Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Genet Med ; 21(9): 2145-2150, 2019 09.
Article in English | MEDLINE | ID: mdl-30728528

ABSTRACT

PURPOSE: Severe hematotoxicity in patients with thiopurine therapy has been associated with genetic polymorphisms in the thiopurine S-methyltransferase (TPMT). While TPMT genetic testing is clinically implemented for dose individualization, alterations in the nudix hydrolase 15 (NUDT15) emerged as independent determinant of thiopurine-related hematotoxicity. Because data for European patients are limited, we investigated the relevance of NUDT15 in Europeans. METHODS: Additionally to TPMT phenotyping/genotyping, we performed in-depth Sanger sequencing analyses of NUDT15 coding region in 107 European patients who developed severe thiopurine-related hematotoxicity as extreme phenotype. Moreover, genotyping for NUDT15 variants in 689 acute lymphoblastic leukemia (ALL) patients was performed. RESULTS: As expected TPMT was the main cause of severe hematotoxicity in 31% of patients, who were either TPMT deficient (10%) or heterozygous carriers of TPMT variants (21%). By comparison, NUDT15 genetic polymorphism was identified in 14 (13%) patients including one novel variant (p.Met1Ile). Six percent of patients with severe toxicity carried variants in both TPMT and NUDT15. Among patients who developed toxicity within 3 months of treatment, 13% were found to be carriers of NUDT15 variants. CONCLUSION: Taken together, NUDT15 and TPMT genetics explain ~50% of severe thiopurine-related hematotoxicity, providing a compelling rationale for additional preemptive testing of NUDT15 genetics not only in Asians, but also in Europeans.


Subject(s)
Drug Hypersensitivity/genetics , Methyltransferases/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Pyrophosphatases/genetics , Adult , Aged , Aged, 80 and over , Drug Hypersensitivity/etiology , Drug Hypersensitivity/pathology , Drug-Related Side Effects and Adverse Reactions/genetics , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Genetic Predisposition to Disease , Genetic Testing/methods , Humans , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Purine-Pyrimidine Metabolism, Inborn Errors/pathology
2.
Mol Genet Metab ; 124(4): 243-253, 2018 08.
Article in English | MEDLINE | ID: mdl-29801986

ABSTRACT

Carbamoyl phosphate synthetase 1 (CPS1) is a urea cycle enzyme that forms carbamoyl phosphate from bicarbonate, ammonia and ATP. Bi-allelic mutations of the CPS1 gene result in a urea cycle disorder presenting with hyperammonemia, often with reduced citrulline, and without orotic aciduria. CPS1 deficiency is particularly challenging to treat and lack of early recognition typically results in early neonatal death. Therapeutic interventions have limited efficacy and most patients develop long-term neurologic sequelae. Using transgenic techniques, we generated a conditional Cps1 knockout mouse. By loxP/Cre recombinase technology, deletion of the Cps1 locus was achieved in adult transgenic animals using a Cre recombinase-expressing adeno-associated viral vector. Within four weeks from vector injection, all animals developed hyperammonemia without orotic aciduria and died. Minimal CPS1 protein was detectable in livers. To investigate the efficacy of gene therapy for CPS deficiency following knock-down of hepatic endogenous CPS1 expression, we injected these mice with a helper-dependent adenoviral vector (HDAd) expressing the large murine CPS1 cDNA under control of the phosphoenolpyruvate carboxykinase promoter. Liver-directed HDAd-mediated gene therapy resulted in survival, normalization of plasma ammonia and glutamine, and 13% of normal Cps1 expression. A gender difference in survival suggests that female mice may require higher hepatic CPS1 expression. We conclude that this conditional murine model recapitulates the clinical and biochemical phenotype detected in human patients with CPS1 deficiency and will be useful to investigate ammonia-mediated neurotoxicity and for the development of cell- and gene-based therapeutic approaches.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/therapy , Genetic Therapy , Hyperammonemia/therapy , Ammonia/metabolism , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/therapeutic use , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/metabolism , Carbamoyl-Phosphate Synthase I Deficiency Disease/pathology , Carbamyl Phosphate/metabolism , Female , Gene Expression Regulation, Enzymologic , Glutamine/metabolism , Humans , Hyperammonemia/genetics , Hyperammonemia/metabolism , Hyperammonemia/pathology , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Knockout , Mutation , Orotate Phosphoribosyltransferase/deficiency , Orotate Phosphoribosyltransferase/genetics , Orotidine-5'-Phosphate Decarboxylase/deficiency , Orotidine-5'-Phosphate Decarboxylase/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/pathology
3.
Ann Biol Clin (Paris) ; 75(4): 445-449, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28751290

ABSTRACT

Myoadenylate deaminase deficit (MAD, MIM#615511) is the most common cause of metabolic myopathies with an estimated prevalence of 1-2% in the general population. We report the case of a 39-year-old man suffering from severe skeletal muscle pain that had developed gradually for 4 years. A moderate increase in creatine kinase (CK) was the only biological sign observed. This study takes a closer look at a common but poorly known pathology and highlights the interest of the dynamic metabolic investigations carried out during exercise stress test with a cycle ergometer. Our non-invasive clinical and biological examination, at the interface between physiology and biology, disclosed the total absence of a physiological increase in plasma ammonia evocative of MAD. However, MAD was later confirmed by histochemistry and molecular studies, which revealed the presence of the recurrent homozygous pathogenic variant affecting the adenosine monophosphate deaminase 1 gene (AMPD1) in most patients with MAD.


Subject(s)
AMP Deaminase/deficiency , Exercise Test , Myalgia/diagnosis , Purine-Pyrimidine Metabolism, Inborn Errors/diagnosis , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , AMP Deaminase/genetics , Adult , Diagnosis, Differential , Exercise Tolerance/genetics , Homozygote , Humans , Male , Mutation , Myalgia/genetics , Myalgia/pathology , Purine-Pyrimidine Metabolism, Inborn Errors/complications , Purine-Pyrimidine Metabolism, Inborn Errors/pathology
4.
Rom J Morphol Embryol ; 58(1): 211-217, 2017.
Article in English | MEDLINE | ID: mdl-28523321

ABSTRACT

The association between two autoimmune diseases is known in the literature as overlap syndrome. We present the case of an 18-year-old boy, diagnosed at the age of 13 with an overlap syndrome between type I autoimmune hepatitis and sclerosing cholangitis. The response to immunosuppressant therapy was hampered by azathioprine-induced toxicity causing severe pancytopenia, as a result of thiopurine methyltransferase enzyme genetic deficiency. Treatment was replaced by mycophenolate mofetil. Although the relapse rate was reduced, the disease progressed to cirrhosis. Specific features of this case were the overlap syndrome, young age of onset, especially for sclerosing cholangitis, azathioprine toxicity that influenced the prognosis and the treatment problems regarding the use and efficiency of alternative immunosuppressant agents in pediatric patients.


Subject(s)
Cholangitis, Sclerosing/complications , Drug Hypersensitivity/complications , Hepatitis, Autoimmune/complications , Purine-Pyrimidine Metabolism, Inborn Errors/complications , Adolescent , CD8-Positive T-Lymphocytes/pathology , Cholangitis, Sclerosing/pathology , Dendritic Cells/pathology , Drug Hypersensitivity/pathology , Hepatitis, Autoimmune/pathology , Humans , Inflammation/pathology , Liver/pathology , Macrophages/pathology , Male , Purine-Pyrimidine Metabolism, Inborn Errors/pathology
5.
PLoS One ; 10(7): e0132972, 2015.
Article in English | MEDLINE | ID: mdl-26207760

ABSTRACT

AIM: Our aim was to evaluate the accuracy of aerobic exercise testing to diagnose metabolic myopathies. METHODS: From December 2008 to September 2012, all the consecutive patients that underwent both metabolic exercise testing and a muscle biopsy were prospectively enrolled. Subjects performed an incremental and maximal exercise testing on a cycle ergometer. Lactate, pyruvate, and ammonia concentrations were determined from venous blood samples drawn at rest, during exercise (50% predicted maximal power, peak exercise), and recovery (2, 5, 10, and 15 min). Biopsies from vastus lateralis or deltoid muscles were analysed using standard techniques (reference test). Myoadenylate deaminase (MAD) activity was determined using p-nitro blue tetrazolium staining in muscle cryostat sections. Glycogen storage was assessed using periodic acid-Schiff staining. The diagnostic accuracy of plasma metabolite levels to identify absent and decreased MAD activity was assessed using Receiver Operating Characteristic (ROC) curve analysis. RESULTS: The study involved 51 patients. Omitting patients with glycogenoses (n = 3), MAD staining was absent in 5, decreased in 6, and normal in 37 subjects. Lactate/pyruvate at the 10th minute of recovery provided the greatest area under the ROC curves (AUC, 0.893 ± 0.067) to differentiate Abnormal from Normal MAD activity. The lactate/rest ratio at the 10th minute of recovery from exercise displayed the best AUC (1.0) for discriminating between Decreased and Absent MAD activities. The resulting decision tree achieved a diagnostic accuracy of 86.3%. CONCLUSION: The present algorithm provides a non-invasive test to accurately predict absent and decreased MAD activity, facilitating the selection of patients for muscle biopsy and target appropriate histochemical analysis.


Subject(s)
AMP Deaminase/deficiency , Algorithms , Exercise Test , Glycogen Storage Disease/diagnosis , Purine-Pyrimidine Metabolism, Inborn Errors/diagnosis , AMP Deaminase/metabolism , Adolescent , Adult , Biopsy , Exercise/physiology , Female , Glycogen Storage Disease/metabolism , Glycogen Storage Disease/pathology , Humans , Male , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Purine-Pyrimidine Metabolism, Inborn Errors/metabolism , Purine-Pyrimidine Metabolism, Inborn Errors/pathology , Young Adult
6.
Pharmacogenomics ; 16(8): 891-903, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26067482

ABSTRACT

Azathioprine and 6-mercaptopurine remain pivotal therapies for the maintenance of disease remission in patients with Crohn's disease and ulcerative colitis. While thiopurine S-methyltransferase deficiency was the first pharmacogenetic phenomenon to be recognized to influence thiopurine toxicity and reliably predict leukopenia, it does not predict other adverse effects, nor does it explain most cases of thiopurine resistance. In recent years, a number of other genetic polymorphisms have received increasing attention in the literature. In particular, SNPs in NUDT15 and in the class II HLA locus have been shown to predict thiopurine-related leukopenia and pancreatitis. The aim of this review is to provide a concise update of genetic variability which may influence patient response to azathioprine and 6-mercaptopurine.


Subject(s)
Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Drug-Related Side Effects and Adverse Reactions/genetics , Inflammatory Bowel Diseases/drug therapy , Azathioprine/therapeutic use , Colitis, Ulcerative/genetics , Crohn Disease/genetics , Drug Hypersensitivity/genetics , Drug Hypersensitivity/pathology , Drug-Related Side Effects and Adverse Reactions/pathology , Genotype , Histocompatibility Antigens Class II/genetics , Humans , Inflammatory Bowel Diseases/genetics , Mercaptopurine/therapeutic use , Pharmacogenetics , Polymorphism, Single Nucleotide , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/pathology , Pyrophosphatases/genetics
7.
J Allergy Clin Immunol ; 134(1): 155-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24767876

ABSTRACT

BACKGROUND: Purine nucleoside phosphorylase (PNP) deficiency is a rare form of autosomal recessive combined primary immunodeficiency caused by a enzyme defect leading to the accumulation of inosine, 2'-deoxy-inosine (dIno), guanosine, and 2'-deoxy-guanosine (dGuo) in all cells, especially lymphocytes. Treatments are available and curative for PNP deficiency, but their efficacy depends on the early approach. PNP-combined immunodeficiency complies with the criteria for inclusion in a newborn screening program. OBJECTIVE: This study evaluate whether mass spectrometry can identify metabolite abnormalities in dried blood spots (DBSs) from affected patients, with the final goal of individuating the disease at birth during routine newborn screening. METHODS: DBS samples from 9 patients with genetically confirmed PNP-combined immunodeficiency, 10,000 DBS samples from healthy newborns, and 240 DBSs from healthy donors of different age ranges were examined. Inosine, dIno, guanosine, and dGuo were tested by using tandem mass spectrometry (TMS). T-cell receptor excision circle (TREC) and kappa-deleting recombination excision circle (KREC) levels were evaluated by using quantitative RT-PCR only for the 2 patients (patients 8 and 9) whose neonatal DBSs were available. RESULTS: Mean levels of guanosine, inosine, dGuo, and dIno were 4.4, 133.3, 3.6, and 3.8 µmol/L, respectively, in affected patients. No indeterminate or false-positive results were found. In patient 8 TREC levels were borderline and KREC levels were abnormal; in patient 9 TRECs were undetectable, whereas KREC levels were normal. CONCLUSION: TMS is a valid method for diagnosis of PNP deficiency on DBSs of affected patients at a negligible cost. TMS identifies newborns with PNP deficiency, whereas TREC or KREC measurement alone can fail.


Subject(s)
Immunologic Deficiency Syndromes/diagnosis , Mutation , Purine-Nucleoside Phosphorylase/deficiency , Purine-Nucleoside Phosphorylase/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/diagnosis , Adolescent , Child, Preschool , DNA Repair , Deoxyguanosine/analysis , Deoxyguanosine/metabolism , Dried Blood Spot Testing , Female , Guanosine/analysis , Guanosine/metabolism , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Infant , Infant, Newborn , Inosine/analogs & derivatives , Inosine/analysis , Inosine/metabolism , Lymphocytes/pathology , Male , Neonatal Screening , Primary Immunodeficiency Diseases , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/pathology , Tandem Mass Spectrometry
8.
Genes Cells ; 17(1): 28-38, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22212473

ABSTRACT

The AMPD2 gene, a member of the AMPD gene family encoding AMP deaminase, is widely expressed in nonmuscle tissues including kidney, although its functions have not been fully elucidated. In this study, we studied the function of the AMPD2 gene by establishing AMPD2-deficient model animal. We established AMPD2 knockout mice by using gene transfer and homologous recombination in murine ES cells and studied phenotypes and functions in the kidneys of these animals. AMPD activity was decreased from 22.9 mIU/mg protein to 2.5 mIU/mg protein in the kidneys of AMPD knockout mice. In addition to changes in nucleotide metabolism in the kidneys, proteinuria was found in 3-week-old AMPD2 knockout mice, followed by a further increment up to a peak level at 6 weeks old (up to 0.6 g/dL). The major protein component in the urine of AMPD2 knockout mice was found to be albumin, indicating that AMPD2 may have a key role in glomerular filtration. Indeed, an ultrastructure study of glomerulus specimens from these mice showed effacement of the podocyte foot processes, resembling minimal-change nephropathy in humans. Based on our results, we concluded that AMPD2 deficiency induces imbalanced nucleotide metabolism and proteinuria, probably due to podocyte dysfunction.


Subject(s)
Kidney Glomerulus/pathology , Kidney/metabolism , Nucleotide Deaminases/metabolism , Nucleotides/metabolism , Proteinuria/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/physiopathology , AMP Deaminase/deficiency , AMP Deaminase/genetics , Animals , Kidney Glomerulus/metabolism , Mice , Mice, Knockout , Proteinuria/metabolism , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/pathology
9.
Eur J Pediatr ; 171(1): 131-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21625931

ABSTRACT

Adenylosuccinate lyase (ADSL) deficiency is a rare autosomal recessive disorder of purine metabolism. Patients may present with a wide range of neurological symptoms. Head imaging abnormalities have been reported only rarely in the scientific literature and include atrophy of the cerebral cortex, corpus callosum, cerebellar vermis, lack of myelination, delayed myelination, anomalies of the white matter, and lissencephaly. The pathogenesis of abnormalities remains unknown. To further the understanding of the spectrum of brain abnormalities associated with ADSL deficiency, we examined the magnetic resonance findings in seven Polish patients with different clinical phenotypes and genotypes. Head MRI showed impaired white matter myelination with various degrees of global supra- and infratentorial white matter loss including widening of the lateral ventricles, enlargement of the subarachnoid spaces, atrophy of the cerebrum, hypoplasia of the cerebellar hemispheres and enlargement of the cisterna magna, and white matter abnormal hyperintense signal on T(2)-weighted sequences. We recommend performing a detailed analysis of urine and plasma purine metabolites in patients who have neurological findings, including developmental delay, microcephaly, autistic features, neonatal encephalopathy, and seizures especially if MRI findings such as delayed or lack of myelination, white matter abnormal signal, and atrophy of the cerebrum and/or cerebellum are also present. Greater awareness of adenylosuccinate lyase deficiency among general pediatricians, neonatologists, pediatric neurologists, and also radiologists is the key to identifying the disorder at an early stage.


Subject(s)
Brain/pathology , Purine-Pyrimidine Metabolism, Inborn Errors/pathology , Adenylosuccinate Lyase/deficiency , Autistic Disorder , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Microcephaly/etiology , Purine-Pyrimidine Metabolism, Inborn Errors/complications , Retrospective Studies
10.
Eur J Paediatr Neurol ; 16(4): 343-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21903433

ABSTRACT

BACKGROUND: Adenylosuccinate lyase (ADSL) deficiency is an autosomal recessive disorder of the purine synthesis which results in accumulation of succinylpurines (succinyladenosine (S-Ado) and succinylamino-imidazole carboxamide riboside (SAICAr)) in body fluids. Patients present developmental delay, often accompanied by epilepsy and autistic spectrum disorders. OBJECTIVES: To describe atypical neurological features in the evolution of three novel unrelated cases of ADSL deficiency. PATIENTS: A 9-year-old boy with severe cognitive impairment and autistic behaviour received d-ribose therapy for one year. Drug withdrawal was associated with acute neurological deterioration, severe brain atrophy and demyelination on MRI. The second patient is a 5.5-year-old girl with mild developmental delay who presented a benign course with moderate cognitive impairment as the only feature in her evolution. The final patient is a 14-year-old boy with severe cognitive impairment who developed drug-resistant epilepsy and bathing reflex seizures, progressive spasticity in the lower limbs and thoracic deformity. METHODS: SAICAr and S-Ado in urine were analysed by HPLC with diode array detection. Diagnosis was confirmed by molecular analysis of the ADSL gene. RESULTS: An elevation of S-Ado and SAICAr excretion in urine was detected in all three patients. The patients were homozygous for the missence change p.I369L and for the novel change p.M389V. CONCLUSION: Drug-resistant epilepsy and specific therapeutic interventions may modify the neurological outcome in ADSL deficiency. d-ribose must be considered with caution as, in our experience, it returns no clinical benefit and drug withdrawal can precipitate status epilepticus and acute neurological deterioration.


Subject(s)
Adenylosuccinate Lyase/deficiency , Purine-Pyrimidine Metabolism, Inborn Errors/psychology , Adenylosuccinate Lyase/genetics , Adolescent , Autistic Disorder , Brain/pathology , Child , Child Behavior Disorders/etiology , Child, Preschool , Cognition Disorders/etiology , Education, Special , Epilepsy/etiology , Epilepsy, Reflex/etiology , Female , Fever/etiology , Humans , Magnetic Resonance Imaging , Male , Movement Disorders/etiology , Muscle Spasticity/etiology , Neuropsychological Tests , Purine-Pyrimidine Metabolism, Inborn Errors/diagnosis , Purine-Pyrimidine Metabolism, Inborn Errors/pathology , Quadriplegia/etiology , Ribose/therapeutic use
11.
Saudi J Kidney Dis Transpl ; 21(2): 328-31, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20228523

ABSTRACT

Hereditary xanthinuria type I, a defect of purine metabolism, results from a genetic deficiency of xanthine oxidase. It is an uncommon cause of stone formation in children. We report here two children with xanthine urolithiasis. The first patient was an 8-year-old boy who presented with repeated episodes of hematuria evaluated with excretory urography, which demonstrated radio-lucent pelvic stone in the right kidney, causing hydronephrosis. He had pyelolithotomy, and the extracted stone consisted of pure xanthine. Family study revealed an asymptomatic xanthinuria in younger brother. The second patient was a 5-year-old boy who had a 2-week history of abdominal pain and gross hematuria. Conventional excretory intravenous urography showed a non-functioning right kidney. Nephrectomy was performed, and histology revealed end-stage pyelonephritis. The calculi consisted of pure xanthine. In both patients, plasma and urinary concentrations of uric acid were low but xanthine and hypoxanthine concentrations were markedly elevated. Xanthine urolithiasis is usually a benign condition, easy to prevent or cure by appropriate alkalinization, forced hydration and restriction of dietary purines. However asymptomatic, and therefore undiagnosed, stones may invade the kidney and urinary tract, resulting in destruction of parenchyma, nephrectomy and renal failure.


Subject(s)
Purine-Pyrimidine Metabolism, Inborn Errors/diagnosis , Urolithiasis/genetics , Xanthine/urine , Abdominal Pain/genetics , Biomarkers/urine , Biopsy , Child , Child, Preschool , Hematuria/genetics , Humans , Hydronephrosis/genetics , Male , Nephrectomy , Purine-Pyrimidine Metabolism, Inborn Errors/complications , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/pathology , Purine-Pyrimidine Metabolism, Inborn Errors/urine , Pyelonephritis/genetics , Treatment Outcome , Urography , Urolithiasis/diagnostic imaging , Urolithiasis/pathology , Urolithiasis/urine , Urologic Surgical Procedures, Male
12.
Neurochem Int ; 56(3): 367-78, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20005278

ABSTRACT

This review is devised to gather the presently known inborn errors of purine metabolism that manifest neurological pediatric syndromes. The aim is to draw a comprehensive picture of these rare diseases, characterized by unexpected and often devastating neurological symptoms. Although investigated for many years, most purine metabolism disorders associated to psychomotor dysfunctions still hide the molecular link between the metabolic derangement and the neurological manifestations. This basically indicates that many of the actual functions of nucleosides and nucleotides in the development and function of several organs, in particular central nervous system, are still unknown. Both superactivity and deficiency of phosphoribosylpyrophosphate synthetase cause hereditary disorders characterized, in most cases, by neurological impairments. The deficiency of adenylosuccinate lyase and 5-amino-4-imidazolecarboxamide ribotide transformylase/IMP cyclohydrolase, both belonging to the de novo purine synthesis pathway, is also associated to severe neurological manifestations. Among catabolic enzymes, hyperactivity of ectosolic 5'-nucleotidase, as well as deficiency of purine nucleoside phosphorylase and adenosine deaminase also lead to syndromes affecting the central nervous system. The most severe pathologies are associated to the deficiency of the salvage pathway enzymes hypoxanthine-guanine phosphoribosyltransferase and deoxyguanosine kinase: the former due to an unexplained adverse effect exerted on the development and/or differentiation of dopaminergic neurons, the latter due to a clear impairment of mitochondrial functions. The assessment of hypo- or hyperuricemic conditions is suggestive of purine enzyme dysfunctions, but most disorders of purine metabolism may escape the clinical investigation because they are not associated to these metabolic derangements. This review may represent a starting point stimulating both scientists and physicians involved in the study of neurological dysfunctions caused by inborn errors of purine metabolism with the aim to find novel therapeutical approaches.


Subject(s)
Brain Diseases, Metabolic, Inborn/metabolism , Brain Diseases, Metabolic, Inborn/physiopathology , Purine-Pyrimidine Metabolism, Inborn Errors/metabolism , Purine-Pyrimidine Metabolism, Inborn Errors/physiopathology , Purines/metabolism , Age Factors , Brain Diseases, Metabolic, Inborn/pathology , Central Nervous System/growth & development , Central Nervous System/metabolism , Central Nervous System/physiopathology , Child , Child, Preschool , Enzymes/metabolism , Humans , Infant , Infant, Newborn , Metabolic Networks and Pathways/physiology , Neurons/metabolism , Neurons/pathology , Nucleotides/metabolism , Purine-Pyrimidine Metabolism, Inborn Errors/pathology
13.
Folia Neuropathol ; 47(4): 314-20, 2009.
Article in English | MEDLINE | ID: mdl-20054783

ABSTRACT

Adenylosuccinate lyase (ADSL) deficiency is an autosomal recessive disorder caused by mutation in the ADSL gene. The disease was identified in 1984 by Jaeken and van der Berghe as the first inborn defect of purine biosynthesis. Affected children revealed encephalopathy with epilepsy and marked psychomotor retardation. A neurological examination showed hypotonia, followed sometimes after years by spasticity. The diagnosis is based on detection in the urine and CSF succinyladenosine (S-Ado) and succinylaminoimidazole carboxamide ribotide (SAICAr). We present brain MR examinations of seven patients with ADSL deficiency in the correlation with their clinical findings. In all cases lack of myelination or of delayed myelination of cerebral white matter was seen. Additionally cerebral and cerebellar atrophy was observed. Neuropathological findings revealed damage of all cellular elements of brain tissue and are cause of observed MR changes. Hypo/dysmyelination seemed to be secondary to damage of oligodendroglia and axons of damaged neuronal cells.


Subject(s)
Adenylosuccinate Lyase/deficiency , Brain/pathology , Demyelinating Diseases/pathology , Encephalitis/pathology , Purine-Pyrimidine Metabolism, Inborn Errors/pathology , Adenylosuccinate Lyase/genetics , Atrophy/pathology , Brain Mapping , Child , Child, Preschool , Demyelinating Diseases/genetics , Encephalitis/genetics , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Muscle Hypotonia/etiology , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Muscle Spasticity/etiology , Muscle Spasticity/genetics , Muscle Spasticity/pathology , Mutation , Neurons/pathology , Poland , Purine-Pyrimidine Metabolism, Inborn Errors/complications , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Seizures/etiology , Seizures/genetics , Seizures/pathology
14.
Folia Neuropathol ; 46(1): 81-91, 2008.
Article in English | MEDLINE | ID: mdl-18368630

ABSTRACT

Adenylosuccinase (ADSL) deficiency is an autosomal recessive disorder affecting mainly the nervous system. The disease causes psychomotor retardation, frequently with autistic features and epilepsy. ADSL deficiency may be diagnosed by detection of two abnormal metabolites in body fluids--succinyladenosine (S-Ado) and succinylaminoimidazole carboxamide riboside (SAICAr). It is assumed that the former metabolite is neurotoxic. We present clinical, biochemical and neuropathological findings of a child affected by a severe form of ADSL deficiency. She had progressive neurological symptoms that started immediately after birth and died at 2.5 months of age. Macroscopically the brain showed signs of moderate atrophy. Histological examination of all grey matter structures showed widespread damage of neurons accompanied by microspongiosis of neuropile. Cerebral white matter showed lack of myelination in the centrum semiovale and diffuse spongiosis of neuropile. Myelination appropriate for the age was visible in posterior limb of internal capsule, in striatum, thalamus and in brain stem structures but diffuse destruction of myelin sheets was seen with severe marked astroglial reaction with signs of destruction of the cells and their processes. Ultrastructural examination showed enormous destruction of all cellular elements, but astonishingly mitochondria were relatively spared. The neuropathological changes can be considered as the neurotoxic result of metabolic disturbances connected with adenylosuccinase deficiency.


Subject(s)
Adenylosuccinate Lyase/deficiency , Brain Diseases, Metabolic, Inborn/pathology , Brain/ultrastructure , Purine-Pyrimidine Metabolism, Inborn Errors/pathology , Adenosine/analogs & derivatives , Adenosine/cerebrospinal fluid , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/cerebrospinal fluid , Brain Diseases, Metabolic, Inborn/cerebrospinal fluid , Brain Diseases, Metabolic, Inborn/physiopathology , Female , Humans , Infant , Infant, Newborn , Poland , Purine-Pyrimidine Metabolism, Inborn Errors/cerebrospinal fluid , Purine-Pyrimidine Metabolism, Inborn Errors/physiopathology , Ribonucleosides/cerebrospinal fluid
15.
Brain Dev ; 29(9): 600-2, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17485188

ABSTRACT

Adenylosuccinate lyase (ADSL) deficiency is a rare disease of de novo purine synthesis. The main symptoms are psychomotor retardation, epilepsy, autistic features, occasionally associated with muscular hypotonia. Diagnosis is made by detection of abnormal purine metabolites (succinyladenosine - S-Ado and succinylaminoimidazole carboxamide riboside - SAICAr) in body fluids. The severity of the clinical features correlates with low S-Ado/SAICAr ratio. We report clinical, biochemical and brain MRI findings of a female infant with severe early epilepsy and hypotonia, who died at the age of 10 weeks.


Subject(s)
Adenylosuccinate Lyase/deficiency , Purine-Pyrimidine Metabolism, Inborn Errors/diagnosis , Female , Humans , Infant, Newborn , Magnetic Resonance Imaging/methods , Poland/ethnology , Purine-Pyrimidine Metabolism, Inborn Errors/pathology
16.
Brain Dev ; 29(2): 124-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16949240

ABSTRACT

Progressive multifocal leukoencephalopathy is a demyelinating disease caused by JC virus, an opportunistic infection of the central nervous system. Although the majority of cases are infected with the human immunodeficiency virus (HIV), other immunocompromised patients are also at risk. Purine nucleoside phosphorylase is an enzyme in the purine salvage pathway that reversibly converts inosine to hypoxanthine and guanosine to guanine. Purine nucleoside phosphorylase deficiency is a combined immunodeficiency with a profound cellular defect. Neurologic abnormalities are salient features of this syndrome. We describe for the first time a patient with this rare disorder presented with progressive multifocal leukoencephalopathy.


Subject(s)
Leukoencephalopathy, Progressive Multifocal/etiology , Purine-Nucleoside Phosphorylase/deficiency , Purine-Pyrimidine Metabolism, Inborn Errors/complications , Child , Humans , Leukoencephalopathy, Progressive Multifocal/pathology , Magnetic Resonance Imaging/methods , Male , Purine-Pyrimidine Metabolism, Inborn Errors/pathology
17.
J Biol Chem ; 273(9): 5093-100, 1998 Feb 27.
Article in English | MEDLINE | ID: mdl-9478961

ABSTRACT

Adenosine deaminase (ADA) deficiency in humans leads to a combined immunodeficiency. The mechanisms involved in the lymphoid specificity of the disease are not fully understood due to the inaccessibility of human tissues for detailed analysis and the absence of an adequate animal model for the disease. We report the use of a two-stage genetic engineering strategy to generate ADA-deficient mice that retain many features associated with ADA deficiency in humans, including a combined immunodeficiency. Severe T and B cell lymphopenia was accompanied by a pronounced accumulation of 2'-deoxyadenosine and dATP in the thymus and spleen, and a marked inhibition of S-adenosylhomocysteine hydrolase in these organs. Accumulation of adenosine was widespread among all tissues examined. ADA-deficient mice also exhibited severe pulmonary insufficiency, bone abnormalities, and kidney pathogenesis. These mice have provided in vivo information into the metabolic basis for the immune phenotype associated with ADA deficiency.


Subject(s)
Adenosine Deaminase/deficiency , Purine-Pyrimidine Metabolism, Inborn Errors/enzymology , Severe Combined Immunodeficiency/genetics , Adenosine/metabolism , Adenosine Deaminase/genetics , Adenosylhomocysteinase , Animals , Antigens, CD/analysis , Antigens, Differentiation , B-Lymphocytes/cytology , Bone and Bones/abnormalities , Deoxyadenosines/metabolism , Genetic Engineering/methods , Hydrolases/analysis , Kidney/abnormalities , Lung/pathology , Lymphocyte Count , Lymphopenia , Mice , Pulmonary Valve Insufficiency , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/pathology , Research Design , Severe Combined Immunodeficiency/enzymology , Spleen/pathology , T-Lymphocytes/cytology , Thymus Gland/pathology , Tissue Distribution
19.
J Inherit Metab Dis ; 13(1): 121-4, 1990.
Article in English | MEDLINE | ID: mdl-2109146

ABSTRACT

We present the clinical and biochemical features of a boy with dihydropyrimidine dehydrogenase deficiency, which seem to underline a disease entity of developmental retardation, epilepsy and muscular hypertonia.


Subject(s)
Oxidoreductases/deficiency , Purine-Pyrimidine Metabolism, Inborn Errors/enzymology , Brain/pathology , Dihydrouracil Dehydrogenase (NADP) , Epilepsy/etiology , Humans , Infant , Intellectual Disability/etiology , Male , Muscle Hypertonia/etiology , Purine-Pyrimidine Metabolism, Inborn Errors/complications , Purine-Pyrimidine Metabolism, Inborn Errors/pathology
20.
Minerva Med ; 80(5): 507-9, 1989 May.
Article in Italian | MEDLINE | ID: mdl-2747979

ABSTRACT

A case of hereditary xanthinuria in a 68-year-old man with congestive heart failure and alcoholic liver disease is presented. Urolithiasis and muscular symptoms were absent, and the metabolic error was revealed by hypouricemia, hypouricosuria and excess of xanthine and hypoxanthine excretion in urine. Xanthine oxidase (EC 1.2.3.2) activity in liver tissue was absent, confirming the diagnosis of xanthinuria.


Subject(s)
Purine-Pyrimidine Metabolism, Inborn Errors/metabolism , Xanthines/urine , Aged , Biopsy, Needle , Heart Failure/metabolism , Heart Failure/pathology , Humans , Hypoxanthines/analysis , Liver/enzymology , Liver/pathology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Male , Purine-Pyrimidine Metabolism, Inborn Errors/pathology , Uric Acid/analysis , Xanthine Oxidase/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...