Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Orphanet J Rare Dis ; 12(1): 162, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-29025426

RESUMO

Phenylketonuria (PKU) is an autosomal recessive inborn error of phenylalanine metabolism caused by deficiency in the enzyme phenylalanine hydroxylase that converts phenylalanine into tyrosine. If left untreated, PKU results in increased phenylalanine concentrations in blood and brain, which cause severe intellectual disability, epilepsy and behavioural problems. PKU management differs widely across Europe and therefore these guidelines have been developed aiming to optimize and standardize PKU care. Professionals from 10 different European countries developed the guidelines according to the AGREE (Appraisal of Guidelines for Research and Evaluation) method. Literature search, critical appraisal and evidence grading were conducted according to the SIGN (Scottish Intercollegiate Guidelines Network) method. The Delphi-method was used when there was no or little evidence available. External consultants reviewed the guidelines. Using these methods 70 statements were formulated based on the highest quality evidence available. The level of evidence of most recommendations is C or D. Although study designs and patient numbers are sub-optimal, many statements are convincing, important and relevant. In addition, knowledge gaps are identified which require further research in order to direct better care for the future.


Assuntos
Fenilcetonúrias/diagnóstico , Fenilcetonúrias/terapia , Guias de Prática Clínica como Assunto , Europa (Continente) , Humanos
3.
JIMD Rep ; 29: 103-107, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26898294

RESUMO

S-adenosyl methionine, which is formed from methionine, is an essential methyl donor within the central nervous system. Methionine is formed by the enzyme methionine synthase for which 5-methyltetrahydrofolate (5-MTHF) and homocysteine are substrates. Patients with severe methylenetetrahydrofolate reductase (MTHFR) deficiency cannot make 5-MTHF and have extremely low levels in the CSF. As a consequence, methylation reactions in the CNS are compromised, and this is likely to play an important role in the neurological abnormalities that occur in MTHFR deficiency. Although treatment with oral betaine can remethylate homocysteine to methionine in the liver, betaine crosses the blood-brain barrier poorly, and CSF levels of methionine remain low. We report three patients with severe MTHFR deficiency (enzyme activity ≤1% of controls) who had undetectable levels of CSF 5-MTHF at diagnosis and while on treatment with either folic acid or calcium folinate. Only treatment with oral 5-MTHF given as calcium mefolinate at doses of 15-60 mg/kg/day resulted in an increase in CSF 5-MTHF.

4.
Eur J Pediatr ; 173(3): 361-6, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24114256

RESUMO

UNLABELLED: Pyruvate carboxylase (PC) deficiency (OMIM 266150) is an autosomal recessive disorder that usually presents with lactic acidaemia and severe neurological dysfunction, leading to death in infancy. Because the enzyme is involved in gluconeogenesis and anaplerosis of the Krebs cycle, therapeutic strategies have included avoiding fasting and attempts to correct the defect of anaplerosis. Triheptanoin is a triglyceride of C7 fatty acids. The oxidation of odd chain fatty acids leads to the production not only of acetyl-CoA but also of propionyl-CoA, which is an anaplerotic substrate for the Krebs cycle. One infant with PC deficiency has previously been treated with triheptanoin as well as citrate and 2-chloropropionate. We report two further patients with PC deficiency, who were treated with triheptanoin, continuously from 11 and 21 days of age. They were also given citrate, aspartate and dichloroacetate. Triheptanoin did not lead to any clinical or biochemical improvement. The plasma and CSF lactate concentrations remained high with episodes of severe ketoacidosis and lactic acidosis. Both patients had severe hearing loss, roving eye movements, seizures and very limited neurodevelopmental progress; they died at the ages of 7 and 8 months. CONCLUSION: Though triheptanoin did not alter the clinical course in our patients, it was well tolerated. It remains possible that less severely affected patients might benefit from this form of therapy.


Assuntos
Doença da Deficiência de Piruvato Carboxilase/tratamento farmacológico , Triglicerídeos/uso terapêutico , Feminino , Humanos , Recém-Nascido , Resultado do Tratamento
5.
J Inherit Metab Dis ; 32(5): 630, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19642010

RESUMO

Objectives Isolated methylmalonic acidurias (MMAurias) are caused by deficiency of methylmalonyl-CoA mutase or by defects in the synthesis of its cofactor 5'-deoxyadenosylcobalamin. The aim of this study was to evaluate which parameters best predicted the long-term outcome. Methods Standardized questionnaires were sent to 20 European metabolic centres asking for age at diagnosis, birth decade, diagnostic work-up, cobalamin responsiveness, enzymatic subgroup (mut(0), mut(-), cblA, cblB) and different aspects of long-term outcome. Results 273 patients were included. Neonatal onset of the disease was associated with increased mortality rate, high frequency of developmental delay, and severe handicap. Cobalamin non-responsive patients with neonatal onset born in the 1970s and 1980s had a particularly poor outcome. A more favourable outcome was found in patients with late onset of symptoms, especially when cobalamin responsive or classified as mut(-). Prevention of neonatal crises in pre-symptomatically diagnosed newborns was identified as a protective factor concerning handicap. Chronic renal failure manifested earlier in mut(0) patients than in other enzymatic subgroups. Conclusion Outcome in MMAurias is best predicted by the enzymatic subgroup, cobalamin responsiveness, age at onset and birth decade. The prognosis is still unfavourable in patients with neonatal metabolic crises and non-responsiveness to cobalamin, in particular mut(0) patients.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Biomarcadores/análise , Metilmalonil-CoA Mutase/deficiência , Adolescente , Adulto , Idade de Início , Erros Inatos do Metabolismo dos Aminoácidos/epidemiologia , Erros Inatos do Metabolismo dos Aminoácidos/genética , Erros Inatos do Metabolismo dos Aminoácidos/mortalidade , Criança , Pré-Escolar , Cobamidas/deficiência , Estudos de Coortes , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Metilmalonil-CoA Mutase/genética , Avaliação de Resultados em Cuidados de Saúde , Prognóstico , Análise de Sobrevida , Adulto Jovem
6.
J Inherit Metab Dis ; 32(2): 214-7, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19255872

RESUMO

Prolonged fasting in children with disorder of fat oxidation or ketone body synthesis can lead not only to hypoglycaemia but also to the accumulation of toxic metabolites. The length of time such patients can be safely fasted is important information for caregivers. Most children with MCAD deficiency when well can tolerate 'normal' periods without food, but in more severe disorders such as LCHAD deficiency even these may be associated with acute or chronic damage. Guidelines have been published for safe fasting periods in MCAD but not in other conditions. In the absence of such recommendations, a rational approach must be based on an understanding of the normal physiology of fasting in children of different ages and the pathophysiology associated with the child's particular disorder. Intercurrent infections pose a particular risk and may significantly reduce fasting tolerance.


Assuntos
Jejum/fisiologia , Corpos Cetônicos/metabolismo , Erros Inatos do Metabolismo Lipídico/fisiopatologia , Acil-CoA Desidrogenase/deficiência , Acil-CoA Desidrogenase/genética , Acil-CoA Desidrogenase/metabolismo , Acil-CoA Desidrogenase de Cadeia Longa/genética , Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Criança , Gorduras na Dieta/metabolismo , Ácidos Graxos/metabolismo , Humanos , Erros Inatos do Metabolismo Lipídico/metabolismo , Erros Inatos do Metabolismo Lipídico/terapia , Oxirredução
7.
J Inherit Metab Dis ; 32(3): 441-6, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19306072

RESUMO

Developments in mutation analysis have led to significant benefits for patients with inherited metabolic disorders and their families. This is particularly the case where new methodologies have prevented the need for invasive tissue biopsies or have allowed carrier detection or first trimester prenatal testing to be undertaken. Whereas in the past it may have only been possible to identify specific 'common' mutations, the availability of techniques, such as automated sequencing, and novel technologies including mutation scanning techniques, multiplex ligation dependent probe amplification, and array technologies, have vastly improved the diagnostic efficiency of molecular testing.


Assuntos
Análise Mutacional de DNA , Genes/fisiologia , Relações Médico-Paciente , Prática Profissional , Relações Profissional-Família , Encefalopatias Metabólicas Congênitas/diagnóstico , Encefalopatias Metabólicas Congênitas/genética , Criança , Análise Mutacional de DNA/métodos , Análise Mutacional de DNA/psicologia , Família , Feminino , Conhecimentos, Atitudes e Prática em Saúde , Humanos , Lactente , Recém-Nascido , Masculino , Pacientes , Linhagem , Médicos , Diagnóstico Pré-Natal/psicologia
8.
J Inherit Metab Dis ; 32(1): 95-101, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19191006

RESUMO

BACKGROUND: In order to test the feasibility of cord blood screening for inherited metabolic disease, a two-year cohort study of births in six obstetric units from five towns in the north of England was undertaken. These towns have a high prevalence of consanguineous marriages, largely among the immigrant Asian community. The purpose of the study was to determine whether early detection of metabolic disease was possible and whether early intervention would improve prognosis. METHODS: Following parental consent, cord blood samples were collected at birth and analysed for acylcarnitine and amino acid profiles by tandem mass spectrometry in one of two laboratories. One laboratory used butylated derivatives, the other used underivatized samples. The same laboratories performed routine blood spot neonatal screening at 5-7 days of age on these babies. Patients with positive results were investigated and treated by a metabolic paediatrician as soon as possible. RESULTS: 24,983 births were examined. 12,952 samples were analysed as butyl derivatives, 12,031 samples were analysed underivatized. The following disorders were detected: medium-chain acyl-CoA dehydrogenase (MCAD) deficiency (1 case), 3-methylcrotonyl-CoA carboxylase (MCC) deficiency (2 cases), maternal carnitine transporter defect (2 cases), maternal MCC (1 case). The following disorders were diagnosed subsequently but were not detected by the cord blood screening: phenylketonuria (PKU) (1 case), maple syrup urine disease (MSUD) (2 cases), argininosuccinic aciduria (1 case), methylmalonic acidaemia (MMA) (1 case), glutaric aciduria type 2 (1 case), MCAD deficiency (2 cases), 3-hydroxy-3-methylglutaryl-CoA lyase deficiency (1 case). Comprehensive reference data for all analytes by both methods were obtained. CONCLUSIONS: Cord blood testing is of limited value in detecting inherited metabolic disease. The metabolites associated with most disorders examined were not elevated in cord blood. Some maternal disorders, carnitine transporter defect and 3-methlycrotonyl-CoA carboxylase deficiency, are detected. These remain of uncertain clinical significance. Comprehensive reference data have been obtained that will facilitate future interpretation of studies in cord blood.


Assuntos
Aminoácidos/sangue , Análise Química do Sangue/normas , Coleta de Amostras Sanguíneas/métodos , Carnitina/análogos & derivados , Sangue Fetal/química , Aminoácidos/análise , Análise Química do Sangue/métodos , Coleta de Amostras Sanguíneas/normas , Carnitina/análise , Carnitina/sangue , Estudos de Coortes , Eficiência , Reações Falso-Negativas , Doenças Fetais/sangue , Doenças Fetais/diagnóstico , Humanos , Recém-Nascido , Doenças Metabólicas/sangue , Doenças Metabólicas/diagnóstico , Mães , Triagem Neonatal/métodos , Triagem Neonatal/normas , Valores de Referência , Fatores de Tempo
9.
J Inherit Metab Dis ; 31 Suppl 2: S419-23, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18985435

RESUMO

3-Methylglutaconic aciduria (MGA) encompasses a heterogeneous group of disorders, often coinciding with elevated levels of urinary 3-methylglutaric acid. Type I MGA is a disorder of leucine metabolism, while the biological basis for the MGA is unclear for the other types (MGA types II-V). MGA type III (Costeff optic atrophy syndrome, autosomal recessive optic atrophy-3 or optic atrophy plus syndrome, OMIM 258501) is distinguished by early bilateral optic atrophy, later-onset spasticity, extrapyramidal dysfunction, ataxia, and occasional cognitive deficits. It is caused by homozygous mutations in the optic atrophy 3 gene (OPA3). We present a case of a patient with MGA who has infantile-onset optic atrophy, ataxia, extrapyramidal movements and spasticity, but with normal intellect. Sequencing of the patient's DNA revealed a homozygous nonsense mutation c.415C>T (p.Q139X) in exon 2 of transcript 2 of the OPA3 gene, as well as a common silent polymorphism c.231T>C in the same exon. This is the first nonsense mutation found in OPA3. The molecular findings in OPA3 are also reviewed, including mutations in OPA3 that result in autosomal dominant optic atrophy and cataract (ADOAC). The recessive mode of inheritance of MGA type III as a result of the p.Q139X mutation is supported by the carrier status of the unaffected father.


Assuntos
Coreia/diagnóstico , Códon sem Sentido , Erros Inatos do Metabolismo/diagnóstico , Atrofia Óptica/diagnóstico , Proteínas/genética , Paraplegia Espástica Hereditária/diagnóstico , Adolescente , Desenvolvimento do Adolescente , Biomarcadores/urina , Coreia/complicações , Coreia/genética , Coreia/urina , Análise Mutacional de DNA , Feminino , Predisposição Genética para Doença , Glutaratos/urina , Heterozigoto , Homozigoto , Humanos , Erros Inatos do Metabolismo/complicações , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/urina , Atrofia Óptica/complicações , Atrofia Óptica/genética , Atrofia Óptica/urina , Linhagem , Fenótipo , Paraplegia Espástica Hereditária/complicações , Paraplegia Espástica Hereditária/genética , Paraplegia Espástica Hereditária/urina
10.
J Inherit Metab Dis ; 31(3): 361-7, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18563634

RESUMO

The long-term outcome of patients with methylmalonic aciduria (MMA) is still uncertain due to a high frequency of complications such as chronic renal failure and metabolic stroke. The understanding of this disease is hampered by a huge variation in the management of these patients. The major aim of this study was to evaluate the current practice in different European metabolic centres. A standardized questionnaire was sent to 20 metabolic centres asking for standard procedures for confirmation of diagnosis, testing cobalamin responsiveness, dietary treatment, pharmacotherapy, and biochemical and clinical monitoring. Sixteen of 20 metabolic centres (80%) returned questionnaires on 183 patients: 89 of the patients were classified as mut(0), 36 as mut(-), 13 as cblA, 7 as cblB, and 38 as cblA/B. (1) Confirmation of diagnosis: All centres investigate enzyme activity by propionate fixation in fibroblasts; six centres also perform mutation analysis. (2) Cobalamin response: Ten centres follow standardized protocols showing large variations. A reliable exclusion of nonspecific effects has not yet been achieved by these protocols. (3) Long-term treatment: In cobalamin-responsive patients, most centres use hydroxocobalamin (1-14 mg/week i.m. or 5-20 mg/week orally), while two centres use cyanocobalamin. All cobalamin-nonresponsive patients and most cobalamin-responsive patients are supplemented with L: -carnitine (50-100 mg/kg per day). Fourteen centres use intestinal decontamination by antibiotic therapy. Most centres follow D-A-CH (n = 6) or Dewey (n = 4) recommendations for protein requirements. Fourteen centres regularly use precursor-free amino acid supplements. Standardized monitoring protocols are available in seven centres, again showing high variability.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Ácido Metilmalônico/urina , Adolescente , Adulto , Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Criança , Pré-Escolar , Humanos , Hidroxocobalamina/uso terapêutico , Lactente , Recém-Nascido , Vitamina B 12/uso terapêutico
11.
J Inherit Metab Dis ; 31 Suppl 2: S219-22, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18338235

RESUMO

Hyperammonaemia is common in neonates with branched-chain organic acidaemias, primarily due to the inhibition of N-acetylglutamate (NAG) synthetase; NAG is an activator for carbamylphosphate synthetase I, the first enzyme of the urea cycle. N-Carbamylglutamate, a NAG analogue, has been reported to correct hyperammonaemia in neonates with organic acidaemias. It is, however, uncertain how the ammonia concentrations in these neonates would have progressed without the drug. We report a neonate with propionic acidaemia, whose plasma ammonia concentration responded dramatically to N-carbamylglutamate, having previously been over 950 µmol/L for 33 h. Our patient presented with poor feeding, hypoglycaemia, acidosis and hyperammonaemia (1044 µmol/L at 65 h of age). The patient was treated with intravenous glucose (12 mg/kg per min), insulin, sodium benzoate, sodium phenylbutyrate, carnitine and continuous veno-venous haemofiltration (CVVH). In spite of these measures, the plasma ammonia concentration remained above 950 µmol/L. After 30 h of CVVH, N-carbamylglutamate (250 mg/kg) was given through a nasogastric tube. Over the following 4 h, the plasma ammonia fell from 1410 µmol/L to 267 µmol/L. Despite stopping CVVH, the ammonia level dropped to 137 µmol/L over the next 2 h and it continued to fall while the intravenous drug doses were reduced. The patient was readmitted, aged 4 weeks, with hyperammonaemia (347 µmol/L) and again this responded to N-carbamylglutamate. In contrast, we report a previous patient with propionic acidaemia who showed no response to a lower dose of N-carbamylglutamate (25 mg/kg).


Assuntos
Amônia/sangue , Glutamatos/administração & dosagem , Hiperamonemia/tratamento farmacológico , Acidemia Propiônica/complicações , Biomarcadores/sangue , Feminino , Humanos , Hiperamonemia/sangue , Hiperamonemia/etiologia , Recém-Nascido , Intubação Gastrointestinal , Masculino , Acidemia Propiônica/sangue , Acidemia Propiônica/terapia , Fatores de Tempo , Resultado do Tratamento
12.
J Inherit Metab Dis ; 31 Suppl 3: 493-6, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18181028

RESUMO

Familial selective vitamin B12 (cobalamin, Cbl) malabsorption (Imerslund-Gräsbeck syndrome, IGS, OMIM 261100) is a group of autosomal recessive disorders characterized by selective malabsorption of Cbl from the terminal ileum in the presence of normal histology. Mutations in the amnionless (AMN) and cubilin (CUBN) genes are known to be causes of IGS. Their gene products combine to form a receptor complex (cubam), which is instrumental in the binding and transport of Cbl in the gut. As opposed to Cbl transport in the terminal ileum, normal transport of Cbl into the CNS is poorly understood and little is known regarding its molecular basis. Studies in adults with neuropsychiatric disease have suggested the presence of an active transport mechanism into the central nervous system constituting a blood-brain barrier (BBB) for Cbl. A child with IGS, compound heterozygous for a missense and a nonsense mutation in the amnionless (AMN) protein gene, was noted to have a high daily cobalamin (Cbl) requirement for neuropsychiatric, but not for systemic metabolic and haematological, remission. Measurements of CSF Cbl revealed evidence that the transport of Cbl into the central nervous system was impaired, and a standard Schilling test was consistent with a dose response of cobalamin transport across the terminal ileum. Amnionless protein is known to be expressed in the fetal and postnatal central nervous system, and is known to be involved in Cbl transport in other tissues such as kidney as well as the gut. It is possible that an active Cbl transport mechanism at the BBB exists, and that the amnionless (AMN) protein may be part of this mechanism, as it is in cobalamin transport in the terminal ileum.

13.
Dev Med Child Neurol ; 48(9): 756-60, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16904023

RESUMO

Pyruvate dehydrogenase (PDH) deficiency is a major cause of neurological dysfunction and lactic acidosis in infancy and early childhood. The great majority of cases (>80%) result from mutations in the X-linked gene for the E1alpha subunit of the complex (PDHA1). Mutations in the genes for the other subunits have all been described, but only dihydrolipoamide dehydrogenase (E3) and E3 binding protein (E3BP) defects contribute significantly to the total number of patients with PDH deficiency. Although previously considered rare, with only 13 reported cases, we have found that mutations in PDX1, the gene for the E3 binding protein, are in fact relatively common. Clinical, biochemical, and genetic features of six new patients (four males, two females; age range 15mo-6y) with mutations in this gene are compared with previously reported cases. All patients with E3BP deficiency identified to date have mutations which completely prevent synthesis of the protein product. However, they are generally less severely affected than patients with PDHA1 mutations, although there is considerable overlap in clinical and neuroradiological features.


Assuntos
Doença da Deficiência do Complexo de Piruvato Desidrogenase/genética , Complexo Piruvato Desidrogenase/genética , Criança , Pré-Escolar , Análise Mutacional de DNA , Feminino , Humanos , Lactente , Masculino , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Complexo Piruvato Desidrogenase/metabolismo , Doença da Deficiência do Complexo de Piruvato Desidrogenase/diagnóstico , Doença da Deficiência do Complexo de Piruvato Desidrogenase/metabolismo , Índice de Gravidade de Doença
14.
J Inherit Metab Dis ; 29(5): 627-30, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16865412

RESUMO

Carnitine transporter defect (CTD) is an autosomal recessive disorder characterized by episodes of non-ketotic hypoglycaemia, hyperammonaemia and liver disease, or by the development of cardiomyopathy, both of which occur in infancy and childhood. Blood carnitine concentrations are extremely low. The diagnosis can be confirmed by finding abnormal fat oxidation and carnitine uptake in skin fibroblasts. The condition has not previously been thought to present later in life or to be benign. We report the identification of four women discovered to have CTD as a consequence of finding low carnitine concentrations in the cord blood or newborn samples from their infants. All four mothers had been asymptomatic and none had a cardiomyopathy.


Assuntos
Carnitina/análogos & derivados , Carnitina/metabolismo , Erros Inatos do Metabolismo/diagnóstico , Erros Inatos do Metabolismo/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Adulto , Carnitina/sangue , Consanguinidade , Feminino , Sangue Fetal/metabolismo , Humanos , Recém-Nascido , Masculino , Espectrometria de Massas , Mães , Proteínas de Transporte de Cátions Orgânicos/fisiologia , Oxigênio/metabolismo , Membro 5 da Família 22 de Carreadores de Soluto
15.
J Inherit Metab Dis ; 29(2-3): 261-74, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16763886

RESUMO

In view of the major improvements in treatment, it has become increasingly important that in order for first-line physicians not to miss a treatable disorder they should be able initiate a simple method of clinical screening, particularly in the emergency room. We present a simplified classification of treatable inborn errors of metabolism in three groups. Group 1 includes inborn errors of intermediary metabolism that give rise to an acute or chronic intoxication. It encompasses aminoacidopathies, organic acidurias, urea cycle disorders, sugar intolerances, metal disorders and porphyrias. Clinical expression can be acute or systemic or can involve a specific organ, and can strike in the neonatal period or later and intermittently from infancy to late adulthood. Most of these disorders are treatable and require the emergency removal of the toxin by special diets, extracorporeal procedures, cleansing drugs or vitamins. Group 2 includes inborn errors of intermediary metabolism that affect the cytoplasmic and mitochondrial energetic processes. Cytoplasmic defects encompass those affecting glycolysis, glycogenosis, gluconeogenesis, hyperinsulinisms, and creatine and pentose phosphate pathways; the latter are untreatable. Mitochondrial defects include respiratory chain disorders, and Krebs cycle and pyruvate oxidation defects, mostly untreatable, and disorders of fatty acid oxidation and ketone bodies that are treatable. Group 3 involves cellular organelles and includes lysosomal, peroxisomal, glycosylation, and cholesterol synthesis defects. Among these, some lysosomal disorders can be efficiently treated by enzyme replacement or substrate reduction therapies. Physicians can be faced with the possibility of a treatable inborn error in an emergency, either in the neonatal period or late in infancy to adulthood, or as chronic and progressive symptoms--general (failure to thrive), neurological, or specific for various organs or systems. These symptoms are summarized in four tables. In addition, an extensive list of medications used in the treatment of inborn errors is presented.


Assuntos
Erros Inatos do Metabolismo/classificação , Erros Inatos do Metabolismo/terapia , Humanos , Recém-Nascido , Redes e Vias Metabólicas/genética , Erros Inatos do Metabolismo/diagnóstico , Erros Inatos do Metabolismo/genética , Triagem Neonatal
16.
Arch Dis Child ; 90(2): 143-6, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15665165

RESUMO

BACKGROUND: The effects of maternal phenylalanine on the fetus include facial dysmorphism, microcephaly, intrauterine growth retardation, developmental delay, and congenital heart disease. AIMS: To evaluate the impact of phenylalanine restricted diet in pregnant women with phenylketonuria (PKU) on their offspring. METHODS: Data on virtually all pregnancies of women with PKU in the United Kingdom between 1978 and 1997 were reported to the United Kingdom PKU Registry. The effect of the use and timing in relation to pregnancy of a phenylalanine restricted diet on birth weight, birth head circumference, the presence or absence of congenital heart disease (CHD), 4 year developmental quotient, and 8 year intelligence quotient were examined. RESULTS: A total of 228 pregnancies resulted in live births (seven twin pregnancies were excluded). In 110 (50%), diet started before conception. For this group mean (SD) birth weight was 3160 (612) g, birth head circumference 33.6 (1.9) cm, 4 year DQ 108.9 (13.2), 8 year IQ 103.4 (15.6), and incidence of CHD was 2.4%. In comparison, for those born where treatment was started during pregnancy (n = 91), birth weight was 2818 (711) g, birth head circumference 32.7 (2.0) cm, 4 year DQ 96.8 (15.0), 8 year IQ 86.5 (13.0), and incidence of CHD was 17%. Month-by-month regression analyses suggested that metabolic control by 12-16 weeks gestation had most influence on outcome. CONCLUSIONS: Many features of the maternal PKU syndrome are preventable by starting a phenylalanine restricted diet. Women with PKU and their carers must be aware of the risks and should start the diet before conception, or as soon after as possible.


Assuntos
Fenilalanina/administração & dosagem , Fenilcetonúria Materna/dietoterapia , Análise de Variância , Peso ao Nascer , Criança , Pré-Escolar , Transtornos Cognitivos/etiologia , Deficiências do Desenvolvimento/etiologia , Feminino , Cabeça/anatomia & histologia , Cardiopatias Congênitas/etiologia , Humanos , Recém-Nascido , Inteligência , Fenilalanina/sangue , Fenilcetonúria Materna/sangue , Gravidez , Sistema de Registros , Fatores de Tempo , Reino Unido
17.
Dev Med Child Neurol ; 46(10): 710-2, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15473177

RESUMO

Two individuals with pyruvate dehydrogenase (PDH) deficiency due to missense mutations in the gene for the E1alpha subunit (PDHA1) presented during childhood with dystonia. The first patient, a male, presented at age 4 years with dystonia affecting the lower limbs, which responded to treatment with combined carbidopa and levodopa. The second patient, a female, was first investigated at age 6 years because of a dystonic gait disorder. In both patients, the main clue to the biochemical diagnosis was a raised concentration of lactate in the cerebrospinal fluid. PDH activity was significantly reduced in cultured fibroblasts in both cases. Dystonia is a previously unrecognized major manifestation of PDH deficiency and is of particular interest as the mutations in the PDHA1 gene in these patients have both been identified previously in individuals with typical presentations of the condition.


Assuntos
Distúrbios Distônicos/diagnóstico , Distúrbios Distônicos/etiologia , Doença da Deficiência do Complexo de Piruvato Desidrogenase/complicações , Doença da Deficiência do Complexo de Piruvato Desidrogenase/diagnóstico , Criança , Pré-Escolar , Distúrbios Distônicos/genética , Feminino , Humanos , Masculino , Mutação de Sentido Incorreto , Piruvato Desidrogenase (Lipoamida)/genética , Doença da Deficiência do Complexo de Piruvato Desidrogenase/genética
18.
J Inherit Metab Dis ; 27(4): 539-40, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15334736

RESUMO

There are few reports of liver transplantation in glycogenosis type Ib (GSD Ib). We present two cases who had dramatic catch-up growth and reduced infections after transplantation, despite persistent neutropenia.


Assuntos
Doença de Depósito de Glicogênio Tipo I/cirurgia , Transplante de Fígado , Antiporters/deficiência , Doença de Depósito de Glicogênio Tipo I/enzimologia , Humanos , Lactente , Fígado/enzimologia , Masculino , Proteínas de Transporte de Monossacarídeos/deficiência , Neutropenia/etiologia , Neutropenia/terapia
19.
Acta Paediatr ; 93(4): 552-3, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15188986

RESUMO

UNLABELLED: Undiluted goats milk should not be given to infants. High protein infant feeds are a forgotten cause of metabolic acidosis. CONCLUSION: The metabolic abnormalities associated with goat's milk ingestion can cause a clinical picture very similar to tyrosinaemia type 1.


Assuntos
Acidose/induzido quimicamente , Alimentos Infantis , Leite/efeitos adversos , Tirosinemias/diagnóstico , Acidose/diagnóstico , Acidose/terapia , Animais , Diagnóstico Diferencial , Cabras , Humanos , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido , Testes de Função Hepática , Masculino , Resultado do Tratamento , Tirosinemias/sangue
20.
J Inherit Metab Dis ; 26(2-3): 181-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12889659

RESUMO

A questionnaire was posted on the electronic mailing-list Metab-1 to determine current practice as regards the use of oral L-carnitine in medium-chain acyl-CoA dehydrogenase (MCAD) deficiency, propionic acidaemia (PA) and methylmalonic acidaemia (MMA). Thirty-one centres replied: L-carnitine was used routinely by 94% of respondents in PA and MMA but by only 36% in MCAD deficiency. A search was made for published papers on the use of L-carnitine in organic acidaemias and in MCAD deficiency. The quality of evidence to support the use of L-carnitine was graded according to the scale published by the Scottish Intercollegiate Guideline Network. No high-quality evidence was identified.


Assuntos
Acil-CoA Desidrogenase/deficiência , Carnitina/uso terapêutico , Erros Inatos do Metabolismo/tratamento farmacológico , Ácido Metilmalônico/sangue , Propionatos/sangue , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA