Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Mol Genet Metab ; 137(1-2): 153-163, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36049366

RESUMEN

BACKGROUND: Arginase 1 Deficiency (ARG1-D) is a rare, progressive, metabolic disorder that is characterized by devastating manifestations driven by elevated plasma arginine levels. It typically presents in early childhood with spasticity (predominately affecting the lower limbs), mobility impairment, seizures, developmental delay, and intellectual disability. This systematic review aims to identify and describe the published evidence outlining the epidemiology, diagnosis methods, measures of disease progression, clinical management, and outcomes for ARG1-D patients. METHODS: A comprehensive literature search across multiple databases such as MEDLINE, Embase, and a review of clinical studies in ClinicalTrials.gov (with results reported) was carried out per PRISMA guidelines on 20 April 2020 with no date restriction. Pre-defined eligibility criteria were used to identify studies with data specific to patients with ARG1-D. Two independent reviewers screened records and extracted data from included studies. Quality was assessed using the modified Newcastle-Ottawa Scale for non-comparative studies. RESULTS: Overall, 55 records reporting 40 completed studies and 3 ongoing studies were included. Ten studies reported the prevalence of ARG1-D in the general population, with a median of 1 in 1,000,000. Frequently reported diagnostic methods included genetic testing, plasma arginine levels, and red blood cell arginase activity. However, routine newborn screening is not universally available, and lack of disease awareness may prevent early diagnosis or lead to misdiagnosis, as the disease has overlapping symptomology with other diseases, such as cerebral palsy. Common manifestations reported at time of diagnosis and assessed for disease progression included spasticity (predominately affecting the lower limbs), mobility impairment, developmental delay, intellectual disability, and seizures. Severe dietary protein restriction, essential amino acid supplementation, and nitrogen scavenger administration were the most commonly reported treatments among patients with ARG1-D. Only a few studies reported meaningful clinical outcomes of these interventions on intellectual disability, motor function and adaptive behavior assessment, hospitalization, or death. The overall quality of included studies was assessed as good according to the Newcastle-Ottawa Scale. CONCLUSIONS: Although ARG1-D is a rare disease, published evidence demonstrates a high burden of disease for patients. The current standard of care is ineffective at preventing disease progression. There remains a clear need for new treatment options as well as improved access to diagnostics and disease awareness to detect and initiate treatment before the onset of clinical manifestations to potentially enable more normal development, improve symptomatology, or prevent disease progression.


Asunto(s)
Hiperargininemia , Discapacidad Intelectual , Recién Nacido , Humanos , Preescolar , Arginasa/genética , Hiperargininemia/diagnóstico , Hiperargininemia/epidemiología , Hiperargininemia/genética , Convulsiones/diagnóstico , Convulsiones/epidemiología , Convulsiones/etiología , Espasticidad Muscular/diagnóstico , Espasticidad Muscular/epidemiología , Espasticidad Muscular/genética , Arginina/uso terapéutico , Aminoácidos Esenciales , Progresión de la Enfermedad , Nitrógeno
2.
Pediatr Int ; 64(1): e14945, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34343381

RESUMEN

BACKGROUND: Arginase-1 deficiency is a rare, autosomal recessively inherited disorder of the urea cycle. In this study, we describe the clinical and molecular details of six patients who were diagnosed with argininemia, and we describe two of the patients with hyperargininemia who carried two novel variations of the Arginase-1 gene. METHODS: The clinical and demographic characteristics of the patients were retrospectively evaluated. RESULTS: The ages of the six patients ranged from 1 day to 20 years, and each patient had consanguineous parents. Neuromotor retardation and spastic paraparesis were found in all patients except one, who was diagnosed prenatally. Hyperargininemia was present in all patients. Urinary orotic acid excretion was increased in four of the six patients. The diagnosis was confirmed by genetic analysis in all the patients. Elevated liver enzymes were detected in three patients and blood urea nitrogen levels were normal in each of the six patients. CONCLUSIONS: In this study, we describe the two patients with hyperargininemia who carried two novel variations of the ARG1 gene. Also, we present a patient with normal neurodevelopment who was diagnosed prenatally and treated at an early stage of the disease.


Asunto(s)
Arginasa , Hiperargininemia , Hepatopatías , Adolescente , Arginasa/genética , Niño , Preescolar , Humanos , Hiperargininemia/diagnóstico , Hiperargininemia/genética , Lactante , Mutación , Estudios Retrospectivos , Adulto Joven
3.
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
4.
Mov Disord ; 34(5): 625-636, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30913345

RESUMEN

The term "cerebral palsy mimic" is used to describe a number of neurogenetic disorders that may present with motor symptoms in early childhood, resulting in a misdiagnosis of cerebral palsy. Cerebral palsy describes a heterogeneous group of neurodevelopmental disorders characterized by onset in infancy or early childhood of motor symptoms (including hypotonia, spasticity, dystonia, and chorea), often accompanied by developmental delay. The primary etiology of a cerebral palsy syndrome should always be identified if possible. This is particularly important in the case of genetic or metabolic disorders that have specific disease-modifying treatment. In this article, we discuss clinical features that should alert the clinician to the possibility of a cerebral palsy mimic, provide a practical framework for selecting and interpreting neuroimaging, biochemical, and genetic investigations, and highlight selected conditions that may present with predominant spasticity, dystonia/chorea, and ataxia. Making a precise diagnosis of a genetic disorder has important implications for treatment, and for advising the family regarding prognosis and genetic counseling. © 2019 International Parkinson and Movement Disorder Society.


Asunto(s)
Parálisis Cerebral/diagnóstico , Diagnóstico Diferencial , Trastornos del Movimiento/diagnóstico , Adenilil Ciclasas/genética , Ataxia/fisiopatología , Ataxia Telangiectasia/diagnóstico , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/fisiopatología , Ataxia Telangiectasia/terapia , Encéfalo/diagnóstico por imagen , Encefalopatías Metabólicas Innatas/diagnóstico , Encefalopatías Metabólicas Innatas/genética , Encefalopatías Metabólicas Innatas/fisiopatología , Encefalopatías Metabólicas Innatas/terapia , Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico , Errores Innatos del Metabolismo de los Carbohidratos/genética , Errores Innatos del Metabolismo de los Carbohidratos/fisiopatología , Errores Innatos del Metabolismo de los Carbohidratos/terapia , Parálisis Cerebral/fisiopatología , Corea/fisiopatología , Creatina/deficiencia , Creatina/genética , Discinesias/diagnóstico , Discinesias/genética , Discinesias/fisiopatología , Discinesias/terapia , Distonía/fisiopatología , Deficiencia de Ácido Fólico/diagnóstico , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/fisiopatología , Deficiencia de Ácido Fólico/terapia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Humanos , Hiperargininemia/diagnóstico , Hiperargininemia/genética , Hiperargininemia/fisiopatología , Hiperargininemia/terapia , Síndrome de Lesch-Nyhan/diagnóstico , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/fisiopatología , Síndrome de Lesch-Nyhan/terapia , Imagen por Resonancia Magnética , Discapacidad Intelectual Ligada al Cromosoma X/diagnóstico , Discapacidad Intelectual Ligada al Cromosoma X/genética , Discapacidad Intelectual Ligada al Cromosoma X/fisiopatología , Discapacidad Intelectual Ligada al Cromosoma X/terapia , Proteínas de Transporte de Monosacáridos/deficiencia , Proteínas de Transporte de Monosacáridos/genética , Trastornos del Movimiento/genética , Trastornos del Movimiento/fisiopatología , Trastornos del Movimiento/terapia , Deficiencia Múltiple de Carboxilasa/diagnóstico , Deficiencia Múltiple de Carboxilasa/genética
5.
J Inherit Metab Dis ; 42(3): 407-413, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30671984

RESUMEN

PURPOSE: We report a patient with a human cationic amino acid transporter 2 (CAT-2) defect discovered due to a suspected arginase 1 deficiency observed in newborn screening (NBS). METHODS: A NBS sample was analyzed using tandem mass spectrometry. Screen results were confirmed by plasma and urine amino acid quantification. Molecular diagnosis was done using clinical exome sequencing. Dimethylated arginines were determined by HPLC and nitrate/nitrite levels by a colorimetric assay. The metabolomic profile was analyzed using 1D nuclear magnetic resonance spectroscopy. RESULTS: A Spanish boy of nonconsanguineous parents had high arginine levels in a NBS blood sample. Plasma and urinary cationic amino acids were high. Arginase enzyme activity in erythrocytes was normal and no pathogenic mutations were identified in the ARG1 gene. Massive parallel sequencing detected two loss-of-function mutations in the SLC7A2 gene. Currently, the child receives a protein-controlled diet of 1.2 g/kg/day with protein-and amino-acid free infant formula, 30 g/day, and is asymptomatic. CONCLUSION: We identified a novel defect in human CAT-2 due to biallelic pathogenic variants in the SLC7A2 gene. The characteristic biochemical profile includes high plasma and urine arginine, ornithine, and lysine levels. NBS centers should know of this disorder since it can be detected in arginase 1 deficiency screening.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/genética , Transportador de Aminoácidos Catiônicos 2/deficiencia , Enfermedades Metabólicas/genética , Arginasa/genética , Dieta con Restricción de Proteínas , Humanos , Hiperargininemia/genética , Recién Nacido , Masculino , Enfermedades Metabólicas/dietoterapia , Mutación , Tamizaje Neonatal
6.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(10): 996-998, 2019 Oct 10.
Artículo en Zh | MEDLINE | ID: mdl-31598944

RESUMEN

OBJECTIVE: To explore the genetic basis for an infant with early-onset argininemia. METHODS: Potential variant was detected with an Ion Torrent semiconductor sequencer using a gene panel for inherited diseases. Suspected variants were verified by Sanger sequencing. RESULTS: Genetic testing indicated that he has carried c.560+2T>C and c.811T>C compound heterozygous variant of the AGR1 gene, which were inherited from his father and mother, respectively. Among these, c.560+2T>C was suspected to be pathogenic, while c.811T>C was of unknown clinical significance, and both were not reported previously. CONCLUSION: The c.560+2T>C and c.811T>C compound heterozygous variants of the AGR1 gene probably underlies the argininemia in this child. Above finding has enriched the variant spectrum of the AGR1 gene.


Asunto(s)
Arginasa/genética , Hiperargininemia/genética , Femenino , Pruebas Genéticas , Humanos , Lactante , Masculino
7.
Metab Brain Dis ; 33(5): 1775-1778, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29961243

RESUMEN

Argininemia is a rare hereditary disease due to a deficiency of hepatic arginase, which is the last enzyme of the urea cycle and hydrolyzes arginine to ornithine and urea. Herein we report a patient with arginase I (ARG1) deficiency who presented with recurrent nonconvulsive status epilepticus and liver failure. A novel homozygous frameshift mutation c.703_707delGGACTinsAGACTGGACC (p.G235Rfs*20) was detected.


Asunto(s)
Arginasa/genética , Hiperargininemia/complicaciones , Fallo Hepático/etiología , Estado Epiléptico/etiología , Encéfalo/diagnóstico por imagen , Preescolar , Femenino , Humanos , Hiperargininemia/diagnóstico por imagen , Hiperargininemia/genética , Fallo Hepático/diagnóstico por imagen , Fallo Hepático/genética , Imagen por Resonancia Magnética , Estado Epiléptico/diagnóstico por imagen , Estado Epiléptico/genética
8.
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
9.
Gene Ther ; 22(2): 111-5, 2015 02.
Artículo en Inglés | MEDLINE | ID: mdl-25474440

RESUMEN

Hyperammonemia is less severe in arginase 1 deficiency compared with other urea cycle defects. Affected patients manifest hyperargininemia and infrequent episodes of hyperammonemia. Patients typically suffer from neurological impairment with cortical and pyramidal tract deterioration, spasticity, loss of ambulation, seizures and intellectual disability; death is less common than with other urea cycle disorders. In a mouse model of arginase I deficiency, the onset of symptoms begins with weight loss and gait instability, which progresses toward development of tail tremor with seizure-like activity; death typically occurs at about 2 weeks of life. Adeno-associated viral vector gene replacement strategies result in long-term survival of mice with this disorder. With neonatal administration of vector, the viral copy number in the liver greatly declines with hepatocyte proliferation in the first 5 weeks of life. Although the animals do survive, it is not known from a functional standpoint how well the urea cycle is functioning in the adult animals that receive adeno-associated virus. In these studies, we administered [1-13C] acetate to both littermate controls and adeno-associated virus-treated arginase 1 knockout animals and examined flux through the urea cycle. Circulating ammonia levels were mildly elevated in treated animals. Arginine and glutamine also had perturbations. Assessment 30 min after acetate administration demonstrated that ureagenesis was present in the treated knockout liver at levels as low at 3.3% of control animals. These studies demonstrate that only minimal levels of hepatic arginase activity are necessary for survival and ureagenesis in arginase-deficient mice and that this level of activity results in control of circulating ammonia. These results may have implications for potential therapy in humans with arginase deficiency.


Asunto(s)
Dependovirus/genética , Hiperargininemia/terapia , Amoníaco/sangre , Animales , Arginasa/genética , Arginasa/metabolismo , Modelos Animales de Enfermedad , Terapia Genética , Hiperamonemia/sangre , Hiperamonemia/genética , Hiperamonemia/terapia , Hiperargininemia/sangre , Hiperargininemia/genética , Hígado/enzimología , Hígado/patología , Ratones , Ratones Noqueados
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.
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
12.
Gene Ther ; 20(8): 785-96, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23388701

RESUMEN

Complete arginase I deficiency is the least severe urea cycle disorder, characterized by hyperargininemia and infrequent episodes of hyperammonemia. Patients suffer from neurological impairment with cortical and pyramidal tract deterioration, spasticity, loss of ambulation and seizures, and is associated with intellectual disability. In mice, onset is heralded by weight loss beginning around day 15; gait instability follows progressing to inability to stand and development of tail tremor with seizure-like activity and death. Here we report that hyperargininemic mice treated neonatally with an adeno-associated virus (AAV)-expressing arginase and followed long-term lack any presentation consistent with brain dysfunction. Behavioral and histopathological evaluation demonstrated that treated mice are indistinguishable from littermates, and that putative compounds associated with neurotoxicity are diminished. In addition, treatment results in near complete resolution of metabolic abnormalities early in life; however, there is the development of some derangement later with decline in transgene expression. Ammonium challenging revealed that treated mice are affected by exogenous loading much greater than littermates. These results demonstrate that AAV-based therapy for hyperargininemia is effective and prevents development of neurological abnormalities and cognitive dysfunction in a mouse model of hyperargininemia; however, nitrogen challenging reveals that these mice remain impaired in the handling of waste nitrogen.


Asunto(s)
Arginasa/genética , Terapia Genética , Hiperargininemia/genética , Enfermedades del Sistema Nervioso/genética , Enfermedades Neurodegenerativas/genética , Animales , Arginasa/metabolismo , Dependovirus , Modelos Animales de Enfermedad , Humanos , Hiperamonemia/genética , Hiperamonemia/patología , Hiperamonemia/terapia , Hiperargininemia/patología , Hiperargininemia/terapia , Ratones , Ratones Transgénicos , Enfermedades del Sistema Nervioso/patología , Enfermedades del Sistema Nervioso/terapia , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia
13.
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
14.
Mol Ther ; 20(10): 1844-51, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22760543

RESUMEN

Arginase deficiency is characterized by hyperargininemia and infrequent episodes of hyperammonemia. Human patients suffer from neurological impairment with spasticity, loss of ambulation, seizures, and severe mental and growth retardation. In a murine model, onset of the phenotypic abnormality is heralded by weight loss beginning around day 15 with death occurring typically by postnatal day 17 with hyperargininemia and markedly elevated ammonia. The goal of this study was to address the development of a gene therapy approach for arginase deficiency beginning in the neonatal period. Lifespan extension, body weight, circulating amino acids and ammonia levels were examined as outcome parameters after gene therapy with an adeno-associated viral vector expressing arginase was administered to mice on the second day of life (DOL). One-hundred percent of untreated arginase-deficient mice died by DOL 24, whereas 89% of the adeno-associated virus (AAV)-treated arginase deficient mice have survived for >8 months. While animals at 8 months demonstrate elevated glutamine levels, ammonia is less than three times that of controls and arginine levels are normal. These studies are the first to demonstrate that AAV-based therapy for arginase deficiency is effective and supports the development of gene therapy for this and the other urea cycle disorders.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Hiperargininemia/terapia , Aminoácidos/sangre , Amoníaco/sangre , Animales , Arginasa/genética , Arginasa/metabolismo , Western Blotting , Femenino , Fertilidad/genética , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Genotipo , Glutamina/sangre , Hiperargininemia/genética , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados
15.
Zhongguo Dang Dai Er Ke Za Zhi ; 15(11): 954-9, 2013 Nov.
Artículo en Zh | MEDLINE | ID: mdl-24229587

RESUMEN

Argininemia is a rare, autosomal recessive, metabolic disorder caused by an hereditary deficiency of hepatocytes arginase due to ARG1 gene defect. Arginase is the final enzyme in the urea cycle, catalyzing the hydrolysis of arginine to ornithine and urea. Research advances in the clinical manifestations, diagnosis, treatment, prenatal diagnosis and genetics of argininemia were reviewed in this paper. The clinical manifestations of patients with argininemia are complicated and nonspecific so that clinical diagnosis is usually difficult and delayed. Progressive spastic tetraplegia, seizures and cerebella atrophy are common clinical features of the disease. Blood amino acids analysis, arginase assay and ARG1 gene analysis are important to the diagnosis of argininemia. Early diagnosis and a protein-restricted diet with citrulline and benzoate supplements can contribute a lot to improve patient prognosis. With the application of liquid chromatography-tandem mass spectrometry in selective screening and newborn screening for inborn errors of metabolism, an ever-increasing number of patients with argininemia are detected at the asymptomatic or early stages.


Asunto(s)
Hiperargininemia/terapia , Arginasa/genética , Humanos , Hiperargininemia/diagnóstico , Hiperargininemia/genética , Biología Molecular , Pronóstico
16.
J Genet ; 1022023.
Artículo en Inglés | MEDLINE | ID: mdl-36722221

RESUMEN

Arginase deficiency is an autosomal recessive urea cycle disorder caused by pathogenic variants in the ARG1 gene. The clinical features of the disease include spasticity, tremour, ataxia, hypotonia, microcephaly and seizures. Growth delay can also be observed in the affected individuals. Here we describe the results from molecular-genetic analysis of two patients with arginase deficiency. In the first case, we reported a novel homozygous missense variant c.775G>A p.(Gly259Ser) in a patient with Bulgarian ethnic origin. In the second case, a novel homozygous splice site variant c.329+1G>A was detected in a patient from a consanguineous family of Roma ethnic origin. A hundred samples of newborns of Roma origin were screened for variant c.329+1G>A and one individual was found to be a heterozygous carrier of variant c.329+1G> A. The results from this study indicated the necessity for screening of the Roma population with respect to the disease arginase deficiency in Bulgaria.


Asunto(s)
Hiperargininemia , Recién Nacido , Humanos , Hiperargininemia/epidemiología , Hiperargininemia/genética , Bulgaria/epidemiología , Ataxia , Consanguinidad , Etnicidad
17.
J Pediatr Endocrinol Metab ; 35(1): 125-129, 2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-34653322

RESUMEN

We described two Japanese siblings with arginase-1 (ARG1) deficiency. A 10-year-old girl (the proband and elder sister) was referred to our hospital complaining about her short stature. We diagnosed her with ARG1 deficiency, possibly with elevated levels of blood ammonia and plasma arginine. Her younger sister was found to have spastic paraparesis in her lower extremities and short stature at the age of 4 years. The younger sister also had high levels of plasma arginine, instead of normal levels of blood ammonia. Interestingly, they also prefer to avoid protein-rich foods such as meat, soybeans, cow milk, and dairy products. Genetic testing identified compound heterozygous mutations (c.121_122insCTT [p.Lys41Thrfs∗2] and c.298G>A [p.Asp100Asn]) in the ARG1 gene. The ARG1 mutation of p.Lys41Thrfs∗2 is a novel pathogenic mutation according to open databases and literature.


Asunto(s)
Arginasa/genética , Mutación del Sistema de Lectura , Hiperargininemia/genética , Adolescente , Amoníaco/sangre , Arginina/sangre , Niño , Femenino , Humanos , Hermanos
18.
Orphanet J Rare Dis ; 17(1): 94, 2022 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-35236361

RESUMEN

BACKGROUND/OBJECTIVE: Arginase 1 Deficiency (ARG1-D) is a rare inherited metabolic disease with progressive, devastating neurological manifestations with early mortality and high unmet need. Information on prevalence is scarce and highly variable due to limited newborn screening (NBS) availability, variability of arginine levels in the first days of life, and high rates of misdiagnosis. US birth prevalence was recently estimated via indirect methods at 1.1 cases per million live births. Due to the autosomal recessive nature of ARG1-D we hypothesize that the global prevalence may be more accurately estimated using genetic population databases. METHODS: MEDLINE and EMBASE were systematically searched for previously reported disease variants. Disease variants in ARG1-D were annotated wherever possible with allele frequencies from gnomAD. Ethnicity-specific prevalence was calculated using the Hardy-Weinberg equation and applied to generate country-specific carrier frequencies for 38 countries. Finally, documented consanguinity rates were applied to establish a birth prevalence for each country. RESULTS: 133 of 228 (58%) known causative alleles were annotated with ethnic-specific frequencies. Global birth prevalence for ARG1-D was estimated at 2.8 cases per million live births (country-specific estimates ranged from 0.92 to 17.5) and population prevalence to be 1.4 cases per million people (approximately 1/726,000 people). Birth prevalence estimates were dependent on population demographics and consanguinity rate. CONCLUSION: Birth prevalence of ARG1-D based on genetic database analysis was estimated to be more frequent than previous NBS studies have indicated. There was a higher degree of confidence in North American and European countries due to availability of genetic databases and mutational analysis versus other regions. These findings suggest the need for greater disease education around signs and manifestations of ARG1-D, as well as more widespread testing and standardization of screening for this severe disease in order to appropriately identify patients prior to disease progression.


Asunto(s)
Arginasa , Hiperargininemia , Alelos , Arginasa/genética , Bases de Datos Genéticas , Frecuencia de los Genes , Humanos , Hiperargininemia/epidemiología , Hiperargininemia/genética , Recién Nacido , Prevalencia
19.
J Coll Physicians Surg Pak ; 32(12): 1629-1631, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36474391

RESUMEN

Arginase 1(ARG1) deficiency is a rare disorder of the urea cycle. The presentation is usually late, leading to loss of intellectual milestones, spasticity and liver involvement. Hyperammonemic crises are rarely encountered. We herein present a case of a 16-year immigrant girl of Syrian origin who was evaluated for acute onset of fever, vomiting, and seizures. Laboratory analyses showed slightly elevated lactate, creatine kinase, and coagulation parameters. Ammonium levels were also moderately increased. On 5th day of admission, she went into an encephalopathic state. Blood amino acid analysis showed highly elevated arginine levels. An increased level of orotic acid was found in urine organic acid analysis. Molecular genetic analysis of ARG1 gene showed a novel homozygous mutation. Although the presentation of ARG1 deficiency is usually chronic in the majority of patients, an acute crisis of encephalopathy due to hyperammonemia may occur and delayed diagnosis may lead to irreversible neurological damage. Key Words: Urea cycle disorder, Hyperammonemia, Argininemia, Encephalopathy.


Asunto(s)
Hiperargininemia , Estado Epiléptico , Humanos , Hiperargininemia/complicaciones , Hiperargininemia/diagnóstico , Hiperargininemia/genética , Estado Epiléptico/diagnóstico , Estado Epiléptico/etiología
20.
Indian J Pediatr ; 88(3): 266-268, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32770317

RESUMEN

Argininemia or hyperargininemia is a urea cycle disorder caused by deficiency of the enzyme arginase 1. It is inherited in an autosomal recessive fashion. It commonly leads to spastic diplegia in childhood, but other important features include cognitive deterioration and epilepsy. Unlike other disorders of the urea cycle, hyperammonemia is not prominent. The authors report three siblings with genetically proven argininemia who presented with diverse phenotypes but with spasticity being a common feature. Sibling 1 developed motor regression in early childhood, sibling 2 developed delayed motor milestones from early infancy, whereas sibling 3 had global developmental delay in late infancy after a period of normal development. All siblings had mild hyperammonemia only. Early recognition is imperative, not only to initiate ammonia scavenging therapy which may lead to definite clinical improvement, but also to provide genetic counselling.


Asunto(s)
Hiperamonemia , Hiperargininemia , Arginasa/genética , Preescolar , Humanos , Hiperamonemia/etiología , Hiperamonemia/genética , Hiperargininemia/diagnóstico , Hiperargininemia/genética , Fenotipo , Hermanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA