Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Med Genet A ; 152A(8): 1994-2001, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20635355

RESUMO

Angelman syndrome (AS) is caused by reduced or absent expression of the maternally inherited ubiquitin protein ligase 3A gene (UBE3A), which maps to chromosome 15q11-q13. UBE3A is subject to genomic imprinting in neurons in most regions of the brain. Expression of UBE3A from the maternal chromosome is essential to prevent AS, because the paternally inherited gene is not expressed, probably mediated by antisense UBE3A RNA. We hypothesized that increasing methylation might reduce expression of the antisense UBE3A RNA, thereby increasing UBE3A expression from the paternal gene and ameliorating the clinical phenotype. We conducted a trial using two dietary supplements, betaine and folic acid to promote global levels of methylation and attempt to activate the paternally inherited UBE3A gene. We performed a number of investigations at regular intervals including general clinical and developmental evaluations, biochemical determinations on blood and urine, and electroencephalographic studies. We report herein the data on 48 children with AS who were enrolled in a double-blind placebo-controlled protocol using betaine and folic acid for 1 year. There were no statistically significant changes between treated and untreated children; however, in a small subset of patients we observed some positive trends.


Assuntos
Síndrome de Angelman/tratamento farmacológico , Betaína/uso terapêutico , Ácido Fólico/uso terapêutico , Adolescente , Síndrome de Angelman/genética , Síndrome de Angelman/patologia , Criança , Pré-Escolar , Cromossomos Humanos Par 15/genética , Metilação de DNA , Método Duplo-Cego , Combinação de Medicamentos , Feminino , Impressão Genômica , Humanos , Lactente , Lipotrópicos/uso terapêutico , Masculino , Fenótipo , Placebos , Ubiquitina-Proteína Ligases/genética , Complexo Vitamínico B/uso terapêutico
2.
BMC Immunol ; 10: 33, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19486531

RESUMO

BACKGROUND: Arginase is significantly upregulated in the lungs in murine models of asthma, as well as in human asthma, but its role in allergic airway inflammation has not been fully elucidated in mice. RESULTS: In order to test the hypothesis that arginase has a role in allergic airway inflammation we generated arginase I-deficient bone marrow (BM) chimeric mice. Following transfer of arginase I-deficient BM into irradiated recipient mice, arginase I expression was not required for hematopoietic reconstitution and baseline immunity. Arginase I deficiency in bone marrow-derived cells decreased allergen-induced lung arginase by 85.8 +/- 5.6%. In contrast, arginase II-deficient mice had increased lung arginase activity following allergen challenge to a similar level to wild type mice. BM-derived arginase I was not required for allergen-elicited sensitization, recruitment of inflammatory cells in the lung, and proliferation of cells. Furthermore, allergen-induced airway hyperresponsiveness and collagen deposition were similar in arginase-deficient and wild type mice. Additionally, arginase II-deficient mice respond similarly to their control wild type mice with allergen-induced inflammation, airway hyperresponsiveness, proliferation and collagen deposition. CONCLUSION: Bone marrow cell derived arginase I is the predominant source of allergen-induced lung arginase but is not required for allergen-induced inflammation, airway hyperresponsiveness or collagen deposition.


Assuntos
Alérgenos/imunologia , Arginase , Células da Medula Óssea/enzimologia , Pulmão/metabolismo , Quimera por Radiação , Hipersensibilidade Respiratória/enzimologia , Animais , Arginase/imunologia , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Colágeno/metabolismo , Hiperargininemia , Imunização , Inflamação , Pulmão/imunologia , Pulmão/patologia , Camundongos , Hipersensibilidade Respiratória/patologia
3.
Mol Genet Metab ; 96(1): 44-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19036621

RESUMO

Citrin deficiency, caused by mutations in SLC25A13, can present with neonatal intrahepatic cholestasis or with adult onset neuropsychiatric, hepatic and pancreatic disease. Until recently, it had been thought to be found mostly in individuals of East Asian ancestry. A key diagnostic feature has been the deficient argininosuccinate synthetase (ASS) activity (E.C. 6.3.4.5) in liver, with normal activity in skin fibroblasts. In this series we describe the clinical presentation of 10 patients referred to our laboratories for sequence analysis of the SCL25A13 gene, including several patients who presented with elevated citrulline on newborn screening. In addition to sequence analysis performed on all patients, ASS enzyme activity, citrulline incorporation and Western blot analysis for ASS and citrin were performed on skin fibroblasts if available. We have found 5 unreported mutations including two apparent founder mutations in three unrelated French-Canadian patients. In marked contrast to previous cases, these patients have a markedly reduced ASS activity in skin fibroblasts. The presence of citrin protein on Western blot in three of our cases reduces the sensitivity of a screening test based on protein immunoblotting. The finding of citrin mutations in patients of Arabic, Pakistani, French Canadian and Northern European origins supports the concept that citrin deficiency is a panethnic disease.


Assuntos
Transtornos Congênitos do Transporte de Aminoácidos/enzimologia , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/genética , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Grupos Raciais/genética , Transtornos Congênitos do Transporte de Aminoácidos/genética , Aminoácidos/sangue , Argininossuccinato Sintase/genética , Argininossuccinato Sintase/metabolismo , Células Cultivadas , Pré-Escolar , Citrulina/metabolismo , Feminino , Fibroblastos/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Proteínas de Transporte da Membrana Mitocondrial , Mutação
4.
Physiol Genomics ; 35(1): 116-22, 2008 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-18628339

RESUMO

Obesity and its comorbidities are taking an increasing toll on human health. Key pathways that were identified with single gene variants in humans and model organisms have led to improved understanding and treatment of rare cases of human obesity. However, similar progress remains elusive for the more common multifactorial cases of metabolic dysfunction and disease. A survey of mouse chromosome substitution strains (CSSs) provided insight into the complex genetic control of diet-induced obesity and related conditions. We now report a survey of 60 traits related to obesity and metabolic syndrome in mice with a single substituted chromosome as well as selected traits measured in congenic strains derived from the substituted strain. We found that each strain that was resistant to diet-induced obesity had a distinct phenotype that uniquely modeled different combinations of traits related to metabolic disease. For example, the chromosome 6 CSS remained insulin resistant in the absence of obesity, demonstrating an atypical relationship between body weight and insulin resistance. These results provide insights into the genetic control of constant components of this mouse model of diet-induced metabolic disease as well as phenotypes that vary depending on genetic background. A better understanding of these genotype-phenotype relationships may enable a more individualized diagnosis and treatment of obesity and the metabolic syndrome.


Assuntos
Cromossomos de Mamíferos/genética , Dieta , Obesidade/genética , Adiposidade/genética , Animais , Cruzamentos Genéticos , Gorduras na Dieta , Feminino , Genótipo , Masculino , Síndrome Metabólica/genética , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Modelos Animais , Obesidade/etiologia , Fenótipo , Locos de Características Quantitativas
5.
Am J Med Genet A ; 146A(22): 2885-90, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18925679

RESUMO

Expanded newborn screening detects patients with modest elevations in citrulline; however it is currently unclear how to treat these patients and how to counsel their parents. In order to begin to address these issues, we compared the clinical, biochemical, and molecular features of 10 patients with mildly elevated citrulline levels. Three patients presented with clinical illness whereas seven came to attention as a result of expanded newborn screening. One patient presented during pregnancy and responded promptly to IV sodium phenylacetate/sodium benzoate and arginine therapy with no long-term adverse effects on mother or fetus. Two children presented with neurocognitive dysfunction, one of these responded dramatically to dietary protein reduction. ASS enzyme activity was not deficient in all patients with biallelic mutations suggesting this test cannot exclude the ASS1 locus in patients with mildly elevated plasma citrulline. Conversely, all symptomatic patients who were tested had deficient activity. We describe four unreported mutations (p.Y291S, p.R272H, p.F72L, and p.L88I), as well as the common p.W179R mutation. In silico algorithms were inconsistent in predicting the pathogenicity of mutations. The cognitive benefit in one patient of protein restriction and the lack of adverse outcome in seven others restricted from birth, suggest a role for protein restriction and continued monitoring to prevent neurocognitive dysfunction.


Assuntos
Argininossuccinato Sintase/sangue , Argininossuccinato Sintase/genética , Citrulinemia/enzimologia , Citrulinemia/genética , Adolescente , Criança , Pré-Escolar , Citrulina/sangue , Citrulinemia/diagnóstico , Citrulinemia/terapia , Análise Mutacional de DNA , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Triagem Neonatal , Gravidez , Complicações na Gravidez/enzimologia , Complicações na Gravidez/genética , Complicações na Gravidez/terapia
6.
Mol Cell Biol ; 22(13): 4491-8, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12052859

RESUMO

Deficiency of liver arginase (AI) causes hyperargininemia (OMIM 207800), a disorder characterized by progressive mental impairment, growth retardation, and spasticity and punctuated by sometimes fatal episodes of hyperammonemia. We constructed a knockout mouse strain carrying a nonfunctional AI gene by homologous recombination. Arginase AI knockout mice completely lacked liver arginase (AI) activity, exhibited severe symptoms of hyperammonemia, and died between postnatal days 10 and 14. During hyperammonemic crisis, plasma ammonia levels of these mice increased >10-fold compared to those for normal animals. Livers of AI-deficient animals showed hepatocyte abnormalities, including cell swelling and inclusions. Plasma amino acid analysis showed the mean arginine level in knockouts to be approximately fourfold greater than that for the wild type and threefold greater than that for heterozygotes; the mean proline level was approximately one-third and the ornithine level was one-half of the proline and ornithine levels, respectively, for wild-type or heterozygote mice--understandable biochemical consequences of arginase deficiency. Glutamic acid, citrulline, and histidine levels were about 1.5-fold higher than those seen in the phenotypically normal animals. Concentrations of the branched-chain amino acids valine, isoleucine, and leucine were 0.4 to 0.5 times the concentrations seen in phenotypically normal animals. In summary, the AI-deficient mouse duplicates several pathobiological aspects of the human condition and should prove to be a useful model for further study of the disease mechanism(s) and to explore treatment options, such as pharmaceutical administration of sodium phenylbutyrate and/or ornithine and development of gene therapy protocols.


Assuntos
Hiperargininemia/genética , Hiperargininemia/fisiopatologia , Animais , Animais Recém-Nascidos , Arginase/genética , Modelos Animais de Doenças , Feminino , Humanos , Rim/enzimologia , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Compostos de Amônio Quaternário/sangue
7.
Physiol Genomics ; 21(3): 404-10, 2005 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-15741506

RESUMO

Increased levels of homocysteine in the blood have been associated with various birth defects and adult diseases. However, the extent to which genetic factors control homocysteine levels in healthy individuals is unclear. Laboratory mice are valuable models for dissecting the genetic and environmental controls of total homocysteine (tHcy) levels. We assessed the inheritance of tHcy levels in two inbred strains, A/J and C57BL/6J (B6), under controlled physiological conditions and assessed the relative importance of genetic, diet, gender, and parental effects. Diet affected mean tHcy levels, whereas gender affected both the mean and variance of tHcy levels. Moreover, gender of the parents influenced mean tHcy levels in reciprocal F1 hybrids, suggesting maternal effects. Finally, gene-diet interactions affected heritability of mean tHcy levels. These studies showed that each of these factors contributes to tHcy levels and provided important clues to understanding homocysteine homeostasis in humans.


Assuntos
Homocisteína/sangue , Homocisteína/genética , Camundongos Endogâmicos A/genética , Camundongos Endogâmicos C57BL/genética , Animais , Feminino , Masculino , Camundongos , Caracteres Sexuais , Especificidade da Espécie
8.
Obstet Gynecol ; 105(5 Pt 2): 1244-6, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15863597

RESUMO

BACKGROUND: Urea cycle disorders are relatively rare but well-established causes of postpartum coma and death. Such clinical presentations have been reported previously in ornithine transcarbamylase and carbamyl phosphate synthetase deficiencies. CASE: We describe a woman, without prior symptoms of metabolic disease, who presented with hyperammonemia and psychiatric symptoms in the postpartum period. Initial diagnoses included acute fatty liver of pregnancy and postpartum psychosis. She was later found to have argininosuccinate synthetase deficiency after further metabolic investigations. Rare heterozygous mutations in the argininosuccinate synthetase gene were identified. CONCLUSION: Urea cycle disorders may present initially with postpartum psychiatric symptoms and may represent an underrecognized cause of "postpartum psychosis." We recommend obtaining metabolic studies in women with neurologic or severe psychiatric symptoms in the postpartum period.


Assuntos
Argininossuccinato Sintase/deficiência , Depressão Pós-Parto/diagnóstico , Doenças Metabólicas/diagnóstico , Adulto , Depressão Pós-Parto/enzimologia , Depressão Pós-Parto/terapia , Diagnóstico Diferencial , Feminino , Seguimentos , Humanos , Doenças Metabólicas/terapia , Período Pós-Parto , Gravidez , Transtornos Puerperais/diagnóstico , Transtornos Puerperais/enzimologia , Transtornos Puerperais/terapia , Fatores de Risco , Índice de Gravidade de Doença
11.
J Neurotrauma ; 20(10): 995-1006, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14588116

RESUMO

Traumatic brain injury causes a reduction in cerebral blood flow, which may cause additional damage to the brain. The purpose of this study was to examine the role of nitric oxide produced by endothelial nitric oxide synthase (eNOS) in these vascular effects of trauma. To accomplish this, cerebral hemodynamics were monitored in mice deficient in eNOS and wild-type control mice that underwent lateral controlled cortical impact injury followed by administration of either L-arginine, 300 mg/kg, or saline at 5 min after the impact injury. The eNOS deficient mice had a greater reduction in laser Doppler flow (LDF) in the contused brain tissue at the impact site after injury, despite maintaining a higher blood pressure. L-Arginine administration increased LDF post-injury only in the wild-type mice. L-Arginine administration also resulted in a reduction in contusion volume, from 2.4 +/- 1.5 to 1.1 +/- 1.2 mm(3) in wild-type mice. Contusion volume in the eNOS deficient mice was not significantly altered by L-arginine administration. These differences in cerebral hemodynamics between the eNOS-deficient and the wild-type mice suggest an important role for nitric oxide produced by eNOS in the preservation of cerebral blood flow in contused brain following traumatic injury, and in the improvement in cerebral blood flow with L-arginine administration.


Assuntos
Lesões Encefálicas/enzimologia , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/enzimologia , Circulação Cerebrovascular/fisiologia , Óxido Nítrico Sintase/fisiologia , Animais , Arginina/farmacologia , Arginina/uso terapêutico , Lesões Encefálicas/tratamento farmacológico , Córtex Cerebral/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase/deficiência , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III
12.
J Chromatogr B Analyt Technol Biomed Life Sci ; 791(1-2): 127-35, 2003 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-12798173

RESUMO

A protocol utilizing gas chromatography with selected ion monitoring mass spectrometric detection (GC-SIM-MS) using a simplified trimethylsilyl (TMS) derivatization protocol was developed and validated for the determination of hydroxylated metabolites of 3-keto-4-ene steroids such as testosterone, progesterone and androstenedione. Hydroxylated metabolites catalyzed by human CYP1B1 were extracted with methylene chloride and derivatized with BSTFA-10% TMCS. To get an optimal derivatizing condition, the effect of various incubation times and temperatures was evaluated. When the incubation temperature and time in the presence of the TMS derivatizing agent were increased, the 3-keto group became derivatized with TMS to form a 3-TMS derivative. To minimize the formation of the TMS ether on the 3-keto group, a reaction condition of 56 degrees C for 10 min was used for the routine measurement of the steroids and their hydroxylated metabolite. Performance studies including linearity of calibration curves, extraction efficiency and precision were performed. Linearity of the calibration curves was satisfactory from 0.125 to 5 microM for most compounds except 21-hydroxyprogesterone and 16alpha-hydroxyandrostenedione which deviated from linearity at the lower concentrations. Mean percentage extraction recoveries were greater than 80% for all compounds. Most compounds showed good precisions with C.V.s of within-day precision of less than 5% and C.V.s of between-day precision of less than 10%. The selected ion chromatograms from the recombinant human CYP1B1 incubations with testosterone, progesterone and androstenedione showed evidence of 6beta-, 16alpha-, 2alpha-, and 15alpha-hydroxytestosterone, 6alpha- and 16alpha-hydroxyprogesterone and 6alpha- and 16alpha-hydroxyandrostenedione, respectively. There was no significant interference associated with Escherichia coli membrane extracts in detecting hydroxylated metabolites. This procedure provides a rapid and sensitive method for the evaluation of steroid hydroxylation by CYP isoenzymes.


Assuntos
Hidrocarboneto de Aril Hidroxilases/metabolismo , Cromatografia Gasosa-Espectrometria de Massas/métodos , Esteroides/metabolismo , Compostos de Trimetilsilil/química , Calibragem , Catálise , Citocromo P-450 CYP1B1 , Humanos , Hidroxilação , Proteínas Recombinantes/metabolismo
13.
Am J Clin Nutr ; 93(6): 1248-54, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21490144

RESUMO

BACKGROUND: Phenylbutyrate is a drug used in patients with urea cycle disorder to elicit alternative pathways for nitrogen disposal. However, phenylbutyrate administration decreases plasma branched-chain amino acid (BCAA) concentrations, and previous research suggests that phenylbutyrate administration may increase leucine oxidation, which would indicate increased protein degradation and net protein loss. OBJECTIVE: We investigated the effects of phenylbutyrate administration on whole-body protein metabolism, glutamine, leucine, and urea kinetics in healthy and ornithine transcarbamylase-deficient (OTCD) subjects and the possible benefits of BCAA supplementation during phenylbutyrate therapy. DESIGN: Seven healthy control and 7 partial-OTCD subjects received either phenylbutyrate or no treatment in a crossover design. In addition, the partial-OTCD and 3 null-OTCD subjects received phenylbutyrate and phenylbutyrate plus BCAA supplementation. A multitracer protocol was used to determine the whole-body fluxes of urea and amino acids of interest. RESULTS: Phenylbutyrate administration reduced ureagenesis by ≈15% without affecting the fluxes of leucine, tyrosine, phenylalanine, or glutamine and the oxidation of leucine or phenylalanine. The transfer of (15)N from glutamine to urea was reduced by 35%. However, a reduction in plasma concentrations of BCAAs due to phenylbutyrate treatment was observed. BCAA supplementation did not alter the respective baseline fluxes. CONCLUSIONS: Prolonged phenylbutyrate administration reduced ureagenesis and the transfer of (15)N from glutamine to urea without parallel reductions in glutamine flux and concentration. There were no changes in total-body protein breakdown and amino acid catabolism, which suggests that phenylbutyrate can be used to dispose of nitrogen effectively without adverse effects on body protein economy.


Assuntos
Aminoácidos de Cadeia Ramificada/sangue , Glutamina/metabolismo , Nitrogênio/metabolismo , Doença da Deficiência de Ornitina Carbomoiltransferase/metabolismo , Fenilbutiratos/farmacologia , Proteínas/metabolismo , Ureia/metabolismo , Adolescente , Adulto , Idoso , Aminoácidos/metabolismo , Aminoácidos/farmacologia , Aminoácidos de Cadeia Ramificada/farmacologia , Criança , Feminino , Humanos , Masculino , Redes e Vias Metabólicas/efeitos dos fármacos , Pessoa de Meia-Idade , Doença da Deficiência de Ornitina Carbomoiltransferase/tratamento farmacológico , Fenilbutiratos/uso terapêutico , Adulto Jovem
14.
Nat Med ; 17(12): 1619-26, 2011 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-22081021

RESUMO

Nitric oxide (NO) is crucial in diverse physiological and pathological processes. We show that a hypomorphic mouse model of argininosuccinate lyase (encoded by Asl) deficiency has a distinct phenotype of multiorgan dysfunction and NO deficiency. Loss of Asl in both humans and mice leads to reduced NO synthesis, owing to both decreased endogenous arginine synthesis and an impaired ability to use extracellular arginine for NO production. Administration of nitrite, which can be converted into NO in vivo, rescued the manifestations of NO deficiency in hypomorphic Asl mice, and a nitric oxide synthase (NOS)-independent NO donor restored NO-dependent vascular reactivity in humans with ASL deficiency. Mechanistic studies showed that ASL has a structural function in addition to its catalytic activity, by which it contributes to the formation of a multiprotein complex required for NO production. Our data demonstrate a previously unappreciated role for ASL in NOS function and NO homeostasis. Hence, ASL may serve as a target for manipulating NO production in experimental models, as well as for the treatment of NO-related diseases.


Assuntos
Argininossuccinato Liase/metabolismo , Acidúria Argininossuccínica/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico/deficiência , Animais , Arginina/farmacologia , Argininossuccinato Sintase/metabolismo , Acidúria Argininossuccínica/genética , Linhagem Celular , Modelos Animais de Doenças , Células Endoteliais , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase/metabolismo , Nitritos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Análise de Sequência de DNA , Suínos
15.
Methods Mol Biol ; 603: 175-85, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20077070

RESUMO

Creatine metabolism disorders include a creatine transporter deficiency, as well as, deficiencies of two enzymes involved in creatine synthesis, arginine-glycine amidinotransferase (AGAT) and guanidinoacetate methyltransferase (GAMT). Laboratory diagnosis of these disorders relies on the determination of creatine and guanidinoacetate in both plasma and urine. Here we describe a rapid HPLC/MS/MS method for these measurements using a normal phase HILIC column after analyte derivatization.


Assuntos
Creatina/sangue , Creatina/urina , Glicina/análogos & derivados , Doenças Metabólicas/diagnóstico , Amidinotransferases/deficiência , Cromatografia Líquida de Alta Pressão/métodos , Glicina/sangue , Glicina/urina , Guanidinoacetato N-Metiltransferase/deficiência , Humanos , Doenças Metabólicas/sangue , Doenças Metabólicas/urina , Espectrometria de Massas em Tandem
16.
J Child Neurol ; 24(5): 551-6, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19225139

RESUMO

We tested the hypothesis that increasing methyl-group pools might promote transcriptional repression by other methyl-binding proteins or by mutant methyl-CpG-binding protein 2 with altered affinity, ameliorating the clinical features of Rett syndrome. A 12-month, double-blind, placebo-controlled folate-betaine trial enrolled 73 methylCpG-binding protein 2 mutation positive female participants meeting consensus criteria for Rett syndrome. Participants were randomized as young (< age 5 years) or old (>or= age 5 years). Structured clinical assessments occurred at baseline, 3, 6, and 12 months. Primary outcome measures included quantitative evaluation of breathing and hand movements during wakefulness, growth, anthropometry, motor/behavioral function, and qualitative evaluations from electroencephalograms and parent questionnaires. In all, 68 participants completed the study. Objective evidence of improvement was not found. Subjective improvement from parent questionnaires was noted for the <5 years group. This study should inform future treatment trials regarding balancing participants with specific mutations and comparable severity to minimize selection bias.


Assuntos
Betaína/uso terapêutico , Ácido Fólico/uso terapêutico , Síndrome de Rett/tratamento farmacológico , Adolescente , Adulto , Betaína/sangue , Criança , Pré-Escolar , Método Duplo-Cego , Feminino , Humanos , Lactente , Proteína 2 de Ligação a Metil-CpG/genética , Síndrome de Rett/sangue , Síndrome de Rett/genética , Resultado do Tratamento , Adulto Jovem
18.
Mol Genet Metab ; 90(2): 217-20, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17005426

RESUMO

Significant heritability has been shown for several plasma amino acid levels, but the results may have been confounded by sampling in a variety of nutritional states. We studied a group of families on a low protein steady-state diet in fasting and non-fasting states. Heritability of individual amino acids varied according to the nutritional state, suggesting the amount of genetic and environmental influences differ among the operative systems that control individual amino acid homeostasis throughout the feed/fast cycle.


Assuntos
Aminoácidos/sangue , Estado Nutricional/genética , Estudos de Casos e Controles , Deficiências Nutricionais/dietoterapia , Deficiências Nutricionais/genética , Dieta com Restrição de Proteínas , Família , Jejum , Feminino , Heterozigoto , Humanos , Masculino , Locos de Características Quantitativas , Valores de Referência
19.
Am J Physiol Cell Physiol ; 293(4): C1296-301, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17686999

RESUMO

The role of ornithine decarboxylase (ODC) in polyamine metabolism has long been established, but the exact source of ornithine has always been unclear. The arginase enzymes are capable of producing ornithine for the production of polyamines and may hold important regulatory functions in the maintenance of this pathway. Utilizing our unique set of arginase single and double knockout mice, we analyzed polyamine levels in the livers, brains, kidneys, and small intestines of the mice at 2 wk of age, the latest timepoint at which all of them are still alive, to determine whether tissue polyamine levels were altered in response to a disruption of arginase I (AI) and II (AII) enzymatic activity. Whereas putrescine was minimally increased in the liver and kidneys from the AII knockout mice, spermidine and spermine were maintained. ODC activity was not greatly altered in the knockout animals and did not correlate with the fluctuations in putrescine. mRNA levels of ornithine aminotransferase (OAT), antizyme 1 (AZ1), and spermidine/spermine-N(1)-acetyltransferase (SSAT) were also measured and only minor alterations were seen, most notably an increase in OAT expression seen in the liver of AI knockout and double knockout mice. It appears that putrescine catabolism may be affected in the liver when AI is disrupted and ornithine levels are highly reduced. These results suggest that endogenous arginase-derived ornithine may not directly contribute to polyamine homeostasis in mice. Alternate sources such as diet may provide sufficient polyamines for maintenance in mammalian tissues.


Assuntos
Arginase/genética , Poliaminas Biogênicas/metabolismo , Homeostase/fisiologia , Acetiltransferases/genética , Amina Oxidase (contendo Cobre)/genética , Animais , Encéfalo/metabolismo , Expressão Gênica , Intestino Delgado/metabolismo , Rim/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ornitina Descarboxilase/metabolismo , Ornitina-Oxo-Ácido Transaminase/genética , Proteínas/genética , Putrescina/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermidina/metabolismo , Espermina/metabolismo
20.
Mol Genet Metab ; 89(1-2): 87-96, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16753325

RESUMO

Knockout mouse models have been created to study the consequences of deficiencies in arginase AI and AII, both individually and combined. The AI knockout animals die by 14 days of age from hyperammonemia, while the AII knockout has no obvious phenotype. The double knockout (AI(-/-)/AII(-/-)) exhibits the phenotype of the AI-deficient mice, with the additional absence of AII not exacerbating the observed phenotype of the AI knockout animals. Plasma amino acid measurements in the double knockout have shown arginine levels increased roughly 100-fold and ornithine decreased roughly 10-fold as compared to wildtype. Liver ornithine levels were reduced to 2% of normal in the double knockout with arginine very highly elevated. Arginine and ornithine were also altered in other tissues in the double knockout mice, such as kidney, brain, and small intestine. This is the first demonstration that the fatal hyperammonemia in the AI knockout mouse is almost certainly due to ornithine deficiency, the amino acid needed to drive the urea cycle. Others have shown that the expression of ornithine aminotransferase (OAT) rapidly decreases in the intestine at the same age when the AI-deficient animals die, indicating that this enzyme is critical to the maintenance of ornithine homeostasis, at least at this early stage of mouse development. Although most human AI-deficient patients have no symptomatic hyperammonemia at birth, it is possible that clinically significant ornithine deficiency is already present.


Assuntos
Arginase/genética , Hiperamonemia/genética , Ornitina/deficiência , Animais , Arginase/análise , Arginina/análise , Arginina/sangue , Ácido Argininossuccínico/análise , Química Encefálica , Regulação para Baixo , Intestino Delgado/química , Intestino Delgado/enzimologia , Rim/química , Fígado/química , Camundongos , Camundongos Knockout , Ornitina/análise , Ornitina/sangue , Ornitina-Oxo-Ácido Transaminase/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA