Entry - *602671 - SOLUTE CARRIER FAMILY 37 (GLUCOSE-6-PHOSPHATE TRANSPORTER), MEMBER 4; SLC37A4 - OMIM
* 602671

SOLUTE CARRIER FAMILY 37 (GLUCOSE-6-PHOSPHATE TRANSPORTER), MEMBER 4; SLC37A4


Alternative titles; symbols

GLUCOSE-6-PHOSPHATE TRANSPORTER 1; G6PT1
GLUCOSE-6-PHOSPHATE TRANSLOCASE
G6P TRANSLOCASE


HGNC Approved Gene Symbol: SLC37A4

Cytogenetic location: 11q23.3     Genomic coordinates (GRCh38): 11:119,024,112-119,030,877 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
11q23.3 Congenital disorder of glycosylation, type IIw 619525 AD 3
Glycogen storage disease Ib 232220 AR 3
Glycogen storage disease Ic 232240 AR 3

TEXT

Description

G6PT1 regulates the rate-limiting step of glucose-6-phosphate (G6P) transport through the endoplasmic reticulum (ER) membrane. It also functions in ATP-mediated calcium sequestration in the ER lumen and as a G6P receptor/sensor (Belkaid et al., 2006).


Cloning and Expression

Gerin et al. (1997) isolated a cDNA from a human bladder tumor cDNA library. The cDNA predicted a 429-amino acid protein with a calculated molecular mass of 46 kD. The protein contains a putative signal for retention in the endoplasmic reticulum. Its nearest homolog was the bacterial gene UhpC, an E. coli glucose-6-phosphate receptor. Northern blot analysis revealed a 2.0-kb mRNA in human liver. The authors concluded that this gene is likely to be a human glucose-6-phosphate translocase.

Using Northern blot analysis, Gerin et al. (1999) detected a G6PT1 transcript of about 2.4 kb in liver and kidney, with weaker expression in leukocytes. They identified a G6PT1 splice variant containing exon 7 in fetal brain EST libraries. Exon 7 introduces 22 amino acids into the luminal loop between transmembrane helices 9 and 10. PCR analysis did not detect the exon 7-containing variant in liver, kidney, or leukocyte mRNA. PCR analysis of mouse tissues revealed exon 7-containing transcripts in brain and heart, but not in liver, kidney, lung, or spleen. In mouse, exon 7 encodes 20 amino acids.

Ihara et al. (2000) investigated the quantitative expression of the G6PT1 gene and its splice variants in human tissues. The G6PT1 gene was strongly expressed in liver, kidney, and hematopoietic progenitor cells. RT-PCR amplification of G6PT1 cDNA revealed tissue-specific expression of several splice variants. The exon 7-containing brain isoform was also identified in heart and skeletal muscle.


Gene Structure

Ihara et al. (1998) determined that the G6P translocase gene spans approximately 5 kb and contains 8 exons. Marcolongo et al. (1998) determined that the G6PT1 gene contains 9 exons and spans about 4 kb. Hou et al. (1999) found that the G6PT1 gene spans 4.5 kb and that all exon/intron boundaries adhere to the canonical ag/gt rule. Gerin et al. (1999) and Hiraiwa et al. (1999) determined that the G6PT1 gene contains 9 exons, and Hiraiwa et al. (1999) determined that the G6PT gene spans 5.3 kb.

Gerin et al. (1999) identified an upstream TATA box and 2 main transcriptional start sites at about -100 and -200 nt with respect to the initiator ATG. They also found 3 Alu sequences at about -3400, -2800, and -1800 nt with respect to the initiator ATG.


Mapping

Kure et al. (1998) mapped the G6PT1 gene to chromosome 11 by study of a DNA panel of human/hamster hybrid cells. Veiga-da-Cunha et al. (1998) used radiation hybrid analysis to map the G6PT1 gene to chromosome 11q23. By fluorescence in situ hybridization, Ihara et al. (1998) refined the localization of the G6PT1 gene to chromosome 11q23.3.


Gene Function

Lin et al. (2000) showed that the exon 7-containing G6PT splice variant, which they designated vG6PT, was active in microsomal G6P transport. They raised the possibility that mutations in exon 7 of the G6PT gene, which would not perturb glucose homeostasis, might have other deleterious effects.

Using small interfering RNA, Belkaid et al. (2006) found that silencing G6PT induced necrosis and late apoptosis in brain tumor-derived U87 glioma cells. The anticancer drug curcumin, which regulates key enzymes involved in carbohydrate metabolism, inhibited G6PT expression by more than 90% and triggered U87 cell death. Overexpression of G6PT rescued cells from curcumin-induced cell death.

Using a combination of enzymologic, cell-culture, and in vivo approaches, Veiga-da-Cunha et al. (2019) demonstrated that G6PT and G6PC3 (611045) collaborate to destroy 1,5-anhydroglucitol-6-phosphate (1,5AG6P), a close structural analog of glucose-6-phosphate and an inhibitor of low-Km hexokinases, which catalyze the first step in glycolysis in most tissues. Veiga-da-Cunha et al. (2019) showed that 1,5AG6P is made by phosphorylation of 1,5-anhydroglucitol (1,5AG), a compound normally present in human plasma, by side activities of ADP-glucokinase and low-Km hexokinases.


Molecular Genetics

Glycogen Storage Disease Ib, Ic, Id

In 4 unrelated Japanese families with glycogen storage disease Ib (GSD1B; 232220), Kure et al. (1998) identified 3 novel mutations in the G6PT1 gene; the W118R mutation (602671.0003) accounted for 4 of 8 mutant alleles, suggesting that it is particularly prevalent among Japanese patients. Ihara et al. (1998) also reported 2 novel mutations in a Japanese patient.

In 2 patients with glycogen storage disease Ib, Gerin et al. (1997) found 2 mutations in the putative glucose-6-phosphate translocase. One patient was homozygous for a gly339-to-cys substitution (602671.0001). The other patient was a compound heterozygote for the gly339-to-cys mutation and a glu355-to-ter (602671.0002) mutation. Both patients showed typical clinical and laboratory findings of GSD Ib, including neutropenia. Neither mutation was present in 4 normal controls.

Veiga-da-Cunha et al. (1998) showed that mutations in the G6PT1 gene account for GSD types Ib and Ic (GSD1C; 232240). Using SSCP analysis and sequencing, they screened this gene for mutations in genomic DNA from 22 families with GSD types Ib and Ic. Of 20 mutations found, 11 resulted in truncated proteins that were probably nonfunctional. Most other mutations resulted in substitutions of conserved or semiconserved residues. The 2 most common mutations, gly339 to cys and 1211-1212delCT (602671.0006), together accounted for approximately 40% of the disease alleles. The fact that the same mutations were found in GSD types Ib and Ic could indicate either that Pi and glucose-6-phosphate are transported in microsomes by the same transporter or that the biochemical assays used to differentiate Pi and glucose-6-phosphate transport defects are not reliable.

In 12 of the 22 families investigated by Veiga-da-Cunha et al. (1998), patients were homozygous for the identified mutation of the G6PT1 gene. In 4 of these families the parents were consanguineous. In 7 of the 10 families in which the patients showed compound heterozygosity, the availability of parental DNA enabled them to show that the 2 mutations were on different chromosomes.

Hiraiwa et al. (1999) identified mutations in the G6PT gene that segregated with GSD Ib. They functionally characterized the recombinant G6PT and demonstrated that mutations found in GSD Ib patients disrupt G6P transport. Hiraiwa et al. (1999) stated that this was the first definition of a molecular basis for functional deficiency in GSD Ib and raised the possibility that defective G6PT contributes to neutropenia and neutrophil/monocyte dysfunctions characteristic of GSD Ib patients.

Hou et al. (1999) studied 5 Japanese patients with GSD type Ib. Two novel homozygous mutations were identified in 2 families: a 3-bp deletion (V235del) in exon 2 in a consanguineous family and a splicing mutation (IVS7DS+1G-T) in intron 7 in a nonconsanguineous family. Two patients were homozygotes for W118R. A fifth patient was a compound heterozygote for W118R and IVS1DS+1G-A. Including their previous study (Kure et al., 1998), this group found a total of 10 W118R alleles in 9 Japanese patients.

Veiga-da-Cunha et al. (1999) analyzed 23 additional families diagnosed as having GSD I non-a (i.e., GSD Ib, Ic, Id). The 9 exons of the G6PT1 gene were amplified by PCR and mutations sought both by SSCP and heteroduplex analysis. Except for one family in which only 1 mutation was found, all patients had 2 allelic mutations in the gene encoding the putative translocase. The mutations were new in 16 and they all were predicted to result in nonfunctional proteins. All investigated patients had some degree of neutropenia or neutrophil dysfunction, and the clinical phenotype of 4 new patients who had been diagnosed as GSD Ic and the one diagnosed as GSD Id was no different from that of the GSD Ib patients. Since these patients, and the 4 type Ic patients from 2 families previously studied, shared several mutations with GSD Ib patients, Veiga-da-Cunha et al. (1999) concluded that their basic defect is in the putative translocase and that they should be reclassified as GSD Ib. Isolated defects in microsomal Pi transporter or in microsomal glucose transporter must be very rare or have phenotypes that are not recognized as GSD I, so that in practice there are only 2 subtypes of GSD I (Ia and Ib).

Galli et al. (1999) reported on the analysis by SSCP and/or DNA sequencing of the exons of the G6PT gene in 14 patients diagnosed as affected with either the GSD Ib or GSD Ic subtype. Mutations in the G6PT gene were found in all patients. Four of the mutations were novel. The results confirmed that the Ib and Ic forms are due to mutations in the same gene. Galli et al. (1999) also showed that the same kind of mutation may or may not be associated with evident clinical complications such as neutrophil impairment. There was no correlation between the type and position of the mutation and the severity of the disease, including the presence of neutropenia.

Janecke et al. (2000) studied 13 patients with glycogen storage disease type I non-A. Analysis of the G6PT gene revealed mutations on both chromosomes in each case, 4 of which were novel.

Chen et al. (2000) demonstrated that 15 missense mutations and a codon deletion mutation abolished microsomal G6P uptake activity and that 2 splicing mutations caused exon skipping in the G6PT gene in patients with glycogen storage disease Ib. Mutation analysis elucidated the structural requirements for stability and transport activity of the G6PT protein.

Using an assay based on an adenoviral vector-mediated expression system, Chen et al. (2002) functionally characterized all 30 codon mutations found in GSD Ib patients. Twenty of the naturally occurring mutations completely abolished microsomal G6P uptake activity, whereas the other 10 mutations (including 5 previously characterized) partially inactivated the transporter. The authors also demonstrated that 5 of the mutations, including val235del (602671.0010), G339C (602671.0001), and G339D (602671.0015), also compromised G6PT stability. The N-terminal domain of G6PT was required for optimal G6P uptake activity. Degradation of both wildtype and mutant G6PT was inhibited by the proteasome inhibitor lactacystin, demonstrating that G6PT is a substrate for proteasome-mediated degradation.

Congenital Disorder of Glycosylation, Type IIw

In a patient with CDG2W, Marquardt et al. (2020) identified a heterozygous mutation (R423X; 602671.0017) in the SLC37A4 gene. The de novo mutation was identified by trio whole-exome sequencing. The mutation was predicted to remove an endoplasmic reticulum retention signal and expose a weak Golgi retention signal. Expression of the mutant protein in HepG2 cells and subsequent immunolocalization studies suggested that it mislocalized to the Golgi. The patient had a CDG type II glycosylation pattern on serum transferrin (190000) isoelectric focusing, and HPLC analysis of serum transferrin showed severe hypoglycosylation.

Wilson et al. (2021) identified heterozygosity for the R423X mutation in a patient with CDG2W. The de novo mutation was identified by whole-exome sequencing and confirmed with Sanger sequencing.

Ng et al. (2021) identified heterozygosity for the R423X mutation in 7 patients from 4 families, including a mother and son from one family and a mother, son, and daughter from another family, with CDG2W. Analysis of Huh7 cells expressing SLC37A4 with the R423X mutation showed that the mutant protein likely localized to an undefined intermediate subcompartment between the total ER and the cis-Golgi, leading to lowered Golgi pH.


Animal Model

Hiraiwa et al. (2001) investigated whether there is a molecular link between HNF1-alpha (HNF1A; 142410) deficiency and function of the G6Pase system. Transactivation studies revealed that HNF1A is required for transcription of the G6PT gene. Hepatic G6PT mRNA levels and microsomal G6P transport activity are also markedly reduced in Hnf1a -/- mice as compared with Hnf1a +/+ and Hnf1a +/- littermates. On the other hand, hepatic G6Pase mRNA expression and activity are upregulated in Hnf1a -/- mice, consistent with observations that G6Pase expression is increased in diabetic animals. Taken together, these results strongly suggest that metabolic abnormalities in Hnf1a-null mice are caused in part by G6PT deficiency and by perturbations of the G6Pase system.

Chen et al. (2003) generated a G6pt-knockout (G6pt -/-) mouse that mimicked all known defects of human GSD Ib. Neutropenia was caused directly by the loss of G6PT activity; chemotaxis and calcium flux, induced by the chemokines KC (MGSA; see 155730) and macrophage inflammatory protein-2 (139110), were defective in G6pt -/- neutrophils; local production of these chemokines and the resultant neutrophil trafficking in vivo were depressed in G6pt -/- ascites during an inflammatory response. The bone and spleen of G6pt -/- mice were developmentally delayed and accompanied by marked hypocellularity of the bone marrow, elevation of myeloid progenitor cell frequencies in both organs, and a corresponding dramatic increase in granulocyte colony-stimulating factor (138970) levels in both GSD Ib mice and humans. The authors concluded that G6PT is not only a G6P transport protein, but is also an important immunomodulatory protein with a role in myeloid complications in GSD Ib.


ALLELIC VARIANTS ( 17 Selected Examples):

.0001 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, GLY339CYS
  
RCV000007330...

In a 22-year-old female patient with type Ib glycogen storage disease (GSD1B; 232220), Gerin et al. (1997) demonstrated homozygosity for a gly339-to-cys (G339C) substitution in the glucose 6-phosphate transporter.


.0002 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, GLU355TER
  
RCV000007331

Gerin et al. (1997) found that a 10-year-old female patient with type Ib glycogen storage disease (GSD1B; 232220) was a compound heterozygote for the G339C substitution (602671.0001) and a glu-355-to-ter (E355X) mutation.


.0003 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, TRP118ARG
  
RCV000007332...

In 4 unrelated Japanese families with glycogen storage disease Ib (GSD1B; 232220), Kure et al. (1998) found that a W118R missense mutation in the G6PT1 gene accounted for 4 of 8 mutant alleles. This same group (Hou et al., 1999) reported that they found a total of 10 W118R alleles in 9 Japanese patients, suggesting that this is an unusually prevalent mutation in Japan.


.0004 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, 4-BP DEL, 2-BP INS, NT1094
  
RCV000007333

In a Japanese patient with glycogen storage disease Ib (GSD1B; 232220), Kure et al. (1998) found homozygosity for a deletion/insertion mutation of the G6PT1 gene. GCTG at nucleotides 1094-1097 were deleted and replaced by TC.


.0005 GLYCOGEN STORAGE DISEASE Ib

GLYCOGEN STORAGE DISEASE Ic, INCLUDED
SLC37A4, 170-BP DEL, NT148
  
RCV000007344...

In a Japanese patient with glycogen storage disease Ib (GSD1B; 232220), Kure et al. (1998) found compound heterozygosity for the W118R mutation (602671.0003) and a G-to-A substitution within a consensus splicing donor site, which resulted in deletion of 170 bp (nucleotides 148-317) and involved the initiation methionine codon.

In a German patient with GSD Ic (GSD1C; 232240), Janecke et al. (1999) identified the same mutation in homozygous state. Genomic sequencing revealed a homozygous 317+1G-T substitution within a consensus splicing donor site.


.0006 GLYCOGEN STORAGE DISEASE Ib

GLYCOGEN STORAGE DISEASE Ic, INCLUDED
SLC37A4, 2-BP DEL, 1211CT
  
RCV000007336...

In 2 families, Veiga-da-Cunha et al. (1998) found that patients with glycogen storage disease Ib (GSD1B; 232220) were homozygous for a 2-bp deletion (1211-1212delCT) in the G6PT1 gene, resulting in a change in reading frame after ala347.

This common frameshift mutation was reported by Veiga-da-Cunha et al. (1998) to be present in 8 GSD Ib patients. In a Turkish patient with GSD Ic (GSD1C; 232240), Janecke et al. (1999) identified the same mutation. Thus, GSD Ib and Ic result from the same mutation of the same gene.


.0007 GLYCOGEN STORAGE DISEASE Ic

SLC37A4, IVS8, 4-BP DEL
  
RCV001733377...

In a Pakistani family in which Fenske et al. (1998) mapped the glycogen storage disease Ic (GSD1C; 232240) locus to 11q23-q24.2, Veiga-da-Cunha et al. (1998) demonstrated a splice site mutation at the exon 8/intron 8 junction in homozygous state in affected individuals.


.0008 GLYCOGEN STORAGE DISEASE Ic

SLC37A4, TRP96TER
  
RCV000007339...

In a female patient with glycogen storage disease Ic (GSD1C; 232240), Veiga-da-Cunha et al. (1998) demonstrated compound heterozygosity for a trp96-to-ter (W96X) nonsense mutation and insertion of a 4-amino acid repeat after met311 (602671.0009) of the G6PT1 gene. They found the latter mutation in compound heterozygous state in a patient diagnosed as GSD Ib (GSD1B; 232220).


.0009 GLYCOGEN STORAGE DISEASE Ib

GLYCOGEN STORAGE DISEASE Ic, INCLUDED
SLC37A4, 12-BP INS, NT1103
  
RCV000169130...

In 2 unrelated patients, 1 with glycogen storage disease Ib (GSD1B; 232220) and 1 with GSD Ic (GSD1C; 232240), Veiga-da-Cunha et al. (1998) observed compound heterozygosity for a 12-nucleotide insertion resulting in a 4-amino acid repeat inserted after met311. In the patient with GSD Ic, the mutation was combined with the missense mutation trp96 to ter (602671.0008); in the patient with GSD Ib, the mutation was combined with a deletion causing a shift of reading frame after ala347. That mutation, due to deletion of nucleotides CT (1211-1212) (602671.0006), was found in homozygosity in 2 families with GSD Ib.


.0010 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, VAL235DEL
  
RCV000007342

In a consanguineous family with GSD Ib (GSD1B; 232220), Hou et al. (1999) identified a 3-bp deletion (val235del) in exon 2 of the G6PT1 gene.


.0011 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, IVS7, G-T, +1
  
RCV000007343

In a nonconsanguineous family with GSD Ib (GSD1B; 232220), Hou et al. (1999) identified a G-to-T change at position +1 of IVS7 of the G6PT1 gene.


.0012 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, IVS1, G-A, +1
   RCV000007344...

Hou et al. (1999) determined that a patient with GSD Ib (GSD1B; 232220) was a compound heterozygote for the W118R mutation (602671.0003) and a G-to-A change at position +1 in intron 1 of the G6PT1 gene.


.0013 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, ARG28HIS
  
RCV000007345...

In 2 of 9 families with GSD Ib (GSD1B; 232220), Hiraiwa et al. (1999) identified a G-to-A transition at nucleotide 252 of the G6PT1 gene, resulting in an arg28-to-his (R28H) mutation. They demonstrated that this mutation resulted in inactive G6P transport.


.0014 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, ARG415TER
  
RCV000007346...

In patients with classic GSD type Ib (GSD1B; 232220), Veiga-da-Cunha et al. (1999) reported a T-to-C transition at nucleotide 415 in exon 8 of the G6PT1 gene, resulting in an arg415-to-ter (R415X) substitution.


.0015 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, GLY339ASP
  
RCV000007347...

In a 25-year-old patient with glycogen storage disease type Ib (GSD1B; 232220) based on enzymatic analysis but no evidence of neutropenia or recurrent infections, Kure et al. (2000) identified an arg415-to-ter mutation (R415X; 606671.0014), which has been reported in patients with neutropenia, in compound heterozygosity with a gly339-to-asp mutation (G339D) due to a G-to-A transition at nucleotide 1185 in exon 7 of the G6PT1 gene.


.0016 GLYCOGEN STORAGE DISEASE Ib

SLC37A4, 794G-A
  
RCV000007348

In a 9-year-old patient with glycogen storage type Ib (GSD1B; 232220) but no evidence of neutropenia or recurrent infections, Kure et al. (2000) identified homozygosity for a G-to-A transition at nucleotide 794 of the G6PT1 gene, which is the last nucleotide of exon 3, resulting in skipping of exon 3 in a proportion of amplified cDNA. Kure et al. (2000) suggested that the residual full-length allele spared the patient from the neutropenia and its complications.


.0017 CONGENITAL DISORDER OF GLYCOSYLATION, TYPE IIw

SLC37A4, ARG423TER
  
RCV001543403...

In a patient with congenital disorder of glycosylation type IIw (CDG2W; 619525), Marquardt et al. (2020) identified a heterozygous c.1267C-T transition in the SLC37A4 gene resulting in an arg423-to-ter (R423X) early termination. The de novo mutation was identified by trio whole-exome sequencing. The mutation was predicted to remove an ER retention signal and expose a weak Golgi retention signal. Expression of the mutant protein in HepG2 cells suggested that it mislocalized to the Golgi and led to significant changes in glycosylation, including reduced tetrasialotransferrin and increased polylactosamines.

In a patient with CDG2W, Wilson et al. (2021) identified heterozygosity for the c.1267C-T transition (c.1267C-T, NM_001164277.2) in the SLC37A4 gene resulting in R423X. The de novo mutation was identified by whole-exome sequencing and confirmed by Sanger sequencing. The patient had a CDG type II glycosylation pattern on serum transferrin isoelectric focusing.

In 7 individuals from 4 families, including a mother and son from one family and a mother, son, and daughter from another family, Ng et al. (2021) identified heterozygosity for the R423X mutation in the SLC37A4 gene. The mutations were identified by a combination of whole-genome sequencing, whole-exome sequencing, and Sanger sequencing. The mutation was absent from the gnomAD, DiscovEHR, and Geno2MP databases. iPSCs generated from one of the patients (patient 7) were differentiated into hepatocytes and shown to accumulate abnormal N-glycans. Analysis of Huh7 cells expressing SLC37A4 with the R423X mutation showed that the mutant protein likely localized to an undefined intermediate subcompartment between the total ER and the cis-Golgi, resulting in lowered Golgi pH.


REFERENCES

  1. Belkaid, A., Copland, I. B., Massillon, D., Annabi, B. Silencing of the human microsomal glucose-6-phosphate translocase induces glioma cell death: potential new anticancer target for curcumin. FEBS Lett. 580: 3746-3752, 2006. [PubMed: 16777101, related citations] [Full Text]

  2. Chen, L.-Y., Lin, B., Pan, C.-J., Hiraiwa, H., Chou, J. Y. Structural requirements for the stability and microsomal transport activity of the human glucose 6-phosphate transporter. J. Biol. Chem. 275: 34280-34286, 2000. [PubMed: 10940311, related citations] [Full Text]

  3. Chen, L.-Y., Pan, C.-J., Shieh, J.-J., Chou, J. Y. Structure-function analysis of the glucose-6-phosphate transporter deficient in glycogen storage disease type Ib. Hum. Molec. Genet. 11: 3199-3207, 2002. [PubMed: 12444104, related citations] [Full Text]

  4. Chen, L.-Y., Shieh, J.-J., Lin, B., Pan, C.-J., Gao, J.-L., Murphy, P. M., Roe, T. F., Moses, S., Ward, J. M., Lee, E. J., Westphal, H., Mansfield, B. C., Chou, J. Y. Impaired glucose homeostasis, neutrophil trafficking and function in mice lacking the glucose-6-phosphate transporter. Hum. Molec. Genet. 12: 2547-2558, 2003. [PubMed: 12925567, related citations] [Full Text]

  5. Fenske, C. D., Jeffery, S., Weber, J. L., Houlston, R. S., Leonard, J. V., Lee, P. Localisation of the gene for glycogen storage disease type Ic by homozygosity mapping to 11q. J. Med. Genet. 35: 269-272, 1998. [PubMed: 9598717, related citations] [Full Text]

  6. Galli, L., Orrico, A., Marcolongo, P., Fulceri, R., Burchell, A., Melis, D., Parini, R., Gatti, R., Lam, C.-W., Benedetti, A., Sorrentino, V. Mutations in the glucose-6-phosphate transporter (G6PT) gene in patients with glycogen storage diseases type 1b and 1c. FEBS Lett. 459: 255-258, 1999. [PubMed: 10518030, related citations] [Full Text]

  7. Gerin, I., Veiga-da-Cunha, M., Achouri, Y., Collet, J.-F., Van Schaftingen, E. Sequence of a putative glucose 6-phosphate translocase, mutated in glycogen storage disease type Ib. FEBS Lett. 419: 235-238, 1997. [PubMed: 9428641, related citations] [Full Text]

  8. Gerin, I., Veiga-da-Cunha, M., Noel, G., Van Schaftingen, E. Structure of the gene mutated in glycogen storage disease type Ib. Gene 227: 189-195, 1999. [PubMed: 10023055, related citations] [Full Text]

  9. Hiraiwa, H., Pan, C.-J., Lin, B., Akiyama, T. E., Gonzalez, F. J., Chou, J. Y. A molecular link between the common phenotypes of type 1 glycogen storage disease and HNF1-alpha-null mice. J. Biol. Chem. 276: 7963-7967, 2001. [PubMed: 11121425, related citations] [Full Text]

  10. Hiraiwa, H., Pan, C.-J., Lin, B., Moses, S. W., Chou, J. Y. Inactivation of the glucose 6-phosphate transporter causes glycogen storage disease type 1b. J. Biol. Chem. 274: 5532-5536, 1999. [PubMed: 10026167, related citations] [Full Text]

  11. Hou, D.-C., Kure, S., Suzuki, Y., Hasegawa, Y., Hara, Y., Inoue, T., Kida, Y., Matsubara, Y., Narisawa, K. Glycogen storage disease type Ib: structural and mutational analysis of the microsomal glucose-6-phosphate transporter gene. Am. J. Med. Genet. 86: 253-257, 1999. [PubMed: 10482875, related citations] [Full Text]

  12. Ihara, K., Kuromaru, R., Hara, T. Genomic structure of the human glucose 6-phosphate translocase gene and novel mutations in the gene of a Japanese patient with glycogen storage disease type Ib. Hum. Genet. 103: 493-496, 1998. [PubMed: 9856496, related citations] [Full Text]

  13. Ihara, K., Nomura, A., Hikino, S., Takada, H., Hara, T. Quantitative analysis of glucose-6-phosphate translocase gene expression in various human tissues and haematopoietic progenitor cells. J. Inherit. Metab. Dis. 23: 583-592, 2000. [PubMed: 11032333, related citations] [Full Text]

  14. Ihara, K., Takabayashi, A., Terasaki, K., Hara, T. Assignment of glucose 6-phosphate translocase (G6PT1) to human chromosome band 11q23.3 by in situ hybridization. Cytogenet. Cell Genet. 83: 50-51, 1998. [PubMed: 9925924, related citations] [Full Text]

  15. Janecke, A. R., Bosshard, N. U., Mayatepek, E., Schulze, A., Gitzelmann, R., Burchell, A., Bartram, C. R., Janssen, B. Molecular diagnosis of type 1c glycogen storage disease. Hum. Genet. 104: 275-277, 1999. [PubMed: 10323254, related citations] [Full Text]

  16. Janecke, A. R., Lindner, M., Erdel, M., Mayatepek, E., Moslinger, D., Podskarbi, T., Fresser, F., Stockler-Ipsiroglu, S., Hoffmann, G. F., Utermann, G. Mutation analysis in glycogen storage disease type 1 non-a. Hum. Genet. 107: 285-289, 2000. [PubMed: 11071391, related citations] [Full Text]

  17. Kure, S., Hou, D.-C., Suzuki, Y., Yamagishi, A., Hiratsuka, M., Fukuda, T., Sugie, H., Kondo, N., Matsubara, Y., Narisawa, K. Glycogen storage disease type Ib without neutropenia. J. Pediat. 137: 253-256, 2000. [PubMed: 10931421, related citations] [Full Text]

  18. Kure, S., Suzuki, Y., Matsubara, Y., Sakamoto, O., Shintaku, H., Isshiki, G., Hoshida, C., Izumi, I., Sakura, N., Narisawa, K. Molecular analysis of glycogen storage disease type Ib: identification of a prevalent mutation among Japanese patients and assignment of a putative glucose-6-phosphate translocase gene to chromosome 11. Biochem. Biophys. Res. Commun. 248: 426-431, 1998. [PubMed: 9675154, related citations] [Full Text]

  19. Lin, B., Pan, C.-J., Chou, J. Y. Human variant glucose-6-phosphate transporter is active in microsomal transport. Hum. Genet. 107: 526-529, 2000. [PubMed: 11140953, related citations] [Full Text]

  20. Marcolongo, P., Barone, V., Priori, G., Pirola, B., Giglio, S., Biasucci, G., Zammarchi, E., Parenti, G., Burchell, A., Benedetti, A., Sorrentino, V. Structure and mutation analysis of the glycogen storage disease type 1b gene. FEBS Lett. 436: 247-250, 1998. Note: Erratum: FEBS Lett. 445: 451 only, 1999. [PubMed: 9781688, related citations] [Full Text]

  21. Marquardt, T., Bzduch, V., Hogrebe, M., Rust, S., Reunert, J., Gruneberg, M., Park, J., Callewaert, N., Lachmann, R., Wada, Y., Engel, T. SLC37A4-CDG: mislocalization of the glucose-6-phosphate transporter to the Golgi causes a new congenital disorder of glycosylation. Molec. Genet. Metab. Rep. 25: 100636, 2020. [PubMed: 32884905, images, related citations] [Full Text]

  22. Ng, B. G., Sosicka, P., Fenaille, F., Harroche, A., Vuillaumier-Barrot, S., Porterfield, M., Xia, Z.-J., Wagner, S., Bamshad, M. J., Vergnes-Boiteux, M.-C., Cholet, S., Dalton, S., and 21 others. A mutation in SLC37A4 causes a dominantly inherited congenital disorder of glycosylation characterized by liver dysfunction. Am. J. Hum. Genet. 108: 1040-1052, 2021. [PubMed: 33964207, related citations] [Full Text]

  23. Veiga-da-Cunha, M., Chevalier, N., Stephenne, X., Defour, J.-P., Paczia, N., Ferster, A., Achouri, Y., Dewulf, J. P., Linster, C. L., Bommer, G. T., Van Schaftingen, E. Failure to eliminate a phosphorylated glucose analog leads to neutropenia in patients with G6PT and G6PC3 deficiency. Proc. Nat. Acad. Sci. 116: 1241-1250, 2019. [PubMed: 30626647, images, related citations] [Full Text]

  24. Veiga-da-Cunha, M., Gerin, I., Chen, Y.-T., de Barsy, T., de Lonlay, P., Dionisi-Vici, C., Fenske, C. D., Lee, P. J., Leonard, J. V., Maire, I., McConkie-Rosell, A., Schweitzer, S., Vikkula, M., Van Schaftingen, E. A gene on chromosome 11q23 coding for a putative glucose-6-phosphate translocase is mutated in glycogen-storage disease types Ib and Ic. Am. J. Hum. Genet. 63: 976-983, 1998. [PubMed: 9758626, related citations] [Full Text]

  25. Veiga-da-Cunha, M., Gerin, I., Chen, Y.-T., Lee, P. J., Leonard, J. V., Maire, I., Wendel, U., Vikkula, M., Van Schaftingen, E. The putative glucose 6-phosphate translocase gene is mutated in essentially all cases of glycogen storage disease type I non-a. Europ. J. Hum. Genet. 7: 717-723, 1999. [PubMed: 10482962, related citations] [Full Text]

  26. Wilson, M. P., Quelhas, D., Leao-Teles, E., Sturiale, L., Rymen, D., Keldermans, L., Race, V., Souche, E., Rodrigues, E., Campos, T., Van Schaftingen, E., Foulquier, F., Garozzo, D., Matthijs, G., Jaeken, J. SLC37A4-CDG: second patient. JIMD Rep. 58: 122-128, 2021. [PubMed: 33728255, images, related citations] [Full Text]


Hilary J. Vernon - updated : 09/10/2021
Ada Hamosh - updated : 11/04/2020
Patricia A. Hartz - updated : 2/13/2009
George E. Tiller - updated : 9/12/2005
George E. Tiller - updated : 9/2/2004
Ada Hamosh - updated : 3/4/2004
Victor A. McKusick - updated : 2/5/2002
Ada Hamosh - updated : 4/20/2001
Victor A. McKusick - updated : 12/19/2000
Ada Hamosh - updated : 10/31/2000
Victor A. McKusick - updated : 10/3/2000
Victor A. McKusick - updated : 1/19/2000
Victor A. McKusick - updated : 11/8/1999
Victor A. McKusick - updated : 10/21/1999
Victor A. McKusick - updated : 4/26/1999
Carol A. Bocchini - updated : 4/5/1999
Ada Hamosh - updated : 3/10/1999
Victor A. McKusick - updated : 10/22/1998
Victor A. McKusick - updated : 9/2/1998
Creation Date:
Jennifer P. Macke : 5/30/1998
alopez : 09/10/2021
alopez : 11/04/2020
mcolton : 04/29/2014
mcolton : 4/28/2014
terry : 11/29/2012
carol : 5/8/2009
carol : 5/8/2009
mgross : 2/17/2009
terry : 2/13/2009
alopez : 10/20/2005
terry : 9/12/2005
carol : 9/3/2004
terry : 9/2/2004
carol : 3/4/2004
terry : 3/4/2004
mgross : 2/19/2002
terry : 2/5/2002
alopez : 4/30/2001
terry : 4/20/2001
mcapotos : 1/4/2001
mcapotos : 1/3/2001
terry : 12/19/2000
mgross : 11/2/2000
mgross : 11/2/2000
terry : 10/31/2000
mcapotos : 10/12/2000
mcapotos : 10/9/2000
terry : 10/3/2000
mcapotos : 1/28/2000
mcapotos : 1/24/2000
terry : 1/19/2000
alopez : 11/12/1999
terry : 11/8/1999
carol : 10/25/1999
terry : 10/21/1999
mgross : 5/7/1999
mgross : 4/29/1999
terry : 4/26/1999
mgross : 4/7/1999
carol : 4/5/1999
alopez : 3/11/1999
alopez : 3/10/1999
carol : 10/26/1998
terry : 10/22/1998
carol : 9/8/1998
alopez : 9/2/1998
alopez : 8/3/1998
alopez : 8/3/1998
alopez : 7/17/1998
alopez : 6/1/1998

* 602671

SOLUTE CARRIER FAMILY 37 (GLUCOSE-6-PHOSPHATE TRANSPORTER), MEMBER 4; SLC37A4


Alternative titles; symbols

GLUCOSE-6-PHOSPHATE TRANSPORTER 1; G6PT1
GLUCOSE-6-PHOSPHATE TRANSLOCASE
G6P TRANSLOCASE


HGNC Approved Gene Symbol: SLC37A4

SNOMEDCT: 237965005, 30102006;  


Cytogenetic location: 11q23.3     Genomic coordinates (GRCh38): 11:119,024,112-119,030,877 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
11q23.3 Congenital disorder of glycosylation, type IIw 619525 Autosomal dominant 3
Glycogen storage disease Ib 232220 Autosomal recessive 3
Glycogen storage disease Ic 232240 Autosomal recessive 3

TEXT

Description

G6PT1 regulates the rate-limiting step of glucose-6-phosphate (G6P) transport through the endoplasmic reticulum (ER) membrane. It also functions in ATP-mediated calcium sequestration in the ER lumen and as a G6P receptor/sensor (Belkaid et al., 2006).


Cloning and Expression

Gerin et al. (1997) isolated a cDNA from a human bladder tumor cDNA library. The cDNA predicted a 429-amino acid protein with a calculated molecular mass of 46 kD. The protein contains a putative signal for retention in the endoplasmic reticulum. Its nearest homolog was the bacterial gene UhpC, an E. coli glucose-6-phosphate receptor. Northern blot analysis revealed a 2.0-kb mRNA in human liver. The authors concluded that this gene is likely to be a human glucose-6-phosphate translocase.

Using Northern blot analysis, Gerin et al. (1999) detected a G6PT1 transcript of about 2.4 kb in liver and kidney, with weaker expression in leukocytes. They identified a G6PT1 splice variant containing exon 7 in fetal brain EST libraries. Exon 7 introduces 22 amino acids into the luminal loop between transmembrane helices 9 and 10. PCR analysis did not detect the exon 7-containing variant in liver, kidney, or leukocyte mRNA. PCR analysis of mouse tissues revealed exon 7-containing transcripts in brain and heart, but not in liver, kidney, lung, or spleen. In mouse, exon 7 encodes 20 amino acids.

Ihara et al. (2000) investigated the quantitative expression of the G6PT1 gene and its splice variants in human tissues. The G6PT1 gene was strongly expressed in liver, kidney, and hematopoietic progenitor cells. RT-PCR amplification of G6PT1 cDNA revealed tissue-specific expression of several splice variants. The exon 7-containing brain isoform was also identified in heart and skeletal muscle.


Gene Structure

Ihara et al. (1998) determined that the G6P translocase gene spans approximately 5 kb and contains 8 exons. Marcolongo et al. (1998) determined that the G6PT1 gene contains 9 exons and spans about 4 kb. Hou et al. (1999) found that the G6PT1 gene spans 4.5 kb and that all exon/intron boundaries adhere to the canonical ag/gt rule. Gerin et al. (1999) and Hiraiwa et al. (1999) determined that the G6PT1 gene contains 9 exons, and Hiraiwa et al. (1999) determined that the G6PT gene spans 5.3 kb.

Gerin et al. (1999) identified an upstream TATA box and 2 main transcriptional start sites at about -100 and -200 nt with respect to the initiator ATG. They also found 3 Alu sequences at about -3400, -2800, and -1800 nt with respect to the initiator ATG.


Mapping

Kure et al. (1998) mapped the G6PT1 gene to chromosome 11 by study of a DNA panel of human/hamster hybrid cells. Veiga-da-Cunha et al. (1998) used radiation hybrid analysis to map the G6PT1 gene to chromosome 11q23. By fluorescence in situ hybridization, Ihara et al. (1998) refined the localization of the G6PT1 gene to chromosome 11q23.3.


Gene Function

Lin et al. (2000) showed that the exon 7-containing G6PT splice variant, which they designated vG6PT, was active in microsomal G6P transport. They raised the possibility that mutations in exon 7 of the G6PT gene, which would not perturb glucose homeostasis, might have other deleterious effects.

Using small interfering RNA, Belkaid et al. (2006) found that silencing G6PT induced necrosis and late apoptosis in brain tumor-derived U87 glioma cells. The anticancer drug curcumin, which regulates key enzymes involved in carbohydrate metabolism, inhibited G6PT expression by more than 90% and triggered U87 cell death. Overexpression of G6PT rescued cells from curcumin-induced cell death.

Using a combination of enzymologic, cell-culture, and in vivo approaches, Veiga-da-Cunha et al. (2019) demonstrated that G6PT and G6PC3 (611045) collaborate to destroy 1,5-anhydroglucitol-6-phosphate (1,5AG6P), a close structural analog of glucose-6-phosphate and an inhibitor of low-Km hexokinases, which catalyze the first step in glycolysis in most tissues. Veiga-da-Cunha et al. (2019) showed that 1,5AG6P is made by phosphorylation of 1,5-anhydroglucitol (1,5AG), a compound normally present in human plasma, by side activities of ADP-glucokinase and low-Km hexokinases.


Molecular Genetics

Glycogen Storage Disease Ib, Ic, Id

In 4 unrelated Japanese families with glycogen storage disease Ib (GSD1B; 232220), Kure et al. (1998) identified 3 novel mutations in the G6PT1 gene; the W118R mutation (602671.0003) accounted for 4 of 8 mutant alleles, suggesting that it is particularly prevalent among Japanese patients. Ihara et al. (1998) also reported 2 novel mutations in a Japanese patient.

In 2 patients with glycogen storage disease Ib, Gerin et al. (1997) found 2 mutations in the putative glucose-6-phosphate translocase. One patient was homozygous for a gly339-to-cys substitution (602671.0001). The other patient was a compound heterozygote for the gly339-to-cys mutation and a glu355-to-ter (602671.0002) mutation. Both patients showed typical clinical and laboratory findings of GSD Ib, including neutropenia. Neither mutation was present in 4 normal controls.

Veiga-da-Cunha et al. (1998) showed that mutations in the G6PT1 gene account for GSD types Ib and Ic (GSD1C; 232240). Using SSCP analysis and sequencing, they screened this gene for mutations in genomic DNA from 22 families with GSD types Ib and Ic. Of 20 mutations found, 11 resulted in truncated proteins that were probably nonfunctional. Most other mutations resulted in substitutions of conserved or semiconserved residues. The 2 most common mutations, gly339 to cys and 1211-1212delCT (602671.0006), together accounted for approximately 40% of the disease alleles. The fact that the same mutations were found in GSD types Ib and Ic could indicate either that Pi and glucose-6-phosphate are transported in microsomes by the same transporter or that the biochemical assays used to differentiate Pi and glucose-6-phosphate transport defects are not reliable.

In 12 of the 22 families investigated by Veiga-da-Cunha et al. (1998), patients were homozygous for the identified mutation of the G6PT1 gene. In 4 of these families the parents were consanguineous. In 7 of the 10 families in which the patients showed compound heterozygosity, the availability of parental DNA enabled them to show that the 2 mutations were on different chromosomes.

Hiraiwa et al. (1999) identified mutations in the G6PT gene that segregated with GSD Ib. They functionally characterized the recombinant G6PT and demonstrated that mutations found in GSD Ib patients disrupt G6P transport. Hiraiwa et al. (1999) stated that this was the first definition of a molecular basis for functional deficiency in GSD Ib and raised the possibility that defective G6PT contributes to neutropenia and neutrophil/monocyte dysfunctions characteristic of GSD Ib patients.

Hou et al. (1999) studied 5 Japanese patients with GSD type Ib. Two novel homozygous mutations were identified in 2 families: a 3-bp deletion (V235del) in exon 2 in a consanguineous family and a splicing mutation (IVS7DS+1G-T) in intron 7 in a nonconsanguineous family. Two patients were homozygotes for W118R. A fifth patient was a compound heterozygote for W118R and IVS1DS+1G-A. Including their previous study (Kure et al., 1998), this group found a total of 10 W118R alleles in 9 Japanese patients.

Veiga-da-Cunha et al. (1999) analyzed 23 additional families diagnosed as having GSD I non-a (i.e., GSD Ib, Ic, Id). The 9 exons of the G6PT1 gene were amplified by PCR and mutations sought both by SSCP and heteroduplex analysis. Except for one family in which only 1 mutation was found, all patients had 2 allelic mutations in the gene encoding the putative translocase. The mutations were new in 16 and they all were predicted to result in nonfunctional proteins. All investigated patients had some degree of neutropenia or neutrophil dysfunction, and the clinical phenotype of 4 new patients who had been diagnosed as GSD Ic and the one diagnosed as GSD Id was no different from that of the GSD Ib patients. Since these patients, and the 4 type Ic patients from 2 families previously studied, shared several mutations with GSD Ib patients, Veiga-da-Cunha et al. (1999) concluded that their basic defect is in the putative translocase and that they should be reclassified as GSD Ib. Isolated defects in microsomal Pi transporter or in microsomal glucose transporter must be very rare or have phenotypes that are not recognized as GSD I, so that in practice there are only 2 subtypes of GSD I (Ia and Ib).

Galli et al. (1999) reported on the analysis by SSCP and/or DNA sequencing of the exons of the G6PT gene in 14 patients diagnosed as affected with either the GSD Ib or GSD Ic subtype. Mutations in the G6PT gene were found in all patients. Four of the mutations were novel. The results confirmed that the Ib and Ic forms are due to mutations in the same gene. Galli et al. (1999) also showed that the same kind of mutation may or may not be associated with evident clinical complications such as neutrophil impairment. There was no correlation between the type and position of the mutation and the severity of the disease, including the presence of neutropenia.

Janecke et al. (2000) studied 13 patients with glycogen storage disease type I non-A. Analysis of the G6PT gene revealed mutations on both chromosomes in each case, 4 of which were novel.

Chen et al. (2000) demonstrated that 15 missense mutations and a codon deletion mutation abolished microsomal G6P uptake activity and that 2 splicing mutations caused exon skipping in the G6PT gene in patients with glycogen storage disease Ib. Mutation analysis elucidated the structural requirements for stability and transport activity of the G6PT protein.

Using an assay based on an adenoviral vector-mediated expression system, Chen et al. (2002) functionally characterized all 30 codon mutations found in GSD Ib patients. Twenty of the naturally occurring mutations completely abolished microsomal G6P uptake activity, whereas the other 10 mutations (including 5 previously characterized) partially inactivated the transporter. The authors also demonstrated that 5 of the mutations, including val235del (602671.0010), G339C (602671.0001), and G339D (602671.0015), also compromised G6PT stability. The N-terminal domain of G6PT was required for optimal G6P uptake activity. Degradation of both wildtype and mutant G6PT was inhibited by the proteasome inhibitor lactacystin, demonstrating that G6PT is a substrate for proteasome-mediated degradation.

Congenital Disorder of Glycosylation, Type IIw

In a patient with CDG2W, Marquardt et al. (2020) identified a heterozygous mutation (R423X; 602671.0017) in the SLC37A4 gene. The de novo mutation was identified by trio whole-exome sequencing. The mutation was predicted to remove an endoplasmic reticulum retention signal and expose a weak Golgi retention signal. Expression of the mutant protein in HepG2 cells and subsequent immunolocalization studies suggested that it mislocalized to the Golgi. The patient had a CDG type II glycosylation pattern on serum transferrin (190000) isoelectric focusing, and HPLC analysis of serum transferrin showed severe hypoglycosylation.

Wilson et al. (2021) identified heterozygosity for the R423X mutation in a patient with CDG2W. The de novo mutation was identified by whole-exome sequencing and confirmed with Sanger sequencing.

Ng et al. (2021) identified heterozygosity for the R423X mutation in 7 patients from 4 families, including a mother and son from one family and a mother, son, and daughter from another family, with CDG2W. Analysis of Huh7 cells expressing SLC37A4 with the R423X mutation showed that the mutant protein likely localized to an undefined intermediate subcompartment between the total ER and the cis-Golgi, leading to lowered Golgi pH.


Animal Model

Hiraiwa et al. (2001) investigated whether there is a molecular link between HNF1-alpha (HNF1A; 142410) deficiency and function of the G6Pase system. Transactivation studies revealed that HNF1A is required for transcription of the G6PT gene. Hepatic G6PT mRNA levels and microsomal G6P transport activity are also markedly reduced in Hnf1a -/- mice as compared with Hnf1a +/+ and Hnf1a +/- littermates. On the other hand, hepatic G6Pase mRNA expression and activity are upregulated in Hnf1a -/- mice, consistent with observations that G6Pase expression is increased in diabetic animals. Taken together, these results strongly suggest that metabolic abnormalities in Hnf1a-null mice are caused in part by G6PT deficiency and by perturbations of the G6Pase system.

Chen et al. (2003) generated a G6pt-knockout (G6pt -/-) mouse that mimicked all known defects of human GSD Ib. Neutropenia was caused directly by the loss of G6PT activity; chemotaxis and calcium flux, induced by the chemokines KC (MGSA; see 155730) and macrophage inflammatory protein-2 (139110), were defective in G6pt -/- neutrophils; local production of these chemokines and the resultant neutrophil trafficking in vivo were depressed in G6pt -/- ascites during an inflammatory response. The bone and spleen of G6pt -/- mice were developmentally delayed and accompanied by marked hypocellularity of the bone marrow, elevation of myeloid progenitor cell frequencies in both organs, and a corresponding dramatic increase in granulocyte colony-stimulating factor (138970) levels in both GSD Ib mice and humans. The authors concluded that G6PT is not only a G6P transport protein, but is also an important immunomodulatory protein with a role in myeloid complications in GSD Ib.


ALLELIC VARIANTS 17 Selected Examples):

.0001   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, GLY339CYS
SNP: rs80356490, gnomAD: rs80356490, ClinVar: RCV000007330, RCV000059116

In a 22-year-old female patient with type Ib glycogen storage disease (GSD1B; 232220), Gerin et al. (1997) demonstrated homozygosity for a gly339-to-cys (G339C) substitution in the glucose 6-phosphate transporter.


.0002   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, GLU355TER
SNP: rs121908975, gnomAD: rs121908975, ClinVar: RCV000007331

Gerin et al. (1997) found that a 10-year-old female patient with type Ib glycogen storage disease (GSD1B; 232220) was a compound heterozygote for the G339C substitution (602671.0001) and a glu-355-to-ter (E355X) mutation.


.0003   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, TRP118ARG
SNP: rs80356489, gnomAD: rs80356489, ClinVar: RCV000007332, RCV000059128

In 4 unrelated Japanese families with glycogen storage disease Ib (GSD1B; 232220), Kure et al. (1998) found that a W118R missense mutation in the G6PT1 gene accounted for 4 of 8 mutant alleles. This same group (Hou et al., 1999) reported that they found a total of 10 W118R alleles in 9 Japanese patients, suggesting that this is an unusually prevalent mutation in Japan.


.0004   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, 4-BP DEL, 2-BP INS, NT1094
SNP: rs2134631556, ClinVar: RCV000007333

In a Japanese patient with glycogen storage disease Ib (GSD1B; 232220), Kure et al. (1998) found homozygosity for a deletion/insertion mutation of the G6PT1 gene. GCTG at nucleotides 1094-1097 were deleted and replaced by TC.


.0005   GLYCOGEN STORAGE DISEASE Ib

GLYCOGEN STORAGE DISEASE Ic, INCLUDED
SLC37A4, 170-BP DEL, NT148
SNP: rs1943672400, ClinVar: RCV000007344, RCV002223174, RCV002496291

In a Japanese patient with glycogen storage disease Ib (GSD1B; 232220), Kure et al. (1998) found compound heterozygosity for the W118R mutation (602671.0003) and a G-to-A substitution within a consensus splicing donor site, which resulted in deletion of 170 bp (nucleotides 148-317) and involved the initiation methionine codon.

In a German patient with GSD Ic (GSD1C; 232240), Janecke et al. (1999) identified the same mutation in homozygous state. Genomic sequencing revealed a homozygous 317+1G-T substitution within a consensus splicing donor site.


.0006   GLYCOGEN STORAGE DISEASE Ib

GLYCOGEN STORAGE DISEASE Ic, INCLUDED
SLC37A4, 2-BP DEL, 1211CT
SNP: rs80356491, gnomAD: rs80356491, ClinVar: RCV000007336, RCV000007337, RCV000601076, RCV000624535, RCV000723824, RCV002279712, RCV002476940

In 2 families, Veiga-da-Cunha et al. (1998) found that patients with glycogen storage disease Ib (GSD1B; 232220) were homozygous for a 2-bp deletion (1211-1212delCT) in the G6PT1 gene, resulting in a change in reading frame after ala347.

This common frameshift mutation was reported by Veiga-da-Cunha et al. (1998) to be present in 8 GSD Ib patients. In a Turkish patient with GSD Ic (GSD1C; 232240), Janecke et al. (1999) identified the same mutation. Thus, GSD Ib and Ic result from the same mutation of the same gene.


.0007   GLYCOGEN STORAGE DISEASE Ic

SLC37A4, IVS8, 4-BP DEL
SNP: rs782612223, gnomAD: rs782612223, ClinVar: RCV001733377, RCV002274201

In a Pakistani family in which Fenske et al. (1998) mapped the glycogen storage disease Ic (GSD1C; 232240) locus to 11q23-q24.2, Veiga-da-Cunha et al. (1998) demonstrated a splice site mutation at the exon 8/intron 8 junction in homozygous state in affected individuals.


.0008   GLYCOGEN STORAGE DISEASE Ic

SLC37A4, TRP96TER
SNP: rs121908976, gnomAD: rs121908976, ClinVar: RCV000007339, RCV000169480, RCV000779044

In a female patient with glycogen storage disease Ic (GSD1C; 232240), Veiga-da-Cunha et al. (1998) demonstrated compound heterozygosity for a trp96-to-ter (W96X) nonsense mutation and insertion of a 4-amino acid repeat after met311 (602671.0009) of the G6PT1 gene. They found the latter mutation in compound heterozygous state in a patient diagnosed as GSD Ib (GSD1B; 232220).


.0009   GLYCOGEN STORAGE DISEASE Ib

GLYCOGEN STORAGE DISEASE Ic, INCLUDED
SLC37A4, 12-BP INS, NT1103
SNP: rs786204477, gnomAD: rs786204477, ClinVar: RCV000169130, RCV002273969, RCV003407627

In 2 unrelated patients, 1 with glycogen storage disease Ib (GSD1B; 232220) and 1 with GSD Ic (GSD1C; 232240), Veiga-da-Cunha et al. (1998) observed compound heterozygosity for a 12-nucleotide insertion resulting in a 4-amino acid repeat inserted after met311. In the patient with GSD Ic, the mutation was combined with the missense mutation trp96 to ter (602671.0008); in the patient with GSD Ib, the mutation was combined with a deletion causing a shift of reading frame after ala347. That mutation, due to deletion of nucleotides CT (1211-1212) (602671.0006), was found in homozygosity in 2 families with GSD Ib.


.0010   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, VAL235DEL
SNP: rs121908977, ClinVar: RCV000007342

In a consanguineous family with GSD Ib (GSD1B; 232220), Hou et al. (1999) identified a 3-bp deletion (val235del) in exon 2 of the G6PT1 gene.


.0011   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, IVS7, G-T, +1
SNP: rs1943574435, ClinVar: RCV000007343

In a nonconsanguineous family with GSD Ib (GSD1B; 232220), Hou et al. (1999) identified a G-to-T change at position +1 of IVS7 of the G6PT1 gene.


.0012   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, IVS1, G-A, +1
ClinVar: RCV000007344, RCV002223174, RCV002496291

Hou et al. (1999) determined that a patient with GSD Ib (GSD1B; 232220) was a compound heterozygote for the W118R mutation (602671.0003) and a G-to-A change at position +1 in intron 1 of the G6PT1 gene.


.0013   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, ARG28HIS
SNP: rs121908978, gnomAD: rs121908978, ClinVar: RCV000007345, RCV000059144, RCV002482839

In 2 of 9 families with GSD Ib (GSD1B; 232220), Hiraiwa et al. (1999) identified a G-to-A transition at nucleotide 252 of the G6PT1 gene, resulting in an arg28-to-his (R28H) mutation. They demonstrated that this mutation resulted in inactive G6P transport.


.0014   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, ARG415TER
SNP: rs121908979, gnomAD: rs121908979, ClinVar: RCV000007346, RCV001196429, RCV002496292, RCV003989281

In patients with classic GSD type Ib (GSD1B; 232220), Veiga-da-Cunha et al. (1999) reported a T-to-C transition at nucleotide 415 in exon 8 of the G6PT1 gene, resulting in an arg415-to-ter (R415X) substitution.


.0015   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, GLY339ASP
SNP: rs121908980, ClinVar: RCV000007347, RCV000059117, RCV002482840

In a 25-year-old patient with glycogen storage disease type Ib (GSD1B; 232220) based on enzymatic analysis but no evidence of neutropenia or recurrent infections, Kure et al. (2000) identified an arg415-to-ter mutation (R415X; 606671.0014), which has been reported in patients with neutropenia, in compound heterozygosity with a gly339-to-asp mutation (G339D) due to a G-to-A transition at nucleotide 1185 in exon 7 of the G6PT1 gene.


.0016   GLYCOGEN STORAGE DISEASE Ib

SLC37A4, 794G-A
SNP: rs1272300904, gnomAD: rs1272300904, ClinVar: RCV000007348

In a 9-year-old patient with glycogen storage type Ib (GSD1B; 232220) but no evidence of neutropenia or recurrent infections, Kure et al. (2000) identified homozygosity for a G-to-A transition at nucleotide 794 of the G6PT1 gene, which is the last nucleotide of exon 3, resulting in skipping of exon 3 in a proportion of amplified cDNA. Kure et al. (2000) suggested that the residual full-length allele spared the patient from the neutropenia and its complications.


.0017   CONGENITAL DISORDER OF GLYCOSYLATION, TYPE IIw

SLC37A4, ARG423TER
SNP: rs2134626266, ClinVar: RCV001543403, RCV001647389, RCV002506653

In a patient with congenital disorder of glycosylation type IIw (CDG2W; 619525), Marquardt et al. (2020) identified a heterozygous c.1267C-T transition in the SLC37A4 gene resulting in an arg423-to-ter (R423X) early termination. The de novo mutation was identified by trio whole-exome sequencing. The mutation was predicted to remove an ER retention signal and expose a weak Golgi retention signal. Expression of the mutant protein in HepG2 cells suggested that it mislocalized to the Golgi and led to significant changes in glycosylation, including reduced tetrasialotransferrin and increased polylactosamines.

In a patient with CDG2W, Wilson et al. (2021) identified heterozygosity for the c.1267C-T transition (c.1267C-T, NM_001164277.2) in the SLC37A4 gene resulting in R423X. The de novo mutation was identified by whole-exome sequencing and confirmed by Sanger sequencing. The patient had a CDG type II glycosylation pattern on serum transferrin isoelectric focusing.

In 7 individuals from 4 families, including a mother and son from one family and a mother, son, and daughter from another family, Ng et al. (2021) identified heterozygosity for the R423X mutation in the SLC37A4 gene. The mutations were identified by a combination of whole-genome sequencing, whole-exome sequencing, and Sanger sequencing. The mutation was absent from the gnomAD, DiscovEHR, and Geno2MP databases. iPSCs generated from one of the patients (patient 7) were differentiated into hepatocytes and shown to accumulate abnormal N-glycans. Analysis of Huh7 cells expressing SLC37A4 with the R423X mutation showed that the mutant protein likely localized to an undefined intermediate subcompartment between the total ER and the cis-Golgi, resulting in lowered Golgi pH.


REFERENCES

  1. Belkaid, A., Copland, I. B., Massillon, D., Annabi, B. Silencing of the human microsomal glucose-6-phosphate translocase induces glioma cell death: potential new anticancer target for curcumin. FEBS Lett. 580: 3746-3752, 2006. [PubMed: 16777101] [Full Text: https://doi.org/10.1016/j.febslet.2006.05.071]

  2. Chen, L.-Y., Lin, B., Pan, C.-J., Hiraiwa, H., Chou, J. Y. Structural requirements for the stability and microsomal transport activity of the human glucose 6-phosphate transporter. J. Biol. Chem. 275: 34280-34286, 2000. [PubMed: 10940311] [Full Text: https://doi.org/10.1074/jbc.M006439200]

  3. Chen, L.-Y., Pan, C.-J., Shieh, J.-J., Chou, J. Y. Structure-function analysis of the glucose-6-phosphate transporter deficient in glycogen storage disease type Ib. Hum. Molec. Genet. 11: 3199-3207, 2002. [PubMed: 12444104] [Full Text: https://doi.org/10.1093/hmg/11.25.3199]

  4. Chen, L.-Y., Shieh, J.-J., Lin, B., Pan, C.-J., Gao, J.-L., Murphy, P. M., Roe, T. F., Moses, S., Ward, J. M., Lee, E. J., Westphal, H., Mansfield, B. C., Chou, J. Y. Impaired glucose homeostasis, neutrophil trafficking and function in mice lacking the glucose-6-phosphate transporter. Hum. Molec. Genet. 12: 2547-2558, 2003. [PubMed: 12925567] [Full Text: https://doi.org/10.1093/hmg/ddg263]

  5. Fenske, C. D., Jeffery, S., Weber, J. L., Houlston, R. S., Leonard, J. V., Lee, P. Localisation of the gene for glycogen storage disease type Ic by homozygosity mapping to 11q. J. Med. Genet. 35: 269-272, 1998. [PubMed: 9598717] [Full Text: https://doi.org/10.1136/jmg.35.4.269]

  6. Galli, L., Orrico, A., Marcolongo, P., Fulceri, R., Burchell, A., Melis, D., Parini, R., Gatti, R., Lam, C.-W., Benedetti, A., Sorrentino, V. Mutations in the glucose-6-phosphate transporter (G6PT) gene in patients with glycogen storage diseases type 1b and 1c. FEBS Lett. 459: 255-258, 1999. [PubMed: 10518030] [Full Text: https://doi.org/10.1016/s0014-5793(99)01248-x]

  7. Gerin, I., Veiga-da-Cunha, M., Achouri, Y., Collet, J.-F., Van Schaftingen, E. Sequence of a putative glucose 6-phosphate translocase, mutated in glycogen storage disease type Ib. FEBS Lett. 419: 235-238, 1997. [PubMed: 9428641] [Full Text: https://doi.org/10.1016/s0014-5793(97)01463-4]

  8. Gerin, I., Veiga-da-Cunha, M., Noel, G., Van Schaftingen, E. Structure of the gene mutated in glycogen storage disease type Ib. Gene 227: 189-195, 1999. [PubMed: 10023055] [Full Text: https://doi.org/10.1016/s0378-1119(98)00614-3]

  9. Hiraiwa, H., Pan, C.-J., Lin, B., Akiyama, T. E., Gonzalez, F. J., Chou, J. Y. A molecular link between the common phenotypes of type 1 glycogen storage disease and HNF1-alpha-null mice. J. Biol. Chem. 276: 7963-7967, 2001. [PubMed: 11121425] [Full Text: https://doi.org/10.1074/jbc.M010523200]

  10. Hiraiwa, H., Pan, C.-J., Lin, B., Moses, S. W., Chou, J. Y. Inactivation of the glucose 6-phosphate transporter causes glycogen storage disease type 1b. J. Biol. Chem. 274: 5532-5536, 1999. [PubMed: 10026167] [Full Text: https://doi.org/10.1074/jbc.274.9.5532]

  11. Hou, D.-C., Kure, S., Suzuki, Y., Hasegawa, Y., Hara, Y., Inoue, T., Kida, Y., Matsubara, Y., Narisawa, K. Glycogen storage disease type Ib: structural and mutational analysis of the microsomal glucose-6-phosphate transporter gene. Am. J. Med. Genet. 86: 253-257, 1999. [PubMed: 10482875] [Full Text: https://doi.org/10.1002/(sici)1096-8628(19990917)86:3<253::aid-ajmg11>3.0.co;2-7]

  12. Ihara, K., Kuromaru, R., Hara, T. Genomic structure of the human glucose 6-phosphate translocase gene and novel mutations in the gene of a Japanese patient with glycogen storage disease type Ib. Hum. Genet. 103: 493-496, 1998. [PubMed: 9856496] [Full Text: https://doi.org/10.1007/s004390050856]

  13. Ihara, K., Nomura, A., Hikino, S., Takada, H., Hara, T. Quantitative analysis of glucose-6-phosphate translocase gene expression in various human tissues and haematopoietic progenitor cells. J. Inherit. Metab. Dis. 23: 583-592, 2000. [PubMed: 11032333] [Full Text: https://doi.org/10.1023/a:1005677912539]

  14. Ihara, K., Takabayashi, A., Terasaki, K., Hara, T. Assignment of glucose 6-phosphate translocase (G6PT1) to human chromosome band 11q23.3 by in situ hybridization. Cytogenet. Cell Genet. 83: 50-51, 1998. [PubMed: 9925924] [Full Text: https://doi.org/10.1159/000015167]

  15. Janecke, A. R., Bosshard, N. U., Mayatepek, E., Schulze, A., Gitzelmann, R., Burchell, A., Bartram, C. R., Janssen, B. Molecular diagnosis of type 1c glycogen storage disease. Hum. Genet. 104: 275-277, 1999. [PubMed: 10323254] [Full Text: https://doi.org/10.1007/s004390050948]

  16. Janecke, A. R., Lindner, M., Erdel, M., Mayatepek, E., Moslinger, D., Podskarbi, T., Fresser, F., Stockler-Ipsiroglu, S., Hoffmann, G. F., Utermann, G. Mutation analysis in glycogen storage disease type 1 non-a. Hum. Genet. 107: 285-289, 2000. [PubMed: 11071391] [Full Text: https://doi.org/10.1007/s004390000371]

  17. Kure, S., Hou, D.-C., Suzuki, Y., Yamagishi, A., Hiratsuka, M., Fukuda, T., Sugie, H., Kondo, N., Matsubara, Y., Narisawa, K. Glycogen storage disease type Ib without neutropenia. J. Pediat. 137: 253-256, 2000. [PubMed: 10931421] [Full Text: https://doi.org/10.1067/mpd.2000.107472]

  18. Kure, S., Suzuki, Y., Matsubara, Y., Sakamoto, O., Shintaku, H., Isshiki, G., Hoshida, C., Izumi, I., Sakura, N., Narisawa, K. Molecular analysis of glycogen storage disease type Ib: identification of a prevalent mutation among Japanese patients and assignment of a putative glucose-6-phosphate translocase gene to chromosome 11. Biochem. Biophys. Res. Commun. 248: 426-431, 1998. [PubMed: 9675154] [Full Text: https://doi.org/10.1006/bbrc.1998.8985]

  19. Lin, B., Pan, C.-J., Chou, J. Y. Human variant glucose-6-phosphate transporter is active in microsomal transport. Hum. Genet. 107: 526-529, 2000. [PubMed: 11140953] [Full Text: https://doi.org/10.1007/s004390000404]

  20. Marcolongo, P., Barone, V., Priori, G., Pirola, B., Giglio, S., Biasucci, G., Zammarchi, E., Parenti, G., Burchell, A., Benedetti, A., Sorrentino, V. Structure and mutation analysis of the glycogen storage disease type 1b gene. FEBS Lett. 436: 247-250, 1998. Note: Erratum: FEBS Lett. 445: 451 only, 1999. [PubMed: 9781688] [Full Text: https://doi.org/10.1016/s0014-5793(98)01129-6]

  21. Marquardt, T., Bzduch, V., Hogrebe, M., Rust, S., Reunert, J., Gruneberg, M., Park, J., Callewaert, N., Lachmann, R., Wada, Y., Engel, T. SLC37A4-CDG: mislocalization of the glucose-6-phosphate transporter to the Golgi causes a new congenital disorder of glycosylation. Molec. Genet. Metab. Rep. 25: 100636, 2020. [PubMed: 32884905] [Full Text: https://doi.org/10.1016/j.ymgmr.2020.100636]

  22. Ng, B. G., Sosicka, P., Fenaille, F., Harroche, A., Vuillaumier-Barrot, S., Porterfield, M., Xia, Z.-J., Wagner, S., Bamshad, M. J., Vergnes-Boiteux, M.-C., Cholet, S., Dalton, S., and 21 others. A mutation in SLC37A4 causes a dominantly inherited congenital disorder of glycosylation characterized by liver dysfunction. Am. J. Hum. Genet. 108: 1040-1052, 2021. [PubMed: 33964207] [Full Text: https://doi.org/10.1016/j.ajhg.2021.04.013]

  23. Veiga-da-Cunha, M., Chevalier, N., Stephenne, X., Defour, J.-P., Paczia, N., Ferster, A., Achouri, Y., Dewulf, J. P., Linster, C. L., Bommer, G. T., Van Schaftingen, E. Failure to eliminate a phosphorylated glucose analog leads to neutropenia in patients with G6PT and G6PC3 deficiency. Proc. Nat. Acad. Sci. 116: 1241-1250, 2019. [PubMed: 30626647] [Full Text: https://doi.org/10.1073/pnas.1816143116]

  24. Veiga-da-Cunha, M., Gerin, I., Chen, Y.-T., de Barsy, T., de Lonlay, P., Dionisi-Vici, C., Fenske, C. D., Lee, P. J., Leonard, J. V., Maire, I., McConkie-Rosell, A., Schweitzer, S., Vikkula, M., Van Schaftingen, E. A gene on chromosome 11q23 coding for a putative glucose-6-phosphate translocase is mutated in glycogen-storage disease types Ib and Ic. Am. J. Hum. Genet. 63: 976-983, 1998. [PubMed: 9758626] [Full Text: https://doi.org/10.1086/302068]

  25. Veiga-da-Cunha, M., Gerin, I., Chen, Y.-T., Lee, P. J., Leonard, J. V., Maire, I., Wendel, U., Vikkula, M., Van Schaftingen, E. The putative glucose 6-phosphate translocase gene is mutated in essentially all cases of glycogen storage disease type I non-a. Europ. J. Hum. Genet. 7: 717-723, 1999. [PubMed: 10482962] [Full Text: https://doi.org/10.1038/sj.ejhg.5200366]

  26. Wilson, M. P., Quelhas, D., Leao-Teles, E., Sturiale, L., Rymen, D., Keldermans, L., Race, V., Souche, E., Rodrigues, E., Campos, T., Van Schaftingen, E., Foulquier, F., Garozzo, D., Matthijs, G., Jaeken, J. SLC37A4-CDG: second patient. JIMD Rep. 58: 122-128, 2021. [PubMed: 33728255] [Full Text: https://doi.org/10.1002/jmd2.12195]


Contributors:
Hilary J. Vernon - updated : 09/10/2021
Ada Hamosh - updated : 11/04/2020
Patricia A. Hartz - updated : 2/13/2009
George E. Tiller - updated : 9/12/2005
George E. Tiller - updated : 9/2/2004
Ada Hamosh - updated : 3/4/2004
Victor A. McKusick - updated : 2/5/2002
Ada Hamosh - updated : 4/20/2001
Victor A. McKusick - updated : 12/19/2000
Ada Hamosh - updated : 10/31/2000
Victor A. McKusick - updated : 10/3/2000
Victor A. McKusick - updated : 1/19/2000
Victor A. McKusick - updated : 11/8/1999
Victor A. McKusick - updated : 10/21/1999
Victor A. McKusick - updated : 4/26/1999
Carol A. Bocchini - updated : 4/5/1999
Ada Hamosh - updated : 3/10/1999
Victor A. McKusick - updated : 10/22/1998
Victor A. McKusick - updated : 9/2/1998

Creation Date:
Jennifer P. Macke : 5/30/1998

Edit History:
alopez : 09/10/2021
alopez : 11/04/2020
mcolton : 04/29/2014
mcolton : 4/28/2014
terry : 11/29/2012
carol : 5/8/2009
carol : 5/8/2009
mgross : 2/17/2009
terry : 2/13/2009
alopez : 10/20/2005
terry : 9/12/2005
carol : 9/3/2004
terry : 9/2/2004
carol : 3/4/2004
terry : 3/4/2004
mgross : 2/19/2002
terry : 2/5/2002
alopez : 4/30/2001
terry : 4/20/2001
mcapotos : 1/4/2001
mcapotos : 1/3/2001
terry : 12/19/2000
mgross : 11/2/2000
mgross : 11/2/2000
terry : 10/31/2000
mcapotos : 10/12/2000
mcapotos : 10/9/2000
terry : 10/3/2000
mcapotos : 1/28/2000
mcapotos : 1/24/2000
terry : 1/19/2000
alopez : 11/12/1999
terry : 11/8/1999
carol : 10/25/1999
terry : 10/21/1999
mgross : 5/7/1999
mgross : 4/29/1999
terry : 4/26/1999
mgross : 4/7/1999
carol : 4/5/1999
alopez : 3/11/1999
alopez : 3/10/1999
carol : 10/26/1998
terry : 10/22/1998
carol : 9/8/1998
alopez : 9/2/1998
alopez : 8/3/1998
alopez : 8/3/1998
alopez : 7/17/1998
alopez : 6/1/1998