Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
JCI Insight ; 8(17)2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37490345

RESUMEN

Nitric oxide (NO) is a critical signaling molecule that has been implicated in the pathogenesis of neurocognitive diseases. Both excessive and insufficient NO production have been linked to pathology. Previously, we have shown that argininosuccinate lyase deficiency (ASLD) is a novel model system to investigate cell-autonomous, nitric oxide synthase-dependent NO deficiency. Humans with ASLD are at increased risk for developing hyperammonemia due to a block in ureagenesis. However, natural history studies have shown that individuals with ASLD have multisystem disease including neurocognitive deficits that can be independent of ammonia. Here, using ASLD as a model of NO deficiency, we investigated the effects of NO on brain endothelial cells in vitro and the blood-brain barrier (BBB) in vivo. Knockdown of ASL in human brain microvascular endothelial cells (HBMECs) led to decreased transendothelial electrical resistance, indicative of increased cell permeability. Mechanistically, treatment with an NO donor or inhibition of Claudin-1 improved barrier integrity in ASL-deficient HBMECs. Furthermore, in vivo assessment of a hypomorphic mouse model of ASLD showed increased BBB leakage, which was partially rescued by NO supplementation. Our results suggest that ASL-mediated NO synthesis is required for proper maintenance of brain microvascular endothelial cell functions as well as BBB integrity.


Asunto(s)
Aciduria Argininosuccínica , Ratones , Animales , Humanos , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/metabolismo , Aciduria Argininosuccínica/patología , Óxido Nítrico/metabolismo , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Claudinas/metabolismo , Modelos Animales de Enfermedad
2.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33373331

RESUMEN

Previous studies have shown that nitric oxide (NO) supplements may prevent bone loss and fractures in preclinical models of estrogen deficiency. However, the mechanisms by which NO modulates bone anabolism remain largely unclear. Argininosuccinate lyase (ASL) is the only mammalian enzyme capable of synthesizing arginine, the sole precursor for nitric oxide synthase-dependent (NOS-dependent) NO synthesis. Moreover, ASL is also required for channeling extracellular arginine to NOS for NO production. ASL deficiency (ASLD) is thus a model to study cell-autonomous, NOS-dependent NO deficiency. Here, we report that loss of ASL led to decreased NO production and impairment of osteoblast differentiation. Mechanistically, the bone phenotype was at least in part driven by the loss of NO-mediated activation of the glycolysis pathway in osteoblasts that led to decreased osteoblast differentiation and function. Heterozygous deletion of caveolin 1, a negative regulator of NO synthesis, restored NO production, osteoblast differentiation, glycolysis, and bone mass in a hypomorphic mouse model of ASLD. The translational significance of these preclinical studies was further reiterated by studies conducted in induced pluripotent stem cells from an individual with ASLD. Taken together, our findings suggest that ASLD is a unique genetic model for studying NO-dependent osteoblast function and that the NO/glycolysis pathway may be a new target to modulate bone anabolism.


Asunto(s)
Aciduria Argininosuccínica/metabolismo , Huesos/metabolismo , Diferenciación Celular , Glucólisis , Ácido Nítrico/metabolismo , Osteoblastos/metabolismo , Adolescente , Adulto , Animales , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/patología , Huesos/patología , Niño , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Osteoblastos/patología
3.
Mol Genet Metab ; 131(4): 390-397, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33288448

RESUMEN

OBJECTIVE: The implementation of newborn screening (NBS) programs for citrullinemia type 1 (CTLN1) and argininosuccinic aciduria (ASA) is subject to controversial debate. The aim of this study was to assess the impact of NBS on the metabolic disease course and clinical outcome of affected individuals. METHODS: In 115 individuals with CTLN1 and ASA, we compared the severity of the initial hyperammonemic episode (HAE) and the frequency of (subsequent) HAEs with the mode of diagnosis. Based on a recently established functional disease prediction model, individuals were stratified according to their predicted severe or attenuated phenotype. RESULTS: Individuals with predicted attenuated forms of CTLN1 and ASA were overrepresented in the NBS group, while those with a predicted severe phenotype were underrepresented compared to individuals identified after the manifestation of symptoms (SX). Identification by NBS was associated with reduced severity of the initial HAE both in individuals with predicted severe and attenuated phenotypes, while it was not associated with lower frequency of (subsequent) HAEs. Similar results were obtained when including some patients diagnosed presymptomatically (i.e. prenatal testing, and high-risk family screening) in this analysis. CONCLUSION: Since one of the major challenges of NBS outcome studies is the potential overrepresentation of individuals with predicted attenuated phenotypes in NBS cohorts, severity-adjusted evaluation of screened and unscreened individuals is important to avoid overestimation of the NBS effect. NBS enables the attenuation of the initial HAE but does not affect the frequency of subsequent metabolic decompensations in individuals with CTLN1 and ASA. Future long-term studies will need to evaluate the clinical impact of this finding, especially with regard to mortality, as well as cognitive outcome and quality of life of survivors.


Asunto(s)
Aciduria Argininosuccínica/diagnóstico , Citrulinemia/diagnóstico , Enfermedades Metabólicas/genética , Trastornos Innatos del Ciclo de la Urea/diagnóstico , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/metabolismo , Aciduria Argininosuccínica/patología , Citrulinemia/genética , Citrulinemia/metabolismo , Citrulinemia/patología , Femenino , Humanos , Hiperamonemia/diagnóstico , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/patología , Recién Nacido , Masculino , Enfermedades Metabólicas/diagnóstico , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Tamizaje Neonatal , Calidad de Vida , Índice de Severidad de la Enfermedad , Trastornos Innatos del Ciclo de la Urea/genética , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/patología
4.
Mol Genet Genomic Med ; 8(7): e1301, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32410394

RESUMEN

BACKGROUND: The urea cycle plays a key role in preventing the accumulation of toxic nitrogenous waste products, including two essential enzymes: ornithine transcarbamylase (OTC) and argininosuccinate lyase (ASL). Ornithine transcarbamylase deficiency (OTCD) results from mutations in the OTC. Meanwhile, argininosuccinate lyase deficiency (ASLD) is caused by mutations in the ASL. METHODS: Blood tandem mass spectrometric analysis and urea organic acidemia screening were performed on five Chinese cases, including three OTCD and two ASLD patients. Next-generation sequencing was then used to make a definite diagnosis, and the related variants were validated by Sanger sequencing. RESULTS: The five patients exhibited severe clinical symptoms, with abnormal biochemical analysis and amino acids profile. Genetic analysis revealed two variants [c.77G>A (p.Arg26Gln); c.116G>T (p.Gly39Val)] in the OTC, as well as two variants [c.1311T>G (p.Tyr437*); c.961T>A (p.Tyr321Asn)] in the ASL. Conservation analysis showed that the amino acids of the two novel mutations were highly conserved in different species and were predicted to be possibly damaging with several in silico prediction programs. 3D-modeling analysis indicated that the two novel missense variants might result in modest distortions of the OTC and ASL protein structures, respectively. CONCLUSIONS: Two novel variants expand the mutational spectrums of the OTC and ASL. All the results may contribute to a better understanding of the clinical course and genetic characteristics of patients with urea cycle disorders.


Asunto(s)
Argininosuccinatoliasa/genética , Aciduria Argininosuccínica/genética , Mutación , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/genética , Ornitina Carbamoiltransferasa/genética , Argininosuccinatoliasa/química , Aciduria Argininosuccínica/patología , Femenino , Humanos , Lactante , Masculino , Simulación de Dinámica Molecular , Ornitina Carbamoiltransferasa/química , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/patología , Linaje , Dominios Proteicos
5.
Stem Cell Rev Rep ; 16(1): 186-197, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31792768

RESUMEN

Previous studies have shown that human liver stem-like cells (HLSCs) may undergo differentiation in vitro into urea producing hepatocytes and in vivo may sustain liver function in models of experimentally induced acute liver injury. The aim of this study was to assess the safety of HLSCs intrahepatic administration in inherited neonatal-onset hyperammonemia. The study was approved by the Agenzia Italiana del Farmaco on favorable opinion of the Italian Institute of Health as an open-label, prospective, uncontrolled, monocentric Phase I study (HLSC 01-11, EudraCT-No. 2012-002120-33). Three patients affected by argininosuccinic aciduria (patient 1) and methylmalonic acidemia (patients 2 and 3) and included in the liver transplantation list were enrolled. In all patients, HLSCs were administered by percutaneous intrahepatic injections (once a week for two consecutive weeks) within the first months of life. The first patient received 125,000 HLSCs x gram of liver/dose while the other two patients received twice this dose. No immunosuppression was administered since HLSCs possess immunomodulatory activities. None of the patients experienced infections, hyperammonemia decompensation, or other adverse events during the whole observation period. No donor specific antibodies (DSA) against HLSCs were detected. Patients were metabolic stable despite an increase (~30%) in protein intake. Two patients underwent liver transplantation after 19 and 11 months respectively, and after explantation, the native livers showed no histological alterations. In conclusion, percutaneous intrahepatic administration of HLSCs was safe in newborn with inherited neonatal-onset hyperammonemia. These data pave the way for Phase II studies in selected inherited and acquired liver disorders.


Asunto(s)
Hiperamonemia/terapia , Trasplante de Hígado , Hígado/metabolismo , Errores Innatos del Metabolismo/terapia , Trasplante de Células Madre , Edad de Inicio , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/patología , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Amoníaco/metabolismo , Aciduria Argininosuccínica/metabolismo , Aciduria Argininosuccínica/patología , Aciduria Argininosuccínica/terapia , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/patología , Recién Nacido , Hígado/crecimiento & desarrollo , Hígado/patología , Masculino , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/metabolismo , Células Madre/metabolismo , Urea/metabolismo
6.
Orphanet J Rare Dis ; 14(1): 203, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31426867

RESUMEN

BACKGROUND: The incidence, prevalence, and molecular epidemiology of urea cycle disorders (UCDs) in Argentina remain underexplored. The present study is the first to thoroughly assess the clinical and molecular profiles of UCD patients examined at a single reference center in Argentina. RESULTS: Forty-nine UCD cases were collected. About half (26/49, 53%) manifested neonatally with classical presentation and had a high mortality (25/26, 96%). Ornithine transcarbamylase deficiency (OTCD) was the most common UCD (26 patients). Argininosuccinate synthetase deficiency (ASSD) was detected in 19 cases, while argininosuccinate lyase deficiency (ASLD) was diagnosed in 4 cases. Molecular genetic analysis revealed 8 private OTC mutations and two large deletion/duplication events in the OTC gene. Most mutations in the ASS1 and ASL genes were recurrent missense changes, and four alterations were novel. The clinical outcome of our UCD cohort was poor, with an overall mortality of 57% (28/49 cases), and a 28% (6/21) disability rate among the survivors. CONCLUSIONS: Most patients in our case series showed severe neonatal onset, with high morbidity/mortality. We detected in total 19 mutations, most of them recurrent and of high frequency worldwide. Noteworthy, we highlight the presence of a geographic cluster with high prevalence of a point mutation in the ASS1 gene. This study suggests that these disorders may be more frequent than commonly assumed, and stresses the need for increased awareness amongst health professionals and greater availability of diagnostic tools for accurate identification, early diagnosis, and timely treatment.


Asunto(s)
Trastornos Innatos del Ciclo de la Urea/epidemiología , Trastornos Innatos del Ciclo de la Urea/genética , Trastornos Innatos del Ciclo de la Urea/patología , Argentina/epidemiología , Aciduria Argininosuccínica/epidemiología , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/patología , Niño , Preescolar , Citrulinemia/epidemiología , Citrulinemia/genética , Citrulinemia/patología , Femenino , Humanos , Hiperamonemia/epidemiología , Hiperamonemia/genética , Hiperamonemia/patología , Lactante , Recién Nacido , Masculino , Mutación/genética , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/epidemiología , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/genética , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/patología
7.
J Inherit Metab Dis ; 42(6): 1077-1087, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30907007

RESUMEN

The urea cycle disorder (UCD) argininosuccinate lyase (ASL) deficiency, caused by a defective ASL enzyme, exhibits a wide range of phenotypes, from life-threatening neonatal hyperammonemia to asymptomatic patients, with only the biochemical marker argininosuccinic acid (ASA) elevated in body fluids. Remarkably, even without ever suffering from hyperammonemia, patients often develop severe cognitive impairment and seizures. The goal of this study was to understand the effect on the known toxic metabolite ASA and the assumed toxic metabolite guanidinosuccinic acid (GSA) on developing brain cells, and to evaluate the potential role of creatine (Cr) supplementation, as it was described protective for brain cells exposed to ammonia. We used an in vitro model, in which we exposed three-dimensional (3D) organotypic rat brain cell cultures in aggregates to different combinations of the metabolites of interest at two time points (representing two different developmental stages). After harvest and cryopreservation of the cell cultures, the samples were analyzed mainly by metabolite analysis, immunohistochemistry, and western blotting. ASA and GSA were found toxic for astrocytes and neurons. This toxicity could be reverted in vitro by Cr. As well, an antiapoptotic effect of ASA was revealed, which could contribute to the neurotoxicity in ASL deficiency. Further studies in human ASL deficiency will be required to understand the biochemical situation in the brain of affected patients, and to investigate the impact of high or low arginine doses on brain Cr availability. In addition, clinical trials to evaluate the beneficial effect of Cr supplementation in ASL deficiency would be valuable.


Asunto(s)
Ácido Argininosuccínico/toxicidad , Aciduria Argininosuccínica/patología , Aciduria Argininosuccínica/prevención & control , Encéfalo/patología , Creatina/farmacología , Síndromes de Neurotoxicidad/patología , Animales , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Humanos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología , Síndromes de Neurotoxicidad/metabolismo , Técnicas de Cultivo de Órganos/métodos , Ratas , Andamios del Tejido/química
8.
J Inherit Metab Dis ; 42(6): 1147-1161, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30723942

RESUMEN

The first patients affected by argininosuccinic aciduria (ASA) were reported 60 years ago. The clinical presentation was initially described as similar to other urea cycle defects, but increasing evidence has shown overtime an atypical systemic phenotype with a paradoxical observation, that is, a higher rate of neurological complications contrasting with a lower rate of hyperammonaemic episodes. The disappointing long-term clinical outcomes of many of the patients have challenged the current standard of care and therapeutic strategy, which aims to normalize plasma ammonia and arginine levels. Interrogations have raised about the benefit of newborn screening or liver transplantation on the neurological phenotype. Over the last decade, novel discoveries enabled by the generation of new transgenic argininosuccinate lyase (ASL)-deficient mouse models have been achieved, such as, a better understanding of ASL and its close interaction with nitric oxide metabolism, ASL physiological role outside the liver, and the pathophysiological role of oxidative/nitrosative stress or excessive arginine treatment. Here, we present a collaborative review, which highlights these recent discoveries and novel emerging concepts about ASL role in human physiology, ASA clinical phenotype and geographic prevalence, limits of current standard of care and newborn screening, pathophysiology of the disease, and emerging novel therapies. We propose recommendations for monitoring of ASA patients. Ongoing research aims to better understand the underlying pathogenic mechanisms of the systemic disease to design novel therapies.


Asunto(s)
Aciduria Argininosuccínica , Animales , Argininosuccinatoliasa/genética , Aciduria Argininosuccínica/diagnóstico , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/patología , Aciduria Argininosuccínica/terapia , Humanos , Hiperamonemia/diagnóstico , Hiperamonemia/genética , Hiperamonemia/terapia , Recién Nacido , Hígado/diagnóstico por imagen , Hígado/patología , Hígado/cirugía , Trasplante de Hígado , Ratones , Ratones Transgénicos , Tamizaje Neonatal/métodos , Tamizaje Neonatal/tendencias , Estrés Oxidativo/fisiología , Fenotipo
9.
J Inherit Metab Dis ; 40(3): 357-368, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28251416

RESUMEN

OBJECTIVES: This UK-wide study defines the natural history of argininosuccinic aciduria and compares long-term neurological outcomes in patients presenting clinically or treated prospectively from birth with ammonia-lowering drugs. METHODS: Retrospective analysis of medical records prior to March 2013, then prospective analysis until December 2015. Blinded review of brain MRIs. ASL genotyping. RESULTS: Fifty-six patients were defined as early-onset (n = 23) if symptomatic < 28 days of age, late-onset (n = 23) if symptomatic later, or selectively screened perinatally due to a familial proband (n = 10). The median follow-up was 12.4 years (range 0-53). Long-term outcomes in all groups showed a similar neurological phenotype including developmental delay (48/52), epilepsy (24/52), ataxia (9/52), myopathy-like symptoms (6/52) and abnormal neuroimaging (12/21). Neuroimaging findings included parenchymal infarcts (4/21), focal white matter hyperintensity (4/21), cortical or cerebral atrophy (4/21), nodular heterotopia (2/21) and reduced creatine levels in white matter (4/4). 4/21 adult patients went to mainstream school without the need of additional educational support and 1/21 lives independently. Early-onset patients had more severe involvement of visceral organs including liver, kidney and gut. All early-onset and half of late-onset patients presented with hyperammonaemia. Screened patients had normal ammonia at birth and received treatment preventing severe hyperammonaemia. ASL was sequenced (n = 19) and 20 mutations were found. Plasma argininosuccinate was higher in early-onset compared to late-onset patients. CONCLUSIONS: Our study further defines the natural history of argininosuccinic aciduria and genotype-phenotype correlations. The neurological phenotype does not correlate with the severity of hyperammonaemia and plasma argininosuccinic acid levels. The disturbance in nitric oxide synthesis may be a contributor to the neurological disease. Clinical trials providing nitric oxide to the brain merit consideration.


Asunto(s)
Aciduria Argininosuccínica/patología , Aciduria Argininosuccínica/terapia , Adolescente , Adulto , Amoníaco/metabolismo , Ácido Argininosuccínico/sangre , Aciduria Argininosuccínica/sangre , Aciduria Argininosuccínica/genética , Niño , Preescolar , Femenino , Estudios de Seguimiento , Genotipo , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/patología , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Mutación/genética , Fenotipo , Estudios Prospectivos , Estudios Retrospectivos , Adulto Joven
10.
J Leukoc Biol ; 97(2): 273-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25492936

RESUMEN

ASS1 is a cytosolic enzyme that plays a role in the conversion of citrulline to arginine. In human and mouse tissues, ASS1 protein is found in several components of the immune system, including the thymus and T cells. However, the role of ASS1 in these tissues remains to be defined. Considerable attention has been focused recently on the role of metabolism in T cell differentiation and function. Based on the expression of ASS1 in the immune system, we hypothesized that ASS1 deficiency would result in T cell defects. To evaluate this question, we characterized immune function in hypomorphic fold/fold mice. Analysis of splenic T cells by flow cytometry showed a marked reduction in T cell numbers with normal expression of activation surface markers. Gene therapy correction of liver ASS1 to enhance survival resulted in a partial recovery of splenic T cells for characterization. In vitro and in vivo studies demonstrated the persistence of the ASS1 enzyme defect in T cells and abnormal T cell differentiation and function. Overall, our work suggests that ASS1 plays a role in T cell function, and deficiency produces primary immune dysfunction. In addition, these data suggest that patients with ASS1 deficiency (citrullinemia type I) may have T cell dysfunction.


Asunto(s)
Aciduria Argininosuccínica/inmunología , Diferenciación Celular/inmunología , Activación de Linfocitos , Linfocitos T/inmunología , Animales , Argininosuccinatoliasa/genética , Argininosuccinatoliasa/inmunología , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/patología , Diferenciación Celular/genética , Humanos , Ratones , Ratones Transgénicos , Bazo/inmunología , Bazo/patología , Linfocitos T/patología
11.
J Biol Chem ; 288(48): 34599-611, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24136197

RESUMEN

Argininosuccinic aciduria (ASA) is an autosomal recessive urea cycle disorder caused by deficiency of argininosuccinate lyase (ASL) with a wide clinical spectrum from asymptomatic to severe hyperammonemic neonatal onset life-threatening courses. We investigated the role of ASL transcript variants in the clinical and biochemical variability of ASA. Recombinant proteins for ASL wild type, mutant p.E189G, and the frequently occurring transcript variants with exon 2 or 7 deletions were (co-)expressed in human embryonic kidney 293T cells. We found that exon 2-deleted ASL forms a stable truncated protein with no relevant activity but a dose-dependent dominant negative effect on enzymatic activity after co-expression with wild type or mutant ASL, whereas exon 7-deleted ASL is unstable but seems to have, nevertheless, a dominant negative effect on mutant ASL. These findings were supported by structural modeling predictions for ASL heterotetramer/homotetramer formation. Illustrating the physiological relevance, the predominant occurrence of exon 7-deleted ASL was found in two patients who were both heterozygous for the ASL mutant p.E189G. Our results suggest that ASL transcripts can contribute to the highly variable phenotype in ASA patients if expressed at high levels. Especially, the exon 2-deleted ASL variant may form a heterotetramer with wild type or mutant ASL, causing markedly reduced ASL activity.


Asunto(s)
Argininosuccinatoliasa/química , Argininosuccinatoliasa/genética , Aciduria Argininosuccínica/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Adulto , Argininosuccinatoliasa/metabolismo , Aciduria Argininosuccínica/enzimología , Aciduria Argininosuccínica/patología , Niño , Exones , Regulación de la Expresión Génica , Genotipo , Células HEK293 , Humanos , Masculino , Mutación , Fenotipo , Isoformas de Proteínas/metabolismo , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/genética
12.
Mol Genet Metab ; 108(3): 161-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23403242

RESUMEN

BACKGROUND: Urea cycle disorders (UCD) are caused by genetic defects in enzymes that constitute the hepatic ammonia detoxification pathway. Patients may present with variable clinical manifestations and with hyperammonaemia. Liver abnormalities have been associated with UCD, but only a few reports on the histopathological findings in the liver of UCD patients have been published. METHODS: We conducted a retrospective review of liver biopsies, ex-planted livers and livers at post-mortem of patients with UCD. A single pathologist reviewed all specimens. RESULTS: There were 18 liver samples from 13 patients with confirmed UCD: four ex-planted livers from patients with Ornithine Transcarbamylase (OTC) (n=3) and Carbamoyl Phosphate Synthetase 1 (CPS 1) (n=1) deficiencies, eight post-mortem samples from patients with CPS 1 (n=2), OTC (n=4), Argininosuccinate Synthetase (ASS) (n=1) and Argininosuccinate Lyase (ASL) (n=1) deficiencies, and six liver biopsies, three of which came from one patient with ASL deficiency. The other three liver biopsies were from patients who subsequently received liver transplantation. Histopathological findings in samples from neonates were non-specific. Samples from three late onset OTC deficient and one ASL deficient patients showed thin fibrous septa with portal to portal bridging fibrosis and focal marked enlargement and pallor of the hepatocytes due to accumulation of glycogen particles, resembling glycogenosis and resulting in a prominent nodular pattern. Serial liver biopsies in four UCD patients with interval between samples ranging from 1 year 2 months to 17 years showed progression in fibrosis in one OTC and one ASL deficient patients. Moderate fatty changes to no progression in liver disease were noted in the two patients (OTC=1 and CPS=1). A variety of non-specific features such as fatty change, mild inflammation, cholestasis and focal necrosis were seen in the other UCD patients. CONCLUSIONS: Histopathological changes in livers from neonates with UCD are non-specific. Older patients with UCD seem to show variable hepatic fibrosis compared to those who died early. Some of these patients also show focal and superficial resemblance to a glycogen storage disorder and cirrhosis. However, progression of these changes seems to be slow. To clarify the long term consequence of these changes, more extensive periods of follow up in a larger population series is needed.


Asunto(s)
Argininosuccinato Sintasa/deficiencia , Aciduria Argininosuccínica/patología , Carbamoil-Fosfato Sintasa (Amoniaco)/deficiencia , Hepatocitos/patología , Hiperamonemia/patología , Hígado/patología , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/patología , Aciduria Argininosuccínica/complicaciones , Aciduria Argininosuccínica/metabolismo , Autopsia , Biopsia , Niño , Preescolar , Ácidos Grasos/metabolismo , Femenino , Hepatocitos/metabolismo , Histocitoquímica , Humanos , Hiperamonemia/complicaciones , Hiperamonemia/metabolismo , Lactante , Recién Nacido , Hígado/metabolismo , Trasplante de Hígado , Masculino , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/complicaciones , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/metabolismo
13.
Spec Care Dentist ; 32(4): 155-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22784324

RESUMEN

An increased level of ammonia in the bloodstream, or hyperammonemia, is a symptom associated with metabolic disorders referred to as inborn errors of metabolism. Urea cycle disorder is a congenital abnormality or absence of one of the six enzymes involved in the elimination of ammonia. Administration of certain medications, high protein diet, excessive exercise, surgical procedures, or trauma can precipitate symptoms of mental confusion, seizure-like activity, and ataxia. This paper reviews the literature with insight into current treatment and management options of the disorder and modification of treatment for the dental patient.


Asunto(s)
Aciduria Argininosuccínica , Atención Dental para Enfermos Crónicos , Enfermedades de la Boca/etiología , Preparaciones Farmacéuticas Odontológicas/efectos adversos , Aciduria Argininosuccínica/complicaciones , Aciduria Argininosuccínica/enzimología , Aciduria Argininosuccínica/patología , Citocromo P-450 CYP3A/metabolismo , Interacciones Farmacológicas , Humanos , Hiperamonemia/complicaciones , Enfermedades de la Boca/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...