Entry - *191350 - DOLICHYL-PHOSPHATE N-ACETYLGLUCOSAMINE PHOSPHOTRANSFERASE; DPAGT1 - OMIM
* 191350

DOLICHYL-PHOSPHATE N-ACETYLGLUCOSAMINE PHOSPHOTRANSFERASE; DPAGT1


Alternative titles; symbols

UDP-GlcNAc:DOLICHYL-PHOSPHATE N-ACETYLGLUCOSAMINEPHOSPHOTRANSFERASE
DPAGT2
GlcNAc-1-P TRANSFERASE


HGNC Approved Gene Symbol: DPAGT1

Cytogenetic location: 11q23.3     Genomic coordinates (GRCh38): 11:119,093,874-119,101,853 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
11q23.3 Congenital disorder of glycosylation, type Ij 608093 AR 3
Myasthenic syndrome, congenital, 13, with tubular aggregates 614750 AR 3

TEXT

Description

N-linked glycosylation is initiated in all eukaryotic cells with the synthesis of lipid-linked oligosaccharides in a cyclic pathway, the dolichol cycle. DPAGT1 (EC 2.7.8.15) catalyzes the first step in the dolichol cycle, the synthesis of N-acetylglucosaminyl-pyrophosphoryldolichol (GlcNAc-PP-dolichol) from dolichol phosphate and UDP-GlcNAc, and can be inhibited by the antibiotic tunicamycin (Eckert et al., 1998).


Cloning and Expression

Rajput et al. (1992) isolated mRNA for the Dpagt1 protein from mouse mammary glands. The mouse cDNA recognized a single mRNA species of about 2 kb in mouse mammary glands when used as a probe in Northern blot analysis.

Eckert et al. (1998) cloned a human DPAGT1 cDNA from a human lung fibroblast cDNA library. The cDNA encodes a deduced 400-amino acid protein with a calculated molecular mass of 44.7 kD. DPAGT1 contains an N-terminal signal peptide, 2 potential dolichol-binding sequences, and 4 sites for N-glycosylation. It shares 93% amino acid homology with hamster Dpagt, including 100% identity in the dolichol-binding region, and 42% homology with S. cerevisiae GlcNAc-1-P transferase.


Gene Function

Protein asparagine-linked glycosylation is a multistep process that is divided into 2 stages. The first stage consists of the synthesis of the lipid-linked oligosaccharide precursor (LLO) and its en bloc transfer to nascent polypeptides in the lumen of the endoplasmic reticulum. This process requires at least 34 genes, of which DPAGT1 is the first. The second stage involves the processing of protein-bound oligosaccharides and requires at least an additional 20 genes to form a bi-antennary sugar chain typical of plasma glycoproteins. Genetic defects in some of these genes, including DPAGT1, cause severe multisystem disorders called congenital disorders of glycosylation (CDGs) (Freeze, 2001)

Eckert et al. (1998) demonstrated that S. cerevisiae expressing recombinant DPAGT1 synthesized GlcNAc- and GlcNAc(2)-PP-dolichol. Expression of human DPAGT1 also complemented a conditional lethal S. cerevisiae strain defective for GlcNAc-1-P transferase. Expression of recombinant DPAGT1 from a multicopy expression vector also conferred a higher tolerance toward tunicamycin due to elevated enzyme synthesis, thus showing a gene dosage effect.


Biochemical Features

Dong et al. (2018) determined the crystal structures of human DPAGT1 and DPAGT1 in complex with UDP-GlcNAc or tunicamycin at 3.1- to 3.6-angstrom resolution. DPAGT1 exists predominantly as a noncovalent dimer in solution, and dimerization is important for its stability. DPAGT1 consists of 10 transmembrane helices (TMHs) with both termini in the endoplasmic reticulum (ER) lumen. The active site is on the cytoplasmic face of the membrane, formed by 4 of the 5 cytoplasmic loops between the TMHs. Three loops are on the ER side of the membrane, and 1 is embedded in the membrane on the ER side. Formation of the DPAGT1-UDP-GlcNAc complex stabilizes the active site of DPAGT1. The authors determined that missense mutations in DPAGT1 alter DPAGT1 function via diverse mechanisms. Structural analysis of the DPAGT1-tunicamycin complex suggested that tunicamycin inhibits DPAGT1 through partial mimicry of the complex formed during catalysis between acceptor phospholipid Dol-P and UDP-GlcNAc. The authors designed semisynthetic and lipid-altered tunicamycin analogs that retained antimicrobial activity but no longer inhibited DPAGT1, thereby circumventing toxicity to eukaryotic cells. These tunicamycin analogs could reduce intracellular bacterial burdens with nanomolar antimicrobial potency and no signs of toxicity, providing leads for tuberculosis antibiotic development.


Mapping

Using FISH and somatic cell hybrid analysis, Smith et al. (1993) mapped the DPAGT1 gene (D11S366) to chromosome 11q23.3.

Using a panel of mouse/hamster somatic cell hybrids and a specific probe derived from the 3-prime noncoding region of the mouse cDNA, Rajput et al. (1992) mapped the mouse Dpagt1 gene to chromosome 17.


Animal Model

Marek et al. (1999) found that Dpagt1-null mice died 4 to 5 days postfertilization, just after implantation, suggesting that DPAGT1 function and protein N-glycosylation are essential in early embryogenesis.


Molecular Genetics

Congenital Disorder of Glycosylation Type Ij

In a patient with CDG Ij (CDGIJ; 608093), Wu et al. (2003) identified a tyr170-to-cys mutation (Y170C; 191350.0001) in the DPAGT1 gene.

Timal et al. (2012) identified compound heterozygosity for 2 mutations in the DPAGT1 gene (191350.0007 and 191350.0008) in a Caucasian boy with CDG Ij. The mutations were found by exome sequencing and confirmed by Sanger sequencing.

In 2 sibs, born of consanguineous Turkish parents, with severe CDG Ij, Wurde et al. (2012) identified a homozygous mutation in the DPAGT1 gene (A114G; 191350.0009). The mutation was found by homozygosity mapping followed by candidate gene sequencing. The unaffected parents were heterozygous for the mutation, which was not found in 100 control alleles of the same ethnic background. RT-PCR analysis of patient cells showed that the mutation also increased the amount of normal aberrant splicing seen in controls, resulting in the skipping of exons 2/3 and a truncated protein. In vitro functional expression assays showed decreased DPAGT1 activity, at 18% of normal values. The patients had a severe disorder characterized by hyperexcitability, intractable seizures, bilateral cataracts, nystagmus, strabismus, and progressive microcephaly. Both died within their first year of life from cardiorespiratory failure.

In a Pakistani brother and sister, born of unrelated patients with a mild from of CDG Ij, Iqbal et al. (2013) identified compound heterozygous mutations in the DPAGT1 gene (I29F; 191350.0010 and L168P; 191350.0011). The mutations were found by exome sequencing, confirmed by Sanger sequencing, segregated with the disorder, and occurred at highly conserved residues. Neither was present in over 200 ethnically matched chromosomes or in the dbSNP, 1000 Genomes Project, or Exome Variant Server databases. Functional studies of the variants were not performed. The patients had normal psychomotor development until ages 2 and 5 years, respectively, when they both developed seizures, hypotonia, and aggressive behavior. As adults, they had moderate intellectual disability, poor speech, aggressive behavior, hypotonia, seizures, and mild facial dysmorphic features.

Congenital Myasthenic Syndrome 13

In 5 patients from 4 families with congenital myasthenic syndrome-13 (CMS13; 614750) with tubular aggregates, Belaya et al. (2012) identified 7 different mutations in the DPAGT1 gene (see, e.g., 191350.0002-191350.0006). All mutations were in the compound heterozygous state. The first 4 mutations were identified by exome sequencing of 2 unrelated patients and were confirmed by Sanger sequencing. The mutations segregated with the disorder in those families with available material. Analyses of motor endplates from 2 patients showed a severe reduction of endplate acetylcholine receptors (AChR). In vitro studies showed that DPAGT1 is required for efficient glycosylation of AChR subunits and for efficient export of AChR receptors to the cell surface. The findings demonstrated the importance of N-linked protein glycosylation for proper functioning of the neuromuscular junction, and suggested that the primary pathogenic mechanism of DPAGT1 mutations is reduced levels of AChR at the endplate region. Laboratory studies of 2 patients showed abnormal glycosylation of transferrin, consistent with a functional defect of DPAGT1. Belaya et al. (2012) postulated that the defect in glycosylation of certain proteins may lead to misfolding and aggregation in the sarcoplasmic reticulum, resulting in formation of tubular aggregates within muscle tissue.


Nomenclature

GPT has been used as an abbreviation for this enzyme, but this runs the risk of confusion with glutamate-pyruvate transaminase (GPT; 138200).


ALLELIC VARIANTS ( 11 Selected Examples):

.0001 CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, TYR170CYS
  
RCV000013090...

In a patient with central disorder of glycosylation type Ij (CDG1J; 608093), Wu et al. (2003) identified reduced DPAGT1 enzymatic activity; sequencing of genomic DNA and cDNAs of the DPAGT1 gene identified, in the paternal allele, a 660A-G transition in exon 5, resulting in a tyr170-to-cys (Y170C) mutation. Although no mutation was identified in the maternal allele, it produced only 12% of the normal amount of mature mRNA; the remainder showed a complex exon skipping pattern that shifted the reading frame and resulted in a truncated nonfunctional protein. The patient had developed infantile spasms at the age of 4 months within 72 hours of receiving DPT immunization. Development was significantly delayed in all aspects with microcephaly, arched palate, micrognathia, and exotropia. She also had fifth finger clinodactyly, single flexion creases of the hands, and skin dimples on the upper thighs. She had severe hypotonia and medically intractable seizures, and at 6 years of age had minimal speech. Abnormal isoelectric focusing pattern of serum transferrin was consistent with the diagnosis of type I CDG.


.0002 MYASTHENIC SYNDROME, CONGENITAL, 13

DPAGT1, VAL117ILE
  
RCV000030601...

In a patient with congenital myasthenic syndrome-13 (CMS13; 614750), Belaya et al. (2012) identified compound heterozygosity for 2 mutations in the DPAGT1 gene: a 349G-A transition resulting in a val117-to-ile (V117I) substitution, and a 324G-C transversion resulting in a met108-to-ile (M108I; 191350.0003) substitution. Another patient was compound heterozygous for V117I and a 1-bp duplication (c.699dup; 191350.0004), resulting in a frameshift, premature termination (Thr234HisfsTer116), and nonsense-mediated mRNA decay. The mutations were found by exome sequencing and confirmed by Sanger sequencing. The 324G-C mutation was found in 2 (0.0186%) of 10,758 control alleles from the general population and 1 (0.0142%) of 7,020 alleles in the European American population. None of the other mutations were found in controls. The patients had onset at age 2.5 and 7 years, respectively, of difficulty walking due to proximal muscle weakness, and showed a favorable response to pyridostigmine. Muscle biopsy showed reduced levels of endplate acetylcholine receptors (AChR). In vitro functional expression studies showed that the c.699dup mutation was unable to restore normal levels of glycosylated AChR in HEK293 cells with DPAGT1 inhibition.


.0003 MYASTHENIC SYNDROME, CONGENITAL, 13

DPAGT1, MET108ILE
  
RCV000030602...

For discussion of the met108-to-ile (M108I) mutation in the DPAGT1 gene that was found in compound heterozygous state in a patient with congenital myasthenic syndrome-13 (CMS13; 614750) by Belaya et al. (2012), see 191350.0002.


.0004 MYASTHENIC SYNDROME, CONGENITAL, 13

DPAGT1, 1-BP DUP, NT699
  
RCV000030603...

For discussion of the 1-bp duplication in the DPAGT1 gene (699dup) that was found in compound heterozygous state in a patient with congenital myasthenic syndrome-13 (CMS13; 614750) by Belaya et al. (2012), see 191350.0002.


.0005 MYASTHENIC SYNDROME, CONGENITAL, 13

DPAGT1, LEU120MET
  
RCV000030604

In 2 sibs with congenital myasthenic syndrome-13 (CMS13; 614750), Belaya et al. (2012) identified compound heterozygosity for 2 mutations in the DPAGT1 gene: a 358C-A transversion resulting in a leu120-to-met (L120M) substitution, and a 791T-G transversion resulting in a val264-to-gly (V264G; 191350.0006) substitution. The patients had onset in the first year of life of hypotonia, poor head control, and delayed motor development. They showed some improvement in muscle power during the teenage years, and both showed a response to pyridostigmine.


.0006 MYASTHENIC SYNDROME, CONGENITAL, 13

DPAGT1, VAL264GLY
  
RCV000030605

For discussion of the val264-to-gly (V264G) mutation in the DPAGT1 gene that was found in compound heterozygous state in patients with congenital myasthenic syndrome-13 (CMS13; 614750) by Belaya et al. (2012), see 191350.0005.


.0007 CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, ILE69ASN
  
RCV000032992

In a patient with congenital disorder of glycosylation type Ij (CDGIJ; 608093), Timal et al. (2012) identified compound heterozygosity for 2 mutations in the DPAGT1 gene: a 206T-A transversion in exon 2 resulting in an ile69-to-asn (I69N) substitution at a highly conserved residue in the highly conserved dolichol recognition motif, and a G-to-A transition in intron 1 (161+5G-A; 191350.0008), which resulted in degradation of the mutant mRNA. The mutations were found by exome sequencing and confirmed by Sanger sequencing. Each unaffected parent was heterozygous for 1 of the mutations. In patient-derived cells, the formation of GlcNAc-PP-dolichol was reduced to 22% of controls. The patient had multisystem problems, including asphyxia at birth, respiratory insufficiency, frequent apneas, jaundice, nuclear cataracts, cryptorchidism, dysmorphic features, hypertonia of the limbs, joint contractures, tremor, and feeding difficulties. Laboratory studies showed chronic anemia, hypoproteinemia, increased liver enzymes, and coagulation defects.


.0008 CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, IVS1DS, G-A, +5
  
RCV000032993

For discussion of the splice site mutation in the DPAGT1 gene (161+5G-A) that was found in compound heterozygous state in a patient with congenital disorder of glycosylation type Ij (CDGIJ; 608093) by Timal et al. (2012), see 191350.0007.


.0009 CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, ALA114GLY
  
RCV000055659...

In 2 sibs, born of consanguineous Turkish parents, with congenital disorder of glycosylation type Ij (CDGIJ; 608093), Wurde et al. (2012) identified a homozygous c.341C-G transversion in exon 3 of the DPAGT1 gene, resulting in an ala114-to-gly (A114G) substitution. The mutation was found by homozygosity mapping followed by candidate gene sequencing. The unaffected parents were heterozygous for the mutation, which was not found in 100 control alleles of the same ethnic background. RT-PCR of patient cells showed that the mutation also increased the normal aberrant splicing seen in controls, resulting in the skipping of exons 2/3 and a truncated protein. In vitro functional expression assays showed decreased DPAGT1 activity, at 18% of normal values. The patients had a severe disorder characterized by hyperexcitability, intractable seizures, bilateral cataracts, nystagmus, strabismus, and progressive microcephaly. Both died within their first year of life from cardiorespiratory failure.


.0010 CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, ILE29PHE
  
RCV000055660...

In 2 Pakistani sibs, born of unrelated parents, with a relatively mild form of congenital disorder of glycosylation type Ij (CDGIJ; 608093), Iqbal et al. (2013) identified compound heterozygosity for 2 mutations in the DPAGT1 gene: a c.85A-T transition resulting in an ile29-to-phe (I29F) substitution, and a c.503T-C transition resulting in a leu168-to-pro (L168P; 191350.0011) substitution. The mutations were found by exome sequencing of 1 of the patients and confirmed by Sanger sequencing in both patients. The mutations segregated with the disorder and occurred at highly conserved residues. Neither was present in over 200 ethnically matched chromosomes or in the dbSNP, 1000 Genomes Project, or Exome Variant Server databases. Functional studies of the variants were not performed.


.0011 CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, LEU168PRO
  
RCV000055661

For discussion of the leu168-to-pro (L168P) mutation in the DPAGT1 gene that was found in compound heterozygous state in patients with congenital disorder of glycosylation type Ij (CDGIJ; 608093) by Iqbal et al. (2013), see 191350.0010.


REFERENCES

  1. Belaya, K., Finlayson, S., Slater, C. R., Cossins, J., Liu, W. W., Maxwell, S., McGowan, S. J., Maslau, S., Twigg, S. R. F., Walls, T. J., Pascual Pascual, S. I., Palace, J., Beeson, D. Mutations in DPAGT1 cause a limb-girdle congenital myasthenic syndrome with tubular aggregates. Am. J. Hum. Genet. 91: 193-201, 2012. [PubMed: 22742743, images, related citations] [Full Text]

  2. Dong, Y. Y., Wang, H., Pike, A. C. W., Cochrane, S. A., Hamedzadeh, S., Wyszynski, F. J., Bushell, S. R., Royer, S. F., Widdick, D. A., Sajid, A., Boshoff, H. I., Park, Y., and 20 others. Structures of DPAGT1 explain glycosylation disease mechanisms and advance TB antibiotic design. Cell 175: 1045-1058, 2018. [PubMed: 30388443, images, related citations] [Full Text]

  3. Eckert, V., Blank, M., Mazhari-Tabrizi, R., Mumberg, D., Funk, M., Schwarz, R. T. Cloning and functional expression of the human GlcNAc-1-P transferase, the enzyme for the committed step of the dolichol cycle, by heterologous complementation in Saccharomyces cerevisiae. Glycobiology 8: 77-85, 1998. [PubMed: 9451016, related citations] [Full Text]

  4. Freeze, H. H. Update and perspectives on congenital disorders of glycosylation. Glycobiology 11: 129R-143R, 2001. [PubMed: 11805072, related citations]

  5. Iqbal, Z., Shahzad, M., Vissers, L. E. L. M., van Scherpenzeel, M., Gilissen, C., Razzaq, A., Zahoor, M. Y., Khan, S. N., Kleefstra, T., Veltman, J. A., de Brouwer, A. P. M., Lefeber, D. J., van Bokhoven, H., Riazuddin, S. A compound heterozygous mutation in DPAGT1 results in a congenital disorder of glycosylation with a relatively mild phenotype. Europ. J. Hum. Genet. 21: 844-849, 2013. [PubMed: 23249953, images, related citations] [Full Text]

  6. Marek, K. W., Vijay, I. K., Marth, J. D. A recessive deletion in the GlcNAc-1-phosphotransferase gene results in peri-implantation embryonic lethality. Glycobiology 9: 1263-1271, 1999. [PubMed: 10536042, related citations] [Full Text]

  7. Rajput, B., Ma, J., Muniappa, N., Schantz, L., Naylor, S. L., Lalley, P. A., Vijay, I. K. Mouse UDP-GlcNAc:dolichyl-phosphate N-acetylglucosaminephosphotransferase: molecular cloning of the cDNA, generation of anti-peptide antibodies and chromosomal localization. Biochem. J. 285: 985-992, 1992. [PubMed: 1323278, related citations] [Full Text]

  8. Smith, M. W., Clark, S. P., Hutchinson, J. S., Wei, Y. H., Churukian, A. C., Daniels, L. B., Diggle, K. L., Gen, M. W., Romo, A. J., Lin, Y., Selleri, L., Mcelligott, D. L., Evans, G. A. A sequence-tagged site map of human chromosome 11. Genomics 17: 699-725, 1993. [PubMed: 8244387, related citations] [Full Text]

  9. Timal, S., Hoischen, A., Lehle, L., Adamowicz, M., Huijben, K., Sykut-Cegielska, J., Paprocka, J., Jamroz, E., van Spronsen, F. J., Korner, C., Gilissen, C., Rodenburg, R. J., Eidhof, I., Van den Heuvel, L., Thiel, C., Wevers, R. A., Morava, E., Veltman, J., Lefeber, D. J. Gene identification in the congenital disorders of glycosylation type I by whole-exome sequencing. Hum. Molec. Genet. 21: 4151-4161, 2012. [PubMed: 22492991, related citations] [Full Text]

  10. Wu, X., Rush, J. S., Karaoglu, D., Krasnewich, D., Lubinsky, M. S., Waechter, C. J., Gilmore, R., Freeze, H. H. Deficiency of UDP-GlcNAc:dolichol phosphate N-acetylglucosamine-1 phosphate transferase (DPAGT1) causes a novel congenital disorder of glycosylation type Ij. Hum. Mutat. 22: 144-150, 2003. [PubMed: 12872255, related citations] [Full Text]

  11. Wurde, A. E., Reunert, J., Rust, S., Hertzberg, C., Haverkamper, S., Nurnberg, G., Nurnberg, P., Lehle, L., Rossi, R., Marquardt, T. Congenital disorder of glycosylation type Ij (CDG-Ij, DPAGT1-CDG): extending the clinical and molecular spectrum of a rare disease. Molec. Genet. Metab. 105: 634-641, 2012. [PubMed: 22304930, related citations] [Full Text]


Bao Lige - updated : 04/05/2019
Cassandra L. Kniffin - updated : 9/18/2013
Cassandra L. Kniffin - updated : 11/8/2012
Cassandra L. Kniffin - updated : 8/1/2012
Patricia A. Hartz - updated : 11/17/2003
Victor A. McKusick - updated : 9/9/2003
Creation Date:
Victor A. McKusick : 11/10/1992
carol : 03/04/2022
mgross : 04/05/2019
carol : 04/12/2017
carol : 03/27/2017
alopez : 04/29/2015
carol : 4/27/2015
ckniffin : 4/21/2015
mcolton : 4/20/2015
carol : 10/15/2013
carol : 9/26/2013
tpirozzi : 9/26/2013
tpirozzi : 9/26/2013
ckniffin : 9/18/2013
carol : 9/17/2013
terry : 11/8/2012
carol : 11/8/2012
ckniffin : 11/8/2012
carol : 8/2/2012
ckniffin : 8/1/2012
joanna : 1/13/2011
terry : 4/4/2005
mgross : 11/17/2003
tkritzer : 9/26/2003
carol : 9/26/2003
tkritzer : 9/12/2003
tkritzer : 9/9/2003
carol : 8/19/1998
mark : 12/22/1997
carol : 11/10/1992

* 191350

DOLICHYL-PHOSPHATE N-ACETYLGLUCOSAMINE PHOSPHOTRANSFERASE; DPAGT1


Alternative titles; symbols

UDP-GlcNAc:DOLICHYL-PHOSPHATE N-ACETYLGLUCOSAMINEPHOSPHOTRANSFERASE
DPAGT2
GlcNAc-1-P TRANSFERASE


HGNC Approved Gene Symbol: DPAGT1

SNOMEDCT: 725079003;  


Cytogenetic location: 11q23.3     Genomic coordinates (GRCh38): 11:119,093,874-119,101,853 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
11q23.3 Congenital disorder of glycosylation, type Ij 608093 Autosomal recessive 3
Myasthenic syndrome, congenital, 13, with tubular aggregates 614750 Autosomal recessive 3

TEXT

Description

N-linked glycosylation is initiated in all eukaryotic cells with the synthesis of lipid-linked oligosaccharides in a cyclic pathway, the dolichol cycle. DPAGT1 (EC 2.7.8.15) catalyzes the first step in the dolichol cycle, the synthesis of N-acetylglucosaminyl-pyrophosphoryldolichol (GlcNAc-PP-dolichol) from dolichol phosphate and UDP-GlcNAc, and can be inhibited by the antibiotic tunicamycin (Eckert et al., 1998).


Cloning and Expression

Rajput et al. (1992) isolated mRNA for the Dpagt1 protein from mouse mammary glands. The mouse cDNA recognized a single mRNA species of about 2 kb in mouse mammary glands when used as a probe in Northern blot analysis.

Eckert et al. (1998) cloned a human DPAGT1 cDNA from a human lung fibroblast cDNA library. The cDNA encodes a deduced 400-amino acid protein with a calculated molecular mass of 44.7 kD. DPAGT1 contains an N-terminal signal peptide, 2 potential dolichol-binding sequences, and 4 sites for N-glycosylation. It shares 93% amino acid homology with hamster Dpagt, including 100% identity in the dolichol-binding region, and 42% homology with S. cerevisiae GlcNAc-1-P transferase.


Gene Function

Protein asparagine-linked glycosylation is a multistep process that is divided into 2 stages. The first stage consists of the synthesis of the lipid-linked oligosaccharide precursor (LLO) and its en bloc transfer to nascent polypeptides in the lumen of the endoplasmic reticulum. This process requires at least 34 genes, of which DPAGT1 is the first. The second stage involves the processing of protein-bound oligosaccharides and requires at least an additional 20 genes to form a bi-antennary sugar chain typical of plasma glycoproteins. Genetic defects in some of these genes, including DPAGT1, cause severe multisystem disorders called congenital disorders of glycosylation (CDGs) (Freeze, 2001)

Eckert et al. (1998) demonstrated that S. cerevisiae expressing recombinant DPAGT1 synthesized GlcNAc- and GlcNAc(2)-PP-dolichol. Expression of human DPAGT1 also complemented a conditional lethal S. cerevisiae strain defective for GlcNAc-1-P transferase. Expression of recombinant DPAGT1 from a multicopy expression vector also conferred a higher tolerance toward tunicamycin due to elevated enzyme synthesis, thus showing a gene dosage effect.


Biochemical Features

Dong et al. (2018) determined the crystal structures of human DPAGT1 and DPAGT1 in complex with UDP-GlcNAc or tunicamycin at 3.1- to 3.6-angstrom resolution. DPAGT1 exists predominantly as a noncovalent dimer in solution, and dimerization is important for its stability. DPAGT1 consists of 10 transmembrane helices (TMHs) with both termini in the endoplasmic reticulum (ER) lumen. The active site is on the cytoplasmic face of the membrane, formed by 4 of the 5 cytoplasmic loops between the TMHs. Three loops are on the ER side of the membrane, and 1 is embedded in the membrane on the ER side. Formation of the DPAGT1-UDP-GlcNAc complex stabilizes the active site of DPAGT1. The authors determined that missense mutations in DPAGT1 alter DPAGT1 function via diverse mechanisms. Structural analysis of the DPAGT1-tunicamycin complex suggested that tunicamycin inhibits DPAGT1 through partial mimicry of the complex formed during catalysis between acceptor phospholipid Dol-P and UDP-GlcNAc. The authors designed semisynthetic and lipid-altered tunicamycin analogs that retained antimicrobial activity but no longer inhibited DPAGT1, thereby circumventing toxicity to eukaryotic cells. These tunicamycin analogs could reduce intracellular bacterial burdens with nanomolar antimicrobial potency and no signs of toxicity, providing leads for tuberculosis antibiotic development.


Mapping

Using FISH and somatic cell hybrid analysis, Smith et al. (1993) mapped the DPAGT1 gene (D11S366) to chromosome 11q23.3.

Using a panel of mouse/hamster somatic cell hybrids and a specific probe derived from the 3-prime noncoding region of the mouse cDNA, Rajput et al. (1992) mapped the mouse Dpagt1 gene to chromosome 17.


Animal Model

Marek et al. (1999) found that Dpagt1-null mice died 4 to 5 days postfertilization, just after implantation, suggesting that DPAGT1 function and protein N-glycosylation are essential in early embryogenesis.


Molecular Genetics

Congenital Disorder of Glycosylation Type Ij

In a patient with CDG Ij (CDGIJ; 608093), Wu et al. (2003) identified a tyr170-to-cys mutation (Y170C; 191350.0001) in the DPAGT1 gene.

Timal et al. (2012) identified compound heterozygosity for 2 mutations in the DPAGT1 gene (191350.0007 and 191350.0008) in a Caucasian boy with CDG Ij. The mutations were found by exome sequencing and confirmed by Sanger sequencing.

In 2 sibs, born of consanguineous Turkish parents, with severe CDG Ij, Wurde et al. (2012) identified a homozygous mutation in the DPAGT1 gene (A114G; 191350.0009). The mutation was found by homozygosity mapping followed by candidate gene sequencing. The unaffected parents were heterozygous for the mutation, which was not found in 100 control alleles of the same ethnic background. RT-PCR analysis of patient cells showed that the mutation also increased the amount of normal aberrant splicing seen in controls, resulting in the skipping of exons 2/3 and a truncated protein. In vitro functional expression assays showed decreased DPAGT1 activity, at 18% of normal values. The patients had a severe disorder characterized by hyperexcitability, intractable seizures, bilateral cataracts, nystagmus, strabismus, and progressive microcephaly. Both died within their first year of life from cardiorespiratory failure.

In a Pakistani brother and sister, born of unrelated patients with a mild from of CDG Ij, Iqbal et al. (2013) identified compound heterozygous mutations in the DPAGT1 gene (I29F; 191350.0010 and L168P; 191350.0011). The mutations were found by exome sequencing, confirmed by Sanger sequencing, segregated with the disorder, and occurred at highly conserved residues. Neither was present in over 200 ethnically matched chromosomes or in the dbSNP, 1000 Genomes Project, or Exome Variant Server databases. Functional studies of the variants were not performed. The patients had normal psychomotor development until ages 2 and 5 years, respectively, when they both developed seizures, hypotonia, and aggressive behavior. As adults, they had moderate intellectual disability, poor speech, aggressive behavior, hypotonia, seizures, and mild facial dysmorphic features.

Congenital Myasthenic Syndrome 13

In 5 patients from 4 families with congenital myasthenic syndrome-13 (CMS13; 614750) with tubular aggregates, Belaya et al. (2012) identified 7 different mutations in the DPAGT1 gene (see, e.g., 191350.0002-191350.0006). All mutations were in the compound heterozygous state. The first 4 mutations were identified by exome sequencing of 2 unrelated patients and were confirmed by Sanger sequencing. The mutations segregated with the disorder in those families with available material. Analyses of motor endplates from 2 patients showed a severe reduction of endplate acetylcholine receptors (AChR). In vitro studies showed that DPAGT1 is required for efficient glycosylation of AChR subunits and for efficient export of AChR receptors to the cell surface. The findings demonstrated the importance of N-linked protein glycosylation for proper functioning of the neuromuscular junction, and suggested that the primary pathogenic mechanism of DPAGT1 mutations is reduced levels of AChR at the endplate region. Laboratory studies of 2 patients showed abnormal glycosylation of transferrin, consistent with a functional defect of DPAGT1. Belaya et al. (2012) postulated that the defect in glycosylation of certain proteins may lead to misfolding and aggregation in the sarcoplasmic reticulum, resulting in formation of tubular aggregates within muscle tissue.


Nomenclature

GPT has been used as an abbreviation for this enzyme, but this runs the risk of confusion with glutamate-pyruvate transaminase (GPT; 138200).


ALLELIC VARIANTS 11 Selected Examples):

.0001   CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, TYR170CYS
SNP: rs28934876, gnomAD: rs28934876, ClinVar: RCV000013090, RCV000694750, RCV001291042, RCV001567586

In a patient with central disorder of glycosylation type Ij (CDG1J; 608093), Wu et al. (2003) identified reduced DPAGT1 enzymatic activity; sequencing of genomic DNA and cDNAs of the DPAGT1 gene identified, in the paternal allele, a 660A-G transition in exon 5, resulting in a tyr170-to-cys (Y170C) mutation. Although no mutation was identified in the maternal allele, it produced only 12% of the normal amount of mature mRNA; the remainder showed a complex exon skipping pattern that shifted the reading frame and resulted in a truncated nonfunctional protein. The patient had developed infantile spasms at the age of 4 months within 72 hours of receiving DPT immunization. Development was significantly delayed in all aspects with microcephaly, arched palate, micrognathia, and exotropia. She also had fifth finger clinodactyly, single flexion creases of the hands, and skin dimples on the upper thighs. She had severe hypotonia and medically intractable seizures, and at 6 years of age had minimal speech. Abnormal isoelectric focusing pattern of serum transferrin was consistent with the diagnosis of type I CDG.


.0002   MYASTHENIC SYNDROME, CONGENITAL, 13

DPAGT1, VAL117ILE
SNP: rs387907243, ClinVar: RCV000030601, RCV001852609

In a patient with congenital myasthenic syndrome-13 (CMS13; 614750), Belaya et al. (2012) identified compound heterozygosity for 2 mutations in the DPAGT1 gene: a 349G-A transition resulting in a val117-to-ile (V117I) substitution, and a 324G-C transversion resulting in a met108-to-ile (M108I; 191350.0003) substitution. Another patient was compound heterozygous for V117I and a 1-bp duplication (c.699dup; 191350.0004), resulting in a frameshift, premature termination (Thr234HisfsTer116), and nonsense-mediated mRNA decay. The mutations were found by exome sequencing and confirmed by Sanger sequencing. The 324G-C mutation was found in 2 (0.0186%) of 10,758 control alleles from the general population and 1 (0.0142%) of 7,020 alleles in the European American population. None of the other mutations were found in controls. The patients had onset at age 2.5 and 7 years, respectively, of difficulty walking due to proximal muscle weakness, and showed a favorable response to pyridostigmine. Muscle biopsy showed reduced levels of endplate acetylcholine receptors (AChR). In vitro functional expression studies showed that the c.699dup mutation was unable to restore normal levels of glycosylated AChR in HEK293 cells with DPAGT1 inhibition.


.0003   MYASTHENIC SYNDROME, CONGENITAL, 13

DPAGT1, MET108ILE
SNP: rs376039938, gnomAD: rs376039938, ClinVar: RCV000030602, RCV001224025, RCV003144114

For discussion of the met108-to-ile (M108I) mutation in the DPAGT1 gene that was found in compound heterozygous state in a patient with congenital myasthenic syndrome-13 (CMS13; 614750) by Belaya et al. (2012), see 191350.0002.


.0004   MYASTHENIC SYNDROME, CONGENITAL, 13

DPAGT1, 1-BP DUP, NT699
SNP: rs397515321, gnomAD: rs397515321, ClinVar: RCV000030603, RCV003764646

For discussion of the 1-bp duplication in the DPAGT1 gene (699dup) that was found in compound heterozygous state in a patient with congenital myasthenic syndrome-13 (CMS13; 614750) by Belaya et al. (2012), see 191350.0002.


.0005   MYASTHENIC SYNDROME, CONGENITAL, 13

DPAGT1, LEU120MET
SNP: rs387907244, ClinVar: RCV000030604

In 2 sibs with congenital myasthenic syndrome-13 (CMS13; 614750), Belaya et al. (2012) identified compound heterozygosity for 2 mutations in the DPAGT1 gene: a 358C-A transversion resulting in a leu120-to-met (L120M) substitution, and a 791T-G transversion resulting in a val264-to-gly (V264G; 191350.0006) substitution. The patients had onset in the first year of life of hypotonia, poor head control, and delayed motor development. They showed some improvement in muscle power during the teenage years, and both showed a response to pyridostigmine.


.0006   MYASTHENIC SYNDROME, CONGENITAL, 13

DPAGT1, VAL264GLY
SNP: rs387907245, gnomAD: rs387907245, ClinVar: RCV000030605

For discussion of the val264-to-gly (V264G) mutation in the DPAGT1 gene that was found in compound heterozygous state in patients with congenital myasthenic syndrome-13 (CMS13; 614750) by Belaya et al. (2012), see 191350.0005.


.0007   CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, ILE69ASN
SNP: rs397514586, gnomAD: rs397514586, ClinVar: RCV000032992

In a patient with congenital disorder of glycosylation type Ij (CDGIJ; 608093), Timal et al. (2012) identified compound heterozygosity for 2 mutations in the DPAGT1 gene: a 206T-A transversion in exon 2 resulting in an ile69-to-asn (I69N) substitution at a highly conserved residue in the highly conserved dolichol recognition motif, and a G-to-A transition in intron 1 (161+5G-A; 191350.0008), which resulted in degradation of the mutant mRNA. The mutations were found by exome sequencing and confirmed by Sanger sequencing. Each unaffected parent was heterozygous for 1 of the mutations. In patient-derived cells, the formation of GlcNAc-PP-dolichol was reduced to 22% of controls. The patient had multisystem problems, including asphyxia at birth, respiratory insufficiency, frequent apneas, jaundice, nuclear cataracts, cryptorchidism, dysmorphic features, hypertonia of the limbs, joint contractures, tremor, and feeding difficulties. Laboratory studies showed chronic anemia, hypoproteinemia, increased liver enzymes, and coagulation defects.


.0008   CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, IVS1DS, G-A, +5
SNP: rs397515322, gnomAD: rs397515322, ClinVar: RCV000032993

For discussion of the splice site mutation in the DPAGT1 gene (161+5G-A) that was found in compound heterozygous state in a patient with congenital disorder of glycosylation type Ij (CDGIJ; 608093) by Timal et al. (2012), see 191350.0007.


.0009   CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, ALA114GLY
SNP: rs397515327, gnomAD: rs397515327, ClinVar: RCV000055659, RCV001291448

In 2 sibs, born of consanguineous Turkish parents, with congenital disorder of glycosylation type Ij (CDGIJ; 608093), Wurde et al. (2012) identified a homozygous c.341C-G transversion in exon 3 of the DPAGT1 gene, resulting in an ala114-to-gly (A114G) substitution. The mutation was found by homozygosity mapping followed by candidate gene sequencing. The unaffected parents were heterozygous for the mutation, which was not found in 100 control alleles of the same ethnic background. RT-PCR of patient cells showed that the mutation also increased the normal aberrant splicing seen in controls, resulting in the skipping of exons 2/3 and a truncated protein. In vitro functional expression assays showed decreased DPAGT1 activity, at 18% of normal values. The patients had a severe disorder characterized by hyperexcitability, intractable seizures, bilateral cataracts, nystagmus, strabismus, and progressive microcephaly. Both died within their first year of life from cardiorespiratory failure.


.0010   CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, ILE29PHE
SNP: rs397515328, gnomAD: rs397515328, ClinVar: RCV000055660, RCV001209851, RCV003236665

In 2 Pakistani sibs, born of unrelated parents, with a relatively mild form of congenital disorder of glycosylation type Ij (CDGIJ; 608093), Iqbal et al. (2013) identified compound heterozygosity for 2 mutations in the DPAGT1 gene: a c.85A-T transition resulting in an ile29-to-phe (I29F) substitution, and a c.503T-C transition resulting in a leu168-to-pro (L168P; 191350.0011) substitution. The mutations were found by exome sequencing of 1 of the patients and confirmed by Sanger sequencing in both patients. The mutations segregated with the disorder and occurred at highly conserved residues. Neither was present in over 200 ethnically matched chromosomes or in the dbSNP, 1000 Genomes Project, or Exome Variant Server databases. Functional studies of the variants were not performed.


.0011   CONGENITAL DISORDER OF GLYCOSYLATION, TYPE Ij

DPAGT1, LEU168PRO
SNP: rs397515329, ClinVar: RCV000055661

For discussion of the leu168-to-pro (L168P) mutation in the DPAGT1 gene that was found in compound heterozygous state in patients with congenital disorder of glycosylation type Ij (CDGIJ; 608093) by Iqbal et al. (2013), see 191350.0010.


REFERENCES

  1. Belaya, K., Finlayson, S., Slater, C. R., Cossins, J., Liu, W. W., Maxwell, S., McGowan, S. J., Maslau, S., Twigg, S. R. F., Walls, T. J., Pascual Pascual, S. I., Palace, J., Beeson, D. Mutations in DPAGT1 cause a limb-girdle congenital myasthenic syndrome with tubular aggregates. Am. J. Hum. Genet. 91: 193-201, 2012. [PubMed: 22742743] [Full Text: https://doi.org/10.1016/j.ajhg.2012.05.022]

  2. Dong, Y. Y., Wang, H., Pike, A. C. W., Cochrane, S. A., Hamedzadeh, S., Wyszynski, F. J., Bushell, S. R., Royer, S. F., Widdick, D. A., Sajid, A., Boshoff, H. I., Park, Y., and 20 others. Structures of DPAGT1 explain glycosylation disease mechanisms and advance TB antibiotic design. Cell 175: 1045-1058, 2018. [PubMed: 30388443] [Full Text: https://doi.org/10.1016/j.cell.2018.10.037]

  3. Eckert, V., Blank, M., Mazhari-Tabrizi, R., Mumberg, D., Funk, M., Schwarz, R. T. Cloning and functional expression of the human GlcNAc-1-P transferase, the enzyme for the committed step of the dolichol cycle, by heterologous complementation in Saccharomyces cerevisiae. Glycobiology 8: 77-85, 1998. [PubMed: 9451016] [Full Text: https://doi.org/10.1093/glycob/8.1.77]

  4. Freeze, H. H. Update and perspectives on congenital disorders of glycosylation. Glycobiology 11: 129R-143R, 2001. [PubMed: 11805072]

  5. Iqbal, Z., Shahzad, M., Vissers, L. E. L. M., van Scherpenzeel, M., Gilissen, C., Razzaq, A., Zahoor, M. Y., Khan, S. N., Kleefstra, T., Veltman, J. A., de Brouwer, A. P. M., Lefeber, D. J., van Bokhoven, H., Riazuddin, S. A compound heterozygous mutation in DPAGT1 results in a congenital disorder of glycosylation with a relatively mild phenotype. Europ. J. Hum. Genet. 21: 844-849, 2013. [PubMed: 23249953] [Full Text: https://doi.org/10.1038/ejhg.2012.257]

  6. Marek, K. W., Vijay, I. K., Marth, J. D. A recessive deletion in the GlcNAc-1-phosphotransferase gene results in peri-implantation embryonic lethality. Glycobiology 9: 1263-1271, 1999. [PubMed: 10536042] [Full Text: https://doi.org/10.1093/glycob/9.11.1263]

  7. Rajput, B., Ma, J., Muniappa, N., Schantz, L., Naylor, S. L., Lalley, P. A., Vijay, I. K. Mouse UDP-GlcNAc:dolichyl-phosphate N-acetylglucosaminephosphotransferase: molecular cloning of the cDNA, generation of anti-peptide antibodies and chromosomal localization. Biochem. J. 285: 985-992, 1992. [PubMed: 1323278] [Full Text: https://doi.org/10.1042/bj2850985]

  8. Smith, M. W., Clark, S. P., Hutchinson, J. S., Wei, Y. H., Churukian, A. C., Daniels, L. B., Diggle, K. L., Gen, M. W., Romo, A. J., Lin, Y., Selleri, L., Mcelligott, D. L., Evans, G. A. A sequence-tagged site map of human chromosome 11. Genomics 17: 699-725, 1993. [PubMed: 8244387] [Full Text: https://doi.org/10.1006/geno.1993.1392]

  9. Timal, S., Hoischen, A., Lehle, L., Adamowicz, M., Huijben, K., Sykut-Cegielska, J., Paprocka, J., Jamroz, E., van Spronsen, F. J., Korner, C., Gilissen, C., Rodenburg, R. J., Eidhof, I., Van den Heuvel, L., Thiel, C., Wevers, R. A., Morava, E., Veltman, J., Lefeber, D. J. Gene identification in the congenital disorders of glycosylation type I by whole-exome sequencing. Hum. Molec. Genet. 21: 4151-4161, 2012. [PubMed: 22492991] [Full Text: https://doi.org/10.1093/hmg/dds123]

  10. Wu, X., Rush, J. S., Karaoglu, D., Krasnewich, D., Lubinsky, M. S., Waechter, C. J., Gilmore, R., Freeze, H. H. Deficiency of UDP-GlcNAc:dolichol phosphate N-acetylglucosamine-1 phosphate transferase (DPAGT1) causes a novel congenital disorder of glycosylation type Ij. Hum. Mutat. 22: 144-150, 2003. [PubMed: 12872255] [Full Text: https://doi.org/10.1002/humu.10239]

  11. Wurde, A. E., Reunert, J., Rust, S., Hertzberg, C., Haverkamper, S., Nurnberg, G., Nurnberg, P., Lehle, L., Rossi, R., Marquardt, T. Congenital disorder of glycosylation type Ij (CDG-Ij, DPAGT1-CDG): extending the clinical and molecular spectrum of a rare disease. Molec. Genet. Metab. 105: 634-641, 2012. [PubMed: 22304930] [Full Text: https://doi.org/10.1016/j.ymgme.2012.01.001]


Contributors:
Bao Lige - updated : 04/05/2019
Cassandra L. Kniffin - updated : 9/18/2013
Cassandra L. Kniffin - updated : 11/8/2012
Cassandra L. Kniffin - updated : 8/1/2012
Patricia A. Hartz - updated : 11/17/2003
Victor A. McKusick - updated : 9/9/2003

Creation Date:
Victor A. McKusick : 11/10/1992

Edit History:
carol : 03/04/2022
mgross : 04/05/2019
carol : 04/12/2017
carol : 03/27/2017
alopez : 04/29/2015
carol : 4/27/2015
ckniffin : 4/21/2015
mcolton : 4/20/2015
carol : 10/15/2013
carol : 9/26/2013
tpirozzi : 9/26/2013
tpirozzi : 9/26/2013
ckniffin : 9/18/2013
carol : 9/17/2013
terry : 11/8/2012
carol : 11/8/2012
ckniffin : 11/8/2012
carol : 8/2/2012
ckniffin : 8/1/2012
joanna : 1/13/2011
terry : 4/4/2005
mgross : 11/17/2003
tkritzer : 9/26/2003
carol : 9/26/2003
tkritzer : 9/12/2003
tkritzer : 9/9/2003
carol : 8/19/1998
mark : 12/22/1997
carol : 11/10/1992