Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
1.
Elife ; 112022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35133277

RESUMO

Adenylosuccinate lyase (ADSL) functions in de novo purine synthesis (DNPS) and the purine nucleotide cycle. ADSL deficiency (ADSLD) causes numerous neurodevelopmental pathologies, including microcephaly and autism spectrum disorder. ADSLD patients have normal serum purine nucleotide levels but exhibit accumulation of dephosphorylated ADSL substrates, S-Ado, and SAICAr, the latter being implicated in neurotoxic effects through unknown mechanisms. We examined the phenotypic effects of ADSL depletion in human cells and their relation to phenotypic outcomes. Using specific interventions to compensate for reduced purine levels or modulate SAICAr accumulation, we found that diminished AMP levels resulted in increased DNA damage signaling and cell cycle delays, while primary ciliogenesis was impaired specifically by loss of ADSL or administration of SAICAr. ADSL-deficient chicken and zebrafish embryos displayed impaired neurogenesis and microcephaly. Neuroprogenitor attrition in zebrafish embryos was rescued by pharmacological inhibition of DNPS, but not increased nucleotide concentration. Zebrafish also displayed phenotypes commonly linked to ciliopathies. Our results suggest that both reduced purine levels and impaired DNPS contribute to neurodevelopmental pathology in ADSLD and that defective ciliogenesis may influence the ADSLD phenotypic spectrum.


Assuntos
Adenilossuccinato Liase/deficiência , Adenilossuccinato Liase/metabolismo , Transtorno Autístico/metabolismo , Neurogênese , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/metabolismo , Animais , Transtorno do Espectro Autista/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Galinhas/metabolismo , Ciliopatias/metabolismo , Dano ao DNA , Humanos , Microcefalia/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fenótipo , Fosfoproteínas/metabolismo , Purinas/metabolismo , Ribonucleotídeos/metabolismo , Peixe-Zebra/metabolismo
2.
Mol Genet Metab ; 136(3): 164-176, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35216884

RESUMO

Inborn errors of purine and pyrimidine (P/P) metabolism are under-reported and rarely mentioned in the general literature or in clinical practice, as well as in reviews dedicated to other inborn errors of metabolism (IEMs). However, their diagnosis is important because genetic counseling can be provided and, in some cases, specific treatment exists that may slow or even reverse clinical signs. The purpose of this review is to provide a practical guideline on the suspicion and investigation of inborn errors of P/P metabolism. Failure of a physician to recognize the presence of these disorders may be devastating for affected infants and children because of its permanent effects in the patient, and for their parents because of implications for future offspring. Diagnosis is crucial because genetic counseling can be provided and, in some cases, specific treatment can be offered that may slow or even reverse clinical symptoms. This review highlights the risk factors in the history, the important examination findings, and the appropriate biochemical investigation of the child. Herein we describe the approach to the diagnosis of P/P disorders and emphasize clinical situations in which physicians should consider these diseases as diagnostic possibilities.


Assuntos
Erros Inatos do Metabolismo da Purina-Pirimidina , Criança , Família , Humanos , Lactente , Erros Inatos do Metabolismo da Purina-Pirimidina/diagnóstico , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Purinas/metabolismo , Pirimidinas/metabolismo , Fatores de Risco
3.
Mol Genet Metab ; 136(3): 190-198, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34998670

RESUMO

Purines are essential molecules that are components of vital biomolecules, such as nucleic acids, coenzymes, signaling molecules, as well as energy transfer molecules. The de novo biosynthesis pathway starts from phosphoribosylpyrophosphate (PRPP) and eventually leads to the synthesis of inosine monophosphate (IMP) by means of 10 sequential steps catalyzed by six different enzymes, three of which are bi-or tri-functional in nature. IMP is then converted into guanosine monophosphate (GMP) or adenosine monophosphate (AMP), which are further phosphorylated into nucleoside di- or tri-phosphates, such as GDP, GTP, ADP and ATP. This review provides an overview of inborn errors of metabolism pertaining to purine synthesis in humans, including either phosphoribosylpyrophosphate synthetase (PRS) overactivity or deficiency, as well as adenylosuccinate lyase (ADSL), 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase (ATIC), phosphoribosylaminoimidazole succinocarboxamide synthetase (PAICS), and adenylosuccinate synthetase (ADSS) deficiencies. ITPase deficiency is being described as well. The clinical spectrum of these disorders is broad, including neurological impairment, such as psychomotor retardation, epilepsy, hypotonia, or microcephaly; sensory involvement, such as deafness and visual disturbances; multiple malformations, as well as muscle presentations or consequences of hyperuricemia, such as gouty arthritis or kidney stones. Clinical signs are often nonspecific and, thus, overlooked. It is to be hoped that this is likely to be gradually overcome by using sensitive biochemical investigations and next-generation sequencing technologies.


Assuntos
Adenilossuccinato Liase , Erros Inatos do Metabolismo da Purina-Pirimidina , Adenilossuccinato Liase/deficiência , Adenilossuccinato Liase/genética , Adenilossuccinato Liase/metabolismo , Transtorno Autístico , Humanos , Inosina Monofosfato , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Purinas
4.
Artigo em Inglês | MEDLINE | ID: mdl-34643162

RESUMO

In June 2021, the Purine and Pyrimidine Society (PPS) organized the 19th biennial symposium on Purine and Pyrimidine metabolism (PP21). Due to the ongoing pandemic, the conference was organized as a webinar over 3 days with sessions dealing with enzymes, cancer, inborn errors, gout among others. The current issue of Nucleosides, Nucleotides & Nucleic Acids is a special issue covering proceedings from PP21-presentations and other PPS-related manuscripts, and in this editorial, we will give an overview of the scientific program of the meeting.


Assuntos
Gota , Erros Inatos do Metabolismo da Purina-Pirimidina , Humanos , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Purinas/metabolismo , Pirimidinas/metabolismo
5.
Front Immunol ; 11: 1257, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32695102

RESUMO

Introduction: Complete or near complete absence of the purine nucleoside phosphorylase (PNP) enzyme causes a profound T cell immunodeficiency and neurological abnormalities that are often lethal in infancy and early childhood. We hypothesized that patients with partial PNP deficiency, characterized by a late and mild phenotype due to residual PNP enzyme, would provide important information about the minimal PNP activity needed for normal development. Methods: Three siblings with a homozygous PNP gene mutation (c.769C>G, p.His257Asp) resulting in partial PNP deficiency were investigated. PNP activity was semi-quantitively assayed by the conversion of [14C]inosine in hemolysates, mononuclear cells, and lymphoblastoid B cells. PNP protein expression was determined by Western Blotting in lymphoblastoid B cells. DNA repair was quantified by measuring viability of lymphoblastoid B cells following ionizing irradiation. Results: A 21-year-old female was referred for recurrent sino-pulmonary infections while her older male siblings, aged 25- and 28- years, did not suffer from significant infections. Two of the siblings had moderately reduced numbers of T, B, and NK cells, while the other had near normal lymphocyte subset numbers. T cell proliferations were normal in the two siblings tested. Hypogammaglobulinemia was noted in two siblings, including one that required immunoglobulin replacement. All siblings had typical (normal) neurological development. PNP activity in various cells from two patients were 8-11% of the normal level. All siblings had normal blood uric acid and increased PNP substrates in the urine. PNP protein expression in cells from the two patients examined was similar to that observed in cells from healthy controls. The survival of lymphoblastoid B cells from 2 partial PNP-deficient patients after irradiation was similar to that of PNP-proficient cells and markedly higher than the survival of cells from a patient with absent PNP activity or a patient with ataxia telangiectasia. Conclusions: Patients with partial PNP deficiency can present in the third decade of life with mild-moderate immune abnormalities and typical development. Near-normal immunity might be achieved with relatively low PNP activity.


Assuntos
Neurogênese , Doenças da Imunodeficiência Primária/imunologia , Doenças da Imunodeficiência Primária/metabolismo , Purina-Núcleosídeo Fosforilase/deficiência , Purina-Núcleosídeo Fosforilase/metabolismo , Erros Inatos do Metabolismo da Purina-Pirimidina/imunologia , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Adulto , Alelos , Análise Mutacional de DNA , Ativação Enzimática , Feminino , Genótipo , Humanos , Imunofenotipagem , Linfócitos/imunologia , Linfócitos/metabolismo , Linfócitos/efeitos da radiação , Masculino , Mutação , Neurogênese/genética , Neurogênese/imunologia , Linhagem , Doenças da Imunodeficiência Primária/genética , Doenças da Imunodeficiência Primária/terapia , Purina-Núcleosídeo Fosforilase/genética , Purina-Núcleosídeo Fosforilase/imunologia , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , Erros Inatos do Metabolismo da Purina-Pirimidina/terapia , Purinas/química , Tolerância a Radiação , Adulto Jovem
6.
Mol Genet Metab ; 129(4): 272-277, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32151545

RESUMO

Methylmalonate semialdehyde dehydrogenase deficiency (MMSDD; MIM 614105) is a rare autosomal recessive defect of valine and pyrimidine catabolism. Four prior MMSDD cases are published. We present a fifth case, along with functional and metabolomic analysis. The patient, born to non-consanguineous parents of East African origin, was admitted at two weeks of age for failure to thrive. She was nondysmorphic, had a normal brain MRI, and showed mild hypotonia. Gastroesophageal reflux occurred with feeding. Urine organic acid assessment identified excess 3-hydroxyisobutyrate and 3-hydroxypropionate, while urine amino acid analysis identified elevated concentrations of ß-aminoisobutyrate and ß-alanine. Plasma amino acids showed an elevated concentration of ß-aminoisobutyrate with undetectable ß-alanine. ALDH6A1 gene sequencing identified a homozygous variant of uncertain significance, c.1261C > T (p.Pro421Ser). Management with valine restriction led to reduced concentration of abnormal analytes in blood and urine, improved growth, and reduced gastroesophageal reflux. Western blotting of patient fibroblast extracts demonstrated a large reduction of methylmalonate semialdehyde dehydrogenase (MMSD) protein. Patient cells displayed compromised mitochondrial function with increased superoxide production, reduced oxygen consumption, and reduced ATP production. Metabolomic profiles from patient fibroblasts demonstrated over-representation of fatty acids and fatty acylcarnitines, presumably due to methylmalonate semialdehyde shunting to ß-alanine and subsequently to malonyl-CoA with ensuing increase of fatty acid synthesis. Previously reported cases of MMSDD have shown variable clinical presentation. Our case continues the trend as clinical phenotypes diverge from prior cases. Recognition of mitochondrial dysfunction and novel metabolites in this patient provide the opportunity to assess future patients for secondary changes that may influence clinical outcome.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Metabolômica , Metilmalonato-Semialdeído Desidrogenase (Acilante)/deficiência , Mitocôndrias/metabolismo , Erros Inatos do Metabolismo da Purina-Pirimidina/diagnóstico , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Biópsia , Linhagem Celular , Feminino , Fibroblastos/metabolismo , Humanos , Recém-Nascido , Metilmalonato-Semialdeído Desidrogenase (Acilante)/metabolismo , Fenótipo , Pele/patologia , Valina/sangue , Valina/metabolismo , Valina/urina
8.
Mol Diagn Ther ; 23(3): 429-438, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30963516

RESUMO

BACKGROUND: Thiopurine S-methyltransferase (TPMT) testing, either by genotyping or phenotyping, can reduce the incidence of adverse severe myelotoxicity episodes induced by azathioprine. The comparative cost-effectiveness of TPMT genotyping and phenotyping are not known. OBJECTIVE: Our aim was to assess the cost-effectiveness of phenotyping-based dosing of TPMT activity, genotyping-based screening and no screening (reference) for patients treated with azathioprine. METHODS: A decision tree was built to compare the conventional weight-based dosing strategy with phenotyping and with genotyping using a micro-simulation model of patients with inflammatory bowel disease from the perspective of the French health care system. The time horizon was set up as 1 year. Only direct medical costs were used. Data used were obtained from previous reports, except for screening test and admission costs, which were from real cases. The main outcome was the cost-effectiveness ratios, with an effectiveness criterion of one averted severe myelotoxicity episode. RESULTS: The total expected cost of the no screening strategy was €409/patient, the total expected cost of the phenotyping strategy was €427/patient, and the total expected cost of the genotyping strategy was €476/patient. The incremental cost-effectiveness ratio was €2602/severe myelotoxicity averted in using the phenotyping strategy, and €11,244/severe myelotoxicity averted in the genotyping strategy compared to the no screening strategy. At prevalence rates of severe myelotoxicity > 1%, phenotyping dominated genotyping and conventional strategies. CONCLUSION: The phenotype-based strategy to screen for TPMT deficiency dominates (cheaper and more effective) the genotype-based screening strategy in France. Phenotype-based screening dominates no screening in populations with a prevalence of severe myelosuppression due to azathioprine of > 1%.


Assuntos
Hipersensibilidade a Drogas/diagnóstico , Hipersensibilidade a Drogas/genética , Genótipo , Metiltransferases/genética , Modelos Biológicos , Fenótipo , Erros Inatos do Metabolismo da Purina-Pirimidina/diagnóstico , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , Antimetabólitos/química , Antimetabólitos/farmacologia , Antimetabólitos/uso terapêutico , Azatioprina/química , Azatioprina/farmacologia , Azatioprina/uso terapêutico , Análise Custo-Benefício , Hipersensibilidade a Drogas/tratamento farmacológico , Hipersensibilidade a Drogas/metabolismo , Testes Genéticos/economia , Testes Genéticos/métodos , Variação Genética , Humanos , Metiltransferases/metabolismo , Erros Inatos do Metabolismo da Purina-Pirimidina/tratamento farmacológico , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo
9.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1182-1191, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30658162

RESUMO

Pyrimidine nucleotides are essential for a vast number of cellular processes and dysregulation of pyrimidine metabolism has been associated with a variety of clinical abnormalities. Inborn errors of pyrimidine metabolism affecting enzymes in the pyrimidine de novo and degradation pathway have been identified but no patients have been described with a deficiency in proteins affecting the cellular import of ribonucleosides. In this manuscript, we report the elucidation of the genetic basis of the observed uridine-cytidineuria in a patient presenting with fever, hepatosplenomegaly, persistent lactate acidosis, severely disturbed liver enzymes and ultimately multi-organ failure. Sequence analysis of genes encoding proteins directly involved in the metabolism of uridine and cytidine showed two variants c.1528C > T (p.R510C) and c.1682G > A (p.R561Q) in SLC28A1, encoding concentrative nucleotide transporter 1 (hCNT1). Functional analysis showed that these variants affected the three-dimensional structure of hCNT1, altered glycosylation and decreased the half-life of the mutant proteins which resulted in impaired transport activity. Co-transfection of both variants, mimicking the trans disposition of c.1528C > T (p.R510C) and c.1682G > A (p.R561Q) in the patient, significantly impaired hCNT1 biological function. Whole genome sequencing identified two pathogenic variants c.50delT; p.(Leu17Argfs*34) and c.853_855del; p.(Lys285del) in the PRF1 gene, indicating that our patient was also suffering from Familial Hemophagocytic Lymphohistiocytosis type 2. The identification of two co-existing monogenic defects might have resulted in a blended phenotype. Thus, the clinical presentation of isolated hCNT1 deficiency remains to be established.


Assuntos
Proteínas de Membrana Transportadoras/deficiência , Insuficiência de Múltiplos Órgãos/metabolismo , Perforina/deficiência , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Pirimidinas/metabolismo , Evolução Fatal , Humanos , Lactente , Recém-Nascido , Masculino , Proteínas de Membrana Transportadoras/genética , Insuficiência de Múltiplos Órgãos/genética , Perforina/genética , Fenótipo , Erros Inatos do Metabolismo da Purina-Pirimidina/genética
10.
PLoS One ; 12(11): e0187266, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29095874

RESUMO

PURPOSE: Current evidence indicates that the common AMPD1 gene variant is associated with improved survival in patients with advanced heart failure. Whilst adenosine has been recognized to mediate the cardioprotective effect of C34T AMPD1, the precise pathophysiologic mechanism involved remains undefined to date. To address this issue, we used cardio-pulmonary exercise testing data (CPX) from subjects with myoadenylate deaminase (MAD) defects. METHODS: From 2009 to 2013, all the patients referred in our laboratory to perform a metabolic exercise testing, i.e. a CPX with measurements of muscle metabolites in plasma during and after exercise testing, were prospectively enrolled. Subjects that also underwent an open muscle biopsy for diagnosis purpose were finally included. The metabolic-chronotropic response was assessed by calculating the slope of the linear relationship between the percent heart rate reserve and the percent metabolic reserve throughout exercise. MAD activity was measured using the Fishbein's technique in muscle biopsy sample. The common AMPD1 mutation was genotyped and the AMPD1 gene was sequenced to screen rare variants from blood DNA. RESULTS: Sixty-seven patients were included in the study; 5 had complete MAD deficiency, 11 had partial MAD deficiency, and 51 had normal MAD activity. Compared with normal MAD activity subjects, MAD deficient subjects appeared to have a lower-than-expected metabolic-chronotopic response during exercise. The metabolic-chronotropic relationship is more closely correlated with MAD activity in skeletal muscle (Rs = 0.57, p = 5.93E-7, Spearman correlation) than the presence of the common AMPD1 gene variant (Rs = 0.34, p = 0.005). Age-predicted O2 pulse ratio is significantly increased in MAD deficient subjects, indicating a greater efficiency of the cardiovascular system to deliver O2 (p < 0.01, Scheffé's post hoc test). CONCLUSION: The metabolic-chronotropic response is decreased in skeletal muscle MAD deficiency, suggesting a biological mechanism by which AMPD1 gene exerts cardiac effect.


Assuntos
AMP Desaminase/deficiência , Mutação , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , AMP Desaminase/genética , AMP Desaminase/metabolismo , Adolescente , Adulto , Biópsia , Teste de Esforço , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo
11.
Am J Med Genet A ; 173(10): 2736-2742, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28742244

RESUMO

Phosphoribosylpyrophosphate synthetase (PRPPS) superactivity (OMIM 300661) is a rare inborn error of purine metabolism that is caused by gain-of-function mutations in the X-chromosomal gene PRPS1 (Xq22.3). Clinical characteristics include congenital hyperuricemia and hyperuricosuria, gouty arthritis, urolithiasis, developmental delay, hypotonia, recurrent infections, short stature, and hearing loss. Only eight families with PRPPS superactivity and PRPS1 gain-of-function mutations have been reported to date. We report on a 7-year-old boy with congenital hyperuricemia, urolithiasis, developmental delay, short stature, hypospadias, and facial dysmorphisms. His mother also suffered from hyperuricemia that was diagnosed at age 13 years. A novel PRPS1 missense mutation (c.573G>C, p.[Leu191Phe]) was detected in the proband and his mother. Enzyme activity analysis confirmed superactivity of PRPP synthetase. Analysis of the crystal structure of human PRPPS suggests that the Leu191Phe mutation affects the architecture of both allosteric sites, thereby preventing the allosteric inhibition of the enzyme. The family reported here broadens the clinical spectrum of PRPPS superactivity and indicates that this rare metabolic disorder might be associated with a recognizable facial gestalt.


Assuntos
Face/anormalidades , Mutação com Ganho de Função , Hiperuricemia/congênito , Hiperuricemia/genética , Ribose-Fosfato Pirofosfoquinase/genética , Criança , Face/patologia , Humanos , Hiperuricemia/patologia , Masculino , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Ribose-Fosfato Pirofosfoquinase/metabolismo
12.
Cardiovasc Hematol Agents Med Chem ; 15(1): 23-30, 2017 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-28552060

RESUMO

BACKGROUND: Thiopurine S-methyltransferase (TPMT) enzyme metabolizes thiopurine drugs which are widely used in various disciplines as well as in leukemias. Individual enzyme activity varies depending on the genetic polymorphisms of TPMT gene located at chromosome 6. Up to 14% of population is known to have a decreased enzyme activity, and if treated with standard doses of thiopurines, these individuals are at a high risk of severe Adverse Drug Reactions (ADR) as myelosuppression, gastrointestinal intolerance, pancreatitis and hypersensitivity. However, TPMT-deficient patients can successfully be treated with decreased thiopurine doses if enzyme status is identified by a prior testing. TPMT status identification is a pioneering experience in application of pharmacogenetic testing in clinical settings. 4 TPMT (*2, *3A, *3B, *3C) alleles are known to account for 80-95% of a decreased enzyme activity, and therefore, identifying the presence of these alleles supported by phenotypic measurement of the enzyme activity can reveal patient's TPMT status. Evaluation of the levels of thiopurine metabolites further supports the practice of appropriate dose adjustment by providing the efficient monitoring of drug cytotoxicity. CONCLUSION: We hereby review the thiopurine pharmacogenetics and the methods applied in common practice to evaluate patient's TPMT status.


Assuntos
Hipersensibilidade a Drogas/genética , Mercaptopurina/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Preparações Farmacêuticas/metabolismo , Farmacogenética/métodos , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , Animais , Hipersensibilidade a Drogas/diagnóstico , Hipersensibilidade a Drogas/metabolismo , Frequência do Gene , Técnicas de Genotipagem/métodos , Humanos , Mercaptopurina/análogos & derivados , Polimorfismo de Nucleotídeo Único , Erros Inatos do Metabolismo da Purina-Pirimidina/diagnóstico , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo
13.
J Inherit Metab Dis ; 40(3): 423-431, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28205048

RESUMO

BACKGROUND: Elevated urinary excretion of orotic acid is associated with treatable disorders of the urea cycle and pyrimidine metabolism. Establishing the correct and timely diagnosis in a patient with orotic aciduria is key to effective treatment. Uridine monophosphate synthase is involved in de novo pyrimidine synthesis. Uridine monophosphate synthase deficiency (or hereditary orotic aciduria), due to biallelic mutations in UMPS, is a rare condition presenting with megaloblastic anemia in the first months of life. If not treated with the pyrimidine precursor uridine, neutropenia, failure to thrive, growth retardation, developmental delay, and intellectual disability may ensue. METHODS AND RESULTS: We identified mild and isolated orotic aciduria in 11 unrelated individuals with diverse clinical signs and symptoms, the most common denominator being intellectual disability/developmental delay. Of note, none had blood count abnormalities, relevant hyperammonemia or altered plasma amino acid profile. All individuals were found to have heterozygous alterations in UMPS. Four of these variants were predicted to be null alleles with complete loss of function. The remaining variants were missense changes and predicted to be damaging to the normal encoded protein. Interestingly, family screening revealed heterozygous UMPS variants in combination with mild orotic aciduria in 19 clinically asymptomatic family members. CONCLUSIONS: We therefore conclude that heterozygous UMPS-mutations can lead to mild and isolated orotic aciduria without clinical consequence. Partial UMPS-deficiency should be included in the differential diagnosis of mild orotic aciduria. The discovery of heterozygotes manifesting clinical symptoms such as hypotonia and developmental delay are likely due to ascertainment bias.


Assuntos
Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Orotato Fosforribosiltransferase/deficiência , Orotidina-5'-Fosfato Descarboxilase/deficiência , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Anemia Megaloblástica/genética , Anemia Megaloblástica/metabolismo , Criança , Pré-Escolar , Feminino , Heterozigoto , Humanos , Lactente , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Masculino , Mutação/genética , Orotato Fosforribosiltransferase/genética , Orotato Fosforribosiltransferase/metabolismo , Ácido Orótico/metabolismo , Orotidina-5'-Fosfato Descarboxilase/genética , Orotidina-5'-Fosfato Descarboxilase/metabolismo , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , Pirimidinas/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/genética , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Uridina/metabolismo
14.
Nucleosides Nucleotides Nucleic Acids ; 35(10-12): 495-501, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27906626

RESUMO

In June, 2015, the Purine and Pyrimidine Society organized the 16th biennial symposium on Purine and Pyrimidine metabolism at the Faculty House of Columbia University, New York City. This exciting meeting focused on these important molecules, new developments in inborn errors of metabolism; therapeutic analogs. In addition, the biochemistry of mammalian and non-mammalian systems were discussed. Due to significant advances in molecular medicine, the boundaries between clinical and basic sciences have merged into exciting translational research, of which a small portion was highlighted in the presymposium.


Assuntos
Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Purinas/metabolismo , Pirimidinas/metabolismo , Animais , Humanos , Erros Inatos do Metabolismo da Purina-Pirimidina/tratamento farmacológico
15.
Nucleosides Nucleotides Nucleic Acids ; 35(10-12): 578-594, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27906631

RESUMO

Carefully balanced deoxynucleoside triphosphate (dNTP) pools are essential for both nuclear and mitochondrial genome replication and repair. Two synthetic pathways operate in cells to produce dNTPs, e.g., the de novo and the salvage pathways. The key regulatory enzymes for de novo synthesis are ribonucleotide reductase (RNR) and thymidylate synthase (TS), and this process is considered to be cytosolic. The salvage pathway operates both in the cytosol (TK1 and dCK) and the mitochondria (TK2 and dGK). Mitochondrial dNTP pools are separated from the cytosolic ones owing to the double membrane structure of the mitochondria, and are formed by the salvage enzymes TK2 and dGK together with NMPKs and NDPK in postmitotic tissues, while in proliferating cells the mitochondrial dNTPs are mainly imported from the cytosol produced by the cytosolic pathways. Imbalanced mitochondrial dNTP pools lead to mtDNA depletion and/or deletions resulting in serious mitochondrial diseases. The mtDNA depletion syndrome is caused by deficiencies not only in enzymes in dNTP synthesis (TK2, dGK, p53R2, and TP) and mtDNA replication (mtDNA polymerase and twinkle helicase), but also in enzymes in other metabolic pathways such as SUCLA2 and SUCLG1, ABAT and MPV17. Basic questions are why defects in these enzymes affect dNTP synthesis and how important is mitochondrial nucleotide synthesis in the whole cell/organism perspective? This review will focus on recent studies on purine and pyrimidine metabolism, which have revealed several important links that connect mitochondrial nucleotide metabolism with amino acids, glucose, and fatty acid metabolism.


Assuntos
Nucleotídeos de Purina/biossíntese , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Nucleotídeos de Pirimidina/biossíntese , Animais , Vias Biossintéticas , Replicação do DNA , DNA Mitocondrial/biossíntese , DNA Mitocondrial/genética , Humanos , Mitocôndrias/metabolismo , Estresse Oxidativo , Erros Inatos do Metabolismo da Purina-Pirimidina/tratamento farmacológico
16.
Clin Chem Lab Med ; 54(4): 627-32, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26466166

RESUMO

BACKGROUND: Purine nucleoside phosphorylase (PNP) deficiency has been recently introduced in the newborn screening program in Tuscany. In order to improve the PNP screening efficiency, we developed a 2nd tier test to quantify PNP primary markers deoxyguanosine (dGuo) and deoxyinosine (dIno). METHODS: Dried blood spots (DBS) samples were extracted with 200 µL of methanol and 100 µL of water (by two steps). Internal standards were added at a final concentration of 10 µmol/L. After extraction, samples were analysed by LC-MS/MS. The chromatographic run was performed in gradient mode by using a Synergi Fusion column. RESULTS: The assay was linear over a concentration range of 0.05-50 µmol/L (R2>0.999) for dGuo and 0.5-50 µmol/L (R2>0.998) for dIno. Intra- and interassay imprecision (mean CVs) for dIno and dGuo ranged from 2.9% to 12%. Limit of quantitaion (LOQ) were found to be 0.05 µmol/L and 0.5 µmol/L for dGuo and dIno, respectively. The reference ranges, obtained by measuring dGuo and dIno concentrations on DBS, were close to zero for both biomarkers. Moreover, DBS samples from seven patients with confirmed PNP were retrospectively evaluated and correctly identified. CONCLUSIONS: The LC-MS/MS method can reliably measure dIno and dGuo in DBS for the diagnosis of PNP. Validation data confirm the present method is characterised by good reproducibility, accuracy and imprecision for the quantitation of dIno and dGuo. The assay also appears suitable for use in monitoring treatment of PNP patients.


Assuntos
Teste em Amostras de Sangue Seco , Triagem Neonatal , Purina-Núcleosídeo Fosforilase/deficiência , Erros Inatos do Metabolismo da Purina-Pirimidina/sangue , Adulto , Cromatografia Líquida , Humanos , Recém-Nascido , Doenças da Imunodeficiência Primária , Purina-Núcleosídeo Fosforilase/sangue , Purina-Núcleosídeo Fosforilase/metabolismo , Erros Inatos do Metabolismo da Purina-Pirimidina/diagnóstico , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Espectrometria de Massas em Tandem
17.
PLoS One ; 10(7): e0132972, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26207760

RESUMO

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.


Assuntos
AMP Desaminase/deficiência , Algoritmos , Teste de Esforço , Doença de Depósito de Glicogênio/diagnóstico , Erros Inatos do Metabolismo da Purina-Pirimidina/diagnóstico , AMP Desaminase/metabolismo , Adolescente , Adulto , Biópsia , Exercício Físico/fisiologia , Feminino , Doença de Depósito de Glicogênio/metabolismo , Doença de Depósito de Glicogênio/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Erros Inatos do Metabolismo da Purina-Pirimidina/patologia , Adulto Jovem
19.
Artigo em Inglês | MEDLINE | ID: mdl-24940665

RESUMO

The biennial 15th symposium on Purine and Pyrimidine metabolism was held in Madrid, June 2013 (PP13). During the meeting, several novel developments on the diagnosis, pathophysiology, and treatment of several inborn errors of purine and pyrimidine metabolism were presented. These ranged from new drugs for gout to enzyme replacement therapies for mitochondrial diseases. A relatively novel aspect in this meeting was the interest in purine and pyrimidine metabolism in nonmammalian systems, such as parasites, mycoplasms, and bacteria. Development of novel analogs for parasite infections, cardiovascular diseases, inflammatory diseases, and cancer were also discussed.


Assuntos
Erros Inatos do Metabolismo da Purina-Pirimidina/tratamento farmacológico , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Purinas/metabolismo , Purinas/uso terapêutico , Pirimidinas/metabolismo , Pirimidinas/uso terapêutico , Distinções e Prêmios , Humanos , Purinas/química , Pirimidinas/química
20.
Artigo em Inglês | MEDLINE | ID: mdl-24798598

RESUMO

Hypouricemia is defined as a serum urate levels less than 2 mg/dL (119 µmol/L). Primary hypouricemia is caused by disorders of purine metabolism and transport. This laboratory finding is sometimes overlooked and, following two genetic defects, should be considered in differential diagnosis of unexplained hypouricemia. Hereditary xanthinuria is autosomal recessive and due to mutations in xanthine oxidase, leading to over-production of xanthine and minimal production of urate. Patients have very low serum urate levels and suffer from elevated levels of xanthine in the urine, leading to xanthine stones, haematuria, and sometimes occult chronic kidney failure. Hypouricemia is the key to diagnosis. Hereditary renal hypouricemia is a new genetic defect of renal transport of uric acid. Two types were distinguished: a) renal hypouricemia type 1, caused by the defects in the SLC22A12 gene coding the human urate transporter 1 (hURAT1) and b) renal hypouricemia type 2, caused by the defects in the SLC2A9 gene, which encodes GLUT9 transporter. This disorder predisposes patients to exercise-induced acute renal failure and/or nephrolithiasis. Diagnosis is based on two markers: hypouricemia (<119 µmol/L) and increased fractional excretion of uric acid (>10%). Over one hundred cases were identified in Japan and and this number is unique worldwide. Several patients were described in Macedonia. We were able to detect four Czech families with hereditary xanthinuria and eight cases of hereditary renal hypouricemia. In conclusion, hereditary xanthinuria and hereditary renal hypouricemia are still unrecognized conditions. Patients with unexplained hypouricemia need detailed purine metabolic investigations.


Assuntos
Predisposição Genética para Doença , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , Purinas/metabolismo , Erros Inatos do Transporte Tubular Renal/genética , Ácido Úrico/metabolismo , Cálculos Urinários/genética , Adolescente , Adulto , Criança , Feminino , Humanos , Masculino , Erros Inatos do Metabolismo da Purina-Pirimidina/metabolismo , Erros Inatos do Transporte Tubular Renal/metabolismo , Cálculos Urinários/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...