Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Mol Biol ; 1321: 323-33, 2015.
Article in English | MEDLINE | ID: mdl-26082232

ABSTRACT

The degree of sialylation of therapeutic glycoproteins affects its circulatory half-life and efficacy because incompletely sialylated glycoproteins are cleared from circulation by asialoglycoprotein receptors present in the liver cells. Mammalian expression systems, often employed in the production of these glycoprotein drugs, produce heterogeneously sialylated products. Here, we describe how to produce highly sialylated glycoproteins using a Chinese hamster ovary (CHO) cell glycosylation mutant called CHO-gmt4 with human erythropoietin (EPO) as a model glycoprotein. The protocol describes how to isolate and characterize the CHO glycosylation mutants and how to assess the sialylation of the recombinant protein using isoelectric focusing (IEF). It further describes how to inactivate the dihydrofolate reductase (DHFR) gene in these cells using zinc finger nuclease (ZFN) technology to enable gene amplification and the generation of stable cell lines producing highly sialylated EPO.


Subject(s)
Glycoproteins/genetics , Mutation/genetics , Plant Lectins/genetics , Recombinant Proteins/genetics , Animals , CHO Cells , Cell Line , Cricetulus , Erythropoietin/genetics , Gene Amplification/genetics , Glycosylation , Humans , Tetrahydrofolate Dehydrogenase/genetics
2.
Bioengineered ; 5(4): 269­73, 2014.
Article in English | MEDLINE | ID: mdl-24911584

ABSTRACT

Recombinant glycoprotein drugs require proper glycosylation for optimal therapeutic efficacy. Glycoprotein therapeutics are rapidly removed from circulation and have reduced efficacy if they are poorly sialylated. Ricinus communis agglutinin-I (RCA-I) was found highly toxic to wild-type CHO-K1 cells and all the mutants that survived RCA-I treatment contained a dysfunctional N-acetylglucosaminyltransferase I (GnT I) gene. These mutants are named CHO-gmt4 cells. Interestingly, upon restoration of GnT I, the sialylation of a model glycoprotein, erythropoietin, produced in CHO-gmt4 cells was shown to be superior to that produced in wild-type CHO-K1 cells. This addendum summarizes the applicability of this cell line, from transient to stable expression of the recombinant protein, and from a lab scale to an industrial scale perfusion bioreactor. In addition, CHO-gmt4 cells can be used to produce glycoproteins with mannose-terminated N-glycans. Recombinant glucocerebrosidase produced by CHO-gmt4 cells will not require glycan remodeling and may be directly used to treat patients with Gaucher disease. CHO-gmt4 cells can also be used to produce other glycoprotein therapeutics which target cells expressing mannose receptors.


Subject(s)
Cell Engineering , Erythropoietin/genetics , Erythropoietin/metabolism , N-Acetylglucosaminyltransferases/metabolism , N-Acetylneuraminic Acid/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Animals , Humans
3.
Biotechnol J ; 9(1): 100-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166780

ABSTRACT

Therapeutic glycoprotein drugs require a high degree of sialylation of their N-glycans for a better circulatory half-life that results in greater efficacy. It has been demonstrated that Chinese hamster ovary (CHO) glycosylation mutants lacking N-acetylglucosaminyltransferase I (GnT I), when restored by introduction of a functional GnT I, produced highly sialylated erythropoietin (EPO). We have now further engineered one of such mutants, JW152, by inactivating the dihydrofolate reductase (DHFR) gene to allow for the amplification of the EPO gene with methotrexate (MTX). Several MTX-amplified clones maintained the ability to produce highly sialylated EPO and one was selected for culture in a perfusion bioreactor that is used in an existing industrial EPO-production bioprocess. Extensive characterization of the EPO produced was performed using total sialic quantification, HPAEC-PAD and MALDI-TOF MS analyses. Our results demonstrated that the EPO produced by the mutant line exhibits superior sialylation compared to the commercially used EPO-producing CHO clone cultured under the same conditions. Therefore, this mutant has the industrial potential for producing highly sialylated recombinant EPO and potentially other recombinant glycoprotein therapeutics.


Subject(s)
Cell Engineering , Erythropoietin/genetics , Erythropoietin/metabolism , N-Acetylglucosaminyltransferases/metabolism , N-Acetylneuraminic Acid/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Animals , Bioreactors , CHO Cells , Cricetinae , Cricetulus , Erythropoietin/chemistry , Gene Amplification/drug effects , Glycosylation , Half-Life , Humans , Methotrexate/pharmacology , Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tetrahydrofolate Dehydrogenase/metabolism
4.
Glycobiology ; 22(7): 897-911, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22492235

ABSTRACT

The GDP-fucose transporter SLC35C1 critically regulates the fucosylation of glycans. Elucidation of its structure-function relationships remains a challenge due to the lack of an appropriate mutant cell line. Here we report a novel Chinese hamster ovary (CHO) mutant, CHO-gmt5, generated by the zinc-finger nuclease technology, in which the Slc35c1 gene was knocked out from a previously reported CHO mutant that has a dysfunctional CMP-sialic acid transporter (CST) gene (Slc35a1). Consequently, CHO-gmt5 harbors double genetic defects in Slc35a1 and Slc35c1 and produces N-glycans deficient in both sialic acid and fucose. The structure-function relationships of SLC35C1 were studied using CHO-gmt5 cells. In contrast to the CST and UDP-galactose transporter, the C-terminal tail of SLC35C1 is not required for its Golgi localization but is essential for generating glycans that are recognized by a fucose-binding lectin, Aleuria aurantia lectin (AAL), suggesting an important role in the transport activity of SLC35C1. Furthermore, we found that this impact can be independently contributed by a cluster of three lysine residues and a Glu-Met (EM) sequence within the C terminus. We also showed that the conserved glycine residues at positions 180 and 277 of SLC35C1 have significant impacts on AAL binding to CHO-gmt5 cells, suggesting that these conserved glycine residues are required for the transport activity of Slc35 proteins. The absence of sialic acid and fucose on Fc N-glycan has been independently shown to enhance the antibody-dependent cellular cytotoxicity (ADCC) effect. By combining these features into one cell line, we postulate that CHO-gmt5 may represent a more advantageous cell line for the production of recombinant antibodies with enhanced ADCC effect.


Subject(s)
Monosaccharide Transport Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Carbohydrate Conformation , Carbohydrate Sequence , Conserved Sequence , Cricetinae , Glycosylation , Golgi Apparatus/metabolism , HeLa Cells , Humans , INDEL Mutation , Lectins/chemistry , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/metabolism , Mutagenesis , Open Reading Frames , Peanut Agglutinin/chemistry , Protein Binding , Protein Structure, Tertiary , Protein Transport , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Staining and Labeling , Zinc Fingers
5.
Biotechnol Bioeng ; 107(3): 516-28, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20521304

ABSTRACT

One of the goals of recombinant glycoprotein production is to achieve consistent glycosylation. Although many studies have examined the changes in the glycosylation quality of recombinant protein with culture, very little has been done to examine the underlying changes in glycosylation gene expression as a culture progresses. In this study, the expression of 24 genes involved in N-glycosylation were examined using quantitative RT PCR to gain a better understanding of recombinant glycoprotein glycosylation during production processes. Profiling of the N-glycosylation genes as well as concurrent analysis of glycoprotein quality was performed across the exponential, stationary and death phases of a fed-batch culture of a CHO cell line producing recombinant human interferon-gamma (IFN-gamma). Of the 24 N-glycosylation genes examined, 21 showed significant up- or down-regulation of gene expression as the fed-batch culture progressed from exponential, stationary and death phase. As the fed-batch culture progressed, there was also an increase in less sialylated IFN-gamma glycoforms, leading to a 30% decrease in the molar ratio of sialic acid to recombinant IFN-gamma. This correlated with decreased expression of genes involved with CMP sialic acid synthesis coupled with increased expression of sialidases. Compared to batch culture, a low glutamine fed-batch strategy appears to need a 0.5 mM glutamine threshold to maintain similar N-glycosylation genes expression levels and to achieve comparable glycoprotein quality. This study demonstrates the use of quantitative real time PCR method to identify possible "bottlenecks" or "compromised" pathways in N-glycosylation and subsequently allow for the development of strategies to improve glycosylation quality.


Subject(s)
Gene Expression Profiling , Glycosyltransferases/biosynthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Glycoproteins/chemistry , Glycoproteins/metabolism , Glycosylation , Humans , Interferon-gamma/chemistry , Interferon-gamma/metabolism , N-Acetylneuraminic Acid/analysis , RNA, Messenger/analysis , RNA, Messenger/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
6.
Metab Eng ; 12(4): 360-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20346410

ABSTRACT

A large number of CHO glycosylation mutants were isolated by Ricinus communis agglutinin-I (RCA-I). Complementation tests revealed that all these mutant lines possessed a dysfunctional N-acetylglucosaminyltransferase I (GnT I) gene. Sequencing analyses on the GnT I cDNAs isolated from 16 mutant lines led to the identification of nine different single base pair mutations. Some mutations result in a premature stop codon whereas others cause a single amino acid substitution in the GnT I protein. Interestingly, expression of the normal GnT I cDNA in mutant cells resulted in enhanced sialylation of N-glycans. The sialylation of recombinant erythropoietin (EPO) produced in mutant cells that were co-transfected with GnT I was enhanced compared to that of EPO produced in wild type CHO cells. The enhanced sialylation of EPO produced by JW152 cells in the presence of GnT I over CHO-K1 cells is a result of increased sialylated glycan structures with higher antennary branching. These findings represent a new strategy that may be utilized by the biotechnology industry to produce highly sialylated therapeutic glycoproteins.


Subject(s)
Erythropoietin/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylneuraminic Acid/metabolism , Plant Lectins/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Glycosylation , Point Mutation , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...