend the utility of SfSWT-1 cells, we have isolated a new baculovirus vector, AcSWT-7B, designed to express two mammalian enzymes that can convert N-acetylmannosamine to CMP-sialic acid during the early phase of infection. AcSWT-7B was also designed to express a model recombinant glycoprotein during the very late phase of infection. Characterization of this new baculovirus vector showed that it induced high levels of intracellular CMP-sialic acid and sialylation of the recombinant N-glycoprotein upon infection of SfSWT-1 cells cultured in serum-free medium supplemented with N-acetylmannosamine. In addition, co-infection of SfSWT-1 cells with AcSWT-7B plus a conventional baculovirus vector encoding human tissue plasminogen activator resulted in sialylation of this recombinant N-glycoprotein under the same culture conditions. These results demonstrate that AcSWT-7B can be used in two different ways to support recombinant N-glycoprotein sialylation by SfSWT-1 cells in serum-free medium. Thus, AcSWT-7B can be used to extend the utility of this previously described transgenic insect cell line for recombinant sialoglycoprotein production.


Subject(s)
Baculoviridae/genetics , Baculoviridae/isolation & purification , Glycoproteins/genetics , Glycoproteins/metabolism , Sialic Acids/metabolism , Spodoptera/genetics , Spodoptera/metabolism , Animals , Cell Line , Culture Media, Serum-Free , Genetic Vectors/genetics , Recombinant Proteins/metabolism
6.
Protein Expr Purif ; 47(2): 571-90, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16427309

ABSTRACT

Sf9, a cell line derived from the lepidopteran insect, Spodoptera frugiperda, is widely used as a host for recombinant glycoprotein expression and purification by baculovirus vectors. Previous studies have shown that this cell line has one or more beta-N-acetylglucosaminidase activities that may be involved in the degradation and/or processing of N-glycoprotein glycans. However, these enzymes and their functions remain poorly characterized. Therefore, the goal of this study was to isolate beta-N-acetylglucosaminidase genes from Sf9 cells, over-express the gene products, and characterize their enzymatic activities. A degenerate PCR approach yielded three Sf9 cDNAs, which appeared to encode two distinct beta-N-acetylglucosaminidases, according to bioinformatic analyses. Baculovirus-mediated expression of these two cDNA products induced membrane-associated beta-N-acetylglucosaminidase activities in Sf9 cells, which cleaved terminal N-acetylglucosamine residues from the alpha-3 and -6 branches of a biantennary N-glycan substrate with acidic pH optima and completely hydrolyzed chitotriose to its constituent N-acetylglucosamine monomers. GFP-tagged forms of both enzymes exhibited punctate cytoplasmic fluorescence, which did not overlap with either lysosomal or Golgi-specific dyes. Together, these results indicated that the two new Sf9 genes identified in this study encode broad-spectrum beta-N-acetylglucosaminidases that appear to have unusual intracellular distributions. Their relative lack of substrate specificity and acidic pH optima are consistent with a functional role for these enzymes in glycoprotein glycan and chitin degradation, but not with a role in N-glycoprotein glycan processing.


Subject(s)
Acetylglucosaminidase/chemistry , Insect Proteins/chemistry , Spodoptera/enzymology , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Animals , Baculoviridae/genetics , Base Sequence , Cell Line , Cytoplasm/chemistry , Cytoplasm/enzymology , Cytoplasm/genetics , Golgi Apparatus/chemistry , Golgi Apparatus/enzymology , Golgi Apparatus/genetics , Insect Proteins/genetics , Lysosomes/chemistry , Lysosomes/enzymology , Lysosomes/genetics , Molecular Sequence Data , Protein Processing, Post-Translational , Protein Transport/physiology , Spodoptera/cytology , Spodoptera/genetics , Substrate Specificity/physiology
7.
Glycobiology ; 13(1): 23-34, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12634321

ABSTRACT

A novel recombinant baculovirus expression vector was used to produce His-tagged human transferrin in a transformed insect cell line (Tn5beta4GalT) that constitutively expresses a mammalian beta-1,4-galactosyltransferase. This virus encoded the His-tagged human transferrin protein in conventional fashion under the control of the very late polyhedrin promoter. In addition, to enhance the synthesis of galactosylated biantennary N-glycans, this virus encoded human beta-1,2- N-acetylglucosaminyltransferase II under the control of an immediate-early (ie1) promoter. Detailed analyses by MALDI-TOF MS, exoglycosidase digestion, and two-dimensional HPLC revealed that the N-glycans on the purified recombinant human transferrin produced by this virus-host system included four different fully galactosylated, biantennary, complex-type glycans. Thus, this study describes a novel baculovirus-host system, which can be used to produce a recombinant glycoprotein with fully galactosylated, biantennary N-glycans.


Subject(s)
N-Acetylglucosaminyltransferases/metabolism , N-Acetyllactosamine Synthase/metabolism , Oligosaccharides/chemistry , Spodoptera/enzymology , Spodoptera/genetics , Transferrin/metabolism , Animals , Baculoviridae/genetics , Carbohydrate Sequence , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , Glycoside Hydrolases/metabolism , Glycosylation , Humans , Molecular Sequence Data , Monosaccharides/chemistry , N-Acetylglucosaminyltransferases/genetics , N-Acetyllactosamine Synthase/genetics , Nucleopolyhedroviruses/genetics , Occlusion Body Matrix Proteins , Promoter Regions, Genetic/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spodoptera/virology , Transferrin/chemistry , Transferrin/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Structural Proteins
8.
Glycobiology ; 13(6): 497-507, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12626399

ABSTRACT

We have previously engineered transgenic insect cell lines to express mammalian glycosyltransferases and showed that these cells can sialylate N-glycoproteins, despite the fact that they have little intracellular sialic acid and no detectable CMP-sialic acid. In the accompanying study, we presented evidence that these cell lines can salvage sialic acids for de novo glycoprotein sialylation from extracellular sialoglycoproteins, such as fetuin, found in fetal bovine serum. This finding led us to create a new transgenic insect cell line designed to synthesize its own sialic acid and CMP-sialic acid. SfSWT-1 cells, which encode five mammalian glycosyltransferases, were transformed with two additional mammalian genes that encode sialic acid synthase and CMP-sialic acid synthetase. The resulting cell line expressed all seven mammalian genes, produced CMP-sialic acid, and sialylated a recombinant glycoprotein when cultured in a serum-free growth medium supplemented with N-acetylmannosamine. Thus the addition of mammalian genes encoding two enzymes involved in CMP-sialic acid biosynthesis yielded a new transgenic insect cell line, SfSWT-3, that can sialylate recombinant glycoproteins in the absence of fetal bovine serum. This new cell line will be widely useful as an improved host for baculovirus-mediated recombinant glycoprotein production.


Subject(s)
Cytidine Monophosphate N-Acetylneuraminic Acid/metabolism , Genetic Engineering , Glycoproteins/chemistry , Glycoproteins/metabolism , Spodoptera , Spodoptera/cytology , Spodoptera/metabolism , Animals , Animals, Genetically Modified , Baculoviridae , Cell Division , Cell Line , Genetic Vectors/genetics , Glycoproteins/biosynthesis , Glycoproteins/genetics , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Oxo-Acid-Lyases/genetics , Oxo-Acid-Lyases/metabolism , Spodoptera/virology
9.
J Virol ; 77(1): 280-90, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12477833

ABSTRACT

Autographa californica multicapsid nucleopolyhedrovirus (AcMNPV), the type species of the Nucleopolyhedrovirus genus (Baculoviridae family), has two highly unusual traits shared by several baculovirus species. First, the occlusion-derived virus (ODV) that establishes primary infection in the midgut following its ingestion by host larvae contains multiple nucleocapsids, all of which enter the same midgut cell. Second, GP64, the envelope fusion protein of the budded virus (BV) that spreads infection beyond the midgut, is synthesized both early and late during infection. We tested the hypothesis that, together, these two traits enable parental ODV nucleocapsids to bud from infected midgut cells, essentially as BV, to establish secondary infections prior to completion of viral replication within the midgut. This "pass-through" strategy would enable the virus to counter the host's principal defense, sloughing of infected midgut cells, by accelerating the onset of systemic infections. To test this hypothesis, we created an AcMNPV recombinant, AcLate21/20-64HB, that can express gp64 only during the late phase of infection (coincident with the other structural proteins). We then compared the virulence of this virus to that of a control recombinant virus that expresses gp64 in a wild-type manner. We found that when administered orally, the control virus was far more virulent and established secondary infection earlier than AcLate21/20-64HB, but when administered intrahemocoelically, infectivity and virulence of the two recombinants were identical. Our results demonstrate that early gp64 expression is a key component of a unique and highly adaptive baculovirus infection strategy.


Subject(s)
Nucleopolyhedroviruses/pathogenicity , Viral Fusion Proteins/physiology , Animals , Base Sequence , Insecta/virology , Larva/virology , Molecular Sequence Data , Nucleopolyhedroviruses/genetics , Recombination, Genetic , Virulence , Virus Assembly
10.
Protein Expr Purif ; 26(3): 438-48, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12460768

ABSTRACT

Insect cells are used routinely to express recombinant mammalian glycoproteins. However, insect protein glycosylation pathways are not well understood and appear to differ from those of mammalian cells. One way to more clearly evaluate the protein glycosylation potential of insect cells is to use the Drosophila melanogaster genome to identify genes that might encode relevant functions. These genes can then be expressed and the functions of the gene products directly evaluated by biochemical assays. In this study, we used this approach to determine the function of a putative Drosophila nucleotide sugar transporter gene. The results showed that this gene encodes a protein that can transport UDP-galactose, but not CMP-sialic acid. Thus, Drosophila encodes at least some of the infrastructure needed to produce glycoproteins with complex glycans, but this particular gene product does not directly support glycoprotein sialylation. These findings are relevant to insect cell biology and to an informed consideration of insect cell expression systems as tools for recombinant glycoprotein production.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cell Line , Cricetinae , Drosophila Proteins/biosynthesis , Drosophila Proteins/chemistry , Drosophila melanogaster/genetics , Fluorescent Antibody Technique , Gene Expression , Genes, Insect/genetics , Genetic Complementation Test , Glycosylation , Humans , Molecular Sequence Data , Monosaccharide Transport Proteins/biosynthesis , Monosaccharide Transport Proteins/chemistry , Phylogeny , Sequence Homology , Spodoptera , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...