Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Genet Metab ; 122(3): 76-84, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28711408

RESUMO

Carnitine transporter defect (CTD; also known as systemic primary carnitine deficiency; MIM 212140) is due to mutations in the SLC22A5 gene and leads to extremely low carnitine levels in blood and tissues. Affected individuals may develop early onset cardiomyopathy, weakness, or encephalopathy, which may be serious or even fatal. The disorder can be suggested by newborn screening. However, markedly low newborn carnitine levels can also be caused by conditions unrelated to CTD, such as the low carnitine levels often associated with normal pregnancies and some metabolic disorders occurring in the mother. In order to clarify the biochemical characteristics most useful for identification of CTD in newborns, we examined California Department of Public Health newborn screening data for CTD from 2005 to 12 and performed detailed chart reviews at six metabolic centers in California. The reviews covered 14 cases of newborn CTD, 14 cases of maternal disorders (CTD, 6 cases; glutaric aciduria, type 1, 5; medium-chain acyl CoA dehydrogenase deficiency, 2; and cobalamin C deficiency, 1), and 154 false-positive cases identified by newborn screening. Our results show that newborns with CTD identified by NBS exhibit different biochemical characteristics, compared to individuals ascertained clinically. Newborns with CTD may have NBS dried blood spot free carnitine near the lower cutoff and confirmatory plasma total and free carnitine levels near the normal lower limit, particularly if obtained within two weeks after birth. These findings raise the concern that true cases of CTD may exist that could have been missed by newborn screening. CTD should be considered as a possible diagnosis in cases with suggestive clinical features, even if CTD was thought to be excluded in the newborn period. Maternal plasma total carnitine and newborn urine total carnitine values are the most important predictors of true CTD in newborns. However, biochemical testing alone does not yield a discriminant rule to distinguish true CTD from low carnitine in newborns due to other causes. Because of this biochemical variability and overlap, molecular genetic testing is imperative to confirm CTD in newborns. Additionally, functional testing of fibroblast carnitine uptake remains necessary for cases in which other confirmatory testing is inconclusive. Even with utilization of all available diagnostic testing methods, confirmation of CTD ascertained by NBS remains lengthy and challenging. Incorporation of molecular analysis as a second tier step in NBS for CTD may be beneficial and should be investigated.


Assuntos
Cardiomiopatias/sangue , Cardiomiopatias/diagnóstico , Carnitina/sangue , Carnitina/deficiência , Carnitina/metabolismo , Hiperamonemia/sangue , Hiperamonemia/diagnóstico , Doenças Musculares/sangue , Doenças Musculares/diagnóstico , Triagem Neonatal/métodos , California , Cardiomiopatias/complicações , Carnitina/análise , Carnitina/química , Carnitina/urina , Teste em Amostras de Sangue Seco , Reações Falso-Positivas , Feminino , Fibroblastos/fisiologia , Humanos , Hiperamonemia/complicações , Recém-Nascido , Limite de Detecção , Masculino , Mães , Doenças Musculares/complicações , Mutação , Análise de Sequência de DNA , Membro 5 da Família 22 de Carreadores de Soluto/deficiência , Membro 5 da Família 22 de Carreadores de Soluto/genética
3.
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
4.
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
5.
Mol Genet Metab ; 90(1): 37-41, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16935537

RESUMO

Hyperargininemia is a urea cycle disorder caused by mutations in the gene for arginase I (AI) resulting in elevated blood arginine and ammonia levels. Sodium phenylacetate and a precursor, sodium phenylbutyrate (NaPB) have been used to lower ammonia, conjugating glutamine to produce phenylacetylglutamine which is excreted in urine. The elevated arginine levels induce the second arginase (AII) in patient kidney and kidney tissue culture. It has been shown that NaPB increases expression of some target genes and we tested its effect on arginase induction. Eight 9-week old male mice fed on chow containing 7.5 g NaPB/kg rodent chow and drank water with 10 g NaPB/L, and four control mice had a normal diet. After one week all mice were sacrificed. The arginase specific activities for control and NaPB mice, respectively, were 38.2 and 59.4 U/mg in liver, 0.33 and 0.42 U/mg in kidney, and 0.29 and 1.19 U/mg in brain. Immunoprecipitation of arginase in each tissue with AI and AII antibodies showed the activity induced by NaPB is mostly AI. AII may also be induced in kidney. AI accounts for the fourfold increased activity in brain. In some cell lines, NaPB increased arginase activity up to fivefold depending on dose (1-5 mM) and exposure time (2-5 days); control and NaPB activities, respectively, are: erythroleukemia, HEL, 0.06 and 0.31 U/mg, and K562, 0.46 and 1.74 U/mg; embryonic kidney, HEK293, 1.98 and 3.58 U/mg; breast adenocarcinoma, MDA-MB-468, 1.11 and 4.06 U/mg; and prostate adenocarcinoma, PC-3, 0.55 and 3.20 U/mg. In MDA-MB-468 and HEK most, but not all, of the induced activity is AI. These studies suggest that NaPB may induce AI when used to treat urea cycle disorders. It is relatively less useful in AI deficiency, although it could have some effect in those patients with missense mutations.


Assuntos
Arginase/biossíntese , Fenilbutiratos/farmacologia , Animais , Linhagem Celular , Indução Enzimática/efeitos dos fármacos , Humanos , Isoenzimas/biossíntese , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
6.
J Neurosci Res ; 66(3): 406-22, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11746358

RESUMO

The two forms of arginase (AI and AII) in man, identical in enzymatic function, are encoded in separate genes and are expressed differentially in various tissues. AI is expressed predominantly in the liver cytosol and is thought to function primarily to detoxify ammonia as part of the urea cycle. AII, in contrast, is predominantly mitochondrial, is more widely expressed, and is thought to function primarily to produce ornithine. Ornithine is a precursor in the synthesis of proline, glutamate, and polyamines. This study was undertaken to explore the cellular and regional distribution of AI and AII expression in brain using in situ hybridization and immunohistochemistry. AI and AII were detected only in neurons and not in glial cells. AI presented stronger expression than AII, but AII was generally coexpressed with AI in most cells studied. Expression was particularly high in the cerebral cortex, cerebellum, pons, medulla, and spinal cord neurons. Glutamic acid decarboxylase 65 and glutamic acid decarboxylase 67, postulated to be related to the risk of glutamate excitotoxic and/or gamma-aminobutyric acid inhibitoxic injury, were similarly ubiquitous in their expression and generally paralleled arginase expression patterns, especially in cerebral cortex, hippocampus, cerebellum, pons, medulla, and spinal cord. This study showed that AI is expressed in the mouse brain, and more strongly than AII, and sheds light on the anatomic basis for the arginine-->ornithine-->glutamate-->GABA pathway.


Assuntos
Arginase/metabolismo , Sistema Nervoso Central/enzimologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Ácido Glutâmico/metabolismo , Neurônios/enzimologia , Ornitina/biossíntese , Ácido gama-Aminobutírico/metabolismo , Animais , Arginase/genética , Arginina/metabolismo , Tronco Encefálico/citologia , Tronco Encefálico/enzimologia , Sistema Nervoso Central/citologia , Cerebelo/citologia , Cerebelo/enzimologia , DNA Complementar , Glutamato Descarboxilase/biossíntese , Imuno-Histoquímica , Isoenzimas/biossíntese , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Precipitina , Prosencéfalo/citologia , Prosencéfalo/enzimologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/citologia , Medula Espinal/enzimologia
7.
J Pediatr ; 138(1 Suppl): S21-9, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11148546

RESUMO

The urea cycle disorders (UCDs) represent a group of inherited metabolic diseases with hyperammonemia as the primary laboratory abnormality. Affected individuals may become comatose or die if not treated rapidly. Diagnosis of a UCD requires a high index of suspicion and judicious use of the laboratory. It is important to rule out other conditions causing hyperammonemia that may require different treatment. The astute clinician may suspect a specific UCD in the appropriate clinical setting, but only laboratory results can confirm a specific diagnosis. The importance of the laboratory in helping the clinician to differentiate among various causes of hyperammonemia, in confirming a specific UCD, in carrier testing, and in prenatal diagnostic testing is highlighted in this review.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Hiperamonemia/diagnóstico , Ureia/metabolismo , Algoritmos , Citrulina/metabolismo , Análise Mutacional de DNA , Humanos , Hiperamonemia/etiologia , Ácido Orótico/urina
8.
J Pediatr ; 138(1 Suppl): S72-80, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11148552

RESUMO

A survey was sent to the American members of the National Urea Cycle Disorders Foundation to ascertain the types and extent of stress imposed on families who have a child with a urea cycle defect. Forty percent of the surveys were returned. The greatest sources of stress were financial, fear of death, and the restrictions imposed by the diet. Other than removal of the economic stress and uncertainty, the results did not suggest that any specific support systems required augmentation. Instructions to mitigate frustrations occurring in emergency situations would, however, be a great help to families.


Assuntos
Adaptação Psicológica , Erros Inatos do Metabolismo dos Aminoácidos , Saúde da Família , Estresse Psicológico , Ureia/metabolismo , Adulto , Erros Inatos do Metabolismo dos Aminoácidos/psicologia , Criança , Feminino , Humanos , Masculino , Apoio Social , Estresse Psicológico/prevenção & controle , Inquéritos e Questionários
9.
Arch Biochem Biophys ; 374(2): 255-60, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10666305

RESUMO

The objective of this study was to elucidate the mechanism by which cyclic AMP increases arginase activity in cultured human Caco-2 tumor cells. Caco-2 cells were incubated for 24 h in the presence of 8-bromo cyclic AMP or forskolin, and the cells were harvested, lysed, and assayed for total arginase activity. Both test agents increased arginase activity by twofold, and this was attributed to the induction of the arginase II isoform. Both arginase II mRNA and protein showed increased expression in response to 8-bromo cyclic AMP and forskolin, and these effects were inhibited by H-89 (protein kinase A inhibitor), enhanced by okadaic acid (phosphatase inhibitor), and enhanced by 1-methyl-3-isobutylxanthine (cyclic nucleotide phosphodiesterase inhibitor). Cyclic GMP did not appear to be involved in arginase II induction. These observations indicate that cyclic AMP stimulates arginase II gene expression by mechanisms involving activation of protein kinase A and consequent activation of appropriate transcription factors.


Assuntos
8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Arginase/genética , Colforsina/farmacologia , AMP Cíclico/fisiologia , Regulação Enzimológica da Expressão Gênica , Sulfonamidas , 1-Metil-3-Isobutilxantina/farmacologia , 4-(3-Butoxi-4-metoxibenzil)-2-imidazolidinona/farmacologia , Arginase/biossíntese , Células CACO-2 , Indução Enzimática , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Isoenzimas/biossíntese , Isoenzimas/genética , Isoquinolinas/farmacologia , Cinética , Ácido Okadáico/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , RNA Mensageiro/genética , Transcrição Gênica/efeitos dos fármacos
11.
Mol Genet Metab ; 64(4): 243-9, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9758714

RESUMO

Hyperargininemia is a rare autosomal recessive disorder that results from a deficiency of hepatic type I arginase. At the genetic level, this deficiency in arginase activity is a consequence of random point mutations throughout the gene that lead to premature termination of the protein or to substitution mutations. Given the high degree of sequence homology between human liver and rat liver enzymes, we have mapped both patient and nonpatient mutations of the human enzyme onto the structure of the rat liver enzyme to rationalize the molecular basis for the low activities of these mutant arginases. Mutations identified in hyperargininemia patients affect the structure and function of the enzyme by compromising active-site residues, packing interactions in the protein scaffolding, and/or quaternary structure by destabilizing the assembly of the arginase trimer.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Arginase/genética , Arginina/sangue , Fígado/enzimologia , Mutação , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Animais , Arginase/química , Sítios de Ligação , Biopolímeros , Humanos , Estrutura Molecular , Ratos
14.
Mol Genet Metab ; 63(3): 168-75, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9608538

RESUMO

Two forms of arginase, both catalyzing the hydrolysis of arginine to ornithine and urea, are found in animals ranging from amphibians to mammals. In humans, inherited deficiency of hepatic or type I arginase results in hyperargininemia, a syndrome characterized by periodic episodes of hyperammonemia, spasticity, and neurological deterioration. In these patients, a second extrahepatic or type II arginase activity is significantly increased, an induction that may partially compensate for the lack of AI activity and apparently mitigates some of the clinical effects of the condition. Cloning and characterization of the human AII cDNA was recently accomplished. The cloning, sequencing, and partial characterization of the mouse and rat AII cDNAs are reported herein. The DNA sequences predicted polypeptides of 354 amino acids, including a N-terminal mitochondrial import signal. Sequence homology to the human type II arginase, arginase activity data, and immunoprecipitation with an anti-AII antibody confirm the identity of these cloned genes as rodent extrahepatic type II arginases.


Assuntos
Arginase/genética , Clonagem Molecular , Sequência de Aminoácidos , Animais , Arginase/química , Arginase/metabolismo , DNA Complementar , Escherichia coli/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Filogenia , Testes de Precipitina , Ratos , Ratos Sprague-Dawley , Homologia de Sequência de Aminoácidos , Ureia/metabolismo
15.
Mol Genet Metab ; 65(4): 264-71, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9889013

RESUMO

A 2-year-old female was well until 12 months of age when she was found to be anemic and had dilated cardiomyopathy. Total plasma carnitine was 6 microM and acylcarnitine analysis while receiving carnitine supplement revealed an increase in the four-carbon species. Urine organic acids were normal. In vitro analysis of the mitochondrial pathways for beta oxidation, and leucine, valine, and isoleucine metabolism was performed in fibroblasts using stable isotope-labeled precursors to these pathways followed by acylcarnitine analysis by tandem mass spectrometry. 16-2H3-palmitate was metabolized normally down to the level of butyryl-CoA thus excluding SCAD deficiency. 13C6-leucine and 13C6-isoleucine were also metabolized normally. 13C5-valine incubation revealed a significant increase in 13C4-isobutyrylcarnitine without any incorporation into propionylcarnitine as is observed normally. These same precursors were also evaluated in fibroblasts with proven ETF-QO deficiency in which acyl-CoA dehydrogenase deficiencies in each of these pathways was clearly identified. These results indicate that in the human, there is an isobutyryl-CoA dehydrogenase which exists as a separate enzyme serving only the valine pathway in addition to the 2-methyl branched-chain dehydrogenase which serves both the valine and the isoleucine pathways in both rat and human.


Assuntos
Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/deficiência , Oxirredutases/metabolismo , Valina/metabolismo , Cardiomiopatias , Carnitina/análogos & derivados , Carnitina/sangue , Carnitina/metabolismo , Carnitina/uso terapêutico , Pré-Escolar , Feminino , Fibroblastos/metabolismo , Humanos , Lactente , Masculino , Ácidos Palmíticos/metabolismo , Ácidos Palmíticos/farmacocinética , Especificidade por Substrato
16.
Neurology ; 49(6): 1655-61, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9409363

RESUMO

Inborn errors of the pyruvate dehydrogenase complex (PDC) are associated with lactic acidosis, neuroanatomic defects, developmental delay, and early death. PDC deficiency is a clinically heterogeneous disorder, with most mutations located in the coding region of the X-linked alpha subunit of the first catalytic component, pyruvate dehydrogenase (E1). Treatment of E1 deficiency hs included cofactor replacement, activation of PDC with dichloroacetate, and ketogenic diets. In this report, we describe the outcome of ketogenic diet treatment in seven boys with E1 deficiency. These patients were divided into two groups based on their mutations (R349H, three patients; and R234G, four patients, two sibling pairs). All seven patients received ketogenic diets with varying degrees of carbohydrate restriction. Clinical outcome was compared within each group and between siblings as related to the intensity and duration of dietary intervention. Subjects who either had the diet initiated earlier in life or who were placed on greater carbohydrate restriction had increased longevity and improved mental development. Based on the improved outcomes of patients with identical mutations, it appears that a nearly carbohydrate-free diet initiated shortly after birth may be useful in the treatment of E1 deficiency.


Assuntos
Corpos Cetônicos/biossíntese , Doença da Deficiência do Complexo de Piruvato Desidrogenase/dietoterapia , Doença da Deficiência do Complexo de Piruvato Desidrogenase/metabolismo , Desenvolvimento Infantil , Carboidratos da Dieta/administração & dosagem , Humanos , Recém-Nascido , Longevidade , Masculino , Mutação , Doença da Deficiência do Complexo de Piruvato Desidrogenase/genética , Resultado do Tratamento
18.
Genomics ; 38(2): 118-23, 1996 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8954792

RESUMO

There are two forms of arginase in humans, both catalyzing the hydrolysis of arginine to ornithine and urea. Recent studies in animal models and in Type I arginase-deficient patients suggest that Type II arginase is inducible and may play an important role in the regulation of extra-urea cycle arginine metabolism by modulating cellular arginine concentrations. We PCR amplified and cloned the human Type II arginase gene, the first nonliver arginase gene reported in mammals. While sequence homology to Type I arginase, arginase activity data, and immunoprecipitation with an anti-AII antibody confirm the identity of this gene, Northern blot analysis demonstrates its differential expression in the brain, prostate, and kidney. Type II arginase may be an important part of the arginine regulatory system affecting nitric oxide synthase, arginine decarboxylase, kyotorphin synthase, and arginine-glycine transaminase activities and polyamine and proline biosynthesis.


Assuntos
Arginase/genética , Sequência de Aminoácidos , Animais , Arginase/classificação , Sequência de Bases , Northern Blotting , Clonagem Molecular , DNA Complementar , Humanos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico
19.
Am J Physiol ; 271(5 Pt 2): H1988-98, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8945918

RESUMO

Rat aortic endothelial cells were found to contain both constitutive and lipopolysaccharide (LPS)-inducible arginase activity. Studies were performed to determine whether induction of nitric oxide synthase (NOS) by LPS and cytokines is accompanied by sufficient arginase induction to render arginine concentrations rate limiting for high-output NO production. Unactivated cells contained abundant arginase activity accompanied by continuous urea formation. LPS induced the formation of both inducible NOS (iNOS) and arginase, and this was accompanied by increased production of NO, citrulline, and urea. Immunoprecipitation experiments revealed the constitutive presence of arginase-I in both unactivated and LPS-activated cells and arginase-II induction by LPS. Arginase-I and iNOS were verified by reverse transcriptase-polymerase chain reaction. Induction of large amounts of iNOS by LPS plus several cytokines resulted in large quantities of NO, citrulline, and NG-hydroxy-L-arginine (NOHA), but urea production was markedly diminished. Decreased urea production was attributed to increased formation of NOHA, the precursor to NO and citrulline and a potent inhibitor of arginase-I activity with an inhibitory constant of 10-12 microM. Inhibition of iNOS activity by NG-methyl-L-arginine decreased NO and NOHA production and increased urea production. This study reveals for the first time that substantial arginase activity is present constitutively in rat aortic endothelial cells, a different isoform of arginase is induced by LPS, and intracellular arginase activity can be markedly inhibited during cytokine induction of iNOS because of NOHA formation. The inhibition of arginase activity that occurs by NOHA during marked iNOS induction may be a mechanism to ensure sufficient arginine availability for high-output production of NO.


Assuntos
Arginase/antagonistas & inibidores , Arginase/metabolismo , Arginina/análogos & derivados , Endotélio Vascular/metabolismo , Óxido Nítrico/biossíntese , Animais , Arginina/farmacologia , Células Cultivadas , Citrulina/biossíntese , Endotélio Vascular/citologia , Indução Enzimática , Masculino , Ratos , Ratos Sprague-Dawley , Ureia/antagonistas & inibidores , Ureia/metabolismo
20.
Somat Cell Mol Genet ; 22(6): 489-98, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9131018

RESUMO

As a toxic metabolic byproduct in mammals, excess ammonia is converted into urea by a series of five enzymatic reactions in the liver that constitute the urea cycle. A portion of this cycle takes place in the mitochondria, while the remainder is cytosolic. Liver arginase (L-arginine ureahydrolase, A1) is the fifth enzyme of the cycle, catalyzing the hydrolysis of arginine to ornithine and urea within the cytosol. Patients deficient in this enzyme exhibit hyperargininemia with episodic hyperammonemia and long-term effects of mental retardation and spasticity. However, the hyperammonemic effects are not so catastrophic in arginase deficiency as compared to other urea cycle defects. Earlier studies have suggested that this is due to the mitigating effect of a second isozyme of arginase (AII) expressed predominantly in the kidney and localized within the mitochondria. In order to explore the curious dual evolution of these two isozymes, and the ways in which the intriguing, aspects of AII physiology might be exploited for gene replacement therapy of AI deficiency, the cloned cDNA for human AI was inserted into an expression vector downstream from the mitochondrial targeting leader sequence for the mitochondrial enzyme ornithine transcarbamylase and transfected into a variety of recipient cell types. AI expression in the target cells was confirmed by northern blot analysis, and competition and immunoprecipitation studies showed successful translocation of the exogenous AI enzyme into the transfected cell mitochondria. Stability studies demonstrated that the translocated enzyme had a longer half-life than either native cytosolic AI or mitochondrial AII. Incubation of the transfected cells with increasing amounts of arginine produced enhanced levels of mitochondrial AI activity, a substrate-induced effect that we have previously seen with native AII but never AI. Along with exploring the basic biological questions of regulation and subcellular localization in this unique dual-enzyme system, these results suggest that the mitochondrial matrix space may be a preferred site for delivery of enzymes in gene replacement therapy.


Assuntos
Arginase/genética , Técnicas de Transferência de Genes , Terapia Genética , Mitocôndrias Hepáticas/genética , Linhagem Celular , Sistemas de Liberação de Medicamentos , Humanos , Mitocôndrias Hepáticas/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...