You are on page 1of 21

Eur J Pediatr (2003) 162: 359–379

DOI 10.1007/s00431-002-1136-0

R EV IE W

T. Marquardt Æ J. Denecke

Congenital disorders of glycosylation: review of their molecular bases,


clinical presentations and specific therapies

Received: 14 March 2002 / Revised: 6 November 2002 / Accepted: 7 November 2002 / Published online: 15 March 2003
 Springer-Verlag 2003

Abstract Congenital disorders of glycosylation (CDG,


formerly named carbohydrate-deficient glycoprotein
Introduction
syndromes) are a rapidly growing family of inherited
Congenital disorders of glycosylation (CDG) are inher-
disorders affecting the assembly or processing of glycans
ited metabolic diseases caused by defects in the biosyn-
on glycoconjugates. The clinical spectrum of the differ-
thesis of glycoconjugates. These disorders, formerly
ent types of CDG discovered so far is variable, ranging
known as carbohydrate-deficient glycoprotein syn-
from severe multisystemic disorders to disorders
dromes (CDG-syndromes, CDGS), were recently
restricted to specific organs. This review deals with
renamed due to limitations of the old nomenclature
clinical, diagnostic, and biochemical aspects of all
[13, 14].
characterized CDGs, including a disorder affecting the
In CDG, hypoglycosylation of different glycoproteins
N-glycosylation of erythrocytes, congenital dyserythro-
and occasionally of other glycoconjugates leads to a
poietic anemia type II (CDA II/HEMPAS), and the first
variety of symptoms affecting multiple systems. In most
disorders affecting O-glycosylation. Since the clinical
cases, statomotor and mental retardation is present.
spectrum of symptoms in CDG is variable and may be
Although about 1% of the human genome codes for
unspecific, a generous selective screening for the pres-
genes involved in glycosylation processes [37], it was not
ence of CDG is recommended.
until 1995 that a disorder in the biosynthesis of N-gly-
cans was first characterized [137]. This review focuses on
Keywords CDG Æ LAD II Æ CDA II Æ HEMPAS Æ
the clinical presentation of the different CDG types,
Glycosylation
their biochemical and molecular basis, and the specific
treatment options known to date. Online information
Abbreviations aa Amino acid Æ CDA Congenital
can be found at our homepage (http://cdg.uni-
dyserythropoietic anemia Æ CDG Congential disorder of
muenster.de/) and a mutation database at http://
glycosylation Æ HEMPAS Hereditary erythroblastic
www.med.kuleuven.ac.be/cdg/.
multinuclearity with positive acidified serum test Æ LLO
Lipid-linked oligosaccharides Æ MRI Magnetic
resonance imaging Æ OST Oligosaccharyltransferase Æ N-glycosylation of proteins
PDO Protein-derived oligosaccharides Æ RER Rough
endoplasmic reticulum Æ Sia Sialic acid Most types of CDG that have been discovered so far
were identified by demonstrating defects in the N-gly-
cosylation pathway of glycoproteins. Although O-gly-
cosylation or the glycosylation of glycolipids is affected
in some of them, the main alterations concern the N-
glycosylation pathway. Understanding the processes
Further information on congenital disorders of glycosylation
(CDG) can be found at: http://cdg.uni-muenster.de/
involved is therefore essential to an understanding of the
biochemical basis of known CDG as well as of types still
T. Marquardt (&) Æ J. Denecke to be discovered.
Klinik und Poliklinik für Kinderheilkunde, Protein translation occurs on ribosomes either in the
Albert-Schweitzer-Str. 33, 48149 cytoplasm or on the membrane of the rough endoplas-
Münster, Germany
E-mail: marquat@uni-muenster.de mic reticulum (RER). Whereas proteins synthesized in
Tel.: +49-251-8358519 the cytoplasm generally remain unglycosylated, the
Fax: +49-251-8356085 majority of proteins that are secreted or destined to be
360
361
362
b tensive research in the past. One of the most important
Fig. 1 Synthesis of dolichol, assembly and processing of N-linked summaries is the review by Kornfeld & Kornfeld [85] but
glycans and molecular defects of known CDG types. Dolichol is a other more recent reviews give an extensive overview as
polyisoprenyl containing 18–21 isoprene units. It serves as an
precursor monosaccharide donor and carrier of the assembled well [59]. Assembly and processing reactions are sum-
oligosaccharide in the ER membrane prior to its transfer to the marized in Fig. 1.
nascent protein. Enzymes catalyze the conversion of isopentenyl- Biosynthesis of the oligosaccharide starts at the cy-
pyrophosphate and farnesylpyrophosphate to polyprenol and toplasmic site of the RER membrane. Dolichol phos-
finally dolichol phosphate (upper part). Assembly of N-glycans is
initiated on the cytoplasmic side of the ER membrane by
phate (Dol-P) is the acceptor for the first assembly
transferring two GlcNAc residues (blue squares) to Dol-P using reaction catalyzed by a GlcNAc 1-P transferase that uses
UDP-GlcNAc as GlcNAc donor. This process is catalyzed by UDP-GlcNAc as GlcNAc donor. The product, Glc-
GlcNAc 1-P transferase and chitobiose synthase. The product NAc-P-P-Dol, is then extended by chitobiose synthase
GlcNAc2-P-P-Dol, is extended by different mannosyltransferases that adds a second GlcNAc in b1,4-linkage, again using
using GDP-mannose (mannose: red dots) as the donor substrate.
After assembly of a biantennary Man5GlcNAc2 structure on the UDP-GlcNAc as donor. This structure is further
cytoplasmic side of the ER-membrane, the oligosaccharide is elongated by different mannosyltransferases that use
translocated to the lumenal side of the organelle (light grey), GDP-mannose as donor substrate. The first mannose is
catalyzed by a flipping enzyme. For the subsequent enzymatic attached in b1,4-linkage to the GlcNAc and sequentially
steps, mannose and glucose attached to Dol-P must be translocated
into the lumen of the ER by a Dol-P-sugar flipping mechanism. the oligosaccharide branches into an a1,3 mannose and
ER-localized mannosyl- and glucosyltransferases elongate the an a1,6 mannose branch. The a1,3 branch is extended by
branches to a Glc3Man9GlcNAc2 oligosaccharide (glucose: yellow two more mannoses in order to build up a biantennary
triangle). This oligosaccharide is transferred by the multi-subunit Man5GlcNAc2 structure. At this stage, the oligosac-
enzyme OST to glycosylation consensus sites of the nascent
protein. After removal of the glucose residues and one mannose
charide is translocated from the cytoplasmic side of the
from the a1,6 branch, the glycoprotein is transferred to the Golgi RER membrane to the lumen where further elongation
by vesicular transport. In the Golgi, mannosidase I cleaves takes place. A flipping enzyme catalyzing this reaction
mannose, enabling the transfer of GlcNAc onto the a1,3 arm by has recently been identified in yeast [58]. The protein,
the GlcNAc-transferase I. Mannosidase II cleaves 2 mannose Rft1, catalyzes the flipping of Man5GlcNAc2-PP-Dol
residues from the a1,6 arm, followed by the transfer of GlcNAc by
GlcNAc-transferase II. Following this step, the structure of but not of Dol-P-Man [58]. Further elongation steps use
N-glycans is characterized by a broad heterogeneity. Fucosyltrans- Dol-P-Man and Dol-P-Glc as substrates since the
ferase 8 may transfer fucose (grey triangle) in a1,6 linkage to the membrane of the RER is impermeable to UDP-GlcNAc
core GlcNAc and multiple galactosyltransferases, GlcNAc-trans- and GDP-mannose. Dolicholphosphate sugars are syn-
ferases, sialyltransferases and fucosyltransferases spread over cis,
median, and trans-Golgi catalyze the formation of complex thesized on the cytoplasmic side and have to flip to the
oligosaccharides. Green rhomb, galactose; pink rhomb, sialic acid. lumen, a step that is probably enzyme-catalyzed. The
The mouse symbol designates the availability of a knock-out mouse, gene product of the human MPDU1 gene was recently
in red are cell lines generated from yeast or from CHO cells and established as being essential for the flipping of Dol-
expressing specific enzymatic defects
P-Man and Dol-P-Glc from the cytoplasmic to the
lumenal side of the ER, although MPDU1 seems not to
be the flippase itself [140]. ER-localized mannosyl- and
membrane proteins are glycosylated. The latter are glucosyltransferases then elongate the oligosaccharide to
synthesized on ribosomes attached to the outer mem- its final structure. The a1,6-branch is further elongated
brane of the RER. Upon the start of translation, the with mannoses generating another a1,3/ a1,6-branching,
N-terminal part of these proteins is translocated across whereas the a1,3-branch is elongated with two glucose
the membrane. At the lumenal side of the membrane, a residues in a1,3-linkage and a terminal glucose in
multimeric enzymatic complex, oligosaccharyl transfer- a1,2-linkage.
ase (OST), recognizes the motif Asn-X-Ser/Thr (gly- This preassembled oligosaccharide, Glc3Man9Glc-
cosylation consensus site; X cannot be proline) and NAc2, is subsequently transferred by oligosaccharyl-
attaches a preassembled oligosaccharide to the aspara- transferase (OST) to glycosylation consensus sites of the
gine. There is only a short time frame for the OST to nascent chain. 9 subunits of OST have been found in
transfer the oligosaccharide. The translocation speed is yeast and four corresponding subunits have been iden-
not constant, and pauses during translocation might tified in humans. OST has a high substrate affinity with
facilitate the glycosylation of these sites. the fully assembled oligosaccharide whereas shorter
The transferred oligosaccharide is a branched struc- species are transferred inefficiently. Immediately after
ture of 14 monosaccharides consisting of two N-acetyl- the transfer, the oligosaccharide is trimmed by the
glucosamines (GlcNAc), nine mannoses (Man) and three membrane-bound glucosidase I that removes the termi-
glucose residues (Glc). Biosynthesis of this structure nal a1,2-linked glucose, followed by removal of the two
occurs at the membrane of the rough endoplasmic re- a1,3-linked glucose residues by soluble a-glucosidase II.
ticulum (RER) in a step-by-step process on a lipid an- High-mannose oligosaccharides with a single glucose
chor, dolichol (Fig. 1). Dozens of enzymes and residue interact with ER chaperones calnexin and cal-
transporters are required for the synthesis of this lipid- reticulin, an interaction that is essential for the so-called
linked oligosaccharide (LLO). The assembly steps of the quality control of newly folded glycoproteins in the
oligosaccharide on dolichol have been the focus of in- RER [110]. After the removal of the first a1,2 mannose
363

from the branched a1,6 arm by one of two ER-based see [51]). Further elongation and modification by
a-mannosidases, the glycoproteins travel to the Golgi by polylactosamine extension, sialylation, fucosylation,
vesicular transport. sulfatation and acetylation produce a broad variety of
In the Golgi, heterogeneity of carbohydrate struc- glycans. Major antigens such as AB0 blood groups and
tures is generated by a variety of enzymes. Mannosidases Lewis antigens are often present on N- and O-glycans
I generate a Man5 structure followed by elongation of [10].
the a1,3 arm with GlcNAc in b1,2-linkage by GlcNAc- Many glycosyltransferases, especially for the synthe-
transferase I (GnT I). Mannosidase II cleaves the two sis of the glycan core structures, are specific to O-gly-
mannose residues from the a1,6 arm, and GlcNAc- cosylation, but there are overlapping enzyme patterns
transferase II (GnT II) adds GlcNAc in b1,2-linkage to for N- and O-glycosylation as well [10, 33, 51, 135].
the other branch. Galactosyltransferase and sialyltrans- Besides these prevailing types of O-glycosylation,
ferase then elongate both branches to the final carbo- there are some deviating pathways for O-glycans: 1)
hydrate structure (Sia2Gal2GlcNAc2Man3GlcNAc2). addition of b-O-GlcNAc to Ser/Thr of various nuclear
Fucosyltransferase 8 (FuT8) catalyzes the a1,6 core fu- and cytoplasmatic proteins by a specific and highly
cosylation present at some glycoproteins [105]. a1,6-FuT conserved GlcNAc-transferase, often regulating the ac-
activity requires an unsubstituted GlcNAc residue of the tivity of transcription factors by competition with
a1,3-antennae of N-glycans [125] and the asialo-, aga- phosphorylation [22, 55, 89], 2) direct glucosylation,
lacto-triantennary structure is the most suitable sub- fucosylation and galactosylation of specific proteins [5,
strate [77, 105]. Besides biantennary oligosaccharides of 53], 3) O-mannosylation [33].
the structure described above, a large variety of different There are four glycosylation disorders concerning
glycans is generated in the Golgi apparatus. O-glycosylation known so far. The progeria variant of
Defects in the early biosynthesis pathway lead to a Ehlers Danlos syndrome, the hereditary multiple exos-
uniform hypoglycosylation phenotype of glycoproteins. toses disease, the muscle eye brain, and the Walker
This is caused by a distinct substrate specificity of OST: Warburg disease. The latter two impair the assembly of
whereas Glc3Man9GlcNAc2 is efficiently transferred O-mannosyl glycans [148]. O-mannosylation is a fre-
by OST, most of the intermediates are not. For this quent and essential type of glycosylation in yeast but
reason, many different defects leading to a truncation only a few proteins of mammals, mainly of brain and
of the LLO will result in a lack of the complete oli- muscle, carry O-mannosyl glycans [33, 47].
gosaccharide on the glycoprotein at some glycosylation The pathway of O-mannosylation in yeast is well
consensus sites. There are conflicting data on whether known. The first and second mannose residues are at-
the non-occupation of certain consensus sites is random tached to the proteins in the ER using Dol-P-Man as
[146] or not (Winchester et al., Glycobiology meeting, substrate donor. Further elongation is performed in the
Boston 2000). Mutations in one of the OST subunits Golgi resulting in linear chains of 1–7 mannose residues
have been shown to have the same effect [123, 147], but [33]. In mammals, the synthesis pathway of O-man-
no corresponding human disorders have been found so nosylation is not fully understood, but there is evidence
far. that the first step of the O-mannosylation in mammals,
catalyzed by the protein-O-mannosyl transferase
(POMT1), is located in the ER, whereas elongation of
O-glycosylation of proteins this glycan is located in the Golgi. In contrast to yeast O-
mannosyl glycans, O-glycans of mammals may carry
O-glycosylation is a widespread type of glycosylation in other sugars besides mannose.
eukaryotic and in part also in prokaryotic cells. Whereas
N-glycosylation starts cotranslationally in the ER and
further processing takes place in the Golgi, O-glycosy- CDG types
lation occurs posttranslationally and is generally initi-
ated in the cis-Golgi [118]. Hydroxyl groups of the Eleven different human disorders in the N-glycosylation
amino acids serine (Ser), threonine (Thr), and sometimes pathway and three in O-glycosylation are known today.
proline are the acceptor sites of the protein. No con- These disorders have different clinical phenotypes and it
sensus sequences of amino acids determining O-gly- is important to realize that some of these diseases have
cosylation sites could be elucidated, although there is little more in common than their names and biochemical
evidence that some structures are glycosylated prefer- bases. Whereas the former nomenclature was based
entially [135]. mainly on the isoelectric focusing pattern of serum
The first step in the most common type of O-gly- transferrin, the new nomenclature is based on the in-
cosylation, mucin-type O-glycosylation, is the transfer of tracellular localization of the molecular defect. All bio-
N-acetylgalactosamine (GalNAc) to Ser/Thr of the synthesis defects up to and including OST are named
protein in the Golgi, forming the so-called Tn-antigen. type I, whereas defects in the processing of the oligo-
Addition of Gal, GalNAc or GlcNAc onto this first saccharide or O-glycosylation defects are named type II.
GalNAc builds up a panel of eight different core struc- The different enzymatic defects are characterized by
tures known so far in mucin-type O-glycans (for review small letters [13, 14].
364

Table 1 CDG types characterized to date

New name Old name Affected protein Affected gene Reference

CDG-I
CDG-Ia CDGS Ia Phosphomannomutase 2 PMM 2 [137]
CDG-Ib CDGS Ib Phosphomannose Isomerase PMI [106]
CDG-Ic CDGS Ic/V Dol-P-Glc: hALG6 [63, 82]
Man9GlcNAc2-Glucosyltransferase
CDG-Id CDGS IV Dol-P-Man: hALG3/Not56L [83]
Man5GlcNAc2-Mannosyltransferase
CDG-Ie CDGS IV Dol-P-Man Synthase 1 DPM 1 [65, 79]
CDG-If – MPDU1/Lec35 [86, 121]
CDG-Ig – Dol-P-Man: hALG12 [134]
Man7GlcNAc2PP-Dol mannosyltransferase
CDG-II
CDG-IIa CDGS II GlcNAc-Transferase 2 (GnT II) MGAT2 [74, 133]
CDG-IIb – Glucosidase I HSAGLUCIE [30]
CDG-IIc LAD II GDP-Fucose Transporter [92, 93]
CDG-IId – b1,4 galactosyltransferase [52]

The previously described CDG syndrome type III CDG-Ia children by decreasing the low PMM activity in
[127] now belongs to the CDG-x group since no their cells even further. PMM2 is widely distributed in
underlying defect is known. Since hardly any protein is tissues, with the highest activity in intestinal mucosa
hypoglycosylated in this disorder, it is doubtful whether [113]. In contrast, a second PMM, PMM1, was found in
it is a primary glycosylation disorder at all. More than rats to be expressed only in lung and brain [113]. Sur-
20% of the CDG patients identified still cannot be as- prisingly, PMM1 accounts for 66% of the total PMM
cribed to one of the known enzyme defects and are activity in rat brain [113]. Under the assumption that
named CDG-x. CDG-Ia to CDG-Ig and CDG II-a to this is similar in humans, it is still unclear why the loss of
CDG-IId are currently characterized (Table 1). PMM2 activity in the human brain is not compensated
for by PMM1.
Inverted nipples and abnormal subcutaneous fat pads
CDG-Ia are characteristics of CDG-Ia and are already present at
birth [21] (Fig. 2b,c). Fat pads disappear with increasing
CDG-Ia (McKusick 212065) was first observed in ho- age [88]. Axial hypotonia, psychomotor retardation and
mozygous twin sisters and published in an abstract by internal strabismus (Fig. 2 a) are common symptoms.
Jaeken and coworkers in 1980 [71]. It is the most frequent Ataxia on the basis of marked cerebellar atrophy is
and best known CDG with more than 500 patients usually present and some children develop epilepsy [70,
worldwide and is caused by a deficiency of phospho- 88]. Tendon reflexes have commonly disappeared by
mannomutase, a cytosolic enzyme that converts mannose 1 year of age [70, 88] and nerve conduction velocity es-
6-P to mannose 1-P [137] (Fig. 1). As a consequence, the pecially at the lower limbs is already decreased by the
GDP-Man pool in CDG-Ia fibroblasts is reduced to 10% age of 6–8 months [69, 88]. Joint contractures and
of normal [119]. Whereas normal fibroblasts mainly skeletal deformities may occur. Most children do not
synthesize Glc3Man9GlcNAc2 oligosaccharides on their learn to walk without support. Feeding problems and
dolichol, CDG-Ia fibroblasts predominantly make failure to thrive often make tube feeding necessary [112].
truncated forms, mainly Man5GlcNAc2 [109]. Impaired night vision and retinitis pigmentosa have been
Many different mutations have been found in the reported [130]. Stroke-like episodes, usually with com-
corresponding gene, PMM2, located on chromosome plete recovery can occur mainly during feverish infec-
16p13 [99]. The most frequent one, R141H, accounts for tions [69, 88]. Sometimes hypoventilation makes
40% of the disease alleles of CDG-Ia patients [122]. It artificial ventilation necessary early on [21]. Hypertro-
has a high carrier frequency of 1/70 in the general phic obstructive cardiomyopathy (HOCM) can develop
population, but has never been found in a homozygous very rapidly after birth and cardial effusion may be de-
form, suggesting that this combination may be lethal [81, tectable [21]. Nonimmune hydrops fetalis is a possible
100, 122]. The second most frequent mutation, F119L, presentation of CDG-Ia in newborns [27]. Ultrasound
has a much lower carrier frequency, estimated at 1/1000
[122]. PMM with the R141H mutation has virtually no c
activity (0.4% of normal) [81, 114]. Whereas wild-type Fig. 2a–d Phenotype of different CDG types. CDG-Ia: internal
PMM has a normal activity at 40C, many mutant forms strabism (a), inverted nipples (b) and abnormal fat pads (c) as
have low thermal stability [81, 114]. V231M has normal characteristics of infants suffering from CDG-Ia. CDG-IIc: short
limbs and a flat face with a broad and depressed nasal bridge are
activity at 30C, but none at 40C [114]. Although these features of patients suffering from CDG-IIc (d). CDG-If: ichthyosis
data are from in vitro experiments, fever might endanger and erythema of the skin in a CDG-If patient (e)
365
366

investigations show hyperechogenic kidneys due to [145], or two dimensional gels [54]. Haptoglobin is nor-
multiple microcysts; sometimes nephrotic syndrome is mally absent [54]. It has been clearly demonstrated that
present early on, while renal tubular function is usually the carbohydrates released from the transferrin of CDG-
normal [129]. In the first years of life, mortality is Ia patients are not truncated and that either one or both
around 20% [70], but the clinical condition stabilizes oligosaccharides are missing in about 50% of the trans-
with increasing age and life-threatening events rarely ferrin population [138, 139, 145]. Non-glycosylation has
occur after the first decade. Hypoglycosylation of co- been reported to occur randomly at either of the two
agulation factors is frequent but only rarely results in glycosylation sites [146], but other authors have found a
thrombosis or hemorrhage. preference for one of the sites (Winchester et al., Gly-
As in other CDGs, serum transaminases are often cobiology Meeting, Boston 2000). The IEF pattern of
elevated [107]. Absolute concentrations of transferrin different glycoproteins is normal during intrauterine life
and a1-antitrypsin in the serum are normal [107]. and directly after birth; hypoglycosylation becomes ap-
Hypoproteinemia, low cholesterol levels and low chol- parent in the 2nd–3rd postnatal week [20].
inesterase levels are common findings [107]. Anti- It is still unclear whether dietary supplementation
thrombin III, factor XI and protein C show reduced with mannose has any beneficial effects in CDG-Ia
activities [136]. children. In CDG-Ia fibroblasts, mannose can correct
Hypoglycosylation of serum transferrin in CDG-Ia the size of the truncated lipid-linked oligosaccharides
was first discovered in 1984 [72]. Based on this observa- (LLO) [84, 109]. Addition of 1 mM mannose to the
tion, the isoelectric focusing (IEF) of serum transferrin culture medium has no effect on the GDP-Man pool in
became the commonly used diagnostic test for most normal fibroblasts, but nearly normalizes the reduced
CDG types (Fig. 3). The IEF of serum transferrin of pool in CDG-Ia fibroblasts [109, 119]. The mechanism
CDG patients shows a cathodal shift towards a higher of this correction is unknown. Based on these experi-
isoelectric point caused by the absence of the terminal ments, mannose therapy has been tried in CDG-Ia pa-
sialic acid residues [126]. Not only transferrin but many tients [80, 96, 102, 103]. Mannose given orally in a dose
different serum proteins visualize hypoglycosylation on of 0.2 g/kg b.w. leads to peak mannose concentrations
one-dimensional IEF [60, 61, 107], SDS-PAGE gels of more than 200 lmol/l after 1 h in healthy subjects,
with a clearing half-time of 4 h [3]. Mannose levels in
Fig. 3 Isoelectric focusing (IEF) of serum transferrin. The IEF of CDG-Ia children after oral incorporation are often
serum transferrin is the commonly used screening test for CDG. In lower (T.Marquardt, manuscript in preparation). No
controls, the major band is tetrasialotransferrin. In CDG-I patients clear effect of mannose therapy in CDG-Ia has been
(shown here: CDG-Ia), additional bands are seen, migrating at the reported so far. A 3-week continuous intravenous
position of disialo- and asialotransferrin. As seen in the diagrams
of possible structures on the right, the IEF does not reveal the mannose infusion with serum mannose levels up to
structural change of the attached oligosaccharide. Since only the 2 mmol/l followed by 6 months of oral mannose treat-
terminal sialic acid residues are charged, abnormal migration can ment failed to correct the hypoglycosylation of different
be due to different structural alterations. In addition, an amino acid glycoproteins [102, 103]. However, no study has yet in-
change in the protein core of the transferrin molecule can mimic
CDG. Blue square, GlcNAc; red circle, mannose; green rhomb,
vestigated the glycosylation status of non-liver-derived
galactose; pink rhomb, sialic acid; grey bar, protein core proteins during therapy.
367

CDG-Ib The clinical presentation is milder than in CDG-Ia.


Muscular hypotonia is a common symptom and nearly
CDG-Ib (McKusick 602579) and its therapy were first all patients have recurrent seizures [50]. In contrast to
described in 1998 by our study group [106] and inde- CDG-Ia, there are no characteristic physical stigmata.
pendently by two other groups in the same year [26, 75]. Brainstem auditory-evoked potentials, tendon reflexes
Our group also proposed a therapeutic approach with and nerve conduction velocities are normal and there are
mannose [106]. The disorder is caused by a deficiency of no obvious cerebral abnormalities in MRI [50]. In early
phosphomannose isomerase that affects the endogenous childhood, internal strabism (as observed in CDG-Ia)
production of mannose 6-P (Fig. 1). Several summaries can occur. At 2–3 years of age, the children are able to
on the first patients with CDG-Ib have been published sit and they usually start walking at 3–4 years of age
[28, 38] and approximately 20 patients are known to date. [50]. Speech development is delayed.
In contrast to CDG-Ia, mental and motor develop- The transferrin IEF pattern looks similar to the one
ment is normal [26, 106]. The predominant symptom of found in CDG-Ia. Activities of AT III and factor XI are
CDG-Ib is chronic diarrhea, commonly starting during considerably reduced. The hypoglycosylation of b-CSF
the first year of life [75]. Cyclic vomiting can be a leading trace protein, a CSF glycoprotein with two biantennary
symptom [6, 26]. Failure to thrive and protein-losing N-linked sugars and a molecular weight of 27 kDa, is
enteropathy can occur [106]. Partial villus atrophy can be less severe than in CDG-Ia patients [49].
present in duodenal biopsies and might lead to suspect
coeliac disease [75, 106]. Hypoglycemia occurs frequently
[6, 75] and inadequately elevated insulin levels have been CDG-Id
found to be causative in some patients but not in others
[6, 29]. Some patients developed hepatic fibrosis [6, 28, CDG-Id (McKusick 601110) is caused by a deficiency of
75]. Hypoalbuminemia, elevated aminotransferases and the first Dol-P-Man-dependent mannosyltransferase
low AT III activity are common findings in CDG-Ib that adds mannose to Man5GlcNAc2-P-P-Dol in the
patients [6]. Thrombotic episodes and severe bleeding endoplasmic reticulum [83]. It was formerly termed
complications may complicate the course [106]. Several CDGS IV [83, 128]. Truncated LLOs of the structure
children died [75] or were in life-threatening conditions. Man5GlcNAc2 accumulate. In the three known patients
CDG-Ib was the first disorder of glycosylation where with this disorder, an ortholog gene to the Sacchar-
a specific therapy was available. Symptoms can be ef- omyces cerevisiae hALG3 gene, the human Not (neighbor
fectively treated with the oral uptake of mannose [106]. of tid) 56-like protein gene, had a homozygous missense
Mannose supplementation increases the depleted man- mutation that reduced the enzymatic activity.
nose 6-P pool, thereby normalizing the hypoglycosyla- Only one patient has been described in the literature,
tion of proteins. Mannose normalizes hypoproteinemia another one can be found in the Leuven mutation
and blood coagulation and effectively treats the symp- database, and the third one will be described shortly
toms of CDG-Ib like protein-losing enteropathy and (Denecke and Marquardt, manuscript in preparation).
hypoglycemia. An oral dose of 1 g mannose per kg body The described patient was suffering from tetraspastic
weight per day divided into five doses is used. Higher paresis, microcephaly, severe and intractable seizures,
doses can induce osmotic diarrhea. Whereas the clinical minimal psychomotor development, and atrophy of the
symptoms disappear rapidly, significant improvement of optic nerve. Dysplastic ears, abnormalities of the uvula,
the transferrin IEF pattern during mannose therapy a high-arched palate and coloboma of the iris were
takes several months of treatment to occur [29, 106]. described. Hypoplasia of the cerebellum similar to
Considering the short biological half-life of transferrin CDG-Ia was present [128]. Transferrin IEF showed an
of approximately 1 week, it is unclear why it takes so increase in disialotransferrin whereas only minimal
long for the IEF pattern to normalize. amounts of asialotransferin were detected.

CDG-Ic CDG-Ie

CDG-Ic (McKusick 603147) is caused by a defect in the CDG-Ie (McKusick 603503) is caused by a defect in the
a1,3-glucosyltransferase and is characterized by accu- dolichol-phosphate-mannose synthase 1 gene (DPM 1),
mulation of Man9GlcNAc2-P-P-Dol [12, 63]. The cor- that codes for the cytosolic subunit of the Dol-P-Man
responding gene has 14 exons [64], codes for a 507 aa synthase multimer (Dol-P-Man synthase) [65, 79]. It was
membrane protein, and was named hALG6, derived formerly also termed CDGS IV, a group that was found
from the name of its ortholog in Saccharomyces cerevi- to consist of two subtypes based on different molecular
siae. A333 V is a common mutation [50] but other mu- defects [79, 128]. Four patients with this disorder have
tations have also been described [141]. The disorder was been described so far [65, 79]. In all patients there is
formerly also called type V [82]. some residual enzymatic activity: approximately 5%.
Psychomotor retardation is the main clinical symp- Considering the severity of the clinical phenotype of
tom of the more than 30 CDG-Ic patients known so far. these patients, mutations causing a complete loss of
368

enzymatic activity might very well be lethal. The affected origin, suffered from delayed psychomotor development,
gene codes for the catalytic subunit of Dol-P-Man syn- convulsions starting at the age of 14 months, micro-
thase, has nine exons, and is located on chromosome cephaly, muscular hypotonia, supragluteal fat pats,
20q13. No mutations have so far been identified in the dysplastic ears and a short filtrum. The partial throm-
gene DPM2, which encodes for the second membrane- boplastin time was prolonged. Transferrin IEF showed a
anchored subunit of the enzyme, or in DPM3 another pattern similar to the one seen in CDG-Ia but with less
subunit of Dol-P-Man synthase. LLO analysis is char- asialotransferrin. Magnetic resonance imaging of the
acterized by an accumulation of Man5GlcNAc2-P-P-Dol brain showed enlarged lateral ventricles.
[65, 79]. It has to be proven if CDG-Ie can be caused by The prevailing oligosaccharide structure on the doli-
heterogenous molecular defects. chol of the patient was Man7GlcNAc2 whereas the ma-
Severe psychomotor retardation combined with ture GlcNAc2Man9Glc3 was reduced to about 5% of
muscular hypotonia and seizures starting in the 1st year total dolichol-linked oligosaccharides, thus indicating a
of life are predominant symptoms in children suffering defect in the gene of the Dol-P-Man:Man7GlcNAc2PP-
from CDG-Ie [65, 79]. An absence of visual fixation was Dol mannosyltransferase. This enzyme transfers a
observed in several patients and cortical blindness was mannosyl residue to Dol-PP-GlcNAc2Man7 using Dol-
diagnosed in some of them [65, 79]. Social contact failed P-Man as substrate [11]. The activity of the patient’s
to develop. Gothic palate, hypertelorism, dysplastic nails enzyme was severely reduced and the coding gene, the
and knee contractures are features of the disorder [65]. human ortholog of the yeast ALG12 gene, showed two
Body weight, length and head circumference might be alleles with different point mutations, one resulting in a
normal at birth, but later on microcephaly is typical of stop codon with a loss of the C-terminal 74 amino acids
CDG-Ie. Magnetic resonance imaging reveals cerebral and one causing a substitution of leucine with proline.
atrophy. Transferrin IEF shows a significant amount of
disialotransferrin, but hardly any asialotransferrin [79].
b-CSF trace protein is hypoglycosylated in CDG-Ie CDG-IIa
patients.
There are conflicting results concerning the effect of CDG-IIa (McKusick 212066) is caused by mutations in
mannose supplementation in CDG-Ie fibroblasts: the MGAT2 gene on chromosome 14q21 that codes for
whereas a correction of LLO size was described by one N-acetylglucosaminyltransferase II (GnT II) localized in
group [79], no effect was recorded by others [65]. the Golgi membrane [133]. The entire coding region is
on one single exon.
Compared to CDG-Ia children, more profound psy-
CDG-If chomotor retardation but no peripheral neuropathy and
normal deep-tendon reflexes are present in the four
CDG-If is based on alterations in an RER-located CDG-IIa patients known so far. A dysmorphic coarse
transmembrane protein called MPDU1/Lec35 face, large low-set ears and widely spaced nipples have
(McKusick 604041) [86, 121]. MPDU1 is the abbrevia- been described [117]. CDG-IIa patients show generalized
tion for mannose-P-dolichol utilization defect 1 and hypotonia, limb weakness and stereotypical behavior.
Lec35 CHO cells have the same molecular defect. The Epilepsy developed in one patient. MRI of the brain
product of MPDU1 is a 27 kDa protein with two putative revealed cortical atrophy, focal white matter lesions and
membrane spanning regions and a cytosolic C-terminal delayed myelinization but no cerebellar atrophy. De-
ER retention signal (KKXX). The N-terminal half of the creased AT III, factor IX and XII activities are present
protein is predicted to have a cytosolic orientation [141]. [73]. Transferrin IEF shows a characteristic pattern with
The function of this protein is unknown, but it seems to disialotransferrin as the most prominent band [73, 117].
play a crucial role in the translocation of mannose and
glucose into the lumen of the RER. Man5GlcNAc2 as
well as Man9GlcNAc2 accumulate on dolichol. CDG-IIb
The four patients identified so far have shown psy-
chomotor retardation, muscular hypotonia, seizures, CDG-IIb (McKusick 606056) is caused by a deficiency
absence of speech development, short stature, failure to of glucosidase I, the first enzyme to act on the oligo-
thrive, feeding problems, impaired vision and retinitis saccharide after its transfer to the nascent chain. The
pigmentosa [86, 121]. Two of them have shown ichthy- activity in cultured skin fibroblasts was reduced to less
osis of the skin (Fig. 2e). than 3% of control values. The enzyme has a molecular
mass of 92 kDa and its gene is localized on chromosome
2p12–13.
CDG-Ig Only one patient with CDG-IIb, a girl of consan-
guineous parents, has been identified so far [30]. Dys-
The first patient suffering from CDG-Ig was described morphic features such as broad nose, retrognathism,
by Thiel et al. [134], closely followed by descriptions by high-arched palate and overlapping fingers were de-
other groups [15, 48]. The first patient, a girl of Indian scribed. Muscular hypotonia was present and motor
369

nerve conduction velocity was reduced. Hypoventilation was absent. In early infancy he had had severe episodes
and apnea made artificial ventilation necessary. Seizures of pneumonia, but later on there was no increased
developed at 3 weeks of age. Increasing hepatomegaly incidence of infections. Nevertheless, his leukocyte
developed and histological findings of liver tissue revealed counts were constantly elevated to around 40,000/ll.
proliferation and dilatation of the bile ducts, fibrosis, fat The major clinical problem was persistent periodontitis.
accumulation, iron storage and multilamellar inclusion In two patients, leukocyte adhesion deficiency could
bodies in hepatocytes. Following a rapid decline and a be effectively treated with oral fucose [62, 95, 98]. Oral
stuporous state, the girl died at 2.5 months of age. fucose supplementation did not correct all fucosylation
A prominent abnormal tetrasaccharide band (Glc- processes. H-antigen was still absent on the erythrocyte
Glc-Glc-Man) found upon thin-layer chromatography surface after 2 years of treatment in one of the patients
of the urine was the pathognomonic finding in the CDG- [95, 98]. Depending on the mutation in the GDP-fucose
IIb patient. It is important to note that the IEF patterns transporter, fucose therapy was thought to be ineffective
of serum transferrin and b-CSF trace protein were in some CDG-IIc patients [131], but a patient with a
normal in the patient. Thus, the standard screening test severe truncation of the GDP-fucose transporter also
for CDG fails to detect CDG-IIb patients. Most likely, a responded to fucose therapy [62].
cleavage of the unprocessed oligosaccharides by an
endo-a1,2-mannosidase in the Golgi [59] releases the
tetrasaccharide found in the urine, allowing further CDG-IId
processing of the carbohydrate.
Interestingly, no hypoglycosylation of serum glyco- CDG-IId (McKusick 607091) is caused by a deficiency
proteins occurs in CDG-IIb. It is intriguing to speculate of b1,4 galactosyltransferase I. Only one patient with
that the absence of monoglucosylated oligosaccharide this disorder is known to date. In this child, a frameshift
intermediates in CDG-IIb might disable ER quality in the coding region caused by an insertion led to a
control, which is normally responsible for the retention protein truncated at the C-terminal end by 50 amino
of misfolded glycoproteins [110]. acids. This protein mislocalized to the ER instead of to
the Golgi [52].
The patient, a 2.5-year-old boy, was suffering from
CDG-IIc muscular hypotonia with increased serum creatinine
kinase levels. He presented a Dandy-Walker malforma-
CDG-IIc (McKusick 266265) was discovered by Etzioni tion with macrocephalus at birth and progressive hy-
in 1992 and named leukocyte adhesion deficiency type II drocephalus later on. CDG-IId was detected with the
(LAD II) [35]. Fucosylated glycoconjugates are severely standard transferrin screening test showing an unusual
diminished in this disorder, due to a defect of GDP- pattern where asialo-, monosialo- and to a lesser extent
fucose import into the Golgi [93]. The absence of fu- disialotransferrin were the major isoforms [111].
cosylated selectin ligands on the surface of neutrophil
granulocytes causes the leukocyte adhesion deficiency
[35, 97]. Addition of fucose to the culture medium re- Other glycosylation defects
stores the expression of fucosylated ligands in LAD II
cells [78, 98]. Walker-Warburg syndrome and muscle–eye–brain
In two of the patients, a defect of the GDP-fucose disease
transporter in the membrane of the Golgi apparatus was
found [93, 131]. Both children have mutations in puta- Recently two disorders of O-glycosylation have been
tive transmembrane regions of the transporter [93]. described [8, 148] (Fig. 4). A defect in the gene of the
Short limbs and stature, a flat face with a broad and protein O-mannosyl b-1,2-N-acetylglucosaminyltrans-
depressed nasal bridge, long eyelashes and broad palms ferase (POMGnT1) causes a disease that has been re-
are dysmorphic features of the five known patients [35, ferred to as muscle–eye–brain (MEB) disease (McKusick
39, 97] (Fig. 2d). Moderate to severe psychomotor re- 253280) [116]. The gene responsible for MEB disease
tardation is present. Increased peripheral leukocyte had previously been mapped to chromosome 1p32–34
counts are the predominant finding and are already [23] and now POMGnT1 could be located to the same
present in newborns. Leucocytosis and immune region. POMGnT1 is a type II membrane protein similar
deficiency are due to the absence of fucosylated selectin to other Golgi glycosyltransferases and catalyzes the b-
ligands, decreasing the adhesion of leukocytes to endo- 1,2 linkage of GlcNAc to O-linked mannose using UDP-
thelial cells and migration of neutrophils to infection GlcNAc as donor substrate. The gene consists of 22
focuses [35, 97]. Since a1,2 fucose is a component of the exons coding for a protein of approximately 80 kDa.
H-antigen, patients lack a detectable ABO blood group Northern blot analysis revealed a broad distribution in
on their erythrocytes (Bombay blood group) [35]. several tissues. Patients showed homozygous and com-
Etzioni described a 5-year follow-up of a 10-year-old pound heterozygous point mutations in the affected gene
boy with LAD II [36]. The height and weight of the child [148]. The other disorder, Walker-Warburg syndrome
were below the 3rd percentile and speech development (McKusick 236670), affects the first enzyme in the same
370

b
Fig. 4 O-mannosylation. O-mannosylation is a posttranslational
protein modification occurring predominantly in nervous tissue
and in the muscle. Mannosylation of a serine or threonine occurs
probably in the RER and further elongation takes place in the
Golgi apparatus. Symbols are the same as in Fig. 1

glycosylation pathway, protein O-mannosyl transferase


I (POMT1), at least in 30% of the patients [8].
The so-called O-mannosyl glycans are common in
fungi but rare in mammals and are limited to few gly-
coproteins, which are located mainly in brain, nerve and
skeletal muscle [33, 132]. An important O-mannosylated
protein, the dystroglycan complex, consists of a b-dys-
troglycan subunit, a transmembrane glycoprotein, and
a-dystroglycan, an extracellular membrane glycoprotein.
a-dystroglycan is heavily glycosylated. The predicted
weight of the protein core is approximately 74 kDa
whereas the glycosylated protein has an apparent mo-
lecular mass of 156 kDa, judged by SDS-PAGE. N-
glycosylation plays a minor role in a-dystroglycan while
sialylated O-glycans with the structure Siaa2–3Galb1–
4GlcNAcb1–2Man seem to play a crucial role in the
binding of a-dystroglycan to the extracellular matrix
protein laminin [17, 34]. Impairment of a-dystroglycan
function due to the lack of O-mannosyl glycans, neces-
sary for neuronal migration and muscular function,
seems to be the crucial mechanism causing MEB disease.
Although a molecular defect of MEB is now character-
ized, the biosynthetic pathway of O-mannosyl glycans in
mammals is not fully understood. The initiation step of
mammalian O-mannosylation is catalyzed by POMT1,
which seems to be located in the ER (Fig. 4). Further
elongation takes place in the Golgi.
The clinical presentation of MEB disease consists of
congenital glaucoma and myopia, retinal hypoplasia,
pallor of the optic discus, congenital muscular dystro-
phy, severe muscular hypotonia, myoclonic jerks,
mental retardation, hydrocephalus, and electroenceph-
alographic abnormalities. MRI shows pachygyria, a flat
brainstem and cerebellar hypoplasia. The clinical pre-
sentation of Walker-Warburg syndrome is more severe
with lissencephaly, encephaloceles, microphthalmos,
and death usually within the first few months of life
[24].

Progeria variant of Ehlers-Danlos syndrome

The progeria variant of Ehlers-Danlos syndrome


(McKusick 130070) was reported in one patient showing
a clinical picture partially overlapping with Ehlers-
Danlos syndrome and progeria showing hypermobile
joints and loose but elastic skin in addition to aged ap-
pearance and scanty scalp hair [87]. Further symptoms
such as short stature, osteopenia of all bones, defective
deciduous teeth, delayed wound healing, hypotonic
muscle, craniofacial disproportion and developmental
delay were clearly distinct from Ehlers-Danlos syndrome
and progeria.
371

Hypoglycosylation of a small proteoglycan resulting CDA II (HEMPAS)


in a core protein carrying only one xylose O-linked to
serine was the biochemical characteristic implying a Congenital dyserythropoietic anemias (CDAs) are
defect of a b-galactosyltransferase. A defect of the cod- inherited disorders of erythropoiesis. They were first
ing gene was shown by Quentin et al. [115]. The subdivided into three different types by Heimpel et al. in
B4GalT7 catalyzes the second step of the glycosamino- 1968 [56]. CDA type II is the most common form, with
glycan synthesis initiated by the addition of xylose to more than 250 cases described. It is also known as he-
serine of the protein. The activity of B4GalT7 was reditary erythroblastic multinuclearity with positive
markedly reduced and the enzyme was abnormally acidified-serum test (HEMPAS) because of hemolysis of
thermolabile. Molecular characterization of the CDA-II erythrocytes by sera of about 30% of healthy
B4GalT7 gene [2, 108] allowed the screening for muta- ABO-compatible donors in a slightly acid pH optimized
tions in the B4GalT7 gene in the patient described by for complement action [25]. Mild to severe normocytic
Kresse et al. This patient was compound heterozygote anemia is the predominant symptom; jaundice, spleno-
for one mutation, resulting in a non-functional protein megaly (Fig. 5a) and gallbladder disease are common.
mislocalized in the cytoplasm (L206P) and another Hemochromatosis and liver cirrhosis rarely complicate
mutation in the same exon resulting in an enzyme with the course of the disease [57, 142]. Due to the often mild
markedly reduced activity (A186D) while the parents clinical presentation, the median age of diagnosis was
were heterozygous for either of the mutations [108]. 15.9 years in a study covering 98 CDA II patients [66].
In CDA II patients, 10–40% of erythroblasts are bi- and
multinucleated (Fig. 5b), an observation that serves as a
Hereditary multiple exostoses major diagnostic criterion [56]. Endoplasmic reticulum
membranes are found close to the cell membrane in
Hereditary multiple exostoses (HME) (McKusick erythroblasts and erythrocytes [9, 66, 142] (Fig. 5d). The
133700 and 133701) is the most frequent type of benign observation that band 3 (anion exchange protein 1) and
bone tumor. The incidence is about 1 to 50,000–100,000 band 4.5 (Glut1), two prevailing membrane proteins of
in Western populations, with an autosomal dominant red blood cells, show a narrower aspect and faster mi-
inheritance. Patients suffer from multiple cartilage-cap- gration in sodium dodecyl sulfate-polyacrylamide gel
ped tumors located primarily at the long bones, often a electrophoresis (SDS-PAGE) [4, 7, 76] revealed that a
short stature and various orthopedic deformities [124]. defect in glycosylation of erythrocyte membrane pro-
The risk of developing malignancies of the cartilage or teins is a common biochemical characteristic of all pa-
bone is significantly increased in HME patients. For- tients suffering from CDA II and may be the cause of the
mation of exostoses starts in early infancy and continues disorder [41, 101, 120]. Glycosylation abnormalities are
until the cessation of growth. limited to erythroblasts [76]. Although a report de-
Mapping studies revealed that the vast majority of scribing hypoglycosylation of serum transferrin in one
HME patients are linked to chromosome 8q24.1 [94] and patient exists [45], glycoproteins synthesized in the liver
chromosome 11p11-p12 [143]. Subsequently candidate and glycoproteins of lymphocytes are generally not
genes for HME could be cloned and were found to be affected [76].
exostosin-1 (EXT1) and exostosin-2 (EXT2) [143, 144], Band 3 has one N-glycosylation site normally carry-
coding for two homolog and well-conserved type-II ing a branched oligosaccharide with many lactosamine
transmembrane glycoproteins expressed ubiquitously in repeats (Galb1,4GlcNAcb1,3) [40]. Western blot analy-
human tissue. Biochemical studies confirmed that EXT1 sis of erythrocyte membranes probed with tomato lectin,
and EXT2 possess glycosyltransferase activity necessary a lectin specific for polylactosamines, showed severely
for the formation of heparan sulfate by adding single D- reduced signals in CDA II patients [76] (Fig. 5c). Mass
glucoronic acid and N-acetylglucosamine. EXT1 and spectrometry demonstrated a truncation of polylactos-
EXT2 are part of a hetero-oligomeric complex with amines and the presence of incompletely processed high
higher glycosyltransferase activity than the single pro- mannose and hybrid glycans [66, 76]. Structural data of
teins [91, 104]. Thus the mutation of either EXT1 or N-glycans suggested a possible defect of a-mannosidase
EXT2 results in the same clinical phenotype. A knock- II affecting the trimming of the a1,6-branch in early
out mouse of the EXT1 gene showed embryonic lethality Golgi processing. An a-mannosidase II knock-out
whereas heterozygous mice with a 50% reduction of mouse developed dyserythropoietic anemia whereas
heparan sulfate had no obvious abnormalities although glycan formation in other tissues was not affected due to
patients with HME develop exostoses in an autosomal the presence of an additional mannosidase [18]. In two
dominant trait [90]. The development of exostoses is CDA II patients, a-mannosidase II was shown to have
proposed to be mediated by a lack of heparan sulfate lower activity and a reduced Northern blot signal; one
proteoglycans that play a crucial role in the negative patient had truncated transcripts of the a-ManII gene.
feedback loop regulating chondrocyte proliferation and However, even in these patients, no mutation in the
maturation [32]. There are no causal therapeutic options a-ManII gene could be found [44]. Two further enzymes
for HME; exostoses and deformities of bones are treated were proposed to be affected in other CDA II patients,
symptomatically. N-acetylglucosaminyltransferase II (activities in two
372
373

b negative charges. Any modification of the carbohydrate


Fig. 5a–d HEMPAS. a The clinical picture of CDA II presenting that affects the terminal sialic acid residues therefore
with anemia, icterus and splenomegaly. b Normal, bi- and results in a change in the isoelectric point of the trans-
multinucleated erythroblasts in the bone marrow of CDA II
patients. c Western blot of CDA II erythrocyte membrane ferrin. The IEF pattern does not allow a distinction
preparations probed with tomato-lectin, specific for polylactosam- between truncated or completely missing oligosaccha-
ines, showing a virtual absence of binding in CDA II. d Electron rides.
microscopy of CDA II erythrocytes showing a double membrane In healthy controls, the major form of transferrin
due to the presence of ER close to the cell membrane (arrow). ER
organelle loss seems to be incomplete in these cells
contains two carbohydrate side-chains with two terminal
sialic acids each (tetrasialo-transferrin, isoelectric point
5.4, 80 kDa). Due to triantennary structures, some
patients 11% and 30% of normal) [42] and galactosyl- higher sialylated isoforms are present (Fig. 3, left lane).
transferase (activity of the membrane-bound form 24% In CDG-Ia, a subpopulation of transferrin carries either
of normal in 1 patient) [43]. The patient with reduced only one carbohydrate side-chain (disialo-transferrin) or
galactosyltransferase activity exhibited many severe no carbohydrate at all (asialo-transferrin) (Fig. 3, right
symptoms which are not typical of HEMPAS, and in the lane). The resulting IEF pattern has been described as
coding gene of the galactosyltransferase no mutation type I pattern. All CDG-I entities described so far show
could be found. Thus, reduced activity is likely to be a the same pattern (although the amount of abnormal
secondary effect (M. Fukuda, personal communication). isoforms varies). Thus, the IEF pattern of transferrin
A defect of the N-acetylglucosaminyltransferase II could does not distinguish between the different subtypes of
be assigned to CDG-IIa described in detail above. The CDG.
activity of these three enzymes, the expression levels and In CDG-II, the number of glycans is normal, but
the sequences of the coding genes revealed no abnor- processing defects of the oligosaccharide lead to trun-
malities in other patients. cated carbohydrate structures. In CDG-IIa, truncation
Linkage analysis excluded the genes of a-mannosi- of one branch of the diantennary carbohydrate occurs.
dase, a-mannosidase IIx and N-acetylglucosaminyl- Nearly the whole population of transferrin molecules
transferase II and located the CDA II gene to shows an isoelectric shift, an IEF pattern called the type
chromosome 20q11.2. However, the same group re- II pattern.
ported that 5–10% of CDA II patients could not be Although the IEF of transferrin is still the best
linked to this locus, suggesting a genetic heterogeneity of screening test for CDG, not all CDG types can be de-
CDA II [46, 67, 68]. tected in this way. CDG-IIb, CDG-IIc, muscle–eye–
Although a truncation of N-glycans is the common brain disease, Walker-Warburg syndrome, hereditary
characteristic of CDA II, the molecular basis of this multiple exostoses, progeria variant of Ehlers-Danlos
disorder still remains an enigma. syndrome, and HEMPAS are not detectable by trans-
ferrin IEF. Whereas thin-layer chromatography of urine
oligosaccharides is the method of choice in the case of
Secondary glycosylation disorders CDG-IIb, CDG-IIc and HEMPAS can be detected by a
loss of lectin binding to specific glycoconjugates. Even
In patients with classic galactosemia (galactose-1-phos- some CDG-Ia patients might be missed by the IEF test.
phate uridyltransferase deficiency) before dietary treat- One of our CDG-Ia patients with a severe phenotype
ment and thus under galactose exposure, truncated and common mutations in the PMM2 gene was followed
glycans are formed mimicking CDG in transferrin IEF. up for 6 years and serum samples for transferrin IEF
The majority of carbohydrates released from serum were drawn at regular intervals. The typical type I hy-
transferrin of these patients are asialo-agalactose struc- poglycosylation pattern was observed in the first 4 years
tures [16]. Hypoglycosylation of transferrin can also of follow-up, but since then the IEF pattern as well as
occur in fructose intolerance [1]. AT III and factor XI activities have normalized. Phos-
phomannomutase activity in leukocytes is still below 5%
of normal. The reason for this phenomenon is unknown.
Diagnostic procedures Currently, there is no single screening test for the de-
tection of all forms of CDG.
Screening test: isoelectric focusing of transferrin Functional antithrombin III (AT III) tests often show
reduced activities but there are known CDG patients
The common diagnostic test for CDG is isoelectric fo- with PMM deficiency or other defects who show normal
cusing (IEF) of serum transferrin (Fig. 3). Before IEF is AT III activities. Therefore, transferrin IEF should be
performed, transferrin has to be saturated with iron. performed in all patients where CDG is suspected.
Transferrin has two biantennary N-linked carbohydrate Children with different types of CDG and especially the
side-chains attached to amino acids 413 and 611. In group of children with not yet identified molecular de-
CDG, the carbohydrate chains are either completely fects (CDG-x) show a broad spectrum of clinical
missing or are truncated. In both cases there is a loss of symptoms, making it difficult to decide who should be
terminal a2,6-bound sialic acids, resulting in a loss of screened for CDG. There are known CDG children with
374

dilative cardiomyopathy as the only symptom. Others Table 2 Clinical symptoms, findings of diagnostic procedures, and
have either cutis laxa, ichthyosis, unexplained throm- laboratory investigations that might be associated with CDG
boembolic events or hyperthermia caused by anesthetics. Neurology
Although the majority of children have multisystem Psychomotor retardation (normally present
disease with psychomotor retardation, there are several in all CDG but NOT in CDG-Ib)
children who show no neurological involvement at all. Epilepsia
Stroke-like episodes (CDG-Ia)
Knock-out mice for mannosidase II or GnT V [19, 31] Ataxia
suggest that some forms of autoimmune disease in hu- Decreased or missing tendon reflexes (CDG-Ia)
mans might also be caused by a primary glycosylation Tetraspastic paresis
defect. Currently, it is impossible to devise a general Microcephaly
guideline about who should be screened for CDG and Cerebellar atrophy (CDG-Ia)
Cerebral atrophy
who should not. Nevertheless it is advisable to screen Abnormal eye movements
any child with psychomotor retardation, unexplained Muscular hypotonia
multisystem disease, autoimmune disorders and chronic Decreased nerve conduction velocity (CDG-Ia)
inflammatory disease, cardiomyopathy, protein-losing Eyes
Strabism (CDG-Ia, -Ic)
enteropathy, cyclic vomiting, anemia with reduced os- Retinitis pigmentosa
motic resistance of the erythrocytes, persistent leukocy- Optical atrophy
tosis or low AT III levels for CDG. Symptoms present in Coloboma
patients with inherited glycosylation disorders are sum- Skeletal/Growth
Failure to thrive
marized in Table 2. Growth retardation
There are some pitfalls to the transferrin IEF test. A Short limbs (CDG-IIc)
number of protein variants of transferrin are known and Joint contractures
some of them affect the isoelectric point of the molecule Delayed puberty
[107]. Often, double bands are an indication of a protein Hypogonadism
Gastrointestinal
variant. If an abnormal IEF pattern is detected, IEF be- Chronic diarrhea (CDG-Ib)
fore and after neuraminidase treatment [107] is necessary Protein-losing enteropathy (CDG-Ib)
in order to rule out protein variants. In addition, SDS- Cyclic vomiting (CDG-Ib)
polyacrylamide gel electrophoresis (SDS-PAGE) or sep- Hepatic fibrosis (CDG-Ib)
Heart
aration of the serum glycoproteins on two-dimensional Cardiomyopathy (dilated or hypertrophic; CDG-Ia, CDG-x)
gels gives valuable supplementary information. Glycosy- Cardial effusion
lation of plasma proteins of a fetus suffering from CDG-Ia Hydrops fetalis
was normal during intrauterine life and became abnormal Renal
Congenital nephrotic syndrome (CDG-Ia)
3 weeks after delivery [20]. We additionally found normal Skin
glycosylation of serum proteins in a fetus suffering from Inverted nipples (CDG-Ia)
CDG-Id (Denecke and Marquardt, manuscript in prep- Abnormal fat pads (CDG-Ia)
aration). Thus diagnostic IEF for CDG should not be Ichthyosis (CDG-If)
performed before 3 weeks of age to avoid false-negative Coagulation
Thrombosis
results. Furthermore, other disorders, including untreat- Bleeding tendency
ed fructose intolerance and galactosemia as well as alco- Decreased AT III activity (characteristic for CDG)
hol abuse might lead to functional inhibition of enzymes Decreased clotting factor XI activity
involved in the carbohydrate biosynthesis or processing, Decreased protein C
Laboratory
thus mimicking CDG. Elevated liver enzymes
In all patients where an abnormal IEF test is present, Hypoproteinemia
the specific enzyme defect has to be determined. Enzyme Low cholesterol levels
activities for CDG-Ia and -Ib can be determined in Low cholinesterase levels
Elevated FSH, LH and prolactin
leukocytes; for all other CDG types, cultured skin fi- Anemia
broblasts from the patient are necessary. Leukocytosis (CDG-IIc)
Bombay blood group (CDG-IIc)

In parenthesis CDG types are listed where the symptom is char-


Specific analyses acteristic. Please note that only the most common CDG types are
listed and that these symptoms can also occur in other CDG types.
In order to investigate CDG patients where the clinical Strabism, for instance, is nearly always present in CDG-Ia, can be
phenotype does not clearly suggest a certain type, analysis present in CDG-Ic, but is also seen in other CDG types
of lipid-linked and protein-derived oligosaccharides syn-
thesized in the patients’ fibroblasts is the method of lease Dol-P-Man into the supernatant and with water in
choice. The method has been described in detail [109] and order to release free oligosaccharides, mannose phos-
the principle can be summarized as follows: fibroblasts are phates and sugar nucleotides. The remaining pellet is ex-
labeled for 30 min in the presence of 3H-mannose. The tracted with chloroform/methanol/water in order to
cells are extracted with chloroform/methanol 2:1 to re- release the lipid-linked oligosaccharides (LLO) (being
375

more polar because of their oligosaccharide). The protein- 4. Anselstetter V, Horstmann HJ, Heimpel H (1977) Congenital
derived oligosaccharides (PDO) are released by treatment dyserythropoietic anaemia, type I and II: aberrant pattern of
erythrocyte membrane proteins in CDA II, as revealed by two-
of the final pellet with PNGase F. The oligosaccharides dimensional polyacrylamide gel electrophoresis. Br J Hae-
are then analyzed by HPLC. matol: 209–217
The majority of known CDG types have been discov- 5. Anttinen H, Hulkko A (1980) Regulation of the glycosyla-
ered on the basis of an abnormal LLO profile. If the LLO tions of collagen hydroxylysine in chick embryo tendon and
cartilage cells. Biochim Biophys Acta 632: 417–427
profile in fibroblasts of a CDG-x patient is normal, ra- 6. Babovic-Vuksanovic D, Patterson MC, Schwenk WF, O’Brien
dioactivity incorporated into LLO should be normalized JF, Vockley J, Freeze HH, Mehta DP, Michels VV (1999)
to the amount of total cellular protein to check the Severe hypoglycemia as a presenting symptom of carbohy-
amount of LLO that has been made. Double-labeling with drate-deficient glycoprotein syndrome. J Pediatr 135: 755–781
3
H-mannose and 35S-methionine and determination of 7. Baines AJ, Banga JPS, Gratzer WB, Linch DC, Huehns ER
(1982) Red cell membrane protein abnormalities in congenital
the amounts of the different isotopes incorporated into dyserythropoietic anaemia, type II (HEMPAS). Br J Hae-
newly made proteins is important for the detection of matol 50: 563–574
oligosaccharyltransferase defects. Analysis of PDO 8. Beltran-Valero De Bernabe D, Currier S, Steinbrecher A, Celli
structures by HPLC should be performed in order to J, Van Beusekom E, Van Der Zwaag B, Kayserili H, Merlini
L, Chitayat D, Dobyns WB, Cormand B, Lehesjoki AE,
check for processing defects. Enzymatic release of glycans Cruces J, Voit T, Walsh CA, Van Bokhoven H, Brunner HG
from proteins separated on one- or two-dimensional gels (2002) Mutations in the O-mannosyltransferase gene POMT1
with subsequent MALDI-TOF analysis of the glycan is give rise to the severe neuronal migration disorder Walker-
another valuable tool for the detection of type II CDG. Warburg syndrome. Am J Hum Genet 71: 1033–1043
9. Breton-Gorius J, Daniel MT, Caluvel JP, Dreyfus B (1973)
Anomalies ultrastructurales des érythryoblastes et des éry-
throcytes dans six cas de dysérythropoièse congenitale. Nouv
Future Rev Fr Hematol 13: 23–50
10. Brockhausen I (1999) Pathways of O-glycan biosynthesis in
cancer cells. Biochim Biophys Acta 1473: 67–95
The current knowledge of the clinical spectrum of CDG 11. Burda P, Aebi M (1999) The dolichol pathway of N-linked
is certainly limited and most likely biased by the fact glycosylation. Biochimica and Biophysica Acta 1426: 239–
that it is mainly children with neurological phenotypes 257
that are screened for CDG. The recent discovery of new 12. Burda P, Borsig L, de Rijk-van Andel J, Wevers R, Jaeken J,
CDG types has proven that CDG may cause clinical Carchon H, Berger EG, Aebi M (1998) A novel carbohydrate-
deficient glycoprotein syndrome characterized by a deficiency
phenotypes very different from the known types of this in glucosylation of the dolichol-linked oligosaccharide. J Clin
disorder. Since more than 20% of all children already Invest 102: 647–652
known to have CDG have none of the known molecular 13. Aebi M, Helenius A, Schenk B, Barone R, Fiumara A, Berger
defects, it seems likely that many more disorders will be EG, Hennet T, Imbach T, Stutz A, Bjursell C, Uller A,
Wahlstrom JG, Briones P, Cardo E, Clayton P, Winchester B,
discovered in the coming years. Cormier-Dalre V, de Lonlay P, Cuer M, Dupre T, Seta N, de
Koning T, Dorland L, de Loos F, Kupers L, et al (1999)
Acknowledgements Supported by grants MA 1229/3-1/-2 from the Carbohydrate-deficient glycoprotein syndromes become con-
Deutsche Forschungsgemeinschaft to T.M. and from Innovative genital disorders of glycosylation: an updated nomenclature
Medizinische Forschung (IMF) Münster to T.M. and J.D. S. for CDG. First International Workshop on CDGS (letter).
Bushuven is acknowledged for initial help with the graphic work. Glycoconj J 16:669–671
This review is dedicated to Prof. Erik Harms for his ongoing 14. Aebi M, Helenius A, Schenk B, Barone R, Fiumara A, Berger
support that made our contribution to this rapidly expanding field EG, Hennet T, Imbach T, Stutz A, Bjursell C, Uller A, Wa-
possible.If CDG is suspected, a transferrin IEF test should be hlstrom JG, Briones P, Cardo E, Clayton P, Winchester B,
performed. Many laboratories in Europe offer this service. For CormierDaire V, deLonlay P, Cuer M, Dupre T, Seta N,
analysis in our laboratory, 0.1 ml serum should be sent by regular deKoning T, Dorland L, deLoos F, Kupers L, et al (2000)
mail to: Stoffwechsellabor der Kinderklinik, Domagkstr. 3b, Carbohydrate-deficient glycoprotein syndromes become con-
48129 Münster, Germany. genital disorders of glycosylation: an updated nomenclature
for CDG (abstract). Glycobiology 10:iii–v
15. Chantret I, Dupre T, Delenda C, Bucher S, Dancourt J,
Barnier A, Charollais A, Heron D, Bader-Meunier B, Danos
References O, Seta N, Durand G, Oriol R, Codogno P, Moore SE (2002)
Congenital disorders of glycosylation type Ig is defined by a
1. Adamowicz M, Pronicka E (1996) Carbohydrate-deficient deficiency in dolichyl-P-mannose:Man7GlcNAc2-PP-dolichyl
glycoprotein syndrome-like transferrin isoelectric focusing mannosyltransferase. J Biol Chem 277: 25815–25822
pattern in untreated fructosaemia. Eur J Pediatr: 347–348 16. Charlwood J, Clayton P, Keir G, Mian N, Winchester B
2. Almeida R, Levery SB, Mandel U, Kresse H, Schwientek T, (1998) Defective galactosylation of serum transferrin in ga-
Bennett EP, Clausen H (1999) Cloning and expression of a lactosemia. Glycobiology 8: 351–357
proteoglycan UDP-galactose:beta-xylose beta1,4-galactosyl- 17. Chiba A, Matsumura K, Yamada H, Inazu T, Shimizu T,
transferase I. A seventh member of the human beta4- Kusunoki S, Kanazawa I, Kobata A, Endo T (1997) Struc-
galactosyltransferase gene family. J Biol Chem 274: 26165– tures of sialylated O-linked oligosaccharides of bovine
26171 peripheral nerve alpha-dystroglycan. The role of a novel
3. Alton G, Kjaergaard S, Etchinson JR, Skovby F, Freeze HH O-mannosyl-type oligosaccharide in the binding of alpha-
(1997) Oral ingestion of mannose elevates blood mannose dystroglycan with laminin. J Biol Chem 272: 2156–2162
levels: a first step toward potential therapy for carbohydrate- 18. Chui D, Oh-Eda M, Liao Y-F, Panneerselvan K, Lal A,
deficient glycoprotein syndrome type I. Biochem Molec Med Marek KW, Freeze HH, Moremen KW, Fukuda MN,
60: 127–133 Marth JD (1997) Alpha-mannosidase-II deficiency results in
376

dyserythropoiesis and unveils an alternate pathway in oligo- 36. Etzioni A, Gershoni-Baruch R, Pollack S, Shehadeh N (1998)
saccharide biosynthesis. Cell 90: 157–167 Leukocyte adhesion deficiency type II: long-term follow-up.
19. Chui D, Sellakumar G, Green R, Sutton-Smith M, McQuis- J Allergy Clin Immunol 102: 323–324
tan T, Marek K, Morris H, Dell A, Marth J (2001) Genetic 37. Freeze HH (1998) Disorders in protein glycosylation and
remodeling of protein glycosylation in vivo induces autoim- potential therapy: tip of an iceberg ? J Pediatr 133: 593–600
mune disease. Proc Natl Acad Sci U S A 98: 1142–1147 38. Freeze HH (1999) New diagnosis and treatment of congenital
20. Clayton P, Winchester B, di Tomaso E, Young E, Keir G, hepatic fibrosis. J Pediatr Gastroenterol Nutr 29: 104–106
Rodeck C (1993) Carbohydrate-deficient glycoprotein syn- 39. Frydman M, Etzioni A, Eidlitz-Markus T, Avidor I, Varsano
drome: normal glycosylation in the fetus. Lancet 341: 956 I, Shechter Y, Orlin JB, Gershoni-Baruch R (1992) Ramban-
21. Clayton PT, Winchester BG, Keir G (1992) Hypertrophic Hasharon syndrome of psychomotor retardation, short stat-
obstructive cardiomyopathy in a neonate with the carbohy- ure, defective neutrophil motility, and Bombay phenotype.
drate-deficient glycoprotein syndrome. J Inher Metab Dis 15: Am J Med Gen 44: 297–302
857–861 40. Fukuda M, Dell A, Fukuda MN (1984) Structure of fetal
22. Comer FI, Hart GW (2000) O-Glycosylation of nuclear and lactosaminoglycan. The carbohydrate moiety of band 3 iso-
cytosolic proteins. Dynamic interplay between O-GlcNAc and lated from human umbilical cord erythrocytes. J Biol Chem
O-phosphate. J Biol Chem 275: 29179–29182 259: 4782–4791
23. Cormand B, Avela K, Pihko H, Santavuori P, Talim B, 41. Fukuda MN, Papayannopoulou T, Gordon-Smith EC,
Topaloglu H, de la Chapelle A, Lehesjoki AE (1999) Rochant H, Testa U (1984) Defect in glycosylation of ery-
Assignment of the muscle-eye-brain disease gene to 1p32-p34 throcyte membrane proteins in congenital dyserythropoietic
by linkage analysis and homozygosity mapping. Am J Hum anaemia type II (HEMPAS). Br J Haematol 56: 55–68
Genet 64: 126–135 42. Fukuda MN, Dell A, Scartezzini P (1987) Primary defect of
24. Cormand B, Pihko H, Bayes M, Valanne L, Santavuori P, congenital dyserythropoietic anemia type II: failure in gly-
Talim B, Gershoni-Baruch R, Ahmad A, van Bokhoven H, cosylation of erythrocyte lactosaminoglycan proteins caused
Brunner HG, Voit T, Topaloglu H, Dobyns WB, Lehesjoki by lowered N-acetylglucosaminyltransferase II. J Biol Chem
AE (2001) Clinical and genetic distinction between Walker- 262: 7195–7206
Warburg syndrome and muscle-eye-brain disease. Neurology 43. Fukuda MN, Masri KA, Dell A, Thonar EJ-M, Klier G,
56: 1059–1069 Lowenthal RM (1989) Defective glycosylation of erythrocyte
25. Crookston JH, Crookston MC, Burnie KL, Francombe WH, membrane glycoconjugates in a variant of congenital dysery-
Dacie JV, Davis JA, Lewis SM (1969) Hereditary erythrob- thropoietic anemia type II: association of low level of
lastic multinuclearity with a positive acid serum test: a type membrane-bound form of galactosyltransferase. Blood 73:
of congenital dyserythropoietic anemia. Br J Haematol 17: 1331–1339
11–26 44. Fukuda MN, Masri KA, Dell A, Luzzatto L, Moremen KW
26. De Koning TJ, Dorland L, van Diggelen OP, de Jong GJ, van (1990) Incomplete synthesis of N-glycans in congenital dys-
Noort WL, De Schryver JEAR, Duran M, van den Berg IET, erythropoietic anemia type II caused by a defect in the gene
Gerwig GJ, Berger R, Poll-The BT (1998) A novel disorder of encoding a-mannosidase II. Proc Natl Acad Sci U S A 87:
N-glycosylation due to phosphomannose isomerase deficien- 7443–7447
cy. Biochem Biophys Res Comm 245: 38–42 45. Fukuda MN, Gaetani GF, Izzo P, Scartezzini P, Dell A (1992)
27. De Koning TJ, Toet M, Dorland L, de Vries LS, van den Berg Incompletely processed N-glycans of serum glycoproteins in
IE, Duran M, Poll-The BT (1998) Recurrent nonimmune congenital dyserythropoietic anaemia type II (HEMPAS). Br
hydrops fetalis associated with carbohydrate-deficient glyco- J Haematol 82: 745–752
protein syndrome. J Inherit Metab Dis 21: 681–682 46. Gasparini P, del Giudice EM, Delaunay J, Totaro A,
28. De Koning TJ, Dorland L, van Berge Henegouwen GP (1999) Granatiero M, Melchionda S, Zelante L, Iolascon A (1997)
Phosphomannose isomerase deficiency as a cause of congen- Localization of congential dyserythropoietic anemia II locus
ital hepatic fibrosis and protein-losing enteropathy. J Hepatol to chromosome 20q11.2 by genomewide search. Am J Hum
31: 557–560 Genet 61: 1112–1116
29. De Lonlay P, Cuer M, Vuillaumier-Barrot S, Beaune G, 47. Gemmill TR, Trimble RB (1999) Overview of N- and
Castelnau P, Kretz M, Durand G, Saudubray J-M, Seta N O-linked oligosaccharide structures found in various yeast
(1999) Hyperinsulinemic hypoglycemia as a presenting sign in species. Biochim Biophys Acta 1426: 227–237
phosphomannose isomerase deficiency: a new manifestation 48. Grubenmann CE, Frank CG, Kjaergaard S, Berger EG, Aebi
of carbohydrate-deficient glycoprotein syndrome treatable M, Hennet T (2002) ALG12 mannosyltransferase defect in
with mannose. J Pediatr 135: 379–383 congenital disorder of glycosylation type lg. Hum Mol Genet
30. De Praeter CM, Gerwig GJ, Bause E, Nuytinck LK, 11: 2331–2339
Vliegenthart JF, Breuer W, Kamerling JP, Espeel MF, Martin 49. Grünewald S, Huyben K, de Jong JGN, Smeitink JAM,
JJ, de Paepe AM, Chan NWC, Dacremont GA, van Coster Rubio E, Boers GHJ, Conradt HS, Wendel U, Wevers RA
RN (2000) A novel disorder caused by defective biosynthesis (1999) b-trace protein in human cerebrospinal fluid: a diag-
of N-linked oligosaccharides due to glucosidase I deficiency. nostic marker for N-glycosylation defects in brain. Biochim
Am J Hum Genet 66: 1744–1756 Biophys Acta 1455: 54–60
31. Demetriou M, Granovsky M, Quaggin S, Dennis JW (2001) 50. Grünewald S, Imbach T, Huijben K, Rubio-Gozalbo ME,
Negative regulation of T-cell activation and autoimmunity by Verrips A, de Klerk JBC, Stroink H, de Rijk-van Andel JF,
Mgat5 N-glycosylation. Nature 409: 733–739 Van Hove JL, Wendel U, Matthijs G, Hennet T, Jaeken J,
32. Duncan G, McCormick C, Tufaro F (2001) The link between Wevers RA (2000) Clinical and biochemical characteristics of
heparan sulfate and hereditary bone disease: finding a congenital disorder of glycosylation type Ic, the first recog-
function for the EXT family of putative tumor suppressor nized endoplasmic reticulum defect in N-glycan synthesis.
proteins. J Clin Invest 108: 511–516 Ann Neurol 47: 776–781
33. Endo T (1999) O-mannosyl glycans in mammals. Biochim 51. Hanisch FG (2001) O-glycosylation of the mucin type. Biol
Biophys Acta 1473: 237–246 Chem 382: 143–149
34. Ervasti JM, Campbell KP (1991) Membrane organization of 52. Hanßke B, Thiel C, Lübke T, Hasilik M, Höning S, Peters V,
the dystrophin-glycoprotein complex. Cell 66: 1121–1131 Heidemann PH, Hoffmann GF, Berger EC, von Figura K,
35. Etzioni A, Frydman M, Pollack S, Avidor I, Phillips ML, Körner C (2002) Deficiency of UDP-galactose: N-acetylglu-
Paulson JC, Gershoni-Baruch R (1992) Recurrent severe cosamine b-1,4-galactosyltransferase I as cause of the con-
infections caused by a novel leukocyte adhesion deficiency. N genital disorder of glycosylation type IId (CDG-IId). J Clin
Engl J Med 327: 1789–1792 Invest 109: 725–733
377

53. Harris RJ, Spellman MW (1993) O-linked fucose and other serum arylsulphatase A and increased CSF protein: a new
post-translational modifications unique to EGF modules. syndrome ? Pediatr Res 14: 179
Glycobiology 3: 219–224 72. Jaeken J, van Eijk HG, van der Heul C, Corbeel L, Eeckels R,
54. Harrison HH, Miller KL, Harbison MD, Slonim AE (1992) Eggermont E (1984) Sialic acid-deficient serum and cereb-
Multiple serum protein abnormalities in carbohydrate- rospinal fluid transferrin in a newly recognized genetic syn-
deficient glycoprotein syndrome: pathognomonic finding of drome. Clin Chim Acta 144: 245–247
two-dimensional electrophoresis? Clin Chem 38: 1390–1392 73. Jaeken J, de Cock P, Stibler H, van Geet C, Kint J, Ramae-
55. Hart GW, Greis KD, Dong LY, Blomberg MA, Chou TY, kers V, Carchon H (1993) Carbohydrate-deficient glycopro-
Jiang MS, Roquemore EP, Snow DM, Kreppel LK, Cole RN tein syndrome type II. J Inher Metabol Dis 16: 1041
et al (1995) O-linked N-acetylglucosamine: the ‘‘yin-yang’’ of 74. Jaeken J, Schachter H, Carchon H, De Cock P, Coddeville B,
Ser/Thr phosphorylation? Nuclear and cytoplasmic glycosy- Spik G (1994) Carbohydrate deficient glycoprotein syndrome
lation. Adv Exp Med Biol 376: 115–123 type II: a deficiency in Golgi localised N-acetyl-glucosami-
56. Heimpel H, Wendt F (1968) Congenital dyserythropoietic nyltransferase II. Arch Dis Child 71: 123–127
anemia with karyorrhexis and multinuclearity of erythro- 75. Jaeken J, Matthijs G, Saudubray J-M, Dionisi-Vici C,
blasts. Helv Med Acta 34: 103–112 Bertini E, De Lonlay P, Henri H, Carchon H, Schollen E,
57. Heimpel H, Maier K, Kohne E (1999) Kongenitale dysery- van Schaftingen E (1998) Phosphomannose isomerase defi-
thropoetische Anämien. Monatsschr Kinderkeilkd 147: ciency: a carbohydrate-deficient glycoprotein syndrome with
992–999 hepatic-intestinal presentation. Am J Hum Genet 62: 1535–
58. Helenius J, Ng DT, Marolda CL, Walter P, Valvano MA, 1539
Aebi M (2002) Translocation of lipid-linked oligosaccharides 76. Kameh H, Landolt-Marticorena C, Charuk JHM, Schachter
across the ER membrane requires Rft1 protein. Nature 415: H, Reithmeier RAF (1998) Structural and functional conse-
447–450 quences of an N-glycosylation mutation (HEMPAS) affecting
59. Herscovics A (1999) Importance of glycosidases in mamma- human erythrocyte membrane glycoproteins. Biochem Cell
lian glycoprotein biosynthesis. Biochem Biophys Acta 1473: Biol 76: 823–835
96–107 77. Kaminska J, Glick MC, Koscielak J (1998) Purification and
60. Heyne K, Weidinger S (1992) Diagnostik und Nosologie der characterization of GDP-L-Fuc: N-acetyl beta-D-glucosami-
Glykanose CDG (Carbohydrate-deficient glycoprotein syn- nide alpha1 fi 6fucosyltransferase from human blood plate-
drome). Monatsschr Kinderheilkd 140: 822–827 lets. Glycoconj J 15: 783–788
61. Heyne K, Weidinger S (1994) Drei neue biochemische Marker 78. Karsan A, Cornejo CJ, Winn RK, Schwartz BR, Way W,
bei Glykanose CDG. Monatsschr Kinderheilkd 142: 199–204 Lannir N, Gershoni-Baruch R, Etzioni A, Ochs HD, Harlan
62. Hidalgo A, Ma S, Peired AJ, Weiss LA, Cunnigham-Rundles JM (1998) Leukocyte adhesion deficiency type II is a gener-
C, Frenette PS (2002) Insights into leukocyte adhesion defi- alized defect of de novo GDP-fucose biosynthesis. Endothelial
ciency type II from a novel mutation in the GDP-fucose cell fucosylation is not required for neutrophil rolling an
transporter gene. Blood (prepublished online, 24 Oct 2002), human nonlymphoid endothelium. J Clin Invest 101: 2438–
10.1182/blood-2002-09-2840 2445
63. Imbach T, Burda P, Kuhnert P, Wevers RA, Aebi M, Berger 79. Kim S, Westphal V, Srikrishna G, Mehta DP, Peterson S,
EG, Hennet T (1999) A mutation in the human ortholog of Filiano J, Karnes PS, Patterson MC, Freeze HH (2000)
the Saccharomyces cerevisiae ALG6 gene causes carbohy- Dolichol phosphate mannose synthase (DPM1) mutations
drate-deficient glycoprotein syndrome type-Ic. Proc Natl define congenital disorder of glycosylation Ie (CDG-Ie). J Clin
Acad Sci U S A 96: 6982–6987 Invest 105: 191–198
64. Imbach T, Grünewald S, Schenk B, Burda P, Schollen E, 80. Kjaergaard S, Kristiansson B, Stibler H, Freeze HH, Schwartz
Wevers RA, Jaeken J, de Klerk JBC, Berger EG, Matthijs G, M, Martinsson T, Skovby F (1998) Failure of short-term
Aebi M, Hennet T (2000) Multi-allelic origin of congenital mannose therapy of patients with carbohydrate-deficient gly-
disorder of glycosylation (CDG)-Ic. Hum Genet 106: 538–545 coprotein syndrome type 1A. Acta Paediatr 87: 884–888
65. Imbach T, Schenk B, Schollen E, Burda P, Stutz A, Grüne- 81. Kjaergaard S, Skovby F, Schwartz M (1999) Carbohydrate-
wald S, Bailie NM, King MD, Jaeken J, Matthijs G, Berger deficient glycoprotein syndrome type 1A: expression and
EG, Aebi M, Hennet T (2000) Deficiency of dolichol-phos- characterization of wild type and mutant PMM2 in E. Coli.
phate-mannose synthase-1 causes congenital disorder of gly- Eur J Hum Genet 7: 884–888
cosylation type Ie. J Clin Invest 105: 233–239 82. Körner C, Knauer R, Holzbach U, Hanefeld F, Lehle L, von
66. Iolascon A, D’Agostaro G, Perrotta S, Izzo P, Tavano R, del Figura K (1998) Carbohydrate-deficient glycoprotein syn-
Giudice M (1996) Congenital dyserythropoietic anemia type drome type V: deficiency of dolichol-P-Glc:Man9GlcNAc2-
II: molecular basis and clinical aspects. Haematologica 81: PP-dolichyl glucosyltransferase. Proc Natl Acad Sci U S A
543–559 95: 13200–13205
67. Iolascon A, de Mattia D, Perrotta S, Carella M, Gasparini P, 83. Körner C, Knauer R, Stephani U, Marquardt T, Lehle L, von
Lambertenghi Deliliers G (1998) Genetic heterogeneity of Figura K (1999) Carbohydrate deficient glycoprotein
congenital dyserythropoietic anemia type II. Blood 92: 2593– syndrome type IV: deficiency of dolichyl-P-Man:Man(5)
2594 GlcNAc(2)-PP-dolichyl mannosyltransferase. EMBO J 18:
68. Iolascon A, Delaunay J, Wickramasinghe SN, Perrotta S, 6816–6822
Gigante M, Camaschella C (2001) Natural history of con- 84. Körner C, Lehle L, von Figura K (1998) Carbohydrate-
genital dyserythropoietic anemia type II. Blood 98: 1258–1260 deficient glycoprotein syndrome type 1: correction of the
69. Jaeken J, Stibler H (1989) A newly recognized inherited neu- glycosylation defect by deprivation of glucose or supplemen-
rological disease with carbohydrate deficient secretory glyco- tation of mannose. Glycoconj J 15: 499–505
proteins. In: L Wetterberg (ed) Genetics of neuropsychiatric 85. Kornfeld R, Kornfeld S (1985) Assembly of asparagine-linked
diseases, Wenner-Gren International Symposium Series, Vol oligosaccarides. Ann Rev Biochem 54: 631–664
5, Macmillian Press, London,pp 69–80 86. Kranz C, Denecke J, Lehrman MA, Ray S, Kienz P, Kreissel
70. Jaeken J, Carchon H (1993) The carbohydrate-deficient gly- G, Sagi D, Peter-Katalinic J, Freeze HH, Schmid T, Jac-
coprotein syndromes: an overview. J Inher Metab Dis 16: kowski-Dohrmann S, Harms E, Marquardt T (2001) A mu-
813–820 tation in the human MPDU1 gene causes congenital disorder
71. Jaeken J, Vanderschueren-Lodeweyckx M, Casaer P, Snoeck of glycosylation If (CDG-If). J Clin Invest 108: 1613–1619
L, Corbeel L, Eggermont E, Eeckels R (1980) Familial psy- 87. Kresse H, Rosthoj S, Quentin E, Hollmann J, Glossl J, Okada
chomotor retardation with markedly fluctuating serum prol- S, Tonnesen T (1987) Glycosaminoglycan-free small proteo-
actin, FSH and GH levels, partial TBG deficiency, increased glycan core protein is secreted by fibroblasts from a patient
378

with a syndrome resembling progeroid. Am J Hum Genet 41: lyzes the synthesis of heparan sulfate. Proc Natl Acad Sci U S
436–453 A 97: 668–673
88. Kristiannson B, Andersson M, Tonnby B, Hagberg B (1989) 105. Miyoshi E, Noda K, Yamaguchi Y, Inoue S, Ikeda Y, Wang
Disialotransferrin developmental deficiency syndrome. Arch W, Ko JH, Uozumi N, Li W, Taniguchi N (1999) The alpha1–
Dis Child 64: 71–76 6-fucosyltransferase gene and its biological significance. Bio-
89. Lefebvre T, Alonso C, Mahboub S, Dupire MJ, Zanetta JP, chim Biophys Acta 1473: 9–20
Caillet-Boudin ML, Michalski JC (1999) Effect of okadaic 106. Niehues R, Hasilik M, Alton G, Körner C, Schiebe-Sukumar
acid on O-linked N-acetylglucosamine levels in a neuroblas- M, Koch HG, Zimmer K-P, Wu R, Harms E, Reiter K, von
toma cell line. Biochim Biophys Acta 1472: 71–81 Figura K, Freeze HH, Harms HK, Marquardt T (1998)
90. Lin X, Wells D (1997) Isolation of the mouse cDNA homol- Carbohydrate-deficient glycoprotein syndrome type Ib.
ogous to the human EXT1 gene responsible for hereditary Phosphomannose isomerase deficiency and mannose therapy.
multiple exostoses. DNA Seq 7: 199–202 J Clin Invest 101: 1414–1420
91. Lind T, Tufaro F, McCormick C, Lindahl U, Lidholt K 107. Ohno K, Yuasa I, Akaboshi S, Itoh M, Yoshida K, Ehara H,
(1998) The putative tumor suppressors EXT1 and EXT2 are Ochiai Y, Takeshita K (1992) The carbohydrate deficient
glycosyltransferases required for the biosynthesis of heparan glycoprotein syndrome in three Japanese children. Brain Dev
sulfate. J Biol Chem 273: 26265–26268 14: 30–35
92. Lübke T, Marquardt T, von Figura K, Körner C (1999) A 108. Okajima T, Fukumoto S, Furukawa K, Urano T (1999)
new type of carbohydrate-deficient glycoprotein syndrome Molecular basis for the progeroid variant of Ehlers-Danlos
due to a decreased import of GDP-fucose into the Golgi. J Biol syndrome. Identification and characterization of two muta-
Chem 274: 25986–25989 tions in galactosyltransferase I gene. J Biol Chem 274: 28841–
93. Lübke T, Marquardt T, Etzioni A, Hartmann E, von Figura 28844
K, Körner C (2001) Complementation cloning identifies 109. Panneerselvam K, Freeze HH (1996) Mannose corrects
CDG-IIc (LAD II), a new type of congenital disorder of altered N-glycosylation in carbohydrate-deficient glycoprotein
glycosylation, as a GDP-fucose transporter deficiency. Nat syndrome fibroblasts. J Clin Invest 97: 1478–1487
Genet 28: 73–76 110. Parodi AJ (2000) Protein glucosylation and its role in protein
94. Ludecke HJ, Wagner MJ, Nardmann J, La Pillo B, Parrish folding. Annu Rev Biochem 69: 69–93
JE, Willems PJ, Haan EA, Frydman M, Hamers GJ, Wells 111. Peters V, Penzien JM, Reiter G, Körner C, Hackler R,
DE et al (1995) Molecular dissection of a contiguous gene Assmann B, Fang J, Schaefer JR, Hoffmann GF, Heidemann
syndrome: localization of the genes involved in the Langer- PH (2002) Congenital disorder of glycosylation IId (CDG-
Giedion syndrome. Hum Mol Genet 4: 31–36 IId) – a new entity: clinical presentation with Dandy-Walker
95. Lühn K, Marquardt T, Harms E, Vestweber D (2001) Dis- malformation and myopathy. Neuropediatrics 33: 27–32
continuation of fucose therapy in LAD II causes rapid loss of 112. Petersen MB, Brostrøm K, Stibler H, Skovby F (1993) Early
selectin ligands and rise of leukocyte counts. Blood 97: 330– manifestations of the carbohydrate-deficient glycoprotein
332 syndrome. J Pediatr 122: 66–70
96. Marquardt T, Hasilik M, Niehues R, Herting M, Muntau A, 113. Pirard M, Acouri Y, Collet J-F, Schollen E, Matthijs G, van
Holzbach U, Hanefeld F, Freeze H, Harms E (1997) Mannose Schaftingen E (1999) Kinetic properties and tissular distribu-
therapy in carbohydrate-deficient glycoprotein syndrome type tion of mammalian phosphomannomutase isoenzymes.
I—first results of the German multicenter study. Amino Acids Biochem J 339: 201–207
12: 389 114. Pirard M, Matthijs G, Heykants L, Schollen E, Grünewald S,
97. Marquardt T, Brune T, Lühn K, Zimmer KP, Körner C, Jaeken J, van Schaftingen E (1999) Effect of mutations found
Fabritz L, van der Werft N, Vormoor J, Freeze HH, Louwen in carbohydrate-deficient glycoprotein syndrome type IA on
F, Biermann B, Harms E, von Figura K, Vestweber D, Koch the activity of phosphomannomutase 2. FEBS Lett 452: 319–
HG (1999) A new patient with leukocyte adhesion deficiency 322
(LAD) II syndrome, a generalized defect in fucose metabo- 115. Quentin E, Gladen A, Roden L, Kresse H (1990) A genetic
lism. J Pediatr 134: 681–688 defect in the biosynthesis of dermatan sulfate proteoglycan:
98. Marquardt T, Lühn K, Srikrishna G, Freeze HH, Harms E, galactosyltransferase I deficiency in fibroblasts from a patient
Vestweber D (1999) Correction of leukocyte adhesion with a progeroid syndrome. Proc Natl Acad Sci U S A 87:
deficiency type II with oral fucose. Blood 94: 3976–3985 1342–1346
99. Matthijs G, Schollen E, Pardon E, Veiga-Da-Cunha M, 116. Raitta C, Lamminen M, Santavuori P, Leisti J (1978) Oph-
Jaeken J, Cassimani J-J, Van Schaftingen E (1997) Mutations thalmological findings in a new syndrome with muscle, eye
in PMM2, a phosphomannomutase gene on chromosome and brain involvement. Acta Ophthalmol (Copenh) 56: 465–
16p13, in carbohydrate-deficient glycoprotein type I syndrome 472
(Jaeken syndrome). Nat Genet 16: 88–92 117. Ramaekers VT, Stibler H, Kint J, Jaeken J (1991) A new
100. Matthijs G, Schollen E, van Schaftingen E, Cassiman J-J, variant of the carbohydrate deficient glycoproteins syndrome.
Jaeken J (1998) Lack of homozygotes for the most frequent J Inher Metab Dis 14: 385–388
disease allele in carbohydrate-deficient glycoprotein syndrome 118. Roth J, Wang Y, Eckhardt AE, Hill RL (1994) Subcellular
type 1A. Am J Hum Genet 62: 542–550 localization of the UDP-N-acetyl-D-galactosamine: polypep-
101. Mawby WJ, Tanner MJA, Anstee DJ, Clamp JR (1983) tide N-acetylgalactosaminyltransferase-mediated O-glycosy-
Incomplete glycosylation of erythrocyte membrane proteins in lation reaction in the submaxillary gland. Proc Natl Acad Sci
congenital dyserythropoietic anaemia type II (CDA II). Br U S A 91: 8935–8939
J Haematol 55: 357–368 119. Rush JS, Panneerselvam K, Waechter CJ, Freeze HH (2000)
102. Mayatepek E, Kohlmüller D (1998) Mannose supplementa- Mannose supplementation corrects GDP-mannose deficiency
tion in carbohydrate-deficient glycoprotein syndrome type I in cultured fibroblasts from some patients with congenital
and phosphomannomutase deficiency. Eur J Pediatr 157: 605– disorders of glycosylation (CDG). Glycobiology 10: 829–835
606 120. Scartezzini P, Forni GL, Baldi M, Izzo C, Sansone G (1982)
103. Mayatepek E, Schröder M, Kohlmüller D, Bieger WP, Nüt- Decreased glycosylation of band 3 and band 4.5 glycoproteins
zenadel W (1997) Continuous mannose infusion in carbohy- of erythrocyte membrane in congenital dyserythropoietic
drate-deficient glycoprotein syndrome type I. Acta Paediatr anaemia type II. Br J Haematol 51: 569–576
86: 1138–1140 121. Schenk B, Imbach T, Frank CG, Grubenmann CE, Raymond
104. McCormick C, Duncan G, Goutsos KT, Tufaro F (2000) The GV, Hurvitz H, Raas-Rotschild A, Luder AS, Jaeken J,
putative tumor suppressors EXT1 and EXT2 form a stable Berger EG, Matthijs G, Hennet T, Aebi M (2001) MPDU1
complex that accumulates in the Golgi apparatus and cata- mutations underlie a novel human congenital disorder of
379

glycosylation, designated type If. J Clin Invest 108: 1687– 136. Van Geet C, Jaeken J (1993) A unique pattern of coagulation
1695 abnormalities in carbohydrate-deficient glycoprotein syn-
122. Schollen E, Kjaergaard S, Legius E, Schwartz M, Matthijs G drome. Pediatr Res 33: 540–541
(2000) Lack of Hard-Weinberg equilibrium for the most 137. Van Schaftingen E, Jaeken J (1995) Phosphomannomutase
prevalent PMM2 mutation in CDG-Ia (congenital disorder of deficiency is a cause of carbohydrate-deficient glycoprotein
glycosylation type Ia). Eur J Hum Genet 8: 367–371 syndrome type I. FEBS Lett 377: 318–320
123. Silberstein S, Collins PG, Kelleher DJ, Rapiejko PJ, Gilmore 138. Wada Y, Nishikawa A, Okamoto N, Inui K, Tsukamoto H,
R (1995) The alpha subunit of the Saccharomyces cerevisiae Okada S, Taniguchi N (1992) Structure of serum transferrin in
oligosaccharyltransferase complex is essential for vegetative carbohydrate-deficient glycoprotein syndrome. Biochem Bio-
growth of yeast and is homologous to mammalian ribophorin phys Res Comm 189: 832–836
I. J Cell Biol 128: 525–536 139. Wada Y, Gu J, Okamoto N, Inui K (1994) Diagnosis of
124. Solomon L (1963) Hereditary multiple exostoses. J Bone carbohydrate-deficient glycoprotein syndrome by matrix-as-
J Surg 45: 292–304 sisted laser desorption time-of-flight mass spectrometry. 23:
125. Staudacher E, Altmann F, Wilson IB, Marz L (1999) Fucose 108–109
in N-glycans: from plant to man. Biochim Biophys Acta 1473: 140. Ware FE, Lehrman MA (1996) Expression cloning of a
216–236 novel suppressor of the Lec15 and Lec35 glycosylation mu-
126. Stibler H, Jaeken J (1990) Carbohydrate deficient serum tations of chinese hamster ovary cells. J Biol Chem 271:
transferrin in a new systemic hereditary syndrome. Arch Dis 13935–13938
Child 65: 107–111 141. Westphal V, Schottstädt C, Marquardt T, Freeze HH (2000)
127. Stibler H, Westerberg B, Hanefeld F, Hagberg B (1993) Analysis of multiple mutations in the hALG6 gene in a patient
Carbohydrate-deficient glycoprotein (CDG) syndrome—a with congenital disorder of glycosylation Ic. Mol Genet
new variant, type III. Neuropediatrics 24: 51–52 Metab 70: 219–223
128. Stibler H, Stephani U, Kutsch U (1995) Carbohydrate- 142. Wickramasinghe SN (1997) Dyserythropoiesis and congenital
deficient glycoprotein syndrome—a fourth subtype. Neuro- dyserythrpoietic anaemias. Br J Hematol 98: 785–797
pediatrics 26: 235–237 143. Wuyts W, Ramlakhan S, Van Hul W, Hecht JT, van den
129. Strøm EH, Strømme P, Westvik K, Pedersen SJ (1993) Renal Ouweland AM, Raskind WH, Hofstede FC, Reyniers E,
cysts in the carbohydrate-deficient glycoprotein syndrome. Wells DE, de Vries B et al (1995) Refinement of the multiple
Pediatr Nephrol 7: 253–255 exostoses locus (EXT2) to a 3-cM interval on chromosome 11.
130. Stromland K, Hagberg B, Kristiansson B (1990) Ocular Am J Hum Genet 57: 382–387
pathology in disialotransferrin developmental deficiency 144. Wuyts W, Van Hul W, Wauters J, Nemtsova M, Reyniers E,
syndrome. Ophthalmic Paediatr Genet 11: 309–313 Van Hul EV, De Boulle K, de Vries BB, Hendrickx J,
131. Sturla L, Puglielli L, Tonetti M, Berninsone P, Hirschberg CB, Herrygers I, Bossuyt P, Balemans W, Fransen E, Vits L,
de Flora A, Etzioni A (2001) Impairment of the Golgi GDP-L- Coucke P, Nowak NJ, Shows TB, Mallet L, van den Ouwe-
fucose transport and unresponsiveness to fucose replacement land AM, McGaughran J, Halley DJ, Willems PJ (1996) Po-
therapy in LAD II patients. Pediatr Res 49: 537–542 sitional cloning of a gene involved in hereditary multiple
132. Takahashi S, Sasaki T, Manya H, Chiba Y, Yoshida A, exostoses. Hum Mol Genet 5: 1547–1557
Mizuno M, Ishida H, Ito F, Inazu T, Kotani N, Takasaki S, 145. Yamashita K, Ideo H, Ohkura T, Fukushima K, Yuasa I,
Takeuchi M, Endo T (2001) A new beta-1,2-N-acetylglucos- Ohno K, Takeshita K (1993) Sugar chains of serum trans-
aminyltransferase that may play a role in the biosynthesis of ferrin from patients with carbohydrate deficient glycoprotein
mammalian O-mannosyl glycans. Glycobiology 11: 37–45 syndrome. J Biol Chem 268: 5783–5789
133. Tan J, Dunn J, Jaeken J, Schachter H (1996) Mutations in the 146. Yamashita K, Takashi O, Ideo H, Ohno K, Kanai M (1994)
MGAT2 gene controlling complex N-glycan synthesis cause Electrospray ionization-mass spectrometric analysis of serum
carbohydrate-deficient glycoprotein syndrome type II, an transferrin isoforms in patients with carbohydrate-deficient
autosomal recessive disease with defective brain development. glycoproteins syndrome. J Biochem 114: 766–769
Am J Human Genet 59: 810–817 147. Yan Q, Lennarz WJ (1999) Oligosaccharyltransferase: a
134. Thiel C, Schwarz M, Hasilik M, Grieben U, Hanefeld F, complex multisubunit enzyme of the endoplasmic reticulum.
Lehle L, Von Figura K, Koerner C (2002) Deficiency of Biochem Biophys Res Commun 266: 684–689
dolichyl-P-Man:Man7GlcNAc2-PP-dolichyl mannosyltrans- 148. Yoshida A, Kobayashi K, Manya H, Taniguchi K, Kano H,
ferase causes congenital disorder of glycosylation-Ig. Biochem Mizuno M, Inazu T, Mitsuhashi H, Takahashi S, Takeuchi
J 367: 195–201 M, Herrmann R, Straub V, Talim B, Voit T, Topaloglu H,
135. Van den Steen P, Rudd PM, Dwek RA, Opdenakker G (1998) Toda T, Endo T (2001) Muscular dystrophy and neuronal
Concepts and principles of O-linked glycosylation. Crit Rev migration disorder caused by mutations in a glycosyltrans-
Biochem Mol Biol 33: 151–208 ferase, POMGnT1. Dev Cell 1: 717–724

You might also like