Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mov Disord ; 34(5): 625-636, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30913345

RESUMO

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.


Assuntos
Paralisia Cerebral/diagnóstico , Diagnóstico Diferencial , Transtornos dos Movimentos/diagnóstico , Adenilil Ciclases/genética , Ataxia/fisiopatologia , Ataxia Telangiectasia/diagnóstico , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/fisiopatologia , Ataxia Telangiectasia/terapia , Encéfalo/diagnóstico por imagem , Encefalopatias Metabólicas Congênitas/diagnóstico , Encefalopatias Metabólicas Congênitas/genética , Encefalopatias Metabólicas Congênitas/fisiopatologia , Encefalopatias Metabólicas Congênitas/terapia , Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Erros Inatos do Metabolismo dos Carboidratos/genética , Erros Inatos do Metabolismo dos Carboidratos/fisiopatologia , Erros Inatos do Metabolismo dos Carboidratos/terapia , Paralisia Cerebral/fisiopatologia , Coreia/fisiopatologia , Creatina/deficiência , Creatina/genética , Discinesias/diagnóstico , Discinesias/genética , Discinesias/fisiopatologia , Discinesias/terapia , Distonia/fisiopatologia , Deficiência de Ácido Fólico/diagnóstico , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/fisiopatologia , Deficiência de Ácido Fólico/terapia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Humanos , Hiperargininemia/diagnóstico , Hiperargininemia/genética , Hiperargininemia/fisiopatologia , Hiperargininemia/terapia , Síndrome de Lesch-Nyhan/diagnóstico , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/fisiopatologia , Síndrome de Lesch-Nyhan/terapia , Imageamento por Ressonância Magnética , Deficiência Intelectual Ligada ao Cromossomo X/diagnóstico , Deficiência Intelectual Ligada ao Cromossomo X/genética , Deficiência Intelectual Ligada ao Cromossomo X/fisiopatologia , Deficiência Intelectual Ligada ao Cromossomo X/terapia , Proteínas de Transporte de Monossacarídeos/deficiência , Proteínas de Transporte de Monossacarídeos/genética , Transtornos dos Movimentos/genética , Transtornos dos Movimentos/fisiopatologia , Transtornos dos Movimentos/terapia , Deficiência Múltipla de Carboxilase/diagnóstico , Deficiência Múltipla de Carboxilase/genética
2.
Semin Pediatr Neurol ; 26: 110-114, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29961498

RESUMO

Inborn errors of metabolism (IEMs) are thought to present in infancy with acute decompensation including feeding intolerance and vomiting, lethargy, and coma. Most practitioners assume that children will be diagnosed in their first months of life. However, certain IEMs present more insidiously, and occasionally children fail to receive newborn screening resulting in delayed diagnoses, as metabolic and genetic disorders are overlooked causes of cognitive and neurologic deficits. Although signs and symptoms may be present but subtle, careful and detailed history taking, particularly of a child's diet and neurologic medical history, in addition to certain physical examination findings may suggest a diagnosis that is later supported by laboratory and radiographic testing. We present the case of an 11-year-old girl who presented with a diagnosis of cerebral palsy, seizure disorder, and concerns of fatigue and increasing seizure frequency. During hospitalization, she was found to have hyperammonemia, and a diagnosis of arginase deficiency was made. More thorough review of her previous records may have raised suspicion for IEM earlier.


Assuntos
Hiperargininemia/diagnóstico , Hiperargininemia/fisiopatologia , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Paralisia Cerebral/diagnóstico , Criança , Diagnóstico Diferencial , Feminino , Humanos , Hiperargininemia/terapia
3.
RNA Biol ; 15(7): 914-922, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29923457

RESUMO

Arginase I (ARG1) deficiency is an autosomal recessive urea cycle disorder, caused by deficiency of the enzyme Arginase I, resulting in accumulation of arginine in blood. Current Standard of Care (SOC) for ARG1 deficiency in patients or those having detrimental mutations of ARG1 gene is diet control. Despite diet and drug therapy with nitrogen scavengers, ~25% of patients suffer from severe mental deficits and loss of ambulation. 75% of patients whose symptoms can be managed through diet therapy continue to suffer neuro-cognitive deficits. In our research, we demonstrate in vitro and in vivo that administration of ARG1 mRNA increased ARG1 protein expression and specific activity in relevant cell types, including ARG1-deficient patient cell lines, as well as in wild type mice for up to 4 days. These studies demonstrate that ARG1 mRNA treatment led to increased functional protein expression of ARG1 and subsequently an increase in urea. Hence, ARG1 mRNA therapy could be a potential treatment option to develop for patients.


Assuntos
Arginase/metabolismo , Arginina/metabolismo , Terapia Biológica/métodos , Hiperargininemia/terapia , RNA Mensageiro/administração & dosagem , Animais , Arginase/genética , Células HeLa , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Ureia/metabolismo
4.
Medicine (Baltimore) ; 97(7): e9880, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29443755

RESUMO

RATIONALE: Argininemia is an autosomal recessive inherited disorder of the urea cycle. Because of its atypical symptoms in early age, diagnosis can be delayed until the typical chronic manifestations - including spastic diplegia, deterioration in cognitive function, and epilepsy - appear in later childhood. PATIENT CONCERNS: A Chinese boy initially presented with severe stunting and partial growth hormone deficiency (PGHD) at 3 years old and was initially treated with growth hormone replacement therapy. Seven years later (at 10 years old), he presented with spastic diplegia, cognitive function lesions, epilepsy, and peripheral neuropathy. DIAGNOSES: Ultimately, the patient was diagnosed with argininemia with homozygous mutation (c.32T>C) of the ARG1 gene at 10 years old. Blood tests showed mildly elevated blood ammonia and creatine kinase, and persistently elevated bilirubin. INTERVENTIONS: Protein intake was limited to 0.8 g/kg/day, citrulline (150-200 mg [kg d]) was prescribed. OUTCOMES: The patient's mental state and vomiting had improved after 3 months treatment. At 10 years and 9 month old, his height and weight had reached 121cm and 22kg, respectively, but his spastic diplegia symptoms had not improved. LESSONS: This case demonstrates that stunting and PGHD that does not respond to growth hormone replacement therapy might hint at inborn errors of metabolism (IEM). IEM should also be considered in patients with persistently elevated bilirubin with or without abnormal liver transaminase, as well as elevated blood ammonia and creatine kinase, in the absence of hepatic disease.


Assuntos
Transtornos do Crescimento , Hiperargininemia , Arginase/genética , Bilirrubina/análise , Criança , Pré-Escolar , Diagnóstico Diferencial , Transtornos do Crescimento/diagnóstico , Transtornos do Crescimento/etiologia , Hormônio do Crescimento/análise , Hormônio do Crescimento/deficiência , Hormônio do Crescimento/uso terapêutico , Terapia de Reposição Hormonal/efeitos adversos , Terapia de Reposição Hormonal/métodos , Humanos , Hiperargininemia/diagnóstico , Hiperargininemia/genética , Hiperargininemia/fisiopatologia , Hiperargininemia/terapia , Masculino , Mutação
5.
Nucleic Acid Ther ; 28(2): 74-85, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29437538

RESUMO

Messenger RNA (mRNA) is a promising new class of therapeutics that has potential for treatment of diseases in fields such as immunology, oncology, vaccines, and inborn errors of metabolism. mRNA therapy has several advantages over DNA-based gene therapy, including the lack of the need for nuclear import and transcription, as well as limited possibility of genomic integration. One drawback of mRNA therapy, especially in cases such as metabolic disorders where repeated dosing will be necessary, is the relatively short in vivo half-life of mRNA (∼6-12 h). We hypothesize that protein engineering designed to improve translation, yielding longer-lasting protein, or modifications that would increase enzymatic activity would be helpful in alleviating this issue. In this study, we present two examples where sequence engineering improved the expression and duration, as well as enzymatic activity of target proteins in vitro. We then confirmed these findings in wild-type mice. This work shows that rational engineering of proteins can lead to improved therapies in vivo.


Assuntos
Arginase/genética , Hiperargininemia/terapia , Hipoxantina Fosforribosiltransferase/genética , Síndrome de Lesch-Nyhan/terapia , RNA Mensageiro/uso terapêutico , Sequência de Aminoácidos , Animais , Arginase/isolamento & purificação , Arginase/metabolismo , Células HeLa , Humanos , Hiperargininemia/sangue , Hipoxantina Fosforribosiltransferase/isolamento & purificação , Hipoxantina Fosforribosiltransferase/metabolismo , Síndrome de Lesch-Nyhan/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Nanopartículas/uso terapêutico , Engenharia de Proteínas , RNA Mensageiro/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência
6.
Sci Rep ; 7(1): 2585, 2017 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-28566761

RESUMO

Arginase-1 deficiency in humans is a rare genetic disorder of metabolism resulting from a loss of arginase-1, leading to impaired ureagenesis, hyperargininemia and neurological deficits. Previously, we generated a tamoxifen-inducible arginase-1 deficient mouse model harboring a deletion of Arg1 exons 7 and 8 that leads to similar biochemical defects, along with a wasting phenotype and death within two weeks. Here, we report a strategy utilizing the Clustered, Regularly Interspaced, Short Palindromic Repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system in conjunction with piggyBac technology to target and reincorporate exons 7 and 8 at the specific Arg1 locus in attempts to restore the function of arginase-1 in induced pluripotent stem cell (iPSC)-derived hepatocyte-like cells (iHLCs) and macrophages in vitro. While successful gene targeted repair was achieved, minimal urea cycle function was observed in the targeted iHLCs compared to adult hepatocytes likely due to inadequate maturation of the cells. On the other hand, iPSC-derived macrophages expressed substantial amounts of "repaired" arginase. Our studies provide proof-of-concept for gene-editing at the Arg1 locus and highlight the challenges that lie ahead to restore sufficient liver-based urea cycle function in patients with urea cycle disorders.


Assuntos
Arginase/genética , Edição de Genes , Hiperargininemia/terapia , Distúrbios Congênitos do Ciclo da Ureia/terapia , Animais , Arginase/uso terapêutico , Sistemas CRISPR-Cas/genética , Modelos Animais de Doenças , Terapia Genética , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Hiperargininemia/genética , Hiperargininemia/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fígado/metabolismo , Fígado/patologia , Camundongos , Distúrbios Congênitos do Ciclo da Ureia/genética , Distúrbios Congênitos do Ciclo da Ureia/patologia
7.
J Neurosci ; 36(25): 6680-90, 2016 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-27335400

RESUMO

UNLABELLED: Arginase 1 deficiency is a urea cycle disorder associated with hyperargininemia, spastic diplegia, loss of ambulation, intellectual disability, and seizures. To gain insight on how loss of arginase expression affects the excitability and synaptic connectivity of the cortical neurons in the developing brain, we used anatomical, ultrastructural, and electrophysiological techniques to determine how single-copy and double-copy arginase deletion affects cortical circuits in mice. We find that the loss of arginase 1 expression results in decreased dendritic complexity, decreased excitatory and inhibitory synapse numbers, decreased intrinsic excitability, and altered synaptic transmission in layer 5 motor cortical neurons. Hepatic arginase 1 gene therapy using adeno-associated virus rescued nearly all these abnormalities when administered to neonatal homozygous knock-out animals. Therefore, gene therapeutic strategies can reverse physiological and anatomical markers of arginase 1 deficiency and therefore may be of therapeutic benefit for the neurological disabilities in this syndrome. SIGNIFICANCE STATEMENT: These studies are one of the few investigations to try to understand the underlying neurological dysfunction that occurs in urea cycle disorders and the only to examine arginase deficiency. We have demonstrated by multiple modalities that, in murine layer 5 cortical neurons, a gradation of abnormalities exists based on the functional copy number of arginase: intrinsic excitability is altered, there is decreased density in asymmetrical and perisomatic synapses, and analysis of the dendritic complexity is lowest in the homozygous knock-out. With neonatal administration of adeno-associated virus expressing arginase, there is near-total recovery of the abnormalities in neurons and cortical circuits, supporting the concept that neonatal gene therapy may prevent the functional abnormalities that occur in arginase deficiency.


Assuntos
Arginase/uso terapêutico , Terapia Genética , Hiperargininemia/patologia , Hiperargininemia/terapia , Córtex Motor/fisiologia , Recuperação de Função Fisiológica/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Amônia/sangue , Animais , Animais Recém-Nascidos , Arginase/genética , Arginase/metabolismo , Modelos Animais de Doenças , Hiperargininemia/sangue , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Córtex Motor/citologia , Córtex Motor/ultraestrutura , Rede Nervosa/patologia , Rede Nervosa/fisiologia , Rede Nervosa/ultraestrutura , Neurônios/fisiologia , Neurônios/ultraestrutura , Picrotoxina/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Sinapses/ultraestrutura , Tetrodotoxina/farmacologia
8.
J Mol Med (Berl) ; 93(12): 1287-96, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26467175

RESUMO

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.


Assuntos
Hiperargininemia/genética , Hiperargininemia/metabolismo , Animais , Arginase/química , Arginase/genética , Arginase/metabolismo , Modelos Animais de Doenças , Estudos de Associação Genética , Humanos , Hiperargininemia/diagnóstico , Hiperargininemia/terapia , Fígado/metabolismo , Mutação , Fenótipo , Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/genética , Distúrbios Congênitos do Ciclo da Ureia/metabolismo
9.
Gene Ther ; 22(2): 111-5, 2015 02.
Artigo em Inglês | MEDLINE | ID: mdl-25474440

RESUMO

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.


Assuntos
Dependovirus/genética , Hiperargininemia/terapia , Amônia/sangue , Animais , Arginase/genética , Arginase/metabolismo , Modelos Animais de Doenças , Terapia Genética , Hiperamonemia/sangue , Hiperamonemia/genética , Hiperamonemia/terapia , Hiperargininemia/sangue , Hiperargininemia/genética , Fígado/enzimologia , Fígado/patologia , Camundongos , Camundongos Knockout
10.
Zhongguo Dang Dai Er Ke Za Zhi ; 15(11): 954-9, 2013 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-24229587

RESUMO

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.


Assuntos
Hiperargininemia/terapia , Arginase/genética , Humanos , Hiperargininemia/diagnóstico , Hiperargininemia/genética , Biologia Molecular , Prognóstico
11.
Gene Ther ; 20(8): 785-96, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23388701

RESUMO

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.


Assuntos
Arginase/genética , Terapia Genética , Hiperargininemia/genética , Doenças do Sistema Nervoso/genética , Doenças Neurodegenerativas/genética , Animais , Arginase/metabolismo , Dependovirus , Modelos Animais de Doenças , Humanos , Hiperamonemia/genética , Hiperamonemia/patologia , Hiperamonemia/terapia , Hiperargininemia/patologia , Hiperargininemia/terapia , Camundongos , Camundongos Transgênicos , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/terapia , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/terapia
12.
Mol Ther ; 20(10): 1844-51, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22760543

RESUMO

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.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Hiperargininemia/terapia , Aminoácidos/sangue , Amônia/sangue , Animais , Arginase/genética , Arginase/metabolismo , Western Blotting , Feminino , Fertilidade/genética , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Genótipo , Glutamina/sangue , Hiperargininemia/genética , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout
13.
J Inherit Metab Dis ; 33 Suppl 3: S503-6, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21229317

RESUMO

Hyperargininemia is a rare inborn error of metabolism due to arginase deficiency, which is inherited in an autossomal recessive manner. Arginase is the final enzyme of the urea cycle and catalyzes the conversion of arginine to urea and ornithine. This condition typically presents in early childhood (between 2 and 4 years of age) with developmental delay associated with progressive spastic paraparesis. Neonatal presentation is very uncommon with a poorly described outcome. Here, we discuss two cases of neonatal cholestasis as initial clinical presentation of hyperargininemia. In case 1, diagnosis was established at 2 months of age upon investigation of the etiology of cholestatic injury pattern and hepatosplenomegaly, and treatment was then initiated at when the patient was 3 months old. Unfortunately, the patient had progressive biliary cirrhosis to end-stage liver disease complicated with portal hypertension for which she underwent successful orthotopic liver transplant at 7 years of age. In case 2, hyperargininemia was identified through newborn screening and treatment was started when patient was 21 days old. Cholestasis was only identified in the patient's further evaluation and it resolved 2 weeks into treatment. The patient is currently 18 months old and her development and neurological examination remain unremarkable. Neonatal cholestasis as first presentation of hyperargininemia is rare, but this disorder should be included in the differential diagnosis of unexplained cholestasis in the neonate. In fact, these two cases suggest that arginase deficiency may be the cause of cholestatic liver disease.


Assuntos
Colestase/etiologia , Hiperargininemia/complicações , Aminoácidos Essenciais/uso terapêutico , Arginase/genética , Arginase/metabolismo , Arginina/sangue , Biomarcadores/sangue , Criança , Desenvolvimento Infantil , Pré-Escolar , Colestase/diagnóstico , Colestase/terapia , Dieta com Restrição de Proteínas , Progressão da Doença , Doença Hepática Terminal/etiologia , Feminino , Predisposição Genética para Doença , Humanos , Hiperargininemia/diagnóstico , Hiperargininemia/enzimologia , Hiperargininemia/genética , Hiperargininemia/terapia , Hipertensão Portal/etiologia , Lactente , Recém-Nascido , Cirrose Hepática Biliar/etiologia , Transplante de Fígado , Triagem Neonatal , Fenótipo , Resultado do Tratamento
14.
Mol Ther ; 17(7): 1155-63, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19367256

RESUMO

Neonatal gene therapy has the potential to ameliorate abnormalities before disease onset. Our gene knockout of arginase I (AI) deficiency is characterized by increasing hyperammonemia, neurological deterioration, and early death. We constructed a helper-dependent adenoviral vector (HDV) carrying AI and examined for correction of this defect. Neonates were administered 5 x 10(9) viral particles/g and analyzed for survival, arginase activity, and ammonia and amino acids levels. The life expectancy of arg(-/-) mice increased to 27 days while controls died at 14 days with hyperammonemia and in extremis. Death correlated with a decrease in viral DNA/RNA per cell as liver mass increased. Arginase assays demonstrated that vector-injected hepatocytes had ~20% activity of heterozygotes at 2 weeks of age. Hepatic arginine and ornithine in treated mice were similar to those of saline-injected heterozygotes at 2 weeks, whereas ammonia was normal. By 26 days, arginase activity in the treated arg(-/-) livers declined to <10%, and arginine and ornithine increased. Ammonia levels began increasing by day 25, suggesting the cause of death to be similar to that of uninjected arg(-/-) mice, albeit at a later time. These studies demonstrate that the AI deficient newborn mouse can be temporarily corrected and rescued using a HDV.


Assuntos
Adenoviridae/genética , Arginase/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Hiperamonemia/terapia , Hiperargininemia/terapia , Aminoácidos/metabolismo , Amônia/metabolismo , Animais , Animais Recém-Nascidos , Arginase/metabolismo , Arginase/fisiologia , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...