Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Anal Biochem ; 636: 114343, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34637785

RESUMEN

Urea cycle disorders (UCD) are inherited diseases resulting from deficiency in one of six enzymes or two carriers that are required to remove ammonia from the body. UCD may be associated with neurological damage encompassing a spectrum from asymptomatic/mild to severe encephalopathy, which results in most cases from Hyperammonemia (HA) and elevation of other neurotoxic intermediates of metabolism. Electroencephalography (EEG), Magnetic resonance imaging (MRI) and Proton Magnetic resonance spectroscopy (MRS) are noninvasive measures of brain function and structure that can be used during HA to guide management and provide prognostic information, in addition to being research tools to understand the pathophysiology of UCD associated brain injury. The Urea Cycle Rare disorders Consortium (UCDC) has been invested in research to understand the immediate and downstream effects of hyperammonemia (HA) on brain using electroencephalogram (EEG) and multimodal brain MRI to establish early patterns of brain injury and to track recovery and prognosis. This review highlights the evolving knowledge about the impact of UCD and HA in particular on neurological injury and recovery and use of EEG and MRI to study and evaluate prognostic factors for risk and recovery. It recognizes the work of others and discusses the UCDC's prior work and future research priorities.


Asunto(s)
Encéfalo , Electroencefalografía , Hiperamonemia , Imagen por Resonancia Magnética , Espectroscopía de Protones por Resonancia Magnética , Trastornos Innatos del Ciclo de la Urea , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/fisiopatología , Historia del Siglo XXI , Hiperamonemia/diagnóstico por imagen , Hiperamonemia/historia , Hiperamonemia/metabolismo , Hiperamonemia/fisiopatología , Trastornos Innatos del Ciclo de la Urea/diagnóstico por imagen , Trastornos Innatos del Ciclo de la Urea/historia , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/fisiopatología
2.
Mol Cell ; 81(18): 3749-3759, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34469752

RESUMEN

The expression of the urea cycle (UC) proteins is dysregulated in multiple cancers, providing metabolic benefits to tumor survival, proliferation, and growth. Here, we review the main changes described in the expression of UC enzymes and metabolites in different cancers at various stages and suggest that these changes are dynamic and should hence be viewed in a context-specific manner. Understanding the evolvability in the activity of the UC pathway in cancer has implications for cancer-immune cell interactions and for cancer diagnosis and therapy.


Asunto(s)
Carcinogénesis/metabolismo , Transformación Celular Neoplásica/metabolismo , Urea/metabolismo , Amoníaco/metabolismo , Línea Celular Tumoral , Proliferación Celular , Expresión Génica/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/fisiopatología
3.
J Inherit Metab Dis ; 44(4): 826-837, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33840128

RESUMEN

Urea cycle disorders (UCDs) are inherited metabolic disorders with impaired nitrogen detoxification caused by defects in urea cycle enzymes. They often manifest with hyperammonemic attacks resulting in significant morbidity or death. We performed a nationwide questionnaire-based study between January 2000 and March 2018 to document all UCDs in Japan, including diagnoses, treatments, and outcomes. A total of 229 patients with UCDs were enrolled in this study: 73 males and 53 females with ornithine transcarbamylase deficiency (OTCD), 33 patients with carbamoylphosphate synthetase 1 deficiency, 48 with argininosuccinate synthetase deficiency, 14 with argininosuccinate lyase deficiency, and 8 with arginase deficiency. Survival rates at 20 years of age of male and female patients with late-onset OTCD were 100% and 97.7%, respectively. Blood ammonia levels and time of onset had a significant impact on the neurodevelopmental outcome (P < .001 and P = .028, respectively). Hemodialysis and liver transplantation did not prevent poor neurodevelopmental outcomes. While treatment including medication, hemodialysis, and liver transplantation may aid in decreasing blood ammonia and/or preventing severe hyperammonemia, a blood ammonia level ≥ 360 µmol/L was found to be a significant indicator for a poor neurodevelopmental outcome. In conclusion, although current therapy for UCDs has advanced and helped saving lives, patients with blood ammonia levels ≥ 360 µmol/L at onset often have impaired neurodevelopmental outcomes. Novel neuroprotective measures should therefore be developed to achieve better neurodevelopmental outcomes in these patients.


Asunto(s)
Hiperamonemia/prevención & control , Trastornos del Neurodesarrollo/etiología , Trastornos Innatos del Ciclo de la Urea/fisiopatología , Trastornos Innatos del Ciclo de la Urea/terapia , Adolescente , Adulto , Amoníaco/sangre , Niño , Preescolar , Femenino , Humanos , Hiperamonemia/etiología , Japón/epidemiología , Trasplante de Hígado , Masculino , Diálisis Renal , Tasa de Supervivencia , Trastornos Innatos del Ciclo de la Urea/sangre , Trastornos Innatos del Ciclo de la Urea/epidemiología , Adulto Joven
4.
Nutrients ; 12(11)2020 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-33138136

RESUMEN

Low-protein diets (LPDs) are the main treatment for urea cycle disorders (UCDs) and organic acidemias (OAs). In most cases, LPDs start in childhood and must be continued into adulthood. The improved life expectancy of patients with UCDs and OAs raises the question of their consequences on nutritional status in adult subjects. As this topic has so far received little attention, we conducted a review of scientific studies that investigated the nutrient intake and nutritional status in adult patients with UCDs and branched chain organic acidemias (BCOAs) on LPD. METHODS: The literature search was conducted in PubMed/MEDLINE, Scopus, EMBASE and Google Scholar from 1 January 2000 to 31 May 2020, focusing on nutrient intake and nutritional status in UCD and OA adult patients. RESULTS: Despite protein restriction is recommended as the main treatment for UCDs and OAs, in these patients, protein intake ranges widely, with many patients who do not reach safety levels. When evaluated, micronutrient intake resulted below recommended values in some patients. Lean body mass resulted in most cases lower than normal range while fat body mass (FM) was often found normal or higher than the controls or reference values. Protein intake correlated inversely with FM both in adult and pediatric UCD patients. CONCLUSIONS: The clinical management of adult patients with UCDs and BCOAs should include an accurate assessment of the nutritional status and body composition. However, as little data is still available on this topic, further studies are needed to better clarify the effects of LPDs on nutritional status in adult UCD and BCOA patients.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/dietoterapia , Aminoácidos de Cadena Ramificada/metabolismo , Dieta con Restricción de Proteínas/métodos , Estado Nutricional , Trastornos Innatos del Ciclo de la Urea/dietoterapia , Adulto , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Composición Corporal , Ingestión de Alimentos/fisiología , Femenino , Humanos , Masculino , Micronutrientes/análisis , Resultado del Tratamiento , Trastornos Innatos del Ciclo de la Urea/fisiopatología
5.
Orphanet J Rare Dis ; 14(1): 208, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31443672

RESUMEN

BACKGROUND: Hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome is a rare disorder of urea cycle characterized by progressive pyramidal and cerebellar dysfunction, whose pathophysiology is not yet fully understood. Here we describe the spectrum of the long fibers involvement in HHH syndrome, attempting a correlation between clinical, electrophysiological and neuro-radiological data. METHODS: Nine HHH patients were longitudinally evaluated by clinical examination, neurophysiological assessment including motor (MEPs), somato-sensory evoked potentials (PESS) and nerve conduction velocity (NCV), brain and spinal cord MRI RESULTS: All patients had pyramidal dysfunction and 3/9 an overt spastic paraplegia. Mild to moderate cerebellar signs were found in 7/9, intellectual disability in 8/9. At lower limbs, MEPs resulted abnormal in 7/8 patients and PESS in 2/8; peripheral sensory-motor neuropathy was found in 1/9. MRI documented atrophic changes in supra-tentorial brain regions in 6/9 patients, cerebellum in 6/9, spinal cord in 3/7. CONCLUSIONS: A predominant corticospinal dysfunction is evident in HHH syndrome, along with milder cerebellar signs, intellectual disability of variable degree and rare peripheral neuropathy. Phenotypical similarities with other disorders affecting the urea cycle (argininemia and pyrroline-5-carboxylate synthetase deficiency) suggest possible common mechanisms contributing in the maintenance of the corticospinal tract integrity. HHH syndrome phenotype largely overlaps with complex Hereditary Spastic Paraplegias (HSPs), in the list of which it should be included, emphasizing the importance to screen all the unsolved cases of HSPs for metabolic biomarkers.


Asunto(s)
Hiperamonemia/metabolismo , Hiperamonemia/patología , Ornitina/deficiencia , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/patología , Adolescente , Adulto , Encéfalo/metabolismo , Encéfalo/fisiología , Niño , Femenino , Humanos , Hiperamonemia/fisiopatología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Mutación/genética , Conducción Nerviosa/fisiología , Ornitina/metabolismo , Paraplejía Espástica Hereditaria/metabolismo , Paraplejía Espástica Hereditaria/patología , Paraplejía Espástica Hereditaria/fisiopatología , Médula Espinal/metabolismo , Médula Espinal/fisiología , Trastornos Innatos del Ciclo de la Urea/fisiopatología , Adulto Joven
6.
Nat Rev Neurol ; 15(5): 253-271, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30914790

RESUMEN

Organic acidurias (OADs) are inherited neurometabolic diseases largely caused by deficiencies in enzymes involved in amino acid degradation, which result in accumulation of organic acids in the brain and other tissues. Disease presentation usually occurs in infancy, although late-onset variants can emerge during childhood or adulthood. Patients predominantly manifest with acute encephalopathy with life-threatening systemic manifestations (classical OADs) or progressive neurological symptoms (cerebral OADs), leading to permanent cerebral abnormalities. Some OADs are treatable, and early diagnosis and treatment implementation have substantially decreased the mortality and overall morbidity from OADs. However, long-term irreversible cerebral and systemic complications are frequent because the therapeutic options are currently limited. The pathophysiology of brain dysfunction is still unclear in most OADs, and further investigation is needed to enable the development of novel therapeutic strategies. This Review focuses on current knowledge of the OADs, including epidemiology, short-term and long-term neurological and systemic features, diagnosis and prognosis, and recent advances in therapy and pathophysiology. The goal of the article is to alert neurologists and related health professionals to the existence and importance of these neurometabolic diseases and to stimulate research into the damaging factors that contribute to their neurodegenerative sequelae.


Asunto(s)
Encefalopatías/etiología , Trastornos Innatos del Ciclo de la Urea/complicaciones , Adulto , Encefalopatías/fisiopatología , Niño , Progresión de la Enfermedad , Humanos , Lactante , Pronóstico , Trastornos Innatos del Ciclo de la Urea/fisiopatología
7.
Transplantation ; 103(9): 1903-1915, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30801523

RESUMEN

BACKGROUND: Regenerative medicine using stem cell technology is an emerging field that is currently tested for inborn and acquired liver diseases. OBJECTIVE: This phase I/II prospective, open label, multicenter, randomized trial aimed primarily at evaluating the safety of Heterologous Human Adult Liver-derived Progenitor Cells (HepaStem) in pediatric patients with urea cycle disorders (UCDs) or Crigler-Najjar (CN) syndrome 6 months posttransplantation. The secondary objective included the assessment of safety up to 12 months postinfusion and of preliminary efficacy. METHODS: Fourteen patients with UCDs and 6 with CN syndrome were divided into 3 cohorts by body weight and intraportally infused with 3 doses of HepaStem. Clinical status, portal vein hemodynamics, morphology of the liver, de novo detection of circulating anti-human leukocyte antigen antibodies, and clinically significant adverse events (AEs) and serious adverse events to infusion were evaluated by using an intent-to-treat analysis. RESULTS: The overall safety of HepaStem was confirmed. For the entire study period, patient-month incidence rate was 1.76 for the AEs and 0.21 for the serious adverse events, of which 38% occurred within 1 month postinfusion. There was a trend of higher events in UCD as compared with CN patients. Segmental left portal vein thrombosis occurred in 1 patient and intraluminal local transient thrombus in a second patient. The other AEs were in line with expectations for catheter placement, cell infusion, concomitant medications, age, and underlying diseases. CONCLUSIONS: This study led to European clinical trial authorization for a phase II study in a homogeneous patient cohort, with repeated infusions and intermediate doses.


Asunto(s)
Síndrome de Crigler-Najjar/tratamiento farmacológico , Trasplante de Hígado , Hígado/metabolismo , Trasplante de Células Madre , Trastornos Innatos del Ciclo de la Urea/cirugía , Adolescente , Factores de Edad , Niño , Preescolar , Síndrome de Crigler-Najjar/sangre , Síndrome de Crigler-Najjar/diagnóstico , Síndrome de Crigler-Najjar/fisiopatología , Europa (Continente) , Femenino , Humanos , Lactante , Hígado/patología , Hígado/fisiopatología , Regeneración Hepática , Trasplante de Hígado/efectos adversos , Masculino , Estudios Prospectivos , Trasplante de Células Madre/efectos adversos , Factores de Tiempo , Trasplante Heterólogo , Resultado del Tratamiento , Trastornos Innatos del Ciclo de la Urea/sangre , Trastornos Innatos del Ciclo de la Urea/diagnóstico , Trastornos Innatos del Ciclo de la Urea/fisiopatología
8.
Nephrology (Carlton) ; 23(10): 957-961, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29888426

RESUMEN

Severe urea cycle defects (UCD), organic acidemias (OA) and maple syrup urine disease (MSUD) are life-threatening disorders presenting in the first days of life. Renal replacement therapy (RRT) is an emergency option in affected newborns, mostly performed as ultima ratio. We report our 10-year experience using emergency RRT in newborns with UCD, OA and MSUD. Twelve newborns (eight with UCD, two with methylmalonic acidemia and two with MSUD) underwent emergency RRT. The overall survival rate to RRT was 58.3%. Hyperammonemic newborns required earlier RRT with respect to MSUD patients (75 (65-102) vs 301 (192-410) h of life, P < 0.01). Hyperammonemic neonates surviving (n = 5) and non-surviving (n = 5) the acute neonatal decompensation showed similar birth weight (P = 0.690), duration of intubation (P = 0.917), ammonia at onset (P = 0.916) and at the start of RRT (P = 0.426), age at RRT (P = 0.999) and duration of coma before RRT (P = 0.691). Remarkably, all survivors quickly responded to RRT, with ammonia concentration less than 300 µmol/L after 8 h of treatment. One patient with UCD successfully treated by neonatal RRT died at 4 months of life because of sepsis. All patients with MSUD had normalized leucine levels after 12 h of RRT, surviving the acute neonatal decompenstation. All long-term survivors (five liver transplanted, one waiting for liver transplantation) currently show normal or near-normal neurological development (48 ± 39 months of age). Early response to RRT was associated with survival irrespective of pre-treatment picture. RRT can be considered even in huge neonatal metabolic decompensations. Early liver transplantation may be an option for select patients.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Enfermedad de la Orina de Jarabe de Arce/terapia , Terapia de Reemplazo Renal/métodos , Trastornos Innatos del Ciclo de la Urea/terapia , Factores de Edad , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/mortalidad , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Desarrollo Infantil , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Trasplante de Hígado , Masculino , Enfermedad de la Orina de Jarabe de Arce/diagnóstico , Enfermedad de la Orina de Jarabe de Arce/mortalidad , Enfermedad de la Orina de Jarabe de Arce/fisiopatología , Recuperación de la Función , Terapia de Reemplazo Renal/efectos adversos , Terapia de Reemplazo Renal/mortalidad , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Trastornos Innatos del Ciclo de la Urea/diagnóstico , Trastornos Innatos del Ciclo de la Urea/mortalidad , Trastornos Innatos del Ciclo de la Urea/fisiopatología
9.
Pan Afr Med J ; 31: 103, 2018.
Artículo en Francés | MEDLINE | ID: mdl-31037164

RESUMEN

In some inherited metabolic diseases, in particular in urea cycle disorders, which are usually diagnosed in neonatal period or in childhood, vomiting is often the first symptom. We report a case of late revelation of urea cycle disorder in a 13 years old female patient hospitalized for convulsions and failure to thrive. The patient underwent an interview revealing chronic vomiting associated with behavioral disorders, ideomotor slowdown and headaches. Clinical examination showed ataxia. Lumbar puncture and head CT scan were normal. The patient had substantially elevated blood ammonia level at 75 micromoles/L (11-50). Chromatography of amino acids in the blood showed increased glutamine and alanine. Chromatography of amino acids in the urine showed increased basic amino acids evoking a deficit of the urea cycle due to deficit of the enzyme argininosuccinate lyase. The patient was treated as an emergency, exclusively with glycolipid-diet and sodium benzoate, allowing improvement of patient's clinical condition and weight resumption. The seizures were controlled by phenobarbital. Family interview revealed that patient's sister, aged 20 years, had a 3-year history of seizures treated with phenobarbital. She underwent metabolic assessment in our department, which showed the same urea cycle abnormality as hes sister. Urea cycle deficiency should be suspected in patients of any age with encephalopathy associated with epilepsy, vomiting, weight stagnation and hyperammonemia. The diagnosis is very often made during severe neurodigestive attack involving vomiting, attack and/or seizures.


Asunto(s)
Convulsiones/etiología , Trastornos Innatos del Ciclo de la Urea/diagnóstico , Vómitos/etiología , Adolescente , Anticonvulsivantes/uso terapéutico , Peso Corporal , Insuficiencia de Crecimiento/etiología , Femenino , Cefalea/etiología , Humanos , Hiperamonemia/etiología , Fenobarbital/uso terapéutico , Convulsiones/tratamiento farmacológico , Trastornos Innatos del Ciclo de la Urea/fisiopatología , Trastornos Innatos del Ciclo de la Urea/terapia
10.
Expert Opin Ther Targets ; 21(4): 391-399, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28281899

RESUMEN

INTRODUCTION: Carbamoyl phosphate synthetase 1 (CPS1) deficiency (CPS1D) is a rare autosomal recessive urea cycle disorder (UCD), which can lead to life-threatening hyperammonemia. Unless promptly treated, it can result in encephalopathy, coma and death, or intellectual disability in surviving patients. Over recent decades, therapies for CPS1D have barely improved leaving the management of these patients largely unchanged. Additionally, in many cases, current management (protein-restriction and supplementation with citrulline and/or arginine and ammonia scavengers) is insufficient for achieving metabolic stability, highlighting the importance of developing alternative therapeutic approaches. Areas covered: After describing UCDs and CPS1D, we give an overview of the structure- function of CPS1. We then describe current management and potential novel treatments including N-carbamoyl-L-glutamate (NCG), pharmacological chaperones, and gene therapy to treat hyperammonemia. Expert opinion: Probably, the first novel CPS1D therapies to reach the clinics will be the already commercial substance NCG, which is the standard treatment for N-acetylglutamate synthase deficiency and has been proven to rescue specific CPS1D mutations. Pharmacological chaperones and gene therapy are under development too, but these two technologies still have key challenges to be overcome. In addition, current experimental therapies will hopefully add further treatment options.


Asunto(s)
Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/terapia , Glutamatos/uso terapéutico , Animales , Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/genética , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/fisiopatología , Terapia Genética/métodos , Humanos , Chaperonas Moleculares/farmacología , Mutación , Trastornos Innatos del Ciclo de la Urea/fisiopatología , Trastornos Innatos del Ciclo de la Urea/terapia
11.
Expert Opin Drug Metab Toxicol ; 13(4): 439-448, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27860485

RESUMEN

INTRODUCTION: Ammonia-scavenging drugs, benzoate and phenylacetate (PA)/phenylbutyrate (PB), modulate hepatic nitrogen metabolism mainly by providing alternative pathways for nitrogen disposal. Areas covered: We review the major findings and potential novel applications of ammonia-scavenging drugs, focusing on urea cycle disorders and liver disease. Expert opinion: For over 40 years, ammonia-scavenging drugs have been used in the treatment of urea cycle disorders. Recently, the use of these compounds has been advocated in acute liver failure and cirrhosis for reducing hyperammonemic-induced hepatic encephalopathy. The efficacy and mechanisms underlying the antitumor effects of these ammonia-scavenging drugs in liver cancer are more controversial and are discussed in the review. Overall, as ammonia-scavenging drugs are usually safe and well tolerated among cancer patients, further studies should be instigated to explore the role of these drugs in liver cancer. Considering the relevance of glutamine metabolism to the progression and resolution of liver disease, we propose that ammonia-scavenging drugs might also be used to non-invasively probe liver glutamine metabolism in vivo. Finally, novel derivatives of classical ammonia-scavenging drugs with fewer and less severe adverse effects are currently being developed and used in clinical trials for the treatment of acute liver failure and cirrhosis.


Asunto(s)
Amoníaco/metabolismo , Hepatopatías/tratamiento farmacológico , Trastornos Innatos del Ciclo de la Urea/tratamiento farmacológico , Animales , Benzoatos/metabolismo , Diseño de Fármacos , Glutamina/metabolismo , Humanos , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/fisiopatología , Hepatopatías/fisiopatología , Fallo Hepático Agudo/tratamiento farmacológico , Fallo Hepático Agudo/fisiopatología , Nitrógeno/metabolismo , Fenilacetatos/metabolismo , Fenilbutiratos/metabolismo , Trastornos Innatos del Ciclo de la Urea/fisiopatología
12.
Mol Genet Metab ; 119(4): 307-310, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27771289

RESUMEN

BACKGROUND: N-acetylglutamate synthase (NAGS) plays a key role in the removal of ammonia via the urea cycle by catalyzing the synthesis of N-acetylglutamate (NAG), the obligatory cofactor in the carbamyl phosphate synthetase 1 reaction. Enzymatic analysis of NAGS in liver homogenates has remained insensitive and inaccurate, which prompted the development of a novel method. METHODS: UPLC-MS/MS was used in conjunction with stable isotope (N-acetylglutamic-2,3,3,4,4-d5 acid) dilution for the quantitative detection of NAG produced by the NAGS enzyme. The assay conditions were optimized using purified human NAGS and the optimized enzyme conditions were used to measure the activity in mouse liver homogenates. RESULTS: A low signal-to-noise ratio in liver tissue samples was observed due to non-enzymatic formation of N-acetylglutamate and low specific activity, which interfered with quantitative analysis. Quenching of acetyl-CoA immediately after the incubation circumvented this analytical difficulty and allowed accurate and sensitive determination of mammalian NAGS activity. The specificity of the assay was validated by demonstrating a complete deficiency of NAGS in liver homogenates from Nags -/- mice. CONCLUSION: The novel NAGS enzyme assay reported herein can be used for the diagnosis of inherited NAGS deficiency and may also be of value in the study of secondary hyperammonemia present in various inborn errors of metabolism as well as drug treatment.


Asunto(s)
N-Acetiltransferasa de Aminoácidos/genética , Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Hiperamonemia/diagnóstico , Trastornos Innatos del Ciclo de la Urea/diagnóstico , Acetilcoenzima A/metabolismo , N-Acetiltransferasa de Aminoácidos/metabolismo , Animales , Carbamoil-Fosfato Sintasa (Amoniaco)/deficiencia , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/fisiopatología , Hígado/enzimología , Ratones , Ratones Noqueados , Espectrometría de Masas en Tándem , Trastornos Innatos del Ciclo de la Urea/genética , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/fisiopatología
13.
Mol Genet Metab ; 112(1): 25-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24721342

RESUMEN

The hyperornithinemia-hyperammonemia-homocitrullinuria syndrome is a rare autosomal recessive disorder caused by the functional deficiency of the mitochondrial ornithine transporter 1 (ORC1). ORC1 is encoded by the SLC25A15 gene and catalyzes the transport of cytosolic ornithine into mitochondria in exchange for citrulline. Although the age of onset and the severity of the symptoms vary widely, the disease usually manifests in early infancy. The typical clinical features include protein intolerance, lethargy, episodic confusion, cerebellar ataxia, seizures and mental retardation. In this study, we identified a novel p.Ala15Val (c.44C>T) mutation by genomic DNA sequencing in a Turkish child presenting severe tantrum, confusion, gait disturbances and loss of speech abilities in addition to hyperornithinemia, hyperammonemia and homocitrullinuria. One hundred Turkish control chromosomes did not possess this variant. The functional effect of the novel mutation was assessed by both complementation of the yeast ORT1 null mutant and transport assays. Our study demonstrates that the A15V mutation dramatically interferes with the transport properties of ORC1 since it was shown to inhibit ornithine transport nearly completely.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Trastornos Innatos del Ciclo de la Urea/complicaciones , Trastornos Innatos del Ciclo de la Urea/genética , Niño , Análisis Mutacional de ADN , Humanos , Masculino , Mitocondrias/fisiología , Proteínas de Transporte de Membrana Mitocondrial , Mutación Puntual , Alineación de Secuencia , Trastornos Innatos del Ciclo de la Urea/fisiopatología
14.
Mol Genet Metab ; 112(1): 17-24, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24630270

RESUMEN

OBJECTIVE: To evaluate glycerol phenylbutyrate (GPB) in the treatment of pediatric patients with urea cycle disorders (UCDs). STUDY DESIGN: UCD patients (n=26) ages 2months through 17years were treated with GPB and sodium phenylbutyrate (NaPBA) in two short-term, open-label crossover studies, which compared 24-hour ammonia exposure (AUC0-24) and glutamine levels during equivalent steady-state dosing of GPB and sodium phenylbutyrate (NaPBA). These 26 patients plus an additional 23 patients also received GPB in one of three 12-month, open label extension studies, which assessed long-term ammonia control, hyperammonemic (HA) crises, amino acid levels, and patient growth. RESULTS: Mean ammonia exposure on GPB was non-inferior to NaPBA in each of the individual crossover studies. In the pooled analyses, it was significantly lower on GPB vs. NaPBA (mean [SD] AUC0-24: 627 [302] vs. 872 [516] µmol/L; p=0.008) with significantly fewer abnormal values (15% on GPB vs. 35% on NaPBA; p=0.02). Mean ammonia levels remained within the normal range during 12months of GPB dosing and, when compared with the 12months preceding enrollment, a smaller percentage of patients (24.5% vs. 42.9%) experienced fewer (17 vs. 38) HA crises. Glutamine levels tended to be lower with GPB than with NaPBA during short-term dosing (mean [SD]: 660.8 [164.4] vs. 710.0 [158.7] µmol/L; p=0.114) and mean glutamine and branched chain amino acid levels, as well as other essential amino acids, remained within the normal range during 12months of GPB dosing. Mean height and weight Z-scores were within normal range at baseline and did not change significantly during 12months of GPB treatment. CONCLUSIONS: Dosing with GPB was associated with 24-hour ammonia exposure that was non-inferior to that during dosing with NaPBA in individual studies and significantly lower in the pooled analysis. Long-term GPB dosing was associated with normal levels of glutamine and essential amino acids, including branched chain amino acids, age-appropriate growth and fewer HA crises as compared with the 12month period preceding enrollment.


Asunto(s)
Fenilbutiratos/administración & dosificación , Fenilbutiratos/efectos adversos , Trastornos Innatos del Ciclo de la Urea/tratamiento farmacológico , Adolescente , Amoníaco/sangre , Niño , Preescolar , Estudios Cruzados , Femenino , Glutamina/orina , Humanos , Lactante , Recién Nacido , Masculino , Fenilbutiratos/uso terapéutico , Trastornos Innatos del Ciclo de la Urea/dietoterapia , Trastornos Innatos del Ciclo de la Urea/fisiopatología
16.
Eur J Pediatr ; 173(12): 1635-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24233332

RESUMEN

Acute hyperammonemia has a variety of etiologies and clinical manifestations. If not treated early in neonates, it leads to irreversible brain damage or death. We present a 7-day-old female patient who was brought to the emergency department with drownsiness and vomiting. Metabolic work-up revealed a blood ammonia level of 290 µmol/L (normal <100 µmol/L in neonates) with a compensated respiratory alkalosis, normal glycaemia and lactate and absence of urinary ketones. Oral feeding was stopped, an infusion of 20 % glucose was started, and sodium benzoate and arginine hydrochloride were given. After a drop of ammonemia within 12 h of treatment, it started rising again. N-carbamylglutamate (NCG) was added resulting in a rapid normalisation of ammonemia. Feedings were progressively reintroduced, the ammonia levels remained low. The results of the metabolic work-up were compatible with carbamyl phosphate synthase 1 (CPS1) or N-acetyl glutamate synthase (NAGS) deficiency. Genetic analysis confirmed the latter diagnosis with a homozygous mutation c. 1450T > C (p.W484R) in exon 6 of the NAGS gene in the patient and a carrier state in both parents. At the age of 9 months, the child is growing well with normal neurological development, under treatment with NCG 100 mg/kg/day and a normal diet. Conclusion: This case highlights the importance of keeping a high index of suspicion and early testing for ammonia levels in neonates/children with unexplained encephalopathy. In neonates with congenital hyperammonemia, NCG should always be started together with the standard management of hyperammonemia until all laboratory investigations are complete or indicate another disease.


Asunto(s)
Encéfalo/fisiopatología , Glutamatos/uso terapéutico , Trastornos Innatos del Ciclo de la Urea/tratamiento farmacológico , N-Acetiltransferasa de Aminoácidos/sangre , Femenino , Humanos , Recién Nacido , Factores de Tiempo , Trastornos Innatos del Ciclo de la Urea/sangre , Trastornos Innatos del Ciclo de la Urea/fisiopatología
17.
Cell Mol Neurobiol ; 33(1): 137-46, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23053545

RESUMEN

Isolated 3-methylcrotonyl-CoA carboxylase deficiency (3MCCD) is an autosomal recessive disorder of leucine metabolism biochemically characterized by accumulation of 3-methylcrotonylglycine (3MCG), 3-methylcrotonic acid (3MCA) and 3-hydroxyisovaleric acid. A considerable number of affected individuals present neurological symptoms with or without precedent crises of metabolic decompensation and brain abnormalities whose pathogenesis is poorly known. We investigated the in vitro effects of 3MCG and 3MCA on important parameters of oxidative stress in cerebral cortex of young rats. 3MCG and 3MCA significantly increased TBA-RS and carbonyl formation, indicating that these compounds provoke lipid and protein oxidation, respectively. In contrast, nitric oxide production was not affected by 3MCG and 3MCA. Furthermore, 3MCG- and 3MCA-induced elevation of TBA-RS values was fully prevented by melatonin, trolox and reduced glutathione, but not by the nitric oxide inhibitor N(ω)-nitro-L-arginine methyl ester or the combination of catalase plus superoxide dismutase, indicating that reactive oxygen species were involved in the oxidative damage caused by these compounds. We also found that the activity of the antioxidant enzymes glutathione peroxidase, catalase, superoxide dismutase and glutathione reductase were not altered in vitro by 3MCG and 3MCA. It is therefore presumed that alterations of the cellular redox homeostasis caused by the major metabolites accumulating in 3MCCD may potentially be involved in the pathophysiology of the neurological dysfunction and structural brain alterations found in patients affected by this disorder.


Asunto(s)
Química Encefálica/fisiología , Ligasas de Carbono-Carbono/deficiencia , Corteza Cerebral/metabolismo , Estrés Oxidativo/fisiología , Factores de Edad , Animales , Corteza Cerebral/efectos de los fármacos , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Trastornos Innatos del Ciclo de la Urea/enzimología , Trastornos Innatos del Ciclo de la Urea/fisiopatología
19.
Orphanet J Rare Dis ; 7: 31, 2012 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-22642865

RESUMEN

BACKGROUND: Isolated 3-methylcrotonyl-CoA carboxylase (MCC) deficiency is an autosomal recessive disorder of leucine metabolism caused by mutations in MCCC1 or MCCC2 encoding the α and ß subunit of MCC, respectively. The phenotype is highly variable ranging from acute neonatal onset with fatal outcome to asymptomatic adults. METHODS: We report clinical, biochemical, enzymatic and mutation data of 88 MCC deficient individuals, 53 identified by newborn screening, 26 diagnosed due to clinical symptoms or positive family history and 9 mothers, identified following the positive newborn screening result of their baby. RESULTS: Fifty-seven percent of patients were asymptomatic while 43% showed clinical symptoms, many of which were probably not related to MCC deficiency but due to ascertainment bias. However, 12 patients (5 of 53 identified by newborn screening) presented with acute metabolic decompensations. We identified 15 novel MCCC1 and 16 novel MCCC2 mutant alleles. Additionally, we report expression studies on 3 MCCC1 and 8 MCCC2 mutations and show an overview of all 132 MCCC1 and MCCC2 variants known to date. CONCLUSIONS: Our data confirm that MCC deficiency, despite low penetrance, may lead to a severe clinical phenotype resembling classical organic acidurias. However, neither the genotype nor the biochemical phenotype is helpful in predicting the clinical course.


Asunto(s)
Trastornos Innatos del Ciclo de la Urea/metabolismo , Ligasas de Carbono-Carbono/deficiencia , Ligasas de Carbono-Carbono/genética , Ligasas de Carbono-Carbono/metabolismo , Línea Celular , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Reacción en Cadena de la Polimerasa , Encuestas y Cuestionarios , Trastornos Innatos del Ciclo de la Urea/genética , Trastornos Innatos del Ciclo de la Urea/patología , Trastornos Innatos del Ciclo de la Urea/fisiopatología
20.
Clin Chim Acta ; 413(13-14): 1151-5, 2012 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-22465082

RESUMEN

BACKGROUND: In hyperornithinemia-hyperammonemia-homocitrullinemia (HHH) syndrome, impaired ornithine transport across the mitochondrial membrane causes ornithine accumulation in cytoplasm. The resulting mitochondrial ornithine deficiency leads to reduced clearance of ammonia through the urea cycle. First described in 1969, no long-term follow-up has been reported. METHODS: Four patients were followed up for 11 to 38y. Diagnosis was made by plasma amino acid analysis using ion exchange chromatography, HPLC orotic acid measurement, and (14)C-ornithine incorporation study using cultured fibroblasts. DNA from fibroblasts was amplified and sequenced. Blood ammonia was controlled by restriction of protein intake. RESULTS: All patients had reduced (14)C-ornithine incorporation. Mutation analysis revealed two novel mutations in the ORNT1 gene. Neurologic outcome included memory loss, low IQ, tremor, spasticity of extremities, bladder incontinence, and abnormal gait. Neuroimaging revealed subcortical, cerebral and cerebellar atrophy, sparing the basal ganglia. Individual examination showed pyramidal signs, cerebellar signs, paraplegia, movement disorder, dystonia, and epilepsy. One patient had 3 pregnancies, one of which resulted in intrauterine growth retardation. CONCLUSIONS: Our patients expand the clinical phenotype of adults with HHH. Long-term follow-up showed serious neurologic outcomes in all patients; three patients clearly exhibited progression of neurologic dysfunction despite control of hyperammonemia. Intracellular ornithine deficiency may adversely affect brain functions.


Asunto(s)
Hiperamonemia/fisiopatología , Trastornos Innatos del Ciclo de la Urea/fisiopatología , Adulto , Sistemas de Transporte de Aminoácidos Básicos/genética , Aminoácidos/sangre , Cromatografía Líquida de Alta Presión , Cromatografía por Intercambio Iónico , Análisis Mutacional de ADN , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Hiperamonemia/sangre , Hiperamonemia/genética , Masculino , Persona de Mediana Edad , Proteínas de Transporte de Membrana Mitocondrial , Ornitina/sangre , Ornitina/deficiencia , Ornitina/genética , Fenotipo , Factores de Tiempo , Trastornos Innatos del Ciclo de la Urea/sangre , Trastornos Innatos del Ciclo de la Urea/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...