Skip to main content
Erschienen in: BMC Pediatrics 1/2021

Open Access 01.12.2021 | Case report

Component of oligomeric Golgi complex 1 deficiency leads to hypoglycemia: a case report and literature review

verfasst von: Yizhou Huang, Han Dai, Gangyi Yang, Lili Zhang, Shiyao Xue, Min Zhu

Erschienen in: BMC Pediatrics | Ausgabe 1/2021

Abstract

Background

Congenital disorders of glycosylation (CDG) are a group of metabolic diseases with clinical and genetic heterogeneity, and CDG-IIg is one of the rare reported types of CDG. The aim of this study is to report the clinical manifestations and gene-phenotype characteristics of a rare case of CDG caused by a COG1 gene mutation and review literatures of CDG disease.

Case presentation

The patient was male, and the main clinical symptoms were developmental retardation, convulsion, strabismus, and hypoglycemia, which is rarely reported in CDG-IIg. We treated the patient with glucose infusion and he was recovered from hypoglycemia. Genetic analysis showed that the patient carried the heterozygous intron mutation c.1070 + 3A > G (splicing) in the coding region of the COG1 gene that was inherited from the mother, and the heterozygous mutation c.2492G > A (p. Arg831Gln) in exon 10 of the COG1 gene that was inherited from the father. The genes interacting with COG1 were mainly involved in the transport and composition of the Golgi. The clinical data and laboratory results from a patient diagnosed with CDG-IIg were analyzed, and the causative gene mutation was identified by high-throughput sequencing. The genes and signal pathways related to COG1 were analyzed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses.

Conclusions

The c.2492G > A (p. Arg831Gln) mutation in exon 10 of the COG1 gene may be a potential pathogenetic variant for CDG-IIg. Because of the various manifestations of CDG in clinical practice, multidisciplinary collaboration is important for the diagnosis and treatment of this disease.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12887-021-02922-7.
Yizhou Huang and Han Dai contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CDG
Congenital disorder of glycosylation
COG1 (gene)
Component of oligomeric golgi complex 1
COG (protein)
Conserved oligomeric golgi
SNARE
Soluble NSF attachment protein receptor
NSF
N-ethylmaleimide-sensitive fusion protein
PMM2
Phosphomannomutase-2
MRI
Magnetic resonance imaging
CT
Computed tomography
EEG
Electroencephalogram
DQ
Developmental quotient
SIFT
sorting intolerant from tolerant
PolyPhen_2
Polymorphism phenotyping v2
REVEL
Rare exome variant ensemble learner
HGMD
Human gene mutation database
STRING
Search tool for the retrieval of interacting genes database
PPI
Protein and protein interaction
GO
Gene ontology
KEGG
Kyoto encyclopedia of genes and genomes
BP
Biological processes
MF
Molecular functions
CC
Cellular components
FDR
False discovery rate
ER
Endoplasmic reticulum
SNAP
Soluble NSF attachment proteins
GEARs
A set of COG-sensitive, integral membrane Golgi proteins
PGM1
Phosphoglucomutase-1
IEF
Isoelectric focusing

Background

Congenital disorders of glycosylation (CDG) are a group of metabolic diseases with clinical and genetic heterogeneity [1]. All the CDG types are autosomal recessive, and they are characterized by defects in glycoprotein synthesis, particularly defects in N- and O- terminal glycosylation [2, 3]. Since the first report of CDG by Jaeken et al. in 1980 [1], more than 150 types of CDG have been reported so far. CDG often involves multiple systems, including the cardiovascular system, digestive system, skeletal system, hematopoietic system and central nervous system [2, 4], and has various clinical manifestations. It has been reported that CDG-Ia caused by PMM2 gene deficiency is the most common type of CDG [5].
Conserved oligomeric Golgi (COG) complex is a hetero-octamer complex composed of eight subunits (COG1–8). Its function is to regulate transport in the Golgi in eukaryotic cells and the integrity of the Golgi [6]. The eight subunits of COG are divided into two lobes, A (COG2–4) and B (COG5–7), and two subunits, COG1 and COG8, that bridge the two lobes [69]. CDG-IIg is a rare type of CDG that is caused by COG1 deficiency, and its clinical features are multiple system damage, growth retardation, hypotonia, psychomotor retardation, cerebellar atrophy, and recurrent infections. At present, there are only three reported cases of CDG-IIg [6, 10].
In the current report, the clinical features of a rare case of CDG-IIg caused by COG1 gene mutation are described. Additionally, genetic analysis was performed to identify the associated genes and shed light on the potential underlying mechanism.

Case presentation

A 6-month-old Chinese male child was admitted to our department with recurring episodes of hypoglycemia for 6 months. His parents were healthy and not consanguineous, and his family history was not notable. The child was first admitted to the hospital because of repeated cyanosis and poor reaction at the age of 2 days. He was diagnosed with hypoglycemia based on his blood glucose level (which was 0.3 mmol/L) and recovered after receiving glucose and glucocorticoid infusion.
At the age of 22 days, the infant appeared pale and had a poor reaction and reduced muscle strength after fasting for 4 h for Magnetic resonance imaging (MRI) examination. His blood glucose was 2.06 mmol/L and he was recovered from glucose infusion again. Subsequently, the child showed increased muscle tension and movement retardation.
At the age of 4 months, the child experienced repeated vomiting and diarrhea for one day, and hypoglycemia reappeared (blood glucose, 4 mmol/L). In addition, the child had an episode of convulsions that lasted for 2–3 min. After intravenous infusion of glucose, blood glucose returned to normal.
At the age of 6 months, the child was admitted to the hospital for a systematic examination. Physical examination at admission showed that his body weight was 6600 g (3th centile); body length, 76.0 cm (97th centile); head circumference, 41.0 cm (3th centile); chest circumference, 43.0 cm (50th centile). He was conscious and had a good mental reaction, nutrition levels, and skin elasticity. He had strabismus in both eyes, but it was prominent in the left eye. He was unable to use the support of his elbows to get up from a reclining position, and when he stood upright, his head was tilted to one side. He could not turn over or bear his weight. He leaned forward fully in the sitting position and flexed his legs in the standing position. He was not actively conscious of grasping objects. He could keep holding objects, but he could not change objects with his hands. He could pronounce vowels, but he was insensitive to locate the sources of sound or light. He had normal muscular strength in both upper limbs, but greater muscular strength in both lower limbs, mainly in the adductor muscles. The testes were not completely lowered into the scrotum, and the result of the transillumination test was negative.
The results of laboratory tests, including blood routine (platelet count, 435 × 10 9/L; hemoglobin, 107 g/L), thyroid function, biochemical tests, stool and urine routine, and blood lactic acid, were normal. In order to rule out other possible causes of hypoglycemia, we had conducted a fasting test. The test, sustained for 4 h, was considered completed when the blood glucose level of patient indicated the hypoglycemia. The results showed that both insulin and cortisol were within the normal range, which could exclude cortisol deficiency and hyperinsulinemia. (Table 1) Computed tomography (CT) of the brain indicated that the brain parenchyma was scattered with low-density shadows, and the bilateral ventricles appeared plump.
Table 1
The biochemical measurements of fasting study
Characteristics
Results
Range
FPG (mmol/l)
2.2
2.2–7.0
COR (nmol/l)
425
138–698
INS (mIU/l)
0.7
<28.4
FPG fasting plasm glucose, COR cortisol, INS insulin
MRI of the brain indicated extensive abnormal signals in both cerebral hemispheres. This was considered to be indicative of atrophic cystic changes caused by encephalomalacia. Electroencephalogram (EEG) showed that multiple spikes/sharp-slow waves were synchronously or asynchronously discharged in the left and right occipital regions during both waking and sleep. A 2–3 Hz increase in δ activity was found in the occipital region, and the dominant rhythm of the occipital region was not significant in the waking state. Echocardiography was indicative of patent ductus arteriosus. The Gesell Developmental Schedules showed that the gross motor skills of the patient at 4 weeks and 3 months were equivalent to those of neonates at 0 months, and the fine motor skills were equivalent to those of neonates at 0–1 month. The Gesell Intelligence Scale showed that the ability to respond to people and the environment was equivalent to that of infants at 6 weeks (Developmental Quotient [DQ] = 35 points), while the ability to respond to physical objects was equivalent to that of infants at 3 weeks (DQ = 18 points). In addition, the patient’s gross motor skills were equivalent to that of infants at 0 weeks (DQ = 0 points); his fine motor skills were equivalent to that of infants at 2 weeks (DQ = 12 points), and the ability to communicate was equivalent to that of infants at 6 weeks (DQ = 35 points).
The study was carried out in accordance with the Declaration of Helsinki of the World.
Medical Association and was approved by the Committee of Medical Ethics of The Second Affiliated Hospital of Chongqing Medical University. Informed consent was obtained from parents. A high-throughput sequencing and genetic analysis, and Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses had been used.
After obtaining the parents’ consent and signatures on the informed consent form, blood samples were taken for a whole exon sequencing analysis. The results revealed the heterozygous intron mutation c.1070 + 3A > G (splicing) in the coding region of the COG1 gene that was inherited from the mother, and the heterozygous mutation c.2492 g > A (p. Arg831Gln) in exon 10 of the COG1 gene that was inherited from the father.
The coding region of c.1070 + 3A > G in the COG1 gene could not be predicted by the Sorting Intolerant From Tolerant (SIFT), Polymorphism Phenotyping v2 (PolyPhen_2), Rare Exome Variant Ensemble Learner (REVEL), and Mutation Taster software [6, 10]. For the coding region of the COG1 gene, c.2492G > A (p.Arg831Gln), the results from the protein function prediction software SIFT, PolyPhen_2, REVEL revealed that it was destructive, unknown, or benign, and has not been reported in Human Gene Mutation Database (HGMD). The prediction software Mutation Taster indicated that it was a pathogenic mutation [11]. The results of other prediction software are shown in Table 2 [1217]. The secondary and tertiary structures of COG1-encoded proteins were predicted by the Phyre2 software (Fig. 1) [18]. Based on the identified heterozygous mutation in exon 10 of the COG1 gene and the patient’s clinical manifestations, we speculated that CDG-IIg in the patient may be caused by the compound heterozygous mutation.
Table 2
The predictions for COG1 mutations using various software
Software
c.1070 + 3A > G (splicing)
c.2492G > A (p.Arg831Gln)
VarSome
Likely Benign
Uncertain significance
DANN
Score:0.8675
Score:0.9912
dbscSNV
ADA Score:0.001334
N/A
Mutation Taster
N/A
Disease-causing
Mutation assessor
N/A
Medium
FATHMM-MKL
N/A
Damaging
LRT
N/A
Deleterious
DEOGEN2
N/A
Tolerated
EIGEN
N/A
Pathogenic
EIGEN PC
N/A
Pathogenic
SIFT
N/A
Damaging
SIFT4G
N/A
Damaging
c. 1070 + 3A > G, the nucleotide of 1070 + 3 in the coding region was mutated from adenine to guanine; c.2492G > A, the nucleotide of 2492 in the coding region was mutated from guanine to adenine; p.Arg831Gln, amino acid 831 changed from arginine to glutamine
To explore the relationship between COG1 and related genes signal pathways, we used the Search Tool for the Retrieval of Interacting Genes database (STRING, 11.0 version) [19]. We constructed the protein and protein interaction (PPI) network of COG1 by using the search tool of the STRING database. An interaction score of 0.9 and a maximum interaction number of 50 were considered as the cut-off criteria (An additional figure file shows this in more detail (see Additional file 1)). The number of genes interact with will be more if the degree of a gene is larger. In the current study, we rank the genes from high to low. High-degree genes in the top fourteen and INS gene were identified as the core genes. We used core genes to perform enrichment analysis and constructed PPI network again. The PPI network is shown in Fig. 2a.
GO and KEGG enrichment analyses were performed using the Cluster profiler package [20]. The biological processes (BP), molecular functions (MF), and cellular components (CC) associated with these genes were identified, and p values <0.05 were considered to indicate statistical significance. In addition, we used the STRING database to perform Reactome pathway enrichment analysis [21]. A false discovery rate (FDR) of <0.05 was considered to indicate statistical significance. The results of the GO analysis are shown in Fig. 2c. With regard to BP, the COG1-related proteins were involved in Golgi vesicle transport, intracellular transport, protein transport, ER-to-Golgi-vesicle-mediated transport, and intra-Golgi vesicle-mediated transport. With regard to CC, the interacting proteins were predominantly enriched in the Golgi membrane, Golgi subcompartment, Golgi apparatus part, trans−Golgi network membrane and Golgi transport complex. With regard to MF, the interacting proteins were involved in soluble N-ethylmaleimide-sensitive fusion attachment proteins (SNAP) receptor activity, soluble NSF protein attachment protein receptor (SNARE) binding and syntaxin binding. The results of KEGG analysis showed that COG1 was mainly enriched in SNARE interactions in vesicular transport (Fig. 2d). To further identify the signal pathways related to COG1, we used the Reactome pathway database for screening, with FDR < 0.05 as the search condition. The top six identified items are shown in Fig. 2b. The identified proteins were involved in intra-Golgi traffic, COPI-mediated anterograde transport, Membrane Trafficking, Retrograde transport at the Trans-Golgi-Network, COPII-mediated vesicle transport, and Cargo concentration in the ER.

Discussion and Conclusions

In this report, we describe a rare case of CDG-IIg that was associated with a compound heterozygous autosomal recessive COG1 mutation. To the best of our knowledge, there are no other reports of CDG-IIg cases in China. Three cases have been reported in other countries, and two were caused by an intron mutation in the COG1 gene.
The patient in the present report had recurrent cyanosis, poor reaction, and hypoglycemia on the second day after birth. During hospitalization, the blood glucose was too low to be detected, and the patient was diagnosed with hypoglycemia. After discharge from the hospital, the patient had several episodes of hypoglycemia and gradually began to exhibit clinical manifestations of CDG-IIg, such as lower limb muscle strength enhancement and retardation, and epilepsy. Gene analysis confirmed that the patient had a compound heterozygous mutation in the COG1 gene. Therefore, the diagnosis of CDG-IIg was confirmed. Further spectrum analysis showed that the CDG-IIg had an autosomal recessive inheritance pattern.
In eukaryotic cells, proteins have biological roles after glycosylation. Glycoproteins are composed of glucose chains and polypeptides, which can be divided into O-glycosidic bonds (that bind the serine or threonine residues of proteins) or N-glycosidic bonds (that bind the asparagine residues of proteins). At present, most types of CDG are caused by protein N-glycosylation disorder, while a few are caused by protein O-glycosylation disorder [22]. Glycosylation of N-glycosidic bonds has an important impact on the post-translational modification and function of many biological enzymes [23]. Therefore, the clinical manifestations of CDG are various. It may involve multiple organs and systems or only a single organ, and may even present with no obvious clinical manifestations. That is, the severity of CDG varies. At present, there are about 150 types of CDG, most of which exhibit neuromuscular involvement, including global developmental delay, speech delay and so on [24] CDG-I is mainly caused by a deficiency in polysaccharide synthesis or transfer in the cytoplasm and endoplasmic reticulum (ER). CDG-II is mainly caused by processing and maturation defects of the ER and Golgi polysaccharides [6]. (Fig. 3, drawn by Cytoscape [25]).
In CDG-IIg disease, Oka et al. [26] found that the steady-state level of the GEARs (a set of COG-sensitive, integral membrane Golgi proteins, including mannosidase II, GOS-28, GS15, GPP130, CASP, giantin, and golgin-84) subset is dependent on the root complex. The loss of any subunit in the COG complex may change the level of GEARs and their distribution in the Golgi complex, and this results in the biological synthesis of polysaccharides [7]. The COG1 gene, located on chromosome 17 (17q25.1), contains 14 exons and encodes 980 amino acids. In patients with COG1 deficiency, the levels of α-mannosidase II and β-1,4-galactosyltransferase I are significantly decreased in the Golgi matrix [6]. Any defect of the COG1 subunit will affect the function of the COG complex, thus destroying the balance of GEAR transportation and dislocation in the glycosylation process [27]. This pathological change maybe the root pathogeny for COG1. However, the connection between this pathological change and symptoms is still unclear.
In the present report, the results of bioinformatics analysis indicated that the proteins interacting with COG1 were mainly enriched in various transport pathways of the Golgi; this is consistent with the findings of previous reports. We suspect the impact of COG1 mutation was displayed on the disorder of Golgi transport function. Meanwhile, we also speculate that COG1 gene mutations may directly or indirectly affect cell biological processes through other genes, and this may result in the occurrence of hereditary disease. In the PPI network, we observed a relationship between COG1 and insulin. Therefore, we speculated that the observed hypoglycemia may be related to the promotion of insulin secretion by the COG1 mutation. A search of the literature showed that there were early reports of hypoglycemia in patients with congenital glycation disorders. It was described as hyperinsulinemic hypoglycemia and was the main symptom of CDG 1b patients [28]. In addition, hypoglycemia has also been reported in patients with CDG 1a and CDG 1d [29, 30]. Because of two obvious symptoms including hyperinsulinemic and hypoglycemia, reasonable conjecture, defective glycosylation of sulphonylurea receptor, was put forward due to the confirmation of sulphonylurea receptor to be a glycoprotein [29]. However, the exact mechanism of hypoglycemia in CDG patients is still unclear. In addition, patients with PGM1-CDG (PGM1, phosphoglucomutase-1) have also been reported to have hypoglycemia [31]. PGM1 is a key enzyme in glycolysis and glycogen production. Hypoglycemia in patients with PGM1 deficiency may be related to insufficient glycogen release or abnormal glycosylation of proteins [32]. However, the patient in the present case was diagnosed with CDG-IIg and had normal serum insulin levels. Therefore, there may be other mechanisms involved in the occurrence of hypoglycemia in this case except for the connection with INS. Further studies involving basic research are still needed to verify our conjecture.
In two of the previously reported cases of CDG-IIg, the first patient’s brain MRI showed mega-cisterna Magna and a hypoplastic vermis. The clinical manifestations included microcephaly, bilateral macular lesions, a short neck, and widely spaced nipples. The second patient showed atrophy of the temporal cortex and floculonodular lobe of the cerebellum, together with dilatation of the lateral ventricle and fourth ventricle and cisterna magna. The clinical manifestations were shortening of the long bones, dysmorphic facial features, ulnar deviation of the fingers, thoracic scoliosis, hypospadias-I, and left-side cryptorchidism. In another reported case, the patient showed mild left ventricular hypertrophy with abnormality of diastolic function of the left ventricle. The clinical manifestations were generalized hypotonia and growth retardation. In the present case, the patient mainly showed growth retardation, convulsion, and hypoglycemia, which is a new symptom of CDG-IIg. Because CDG is extremely rare, its diagnosis depends on clinicians’ awareness and vigilance. When the above clinical manifestations, especially those involving the nervous system, cannot be explained by other diseases, the possibility of CDG should be considered [24].
At present, serum transferrin isoelectric focusing (IEF) is still a common method for the diagnosis of CDG, but the IEF results of some CDG patients can be normal. Gene technology (whole exon and genome sequencing), enzyme analysis, and glycomics are emerging as reliable methods in the diagnosis of CDG [33, 34]. In the present case, CDG was diagnosed based on high-throughput sequencing and genetic analysis. With regard to treatment, no specific treatment for CDG-IIg has been reported so far, and symptomatic treatment is widely used in the clinic. In the present patient, intravenous infusion of glucose solution was used to correct hypoglycemia. What’s more, body rehabilitation was also applied for the improvement of growth retardation due to the hereditary nature of this disease. Similarly, the treatment of specific symptoms has been reported for other types of CDG. PGM1-CDG patients can be treated with oral galactose or nutritional therapy, which is known to be effective [35]. Additionally, stem cell transplantation can correct neutropenia and lymphopenia in patients with PGM3-CDG (PGM3, phosphoglucomutase 3) [36], and patients with phosphomannose isomerase deficiency (MPI-CDG) can be treated by mannose and liver transplantation [37]. However, CDG is a chronic genetic disease that often involves multiple organs and systems. Therefore, patients with CDG need to be managed and treated systematically. In addition, recombinant enzymes, drug chaperones, active sugar compounds, and gene therapy might have potential as effective therapeutic methods in the future [1, 38]. Importantly, genetic screening is performed in the families of patients to ensure early and rapid diagnosis and, thus, improve prognosis. Furthermore, multidisciplinary collaboration is important for the diagnosis and treatment of this disease.

Acknowledgements

Not applicable.

Declarations

We have obtained the parents’ consent and signatures on the informed consent form. The study was carried out in accordance with the Declaration of Helsinki of the World and was approved by the Committee of Medical Ethics of The Second Affiliated Hospital of Chongqing Medical University.
The consent for publication from the patient’s parents had been acquired. The informed consent form with parents’ signatures was obtained from the patients’ parents.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Verheijen J, Tahata S, Kozicz T, Witters P, Morava E. Therapeutic approaches in Congenital Disorders of Glycosylation (CDG) involving N-linked glycosylation: an update. Genet Med. 2020;22:268–79.CrossRef Verheijen J, Tahata S, Kozicz T, Witters P, Morava E. Therapeutic approaches in Congenital Disorders of Glycosylation (CDG) involving N-linked glycosylation: an update. Genet Med. 2020;22:268–79.CrossRef
2.
Zurück zum Zitat Footitt EJ, Karimova A, Burch M, Yayeh T, Dupré T, Vuillaumier-Barrot S, et al. Cardiomyopathy in the congenital disorders of glycosylation (CDG): a case of late presentation and literature review. J Inherit Metab Dis. 2009;32:313–9.CrossRef Footitt EJ, Karimova A, Burch M, Yayeh T, Dupré T, Vuillaumier-Barrot S, et al. Cardiomyopathy in the congenital disorders of glycosylation (CDG): a case of late presentation and literature review. J Inherit Metab Dis. 2009;32:313–9.CrossRef
3.
Zurück zum Zitat Freeze HH. Update and perspectives on congenital disorders of glycosylation. Glycobiology. 2001;11:129R–43R.CrossRef Freeze HH. Update and perspectives on congenital disorders of glycosylation. Glycobiology. 2001;11:129R–43R.CrossRef
4.
Zurück zum Zitat Coman D, Irving M, Kannu P, Jaeken J, Savarirayan R. The skeletal manifestations of the congenital disorders of glycosylation. Clin Genet. 2008;73:507–15.CrossRef Coman D, Irving M, Kannu P, Jaeken J, Savarirayan R. The skeletal manifestations of the congenital disorders of glycosylation. Clin Genet. 2008;73:507–15.CrossRef
5.
Zurück zum Zitat Lao JP, DiPrimio N, Prangley M, Sam FS, Mast JD, Perlstein EO. Yeast Models Of Phosphomannomutase 2 Deficiency, A Congenital Disorder Of Glycosylation. G3. 2019;9:413–23.CrossRef Lao JP, DiPrimio N, Prangley M, Sam FS, Mast JD, Perlstein EO. Yeast Models Of Phosphomannomutase 2 Deficiency, A Congenital Disorder Of Glycosylation. G3. 2019;9:413–23.CrossRef
6.
Zurück zum Zitat Foulquier F, Vasile E, Schollen E, Callewaert N, Raemaekers T, Quelhas D, et al. Conserved oligomeric Golgi complex subunit 1 deficiency reveals a previously uncharacterized congenital disorder of glycosylation type II. Proc Natl Acad Sci U S A. 2006;103:3764–9.CrossRef Foulquier F, Vasile E, Schollen E, Callewaert N, Raemaekers T, Quelhas D, et al. Conserved oligomeric Golgi complex subunit 1 deficiency reveals a previously uncharacterized congenital disorder of glycosylation type II. Proc Natl Acad Sci U S A. 2006;103:3764–9.CrossRef
7.
Zurück zum Zitat Jaeken J. Congenital disorders of glycosylation. Ann N Y Acad Sci. 2010;1214:190–8.CrossRef Jaeken J. Congenital disorders of glycosylation. Ann N Y Acad Sci. 2010;1214:190–8.CrossRef
8.
Zurück zum Zitat Oka T, Vasile E, Penman M, Novina CD, Dykxhoorn DM, Ungar D, et al. Genetic Analysis of the Subunit Organization and Function of the Conserved Oligomeric Golgi (COG) Complex. J Biol Chem. 2005;280:32736–45.CrossRef Oka T, Vasile E, Penman M, Novina CD, Dykxhoorn DM, Ungar D, et al. Genetic Analysis of the Subunit Organization and Function of the Conserved Oligomeric Golgi (COG) Complex. J Biol Chem. 2005;280:32736–45.CrossRef
9.
Zurück zum Zitat Ungar D, Oka T, Vasile E, Krieger M, Hughson FM. Subunit architecture of the conserved oligomeric Golgi complex. J Biol Chem. 2005;280:32729–35.CrossRef Ungar D, Oka T, Vasile E, Krieger M, Hughson FM. Subunit architecture of the conserved oligomeric Golgi complex. J Biol Chem. 2005;280:32729–35.CrossRef
10.
Zurück zum Zitat Zeevaert R, Foulquier F, Dimitrov B, Reynders E, Van Damme-Lombaerts R, Simeonov E, et al. Cerebrocostomandibular-like syndrome and a mutation in the conserved oligomeric Golgi complex, subunit 1. Hum Mol Genet. 2008;18:517–24.CrossRef Zeevaert R, Foulquier F, Dimitrov B, Reynders E, Van Damme-Lombaerts R, Simeonov E, et al. Cerebrocostomandibular-like syndrome and a mutation in the conserved oligomeric Golgi complex, subunit 1. Hum Mol Genet. 2008;18:517–24.CrossRef
11.
Zurück zum Zitat Lubeck E, Coskun AF, Zhiyentayev T, Ahmad M, Cai L. Single-cell in situ RNA profiling by sequential hybridization. Nat Methods. 2014;11:360–1.CrossRef Lubeck E, Coskun AF, Zhiyentayev T, Ahmad M, Cai L. Single-cell in situ RNA profiling by sequential hybridization. Nat Methods. 2014;11:360–1.CrossRef
12.
Zurück zum Zitat Kopanos C, Tsiolkas V, Kouris A, Chapple CE, Albarca AM, Meyer R, et al. VarSome: the human genomic variant search engine. Bioinformatics. 2019;35:1978–80.CrossRef Kopanos C, Tsiolkas V, Kouris A, Chapple CE, Albarca AM, Meyer R, et al. VarSome: the human genomic variant search engine. Bioinformatics. 2019;35:1978–80.CrossRef
13.
Zurück zum Zitat Quang D, Chen Y, Xie X. DANN: a deep learning approach for annotating the pathogenicity of genetic variants. Bioinformatics. 2015;31:761–3.CrossRef Quang D, Chen Y, Xie X. DANN: a deep learning approach for annotating the pathogenicity of genetic variants. Bioinformatics. 2015;31:761–3.CrossRef
14.
Zurück zum Zitat Jian X, Boerwinkle E, Liu X. In silico prediction of splice-altering single nucleotide variants in the human genome. Nucleic Acids Res. 2014;42:13534–44.CrossRef Jian X, Boerwinkle E, Liu X. In silico prediction of splice-altering single nucleotide variants in the human genome. Nucleic Acids Res. 2014;42:13534–44.CrossRef
15.
Zurück zum Zitat Dong C, Wei P, Jian X, Gibbs R, Boerwinkle E, Wang K, et al. Comparison and integration of deleteriousness prediction methods for nonsynonymous SNVs in whole exome sequencing studies. Hum Mol Genet. 2015;24:2125–37.CrossRef Dong C, Wei P, Jian X, Gibbs R, Boerwinkle E, Wang K, et al. Comparison and integration of deleteriousness prediction methods for nonsynonymous SNVs in whole exome sequencing studies. Hum Mol Genet. 2015;24:2125–37.CrossRef
16.
Zurück zum Zitat Liu X, Wu C, Li C, Boerwinkle E. dbNSFP v3.0: A One-Stop Database of Functional Predictions and Annotations for Human Nonsynonymous and Splice-Site SNVs. Hum Mutat. 2016;37:235–41.CrossRef Liu X, Wu C, Li C, Boerwinkle E. dbNSFP v3.0: A One-Stop Database of Functional Predictions and Annotations for Human Nonsynonymous and Splice-Site SNVs. Hum Mutat. 2016;37:235–41.CrossRef
17.
Zurück zum Zitat Liu X, Jian X, Boerwinkle E. dbNSFP: A lightweight database of human nonsynonymous SNPs and their functional predictions. Hum Mutat. 2011;32:894–9.CrossRef Liu X, Jian X, Boerwinkle E. dbNSFP: A lightweight database of human nonsynonymous SNPs and their functional predictions. Hum Mutat. 2011;32:894–9.CrossRef
18.
Zurück zum Zitat Kelley LA, Mezulis S, Yates CM, Wass MN, Sternberg MJE. The Phyre2 web portal for protein modeling, prediction and analysis. Nat Protoc. 2015;10:845–58.CrossRef Kelley LA, Mezulis S, Yates CM, Wass MN, Sternberg MJE. The Phyre2 web portal for protein modeling, prediction and analysis. Nat Protoc. 2015;10:845–58.CrossRef
19.
Zurück zum Zitat Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, et al. The STRING database in 2017: quality-controlled protein–protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45:D362–8.CrossRef Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, et al. The STRING database in 2017: quality-controlled protein–protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45:D362–8.CrossRef
20.
Zurück zum Zitat Yu G, Wang L, Han Y, He Q. clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters. OMICS A J Integr Biol. 2012;16:284–7.CrossRef Yu G, Wang L, Han Y, He Q. clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters. OMICS A J Integr Biol. 2012;16:284–7.CrossRef
21.
Zurück zum Zitat Jassal B, Matthews L, Viteri G, Gong C, Lorente P, Fabregat A, et al. The reactome pathway knowledgebase. Nucleic Acids Res. 2020;48:D498–503.PubMed Jassal B, Matthews L, Viteri G, Gong C, Lorente P, Fabregat A, et al. The reactome pathway knowledgebase. Nucleic Acids Res. 2020;48:D498–503.PubMed
22.
Zurück zum Zitat Höck M, Wegleiter K, Ralser E, Kiechl-Kohlendorfer U, Scholl-Bürgi S, Fauth C, et al. ALG8-CDG: novel patients and review of the literature. Orphanet J Rare Dis. 2015;10:73.CrossRef Höck M, Wegleiter K, Ralser E, Kiechl-Kohlendorfer U, Scholl-Bürgi S, Fauth C, et al. ALG8-CDG: novel patients and review of the literature. Orphanet J Rare Dis. 2015;10:73.CrossRef
23.
Zurück zum Zitat Shental-Bechor D, Levy Y. Folding of glycoproteins: toward understanding the biophysics of the glycosylation code. Curr Opin Struct Biol. 2009;19:524–33.CrossRef Shental-Bechor D, Levy Y. Folding of glycoproteins: toward understanding the biophysics of the glycosylation code. Curr Opin Struct Biol. 2009;19:524–33.CrossRef
24.
Zurück zum Zitat Barone R, Fiumara A, Jaeken J. Congenital Disorders of Glycosylation with Emphasis on Cerebellar Involvement. Semin Neurol. 2014;34:357–66.CrossRef Barone R, Fiumara A, Jaeken J. Congenital Disorders of Glycosylation with Emphasis on Cerebellar Involvement. Semin Neurol. 2014;34:357–66.CrossRef
25.
Zurück zum Zitat Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13:2498–504.CrossRef Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13:2498–504.CrossRef
26.
Zurück zum Zitat Oka T, Ungar D, Hughson FM, Krieger M. The COG and COPI complexes interact to control the abundance of GEARs, a subset of Golgi integral membrane proteins. Mol Biol Cell. 2004;15:2423–35.CrossRef Oka T, Ungar D, Hughson FM, Krieger M. The COG and COPI complexes interact to control the abundance of GEARs, a subset of Golgi integral membrane proteins. Mol Biol Cell. 2004;15:2423–35.CrossRef
27.
Zurück zum Zitat Zeevaert R, Foulquier F, Jaeken J, Matthijs G. Deficiencies in subunits of the Conserved Oligomeric Golgi (COG) complex define a novel group of Congenital Disorders of Glycosylation. Mol Genet Metab. 2008;93:15–21.CrossRef Zeevaert R, Foulquier F, Jaeken J, Matthijs G. Deficiencies in subunits of the Conserved Oligomeric Golgi (COG) complex define a novel group of Congenital Disorders of Glycosylation. Mol Genet Metab. 2008;93:15–21.CrossRef
28.
Zurück zum Zitat Babovic-Vuksanovic D, Patterson MC, Schwenk WF, O'Brien JF, Vockley J, Freeze HH, et al. Severe hypoglycemia as a presenting symptom of carbohydrate-deficient glycoprotein syndrome. J Pediatr. 1999;135:775–81.CrossRef Babovic-Vuksanovic D, Patterson MC, Schwenk WF, O'Brien JF, Vockley J, Freeze HH, et al. Severe hypoglycemia as a presenting symptom of carbohydrate-deficient glycoprotein syndrome. J Pediatr. 1999;135:775–81.CrossRef
29.
Zurück zum Zitat Bohles H, Sewell AA, Gebhardt B, Reinecke-Luthge A, Kloppel G, Marquardt T. Hyperinsulinaemic hypoglycaemia--leading symptom in a patient with congenital disorder of glycosylation Ia (phosphomannomutase deficiency). J Inherit Metab Dis. 2001;24:858–62.CrossRef Bohles H, Sewell AA, Gebhardt B, Reinecke-Luthge A, Kloppel G, Marquardt T. Hyperinsulinaemic hypoglycaemia--leading symptom in a patient with congenital disorder of glycosylation Ia (phosphomannomutase deficiency). J Inherit Metab Dis. 2001;24:858–62.CrossRef
30.
Zurück zum Zitat Sun L, Eklund EA, Chung WK, Wang C, Cohen J, Freeze HH. Congenital Disorder of Glycosylation Id Presenting with Hyperinsulinemic Hypoglycemia and Islet Cell Hyperplasia. J Clin Endocrinol Metab. 2005;90:4371–5.CrossRef Sun L, Eklund EA, Chung WK, Wang C, Cohen J, Freeze HH. Congenital Disorder of Glycosylation Id Presenting with Hyperinsulinemic Hypoglycemia and Islet Cell Hyperplasia. J Clin Endocrinol Metab. 2005;90:4371–5.CrossRef
31.
Zurück zum Zitat Tegtmeyer LC, Rust S, van Scherpenzeel M, Ng BG, Losfeld ME, Timal S, et al. Multiple phenotypes in phosphoglucomutase 1 deficiency. N Engl J Med. 2014;370:533–42.CrossRef Tegtmeyer LC, Rust S, van Scherpenzeel M, Ng BG, Losfeld ME, Timal S, et al. Multiple phenotypes in phosphoglucomutase 1 deficiency. N Engl J Med. 2014;370:533–42.CrossRef
32.
Zurück zum Zitat Morava E. Galactose supplementation in phosphoglucomutase-1 deficiency; review and outlook for a novel treatable CDG. Mol Genet Metab. 2014;112:275–9.CrossRef Morava E. Galactose supplementation in phosphoglucomutase-1 deficiency; review and outlook for a novel treatable CDG. Mol Genet Metab. 2014;112:275–9.CrossRef
33.
Zurück zum Zitat Jaeken J, Péanne R. What is new in CDG? J Inherit Metab Dis. 2017;40:569–86.CrossRef Jaeken J, Péanne R. What is new in CDG? J Inherit Metab Dis. 2017;40:569–86.CrossRef
34.
Zurück zum Zitat Fang J, Peters V, Körner C, Hoffmann GF. Improvement of CDG diagnosis by combined examination of several glycoproteins. J Inherit Metab Dis. 2004 2004-01-01;27:581–90.CrossRef Fang J, Peters V, Körner C, Hoffmann GF. Improvement of CDG diagnosis by combined examination of several glycoproteins. J Inherit Metab Dis. 2004 2004-01-01;27:581–90.CrossRef
35.
Zurück zum Zitat Radenkovic S, Bird MJ, Emmerzaal TL, Wong SY, Felgueira C, Stiers KM, et al. The Metabolic Map into the Pathomechanism and Treatment of PGM1-CDG. Am J Hum Genet. 2019;104:835–46.CrossRef Radenkovic S, Bird MJ, Emmerzaal TL, Wong SY, Felgueira C, Stiers KM, et al. The Metabolic Map into the Pathomechanism and Treatment of PGM1-CDG. Am J Hum Genet. 2019;104:835–46.CrossRef
36.
Zurück zum Zitat Stray-Pedersen A, Backe PH, Sorte HS, Mørkrid L, Chokshi NY, Erichsen HC, et al. PGM3 Mutations Cause a Congenital Disorder of Glycosylation with Severe Immunodeficiency and Skeletal Dysplasia. Am J Hum Genet. 2014;95:96–107.CrossRef Stray-Pedersen A, Backe PH, Sorte HS, Mørkrid L, Chokshi NY, Erichsen HC, et al. PGM3 Mutations Cause a Congenital Disorder of Glycosylation with Severe Immunodeficiency and Skeletal Dysplasia. Am J Hum Genet. 2014;95:96–107.CrossRef
37.
Zurück zum Zitat Janssen MC, de Kleine RH, van den Berg AP, Heijdra Y, van Scherpenzeel M, Lefeber DJ, et al. Successful liver transplantation and long-term follow-up in a patient with MPI-CDG. Pediatrics. 2014;134:e279–83.CrossRef Janssen MC, de Kleine RH, van den Berg AP, Heijdra Y, van Scherpenzeel M, Lefeber DJ, et al. Successful liver transplantation and long-term follow-up in a patient with MPI-CDG. Pediatrics. 2014;134:e279–83.CrossRef
38.
Zurück zum Zitat Ramírez AS, Boilevin J, Lin C, Ha Gan B, Janser D, Aebi M, et al. Chemo-enzymatic synthesis of lipid-linked GlcNAc2Man5 oligosaccharides using recombinant Alg1, Alg2 and Alg11 proteins. Glycobiology. 2017;27:726–33.CrossRef Ramírez AS, Boilevin J, Lin C, Ha Gan B, Janser D, Aebi M, et al. Chemo-enzymatic synthesis of lipid-linked GlcNAc2Man5 oligosaccharides using recombinant Alg1, Alg2 and Alg11 proteins. Glycobiology. 2017;27:726–33.CrossRef
Metadaten
Titel
Component of oligomeric Golgi complex 1 deficiency leads to hypoglycemia: a case report and literature review
verfasst von
Yizhou Huang
Han Dai
Gangyi Yang
Lili Zhang
Shiyao Xue
Min Zhu
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
BMC Pediatrics / Ausgabe 1/2021
Elektronische ISSN: 1471-2431
DOI
https://doi.org/10.1186/s12887-021-02922-7

Weitere Artikel der Ausgabe 1/2021

BMC Pediatrics 1/2021 Zur Ausgabe

Neuer Typ-1-Diabetes bei Kindern am Wochenende eher übersehen

23.04.2024 Typ-1-Diabetes Nachrichten

Wenn Kinder an Werktagen zum Arzt gehen, werden neu auftretender Typ-1-Diabetes und diabetische Ketoazidosen häufiger erkannt als bei Arztbesuchen an Wochenenden oder Feiertagen.

Neue Studienergebnisse zur Myopiekontrolle mit Atropin

22.04.2024 Fehlsichtigkeit Nachrichten

Augentropfen mit niedrig dosiertem Atropin können helfen, das Fortschreiten einer Kurzsichtigkeit bei Kindern zumindest zu verlangsamen, wie die Ergebnisse einer aktuellen Studie mit verschiedenen Dosierungen zeigen.

Spinale Muskelatrophie: Neugeborenen-Screening lohnt sich

18.04.2024 Spinale Muskelatrophien Nachrichten

Seit 2021 ist die Untersuchung auf spinale Muskelatrophie Teil des Neugeborenen-Screenings in Deutschland. Eine Studie liefert weitere Evidenz für den Nutzen der Maßnahme.

Fünf Dinge, die im Kindernotfall besser zu unterlassen sind

18.04.2024 Pädiatrische Notfallmedizin Nachrichten

Im Choosing-Wisely-Programm, das für die deutsche Initiative „Klug entscheiden“ Pate gestanden hat, sind erstmals Empfehlungen zum Umgang mit Notfällen von Kindern erschienen. Fünf Dinge gilt es demnach zu vermeiden.

Update Pädiatrie

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