Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 298
Filter
1.
Stem Cell Reports ; 16(7): 1749-1762, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34214487

ABSTRACT

Mutations in HPRT1, a gene encoding a rate-limiting enzyme for purine salvage, cause Lesch-Nyhan disease which is characterized by self-injury and motor impairments. We leveraged stem cell and genetic engineering technologies to model the disease in isogenic and patient-derived forebrain and midbrain cell types. Dopaminergic progenitor cells deficient in HPRT showed decreased intensity of all developmental cell-fate markers measured. Metabolic analyses revealed significant loss of all purine derivatives, except hypoxanthine, and impaired glycolysis and oxidative phosphorylation. real-time glucose tracing demonstrated increased shunting to the pentose phosphate pathway for de novo purine synthesis at the expense of ATP production. Purine depletion in dopaminergic progenitor cells resulted in loss of RHEB, impairing mTORC1 activation. These data demonstrate dopaminergic-specific effects of purine salvage deficiency and unexpectedly reveal that dopaminergic progenitor cells are programmed to a high-energy state prior to higher energy demands of terminally differentiated cells.


Subject(s)
Dopaminergic Neurons/metabolism , Energy Metabolism , Lesch-Nyhan Syndrome/metabolism , Lesch-Nyhan Syndrome/pathology , Mesencephalon/pathology , Biomarkers/metabolism , Cell Lineage , Cerebral Cortex/pathology , Glucose/metabolism , Glycolysis , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome/enzymology , Mechanistic Target of Rapamycin Complex 1/metabolism , Neural Stem Cells/metabolism , Oxidative Phosphorylation , Pentose Phosphate Pathway , Purines/metabolism
2.
Nucleosides Nucleotides Nucleic Acids ; 36(11): 704-711, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29185864

ABSTRACT

Lesch-Nyhan disease (LND) is a rare X-linked inherited neurogenetic disorder of purine metabolism in which the enzyme, hypoxanthine-guanine phosphoribosyltransferase (HGprt) is defective. The authors report a novel point mutation that led to HGprt-related neurological dysfunction (HND) in a family in which there was a missense mutation in exon 6 of the coding region of the HPRT1 gene: g.34938G>T, c.403G>T, p.D135Y. Molecular diagnosis is consistent with the genetic heterogeneity of the HPRT1 gene responsible for HGprt deficiency. It allows fast, accurate carrier detection and genetic counseling.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Lesch-Nyhan Syndrome/genetics , Mutation, Missense , Base Sequence , Child, Preschool , Exons/genetics , Female , Humans , Hypoxanthine Phosphoribosyltransferase/metabolism , Male , Pedigree
3.
Nucleosides Nucleotides Nucleic Acids ; 36(7): 452-462, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28524722

ABSTRACT

Lesch-Nyhan disease (LND) is a rare X-linked inherited neurogenetic disorder of purine metabolism in which the enzyme, hypoxanthine-guanine phosphoribosyltransferase (HGprt) is defective. The authors report two independent point mutations leading to splicing errors: IVS 2 +1G>A, c.134 +1G>A, and IVS 3 +1G>A, c.318 +1G>A in the hypoxanthine-phosphoribosyltransferase1 (HPRT1) gene which result in exclusion of exon 2 and exon 3 respectively, in the HGprt enzyme protein from different members of two Chiloé Island families. Molecular analysis has revealed the heterogeneity of genetic mutation of the HPRT1 gene responsible for the HGprt deficiency. It allows fast, accurate carrier detection and genetic counseling.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Islands , Lesch-Nyhan Syndrome/enzymology , Lesch-Nyhan Syndrome/genetics , Mutation , Pedigree , Adolescent , Adult , Base Sequence , Chile , Exons/genetics , Female , Humans , Male , Young Adult
4.
Ann Neurol ; 76(1): 95-107, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24891139

ABSTRACT

OBJECTIVE: Lesch-Nyhan disease (LND) is caused by congenital deficiency of the purine recycling enzyme, hypoxanthine-guanine phosphoribosyltransferase (HGprt). Affected patients have a peculiar neurobehavioral syndrome linked with reductions of dopamine in the basal ganglia. The purpose of the current studies was to determine the anatomical basis for the reduced dopamine in human brain specimens collected at autopsy. METHODS: Histopathological studies were conducted using autopsy tissue from 5 LND cases and 6 controls. Specific findings were replicated in brain tissue from an HGprt-deficient knockout mouse using immunoblots, and in a cell model of HGprt deficiency by flow-activated cell sorting (FACS). RESULTS: Extensive histological studies of the LND brains revealed no signs suggestive of a degenerative process or other consistent abnormalities in any brain region. However, neurons of the substantia nigra from the LND cases showed reduced melanization and reduced immunoreactivity for tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine synthesis. In the HGprt-deficient mouse model, immunohistochemical stains for TH revealed no obvious loss of midbrain dopamine neurons, but quantitative immunoblots revealed reduced TH expression in the striatum. Finally, 10 independent HGprt-deficient mouse MN9D neuroblastoma lines showed no signs of impaired viability, but FACS revealed significantly reduced TH immunoreactivity compared to the control parent line. INTERPRETATION: These results reveal an unusual phenomenon in which the neurochemical phenotype of dopaminergic neurons is not linked with a degenerative process. They suggest an important relationship between purine recycling pathways and the neurochemical integrity of the dopaminergic phenotype.


Subject(s)
Dopamine/deficiency , Dopaminergic Neurons/pathology , Lesch-Nyhan Syndrome/genetics , Lesch-Nyhan Syndrome/pathology , Mesencephalon/enzymology , Mesencephalon/pathology , Phenotype , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Child , Child, Preschool , Corpus Striatum/enzymology , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/genetics , Dopaminergic Neurons/enzymology , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Substantia Nigra/enzymology , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/deficiency , Tyrosine 3-Monooxygenase/genetics , Young Adult
5.
Article in English | MEDLINE | ID: mdl-24940672

ABSTRACT

Mutation of hypoxanthine guanine phosphoribosyltransferase (HPRT) gives rise to Lesch-Nyhan syndrome, which is characterized by hyperuricemia, severe motor disability, and self-injurious behavior, or HPRT-related gout with hyperuricemia. Four mutations were detected in two Lesch-Nyhan families and two families with partial deficiency since our last report. A new mutation of G to TT (c.456delGinsTT) resulting in a frameshift (p.Q152Hfs*3) in exon 3 has been identified in one Lesch-Nyhan family. In the other Lesch-Nyhan family, a new point mutation in intron 7 (c.532+5G>T) causing splicing error (exon 7 excluded, p.L163Cfs*4) was detected. In the two partial deficiency cases with hyperuricemia, two missense mutations of p.D20V (c.59A>T) and p.H60R (c.179A>G) were found. An increase of erythrocyte PRPP concentration was observed in the respective phenotypes and seems to be correlated with disease severity.


Subject(s)
Asian People/genetics , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/blood , Lesch-Nyhan Syndrome/genetics , Mutation , Pedigree , Ribose-Phosphate Pyrophosphokinase/blood , Erythrocytes/enzymology , Female , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome/enzymology , Male
6.
Biochem Biophys Res Commun ; 446(4): 1091-5, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24680827

ABSTRACT

Lesch-Nyhan syndrome (LNS) is a neurogenetic disorder of purine metabolism in which the enzyme, hypoxanthine-guanine phosphoribosyltransferase (HPRT) is defective. A major unsolved question is how the loss of HPRT enzyme function affects the brain to cause the neurobehavioural syndrome in LNS and its attenuated variants (LNVs). To address this issue, a search for a link between LNS and the amyloid precursor protein (APP) is developed. Here, I identified, for the first time in fibroblasts from normal subjects as well as from LNS and LNV patients: (a) several APP-mRNA isoforms encoding divers APP protein isoforms ranging from 120 to 770 amino acids (with or without mutations and/or deletions) accounted for epigenetic mechanisms in the regulation of alternative APP pre-mRNA splicing and (b) five novel independent polymorphisms in the APP promoter: -956A>G, -1023T>C, -1161A>G, -2224G>A, -2335C>T relative to the transcription start site. A role for epistasis between mutated HPRT and APP genes affecting the regulation of alternative APP pre-mRNA splicing in LNS is suggested. An accurate quantification of various APP isoforms in brain tissues for detection of initial pathological changes or pathology development is needed. My findings may provide new directions not only for investigating the role of APP in neuropathology associated with HPRT-deficiency in LNS but also for the research in neurodevelopmental and neurodegenerative disorders by which various APP isoforms involved in the pathogenesis of the diseases such as Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Lesch-Nyhan Syndrome/genetics , Epigenesis, Genetic , Fibroblasts/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Male , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Protein Isoforms/genetics , RNA Precursors/genetics , RNA Splicing
7.
BMJ Case Rep ; 20132013 Dec 10.
Article in English | MEDLINE | ID: mdl-24326440

ABSTRACT

A 30-year-old man was referred for investigation and management of hyperuricaemia. History included recurrent nephrolithiasis and chronic gout with poor response to medical management. Hypoxanthine-guanine phosphoribosyltransferase (HGPRT) enzyme activity was investigated and found to be deficient confirming the diagnosis of Lesch-Nyhan disease. Hyperuricaemia was treated with allopurinol. To prevent nephrolithiasis, the patient was instructed to avoid dehydration and aim for a minimum urine output of 2 L/day. Urinary alkalinisation with potassium citrate was started. The patient was referred for genetic counselling. This case discusses the genetics, pathophysiology, clinical manifestations, diagnosis and management of HGPRT deficiency.


Subject(s)
Hyperuricemia/genetics , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome , Adult , Allopurinol/therapeutic use , Diuretics/therapeutic use , Genetic Counseling , Gout/etiology , Gout Suppressants/therapeutic use , Humans , Hyperuricemia/diagnosis , Hyperuricemia/drug therapy , Hyperuricemia/enzymology , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/blood , Lesch-Nyhan Syndrome/diagnosis , Lesch-Nyhan Syndrome/drug therapy , Lesch-Nyhan Syndrome/enzymology , Male , Nephrolithiasis/etiology , Nephrolithiasis/prevention & control , Potassium Citrate/therapeutic use , Uric Acid/blood
8.
Hum Mol Genet ; 22(22): 4502-15, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23804752

ABSTRACT

Lesch-Nyhan syndrome (LNS) is a neurodevelopmental disorder caused by mutations in the gene encoding the purine metabolic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT). A series of motor, cognitive and neurobehavioral anomalies characterize this disease phenotype, which is still poorly understood. The clinical manifestations of this syndrome are believed to be the consequences of deficiencies in neurodevelopmental pathways that lead to disordered brain function. We have used microRNA array and gene ontology analysis to evaluate the gene expression of differentiating HPRT-deficient human neuron-like cell lines. We set out to identify dysregulated genes implicated in purine-based cellular functions. Our approach was based on the premise that HPRT deficiency affects preeminently the expression and the function of purine-based molecular complexes, such as guanine nucleotide exchange factors (GEFs) and small GTPases. We found that several microRNAs from the miR-17 family cluster and genes encoding GEF are dysregulated in HPRT deficiency. Most notably, our data show that the expression of the exchange protein activated by cAMP (EPAC) is blunted in HPRT-deficient human neuron-like cell lines and fibroblast cells from LNS patients, and is altered in the cortex, striatum and midbrain of HPRT knockout mouse. We also show a marked impairment in the activation of small GTPase RAP1 in the HPRT-deficient cells, as well as differences in cytoskeleton dynamics that lead to increased motility for HPRT-deficient neuron-like cell lines relative to control. We propose that the alterations in EPAC/RAP1 signaling and cell migration in HPRT deficiency are crucial for neuro-developmental events that may contribute to the neurological dysfunctions in LNS.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Lesch-Nyhan Syndrome/genetics , MicroRNAs/genetics , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Line , Cell Movement/physiology , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Cytoskeleton/metabolism , Gene Ontology , Guanine Nucleotide Exchange Factors/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Male , Mesencephalon/metabolism , Mice , Mice, Knockout , MicroRNAs/metabolism , Multigene Family , Oligonucleotide Array Sequence Analysis , Signal Transduction , rap1 GTP-Binding Proteins/genetics
9.
PLoS One ; 8(5): e63333, 2013.
Article in English | MEDLINE | ID: mdl-23691025

ABSTRACT

Lesch-Nyhan Disease (LND) is the result of mutations in the X-linked gene encoding the purine metabolic enzyme, hypoxanthine guanine phosphoribosyl transferase (HPRT). LND gives rise to severe neurological anomalies including mental retardation, dystonia, chorea, pyramidal signs and a compulsive and aggressive behavior to self injure. The neurological phenotype in LND has been shown to reflect aberrant dopaminergic signaling in the basal ganglia, however there are little data correlating the defect in purine metabolism to the neural-related abnormalities. In the present studies, we find that HPRT-deficient neuronal cell lines have reduced CREB (cAMP response element-binding protein) expression and intracellular cyclic AMP (cAMP), which correlates with attenuated CREB-dependent transcriptional activity and a reduced phosphorylation of protein kinase A (PKA) substrates such as synapsin (p-syn I). Of interest, we found increased expression of phosphodiesterase 10A (PDE10A) in HPRT-deficient cell lines and that the PDE10 inhibitor papaverine and PDE10A siRNA restored cAMP/PKA signaling. Furthermore, reconstitution of HPRT expression in mutant cells partly increased cAMP signaling synapsin phosphorylation. In conclusion, our data show that HPRT-deficiency alters cAMP/PKA signaling pathway, which is in part due to the increased of PDE10A expression and activity. These findings suggest a mechanistic insight into the possible causes of LND and highlight PDE10A as a possible therapeutic target for this intractable neurological disease.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Gene Expression Regulation, Enzymologic , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome/drug therapy , Phosphoric Diester Hydrolases/metabolism , Signal Transduction , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Knockdown Techniques , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Lesch-Nyhan Syndrome/pathology , MicroRNAs/genetics , Molecular Targeted Therapy , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synapsins/genetics , Transcription, Genetic
10.
Article in English | MEDLINE | ID: mdl-23473102

ABSTRACT

Inherited mutation of the purine salvage enzyme, hypoxanthine guanine phosphoribosyltransferase (HPRT) gives rise to Lesch-Nyhan syndrome (LNS) or Lesch-Nyhan variants (LNVs). We report three novel independent mutations in the coding region of HPRT gene: exon 3: c.141delA, p.D47fs53X; exon 5: c.400G>A, p.E134K; exon 7: c.499A>G, p.R167G from three LNS affected male patients.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Lesch-Nyhan Syndrome/genetics , Mutation , Base Sequence , Humans , Male , Sequence Analysis, DNA
11.
Joint Bone Spine ; 80(1): 93-5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22999896

ABSTRACT

The hypoxanthine-guanine phosphoribosyltransferase deficiency is an inborn error of purine metabolism, linked to the X chromosome. The clinical phenotypes associated with HPRT deficiency varied according to the level of enzyme deficiency, with a large spectrum of neurologic features like self-injurious behaviour in patients with complete deficiency. We report a 20-year-old man who had asymmetric polyarthritis, tophi, hyperuricemia, nephrolithiasis and mild neurologic symptoms with undetectable levels of HPRT activity in lysed erythrocytes. The genetic study identified the c.143G>A mutation in exon 3, GAA CGT (CTT>GAA CAT CTT (48arg>his). The presence of gouty arthropathy and chronic hyperuricemia in a young patient with neurological symptoms, suggests HPRT deficiency for which it is necessary its enzyme and molecular determination.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/genetics , Humans , Lesch-Nyhan Syndrome/diagnosis , Lesch-Nyhan Syndrome/drug therapy , Lesch-Nyhan Syndrome/enzymology , Male , Mutation , Young Adult
12.
Mol Genet Metab ; 106(4): 498-501, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22766437

ABSTRACT

Inherited mutation of the purine salvage enzyme, hypoxanthine guanine phosphoribosyltransferase (HPRT) gives rise to Lesch-Nyhan syndrome (LNS) or Lesch-Nyhan variants (LNV). We report a case of two LNS affected members of a family with deficiency of activity of HPRT in intact cultured fibroblasts in whom mutation could not be found in the HPRT coding sequence but there was markedly decreased HPRT expression of mRNA.


Subject(s)
DNA/genetics , Gene Expression Regulation, Enzymologic , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Lesch-Nyhan Syndrome/genetics , Open Reading Frames/genetics , Adolescent , Enzyme Assays , Genome, Human/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/metabolism , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reference Standards , Regulatory Sequences, Nucleic Acid/genetics
13.
Proc Natl Acad Sci U S A ; 109(9): 3377-82, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22331909

ABSTRACT

Lesch-Nyhan disease (LND) is an X-linked genetic disorder caused by mutations of the hypoxanthine guanine phosphoribosyltransferase (HPRT) purine biosynthesis gene and characterized by aberrant purine metabolism, deficient basal ganglia dopamine levels, dystonia, and severe neurobehavioral manifestations, including compulsive self-injurious behavior. Although available evidence has identified important roles for purinergic signaling in brain development, the mechanisms linking HPRT deficiency, purinergic pathways, and neural dysfunction of LND are poorly understood. In these studies aimed at characterizing purinergic signaling in HPRT deficiency, we used a lentivirus vector stably expressing an shRNA targeted to the HPRT gene to produce HPRT-deficient human CVB induced pluripotent stem cells and human HUES11 embryonic stem cells. Both CVB and HUES11 cells show >99% HPRT knockdown and demonstrate markedly decreased expression of the purinergic P2Y1 receptor mRNA. In CVB cells, P2Y1 mRNA and protein down-regulation by HPRT knockdown is refractory to activation by the P2Y1 receptor agonist ATP and shows aberrant purinergic signaling, as reflected by marked deficiency of the transcription factor pCREB and constitutive activation of the MAP kinases phospho-ERK1/2. Moreover, HPRT-knockdown CVB cells also demonstrate marked reduction of phosphorylated ß-catenin. These results indicate that the housekeeping gene HPRT regulates purinergic signaling in pluripotent human stem cells, and that this regulation occurs at least partly through aberrant P2Y1-mediated expression and signaling. We propose that such mechanisms may play a role in the neuropathology of HPRT-deficiency LND and may point to potential molecular targets for modulation of this intractable neurological phenotype.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/physiology , Neurogenesis/physiology , Pluripotent Stem Cells/enzymology , Purines/metabolism , Adenosine Triphosphate/pharmacology , Cell Line , Fibroblasts/enzymology , Gene Knockdown Techniques , Genes, Essential , Genetic Vectors/genetics , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Humans , Lentivirus/genetics , Lesch-Nyhan Syndrome/enzymology , MAP Kinase Signaling System/physiology , Male , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Purinergic P2Y Receptor Agonists/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/physiology , beta Catenin/metabolism
14.
J Biol Chem ; 287(5): 2997-3008, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22157001

ABSTRACT

Lesch-Nyhan disease and its attenuated variants are caused by mutations in the HPRT1 gene, which encodes the purine recycling enzyme hypoxanthine-guanine phosphoribosyltransferase. The mutations are heterogeneous, with more than 400 different mutations already documented. Prior efforts to correlate variations in the clinical phenotype with different mutations have suggested that milder phenotypes typically are associated with mutants that permit some residual enzyme function, whereas the most severe phenotype is associated with null mutants. However, multiple exceptions to this concept have been reported. In the current studies 44 HPRT1 mutations associated with a wide spectrum of clinical phenotypes were reconstructed by site-directed mutagenesis, the mutant enzymes were expressed in vitro and purified, and their kinetic properties were examined toward their substrates hypoxanthine, guanine, and phosphoribosylpyrophosphate. The results provide strong evidence for a correlation between disease severity and residual catalytic activity of the enzyme (k(cat)) toward each of its substrates as well as several mechanisms that result in exceptions to this correlation. There was no correlation between disease severity and the affinity of the enzyme for its substrates (K(m)). These studies provide a valuable model for understanding general principles of genotype-phenotype correlations in human disease, as the mechanisms involved are applicable to many other disorders.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/chemistry , Lesch-Nyhan Syndrome/enzymology , Genotype , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Kinetics , Lesch-Nyhan Syndrome/genetics , Mutagenesis, Site-Directed , Mutation , Phenotype , Substrate Specificity/genetics
15.
Nucleosides Nucleotides Nucleic Acids ; 30(12): 1266-71, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22132985

ABSTRACT

Lesch-Nyhan syndrome is caused by a deficiency of hypoxanthine phosphoribosyltransferase (HPRT) encoded by HPRT1. About 20% of patients have a deletion of HPRT1 and large deletions of HPRT1 are not always fully characterized at the molecular level. Here, we report on a case of Lesch-Nyhan syndrome with a 33-kb deletion involving exon 1 of HPRT1. This novel mutation is caused by a nonhomologous recombination between different classes of interspersed repetitive DNA.


Subject(s)
Gene Deletion , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Lesch-Nyhan Syndrome/genetics , Adolescent , Base Sequence , Chromosome Breakpoints , Humans , Male , Molecular Sequence Data
16.
Nucleosides Nucleotides Nucleic Acids ; 30(12): 1272-5, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22132986

ABSTRACT

Mutations of two enzyme genes, HPRT1 encoding hypoxanthine guanine phosphoribosyltransferase (HPRT) and PRPS1 encoding a catalytic subunit (PRS-I) of phosphoribosylpyrophosphate synthetase, cause X-linked inborn errors of purine metabolism. Analyzing these two genes, we have identified three HPRT1 mutations in Lesch-Nyhan families following our last report. One of them, a new mutation involving the deletion of 4224 bp from intron 4 to intron 5 and the insertion of an unknown 28 bp, has been identified. This mutation resulted in an enzyme polypeptide with six amino acids deleted due to abnormal mRNA skipping exon 5. The other HPRT1 mutations, a single base deletion (548delT, 183fs189X), and a point mutation causing a splicing error (532+1G>A, 163fs165X) were detected first in Japanese patients but have been reported in European families. On the other hand, in the analysis of PRPS1, no mutation was identified in any patient.


Subject(s)
Genetic Diseases, X-Linked/enzymology , Genetic Diseases, X-Linked/genetics , Hypoxanthine Phosphoribosyltransferase/genetics , Mutation/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/enzymology , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Ribose-Phosphate Pyrophosphokinase/genetics , Humans , Lesch-Nyhan Syndrome/enzymology , Lesch-Nyhan Syndrome/genetics
17.
Nucleosides Nucleotides Nucleic Acids ; 30(6): 440-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21780909

ABSTRACT

Inherited mutation of a purine salvage enzyme, hypoxanthine guanine phosphoribosyltransferase (HPRT), gives rise to Lesch-Nyhan Syndrome (LNS) or HPRT-related gout. Here, we report five novel independent mutations in the coding region of the HPRT gene from five unrelated male patients manifesting different clinical phenotypes associated with LNS: exon 2: c.133A > G, p.45R > G; c.35A > C, p.12D > A; c.88delG; exon 7: c.530A > T, p.177D > V; and c.318 + 1G > C: IVS3 + 1G > C splice site mutation.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Lesch-Nyhan Syndrome/genetics , Mutation , Base Sequence , Humans , Male , Mutation, Missense
20.
Hum Mol Genet ; 18(13): 2317-27, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19342420

ABSTRACT

Hypoxanthine-guanine phosphoribosyltransferase (HPRT) deficiency results in Lesch-Nyhan disease (LND), where affected individuals exhibit a characteristic neurobehavioral disorder that has been linked with dysfunction of dopaminergic pathways of the basal ganglia. Since the functions of HPRT, a housekeeping enzyme responsible for recycling purines, have no direct relationships with the dopaminergic pathways, the mechanisms whereby HPRT deficiency affect them remain unknown. The current studies demonstrate that HPRT deficiency influences early developmental processes controlling the dopaminergic phenotype, using several different cell models for HPRT deficiency. Microarray methods and quantitative PCR were applied to 10 different HPRT-deficient (HPRT(-)) sublines derived from the MN9D cell line. Despite the variation inherent in such mutant sublines, several consistent abnormalities were evident. Most notable were increases in the mRNAs for engrailed 1 and 2, transcription factors known to play a key role in the specification and survival of dopamine neurons. The increases in mRNAs were accompanied by increases in engrailed proteins, and restoration of HPRT reverted engrailed expression towards normal levels, demonstrating a functional relationship between HPRT and engrailed. The functional relevance of the abnormal developmental molecular signature of the HPRT(-) MN9D cells was evident in impoverished neurite outgrowth when the cells were forced to differentiate chemically. To verify that these abnormalities were not idiosyncratic to the MN9D line, HPRT(-) sublines from the SK-N-BE(2) M17 human neuroblastoma line were evaluated and an increased expression of engrailed mRNAs was also seen. Over-expression of engrailed occurred even in primary fibroblasts from patients with LND in a manner that suggested a correlation with disease severity. These results provide novel evidence that HPRT deficiency may affect dopaminergic neurons by influencing early developmental mechanisms.


Subject(s)
Dopamine/metabolism , Gene Expression Regulation, Developmental , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome/enzymology , Neurogenesis , Neurons/metabolism , Animals , Cell Line , Cells, Cultured , Fibroblasts/enzymology , Fibroblasts/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/metabolism , Lesch-Nyhan Syndrome/pathology , Male , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...