Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biomolecules ; 9(8)2019 08 15.
Article in English | MEDLINE | ID: mdl-31443181

ABSTRACT

System-wide quantitative characterization of human neonatal Fc receptor (FcRn) properties is critical for understanding and predicting human PK (pharmacokinetics) as well as the distribution of mAbs and Fc-fusion proteins using PBPK (physiologically-based pharmacokinetic) modeling. To this end, tissue-specific FcRn expression and half-life are important model inputs. Herein, human FcRn tissue expression was measured by peptide immunoaffinity chromatography coupled with high-resolution mass spectrometry. FcRn concentrations across 14 human tissues ranged from low to 230 pmol per gram of tissue. Furthermore, the FcRn half-life was determined to be 11.1 h from a human stable isotope labelled leucine pulse labeling experiment. The spatial and temporal quantitative human FcRn data now promise to enable a refined PBPK model with improved accuracy of human PK predictions for Fc-containing biotherapeutics.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Histocompatibility Antigens Class I/genetics , Receptors, Fc/genetics , Animals , Antibodies, Monoclonal/metabolism , Gene Expression Profiling , Half-Life , Healthy Volunteers , Histocompatibility Antigens Class I/metabolism , Humans , Ligands , Rabbits , Receptors, Fc/metabolism
3.
MAbs ; 8(6): 1064-78, 2016.
Article in English | MEDLINE | ID: mdl-27232760

ABSTRACT

Therapeutic antibodies continue to develop as an emerging drug class, with a need for preclinical tools to better predict in vivo characteristics. Transgenic mice expressing human neonatal Fc receptor (hFcRn) have potential as a preclinical pharmacokinetic (PK) model to project human PK of monoclonal antibodies (mAbs). Using a panel of 27 mAbs with a broad PK range, we sought to characterize and establish utility of this preclinical animal model and provide guidance for its application in drug development of mAbs. This set of mAbs was administered to both hemizygous and homozygous hFcRn transgenic mice (Tg32) at a single intravenous dose, and PK parameters were derived. Higher hFcRn protein tissue expression was confirmed by liquid chromatography-high resolution tandem mass spectrometry in Tg32 homozygous versus hemizygous mice. Clearance (CL) was calculated using non-compartmental analysis and correlations were assessed to historical data in wild-type mouse, non-human primate (NHP), and human. Results show that mAb CL in hFcRn Tg32 homozygous mouse correlate with human (r(2) = 0.83, r = 0.91, p < 0.01) better than NHP (r(2) = 0.67, r = 0.82, p < 0.01) for this dataset. Applying simple allometric scaling using an empirically derived best-fit exponent of 0.93 enabled the prediction of human CL from the Tg32 homozygous mouse within 2-fold error for 100% of mAbs tested. Implementing the Tg32 homozygous mouse model in discovery and preclinical drug development to predict human CL may result in an overall decreased usage of monkeys for PK studies, enhancement of the early selection of lead molecules, and ultimately a decrease in the time for a drug candidate to reach the clinic.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Drug Discovery/methods , Histocompatibility Antigens Class I/genetics , Receptors, Fc/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/analysis , Chromatography, Liquid , Hemizygote , Homozygote , Humans , Macaca fascicularis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Predictive Value of Tests , Tandem Mass Spectrometry
4.
MAbs ; 8(5): 848-53, 2016 07.
Article in English | MEDLINE | ID: mdl-27104806

ABSTRACT

The neonatal Fc receptor (FcRn) is a homeostatic receptor responsible for prolonging immunoglobulin G (IgG) half-life by protecting it from lysosomal degradation and recycling it to systemic circulation. Tissue-specific FcRn expression is a critical parameter in physiologically-based pharmacokinetic (PBPK) modeling for translational pharmacokinetics of Fc-containing biotherapeutics. Using online peptide immuno-affinity chromatography coupled with high resolution mass spectrometry, we established a quantitative FcRn tissue protein expression profile in human FcRn (hFcRn) transgenic mice, Tg32 homozygous and hemizygous strains. The concentration of hFcRn across 14 tissues ranged from 3.5 to 111.2 pmole per gram of tissue. Our hFcRn quantification data from Tg32 mice will enable a more refined PBPK model to improve the accuracy of human PK predictions for Fc-containing biotherapeutics.


Subject(s)
Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class I/biosynthesis , Receptors, Fc/analysis , Receptors, Fc/biosynthesis , Animals , Chromatography, Affinity/methods , Humans , Mass Spectrometry/methods , Mice , Mice, Transgenic
5.
Anal Chem ; 88(8): 4239-47, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27012525

ABSTRACT

Neonatal Fc receptor (FcRn) is the homeostatic receptor responsible for the long half-life of endogenous IgG by protecting it from lysosomal degradation. Understanding systemic FcRn tissue expression is important to predict and design the half-life of therapeutic antibodies and Fc-coupled biotherapeutics. To this end, we measured human FcRn (hFcRn) tissue expression in Tg32, a human FcRn knock-in transgenic mouse model, for which a strong correlation of drug clearance to humans has been demonstrated. Building an hFcRn tissue expression profile in Tg32 was enabled by the development of a tissue preparation procedure composed of bead-based protein extraction and protein precipitation using acetone followed by pellet digestion with trypsin. Digests were then loaded onto an online peptide immuno-affinity flow configuration hyphenated with reversed phase nanoflow chromatography and coupled with high resolution mass spectrometry to quantify hFcRn derived peptides. The workflow allowed bypassing some of the challenges typically associated with membrane protein analysis. We demonstrated acceptable precision and bias for measuring hFcRn in tissue matrices, typically within 20% coefficient of variation and relative error. We also report hFcRn expression in several Tg32 tissues. We anticipate that establishing a quantitative approach for hFcRn in tissues will enable the systematic measurement of hFcRn concentrations to further increase the accuracy of physiologically based pharmacokinetic (PBPK) models for PK prediction of Fc-containing biotherapeutics. This is anticipated to improve the translation of pharmacokinetic data from preclinical model systems to humans.


Subject(s)
Histocompatibility Antigens Class I/analysis , Immunoassay , Peptides/immunology , Receptors, Fc/analysis , Tissue Array Analysis , Animals , Antibodies/immunology , Chromatography, Liquid , Histocompatibility Antigens Class I/immunology , Humans , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Fc/immunology
6.
J Biol Chem ; 289(4): 2170-9, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24275653

ABSTRACT

N-Linked protein glycosylation is a frequent post-translational modification that can be found in all three domains of life. In a canonical, highly conserved pathway, an oligosaccharide is transferred by a membrane-bound oligosaccharyltransferase from a lipid donor to asparagines in the sequon NX(S/T) of secreted polypeptides. The δ-proteobacterium Actinobacillus pleuropneumoniae encodes an unusual pathway for N-linked protein glycosylation. This pathway takes place in the cytoplasm and is mediated by a soluble N-glycosyltransferase (NGT) that uses nucleotide-activated monosaccharides to glycosylate asparagine residues. To characterize the process of cytoplasmic N-glycosylation in more detail, we studied the glycosylation in A. pleuropneumoniae and functionally transferred the glycosylation system to Escherichia coli. N-Linked glucose specific human sera were used for the analysis of the glycosylation process. We identified autotransporter adhesins as the preferred protein substrate of NGT in vivo, and in depth analysis of the modified sites in E. coli revealed a surprisingly relaxed peptide substrate specificity. Although NX(S/T) is the preferred acceptor sequon, we detected glycosylation of alternative sequons, including modification of glutamine and serine residues. We also demonstrate the use of NGT to glycosylate heterologous proteins. Therefore, our study could provide the basis for a novel route for the engineering of N-glycoproteins in bacteria.


Subject(s)
Actinobacillus pleuropneumoniae/enzymology , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Hexosyltransferases/metabolism , Membrane Proteins/metabolism , Actinobacillus pleuropneumoniae/genetics , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Glycosylation , Hexosyltransferases/genetics , Humans , Membrane Proteins/genetics , Protein Engineering , Substrate Specificity/physiology
7.
Nat Commun ; 4: 2627, 2013.
Article in English | MEDLINE | ID: mdl-24149797

ABSTRACT

The initial glycan transfer in asparagine-linked protein glycosylation is catalysed by the integral membrane enzyme oligosaccharyltransferase (OST). Here we study the mechanism of the bacterial PglB protein, a single-subunit OST, using chemically synthesized acceptor peptide analogues. We find that PglB can glycosylate not only asparagine but also glutamine, homoserine and the hydroxamate Asp(NHOH), although at much lower rates. In contrast, N-methylated asparagine or 2,4-diaminobutanoic acid (Dab) are not glycosylated. We find that of the various peptide analogues, only asparagine- or Dab-containing peptides bind tightly to PglB. Glycopeptide products are unable to bind, providing the driving force of product release. We find no suitably positioned residues near the active site of PglB that can activate the acceptor asparagine by deprotonation, making a general base mechanism unlikely and leaving carboxamide twisting as the most likely mechanistic proposal for asparagine activation.


Subject(s)
Amides/metabolism , Asparagine/metabolism , Bacterial Proteins/metabolism , Campylobacter lari/enzymology , Hexosyltransferases/metabolism , Membrane Proteins/metabolism , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Campylobacter jejuni/enzymology , Campylobacter jejuni/genetics , Campylobacter lari/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Expression , Glutamine/metabolism , Glycosylation , Hexosyltransferases/chemistry , Hexosyltransferases/genetics , Homoserine/metabolism , Kinetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
8.
Mol Microbiol ; 87(1): 112-31, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23163552

ABSTRACT

Bacterial carbohydrate structures play a central role in mediating a variety of host-pathogen interactions. Glycans can either elicit protective immune response or lead to escape of immune surveillance by mimicking host structures. Lipopolysaccharide (LPS), a major component on the surface of Gram-negative bacteria, is composed of a lipid A-core and the O-antigen polysaccharide. Pathogens like Neisseria meningitidis expose a lipooligosaccharide (LOS), which outermost glycans mimick mammalian epitopes to avoid immune recognition. Lewis X (Galß1-4(Fucα1-3)GlcNAc) antigens of Helicobacter pylori or of the helminth Schistosoma mansoni modulate the immune response by interacting with receptors on human dendritic cells. In a glycoengineering approach we generate human carbohydrate structures on the surface of recombinant Gram-negative bacteria, such as Escherichia coli and Salmonella enterica sv. Typhimurium that lack O-antigen. A ubiquitous building block in mammalian N-linked protein glycans is Galß1-4GlcNAc, referred to as a type-2 N-acetyllactosamine, LacNAc, sequence. Strains displaying polymeric LacNAc were generated by introducing a combination of glycosyltransferases that act on modified lipid A-cores, resulting in efficient expression of the carbohydrate epitope on bacterial cell surfaces. The poly-LacNAc scaffold was used as an acceptor for fucosylation leading to polymers of Lewis X antigens. We analysed the distribution of the carbohydrate epitopes by FACS, microscopy and ELISA and confirmed engineered LOS containing LacNAc and Lewis X repeats by MALDI-TOF and NMR analysis. Glycoengineered LOS induced pro-inflammatory response in murine dendritic cells. These bacterial strains can thus serve as tools to analyse the role of defined carbohydrate structures in different biological processes.


Subject(s)
Antigens, Surface/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Lewis X Antigen/genetics , Lewis X Antigen/metabolism , Animals , Dendritic Cells/immunology , Dendritic Cells/microbiology , Disaccharides/immunology , Escherichia coli/metabolism , Helicobacter pylori/metabolism , Host-Pathogen Interactions , Lewis X Antigen/immunology , Lipopolysaccharides/immunology , Mice , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/metabolism , Schistosoma mansoni/metabolism
9.
Nat Chem Biol ; 8(5): 434-6, 2012 Mar 25.
Article in English | MEDLINE | ID: mdl-22446837

ABSTRACT

We performed bottom-up engineering of a synthetic pathway in Escherichia coli for the production of eukaryotic trimannosyl chitobiose glycans and the transfer of these glycans to specific asparagine residues in target proteins. The glycan biosynthesis was enabled by four eukaryotic glycosyltransferases, including the yeast uridine diphosphate-N-acetylglucosamine transferases Alg13 and Alg14 and the mannosyltransferases Alg1 and Alg2. By including the bacterial oligosaccharyltransferase PglB from Campylobacter jejuni, we successfully transferred glycans to eukaryotic proteins.


Subject(s)
Disaccharides/biosynthesis , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Polysaccharides/biosynthesis , Protein Engineering , Campylobacter jejuni/enzymology , Glycosylation , Hexosyltransferases/metabolism , Mannosyltransferases/metabolism , Membrane Proteins/metabolism , N-Acetylglucosaminyltransferases/metabolism , Saccharomyces cerevisiae Proteins/metabolism
10.
J Biol Chem ; 286(40): 35267-74, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21852240

ABSTRACT

N-Linked glycosylation is a frequent protein modification that occurs in all three domains of life. This process involves the transfer of a preassembled oligosaccharide from a lipid donor to asparagine side chains of polypeptides and is catalyzed by the membrane-bound oligosaccharyltransferase (OST). We characterized an alternative bacterial pathway wherein a cytoplasmic N-glycosyltransferase uses nucleotide-activated monosaccharides as donors to modify asparagine residues of peptides and proteins. N-Glycosyltransferase is an inverting glycosyltransferase and recognizes the NX(S/T) consensus sequence. It therefore exhibits similar acceptor site specificity as eukaryotic OST, despite the unrelated predicted structural architecture and the apparently different catalytic mechanism. The identification of an enzyme that integrates some of the features of OST in a cytoplasmic pathway defines a novel class of N-linked protein glycosylation found in pathogenic bacteria.


Subject(s)
Actinobacillus pleuropneumoniae/enzymology , Bacterial Proteins/chemistry , Cytoplasm/metabolism , Glucosyltransferases/chemistry , Binding Sites , Escherichia coli/metabolism , Glucose/chemistry , Glycosylation , Hexosyltransferases/chemistry , Magnetic Resonance Spectroscopy/methods , Mass Spectrometry/methods , Membrane Proteins/chemistry , Monosaccharides/chemistry , Plasmids/metabolism , Protein Processing, Post-Translational , Protein Structure, Tertiary
11.
Bioconjug Chem ; 22(3): 488-96, 2011 Mar 16.
Article in English | MEDLINE | ID: mdl-21319730

ABSTRACT

Glycosylation is the predominant protein modification to diversify the functionality of proteins. In particular, N-linked protein glycosylation can increase the biophysical and pharmacokinetic properties of therapeutic proteins. However, the major challenges in studying the consequences of protein glycosylation on a molecular level are caused by glycan heterogeneities of currently used eukaryotic expression systems, but the discovery of the N-linked protein glycosylation system in the ε-proteobacterium Campylobacter jejuni and its functional transfer to Escherichia coli opened up the possibility to produce glycoproteins in bacteria. Toward this goal, we elucidated whether antibody fragments, a potential class of therapeutic proteins, are amenable to bacterial N-linked glycosylation, thereby improving their biophysical properties. We describe a new strategy for glycoengineering and production of quantitative amounts of glycosylated scFv 3D5 at high purity. The analysis revealed the presence of a homogeneous N-glycan that significantly increased the stability and the solubility of the 3D5 antibody fragment. The process of bacterial N-linked glycosylation offers the possibility to specifically address and alter the biophysical properties of proteins.


Subject(s)
Escherichia coli/metabolism , Nitrogen/metabolism , Single-Chain Antibodies/metabolism , Amino Acid Sequence , Animals , Antigens/immunology , Glycoproteins/immunology , Glycoproteins/metabolism , Glycosylation , Humans , Male , Mice , Models, Molecular , Molecular Sequence Data , Polysaccharides/metabolism , Protein Conformation , Protein Engineering , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacokinetics , Solubility
12.
Glycobiology ; 21(1): 45-54, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20847188

ABSTRACT

A number of proteobacteria carry the genetic information to perform N-linked glycosylation, but only the protein glycosylation (pgl) pathway of Campylobacter jejuni has been studied to date. Here, we report that the pgl gene cluster of Campylobacter lari encodes for a functional glycosylation machinery that can be reconstituted in Escherichia coli. We determined that the N-glycan produced in this system consisted of a linear hexasaccharide. We found that the oligosaccharyltransferase (OST) of C. lari conserved a predominant specificity for the primary sequence D/E-X(-1)-N-X(+1)-S/T (where X(-1) and X(+1) can be any amino acid but proline). At the same time, we observed that this enzyme exhibited a relaxed specificity toward the acceptor site and modified asparagine residues of a protein at sequences DANSG and NNNST. Moreover, C. lari pgl glycosylated a native E. coli protein. Bacterial N-glycosylation appears as a useful tool to establish a molecular description of how single-subunit OSTs perform selection of glycosyl acceptor sites.


Subject(s)
Campylobacter jejuni/enzymology , Hexosyltransferases/chemistry , Membrane Proteins/chemistry , Campylobacter lari/enzymology , Escherichia coli/enzymology , Glycosylation , Hexosyltransferases/metabolism , Membrane Proteins/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism
13.
J Biol Chem ; 284(52): 36223-36233, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19858195

ABSTRACT

Galectin CGL2 from the ink cap mushroom Coprinopsis cinerea displays toxicity toward the model nematode Caenorhabditis elegans. A mutation in a putative glycosyltransferase-encoding gene resulted in a CGL2-resistant C. elegans strain characterized by N-glycans lacking the beta1,4-galactoside linked to the alpha1,6-linked core fucose. Expression of the corresponding GALT-1 protein in insect cells was used to demonstrate a manganese-dependent galactosyltransferase activity. In vitro, the GALT-1 enzyme showed strong selectivity for acceptors with alpha1,6-linked N-glycan core fucosides and required Golgi- dependent modifications on the oligosaccharide antennae for optimal synthesis of the Gal-beta1,4-fucose structure. Phylogenetic analysis of the GALT-1 protein sequence identified a novel glycosyltransferase family (GT92) with members widespread among eukarya but absent in mammals.


Subject(s)
Caenorhabditis elegans/enzymology , Galactosyltransferases/metabolism , Oligosaccharides/biosynthesis , Phylogeny , Animals , Caenorhabditis elegans/genetics , Fucose/genetics , Fucose/metabolism , Galactosyltransferases/genetics , Mutation , Oligosaccharides/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity/physiology
14.
Glycobiology ; 19(4): 418-27, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19136585

ABSTRACT

In general, an elevated expression of beta 3-galactosyltransferase (beta 3GalT) activity contributed by beta 3GalT5 correlates well with increased biosynthesis and expression of type 1 chain (Gal beta 1-3GlcNAc beta 1-) derivatives such as Lewis A and sialyl Lewis A, which are mostly recognized as terminal epitopes and not further extended. Most known beta 3-N-acetylglucosaminyltransferases show a higher activity toward extending type 2 chain (Gal beta 1-4GlcNAc beta 1-), and an over-expression of beta 3GalT5 could suppress the formation of the type 2 chain poly-N-acetyllactosaminoglycans. The potential of extending instead the predominant type 1 chain termini synthesized under such circumstances was, however, not investigated, partly due to technical difficulty in unambiguous identification of extended type 1 chains. Using an advanced mass spectrometry-based glycomic mapping and glycan sequencing approach, we show here that type 1 chains carried on the lacto-series glycosphingolipids of colonic carcinoma cells can be extended when the endogenous beta 3GalT activity relative to competing beta 4GalT activity, as defined against a common GlcNAc beta 1-3Gal beta 1-4Glc acceptor, is sufficiently high, as found in Colo205 and SW1116, but not in DLD-1 cells. In support of this positive correlation, the lacto-series glycosphingolipids isolated from stably transfected DLD-1 clones over-expressing beta 3GalT5 were shown to comprise fucosylated dimeric type 1 chains, whereas a mock transfectant and the DLD-1 parent carried only fucosylated dimeric type 2 chains on their lactosylceramides. It suggests that while the natural expression of extended type 1 chain is likely to be determined by many contributing factors including the relative amounts of competing glycosyltransferases and the UDP-Gal level, the enhanced expression of beta 3GalT5 is sufficient to promote in vivo extension of type 1 chains by furnishing a significantly higher amount of type 1 chain precursors relative to competing type 2 chains.


Subject(s)
Antigens, CD/biosynthesis , Galactosyltransferases/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lactosylceramides/biosynthesis , Antigens, CD/genetics , Carbohydrate Conformation , Cell Line, Tumor , Colonic Neoplasms , Galactosyltransferases/genetics , Humans , Lactosylceramides/genetics , Lewis Blood Group Antigens , Mass Spectrometry , Oligosaccharides/biosynthesis , Oligosaccharides/genetics
15.
J Proteome Res ; 7(8): 3293-303, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18549263

ABSTRACT

Inbred BALB/c mouse implanted with murine tumors serves as an attractive model system for the studies of cancer biology in immuno-competent individuals. It is anticipated that tumor progression would induce notable pathophysiological consequences, some of which manifested as alteration in serum proteomic and glycomic profiles. Similar to sera derived from human cancer patients and immuno-compromised mice bearing human tumors, we show in this work that BALB/c mice of the same genetic background but bearing two distinct tumor origins both exhibited elevated expression levels of acute phase proteins including haptoglobin and serum amyloid P protein, in response to tumor progression. Such common traits are generally not informative nor qualifying as biomarkers. Additional mass spectrometry (MS)-based glycomic mapping nevertheless detected distinctive changes of sialylation pattern on the complex type N-glycans. MALDI MS/MS sequencing afforded a facile but definitive identification of an increase in internal Neu5Gcalpha2-6 sialylation on the GlcNAc of the Neu5Gc2-3Gal1-3GlcNAc terminal sequence as a common feature whereas a substitution of Neu5Gc by Neu5Ac was found to be induced by colonic but not breast tumor. A more pronounced change was similarly detected on N-glycans derived from ascitic fluids representing late tumor progression stages. We next demonstrated that such distinct change in glycotope expression can be localized to a particular protein carrier by LC-MS/MS analysis of glycopeptides. Serotransferrin was identified as one such abundant serum glycoprotein, which changed significantly not in protein expression level but in terminal glycosylation pattern.


Subject(s)
Acute-Phase Proteins/metabolism , Colonic Neoplasms/metabolism , Glycoproteins/metabolism , Mammary Neoplasms, Experimental/metabolism , Sialic Acids/metabolism , Animals , Chromatography, Liquid , Colonic Neoplasms/pathology , Female , Glycomics , Glycopeptides/metabolism , Glycosylation , Male , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
16.
J Biol Chem ; 283(24): 16455-68, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18413313

ABSTRACT

Mammalian glycan chain elongation is mostly based on extending the type 2 chain, Galbeta1-4GlcNAc, whereas the corresponding type 1 chain, Galbeta1-3GlcNAc, is not normally extended. In a broader context of developing high sensitivity mass spectrometry methodologies for glycomic identification of Le(a) versus Le(x) and linear versus branched poly-N-acetyllactosamine (polyLacNAc), we have now shown that the dimeric type 1 glycan chain, as carried on the lactosylceramides of a human colonic adenocarcinoma cell line, Colo205, not only can be further extended linearly but can likewise be branched at C6 of 3-linked Gal in a manner similar to polyLacNAc. A combination of chemical and enzymatic derivatization coupled with advanced mass spectrometry analyses afforded unambiguous identification of a complex mixture of type 1 and 2 hybrids as well as those fucosylated variants founded exclusively on linear and branched trimeric type 1 chain. We further showed by in vitro enzymatic synthesis that extended type 1 and the hybrid chains can be branched by all three forms of the human I branching enzymes (IGnT) currently identified but with lower efficiency and stringency with respect to branching site preference. Importantly, it was found that a better substrate is one that carries a Gal site for branching that is extended at the non-reducing end by a type 2 and not a type 1 unit, whereas the IGnTs are less discriminative with respect to whether the targeted Gal site is itself beta3- or beta4-linked to GlcNAc at the reducing end.


Subject(s)
Adenocarcinoma/metabolism , Colonic Neoplasms/metabolism , Lactosylceramides/chemistry , Cell Line, Tumor , Dimerization , Exons , Fucose/chemistry , Humans , Mass Spectrometry/methods , Polysaccharides/chemistry , Polysaccharides/metabolism , RNA, Messenger/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Subcellular Fractions/metabolism , Tandem Mass Spectrometry/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...