Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Inherit Metab Dis ; 44(4): 847-856, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33325055

RESUMEN

Hyperargininemia in patients with arginase 1 deficiency (ARG1-D) is considered a key driver of disease manifestations, including spasticity, developmental delay, and seizures. Pegzilarginase (AEB1102) is an investigational enzyme therapy which is being developed as a novel arginine lowering approach. We report the safety and efficacy of intravenously (IV) administered pegzilarginase in pediatric and adult ARG1-D patients (n = 16) from a Phase 1/2 study (101A) and the first 12 weeks of an open-label extension study (102A). Substantial disease burden at baseline included lower-limb spasticity, developmental delay, and previous hyperammonemic episodes in 75%, 56%, and 44% of patients, respectively. Baseline plasma arginine (pArg) was elevated (median 389 µM, range 238-566) on standard disease management. Once weekly repeat dosing resulted in a median decrease of pArg of 277 µM after 20 cumulative doses (n = 14) with pArg in the normal range (40 to 115 µM) in 50% of patients at 168 hours post dose (mean pegzilarginase dose 0.10 mg/kg). Lowering pArg was accompanied by improvements in one or more key mobility assessments (6MWT, GMFM-D & E) in 79% of patients. In 101A, seven hypersensitivity reactions occurred in four patients (out of 162 infusions administered). Other common treatment-related adverse events (AEs) included vomiting, hyperammonemia, pruritus, and abdominal pain. Treatment-related serious AEs that occurred in five patients were all observed in 101A. Pegzilarginase was effective in lowering pArg levels with an accompanying clinical response in patients with ARG1-D. The improvements with pegzilarginase occurred in patients receiving standard treatment approaches, which suggests that pegzilarginase could offer benefit over existing disease management.


Asunto(s)
Arginasa/genética , Arginasa/uso terapéutico , Arginina/sangre , Hiperargininemia/tratamiento farmacológico , Adolescente , Adulto , Arginasa/efectos adversos , Arginasa/sangre , Arginina/metabolismo , Niño , Preescolar , Manejo de la Enfermedad , Femenino , Humanos , Hiperamonemia/etiología , Hiperargininemia/sangre , Hiperargininemia/genética , Hiperargininemia/metabolismo , Masculino , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Estados Unidos , Vómitos/etiología , Adulto Joven
2.
Int J Mol Sci ; 21(20)2020 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-33053818

RESUMEN

Arginine is one of the most important nutrients of living organisms as it plays a major role in important biological pathways. However, the accumulation of arginine as consequence of metabolic defects causes hyperargininemia, an autosomal recessive disorder. Therefore, the efficient detection of the arginine is a field of relevant biomedical/biotechnological interest. Here, we developed protein variants suitable for arginine sensing by mutating and dissecting the multimeric and multidomain structure of Thermotoga maritima arginine-binding protein (TmArgBP). Indeed, previous studies have shown that TmArgBP domain-swapped structure can be manipulated to generate simplified monomeric and single domain scaffolds. On both these stable scaffolds, to measure tryptophan fluorescence variations associated with the arginine binding, a Phe residue of the ligand binding pocket was mutated to Trp. Upon arginine binding, both mutants displayed a clear variation of the Trp fluorescence. Notably, the single domain scaffold variant exhibited a good affinity (~3 µM) for the ligand. Moreover, the arginine binding to this variant could be easily reverted under very mild conditions. Atomic-level data on the recognition process between the scaffold and the arginine were obtained through the determination of the crystal structure of the adduct. Collectively, present data indicate that TmArgBP scaffolds represent promising candidates for developing arginine biosensors.


Asunto(s)
Arginina/química , Arginina/metabolismo , Fenómenos Fisiológicos Bacterianos , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Thermotoga maritima/metabolismo , Proteínas Portadoras/genética , Hiperargininemia/diagnóstico , Hiperargininemia/etiología , Hiperargininemia/metabolismo , Ligandos , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad , Thermotoga maritima/genética
3.
J Clin Invest ; 130(11): 5703-5720, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32721946

RESUMEN

Arginase 1 (Arg1), which converts l-arginine into ornithine and urea, exerts pleiotropic immunoregulatory effects. However, the function of Arg1 in inflammatory bowel disease (IBD) remains poorly characterized. Here, we found that Arg1 expression correlated with the degree of inflammation in intestinal tissues from IBD patients. In mice, Arg1 was upregulated in an IL-4/IL-13- and intestinal microbiota-dependent manner. Tie2-Cre Arg1fl/fl mice lacking Arg1 in hematopoietic and endothelial cells recovered faster from colitis than Arg1-expressing (Arg1fl/fl) littermates. This correlated with decreased vessel density, compositional changes in intestinal microbiota, diminished infiltration by myeloid cells, and an accumulation of intraluminal polyamines that promote epithelial healing. The proresolving effect of Arg1 deletion was reduced by an l-arginine-free diet, but rescued by simultaneous deletion of other l-arginine-metabolizing enzymes, such as Arg2 or Nos2, demonstrating that protection from colitis requires l-arginine. Fecal microbiota transfers from Tie2-Cre Arg1fl/fl mice into WT recipients ameliorated intestinal inflammation, while transfers from WT littermates into Arg1-deficient mice prevented an advanced recovery from colitis. Thus, an increased availability of l-arginine as well as altered intestinal microbiota and metabolic products accounts for the accelerated resolution from colitis in the absence of Arg1. Consequently, l-arginine metabolism may serve as a target for clinical intervention in IBD patients.


Asunto(s)
Arginasa/metabolismo , Microbioma Gastrointestinal , Hiperargininemia , Enfermedades Inflamatorias del Intestino , Metaboloma , Animales , Arginasa/genética , Arginina/genética , Arginina/metabolismo , Células Endoteliales/enzimología , Células Endoteliales/patología , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/patología , Hiperargininemia/genética , Hiperargininemia/metabolismo , Hiperargininemia/microbiología , Hiperargininemia/patología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Ratones , Ratones Noqueados
4.
J Genet Genomics ; 47(3): 145-156, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32305173

RESUMEN

Arginine catabolism involves enzyme-dependent reactions in both mitochondria and the cytosol, defects in which may lead to hyperargininemia, a devastating developmental disorder. It is largely unknown if defective arginine catabolism has any effects on mitochondria. Here we report that normal arginine catabolism is essential for mitochondrial homeostasis in Caenorhabditiselegans. Mutations of the arginase gene argn-1 lead to abnormal mitochondrial enlargement and reduced adenosine triphosphate (ATP) production in C. elegans hypodermal cells. ARGN-1 localizes to mitochondria and its loss causes arginine accumulation, which disrupts mitochondrial dynamics. Heterologous expression of human ARG1 or ARG2 rescued the mitochondrial defects of argn-1 mutants. Importantly, genetic inactivation of the mitochondrial basic amino acid transporter SLC-25A29 or the mitochondrial glutamate transporter SLC-25A18.1 fully suppressed the mitochondrial defects caused by argn-1 mutations. These findings suggest that mitochondrial damage probably contributes to the pathogenesis of hyperargininemia and provide clues for developing therapeutic treatments for hyperargininemia.


Asunto(s)
Arginasa/genética , Arginina/metabolismo , Mitocondrias/genética , Adenosina Trifosfato/genética , Animales , Arginina/genética , Caenorhabditis elegans/genética , Citosol/enzimología , Modelos Animales de Enfermedad , Homeostasis/genética , Humanos , Hiperargininemia/genética , Hiperargininemia/metabolismo , Mutación
5.
JCI Insight ; 4(17)2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31484827

RESUMEN

Deficiency of arginase is associated with hyperargininemia, and prominent features include spastic diplegia/tetraplegia, clonus, and hyperreflexia; loss of ambulation, intellectual disability and progressive neurological decline are other signs. To gain greater insight into the unique neuromotor features, we performed gene expression profiling of the motor cortex of a murine model of the disorder. Coexpression network analysis suggested an abnormality with myelination, which was supported by limited existing human data. Utilizing electron microscopy, marked dysmyelination was detected in 2-week-old homozygous Arg1-KO mice. The corticospinal tract was found to be adversely affected, supporting dysmyelination as the cause of the unique neuromotor features and implicating oligodendrocyte impairment in a deficiency of hepatic Arg1. Following neonatal hepatic gene therapy to express Arg1, the subcortical white matter, pyramidal tract, and corticospinal tract all showed a remarkable recovery in terms of myelinated axon density and ultrastructural integrity with active wrapping of axons by nearby oligodendrocyte processes. These findings support the following conclusions: arginase deficiency is a leukodystrophy affecting the brain and spinal cord while sparing the peripheral nervous system, and neonatal AAV hepatic gene therapy can rescue the defects associated with myelinated axons, strongly implicating the functional recovery of oligodendrocytes after restoration of hepatic arginase activity.


Asunto(s)
Arginasa/genética , Predisposición Genética a la Enfermedad/genética , Hiperargininemia/genética , Hiperargininemia/metabolismo , Hígado/enzimología , Hígado/metabolismo , Animales , Arginasa/metabolismo , Axones/metabolismo , Axones/patología , Sistema Nervioso Central/diagnóstico por imagen , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Terapia Genética , Homocigoto , Hiperargininemia/patología , Masculino , Ratones , Ratones Noqueados , Oligodendroglía/metabolismo , Transcriptoma
6.
Proc Natl Acad Sci U S A ; 116(42): 21150-21159, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31501335

RESUMEN

Arginase deficiency is caused by biallelic mutations in arginase 1 (ARG1), the final step of the urea cycle, and results biochemically in hyperargininemia and the presence of guanidino compounds, while it is clinically notable for developmental delays, spastic diplegia, psychomotor function loss, and (uncommonly) death. There is currently no completely effective medical treatment available. While preclinical strategies have been demonstrated, disadvantages with viral-based episomal-expressing gene therapy vectors include the risk of insertional mutagenesis and limited efficacy due to hepatocellular division. Recent advances in messenger RNA (mRNA) codon optimization, synthesis, and encapsulation within biodegradable liver-targeted lipid nanoparticles (LNPs) have potentially enabled a new generation of safer, albeit temporary, treatments to restore liver metabolic function in patients with urea cycle disorders, including ARG1 deficiency. In this study, we applied such technologies to successfully treat an ARG1-deficient murine model. Mice were administered LNPs encapsulating human codon-optimized ARG1 mRNA every 3 d. Mice demonstrated 100% survival with no signs of hyperammonemia or weight loss to beyond 11 wk, compared with controls that perished by day 22. Plasma ammonia, arginine, and glutamine demonstrated good control without elevation of guanidinoacetic acid, a guanidino compound. Evidence of urea cycle activity restoration was demonstrated by the ability to fully metabolize an ammonium challenge and by achieving near-normal ureagenesis; liver arginase activity achieved 54% of wild type. Biochemical and microscopic data showed no evidence of hepatotoxicity. These results suggest that delivery of ARG1 mRNA by liver-targeted nanoparticles may be a viable gene-based therapeutic for the treatment of arginase deficiency.


Asunto(s)
Hiperargininemia/tratamiento farmacológico , Lípidos/farmacología , Hepatopatías/tratamiento farmacológico , Hígado/efectos de los fármacos , Nanopartículas/administración & dosificación , ARN Mensajero/metabolismo , Amoníaco/metabolismo , Animales , Arginasa/metabolismo , Arginina/metabolismo , Codón/metabolismo , Modelos Animales de Enfermedad , Glutamina/metabolismo , Hiperamonemia/tratamiento farmacológico , Hiperamonemia/metabolismo , Hiperargininemia/metabolismo , Hígado/metabolismo , Hepatopatías/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Urea/metabolismo
7.
J Biochem Mol Toxicol ; 31(1): 1-7, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27489181

RESUMEN

We investigated the in vitro and in vivo effects of arginine (Arg) on thiobarbituric acid-reactive substances (TBA-RS) and on the activities of catalase (CAT), glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) in renal tissues of rats. We also studied the influence of antioxidants (α-tocopherol plus ascorbic acid) and nitric oxide synthase inhibitor NG -nitro-l-arginine methyl ester (l-NAME) on the effects elicited by Arg. Results showed that Arg in vitro (1.5 mM) decreased SOD activity and increased the levels of TBA-RS in the renal medulla. Acute administration of Arg [0.8 g/kg, intraperitoneal injection] decreased CAT activity, increased SOD activity and TBA-RS levels in the renal medulla, and decreased CAT activity in the renal cortex of rats. Most results were prevented by antioxidants and/or l-NAME. Data indicate that Arg causes an oxidative imbalance in the renal tissues studied; however, in the presence of antioxidants and l-NAME, some of these alterations in oxidative stress were prevented.


Asunto(s)
Antioxidantes/farmacología , Hiperargininemia/prevención & control , Riñón/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Estrés Oxidativo/efectos de los fármacos , alfa-Tocoferol/farmacología , Animales , Catalasa/metabolismo , Glutatión Peroxidasa/metabolismo , Hiperargininemia/inducido químicamente , Hiperargininemia/metabolismo , Masculino , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo
8.
J Mol Med (Berl) ; 93(12): 1287-96, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26467175

RESUMEN

Arginase-1 (ARG1) deficiency is a rare autosomal recessive disorder that affects the liver-based urea cycle, leading to impaired ureagenesis. This genetic disorder is caused by 40+ mutations found fairly uniformly spread throughout the ARG1 gene, resulting in partial or complete loss of enzyme function, which catalyzes the hydrolysis of arginine to ornithine and urea. ARG1-deficient patients exhibit hyperargininemia with spastic paraparesis, progressive neurological and intellectual impairment, persistent growth retardation, and infrequent episodes of hyperammonemia, a clinical pattern that differs strikingly from other urea cycle disorders. This review briefly highlights the current understanding of the etiology and pathophysiology of ARG1 deficiency derived from clinical case reports and therapeutic strategies stretching over several decades and reports on several exciting new developments regarding the pathophysiology of the disorder using ARG1 global and inducible knockout mouse models. Gene transfer studies in these mice are revealing potential therapeutic options that can be exploited in the future. However, caution is advised in extrapolating results since the lethal disease phenotype in mice is much more severe than in humans indicating that the mouse models may not precisely recapitulate human disease etiology. Finally, some of the functions and implications of ARG1 in non-urea cycle activities are considered. Lingering questions and future areas to be addressed relating to the clinical manifestations of ARG1 deficiency in liver and brain are also presented. Hopefully, this review will spark invigorated research efforts that lead to treatments with better clinical outcomes.


Asunto(s)
Hiperargininemia/genética , Hiperargininemia/metabolismo , Animales , Arginasa/química , Arginasa/genética , Arginasa/metabolismo , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Humanos , Hiperargininemia/diagnóstico , Hiperargininemia/terapia , Hígado/metabolismo , Mutación , Fenotipo , Urea/metabolismo , Trastornos Innatos del Ciclo de la Urea/genética , Trastornos Innatos del Ciclo de la Urea/metabolismo
9.
Hum Mol Genet ; 24(22): 6417-27, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26358771

RESUMEN

Arginase deficiency is caused by deficiency of arginase 1 (ARG1), a urea cycle enzyme that converts arginine to ornithine. Clinical features of arginase deficiency include elevated plasma arginine levels, spastic diplegia, intellectual disability, seizures and growth deficiency. Unlike other urea cycle disorders, recurrent hyperammonemia is typically less severe in this disorder. Normalization of plasma arginine levels is the consensus treatment goal, because elevations of arginine and its metabolites are suspected to contribute to the neurologic features. Using data from patients enrolled in a natural history study conducted by the Urea Cycle Disorders Consortium, we found that 97% of plasma arginine levels in subjects with arginase deficiency were above the normal range despite conventional treatment. Recently, arginine-degrading enzymes have been used to deplete arginine as a therapeutic strategy in cancer. We tested whether one of these enzymes, a pegylated human recombinant arginase 1 (AEB1102), reduces plasma arginine in murine models of arginase deficiency. In neonatal and adult mice with arginase deficiency, AEB1102 reduced the plasma arginine after single and repeated doses. However, survival did not improve likely, because this pegylated enzyme does not enter hepatocytes and does not improve hyperammonemia that accounts for lethality. Although murine models required dosing every 48 h, studies in cynomolgus monkeys indicate that less frequent dosing may be possible in patients. Given that elevated plasma arginine rather than hyperammonemia is the major treatment challenge, we propose that AEB1102 may have therapeutic potential as an arginine-reducing agent in patients with arginase deficiency.


Asunto(s)
Arginasa/uso terapéutico , Arginina/sangre , Hiperargininemia/tratamiento farmacológico , Animales , Arginasa/sangre , Arginasa/genética , Arginina/metabolismo , Encéfalo/metabolismo , Niño , Preescolar , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Humanos , Hiperamonemia/sangre , Hiperamonemia/metabolismo , Hiperargininemia/sangre , Hiperargininemia/genética , Hiperargininemia/metabolismo , Estudios Longitudinales , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/uso terapéutico , Convulsiones/sangre , Convulsiones/metabolismo
10.
Amino Acids ; 47(9): 1751-62, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26123990

RESUMEN

Hyperargininemia is caused by deficiency of arginase 1, which catalyzes the hydrolysis of L-arginine to urea as the final enzyme in the urea cycle. In contrast to other urea cycle defects, arginase 1 deficiency usually does not cause catastrophic neonatal hyperammonemia but rather presents with progressive neurological symptoms including seizures and spastic paraplegia in the first years of life and hepatic pathology, such as neonatal cholestasis, acute liver failure, or liver fibrosis. Some patients have developed hepatocellular carcinoma. A usually mild or moderate hyperammonemia may occur at any age. The pathogenesis of arginase I deficiency is yet not fully understood. However, the accumulation of L-arginine and the resulting abnormalities in the metabolism of guanidine compounds and nitric oxide have been proposed to play a major pathophysiological role. This article provides an update on the first patients ever described, gives an overview of the distinct clinical characteristics, biochemical as well as genetical background and discusses treatment options.


Asunto(s)
Arginasa , Arginina/metabolismo , Hiperargininemia , Arginina/genética , Preescolar , Femenino , Guanidina/metabolismo , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/patología , Hiperamonemia/fisiopatología , Hiperargininemia/genética , Hiperargininemia/metabolismo , Hiperargininemia/patología , Hiperargininemia/fisiopatología , Lactante , Hepatopatías/genética , Hepatopatías/metabolismo , Hepatopatías/patología , Hepatopatías/fisiopatología , Paraplejía/genética , Paraplejía/metabolismo , Paraplejía/patología , Paraplejía/fisiopatología , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/patología , Convulsiones/fisiopatología
11.
J Hepatol ; 62(2): 412-20, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25234945

RESUMEN

BACKGROUND & AIMS: Innate immune activation has been postulated as a central mechanism for disease progression from hepatic steatosis to steatohepatitis in obesity-related fatty liver disease. Arginase 2 competes with inducible nitric oxide synthase (iNOS) for its substrate and the balance between these two enzymes plays a crucial role in regulating immune responses and macrophage activation. Our aim was to test the hypothesis that arginase 2 deficiency in mice favours progression from isolated hepatic steatosis, induced by high fat feeding, to steatohepatitis. METHODS: Arginase 2-knockout (Arg2(-/-)) mice were studied for changes in liver histology and metabolic phenotype at baseline and after a short term course (7 week) feeding with a high fat (HFAT) diet. In additional experiments, Arg2(-/-) mice received tail vein injections of liposome-encapsulated clodronate (CLOD) over a three-week period to selectively deplete liver macrophages. RESULTS: Unexpectedly, Arg2(-/-) mice showed profound changes in their livers at baseline, characterized by significant steatosis as demonstrated with histological and biochemical analysis. These changes were independent of systemic metabolic parameters and associated with marked mRNA level increases of genes involved in hepatic de novo lipogenesis. Liver injury and inflammation were present with elevated serum ALT, marked infiltration of F4/80 positive cells, and increased mRNA levels of inflammatory genes. HFAT feeding exacerbated these changes. Macrophage depletion after CLOD injection significantly attenuated lipid deposition and normalized lipogenic mRNA profile of livers from Arg2(-/-) mice. CONCLUSIONS: This study identifies arginase 2 as a novel link between innate immune responses, hepatic lipid deposition, and liver injury.


Asunto(s)
Arginasa/metabolismo , Hígado Graso/inmunología , Hiperargininemia/complicaciones , Inmunidad Innata , Macrófagos del Hígado/inmunología , Metabolismo de los Lípidos , Lipogénesis/inmunología , Animales , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/metabolismo , Hiperargininemia/inmunología , Hiperargininemia/metabolismo , Immunoblotting , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL
12.
Cell Biochem Funct ; 32(6): 511-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25042906

RESUMEN

In the present investigation, we initially evaluated the in vitro effect of N-acetylarginine on thiobarbituric acid-reactive substances (TBA-RS), total sulfhydryl content and on the activities of antioxidant enzymes such as catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) in the blood, kidney and liver of rats. Results showed that N-acetylarginine, at a concentration of 5.0 µM, decreased the activity of CAT in erythrocytes, enhanced TBA-RS in the renal cortex, decreased CAT and SOD activities in the renal medulla and decreased CAT and increased SOD and GSH-Px activities in the liver of 60-day-old rats. Furthermore, we tested the influence of the antioxidants, trolox and ascorbic acid, as well as of the N(ω) -nitro-L-arginine methyl ester (L-NAME) on the effects elicited by N-acetylarginine on the parameters tested. Antioxidants and L-NAME prevented most of the alterations caused by N-acetylarginine on the oxidative stress parameters evaluated. Data indicate that oxidative stress induction is probably mediated by the generation of NO and/or ONOO(-) and other free radicals because L-NAME and antioxidants prevented the effects caused by N-acetylarginine in the blood, renal tissues and liver of rats. Our findings lend support to a potential therapeutic strategy for this condition, which may include the use of appropriate antioxidants for ameliorating the damage caused by N-acetylarginine.


Asunto(s)
Arginina/análogos & derivados , Hiperargininemia/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Vitaminas/farmacología , Animales , Antioxidantes/farmacología , Arginina/metabolismo , Arginina/farmacología , Ácido Ascórbico/farmacología , Cromanos/farmacología , Corteza Renal/metabolismo , Médula Renal/metabolismo , Hígado/metabolismo , Ratas Wistar , Compuestos de Sulfhidrilo/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , alfa-Tocoferol/farmacología
13.
Mol Ther ; 22(10): 1792-802, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24888478

RESUMEN

Human arginase deficiency is characterized by hyperargininemia and infrequent episodes of hyperammonemia that cause neurological impairment and growth retardation. We previously developed a neonatal mouse adeno-associated viral vector (AAV) rh10-mediated therapeutic approach with arginase expressed by a chicken ß-actin promoter that controlled plasma ammonia and arginine, but hepatic arginase declined rapidly. This study tested a codon-optimized arginase cDNA and compared the chicken ß-actin promoter to liver- and muscle-specific promoters. ARG1(-/-) mice treated with AAVrh10 carrying the liver-specific promoter also exhibited long-term survival and declining hepatic arginase accompanied by the loss of AAV episomes during subsequent liver growth. Although arginase expression in striated muscle was not expected to counteract hyperammonemia, due to muscle's lack of other urea cycle enzymes, we hypothesized that the postmitotic phenotype in muscle would allow vector genomes to persist, and hence contribute to decreased plasma arginine. As anticipated, ARG1(-/-) neonatal mice treated with AAVrh10 carrying a modified creatine kinase-based muscle-specific promoter did not survive longer than controls; however, their plasma arginine levels remained normal when animals were hyperammonemic. These data imply that plasma arginine can be controlled in arginase deficiency by muscle-specific expression, thus suggesting an alternative approach to utilizing the liver for treating hyperargininemia.


Asunto(s)
Arginasa/genética , Regulación de la Expresión Génica , Hiperamonemia/genética , Hiperargininemia/genética , Células Musculares/metabolismo , Animales , Arginasa/metabolismo , Línea Celular , Codón , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/genética , Hepatocitos/metabolismo , Humanos , Hiperamonemia/metabolismo , Hiperargininemia/metabolismo , Hiperargininemia/mortalidad , Masculino , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/metabolismo , Mioblastos Cardíacos/metabolismo , Especificidad de Órganos/genética , Regiones Promotoras Genéticas
14.
J Neuroimaging ; 24(2): 155-60, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-22928720

RESUMEN

BACKGROUND AND PURPOSE: Hyperargininemia (HA) is a rare autosomal recessive metabolic disorder and the neuroimaging features of this disease have seldom been reported. Hyperammonemic encephalopathy is uncommon in HA, and the clinical presentation of HA is distinct from other urea cycle disorders. This paper describes the brain MRI findings and a magnetic resonance spectroscopy (MRS) study of a series of Brazilian HA patients. METHODS: Brain MR images were obtained in eight male and two female patients with the classic HA phenotype. Six patients were evaluated twice. Single-voxel (1)H-MRS was also performed in six of the patients. RESULTS: Only 1 patient, with less severe neurological symptoms, had normal MRI images. A variable degree of cerebral atrophy was noted in the other patients, and 3 patients also presented mild symptoms of cerebellar atrophy. MRS indicated no metabolic abnormalities in any patient. CONCLUSIONS: We present the MRI and MRS findings of a large series of HA patients. Variable degrees of brain atrophy and mild cerebellar atrophy were observed, and these findings were not specific. No metabolic abnormality was observed using MRS in this series of patients.


Asunto(s)
Arginina/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Hiperargininemia/diagnóstico , Hiperargininemia/metabolismo , Imagen por Resonancia Magnética/métodos , Espectroscopía de Protones por Resonancia Magnética/métodos , Adolescente , Adulto , Atrofia , Biomarcadores/metabolismo , Niño , Femenino , Humanos , Masculino , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Adulto Joven
15.
PLoS One ; 8(11): e80001, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24224027

RESUMEN

Arginase deficiency is a rare autosomal recessive disorder resulting from a loss of the liver arginase isoform, arginase 1 (ARG1), which is the final step in the urea cycle for detoxifying ammonia. ARG1 deficiency leads to hyperargininemia, characterized by progressive neurological impairment, persistent growth retardation and infrequent episodes of hyperammonemia. Using the Cre/loxP-directed conditional gene knockout system, we generated an inducible Arg1-deficient mouse model by crossing "floxed" Arg1 mice with CreER(T2) mice. The resulting mice (Arg-Cre) die about two weeks after tamoxifen administration regardless of the starting age of inducing the knockout. These treated mice were nearly devoid of Arg1 mRNA, protein and liver arginase activity, and exhibited symptoms of hyperammonemia. Plasma amino acid analysis revealed pronounced hyperargininemia and significant alterations in amino acid and guanidino compound metabolism, including increased citrulline and guanidinoacetic acid. Despite no alteration in ornithine levels, concentrations of other amino acids such as proline and the branched-chain amino acids were reduced. In summary, we have generated and characterized an inducible Arg1-deficient mouse model exhibiting several pathologic manifestations of hyperargininemia. This model should prove useful for exploring potential treatment options of ARG1 deficiency.


Asunto(s)
Aminoácidos/metabolismo , Arginasa/metabolismo , Hiperargininemia/enzimología , Hiperargininemia/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Animales , Arginasa/genética , Femenino , Hiperargininemia/genética , Masculino , Ratones , Ratones Noqueados
16.
Mol Genet Metab ; 110(3): 222-30, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23920045

RESUMEN

Human arginase deficiency is characterized by hyperargininemia and infrequent episodes of hyperammonemia, which lead to neurological impairment with spasticity, loss of ambulation, seizures, and severe mental and growth retardation; uncommonly, patients suffer early death from this disorder. In a murine targeted knockout model, onset of the phenotypic abnormality is heralded by weight loss at around day 15, and death occurs typically by postnatal day 17 with hyperargininemia and markedly elevated ammonia. This discrepancy between the more attenuated juvenile-onset human disease and the lethal neonatal murine model has remained suboptimal for studying and developing therapy for the more common presentation of arginase deficiency. These investigations aimed to address this issue by creating an adult conditional knockout mouse to determine whether later onset of arginase deficiency also resulted in lethality. Animal survival and ammonia levels, body weight, circulating amino acids, and tissue arginase levels were examined as outcome parameters after widespread Cre-recombinase activation in a conditional knockout model of arginase 1 deficiency. One hundred percent of adult female and 70% of adult male mice died an average of 21.0 and 21.6 days, respectively, after the initiation of tamoxifen administration. Animals demonstrated elevated circulating ammonia and arginine at the onset of phenotypic abnormalities. In addition, brain and liver amino acids demonstrated abnormalities. These studies demonstrate that (a) the absence of arginase in adult animals results in a disease profile (leading to death) similar to that of the targeted knockout and (b) the phenotypic abnormalities seen in the juvenile-onset model are not exclusive to the age of the animal but instead to the biochemistry of the disorder. This adult model will be useful for developing gene- and cell-based therapies for this disorder that will not be limited by the small animal size of neonatal therapy and for developing a better understanding of the characteristics of hyperargininemia.


Asunto(s)
Genes Letales , Hiperargininemia/genética , Hiperargininemia/metabolismo , Fenotipo , Aminoácidos/sangre , Aminoácidos/metabolismo , Animales , Arginasa/genética , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Genotipo , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperargininemia/tratamiento farmacológico , Hiperargininemia/mortalidad , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Tamoxifeno/administración & dosificación , Tamoxifeno/farmacología , Pérdida de Peso
17.
Mol Genet Metab ; 93(2): 172-8, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17997338

RESUMEN

In humans, arginase I (AI)-deficiency results in hyperargininemia, a metabolic disorder with symptoms of progressive neurological and intellectual impairment, spasticity, persistent growth retardation, and episodic hyperammonemia. A deficiency of arginase II (AII) has never been detected and the clinical disorder, if any, associated with its deficiency has not been defined. Since the spasticity and paucity of hyperammonemic crises seen in human AI-deficient patients are not features of the other urea cycle disorders, the likelihood of ammonia as the main neuropathogenic agent becomes extremely low, and the modest elevations of arginine seen in the brains of our mouse model of hyperargininemia make it an unlikely candidate as well. Specific guanidino compounds, direct or indirect metabolites of arginine, are elevated in the blood of patients with uremia. Other guanidino compounds are also increased in plasma and cerebrospinal fluid of hyperargininemic patients making them plausible as neurotoxins in these disorders. We analyzed several guanidino compounds in our arginase single and double knockout animals and found that alpha-keto-delta-guanidinovaleric acid, alpha-N-acetylarginine, and argininic acid were increased in the brain tissue from the AI knockout and double knockout animals. Several compounds were also increased in the plasma, liver, and kidneys. This is the first time that several of the guanidino compounds have been shown to be elevated in the brain tissue of an arginase-deficient mammal, and it further supports their possible role as the neuropathogenic agents responsible for the complications seen in arginase deficiency.


Asunto(s)
Guanidinas/sangre , Hiperargininemia/sangre , Animales , Arginasa/genética , Barrera Hematoencefálica , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Guanidinas/metabolismo , Humanos , Hiperargininemia/genética , Hiperargininemia/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
18.
J Nutr ; 137(6 Suppl 2): 1656S-1661S, 2007 06.
Artículo en Inglés | MEDLINE | ID: mdl-17513443

RESUMEN

The urinary excretion of orotic acid, an intermediate in the pyrimidine biosynthetic pathway, is markedly increased in many inborn errors of the urea cycle and in a number of other disorders involving arginine metabolism. Carbamoyl phosphate, which accumulates within hepatic mitochondria in patients with ornithine transcarbamoylase deficiency, can diffuse to the cytosol and enter the pyrimidine pathway, resulting in greatly increased orotic acid production and excretion. This orotic aciduria also occurs in inborn errors of the mitochondrial ornithine/citrulline transporter, arginase, argininosuccinate synthetase, and argininosuccinate lyase. Increased orotic acid excretion is also found in a number of hypoargininemic states, such as lysinuric protein intolerance. However, orotic aciduria should not be used uncritically as an index of arginine deficiency because it is found in patients with arginase deficiency who exhibit hyperargininemia. Increased orotic acid excretion can also arise as a result of impairments of pyrimidine synthesis, whether brought about by a genetic defect (e.g., in UMP synthase) or by drugs that inhibit the terminal part of the pathway (e.g., allopurinol or 6-azauridine). When used appropriately, measurement of urinary orotic acid is a valuable tool for the study of many derangements of arginine metabolism, including arginine depletion, and to assess the efficacy of therapies used to replete this amino acid.


Asunto(s)
Arginina/metabolismo , Hiperargininemia/metabolismo , Ácido Orótico/orina , Humanos
19.
Cell Mol Neurobiol ; 26(2): 177-89, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16619133

RESUMEN

1. We have previously demonstrated that arginine administration induces oxidative stress and compromises energy metabolism in rat hippocampus. In the present study we initially investigated the influence of pretreatment with alpha-tocopherol and ascorbic acid on the effects produced by arginine on hippocampus energy metabolism. We also tested the effect of acute administration of arginine on various parameters of energy metabolism, namely glucose uptake, lactate release and on the activities of succinate dehydrogenase, complex II and cytochrome c oxidase in rat cerebellum, as well as the influence of pretreatment with alpha-tocopherol and ascorbic acid on the effects elicited by arginine on this structure. 2. Sixty-day-old female Wistar rats were treated with a single i.p. injection of saline (control) or arginine (0.8 g/kg) and were killed 1 h later. In another set of experiments, the animals were pretreated for 1 week with daily i.p. administration of saline (control) or alpha-tocopherol (40 mg/kg) and ascorbic acid (100 mg/kg). Twelve hours after the last injection of the antioxidants the rats received one i.p. injection of arginine (0.8 g/kg) or saline and were killed 1 h later. 3. Results showed that arginine administration significantly increased lactate release and diminished glucose uptake and the activities of succinate dehydrogenase and complex II in rat cerebellum. In contrast, complex IV (cytochrome c oxidase) activity was not changed by this amino acid. Furthermore, pretreatment with alpha-tocopherol and ascorbic acid prevented the impairment of energy metabolism caused by hyperargininemia in cerebellum and hippocampus of rats.


Asunto(s)
Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Metabolismo Energético/efectos de los fármacos , Hipocampo , Hiperargininemia/metabolismo , alfa-Tocoferol/farmacología , Animales , Antioxidantes/administración & dosificación , Arginina/administración & dosificación , Arginina/farmacología , Ácido Ascórbico/administración & dosificación , Complejo II de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Glucosa/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Ácido Láctico/metabolismo , Ratas , Ratas Wistar , Succinato Deshidrogenasa/metabolismo , alfa-Tocoferol/administración & dosificación
20.
Neuroreport ; 13(10): 1301-4, 2002 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-12151791

RESUMEN

Hyperargininemia is an inherited neurometabolic disorder biochemically characterized by tissue accumulation of arginine and clinically by severe neurological symptoms whose pathophysiology is poorly understood. In the present study we investigated the effect of arginine administration on the antioxidant enzyme activities catalase, glutathione peroxidase and superoxide dismutase in rat midbrain. We also tested the effect of L-NAME on the effects produced by Arg. The results showed that arginine decreased catalase activity, without altering the other two activities. L-NAME had no effect on catalase activity, but prevented the reduction of this enzyme provoked by arginine, suggesting that NO formation is involved in the reduction of catalase activity caused by the amino acid. If these findings also occur in the human condition, it may be presumed that oxidative stress contributes to the brain dysfunction observed in hyperargininemia.


Asunto(s)
Arginina/farmacología , Catalasa/metabolismo , Mesencéfalo/efectos de los fármacos , Mesencéfalo/enzimología , Animales , Inhibidores Enzimáticos/farmacología , Glutatión Peroxidasa/metabolismo , Hiperargininemia/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA