ArticlePDF Available

Congenital disorder of glycosylphosphatidylinositol (GPI)-anchor biosynthesis-The phenotype of two patients with novel mutations in the PIGN and PGAP2 genes

Authors:

Abstract and Figures

Background: Glycosylphosphatidylinositol (GPI)-anchor deficiencies are a new subclass of congenital disorders of glycosylation. About 26 genes are involved in the GPI-anchor biosynthesis and remodeling pathway, of which mutations in thirteen have been reported to date as causative of a diverse spectrum of intellectual disabilities. Since the clinical phenotype of these disorders varies and the number of described individuals is limited, we present new patients with inherited GPI-anchor deficiency (IGD) caused by mutations in the PGAP2 and PIGN genes. Patients and methods: The first girl presented with profound psychomotor retardation, low birth parameters, and chest deformities already existing in neonatal period. The disease course was slowly progressive with severe hypotonia, chronic fever, and respiration insufficiency at the age of 6. The second girl showed profound psychomotor retardation, marked hypotonia, and high birth weight (97 centile). Dysmorphy was mild or absent in both girls. Whole exome sequencing revealed novel variants in the genes PGAP2 (c.2T>G and c.221G>A) and PIGN (c.790G>A and c.932T>G). Impaired GPI binding were was subsequently uncovered, although the hyperactivity of alkaline phosphatase (a GPI-anchored protein) occurred only in first case. Conclusions: Based on our results we can conclude that: 1. GPI-anchor biosynthesis disorders may represent a relatively frequent and overlooked metabolic defect; 2. The utility of GPI binding assessment as a screening test for this group of rare diseases requires further studies.
Content may be subject to copyright.
Case Study
Congenital disorder of
glycosylphosphatidylinositol (GPI)-anchor
biosynthesisdThe phenotype of two patients with
novel mutations in the PIGN and PGAP2 genes
Aleksandra Jezela-Stanek
a,*,i
,El
_
zbieta Ciara
a,i
,
Dorota Piekutowska-Abramczuk
a,i
, Joanna Trubicka
a
,
El_
zbieta Jurkiewicz
b
, Dariusz Rokicki
c
, Hanna Mierzewska
d
,
Justyna Spychalska
e
, Małgorzata Uhrynowska
e
,
Marta Szwarc-Bronikowska
c
, Piotr Buda
c
, Abdul Rahim Said
f
,
Ewa Jamroz
g
, Małgorzata Rydzanicz
h
, Rafał Płoski
h
,
Małgorzata Krajewska-Walasek
a
, Ewa Pronicka
a,c
a
Department of Medical Genetics, The Children's Memorial Health Institute, Warsaw, Poland
b
Department of Diagnostic Imaging, The Children's Memorial Health Institute, Warsaw, Poland
c
Department of Pediatrics, Nutrition and Metabolic Diseases, The Children's Memorial Health Institute, Warsaw,
Poland
d
Department of Child and Adolescent Neurology, Institute of Mother and Child, Warsaw, Poland
e
Department of Immunology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
f
Children's Home Hospice, Opole, Poland
g
Department of Pediatrics and Developmental Age Neurology, Medical University of Silesia, Katowice, Poland
h
Department of Medical Genetics, Warsaw Medical University, Warsaw, Poland
article info
Article history:
Received 8 October 2015
Received in revised form
10 January 2016
Accepted 11 January 2016
Keywords:
GPI-anchor deficiency
PGAP2 gene
abstract
Background: Glycosylphosphatidylinositol (GPI)-anchor deficiencies are a new subclass of
congenital disorders of glycosylation. About 26 genes are involved in the GPI-anchor
biosynthesis and remodeling pathway, of which mutations in thirteen have been reported
to date as causative of a diverse spectrum of intellectual disabilities. Since the clinical
phenotype of these disorders varies and the number of described individuals is limited, we
present new patients with inherited GPI-anchor deficiency (IGD) caused by mutations in
the PGAP2 and PIGN genes.
Patients and methods: The first girl presented with profound psychomotor retardation, low
birth parameters, and chest deformities already existing in neonatal period. The disease
*Corresponding author. Department of Medical Genetics, The Children's Memorial Health Institute, Aleja Dzieci Polskich 20, 04-730
Warsaw, Poland. Tel.: þ48 228157452; fax: þ48 22 8157457.
E-mail address: jezela@gmail.com (A. Jezela-Stanek).
i
Equal contribution.
Official Journal of the European Paediatric Neurology Society
european journal of paediatric neurology 20 (2016) 462e473
http://dx.doi.org/10.1016/j.ejpn.2016.01.007
1090-3798/©2016 European Paediatric Neurology Society. Published by Elsevier Ltd. All rights reserved.
PIGN gene
Congenital disorders of
glycosylation
Flow cytometry
course was slowly progressive with severe hypotonia, chronic fever, and respiration
insufficiency at the age of 6. The second girl showed profound psychomotor retardation,
marked hypotonia, and high birth weight (97 centile). Dysmorphy was mild or absent in
both girls. Whole exome sequencing revealed novel variants in the genes PGAP2 (c.2T>G
and c.221G>A) and PIGN (c.790G>A and c.932T>G). Impaired GPI binding were was subse-
quently uncovered, although the hyperactivity of alkaline phosphatase (a GPI-anchored
protein) occurred only in first case.
Conclusions: Based on our results we can conclude that: 1. GPI-anchor biosynthesis disorders
may represent a relatively frequent and overlooked metabolic defect; 2. The utility of GPI
binding assessment as a screening test for this group of rare diseases requires further studies.
©2016 European Paediatric Neurology Society. Published by Elsevier Ltd. All rights
reserved.
1. Introduction
Glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-
APs) are glycolipids composed of phosphatidylinositol,
carbohydrate residues, glucosamine, and mannose. About
150 proteins with various functions are GPI anchored,
including membrane-associated enzymes, adhesion mole-
cules, activation antigens, differentiation markers, immu-
nologically important proteins, including complement
regulatory proteins, and other miscellaneous glycoproteins.
The exact role of GPI-anchored proteins remains unclear.
However, while they function as hydrolytic enzymes,
adhesion molecules, receptors, protease inhibitors, and
complement regulatory proteins, they share similar char-
acteristics based on their common glycolipid membrane
anchors, and can act as receptors in cell adhesion, differ-
entiation, and hostepathogen interactions
28,31,38
Recent
studies showed the importance of GPI-APs for normal
neuronal and embryonic development.
14
GPI-AP deficiencies are a recently emerging group of
diseases within congenital disorders of glycosylation. They
are caused by mutations in two different classes of genes:
PIG (Phosphatidyl Inositol Glycan) involved in the biosyn-
thesis and transfer of GPI, and PGAP (Post GPI Attachment to
Proteins) involved in structural remodeling of GPI after its
attachment to proteins. About 26 PIG and PGAP genes are
involved in the GPI anchor biosynthesis and remodeling
pathway.
18
To date, inherited GPI deficiencies caused by
mutations in at least 13 PIG and PGAP genes have been re-
ported (Table 1). Only PIGA is X-linked recessive; all of the
others are autosomal recessive. Clinical symptoms of in-
dividuals with inherited GPI-anchor deficiencies (IGDs) are
within a wide range, leading to difficulties in their pheno-
typic recognition.
Herein we present the clinical, biochemical and molecular
characteristics of new cases with a congenital disorder of GPI
anchor biosynthesis and remodeling, in whom exome
sequencing revealed mutations in the genes PGAP2 and PIGN.
In comparison with the previously described PGAP2 and PIGN
affected individuals, our patients show the more severe and
the mildest phenotypes, respectively.
2. Case presentation
2.1. Patient 1
The girl, the only child of healthy unrelated parents, was born
at 39 weeks gestation following an uncomplicated pregnancy
with a birth weight of 2940 g (1.57 SD), head circumference of
32 cm (3.01 SD), and 8 points on the Apgar scale. Her family
history was unremarkable. She was born with a flat occiput
and pectus excavatum. Hyperbilirubinemia was noted during
the neonatal period (max. 18 mg/dl), but intrauterine infection
was excluded. In the routine ultrasound examination of the
brain, II degree intraventricular hemorrhage was found,
otherwise the brain structure was normal. At 3 months of life,
psychomotor retardation and generalized weakness were
observed.
Significantly elevated alkaline phosphatase activity (AP)
(1700 U/l) and low normal calcium concentration (2.3 mmol/l)
led to suspicion of rickets, which was later excluded based on
normal simultaneously measured concentrations of phos-
phates (1.85 mmol/l), parathyroid hormone, and 25-OH
vitamin D.
At 4 months of life West-like epileptic attacks with
typical hypsarrhythmia and suppression-burst EEG pattern
were noted, followed by progressive neurological deterio-
ration, hyporeflexia, and hypotonia. Moreover, from 8
months of life persistent, refractory to treatment hyper-
thermia was observed. No acquired immunodeficiency
disorder was found, as TORCH, HIV, and Mycobacterium
tuberculosis infections were excluded. Due to progressive
neurological deterioration and recurrent aspiration pneu-
monia, the patient underwent gastrostomy tube place-
ment. Nerve conduction study showed decreased neural
conduction, electromyography (EMG) examination was
normal. Further EMG examination revealed features of
european journal of paediatric neurology 20 (2016) 462e473 463
Table 1 ePhenotypes and mutations in thirteen GPI-APs genes.
Gene defect Alkaline phosphatase Disease Inheritance Clinical and biochemical
features
Reference
PIGA (MIM 311770) Not high/mildly elevated MCAHS2 (multiple congenital
anomalies-hypotonia-seizures
syndrome 2)
XLR Seizures, brain malformations,
cleft palate, hypotonia, neonatal
seizures, contractures
17
PIGL (MIM 605947) Not high CHIME syndrome (Zunich
neuroectodermal syndrome)
AR Colobomas, Heart disease,
Ichthyosiform dermatosis,
Mental retardation, and Ear
anomalies (CHIME)
34
PIGM (MIM 610273) Not high PIGMeCDG (PIGMecongenital
disorder of glycosylation)
AR Portal venous thrombosis,
absence seizures
1
PIGV (MIM 610274) High HPMRS (hyperphosphatasia with
mental retardation syndrome,
Mabry syndrome)
AR Brachytelephalangy,
hyperphosphatasia, intellectual
disability, seizures, facial
dysmorphism
13
PIGN (MIM 606097) Not high MCAHS (multiple congenital
anomalies-hypotonia-seizures
syndrome)
AR Hypotonia, seizures, multiple
congenital anomalities,
progressive cerebellar atrophy
26
PIGO (MIM 614730) High HPMRS (Mabry syndrome) AR Brachytelephalangy, intellectual
disability, seizures
19
PGAP2 (MIM 614207) High PGAP2eCDG (PGAP2econgenital
disorders of glycosylation)
AR Non-syndromic intellectual
disability, seizures, distinctive
facial gestalt
10
PGAP3 (MIM 615716) High PGAP3eCDG (PGAP3econgenital
disorders of glycosylation)
AR Intellectual disability, seizures,
distinctive facial gestalt
14
PIGT (MIM 615395) Not high/low MCAHS3 (multiple congenital
anomalies-hypotonia-seizures
syndrome 3)
AR Intellectual disability, hypotonia,
seizures, and dysmorphic facial
features in combination with
abnormal skeletal, endocrine,
and ophthalmologic findings,
hypophosphatasia
22
PIGW (MIM 616025) High HPMRS5 (Hyperphosphatasia
with mental retardation
syndrome 5)
AR Delayed development, West
syndrome, dysmorphic facial
features
5
PIGQ (MIM 614749) High HPMRS2 (Hyperphosphatasia
with mental retardation
syndrome 2)
AR Mental retardation, hypotonia,
generalized seizures, nail
hypoplasia, dysmorphic
features, congenital heart defect
25
PGAP1 (MIM 615802) No data MRT42 (Mental retardation,
autosomal recessive 42)
AR Neonatal hypotonia, severely
delayed psychomotor
development, epilepsy with
stereotypic movements,
dysmorphic features
32
PIGY (MIM 610662) Mildly elevated Different phenotypes AR Developmental delay, seizures,
cataracts, dysmorphy,
microcephaly
16
european journal of paediatric neurology 20 (2016) 462e473464
neurogenic damage; sensory-evoked potentials showed
features of demyelination. Lysosomal storage disorders,
peroxisomal diseases, and organic acidurias were excluded
in further investigations. Gas chromatographyemass
spectrometry urine test showed ketosis with marked
C6eC10 dicarboxylic aciduria. Plasma lactate concentra-
tions were normal or high borderline. The transferrin
glycosylation pattern was normal.
Muscle biopsy was done at the age of 26 months. Histo-
chemistry revealed a generalized decrease in cytochrome
oxidase activity, lipid accumulation, and unspecific changes
of muscle fiber differentiation.
At the age of 6 years, respiratory insufficiency with carbon
dioxide accumulation gradually developed. Tracheostomy
had to be performed and assisted breathing was begun under
a national program of mechanical ventilation in the home
(Fig. 1).
The girl has remained in recumbent position from birth.
She has never sat or begun to speak. Presently at the age of 13,
she is still on artificial ventilation, fed by gastrostomy,
continuously treated for drug-resistant epilepsy. According to
the mother's observations, introduction of a ketogenic diet
positively influenced her quality of life, decreasing anxiety
and thermoregulation dysfunction. The occurrence of hyper-
phosphatasemia was never taken into account in differential
diagnostics before finding PGAP2 mutations. Reassessment of
her medical documentation showed that AP activity was
continuously elevated (1496e2780 U/l, ref. 100e550 U/l, 11
measurements on follow-up).
2.2. Patient 2
This proband is the first child of healthy and non-
consanguineous parents with an unremarkable family his-
tory. The pregnancy was complicated by recurrent urinary
tract infections and bacterial pharyngitis at 7 weeks gesta-
tion. The girl was born at term with a birth weight of 4300 g
(þ0.98 SD), head circumference of 34 cm (1.41 SD) , and 10
points on the Apgar scale. Pericranial hematoma was noted.
At the age of 2 months she was hospitalized because of
myoclonic jerks of limbs and suspicion of epilepsy. Brain MRI
revealed slightly widened extra axial spaces around the
frontal lobe; the EEG pattern at that time was normal. The
proband was then diagnosed for gastroesophageal reflux
with a negative result.
Neurological examination during subsequent hospitaliza-
tion at the age of 5 months revealed: head circumference of
42 cm (75e90 centile), generalized hypotonia, vertical
nystagmus, tongue tremor, multiple dyskinesias, and pres-
ence of tendon and postural reflexes. Laboratory tests results
were within normal ranges, except for plasma lactate con-
centrations (max 3.7 mmol/l; ref. <2.0 mmol/l). The EEG
pattern revealed generalized paroxysmal bioelectric activity,
consistent with video-EEG where correlation of myoclonic
seizures with bioelectric abnormalities was found. Subse-
quent MRI showed cerebellar atrophy, particularly significant
cerebellar vermis atrophy, and enlarged lateral ventricles,
more pronounced on the left side. Brain MRI scans are pre-
sented in Figs. 2 and 3, and Table 2.
Although no specific facial dysmorphy was seen (Fig. 4),
taking into account the clinical course of the patient, Angel-
man and Rett syndrome were considered in differential di-
agnoses, but not confirmed by genetic tests.
The patient has undergone extensive diagnostics,
including searching for inborn errors of metabolism. The
performed genetic tests allowed exclusion of spinal muscular
atrophy (no deletion in the SMN1 gene), chromosomal aber-
rations (normal result of 135k array CGH), as well as mito-
chondrial DNA pathology (negative screening for common
mutations in mitochondrial DNA). Krabbe disease, suspected
due to low galactosylceramidase activity (1.1 and 2.7 mmol/
mg protein/18 h [ref. 6.7 ±2.7]), was also excluded by the lack
of the most frequent mutation in the GALC gene.
Both patients were included into a whole exome
sequencing (WES) study based on suspicion of mitochondrial
pathology (MD). The probability of MD was assessed according
to the twelve-step Nijmegen scale
30
and was found to be 3
points for Patient 1 and 4 points for Patient 2, which corre-
sponds to a moderate level of probability (mitochondrial
disease possible).
Fig. 1 eFacial phenotype of Patient 1 with PGAP2 mutations (at the age of 13 y and in early infancy).
european journal of paediatric neurology 20 (2016) 462e473 465
This investigation conforms to the principles outlined in
the Declaration of Helsinki, and the study protocol was
approved by the Bioethics Commission of The Children's Me-
morial Health Institute. Written informed consent was ob-
tained from the parents of both affected girls.
2.3. Methods and results
2.3.1. Whole exome sequencing
DNA was extracted from the peripheral blood and used for
massively parallel exome sequencing. WES Library
Fig. 2 eMRI examination in Patient 2 (with PING mutations). eat the age of 9 weeks, upper row. a., b. sagittal and c. coronal
T2-weighted images show cerebellar hemispheres and vermis in normal limits, d. axial T2-weighted image shows slightly
widened extra axial spaces around the frontal lobes (more visible on the left side), and anterior part of the interhemispheric
fissure. Minimal asymmetry of the lateral ventricles eL>R. efollow-up at the age of 27 months, bottom row. e, f. sagittal T2-
weighted and g. coronal T1-weighted images show significant atrophy of the vermis. Mild atrophy of the cerebellar
hemispheres with enlarged and deep cerebellar fissures. Widening of the fourth ventricle and cisterna magna. Thin
superior cerebellar peduncles. Thin corpus callosum. h. axial T2-weighted image shows progressive frontal cortical atrophy
with enlarged fissures in the frontal lobes and widened extra axial spaces around the frontal lobes. Asymmetric widening of
the lateral ventricles eL>R.
Fig. 3 eAxial diffusion-weighted MR images of Patient 2 (PING mutations) at the age of 5 months show bilateral symmetrical
hyperintensity of the central tegmental tract.
european journal of paediatric neurology 20 (2016) 462e473466
preparation was performed using Nextera Rapid Capture
Exome kits (Illumina). The samples were run on ¼lane each
on HiSeq 1500 using 2 75 bp paired-end reads. Bioinfor-
matics analysis was performed as previously described.
36
Briefly, after initial processing by CASAVA, the generated
reads were aligned to the hg19 reference genome with the
Burrows-Wheeler Alignment Tool
23
and further processed by
the Genome Analysis Toolkit.
27
Base quality score recalibra-
tion, indel realignment, duplicate removal, and SNP/INDEL
calling were done as described.
8
The detected variants were
annotated using Annovar
40
and converted to Microsoft Access
format date for final manual analyses. Alignments were
viewed with the Integrative Genomics Viewer.
37
2.3.2. PGAP2 gene molecular results
The sequencing run for Patient 1's sample achieved 78,755,538
reads, the 10-fold target coverage was 92.2%, and the 20-fold
coverage was 78.2%, yielding a total of 280,442 revealed variants.
Two novel substitutions in the PGAP2 gene
(NM_001256240.1) were identified in this proband, including
c.2T>G (p.?) in exon 2 and c.221G>A (p.Arg74His) in exon 3
resulting in a compound heterozygous state (Fig. 5A, Table 3).
The c.2T>G molecular variant affects the initiating methio-
nine and results in start loss and possible activation of a new
potential translation initiation site or no protein translation.
Additionally, it was predicted to be deleterious by five in silico
predictive software packages, i.e., CADD, PolyPhen-2, LRT,
MutationTaster, and SIFT. The c.221G>A variant is a missense
change from arginine to histidine at residue 74 of the protein,
predicted as deleterious by six algorithms (CADD, PolyPhen-2,
MutationAssessor, LRT, MutationTaster, and SIFT). The
c.221G>A variant was reported in the ExAC database (http://
exac.broadinstitute.org) with minor allele frequency (MAF)
0.00001626, and in ESP 6500 (http://evs.gs.washington.edu/
EVS) with MAF 0.00008. Both variants were validated by
Sanger sequencing confirming their parental inheritance, the
c.2T>G variant was present in the mother and the c.221G>A
variant was inherited from the father.
2.3.3. PIGN gene molecular results
The sequencing run for Patient 2's sample achieved 55,799,742
reads with a total of 110,665 detected variants. Above 81% and
Table 2 eMRI changes observed in Patient 2 with mutation in PIGN gene.
Proband's age Brain MRI results
9 weeks Slightly widened of the extra axial spaces around frontal lobes,
Slightly widened of the both lateral fissures and anterior part of the
interhemispheric fissure,
Myelination appropriate for age,
Cerebellar hemispheres and vermis within normal limits,
Corpus callosum without pathology,
Minimal asymmetry of the slight widening of the lateral ventricle eL>R,
Symmetrical abnormal signal of the central tegmental tracts seen on T2-
weighted and diffusion weighted images in the midbrain and pons.
5 months Slightly progression:
Symmetrical abnormal signal of the central tegmental tracts seen on T2-
weighted and diffusion weighted images ein the midbrain, pons and
medulla oblongata (olivary nuclei).
Widened of the extra axial spaces around frontal lobes.
Widened of the both lateral fissures and anterior part of the
interhemispheric fissure.
Cerebellar hemispheres and vermis within normal limits
Myelination appropriate for age
7 months Without differences
27 months Progression:
Significant atrophy of the vermis
Mild atrophy of the cerebellar hemispheres
Slightly enlargement of the ventricular system, asymmetry of the lateral
ventricles L>R), IV ventricle is also enlarged
Widened of the extra axial spaces around frontal lobes, both lateral
fissures and anterior part of the interhemispheric fissure as in the
previous examination.
Myelination appropriate for age
Abnormal signal of the central tegmental tracts is very week
Fig. 4 eFacial phenotype of Patient 2 with PIGN mutations
(at the age of 3 4/12 y).
european journal of paediatric neurology 20 (2016) 462e473 467
91% of the target region was covered min. 20 and 10 times,
respectively.
Two nucleotide substitutions c.790G>A (p.Gly264Arg) and
c.932T>G (p.Leu311Trp) in the PIGN gene (NM_176787.4) were
identified by WES and confirmed by bidirectional Sanger
sequencing (Fig. 5B, Table 3). Parental studies determined that
the c.790G>A variant was inherited from the unaffected father
and the c.932T>G variant was inherited from the unaffected
Fig. 5 eIntegrative Genomics Viewer view of heterozygous mutations found in the PGAP2 gene (A) and the PIGN gene (B) by
whole-exome sequencing. Nomenclature of mutations following the guidelines of the Human Genome Variation Society
using NM_001256240.1 as a reference cDNA sequence for PGAP2 and NM_176787.4 for PIGN.
european journal of paediatric neurology 20 (2016) 462e473468
mother, consistent with a trans allelic pattern of inheritance
and compound heterozygosity for these changes. Both of the
variants are apparently novel missense mutations that affect
highly or mostly conserved amino acids and are located
within the phosphodiesterase functional domain of the PIGN
gene. In addition, they were predicted by in silico analysis
(Table 3) as probably damaging GPI-anchor biosynthesis,
which suggests that they are disease-causing genetic factors.
The variants were not found in the ClinVar, dbSNP, LOVD, or
PubMed database. Only the c.790G>A substitution had been
observed with a frequency of 0.00007458 in the ExAC database,
indicating that these changes are rare.
2.3.4. GPI functional impairment
The surface expression of GPI molecules and various GPI-APs
was tested in EDTA anticoagulated peripheral blood samples
collected from the patients and as a control esix healthy
blood donors (reference level). Samples were transported and
stored at þ4C. Flow cytometric tests were performed within
48 h following blood collection. The expression of GPI anchors
was determined by staining granulocytes and monocytes with
fluorescently-labeled aerolysin (FLAER Alexa 488, Cedarlane).
For determination the expression of GPI-APs cells were
stained with fluorescently conjugated monoclonal antibodies:
granulocytes with CD24 PE (clone ALB9, Beckman Coulter) and
CD66b FITC (clone G10F5, BD Pharmingen), monocytes with
CD14 APC (MФP9, Becton Dickinson) and CD157 PE (clone
SY11B5, eBioscience), and erythrocytes with CD59 PE (MEM-43,
Invitrogen).
2
Acquisition and analysis stained blood cells were per-
formed on flow cytometer FACSCalibur (BD Biosciences, San
Jose) with CellQuest Pro version 5.2.1 software (Becton Dick-
inson). GPI and GPI-APs expression was quantified by the
determination of median fluorescence intensity (Md) labeled
cells. Reference level was calculated as an average of Md
fluorescence stained cells from 6 donors.
Results of flow cytometry (FACS) analysis for Patient 1 and
for Patient 2 represent Figs. 6 and 7, respectively. The surface
level GPI anchors examined by FLAER binding on granulocytes
and monocytes from the Patient 1 was similar as mean value
on the reference cells (Fig. 6A and C). The cell-surface GPI-APs
tested by monoclonal antibodies: CD24 on granulocytes,
CD157 on monocytes and CD59 on erythrocytes showed
normal expression (Fig. 6B, E and F). However the expression
CD14 GPI-AP on monocytes from Patient 1 demonstrated sig-
nificant reduction (52%) compared with the reference mono-
cytes (Fig. 6D).
The surface expression of GPI anchors tested by FLAER, and
the expression of GPI-AP CD24 and CD66b tested by mono-
clonal antibodies was significantly decreased on the Patient 2
granulocytes compared with the reference cells (34.21 and
31% respectively) (Fig. 7A, B and C). FLAER binding to
Table 3 eDetails of molecular variants identified in PIGN and PGAP2 genes.
PIGN PGAP2
Mutant allele 1 Mutant allele 2 Mutant allele 1 Mutant allele 2
Chromosome 18 18 11 11
RefSeq cDNA NM_176787.4 NM_176787.4 NM_001256240.1 NM_001256240.1
Nucleotide change c.790G>A c.932T>G c.2T>G c.221G>A
Exon 9 11 2 3
IGV reads 41/81 41/111 6/10 16/29
RefSeq protein NP_065905.2 NP_065905.2 NP_001243169.1 NP_001243169.1
Amino acid change p.Gly264Arg p.Leu311Trp p.? p.Arg74His
Mutation
Type Missense Missense Missense Missense
Stage Heterozygous Heterozygous Heterozygous Heterozygous
Status Novel Novel Novel Novel
Parental origin Paternal Maternal Maternal Paternal
Pathogenicity prediction
CADD
a
28 22 17 24
Polyphen2 Probably damaging Probably damaging Probably damaging Probably damaging
Mutationassessor High (functional) Moderate (functional) nd Moderate (functional)
LRT Deleterious Deleterious Deleterious Deleterious
Mutationtaster Disease causing Disease causing Disease causing Disease causing
MAF data
b
ExAC 65000 0.00007458 0 0 0.00001626
ESP 6500 0 0 0 0
EUR 1000 0 0 0 0
POL 300 0 0 0 0
Dept of WES 67 94 10 27
a
Higher scores are more deleterious.
b
MAF eminor allele frequency; ExAC eexome aggregation consortium; EUR 1000 e1000 genomes project; ESP 6500 eexome sequencing
project; POL eproject of 300 exomes from Polish individuals with unrelated diseases.
european journal of paediatric neurology 20 (2016) 462e473 469
monocytes was not significantly changed (85% of ref.) (Fig. 7D),
but the expression of CD14 protein on these cells tested by
monoclonal antibodies was affected (42% of ref.) (Fig. 7E). The
expression GPI-AP CD59 on erythrocytes was also reduced
(76% of ref.) (Fig. 7F).
3. Discussion
In two sporadic patients presenting with a neurogenetic dis-
order from birth, whole exome sequencing performed after
long-term observation (at 3 and 12 years of age) revealed
compound heterozygous pathogenic variants of the PIGN and
PGAP2 genes, both involved in the biosynthesis of
glycosylphosphatidylinositol-anchored proteins. In addition
to several similarities to previously reported cases, our pa-
tients differ significantly, broadening the scope of knowledge
on inherited GPI deficiency.
The PGAP2 gene identified in 2013 as the cause of hyper-
phosphatasia, mental retardation syndrome-3(HPMRS3;
OMIM 614080) has been found in nine individuals from four
families to date.
10,20
The gene is localized on chromosome
11p15.4 and encodes a Golgi/ER-resident membrane protein
that is involved in the final step of lyso-phosphatidylinositol
(lyso-PI) remodeling to phosphatidylinositol (PI) containing
saturated fatty acids. Deficiency of this protein results in
transport to the cell surface of lyso-GPI-APs that are more
prone to cleavage by phospholipase D.
24,39
Similarly as in
other individuals with this defect, continuous (although
overlooked) increases in serum alkaline phosphatase (AP)
activity were demonstrated from infancy in Patient 1 with a
PGAP2 mutation. However, compared with the others, our
patient presented the most severe disease course with
untreatable epilepsy, dependence on artificial ventilation, and
persistent pyrexia. It cannot be excluded that at least persis-
tent pyrexia can be due to a latent infection, e.g.,
osteomyelitis.
9
The PIGN gene was reported in 2011
26
as related to multiple
congenital anomalies-hypotonia-seizures syndrome 100
(MCAHS1; OMIM 614080). The PIGN (phosphatidylinositol
glycan anchor biosynthesis, class N) gene maps to chromosome
band 18q21.33 and encodes glycosylphosphatidylinositol etha-
nolamine phosphate transferase 1, a protein expressed in the
endoplasmic reticulum that is involved in biosynthesis and
transfer of phosphoethanolamine (EtNP) to the first mannose of
the GPI anchor.
26
To date 11 cases from four families have been
reported
3,6,26
and most of them presented with various severe
congenital malformations that led to death. In contrast, our
Fig. 6 ePatient 1 ethe surface expression of GPI anchors and various GPI-anchored proteins tested by flow cytometry:
binding FLAER to GPI structures itself on granulocytes (A) and monocytes (C), binding monoclonal antibodies to CD24 (B), on
granulocytes, CD14 (D) and CD157 on monocytes (E), CD59 on erythrocytes (F). Black line epatient, gray line eblood donor,
gray dotted line enegative control. Md emedian of fluorescence intensity.
european journal of paediatric neurology 20 (2016) 462e473470
Patient 2 with PIGN mutations did not present any malforma-
tions and she demonstrated the mildest disease history as
compared with the reported cases. The course of disease re-
sembles to some extent the reported Japanese siblings,
35
as
they all show severe delay in psychomotor development, hy-
potonia, vertical nystagmus, and seizures. High birth weight
(95 centile) and head circumference (90 centile) found in
several patients with PIGN mutations
26
were also seen in our
Patient 2.
Occurrence of dysmorphic features and even a distinctive
facial gestalt was suggested in some descriptions of patients
with IGD. Abnormal facial features were noted in all eleven
patients with PIGN mutations
3,6,26
and were also considered in
those with PGAP2 mutations.
10,20
This seems not to be the case
in both of our patients, as a thorough re-assessment their
photographs over the years did not document a distinctive
facial gestalt. The facial features of Patient 1 had changed with
age; they were very subtle in childhood and did not have a
specific and recognizable dysmorphic pattern. Hansen et al.
came to a similar conclusion.
10
However, it may be doubtful
whether the facial phenotype of our Patient 1 is only sec-
ondary to her neurologic condition. Longer observations of
subsequent patients with IGD are necessary to arrive at firm
conclusions.
The various congenital malformations described
frequently in IGD are evidence of prenatal onset of patholog-
ical processes at the level of organ differentiation. It is not
clear, however, whether these processes, including brain
damage, cease after birth. The brain MRI of Patient 2 revealed
progressive cerebellar atrophy over time. A similar observa-
tion was reported by Ohba et al.
35
; but more data is needed for
final conclusions.
Although IGD should be classified as an inborn metabolic
disease(IMD),
29
an easy and reliable metabolic test that would
improve (accelerate) the differential diagnosis in suspected pa-
tients is lacking. This was a cause of diagnostic delay in all
known cases before next-generation sequencing became avail-
able. Alkaline phosphatase activity measurement is useful for
recognition of some IGDs (Table 1), but is highly unspecific.
An attempt to measure GPI-AP levels on the cell membrane in
available tissues (blood cells, fibroblasts) by FACS shown prom-
ising results.
14,26,35
The method is used routinely in hematology
for diagnosis of paroxysmal nocturnal hemoglobinuria, a defect
caused by somatic and germline mutations in the PIGA gene.
2
In Patient 1, demonstrating constant elevation of AP ac-
tivity, the FLAER binding on granulocytes and monocytes e
which characterize the GPI expression ewas normal. The
expression of GPI-APs: CD24 on granulocytes, CD157 on
Fig. 7 ePatient 2 ethe surface expression of GPI anchors and various GPI-anchored proteins tested by flow cytometry:
binding FLAER to GPI structures itself on granulocytes (A) and monocytes (D), binding monoclonal antibodies to CD24 (B) and
CD66b (C) on granulocytes, CD14 on monocytes (E), CD59 on erythrocytes (F). Black line epatient, gray line eblood donor,
gray dotted line enegative control. Md emedian of fluorescence intensity.
european journal of paediatric neurology 20 (2016) 462e473 471
monocytes and CD59 on erythrocytes was similar as that in
healthy blood donors. By comparison, analysis the expression
GPI-APs decay accelerating factor (DAF, CD55) and CD59 on
lymphoblastoid cell lines from patients with PGAP2 mutations
also showed no significant difference compared to healthy
donors.
10
In contrast, in vitro functional analysis performed in
PGAP2-deficient Chinese hamster ovary cell lines showed
reduced DAF and CD59 expression.
10
The lower expression of
cell-surface CD14 protein on monocytes (52% of reference
level) of Patient 1 is difficult to explain. It might be resulted
from an unspecific cell activation or individual variability.
11,15
However it may be a result of release of this protein from
plasma membrane to the medium in PGAP2 mutated mono-
cytes
39
similarly to the AP release. Davis et al. demonstrated
experiments with haploid genetic screens PGAP2 knockout
cells suggesting that GPI modifications have markedly distinct
effects on individual GPI-AP pathways. Moreover, these data
indicate that incomplete GPI anchor modification can result in
intracellular retention of certain GPI-APs.
7
The GPI anchor assessment in our Patient 2 revealed
remarkable abnormalities including decreased binding of
FLAER and the decreased expression of some GPI-APs detected
by monoclonal antibodies comparing to the healthy in-
dividuals. The most significant differences were noticed in the
GPI anchors and two GPI-APs CD24 and CD66b on granulocytes
that were expressed on 21e34% of reference levels. Ohba et al.
reported similar results on PIGN mutated granulocytes; they
found expression of CD16 and CD24 decreased to 26e54% of
control values. However, they noticed no differences in the
expression of CD59, CD55, CD48, or GPI tested by FLAER
binding on B lymphoblasts in their patients as compared with
a control group.
35
Only a slight difference between our Patient 2 and healthy
individuals in FLAER binding to GPI anchors on monocytes
(85% of control expression), except CD14 (expression reduced
to 42% of reference levels) was demonstrated. Furthermore,
we found decreased (76%) CD59 expression on erythrocytes, a
finding similar to that observed by Maydan et al.
26
on fibro-
blasts in a patient with a homozygous PIGN mutation.
The flow cytometry results demonstrated functional
impairment of the PIGN mutations on GPI and GPI-anchored
protein expression in blood cells, however, the consequences
of this phenomenon are not known. They might depend on the
involved tissue or the function of the protein that is bound to
GPI. As suggested by Ohba et al., changes to a subset of GPI-
anchored proteins can be sufficient to cause neurological de-
fects and (or) an abnormal structure of the GPI moiety in PIGN
mutated cells might affect the function of these proteins.
GPI-anchor deficiencies cannot be diagnosed or even
screened with transferrin isoform or APOCIII glycosylation
assay. Furthermore results in unrecognizable and unspecific
clinical phenotype underline the importance of whole-exome
sequencing testing in diagnostic procedures.
14
However,
based on our results we suggest that the further studies
leading to the development of the screening test should be
concentrated on the analysis of granulocytes and monocytes
for expression of both GPI anchors and GPI-APs performed
simultaneously. Evaluation of erythrocytes is unlikely to be
useful (too small differences in expression between patients
and healthy controls were observed).
This study was partially financed by CMHI project S136/13
and NCN Project Harmonia 4 No. UMO-2013/08/M/NZ5/00978.
Conflict of interest
None.
Acknowledgments
The authors dedicate this work to their late colleague, Dr.
Maciej Adamowicz, a biochemist with a keen and inquisitive
mind, a humble and benevolent man who patiently taught us
all in Poland about disorders of glycosylation. We missed
Maciek so much at the time of writing this paper.
Many thanks to the parents of both girls for their invalu-
able contribution describing the details of the clinical course
of this very rare disorder.
references
1. Almeida AM, Murakami Y, Layton DM, Hillmen P, Sellick GS,
Maeda Y, Richards S, Patterson S, Kotsianidis I, Mollica L,
Crawford DH, Baker A, Ferguson M, Roberts I, Houlston R,
Kinoshita T, Karadimitris A. Hypomorphic promoter
mutation in PIGM causes inherited
glycosylphosphatidylinositol deficiency. Nat Med
2006;12(7):846e51.
2. Borowitz MJ, Craig FE, DiGiuseppe JA, Illingworth AJ, Rosse W,
Sutherland DR, Wittwer CT, Richards SJ, Clinical Cytometry
Society. Guidelines for the diagnosis and monitoring of
paroxysmal nocturnal hemoglobinuria and related disorders
by flow cytometry. Cytometry Part B Clin Cytometry
2010;78B(4):211e28.
3. Brady PD, Moerman P, De Catte L, Deprest J, Devriendt K,
Vermeesch JR. Exome sequencing identifies a recessive PIGN
splice site mutation as a cause of syndromic congenital
diaphragmatic hernia. Eur J Med Genet 2014;57(9):487e93.
5. Chiyonobu T, Inoue N, Morimoto M, Kinoshita T, Murakami Y.
Glycosylphosphatidylinositol (GPI) anchor deficiency caused
by mutations in PIGW is associated with west syndrome and
hyperphosphatasia with mental retardation syndrome. J Med
Genet 2014;51:203e7.
6. Couser NL, Masood MM, Strande NT, Foreman AK, Crooks K,
Weck KE, Lu M, Wilhelmsen KC, Roche M, Evans JP, Berg JS,
Powell CM. The phenotype of multiple congenital anomalies-
hypotonia-seizures syndrome 1: report and review. Am J Med
Genet A 2015;167(9):2176e81.
7. Davis EM, Kim J, Menasche BL, Sheppard J, Liu X, Tan AC,
Shen J. Comparative haploid genetic screens reveal divergent
pathways in the biogenesis and trafficking of
glycophosphatidylinositol-anchored proteins. Cell Rep
2015;11(11):1727e36.
8. DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR,
Hartl C, Philippakis AA, del Angel G, Rivas MA, Hanna M,
McKenna A, Fennell TJ, Kernytsky AM, Sivachenko AY,
Cibulskis K, Gabriel SB, Altshuler D, Daly MJ. A framework
for variation discovery and genotyping using next-
generation DNA sequencing data. Nat Genet
2011;43:491e8.
9. Golla A, Jansson A, Ramser J, Hellebrand H, Zahn R,
Meitinger T, Belohradsky BH, Meindl A. Chronic recurrent
european journal of paediatric neurology 20 (2016) 462e473472
multifocal osteomyelitis (CRMO): evidence for a susceptibility
gene located on chromosome 18q21.3e18q22. Eur J Hum Genet
2002;10(3):217e21.
10. Hansen L, Tawamie H, Murakami Y, Mang Y, ur Rehman S,
Buchert R, Schaffer S, Muhammad S, Bak M, N
othen MM,
Bennett EP, Maeda Y, Aigner M, Reis A, Kinoshita T,
Tommerup N, Baig SM, Abou Jamra R. Hypomorphic
mutations in PGAP2, encoding a GPI-anchor-remodeling
protein, cause autosomal-recessive intellectual disability. Am
J Hum Genet 2013;92(4):575e83.
11. Hernandez-Campo PM, Almeida J, Sanchez ML, Malvezzi M,
Orfao A. Normal patterns of expression of
glycosylphosphatidylinositol-anchored proteins on different
subsets of peripheral blood cells: a frame of reference for the
diagnosis of paroxysmal nocturnal hemoglobinuria.
Cytometry Part B Clin Cytometry 2006;70B:71e81.
13. Horn D, Krawitz P, Mannhardt A, Korenke GC, Meinecke P.
Hyperphosphatasia-mental retardation syndrome due to
PIGV mutations: expanded clinical spectrum. Am J Med Genet
A2011;155A(8):1917e22.
14. Howard MF, Murakami Y, Pagnamenta AT, Daumer-Haas C,
Fischer B, Hecht J, Keays DA, Knight SJ, K
olsch U, Kru
¨ger U,
Leiz S, Maeda Y, Mitchell D, Mundlos S, Phillips 3rd JA,
Robinson PN, Kini U, Taylor JC, Horn D, Kinoshita T,
Krawitz PM. Mutations in PGAP3 impair GPI-anchor
maturation, causing a subtype of hyperphosphatasia with
mental retardation. Am J Hum Genet 2014;94(2):278e87.
15. Hudig D, Hunter KW, Diamond WJ, Redelman D. Properties of
human blood monocytes. II. Monocytes from healthy adults
are highly heterogeneous within and among individuals.
Cytometry Part B Clin Cytometry 2014;86:121e34.
16. Ilkovski B, Pagnamenta AT, O'Grady GL, Kinoshita T,
Howard MF, Lek M, Thomas B, Turner A, Christodoulou J,
Sillence D, Knight SJL, Popitsch N, Keays DA, Anzilotti C,
Goriely A, Waddell LB, Brilot F, North KN, Kanzawa N,
Macarthur DG, Taylor JC, Kini U, Murakami Y, Clarke NF.
Mutations in PIGY: expanding the phenotype of inherited
glycosylphosphatidylinositol deficiencies. Hum Mol Genet
2015;24(21):6146e59.
17. Johnston JJ, Gropman AL, Sapp JC, Teer JK, Martin JM, Liu CF,
Yuan X, Ye Z, Cheng L, Brodsky RA, Biesecker LG. The
phenotype of a germline mutation in PIGA: the gene
somatically mutated in paroxysmal nocturnal
hemoglobinuria. Am J Hum Genet 2012;90(2):295e300.
18. Kinoshita T. Biosynthesis and deficiencies of
glycosylphosphatidylinositol. Proc Jpn Acad Ser B Phys Biol Sci
2014;90(4):130e43.
19. Krawitz PM, Murakami Y, Hecht J, Kru
¨ger U, Holder SE,
Mortier GR, Delle Chiaie B, De Baere E, Thompson MD,
Roscioli T, Kielbasa S, Kinoshita T, Mundlos S, Robinson PN,
Horn D. Mutations in PIGO, a member of the GPI-anchor-
synthesis pathway, cause hyperphosphatasia with mental
retardation. Am J Hum Genet 2012;91(1):146e51.
20. Krawitz PM, Murakami Y, Rieß A, Hietala M, Kru
¨ger U, Zhu N,
Kinoshita T, Mundlos S, Hecht J, Robinson PN, Horn D. PGAP2
mutations, affecting the GPI-anchor-synthesis pathway,
cause hyperphosphatasia with mental retardation syndrome.
Am J Hum Genet 2013;92(4):584e9.
22. Kvarnung M, Nilsson D, Lindstrand A, Korenke GC, Chiang SC,
Blennow E, Bergmann M, St
odberg T, M
akitie O, Anderlid BM,
Bryceson YT, Nordenskj
old M, Nordgren A. A novel
intellectual disability syndrome caused by GPI anchor
deficiency due to homozygous mutations in PIGT. J Med Genet
2013;50(8):521e8.
23. Li H, Durbin R. Fast and accurate short read alignment with
burrows-wheeler transform. Bioinformatics 2009;25:1754e60.
24. Maeda Y, Tashima Y, Houjou T, Fujita M, Yoko-o T, Jigami Y,
Taguchi R, Kinoshita T. Fatty acid remodeling of GPI-
anchored proteins is required for their raft association. Mol
Biol Cell 2007;18(4):1497e506.
25. Martin HC, Kim GE, Pagnamenta AT, Murakami Y, Carvill GL,
Meyer E, Copley RR, Rimmer A, Barcia G, Fleming MR,
Kronengold J, Brown MR, Hudspith KA, Broxholme J,
Kanapin A, Cazier JB, Kinoshita T, Nabbout R, WGS500
Consortium, Bentley D, McVean G, Heavin S, Zaiwalla Z,
McShane T, Mefford HC, Shears D, Stewart H, Kurian MA,
Scheffer IE, Blair E, Donnelly P, Kaczmarek LK, Taylor JC.
Clinical whole-genome sequencing in severe early-onset
epilepsy reveals new genes and improves molecular
diagnosis. Hum Mol Genet 2014;23(12):3200e11.
26. Maydan G, Noyman I, Har-Zahav A, Neriah ZB, Pasmanik-
Chor M, Yeheskel A, Albin-Kaplanski A, Maya I, Magal N,
Birk E, Simon AJ, Halevy A, Rechavi G, Shohat M,
Straussberg R, Basel-Vanagaite L. Multiple congenital
anomalies-hypotonia-seizures syndrome is caused by a
mutation in PIGN. J Med Genet 2011;48(6):383e9.
27. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K,
Kernytsky A, Garimella K, Altshuler D, Gabriel S, Daly M,
DePristo MA. The genome analysis toolkit: a MapReduce
framework for analyzing next-generation DNA sequencing
data. Genome Res 2010;20:1297e303.
28. Menon, Kinoshita, Orlean, Tamanoi.
Glycosylphosphatidylinositol (GPI) anchoring of proteins. Imprint.
1st ed. Academic Press; 2009, ISBN 9780123749635 [eBook
ISBN: 9780080960951].
29. Morava E, Rahman S, Peters V, Baumgartner MR, Patterson M,
Zschocke J. Quo vadis: the re-definition of inborn metabolic
diseases.J Inherit Metab Dis 2015;38(6):1003e6.
30. Morava E, van den Heuvel L, Hol F, de Vries MC, Hogeveen M,
Rodenburg RJ, Smeitink JA. Mitochondrial disease criteria.
Diagnostic application in children. Neurology 2006;67:1823e6.
31. Mu~
niz M, Riezman H. Trafficking of glycosylphosphatidyl
inositol anchored proteins from the endoplasmic reticulum
to the cell surface. J Lipid Res 2015. pii: jlr.R062760.
32. Murakami Y, Tawamie H, Maeda Y, Bu
¨ttner C, Buchert R,
Radwan F, Schaffer S, Sticht H, Aigner M, Reis A, Kinoshita T,
Jamra RA. Null mutation in PGAP1 impairing Gpi-anchor
maturation in patients with intellectual disability and
encephalopathy. PLoS Genet 2014;10(5):e1004320.
34. Ng BG, Freeze HH. Human genetic disorders involving
glycosylphosphatidylinositol (GPI) anchors and
glycosphingolipids (GSL). J Inherit Metab Dis 2015;38(1):171e8.
35. Ohba C, Okamoto N, Murakami Y, Suzuki Y, Tsurusaki Y,
Nakashima M, Miyake N, Tanaka F, Kinoshita T,
Matsumoto N, Saitsu H. PIGN mutations cause congenital
anomalies, developmental delay, hypotonia, epilepsy, and
progressive cerebellar atrophy. Neurogenetics 2014;15(2):85e92
[Erratum in: Neurogenetics. 2014;15(2):93].
36. Ploski R, Pollak A, Mu
¨ller S, Franaszczyk M, Michalak E,
Kosinska J, Stawinski P, Spiewak M, Seggewiss H, Bilinska ZT.
Does p.Q247X in TRIM63 cause human hypertrophic
cardiomyopathy? Circ Res 2014;114(2):e2e5.
37. Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M,
Lander ES, Getz G, Mesirov JP. Integrative genomics viewer.
Nat Biotechnol 2011;29:24e6.
38. Sharom F. GPI-anchored proteins: biophysical behaviour and
cleavage by Pi-specific phospholipases. In: Dangerfield John
A, Metzner Christoph, editors. GPI membrane anchors ethe
much needed link; 2010 [eISBN: 978-1-60805-123-6; ISBN: 978-1-
60805-375-9].
39. TashimaY, Taguchi R, MurataC, Ashida H, KinoshitaT, Maeda Y.
PGAP2 is essential for correct processing and stable expression
of GPI-anchored proteins. Mol Biol Cell 2006;17(3):1410e20.
40. Wang K, Li M, Hakonarson H. ANNOVAR: functional
annotation of genetic variants from high-throughput
sequencing data. Nucleic Acids Res 2010;38:e164.
european journal of paediatric neurology 20 (2016) 462e473 473
... Both DNA sequence variants identified in family 1 have not been previously reported in the literature. Missense and splicing variants of family 2 have been reported in several studies [6,9,[32][33][34]. In silico analysis with SpliceAI predicted that the splice site c.1674+1G>C variant would most probably affect pre-mRNA splicing. ...
... To our knowledge, PIGN-altering variants have been reported in 33 males and 43 females from 66 unrelated families with a wide range of clinical manifestations. The data from several previous publications suggest that congenital anomalies should not be considered one of the main features of PIGN-related disease [11,32,[35][36][37]. Thiffault et al. (2017) reported on a male who exhibited intellectual disability, hypotonia, and seizures without congenital anomalies or any obvious dysmorphic features [11]. ...
... )[9,[32][33][34].Bayat et al. (2022) reported the same compound heterozygous NG_033144.1(NM_176787.5):c.[932T>G];[1674+1G>C] PIGN genotype in a female 6.5 years of age who demonstrated similar global developmental delay, seizures, and hypotonia. ...
Article
Full-text available
Background and Objectives: Pathogenic variants of PIGN are a known cause of multiple congenital anomalies-hypotonia-seizures syndrome 1 (MCAHS1). Many affected individuals have clinical features overlapping with Fryns syndrome and are mainly characterised by developmental delay, congenital anomalies, hypotonia, seizures, and specific minor facial anomalies. This study investigates the clinical and molecular data of three individuals from two unrelated families, the clinical features of which were consistent with a diagnosis of MCAHS1. Materials and Methods: Next-generation sequencing (NGS) technology was used to identify the changes in the DNA sequence. Sanger sequencing of gDNA of probands and their parents was used for validation and segregation analysis. Bioinformatics tools were used to investigate the consequences of pathogenic or likely pathogenic PIGN variants at the protein sequence and structure level. Results: The analysis of NGS data and segregation analysis revealed a compound heterozygous NM_176787.5:c.[1942G>T];[1247_1251del] PIGN genotype in family 1 and NG_033144.1(NM_176787.5):c.[932T>G];[1674+1G>C] PIGN genotype in family 2. In silico, c.1942G>T (p.(Glu648Ter)), c.1247_1251del (p.(Glu416GlyfsTer22)), and c.1674+1G>C (p.(Glu525AspfsTer68)) variants are predicted to result in a premature termination codon that leads to truncated and functionally disrupted protein causing the phenotype of MCAHS1 in the affected individuals. Conclusions: PIGN-related disease represents a wide spectrum of phenotypic features, making clinical diagnosis inaccurate and complicated. The genetic testing of every individual with this phenotype provides new insights into the origin and development of the disease.
... Biallelic variants have been reported in two genes encoding post-GPI attachment to proteins (PGAP) enzymes that act in the Golgi to stabilize membrane attachment of GPI-anchored proteins (GPI-AP). HPMRS3 [MIM: 614207], or GPIBD8, has been associated with biallelic variants of PGAP2, a gene encoding a membrane protein required, during fatty-acid remodeling, for reacylation of the GPI-lyso intermediate with a saturated fatty acid, such as stearic acid [45][46][47][48]. HPMRS4 [MIM: 615716], or GPIBD10, has been associated with biallelic variants of PGAP3, the gene encoding a seven transmembrane protein that removes an unsaturated fatty acid from the sn-2 position of GPI prior to reacetylation by PGAP2 [49,50]. ...
... The unsaturated fatty acid is cleaved by phospholipase C [49,50], resulting in hyperphosphatasia. By contrast, pathogenic PGAP2 variants [45][46][47][48] are unable to reacetylate, with saturated stearic acid, the lyso-GPI intermediate generated by the action of PGAP3. Phospholipase D (PLD) [62], in turn, cleaves the lyso-GPI intermediate, resulting in transport of the unstable anchor and it's attached protein, alkaline phosphatase, to the extracellular compartment. ...
Article
Full-text available
The case report by Mabry et al. (1970) of a family with four children with elevated tissue non-specific alkaline phosphatase, seizures and profound developmental disability, became the basis for phenotyping children with the features that became known as Mabry syndrome. Aside from improvements in the services available to patients and families, however, the diagnosis and treatment of this, and many other developmental disabilities, did not change significantly until the advent of massively parallel sequencing. As more patients with features of the Mabry syndrome were identified, exome and genome sequencing were used to identify the glycophosphatidylinositol (GPI) biosynthesis disorders (GPIBDs) as a group of congenital disorders of glycosylation (CDG). Biallelic variants of the phosphatidylinositol glycan (PIG) biosynthesis, type V (PIGV) gene identified in Mabry syndrome became evidence of the first in a phenotypic series that is numbered HPMRS1-6 in the order of discovery. HPMRS1 [MIM: 239300] is the phenotype resulting from inheritance of biallelic PIGV variants. Similarly, HPMRS2 (MIM 614749), HPMRS5 (MIM 616025) and HPMRS6 (MIM 616809) result from disruption of the PIGO, PIGW and PIGY genes expressed in the endoplasmic reticulum. By contrast, HPMRS3 (MIM 614207) and HPMRS4 (MIM 615716) result from disruption of post attachment to proteins PGAP2 (HPMRS3) and PGAP3 (HPMRS4). The GPI biosynthesis disorders (GPIBDs) are currently numbered GPIBD1-21. Working with Dr. Mabry, in 2020, we were able to use improved laboratory diagnostics to complete the molecular diagnosis of patients he had originally described in 1970. We identified biallelic variants of the PGAP2 gene in the first reported HPMRS patients. We discuss the longevity of the Mabry syndrome index patients in the context of the utility of pyridoxine treatment of seizures and evidence for putative glycolipid storage in patients with HPMRS3. From the perspective of the laboratory innovations made that enabled the identification of the HPMRS phenotype in Dr. Mabry’s patients, the need for treatment innovations that will benefit patients and families affected by developmental disabilities is clear.
... 14 The clinical phenotype is characterized by varying degrees of developmental delay, intellectual disability and elevated ALP, but unlike other HPMRS types, epilepsy, hypotonia, dysmorphisms and organ anomalies are less common (Table 3). 8,11,12,[14][15][16] Including our patient, 20 individuals with homozygous or compound heterozygous missense variants have been described to date (Table 2). 8,12,[14][15][16] Their clinical characteristics are described in Table 3. ...
... 8,11,12,[14][15][16] Including our patient, 20 individuals with homozygous or compound heterozygous missense variants have been described to date (Table 2). 8,12,[14][15][16] Their clinical characteristics are described in Table 3. ...
Article
Full-text available
Glycosylphosphatidylinositol anchored proteins (GPI‐APs) represent a class of molecules attached to the external leaflet of the plasma membrane by the GPI anchor where they play important roles in numerous cellular processes including neurogenesis, cell adhesion, immune response and signalling. Within the group of GPI anchor defects, six present with the clinical phenotype of Hyperphosphatasia with Mental Retardation Syndrome (HPMRS, Mabry Syndrome) characterized by moderate to severe intellectual disability, dysmorphic features, hypotonia, seizures and persistent hyperphosphatasia. We report the case of a 5‐year‐old female with global developmental delay associated with precocious puberty and persistently raised plasma alkaline phosphatase. Targeted next generation sequencing analysis of the HPMRS genes identified novel compound heterozygous variants in the PGAP2 gene (c.103del p.(Leu35Serfs*90)and c.134A > Gp.(His45Arg)) consistent with the diagnosis of HPMRS type 3. Cerebrospinal fluid (CSF) neurotransmitter analysis showed low levels of pyridoxal phosphate and 5‐methyltetrahydrofolate and raised homovanillic acid. Supplementation with pyridoxine and folinic acid led to normalization of biochemical abnormalities. The patient continues to make developmental progress with significant improvement in speech and fine motor skills. Our reported case expands the clinical spectrum of HPMRS3 in which multisystem involvement is being increasingly recognized. Furthermore, it shows that miss‐targeting GPI‐APs and the effect on normal cellular function could provide a physiopathologic explanation for the CSF biochemical abnormalities with management implications for a group of disorders that currently has no treatment that can lead possibly to improved clinical outcomes.
... Introduction encode proteins involved in the biosynthesis, transfer, and modification of GPI-anchors. 1 The phenotypic expression can include congenital anomalies, intellectual disability, epilepsy, and characteristic facial features, alongside reduced in vivo expression of GPI-APs at the cell surface. [3][4][5][6] This clinical variety has been proposed to be partially related to the position of the defect in this pathway. 1 Collectively these genetic conditions are known as inherited GPI deficiencies (IGDs) and are part of the broader group of conditions known as congenital disorders of glycosylation. ...
... To date, 40 individuals have been reported with biallelic variants in PIGN. [3][4][5][6][9][10][11][12][13][14][19][20][21][22][23] Some of these reports have suggested a possible correlation between the effects of variants on residual PIGN function and clinical severity. 6,9,13 In this article, we report a further 21 patients and compare their clinical and molecular features with those of all previously reported cases, identifying many novel variants and gaining previously unreported insights into genotype-phenotype correlations. ...
Article
Purpose: Biallelic PIGN variants have been described in Fryns syndrome, multiple congenital anomalies-hypotonia-seizure syndrome (MCAHS), and neurologic phenotypes. The full spectrum of clinical manifestations in relation to the genotypes is yet to be reported. Methods: Genotype and phenotype data were collated and analyzed for 61 biallelic PIGN cases: 21 new and 40 previously published cases. Functional analysis was performed for 2 recurrent variants (c.2679C>G p.Ser893Arg and c.932T>G p.Leu311Trp). Results: Biallelic-truncating variants were detected in 16 patients-10 with Fryns syndrome, 1 with MCAHS1, 2 with Fryns syndrome/MCAHS1, and 3 with neurologic phenotype. There was an increased risk of prenatal or neonatal death within this group (6 deaths were in utero or within 2 months of life; 6 pregnancies were terminated). Incidence of polyhydramnios, congenital anomalies (eg, diaphragmatic hernia), and dysmorphism was significantly increased. Biallelic missense or mixed genotype were reported in the remaining 45 cases-32 showed a neurologic phenotype and 12 had MCAHS1. No cases of diaphragmatic hernia or abdominal wall defects were seen in this group except patient 1 in which we found the missense variant p.Ser893Arg to result in functionally null alleles, suggesting the possibility of an undescribed functionally important region in the final exon. For all genotypes, there was complete penetrance for developmental delay and near-complete penetrance for seizures and hypotonia in patients surviving the neonatal period. Conclusion: We have expanded the described spectrum of phenotypes and natural history associated with biallelic PIGN variants. Our study shows that biallelic-truncating variants usually result in the more severe Fryns syndrome phenotype, but neurologic problems, such as developmental delay, seizures, and hypotonia, present across all genotypes. Functional analysis should be considered when the genotypes do not correlate with the predicted phenotype because there may be other functionally important regions in PIGN that are yet to be discovered.
... The PIGN (phosphatidylinositol glycan anchor biosynthesis class N) gene (OMIM* 606097) is located on chromosome 18q21.33 and is composed of 31 exons (29 coding) spanning 142.8 kb, and encoding a glycosylphosphatidylinositol ethanolamine phosphate transferase 1, a protein with 931 amino acids involved in the glycosylphosphatidylinositol (GPI)-anchor biosynthesis pathway, a deeply conserved process that enables proteins to bind to the cell surface membrane [6][7][8][9][10]. PIGN is responsible for the addition of phosphoethanolamine to the first mannose in the GPI [8]. ...
Article
Full-text available
Glycosylation is the most common protein and lipid post-translational modification in humans. Congenital disorders of glycosylation (CDG) are characterized by both genetic and clinical heterogeneity, presenting multisystemic manifestations, and in most cases are autosomal recessive in inheritance. The PIGN gene is responsible for the addition of phosphoethanolamine to the first mannose in the glycosylphosphatidylinositol (GPI)-anchor biosynthesis pathway, a highly conserved process that enables proteins to bind to the cell surface membrane. Here, we report a family with two siblings pediatric cases with the exact same compound heterozygous variants in PIGN. The (c.776T > C) variant of uncertain significance (VUS) together with a known pathogenic variant (c.932T > G), resulting in clinical features compatible with PIGN-related conditions, more specific the CDG. This is the first time that PIGN variant c.776T > C is reported in literature in individuals with PIGN-congenital disorder of glycosylation (PIGN-CDG), and the current submission in ClinVar by Invitae® is specifically of our case. Detailed clinical information and molecular analyses are presented. Here, we show for the first time two affected siblings with one pathogenic variant (c.932T > G) and the c.776T > C VUS in trans. In honor of the family, we propose the name Bella-Noah Syndrome for disorder.
... HPMRS phenotypes can also result from bi-allelic inheritance of variants in two genes encoding post-GPI attachment to proteins (PGAP) that, expressed in the Golgi, help, to stabilize membrane attachment of GPI-anchored proteins (GPI-AP): HPMRS3 (MIM: 614207), or GPIBD8, resulting from PGAP2 variants [21][22][23][24] and HPMRS4 (MIM: 615716), or GPIBD10, resulting from PGAP3 variants [25,26] (https://ncbi.nlm.nih.gov/gtr/conditions/C1853205/) (accessed on 1 December 2022). ...
Article
Full-text available
Unlabelled: We present a case report of a child with features of hyperphosphatasia with neurologic deficit (HPMRS) or Mabry syndrome (MIM 239300) with variants of unknown significance in two post-GPI attachments to proteins genes, PGAP2 and PGAP3, that underlie HPMRS 3 and 4. Background: In addition to HPMRS 3 and 4, disruption of four phosphatidylinositol glycan (PIG) biosynthesis genes, PIGV, PIGO, PIGW and PIGY, result in HPMRS 1, 2, 5 and 6, respectively. Methods: Targeted exome panel sequencing identified homozygous variants of unknown significance (VUS) in PGAP2 c:284A>G and PGAP3 c:259G>A. To assay the pathogenicity of these variants, we conducted a rescue assay in PGAP2 and PGAP3 deficient CHO cell lines. Results: Using a strong (pME) promoter, the PGAP2 variant did not rescue activity in CHO cells and the protein was not detected. Flow cytometric analysis showed that CD59 and CD55 expression on the PGAP2 deficient cell line was not restored by variant PGAP2. By contrast, activity of the PGAP3 variant was similar to wild-type. Conclusions: For this patient with Mabry syndrome, the phenotype is likely to be predominantly HPMRS3: resulting from autosomal recessive inheritance of NM_001256240.2 PGAP2 c:284A>G, p.Tyr95Cys. We discuss strategies for establishing evidence for putative digenic inheritance in GPI deficiency disorders.
Article
PGAP2 gene has been known to be the cause of “hyperphosphatasia, mental retardation syndrome-3” (HPMRS3). To date, 14 pathogenic variants in PGAP2 have been identified as the cause of this syndrome in 24 patients described in single-case reports or small clinical series with pan-ethnic distribution. We aim to present a pediatric PGAP2-mutated case, intending to further expand the clinical phenotype of the syndrome and to report our experience on a therapeutic approach to drug-resistant epilepsy. We present the clinical, neuroradiological, and genetic characterization of a Caucasian pediatric subject with biallelic pathogenic variants in the PGAP2 gene revealed by next generation sequencing analysis. We identified a subject who presented with global developmental delay and visual impairment. Brain magnetic resonance imaging showed mild hypoplasia of the inferior cerebellar vermis and corpus callosum and mild white matter reduction. Laboratory investigations detected an increase in alkaline phosphatase. At the age of 13 months, he began to present epileptic focal seizures with impaired awareness, which did not respond to various antiseizure medications. Electroencephalogram (EEG) showed progressive background activity disorganization and multifocal epileptic abnormalities. Treatment with high-dose pyridoxine showed partial benefit, but the persistence of seizures and the lack of EEG amelioration prompted us to introduce ketogenic diet treatment. Our case provides a further phenotypical expansion of HPMRS3 to include developmental and epileptic encephalopathy. Due to the limited number of patients reported so far, the full delineation of the clinical spectrum of HPMRS3 and indications for precision medicine would benefit from the description of new cases and their follow-up evaluations.
Article
Full-text available
Glycosylphosphatidylinositols (GPIs) anchor over 150 proteins as GPI-anchored proteins (GPI-APs) with crucial roles in diverse biological processes. The highly conserved biosynthesis of GPI-APs involves precise steps with at least 21 genes, categorized as PIG and PGAP genes. Pathogenic variants in these genes are linked to human diseases, highlighting the importance of each biosynthesis step. PGAP2 stands out among these genes due to its association with an expanded clinical spectrum of neurodevelopmental disorder (NDD) phenotypes with biallelic pathogenic variants. We present four patients from two families, one consanguineous and the other nonconsanguineous, each displaying distinct clinical presentations, including intellectual disability, hyperphosphatasia, hearing impairment, and epilepsy, as well as craniofacial and digital anomalies. Genetic analyses revealed homozygous and novel compound heterozygous missense variants in PGAP2 in four affected individuals, confirming the molecular diagnosis of hyperphosphatasia with impaired intellectual development syndrome 3 (HPMRS3). Importantly, the three amino acids affected by missense variants exhibit complete conservation in 10 vertebrate species, illuminating their crucial role in the gene’s functionality. Protein modeling provided additional evidence for the pathogenicity of the three substitutions, demonstrating their detrimental impact on protein folding and putative protein-protein interactions, ultimately leading to impaired protein function. The four patients in our study displayed common phenotypic features, such as brachydactyly, camptodactyly, and syndactyly, which have not been previously documented in individuals with PGAP2 variants. Notably, the occurrence of macrocephaly in two affected brothers from a consanguineous Pakistani family represents a novel finding. These previously unreported digital anomalies, along with macrocephaly and the identification of novel compound heterozygous variants, contribute to the expansion of the phenotypic and genotypic spectrum of HPMRS3 associated with PGAP2 variants.
Article
Full-text available
In eukaryotes, many cell surface proteins are attached to the plasma membrane via a glycolipid, glycosylphosphatidylinositol (GPI) anchor. GPI-anchored proteins (GPI-APs) receive the GPI anchor as a conserved posttranslational modification in the lumen of the endoplasmic reticulum (ER). After anchor attachment, the GPI anchor is structurally remodeled to function as a transport signal that actively triggers the delivery of GPI-APs from the ER to the plasma membrane, via the Golgi apparatus. The structure and composition of the GPI anchor confer a special mode of interaction with membranes of GPI-APs within the lumen of secretory organelles that lead them to be differentially trafficked from other secretory membrane proteins. In this review, we examine the mechanisms by which GPI-APs are selectively transported through the secretory pathway, with special focus on the recent progress made in their actively regulated export from the ER and the trans-Golgi network.
Article
Full-text available
Glycosylphosphatidylinositol (GPI) anchored proteins are ubiquitously expressed in the human body and are important for various functions at the cell surface. Mutations in many GPI biosynthesis genes have been described to date in patients with multi-system disease and together these constitute a subtype of congenital disorders of glycosylation. We used whole exome sequencing in two families to investigate the genetic basis of disease and used RNA and cellular studies to investigate the functional consequences of sequence variants in the PIGY gene. Two families with different phenotypes had homozygous recessive sequence variants in the GPI biosynthesis gene PIGY. Two sisters with c.137T>C (p.Leu46Pro) PIGY variants had multi-system disease including dysmorphism, seizures, severe developmental delay, cataracts and early death. There were significantly reduced levels of GPI-anchored proteins (CD55 and CD59) on the surface of patient-derived skin fibroblasts (∼20-50% compared to controls). In a second, consanguineous family, two siblings had moderate development delay and microcephaly. A homozygous PIGY promoter variant (c.-540G>A) was detected within a 7.7 Mb region of autozygosity. This variant was predicted to disrupt a SP1 consensus binding site and was shown to be associated with reduced gene expression. Mutations in PIGY can occur in coding and non-coding regions of the gene and cause variable phenotypes. This paper contributes to understanding of the range of disease phenotypes and disease genes associated with deficiencies of the GPI-anchor biosynthesis pathway and also serves to highlight the potential importance of analysing variants detected in 5'-UTR regions despite their typically low coverage in exome data. © The Author 2015. Published by Oxford University Press.
Article
Full-text available
Glycophosphatidylinositol-anchored proteins (GPI-APs) play essential roles in physiology, but their biogenesis and trafficking have not been systematically characterized. Here, we took advantage of the recently available haploid genetics approach to dissect GPI-AP pathways in human cells using prion protein (PrP) and CD59 as model molecules. Our screens recovered a large number of common and unexpectedly specialized factors in the GPI-AP pathways. PIGN, PGAP2, and PIGF, which encode GPI anchor-modifying enzymes, were selectively isolated in the CD59 screen, suggesting that GPI anchor composition significantly influences the biogenesis of GPI-APs in a substrate-dependent manner. SEC62 and SEC63, which encode components of the ER-targeting machinery, were selectively recovered in the PrP screen, indicating that they do not constitute a universal route for the biogenesis of mammalian GPI-APs. Together, these comparative haploid genetic screens demonstrate that, despite their similarity in overall architecture and subcellular localization, GPI-APs follow markedly distinct biosynthetic and trafficking pathways. Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.
Article
Full-text available
The Multiple Congenital Anomalies-Hypotonia-Seizures Syndrome 1 (MCAHS1) has been described in two families to date. We describe a 2-year-old Mexican American boy with the syndrome and additional manifestations not yet reported as part of the phenotype. The patient presented with severe hypotonia, microphallus and left cryptorchidism, and was later diagnosed with epilepsy and severe cortical visual impairment. He also had supernumerary nipples, pectus excavatum, a short upturned nose, fleshy ear lobes, and a right auricular pit. Massively parallel exome sequencing and analysis revealed two novel compound heterozygous missense (Trp136Gly and Ser859Thr) variants in the PIGN gene. This report extends and further defines the phenotype of this syndrome. © 2015 Wiley Periodicals, Inc. © 2015 Wiley Periodicals, Inc.
Article
Full-text available
Many eukaryotic cell-surface proteins are anchored to the membrane via glycosylphosphatidylinositol (GPI). There are at least 26 genes involved in biosynthesis and remodeling of GPI anchors. Hypomorphic coding mutations in seven of these genes have been reported to cause decreased expression of GPI anchored proteins (GPI-APs) on the cell surface and to cause autosomal-recessive forms of intellectual disability (ARID). We performed homozygosity mapping and exome sequencing in a family with encephalopathy and non-specific ARID and identified a homozygous 3 bp deletion (p.Leu197del) in the GPI remodeling gene PGAP1. PGAP1 was not described in association with a human phenotype before. PGAP1 is a deacylase that removes an acyl-chain from the inositol of GPI anchors in the endoplasmic reticulum immediately after attachment of GPI to proteins. In silico prediction and molecular modeling strongly suggested a pathogenic effect of the identified deletion. The expression levels of GPI-APs on B lymphoblastoid cells derived from an affected person were normal. However, when those cells were incubated with phosphatidylinositol-specific phospholipase C (PI-PLC), GPI-APs were cleaved and released from B lymphoblastoid cells from healthy individuals whereas GPI-APs on the cells from the affected person were totally resistant. Transfection with wild type PGAP1 cDNA restored the PI-PLC sensitivity. These results indicate that GPI-APs were expressed with abnormal GPI structure due to a null mutation in the remodeling gene PGAP1. Our results add PGAP1 to the growing list of GPI abnormalities and indicate that not only the cell surface expression levels of GPI-APs but also the fine structure of GPI-anchors is important for the normal neurological development.
Article
Rapid improvements in sequencing and array-based platforms are resulting in a flood of diverse genome-wide data, including data from exome and whole-genome sequencing, epigenetic surveys, expression profiling of coding and noncoding RNAs, single nucleotide polymorphism (SNP) and copy number profiling, and functional assays. Analysis of these large, diverse data sets holds the promise of a more comprehensive understanding of the genome and its relation to human disease. Experienced and knowledgeable human review is an essential component of this process, complementing computational approaches. This calls for efficient and intuitive visualization tools able to scale to very large data sets and to flexibly integrate multiple data types, including clinical data. However, the sheer volume and scope of data pose a significant challenge to the development of such tools.
Article
Glycosylation - enabling genes are thought to comprise approximately 1-2 % of the human genome, thus, it is not surprising that more than 100 genetic disorders have been identified in this complex multi-pathway cellular process. Recent advances in next generation sequencing technology (NGS) have led to the discovery of genetic causes of many new disorders and importantly highlighted the broad phenotypes that occur. Here we will focus on two glycosylation pathways that involve lipids; glycosylphosphatidylinositol (GPI) anchors and glycosphingolipids (GSL) with emphasis on the specific gene defects, their biochemical properties, and their expanding clinical spectra. These disorders involve the intersection of two pathways: lipids and carbohydrates. Studies of both pathways were founded on structural biochemistry. Those methods and their more refined and sensitive descendants can both identify the specific genes that cause the disorders and validate the importance of the specific mutations.
Article
Using exome sequencing we identify a homozygous splice site mutation in the PIGN gene in a foetus with multiple congenital anomalies including bilateral diaphragmatic hernia, cardiovascular anomalies, segmental renal dysplasia, facial dysmorphism, cleft palate, and oligodactyly. This finding expands the phenotypic spectrum associated with homozygous loss of function mutations in PIGN, and adds further support for defective GPI anchor biosynthesis as a cause of developmental abnormalities. We demonstrate that exome sequencing is a valuable approach for the identification of a genetic cause in sporadic cases of MCA due to inherited mutations.