Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Mol Genet Metab ; 120(4): 370-377, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28189603

RESUMEN

BACKGROUND: Long-chain fatty acid oxidation disorders (LC-FAOD) lead to accumulation of high concentrations of potentially toxic fatty acid intermediates. Newborn screening and early intervention have reduced mortality, but most patients continue to experience frequent hospitalizations and significant morbidity despite treatment. The deficient energy state can cause serious liver, muscle, and heart disease, and may be associated with an increased risk of sudden death. Triheptanoin is a medium odd-chain fatty acid. Anaplerotic metabolites of triheptanoin have the potential to replace deficient tricarboxylic acid (TCA) cycle intermediates, resulting in net glucose production as a novel energy source for the treatment of LC-FAOD. STUDY DESIGN: A single-arm, open-label, multicenter Phase 2 safety and efficacy study evaluated patients with severe LC-FAOD evidenced by ongoing related musculoskeletal, cardiac, and/or hepatic events despite treatment. After a four-week run-in on current regimen, investigational triheptanoin (UX007) was titrated to a target dose of 25-35% of total daily caloric intake. Patients were evaluated on several age/condition-eligible endpoints, including submaximal exercise tests to assess muscle function/endurance (12-minute walk test; 12MWT) and exercise tolerance (cycle ergometry), and health related quality of life (HR-QoL). Results through 24weeks of treatment are presented; total study duration is 78weeks. RESULTS: Twenty-nine patients (0.8 to 58years) were enrolled; most qualified based on severe musculoskeletal disease. Twenty-five patients (86%) completed the 24-week treatment period. At Week 18, eligible patients (n=8) demonstrated a 28% increase (LS mean=+181.9 meters; p=0.087) from baseline (673.4meters) in 12MWT distance. At Week 24, eligible patients (n=7) showed a 60% increase in watts generated (LS mean=+409.3W; p=0.149) over baseline (744.6W) for the exercise tolerance test. Improvements in exercise tests were supported by significant improvements from baseline in the adult (n=5) self-reported SF-12v2 physical component summary score (LS mean=+8.9; p<0.001). No difference from baseline was seen in pediatric parent-reported (n=5) scores (SF-10) at Week 24. Eighteen patients (62%) had treatment-related adverse events, predominantly gastrointestinal (55%), mild-to-moderate in severity, similar to that seen with prior treatment with medium chain triglyceride (MCT) oil. One patient experienced a treatment-related serious adverse event of gastroenteritis. One patient discontinued from study due to diarrhea of moderate severity; the majority of patients (25/29; 86%) elected to continue treatment in the extension period. CONCLUSIONS: In patients with severe LC-FAOD, UX007 interim study results demonstrated improved exercise endurance and tolerance, and were associated with positive changes in self-reported HR-QoL.


Asunto(s)
Ácidos Grasos/toxicidad , Errores Innatos del Metabolismo Lipídico/tratamiento farmacológico , Resistencia Física/efectos de los fármacos , Triglicéridos/administración & dosificación , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Errores Innatos del Metabolismo Lipídico/metabolismo , Errores Innatos del Metabolismo Lipídico/fisiopatología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Calidad de Vida , Resultado del Tratamiento , Triglicéridos/farmacología , Prueba de Paso , Adulto Joven
2.
Mol Genet Metab ; 119(3): 223-231, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27590926

RESUMEN

Long-chain fatty acid oxidation disorders (LC-FAOD) can cause cardiac hypertrophy and cardiomyopathy, often presenting in infancy, typically leading to death or heart transplant despite ongoing treatment. Previous data on triheptanoin treatment of cardiomyopathy in LC-FAOD suggested a clinical benefit on heart function during acute failure. An additional series of LC-FAOD patients with critical emergencies associated with cardiomyopathy was treated with triheptanoin under emergency treatment or compassionate use protocols. Case reports from 10 patients (8 infants) with moderate or severe cardiomyopathy associated with LC-FAOD are summarized. The majority of these patients were detected by newborn screening, with follow up confirmatory testing, including mutation analysis; all patients were managed with standard treatment, including medium chain triglyceride (MCT) oil. While on this regimen, they presented with acute heart failure requiring hospitalization and cardiac support (ventilation, ECMO, vasopressors) and, in some cases, resuscitation. The patients discontinued MCT oil and began treatment with triheptanoin, an investigational drug. Triheptanoin is expected to provide anaplerotic metabolites, to replace deficient TCA cycle intermediates and improve effective energy metabolism. Cardiac function was measured by echocardiography and ejection fraction (EF) was assessed. EF was moderately to severely impaired prior to triheptanoin treatment, ranging from 12-45%. Improvements in EF began between 2 and 21days following initiation of triheptanoin, and peaked at 33-71%, with 9 of 10 patients achieving EF in the normal range. Continued treatment was associated with longer-term stabilization of clinical signs of cardiomyopathy. The most common adverse event observed was gastrointestinal distress. Of the 10 patients, 7 have continued on treatment, 1 elected to discontinue due to tolerability issues, and 2 patients died from other causes. Two of the case histories illustrate that cardiomyopathy may also develop later in childhood and/or persist into adulthood. Overall, the presented cases suggest a therapeutic effect of triheptanoin in the management of acute cardiomyopathy associated with LC-FAOD.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Errores Innatos del Metabolismo Lipídico/tratamiento farmacológico , Triglicéridos/administración & dosificación , Adolescente , Cardiomiopatías/complicaciones , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Niño , Preescolar , Ensayos Clínicos como Asunto , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos/metabolismo , Femenino , Humanos , Lactante , Recién Nacido , Errores Innatos del Metabolismo Lipídico/complicaciones , Errores Innatos del Metabolismo Lipídico/patología , Masculino , Oxidación-Reducción/efectos de los fármacos , Triglicéridos/efectos adversos
3.
Mol Genet Metab ; 119(1-2): 1-7, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26822703

RESUMEN

Phenylalanine hydroxylase deficient phenylketonuria (PKU) is the paradigm for a treatable inborn error of metabolism where maintaining plasma phenylalanine (Phe) in the therapeutic range relates to improved clinical outcomes. While Phe is the presumed intoxicating analyte causal in neurologic damage, the mechanism(s) of Phe toxicity has remained elusive. Altered DNA methylation is a recognized response associated with exposure to numerous small molecule toxic agents. Paralleling this effect, we hypothesized that chronic Phe over-exposure in the brain would lead to aberrant DNA methylation with secondary influence upon gene regulation that would ultimately contribute to PKU neuropathology. The PAH(enu2) mouse models human PKU with intrinsic hyperphenylalaninemia, abnormal response to Phe challenge, and neurologic deficit. To examine this hypothesis, we assessed DNA methylation patterns in brain tissues using methylated DNA immunoprecipitation and paired end sequencing in adult PAH(enu2) animals maintained under either continuous dietary Phe restriction or chronic hyperphenylalaninemia. Heterozygous PAH(enu2/WT) litter mates served as controls for normal Phe exposure. Extensive repatterning of DNA methylation was observed in brain tissue of hyperphenylalaninemic animals while Phe restricted animals displayed an attenuated pattern of aberrant DNA methylation. Affected gene coding regions displayed aberrant hypermethylation and hypomethylation. Gene body methylation of noncoding RNA genes was observed and among these microRNA genes were prominent. Of particular note, observed only in hyperphenylalaninemic animals, was hypomethylation of miRNA genes within the imprinted Dlk1-Dio3 locus on chromosome 12. Aberrant methylation of microRNA genes influenced their expression which has secondary effects upon the expression of targeted protein coding genes. Differential hypermethylation of gene promoters was exclusive to hyperphenylalaninemic PAH(enu2) animals. Genes with synaptic involvement were targets of promoter hypermethylation that resulted in down-regulation of their expression. Gene dysregulation secondary to abnormal DNA methylation may be contributing to PKU neuropathology. These results suggest drugs that prevent or correct aberrant DNA methylation may offer a novel therapeutic option to management of neurological symptoms in PKU patients.


Asunto(s)
Metilación de ADN/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Yoduro Peroxidasa/genética , Fenilalanina Hidroxilasa/genética , Fenilcetonurias/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Unión al Calcio , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Impresión Genómica , Humanos , Hígado/metabolismo , Hígado/patología , Ratones , Fenilalanina/genética , Fenilalanina/metabolismo , Fenilalanina Hidroxilasa/deficiencia , Regiones Promotoras Genéticas , ARN no Traducido/genética
4.
Clin Pharmacol Ther ; 99(2): 198-207, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26536838

RESUMEN

High throughput molecular and functional profiling of patients is a key driver of precision medicine. DNA and RNA characterization has been enabled at unprecedented cost and scale through rapid, disruptive progress in sequencing technology, but challenges persist in data management and interpretation. We analyze the state-of-the-art of large-scale unbiased sequencing in drug discovery and development, including technology, application, ethical, regulatory, policy and commercial considerations, and discuss issues of LUS implementation in clinical and regulatory practice.


Asunto(s)
Bases de Datos Factuales/tendencias , Descubrimiento de Drogas/tendencias , Farmacogenética/tendencias , Bases de Datos Factuales/legislación & jurisprudencia , Bases de Datos Factuales/normas , Atención a la Salud/tendencias , Descubrimiento de Drogas/legislación & jurisprudencia , Descubrimiento de Drogas/normas , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Farmacogenética/legislación & jurisprudencia , Farmacogenética/normas , Medicina de Precisión , Estados Unidos , United States Food and Drug Administration
5.
Cell Death Dis ; 4: e670, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23764851

RESUMEN

The c-Myc (Myc) oncoprotein regulates numerous phenotypes pertaining to cell mass, survival and metabolism. Glycolysis, oxidative phosphorylation (OXPHOS) and mitochondrial biogenesis are positively controlled by Myc, with myc-/- rat fibroblasts displaying atrophic mitochondria, structural and functional defects in electron transport chain (ETC) components, compromised OXPHOS and ATP depletion. However, while Myc influences mitochondrial structure and function, it is not clear to what extent the reverse is true. To test this, we induced a state of mitochondrial hyper-fission in rat fibroblasts by de-regulating Drp1, a dynamin-like GTPase that participates in the terminal fission process. The mitochondria from these cells showed reduced mass and interconnectivity, a paucity of cristae, a marked reduction in OXPHOS and structural and functional defects in ETC Complexes I and V. High rates of abortive mitochondrial fusion were observed, likely reflecting ongoing, but ultimately futile, attempts to normalize mitochondrial mass. Cellular consequences included reduction of cell volume, ATP depletion and activation of AMP-dependent protein kinase. In response to Myc deregulation, apoptosis was significantly impaired both in the absence and presence of serum, although this could be reversed by increasing ATP levels by pharmacologic means. The current work demonstrates that enforced mitochondrial fission closely recapitulates a state of Myc deficiency and that mitochondrial integrity and function can affect Myc-regulated cellular behaviors. The low intracellular ATP levels that are frequently seen in some tumors as a result of inadequate vascular perfusion could favor tumor survival by countering the pro-apoptotic tendencies of Myc overexpression.


Asunto(s)
Dinaminas/fisiología , Dinámicas Mitocondriales , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Adenosina Trifosfato/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Animales , Apoptosis , Línea Celular , Proliferación Celular , Supervivencia Celular , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Humanos , Fosforilación Oxidativa , Fenotipo , Proteínas Proto-Oncogénicas c-myc/genética , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptores de Estrógenos/biosíntesis , Receptores de Estrógenos/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Ribonucleótidos/fisiología
6.
Mol Genet Metab ; 107(3): 308-14, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22958974

RESUMEN

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.


Asunto(s)
Amoníaco/orina , Glutamina/análogos & derivados , Glicerol/análogos & derivados , Fenilacetatos/orina , Fenilbutiratos/orina , Trastornos Innatos del Ciclo de la Urea/tratamiento farmacológico , Trastornos Innatos del Ciclo de la Urea/orina , Adolescente , Adulto , Amoníaco/sangre , Biomarcadores Farmacológicos/sangre , Biomarcadores Farmacológicos/orina , Niño , Estudios Cruzados , Esquema de Medicación , Femenino , Glutamina/sangre , Glutamina/orina , Glicerol/sangre , Glicerol/farmacocinética , Glicerol/orina , Humanos , Masculino , Fenilacetatos/sangre , Fenilbutiratos/sangre , Fenilbutiratos/farmacocinética , Trastornos Innatos del Ciclo de la Urea/sangre
7.
Pediatr Transplant ; 15(1): 58-64, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20946191

RESUMEN

MSUD is a complex metabolic disorder that has been associated with central nervous system damage, developmental delays, and neurocognitive deficits. Although liver transplantation provides a metabolic cure for MSUD, changes in cognitive and adaptive functioning following transplantation have not been investigated. In this report, we present data from 14 patients who completed cognitive and adaptive functioning testing pre- and one yr and/or three yr post-liver transplantation. Findings show either no significant change (n=8) or improvement (n=5) in IQ scores pre- to post-liver transplantation. Greater variability was observed in adaptive functioning scores, but the majority of patients evidenced no significant change (n=8) in adaptive scores. In general, findings indicate that liver transplantation minimizes the likelihood of additional central nervous system damage, providing an opportunity for possible stabilization or improvement in neurocognitive functioning.


Asunto(s)
Trasplante de Hígado/métodos , Enfermedad de la Orina de Jarabe de Arce/complicaciones , Enfermedad de la Orina de Jarabe de Arce/terapia , Adaptación Psicológica , Adolescente , Niño , Preescolar , Cognición , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/etiología , Femenino , Humanos , Pruebas de Inteligencia , Masculino , Pruebas Neuropsicológicas , Reproducibilidad de los Resultados , Resultado del Tratamiento
8.
Biotech Histochem ; 86(4): 226-31, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20297946

RESUMEN

Magnetic force combined with magnetic nanoparticles recently has shown potential for enhancing nucleic acid delivery. Achieving effective siRNA delivery into primary cultured cells is challenging. We compared the utility of magnetofection with lipofection procedures for siRNA delivery to primary and immortalized mammalian fibroblasts. Transfection efficiency and cell viability were analyzed by flow cytometry and effects of gene knockdown were quantified by real-time PCR. Lipofectamine 2000 and magnetofection achieved high transfection efficiencies comparable to similar gene silencing effects of about 80%; the cytotoxic effect of magnetofection, however, was significantly less. Magnetofection is a reliable and gentle alternative method with low cytotoxicity for siRNA delivery into difficult to transfect cells such as mammalian fibroblasts. These features are especially advantageous for functional end point analyses of gene silencing, e.g., on the metabolite level.


Asunto(s)
Nanopartículas de Magnetita/administración & dosificación , ARN Mensajero/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transfección/métodos , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Supervivencia Celular , Fibroblastos , Silenciador del Gen , Lípidos/administración & dosificación , Lípidos/química , Ratones , Interferencia de ARN/fisiología
9.
J Inherit Metab Dis ; 31 Suppl 2: S453-6, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19089597

RESUMEN

We report a 16-month-old asymptomatic male with enzyme confirmed isovaleric acidaemia (IVA; isovaleryl-CoA dehydrogenase deficiency; OMIM 243500) who, upon routine nutritional follow-up, presented evidence of peroxisomal dysfunction. The newborn screen (2 days of life) revealed elevated C(5)-carnitine (2.95 µmol/L; cutoff <0.09 µmol/L) and IVA was subsequently confirmed by metabolic profiling and in vitro enzymology. Plasma essential fatty acid (EFA) analysis, assessed to evaluate nutritional status during protein restriction and L: -carnitine supplementation, revealed elevated C(26:0) (5.0 µmol/L; normal <1.3). Subsequently, metabolic profiling and molecular genetic analysis confirmed X-linked adrenoleukodystrophy (XALD). Identification of co-inherited XALD with IVA in this currently asymptomatic patient holds significant treatment ramifications for the proband prior to the onset of neurological sequelae, and critically important counselling implications for this family.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Ácidos Grasos Esenciales/sangre , Evaluación Nutricional , Trastorno Peroxisomal/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/sangre , Errores Innatos del Metabolismo de los Aminoácidos/complicaciones , Errores Innatos del Metabolismo de los Aminoácidos/genética , Biomarcadores/sangre , Análisis Mutacional de ADN , Humanos , Lactante , Recién Nacido , Isovaleril-CoA Deshidrogenasa/sangre , Isovaleril-CoA Deshidrogenasa/deficiencia , Isovaleril-CoA Deshidrogenasa/genética , Masculino , Tamizaje Neonatal , Trastorno Peroxisomal/sangre , Trastorno Peroxisomal/complicaciones , Trastorno Peroxisomal/genética , Valor Predictivo de las Pruebas
10.
J Inherit Metab Dis ; 31(6): 761-8, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18937046

RESUMEN

Dimethylglycine dehydrogenase (DMGDH) is a mitochondrial matrix flavoprotein that catalyses the demethylation of dimethylglycine to form sarcosine, accompanied by the reduction of the covalently bound FAD cofactor. Electron-transfer flavoprotein reoxidizes the reduced flavin and transfers reducing equivalents to the main mitochondrial respiratory chain through the enzyme ETF-ubiquinone oxidoreductase. DMGDH plays a prominent role in choline and 1-carbon metabolism. We have expressed the mature form of human DMGDH and the H109R variant identified in a DMGDH-deficient patient as N-terminally His(6)-tagged proteins in E. coli. The enzymes were purified to homogeneity by nickel affinity and anion exchange chromatography. The presence of FAD in the wild-type enzyme was confirmed by spectrophotometric analysis. The H109R variant, however, had only 47% of the wild-type level of bound flavin as expressed in E. coli, indicating its reduced affinity for FAD As previously described for rat enzyme studies, the wild-type human enzyme exhibited two K (m) values for N,N-dimethylglycine (K (m1) = 0.039 +/- 0.010 mmol/L and K(m2) = 15.4 +/- 1.2 mmol/L). The addition of 4 micromol/L tetrahydrofolate resulted in a slight decrease in specific activity and a substantial decrease in K (m2) (1.10 +/- 0.55 mmol/L). The flavinated H109R variant protein exhibited a 27-fold decrease in specific activity and a 65-fold increase in K (m), explaining its pathogenicity. Additionally, the current expression system represents a significant improvement over a previously described rat DMGDH expression system and will enhance our ability to further study this important metabolic enzyme.


Asunto(s)
Dimetilglicina-Deshidrogenasa/deficiencia , Dimetilglicina-Deshidrogenasa/genética , Errores Innatos del Metabolismo/genética , Cromatografía por Intercambio Iónico/métodos , Dimetilglicina-Deshidrogenasa/química , Relación Dosis-Respuesta a Droga , Electrones , Flavina-Adenina Dinucleótido/metabolismo , Flavoproteínas/metabolismo , Humanos , Cinética , Errores Innatos del Metabolismo/diagnóstico , Modelos Químicos , Mutación , Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Sarcosina-Deshidrogenasa/metabolismo , Espectrofotometría/métodos
11.
Am J Hum Genet ; 81(1): 87-103, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17564966

RESUMEN

The acyl-CoA dehydrogenases are a family of multimeric flavoenzymes that catalyze the alpha,beta -dehydrogenation of acyl-CoA esters in fatty acid beta -oxidation and amino acid catabolism. Genetic defects have been identified in most of the acyl-CoA dehydrogenases in humans. Acyl-CoA dehydrogenase 9 (ACAD9) is a recently identified acyl-CoA dehydrogenase that demonstrates maximum activity with unsaturated long-chain acyl-CoAs. We now report three cases of ACAD9 deficiency. Patient 1 was a 14-year-old, previously healthy boy who died of a Reye-like episode and cerebellar stroke triggered by a mild viral illness and ingestion of aspirin. Patient 2 was a 10-year-old girl who first presented at age 4 mo with recurrent episodes of acute liver dysfunction and hypoglycemia, with otherwise minor illnesses. Patient 3 was a 4.5-year-old girl who died of cardiomyopathy and whose sibling also died of cardiomyopathy at age 21 mo. Mild chronic neurologic dysfunction was reported in all three patients. Defects in ACAD9 mRNA were identified in the first two patients, and all patients manifested marked defects in ACAD9 protein. Despite a significant overlap of substrate specificity, it appears that ACAD9 and very-long-chain acyl-CoA dehydrogenase are unable to compensate for each other in patients with either deficiency. Studies of the tissue distribution and gene regulation of ACAD9 and very-long-chain acyl-CoA dehydrogenase identify the presence of two independently regulated functional pathways for long-chain fat metabolism, indicating that these two enzymes are likely to be involved in different physiological functions.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga/genética , Ácidos Grasos/metabolismo , Errores Innatos del Metabolismo Lipídico/genética , Enfermedades Mitocondriales/genética , Acil-CoA Deshidrogenasa de Cadena Larga/análisis , Acil-CoA Deshidrogenasa de Cadena Larga/química , Acil-CoA Deshidrogenasa de Cadena Larga/aislamiento & purificación , Adolescente , Secuencia de Bases , Encéfalo/enzimología , Niño , Análisis Mutacional de ADN , Femenino , Regulación de la Expresión Génica , Genoma Humano , Humanos , Masculino , Datos de Secuencia Molecular , Músculo Esquelético/enzimología , Regiones Promotoras Genéticas , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Especificidad por Sustrato , Distribución Tisular
12.
Am J Med Genet A ; 143A(9): 985-94, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17394213

RESUMEN

Simultaneous occurrence of two unrelated cytogenetic events is rare. We present a case of Angelman Syndrome (AS) deletion and 12q duplication in a child with a history of developmental delay, microcephaly, cerebral palsy, and seizures. Traditional cytogenetic studies showed a normal 46,XY karyotype. Fluorescence in situ hybridization (FISH) using probe D15S10 (AS region/15q11.2) revealed a deletion. In addition, we serendipitously detected 12q24.3 duplication by FISH with 12q subtelomere probe. He inherited this duplication from the mother who presented with a balanced translocation karyotype 46,XX,add(12)(q24.3).ish t(12;13)(q24.3;p11.2)(12qtel-;12qtel+,D13Z1/D21Z1+,RB1+). Array comparative genomic hybridization (array-CGH) revealed a duplication of three bacterial artificial chromosome (BAC) clones (RP11-46H11, RP11-386I8, and RP11-309H3) covering about 423 Kb of DNA sequence. The published 12q terminal duplication cases had a detectable segment by classical banded cytogenetics techniques. To our knowledge, this is the smallest 12q cryptic rearrangement characterized by array-CGH and confirmed by BAC-clone FISH analysis. Based on these findings, we attempted to separate the clinical features associated with AS deletion and those features that are probably due to partial 12q duplication. We then reviewed the genes mapped in the duplicated region using the human genome database to understand the clinical significance. A subsequent pregnancy in the mother revealed an apparently balanced t(12;13) karyotype. We compare our case with the published cases, and discuss the implications of our findings and its relevance in addressing genetic counseling issues.


Asunto(s)
Síndrome de Angelman/genética , Cromosomas Humanos Par 12 , Duplicación de Gen , Preescolar , Bandeo Cromosómico , Humanos , Hibridación Fluorescente in Situ , Masculino
13.
J Inherit Metab Dis ; 26(1): 37-42, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12872838

RESUMEN

We report identification of short-chain acyl-CoA dehydrogenase (SCAD) deficiency in a 12-year-old boy who suffered from recurrent attacks of vomiting once or twice a year from infancy. Growth and development were normal and there were no muscular symptoms. Metabolic screening was performed during a hospitalization at 8 years of age and revealed an increased excretion of ethylmalonic acid (EMA; 45-80 mmol/mol creatinine, normal 0.2-6.6), suggesting a degradation defect of short-chain fatty acids. An increased n-butyrylcarnitine was found in freshly collected serum (0.9 micromol/L; normal <0.4) but not in dry blood spots. Neither of the frequent SCAD gene variants 625G>A and 511C>T was present, but direct sequencing of the promoter and coding regions of the SCAD gene revealed that the patient had mutations on both alleles: 417G>C (Trpl15Cys) and 1095G>T (Gln341His). Neither mutation has been described before in compound heterozygosity or homozygosity. Enzymatic investigations subsequently confirmed a defect of SCAD in both fibroblasts and muscle extracts. Furthermore, expression studies of both mutations demonstrated impaired enzyme function or structure. To our knowledge, this case is the first description of a patient with proven SCAD deficiency presenting with recurrent emesis but without other symptoms, and emphasizes the wide clinical phenotype of this disorder.


Asunto(s)
Butiril-CoA Deshidrogenasa/genética , Malonatos/orina , Mutación/genética , Vómitos/etiología , Vómitos/genética , Alelos , Células Cultivadas , ADN Complementario/genética , Fibroblastos , Humanos , Recién Nacido , Masculino , Músculo Esquelético/enzimología , Mutación/fisiología , Oxidación-Reducción , Fenotipo , Recurrencia
14.
Mol Genet Metab ; 74(1-2): 226-37, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11592819

RESUMEN

Isovaleryl-CoA dehydrogenase (IVD) is a homotetrameric flavoenzyme, which catalyzes the conversion of isovaleryl-CoA to 3-methylcrotonyl-CoA and transfers electrons to the electron-transferring flavoprotein, and is a member of the acyl-CoA dehydrogenase (ACD) enzyme family. Human IVD crystal structure with a bound substrate analogue shows the guanidino group of Arg387, a conserved residue among other members of the ACD enzyme family, juxtaposed to a phosphate oxygen of the 4'-phosphopantothiene moiety of the substrate analogue. Site-directed mutagenesis was used to investigate the role of Arg387 in substrate binding and enzyme function. Replacing this residue with Lys, Ala, Gln, or Glu resulted in stable proteins. Spectrophotometric substrate binding assays indicated that the Arg387Lys mutant was able to form the charge-transfer complex intermediate with similar efficiency to wild type, while the rest of the mutants were significantly less able to properly form this intermediate. However, the Km of the isovaleryl-CoA for the Arg387Lys mutant was 20.3 compared to 1.5 microM for the wild type. The Km for the rest of the mutants were 75.6, 195, and 550 microM, respectively. The catalytic efficiency per mole of FAD was 20.3, 3.3, 2.0, and 0.34 for the mutants, respectively, compared to 260 microM(-1) x min(-1) for the wild type. These results substantiate the important role of Arg387 in anchoring the substrate, and are consistent with the hypothesis that residues distant from the active site are important for stabilizing the enzyme:substrate/product complex, and could play an important role in the mechanism of the enzyme-catalyzed reaction.


Asunto(s)
Acilcoenzima A/metabolismo , Arginina/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Oxidorreductasas/fisiología , Especificidad por Sustrato/fisiología , Sustitución de Aminoácidos/genética , Arginina/genética , Sitios de Unión/genética , Transferencia de Energía/genética , Estabilidad de Enzimas/genética , Humanos , Isovaleril-CoA Deshidrogenasa , Lisina/genética , Modelos Moleculares , Mutación , Oxidorreductasas/biosíntesis , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Unión Proteica/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Espectrometría de Fluorescencia , Especificidad por Sustrato/genética
15.
Hum Mol Genet ; 10(19): 2069-77, 2001 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11590124

RESUMEN

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.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Carnitina/análogos & derivados , Errores Innatos del Metabolismo Lipídico/enzimología , Acil-CoA Deshidrogenasa de Cadena Larga/metabolismo , Animales , Carnitina/metabolismo , Ácidos Grasos/metabolismo , Femenino , Humanos , Errores Innatos del Metabolismo Lipídico/patología , Tamaño de la Camada , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Cardíacas/enzimología , Mitocondrias Hepáticas/enzimología , Mitocondrias Musculares/enzimología
16.
Cancer ; 92(2): 395-405, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11466695

RESUMEN

BACKGROUND: The characterization of differentially expressed genes between cancerous and normal tissues is an important step in the understanding of tumorigenesis. Global gene expression profiling with microarrays has now offered a powerful tool to measure the changes of thousands of genes in any carcinoma tissues in an effort to identify these key disease-related genes. To compare the gene expression of a primary liver carcinoma, metastatic carcinoma to the liver, and normal liver, the authors analyzed tissue from six primary hepatocellular carcinomas (HCCs), five colorectal adenocarcinoma metastases to the liver, and eight normal livers. METHODS: Samples were processed from total RNA to fragmented cRNA and hybridized onto Affymetrix GeneChip(R) expression arrays. Analyses were performed to determine the consensus pattern of gene expression for primary liver carcinoma, metastatic liver carcinoma, and normal liver tissue and their changes in expression level. RESULTS: In hepatocellular carcinoma, 842 genes were overexpressed, and 393 genes were underexpressed in comparison with genes of normal liver tissue. Of note, 7 of the 20 most increased identified known genes previously have been associated with liver carcinoma or other types of cancers. The 13 additional identified genes until now have not previously shown strong association with cancers. Furthermore, the authors identified 42 genes and 24 expressed sequence tags that are expressed at a significant level in both HCC and metastastic tumors, presenting a list of marker genes indicative of cancerous liver tissue. CONCLUSIONS: In this study, genes that can be involved in the production of and maintenance of hepatic carcinomas were identified. These data offer new insight into genes that are potentially important in the pathogenesis of liver carcinoma, as well as additional targets for new strategies for cancer therapy and treatment.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Colorrectales/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Adulto , Anciano , Transformación Celular Neoplásica , Femenino , Humanos , Hígado/fisiología , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos
17.
Mol Genet Metab ; 73(2): 126-37, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11386848

RESUMEN

Isovaleryl-CoA dehydrogenase (IVD) is a flavoenzyme, which catalyzes the conversion of isovaleryl-CoA to 3-methylcrotonyl-CoA in the leucine catabolism pathway and transfers electrons to the electron-transferring flavoprotein (ETF). IVDs from human and rat have been identified and characterized previously. In this study, the gene coding for Caenorhabditis elegans IVD has been identified from a published cDNA sequence and molecular modeling has been performed using the human IVD atomic coordinates. The coding sequence for the mature form of the enzyme was expressed in Escherichia coli, and the recombinant nematode IVD enzyme was purified to essential homogeneity. Its spectrum is typical of recombinant FAD-containing acyl-CoA dehydrogenases and shows a minor broad absorption band at 650-700 nm characteristic of an IVD:CoA persulfide charge-transfer complex. Following treatment of the enzyme with sodium dithionite to remove the bound CoA persulfide, the K(m) values for isovaleryl-, butyryl-, valeryl-, and hexanoyl-CoA were estimated to be 2.5, 36.2, 10.5, and 33.8 microM, respectively, using the ETF fluorescence reduction assay. The catalytic efficiency (k(cat)/K(m)) for these substrates was 56.9, 1.3, 13.7, and 3.2 microM(-1). min(-1) per mole of FAD, respectively. The apparent binding constant (K(D app)) of the recombinant IVD determined spectrally for isovaleryl-CoA was 0.34 microM. These kinetic parameters confirm that isovaleryl-CoA is the preferred substrate for the purified enzyme. The variability in the protein structure among known and putative IVDs from various species is discussed in the context of possible mechanisms for modulating enzyme activity.


Asunto(s)
Caenorhabditis elegans/genética , Ácido Graso Desaturasas/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Oxidorreductasas/genética , Acilcoenzima A/metabolismo , Acil-CoA Deshidrogenasa , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/enzimología , ADN Complementario/química , ADN Complementario/genética , Isovaleril-CoA Deshidrogenasa , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Estructura Terciaria de Proteína , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Espectrofotometría , Especificidad por Sustrato
18.
Gene ; 270(1-2): 253-7, 2001 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-11404023

RESUMEN

Isovaleryl-CoA dehydrogenase (IVD) is an intramitochondrial homotetrameric flavoenzyme that catalyzes the conversion of isovaleryl-CoA to 3-methylcrotonyl-CoA in the leucine catabolism pathway. Deficiency of IVD in humans causes isovaleric acidemia, which shows tremendous clinical variability for reasons that are unknown. To help better understand this disorder, we have cloned and sequenced the mouse IVD genomic and cDNAs. The mouse IVD gene spans approximately 17 kb and contains 12 coding exons organized identically to the human gene. It maps to mouse chromosome 2 in the area of band 2E4-E5, corresponding to the syntenic region of human chromosome 15. Mouse IVD predicted amino acid sequences are 95.8 and 89.6% identical to that of the rat and human sequences, respectively, with conservation of key functional residues. We have now identified IVD sequences from seven species. Comparison of these sequences shows that the rat and mouse proteins are the most closely related, both of which, in turn, share highest homology to human. All of the mammalian enzymes appear to be more closely related than any of the IVDs on other branches of the phylogram, while the fly and worm IVDs are the most divergent. The invertebrate IVDs are more closely related to the mammalian enzymes than to those from two plant species.


Asunto(s)
ADN Complementario/genética , ADN/genética , Evolución Molecular , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Oxidorreductasas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , ADN/química , ADN Complementario/química , Exones , Genes/genética , Intrones , Isovaleril-CoA Deshidrogenasa , Ratones , Datos de Secuencia Molecular , Filogenia , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido
19.
J Pediatr ; 138(4): 585-8, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11295727

RESUMEN

There is a correlation between pregnancy complications such as acute fatty liver of pregnancy and long-chain 3-hydroxyacyl-coenzyme A dehydrogenase (LCHAD) deficiency. We diagnosed another fatty acid beta-oxidation defect, short-chain acyl-coenzyme A dehydrogenase deficiency, in an infant when evaluating him because his mother had acute fatty liver of pregnancy. Other beta-oxidation defects, in addition to LCHAD deficiency, should be considered in children born after pregnancies complicated by liver disease.


Asunto(s)
Ácido Graso Desaturasas/deficiencia , Hígado Graso/complicaciones , Complicaciones del Embarazo , Adulto , Ácido Graso Desaturasas/genética , Hígado Graso/diagnóstico , Femenino , Edad Gestacional , Humanos , Recién Nacido , Masculino , Embarazo , Complicaciones del Embarazo/diagnóstico
20.
Biochem Biophys Res Commun ; 282(1): 297-305, 2001 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-11264007

RESUMEN

Cofactor associations within the electron transferring flavoprotein (ETF) were studied in real time using microelectrospray ionization-mass spectrometry (muESI-MS). Initial analysis of porcine (pETF) and human ETF (hETF) revealed only the holoprotein. When muESI-MS source energies were increased, both pETF and hETF readily lost AMP. Analysis of hETF and pETF in methanol revealed intact alpha- and beta-subunits, and beta-subunit with AMP. The pETF also contained beta-subunit with FAD and beta-subunit with both cofactors. In contrast to crystal structure predictions, AMP dissociates more readily than FAD, and the pETF beta-subunit has an intimate association with FAD. This work demonstrates the complementarity of muESI-MS with NMR X-ray and optical spectroscopy in the analysis of noncovalent complexes.


Asunto(s)
Flavoproteínas/química , Espectrometría de Masa por Ionización de Electrospray/métodos , Animales , Flavoproteínas Transportadoras de Electrones , Flavoproteínas/metabolismo , Humanos , Hígado/química , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA