Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Katrine T. Schjoldager is active.

Publication


Featured researches published by Katrine T. Schjoldager.


Biochimica et Biophysica Acta | 2012

Site-specific protein O-glycosylation modulates proprotein processing — Deciphering specific functions of the large polypeptide GalNAc-transferase gene family

Katrine T. Schjoldager; Henrik Clausen

BACKGROUND Posttranslational modifications (PTMs) greatly expand the function and regulation of proteins, and glycosylation is the most abundant and diverse PTM. Of the many different types of protein glycosylation, one is quite unique; GalNAc-type (or mucin-type) O-glycosylation, where biosynthesis is initiated in the Golgi by up to twenty distinct UDP-N-acetyl-α-d-galactosamine:polypeptide N-acetylgalactosaminyltransferases (GalNAc-Ts). These GalNAc-Ts are differentially expressed in cells and have different (although partly overlapping) substrate specificities, which provide for both unique functions and considerable redundancy. Recently we have begun to uncover human diseases associated with deficiencies in GalNAc-T genes (GALNTs). Thus deficiencies in individual GALNTs produce cell and protein specific effects and subtle distinct phenotypes such as hyperphosphatemia with hyperostosis (GALNT3) and dysregulated lipid metabolism (GALNT2). These phenotypes appear to be caused by deficient site-specific O-glycosylation that co-regulates proprotein convertase (PC) processing of FGF23 and ANGPTL3, respectively. SCOPE OF REVIEW Here we summarize recent progress in uncovering the interplay between human O-glycosylation and protease regulated processing and describes other important functions of site-specific O-glycosylation in health and disease. MAJOR CONCLUSIONS Site-specific O-glycosylation modifies pro-protein processing and other proteolytic events such as ADAM processing and thus emerges as an important co-regulator of limited proteolytic processing events. GENERAL SIGNIFICANCE Our appreciation of this function may have been hampered by our sparse knowledge of the O-glycoproteome and in particular sites of O-glycosylation. New strategies for identification of O-glycoproteins have emerged and recently the concept of SimpleCells, i.e. human cell lines made deficient in O-glycan extension by zinc finger nuclease gene targeting, was introduced for broad O-glycoproteome analysis.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Immature truncated O-glycophenotype of cancer directly induces oncogenic features

Prakash Radhakrishnan; Sally Dabelsteen; Frey Brus Madsen; Chiara Francavilla; Katharina L. Kopp; Catharina Steentoft; Sergey Y. Vakhrushev; J. Olsen; Lars Kai Hansen; Eric P. Bennett; Anders Woetmann; Guangliang Yin; Longyun Chen; Haiyan Song; Mads Bak; Ryan A. Hlady; Staci L. Peters; Rene Opavsky; Christenze Thode; Klaus Qvortrup; Katrine T. Schjoldager; Henrik Clausen; Michael A. Hollingsworth; Hans H. Wandall

Significance Cancer cells characteristically express proteins with immature O-glycosylation, but how and why cancer cells express immature O-glycans has remained poorly understood. Here, we report that one prevalent mechanism in pancreatic cancer is epigenetic silencing, rather than somatic mutations in a key chaperone, core 1 β3-Gal-T-specific molecular chaperone (COSMC), required for mature elongated O-glycosylation. We also demonstrate, with the use of well-defined cell systems generated by precise gene editing, that the aberrant O-glycophenotype by itself induces oncogenic features with enhanced growth and invasion. Our study suggests that the characteristic aberrant O-glycophenotype is critical for the development and behavior of cancer and further provides support for immunotherapeutic strategies that target aberrant O-glycans. Aberrant expression of immature truncated O-glycans is a characteristic feature observed on virtually all epithelial cancer cells, and a very high frequency is observed in early epithelial premalignant lesions that precede the development of adenocarcinomas. Expression of the truncated O-glycan structures Tn and sialyl-Tn is strongly associated with poor prognosis and overall low survival. The genetic and biosynthetic mechanisms leading to accumulation of truncated O-glycans are not fully understood and include mutation or dysregulation of glycosyltransferases involved in elongation of O-glycans, as well as relocation of glycosyltransferases controlling initiation of O-glycosylation from Golgi to endoplasmic reticulum. Truncated O-glycans have been proposed to play functional roles for cancer-cell invasiveness, but our understanding of the biological functions of aberrant glycosylation in cancer is still highly limited. Here, we used exome sequencing of most glycosyltransferases in a large series of primary and metastatic pancreatic cancers to rule out somatic mutations as a cause of expression of truncated O-glycans. Instead, we found hypermethylation of core 1 β3-Gal-T-specific molecular chaperone, a key chaperone for O-glycan elongation, as the most prevalent cause. We next used gene editing to produce isogenic cell systems with and without homogenous truncated O-glycans that enabled, to our knowledge, the first polyomic and side-by-side evaluation of the cancer O-glycophenotype in an organotypic tissue model and in xenografts. The results strongly suggest that truncation of O-glycans directly induces oncogenic features of cell growth and invasion. The study provides support for targeting cancer-specific truncated O-glycans with immunotherapeutic measures.


Journal of Biological Chemistry | 2010

O-glycosylation modulates proprotein convertase activation of angiopoietin-like protein 3: possible role of polypeptide GalNAc-transferase-2 in regulation of concentrations of plasma lipids.

Katrine T. Schjoldager; Malene Bech Vester-Christensen; Eric P. Bennett; Steven B. Levery; Tilo Schwientek; Wu Yin; Ola Blixt; Henrik Clausen

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 (RAPR224↓TT) for activation, and the processing site contains two potential GalNAc O-glycosylation sites immediately C-terminal (TT226). 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 Thr226 adjacent to the proprotein convertase processing site in ANGPTL3. We demonstrated that GalNAc-T2 glycosylation of Thr226 in a peptide with the RAPR224↓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.


Journal of Biological Chemistry | 2011

A systematic study of site-specific GalNAc-Type O-glycosylation modulating proprotein convertase processing

Katrine T. Schjoldager; Malene Bech Vester-Christensen; Christoffer K. Goth; Thomas Nordahl Petersen; Søren Brunak; Eric P. Bennett; Steven B. Levery; Henrik Clausen

Background: GalNAc-type O-glycosylation is emerging as a co-regulator of proprotein convertase processing of proteins. Results: O-Glycosylation within at least ±3 residues of the RXXR substrate motif for furin affected processing. Conclusion: Site-specific O-glycosylation by 20 polypeptide GalNAc transferases have wide co-regulatory functions in proprotein processing. Significance: This is the first systematic study that paves the way for wider co-regulatory functions of O-glycosylation in protein processing. 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.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Probing isoform-specific functions of polypeptide GalNAc-transferases using zinc finger nuclease glycoengineered SimpleCells

Katrine T. Schjoldager; Sergey Y. Vakhrushev; Yun Kong; Catharina Steentoft; Aaron S. Nudelman; Nis Borbye Pedersen; Hans H. Wandall; Ulla Mandel; Eric P. Bennett; Steven B. Levery; Henrik Clausen

Our knowledge of the O-glycoproteome [N-acetylgalactosamine (GalNAc) type] is highly limited. The O-glycoproteome is differentially regulated in cells by dynamic expression of a subset of 20 polypeptide GalNAc-transferases (GalNAc-Ts), and methods to identify important functions of individual GalNAc-Ts are largely unavailable. We recently introduced SimpleCells, i.e., human cell lines made deficient in O-glycan extension by zinc finger nuclease targeting of a key gene in O-glycan elongation (Cosmc), which allows for proteome-wide discovery of O-glycoproteins. Here we have extended the SimpleCell concept to include proteome-wide discovery of unique functions of individual GalNAc-Ts. We used the GalNAc-T2 isoform implicated in dyslipidemia and the human HepG2 liver cell line to demonstrate unique functions of this isoform. We confirm that GalNAc-T2–directed site-specific O-glycosylation inhibits proprotein activation of the lipase inhibitor ANGPTL3 in HepG2 cells and further identify eight O-glycoproteins exclusively glycosylated by T2 of which one, ApoC-III, is implicated in dyslipidemia. Our study supports an essential role for GalNAc-T2 in lipid metabolism, provides serum biomarkers for GalNAc-T2 enzyme function, and validates the use of GALNT gene targeting with SimpleCells for broad discovery of disease-causing deficiencies in O-glycosylation. The presented glycoengineering strategy opens the way for proteome-wide discovery of functions of GalNAc-T isoforms and their role in congenital diseases and disorders.


Glycobiology | 2015

Probing polypeptide GalNAc-transferase isoform substrate specificities by in vitro analysis

Yun Kong; Hiren J. Joshi; Katrine T. Schjoldager; Thomas Daugbjerg Madsen; Thomas A. Gerken; Malene Bech Vester-Christensen; Hans H. Wandall; Eric P. Bennett; Steven B. Levery; Sergey Y. Vakhrushev; Henrik Clausen

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.


Proceedings of the National Academy of Sciences of the United States of America | 2015

A systematic study of modulation of ADAM-mediated ectodomain shedding by site-specific O-glycosylation.

Christoffer K. Goth; Adnan Halim; Sumeet A. Khetarpal; Daniel J. Rader; Henrik Clausen; Katrine T. Schjoldager

Significance Ectodomain shedding is a central event in a range of biological processes and pathways, however the underlying mechanisms are still not fully understood. Here we present evidence that site-specific O-glycosylation regulated by individual GalNAc-Transferase isoforms, serve to coregulate ectodomain shedding, predominantly through blocking of cleavage. Our general finding is exemplified by the specific role of a single GalNAc-T isoform (GalNAc-T2) in coregulating TNF-alpha release in vitro, ex vivo in isogenic cell models, and in vivo in mouse Galnt2 knockouts. Adding the large family of GalNAc-T isoforms to regulation of ectodomain shedding substantially increase the ability to fine-tune this important process on a substrate level. Regulated shedding of the ectodomain of cell membrane proteins by proteases is a common process that releases the extracellular domain from the cell and activates cell signaling. Ectodomain shedding occurs in the immediate extracellular juxtamembrane region, which is also where O-glycosylation is often found and examples of crosstalk between shedding and O-glycosylation have been reported. Here, we systematically investigated the potential of site-specific O-glycosylation mediated by distinct polypeptide GalNAc-transferase (GalNAc-T) isoforms to coregulate ectodomain shedding mediated by the A Disintegrin And Metalloproteinase (ADAM) subfamily of proteases and in particular ADAM17. We analyzed 25 membrane proteins that are known to undergo ADAM17 shedding and where the processing sites included Ser/Thr residues within ± 4 residues that could represent O-glycosites. We used in vitro GalNAc-T enzyme and ADAM cleavage assays to demonstrate that shedding of at least 12 of these proteins are potentially coregulated by O-glycosylation. Using TNF-α as an example, we confirmed that shedding mediated by ADAM17 is coregulated by O-glycosylation controlled by the GalNAc-T2 isoform both ex vivo in isogenic cell models and in vivo in mouse Galnt2 knockouts. The study provides compelling evidence for a wider role of site-specific O-glycosylation in ectodomain shedding.


EMBO Reports | 2015

Deconstruction of O-glycosylation—GalNAc-T isoforms direct distinct subsets of the O-glycoproteome

Katrine T. Schjoldager; Hiren J. Joshi; Yun Kong; Christoffer K. Goth; Sarah Louise King; Hans H. Wandall; Eric P. Bennett; Sergey Y. Vakhrushev; Henrik Clausen

GalNAc‐type O‐glycosylation is found on most proteins trafficking through the secretory pathway in metazoan cells. The O‐glycoproteome is regulated by up to 20 polypeptide GalNAc‐Ts and the contributions and biological functions of individual GalNAc‐Ts are poorly understood. Here, we used a zinc‐finger nuclease (ZFN)‐directed knockout strategy to probe the contributions of the major GalNAc‐Ts (GalNAc‐T1 and GalNAc‐T2) in liver cells and explore how the GalNAc‐T repertoire quantitatively affects the O‐glycoproteome. We demonstrate that the majority of the O‐glycoproteome is covered by redundancy, whereas distinct subsets of substrates are modified by non‐redundant functions of GalNAc‐T1 and GalNAc‐T2. The non‐redundant O‐glycoproteome subsets and specific transcriptional responses for each isoform are related to different cellular processes; for the GalNAc‐T2 isoform, these support a role in lipid metabolism. The results demonstrate that GalNAc‐Ts have different non‐redundant glycosylation functions, which may affect distinct cellular processes. The data serves as a comprehensive resource for unique GalNAc‐T substrates. Our study provides a new view of the differential regulation of the O‐glycoproteome, suggesting that the plurality of GalNAc‐Ts arose to regulate distinct protein functions and cellular processes.


Journal of Biological Chemistry | 2011

Lectin domains of polypeptide GalNAc-transferases exhibit glycopeptide binding specificity

Johannes W. Pedersen; Eric P. Bennett; Katrine T. Schjoldager; Morten Meldal; Andreas P. Holmér; Ola Blixt; Emiliano Cló; Steven B. Levery; Henrik Clausen; Hans H. Wandall

UDP-GalNAc:polypeptide α-N-acetylgalactosaminyltransferases (GalNAc-Ts) constitute a family of up to 20 transferases that initiate mucin-type O-glycosylation. The transferases are structurally composed of catalytic and lectin domains. Two modes have been identified for the selection of glycosylation sites by GalNAc-Ts: confined sequence recognition by the catalytic domain alone, and concerted recognition of acceptor sites and adjacent GalNAc-glycosylated sites by the catalytic and lectin domains, respectively. Thus far, only the catalytic domain has been shown to have peptide sequence specificity, whereas the primary function of the lectin domain is to increase affinity to previously glycosylated substrates. Whether the lectin domain also has peptide sequence selectivity has remained unclear. Using a glycopeptide array with a library of synthetic and recombinant glycopeptides based on sequences of mucins MUC1, MUC2, MUC4, MUC5AC, MUC6, and MUC7 as well as a random glycopeptide bead library, we examined the binding properties of four different lectin domains. The lectin domains of GalNAc-T1, -T2, -T3, and -T4 bound different subsets of small glycopeptides. These results indicate an additional level of complexity in the initiation step of O-glycosylation by GalNAc-Ts.


Molecular & Cellular Proteomics | 2014

The GalNAc-type O-Glycoproteome of CHO Cells Characterized by the SimpleCell Strategy

Zhang Yang; Adnan Halim; Yoshiki Narimatsu; Hiren J. Joshi; Catharina Steentoft; Katrine T. Schjoldager; Morten Alder Schulz; Natalie Sealover; Kevin J. Kayser; Eric P. Bennett; Steven B. Levery; Sergey Y. Vakhrushev; Henrik Clausen

The Chinese hamster ovary cell (CHO) is the major host cell factory for recombinant production of biological therapeutics primarily because of its “human-like” glycosylation features. CHO is used for production of several O-glycoprotein therapeutics including erythropoietin, coagulation factors, and chimeric receptor IgG1-Fc-fusion proteins, however, some O-glycoproteins are not produced efficiently in CHO. We have previously shown that the capacity for O-glycosylation of proteins can be one limiting parameter for production of active proteins in CHO. Although the capacity of CHO for biosynthesis of glycan structures (glycostructures) on glycoproteins are well established, our knowledge of the capacity of CHO cells for attaching GalNAc-type O-glycans to proteins (glycosites) is minimal. This type of O-glycosylation is one of the most abundant forms of glycosylation, and it is differentially regulated in cells by expression of a subset of homologous polypeptide GalNAc-transferases. Here, we have genetically engineered CHO cells to produce homogeneous truncated O-glycans, so-called SimpleCells, which enabled lectin enrichment of O-glycoproteins and characterization of the O-glycoproteome. We identified 738 O-glycoproteins (1548 O-glycosites) in cell lysates and secretomes providing the first comprehensive insight into the O-glycosylation capacity of CHO (http://glycomics.ku.dk/o-glycoproteome_db/).

Collaboration


Dive into the Katrine T. Schjoldager's collaboration.

Top Co-Authors

Avatar

Henrik Clausen

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiren J. Joshi

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adnan Halim

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge