Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 80(4): 90, 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36922433

RESUMEN

Patients with liver cirrhosis show hyperammonemia and peripheral inflammation and may show hepatic encephalopathy with cognitive impairment, reproduced by rats with chronic hyperammonemia. Peripheral inflammation induces neuroinflammation in hippocampus of hyperammonemic rats, altering neurotransmission and leading to cognitive impairment. Extracellular vesicles (EVs) may transmit pathological effects from the periphery to the brain. We hypothesized that EVs from peripheral blood would contribute to cognitive alterations in hyperammonemic rats. The aims were to assess whether EVs from plasma of hyperammonemic rats (HA-EVs) induce cognitive impairment and to identify the underlying mechanisms. Injection of HA-EVs impaired learning and memory, induced microglia and astrocytes activation and increased TNFα and IL-1ß. Ex vivo incubation of hippocampal slices from control rats with HA-EVs reproduced these alterations. HA-EVs increased membrane expression of TNFR1, reduced membrane expression of TGFßR2 and Smad7 and IκBα levels and increased IκBα phosphorylation. This led to increased activation of NF-κB and IL-1ß production, altering membrane expression of NR2B, GluA1 and GluA2 subunits, which would be responsible for cognitive impairment. All these effects of HA-EVs were prevented by blocking TNFα, indicating that they were mediated by enhanced activation of TNFR1 by TNFα. We show that these mechanisms are very different from those leading to motor incoordination, which is due to altered GABAergic neurotransmission in cerebellum. This demonstrates that peripheral EVs play a key role in the transmission of peripheral alterations to the brain in hyperammonemia and hepatic encephalopathy, inducing neuroinflammation and altering neurotransmission in hippocampus, which in turn is responsible for the cognitive deficits.


Asunto(s)
Vesículas Extracelulares , Encefalopatía Hepática , Hiperamonemia , Ratas , Animales , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/farmacología , Enfermedades Neuroinflamatorias , Inhibidor NF-kappaB alfa/metabolismo , Inhibidor NF-kappaB alfa/farmacología , Encefalopatía Hepática/metabolismo , Encefalopatía Hepática/patología , Hiperamonemia/metabolismo , Hiperamonemia/patología , Inflamación/metabolismo , Cognición , Vesículas Extracelulares/metabolismo , Hipocampo/metabolismo
2.
Mol Genet Metab ; 132(1): 19-26, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33388234

RESUMEN

BACKGROUND/AIMS: Neonatal onset Urea cycle disorders (UCDs) can be life threatening with severe hyperammonemia and poor neurological outcomes. Glycerol phenylbutyrate (GPB) is safe and effective in reducing ammonia levels in patients with UCD above 2 months of age. This study assesses safety, ammonia control and pharmacokinetics (PK) of GPB in UCD patients below 2 months of age. METHODS: This was an open-label study in UCD patients aged 0 - 2 months, consisting of an initiation/transition period (1 - 4 days) to GPB, followed by a safety extension period (6 months to 2 years). Patients presenting with a hyperammonemic crisis (HAC) did not initiate GPB until blood ammonia levels decreased to below 100 µmol/L while receiving sodium phenylacetate/sodium benzoate and/or hemodialysis. Ammonia levels, PK analytes and safety were evaluated during transition and monthly during the safety extension for 6 months and every 3 months thereafter. RESULTS: All 16 patients with UCD (median age 0.48 months, range 0.1 to 2.0 months) successfully transitioned to GPB within 3 days. Average plasma ammonia level excluding HAC was 94.3 µmol/L at baseline and 50.4 µmol/L at the end of the transition period (p = 0.21). No patient had a HAC during the transition period. During the safety extension, the majority of patients had controlled ammonia levels, with mean plasma ammonia levels lower during GPB treatment than baseline. Mean glutamine levels remained within normal limits throughout the study. PK analyses indicate that UCD patients <2 months are able to hydrolyze GPB with subsequent absorption of phenylbutyric acid (PBA), metabolism to phenylacetic acid (PAA) and conjugation with glutamine. Plasma concentrations of PBA, PAA, and phenylacetylglutamine (PAGN) were stable during the safety extension phase and mean plasma phenylacetic acid: phenylacetylglutamine ratio remained below 2.5 suggesting no accumulation of GPB. All patients reported at least 1 treatment emergent adverse event with gastroesophageal reflux disease, vomiting, hyperammonemia, diaper dermatitis (37.5% each), diarrhea, upper respiratory tract infection and rash (31.3% each) being the most frequently reported. CONCLUSIONS: This study supports safety and efficacy of GPB in UCD patients aged 0 -2 months who cannot be managed by dietary protein restriction and/or amino acid supplementation alone. GPB undergoes intestinal hydrolysis with no accumulation in this population.


Asunto(s)
Glicerol/análogos & derivados , Hiperamonemia/tratamiento farmacológico , Fenilbutiratos/administración & dosificación , Trastornos Innatos del Ciclo de la Urea/tratamiento farmacológico , Edad de Inicio , Amoníaco/sangre , Preescolar , Femenino , Glicerol/administración & dosificación , Humanos , Hiperamonemia/sangre , Hiperamonemia/patología , Lactante , Recién Nacido , Masculino , Pediatría , Fenilacetatos/administración & dosificación , Diálisis Renal , Trastornos Innatos del Ciclo de la Urea/sangre , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/patología
3.
Mol Genet Metab ; 133(2): 182-184, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34020866

RESUMEN

Carnitine palmitoyl transferase II (CPT II) catalyzes the release of activated long-chain fatty acids from acylcarnitines into mitochondria for subsequent fatty acid oxidation. Depending on residual enzyme activity, deficiency of this enzyme leads to a spectrum of symptoms from early onset hypoglycemia, hyperammonemia, cardiomyopathy and death to onset of recurrent rhabdomyolysis in adolescents and young adults. We present a case of successful orthotopic heart transplantation in a patient with severe infantile onset cardiomyopathy due to CPT II deficiency identified through newborn screening. Excellent cardiac function is preserved 12 years post-transplantation; however, the patient has developed intermittent episodes of hyperammonemia and rhabdomyolysis later in childhood and early adolescence readily resolved with intravenous glucose. Successful heart transplant in this patient demonstrates the feasibility of this management option in patients with even severe forms of long chain fatty acid oxidation disorders.


Asunto(s)
Carnitina O-Palmitoiltransferasa/deficiencia , Carnitina O-Palmitoiltransferasa/genética , Trasplante de Corazón/métodos , Corazón/fisiopatología , Errores Innatos del Metabolismo/terapia , Adolescente , Adulto , Edad de Inicio , Cardiomiopatías/genética , Cardiomiopatías/patología , Cardiomiopatías/terapia , Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Humanos , Hiperamonemia/genética , Hiperamonemia/patología , Hiperamonemia/terapia , Hipoglucemia/genética , Hipoglucemia/patología , Hipoglucemia/terapia , Recién Nacido , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/metabolismo , Errores Innatos del Metabolismo/patología , Tamizaje Neonatal , Rabdomiólisis/genética , Rabdomiólisis/patología , Rabdomiólisis/terapia , Adulto Joven
4.
Mol Genet Metab ; 133(2): 148-156, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33846069

RESUMEN

BACKGROUND: Urea cycle disorders (UCDs) are among the most common inborn errors of liver metabolism. As therapies for hyperammonemia associated with urea cycle dysfunction have improved, chronic complications, such as liver disease, have become increasingly apparent in individuals with UCDs. Liver disease in UCDs may be associated with hepatic inflammation, hepatic fibrosis, portal hypertension, liver cancer and even liver failure. However, except for monitoring serum aminotransferases, there are no clear guidelines for screening and/or monitoring individuals with UCDs for liver disease. Thus, we systematically evaluated the potential utility of several non-invasive biomarkers for liver fibrosis in UCDs. METHODS: We evaluated grey-scale ultrasonography, liver stiffness obtained from shear wave elastography (SWE), and various serum biomarkers for hepatic fibrosis and necroinflammation, in a cohort of 28 children and adults with various UCDs. RESULTS: Overall, we demonstrate a high burden of liver disease in our participants with 46% of participants having abnormal grey-scale ultrasound pattern of the liver parenchyma, and 52% of individuals having increased liver stiffness. The analysis of serum biomarkers revealed that 32% of participants had elevated FibroTest™ score, a marker for hepatic fibrosis, and 25% of participants had increased ActiTest™ score, a marker for necroinflammation. Interestingly, liver stiffness did not correlate with ultrasound appearance or FibroTest™. CONCLUSION: Overall, our results demonstrate the high overall burden of liver disease in UCDs and highlights the need for further studies exploring new tools for identifying and monitoring individuals with UCDs who are at risk for this complication. TRIAL REGISTRATION: This study has been registered in ClinicalTrials.gov (NCT03721367).


Asunto(s)
Argininosuccinatoliasa/sangre , Enfermedades Genéticas Congénitas/sangre , Cirrosis Hepática/sangre , Hepatopatías/sangre , Trastornos Innatos del Ciclo de la Urea/sangre , Adolescente , Adulto , Biomarcadores/sangre , Niño , Preescolar , Diagnóstico por Imagen de Elasticidad , Femenino , Enfermedades Genéticas Congénitas/diagnóstico por imagen , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Humanos , Hiperamonemia/sangre , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/patología , Hígado/diagnóstico por imagen , Hígado/patología , Cirrosis Hepática/diagnóstico por imagen , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Hepatopatías/genética , Hepatopatías/metabolismo , Hepatopatías/patología , Masculino , Errores Innatos del Metabolismo/genética , Persona de Mediana Edad , Ultrasonografía , Trastornos Innatos del Ciclo de la Urea/genética , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/patología , Adulto Joven
5.
Am J Med Genet A ; 185(7): 2026-2036, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33851512

RESUMEN

Urea cycle disorders (UCDs) are inherited metabolic diseases that lead to hyperammonemia with variable clinical manifestations. Using data from a nationwide study, we investigated the onset time, gene variants, clinical manifestations, and treatment of patients with UCDs in Japan. Of the 229 patients with UCDs diagnosed and/or treated between January 2000 and March 2018, identified gene variants and clinical information were available for 102 patients, including 62 patients with ornithine transcarbamylase (OTC) deficiency, 18 patients with carbamoyl phosphate synthetase 1 (CPS1) deficiency, 16 patients with argininosuccinate synthetase (ASS) deficiency, and 6 patients with argininosuccinate lyase (ASL) deficiency. A total of 13, 10, 4, and 5 variants in the OTC, CPS1, ASS, and ASL genes were respectively identified as novel variants, which were neither registered in ClinVar databases nor previously reported. The onset time and severity in patients with UCD could be predicted based on the identified gene variants in each patient from this nationwide study and previous studies. This genetic information may help in predicting the long-term outcome and determining specific treatment strategies such as liver transplantation in patients with UCDs.


Asunto(s)
Argininosuccinatoliasa/genética , Argininosuccinato Sintasa/genética , Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Ornitina Carbamoiltransferasa/genética , Trastornos Innatos del Ciclo de la Urea/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Variación Genética/genética , Humanos , Hiperamonemia/enzimología , Hiperamonemia/genética , Hiperamonemia/patología , Lactante , Masculino , Enfermedades Metabólicas/enzimología , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/patología , Trastornos Innatos del Ciclo de la Urea/enzimología , Trastornos Innatos del Ciclo de la Urea/patología , Adulto Joven
6.
Am J Med Genet A ; 185(10): 2976-2985, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34155781

RESUMEN

Reduced muscle tone, muscle weakness, and physical fatigue can impact considerably on quality of life for children with neurofibromatosis type 1 (NF1). Human muscle biopsies and mouse models of NF1 deficiency in muscle show intramyocellular lipid accumulation, and preclinical data have indicated that L-carnitine supplementation can ameliorate this phenotype. The aim of this study is to examine whether daily L-carnitine supplementation is safe and feasible, and will improve muscle strength and reduce fatigue in children with NF1. A 12-week Phase 2a trial was conducted using 1000 mg daily oral levocarnitine tartrate supplementation. Recruited children were between 8 and 12 years old with a clinical diagnosis of NF1, history of muscle weakness and fatigue, and naïve to L-carnitine. Primary outcomes were safety (self-reporting, biochemical testing) and compliance. Secondary outcomes included plasma acylcarnitine profiles, functional measures (muscle strength, long jump, handwriting speed, 6-minute-walk test [6MWT]), and parent-reported questionnaires (PedsQL™, CBCL/6-18). Six children completed the trial with no self-reported adverse events. Biochemical tests for kidney and liver function were normal, and the average compliance was 95%. Plasma acylcarnitine levels were low, but within a range not clinically linked to carnitine deficiency. For strength measures, there was a mean 53% increase in dorsiflexion strength (95% confidence interval [CI] 8.89-60.75; p = 0.02) and mean 66% increase in plantarflexion strength (95% CI 12.99-134.1; p = 0.03). In terms of muscle performance, there was a mean 10% increase in long jump distance (95% CI 2.97-16.03; p = 0.01) and 6MWT distance (95% CI 5.88-75.45; p = 0.03). Comparison with the 1000 Norms Project data showed a significant improvement in Z-score for all of these measures. Parent reports showed no negative impact on quality of life, and the perceived benefits led to the majority of individuals remaining on L-carnitine after the study. Twelve weeks of L-carnitine supplementation is safe and feasible in children with NF1, and a Phase 3 trial should confirm the efficacy of treatment.


Asunto(s)
Carnitina/administración & dosificación , Fatiga/dietoterapia , Debilidad Muscular/dietoterapia , Neurofibromatosis 1/dietoterapia , Cardiomiopatías/dietoterapia , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Carnitina/efectos adversos , Carnitina/deficiencia , Carnitina/metabolismo , Niño , Suplementos Dietéticos/efectos adversos , Fatiga/genética , Fatiga/patología , Femenino , Humanos , Hiperamonemia/dietoterapia , Hiperamonemia/metabolismo , Hiperamonemia/patología , Masculino , Fuerza Muscular/efectos de los fármacos , Debilidad Muscular/metabolismo , Debilidad Muscular/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Enfermedades Musculares/dietoterapia , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/patología , Calidad de Vida
7.
Metab Brain Dis ; 36(7): 2169-2172, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34427841

RESUMEN

Biallelic pathogenic variants in the neuroblastoma amplified sequence (NBAS) gene were firstly (2015) identified as a cause of fever-triggered recurrent acute liver failure (RALF). Since then, some patients with NBAS deficiency presenting with neurologic features, including a motor delay, intellectual disability, muscular hypotonia and a mild brain atrophy, have been reported. Here, we describe a case of pediatric patient diagnosed with NBAS deficiency due to a homozygous c.2809C > G, p.(Pro937Ala) variant presenting with RALF with severe hyperammonemia, acquired microcephaly and progressive brain atrophy. Not reported in the literature findings include severe hyperammonemia during ALF episode, and neurologic features in the form of acquired progressive microcephaly with brain atrophy. The latter raises the hypothesis about a primary neurologic phenotype in NBAS deficiency.


Asunto(s)
Hiperamonemia , Fallo Hepático Agudo , Microcefalia , Neuroblastoma , Atrofia/genética , Atrofia/patología , Encéfalo/metabolismo , Niño , Humanos , Hiperamonemia/genética , Hiperamonemia/patología , Fallo Hepático Agudo/genética , Fallo Hepático Agudo/patología , Microcefalia/complicaciones , Microcefalia/diagnóstico por imagen , Microcefalia/genética , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
8.
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
9.
J Pediatr Hematol Oncol ; 42(2): e114-e116, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-30789458

RESUMEN

Idiopathic hyperammonemia is a rare, poorly understood, and often lethal condition that has been described in immunocompromised patients. This report describes an immunocompromised patient with acute myelogenous leukemia who developed persistent hyperammonemia up to 705 µmol/L (normal, 0 to 47 µmol/L) refractory to multiple different therapies. However, after beginning azithromycin and then doxycycline therapy for Ureaplasma species infection, the patient showed immediate and sustained clinical improvement and resolution of ammonia levels. Recognizing disseminated Ureaplasma species infection as a potential cause of idiopathic hyperammonemia, an unexplained, often fatal condition in immunocompromised patients, and empirically treating for this infection could potentially be lifesaving.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Hiperamonemia/etiología , Huésped Inmunocomprometido/efectos de los fármacos , Quimioterapia de Inducción/efectos adversos , Leucemia Mieloide Aguda/tratamiento farmacológico , Infecciones por Ureaplasma/complicaciones , Ureaplasma/efectos de los fármacos , Adolescente , Antibacterianos/uso terapéutico , Doxiciclina/uso terapéutico , Femenino , Humanos , Hiperamonemia/tratamiento farmacológico , Hiperamonemia/patología , Leucemia Mieloide Aguda/microbiología , Leucemia Mieloide Aguda/patología , Pronóstico , Infecciones por Ureaplasma/inducido químicamente , Infecciones por Ureaplasma/microbiología
10.
Blood Purif ; 49(3): 382-384, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31910419

RESUMEN

We describe a 9-year-old boy with acute liver failure of unknown etiology, unresponsive to standard medical therapy, with increasing hyperammonemia blood level, lactate elevation, a pediatric end liver stage of 20, a hepatic encephalopathy (HE) score of 2, and scheduled for emergent liver transplantation on the waiting list. We admitted him in the pediatric intensive care unit and managed him in the early stages with continuous renal replacement therapy and therapeutic plasma exchange as soon as neurologic impairment started to worsen. He recovered from his HE after 3 days of blood purification and was removed from the transplantation waiting list due to progressive liver function improvement.


Asunto(s)
Terapia de Reemplazo Renal Híbrido , Fallo Hepático Agudo/terapia , Hígado/patología , Niño , Encefalopatía Hepática/complicaciones , Encefalopatía Hepática/patología , Encefalopatía Hepática/terapia , Humanos , Terapia de Reemplazo Renal Híbrido/métodos , Hiperamonemia/complicaciones , Hiperamonemia/patología , Hiperamonemia/terapia , Fallo Hepático Agudo/complicaciones , Fallo Hepático Agudo/patología , Masculino , Intercambio Plasmático/métodos
11.
Metab Brain Dis ; 35(4): 559-578, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32146658

RESUMEN

Hepatic encephalopathy (HE) may occur in patients with liver failure. The most critical pathophysiologic mechanism of HE is cerebral edema following systemic hyperammonemia. The dysfunctional liver cannot eliminate circulatory ammonia, so its plasma and brain levels rise sharply. Astrocytes, the only cells that are responsible for ammonia detoxification in the brain, are dynamic cells with unique phenotypic properties that enable them to respond to small changes in their environment. Any pathological changes in astrocytes may cause neurological disturbances such as HE. Astrocyte swelling is the leading cause of cerebral edema, which may cause brain herniation and death by increasing intracranial pressure. Various factors may have a role in astrocyte swelling. However, the exact molecular mechanism of astrocyte swelling is not fully understood. This article discusses the possible mechanisms of astrocyte swelling which related to hyperammonia, including the possible roles of molecules like glutamine, lactate, aquaporin-4 water channel, 18 KDa translocator protein, glial fibrillary acidic protein, alanine, glutathione, toll-like receptor 4, epidermal growth factor receptor, glutamate, and manganese, as well as inflammation, oxidative stress, mitochondrial permeability transition, ATP depletion, and astrocyte senescence. All these agents and factors may be targeted in therapeutic approaches to HE.


Asunto(s)
Astrocitos/metabolismo , Edema Encefálico/metabolismo , Encefalopatía Hepática/metabolismo , Hiperamonemia/metabolismo , Amoníaco/metabolismo , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Edema Encefálico/patología , Tamaño de la Célula , Encefalopatía Hepática/patología , Humanos , Hiperamonemia/patología , Estrés Oxidativo/fisiología
12.
Am J Transplant ; 19(7): 2116-2121, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30868732

RESUMEN

Secondary protein-losing enteropathy (PLE) is a rare complication following pediatric liver transplantation (LT), mostly related to venous outflow obstruction of the liver. Here, we discuss a thus far unknown cause of secondary PLE following pediatric LT. A 7-month-old boy underwent LT with biliary anastomosis using a Roux-en-Y jejunal loop. Eleven months later he developed PLE. Routine diagnostic workup was negative. No hepatic outflow obstruction was detected during catheterization. Although the hepatic venous pressure gradient was slightly increased (10 mm Hg), there were no clinical signs of portal hypertension. Albumin scintigraphy with specific early recordings suggested focal albumin intestinal entry in the jejunal Roux-en-Y loop. Local bacterial overgrowth or local lymphangiectasia, possibly due to (venous) congestion, was considered. Treatment with metronidazole did not improve albumin loss. Next, surgical revision of the jejunal Roux-en-Y loop was performed. The explanted loop contained a small abnormal area with a thin hyperemic mucosa, near the former anastomosis. Histopathological analysis showed changes both in the blood vessels and the lymphatic vessels with focal deeper chronic active inflammation resulting in congestion of vessels, hampering lymphatic outflow leading to lymphangiectasia and patchy distortion of lymphatic vessels. Following surgical revision, secondary PLE disappeared, up to now, 1.5 year post revision.


Asunto(s)
Anastomosis en-Y de Roux/métodos , Encefalopatías/cirugía , Hiperamonemia/cirugía , Trasplante de Hígado/efectos adversos , Enteropatías Perdedoras de Proteínas/cirugía , Encefalopatías/patología , Humanos , Hiperamonemia/patología , Lactante , Masculino , Pronóstico , Enteropatías Perdedoras de Proteínas/etiología , Enteropatías Perdedoras de Proteínas/patología , Recuperación de la Función
13.
Liver Int ; 39(4): 676-683, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30394651

RESUMEN

BACKGROUND: Loss of muscle mass and strength is common in cirrhosis and increases the risk of hyperammonaemia and hepatic encephalopathy. Resistance training optimizes muscle mass and strength in several chronic diseases. However, the beneficial effects of resistance training in cirrhosis remain to be investigated. Bile duct-ligated (BDL) rats develop chronic liver disease, hyperammonaemia, reduced muscle mass and strength. Our aim was to test the effects of resistance training on muscle mass, function and ammonia metabolism in BDL-rats. METHODS: A group of BDL-rats underwent a progressive resistance training programme and a group of non-exercise BDL-rats served as controls. Resistance training comprised of ladder climbing with a progressive increase in carrying weights attached to the tail. Training was performed 5 days a week during 4 weeks. Muscle strength and body composition were assessed using grip strength and EchoMRI. Weight and circumference of the gastrocnemius muscle (normalized to bodyweight), plasma ammonia and glutamine synthetase protein expression and activity were assessed. RESULTS: BDL + exercise rats had significantly larger gastrocnemius circumference compared to non-exercise BDL-rats: ratio 0.082 vs 0.075 (P < 0.05). Gastrocnemius muscle weight was higher in exercisers than controls: 0.006 vs 0.005 (P < 0.05). A tendency towards a lower plasma ammonia in the exercise group compared to controls was observed (P = 0.10). There were no differences in lean body mass, GS protein expression and activity between the groups. CONCLUSION: Resistance training in rats with chronic liver disease beneficially effects muscle mass and strength. The effects were followed by non-significant reduction in blood ammonia; however, a tendency was observed.


Asunto(s)
Cirrosis Hepática Experimental/patología , Músculo Esquelético/patología , Condicionamiento Físico Animal/métodos , Entrenamiento de Fuerza , Amoníaco/sangre , Animales , Conductos Biliares/cirugía , Composición Corporal , Modelos Animales de Enfermedad , Encefalopatía Hepática , Hiperamonemia/etiología , Hiperamonemia/patología , Ligadura , Masculino , Proteínas Musculares/metabolismo , Ratas , Ratas Sprague-Dawley , Aumento de Peso
14.
Anticancer Drugs ; 30(3): 313-317, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30531368

RESUMEN

For several decades, 5-Fluorouracil (5-FU) has been the backbone of many chemotherapy regimens for various tumor types. Its most common side effects are gastrointestinal disorders, mucositis, myelosuppression, hand-foot syndrome, and rarely cardiac toxicity. More rarely, 5-FU infusion can induce hyperammonemic encephalopathy. 5-FU toxicities can be worsened by complete or partial genetic and/or phenotypic dihydropyrimidine dehydrogenase deficiency. Here, we report the case of a patient who initially developed a 5-FU-induced hyperammonemic encephalopathy after receiving FOLFIRINOX (oxaliplatin, irinotecan, folinic acid, and 5-FU) chemotherapy with bevacizumab to treat a metastatic gastrointestinal cancer of unknown primary. Thereafter, the patient was rechallenged successfully by the same chemotherapy regimen (FOLFIRINOX) for more than 6 months with a protocol consisting in a free protein diet, and administration of ammonium chelators, and Krebs and urea cycle intermediates, to prevent further hyperammonemia. We also present a review of the literature on 5-FU rechallenge after 5-FU-induced hyperammonemic encephalopathy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Encefalopatías/tratamiento farmacológico , Fluorouracilo/efectos adversos , Neoplasias Gastrointestinales/tratamiento farmacológico , Hiperamonemia/tratamiento farmacológico , Neoplasias Primarias Desconocidas/tratamiento farmacológico , Retratamiento/estadística & datos numéricos , Adulto , Bevacizumab/administración & dosificación , Encefalopatías/inducido químicamente , Encefalopatías/patología , Femenino , Fluorouracilo/administración & dosificación , Neoplasias Gastrointestinales/patología , Humanos , Hiperamonemia/inducido químicamente , Hiperamonemia/patología , Irinotecán/administración & dosificación , Leucovorina/administración & dosificación , Neoplasias Primarias Desconocidas/patología , Oxaliplatino/administración & dosificación , Pronóstico
15.
J Inherit Metab Dis ; 42(6): 1064-1076, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30714172

RESUMEN

The most common ureagenesis defect is X-linked ornithine transcarbamylase (OTC) deficiency which is a main target for novel therapeutic interventions. The spf ash mouse model carries a variant (c.386G>A, p.Arg129His) that is also found in patients. Male spf ash mice have a mild biochemical phenotype with low OTC activity (5%-10% of wild-type), resulting in elevated urinary orotic acid but no hyperammonemia. We recently established a dried blood spot method for in vivo quantification of ureagenesis by Gas chromatography-mass spectrometry (GC-MS) using stable isotopes. Here, we applied this assay to wild-type and spf ash mice to assess ureagenesis at different ages. Unexpectedly, we found an age-dependency with a higher capacity for ammonia detoxification in young mice after weaning. A parallel pattern was observed for carbamoylphosphate synthetase 1 and OTC enzyme expression and activities, which may act as pacemaker of this ammonia detoxification pathway. Moreover, high ureagenesis in younger mice was accompanied by elevated periportal expression of hepatic glutamine synthetase, another main enzyme required for ammonia detoxification. These observations led us to perform a more extensive analysis of the spf ash mouse in comparison to the wild-type, including characterization of the corresponding metabolites, enzyme activities in the liver and plasma and the gut microbiota. In conclusion, the comprehensive enzymatic and metabolic analysis of ureagenesis performed in the presented depth was only possible in animals. Our findings suggest such analyses being essential when using the mouse as a model and revealed age-dependent activity of ammonia detoxification.


Asunto(s)
Envejecimiento/fisiología , Amoníaco/metabolismo , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/metabolismo , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/patología , Ornitina Carbamoiltransferasa/genética , Urea/metabolismo , Factores de Edad , Animales , Modelos Animales de Enfermedad , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/patología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Transgénicos , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/genética
16.
J Inherit Metab Dis ; 42(6): 1097-1104, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30671986

RESUMEN

(Macro)autophagy/autophagy is a highly regulated lysosomal degradative process by which cells recycle their own nutrients, such as amino acids and other metabolites, to be reused in different biosynthetic pathways. Ammonia is a diffusible compound generated daily from catabolism of nitrogen-containing molecules and from gastrointestinal microbiome. Ammonia homeostasis is tightly controlled in humans and ammonia is efficiently converted by the healthy liver into non-toxic urea (through ureagenesis) and glutamine (through glutamine synthetase). Impaired ammonia detoxification leads to systemic hyperammonemia, a life-threatening condition resulting in detrimental effects on central nervous system. Here, we review current understanding on the role of ammonia in modulation of autophagy and the potential implications in the pathogenesis and treatment of disorders with hyperammonemia.


Asunto(s)
Amoníaco/metabolismo , Autofagia/fisiología , Hiperamonemia/etiología , Animales , Glutamato-Amoníaco Ligasa/metabolismo , Glutamina/metabolismo , Homeostasis , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/patología , Hígado/metabolismo , Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/complicaciones , Trastornos Innatos del Ciclo de la Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/patología
17.
J Inherit Metab Dis ; 42(6): 1128-1135, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30724386

RESUMEN

The urea cycle and glutamine synthetase (GS) are the two main pathways for waste nitrogen removal and their deficiency results in hyperammonemia. Here, we investigated the efficacy of liver-specific GS overexpression for therapy of hyperammonemia. To achieve hepatic GS overexpression, we generated a helper-dependent adenoviral (HDAd) vector expressing the murine GS under the control of a liver-specific expression cassette (HDAd-GS). Compared to mice injected with a control vector expressing an unrelated reporter gene (HDAd-alpha-fetoprotein), wild-type mice with increased hepatic GS showed reduced blood ammonia levels and a concomitant increase of blood glutamine after intraperitoneal injections of ammonium chloride, whereas blood urea was unaffected. Moreover, injection of HDAd-GS reduced blood ammonia levels at baseline and protected against acute hyperammonemia following ammonia challenge in a mouse model with conditional hepatic deficiency of carbamoyl phosphate synthetase 1 (Cps1), the initial and rate-limiting step of ureagenesis. In summary, we found that upregulation of hepatic GS reduced hyperammonemia in wild-type and Cps1-deficient mice, thus confirming a key role of GS in ammonia detoxification. These results suggest that hepatic GS augmentation therapy has potential for treatment of both primary and secondary forms of hyperammonemia.


Asunto(s)
Amoníaco/metabolismo , Terapia Genética/métodos , Glutamato-Amoníaco Ligasa/genética , Hiperamonemia/genética , Hiperamonemia/terapia , Hígado/metabolismo , Amoníaco/toxicidad , Animales , Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/genética , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/metabolismo , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/terapia , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Glutamato-Amoníaco Ligasa/metabolismo , Hiperamonemia/metabolismo , Hiperamonemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos/genética
18.
Klin Padiatr ; 231(2): 74-79, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30870873

RESUMEN

BACKGROUND: Hyperammonemic encephalopathy in newborns with urea cycle disorders and certain organic acidurias can cause severe brain injury, coma and death. Standard therapy includes protein restriction, nitrogen-scavenging drugs, prevention of catabolism and hemodialysis. Neuroprotective hypothermia as part of the treatment has been reported only 3 times. It has been suggested that mild systemic hypothermia can contribute to better neurological outcomes in hyperammonemic encephalopathy. However, the limited experience precludes accurate conclusions on safety and efficacy. METHODS: Whole body therapeutic hypothermia was included in the standard treatment of hyperammonemic encephalopathy in 4 neonates with urea cycle disorder or organic aciduria. RESULTS: Two patients survived the initial crisis. One patient has a developmental quotient of 0.8, while the other shows severe developmental delay. The cooling protocol had to be discontinued in 3 patients due to the otherwise untreatable complications (hypotension and hemorrhage). CONCLUSION: The efficacy and safety of therapeutic hypothermia in the treatment of neonatal hyperammonemic encephalopathy depend on various factors, requiring further evaluation.


Asunto(s)
Hiperamonemia/terapia , Hipotermia Inducida/métodos , Hipoxia-Isquemia Encefálica/terapia , Trastornos Innatos del Ciclo de la Urea/terapia , Urea/metabolismo , Humanos , Hiperamonemia/patología , Hipoxia-Isquemia Encefálica/complicaciones , Recién Nacido , Resultado del Tratamiento , Trastornos Innatos del Ciclo de la Urea/complicaciones , Trastornos Innatos del Ciclo de la Urea/genética , Trastornos Innatos del Ciclo de la Urea/patología
19.
Int J Mol Sci ; 20(21)2019 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-31653080

RESUMEN

Lysinuric protein intolerance (LPI) is a rare autosomal disease caused by defective cationic amino acid (CAA) transport due to mutations in SLC7A7, which encodes for the y+LAT1 transporter. LPI patients suffer from a wide variety of symptoms, which range from failure to thrive, hyperammonemia, and nephropathy to pulmonar alveolar proteinosis (PAP), a potentially life-threatening complication. Hyperammonemia is currently prevented by citrulline supplementation. However, the full impact of this treatment is not completely understood. In contrast, there is no defined therapy for the multiple reported complications of LPI, including PAP, for which bronchoalveolar lavages do not prevent progression of the disease. The lack of a viable LPI model prompted us to generate a tamoxifen-inducible Slc7a7 knockout mouse (Slc7a7-/-). The Slc7a7-/- model resembles the human LPI phenotype, including malabsorption and impaired reabsorption of CAA, hypoargininemia and hyperammonemia. Interestingly, the Slc7a7-/- mice also develops PAP and neurological impairment. We observed that citrulline treatment improves the metabolic derangement and survival. On the basis of our findings, the Slc7a7-/- model emerges as a promising tool to further study the complexity of LPI, including its immune-like complications, and to design evidence-based therapies to halt its progression.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/patología , Sistema de Transporte de Aminoácidos y+L/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Sistema de Transporte de Aminoácidos y+L/genética , Aminoácidos/metabolismo , Animales , Citrulina/uso terapéutico , Modelos Animales de Enfermedad , Hiperamonemia/metabolismo , Hiperamonemia/patología , Mucosa Intestinal/metabolismo , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteinosis Alveolar Pulmonar/metabolismo , Proteinosis Alveolar Pulmonar/patología
20.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 286-295, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29107806

RESUMEN

Hyperammonemia contributes to altered neurotransmission and cognition in patients with hepatic encephalopathy. Hyperammonemia in rats affects differently high- and low-affinity AMPA receptors (AMPARs) in cerebellum. We hypothesized that hyperammonemia would alter differently membrane expression of AMPARs GluA1 and GluA2 subunits by altering its phosphorylation. This work aims were: 1) assess if hyperammonemia alters GluA1 and GluA2 subunits membrane expression in cerebellum and 2) analyze the underlying mechanisms. Hyperammonemia reduces membrane expression of GluA2 and enhances membrane expression of GluA1 in vivo. We show that changes in GluA2 and GluA1 membrane expression in hyperammonemia would be due to enhanced NMDA receptors activation which reduces cGMP levels and phosphodiesterase 2 (PDE2) activity, resulting in increased cAMP levels. This leads to increased protein kinase A (PKA) activity which activates phospholipase C (PLC) and protein kinase C (PKC) thus increasing phosphorylation of GluA2 in Ser880, which reduces GluA2 membrane expression, and phosphorylation of GluA1 in Ser831, which increases GluA1 membrane expression. Blocking NMDA receptors or inhibiting PKA, PLC or PKC normalizes GluA2 and GluA1 phosphorylation and membrane expression in hyperammonemic rats. Altered GluA2 and GluA1 membrane expression would alter signal transduction which may contribute to cognitive and motor alterations in hyperammonemia and hepatic encephalopathy.


Asunto(s)
Membrana Celular/metabolismo , Hiperamonemia/genética , Receptores AMPA/genética , Animales , Membrana Celular/patología , Enfermedad Crónica , Encefalopatía Hepática/genética , Encefalopatía Hepática/metabolismo , Encefalopatía Hepática/patología , Hiperamonemia/metabolismo , Hiperamonemia/patología , Masculino , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Wistar , Receptores AMPA/metabolismo , Transducción de Señal/genética , Transmisión Sináptica/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA