Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Mol Genet Metab ; 117(1): 27-32, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26586473

RESUMO

UNLABELLED: Blood ammonia and glutamine levels are used as biomarkers of control in patients with urea cycle disorders (UCDs). This study was undertaken to evaluate glutamine variability and utility as a predictor of hyperammonemic crises (HACs) in UCD patients. METHODS: The relationships between glutamine and ammonia levels and the incidence and timing of HACs were evaluated in over 100 adult and pediatric UCD patients who participated in clinical trials of glycerol phenylbutyrate. RESULTS: The median (range) intra-subject 24-hour coefficient of variation for glutamine was 15% (8-29%) as compared with 56% (28%-154%) for ammonia, and the correlation coefficient between glutamine and concurrent ammonia levels varied from 0.17 to 0.29. Patients with baseline (fasting) glutamine values >900 µmol/L had higher baseline ammonia levels (mean [SD]: 39.6 [26.2]µmol/L) than patients with baseline glutamine ≤ 900 µmol/L (26.6 [18.0]µmol/L). Glutamine values >900 µmol/L during the study were associated with an approximately 2-fold higher HAC risk (odds ratio [OR]=1.98; p=0.173). However, glutamine lost predictive significance (OR=1.47; p=0.439) when concomitant ammonia was taken into account, whereas the predictive value of baseline ammonia ≥ 1.0 upper limit of normal (ULN) was highly statistically significant (OR=4.96; p=0.013). There was no significant effect of glutamine >900 µmol/L on time to first HAC crisis (hazard ratio [HR]=1.14; p=0.813), but there was a significant effect of baseline ammonia ≥ 1.0 ULN (HR=4.62; p=0.0011). CONCLUSIONS: The findings in this UCD population suggest that glutamine is a weaker predictor of HACs than ammonia and that the utility of the predictive value of glutamine will need to take into account concurrent ammonia levels.


Assuntos
Amônia/sangue , Glutamina/sangue , Hiperamonemia/sangue , Distúrbios Congênitos do Ciclo da Ureia/sangue , Adolescente , Adulto , Biomarcadores/sangue , Criança , Pré-Escolar , Jejum , Feminino , Glicerol/análogos & derivados , Glicerol/uso terapêutico , Humanos , Hiperamonemia/etiologia , Masculino , Fenilbutiratos/uso terapêutico , Valor Preditivo dos Testes , Distúrbios Congênitos do Ciclo da Ureia/tratamento farmacológico , Adulto Jovem
2.
Clin Genet ; 86(2): 142-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23889335

RESUMO

Peters plus syndrome (PPS) is a rare autosomal-recessive disorder characterized by Peters anomaly of the eye, short stature, brachydactyly, dysmorphic facial features, developmental delay, and variable other systemic abnormalities. In this report, we describe screening of 64 patients affected with PPS, isolated Peters anomaly and PPS-like phenotypes. Mutations in the coding region of B3GALTL were identified in nine patients; six had a documented phenotype of classic PPS and the remaining three had a clinical diagnosis of PPS with incomplete clinical documentation. A total of nine different pathogenic alleles were identified. Five alleles are novel including one frameshift, c.168dupA, p.(Gly57Argfs*11), one nonsense, c.1234C>T, p.(Arg412*), two missense, c.1045G>A, p.(Asp349Asn) and c.1181G>A, p.(Gly394Glu), and one splicing, c.347+5G>T, mutations. Consistent with previous reports, the c.660+1G>A mutation was the most common mutation identified, seen in eight of the nine patients and accounting for 55% of pathogenic alleles in this study and 69% of all reported pathogenic alleles; while two patients were homozygous for this mutation, the majority had a second rare pathogenic allele. We also report the absence of B3GALTL mutations in 55 cases of PPS-like phenotypes or isolated Peters anomaly, further establishing the strong association of B3GALTL mutations with classic PPS only.


Assuntos
Fenda Labial/genética , Córnea/anormalidades , Galactosiltransferases/genética , Glucosiltransferases/genética , Transtornos do Crescimento/genética , Deformidades Congênitas dos Membros/genética , Mutação/genética , Estudos de Coortes , Feminino , Frequência do Gene/genética , Estudos de Associação Genética , Humanos , Masculino , Fenótipo
3.
Mol Genet Metab ; 110(4): 446-53, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24144944

RESUMO

BACKGROUND: Phenylacetic acid (PAA) is the active moiety in sodium phenylbutyrate (NaPBA) and glycerol phenylbutyrate (GPB, HPN-100). Both are approved for treatment of urea cycle disorders (UCDs) - rare genetic disorders characterized by hyperammonemia. PAA is conjugated with glutamine in the liver to form phenylacetyleglutamine (PAGN), which is excreted in urine. PAA plasma levels ≥ 500 µg/dL have been reported to be associated with reversible neurological adverse events (AEs) in cancer patients receiving PAA intravenously. Therefore, we have investigated the relationship between PAA levels and neurological AEs in patients treated with these PAA pro-drugs as well as approaches to identifying patients most likely to experience high PAA levels. METHODS: The relationship between nervous system AEs, PAA levels and the ratio of plasma PAA to PAGN were examined in 4683 blood samples taken serially from: [1] healthy adults [2], UCD patients of ≥ 2 months of age, and [3] patients with cirrhosis and hepatic encephalopathy (HE). The plasma ratio of PAA to PAGN was analyzed with respect to its utility in identifying patients at risk of high PAA values. RESULTS: Only 0.2% (11) of 4683 samples exceeded 500 µg/ml. There was no relationship between neurological AEs and PAA levels in UCD or HE patients, but transient AEs including headache and nausea that correlated with PAA levels were observed in healthy adults. Irrespective of population, a curvilinear relationship was observed between PAA levels and the plasma PAA:PAGN ratio, and a ratio>2.5 (both in µg/mL) in a random blood draw identified patients at risk for PAA levels>500 µg/ml. CONCLUSIONS: The presence of a relationship between PAA levels and reversible AEs in healthy adults but not in UCD or HE patients may reflect intrinsic differences among the populations and/or metabolic adaptation with continued dosing. The plasma PAA:PAGN ratio is a functional measure of the rate of PAA metabolism and represents a useful dosing biomarker.


Assuntos
Glutamina/análogos & derivados , Encefalopatia Hepática/sangue , Fenilacetatos/sangue , Distúrbios Congênitos do Ciclo da Ureia/sangue , Biomarcadores/sangue , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/sangue , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Glutamina/administração & dosagem , Glutamina/sangue , Glicerol/administração & dosagem , Glicerol/análogos & derivados , Encefalopatia Hepática/etiologia , Encefalopatia Hepática/patologia , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Fenilacetatos/administração & dosagem , Fenilbutiratos/administração & dosagem , Ensaios Clínicos Controlados Aleatórios como Assunto , Distúrbios Congênitos do Ciclo da Ureia/epidemiologia , Distúrbios Congênitos do Ciclo da Ureia/etiologia , Distúrbios Congênitos do Ciclo da Ureia/patologia
4.
J Clin Pharmacol ; 53(7): 699-710, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23775211

RESUMO

Sodium phenylbutyrate and glycerol phenylbutyrate mediate waste nitrogen excretion in the form of urinary phenylacetylglutamine (PAGN) in patients with urea cycle disorders (UCDs); rare genetic disorders characterized by impaired urea synthesis and hyperammonemia. Sodium phenylbutyrate is approved for UCD treatment; the development of glycerol phenylbutyrate afforded the opportunity to characterize the pharmacokinetics (PK) of both compounds. A population PK model was developed using data from four Phase II/III trials that collectively enrolled patients ages 2 months to 72 years. Dose simulations were performed with particular attention to phenylacetic acid (PAA), which has been associated with adverse events in non-UCD populations. The final model described metabolite levels in plasma and urine for both drugs and was characterized by (a) partial presystemic metabolism of phenylbutyric acid (PBA) to PAA and/or PAGN, (b) slower PBA absorption and greater presystemic conversion with glycerol phenylbutyrate, (c) similar systemic disposition with saturable conversion of PAA to PAGN for both drugs, and (d) body surface area (BSA) as a significant covariate accounting for age-related PK differences. Dose simulations demonstrated similar PAA exposure following mole-equivalent PBA dosing of both drugs and greater PAA exposure in younger patients based on BSA.


Assuntos
Glicerol/análogos & derivados , Modelos Biológicos , Fenilbutiratos/administração & dosagem , Fenilbutiratos/farmacocinética , Distúrbios Congênitos do Ciclo da Ureia/tratamento farmacológico , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Adulto , Criança , Pré-Escolar , Ensaios Clínicos Fase II como Assunto , Ensaios Clínicos Fase III como Assunto , Simulação por Computador , Feminino , Glutamina/análogos & derivados , Glutamina/urina , Glicerol/administração & dosagem , Glicerol/farmacocinética , Humanos , Masculino , Nitrogênio/urina , Ensaios Clínicos Controlados Aleatórios como Assunto , Doenças Raras/tratamento farmacológico , Doenças Raras/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/urina
5.
Mol Genet Metab ; 107(3): 308-14, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22958974

RESUMO

UNLABELLED: We have analyzed pharmacokinetic data for glycerol phenylbutyrate (also GT4P or HPN-100) and sodium phenylbutyrate with respect to possible dosing biomarkers in patients with urea cycle disorders (UCD). STUDY DESIGN: These analyses are based on over 3000 urine and plasma data points from 54 adult and 11 pediatric UCD patients (ages 6-17) who participated in three clinical studies comparing ammonia control and pharmacokinetics during steady state treatment with glycerol phenylbutyrate or sodium phenylbutyrate. All patients received phenylbutyric acid equivalent doses of glycerol phenylbutyrate or sodium phenylbutyrate in a cross over fashion and underwent 24-hour blood samples and urine sampling for phenylbutyric acid, phenylacetic acid and phenylacetylglutamine. RESULTS: Patients received phenylbutyric acid equivalent doses of glycerol phenylbutyrate ranging from 1.5 to 31.8 g/day and of sodium phenylbutyrate ranging from 1.3 to 31.7 g/day. Plasma metabolite levels varied widely, with average fluctuation indices ranging from 1979% to 5690% for phenylbutyric acid, 843% to 3931% for phenylacetic acid, and 881% to 1434% for phenylacetylglutamine. Mean percent recovery of phenylbutyric acid as urinary phenylacetylglutamine was 66.4 and 69.0 for pediatric patients and 68.7 and 71.4 for adult patients on glycerol phenylbutyrate and sodium phenylbutyrate, respectively. The correlation with dose was strongest for urinary phenylacetylglutamine excretion, either as morning spot urine (r = 0.730, p < 0.001) or as total 24-hour excretion (r = 0.791 p<0.001), followed by plasma phenylacetylglutamine AUC(24-hour), plasma phenylacetic acid AUC(24-hour) and phenylbutyric acid AUC(24-hour). Plasma phenylacetic acid levels in adult and pediatric patients did not show a consistent relationship with either urinary phenylacetylglutamine or ammonia control. CONCLUSION: The findings are collectively consistent with substantial yet variable pre-systemic (1st pass) conversion of phenylbutyric acid to phenylacetic acid and/or phenylacetylglutamine. The variability of blood metabolite levels during the day, their weaker correlation with dose, the need for multiple blood samples to capture trough and peak, and the inconsistency between phenylacetic acid and urinary phenylacetylglutamine as a marker of waste nitrogen scavenging limit the utility of plasma levels for therapeutic monitoring. By contrast, 24-hour urinary phenylacetylglutamine and morning spot urine phenylacetylglutamine correlate strongly with dose and appear to be clinically useful non-invasive biomarkers for compliance and therapeutic monitoring.


Assuntos
Amônia/urina , Glutamina/análogos & derivados , Glicerol/análogos & derivados , Fenilacetatos/urina , Fenilbutiratos/urina , Distúrbios Congênitos do Ciclo da Ureia/tratamento farmacológico , Distúrbios Congênitos do Ciclo da Ureia/urina , Adolescente , Adulto , Amônia/sangue , Biomarcadores Farmacológicos/sangue , Biomarcadores Farmacológicos/urina , Criança , Estudos Cross-Over , Esquema de Medicação , Feminino , Glutamina/sangue , Glutamina/urina , Glicerol/sangue , Glicerol/farmacocinética , Glicerol/urina , Humanos , Masculino , Fenilacetatos/sangue , Fenilbutiratos/sangue , Fenilbutiratos/farmacocinética , Distúrbios Congênitos do Ciclo da Ureia/sangue
6.
Mol Genet Metab ; 103(4): 323-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21612962

RESUMO

UNLABELLED: Twenty four hour ammonia profiles and correlates of drug effect were examined in a phase 2 comparison of sodium phenylbutyrate (NaPBA) and glycerol phenylbutyrate (GPB or HPN-100), an investigational drug being developed for urea cycle disorders (UCDs). STUDY DESIGN: Protocol HPN-100-005 involved open label fixed-sequence switch-over from the prescribed NaPBA dose to a PBA-equimolar GPB dose with controlled diet. After 7 days on NaPBA or GPB, subjects underwent 24-hour blood sampling for ammonia and drug metabolite levels as well as measurement of 24-hour urinary phenyacetylglutamine (PAGN). Adverse events (AEs), safety labs and triplicate ECGs were monitored. RESULTS: Eleven subjects (9 OTC, 1 ASS, 1 ASL) enrolled and completed the switch-over from NaPBA (mean dose=12.4 g/d or 322 mg/kg/d; range=198-476 mg/kg/d) to GPB (mean dose=10.8 mL or 0.284 mL/kg/d or 313 mg/kg/d; range=192-449 mg/kg/d). Possibly-related AEs were reported in 2 subjects on NaPBA and 4 subjects on GPB. All were mild, except for one moderate AE of vomiting on GPB related to an intercurrent illness. No clinically significant laboratory or ECG changes were observed. Ammonia was lowest after overnight fast, peaked postprandially in the afternoon to early evening and varied widely over 24h with occasional values >100 µmol/L without symptoms. Ammonia values were ~25% lower on GPB vs. NaPBA (p≥0.1 for ITT and p<0.05 for per protocol population). The upper 95% confidence interval for the difference between ammonia on GPB vs. NaPBA in the ITT population (95% CI 0.575, 1.061; p=0.102) was less than the predefined non-inferiority margin of 1.25 and less than 1.0 in the pre-defined per-protocol population (95% CI 0.516, 0.958; p<0.05). No statistically significant differences were observed in plasma phenylacetic acid and PAGN exposure during dosing with GPB vs. NaPBA, and the percentage of orally administered PBA excreted as PAGN (66% for GPB vs. 69% for NaPBA) was very similar. GPB and NaPBA dose correlated best with urinary-PAGN. CONCLUSIONS: These findings suggest that GPB is at least equivalent to NaPBA in terms of ammonia control, has potential utility in pediatric UCD patients and that U-PAGN is a clinically useful biomarker for dose selection and monitoring.


Assuntos
Amônia/sangue , Glicerol/análogos & derivados , Fenilbutiratos/uso terapêutico , Distúrbios Congênitos do Ciclo da Ureia/tratamento farmacológico , Ureia/metabolismo , Adolescente , Criança , Relação Dose-Resposta a Droga , Glicerol/uso terapêutico , Humanos , Masculino , Distúrbios Congênitos do Ciclo da Ureia/metabolismo
7.
J Inherit Metab Dis ; 32(1): 65-72, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18949578

RESUMO

Rabies is a fatal viral encephalitis characterized by a clinically acute and progressive course. With rare exceptions, there is a discrepancy between clinical outcome and frank histological alterations in rabies. Investigators have postulated that rabies virus may modify neurotransmission through occupancy of cellular receptors or alteration of ion channels. We took advantage of these observations to improvise a successful therapy for rabies. The Milwaukee protocol ( www.mcw.edu/rabies ) was further modified to treat two German patients. We measured pterins and monoamine neurotransmitter metabolites in the CSF of patients with rabies by HPLC with electrochemical or fluorescent detection. We report loss of tetrahydrobiopterin (BH(4)) and associated pathological decrease of dopaminergic and serotoninergic neurotransmission in three successive patients with rabies. CSF levels of BH(4) and neurotransmitter metabolites increased in two patients who were supplemented. Our findings support the long-standing speculation of modified neurotransmission in the pathogenesis of rabies, but by another mechanism. Brain turnover of dopamine and serotonin is reduced following rabies-acquired BH(4) deficiency. Neuronal nitric oxide synthase is BH(4)-dependent and may also be involved, possibly causing cerebrovascular insufficiency in one patient. This work must be carefully replicated in animal models and future patients. We are cautiously optimistic at the prospect of readily available, metabolically specific, enteral therapy for rabies.


Assuntos
Biopterinas/análogos & derivados , Fenilcetonúrias/complicações , Raiva/complicações , Adolescente , Adulto , Biopterinas/deficiência , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos dos Movimentos/diagnóstico , Transtornos dos Movimentos/etiologia , Raiva/diagnóstico , Raiva/transmissão , Distúrbios da Fala/diagnóstico , Distúrbios da Fala/etiologia
8.
Mol Genet Metab ; 92(1-2): 109-14, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17689999

RESUMO

A newborn female presented on the first day of life with clinical and biochemical findings consistent with multiple acyl-CoA dehydrogenase deficiency (MADD). Riboflavin supplementation corrected the biochemical abnormalities 24 h after commencing the vitamin. In vitro acylcarnitine profiling in intact fibroblasts both in normal and riboflavin depleted media showed normal oxidation of fatty acids excluding defects in electron transfer flavoprotein (ETF), or ETF ubiquinone oxidoreductase (ETF:QO), or a genetic abnormality in flavin metabolism. In addition, sequencing of the genes encoding ETF and ETF:QO in the proband did not reveal any pathogenic mutations. Determination of the maternal riboflavin status after delivery showed that the mother was riboflavin deficient. Repeat testing done two years after the infant's birth and while on a normal diet showed that the mother was persistently riboflavin deficient and showed a typical MADD profile on plasma acylcarnitine testing. A possible genetic defect in riboflavin transport of metabolism in the mother is postulated to be the cause of the transient MADD seen in the infant. Sequencing of the SLC16A12, RFK and FLAD1 genes encoding key enzymes in riboflavin transport of metabolism in the mother did not identify any pathogenic mutations. The underlying molecular basis of the mother's defect in riboflavin metabolism remains to be established.


Assuntos
Acil-CoA Desidrogenase/deficiência , Erros Inatos do Metabolismo Lipídico/diagnóstico , Deficiência de Riboflavina/genética , Carnitina/análogos & derivados , Carnitina/sangue , Flavoproteínas Transferidoras de Elétrons/genética , Feminino , Fibroblastos/citologia , Fibroblastos/enzimologia , Fibroblastos/patologia , Humanos , Recém-Nascido , Erros Inatos do Metabolismo Lipídico/genética , Desnutrição , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Mães , Oxirredução , Deficiência de Riboflavina/metabolismo , Deficiência de Riboflavina/patologia , Pele/enzimologia , Pele/patologia , Simportadores , Vitaminas/administração & dosagem
9.
J Inherit Metab Dis ; 25(5): 333-46, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12408183

RESUMO

During 1967-1983, the Maternal and Child Health Division of the Public Health Services funded a collaborative study of 211 newborn infants identified on newborn screening as having phenylketonuria (PKU). Subsequently, financial support was provided by the National Institute of Child Health and Human Development (NICHD). The infants were treated with a phenylalanine (Phe)-restricted diet to age 6 years and then randomized either to continue the diet or to discontinue dietary treatment altogether. One hundred and twenty-five of the 211 children were then followed until 10 years of age. In 1998, NICHD scheduled a Consensus Development Conference on Phenylketonuria and initiated a study to follow up the participants from the original Collaborative Study to evaluate their present medical, nutritional, psychological, and socioeconomic status. Fourteen of the original clinics (1967-1983) participated in the Follow-up Study effort. Each clinic director was provided with a list of PKU subjects who had completed the original study (1967-1983), and was asked to evaluate as many as possible using a uniform protocol and data collection forms. In a subset of cases, magnetic resonance imaging and spectroscopy (MRI/MRS) were performed to study brain Phe concentrations. The medical evaluations revealed that the subjects who maintained a phenylalanine-restricted diet reported fewer problems than the diet discontinuers, who had an increased rate of eczema, asthma, mental disorders, headache, hyperactivity and hypoactivity. Psychological data showed that lower intellectual and achievement test scores were associated with dietary discontinuation and with higher childhood and adult blood Phe concentrations. Abnormal MRI results were associated with higher brain Phe concentrations. Early dietary discontinuation for subjects with PKU is associated with poorer outcomes not only in intellectual ability, but also in achievement test scores and increased rates of medical and behavioural problems.


Assuntos
Fenilcetonúrias , Adulto , Química Encefálica , Criança , Continuidade da Assistência ao Paciente , Escolaridade , Seguimentos , Humanos , Imageamento por Ressonância Magnética , Fenilalanina/administração & dosagem , Fenilalanina/análise , Fenilalanina/sangue , Fenilalanina Hidroxilase/genética , Fenilcetonúrias/complicações , Fenilcetonúrias/diagnóstico , Fenilcetonúrias/dietoterapia , Fenilcetonúrias/psicologia , Análise de Regressão , Classe Social , Escalas de Wechsler
10.
Hum Mol Genet ; 10(19): 2069-77, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11590124

RESUMO

Although many patients have been found to have very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency, none have been documented with long-chain acyl-CoA dehydrogenase (LCAD) deficiency. In order to understand the metabolic pathogenesis of long-chain fatty acid oxidation disorders, we generated mice with VLCAD deficiency (VLCAD(-/-)) and compared their pathologic and biochemical phenotypes of mice with LCAD deficiency (LCAD(-/-)) and wild-type mice. VLCAD(-/-) mice had milder fatty change in liver and heart. Dehydrogenation of various acyl-CoA substrates by liver, heart and skeletal muscle mitochondria differed among the three genotypes. The results for liver were most informative as VLCAD(-/-) mice had a reduction in activity toward palmitoyl-CoA and oleoyl-CoA (58 and 64% of wild-type, respectively), whereas LCAD(-/-) mice showed a more profoundly reduced activity toward these substrates (35 and 32% of wild-type, respectively), with a significant reduction of activity toward the branched chain substrate 2,6-dimethylheptanoyl-CoA. C(16) and C(18) acylcarnitines were elevated in bile, blood and serum of fasted VLCAD(-/-) mice, whereas abnormally elevated C(12) and C(14) acylcarnitines were prominent in LCAD(-/-) mice. Progeny with the combined LCAD(+/+)//VLCAD(+/-) genotype were over-represented in offspring from sires and dams heterozygous for both LCAD and VLCAD mutations. In contrast, no live mice with a compound LCAD(-/-)//VLCAD(-/-) genotype were detected.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/deficiência , Carnitina/análogos & derivados , Erros Inatos do Metabolismo Lipídico/enzimologia , Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Animais , Carnitina/metabolismo , Ácidos Graxos/metabolismo , Feminino , Humanos , Erros Inatos do Metabolismo Lipídico/patologia , Tamanho da Ninhada de Vivíparos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Cardíacas/enzimologia , Mitocôndrias Hepáticas/enzimologia , Mitocôndrias Musculares/enzimologia
11.
Pediatr Res ; 49(1): 18-23, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11134486

RESUMO

ABSTRACT Short-chain acyl-CoA dehydrogenase (SCAD) deficiency is considered a rare inherited mitochondrial fatty acid oxidation disorder. Less than 10 patients have been reported, diagnosed on the basis of ethylmalonic aciduria and low SCAD activity in cultured fibroblast. However, mild ethylmalonic aciduria, a biochemical marker of functional SCAD deficiency in vivo, is a common finding in patients suspected of having metabolic disorders. Based on previous observations, we have proposed that ethylmalonic aciduria in a small proportion of cases is caused by pathogenic SCAD gene mutations, and SCAD deficiency can be demonstrated in fibroblasts. Another - much more frequent - group of patients with mild ethylmalonic aciduria has functional SCAD deficiency due to the presence of susceptibility SCAD gene variations, i.e. 625G>A and 511C>T, in whom a variable or moderately reduced SCAD activity in fibroblasts may still be clinically relevant. To substantiate this notion we performed sequence analysis of the SCAD gene in 10 patients with ethylmalonic aciduria and diagnosed with SCAD deficiency in fibroblasts. Surprisingly, only one of the 10 patients carried pathogenic mutations in both alleles, while five were double heterozygotes for a pathogenic mutation in one allele and the 625G>A susceptibility variation in the other. The remaining four patients carried only either the 511C>T or the 625G>A variations in each allele. Our findings document that patients carrying these SCAD gene variations may develop clinically relevant SCAD deficiency, and that patients with even mild ethylmalonic aciduria should be tested for these variations.


Assuntos
Acil-CoA Desidrogenases/genética , Variação Genética , Erros Inatos do Metabolismo Lipídico/genética , Acil-CoA Desidrogenase , DNA Complementar , Feminino , Haplótipos , Humanos , Lactente , Recém-Nascido , Masculino , Mutagênese Sítio-Dirigida , Mutação
13.
Am J Obstet Gynecol ; 180(4): 986-94, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10203668

RESUMO

OBJECTIVES: The aims of this article are to report on a review of cases of maternal phenylketonuria in the International Maternal Phenylketonuria Collaborative Study that were initially diagnosed during or after a pregnancy, to alert health care practitioners to the possible existence of women with undiagnosed phenylketonuria whose fetuses are at risk, and to emphasize that not all adults with untreated phenylketonuria are mentally retarded. STUDY DESIGN: The study was conducted through retrospective database review. RESULTS: Of 414 women with live-born infants, 17 fulfilled our criteria. Six had phenylketonuria diagnosed after they had produced >/=1 affected offspring, 2 had phenylketonuria diagnosed as a result of transient postnatal hyperphenylalaninemia in an offspring, and 9 had phenylketonuria diagnosed by prenatal screening. Undiagnosed maternal phenylketonuria in North America and Europe is currently estimated at 1 case/100,000 births; this rate could be higher elsewhere. CONCLUSIONS: Physicians and midwives should consider a protocol of selective prenatal screening or case finding to detect undiagnosed phenylketonuria among their patients.


Assuntos
Doenças Fetais/diagnóstico , Fenilcetonúria Materna/diagnóstico , Diagnóstico Pré-Natal , Adolescente , Adulto , Feminino , Humanos , Fenilalanina/sangue , Gravidez , Resultado da Gravidez , Estudos Retrospectivos
14.
Neurology ; 52(2): 366-72, 1999 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9932958

RESUMO

OBJECTIVE: To determine an underlying genetic defect within the differential diagnosis of congenital multicore myopathy. BACKGROUND: A 13.5-year-old girl presented with congenital-onset facial and neck weakness, slowly progressive severe limb girdle and axial myopathy, respiratory weakness, cardiomyopathy, progressive joint contractures, lumbar lordosis, progressive external ophthalmoplegia with ptosis, and cataracts. Muscle biopsy at 3 years revealed type I fiber predominance and hypotrophy, multicores with a focal decrease in mitochondria and oxidative enzymes, and internal nuclei. METHODS AND RESULTS: Serum carnitine was decreased (total, 18.2 micromol/L; free, 11.7 micromol/L). Urine organic acids intermittently revealed very large amounts of ethylmalonic and methylsuccinic acids intermittently, with elevated butyrylglycine, 2-methylbutyrylglycine, and tiglylglycine. Fibroblast acylcarnitine profiles revealed marked butyrylcarnitine elevation. Electron-transferring flavoprotein-linked reduction enzymatic assay of fibroblasts with butyryl-coenzyme A (CoA) as substrate, after immunoinactivation of medium-chain acyl-CoA dehydrogenase activity, revealed a complete absence of short-chain acyl-CoA dehydrogenase (SCAD) activity. No SCAD protein was detectable with Western blot analysis. CONCLUSIONS: This patient expands the clinical phenotype of SCAD deficiency and emphasizes the need for its consideration in the differential diagnosis of progressive external ophthalmoplegia and congenital multicore myopathy.


Assuntos
Acil-CoA Desidrogenases/deficiência , Miopatias Mitocondriais/diagnóstico , Oftalmoplegia Externa Progressiva Crônica/diagnóstico , Acil-CoA Desidrogenase , Acil-CoA Desidrogenases/genética , Adolescente , Carnitina/análogos & derivados , Carnitina/sangue , Células Cultivadas , Diagnóstico Diferencial , Feminino , Fibroblastos/enzimologia , Humanos , Miopatias Mitocondriais/enzimologia , Miopatias Mitocondriais/genética , Oftalmoplegia Externa Progressiva Crônica/enzimologia , Oftalmoplegia Externa Progressiva Crônica/genética , Fenótipo
15.
Adv Exp Med Biol ; 466: 395-402, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10709668

RESUMO

The SCAD deficient mouse model has been useful to investigate mechanisms of deficient fatty acid oxidation disease in human patients. This mouse model has been thoroughly characterized and is readily available from the Jackson Laboratory. Using the new technologies of gene-knockout mouse modeling, we envisage developing additional members of the acyl-CoA dehydrogenase family of enzyme deficiencies in mice and furthering our understanding of fatty acid metabolism in health and disease.


Assuntos
Acil-CoA Desidrogenases/deficiência , Acil-CoA Desidrogenases/genética , Erros Inatos do Metabolismo Lipídico/enzimologia , Acil-CoA Desidrogenase , Acil-CoA Desidrogenases/metabolismo , Animais , Modelos Animais de Doenças , Éxons , Humanos , Íntrons , Erros Inatos do Metabolismo Lipídico/genética , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Deleção de Sequência
16.
Proc Natl Acad Sci U S A ; 95(26): 15592-7, 1998 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-9861014

RESUMO

Abnormalities of fatty acid metabolism are recognized to play a significant role in human disease, but the mechanisms remain poorly understood. Long-chain acyl-CoA dehydrogenase (LCAD) catalyzes the initial step in mitochondrial fatty acid oxidation (FAO). We produced a mouse model of LCAD deficiency with severely impaired FAO. Matings between LCAD +/- mice yielded an abnormally low number of LCAD +/- and -/- offspring, indicating frequent gestational loss. LCAD -/- mice that reached birth appeared normal, but had severely reduced fasting tolerance with hepatic and cardiac lipidosis, hypoglycemia, elevated serum free fatty acids, and nonketotic dicarboxylic aciduria. Approximately 10% of adult LCAD -/- males developed cardiomyopathy, and sudden death was observed in 4 of 75 LCAD -/- mice. These results demonstrate the crucial roles of mitochondrial FAO and LCAD in vivo.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/deficiência , Acil-CoA Desidrogenase de Cadeia Longa/genética , Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Animais , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/sangue , Ácidos Graxos não Esterificados/metabolismo , Humanos , Erros Inatos do Metabolismo Lipídico/enzimologia , Erros Inatos do Metabolismo Lipídico/genética , Erros Inatos do Metabolismo Lipídico/patologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/enzimologia , Músculo Esquelético/metabolismo , Especificidade por Substrato
17.
J Pediatr ; 133(5): 697-700, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9821433

RESUMO

We report siblings with a variant of carbohydrate-deficient glycoprotein syndrome, type 1 (CDGS1), characterized by normal phosphomannomutase and phosphomannose isomerase activities, severe thrombocytopenia, and respiratory compromise. Each infant died after a course of intensive care, suggesting that infants with CDGS1 and normal phosphomannomutase and phosphomannose isomerase activities may have a more severe CDGS1 phenotype.


Assuntos
Defeitos Congênitos da Glicosilação/diagnóstico , Doenças do Prematuro/diagnóstico , Manose-6-Fosfato Isomerase/sangue , Fosfotransferases (Fosfomutases)/sangue , Trombocitopenia/diagnóstico , Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/genética , Evolução Fatal , Feminino , Humanos , Recém-Nascido , Doenças do Prematuro/enzimologia , Doenças do Prematuro/genética , Masculino , Gravidez , Diagnóstico Pré-Natal , Trombocitopenia/enzimologia , Trombocitopenia/genética
18.
Pediatr Res ; 44(2): 210-4, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9702916

RESUMO

Acylcarnitines are important diagnostic markers for inborn errors of fatty acid oxidation, but their analysis in body fluids may not always be reliable. Recently, disease-specific acylcarnitine profiles generated by cultured skin fibroblasts were reported to facilitate the diagnosis by localizing a specific enzymatic defect in the mitochondrial beta-oxidation pathway. Using a novel methodologic approach, fibroblasts from 16 patients with inborn errors of fatty acid oxidation and 13 control subjects were preincubated with L-[3H]carnitine to label the intracellular carnitine pool. Cells were subsequently incubated with unlabeled palmitic acid and, after methanol extraction of cells and media, labeled free carnitine and acylcarnitines were analyzed by radio-HPLC. Quantitation was based on the integrated radioactivity of individual peaks relative to the total radioactivity recovered. In control cell lines, all saturated acylcarnitines were detected, and reference values were established. With the exception of one cell line deficient in electron transfer flavoprotein, all mutant cell lines showed abnormal and disease-specific relative concentrations of acylcarnitines. Advantages of the method include use of a small number of cells, no need for trypsinization and permeabilization of cells before incubation, simple extraction without purification of the specimen before HPLC, and relatively inexpensive equipment. The method allows a focused approach to the subsequent, more laborious confirmation of a particular disease by direct enzymatic and/or molecular analysis. It remains to be established whether the method can replace widely used global measurements of fatty acid oxidation rates in vitro that do not provide specific information about the enzyme deficiency involved.


Assuntos
Carnitina/análise , Cromatografia Líquida de Alta Pressão/métodos , Ácidos Graxos/metabolismo , Mitocôndrias/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Pele/química , Acil-CoA Desidrogenase , Acil-CoA Desidrogenase de Cadeia Longa , Carnitina O-Palmitoiltransferase/metabolismo , Células Cultivadas , Ácidos Graxos Dessaturases/metabolismo , Fibroblastos/química , Humanos , Erros Inatos do Metabolismo Lipídico/metabolismo , Mitocôndrias/enzimologia , Oxirredução , Oxirredutases/metabolismo , Ácido Palmítico/metabolismo , Pele/citologia
19.
Pediatr Res ; 42(5): 583-7, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9357927

RESUMO

Primary carnitine deficiency is associated with deficient blood and tissue carnitine concentrations. The clinical syndrome is dominated by heart and skeletal muscle symptoms, and the clinical response to oral carnitine supplementation is life-saving. Carnitine uptake has been shown to be defective in cultured skin fibroblasts and leukocytes obtained from patients with this condition. We report a new case of primary carnitine deficiency and offer direct evidence consistent with an impairment of carnitine uptake in differentiating muscle culture. The patient presented with severe and progressive cardiomyopathy and moderate proximal limb weakness. Plasma and muscle carnitine levels were very low, and the maximal rate of carnitine transport in cultured fibroblasts was deficient. An asymptomatic sister with intermediate levels of carnitine in plasma showed partially deficient carnitine uptake in fibroblasts, indicating heterozygosity. The patient's condition improved dramatically with oral carnitine therapy. Further studies were performed in cultured muscle cells at different stages of maturation, which demonstrated deficient maximal rates of carnitine uptake. Our findings are consistent with the concept that primary carnitine deficiency is the result of a generalized defect involving carnitine transport across tissue membranes.


Assuntos
Carnitina/farmacocinética , Músculo Esquelético/metabolismo , Transporte Biológico/fisiologia , Carnitina/deficiência , Células Cultivadas , Criança , Creatina Quinase/análise , Ácidos Graxos/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Isoenzimas , Oxirredução
20.
Eur J Pediatr ; 156(10): 800-2, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9365073

RESUMO

UNLABELLED: Inspiratory stridor of unknown origin was the leading clinical symptom in an 11-month-old boy. The stridor increased over a period of 4 weeks, and assisted ventilation became necessary. Selective urinary screening by gas chromatography/mass spectrometry analysis revealed excretion of ethylmalonic and 3-OH-isovaleric acid and of N-isobutyryl-, N-2-methylbutyryl-, N-isovaleryl-, N-hexanoyl- and N-suberylglycine. Neither hypoglycaemia nor metabolic acidosis were noticed. Treatment with 200 mg of riboflavin per day led to a dramatic clinical improvement with restoration of normal respiration and an increase in muscular tone within 2 months. During this period, metabolite excretion in urine completely normalized. Riboflavin-sensitive multiple acyl-CoA dehydrogenation deficiency was confirmed in cultured fibroblasts. With riboflavin supplementation, the development of the child has been favourable, with normal school attendance now at an age of 9 years. CONCLUSION: As respiratory symptoms might precede other symptoms in disorders of mitochondrial oxidation, we propose determination of urinary organic acids in all cases of unexplained laryngeal stridor.


Assuntos
Ácidos Graxos Dessaturases/deficiência , Miopatias Mitocondriais/diagnóstico , Hipotonia Muscular/diagnóstico , Sons Respiratórios/etiologia , Riboflavina/uso terapêutico , Adipatos/urina , Criança , Pré-Escolar , Fibroblastos/enzimologia , Seguimentos , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Lactente , Recém-Nascido , Masculino , Malonatos/urina , Miopatias Mitocondriais/enzimologia , Miopatias Mitocondriais/genética , Hipotonia Muscular/enzimologia , Hipotonia Muscular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...