Skip to main content
Erschienen in: Journal of Inherited Metabolic Disease 3/2018

Open Access 01.03.2018 | Glycomics

Clinical glycomics for the diagnosis of congenital disorders of glycosylation

verfasst von: Nurulamin Abu Bakar, Dirk J. Lefeber, Monique van Scherpenzeel

Erschienen in: Journal of Inherited Metabolic Disease | Ausgabe 3/2018

Abstract

Clinical glycomics comprises a spectrum of different analytical methodologies to analyze glycan structures, which provides insights into the mechanisms of glycosylation. Within clinical diagnostics, glycomics serves as a functional readout of genetic variants, and can form a basis for therapy development, as was described for PGM1-CDG. Integration of glycomics with genomics has resulted in the elucidation of previously unknown disorders of glycosylation, namely CCDC115-CDG, TMEM199-CDG, ATP6AP1-CDG, MAN1B1-CDG, and PGM1-CDG. This review provides an introduction into protein glycosylation and presents the different glycomics methodologies ranging from gel electrophoresis to mass spectrometry (MS) and from free glycans to intact glycoproteins. The role of glycomics in the diagnosis of congenital disorders of glycosylation (CDG) is presented, including a diagnostic flow chart and an overview of glycomics data of known CDG subtypes. The review ends with some future perspectives, showing upcoming technologies as system wide mapping of the N- and O-glycoproteome, intact glycoprotein profiling and analysis of sugar metabolism. These new advances will provide additional insights and opportunities to develop personalized therapy. This is especially true for inborn errors of metabolism, which are amenable to causal therapy, because interventions through supplementation therapy can directly target the pathogenesis at the molecular level.
Hinweise
Responsible Editor: Jaak Jaeken
Abkürzungen
2-AA
2-aminobenzoic acid
2-AB
2-aminobenzamide
2-AP
2-aminopyridine
AFP-L3
fucosylated serum alpha-fetoprotein
Apo C-III
apolipoprotein C-III
ARCL2
autosomal recessive cutis laxa type-2
Asn
asparagine
CDG
congenital disorders of glycosylation
CDG-I
CDG type 1
CDG-II
CDG type 2
COG
conserved oligomeric Golgi
COPD
chronic obstructive pulmonary disease (COPD)
ER
endoplasmic Reticulum
ESI
electrospray Ionization
FDA
Food and Drug Administration
FT
Fourier transform
Fuc
fucose
GA
Golgi Apparatus
GAGs
glycosaminoglycans
Gal
galactose
GalNAc
N-acetylgalactosamine
Glc
glucose
GlcA
glucuronic acid
GlcNAc
N-acetylglucosamine
HILIC
hydrophilic interaction liquid chromatography
IEF
isoelectric focusing
LC
liquid chromatography
LLO
lipid-linked oligosaccharides
m/z
mass over charge ratio
MALDI
matrix assisted laser desorption ionization
Man
mannose
MS
mass spectrometry
NGS
next-generation sequencing
PGC
porous graphitized carbon
PNGaseF
N-glycosidase F
QTOF
quadrupole time-of-flight
Ser
serine
Sia
sialic acid
Thr
threonine
UV
ultraviolet
TOF
Time Of Flight
WES
whole exome sequencing

Introduction

Technological advances in science are creating a revolution in the world of clinical diagnostics for rare metabolic disorders. In many cases, establishing a diagnosis via the traditional care route is a complex, lengthy process involving multiple consultations by various clinical specialists. The fact that an omics technique greatly reduces this turnaround time and increases the diagnostic yield has recently been shown for a cohort of 150 patients presenting with complex neurological disorders of suspected genetic origin. They compared the number of solved cases applying either whole exome sequencing (WES) alone (29.3%) or the standard care pathway (7.3%). This improvement in diagnostic yield is significant, without increasing costs compared to the standard diagnostic trajectory (Vissers et al 2017). However, the percentage of solved cases of about 30% is still quite low, which is likely caused by either the lack of sequence coverage of the variant, by disease causes outside the coding sequences, or the presence of too many “variants of unknown significance”. In this light, the important connection of genomics with functional -omics methodologies in the diagnosis of metabolic disorders is recognized more and more. For example, a combination of WES and deep clinical phenotyping was applied to 41 patients with intellectual developmental disorder and unexplained metabolic abnormalities, which resulted in a diagnosis for 28 patients (68%), and a test for targeted intervention on 18 patients (44%) (Van Karnebeek et al 2016). For congenital disorders of glycosylation (CDG), many cases were unsolved until the inclusion of glycomics into clinical practice, to present the functional defect (Jansen et al 2016a; Jansen et al 2016b; Jansen et al 2016c; Van Damme et al 2016; Van Scherpenzeel et al 2014; Tegtmeyer et al 2014; Carss et al 2013; Iqbal et al 2013). The most important advantage of integrating functional omics with genomics in the field of inherited metabolic disorders is the opportunities for therapy, which do arise from insights into functional, biochemical pathways. Initial evidence was published for PGM1-CDG (Tegtmeyer et al 2014; Wong et al 2017). This review discusses glycomics, its role in CDG diagnostics, presents different methodologies, and ends with future perspectives, in which the horizon of the diagnostic laboratory needs to be broadened to functionally understand new genetic defects.

Introduction in glycosylation

Protein glycosylation is considered to be the most common post-translational modification and is ubiquitously present (Moremen et al 2012). Glycosylation is a non-template driven process involving multiple competing enzymes (e.g., glycosidases and glycosyltransferases) in the endoplasmic reticulum (ER) and the Golgi apparatus (GA) as glycoproteins traffic and mature through the secretory pathway. Therefore, glycan structures are highly diverse, with multiple possibilities for branching and linkage (micro-heterogeneity) and differences in site occupancy (macro-heterogeneity). Glycans are known to have many important biological functions, such as cell-cell, macromolecular (e.g., antibody) and pathogen interactions, protein secretion, protein signaling, and protein folding (Moremen et al 2012; Defaus et al 2014).
Glycosylation changes have been identified in various diseases, ranging from monogenetic inherited disorders such as CDG (Jaeken 2011) to multiple types of malignancies; e.g., ovarian (Leiserowitz et al 2007), colon (Sethi et al 2014) and breast cancer (Lee et al 2014), but also observed in other pathological situations, such as cirrhosis, hepatitis, and neurodegenerative diseases such as Alzheimer’s (Palmigiano et al 2016) and Parkinson disease (Russell et al 2017). Therefore, the analysis of glycosylation in complex biological matrices, which is called glycomics, has become one of the popular -omics after the era of genomics and proteomics, especially for biomarker discovery, treatment monitoring, and also to understand disease mechanisms. This has become more relevant and promising since the majority of tumor biomarkers endorsed by the Food and Drug Administration (FDA) are glycoproteins (Fuzery et al 2013). Most of the markers are only judged based on their expression level. However, for two glycoproteins, glycosylation is included as a biomarker in routine clinical diagnostics, namely carbohydrate-deficient transferrin for the detection of CDG and alcohol abuse, and fucosylated serum alpha-fetoprotein (AFP-L3) for the early diagnosis of hepatocellular carcinoma (Li et al 2001; Leerapun et al 2007). It was shown that by including glycosylation in the analysis, specificity and sensitivity were increased over the native alpha-fetoprotein, and intact transferrin mass spectrometry resulted in the direct diagnosis of several CDG-II subtypes (van Scherpenzeel et al 2015).
There are three types of protein glycosylation in mammalian cells, namely N-, O-, and C-linked glycosylation each having their own subclasses (Moremen et al 2012). The first two are the most common types of protein glycosylation in human cells (Wang et al 2014), and are therefore also the most studied and best characterized types. This review on clinical glycomics will be restricted to these two most common types. N-linked glycans are attached to the polypeptide via amide linkages to asparagine (Asn) side chains, while O-glycans are attached through glycosidic linkages to side chains of serine (Ser) or threonine (Thr). N-glycans consist of several monosaccharides, such as N-acetylglucosamine (GlcNAc), mannose (Man), fucose (Fuc), galactose (Gal), glucose (Glc), and sialic acid (Sia), which are built in a specific order (Cummings 2009). Briefly, N-glycans are assembled in the ER before being further processed and modified in the GA producing three types of N-glycans, namely complex, hybrid, and high mannose (Fig. 1a).
In contrast to N-glycosylation, O-glycosylation is assembled directly onto serine or threonine residues without pre-assembly. Mucin type O-glycans are the most well-known type of O-glycans with N-acetylgalactosamine (GalNAc) at the reducing end. Another large group of O-glycans are the glycosaminoglycans (GAGs) on proteoglycans. GAGs are long, unbranched carbohydrates containing repeated GalNAc or GlcNAc residues combined with glucuronic acid (GlcA) or Gal residues. The other five types of O-glycosylation are O-linked GlcNAc, -Gal, −Man, −Glc, and -Fuc. In all of these classes, one or more genetic deficiencies have been identified (Wopereis et al 2006, Hennet 2012, Freeze et al 2014).

Clinical glycomics methodologies

For protein N-glycan analysis, the sample preparation usually starts with the addition of the enzyme N-glycosidase F (PNGaseF), to cleave the N-glycans from the proteins. The method is applicable to purified proteins, as well as on complex biological samples, such as blood, urine, and CSF. In addition to the analysis of protein-released N-glycans, advances in technology allow the analysis of intact glycoproteins, which is fast, robust, does not require enzymatic digestion, and enables high-throughput analysis (Fig. 1b).
Traditionally, derivatization is the common step after releasing N-glycans from proteins, because this type of modification increases the sensitivity of glycan detection as native N-glycans have no significant ultraviolet (UV) absorbance. One could make use of the single reactive carbonyl group at the reducing end of the glycan and perform reductive amination with UV or fluorescent tags, such as 2-aminobenzoic acid (2-AA), 2-aminobenzamide (2-AB), and 2-aminopyridine (2-AP) (Ruhaak et al 2010; Pabst et al 2009), or permethylate glycans (Ciucanu and Kerek 1984; Kang et al 2008), thereby replacing all the hydroxyl groups with methyl ethers. Recently, a method for sialic acid esterification was reported, which not only enhances the stability of acidic glycans during analysis, but also distinguishes alpha2,3 and alpha2,6 sialic acid linkage (Wheeler et al 2009; Reiding et al 2014). Disadvantages of derivatization steps are that they could be incomplete, which creates a biased analysis, as well as the need for additional purification procedures which can cause sample loss (Pabst and Altmann 2011).
Several electrophoretic and chromatographic techniques have been established to separate complex N-glycan mixtures, such as capillary electrophoresis, ion exchange chromatography, hydrophilic interaction liquid chromatography (HILIC), liquid chromatography (LC), and porous graphitized carbon (PGC) (Melmer et al 2011). Recently, PGC-LC-MS has emerged as a popular platform to efficiently separate native glycans without derivatization steps which greatly reduced turnaround time of glycomics sample preparation and enabled the separation of isomeric N-glycans (alpha and beta anomers). Native N-glycan analysis using the PGC-LC approach has been successfully applied for glycan biomarker discovery in ovarian cancer (Hua et al 2013), colorectal cancer (Sethi et al 2015), and lung cancer (Ruhaak et al 2016).
Electrospray ionization (ESI) and matrix assisted laser desorption ionization (MALDI) are the two most common ionization techniques for MS glycan analysis. Both ionization techniques are very sensitive to analyze proteins, peptides, glycans, and lipids as low as picomolar concentrations. ESI is considered as a soft ionization technique, and is able to create multiple charged ions for biomolecules, such as proteins and peptides, which enables these large molecules to be analyzed within the mass over charge ratio (m/z) ranges of the instrument. When coupled to LC and including standards for calibration, ESI is more reliable for quantitative analysis than MALDI (El-Aneed et al 2009; Sturiale et al 2011).
In comparison to ESI, MALDI has the advantages of being robust, fast, and easy to operate. Additionally, MALDI allows analyzing the acidic N-glycans in relative quantities compared to the neutral glycans when derivatization steps, such as permethylation and ethyl esterification, were performed prior to MS to neutralize and stabilize sialic acids. Fourier transform (FT-) and time of flight (TOF)-MS are currently the detectors with the highest resolution (Wuhrer et al 2004; Pabst and Altmann 2008). The second might be preferable considering its capability to analyze large molecules like intact proteins, and the high maintenance costs of FT detectors.
Unfortunately, there is no general O-glycosidase available for enzymatic release of all species of O-glycans. For O-glycan profiling, chemical release by hydrazinolysis or (reductive) beta-elimination needs to be performed, which are harsh methods that always yield some side products. The reductive beta-elimination is the most clean and therefore the most commonly used method, since the simultaneous reduction of the terminal sugar prevents the peeling of the glycan due to the alkaline conditions (degradation from its reducing end) (Thaysen-Andersen and Packer 2014). There are few methods published to simultaneously profile plasma N- and O-linked glycosylation of CDG patients (Faid et al 2007; Xia et al 2013). Because of their limited applicability so far, congenital disorders in the biosynthesis of O-glycans have been identified by genetic approaches. However, for the mucin type O-glycans, intact apolipoprotein C-III (Apo C-III) profiling is available in a diagnostic setting (Wopereis et al 2003). One way to circumvent the need to use harsh conditions to cleave O-glycans is to analyze native glycopeptides, with the additional advantage of keeping the information on the attachment site intact (Hoffmann et al 2016). System-wide mapping of the N- and O-glycoproteome is envisioned in a good review of Thaysen-Andersen and Packer in 2014 (Thaysen-Andersen and Packer 2014). This new frontier in proteomics has the merits of high-resolution MS, complementary fragmentation techniques, and bioinformatic tools.

Application of clinical glycomics for CDG diagnostics

CDG are a group of genetic defects with abnormal glycosylation of proteins, lipids or both. According to the current guidelines (Jaeken et al 2009), CDG is classified into: protein N-glycosylation defects, protein O-glycosylation defects, glycosphingolipid and glycosylphosphatidylinositol anchor glycosylation defects, and multiple glycosylation pathway defects. Currently, 105 distinct types of CDG have been reported (Jaeken and Peanne 2017; Peanne et al 2017). However, with the emergence of next generation genomics technologies, there were also genes identified that do not directly involve the glycosylation biosynthesis pathway, for example defects in nucleotide sugar transport, defects of vesicular transport, defects in O-mannosylation, O-GlcNAcylation or defects in dolichol biosynthesis, which lead to abnormal protein glycosylation (Hennet 2012). CDG are commonly classified by the localization of the genetic defect (Jaeken et al 2009), in which CDG type 1 (CDG-I) refers to deficient synthesis of the precursor glycan in the ER, also including defects in the cytosol (e.g., PMM2- and PMI-CDG) and the transfer of the LLO to the protein, including various OST defects (e.g., DDOST, SST3A, and SST3B). CDG type 2 (CDG-2) occurs in the GA resulting in modified glycans on the glycoprotein (Lefeber et al 2011). Since the majority of these glycosylation disorders showed a defect in the biosynthetic pathway of protein N-glycosylation (Freeze et al 2014), the analysis of plasma N-glycans using MS plays a significant role in CDG research and diagnostics. The two most common clinical glycomics approaches for CDG characterization are global (N-glycans released from whole serum or plasma glycoproteins, further abbreviated as plasma glycomics) and protein-specific (e.g., intact transferrin MS & Apo C-III MS) glycoprofiling (Fig. 1).
The first application of glycomics was ESI-MS for CDG-I characterization. Transferrin profiles using ESI-MS on PMM2-CDG patients clearly showed two abnormal peaks corresponding to lack of one and both complete glycans (Wada et al 1992a; Wada et al 1992b). ESI-MS of transferrin became more ‘mature’ in 2001, when coupled to LC-MS to create a fast and high-throughput screening test for CDG (Lacey et al 2001). A decade later, the emergence of advanced quadrupole time-of-flight (QTOF) detection in combination with nanoLC-ESI-MS enabled the development of high resolution intact transferrin glycoprofiling which in turn improved the CDG diagnostics (van Scherpenzeel et al 2015). Normal transferrin IEF profiles have been observed in some CDG-I and -II cases, such as ALG14-CDG, ALG11-CDG, MOGS-CDG, SLC35A3-CDG, and SLC35C1-CDG (Lefeber 2016; Al Teneiji et al 2017), as well as in some defects in sugar metabolism like GNE-CDG (Voermans et al 2010), NANS-CDG (Van Karnebeek et al 2016), PGM3-CDG (Stray-Pedersen et al 2014), and also in a tissue-specific and GA homeostasis defect of VPS13B-CDG/Cohen syndrome (Duplomb et al 2014).
Transferrin glycopeptide analysis, which was established by Wada et al in 2004 is important to reveal the structural information on both glycans and proteins. For example, the hybrid type glycan which is diagnostic for GA mannosidase defects of MAN1B1-CDG can only be found at Asn-432 but not on Asn-630 (Wada 2016). For O-glycan analysis, Wada et al developed plasma Apo C-III MALDI MS (Wada et al 2012).
For plasma glycomics, MALDI MS was used to analyze permethylated total plasma N-glycans (Guillard et al 2009), or combined plasma N-glycan and O-glycans MS (Xia et al 2013). MALDI MS was also suitable to analyze N-glycans released from fibroblast homogenates of SLC35A3 and healthy controls (Edvardson et al 2013), which is a useful alternative when transferrin glycosylation turned out to be normal. The patient cell homogenate showed a shift toward decreased branching of glycans compared to control, likely due to the lack of UDP-GlcNAc, which is a critical factor in the production of β1,6-branched (tetra-antennary) structures (Sasai et al 2002).
Both total plasma and intact transferrin glycoprofiling have their own advantages in the diagnosis of CDG. Recently, intact transferrin MS has successfully identified a series of novel CDG, such as PGM1-CDG (Tegtmeyer et al 2014) and MAN1B1-CDG (van Scherpenzeel et al 2014). A unique combination of CDG-I (lack of complete glycans) and CDG-II (truncated glycans especially lacking Gal residue) in PGM1-CDG is easily seen by intact transferrin glycoprofiling. Moreover, it is also used to follow-up the biochemical improvement of several patients that underwent an oral D-galactose supplementation (Voermans et al 2017, Wong et al 2017). It has become the primary diagnostic test for CDG, also including fast identification of B4GALT1-CDG, MGAT2-CDG, SLC35A1-CDG, and SLC35A2-CDG (van Scherpenzeel et al 2015).
However, there are several CDG types which cannot be discriminated by intact transferrin glycoprofiling which required total (released) plasma N-glycans for structural insights into the complete mixture of plasma glycoproteins. For example, for diagnosis of SLC35C1-CDG (GDP-fucose transporter defect), global N-glycan mapping is more suitable, because of its higher degree of fucosylation than intact transferrin (Guillard et al 2011). It is also able to detect the N-tetrasaccharide for ALG1-CDG diagnosis and highly abundant abnormal high mannose species of 3 Man- and 4 Man-glycans in PMM2-CDG and MPI-CDG (Zhang et al 2016), and to show abnormal profiles for tissue-specific glycosylation defects for VPS13B-CDG, also called Cohen syndrome (Duplomb et al 2014). In the case of MOGS-CDG which showed a normal transferrin profile, accumulation of several high mannose species from total IgG N-glycans were detected by plasma glycomics (Sadat et al 2014). In summary, Table 1 provides a detailed overview of the glycoprofiling data of total plasma N-glycans and intact transferrin for diagnosis of different CDG subtypes.
Table 1
Overview of plasma glycosylation features from total (released) N-glycans and intact transferrin MS profiling for CDG diagnosis
https://static-content.springer.com/image/art%3A10.1007%2Fs10545-018-0144-9/MediaObjects/10545_2018_144_Tab1a_HTML.gif https://static-content.springer.com/image/art%3A10.1007%2Fs10545-018-0144-9/MediaObjects/10545_2018_144_Tab1b_HTML.gif https://static-content.springer.com/image/art%3A10.1007%2Fs10545-018-0144-9/MediaObjects/10545_2018_144_Tab1c_HTML.gif https://static-content.springer.com/image/art%3A10.1007%2Fs10545-018-0144-9/MediaObjects/10545_2018_144_Tab1d_HTML.gif

Overview of the current CDG diagnostic workflow

As one of the most abundant glycoproteins in human plasma, transferrin has been used traditionally as a biomarker for N-glycosylation defects. Plasma transferrin isoelectric focusing (IEF) is recognized as the classical laboratory method (Jaeken et al 1984) and widely used as a routine screening test for CDG. Human transferrin has two glycosylation sites on Asn-432 and Asn-630, carrying both a bi-antennary glycan with two terminal, negatively charged sialic acid residues. In normal conditions, most transferrin molecules consist of a total of four sialic acid residues which are displayed as a single major band of tetrasialo-transferrin in the IEF pattern (Fig. 2a). The CDG-I IEF pattern shows an increased band of disialo-transferrin and asialo-transferrin, owing to the occurrence of transferrin isoforms with two sialic acids and no sialic acid, respectively (Fig. 2b). CDG-II IEF patterns show high variability as depicted for MGAT2-CDG (Fig. 2c), MAN1B1-CDG (Fig. 2d), and B4GALT1-CDG (Fig. 2e), with increased trisialo-transferrin or increased asialo- and monosialo-transferrin, respectively. Untreated hereditary fructosemia and galactosemia, alcohol abuse, hepatopathy, and bacterial sialidases are known as secondary causes of abnormal transferrin glycosylation.
The diagnostics follow-up for CDG-I profiles started with an enzyme assay in fibroblasts or leukocytes for diagnosis of PMM2-CDG and PMI-CDG. If negative, the next step was to perform lipid-linked oligosaccharides (LLO) in fibroblasts (Gao and Lehrman 2002), or recently this has been replaced by WES using a filter for CDG-I genes and targeted sequencing of a CDG-I panel (Timal et al 2012). The recent discovery of a novel sialylated N-tetrasaccharide for ALG1-CDG and abnormal small high mannose glycan structures in PMM2-CDG and MPI-CDG has proved plasma N-glycan analysis to also be relevant for CDG-I diagnosis. Hence, plasma glycomics is highly useful especially for ALG1-CDG, since it is challenging to identify the defect by genetics due to 14 pseudo genes that can complicate the analysis (Zhang et al 2016). Another upcoming diagnostic choice is applying WES directly and Sanger sequencing to confirm the gene defect, but up till now it takes more time to result and is more expensive than conventional CDG screening (Van Scherpenzeel et al 2016).
The diagnostic follow-up approach for CDG-II profiles is an IEF assay of Apo C-III to profile the mucin type O-glycans (Wopereis et al 2003), to distinguish between an exclusive N-glycosylation defect and a combined disorder of N- and O-glycosylation (Wopereis et al 2007). Apo C-III has only a single O-glycan on Thr-74 which is terminally modified by up to two sialic acids to generate three main IEF isoforms: Apo C-III0 (no sialic acid), Apo C-III1 (one sialic acid), and Apo C-III2 (two sialic acids). Decreased sialylation on Apo C-III profiles has been reported in conserved oligomeric Golgi (COG) defects (Spaapen et al 2005; Foulquier et al 2006; Foulquier et al 2007; Kranz et al 2007; Morava et al 2007; Wopereis et al 2007; Zeevaert et al 2008; Ng et al 2011; Palmigiano et al 2017) and autosomal recessive cutis laxa type-2 (ARCL2) due to ATP6V0A2 dysfunctions (Morava et al 2005; Kornak et al 2008). The limitation of Apo C-III IEF is that it is not able to differentiate between the three possible Apo C-III0 isoforms; the “real unglycosylated Apo C-III” and Apo C-III with two non-sialylated monosaccharides namely Gal and GalNAc. They can easily be separated by Apo C-III MALDI MS (Wada 2016).
So for more structural insight in glycans especially in CDG-II patients, MS is always the method of choice. This rapid profiling of abnormal glycans can be linked to potential gene defects based on the knowledge of glycosylation pathways. For example (Fig. 2): Detection of N-tetrasaccharide glycans in ALG1-CDG might be explained by the modification (galactosylation and sialylation) of the chitobiose glycan core, which accumulates due to cytosolic mannosyltransferase defects; Accumulation of truncated N-glycans lacking GlcNAc in MGAT2-CDG is likely due to the N-acetylglucosaminyltransferase defects in GA; Accumulation of truncated N-glycans lacking Gal could be due to B4GALT1 gene defect, encoding for galactosyltransferase in GA. Accumulation of hybrid N-glycans in MAN1B1-CDG is due to the ER mannosyltransferase defects. The structural information is important to narrow down the number of candidate genes, thereby greatly minimizing the time to diagnosis. The complete diagnostic workflow as described above is depicted in Fig. 3.

Outlook

Based on the identification of several new genetic defects over the past four years by a combination of high-resolution intact transferrin analysis with WES, it might be concluded that clinical glycomics and NGS technology work in synergy by reducing the number of candidate genes and turnaround time to identify a CDG subtype. This approach creates opportunities to identify new genetic defects, but beyond this observation, glycomics has the benefit that the data do not only reflect the genes, but also environmental influences, which create possibilities for therapy monitoring and intervention. The first example was PGM1-CDG (Voermans et al 2017), but nowadays, several new genetic defects appear to be involved in sugar metabolism. Surprisingly, not all these genetic defects show abnormal transferrin glycosylation. Examples are NANS, GNE, PGM3, and Cohen syndrome. NANS and GNE are genetic defects in the sialic acid biosynthesis pathway, and Cohen syndrome patients have a mutation in VPS13B, a protein important for proper GA function. It is known that there are tissue-specific glycosylation processes, for example gamma-glutamyl transpeptidase synthetized by the liver or the kidney (West et al 2010), and that there will be other proteins than transferrin which do show abnormal glycosylation, e.g., muscle proteins for GNE patients.
Native glycopeptide profiling would be an attractive method to obtain insight into protein-specific glycosylation. For some isolated proteins like human transferrin, human alpha-1-acid glycoprotein, influenza A virus hemagglutinin, and human IgG, this has been performed (Khatri et al 2014). If this could work for highly complex protein mixtures like serum or plasma (Zielinska et al 2010), an enormous amount of data would show up and provide us with new mechanistic insights into tissue-specific glycosylation and likely yield several glycoprotein biomarkers for CDG and also common disorders (Yang et al 2017). Knowledge about the human glycoproteome is still very limited, but this new system-wide mapping technology will allow the study of fundamental questions in glycobiology, such as dynamics, macro- and microheterogeneity, tissue-specific glycosylation, and the function of certain glycans in specific biological contexts (Thaysen-Andersen and Packer 2014). In this context, genetic deficiencies beyond the classical CDG, which are in O-glycosylation, e.g., in O-GalNAc, O-mannose, and O-fucose glycosylation or in glycolipid or glycosaminoglycan biosynthesis (comprehensive review in Hennet 2012), have to be mentioned. There is no general diagnostic test available for these rare genetic disorders because of the structural heterogeneity of O-glycans and their tissue-specific expression. Because of the advancement in genetics and bioinformatics, we foresee glycomics becoming interlinked with metabolomics and proteomics, thereby opening research avenues to unravel protein glycosylation in a tissue- or cell-specific manner, to understand the biochemical mechanisms of glycosylation, and to ultimately develop or improve new (sugar based) therapies for this group of so far mainly untreatable disorders.
Another advance in technology is intact protein profiling. Both in the chromatographic part as well as the data analysis there are recent developments which enable the analysis of protein mixtures to have multiple biomarkers in one assay. A large number of biomarkers used in clinical laboratories are glycopeptides of which variations in glycosylation are not taken into account, and only protein expression levels are measured. Examples are alpha-1-antitrypsin for chronic obstructive pulmonary disease (COPD), haptoglobin for gastric cancer, and human chorionic gonadotrophin for ovarian and testicular tumors (Van Scherpenzeel et al 2016). For two intact plasma glycoproteins, namely alpha-fetoprotein and transferrin, it is already known that it is essential to include glycosylation analysis for specificity and sensitivity of the marker. There is an enormous potential to improve first line diagnosis when the level of glycosylation is taken into account on top of the current protein expression levels. With the upcoming use of targeted mass spectrometry in clinical laboratories, the glycan part of the biomarker might easily be included to improve sensitivity and specificity of the marker without additional time-to-result.
With the example of effective D-galactose supplementation on the improvement of glycosylation in PGM1-CDG, which was derived from the intact transferrin glycosylation profile (Tegtmeyer et al 2014), there is emerging interest in the application of sugars as supplemental therapies for metabolic disorders or as supportive therapy to improve the mechanism of action of known therapies, such as chemotherapy, for which 2-deoxyhexose was used to enhance the therapeutic effect by inhibiting glycolysis and even induce an effective antitumor immune response (Beneteau et al 2012). The way to obtain insight into these mechanisms will be one new glycomics area, in which the building blocks of glycosylation, comprising sugar-phosphates and nucleotide sugars, are analyzed. By studying the flux through sugar metabolism, dynamic insights will be obtained, which could lead to the development of new therapies and improve current ones. Bioinformatics will become increasingly important to extract the relevant information out of these big data sets, to visualize, and potentially integrate with other omics layers of information, for better understanding of the complex field of glycobiology.

Acknowledgements

We acknowledge support of the Dutch Organization for Scientific Research (ZONMW Medium Investment Grant 40-00506-98-9001 and VIDI Grant 91713359 to DJL; VENI grant 722015012 to MvS) and the Malaysian government grant (R02087 to NAB).

Compliance with ethical standards

Conflict of interest

None.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
Zurück zum Zitat Ciucanu I, Kerek F (1984) A simple and rapid method for the permethylation of carbohydrate. Carbohydr Res 131:209–217CrossRef Ciucanu I, Kerek F (1984) A simple and rapid method for the permethylation of carbohydrate. Carbohydr Res 131:209–217CrossRef
Zurück zum Zitat Cummings RD (2009) The repertoire of glycan determinants in the human glycome. Mol BioSyst 5:1087–1104CrossRefPubMed Cummings RD (2009) The repertoire of glycan determinants in the human glycome. Mol BioSyst 5:1087–1104CrossRefPubMed
Zurück zum Zitat Defaus S, Gupta P, Andreu D, Gutierrez-Gallego R (2014) Mammalian protein glycosylation-structure versus function. Analyst 139:2944–2967CrossRefPubMed Defaus S, Gupta P, Andreu D, Gutierrez-Gallego R (2014) Mammalian protein glycosylation-structure versus function. Analyst 139:2944–2967CrossRefPubMed
Zurück zum Zitat Dorre K, Olczak M, Wada Y et al (2015) A new case of UDP-galactose transporter deficiency (SLC35A2-CDG): molecular basis, clinical phenotype, and therapeutic approach. J Inherit Metab Dis 38:931–940CrossRefPubMed Dorre K, Olczak M, Wada Y et al (2015) A new case of UDP-galactose transporter deficiency (SLC35A2-CDG): molecular basis, clinical phenotype, and therapeutic approach. J Inherit Metab Dis 38:931–940CrossRefPubMed
Zurück zum Zitat Duplomb L, Duvet S, Picot D et al (2014) Cohen syndrome is associated with major glycosylation defects. Hum Mol Genet 23:2391–2399CrossRefPubMed Duplomb L, Duvet S, Picot D et al (2014) Cohen syndrome is associated with major glycosylation defects. Hum Mol Genet 23:2391–2399CrossRefPubMed
Zurück zum Zitat Edvardson S, Ashikov A, Jalas C et al (2013) Mutations in SLC35A3 cause autism spectrum disorder, epilepsy and arthrogryposis. J Med Genet 50:733–739CrossRefPubMed Edvardson S, Ashikov A, Jalas C et al (2013) Mutations in SLC35A3 cause autism spectrum disorder, epilepsy and arthrogryposis. J Med Genet 50:733–739CrossRefPubMed
Zurück zum Zitat El-Aneed A, Cohen A, Banoub J (2009) Mass spectrometry, review of the basics: electrospray, MALDI, and commonly used mass analyzers. Appl Spectrosc Rev 44:210–230CrossRef El-Aneed A, Cohen A, Banoub J (2009) Mass spectrometry, review of the basics: electrospray, MALDI, and commonly used mass analyzers. Appl Spectrosc Rev 44:210–230CrossRef
Zurück zum Zitat Faid V, Chirat F, Seta N, Foulquier F, Morelle W (2007) A rapid mass spectrometric strategy for the characterization of N- and O-glycan chains in the diagnosis of defects in glycan biosynthesis. Proteomics 7:1800–1813CrossRefPubMed Faid V, Chirat F, Seta N, Foulquier F, Morelle W (2007) A rapid mass spectrometric strategy for the characterization of N- and O-glycan chains in the diagnosis of defects in glycan biosynthesis. Proteomics 7:1800–1813CrossRefPubMed
Zurück zum Zitat Foulquier F, Ungar D, Reynders E et al (2007) A new inborn error of glycosylation due to a Cog8 deficiency reveals a critical role for the Cog1-Cog8 interaction in COG complex formation. Hum Mol Genet 16:717–730CrossRefPubMed Foulquier F, Ungar D, Reynders E et al (2007) A new inborn error of glycosylation due to a Cog8 deficiency reveals a critical role for the Cog1-Cog8 interaction in COG complex formation. Hum Mol Genet 16:717–730CrossRefPubMed
Zurück zum Zitat Foulquier F, Vasile E, Schollen E et al (2006) Conserved oligomeric Golgi complex subunit 1 deficiency reveals a previously uncharacterized congenital disorder of glycosylation type II. Proc Natl Acad Sci U S A 103:3764–3769CrossRefPubMedPubMedCentral Foulquier F, Vasile E, Schollen E et al (2006) Conserved oligomeric Golgi complex subunit 1 deficiency reveals a previously uncharacterized congenital disorder of glycosylation type II. Proc Natl Acad Sci U S A 103:3764–3769CrossRefPubMedPubMedCentral
Zurück zum Zitat Freeze HH, Chong JX, Bamshad MJ, Ng BG (2014) Solving glycosylation disorders: fundamental approaches reveal complicated pathways. Am J Hum Genet 94:161–175CrossRefPubMedPubMedCentral Freeze HH, Chong JX, Bamshad MJ, Ng BG (2014) Solving glycosylation disorders: fundamental approaches reveal complicated pathways. Am J Hum Genet 94:161–175CrossRefPubMedPubMedCentral
Zurück zum Zitat Fuzery AK, Levin J, Chan MM, Chan DW (2013) Translation of proteomic biomarkers into FDA approved cancer diagnostics: issues and challenges. Clin Proteomics 10:13CrossRefPubMedPubMedCentral Fuzery AK, Levin J, Chan MM, Chan DW (2013) Translation of proteomic biomarkers into FDA approved cancer diagnostics: issues and challenges. Clin Proteomics 10:13CrossRefPubMedPubMedCentral
Zurück zum Zitat Gao N, Lehrman MA (2002) Analyses of dolichol pyrophosphate–linked oligosaccharides in cell cultures and tissues by fluorophore-assisted carbohydrate electrophoresis. Glycobiology 12:353–360CrossRefPubMed Gao N, Lehrman MA (2002) Analyses of dolichol pyrophosphate–linked oligosaccharides in cell cultures and tissues by fluorophore-assisted carbohydrate electrophoresis. Glycobiology 12:353–360CrossRefPubMed
Zurück zum Zitat Guillard M, Gloerich J, Wessels HJ, Morava E, Wevers RA, Lefeber DJ (2009) Automated measurement of permethylated serum N-glycans by MALDI-linear ion trap mass spectrometry. Carbohydr Res 344:1550–1557CrossRefPubMed Guillard M, Gloerich J, Wessels HJ, Morava E, Wevers RA, Lefeber DJ (2009) Automated measurement of permethylated serum N-glycans by MALDI-linear ion trap mass spectrometry. Carbohydr Res 344:1550–1557CrossRefPubMed
Zurück zum Zitat Guillard M, Morava E, van Delft FL et al (2011) Plasma N-glycan profiling by mass spectrometry for congenital disorders of glycosylation type II. Clin Chem 57:593–602CrossRefPubMed Guillard M, Morava E, van Delft FL et al (2011) Plasma N-glycan profiling by mass spectrometry for congenital disorders of glycosylation type II. Clin Chem 57:593–602CrossRefPubMed
Zurück zum Zitat Hennet T (2012) Diseases of glycosylation beyond classical congenital disorders of glycosylation. Biochim Biophys Acta 1820:1306–1317CrossRefPubMed Hennet T (2012) Diseases of glycosylation beyond classical congenital disorders of glycosylation. Biochim Biophys Acta 1820:1306–1317CrossRefPubMed
Zurück zum Zitat Hoffmann M, Marx K, Reichl U, Wuhrer M, Rapp E (2016) Site specific O-glycosylation analysis of human blood plasma proteins. Mol Cell Proteomics 15:624–641CrossRefPubMed Hoffmann M, Marx K, Reichl U, Wuhrer M, Rapp E (2016) Site specific O-glycosylation analysis of human blood plasma proteins. Mol Cell Proteomics 15:624–641CrossRefPubMed
Zurück zum Zitat Hua S, Williams CC, Dimapasoc LM et al (2013) Isomer-specific chromatographic profiling yields highly sensitive and specific potential N-glycan biomarkers for epithelial ovarian cancer. J Chromatogr A 1279:58–67CrossRefPubMedPubMedCentral Hua S, Williams CC, Dimapasoc LM et al (2013) Isomer-specific chromatographic profiling yields highly sensitive and specific potential N-glycan biomarkers for epithelial ovarian cancer. J Chromatogr A 1279:58–67CrossRefPubMedPubMedCentral
Zurück zum Zitat Huybrechts S, De Laet C, Bontems P et al (2012) Deficiency of subunit 6 of the conserved oligomeric Golgi complex (COG6-CDG): second patient, different phenotype. JIMD Rep 4:103–108CrossRefPubMed Huybrechts S, De Laet C, Bontems P et al (2012) Deficiency of subunit 6 of the conserved oligomeric Golgi complex (COG6-CDG): second patient, different phenotype. JIMD Rep 4:103–108CrossRefPubMed
Zurück zum Zitat Iqbal Z, Shahzad M, Vissers LE et al (2013) A compound heterozygous mutation in DPAGT1 results in a congenital disorder of glycosylation with a relatively mild phenotype. Eur J Hum Genet 21:844–849CrossRefPubMed Iqbal Z, Shahzad M, Vissers LE et al (2013) A compound heterozygous mutation in DPAGT1 results in a congenital disorder of glycosylation with a relatively mild phenotype. Eur J Hum Genet 21:844–849CrossRefPubMed
Zurück zum Zitat Jaeken J (2011) Congenital disorders of glycosylation (CDG): it's (nearly) all in it! J Inherit Metab Dis 34:853–858CrossRefPubMed Jaeken J (2011) Congenital disorders of glycosylation (CDG): it's (nearly) all in it! J Inherit Metab Dis 34:853–858CrossRefPubMed
Zurück zum Zitat Jaeken J, Van Eijk HG, van der Heul C, Corbeel L, Eeckels R, Eggermont E (1984) Sialic acid-deficient serum and cerebrospinal fluid transferrin in a newly recognized genetic syndrome. Clin Chim Acta 144:245–247CrossRefPubMed Jaeken J, Van Eijk HG, van der Heul C, Corbeel L, Eeckels R, Eggermont E (1984) Sialic acid-deficient serum and cerebrospinal fluid transferrin in a newly recognized genetic syndrome. Clin Chim Acta 144:245–247CrossRefPubMed
Zurück zum Zitat Jansen EJ, Timal S, Ryan M et al (2016a) ATP6AP1 deficiency causes an immunodeficiency with hepatopathy, cognitive impairment and abnormal protein glycosylation. Nat Commun 7:11600CrossRefPubMedPubMedCentral Jansen EJ, Timal S, Ryan M et al (2016a) ATP6AP1 deficiency causes an immunodeficiency with hepatopathy, cognitive impairment and abnormal protein glycosylation. Nat Commun 7:11600CrossRefPubMedPubMedCentral
Zurück zum Zitat Jansen JC, Cirak S, van Scherpenzeel M et al (2016c) CCDC115 deficiency causes a disorder of Golgi homeostasis with abnormal protein glycosylation. Am J Hum Genet 98:310–321CrossRefPubMedPubMedCentral Jansen JC, Cirak S, van Scherpenzeel M et al (2016c) CCDC115 deficiency causes a disorder of Golgi homeostasis with abnormal protein glycosylation. Am J Hum Genet 98:310–321CrossRefPubMedPubMedCentral
Zurück zum Zitat Jansen JC, Timal S, van Scherpenzeel M et al (2016b) TMEM199 deficiency is a disorder of Golgi homeostasis characterized by elevated aminotransferases, alkaline phosphatase, and cholesterol and abnormal glycosylation. Am J Hum Genet 98:322–330CrossRefPubMedPubMedCentral Jansen JC, Timal S, van Scherpenzeel M et al (2016b) TMEM199 deficiency is a disorder of Golgi homeostasis characterized by elevated aminotransferases, alkaline phosphatase, and cholesterol and abnormal glycosylation. Am J Hum Genet 98:322–330CrossRefPubMedPubMedCentral
Zurück zum Zitat Kang P, Mechref Y, Novotny MV (2008) High-throughput solid-phase permethylation of glycans prior to mass spectrometry. Rapid Commun Mass Spectrom 22:721–734CrossRefPubMed Kang P, Mechref Y, Novotny MV (2008) High-throughput solid-phase permethylation of glycans prior to mass spectrometry. Rapid Commun Mass Spectrom 22:721–734CrossRefPubMed
Zurück zum Zitat Khatri K, Staples GO, Leymarie N et al (2014) Confident assignment of site-specific glycosylation in complex glycoproteins in a single step. J Proteome Res 13:4347–4355CrossRefPubMedPubMedCentral Khatri K, Staples GO, Leymarie N et al (2014) Confident assignment of site-specific glycosylation in complex glycoproteins in a single step. J Proteome Res 13:4347–4355CrossRefPubMedPubMedCentral
Zurück zum Zitat Kornak U, Reynders E, Dimopoulou A et al (2008) Impaired glycosylation and cutis laxa caused by mutations in the vesicular H+-ATPase subunit ATP6V0A2. Nat Genet 40:32–34CrossRefPubMed Kornak U, Reynders E, Dimopoulou A et al (2008) Impaired glycosylation and cutis laxa caused by mutations in the vesicular H+-ATPase subunit ATP6V0A2. Nat Genet 40:32–34CrossRefPubMed
Zurück zum Zitat Kranz C, Ng BG, Sun L et al (2007) COG8 deficiency causes new congenital disorder of glycosylation type IIh. Hum Mol Genet 16:731–741CrossRefPubMed Kranz C, Ng BG, Sun L et al (2007) COG8 deficiency causes new congenital disorder of glycosylation type IIh. Hum Mol Genet 16:731–741CrossRefPubMed
Zurück zum Zitat Lacey JM, Bergen HR, Magera MJ, Naylor S, O’Brien JF (2001) Rapid determination of transferrin isoforms by immunoaffinity liquid chromatography and electrospray mass spectrometry. Clin Chem 47:513–518PubMed Lacey JM, Bergen HR, Magera MJ, Naylor S, O’Brien JF (2001) Rapid determination of transferrin isoforms by immunoaffinity liquid chromatography and electrospray mass spectrometry. Clin Chem 47:513–518PubMed
Zurück zum Zitat Lee LY, Thaysen-Andersen M, Baker MS, Packer NH, Hancock WS, Fanayan S (2014) Comprehensive N-glycome profiling of cultured human epithelial breast cells identifies unique secretome N-glycosylation signatures enabling tumorigenic subtype classification. J Proteome Res 13:4783–4795CrossRefPubMed Lee LY, Thaysen-Andersen M, Baker MS, Packer NH, Hancock WS, Fanayan S (2014) Comprehensive N-glycome profiling of cultured human epithelial breast cells identifies unique secretome N-glycosylation signatures enabling tumorigenic subtype classification. J Proteome Res 13:4783–4795CrossRefPubMed
Zurück zum Zitat Leerapun A, Suravarapu SV, Bida JP et al (2007) The utility of Lens Culinaris agglutinin-reactive alpha-fetoprotein in the diagnosis of hepatocellular carcinoma: evaluation in a United States referral population. Clin Gastroenterol Hepatol 5:394–402CrossRefPubMedPubMedCentral Leerapun A, Suravarapu SV, Bida JP et al (2007) The utility of Lens Culinaris agglutinin-reactive alpha-fetoprotein in the diagnosis of hepatocellular carcinoma: evaluation in a United States referral population. Clin Gastroenterol Hepatol 5:394–402CrossRefPubMedPubMedCentral
Zurück zum Zitat Lefeber DJ (2016) Protein-specific glycoprofiling for patient diagnostics. Clin Chem 62:9–11CrossRefPubMed Lefeber DJ (2016) Protein-specific glycoprofiling for patient diagnostics. Clin Chem 62:9–11CrossRefPubMed
Zurück zum Zitat Leiserowitz GS, Lebrilla C, Miyamoto S et al (2007) Glycomics analysis of serum: a potential new biomarker for ovarian cancer? Int J Gynecol Cancer 18:470–475CrossRefPubMedPubMedCentral Leiserowitz GS, Lebrilla C, Miyamoto S et al (2007) Glycomics analysis of serum: a potential new biomarker for ovarian cancer? Int J Gynecol Cancer 18:470–475CrossRefPubMedPubMedCentral
Zurück zum Zitat Li D, Mallory T, Satomura S (2001) AFP-L3: a new generation of tumor marker for hepatocellular carcinoma. Clin Chim Acta 313:15–19CrossRefPubMed Li D, Mallory T, Satomura S (2001) AFP-L3: a new generation of tumor marker for hepatocellular carcinoma. Clin Chim Acta 313:15–19CrossRefPubMed
Zurück zum Zitat Melmer M, Stangler T, Premstaller A, Lindner W (2011) Comparison of hydrophilic-interaction, reversed-phase and porous graphitic carbon chromatography for glycan analysis. J Chromatogr A 1218:118–123CrossRefPubMed Melmer M, Stangler T, Premstaller A, Lindner W (2011) Comparison of hydrophilic-interaction, reversed-phase and porous graphitic carbon chromatography for glycan analysis. J Chromatogr A 1218:118–123CrossRefPubMed
Zurück zum Zitat Mohamed M, Ashikov A, Guillard M et al (2013) Intellectual disability and bleeding diathesis due to deficient CMP–sialic acid transport. Neurology 81:681–687CrossRefPubMed Mohamed M, Ashikov A, Guillard M et al (2013) Intellectual disability and bleeding diathesis due to deficient CMP–sialic acid transport. Neurology 81:681–687CrossRefPubMed
Zurück zum Zitat Morava E, Wopereis S, Coucke P et al (2005) Defective protein glycosylation in patients with cutis laxa syndrome. Eur J Hum Genet 13:414–421CrossRefPubMed Morava E, Wopereis S, Coucke P et al (2005) Defective protein glycosylation in patients with cutis laxa syndrome. Eur J Hum Genet 13:414–421CrossRefPubMed
Zurück zum Zitat Morava E, Zeevaert R, Korsch E et al (2007) A common mutation in the COG7 gene with a consistent phenotype including microcephaly, adducted thumbs, growth retardation, VSD and episodes of hyperthermia. Eur J Hum Genet 15:638–645CrossRefPubMed Morava E, Zeevaert R, Korsch E et al (2007) A common mutation in the COG7 gene with a consistent phenotype including microcephaly, adducted thumbs, growth retardation, VSD and episodes of hyperthermia. Eur J Hum Genet 15:638–645CrossRefPubMed
Zurück zum Zitat Ng BG, Sharma V, Sun L et al (2011) Identification of the first COG-CDG patient of Indian origin. Mol Genet Metab 102:364–367CrossRefPubMed Ng BG, Sharma V, Sun L et al (2011) Identification of the first COG-CDG patient of Indian origin. Mol Genet Metab 102:364–367CrossRefPubMed
Zurück zum Zitat Pabst M, Altmann F (2008) Influence of electrosorption, solvent, temperature, and ion polarity on the performance of LC-ESI-MS using graphitic carbon for acidic oligosaccharides. Anal Chem 80:7534–7542CrossRefPubMed Pabst M, Altmann F (2008) Influence of electrosorption, solvent, temperature, and ion polarity on the performance of LC-ESI-MS using graphitic carbon for acidic oligosaccharides. Anal Chem 80:7534–7542CrossRefPubMed
Zurück zum Zitat Pabst M, Altmann F (2011) Glycan analysis by modern instrumental methods. Proteomics 11:631–643CrossRefPubMed Pabst M, Altmann F (2011) Glycan analysis by modern instrumental methods. Proteomics 11:631–643CrossRefPubMed
Zurück zum Zitat Pabst M, Kolarich D, Poltl G et al (2009) Comparison of fluorescent labels for oligosaccharides and introduction of a new postlabeling purification method. Anal Biochem 384:263–273CrossRefPubMed Pabst M, Kolarich D, Poltl G et al (2009) Comparison of fluorescent labels for oligosaccharides and introduction of a new postlabeling purification method. Anal Biochem 384:263–273CrossRefPubMed
Zurück zum Zitat Paesold-Burda P, Maag C, Troxler H et al (2009) Deficiency in COG5 causes a moderate form of congenital disorders of glycosylation. Hum Mol Genet 18:4350–4356CrossRefPubMed Paesold-Burda P, Maag C, Troxler H et al (2009) Deficiency in COG5 causes a moderate form of congenital disorders of glycosylation. Hum Mol Genet 18:4350–4356CrossRefPubMed
Zurück zum Zitat Palmigiano A, Barone R, Sturiale L et al (2016) CSF N-glycoproteomics for early diagnosis in Alzheimer’s disease. J Proteome 131:29–37CrossRef Palmigiano A, Barone R, Sturiale L et al (2016) CSF N-glycoproteomics for early diagnosis in Alzheimer’s disease. J Proteome 131:29–37CrossRef
Zurück zum Zitat Palmigiano A, Bua RO, Barone R et al (2017) MALDI-MS profiling of serum O- and N-glycosylation in COG5-CDG. J Mass Spectrom 52:372–377CrossRefPubMed Palmigiano A, Bua RO, Barone R et al (2017) MALDI-MS profiling of serum O- and N-glycosylation in COG5-CDG. J Mass Spectrom 52:372–377CrossRefPubMed
Zurück zum Zitat Park JH, Hogrebe M, Gruneberg M et al (2015) SLC39A8 deficiency: a disorder of manganese transport and glycosylation. Am J Hum Genet 97:894–903CrossRefPubMedPubMedCentral Park JH, Hogrebe M, Gruneberg M et al (2015) SLC39A8 deficiency: a disorder of manganese transport and glycosylation. Am J Hum Genet 97:894–903CrossRefPubMedPubMedCentral
Zurück zum Zitat Reiding KR, Blank D, Kuijper DM, Deelder AM, Wuhrer M (2014) High-throughput profiling of protein N-glycosylation by MALDI-TOF-MS employing linkage-specific sialic acid esterification. Anal Chem 86:5784–5793CrossRefPubMed Reiding KR, Blank D, Kuijper DM, Deelder AM, Wuhrer M (2014) High-throughput profiling of protein N-glycosylation by MALDI-TOF-MS employing linkage-specific sialic acid esterification. Anal Chem 86:5784–5793CrossRefPubMed
Zurück zum Zitat Reynders E, Foulquier F, Leao Teles E et al (2009) Golgi function and dysfunction in the first COG4-deficient CDG type II patient. Hum Mol Genet 18:3244–3256CrossRefPubMedPubMedCentral Reynders E, Foulquier F, Leao Teles E et al (2009) Golgi function and dysfunction in the first COG4-deficient CDG type II patient. Hum Mol Genet 18:3244–3256CrossRefPubMedPubMedCentral
Zurück zum Zitat Ruhaak LR, Stroble C, Dai J et al (2016) Serum glycans as risk markers for non-small cell lung cancer. Cancer Prev Res (Phila) 9:317–323CrossRef Ruhaak LR, Stroble C, Dai J et al (2016) Serum glycans as risk markers for non-small cell lung cancer. Cancer Prev Res (Phila) 9:317–323CrossRef
Zurück zum Zitat Ruhaak LR, Zauner G, Huhn C, Bruggink C, Deelder AM, Wuhrer M (2010) Glycan labeling strategies and their use in identification and quantification. Anal Bioanal Chem 397:3457–3481CrossRefPubMedPubMedCentral Ruhaak LR, Zauner G, Huhn C, Bruggink C, Deelder AM, Wuhrer M (2010) Glycan labeling strategies and their use in identification and quantification. Anal Bioanal Chem 397:3457–3481CrossRefPubMedPubMedCentral
Zurück zum Zitat Russell AC, Simurina M, Garcia MT et al (2017) The N-glycosylation of immunoglobulin G as a novel biomarker of Parkinson’s disease. Glycobiology 27:501–510CrossRefPubMed Russell AC, Simurina M, Garcia MT et al (2017) The N-glycosylation of immunoglobulin G as a novel biomarker of Parkinson’s disease. Glycobiology 27:501–510CrossRefPubMed
Zurück zum Zitat Sasai K, Ikeda Y, Fujii T, Tsuda T, Taniguchi N (2002) UDP-GlcNAc concentration is an important factor in the biosynthesis of beta1,6-branched oligosaccharides: regulation based on the kinetic properties of N-acetylglucosaminyltransferase V. Glycobiology 12:119–127CrossRefPubMed Sasai K, Ikeda Y, Fujii T, Tsuda T, Taniguchi N (2002) UDP-GlcNAc concentration is an important factor in the biosynthesis of beta1,6-branched oligosaccharides: regulation based on the kinetic properties of N-acetylglucosaminyltransferase V. Glycobiology 12:119–127CrossRefPubMed
Zurück zum Zitat Sethi MK, Kim H, Park CK et al (2015) In-depth N-glycome profiling of paired colorectal cancer and non-tumorigenic tissues reveals cancer-, stage- and EGFR-specific protein N-glycosylation. Glycobiology 25:1064–1078CrossRefPubMed Sethi MK, Kim H, Park CK et al (2015) In-depth N-glycome profiling of paired colorectal cancer and non-tumorigenic tissues reveals cancer-, stage- and EGFR-specific protein N-glycosylation. Glycobiology 25:1064–1078CrossRefPubMed
Zurück zum Zitat Sethi MK, Thaysen-Andersen M, Smith JT et al (2014) Comparative N-glycan profiling of colorectal cancer cell lines reveals unique bisecting GlcNAc and alpha-2,3-linked sialic acid determinants are associated with membrane proteins of the more metastatic/aggressive cell lines. J Proteome Res 13:277–288CrossRefPubMed Sethi MK, Thaysen-Andersen M, Smith JT et al (2014) Comparative N-glycan profiling of colorectal cancer cell lines reveals unique bisecting GlcNAc and alpha-2,3-linked sialic acid determinants are associated with membrane proteins of the more metastatic/aggressive cell lines. J Proteome Res 13:277–288CrossRefPubMed
Zurück zum Zitat Spaapen LJ, Bakker JA, van der Meer SB et al (2005) Clinical and biochemical presentation of siblings with COG-7 deficiency, a lethal multiple O- and N-glycosylation disorder. J Inherit Metab Dis 28:707–714CrossRefPubMed Spaapen LJ, Bakker JA, van der Meer SB et al (2005) Clinical and biochemical presentation of siblings with COG-7 deficiency, a lethal multiple O- and N-glycosylation disorder. J Inherit Metab Dis 28:707–714CrossRefPubMed
Zurück zum Zitat Stray-Pedersen A, Backe PH, Sorte HS et al (2014) PGM3 mutations cause a congenital disorder of glycosylation with severe immunodeficiency and skeletal dysplasia. Am J Hum Genet 95:96–107CrossRefPubMedPubMedCentral Stray-Pedersen A, Backe PH, Sorte HS et al (2014) PGM3 mutations cause a congenital disorder of glycosylation with severe immunodeficiency and skeletal dysplasia. Am J Hum Genet 95:96–107CrossRefPubMedPubMedCentral
Zurück zum Zitat Sturiale L, Barone R, Garozzo D (2011) The impact of mass spectrometry in the diagnosis of congenital disorders of glycosylation. J Inherit Metab Dis 34:891–899CrossRefPubMed Sturiale L, Barone R, Garozzo D (2011) The impact of mass spectrometry in the diagnosis of congenital disorders of glycosylation. J Inherit Metab Dis 34:891–899CrossRefPubMed
Zurück zum Zitat Thaysen-Andersen M, Packer NH (2014) Advances in LC-MS/MS-based glycoproteomics: getting closer to system-wide site-specific mapping of the N- and O-glycoproteome. Biochim Biophys Acta 1844:1437–1452CrossRefPubMed Thaysen-Andersen M, Packer NH (2014) Advances in LC-MS/MS-based glycoproteomics: getting closer to system-wide site-specific mapping of the N- and O-glycoproteome. Biochim Biophys Acta 1844:1437–1452CrossRefPubMed
Zurück zum Zitat Timal S, Hoischen A, Lehle L et al (2012) Gene identification in the congenital disorders of glycosylation type I by whole exome sequencing. Hum Mol Genet 21:4151–4161CrossRefPubMed Timal S, Hoischen A, Lehle L et al (2012) Gene identification in the congenital disorders of glycosylation type I by whole exome sequencing. Hum Mol Genet 21:4151–4161CrossRefPubMed
Zurück zum Zitat Van Damme T, Gardeitchik T, Mohamed M et al (2016) Mutations in ATP6V1E1 or ATP6V1A cause autosomal-recessive cutis laxa. Am J Hum Genet 100:216–227CrossRef Van Damme T, Gardeitchik T, Mohamed M et al (2016) Mutations in ATP6V1E1 or ATP6V1A cause autosomal-recessive cutis laxa. Am J Hum Genet 100:216–227CrossRef
Zurück zum Zitat Van Karnebeek CD, Bonafe L, Wen XY et al (2016) NANS-mediated synthesis of sialic acid is required for brain and skeletal development. Nat Genet 48:777–784CrossRefPubMed Van Karnebeek CD, Bonafe L, Wen XY et al (2016) NANS-mediated synthesis of sialic acid is required for brain and skeletal development. Nat Genet 48:777–784CrossRefPubMed
Zurück zum Zitat Van Scherpenzeel M, Steenbergen G, Morava E, Wevers RA, Lefeber DJ (2015) High-resolution mass spectrometry glycoprofiling of intact transferrin for diagnosis and subtype identification in the congenital disorders of glycosylation. Transl Res 166:639–649CrossRefPubMed Van Scherpenzeel M, Steenbergen G, Morava E, Wevers RA, Lefeber DJ (2015) High-resolution mass spectrometry glycoprofiling of intact transferrin for diagnosis and subtype identification in the congenital disorders of glycosylation. Transl Res 166:639–649CrossRefPubMed
Zurück zum Zitat Van Scherpenzeel M, Timal S, Rymen D et al (2014) Diagnostic serum glycosylation profile in patients with intellectual disability as a result of MAN1B1 deficiency. Brain 137:1030–1038CrossRefPubMed Van Scherpenzeel M, Timal S, Rymen D et al (2014) Diagnostic serum glycosylation profile in patients with intellectual disability as a result of MAN1B1 deficiency. Brain 137:1030–1038CrossRefPubMed
Zurück zum Zitat Van Scherpenzeel M, Willems E, Lefeber DJ (2016) Clinical diagnostics and therapy monitoring in the congenital disorders of glycosylation. Glyconj J 33:345–358CrossRef Van Scherpenzeel M, Willems E, Lefeber DJ (2016) Clinical diagnostics and therapy monitoring in the congenital disorders of glycosylation. Glyconj J 33:345–358CrossRef
Zurück zum Zitat Vissers L, van Nimwegen KJM, Schieving JH (2017) A clinical utility study of exome sequencing versus conventional genetic testing in pediatric neurology. Genet Med 19:1055–1063CrossRefPubMedPubMedCentral Vissers L, van Nimwegen KJM, Schieving JH (2017) A clinical utility study of exome sequencing versus conventional genetic testing in pediatric neurology. Genet Med 19:1055–1063CrossRefPubMedPubMedCentral
Zurück zum Zitat Voermans NC, Guillard M, Doedee R et al (2010) Clinical features, lectin staining, and a novel GNE frameshift mutation in hereditary inclusion body myopathy. Clin Neuropathol 29:71–77PubMedPubMedCentral Voermans NC, Guillard M, Doedee R et al (2010) Clinical features, lectin staining, and a novel GNE frameshift mutation in hereditary inclusion body myopathy. Clin Neuropathol 29:71–77PubMedPubMedCentral
Zurück zum Zitat Voermans NC, Preisler N, Madsen KL et al (2017) PGM1 deficiency: substrate use during exercise and effect of treatment with galactose. Neuromuscul Disord 27:370–376CrossRefPubMed Voermans NC, Preisler N, Madsen KL et al (2017) PGM1 deficiency: substrate use during exercise and effect of treatment with galactose. Neuromuscul Disord 27:370–376CrossRefPubMed
Zurück zum Zitat Wada Y (2016) Mass spectrometry of transferrin and apolipoprotein C-III for diagnosis and screening of congenital disorder of glycosylation. Glycoconj J 33:297–307CrossRefPubMed Wada Y (2016) Mass spectrometry of transferrin and apolipoprotein C-III for diagnosis and screening of congenital disorder of glycosylation. Glycoconj J 33:297–307CrossRefPubMed
Zurück zum Zitat Wada Y, Kadoya M, Okamoto N (2012) Mass spectrometry of apolipoprotein C-III, a simple analytical method for mucin-type O-glycosylation and its application to an autosomal recessive cutis laxa type-2 (ARCL2) patient. Glycobiology 22:1140–1144CrossRefPubMed Wada Y, Kadoya M, Okamoto N (2012) Mass spectrometry of apolipoprotein C-III, a simple analytical method for mucin-type O-glycosylation and its application to an autosomal recessive cutis laxa type-2 (ARCL2) patient. Glycobiology 22:1140–1144CrossRefPubMed
Zurück zum Zitat Wada Y, Nishikawa A, Okamoto N et al (1992b) Structure of serum transferrin in carbohydrate-deficient glycoprotein syndrome. Biochem Biophys Res Commun 189:832–836CrossRefPubMed Wada Y, Nishikawa A, Okamoto N et al (1992b) Structure of serum transferrin in carbohydrate-deficient glycoprotein syndrome. Biochem Biophys Res Commun 189:832–836CrossRefPubMed
Zurück zum Zitat Wada Y, Tajiri M, Yoshida S (2004) Hydrophilic affinity isolation and MALDI multiple-stage tandem mass spectrometry of glycopeptides for glycoproteomics. Anal Chem 76:6560–6565CrossRefPubMed Wada Y, Tajiri M, Yoshida S (2004) Hydrophilic affinity isolation and MALDI multiple-stage tandem mass spectrometry of glycopeptides for glycoproteomics. Anal Chem 76:6560–6565CrossRefPubMed
Zurück zum Zitat Wada Y, Tamura J, Musselman BD, Kassel DB, Sakurai T, Matsuo T (1992a) Electrospray ionization mass spectra of hemoglobin and transferrin by a magnetic sector mass spectrometer. Comparison with theoretical isotopic distributions. Rapid Commun Mass Spectrom 6:9–13CrossRefPubMed Wada Y, Tamura J, Musselman BD, Kassel DB, Sakurai T, Matsuo T (1992a) Electrospray ionization mass spectra of hemoglobin and transferrin by a magnetic sector mass spectrometer. Comparison with theoretical isotopic distributions. Rapid Commun Mass Spectrom 6:9–13CrossRefPubMed
Zurück zum Zitat Wang YC, Peterson SE, Loring JF (2014) Protein post-translational modifications and regulation of pluripotency in human stem cells. Cell Res 24:143–160CrossRefPubMed Wang YC, Peterson SE, Loring JF (2014) Protein post-translational modifications and regulation of pluripotency in human stem cells. Cell Res 24:143–160CrossRefPubMed
Zurück zum Zitat West MB, Segu ZM, Feasley CL et al (2010) Analysis of site-specific glycosylation of renal and hepatic gamma-glutamyl transpeptidase from normal human tissue. J Biol Chem 285:29511–29524CrossRefPubMedPubMedCentral West MB, Segu ZM, Feasley CL et al (2010) Analysis of site-specific glycosylation of renal and hepatic gamma-glutamyl transpeptidase from normal human tissue. J Biol Chem 285:29511–29524CrossRefPubMedPubMedCentral
Zurück zum Zitat Wheeler SF, Domann P, Harvey DJ (2009) Derivatization of sialic acids for stabilization in matrix-assisted laser desorption/ionization mass spectrometry and concomitant differentiation of alpha(2 --> 3)- and alpha(2 --> 6)-isomers. Rapid Commun Mass Spectrom 23:303–312CrossRefPubMed Wheeler SF, Domann P, Harvey DJ (2009) Derivatization of sialic acids for stabilization in matrix-assisted laser desorption/ionization mass spectrometry and concomitant differentiation of alpha(2 --> 3)- and alpha(2 --> 6)-isomers. Rapid Commun Mass Spectrom 23:303–312CrossRefPubMed
Zurück zum Zitat Wopereis S, Grunewald S, Huijben KM et al (2007) Transferrin and apolipoprotein C-III isofocusing are complementary in the diagnosis of N- and O-glycan biosynthesis defects. Clin Chem 53:180–187CrossRefPubMed Wopereis S, Grunewald S, Huijben KM et al (2007) Transferrin and apolipoprotein C-III isofocusing are complementary in the diagnosis of N- and O-glycan biosynthesis defects. Clin Chem 53:180–187CrossRefPubMed
Zurück zum Zitat Wopereis S, Grunewald S, Morava E et al (2003) Apolipoprotein C-III isofocusing in the diagnosis of genetic defects in O-glycan biosynthesis. Clin Chem 49:1839–1845CrossRefPubMed Wopereis S, Grunewald S, Morava E et al (2003) Apolipoprotein C-III isofocusing in the diagnosis of genetic defects in O-glycan biosynthesis. Clin Chem 49:1839–1845CrossRefPubMed
Zurück zum Zitat Wopereis S, Lefeber DJ, Morava E, Wevers RA (2006) Mechanisms in protein O-glycan biosynthesis and clinical and molecular aspects of protein O-glycan biosynthesis defects: a review. Clin Chem 52:574–600CrossRefPubMed Wopereis S, Lefeber DJ, Morava E, Wevers RA (2006) Mechanisms in protein O-glycan biosynthesis and clinical and molecular aspects of protein O-glycan biosynthesis defects: a review. Clin Chem 52:574–600CrossRefPubMed
Zurück zum Zitat Wuhrer M, Koeleman CA, Deelder AM, Hokke CH (2004) Normal-phase nanoscale liquid chromatography−mass spectrometry of underivatized oligosaccharides at low-femtomole sensitivity. Anal Chem 76:833–838CrossRefPubMed Wuhrer M, Koeleman CA, Deelder AM, Hokke CH (2004) Normal-phase nanoscale liquid chromatography−mass spectrometry of underivatized oligosaccharides at low-femtomole sensitivity. Anal Chem 76:833–838CrossRefPubMed
Zurück zum Zitat Xia B, Zhang W, Li X et al (2013) Serum N-glycan and O-glycan analysis by mass spectrometry for diagnosis of congenital disorder of glycosylation. Anal Biochem 442:178–185CrossRefPubMed Xia B, Zhang W, Li X et al (2013) Serum N-glycan and O-glycan analysis by mass spectrometry for diagnosis of congenital disorder of glycosylation. Anal Biochem 442:178–185CrossRefPubMed
Zurück zum Zitat Yang Y, Franc V, Heck AJR (2017) Glycoproteomics: a balance between high-throughput and in-depth analysis. Trends Biotechnol 35:598–609CrossRefPubMed Yang Y, Franc V, Heck AJR (2017) Glycoproteomics: a balance between high-throughput and in-depth analysis. Trends Biotechnol 35:598–609CrossRefPubMed
Zurück zum Zitat Zeevaert R, Foulquier F, Jaeken J, Matthijs G (2008) Deficiencies in subunits of the conserved oligomeric Golgi (COG) complex define a novel group of congenital disorders of glycosylation. Mol Genet Metab 93:15–21CrossRefPubMed Zeevaert R, Foulquier F, Jaeken J, Matthijs G (2008) Deficiencies in subunits of the conserved oligomeric Golgi (COG) complex define a novel group of congenital disorders of glycosylation. Mol Genet Metab 93:15–21CrossRefPubMed
Zurück zum Zitat Zhang W, James PM, Ng BG et al (2016) A novel N-tetrasaccharide in patients with congenital disorders of glycosylation, including asparagine-linked glycosylation protein 1, phosphomannomutase 2, and mannose phosphate isomerase deficiencies. Clin Chem 62:208–217CrossRefPubMed Zhang W, James PM, Ng BG et al (2016) A novel N-tetrasaccharide in patients with congenital disorders of glycosylation, including asparagine-linked glycosylation protein 1, phosphomannomutase 2, and mannose phosphate isomerase deficiencies. Clin Chem 62:208–217CrossRefPubMed
Zurück zum Zitat Zielinska DF, Gnad F, Wisniewski JR, Mann M (2010) Precision mapping of an in vivo N-glycoproteome reveals rigid topological and sequence constraints. Cell 141:897–899CrossRefPubMed Zielinska DF, Gnad F, Wisniewski JR, Mann M (2010) Precision mapping of an in vivo N-glycoproteome reveals rigid topological and sequence constraints. Cell 141:897–899CrossRefPubMed
Metadaten
Titel
Clinical glycomics for the diagnosis of congenital disorders of glycosylation
verfasst von
Nurulamin Abu Bakar
Dirk J. Lefeber
Monique van Scherpenzeel
Publikationsdatum
01.03.2018
Verlag
Springer Netherlands
Erschienen in
Journal of Inherited Metabolic Disease / Ausgabe 3/2018
Print ISSN: 0141-8955
Elektronische ISSN: 1573-2665
DOI
https://doi.org/10.1007/s10545-018-0144-9

Weitere Artikel der Ausgabe 3/2018

Journal of Inherited Metabolic Disease 3/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.