Entry - *311850 - PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE I; PRPS1 - OMIM
* 311850

PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE I; PRPS1


HGNC Approved Gene Symbol: PRPS1

Cytogenetic location: Xq22.3     Genomic coordinates (GRCh38): X:107,628,510-107,651,026 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Xq22.3 Arts syndrome 301835 XLR 3
Charcot-Marie-Tooth disease, X-linked recessive, 5 311070 XLR 3
Deafness, X-linked 1 304500 XL 3
Gout, PRPS-related 300661 XLR 3
Phosphoribosylpyrophosphate synthetase superactivity 300661 XLR 3

TEXT

Description

Phosphoribosylpyrophosphate synthetase (PRPS; EC 2.7.6.1) catalyzes the phosphoribosylation of ribose 5-phosphate to 5-phosphoribosyl-1-pyrophosphate, which is necessary for the de novo and salvage pathways of purine and pyrimidine biosynthesis (Roessler et al., 1990). Three PRPS genes have been identified: the widely expressed PRPS1 and PRPS2 (311860) genes, which map to chromosome Xq22-q24 and Xp22, respectively, and PRPS3 (PRPS1L1; 611566), which maps to chromosome 7 and appears to be transcribed only in testis (Becker, 2001).


Cloning and Expression

Roessler et al. (1990) isolated a partial clone corresponding to the PRPS1 gene from a human lymphoblast cDNA library. The deduced PRPS1 protein has 318 amino acids and shares 95% amino acid homology with PRPS2. Becker et al. (1990) also cloned the PRPS1 gene and detected a 2.3-kb mRNA transcript.

By Northern blot analysis using rat Prps1 as probe, Taira et al. (1989) detected a 2.3-kb transcript in human adipose tissue, testis, and placenta and in 2 human cell lines.

Kim et al. (2007) demonstrated that the PRPS1 amino acid sequence shows an exceptionally high degree of conservation, with homologies greater than 95% across different species from zebrafish to human.


Gene Structure

The PRPS1 gene spans over 30 kb and contains 7 exons (Becker, 2001).


Mapping

By the Goss-Harris method, Becker et al. (1978) concluded that the order of loci on chromosome Xq is G6PD (305900)--HPRT1 (308000)--PRPS1--alpha-GAL (GLA; 300644)--PGK1 (311800)--centromere. Becker et al. (1979) assigned the PRPS1 locus to a position between the GLA and HPRT1 loci, particularly close to the latter, and discussed the functional significance of the proximity of the genes for their biochemically related functions.

Becker et al. (1990) mapped PRPS1 to Xq22-q24 by a combination of in situ hybridization and study of human/rodent somatic cell hybrids.

Pseudogene

By in situ chromosomal hybridization, Becker et al. (1990) identified a PRPS1-related gene or pseudogene (PRPS1L2) on chromosome 9q33-q34.


Molecular Genetics

De Brouwer et al. (2010) provided a review of the clinical and molecular features of the 4 distinct syndromes caused by mutation in the PRPS1 gene: PRPS1 superactivity (300661), X-linked Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Arts syndrome (301835), and isolated X-linked sensorineural deafness (DFNX1; 304500). The neurologic phenotype in all 4 PRPS1-related disorders seems to result primarily from reduced levels of GTP and possibly other purine nucleotides including ATP, suggesting that these disorders belong to the same disease spectrum. Preliminary results of S-adenosylmethionine (SAM) supplementation in 2 Australian brothers with Arts syndrome revealed some improvement of their condition, suggesting that SAM supplementation could potentially alleviate some of the symptoms of patients with PRPS1 spectrum diseases by replenishing purine nucleotides.

Phosphoribosylpyrophosphate Synthetase Superactivity

In patients with phosphoribosylpyrophosphate synthetase superactivity (300661), Roessler et al. (1991, 1993) and Becker et al. (1995) identified mutations in the PRPS1 gene (311850.0001-311850.0008). All patients except 1 had hyperuricemia, neurodevelopmental abnormalities, and sensorineural deafness; the other patient had only hyperuricemia and gout. Functional expression studies of all mutations showed that enzyme overactivity was due to alteration of allosteric feedback mechanisms.

Charcot-Marie-Tooth Disease, X-linked Recessive, 5

In affected males with X-linked recessive Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Kim et al. (2007) identified mutations in the PRPS1 gene (311850.0009; 311850.0010). The phenotype includes peripheral neuropathy, sensorineural deafness, and visual impairment. Kim et al. (2007) used a positional cloning technique and evaluation of candidate genes known to be expressed in the cochlea to identify the PRPS1 gene for study. The mutations were shown to result in decreased enzyme activity; none of the affected individuals had increased uric acid or gout. Kim et al. (2007) noted that both PRPS1 superactivity and CMTX5 phenotypes share neurologic features.

Arts Syndrome

Arts syndrome (ARTS; 301835) is an X-linked disorder characterized by mental retardation, early-onset hypotonia, ataxia, delayed motor development, hearing impairment, and optic atrophy. Using oligonucleotide microarray expression profiling of fibroblasts from 2 probands in a Dutch family with Arts syndrome, de Brouwer et al. (2007) found reduced expression levels of PRPS1. Sequencing of PRPS1 led to the identification of 2 different missense mutations: L152P (311850.0011) in the Dutch family and Q133P (311850.0012) in the Australian family. Both mutations resulted in a loss of PRPS1 activity, as was shown in silico by molecular modeling and was shown in vitro by enzyme assays in erythrocytes and fibroblasts from patients. This was in contrast to the gain-of-function mutations in PRPS1 identified in PRPS-related gout. The loss-of-function mutations of PRPS1 probably result in impaired purine biosynthesis, which was supported by the undetectable hypoxanthine in urine and the reduced uric acid levels in serum from patients. De Brouwer et al. (2007) suggested that treatment with S-adenosylmethionine (SAM) theoretically could have therapeutic efficacy to replenish low levels of purine, and a clinical trial involving the 2 affected Australian brothers was underway. De Brouwer et al. (2010) reported preliminary results of the 2 Australian brothers with Arts syndrome.

In a German family with variable manifestations of PRPS1 deficiency, including a man with a protracted form of Arts syndrome and features of CMTX5 and his sister with X-linked deafness-1 (DFNX1; 304500), Synofzik et al. (2014) identified a missense mutation in the PRPS1 gene (Q277P; 311850.0019). The mother of these sibs, who had no hearing deficit or neurologic dysfunction at age 66, also carried the mutation; the mutation was heterozygous in the females and hemizygous in the male proband. Erythrocyte PRPS1 activity was not detectable in the proband, was decreased in the sister, and was normal in the mother. X-chromosome inactivation was extremely skewed in the sister with DFNX1 (94%; 6%), but only moderately skewed in the mother (80%; 20%). The findings illustrated that PRPS1 deficiency can present as a continuous spectrum of clinical features, even within the same family.

X-linked Deafness 1

In a large 5-generation Chinese family segregating X-linked nonsyndromic hearing loss (NSHL) mapping to the DFN2 locus (DFNX1; 304500) on chromosome Xq22, Liu et al. (2010) analyzed 14 candidate genes and identified a missense mutation in the PRPS1 gene (D65N; 311850.0013) that cosegregated with the phenotype. Analysis of the PRPS1 gene in a British American DFN2 family, previously reported by Tyson et al. (1996), revealed a different missense mutation (A87T; 311850.0014); missense mutations were also detected in DFN2 families previously reported by Manolis et al. (1999) and Cui et al. (2004) (311850.0015 and 311850.0016, respectively). Liu et al. (2010) stated that none of the mutations were predicted to result in a major structural change in the PRPS1 protein, which might explain why the disease phenotype was limited to NSHL.


Animal Model

In a study of 1,751 knockout alleles created by the International Mouse Phenotyping Consortium (IMPC), Dickinson et al. (2016) found that knockout of the mouse homolog of human PRPS1 is homozygous-lethal (defined as absence of homozygous mice after screening of at least 28 pups before weaning).


History

Wada et al. (1974) and Iinuma et al. (1975) reported a Japanese infant with mental retardation, hypouricemia, megaloblastic changes in the bone marrow, and orotic aciduria associated with erythrocyte PRPS deficiency. Hypsarrhythmia was first observed at 10 months of age and markedly improved with ACTH therapy concomitant with an increase in red cell PRPS activity. However, studies in fibroblasts from this patient did not confirm enzyme deficiency (Becker, 2001).


ALLELIC VARIANTS ( 23 Selected Examples):

.0001 PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, ASN114SER
  
RCV000010606...

In a boy with hyperuricemia, sensorineural deafness, ataxia, and secondary renal insufficiency associated with PRPS1 superactivity (300661) reported by Becker et al. (1986), Roessler et al. (1991, 1993) identified a 341A-G transition in the PRPS1 gene, resulting in an asn113-to-ser (N113S) substitution. Biochemical studies in fibroblasts were consistent with PRPS superactivity and purine nucleotide feedback-resistance. The nucleotide sequence of PRPS2 cDNA was normal. Becker et al. (1995) numbered the variant based on the mature protein.

By in vitro functional expression studies in E. coli, Becker et al. (1995) demonstrated that the N113S mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate.


.0002 PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, ASP183HIS
  
RCV000010607...

In a boy with hyperuricemia, mental retardation, and sensorineural deafness associated with PRPS1 superactivity (300661) reported by Becker et al. (1980), Roessler et al. (1991, 1993) identified a 547C-G transversion in the PRPS1 gene, resulting in an asp182-to-his (D182H) substitution. His affected mother had gout, uric acid urolithiasis, and significant hearing loss. The nucleotide sequence of PRPS2 cDNA was normal. Fibroblast studies of this patient and his mother (Becker et al., 1980) indicated that the mutant enzyme had both regulatory and catalytic defects. The enzyme showed 4- to 5-fold greater than normal resistance to feedback inhibition and, in addition, increased maximal velocity of the enzyme reaction. The son was hemizygous, and his mother heterozygous, for the defect. Becker et al. (1995) numbered the variant based on the mature protein.

By in vitro functional expression studies in E. coli, Becker et al. (1995) demonstrated that the D182H mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate.


.0003 PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, ASP52HIS
  
RCV000010608

In a man with gout due to PRPS1 superactivity (300661) resulting from purine nucleotide feedback-resistance (Zoref et al., 1975), Becker et al. (1995) identified a 154G-C transversion in the PRPS1 gene, resulting in an asp51-to-his (D51H) substitution. The patient had recurrent uric acid lithiasis since age 14 years and severe gouty arthritis since age 20 years. His mother had increased uric acid excretion. In vitro functional expression studies in E. coli demonstrated that the D51H mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate. Becker et al. (1995) numbered the variant based on the mature protein.


.0004 REMOVED FROM DATABASE


.0005 PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, LEU129ILE
  
RCV000010609

In a boy with hyperuricemia, mental retardation, and sensorineural deafness associated with PRPS1 superactivity (300661), Becker et al. (1995) identified a 385C-A transversion in the PRPS1 gene, resulting in a leu128-to-ile (L128I) substitution. In vitro functional expression studies in E. coli demonstrated that the L128I mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate. Becker et al. (1995) numbered the variant based on the mature protein.


.0006 REMOVED FROM DATABASE


.0007 PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, ALA190VAL
  
RCV000010610

In a boy with hyperuricemia, mental retardation, and sensorineural deafness associated with PRPS1 superactivity (300661), Becker et al. (1995) identified a 569C-T transition in the PRPS1 gene, resulting in an ala189-to-val (A189V) substitution. In vitro functional expression studies in E. coli demonstrated that the A189V mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate. Becker et al. (1995) numbered the variant based on the mature protein.


.0008 PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, HIS193GLN
  
RCV000010611

In a boy with hyperuricemia, mental retardation, and sensorineural deafness associated with PRPS1 superactivity (300661), Becker et al. (1995) identified a 579C-G transversion in the PRPS1 gene, resulting in a his192-to-gln (H192Q) substitution. In vitro functional expression studies in E. coli demonstrated that the H192Q mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate. Becker et al. (1995) numbered the variant based on the mature protein.


.0009 CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, GLU43ASP
  
RCV000010612

In 2 affected brothers with X-linked recessive Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Kim et al. (2007) identified a 129A-C transversion in exon 2 of the PRPS1 gene, resulting in a glu43-to-asp (E43D) substitution on the 'flag' region of the N-terminal domain. The affected residue is highly conserved from zebrafish to human, and the mutation was not observed in 50 unrelated Caucasian individuals or in 1,103 Korean control chromosomes. None of the affected individuals had increased uric acid production or gout.


.0010 CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, MET115THR
  
RCV000010613...

In affected members of a Korean family with X-linked Charcot-Marie-Tooth disease (CMTX5; 311070), Kim et al. (2007) identified a 344T-C transition in exon 3 of the PRPS1 gene, resulting in a met115-to-thr (M115T) substitution in the alpha-helix of the N-terminal domain. The affected residue is highly conserved from zebrafish to human, and the mutation was not observed in 1,103 Korean control chromosomes. In vitro functional expression studies showed that the M115T mutation resulted in partial loss of enzyme function. None of the affected individuals had increased uric acid production or gout.


.0011 ARTS SYNDROME

PRPS1, LEU152PRO
  
RCV000010614...

In a Dutch family with Arts syndrome (ARTS; 301835) originally reported by Arts et al. (1993), de Brouwer et al. (2007) found that the disorder was associated with a 455T-C transition in exon 4 of the PRPS1 gene that resulted in a leu152-to-pro (L152P) substitution.


.0012 ARTS SYNDROME

PRPS1, GLN133PRO
  
RCV000010615

In an Australian family with Arts syndrome (ARTS; 301835), de Brouwer et al. (2007) found that the disorder was caused by a 398A-C transversion in exon 3 of the PRPS1 gene that resulted in a gln133-to-pro (Q133P) substitution. Enzyme assays and molecular modeling demonstrated loss of function of the mutant protein.


.0013 DEAFNESS, X-LINKED 1

PRPS1, ASP65ASN
  
RCV000010616

In affected members of a large 5-generation Chinese family segregating X-linked deafness-1 (DFNX1; 304500), Liu et al. (2010) identified a 193G-A transition in exon 2 of the PRPS1 gene, resulting in an asp65-to-asn (D65N) substitution at a highly conserved residue in the alpha helix at the N terminus. The mutation was not found in 1,025 ethnically matched control chromosomes. Enzymatic activity assays showed reductions of PRPS1 activity in patient erythrocytes of approximately 40 to 70% and in patient fibroblasts of approximately 50 to 60% compared to controls.


.0014 DEAFNESS, X-LINKED 1

PRPS1, ALA87THR
  
RCV000010617...

In affected members of a British American family segregating X-linked deafness-1 (DFNX1; 304500), previously reported by Tyson et al. (1996), Liu et al. (2010) identified a 259G-A transition in exon 2 of the PRPS1 gene, resulting in an ala87-to-thr (A87T) substitution at a highly conserved residue. The mutation was not found in 1,475 Chinese control chromosomes or 450 chromosomes of European descent.


.0015 DEAFNESS, X-LINKED 1

PRPS1, GLY306ARG
  
RCV000010618

In affected members of an American family segregating X-linked deafness-1 (DFNX1; 304500), previously reported by Manolis et al. (1999), Liu et al. (2010) identified a 916G-A transition in exon 7 of the PRPS1 gene, resulting in a gly306-to-arg (G306R) substitution at a highly conserved residue. The mutation was not found in 1,475 Chinese control chromosomes or 450 chromosomes of European descent.


.0016 DEAFNESS, X-LINKED 1

PRPS1, ILE290THR
  
RCV000010619

In affected members from a Chinese family segregating X-linked postlingual nonsyndromic hearing loss (DFNX1; 304500), previously reported by Cui et al. (2004), Liu et al. (2010) identified an 869T-C transition in exon 7 of the PRPS1 gene, resulting in an ile290-to-thr (I290T) substitution at a highly conserved residue. The mutation was not found in 1,025 ethnically matched control chromosomes.


.0017 ARTS SYNDROME AND PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, VAL142LEU
  
RCV000022882

In a boy with a complex phenotype comprising Arts syndrome and PRPS1 superactivity (see 301835), Moran et al. (2012) identified a 424G-C transversion in exon 4 of the PRPS1 gene, resulting in a val142-to-leu (V142L) substitution at a highly conserved residue. Both the mother and grandmother were heterozygous for the mutation, which was not found in 202 control alleles. The patient had developmental delay, hypotonia, areflexia, motor neuropathy, sensorineural hearing loss, and a Chiari I malformation. Laboratory studies showed increased serum uric acid and increased urinary hypoxanthine consistent with PRPS1 superactivity, but he did not have gout. In addition, he had recurrent infections and early death at age 27 months from infection, consistent with Arts syndrome. A maternal uncle with similar symptoms had died of pneumonia at age 2. Molecular modeling predicted that the substitution would disrupt allosteric sites involved in inhibition of PRPS1, resulting in a gain of enzyme function, and the ATP-binding site, resulting in a loss of enzyme function. Patient fibroblasts showed normal PRPP synthetase activity, whereas erythrocytes showed a loss of enzyme activity, suggesting that the effect of the V142L mutation on protein activity depends on cell type. Moran et al. (2012) postulated a gain-of-function effect in proliferating cells and a loss-of-function effect in postmitotic cells. The report indicated that PRPS1 missense mutations can cause a continuous spectrum of features ranging from progressive nonsyndromic postlingual hearing impairment to uric acid overproduction, neuropathy, and recurrent infections depending on the functional sites affected.


.0018 CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, ALA121GLY
  
RCV000087131

In a young Korean man with X-linked recessive Charcot-Marie-Tooth disease (CMTX5; 311070) with early-onset sensorineural hearing loss but without optic atrophy or visual disturbance, Park et al. (2013) identified a hemizygous c.362C-G transversion in exon 3 of the PRPS1 gene, resulting in an ala121-to-gly (A121G) substitution at a highly conserved residue. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the dbSNP (build 135) or 1000 Genomes Project databases, or in 250 healthy controls. The patient's unaffected mother was heterozygous for the mutation. Two maternally related male relatives had a similar disorder, but DNA was not available from these individuals. Functional studies of the variant were not performed.


.0019 ARTS SYNDROME

DEAFNESS, X-LINKED 1, INCLUDED
PRPS1, GLN277PRO
  
RCV000208721...

In a German family with variable manifestations of PRPS1 deficiency, including a man with a protracted form of Arts syndrome (ARTS; 301835) and his sister with X-linked deafness-1 (DFNX1; 304500), Synofzik et al. (2014) identified a c.830A-C transversion in the PRPS1 gene, resulting in a gln277-to-pro (Q277P) substitution at a highly conserved residue in the C-terminal domain close to the catalytic site. The mutation was not found in the Exome Variant Server database or in 4,200 ethnically matched control X chromosomes. The mutation was predicted to disrupt the region surrounding the ribose-5-phosphate binding site and thus affect the catalytic site. The mother of these sibs, who had no hearing deficit or neurologic dysfunction at age 66, also carried the mutation; the mutation was heterozygous in the females and hemizygous in the male proband. Erythrocyte PRPS1 activity was not detectable in the proband, was decreased in the sister, and was normal in the mother. X-chromosome inactivation was extremely skewed in the sister with DFNX1 (94%; 6%), but only moderately skewed in the mother (80%; 20%). The findings illustrated that PRPS1 deficiency can present as a continuous spectrum of clinical features, even within the same family.


.0020 CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, SER16PRO
  
RCV000208733

In 4 females from a 3-generation Spanish family (RP-0482) with variable manifestations of X-linked recessive Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Almoguera et al. (2014) identified a heterozygous c.46T-C transition (c.46T-C, NM002764) in exon 1 of the PRPS1 gene, resulting in a ser16-to-pro (S16P) substitution at a conserved residue in the first alpha helix in the N-terminal domain. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was filtered against the 1000 Genomes Project (April 2012 release) and Exome Sequencing Project databases and 669 in-house exomes. The mutation segregated with the disorder in the family and was not found in 258 control X chromosomes. Erythrocyte PRPS1 activity from 3 affected females showed variably decreased levels, which correlated with the age at onset. The proband (IV-3), who was the most severely affected, had significantly skewed X inactivation (82%) of the paternal allele and showed lack of expression of the wildtype allele in lymphocyte mRNA. The proband's sister and mother, who carried the mutation but were less severely affected clinically, did not show significantly skewed X inactivation.


.0021 DEAFNESS, X-LINKED 1

PRPS1, ALA113SER
  
RCV000143857

In 2 Italian brothers (family 1) with postlingual X-linked deafness-1 (DFNX1; 304500), Robusto et al. (2015) identified a hemizygous c.337G-T transversion (c.337G-T, NM_002764.3) in exon 3 of the PRPS1 gene, resulting in an ala113-to-ser (A113S) substitution in an alpha-helix participating in trimer interface, predicted to destabilize the surrounding environment and possibly affect ATP binding. The mother, who had late-onset moderate hearing loss, was heterozygous for the mutation. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was filtered against the dbSNP (build 130), 1000 Genomes Project, and Exome Variant Server databases and was not found in 123 Italian controls. Erythrocyte PRPS1 activity was mildly decreased in the 2 affected males (25-35% of normal controls).


.0022 CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, MET115VAL
  
RCV000143858...

In affected members of an Italian family (family 2) with X-linked recessive Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Robusto et al. (2015) identified a c.343A-G transition (c.343A-G, NM_002764.3) in exon 3 of the PRPS1 gene, resulting in a met115-to-val (M115V) substitution at a highly conserved residue in an alpha-helix participating in trimer interface, predicted to destabilize the ATP-binding site and the allosteric site I. The mutation segregated with the disorder in the family and was not found in the dbSNP (build 130), 1000 Genomes Project, or Exome Variant Server databases. The family was ascertained through a 12-year-old male who had sensorineural hearing loss; subsequent examination of family members showed that those with the mutation had hearing loss as well as subclinical signs of a peripheral neuropathy, which was more evident in males compared to females. Erythrocyte PRPS1 activity was significantly decreased in the 2 affected males (5-10% of normal controls) and mildly decreased in 3 carrier females (34.7-47.7% of normal controls). X-inactivation studies in 1 carrier female showed a normal ratio, but there was preferential expression of the mutant allele (60% vs 40%).


.0023 CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, VAL309PHE
  
RCV000143860

In affected members of a Peruvian family (family 3) with X-linked recessive Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Robusto et al. (2015) identified a c.925G-T transversion (c.925G-T, NM_002764.3) in exon 7 of the PRPS1 gene, resulting in a val309-to-phe (V309F) substitution at a highly conserved residue at the trimer interface, predicted to disturb the allosteric type I function as well as hexameric assembly. The mutation segregated with the disorder in the family and was not found in the dbSNP (build 130), 1000 Genomes Project, or Exome Variant Server databases. The family was ascertained through a 14-year-old male who had sensorineural hearing loss and was subsequently found to have subclinical signs of a peripheral neuropathy. The patient's uncle and mother also carried the mutation; the uncle had sensorineural hearing loss and subtle peripheral neuropathy, whereas the mother had mild peripheral neuropathy without hearing loss. Erythrocyte PRPS1 activity was significantly decreased in the male proband (4.05% of normal controls) and mildly decreased in the mother (18.3% of normal controls). The mother had skewed X inactivation with almost exclusive expression of the wildtype allele (85% vs 15%).


See Also:

REFERENCES

  1. Almoguera, B., He, S., Corton, M., Fernandez-San Jose, P., Blanco-Kelly, F., Lopez-Molina, M. I., Garcia-Sandoval, B., del Val, J., Guo, Y., Tian, L., Liu, X., Guan, L., Torres, R. J., Puig, J. G., Hakonarson, H., Xu, X., Keating, B., Ayuso, C. Expanding the phenotype of PRPS1 syndromes in females: neuropathy, hearing loss and retinopathy. Orphanet J. Rare Dis. 9: 190, 2014. Note: Electronic Article. [PubMed: 25491489, images, related citations] [Full Text]

  2. Arts, W. F. M., Loonen, M. C. B., Sengers, R. C. A., Slooff, J. L. X-linked ataxia, weakness, deafness, and loss of vision in early childhood with a fatal course. Ann. Neurol. 33: 535-539, 1993. [PubMed: 8498830, related citations] [Full Text]

  3. Becker, M. A., Heidler, S. A., Bell, G. I., Seino, S., Le Beau, M. M., Westbrook, C. A., Neuman, W., Shapiro, L. J., Mohandas, T. K., Roessler, B. J., Palella, T. D. Cloning of cDNAs for human phosphoribosylpyrophosphate synthetases 1 and 2 and X chromosome localization of PRPS1 and PRPS2 genes. Genomics 8: 555-561, 1990. [PubMed: 1962753, related citations] [Full Text]

  4. Becker, M. A., Losman, M. J., Wilson, J., Simmonds, H. A. Superactivity of human phosphoribosyl pyrophosphate synthetase due to altered regulation by nucleotide inhibitors and inorganic phosphate. Biochim. Biophys. Acta 882: 168-176, 1986. [PubMed: 2423135, related citations] [Full Text]

  5. Becker, M. A., Raivio, K. O., Bakay, B., Adams, W. B., Nyhan, W. L. Variant human phosphoribosylpyrophosphate synthetase altered in regulatory and catalytic functions. J. Clin. Invest. 65: 109-120, 1980. [PubMed: 6243137, related citations] [Full Text]

  6. Becker, M. A., Smith, P. R., Taylor, W., Mustafi, R., Switzer, R. L. The genetic and functional basis of purine nucleotide feedback-resistant phosphoribosylpyrophosphate synthetase superactivity. J. Clin. Invest. 96: 2133-2141, 1995. [PubMed: 7593598, related citations] [Full Text]

  7. Becker, M. A., Yen, R. C. K., Goss, S. J., Seegmiller, J. E., Itkin, P., Lazar, C., Adams, W. B. Localization of the structural gene for human phosphoribosylpyrophosphate synthetase on the X-chromosome. (Abstract) Clin. Res. 26: 500A, 1978.

  8. Becker, M. A., Yen, R. C. K., Itkin, P., Goss, S. J., Seegmiller, J. E., Bakay, B. Regional localization of the gene for human phosphoribosylpyrophosphate synthetase on the X-chromosome. Science 203: 1016-1019, 1979. [PubMed: 218284, related citations] [Full Text]

  9. Becker, M. A. Hyperuricemia and Gout. In: Scriver, C. R.; Beaudet, A. L.; Sly, W. S.; Valle, D. (eds.): The Metabolic and Molecular Bases of Inherited Disease. Vol. II. (8th ed.) New York: McGraw-Hill (pub.) 2001. P. 2625.

  10. Cui, B., Zhang, H., Lu, Y., Zhong, W., Pei, G., Kong, X., Hu, L. Refinement of the locus for non-syndromic sensorineural deafness (DFN2) J. Genet. 83: 35-38, 2004. [PubMed: 15240907, related citations] [Full Text]

  11. de Brouwer, A. P. M., van Bokhoven, H., Nabuurs, S. B., Arts, W. F., Christodoulou, J., Duley, J. PRPS1 mutations: four distinct syndromes and potential treatment. Am. J. Hum. Genet. 86: 506-518, 2010. [PubMed: 20380929, images, related citations] [Full Text]

  12. de Brouwer, A. P. M., Williams, K. L., Duley, J. A., van Kuilenburg, A. B. P., Nabuurs, S. B., Egmont-Petersen, M., Lugtenberg, D., Zoetekouw, L., Banning, M. J. G., Roeffen, M., Hamel, B. C. J., Weaving, L., Ouvrier, R. A., Donald, J. A., Wevers, R. A., Christodoulou, J., van Bokhoven, H. Arts syndrome is caused by loss-of-function mutations in PRPS1. Am. J. Hum. Genet. 81: 507-518, 2007. [PubMed: 17701896, images, related citations] [Full Text]

  13. Dickinson, M. E., Flenniken, A. M., Ji, X., Teboul, L., Wong, M. D., White, J. K., Meehan, T. F., Weninger, W. J., Westerberg, H., Adissu, H., Baker, C. N., Bower, L., and 73 others. High-throughput discovery of novel developmental phenotypes. Nature 537: 508-514, 2016. Note: Erratum: Nature 551: 398 only, 2017. [PubMed: 27626380, images, related citations] [Full Text]

  14. Iinuma, K., Wada, Y., Onuma, A., Tanabu, M. Electroencephalographic study of an infant with phosphoribosylpyrophosphate synthetase deficiency. Tohoku J. Exp. Med. 116: 53-55, 1975. [PubMed: 168665, related citations] [Full Text]

  15. Kim, H.-J., Sohn, K.-M., Shy, M. E., Krajewski, K. M., Hwang, M., Park, J.-H., Jang, S.-Y., Won, H.-H., Choi, B.-O., Hong, S. H., Kim, B.-J., Suh, Y.-L., Ki, C.-S., Lee, S.-Y., Kim, S.-H., Kim, J.-W. Mutations in PRPS1, which encodes the phosphoribosyl pyrophosphate synthetase enzyme critical for nucleotide biosynthesis, cause hereditary peripheral neuropathy with hearing loss and optic neuropathy (CMT5X). Am. J. Hum. Genet. 81: 552-558, 2007. [PubMed: 17701900, images, related citations] [Full Text]

  16. Lebo, R. V., Martin, D. W., Jr. Electrophoretic heterogeneity of 5-phosphoribosyl-1-pyrophosphate synthetase within and among humans. Biochem. Genet. 16: 905-916, 1978. [PubMed: 217337, related citations] [Full Text]

  17. Liu, X., Han, D., Li, J., Han, B., Ouyang, X., Cheng, J., Li, X., Jin, Z., Wang, Y., Bitner-Glindzicz, M., Kong, X., Xu, H., and 10 others. Loss-of-function mutations in the PRPS1 gene cause a type of nonsyndromic X-linked sensorineural deafness, DFN2. Am. J. Hum. Genet. 86: 65-71, 2010. [PubMed: 20021999, images, related citations] [Full Text]

  18. Manolis, E. N., Eavey, R. D., Sangwatanaroj, S., Halpin, C., Rosenbaum, S., Watkins, H., Jarcho, J., Seidman, C. E., Seidman, J. G. Hereditary postlingual sensorineural hearing loss mapping to chromosome Xq21. Am. J. Otol. 20: 621-626, 1999. [PubMed: 10503584, related citations]

  19. Moran, R., Kuilenburg, A. B. P., Duley, J., Nabuurs, S. B., Retno-Fitri, A., Christodoulou, J., Roelofsen, J., Yntema, H. G., Friedman, N. R., van Bokhoven, H., de Brouwer, A. P. M. Phosphoribosylpyrophosphate synthetase superactivity and recurrent infections is caused by a p.val142-to-leu mutation in PRS-I. Am. J. Med. Genet. 158A: 455-460, 2012. [PubMed: 22246954, related citations] [Full Text]

  20. Park, J., Hyun, Y. S., Kim, Y. J., Nam, S. H., Kim, S., Hong, Y. B., Park, J.-M., Chung, K. W., Choi, B.-O. Exome sequencing reveals a novel PRPS1 mutation in a family with CMTX5 without optic atrophy. J. Clin. Neurol. 9: 283-288, 2013. [PubMed: 24285972, related citations] [Full Text]

  21. Robusto, M., Fang, M., Asselta, R., Castorina, P., Previtali, S. C., Caccia, S., Benzoni, E., De Cristofaro, R., Yu, C., Cesarani, A., Liu, X., Li, W., Primignani, P., Ambrosetti, U., Xu, X., Duga, S., Solda, G. The expanding spectrum of PRPS1-associated phenotypes: three novel mutations segregating with X-linked hearing loss and mild peripheral neuropathy. Europ. J. Hum. Genet. 23: 766-773, 2015. [PubMed: 25182139, related citations] [Full Text]

  22. Roessler, B. J., Bell, G., Heidler, S., Seino, S., Becker, M., Palella, T. D. Cloning of two distinct copies of human phosphoribosylpyrophosphate synthetase cDNA. Nucleic Acids Res. 18: 193 only, 1990. [PubMed: 2155397, related citations] [Full Text]

  23. Roessler, B. J., Nosal, J. M., Smith, P. R., Heidler, S. A., Palella, T. D., Switzer, R. L., Becker, M. A. Human X-linked phosphoribosylpyrophosphate synthetase superactivity is associated with distinct point mutations in the PRPS1 gene. J. Biol. Chem. 268: 26476-26481, 1993. [PubMed: 8253776, related citations]

  24. Roessler, B. J., Palella, T. D., Heidler, S., Becker, M. A. Identification of distinct PRPS1 mutations in two patients with X-linked phosphoribosylpyrophosphate synthetase superactivity. (Abstract) Clin. Res. 39: 267A, 1991.

  25. Synofzik, M., Muller vom Hagen, J., Haack, T. B., Wilhelm, C., Lindig, T., Beck-Wodl, S., Nabuurs, S. B., van Kuilenburg, A. B. P., de Brouwer, A. P. M., Schols, L. X-linked Charcot-Marie-Tooth disease, Arts syndrome, and prelingual non-syndromic deafness form a disease continuum: evidence from a family with a novel PRPS1 mutation. Orphanet J. Rare Dis. 9: 24, 2014. Note: Electronic Article. [PubMed: 24528855, images, related citations] [Full Text]

  26. Taira, M., Iizasa, T., Yamada, K., Shimada, H., Tatibana, M. Tissue-differential expression of two distinct genes for phosphoribosyl pyrophosphate synthetase and existence of the testis-specific transcript. Biochim. Biophys. Acta 1007: 203-208, 1989. [PubMed: 2537655, related citations] [Full Text]

  27. Tyson, J., Bellman, S., Newton, V., Simpson, P., Malcolm, S., Pembrey, M. E., Bitner-Glindzicz, M. Mapping of DFN2 to Xq22. Hum. Molec. Genet. 5: 2055-2060, 1996. [PubMed: 8968763, related citations] [Full Text]

  28. Wada, Y., Nishimura, Y., Tanabu, M., Yoshimura, Y., Iinuma, K., Yoshida, T., Arakawa, T. Hypouricemic, mentally retarded infant with a defect of 5-phosphoribosyl-1-pyrophosphate synthetase of erythrocytes. Tohoku J. Exp. Med. 113: 149-157, 1974. [PubMed: 4373874, related citations] [Full Text]

  29. Zoref, E., De Vries, A., Sperling, O. Mutant feedback-resistant phosphoribosylpyrophosphate synthetase associated with purine overproduction and gout: phosphoribosylpyrophosphate and purine metabolism in cultured fibroblasts. J. Clin. Invest. 56: 1093-1099, 1975. [PubMed: 171280, related citations] [Full Text]


Ada Hamosh - updated : 02/21/2017
Cassandra L. Kniffin - updated : 2/10/2016
Cassandra L. Kniffin - updated : 2/19/2014
Cassandra L. Kniffin - updated : 4/9/2012
Cassandra L. Kniffin - updated : 4/30/2010
Marla J. F. O'Neill - updated : 2/26/2010
Patricia A. Hartz - updated : 10/24/2007
Victor A. McKusick - updated : 8/22/2007
Cassandra L. Kniffin - reorganized : 8/17/2007
Cassandra L. Kniffin - updated : 8/16/2007
Rebekah S. Rasooly - updated : 2/26/1999
Creation Date:
Victor A. McKusick : 6/4/1986
alopez : 03/13/2023
carol : 01/29/2018
alopez : 02/21/2017
alopez : 09/23/2016
carol : 07/09/2016
carol : 3/1/2016
carol : 3/1/2016
ckniffin : 2/10/2016
carol : 1/13/2016
carol : 2/25/2014
mcolton : 2/21/2014
ckniffin : 2/19/2014
alopez : 4/20/2012
alopez : 4/13/2012
alopez : 4/12/2012
terry : 4/10/2012
ckniffin : 4/9/2012
carol : 4/12/2011
wwang : 5/4/2010
ckniffin : 4/30/2010
terry : 4/2/2010
carol : 2/26/2010
mgross : 10/30/2007
terry : 10/24/2007
alopez : 8/22/2007
carol : 8/17/2007
ckniffin : 8/16/2007
alopez : 9/10/2004
terry : 6/25/2004
psherman : 3/1/1999
psherman : 2/26/1999
dkim : 7/7/1998
terry : 11/18/1996
terry : 11/4/1996
mark : 2/2/1996
terry : 1/26/1996
terry : 5/10/1994
warfield : 4/20/1994
carol : 4/11/1994
mimadm : 2/28/1994
carol : 12/16/1992
carol : 12/3/1992

* 311850

PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE I; PRPS1


HGNC Approved Gene Symbol: PRPS1

SNOMEDCT: 702441001, 723454008, 763460007;  


Cytogenetic location: Xq22.3     Genomic coordinates (GRCh38): X:107,628,510-107,651,026 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Xq22.3 Arts syndrome 301835 X-linked recessive 3
Charcot-Marie-Tooth disease, X-linked recessive, 5 311070 X-linked recessive 3
Deafness, X-linked 1 304500 X-linked 3
Gout, PRPS-related 300661 X-linked recessive 3
Phosphoribosylpyrophosphate synthetase superactivity 300661 X-linked recessive 3

TEXT

Description

Phosphoribosylpyrophosphate synthetase (PRPS; EC 2.7.6.1) catalyzes the phosphoribosylation of ribose 5-phosphate to 5-phosphoribosyl-1-pyrophosphate, which is necessary for the de novo and salvage pathways of purine and pyrimidine biosynthesis (Roessler et al., 1990). Three PRPS genes have been identified: the widely expressed PRPS1 and PRPS2 (311860) genes, which map to chromosome Xq22-q24 and Xp22, respectively, and PRPS3 (PRPS1L1; 611566), which maps to chromosome 7 and appears to be transcribed only in testis (Becker, 2001).


Cloning and Expression

Roessler et al. (1990) isolated a partial clone corresponding to the PRPS1 gene from a human lymphoblast cDNA library. The deduced PRPS1 protein has 318 amino acids and shares 95% amino acid homology with PRPS2. Becker et al. (1990) also cloned the PRPS1 gene and detected a 2.3-kb mRNA transcript.

By Northern blot analysis using rat Prps1 as probe, Taira et al. (1989) detected a 2.3-kb transcript in human adipose tissue, testis, and placenta and in 2 human cell lines.

Kim et al. (2007) demonstrated that the PRPS1 amino acid sequence shows an exceptionally high degree of conservation, with homologies greater than 95% across different species from zebrafish to human.


Gene Structure

The PRPS1 gene spans over 30 kb and contains 7 exons (Becker, 2001).


Mapping

By the Goss-Harris method, Becker et al. (1978) concluded that the order of loci on chromosome Xq is G6PD (305900)--HPRT1 (308000)--PRPS1--alpha-GAL (GLA; 300644)--PGK1 (311800)--centromere. Becker et al. (1979) assigned the PRPS1 locus to a position between the GLA and HPRT1 loci, particularly close to the latter, and discussed the functional significance of the proximity of the genes for their biochemically related functions.

Becker et al. (1990) mapped PRPS1 to Xq22-q24 by a combination of in situ hybridization and study of human/rodent somatic cell hybrids.

Pseudogene

By in situ chromosomal hybridization, Becker et al. (1990) identified a PRPS1-related gene or pseudogene (PRPS1L2) on chromosome 9q33-q34.


Molecular Genetics

De Brouwer et al. (2010) provided a review of the clinical and molecular features of the 4 distinct syndromes caused by mutation in the PRPS1 gene: PRPS1 superactivity (300661), X-linked Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Arts syndrome (301835), and isolated X-linked sensorineural deafness (DFNX1; 304500). The neurologic phenotype in all 4 PRPS1-related disorders seems to result primarily from reduced levels of GTP and possibly other purine nucleotides including ATP, suggesting that these disorders belong to the same disease spectrum. Preliminary results of S-adenosylmethionine (SAM) supplementation in 2 Australian brothers with Arts syndrome revealed some improvement of their condition, suggesting that SAM supplementation could potentially alleviate some of the symptoms of patients with PRPS1 spectrum diseases by replenishing purine nucleotides.

Phosphoribosylpyrophosphate Synthetase Superactivity

In patients with phosphoribosylpyrophosphate synthetase superactivity (300661), Roessler et al. (1991, 1993) and Becker et al. (1995) identified mutations in the PRPS1 gene (311850.0001-311850.0008). All patients except 1 had hyperuricemia, neurodevelopmental abnormalities, and sensorineural deafness; the other patient had only hyperuricemia and gout. Functional expression studies of all mutations showed that enzyme overactivity was due to alteration of allosteric feedback mechanisms.

Charcot-Marie-Tooth Disease, X-linked Recessive, 5

In affected males with X-linked recessive Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Kim et al. (2007) identified mutations in the PRPS1 gene (311850.0009; 311850.0010). The phenotype includes peripheral neuropathy, sensorineural deafness, and visual impairment. Kim et al. (2007) used a positional cloning technique and evaluation of candidate genes known to be expressed in the cochlea to identify the PRPS1 gene for study. The mutations were shown to result in decreased enzyme activity; none of the affected individuals had increased uric acid or gout. Kim et al. (2007) noted that both PRPS1 superactivity and CMTX5 phenotypes share neurologic features.

Arts Syndrome

Arts syndrome (ARTS; 301835) is an X-linked disorder characterized by mental retardation, early-onset hypotonia, ataxia, delayed motor development, hearing impairment, and optic atrophy. Using oligonucleotide microarray expression profiling of fibroblasts from 2 probands in a Dutch family with Arts syndrome, de Brouwer et al. (2007) found reduced expression levels of PRPS1. Sequencing of PRPS1 led to the identification of 2 different missense mutations: L152P (311850.0011) in the Dutch family and Q133P (311850.0012) in the Australian family. Both mutations resulted in a loss of PRPS1 activity, as was shown in silico by molecular modeling and was shown in vitro by enzyme assays in erythrocytes and fibroblasts from patients. This was in contrast to the gain-of-function mutations in PRPS1 identified in PRPS-related gout. The loss-of-function mutations of PRPS1 probably result in impaired purine biosynthesis, which was supported by the undetectable hypoxanthine in urine and the reduced uric acid levels in serum from patients. De Brouwer et al. (2007) suggested that treatment with S-adenosylmethionine (SAM) theoretically could have therapeutic efficacy to replenish low levels of purine, and a clinical trial involving the 2 affected Australian brothers was underway. De Brouwer et al. (2010) reported preliminary results of the 2 Australian brothers with Arts syndrome.

In a German family with variable manifestations of PRPS1 deficiency, including a man with a protracted form of Arts syndrome and features of CMTX5 and his sister with X-linked deafness-1 (DFNX1; 304500), Synofzik et al. (2014) identified a missense mutation in the PRPS1 gene (Q277P; 311850.0019). The mother of these sibs, who had no hearing deficit or neurologic dysfunction at age 66, also carried the mutation; the mutation was heterozygous in the females and hemizygous in the male proband. Erythrocyte PRPS1 activity was not detectable in the proband, was decreased in the sister, and was normal in the mother. X-chromosome inactivation was extremely skewed in the sister with DFNX1 (94%; 6%), but only moderately skewed in the mother (80%; 20%). The findings illustrated that PRPS1 deficiency can present as a continuous spectrum of clinical features, even within the same family.

X-linked Deafness 1

In a large 5-generation Chinese family segregating X-linked nonsyndromic hearing loss (NSHL) mapping to the DFN2 locus (DFNX1; 304500) on chromosome Xq22, Liu et al. (2010) analyzed 14 candidate genes and identified a missense mutation in the PRPS1 gene (D65N; 311850.0013) that cosegregated with the phenotype. Analysis of the PRPS1 gene in a British American DFN2 family, previously reported by Tyson et al. (1996), revealed a different missense mutation (A87T; 311850.0014); missense mutations were also detected in DFN2 families previously reported by Manolis et al. (1999) and Cui et al. (2004) (311850.0015 and 311850.0016, respectively). Liu et al. (2010) stated that none of the mutations were predicted to result in a major structural change in the PRPS1 protein, which might explain why the disease phenotype was limited to NSHL.


Animal Model

In a study of 1,751 knockout alleles created by the International Mouse Phenotyping Consortium (IMPC), Dickinson et al. (2016) found that knockout of the mouse homolog of human PRPS1 is homozygous-lethal (defined as absence of homozygous mice after screening of at least 28 pups before weaning).


History

Wada et al. (1974) and Iinuma et al. (1975) reported a Japanese infant with mental retardation, hypouricemia, megaloblastic changes in the bone marrow, and orotic aciduria associated with erythrocyte PRPS deficiency. Hypsarrhythmia was first observed at 10 months of age and markedly improved with ACTH therapy concomitant with an increase in red cell PRPS activity. However, studies in fibroblasts from this patient did not confirm enzyme deficiency (Becker, 2001).


ALLELIC VARIANTS 23 Selected Examples):

.0001   PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, ASN114SER
SNP: rs137852540, ClinVar: RCV000010606, RCV000622306, RCV001328985

In a boy with hyperuricemia, sensorineural deafness, ataxia, and secondary renal insufficiency associated with PRPS1 superactivity (300661) reported by Becker et al. (1986), Roessler et al. (1991, 1993) identified a 341A-G transition in the PRPS1 gene, resulting in an asn113-to-ser (N113S) substitution. Biochemical studies in fibroblasts were consistent with PRPS superactivity and purine nucleotide feedback-resistance. The nucleotide sequence of PRPS2 cDNA was normal. Becker et al. (1995) numbered the variant based on the mature protein.

By in vitro functional expression studies in E. coli, Becker et al. (1995) demonstrated that the N113S mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate.


.0002   PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, ASP183HIS
SNP: rs137852541, ClinVar: RCV000010607, RCV003147280

In a boy with hyperuricemia, mental retardation, and sensorineural deafness associated with PRPS1 superactivity (300661) reported by Becker et al. (1980), Roessler et al. (1991, 1993) identified a 547C-G transversion in the PRPS1 gene, resulting in an asp182-to-his (D182H) substitution. His affected mother had gout, uric acid urolithiasis, and significant hearing loss. The nucleotide sequence of PRPS2 cDNA was normal. Fibroblast studies of this patient and his mother (Becker et al., 1980) indicated that the mutant enzyme had both regulatory and catalytic defects. The enzyme showed 4- to 5-fold greater than normal resistance to feedback inhibition and, in addition, increased maximal velocity of the enzyme reaction. The son was hemizygous, and his mother heterozygous, for the defect. Becker et al. (1995) numbered the variant based on the mature protein.

By in vitro functional expression studies in E. coli, Becker et al. (1995) demonstrated that the D182H mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate.


.0003   PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, ASP52HIS
SNP: rs137852542, ClinVar: RCV000010608

In a man with gout due to PRPS1 superactivity (300661) resulting from purine nucleotide feedback-resistance (Zoref et al., 1975), Becker et al. (1995) identified a 154G-C transversion in the PRPS1 gene, resulting in an asp51-to-his (D51H) substitution. The patient had recurrent uric acid lithiasis since age 14 years and severe gouty arthritis since age 20 years. His mother had increased uric acid excretion. In vitro functional expression studies in E. coli demonstrated that the D51H mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate. Becker et al. (1995) numbered the variant based on the mature protein.


.0004   REMOVED FROM DATABASE


.0005   PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, LEU129ILE
SNP: rs137852543, ClinVar: RCV000010609

In a boy with hyperuricemia, mental retardation, and sensorineural deafness associated with PRPS1 superactivity (300661), Becker et al. (1995) identified a 385C-A transversion in the PRPS1 gene, resulting in a leu128-to-ile (L128I) substitution. In vitro functional expression studies in E. coli demonstrated that the L128I mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate. Becker et al. (1995) numbered the variant based on the mature protein.


.0006   REMOVED FROM DATABASE


.0007   PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, ALA190VAL
SNP: rs137852544, ClinVar: RCV000010610

In a boy with hyperuricemia, mental retardation, and sensorineural deafness associated with PRPS1 superactivity (300661), Becker et al. (1995) identified a 569C-T transition in the PRPS1 gene, resulting in an ala189-to-val (A189V) substitution. In vitro functional expression studies in E. coli demonstrated that the A189V mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate. Becker et al. (1995) numbered the variant based on the mature protein.


.0008   PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, HIS193GLN
SNP: rs137852545, ClinVar: RCV000010611

In a boy with hyperuricemia, mental retardation, and sensorineural deafness associated with PRPS1 superactivity (300661), Becker et al. (1995) identified a 579C-G transversion in the PRPS1 gene, resulting in a his192-to-gln (H192Q) substitution. In vitro functional expression studies in E. coli demonstrated that the H192Q mutation resulted in alteration of the allosteric mechanisms regulating both enzyme inhibition by purine nucleotides and activation by inorganic phosphate. Becker et al. (1995) numbered the variant based on the mature protein.


.0009   CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, GLU43ASP
SNP: rs80338731, ClinVar: RCV000010612

In 2 affected brothers with X-linked recessive Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Kim et al. (2007) identified a 129A-C transversion in exon 2 of the PRPS1 gene, resulting in a glu43-to-asp (E43D) substitution on the 'flag' region of the N-terminal domain. The affected residue is highly conserved from zebrafish to human, and the mutation was not observed in 50 unrelated Caucasian individuals or in 1,103 Korean control chromosomes. None of the affected individuals had increased uric acid production or gout.


.0010   CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, MET115THR
SNP: rs80338732, ClinVar: RCV000010613, RCV000695028

In affected members of a Korean family with X-linked Charcot-Marie-Tooth disease (CMTX5; 311070), Kim et al. (2007) identified a 344T-C transition in exon 3 of the PRPS1 gene, resulting in a met115-to-thr (M115T) substitution in the alpha-helix of the N-terminal domain. The affected residue is highly conserved from zebrafish to human, and the mutation was not observed in 1,103 Korean control chromosomes. In vitro functional expression studies showed that the M115T mutation resulted in partial loss of enzyme function. None of the affected individuals had increased uric acid production or gout.


.0011   ARTS SYNDROME

PRPS1, LEU152PRO
SNP: rs80338676, ClinVar: RCV000010614, RCV000178182

In a Dutch family with Arts syndrome (ARTS; 301835) originally reported by Arts et al. (1993), de Brouwer et al. (2007) found that the disorder was associated with a 455T-C transition in exon 4 of the PRPS1 gene that resulted in a leu152-to-pro (L152P) substitution.


.0012   ARTS SYNDROME

PRPS1, GLN133PRO
SNP: rs80338675, ClinVar: RCV000010615

In an Australian family with Arts syndrome (ARTS; 301835), de Brouwer et al. (2007) found that the disorder was caused by a 398A-C transversion in exon 3 of the PRPS1 gene that resulted in a gln133-to-pro (Q133P) substitution. Enzyme assays and molecular modeling demonstrated loss of function of the mutant protein.


.0013   DEAFNESS, X-LINKED 1

PRPS1, ASP65ASN
SNP: rs180177151, ClinVar: RCV000010616

In affected members of a large 5-generation Chinese family segregating X-linked deafness-1 (DFNX1; 304500), Liu et al. (2010) identified a 193G-A transition in exon 2 of the PRPS1 gene, resulting in an asp65-to-asn (D65N) substitution at a highly conserved residue in the alpha helix at the N terminus. The mutation was not found in 1,025 ethnically matched control chromosomes. Enzymatic activity assays showed reductions of PRPS1 activity in patient erythrocytes of approximately 40 to 70% and in patient fibroblasts of approximately 50 to 60% compared to controls.


.0014   DEAFNESS, X-LINKED 1

PRPS1, ALA87THR
SNP: rs180177152, ClinVar: RCV000010617, RCV001851782

In affected members of a British American family segregating X-linked deafness-1 (DFNX1; 304500), previously reported by Tyson et al. (1996), Liu et al. (2010) identified a 259G-A transition in exon 2 of the PRPS1 gene, resulting in an ala87-to-thr (A87T) substitution at a highly conserved residue. The mutation was not found in 1,475 Chinese control chromosomes or 450 chromosomes of European descent.


.0015   DEAFNESS, X-LINKED 1

PRPS1, GLY306ARG
SNP: rs180177154, ClinVar: RCV000010618

In affected members of an American family segregating X-linked deafness-1 (DFNX1; 304500), previously reported by Manolis et al. (1999), Liu et al. (2010) identified a 916G-A transition in exon 7 of the PRPS1 gene, resulting in a gly306-to-arg (G306R) substitution at a highly conserved residue. The mutation was not found in 1,475 Chinese control chromosomes or 450 chromosomes of European descent.


.0016   DEAFNESS, X-LINKED 1

PRPS1, ILE290THR
SNP: rs180177153, ClinVar: RCV000010619

In affected members from a Chinese family segregating X-linked postlingual nonsyndromic hearing loss (DFNX1; 304500), previously reported by Cui et al. (2004), Liu et al. (2010) identified an 869T-C transition in exon 7 of the PRPS1 gene, resulting in an ile290-to-thr (I290T) substitution at a highly conserved residue. The mutation was not found in 1,025 ethnically matched control chromosomes.


.0017   ARTS SYNDROME AND PHOSPHORIBOSYLPYROPHOSPHATE SYNTHETASE SUPERACTIVITY

PRPS1, VAL142LEU
SNP: rs398122855, ClinVar: RCV000022882

In a boy with a complex phenotype comprising Arts syndrome and PRPS1 superactivity (see 301835), Moran et al. (2012) identified a 424G-C transversion in exon 4 of the PRPS1 gene, resulting in a val142-to-leu (V142L) substitution at a highly conserved residue. Both the mother and grandmother were heterozygous for the mutation, which was not found in 202 control alleles. The patient had developmental delay, hypotonia, areflexia, motor neuropathy, sensorineural hearing loss, and a Chiari I malformation. Laboratory studies showed increased serum uric acid and increased urinary hypoxanthine consistent with PRPS1 superactivity, but he did not have gout. In addition, he had recurrent infections and early death at age 27 months from infection, consistent with Arts syndrome. A maternal uncle with similar symptoms had died of pneumonia at age 2. Molecular modeling predicted that the substitution would disrupt allosteric sites involved in inhibition of PRPS1, resulting in a gain of enzyme function, and the ATP-binding site, resulting in a loss of enzyme function. Patient fibroblasts showed normal PRPP synthetase activity, whereas erythrocytes showed a loss of enzyme activity, suggesting that the effect of the V142L mutation on protein activity depends on cell type. Moran et al. (2012) postulated a gain-of-function effect in proliferating cells and a loss-of-function effect in postmitotic cells. The report indicated that PRPS1 missense mutations can cause a continuous spectrum of features ranging from progressive nonsyndromic postlingual hearing impairment to uric acid overproduction, neuropathy, and recurrent infections depending on the functional sites affected.


.0018   CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, ALA121GLY
SNP: rs587777150, ClinVar: RCV000087131

In a young Korean man with X-linked recessive Charcot-Marie-Tooth disease (CMTX5; 311070) with early-onset sensorineural hearing loss but without optic atrophy or visual disturbance, Park et al. (2013) identified a hemizygous c.362C-G transversion in exon 3 of the PRPS1 gene, resulting in an ala121-to-gly (A121G) substitution at a highly conserved residue. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the dbSNP (build 135) or 1000 Genomes Project databases, or in 250 healthy controls. The patient's unaffected mother was heterozygous for the mutation. Two maternally related male relatives had a similar disorder, but DNA was not available from these individuals. Functional studies of the variant were not performed.


.0019   ARTS SYNDROME

DEAFNESS, X-LINKED 1, INCLUDED
PRPS1, GLN277PRO
SNP: rs869025593, ClinVar: RCV000208721, RCV000208743

In a German family with variable manifestations of PRPS1 deficiency, including a man with a protracted form of Arts syndrome (ARTS; 301835) and his sister with X-linked deafness-1 (DFNX1; 304500), Synofzik et al. (2014) identified a c.830A-C transversion in the PRPS1 gene, resulting in a gln277-to-pro (Q277P) substitution at a highly conserved residue in the C-terminal domain close to the catalytic site. The mutation was not found in the Exome Variant Server database or in 4,200 ethnically matched control X chromosomes. The mutation was predicted to disrupt the region surrounding the ribose-5-phosphate binding site and thus affect the catalytic site. The mother of these sibs, who had no hearing deficit or neurologic dysfunction at age 66, also carried the mutation; the mutation was heterozygous in the females and hemizygous in the male proband. Erythrocyte PRPS1 activity was not detectable in the proband, was decreased in the sister, and was normal in the mother. X-chromosome inactivation was extremely skewed in the sister with DFNX1 (94%; 6%), but only moderately skewed in the mother (80%; 20%). The findings illustrated that PRPS1 deficiency can present as a continuous spectrum of clinical features, even within the same family.


.0020   CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, SER16PRO
SNP: rs869025594, ClinVar: RCV000208733

In 4 females from a 3-generation Spanish family (RP-0482) with variable manifestations of X-linked recessive Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Almoguera et al. (2014) identified a heterozygous c.46T-C transition (c.46T-C, NM002764) in exon 1 of the PRPS1 gene, resulting in a ser16-to-pro (S16P) substitution at a conserved residue in the first alpha helix in the N-terminal domain. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was filtered against the 1000 Genomes Project (April 2012 release) and Exome Sequencing Project databases and 669 in-house exomes. The mutation segregated with the disorder in the family and was not found in 258 control X chromosomes. Erythrocyte PRPS1 activity from 3 affected females showed variably decreased levels, which correlated with the age at onset. The proband (IV-3), who was the most severely affected, had significantly skewed X inactivation (82%) of the paternal allele and showed lack of expression of the wildtype allele in lymphocyte mRNA. The proband's sister and mother, who carried the mutation but were less severely affected clinically, did not show significantly skewed X inactivation.


.0021   DEAFNESS, X-LINKED 1

PRPS1, ALA113SER
SNP: rs587781261, ClinVar: RCV000143857

In 2 Italian brothers (family 1) with postlingual X-linked deafness-1 (DFNX1; 304500), Robusto et al. (2015) identified a hemizygous c.337G-T transversion (c.337G-T, NM_002764.3) in exon 3 of the PRPS1 gene, resulting in an ala113-to-ser (A113S) substitution in an alpha-helix participating in trimer interface, predicted to destabilize the surrounding environment and possibly affect ATP binding. The mother, who had late-onset moderate hearing loss, was heterozygous for the mutation. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was filtered against the dbSNP (build 130), 1000 Genomes Project, and Exome Variant Server databases and was not found in 123 Italian controls. Erythrocyte PRPS1 activity was mildly decreased in the 2 affected males (25-35% of normal controls).


.0022   CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, MET115VAL
SNP: rs587781262, ClinVar: RCV000143858, RCV000143859

In affected members of an Italian family (family 2) with X-linked recessive Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Robusto et al. (2015) identified a c.343A-G transition (c.343A-G, NM_002764.3) in exon 3 of the PRPS1 gene, resulting in a met115-to-val (M115V) substitution at a highly conserved residue in an alpha-helix participating in trimer interface, predicted to destabilize the ATP-binding site and the allosteric site I. The mutation segregated with the disorder in the family and was not found in the dbSNP (build 130), 1000 Genomes Project, or Exome Variant Server databases. The family was ascertained through a 12-year-old male who had sensorineural hearing loss; subsequent examination of family members showed that those with the mutation had hearing loss as well as subclinical signs of a peripheral neuropathy, which was more evident in males compared to females. Erythrocyte PRPS1 activity was significantly decreased in the 2 affected males (5-10% of normal controls) and mildly decreased in 3 carrier females (34.7-47.7% of normal controls). X-inactivation studies in 1 carrier female showed a normal ratio, but there was preferential expression of the mutant allele (60% vs 40%).


.0023   CHARCOT-MARIE-TOOTH DISEASE, X-LINKED RECESSIVE, 5

PRPS1, VAL309PHE
SNP: rs587781263, ClinVar: RCV000143860

In affected members of a Peruvian family (family 3) with X-linked recessive Charcot-Marie-Tooth disease-5 (CMTX5; 311070), Robusto et al. (2015) identified a c.925G-T transversion (c.925G-T, NM_002764.3) in exon 7 of the PRPS1 gene, resulting in a val309-to-phe (V309F) substitution at a highly conserved residue at the trimer interface, predicted to disturb the allosteric type I function as well as hexameric assembly. The mutation segregated with the disorder in the family and was not found in the dbSNP (build 130), 1000 Genomes Project, or Exome Variant Server databases. The family was ascertained through a 14-year-old male who had sensorineural hearing loss and was subsequently found to have subclinical signs of a peripheral neuropathy. The patient's uncle and mother also carried the mutation; the uncle had sensorineural hearing loss and subtle peripheral neuropathy, whereas the mother had mild peripheral neuropathy without hearing loss. Erythrocyte PRPS1 activity was significantly decreased in the male proband (4.05% of normal controls) and mildly decreased in the mother (18.3% of normal controls). The mother had skewed X inactivation with almost exclusive expression of the wildtype allele (85% vs 15%).


See Also:

Lebo and Martin (1978)

REFERENCES

  1. Almoguera, B., He, S., Corton, M., Fernandez-San Jose, P., Blanco-Kelly, F., Lopez-Molina, M. I., Garcia-Sandoval, B., del Val, J., Guo, Y., Tian, L., Liu, X., Guan, L., Torres, R. J., Puig, J. G., Hakonarson, H., Xu, X., Keating, B., Ayuso, C. Expanding the phenotype of PRPS1 syndromes in females: neuropathy, hearing loss and retinopathy. Orphanet J. Rare Dis. 9: 190, 2014. Note: Electronic Article. [PubMed: 25491489] [Full Text: https://doi.org/10.1186/s13023-014-0190-9]

  2. Arts, W. F. M., Loonen, M. C. B., Sengers, R. C. A., Slooff, J. L. X-linked ataxia, weakness, deafness, and loss of vision in early childhood with a fatal course. Ann. Neurol. 33: 535-539, 1993. [PubMed: 8498830] [Full Text: https://doi.org/10.1002/ana.410330519]

  3. Becker, M. A., Heidler, S. A., Bell, G. I., Seino, S., Le Beau, M. M., Westbrook, C. A., Neuman, W., Shapiro, L. J., Mohandas, T. K., Roessler, B. J., Palella, T. D. Cloning of cDNAs for human phosphoribosylpyrophosphate synthetases 1 and 2 and X chromosome localization of PRPS1 and PRPS2 genes. Genomics 8: 555-561, 1990. [PubMed: 1962753] [Full Text: https://doi.org/10.1016/0888-7543(90)90043-t]

  4. Becker, M. A., Losman, M. J., Wilson, J., Simmonds, H. A. Superactivity of human phosphoribosyl pyrophosphate synthetase due to altered regulation by nucleotide inhibitors and inorganic phosphate. Biochim. Biophys. Acta 882: 168-176, 1986. [PubMed: 2423135] [Full Text: https://doi.org/10.1016/0304-4165(86)90151-0]

  5. Becker, M. A., Raivio, K. O., Bakay, B., Adams, W. B., Nyhan, W. L. Variant human phosphoribosylpyrophosphate synthetase altered in regulatory and catalytic functions. J. Clin. Invest. 65: 109-120, 1980. [PubMed: 6243137] [Full Text: https://doi.org/10.1172/JCI109640]

  6. Becker, M. A., Smith, P. R., Taylor, W., Mustafi, R., Switzer, R. L. The genetic and functional basis of purine nucleotide feedback-resistant phosphoribosylpyrophosphate synthetase superactivity. J. Clin. Invest. 96: 2133-2141, 1995. [PubMed: 7593598] [Full Text: https://doi.org/10.1172/JCI118267]

  7. Becker, M. A., Yen, R. C. K., Goss, S. J., Seegmiller, J. E., Itkin, P., Lazar, C., Adams, W. B. Localization of the structural gene for human phosphoribosylpyrophosphate synthetase on the X-chromosome. (Abstract) Clin. Res. 26: 500A, 1978.

  8. Becker, M. A., Yen, R. C. K., Itkin, P., Goss, S. J., Seegmiller, J. E., Bakay, B. Regional localization of the gene for human phosphoribosylpyrophosphate synthetase on the X-chromosome. Science 203: 1016-1019, 1979. [PubMed: 218284] [Full Text: https://doi.org/10.1126/science.218284]

  9. Becker, M. A. Hyperuricemia and Gout. In: Scriver, C. R.; Beaudet, A. L.; Sly, W. S.; Valle, D. (eds.): The Metabolic and Molecular Bases of Inherited Disease. Vol. II. (8th ed.) New York: McGraw-Hill (pub.) 2001. P. 2625.

  10. Cui, B., Zhang, H., Lu, Y., Zhong, W., Pei, G., Kong, X., Hu, L. Refinement of the locus for non-syndromic sensorineural deafness (DFN2) J. Genet. 83: 35-38, 2004. [PubMed: 15240907] [Full Text: https://doi.org/10.1007/BF02715827]

  11. de Brouwer, A. P. M., van Bokhoven, H., Nabuurs, S. B., Arts, W. F., Christodoulou, J., Duley, J. PRPS1 mutations: four distinct syndromes and potential treatment. Am. J. Hum. Genet. 86: 506-518, 2010. [PubMed: 20380929] [Full Text: https://doi.org/10.1016/j.ajhg.2010.02.024]

  12. de Brouwer, A. P. M., Williams, K. L., Duley, J. A., van Kuilenburg, A. B. P., Nabuurs, S. B., Egmont-Petersen, M., Lugtenberg, D., Zoetekouw, L., Banning, M. J. G., Roeffen, M., Hamel, B. C. J., Weaving, L., Ouvrier, R. A., Donald, J. A., Wevers, R. A., Christodoulou, J., van Bokhoven, H. Arts syndrome is caused by loss-of-function mutations in PRPS1. Am. J. Hum. Genet. 81: 507-518, 2007. [PubMed: 17701896] [Full Text: https://doi.org/10.1086/520706]

  13. Dickinson, M. E., Flenniken, A. M., Ji, X., Teboul, L., Wong, M. D., White, J. K., Meehan, T. F., Weninger, W. J., Westerberg, H., Adissu, H., Baker, C. N., Bower, L., and 73 others. High-throughput discovery of novel developmental phenotypes. Nature 537: 508-514, 2016. Note: Erratum: Nature 551: 398 only, 2017. [PubMed: 27626380] [Full Text: https://doi.org/10.1038/nature19356]

  14. Iinuma, K., Wada, Y., Onuma, A., Tanabu, M. Electroencephalographic study of an infant with phosphoribosylpyrophosphate synthetase deficiency. Tohoku J. Exp. Med. 116: 53-55, 1975. [PubMed: 168665] [Full Text: https://doi.org/10.1620/tjem.116.53]

  15. Kim, H.-J., Sohn, K.-M., Shy, M. E., Krajewski, K. M., Hwang, M., Park, J.-H., Jang, S.-Y., Won, H.-H., Choi, B.-O., Hong, S. H., Kim, B.-J., Suh, Y.-L., Ki, C.-S., Lee, S.-Y., Kim, S.-H., Kim, J.-W. Mutations in PRPS1, which encodes the phosphoribosyl pyrophosphate synthetase enzyme critical for nucleotide biosynthesis, cause hereditary peripheral neuropathy with hearing loss and optic neuropathy (CMT5X). Am. J. Hum. Genet. 81: 552-558, 2007. [PubMed: 17701900] [Full Text: https://doi.org/10.1086/519529]

  16. Lebo, R. V., Martin, D. W., Jr. Electrophoretic heterogeneity of 5-phosphoribosyl-1-pyrophosphate synthetase within and among humans. Biochem. Genet. 16: 905-916, 1978. [PubMed: 217337] [Full Text: https://doi.org/10.1007/BF00483742]

  17. Liu, X., Han, D., Li, J., Han, B., Ouyang, X., Cheng, J., Li, X., Jin, Z., Wang, Y., Bitner-Glindzicz, M., Kong, X., Xu, H., and 10 others. Loss-of-function mutations in the PRPS1 gene cause a type of nonsyndromic X-linked sensorineural deafness, DFN2. Am. J. Hum. Genet. 86: 65-71, 2010. [PubMed: 20021999] [Full Text: https://doi.org/10.1016/j.ajhg.2009.11.015]

  18. Manolis, E. N., Eavey, R. D., Sangwatanaroj, S., Halpin, C., Rosenbaum, S., Watkins, H., Jarcho, J., Seidman, C. E., Seidman, J. G. Hereditary postlingual sensorineural hearing loss mapping to chromosome Xq21. Am. J. Otol. 20: 621-626, 1999. [PubMed: 10503584]

  19. Moran, R., Kuilenburg, A. B. P., Duley, J., Nabuurs, S. B., Retno-Fitri, A., Christodoulou, J., Roelofsen, J., Yntema, H. G., Friedman, N. R., van Bokhoven, H., de Brouwer, A. P. M. Phosphoribosylpyrophosphate synthetase superactivity and recurrent infections is caused by a p.val142-to-leu mutation in PRS-I. Am. J. Med. Genet. 158A: 455-460, 2012. [PubMed: 22246954] [Full Text: https://doi.org/10.1002/ajmg.a.34428]

  20. Park, J., Hyun, Y. S., Kim, Y. J., Nam, S. H., Kim, S., Hong, Y. B., Park, J.-M., Chung, K. W., Choi, B.-O. Exome sequencing reveals a novel PRPS1 mutation in a family with CMTX5 without optic atrophy. J. Clin. Neurol. 9: 283-288, 2013. [PubMed: 24285972] [Full Text: https://doi.org/10.3988/jcn.2013.9.4.283]

  21. Robusto, M., Fang, M., Asselta, R., Castorina, P., Previtali, S. C., Caccia, S., Benzoni, E., De Cristofaro, R., Yu, C., Cesarani, A., Liu, X., Li, W., Primignani, P., Ambrosetti, U., Xu, X., Duga, S., Solda, G. The expanding spectrum of PRPS1-associated phenotypes: three novel mutations segregating with X-linked hearing loss and mild peripheral neuropathy. Europ. J. Hum. Genet. 23: 766-773, 2015. [PubMed: 25182139] [Full Text: https://doi.org/10.1038/ejhg.2014.168]

  22. Roessler, B. J., Bell, G., Heidler, S., Seino, S., Becker, M., Palella, T. D. Cloning of two distinct copies of human phosphoribosylpyrophosphate synthetase cDNA. Nucleic Acids Res. 18: 193 only, 1990. [PubMed: 2155397] [Full Text: https://doi.org/10.1093/nar/18.1.193]

  23. Roessler, B. J., Nosal, J. M., Smith, P. R., Heidler, S. A., Palella, T. D., Switzer, R. L., Becker, M. A. Human X-linked phosphoribosylpyrophosphate synthetase superactivity is associated with distinct point mutations in the PRPS1 gene. J. Biol. Chem. 268: 26476-26481, 1993. [PubMed: 8253776]

  24. Roessler, B. J., Palella, T. D., Heidler, S., Becker, M. A. Identification of distinct PRPS1 mutations in two patients with X-linked phosphoribosylpyrophosphate synthetase superactivity. (Abstract) Clin. Res. 39: 267A, 1991.

  25. Synofzik, M., Muller vom Hagen, J., Haack, T. B., Wilhelm, C., Lindig, T., Beck-Wodl, S., Nabuurs, S. B., van Kuilenburg, A. B. P., de Brouwer, A. P. M., Schols, L. X-linked Charcot-Marie-Tooth disease, Arts syndrome, and prelingual non-syndromic deafness form a disease continuum: evidence from a family with a novel PRPS1 mutation. Orphanet J. Rare Dis. 9: 24, 2014. Note: Electronic Article. [PubMed: 24528855] [Full Text: https://doi.org/10.1186/1750-1172-9-24]

  26. Taira, M., Iizasa, T., Yamada, K., Shimada, H., Tatibana, M. Tissue-differential expression of two distinct genes for phosphoribosyl pyrophosphate synthetase and existence of the testis-specific transcript. Biochim. Biophys. Acta 1007: 203-208, 1989. [PubMed: 2537655] [Full Text: https://doi.org/10.1016/0167-4781(89)90040-7]

  27. Tyson, J., Bellman, S., Newton, V., Simpson, P., Malcolm, S., Pembrey, M. E., Bitner-Glindzicz, M. Mapping of DFN2 to Xq22. Hum. Molec. Genet. 5: 2055-2060, 1996. [PubMed: 8968763] [Full Text: https://doi.org/10.1093/hmg/5.12.2055]

  28. Wada, Y., Nishimura, Y., Tanabu, M., Yoshimura, Y., Iinuma, K., Yoshida, T., Arakawa, T. Hypouricemic, mentally retarded infant with a defect of 5-phosphoribosyl-1-pyrophosphate synthetase of erythrocytes. Tohoku J. Exp. Med. 113: 149-157, 1974. [PubMed: 4373874] [Full Text: https://doi.org/10.1620/tjem.113.149]

  29. Zoref, E., De Vries, A., Sperling, O. Mutant feedback-resistant phosphoribosylpyrophosphate synthetase associated with purine overproduction and gout: phosphoribosylpyrophosphate and purine metabolism in cultured fibroblasts. J. Clin. Invest. 56: 1093-1099, 1975. [PubMed: 171280] [Full Text: https://doi.org/10.1172/JCI108183]


Contributors:
Ada Hamosh - updated : 02/21/2017
Cassandra L. Kniffin - updated : 2/10/2016
Cassandra L. Kniffin - updated : 2/19/2014
Cassandra L. Kniffin - updated : 4/9/2012
Cassandra L. Kniffin - updated : 4/30/2010
Marla J. F. O'Neill - updated : 2/26/2010
Patricia A. Hartz - updated : 10/24/2007
Victor A. McKusick - updated : 8/22/2007
Cassandra L. Kniffin - reorganized : 8/17/2007
Cassandra L. Kniffin - updated : 8/16/2007
Rebekah S. Rasooly - updated : 2/26/1999

Creation Date:
Victor A. McKusick : 6/4/1986

Edit History:
alopez : 03/13/2023
carol : 01/29/2018
alopez : 02/21/2017
alopez : 09/23/2016
carol : 07/09/2016
carol : 3/1/2016
carol : 3/1/2016
ckniffin : 2/10/2016
carol : 1/13/2016
carol : 2/25/2014
mcolton : 2/21/2014
ckniffin : 2/19/2014
alopez : 4/20/2012
alopez : 4/13/2012
alopez : 4/12/2012
terry : 4/10/2012
ckniffin : 4/9/2012
carol : 4/12/2011
wwang : 5/4/2010
ckniffin : 4/30/2010
terry : 4/2/2010
carol : 2/26/2010
mgross : 10/30/2007
terry : 10/24/2007
alopez : 8/22/2007
carol : 8/17/2007
ckniffin : 8/16/2007
alopez : 9/10/2004
terry : 6/25/2004
psherman : 3/1/1999
psherman : 2/26/1999
dkim : 7/7/1998
terry : 11/18/1996
terry : 11/4/1996
mark : 2/2/1996
terry : 1/26/1996
terry : 5/10/1994
warfield : 4/20/1994
carol : 4/11/1994
mimadm : 2/28/1994
carol : 12/16/1992
carol : 12/3/1992