Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 658
Filter
1.
Elife ; 122024 May 03.
Article in English | MEDLINE | ID: mdl-38700995

ABSTRACT

Adenine phosphoribosyltransferase (APRT) and hypoxanthine-guanine phosphoribosyltransferase (HGPRT) are two structurally related enzymes involved in purine recycling in humans. Inherited mutations that suppress HGPRT activity are associated with Lesch-Nyhan disease (LND), a rare X-linked metabolic and neurological disorder in children, characterized by hyperuricemia, dystonia, and compulsive self-injury. To date, no treatment is available for these neurological defects and no animal model recapitulates all symptoms of LND patients. Here, we studied LND-related mechanisms in the fruit fly. By combining enzymatic assays and phylogenetic analysis, we confirm that no HGPRT activity is expressed in Drosophila melanogaster, making the APRT homolog (Aprt) the only purine-recycling enzyme in this organism. Whereas APRT deficiency does not trigger neurological defects in humans, we observed that Drosophila Aprt mutants show both metabolic and neurobehavioral disturbances, including increased uric acid levels, locomotor impairments, sleep alterations, seizure-like behavior, reduced lifespan, and reduction of adenosine signaling and content. Locomotor defects could be rescued by Aprt re-expression in neurons and reproduced by knocking down Aprt selectively in the protocerebral anterior medial (PAM) dopaminergic neurons, the mushroom bodies, or glia subsets. Ingestion of allopurinol rescued uric acid levels in Aprt-deficient mutants but not neurological defects, as is the case in LND patients, while feeding adenosine or N6-methyladenosine (m6A) during development fully rescued the epileptic behavior. Intriguingly, pan-neuronal expression of an LND-associated mutant form of human HGPRT (I42T), but not the wild-type enzyme, resulted in early locomotor defects and seizure in flies, similar to Aprt deficiency. Overall, our results suggest that Drosophila could be used in different ways to better understand LND and seek a cure for this dramatic disease.


Subject(s)
Drosophila melanogaster , Lesch-Nyhan Syndrome , Animals , Drosophila melanogaster/physiology , Drosophila melanogaster/genetics , Lesch-Nyhan Syndrome/genetics , Lesch-Nyhan Syndrome/metabolism , Purines/metabolism , Disease Models, Animal , Behavior, Animal , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Hypoxanthine Phosphoribosyltransferase/deficiency , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Locomotion
2.
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
3.
J Radiat Res ; 62(4): 582-589, 2021 Jul 10.
Article in English | MEDLINE | ID: mdl-33899106

ABSTRACT

Tritium is a low energy beta emitter and is discharged into the aquatic environment primarily in the form of tritiated water (HTO) from nuclear power plants or from nuclear fuel reprocessing plants. Although the biological effects of HTO exposures at significant doses or dose rates have been extensively studied, there are few reports concerning the biological effects of HTO exposures at very low dose rates. In the present study using a hyper-sensitive assay system, we investigated the dose rate effect of HTO on the induction of mutations. Confluent cell populations were exposed to HTO for a total dose of 0.2 Gy at dose rates between 4.9 mGy/day and 192 mGy/day by incubating cells in medium containing HTO. HTO-induced mutant frequencies and mutation spectra were then investigated. A significant inflection point for both the mutant frequency and mutation spectra was found between 11 mGy/day and 21.6 mGy/day. Mutation spectra analysis revealed that a mechanistic change in the nature of the mutation events occurred around 11 mGy/day. The present observations and published experimental results from oral administrations of HTO to mice suggest that a threshold dose-rate for HTO exposures might exist between 11 mGy/day and 21.6 mGy/day where the nature of the mutation events induced by HTO becomes similar to those seen in spontaneous events.


Subject(s)
Mutation/genetics , Tritium/chemistry , Water/chemistry , Animals , Cell Line , Cell Survival/radiation effects , Chromosomes, Human, X/genetics , Clone Cells , Cricetinae , Dose-Response Relationship, Radiation , Genetic Markers , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics
4.
Mol Reprod Dev ; 87(9): 930-933, 2020 09.
Article in English | MEDLINE | ID: mdl-32853477

ABSTRACT

The purine hypoxanthine plays important role in regulating oocyte maturation and early embryonic development. The enzyme hypoxanthine phosphoribosyltransferase (HPRT) recycles hypoxanthine to generate substrates for nucleotide synthesis and key metabolites, and here we show that HPRT deficiency in the rat disrupts early embryonic development and causes infertility in females.


Subject(s)
Infertility, Female/etiology , Lesch-Nyhan Syndrome/complications , Animals , Embryonic Development/genetics , Female , Fertility/genetics , Fetal Viability/genetics , Hypoxanthine/metabolism , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Infertility, Female/genetics , Lesch-Nyhan Syndrome/genetics , Lesch-Nyhan Syndrome/pathology , Pregnancy , Purines/metabolism , Rats
5.
CEN Case Rep ; 9(3): 210-214, 2020 08.
Article in English | MEDLINE | ID: mdl-32128695

ABSTRACT

Unlike complete deficiency of hypoxanthine phosphoribosyltransferase (HPRT) (i.e., Lesch-Nyhan syndrome), partial HPRT deficiency causes HPRT-related hyperuricemia without neurological symptoms. Herein, we describe a 22-year-old man without neurological symptoms that presented gout, hyperuricemia (serum urate level, 12.2 mg/dL), multiple renal microcalculi, and a family history of juvenile gout that was exhibited by his brother and grandfather. Genetic testing revealed a novel missense mutation, c.103G>A (p.V35M), in the HPRT1 gene, and biochemical testing (conducted using the patient's erythrocytes) showed that the patient retained only 12.4% HPRT enzymatic activity compared to that exhibited by a healthy control subject. We thus diagnosed the patient with HPRT-related hyperuricemia caused by partial HPRT deficiency. After his serum urate level was controlled via treatment with febuxostat, his gout did not recur. Thus, this study emphasizes that HPRT deficiency should be considered as a potential cause of familial juvenile gout, even in the absence of neurological symptoms.


Subject(s)
Gout/genetics , Hyperuricemia/genetics , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Kidney Diseases/genetics , Febuxostat/administration & dosage , Febuxostat/therapeutic use , Gout/complications , Gout/diagnosis , Gout/drug therapy , Gout Suppressants/administration & dosage , Gout Suppressants/therapeutic use , Humans , Hyperuricemia/blood , Hyperuricemia/diagnosis , Hyperuricemia/drug therapy , Hyperuricemia/etiology , Kidney Calculi/diagnosis , Kidney Calculi/etiology , Kidney Calculi/pathology , Kidney Diseases/diagnosis , Kidney Diseases/drug therapy , Male , Mutation, Missense/genetics , Treatment Outcome , Young Adult
6.
Mol Genet Metab ; 127(2): 147-157, 2019 06.
Article in English | MEDLINE | ID: mdl-31182398

ABSTRACT

BACKGROUND: HPRT deficiency is a rare disorder of purine metabolism whose natural history is not fully understood. No optimal management recommendations exist. The objective of the present study is to characterize a large cohort of patients with HPRT deficiency, comparing Lesch-Nyhan Disease (LND) and its attenuated variants, with the purpose of helping clinicians in disease management and prognostic definition. METHODS: Genetic and clinical features of French and Italian patients with a confirmed diagnosis of HPRT deficiency were collected. RESULTS: A hundred and one patients were studied, including 66 LND, 22 HND (HPRT-related Neurological Dysfunction) and 13 HRH (HPRT-Related Hyperuricemia) patients. The clinical manifestations at onset were not specific, but associated with an orange coloration of diapers in 22% of patients. The overall neurological involvement was more severe in LND than in HND patients. Behavioural disturbances were not limited to self-injuries and were not exclusive of LND. Median age of involuntary movements and self-injuries appearance in LND was 1.0 and 3 years, respectively. Renal manifestations (66.3% of patients) occurred at any age with a median onset age of 1.1 years, while gout (25.7% of patients) appeared later in disease course (median onset age 18 years) and was more frequent in attenuated variants than in LND. HPRT activity and genotype showed a significant correlation with the severity of the neurological disease. On the contrary, there were no significant differences in the development of nephropathy or gout. For the treatment of neurological aspects, botulinum toxin injections, oral or intrathecal baclofen and gabapentin were partially efficacious and well tolerated, while deep brain stimulation was associated to a worsening of patients' condition. CONCLUSIONS: The present study improves the knowledge of the natural history of HPRT deficiency and could represent a starting point for the development of future management guidelines.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/genetics , Adolescent , Adult , Child , Disease Management , Female , France , Humans , Italy , Lesch-Nyhan Syndrome/complications , Lesch-Nyhan Syndrome/diagnosis , Male , Mutation , Prognosis , Retrospective Studies , Young Adult
7.
Stem Cells ; 37(7): 876-887, 2019 07.
Article in English | MEDLINE | ID: mdl-30895693

ABSTRACT

In spite of the progress in gene editing achieved in recent years, a subset of genetic diseases involving structural chromosome abnormalities, including aneuploidies, large deletions and complex rearrangements, cannot be treated with conventional gene therapy approaches. We have previously devised a strategy, dubbed chromosome transplantation (CT), to replace an endogenous mutated chromosome with an exogenous normal one. To establish a proof of principle for our approach, we chose as disease model the chronic granulomatous disease (CGD), an X-linked severe immunodeficiency due to abnormalities in CYBB (GP91) gene, including large genomic deletions. We corrected the gene defect by CT in induced pluripotent stem cells (iPSCs) from a CGD male mouse model. The Hprt gene of the endogenous X chromosome was inactivated by CRISPR/Cas9 technology thus allowing the exploitation of the hypoxanthine-aminopterin-thymidine selection system to introduce a normal donor X chromosome by microcell-mediated chromosome transfer. X-transplanted clones were obtained, and diploid XY clones which spontaneously lost the endogenous X chromosome were isolated. These cells were differentiated toward the myeloid lineage, and functional granulocytes producing GP91 protein were obtained. We propose the CT approach to correct iPSCs from patients affected by other X-linked diseases with large deletions, whose treatment is still unsatisfactory. Stem Cells 2019;37:876-887.


Subject(s)
Chromosomes, Mammalian , Genetic Therapy/methods , Granulocytes/metabolism , Granulomatous Disease, Chronic/therapy , Hypoxanthine Phosphoribosyltransferase/genetics , Induced Pluripotent Stem Cells/metabolism , NADPH Oxidase 2/genetics , Aminopterin/metabolism , Aminopterin/pharmacology , Animals , Base Sequence , CRISPR-Cas Systems , Cell Differentiation , Clone Cells , Culture Media/chemistry , Disease Models, Animal , Gene Editing/methods , Granulocytes/cytology , Granulocytes/drug effects , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/metabolism , Granulomatous Disease, Chronic/pathology , Humans , Hypoxanthine/metabolism , Hypoxanthine/pharmacology , Hypoxanthine Phosphoribosyltransferase/deficiency , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Male , Mice , NADPH Oxidase 2/deficiency , Proof of Concept Study , Sequence Deletion , Thioguanine/metabolism , Thioguanine/pharmacology , Thymidine/metabolism , Thymidine/pharmacology , X Chromosome/chemistry , X Chromosome/metabolism
8.
Comput Biol Med ; 107: 161-171, 2019 04.
Article in English | MEDLINE | ID: mdl-30831305

ABSTRACT

The nucleotide salvage pathway is used to recycle degraded nucleotides (purines and pyrimidines); one of the enzymes that helps to recycle purines is hypoxanthine guanine phosphoribosyl transferase 1 (HGPRT1). Therefore, defects in this enzyme lead to the accumulation of DNA and nucleotide lesions and hence replication errors and genetic disorders. Missense mutations in hypoxanthine phosphoribosyl transferase 1 (HPRT1) are associated with deficiencies such as Lesch-Nyhan disease and chronic gout, which have manifestations such as arthritis, neurodegeneration, and cognitive disorders. In the present study, we collected 88 non-synonymous single nucleotide polymorphisms (nsSNPs) from the UniProt, dbSNP, ExAC, and ClinVar databases. We used a series of sequence-based and structure-based in silico tools to prioritize and characterize the most pathogenic and stabilizing or destabilizing nsSNPs. Moreover, to obtain the structural impact of the pathogenic mutations, we mapped the mutations to the crystal structure of the HPRT protein. We further subjected these mutant proteins to a 50 ns molecular dynamics simulation (MDS). The MDS trajectory showed that all mutant proteins altered the structural conformation and dynamic behavior of the HPRT protein and corroborated its association with LND and gout. This study provides essential information regarding the use of HPRT protein mutants as potential targets for therapeutic development.


Subject(s)
Gout , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome , Mutation, Missense/genetics , DNA Mutational Analysis , Gout/genetics , Gout/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/chemistry , Hypoxanthine Phosphoribosyltransferase/metabolism , Lesch-Nyhan Syndrome/genetics , Lesch-Nyhan Syndrome/metabolism , Molecular Dynamics Simulation
10.
Urol Int ; 102(2): 233-237, 2019.
Article in English | MEDLINE | ID: mdl-30384379

ABSTRACT

Kelley-Seegmiller syndrome (KSS) is a disorder that occurs when there is a partial deficiency of the enzyme hypoxanthine guanine phosphoribosyl transferase. It is involved in the metabolism of purines, clinically manifesting as hyperuricemia, hyperuricosuria, gout arthritis, and urolithiasis. The aim of this article is to present the case of a 33-year-old male with KSS, with left ureteral colic, and a 5-mm, 323-HU ureteral calculi, successfully managed with conservative management. It is critical to recognize that most urologists are not familiar with this inborn metabolic error and 75% of these patients will be affected by urolithiasis, thus making it a very critical and significant disease in our practice.


Subject(s)
Conservative Treatment , Gout/therapy , Hypoxanthine Phosphoribosyltransferase/deficiency , Kidney/metabolism , Renal Colic/therapy , Ureteral Calculi/therapy , Uric Acid/metabolism , Urologists , Adult , Gout/diagnosis , Gout/genetics , Gout/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Kidney/diagnostic imaging , Male , Professional Role , Renal Colic/diagnosis , Renal Colic/genetics , Renal Colic/metabolism , Tomography, X-Ray Computed , Treatment Outcome , Ureteral Calculi/diagnosis , Ureteral Calculi/genetics , Ureteral Calculi/metabolism
11.
Auton Neurosci ; 210: 55-64, 2018 03.
Article in English | MEDLINE | ID: mdl-29305058

ABSTRACT

Mutations in the HGPRT1 gene, which encodes hypoxanthine-guanine phosphoribosyltransferase (HGprt), housekeeping enzyme responsible for recycling purines, lead to Lesch-Nyhan disease (LND). Clinical expression of LND indicates that HGprt deficiency has adverse effects on gastrointestinal motility. Therefore, we aimed to evaluate intestinal motility in HGprt knockout mice (HGprt¯). Spontaneous and neurally evoked mechanical activity was recorded in vitro as changes in isometric tension in circular muscle strips of distal colon. HGprt¯ tissues showed a lower in amplitude spontaneous activity and atropine-sensitivity neural contraction compared to control mice. The responses to carbachol and to high KCl were reduced, demonstrating a widespread impairment of contractility. L-NAME was not able in the HGprt¯ tissues to restore the large amplitude contractile activity typical of control. In HGprt¯ colon, a reduced expression of dopaminergic D1 receptor was observed together with the loss of its tonic inhibitory activity present in control-mice. The analysis of inflammatory and oxidative stress in colonic tissue of HGprt¯ mice revealed a significant increase of lipid peroxidation associated with over production of oxygen free radicals. In conclusion, HGprt deficiency in mice is associated with a decrease in colon contractility, not dependent upon reduction of acetylcholine release from the myenteric plexus or hyperactivity of inhibitory signalling. By contrast the increased levels of oxidative stress could partially explain the reduced colon motility in HGprt¯ mice. Colonic dysmotility observed in HGprt¯ mice may mimic the gastrointestinal dysfunctions symptoms of human syndrome, providing a useful animal model to elucidate the pathophysiology of this problem in the LND.


Subject(s)
Gastrointestinal Motility/genetics , Gene Expression Regulation/genetics , Lesch-Nyhan Syndrome/complications , Muscle, Smooth/physiopathology , Animals , Atropine/pharmacology , Benzazepines/pharmacology , Brain/drug effects , Brain/metabolism , Carbachol/pharmacology , Cytokines/metabolism , Disease Models, Animal , Dopamine/metabolism , Enzyme Inhibitors/pharmacology , Evoked Potentials/drug effects , Evoked Potentials/genetics , Face , Gastrointestinal Motility/drug effects , Gene Expression Regulation/drug effects , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/metabolism , In Vitro Techniques , Lesch-Nyhan Syndrome/genetics , Lesch-Nyhan Syndrome/pathology , Lesch-Nyhan Syndrome/physiopathology , Lipid Peroxidation/drug effects , Lipid Peroxidation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Neurotransmitter Agents/pharmacology , Reactive Oxygen Species/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
12.
J Radiat Res ; 59(suppl_2): ii11-ii17, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29053826

ABSTRACT

It is difficult to distinguish radiation-induced events from spontaneous events during induction of stochastic effects, especially in the case of low-dose or low-dose-rate exposures. By using a hypersensitive system for detecting somatic mutations at the HPRT1 locus, we investigated the frequency and spectrum of mutations induced by low-dose X-rays. The mutant frequencies induced by doses of >0.15 Gy were statistically significant when compared with the spontaneous frequency, and a clear dose dependency was also observed for mutant frequencies at doses of >0.15 Gy. In contrast, mutant frequencies at doses of <0.1 Gy occurred at non-significant levels. The mutation spectrum in HPRT-deficient mutants revealed that the type of mutations induced by low-dose exposures was similar to that seen in spontaneous mutants. An apparent change in mutation type was observed for mutants induced by doses of >0.2 Gy. Our observations suggest that there could be a critical dose for mutation induction at between 0.1 Gy and 0.2 Gy, where mutagenic events are induced by multiple DNA double-strand breaks (DSBs). These observations also suggest that low-dose radiation delivered at doses of <0.1 Gy may not result in DSB-induced mutations but may enhance spontaneous mutagenesis events.


Subject(s)
Mutation/genetics , Radiation , Animals , Cell Line , Chromosomes, Human, X/genetics , Cricetinae , Dose-Response Relationship, Radiation , Genetic Loci , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Models, Genetic , Mutagenesis , Mutation Rate , X-Rays
13.
Clin Chim Acta ; 472: 136-138, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28782500

ABSTRACT

Complete deficiency of hypoxanthine-guanine phosphoribosyltransferase (HPRT) activity causes Lesch Nyhan disease (LND), characterized by hyperuricemia, severe action dystonia, choreoathetosis, ballismus, cognitive and attention deficit and self-injurious behavior. Partial HPRT deficiency is present in patients with Lesch-Nyhan variant (LNV), who present with HPRT-related gout and a variable degree of neurological involvement. The diagnosis of HPRT deficiency relies on clinical, biochemical, enzymatic and molecular data. Patients with HPRT deficiency present low or undetectable HPRT activity in hemolysates, with increased adenine phosphoribosyltransferase (APRT) activity. We present a 9-year-old boy who experienced an episode of macroscopic hematuria with dysuria and left flank pain. He presented hyperuricemia and hyperuricosuria. HPRT and APRT activities were both normal in hemolysate; however, HPRT activity assayed in intact erythrocytes was 50% of control levels. A new missense point mutation c.424 A>G (T142A) was found in the HPRT1 gene. The apparent Michaelis constant (Km) for 5-phosphoribosyl-pyrophosphate assayed in patient hemolysate was 20-fold of control levels. In conclusion, we report a patient with HPRT deficiency who presented with both normal HPRT and APRT activity in hemolysate, in which the enzyme activity determined in intact erythrocytes was of diagnostic utility.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/deficiency , Child , Erythrocytes/metabolism , Hemolysis , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Male , Mutation, Missense
14.
Pharm Res ; 34(7): 1477-1490, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28508122

ABSTRACT

PURPOSE: Because of the evolutionary loss of the uricolytic pathway, humans accumulate poorly soluble urate as the final product of purine catabolism. Restoration of uricolysis through enzyme therapy is a promising treatment for severe hyperuricemia caused by deficiency of hypoxanthine-guanine phosphoribosyltransferase (HPRT). To this end, we studied the effect of PEG conjugation on the activity and stability of the enzymatic complement required for conversion of urate into the more soluble (S)-allantoin. METHODS: We produced in recombinant form three zebrafish enzymes required in the uricolytic pathway. We carried out a systematic study of the effect of PEGylation on the function and stability of the three enzymes by varying PEG length, chemistry and degree of conjugation. We assayed in vitro the uricolytic activity of the PEGylated enzymatic triad. RESULTS: We defined conditions that allow PEGylated enzymes to retain native-like enzymatic activity even after lyophilization or prolonged storage. A combination of the three enzymes in an appropriate ratio allowed efficient conversion of urate to (S)-allantoin with no accumulation of intermediate metabolites. CONCLUSIONS: Pharmaceutical restoration of the uricolytic pathway is a viable approach for the treatment of severe hyperuricemia.


Subject(s)
Amidohydrolases/chemistry , Carboxy-Lyases/chemistry , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome/drug therapy , Polyethylene Glycols/chemistry , Urate Oxidase/chemistry , Uricosuric Agents/chemistry , Allantoin/chemistry , Animals , Enzyme Therapy , Humans , Hyperuricemia/drug therapy , Molecular Weight , Recombinant Proteins/chemistry , Solubility , Stereoisomerism , Uric Acid/chemistry , Zebrafish
15.
Nucleosides Nucleotides Nucleic Acids ; 36(2): 151-157, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28045594

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 three novel independent mutations in the coding region of the HPRT1 gene from genomic DNA of (a) a carrier sister of two male patients with LND: c.569G>C, p.G190A in exon 8; and (b) two LND affected male patients unrelated to her who had two mutations: c.648delC, p.Y216X, and c.653C>G, p.A218G in exon 9. Molecular analysis reveals the heterogeneity of genetic mutation of the HPRT1 gene responsible for the HGprt deficiency. It allows fast, accurate detection of carriers and genetic counseling.


Subject(s)
Amino Acid Substitution , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/genetics , Adult , Alanine , Child, Preschool , Exons , Female , Glycine , Heterozygote , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Infant , Male , Mutation
16.
Clin Chim Acta ; 460: 46-9, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27288985

ABSTRACT

Hyperuricemia depends on the balance of endogenous production and renal excretion of uric acid. Transporters for urate are located in the proximal tubule where uric acid is secreted and extensively reabsorbed: secretion is principally ensured by the highly variable ABCG2 gene. Enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT) plays a central role in purine metabolism and its deficiency is an X-linked inherited metabolic disorder associated with clinical manifestations of purine overproduction. Here we report the case of a middle-aged man with severe chronic tophaceous gout with a poor response to allopurinol and requiring repeated surgical intervention. We identified the causal mutations in the HPRT1 gene, variant c.481G>T (p.A161S), and in the crucial urate transporter ABCG2, a heterozygous variant c.421C>A (p.Q141K). This case shows the value of an analysis of the genetic background of serum uric acid.


Subject(s)
Genetic Background , Gout/genetics , Uric Acid/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Chronic Disease , Gout/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Male , Middle Aged , Mutation , Neoplasm Proteins/genetics , Organic Anion Transporters/genetics
17.
Life Sci ; 156: 68-73, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27221022

ABSTRACT

AIMS: Lesch-Nyhan disease (LND) is characterized by hyperuricemia as well as neurological and neuropsychiatric symptoms including repetitive self-injurious behavior. Symptoms are caused by a deficiency of the enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT) as a result of a mutation on the X chromosome. To elucidate the pathophysiology of LND, we performed a metabolite screening for brain and serum extracts from HPRT knockout mice as an animal model for LND. MAIN METHODS: Analyses were performed by high performance liquid chromatography (HPLC)-coupled quadrupole time-of-flight mass spectrometry (QTOF-MS). KEY FINDINGS: In brain extracts, we found six metabolites with significantly different contents in wild-type and HPRT-deficient mice. Two compounds we could identify as 5-aminoimidazole-4-carboxamide ribotide (AICAR) and 1-methylimidazole-4-acetic acid (1-MI4AA). Whereas AICAR was accumulated in brains of HPRT knockout mice, 1-MI4AA was decreased in these mice. SIGNIFICANCE: Both metabolites play a role in histidine metabolism and, as a consequence, histamine metabolism. AICAR, in addition, is part of the purine metabolism. Our findings may help to better understand the mechanisms leading to the behavioral phenotype of LND.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/deficiency , Mass Spectrometry/methods , Metabolomics/methods , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Biomarkers/metabolism , Brain/metabolism , Hypoxanthine Phosphoribosyltransferase/metabolism , Imidazoles/pharmacology , Mice, Knockout , Principal Component Analysis , Ribonucleotides/pharmacology
18.
J Neurol Sci ; 365: 169-74, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27206901

ABSTRACT

Lesch-Nyhan syndrome (LNS) is characterized by uric acid overproduction and severe neurobehavioral symptoms, such as recurrent self-mutilative behavior. To learn more about the pathophysiology of the disease, we quantified neurotransmitters and their metabolites in the cerebral hemisphere, cerebellum and the medulla oblongata of HPRT knockout mice, an animal model for LNS, in comparison to the corresponding wild-type. Our analyses included l-glutamate, 4-aminobutanoic acid (GABA), acetylcholine, serotonin, 5-hydroxyindoleacetic acid (5-HIAA), norepinephrine, l-normetanephrine, epinephrine and l-metanephrine and were conducted via high performance liquid chromatography (HPLC) coupled to tandem mass spectrometry (MS/MS). Among these neurotransmitter systems, we did not find any abnormalities in the HPRT knockout mouse brains. On one side, this might indicate that HPRT deficiency most severely affects dopamine signaling, while brain functioning based on other neurotransmitters is more or less spared. On the other hand, our findings may reflect a compensating mechanism for impaired purine salvage that protects the brain in HPRT-deficient mice but not in LNS patients.


Subject(s)
Brain/metabolism , Gene Expression Regulation/genetics , Hypoxanthine Phosphoribosyltransferase/deficiency , Neurotransmitter Agents/metabolism , Animals , Chromatography, High Pressure Liquid , Hypoxanthine Phosphoribosyltransferase/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Tandem Mass Spectrometry
19.
Sci Rep ; 6: 25592, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27185277

ABSTRACT

Lesch-Nyhan disease (LND) is a severe neurological disorder caused by loss-of-function mutations in the gene encoding hypoxanthine phosphoribosyltransferase (HPRT), an enzyme required for efficient recycling of purine nucleotides. Although this biochemical defect reconfigures purine metabolism and leads to elevated levels of the breakdown product urea, it remains unclear exactly how loss of HPRT activity disrupts brain function. As the rat is the preferred rodent experimental model for studying neurobiology and diseases of the brain, we used genetically-modified embryonic stem cells to generate an HPRT knock-out rat. Male HPRT-deficient rats were viable, fertile and displayed normal caged behaviour. However, metabolomic analysis revealed changes in brain biochemistry consistent with disruption of purine recycling and nucleotide metabolism. Broader changes in brain biochemistry were also indicated by increased levels of the core metabolite citrate and reduced levels of lipids and fatty acids. Targeted MS/MS analysis identified reduced levels of dopamine in the brains of HPRT-deficient animals, consistent with deficits noted previously in human LND patients and HPRT knock-out mice. The HPRT-deficient rat therefore provides a new experimental platform for future investigation of how HPRT activity and disruption of purine metabolism affects neural function and behaviour.


Subject(s)
Brain/metabolism , Disease Models, Animal , Dopamine/metabolism , Lesch-Nyhan Syndrome/metabolism , Animals , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/genetics , Male , Metabolomics/methods , Mice, Knockout , Mutation , Purine Nucleotides/metabolism , Rats, Transgenic , Rodentia , Tandem Mass Spectrometry
20.
J Inherit Metab Dis ; 39(1): 85-91, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26067813

ABSTRACT

Lesch-Nyhan disease (LND) is a rare, X-linked recessive neurodevelopmental disorder caused by deficiency of hypoxanthine-guanine phosphoribosyltransferase (HGprt), an enzyme in the purine salvage pathway. HGprt has two functions; it recycles hypoxanthine and guanine. Which of these two functions is more relevant for pathogenesis is unclear because some evidence points to hypoxanthine recycling, but other evidence points to guanine recycling. In this study, we selectively assayed hypoxanthine (Hprt) and guanine (Gprt) recycling in skin fibroblasts from 17 persons with LND, 11 with an attenuated variant of the disease (LNV), and 19 age-, sex-, and race-matched healthy controls (HC). Activity levels of both enzymes differed across groups (p < 0.0001), but only Gprt distinguished patients with LND from those with LNV (p < 0.05). Gprt also showed slightly stronger correlations than Hprt with 13 of 14 measures of the clinical phenotype, including the severity of dystonia, cognitive impairment, and behavioral abnormalities. These findings suggest that loss of guanine recycling might be more closely linked to the LND/LNV phenotype than loss of hypoxanthine recycling.


Subject(s)
Guanine/metabolism , Hypoxanthine/metabolism , Lesch-Nyhan Syndrome/metabolism , Adolescent , Adult , Case-Control Studies , Fibroblasts/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/metabolism , Lesch-Nyhan Syndrome/genetics , Male , Phenotype , Purines/metabolism , Skin/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...