Sie sind auf Seite 1von 6

Biochemical and Biophysical Research Communications 361 (2007) 445–450

www.elsevier.com/locate/ybbrc

Null mutations and lethal congenital form of glycogen


storage disease type IV
Stefania Assereto a, Otto P. van Diggelen b, Luisa Diogo c, Eva Morava d,
Denise Cassandrini a, Isabel Carreira c, Willem-Pieter de Boode d, Jildau Dilling d,
Paula Garcia c, Margarida Henriques c, Olinda Rebelo c, Henk ter Laak d,
Carlo Minetti a, Claudio Bruno a,*
a
Muscular and Neurodegenerative Disease Unit, Department of Pediatrics, Istituto Giannina Gaslini,
University of Genova, Largo G. Gaslini 5, I-16147 Genova, Italy
b
Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
c
Unidade de Doencas Metabolicas, Hospital Pediatrico de Coimbra, Coimbra, Portugal
d
Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands

Received 6 July 2007


Available online 24 July 2007

Abstract

Glycogen branching enzyme deficiency (glycogen storage disease type IV, GSD-IV) is a rare autosomal recessive disorder of the gly-
cogen synthesis with high mortality. Two female newborns showed severe hypotonia at birth and both died of cardiorespiratory failure,
at 4 and 12 weeks, respectively. In both patients, muscle biopsies showed deposits of PAS-positive diastase-resistant material and bio-
chemical analysis in cultured fibroblasts showed markedly reduced glycogen branching enzyme activity. Direct sequencing of GBE1 gene
revealed that patient 1 was homozygous for a novel c.691 + 5 g > c in intron 5 (IVS5 + 5 g > c). RT-PCR analysis of GBE1 transcripts
from fibroblasts cDNA showed that this mutation produce aberrant splicing. Patient 2 was homozygous for a novel c.1643G > A muta-
tion leading to a stop at codon 548 in exon 13 (p.W548X). These data underscore that in GSD-IV a severe phenotype correlates with null
mutations, and indicate that RNA analysis is necessary to characterize functional consequences of intronic mutations.
 2007 Published by Elsevier Inc.

Keywords: Glycogen storage disease type IV; Glycogen branching enzyme deficiency; GBE1 gene; Polyglucosan body

Glycogen storage disease type IV (GSD-IV; MIM# The clinical presentation is variable and involves either
232500) is a rare autosomal recessive disorder due to defi- the liver, leading to cirrhosis and death in early childhood,
ciency of the enzyme which catalyzes the last step in glyco- or the neuromuscular system, in its three variants, congen-
gen biosynthesis, the glycogen branching enzyme (GBE; ital, juvenile and adult [1]. The diagnosis is confirmed by
EC 2.4.1.18) [1,2]. Its deficiency produces accumulation the determination of the activity of GBE in affected tissues
of abnormal glycogen consisting of long chains of glucose [3]. The human GBE1 gene, which has been cloned,
units infrequently branched, resembling starch, and known sequenced and localized on chromosome 3p14, codify for
as polyglucosan bodies (PBs). PBs, which stain strongly a 702 amino acids protein [4].
with periodic acid-Schiff, and are partially resistant to dia- Different mutations in the GBE1 gene have been identi-
stase digestion due to their dense packing, accumulate in fied in all different GSD-IV phenotypes [5–14], and in par-
almost all tissues, but to a different degree [1]. ticular, 13 infants with the fatal congenital form have been
characterized genetically in the last few years [5,8–12].
*
Corresponding author. Fax: +39 010 3538265. Here, we report two additional infants with the lethal
E-mail address: claudiobruno@ospedale-gaslini.ge.it (C. Bruno). congenital neuromuscular form of glycogen branching

0006-291X/$ - see front matter  2007 Published by Elsevier Inc.


doi:10.1016/j.bbrc.2007.07.074
446 S. Assereto et al. / Biochemical and Biophysical Research Communications 361 (2007) 445–450

enzyme deficiency, who had novel mutations in the GBE1 and urine organic acids were all normal. Muscle biopsy
gene. showed basophilic intracytoplasmic inclusions in several
fibers deposits (Fig. 1A), and PAS-positive diastase-resis-
Patient reports tant material, compatible with glycogen storage disease.
The baby died at 4 weeks of age due to cardiorespiratory
Patient 1 failure. At autopsy, PAS-positive, diastase-resistant depos-
its were present in almost all tissues (Fig. 1C–F).
This infant girl was the second child of healthy non-con-
sanguineous parents. She was born at 34 weeks of gestation Patient 2
after a pregnancy complicated by polyhydramnios and by
reduced fetal movements. At birth, she presented with This infant girl was the third child of healthy non-con-
severe hypotonia, hyporeflexia, and had to be intubated sanguineous parents. The first two pregnancies resulted in
and mechanically ventilated. In addition, she had equino- healthy siblings. The pregnancy was complicated with
varus feet with flexion contractures. There was no organo- decreased fetal movements and polyhydramnios. She was
megaly. Electrocardiogram and 2D echocardiogram did born at 34 weeks of gestation with a birth weight of
not show any cardiac dysfunctions, and cerebral ultra- 1840 g by caesarean section due to fetal bradycardia.
sound was normal. Extensive laboratory investigations Apgar scores were 1, 3, and 7 at 1, 5, and 10 min on full
did not reveal any abnormalities. Karyotype, screening cardiorespiratory resuscitation, including adrenalin treat-
for metabolic diseases, quantitative serum amino acids ment for severe bradycardia. The patient was immediately

Fig. 1. Hematoxylin and eosin (H&E) staining in muscle biopsies of patient 1 (A) and patient 2 (B). Basophilic intracytoplasmic inclusions in several
fibers. PAS-diastase in liver (C), muscle (D), myocardium (E), and brain (F) of patient 1. PAS-positive and diastase-resistant pale eosinophilic inclusions
(arrows). Original magnification, (A) and (B), 40·; (C) and (E), 400·; (D) and (F), 1000·.
S. Assereto et al. / Biochemical and Biophysical Research Communications 361 (2007) 445–450 447

intubated due to the absence of spontaneous respiratory chemiluminescence detection system (Amersham Pharmacia Biotech). For
activity. The patient had no liver function anomalies, and control protein was used a mouse anti-b-actin monoclonal antibody at a
1:5000 dilution.
no cardiomyopathy was present. The EMG showed no
characteristic alterations or abnormalities comparable to
Werdnig-Hoffman syndrome. Muscle biopsy showed baso- Results
philic intracytoplasmic inclusions in several fibers deposits
(Fig. 1B), and accumulation of periodic acid Schiff-posi- Branching enzyme activity
tive, diastase-resistant storage material. The patient died
at the age of 12 weeks due to cardiorespiratory insuffi- In cultured skin fibroblasts of both patients, GBE activ-
ciency. Autopsy was not performed. ity was less than 5% of normal activity.
Blood, fibroblasts, muscle, and autoptic samples were
collected after fulfilments of our institutional guidelines Molecular analysis
for genetic analysis.
In patient 1, direct sequencing of the promoter region,
Methods the entire coding region sequence, and exon–intron bound-
aries of the GBE1 gene identified an intronic homozygous
Cell culture. Primary cultures of fibroblasts from skin biopsy of g > c transversion 5-bp downstream of exon 5
patients were grown in RPMI 1640 medium containing 20% FCS, 100 U/ (c.691 + 5 g > c, IVS5 + 5 g > c) (Fig. 2A). Both parents
ml penicillin, and 0.1 mg/ml streptomycin. Cells were maintained in an resulted to be heterozygotes for the same mutation.
humidified atmosphere of 5% CO2 at 37 C.
By RT-PCR of exon 3–7 (fragment C), we obtained
Branching enzyme activity. In both patients, GBE activity was assayed
in cultured fibroblasts, as previously described [9]. PCR products with size of 382 and 291-bp, being the
Molecular analysis. Genomic DNA and total RNA were extracted
from the patients’ fibroblasts according to standard protocols or using a
ToTALLY RNA Total RNA Isolation Kit (Ambion, Inc., Austin,
USA), respectively.
For mutation analysis at the genomic DNA level, direct sequencing of
the GBE1 gene was performed after amplification of all exons and flanking
intronic sequences using primer sets and conditions as previously descri-
bed [9].
In addition, mRNA was amplified by RT-PCR using the Superscript
First strand System (Invitrogen Life Technologies, Carlsbad, CA), and
subjected to agarose gel electrophoresis as previously described. The
GBE1 cDNA was amplified in six overlapping fragments (A–F) using the
primers listed in Supplementary Table 1.
The relevant fragments were subcloned using the TOPO TA Cloning
Kit (Invitrogen Life Technologies, Carlsbad, CA) and directly sequenced
with the ABI PRISM 3100 Genetic analyzer (Applied Biosystems, Foster
City, CA).
Sequences of analyzed fragments were compared with the GBE1
cDNA sequence where the A of the ATG translation initiation codon
represents nucleotide +1. The initiation codon is numbered as codon +1.
Nomenclature of mutations follows the recommendations of the
Nomenclature Working Group [15].
To confirm identified mutations and assess parental origin, specific
primer extension techniques were employed using minisequencing analysis
(ABI Prism SNaPshot multiplex kit) [16].
Western blot analysis. Fibroblasts cells of patients and controls were
lysed in 100 ll of RIPA lysis buffer (10 mM Tris–HCl pH 8, 140 mM
NaCl, 1% Triton X-100, 1% Na-desoxycholate, 0.1% SDS, 1 mg/ml
PMSF, 5 ll/ml Protease Inhibitor Cocktail, 1 mM Na3VO4, 1 mM NaF,
and 1 mM EDTA), by sonication and centrifuged at 10,000 rpm for
15 min at 4 C. Supernatants were collected and proteins concentration
was determined by using Bradford protein assay method. Equal amount of
proteins (40 lg) were loaded into each lane and resolved in a 10% SDS–
polyacrylamide gel.
The proteins were blotted onto nitrocellulose membrane (Immobilon
PVDF Millipore).
The membrane was blocked in 5% Non-Fat Dry Milk powder in PBS-
tween 0.1% (PBSt). A Goat anti-rabbit liver GBE polyclonal antibody
(generous gift from Dr. JR Mickelson, University of Minnesota, St. Paul, Fig. 2. (A) Electropherogram of the GBE1 genomic DNA of a normal
MN), diluted 1/400, was used as primary antibody. The secondary anti- control (NC) and of patient 1 (P1), encompassing the exon/intron 5
body was an anti-goat Ig biotinylated from donkey at a 1:750 dilution, and boundary, showing an homozygous mutation (arrow). (B) Electrophero-
an incubation with streptavidin–horseradish peroxidase conjugate (1:6000 gram of the GBE1 genomic DNA of a normal control (NC) and of patient
dilution) was performed [17]. The signal was developed by using the ECL 2 (P2), encompassing exon 13, showing an homozygous mutation (arrow).
448 S. Assereto et al. / Biochemical and Biophysical Research Communications 361 (2007) 445–450

expected size of the wild-type fragment of 518-bp. These Discussion


PCR products have been subcloned and direct sequenced.
Sequencing comparison revealed alternatively spliced prod- GSD-IV is a rare recessive disorder involving the liver or
ucts: a transcript that lacks exon 5, and a transcript lacking the neuromuscular system with different degree of severity
exon 5 plus 6. Of the two mRNA species, the one with skip- depending on the age of onset [1,2].
ping of exon 5 was more prevalent (21 clones against 10 A congenital phenotype can be characterized by severe
clones of the other transcript) (Fig. 3). All other fragments perinatal disorder presenting as fetal akinesia deformation
showed no differences in sequence from that of a normal sequence (FADS) with multiple congenital contractures,
control.
Direct sequencing of patient 2 DNA revealed homozy-
gosity for a G-to-A transition in exon 13 (c.1643G > A), NC1 NC2 P1 P2
leading to a premature stop (p.W548X) (Fig. 2B). By
RT-PCR, mRNA was normally amplified and the muta-
tion was confirmed at cDNA level. Both parents of patient
2 were shown to be carriers of the mutation.
80 kD- GBE
Western blot analysis

To explore the effect on the protein of the identified


mutations in the patients, we performed Western blot β-actin
analysis for glycogen branching enzyme (GBE) from
human fibroblasts of patients and normal controls.
Immunoblot analysis showed that the normal Fig. 4. Immunoblot analysis for glycogen branching enzyme (GBE) on
80.4 kDa protein was absent in both patients, whereas total lysates of human fibroblasts. NC1 and NC2, normal controls; P1,
patient 1; P2, patient 2. In patients 1 and 2, GBE protein levels were
it was present in the wild-type fibroblasts lysates
completely absent. Equal loading control was assessed with an antibody
(Fig. 4). against b-actin (42 kDa).

Fig. 3. Exon skipping caused by the c.691 + 5 g > c (IVS5 + 5 g > c). Fragment C of cDNA from patient 1 (P1) and normal control (NC) fibroblasts were
amplified and analyzed by agarose gel electrophoresis. The PCR products were then subcloned and direct sequenced. The normal sequence for fragment C
of 518-bp is illustrated in (A). Patient 1 showed different bands: (i) a 382-bp band indicative of exon 5 skipping leading to premature termination 7 codon
downstream (B) and (ii) a 291-bp band indicative of exon 5 plus 6 skipping leading to premature termination 6 codon downstream (C).
S. Assereto et al. / Biochemical and Biophysical Research Communications 361 (2007) 445–450 449

hydrops fetalis, and perinatal death [17–20], or by severe phenotype correlates with null mutations. We underline
myopathy, often simulating Werding-Hoffman disease, the importance to evaluate intronic variants as putative
and inconsistently associated with cardiopathy [21,22]. mutations, especially in those cases where the clinical and
Liver involvement usually includes failure to thrive, hepa- biochemical data are strongly diagnostic. Thus, the cDNA
tosplenomegaly, and progressive liver cirrhosis leading to analysis can help to identify unexpected splicing aberra-
death in early childhood [23,24]. The juvenile phenotype tions due to intronic mutation, allowing the genetic
is characterized by myopathy or by cardiopathy, while in diagnosis.
adulthood the defect is associated with central and periph- Finally, because of the high lethality associated with this
eral nervous system dysfunction (adult polyglucosan body disorder, molecular characterization of these patients is of
disease, APBD) [25–29]. particular value for genetic counselling of patients and
Mutations in the GBE1 gene have been identified in all their families.
the different clinical variants, and the same mutation have
been identified in unrelated patients with different clinical Appendix A. Supplementary data
presentations. These data suggest to consider GSD-IV as
a clinical continuum, with different degrees of involvement Supplementary data associated with this article can be
of each organ system, rather than splitting the disease in found, in the online version, at doi:10.1016/j.bbrc.
separate clinical variants, and provide evidence that factors 2007.07.074.
other than the single variants in the GBE1 gene might influ-
ence clinical presentation in the patients. References
To date, in humans, 24 mutations have been described
in the GBE1 gene, distributed throughout the gene [5–14]; [1] S. DiMauro, A.P. Hays, S. Tsujino, Nonlysosomal glycogenoses, in:
Human Gene Mutation Database (http://www.hgmd.cf.a- A.G. Engel, C. Franzini-Armstrong (Eds.), Myology, McGraw-Hill,
c.uk) accessed June 2007] (see Supplementary Table 2). New York, 2004, pp. 1535–1558.
[2] S.W. Moses, R. Parvari, The variable presentations of glycogen
All cases characterized by perinatal death or by fatal infan-
storage disease type IV: a review of clinical, enzymatic and molecular
tile hypotonia have been associated with almost complete studies, Curr. Mol. Med. 2 (2002) 177–188.
absence of GBE activity and with severe mutations in the [3] B.I. Brown, D.H. Brown, Branching enzyme activity of cultured
GBE1 gene [5,8–12]. Reduced enzyme activity and mild amniocytes and chorionic villi: prenatal testing for type IV glycogen
or heterozygous GBE1 mutations result in mild congenital storage disease, Am. J. Hum. Genet. 44 (1989) 378–381.
[4] V.J. Thon, M. Khalil, J.F. Cannon, Isolation of human glycogen
hypotonia and in APBD [6,7,13,14].
branching enzyme cDNAs by screening complementation in yeast, J.
We report two unrelated patients with the lethal congen- Biol. Chem. 268 (1993) 7509–7513.
ital form of GBE deficiency caused by two homozygous [5] Y. Bao, P. Kishnani, J.Y. Wu, Y.T. Chen, Hepatic and neuromus-
null mutations, c.691 + 5 g > c, and c.1643G > A, respec- cular forms of glycogen storage disease type IV caused by mutations
tively. Both mutations have not been previously described in the same glycogen-branching enzyme gene, J. Clin. Invest. 97
(1996) 941–948.
and were not identified in 200 chromosomes from normal
[6] A. Lossos, Z. Meiner, V. Barash, D. Soffer, I. Schlesinger, O.
controls. Abramsky, Z. Argov, S. Shpitzen, V. Meiner, Adult polyglucosan
The intronic base alteration, c.691 + 5 g > c, affect a body disease in Ashkenazi Jewish patients carrying the Tyr329Ser
conserved nucleotide at the donor splice site consensus mutation in the glycogen-branching enzyme gene, Ann. Neurol. 44
sequence (gt[a/g]agt), with a maximal predicted splicing (1998) 867–872.
[7] F. Ziemssen, E. Sindern, J.M. Schroder, Y.S. Shin, J. Zange, M.W.
score of 0.99 [http://www.fruitfly.org/seq_tools/splice.
Kilimann, J.P. Malin, M. Vorgerd, Novel missense mutations in the
html]. glycogen-branching enzyme gene in adult polyglucosan body disease,
To determine the potential pathogenicity of the Ann. Neurol. 47 (2000) 536–540.
c.691 + 5 g > c mutation, we analyzed GBE transcripts [8] M. Nambu, K. Kawabe, T. Fukuda, T.B. Okuno, S. Ohta, I. Nonaka,
from the patient’s fibroblasts, which revealed skipping of H. Sugie, I. Nishino, A neonatal form of glycogen storage disease
type IV, Neurology 61 (2003) 392–394.
exon 5 or skipping of both exons 5 and 6. Skipping of exon
[9] C. Bruno, O.P. van Diggelen, D. Cassandrini, M. Gimpelev, B.
5 (136-bp) leads to a frameshift and premature termination Giuffre, M.A. Donati, P. Introvini, A. Alegria, S. Assereto, L.
7 codons downstream. Skipping of both exons 5 and 6 Morandi, M. Mora, E. Tonoli, S. Mascelli, M. Traverso, E. Pasquini,
(227-bp) leads to a frameshift and premature termination M. Bado, L. Vilarinho, G. van Noort, F. Mosca, S. DiMauro, F.
6 codons downstream. Zara, C. Minetti, Clinical and genetic heterogeneity of branching
enzyme deficiency (glycogenosis type IV), Neurology 63 (2004)
The nonsense mutation, c.1643G > A, cause premature
1053–1058.
termination of translation, generating truncated 548-amino [10] S.K. Tay, H.O. Akman, W.K. Chung, M.G. Pike, F. Muntoni,
acid peptide. A.P. Hays, S. Shanske, S.J. Valberg, J.R. Mickelson, K. Tanji, S.
Both of the reported mutations are therefore predicted DiMauro, Fatal infantile neuromuscular presentation of glycogen
to produce truncated protein. The lack of the proteins storage disease type IV, Neuromuscul. Disord. 14 (2004)
253–260.
showed by Western blot analysis suggest that these proteins
[11] A.R. Janecke, S. Dertinger, U.P. Ketelsen, L. Bereuter, B. Simma, T.
are likely to be degraded. Muller, W. Vogel, F.A. Offner, Neonatal type IV glycogen storage
In conclusion, the identified mutations enlarge the list of disease associated with ‘‘null’’ mutations in glycogen branching
GBE1 mutations, and confirm that in GSD-IV a severe enzyme 1, J. Pediatr. 145 (2004) 705–709.
450 S. Assereto et al. / Biochemical and Biophysical Research Communications 361 (2007) 445–450

[12] A. L’hermine-Coulomb, F. Beuzen, R. Bouvier, M.O. Rolland, R. [21] H. Zellweger, S. Mueller, V. Ionasescu, S.S. Schochet, W.F. McCor-
Froissart, F. Menez, F. Audibert, P. Labrune, Fetal type IV glycogen mick, Glycogenosis IV. A new cause of infantile hypotonia, J. Pediatr.
storage disease: clinical, enzymatic, and genetic data of a pure 80 (1972) 842–844.
muscular form with variable and early antenatal manifestations in the [22] T.T. Tang, A.D. Segura, Y.T. Chen, L.M. Ricci, R.A. Franciosi,
same family, Am. J. Med. Genet. A 139 (2005) 118–122. M.L. Splaingard, M.S. Lubinsky, Neonatal hypotonia and cardio-
[13] E.E. Ubogu, S.T. Hong, H.O. Akman, S. DiMauro, B. Katirji, D.C. myopathy secondary to type IV glycogenosis, Acta Neuropathol.
Preston, B.E. Shapiro, Adult polyglucosan body disease: a case report (Berl.) 87 (1994) 531–536.
of a manifesting heterozygote, Muscle Nerve 32 (2005) 675–681. [23] D.H. Andersen, Familial cirrhosis of the liver with storage of
[14] T.A. Burrow, R.J. Hopkin, K.E. Bove, L. Miles, B.L. Wong, A. abnormal glycogen, Lab. Invest. 5 (1956) 11–20.
Choudhary, D. Bali, S.C. Li, Y.T. Chen, Non-lethal congenital [24] H.L. Greene, B.I. Brown, D.T. McClenahan, R.M. Agostini Jr., S.R.
hypotonia due to glycogen storage disease type IV, Am. J. Med. Taylor, A new variant of type IV glycogenosis: deficiency of
Genet. A 140 (2006) 878–882. branching enzyme activity without apparent progressive liver disease,
[15] J.T. den Dunnen, M.H. Paalman, Standardizing mutation nomen- Hepatology 8 (1988) 302–306.
clature: why bother? Hum. Mutat. 22 (2003) 181–182. [25] S. Servidei, R.E. Riepe, C. Langston, L.Y. Tani, J.T. Bricker, N.
[16] D. Cassandrini, M.G. Calevo, A. Tessa, G. Manfredi, F. Fattori, Crisp-Lindgren, H. Travers, D. Armstrong, S. DiMauro, Severe
M.C. Meschini, R. Carrozzo, E. Tonoli, M. Pedemonte, C. Minetti, cardiopathy in branching enzyme deficiency, J. Pediatr. 111 (1987)
F. Zara, F.M. Santorelli, C. Bruno, A new method for analysis of 51–56.
mitochondrial DNA point mutations and assess levels of hetero- [26] E. Reusche, F. Aksu, H.H. Goebel, Y.S. Shin, T. Yokota, H.
plasmy, Biochem. Biophys. Res. Commun. 342 (2006) 387–393. Reichmann, A mild juvenile variant of type IV glycogenosis, Brain
[17] G. van Noort, W. Straks, O.P. van Diggelen, R.C. Hennekam, A Dev. 14 (1992) 36–43.
congenital variant of glycogenosis type IV, Pediatr. Pathol. 13 (1993) [27] H.H. Goebel, Y.S. Shin, F. Gullotta, T. Yokota, J. Alroy, T. Voit, P.
685–698. Haller, A. Schulz, Adult polyglucosan body myopathy, J. Neuropa-
[18] E. Uro-Coste, M.C. Lelong-Tissier, I. Maire, C. Ceuterick, F. thol. Exp. Neurol. 51 (1992) 24–35.
Chausseray, M.B. Delisle, Congenital variant of type IV glycogenosis. [28] A. Lossos, V. Barash, D. Soffer, Z. Argov, M. Gomori, Z. Ben-
Anatomoclinical report of a case, Ann. Pathol. 16 (1996) 449–452. Nariah, O. Abramsky, I. Steiner, Hereditary branching enzyme
[19] A. Alegria, E. Martins, M. Dias, A. Cunha, M.L. Cardoso, I. Maire, dysfunction in adult polyglucosan body disease: a possible
Glycogen storage disease type IV presenting as hydrops fetalis, J. metabolic cause in two patients, Ann. Neurol. 30 (1991)
Inherit. Metab. Dis. 22 (1999) 330–332. 655–662.
[20] P.M. Cox, L.A. Brueton, K.W. Murphy, V.C. Worthington, P. [29] C. Bruno, S. Servidei, S. Shanske, G. Karpati, S. Carpenter, D.
Bjelogrlic, E.J. Lazda, N.J. Sabire, C.A. Sewry, Early-onset fetal McKee, R.J. Barhon, M. Hirano, Z. Rifai, S. DiMauro, Glycogen
hydrops and muscle degeneration in siblings due to a novel variant of branching enzyme deficiency in adult polyglucosan body disease,
type IV glycogenosis, Am. J. Med. Genet. 86 (1999) 187–193. Ann. Neurol. 33 (1993) 88–93.

Das könnte Ihnen auch gefallen