Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Glycobiology ; 29(9): 645-656, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31172184

ABSTRACT

Complex carbohydrates serve a wide range of biological functions in cells and tissues, and their biosynthesis involves more than 200 distinct glycosyltransferases (GTfs) in human cells. The kinetic properties, cellular expression patterns and subcellular topology of the GTfs direct the glycosylation capacity of a cell. Most GTfs are ER or Golgi resident enzymes, and their specific subcellular localization is believed to be distributed in the secretory pathway according to their sequential role in the glycosylation process, although detailed knowledge for individual enzymes is still highly fragmented. Progress in quantitative transcriptome and proteome analyses has greatly advanced our understanding of the cellular expression of this class of enzymes, but availability of appropriate antibodies for in situ monitoring of expression and subcellular topology have generally been limited. We have previously used catalytically active GTfs produced as recombinant truncated secreted proteins in insect cells for generation of mouse monoclonal antibodies (mAbs) to human enzymes primarily involved in mucin-type O-glycosylation. These mAbs can be used to probe subcellular topology of active GTfs in cells and tissues as well as their presence in body fluids. Here, we present several new mAbs to human GTfs and provide a summary of our entire collection of mAbs, available to the community. Moreover, we present validation of specificity for many of our mAbs using human cell lines with CRISPR/Cas9 or zinc finger nuclease (ZFN) knockout and knockin of relevant GTfs.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity , Glycosyltransferases/immunology , Glycosyltransferases/metabolism , Mucins/metabolism , Animals , Glycosylation , Glycosyltransferases/deficiency , Glycosyltransferases/genetics , HEK293 Cells , Humans , Mice , Reproducibility of Results
2.
Eng Life Sci ; 19(7): 490-501, 2019 Jul.
Article in English | MEDLINE | ID: mdl-32625026

ABSTRACT

Single-chain variable fragments (scFv) are widely used in several fields. However, they can be challenging to purify unless using expensive Protein L-based affinity adsorbents or affinity tags. In this work, a purification process for a scFv using mixed-mode (MM) chromatography was developed by design of experiments (DoE) and proteomics for host cell protein (HCP) quantification. Capture of scFv from human embryonic kidney 293 (HEK293) cell feedstocks was performed by hydrophobic charge induction chromatography (MEP HyperCel™), whereafter polishing was performed by anion hydrophobic MM chromatography (Capto Adhere™). The DoE designs of the polishing step included both binding and flow-through modes, the latter being the standard mode for HCP removal. Chromatography with Capto Adhere™ in binding-mode with elution by linear salt gradient at pH 7.5 resulted in optimal yield, purity and HCP reduction factor of 98.9 > 98.5%, and 14, respectively. Totally, 258 different HCPs were removed, corresponding to 84% of identified HCPs. The optimized conditions enabled binding of the scFv to Capto Adhere™ below its theoretical pI, while the majority of HCPs were in the flow-through. Surface property maps indicated the presence of hydrophobic patches in close proximity to negatively charged patches that could potentially play a role in this unique selectivity.

3.
Nat Biotechnol ; 33(8): 842-4, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26192319

ABSTRACT

Production of glycoprotein therapeutics in Chinese hamster ovary (CHO) cells is limited by the cells' generic capacity for N-glycosylation, and production of glycoproteins with desirable homogeneous glycoforms remains a challenge. We conducted a comprehensive knockout screen of glycosyltransferase genes controlling N-glycosylation in CHO cells and constructed a design matrix that facilitates the generation of desired glycosylation, such as human-like α2,6-linked sialic acid capping. This engineering approach will aid the production of glycoproteins with improved properties and therapeutic potential.


Subject(s)
Glycoproteins , Protein Engineering/methods , Recombinant Proteins , Animals , CHO Cells , Cricetinae , Cricetulus , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
4.
Mol Cell Proteomics ; 14(6): 1616-29, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25813380

ABSTRACT

Circulating O-glycoproteins shed from cancer cells represent important serum biomarkers for diagnostic and prognostic purposes. We have recently shown that selective detection of cancer-associated aberrant glycoforms of circulating O-glycoprotein biomarkers can increase specificity of cancer biomarker assays. However, the current knowledge of secreted and circulating O-glycoproteins is limited. Here, we used the COSMC KO "SimpleCell" (SC) strategy to characterize the O-glycoproteome of two gastric cancer SimpleCell lines (AGS, MKN45) as well as a gastric cell line (KATO III) which naturally expresses at least partially truncated O-glycans. Overall, we identified 499 O-glycoproteins and 1236 O-glycosites in gastric cancer SimpleCells, and a total 47 O-glycoproteins and 73 O-glycosites in the KATO III cell line. We next modified the glycoproteomic strategy to apply it to pools of sera from gastric cancer and healthy individuals to identify circulating O-glycoproteins with the STn glycoform. We identified 37 O-glycoproteins in the pool of cancer sera, and only nine of these were also found in sera from healthy individuals. Two identified candidate O-glycoprotein biomarkers (CD44 and GalNAc-T5) circulating with the STn glycoform were further validated as being expressed in gastric cancer tissue. A proximity ligation assay was used to show that CD44 was expressed with the STn glycoform in gastric cancer tissues. The study provides a discovery strategy for aberrantly glycosylated O-glycoproteins and a set of O-glycoprotein candidates with biomarker potential in gastric cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Glycoproteins/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Stomach Neoplasms/metabolism , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Cell Line, Tumor , Female , Glycoproteins/blood , Humans , Male , Middle Aged , N-Acetylgalactosaminyltransferases/blood , Proteome , Stomach Neoplasms/blood , Polypeptide N-acetylgalactosaminyltransferase
5.
J Biol Chem ; 290(20): 12614-29, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25792743

ABSTRACT

Molecular details underlying regulation of starch mobilization in cereal seed endosperm remain unknown despite the paramount role of this process in plant growth. The structure of the complex between the starch debranching enzyme barley limit dextrinase (LD), hydrolyzing α-1,6-glucosidic linkages, and its endogenous inhibitor (LDI) was solved at 2.7 Å. The structure reveals an entirely new and unexpected binding mode of LDI as compared with previously solved complex structures of related cereal type family inhibitors (CTIs) bound to glycoside hydrolases but is structurally analogous to binding of dual specificity CTIs to proteases. Site-directed mutagenesis establishes that a hydrophobic cluster flanked by ionic interactions in the protein-protein interface is vital for the picomolar affinity of LDI to LD as assessed by analysis of binding by using surface plasmon resonance and also supported by LDI inhibition of the enzyme activity. A phylogenetic analysis identified four LDI-like proteins in cereals among the 45 sequences from monocot databases that could be classified as unique CTI sequences. The unprecedented binding mechanism shown here for LDI has likely evolved in cereals from a need for effective inhibition of debranching enzymes having characteristic open active site architecture. The findings give a mechanistic rationale for the potency of LD activity regulation and provide a molecular understanding of the debranching events associated with optimal starch mobilization and utilization during germination. This study unveils a hitherto not recognized structural basis for the features endowing diversity to CTIs.


Subject(s)
Enzyme Inhibitors/chemistry , Glycoside Hydrolases/chemistry , Hordeum/enzymology , Plant Proteins/chemistry , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Germination/physiology , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Hordeum/genetics , Mutagenesis, Site-Directed , Plant Proteins/antagonists & inhibitors , Plant Proteins/genetics , Plant Proteins/metabolism , Starch/chemistry , Starch/genetics , Starch/metabolism , Structure-Activity Relationship
6.
Nucleic Acids Res ; 43(9): e59, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25753669

ABSTRACT

The nuclease-based gene editing tools are rapidly transforming capabilities for altering the genome of cells and organisms with great precision and in high throughput studies. A major limitation in application of precise gene editing lies in lack of sensitive and fast methods to detect and characterize the induced DNA changes. Precise gene editing induces double-stranded DNA breaks that are repaired by error-prone non-homologous end joining leading to introduction of insertions and deletions (indels) at the target site. These indels are often small and difficult and laborious to detect by traditional methods. Here we present a method for fast, sensitive and simple indel detection that accurately defines indel sizes down to ±1 bp. The method coined IDAA for Indel Detection by Amplicon Analysis is based on tri-primer amplicon labelling and DNA capillary electrophoresis detection, and IDAA is amenable for high throughput analysis.


Subject(s)
DNA Mutational Analysis/methods , INDEL Mutation , Animals , CHO Cells , CRISPR-Cas Systems , Cell Line , Cricetulus , Electrophoresis, Capillary , Gene Targeting , Guinea Pigs , Humans , Mice , Polymerase Chain Reaction , Sequence Analysis, DNA
7.
Glycobiology ; 25(1): 55-65, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25155433

ABSTRACT

N-acetylgalactosaminyltransferase (GalNAc)-type (mucin-type) O-glycosylation is an abundant and highly diverse modification of proteins. This type of O-glycosylation is initiated in the Golgi by a large family of up to 20 homologous polypeptide GalNAc-T isoenzymes that transfer GalNAc to Ser, Thr and possibly Tyr residues. These GalNAc residues are then further elongated by a large set of glycosyltransferases to build a variety of complex O-glycan structures. What determines O-glycan site occupancy is still poorly understood, although it is clear that the substrate specificities of individual isoenzymes and the repertoire of GalNAc-Ts in cells are key parameters. The GalNAc-T isoenzymes are differentially expressed in cells and tissues in principle allowing cells to produce unique O-glycoproteomes dependent on the specific subset of isoforms present. In vitro analysis of acceptor peptide substrate specificities using recombinant expressed GalNAc-Ts has been the method of choice for probing activities of individual isoforms, but these studies have been hampered by biological validation of actual O-glycosylation sites in proteins and number of substrate testable. Here, we present a systematic analysis of the activity of 10 human GalNAc-T isoenzymes with 195 peptide substrates covering known O-glycosylation sites and provide a comprehensive dataset for evaluating isoform-specific contributions to the O-glycoproteome.


Subject(s)
N-Acetylgalactosaminyltransferases/chemistry , Peptides/chemistry , Polysaccharides/chemistry , Carbohydrate Sequence , Enzyme Assays , Gene Expression Regulation , Glycomics , Glycosylation , Golgi Apparatus/chemistry , Golgi Apparatus/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Sequence Data , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Peptides/chemical synthesis , Polysaccharides/metabolism , Proteomics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Polypeptide N-acetylgalactosaminyltransferase
8.
Proc Natl Acad Sci U S A ; 110(52): 21018-23, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24101494

ABSTRACT

The metazoan O-mannose (O-Man) glycoproteome is largely unknown. It has been shown that up to 30% of brain O-glycans are of the O-Man type, but essentially only alpha-dystroglycan (α-DG) of the dystrophin-glycoprotein complex is well characterized as an O-Man glycoprotein. Defects in O-Man glycosylation underlie congenital muscular dystrophies and considerable efforts have been devoted to explore this O-glycoproteome without much success. Here, we used our SimpleCell strategy using nuclease-mediated gene editing of a human cell line (MDA-MB-231) to reduce the structural heterogeneity of O-Man glycans and to probe the O-Man glycoproteome. In this breast cancer cell line we found that O-Man glycosylation is primarily found on cadherins and plexins on ß-strands in extracellular cadherin and Ig-like, plexin and transcription factor domains. The positions and evolutionary conservation of O-Man glycans in cadherins suggest that they play important functional roles for this large group of cell adhesion glycoproteins, which can now be addressed. The developed O-Man SimpleCell strategy is applicable to most types of cell lines and enables proteome-wide discovery of O-Man protein glycosylation.


Subject(s)
Cadherins/chemistry , Glycoproteins/chemistry , Mannose/chemistry , Proteome/chemistry , Proteomics/methods , Cell Adhesion Molecules/chemistry , Cell Line, Tumor , Glycosylation , Humans , Mass Spectrometry , Nerve Tissue Proteins/chemistry
9.
PLoS One ; 8(9): e72413, 2013.
Article in English | MEDLINE | ID: mdl-24039759

ABSTRACT

Membrane bound mucins are up-regulated and aberrantly glycosylated during malignant transformation in many cancer cells. This results in a negatively charged glycoprotein coat which may protect cancer cells from immune surveillance. However, only limited data have so far demonstrated the critical steps in glycan elongation that make aberrantly glycosylated mucins affect the interaction between cancer cells and cytotoxic effector cells of the immune system. Tn (GalNAc-Ser/Thr), STn (NeuAcα2-6GalNAc-Ser/Thr), T (Galß1-3GalNAc-Ser/Thr), and ST (NeuAcα2-6Galß1-3GalNAc-Ser/Thr) antigens are recognized as cancer associated truncated glycans, and are expressed in many adenocarcinomas, e.g. breast- and pancreatic cancer cells. To investigate the role of the cancer associated glycan truncations in immune-mediated killing we created glyco-engineered breast- and pancreatic cancer cells expressing only the shortest possible mucin-like glycans (Tn and STn). Glyco-engineering was performed by zinc finger nuclease (ZFN) knockout (KO) of the Core 1 enzyme chaperone COSMC, thereby preventing glycan elongation beyond the initial GalNAc residue in O-linked glycans. We find that COSMC KO in the breast and pancreatic cancer cell lines T47D and Capan-1 increases sensitivity to both NK cell mediated antibody-dependent cellular-cytotoxicity (ADCC) and cytotoxic T lymphocyte (CTL)-mediated killing. In addition, we investigated the association between total cell surface expression of MUC1/MUC16 and NK or CTL mediated killing, and observed an inverse correlation between MUC16/MUC1 expression and the sensitivity to ADCC and CTL-mediated killing. Together, these data suggest that up-regulation of membrane bound mucins protects cells from immune mediated killing, and that particular glycosylation steps, as demonstrated for glycan elongation beyond Tn and STn, can be important for fine tuning of the immune escape mechanisms in cancer cells.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Antigens, Tumor-Associated, Carbohydrate/metabolism , CA-125 Antigen/metabolism , Membrane Proteins/metabolism , Mucin-1/metabolism , T-Lymphocytes, Cytotoxic/physiology , Tumor Escape , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival , Cetuximab , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gene Knockout Techniques , Glycosylation , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Polysaccharides/metabolism
10.
EMBO J ; 32(10): 1478-88, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23584533

ABSTRACT

Glycosylation is the most abundant and diverse posttranslational modification of proteins. While several types of glycosylation can be predicted by the protein sequence context, and substantial knowledge of these glycoproteomes is available, our knowledge of the GalNAc-type O-glycosylation is highly limited. This type of glycosylation is unique in being regulated by 20 polypeptide GalNAc-transferases attaching the initiating GalNAc monosaccharides to Ser and Thr (and likely some Tyr) residues. We have developed a genetic engineering approach using human cell lines to simplify O-glycosylation (SimpleCells) that enables proteome-wide discovery of O-glycan sites using 'bottom-up' ETD-based mass spectrometric analysis. We implemented this on 12 human cell lines from different organs, and present a first map of the human O-glycoproteome with almost 3000 glycosites in over 600 O-glycoproteins as well as an improved NetOGlyc4.0 model for prediction of O-glycosylation. The finding of unique subsets of O-glycoproteins in each cell line provides evidence that the O-glycoproteome is differentially regulated and dynamic. The greatly expanded view of the O-glycoproteome should facilitate the exploration of how site-specific O-glycosylation regulates protein function.


Subject(s)
Glycoproteins/analysis , N-Acetylgalactosaminyltransferases/metabolism , Proteomics/methods , Algorithms , Amino Acid Motifs , Cell Line, Tumor , Genetic Engineering/methods , Glycoproteins/metabolism , Glycosylation , Humans , N-Acetylgalactosaminyltransferases/genetics , Polypeptide N-acetylgalactosaminyltransferase
11.
J Biol Chem ; 288(20): 14636-14646, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23546879

ABSTRACT

The colonic epithelial surface is protected by an inner mucus layer that the commensal microflora cannot penetrate. We previously demonstrated that Entamoeba histolytica secretes a protease capable of dissolving this layer that is required for parasite penetration. Here, we asked whether there are bacteria that can secrete similar proteases. We screened bacterial culture supernatants for such activity using recombinant fragments of the MUC2 mucin, the major structural component, and the only gel-forming mucin in the colonic mucus. MUC2 has two central heavily O-glycosylated mucin domains that are protease-resistant and has cysteine-rich N and C termini responsible for polymerization. Culture supernatants of Porphyromonas gingivalis, a bacterium that secretes proteases responsible for periodontitis, cleaved the MUC2 C-terminal region, whereas the N-terminal region was unaffected. The active enzyme was isolated and identified as Arg-gingipain B (RgpB). Two cleavage sites were localized to IR↓TT and NR↓QA. IR↓TT cleavage will disrupt the MUC2 polymers. Because this site has two potential O-glycosylation sites, we tested whether recombinant GalNAc-transferases (GalNAc-Ts) could glycosylate a synthetic peptide covering the IRTT sequence. Only GalNAc-T3 was able to glycosylate the second Thr in IRTT, rendering the sequence resistant to cleavage by RgpB. Furthermore, when GalNAc-T3 was expressed in CHO cells expressing the MUC2 C terminus, the second threonine was glycosylated, and the protein became resistant to RgpB cleavage. These findings suggest that bacteria can produce proteases capable of dissolving the inner protective mucus layer by specific cleavages in the MUC2 mucin and that this cleavage can be modulated by site-specific O-glycosylation.


Subject(s)
Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/metabolism , Mucin-2/metabolism , Porphyromonas gingivalis/enzymology , Amino Acid Sequence , Animals , CHO Cells , Chromatography , Colitis/microbiology , Colon/metabolism , Cricetinae , Epithelium/metabolism , Gingipain Cysteine Endopeptidases , Glycosylation , Humans , Mass Spectrometry , Molecular Sequence Data
12.
Mol Cell Proteomics ; 12(4): 932-44, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23399548

ABSTRACT

Characterizing protein GalNAc-type O-glycosylation has long been a major challenge, and as a result, our understanding of this glycoproteome is particularly poor. Recently, we presented a novel strategy for high throughput identification of O-GalNAc glycosites using zinc finger nuclease gene-engineered "SimpleCell" lines producing homogeneous truncated O-glycosylation. Total lysates of cells were trypsinized and subjected to lectin affinity chromatography enrichment, followed by identification of GalNAc O-glycopeptides by nLC-MS/MS, with electron transfer dissociation employed to specify sites of O-glycosylation. Here, we demonstrate a substantial improvement in the SimpleCell strategy by including an additional stage of lectin affinity chromatography on secreted glycoproteins from culture media (secretome) and by incorporating pre-fractionation of affinity-enriched glycopeptides via IEF before nLC-MS/MS. We applied these improvements to three human SimpleCells studied previously, and each yielded a substantial increase in the number of O-glycoproteins and O-glycosites identified. We found that analysis of the secretome was an important independent factor for increasing identifications, suggesting that further substantial improvements can also be sought through analysis of subcellular organelle fractions. In addition, we uncovered a substantial nonoverlapping set of O-glycoproteins and O-glycosites using an alternative protease digestion (chymotrypsin). In total, the improvements led to identification of 259 glycoproteins, of which 152 (59%) were novel compared with our previous strategy using the same three cell lines. With respect to individual glycosites, we identified a total of 856 sites, of which 508 (59%) were novel compared with our previous strategy; this includes four new identifications of O-GalNAc attached to tyrosine. Furthermore, we uncovered ≈ 220 O-glycosites wherein the peptides were clearly identified, but the glycosites could not be unambiguously assigned to specific positions. The improved strategy should greatly facilitate high throughput characterization of the human GalNAc-type O-glycoproteome as well as be applicable to analysis of other O-glycoproteomes.


Subject(s)
Acetylgalactosamine/metabolism , Glycoproteins/metabolism , Protein Processing, Post-Translational , Proteome/metabolism , Tandem Mass Spectrometry/methods , Acetylgalactosamine/chemistry , Amino Acid Sequence , Carbohydrate Conformation , Carbohydrate Sequence , Cell Line, Tumor , Chymotrypsin/chemistry , Glycoproteins/chemistry , Glycoproteins/isolation & purification , Glycosylation , Humans , Isoelectric Focusing , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptide Mapping , Proteolysis , Proteome/chemistry , Proteome/isolation & purification , Trypsin/chemistry , Tyrosine/metabolism
13.
Nat Methods ; 8(11): 977-82, 2011 Oct 09.
Article in English | MEDLINE | ID: mdl-21983924

ABSTRACT

Zinc-finger nuclease (ZFN) gene targeting is emerging as a versatile tool for engineering of multiallelic gene deficiencies. A longstanding obstacle for detailed analysis of glycoproteomes has been the extensive heterogeneities in glycan structures and attachment sites. Here we applied ZFN targeting to truncate the O-glycan elongation pathway in human cells, generating stable 'SimpleCell' lines with homogenous O-glycosylation. Three SimpleCell lines expressing only truncated GalNAcα or NeuAcα2-6GalNAcα O-glycans were produced, allowing straightforward isolation and sequencing of GalNAc O-glycopeptides from total cell lysates using lectin chromatography and nanoflow liquid chromatography-mass spectrometry (nLC-MS/MS) with electron transfer dissociation fragmentation. We identified >100 O-glycoproteins with >350 O-glycan sites (the great majority previously unidentified), including a GalNAc O-glycan linkage to a tyrosine residue. The SimpleCell method should facilitate analyses of important functions of protein glycosylation. The strategy is also applicable to other O-glycoproteomes.


Subject(s)
Carbohydrates/chemistry , Proteome , Amino Acid Sequence , Base Sequence , Cell Line , Chromatography, Liquid , Glycosylation , Humans , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Tandem Mass Spectrometry
14.
J Biol Chem ; 286(46): 40122-32, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21937429

ABSTRACT

Site-specific GalNAc-type O-glycosylation is emerging as an important co-regulator of proprotein convertase (PC) processing of proteins. PC processing is crucial in regulating many fundamental biological pathways and O-glycans in or immediately adjacent to processing sites may affect recognition and function of PCs. Thus, we previously demonstrated that deficiency in site-specific O-glycosylation in a PC site of the fibroblast growth factor, FGF23, resulted in marked reduction in secretion of active unprocessed FGF23, which cause familial tumoral calcinosis and hyperostosis hyperphosphatemia. GalNAc-type O-glycosylation is found on serine and threonine amino acids and up to 20 distinct polypeptide GalNAc transferases catalyze the first addition of GalNAc to proteins making this step the most complex and differentially regulated steps in protein glycosylation. There is no reliable prediction model for O-glycosylation especially of isolated sites, but serine and to a lesser extent threonine residues are frequently found adjacent to PC processing sites. In the present study we used in vitro enzyme assays and ex vivo cell models to systematically address the boundaries of the region within site-specific O-glycosylation affect PC processing. The results demonstrate that O-glycans within at least ±3 residues of the RXXR furin cleavage site may affect PC processing suggesting that site-specific O-glycosylation is a major co-regulator of PC processing.


Subject(s)
Fibroblast Growth Factors/metabolism , Furin/metabolism , Protein Modification, Translational/physiology , Protein Processing, Post-Translational/physiology , Amino Acid Motifs , Animals , CHO Cells , Cricetinae , Cricetulus , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Furin/genetics , Glycosylation , Humans , Proteolysis
15.
J Biol Chem ; 285(47): 36293-303, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20837471

ABSTRACT

The angiopoietin-like protein 3 (ANGPTL3) is an important inhibitor of the endothelial and lipoprotein lipases and a promising drug target. ANGPTL3 undergoes proprotein convertase processing (RAPR(224)↓TT) for activation, and the processing site contains two potential GalNAc O-glycosylation sites immediately C-terminal (TT(226)). We developed an in vivo model system in CHO ldlD cells that was used to show that O-glycosylation in the processing site blocked processing of ANGPTL3. Genome-wide SNP association studies have identified the polypeptide GalNAc-transferase gene, GALNT2, as a candidate gene for low HDL and high triglyceride blood levels. We hypothesized that the GalNAc-T2 transferase performed critical O-glycosylation of proteins involved in lipid metabolism. Screening of a panel of proteins known to affect lipid metabolism for potential sites glycosylated by GalNAc-T2 led to identification of Thr(226) adjacent to the proprotein convertase processing site in ANGPTL3. We demonstrated that GalNAc-T2 glycosylation of Thr(226) in a peptide with the RAPR(224)↓TT processing site blocks in vitro furin cleavage. The study demonstrates that ANGPTL3 activation is modulated by O-glycosylation and that this step is probably controlled by GalNAc-T2.


Subject(s)
Angiopoietins/metabolism , Glycopeptides/metabolism , Lipids/blood , N-Acetylgalactosaminyltransferases/metabolism , Proprotein Convertases/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins , Angiopoietins/genetics , Animals , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Furin/metabolism , Glycosylation , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Polypeptide N-acetylgalactosaminyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL
...