Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Epilepsy Behav ; 145: 109338, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37453291

RESUMO

The global birth prevalence of all inborn errors of metabolism (IEMs) in children (49 studies, 1980-2017) is approximately 50.9/100,000 live births. Regional pooled birth prevalence showed higher rates in Eastern Mediterranean regions (75.7/100,000 live births) and highest in Saudi Arabia (169/100,000) with higher parental consanguinity rates of ∼60%. Case fatality rates globally are estimated to be 33% or higher. IEMs are a group of >600 heterogeneous disorders often presenting in newborns and infants with drug-resistant seizures and/or encephalopathy. Early diagnosis and treatments are key in the prevention of morbidity, early mortality, and high lifetime health care costs, such as the early recognition of the newborn with pyridoxine- or pyridoxal-L-phosphate-dependent seizures which do not respond to standard antiepileptic drugs. The earlier the recognition and intervention in the specific cofactor- or vitamin-responsive epilepsies, the better the outcome and prevention of intractable seizures and encephalopathy leading to irreversible neurologic injury. In recent years, the genetics of IEMs has been transformed by the emergence of new molecular genetic technologies. Depending upon the clinical phenotype, current genetic testing may include chromosomal microarray (deletion/duplication analysis), single target gene sequencing, gene panels (sequencing and deletion/duplication analysis), DNA methylation analysis, mitochondrial nuclear gene panel, and mtDNA sequencing and/or trio WES or WGS (which have reduced in costs). A meta-analysis, showed WES and epilepsy gene panels to be the most cost-effective genetic tests for unknown epilepsies versus chromosomal microarray. Most recently, rapid genomic sequencing (RGS) has been associated with a shorter time to diagnosis (3 days) and increased diagnostic yield when compared with standard-of-care testing, including gene panels and microarrays. A randomized controlled trial (RCT) of rapid(r) WGS or rWES in acutely ill infants with diseases of unknown etiology in pediatric ICUs in San Diego, California found RGS to be highly clinically useful for 77% of 201 infants. RGS changed clinical management in 28% of infants and outcomes in 15%. An Australian study of ultra-rapid (ur) exome sequencing (mean time to genomic test report of 3.3 days) in 108 critically ill infants and children with suspected monogenic conditions, had a molecular diagnostic yield of 51% with 20% requiring further genetic analysis. In 42/55 (76%), ur exome sequencing was felt to have influenced clinical management for targeted treatments, surveillance, or palliative care, however, the study was not designed or powered to measure differences in major clinical outcomes compared to standard care of critically ill patients. Further research is needed to understand this tool's clinical value and generalizability balanced against its high costs. A paradigm shift is evolving from pattern- and evidence-based medicine toward algorithm-based, precision medicine targeted to individual mutations. Meticulous clinical phenotyping and pedigree analysis, combined with advances in high-throughput metabolomics, proteomics, transcriptomics (RNAseq in clinically relevant tissues), and genomics, have expedited the identification of novel pathomechanisms and new therapeutic targets. Evaluation of these therapies in IEMs, many of which manifest with encephalopathy and epilepsy, will depend on international registries of well-characterized phenotypes in RCTs and measurement of clinically relevant endpoints. The earlier the recognition and diagnosis and intervention with targeted therapies, the better the overall outcome in terms of the impact on intellectual disability and the effective management of the associated epilepsy.


Assuntos
Encefalopatias , Epilepsia , Humanos , Estado Terminal , Austrália , Testes Genéticos , Epilepsia/diagnóstico , Epilepsia/genética , Encefalopatias/diagnóstico , Encefalopatias/genética , Convulsões/genética , Ensaios Clínicos Controlados Aleatórios como Assunto
2.
Mol Genet Metab ; 129(3): 213-218, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31864849

RESUMO

Carnitine Uptake Defect (CUD) is an autosomal recessive disorder due to mutations in the SLC22A5 gene. Classically patients present in infancy with profound muscle weakness and cardiomyopathy with characteristic EKG findings. Later presentations include recurrent hypoketotic hypoglycemia, proximal limb girdle myopathy,and/or recurrent muscle pain. Newborn screening detects most of these clinical variants but in addition has identified maternal CUD often in asymptomatic women. We describe a family ascertained through 3 newborn screening (NBS) positive infants found to be unaffected themselves but in whom the mothers (sisters) were affected. There were also two affected children born to an affected male and his heterozygous wife who were false negatives on NBS but had increased fractional excretion of free carnitine in the urine. Analysis on a Next Generation Sequencing panel specifically designed to fully cover newborn screening disease targets showed a homozygous change in the five probands (SLC22A5; NM_003060:c.-149G > A; p.?). The mutation segregates with the CUD within the family. It is in the 5' UTR and has a frequency within the gnomAd database of 0.001198. Plasma carnitine was decreased and fractional excretion of free carnitine was increased in all affected individuals. Functional carnitine uptake studies in cultured skin fibroblasts of one proband showed carnitine uptake at the 5 µM concentration to be 6% of controls. Relative expression of OCTN2 mRNA to beta-actin mRNA by qRT-PCR was increased in a proband relative to controls by a factor of 465-fold. Western blotting revealed a 120 kDa protein band, as well as a weaker 240 kDa band in the proband, the significance of which is unknown at this time.


Assuntos
Regiões 5' não Traduzidas/genética , Cardiomiopatias/diagnóstico , Cardiomiopatias/genética , Carnitina/sangue , Carnitina/deficiência , Hiperamonemia/diagnóstico , Hiperamonemia/genética , Doenças Musculares/diagnóstico , Doenças Musculares/genética , Membro 5 da Família 22 de Carreadores de Soluto/genética , Actinas/genética , Actinas/metabolismo , Transporte Biológico Ativo/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/fisiopatologia , Carnitina/genética , Carnitina/metabolismo , Células Cultivadas , Criança , Pré-Escolar , Feminino , Fibroblastos/metabolismo , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Homozigoto , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/fisiopatologia , Lactente , Recém-Nascido , Masculino , Doenças Musculares/metabolismo , Doenças Musculares/fisiopatologia , Mutação , Triagem Neonatal , Linhagem , Pele/citologia , Pele/metabolismo , Membro 5 da Família 22 de Carreadores de Soluto/metabolismo , Sequenciamento do Exoma
3.
J Inherit Metab Dis ; 40(3): 403-414, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28303425

RESUMO

OBJECTIVES: The common data elements (CDE) project was developed by the National Institute of Neurological Disorders and Stroke (NINDS) to provide clinical researchers with tools to improve data quality and allow for harmonization of data collected in different research studies. CDEs have been created for several neurological diseases; the aim of this project was to develop CDEs specifically curated for mitochondrial disease (Mito) to enhance clinical research. METHODS: Nine working groups (WGs), composed of international mitochondrial disease experts, provided recommendations for Mito clinical research. They initially reviewed existing NINDS CDEs and instruments, and developed new data elements or instruments when needed. Recommendations were organized, internally reviewed by the Mito WGs, and posted online for external public comment for a period of eight weeks. The final version was again reviewed by all WGs and the NINDS CDE team prior to posting for public use. RESULTS: The NINDS Mito CDEs and supporting documents are publicly available on the NINDS CDE website ( https://commondataelements.ninds.nih.gov/ ), organized into domain categories such as Participant/Subject Characteristics, Assessments, and Examinations. CONCLUSION: We developed a comprehensive set of CDE recommendations, data definitions, case report forms (CRFs), and guidelines for use in Mito clinical research. The widespread use of CDEs is intended to enhance Mito clinical research endeavors, including natural history studies, clinical trial design, and data sharing. Ongoing international collaboration will facilitate regular review, updates and online publication of Mito CDEs, and support improved consistency of data collection and reporting.


Assuntos
Elementos de Dados Comuns/normas , Doenças Mitocondriais/patologia , Doenças do Sistema Nervoso/patologia , Acidente Vascular Cerebral/patologia , Pesquisa Biomédica/normas , Coleta de Dados/normas , Humanos , National Institute of Neurological Disorders and Stroke (USA) , Projetos de Pesquisa/normas , Estados Unidos
6.
Muscle Nerve ; 46(5): 767-72, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23055315

RESUMO

INTRODUCTION: Muscle and cardiac metabolism are dependent on the oxidation of fats and glucose for adenosine triphosphate production, for which L-carnitine is an essential cofactor. METHODS: We measured muscle carnitine concentrations in skeletal muscles, diaphragm, and ventricles of C57BL/10ScSn-DMDmdx/J mice (n = 10) and compared them with wild-type C57BL/6J (n = 3), C57BL/10 (n = 10), and C3H (n = 12) mice. Citrate synthase (CS) activity was measured in quadriceps/gluteals and ventricles of mdx and wild-type mice. RESULTS: We found significantly lower tissue carnitine in quadriceps/gluteus (P < 0.05) and ventricle (P < 0.05), but not diaphragm of mdx mice, when compared with controls. CS activity was increased in mdx quadriceps/gluteus (P < 0.03) and ventricle (P < 0.02). This suggests compensatory mitochondrial biogenesis. CONCLUSIONS: Decreased tissue carnitine has implications for reduced fatty acid and glucose oxidation in mdx quadriceps/gluteus and ventricle. The mdx mouse may be a useful model for studying the role of muscle carnitine deficiency in DMD bioenergetic insufficiency and providing a targeted and timed rationale for L-carnitine therapy.


Assuntos
Carnitina/deficiência , Modelos Animais de Doenças , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Deficiência de Vitaminas do Complexo B/genética , Deficiência de Vitaminas do Complexo B/metabolismo , Animais , Carnitina/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/etiologia , Músculo Quadríceps/fisiopatologia , Deficiência de Vitaminas do Complexo B/complicações
7.
Neurohospitalist ; 12(1): 67-73, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34950389

RESUMO

Mitochondrial encephalomyopathy, lactic acidosis and stroke like episodes (MELAS) syndrome is a maternally inherited mitochondrial disorder with recurrent non-arterial distribution stroke-like episodes (SLEs). A 17 yr old boy with MELAS (m.3243A>G tRNALeu(UUR)) presented with SLEs at ages 8 and 10 yrs. At 11 yrs, he suffered a third SLE involving left parietotemporal lobes with dense right hemiplegia and aphasia persistent for 1 week without improvement. On high dose IV L-Arginine (L-Arg) (0.5 g/kg/day divided TID) he had rapid recovery within 48 hours and was rapidly weaned. With emesis of oral L-Arg, his SLE recurred and he was again treated with high dose IV L-Arg with similar rapid recovery and discharged on a slow wean over 6 wks to 0.1 g/kg/day. On maintenance L-Arg he suffered only 2 SLEs at ages 13 and 16 yrs; both resolved rapidly with high dose IV L-Arg without recurrence during a slow wean to maintenance. His phenotype included seizures, ptosis, ophthalmoplegia, facial diplegia, sensorineural hearing loss, ataxia, myopathy, exercise intolerance, peripheral sensorimotor neuropathy, hypertrophic cardiomyopathy, hypertension, and failure to thrive. At 16 yrs he developed end-stage renal disease, due to MELAS, requiring hemodialysis and at 17 yrs he underwent cadaveric renal transplantation. His peri-operative protocol included strict maintenance of perfusion, oxygenation, normothermia, biochemical homeostasis and serum arginine concentrations during which time there were no neurologic decompensations. He was transitioned to oral L-citrulline maintenance therapy which maintained higher serum arginine concentrations with better tolerance. He had no SLEs or seizures in the ensuing 2 yrs.

8.
Biochem Biophys Res Commun ; 404(4): 1010-5, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21187065

RESUMO

BACKGROUND: Transport of L-carnitine, essential cofactor of fatty acid metabolism, into breast milk is critical for the normal growth and development of the suckling infant. OBJECTIVE: To increase understanding of developmental expression of carnitine/organic cation (Octn) transporter family at different stages of murine breast development for carnitine delivery. METHODS: We applied our transporter-specific antibodies to mOctn1, mOctn2 and mOctn3 to sections of mammary glands of virginal non-lactating, pregnant, late lactating and post-lactating C3H females. RESULTS: We demonstrated differential expression of mOctn1, -2 and -3 in epithelial ducts, specialized myoepithelial cells and fatty stroma. There was notable upregulation of all three Octns and mRNA by RT-PCR concurrent with an increase in epithelial ducts in breasts of pregnant (15days gestation) and lactating mice (15-days post-partum) compared to virginal 6 week old females, and notable downregulation in expression of Octns 15 days after cessation of lactation. In lactating murine mammary gland at 15 days post-partum, there was a marked increase of fat globules in epithelial ducts. Octn1 and Octn2 had similar expression patterns in lactating gland cells which formed fat globules that were exocytosed into the lumen of alveoli along with transporters Octn1 and Octn2. Octn3 was primarily localized to myoepithelial cells surrounding the ducts at all stages of breast development. CONCLUSIONS: There is a dynamic upregulation of the Octn family in pregnant and lactating breasts which likely provides the suckling infant with adequate carnitine for the rapid postnatal upregulation of fatty acid oxidation and ketogenesis critical for cerebral energy metabolism during fasting hypoglycemia.


Assuntos
Carnitina/metabolismo , Homeostase , Lactação , Glândulas Mamárias Animais/crescimento & desenvolvimento , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Animais , Animais Lactentes , Feminino , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Proteínas de Transporte de Cátions Orgânicos/genética , Regulação para Cima
9.
Mol Genet Metab ; 104(1-2): 48-60, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21704546

RESUMO

Antiquitin (ATQ) deficiency is the main cause of pyridoxine dependent epilepsy characterized by early onset epileptic encephalopathy responsive to large dosages of pyridoxine. Despite seizure control most patients have intellectual disability. Folinic acid responsive seizures (FARS) are genetically identical to ATQ deficiency. ATQ functions as an aldehyde dehydrogenase (ALDH7A1) in the lysine degradation pathway. Its deficiency results in accumulation of α-aminoadipic semialdehyde (AASA), piperideine-6-carboxylate (P6C) and pipecolic acid, which serve as diagnostic markers in urine, plasma, and CSF. To interrupt seizures a dose of 100 mg of pyridoxine-HCl is given intravenously, or orally/enterally with 30 mg/kg/day. First administration may result in respiratory arrest in responders, and thus treatment should be performed with support of respiratory management. To make sure that late and masked response is not missed, treatment with oral/enteral pyridoxine should be continued until ATQ deficiency is excluded by negative biochemical or genetic testing. Long-term treatment dosages vary between 15 and 30 mg/kg/day in infants or up to 200 mg/day in neonates, and 500 mg/day in adults. Oral or enteral pyridoxal phosphate (PLP), up to 30 mg/kg/day can be given alternatively. Prenatal treatment with maternal pyridoxine supplementation possibly improves outcome. PDE is an organic aciduria caused by a deficiency in the catabolic breakdown of lysine. A lysine restricted diet might address the potential toxicity of accumulating αAASA, P6C and pipecolic acid. A multicenter study on long term outcomes is needed to document potential benefits of this additional treatment. The differential diagnosis of pyridoxine or PLP responsive seizure disorders includes PLP-responsive epileptic encephalopathy due to PNPO deficiency, neonatal/infantile hypophosphatasia (TNSALP deficiency), familial hyperphosphatasia (PIGV deficiency), as well as yet unidentified conditions and nutritional vitamin B6 deficiency. Commencing treatment with PLP will not delay treatment in patients with pyridox(am)ine phosphate oxidase (PNPO) deficiency who are responsive to PLP only.


Assuntos
Aldeído Desidrogenase/deficiência , Epilepsia/diagnóstico , Epilepsia/terapia , Guias de Prática Clínica como Assunto , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Biomarcadores/metabolismo , Epilepsia/genética , Epilepsia/fisiopatologia , Seguimentos , Humanos , Vitamina B 6/uso terapêutico
10.
Ann Neurol ; 67(6): 802-8, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20517942

RESUMO

OBJECTIVE: Individuals with cystic fibrosis (CF) have exercise intolerance and skeletal muscle weakness not solely attributable to physical inactivity or pulmonary function abnormalities. CF transmembrane conductance regulator (CFTR) has been demonstrated in human bronchial smooth and cardiac muscle. Using (31)P-magnetic resonance spectroscopy of skeletal muscle, we showed CF patients to have lower resting muscle adenosine triphosphate and delayed phosphocreatine recovery times after high-intensity exercise, suggesting abnormal muscle aerobic metabolism; and higher end-exercise pH values, suggesting altered bicarbonate transport. Our objective was to study CFTR expression in human skeletal muscle. METHODS AND RESULTS: We studied CFTR expression in human skeletal muscle by Western blot with anti-CFTR antibody (Ab) L12B4 and demonstrated a single band with expected molecular weight of 168kDa. We isolated the cDNA by reverse transcription polymerase chain reaction and directly sequenced a 975bp segment (c. 3,600-4,575) that was identical to the human CFTR sequence. We showed punctate staining of CFTR in sarcoplasm and sarcolemma by immunofluorescence microscopy with L12B4 Ab and secondary Alexa 488-labeled Ab. We confirmed CFTR expression in the sarcotubular network and sarcolemma by electron microscopy, using immunogold-labeled anti-CFTR Ab. We observed activation of CFTR Cl(-) channels with iodide efflux, on addition of forskolin, 3-isobutyl-1-methyl-xanthine, and 8-chlorphenylthio-cyclic adenosine monophosphate, in wild-type C57BL/6J isolated muscle fibers in contrast to no efflux from mutant F508del-CFTR muscle. INTERPRETATION: We speculate that a defect in sarcoplasmic reticulum CFTR Cl(-) channels could alter the electrochemical gradient, causing dysregulation of Ca(2+) homeostasis, for example, ryanodine receptor or sarco(endo)plasmic reticulum Ca(2+) adenosine triphosphatases essential to excitation-contraction coupling leading to exercise intolerance and muscle weakness in CF.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/patologia , Fibrose Cística/fisiopatologia , Exercício Físico/fisiologia , Músculo Esquelético/metabolismo , Animais , Colforsina/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Imunoeletrônica/métodos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Mutação/genética , Inibidores de Fosfodiesterase/farmacologia , Sarcolema/metabolismo , Sarcolema/ultraestrutura , Frações Subcelulares/metabolismo
11.
Pediatr Res ; 69(1): 40-5, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20938370

RESUMO

Previous studies have reported differences in muscle function and metabolism between patients with cystic fibrosis (CF) and healthy controls (HC), but it is currently unknown whether these abnormalities are specific to CF or also seen in other airway diseases. In this study, we used magnetic resonance spectroscopy (MRS) during exercise to assess muscle metabolism in CF patients. Twenty patients with CF and 20 age, gender, and habitual activity-matched HCs and a respiratory disease comparison group with primary ciliary dyskinesia (PCD; n = 10) were studied. Phosphorus MRS (P-MRS) was used to characterize muscle bioenergetic metabolism at rest and after high-, moderate-, and low-intensity exercise. CF patients exhibited lower resting ATP/phosphocreatine (PCr) ratio and significantly higher end-exercise pH values compared with both HC and PCD patients. Both CF and PCD patients demonstrated significantly slower PCr recovery time constants after high-intensity exercise. Our results suggest that not only there are specific abnormalities of muscle metabolism in CF patients but also there is a nonspecific impact of respiratory disease on muscle function.


Assuntos
Fibrose Cística/metabolismo , Metabolismo Energético/fisiologia , Exercício Físico/fisiologia , Síndrome de Kartagener/metabolismo , Músculo Esquelético/metabolismo , Trifosfato de Adenosina/metabolismo , Adolescente , Feminino , Humanos , Concentração de Íons de Hidrogênio , Análise dos Mínimos Quadrados , Espectroscopia de Ressonância Magnética , Masculino , Fosfocreatina/metabolismo
13.
J Rheumatol ; 48(3): 434-441, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32739897

RESUMO

OBJECTIVE: To evaluate the feasibility of studying creatine in juvenile dermatomyositis (JDM). Secondary objectives were to determine the effect of creatine on muscle function and metabolism, aerobic capacity, fatigue, physical activity, and quality of life (QOL), as well as its safety. METHODS: We conducted a 6-month, double-blind, randomized, multiple-baseline design; patients were assigned to creatine or placebo. Feasibility was assessed using attended study visits, completed study procedures, and adherence. Muscle function, aerobic capacity, and muscle strength were assessed with standardized exercise tests. Muscle metabolism was assessed using a 31-Phosphorus Magnetic Resonance Spectroscopy protocol. Fatigue, physical activity, and QOL were assessed by questionnaires. Statistical significance was estimated using a randomization (permutation) test. Changes in outcome measures taken at baseline and end-of-study were calculated using paired t-tests. RESULTS: Median (range) adherence to the study drug was 88.5% (20.5-95.5%) and the proportion of subjects with 80% adherence or higher was 76.9%. There were no missed study visits. There were no statistically significant changes in muscle function, strength, aerobic capacity, disease activity, fatigue, physical activity, or QOL while subjects were receiving creatine compared to placebo. There were statistically significant adaptations in muscle metabolism (e.g., decrease in change in muscle pH following exercise, and decrease in phosphate/phosphocreatine ratio) at the end-of-study compared to baseline. There were no significant adverse effects. CONCLUSION: Creatine supplementation in children with JDM is feasible to study, and is safe and well-tolerated; it may lead to improvements in muscle metabolism.


Assuntos
Creatina , Miosite , Criança , Suplementos Nutricionais , Método Duplo-Cego , Estudos de Viabilidade , Humanos , Força Muscular , Músculo Esquelético , Músculos , Qualidade de Vida
14.
J Inherit Metab Dis ; 33(3): 211-22, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20443061

RESUMO

Mitochondrial dysfunction and oxidative stress are central to the molecular basis of several human diseases associated with neuromuscular disabilities. We hypothesize that mitochondrial dysfunction also contributes to the neuromuscular symptoms observed in patients with ethylmalonic aciduria and homozygosity for ACADS c.625G>A-a common variant of the short-chain acyl-coenzyme A (CoA) dehydrogenase (SCAD) enzyme in the mitochondrial fatty acid oxidation pathway. This study sought to identify the specific factors that initiate cell dysfunction in these patients. We investigated fibroblast cultures from 10 patients with neuromuscular disabilities, elevated levels of ethylmalonic acid (EMA) (>50 mmol/mol creatinine), and ACADS c.625G>A homozygosity. Functional analyses, i.e., ACADS gene and protein expression as well as SCAD enzyme activity measurements, were performed together with a global nano liquid chromatography tandem mass spectroscopy (nano-LC-MS/MS)-based screening of the mitochondrial proteome in patient fibroblasts. Moreover, cell viability of patient fibroblasts exposed to menadione-induced oxidative stress was evaluated. Loss of SCAD function was detected in the patient group, most likely due to decreased ACADS gene expression and/or elimination of misfolded SCAD protein. Analysis of the mitochondrial proteome in patient fibroblasts identified a number of differentially expressed protein candidates, including reduced expression of the antioxidant superoxide dismutase 2 (SOD2). Additionally, patient fibroblasts demonstrated significantly higher sensitivity to oxidative stress than control fibroblasts. We propose that reduced mitochondrial antioxidant capacity is a potential risk factor for ACADS c.625G>A-associated ethylmalonic aciduria and that mitochondrial dysfunction contributes to the neurotoxicity observed in patients.


Assuntos
Antioxidantes/metabolismo , Malonatos/urina , Erros Inatos do Metabolismo/diagnóstico , Butiril-CoA Desidrogenase/genética , Sobrevivência Celular , Pré-Escolar , Cromatografia Líquida/métodos , Fibroblastos/metabolismo , Homozigoto , Humanos , Espectrometria de Massas/métodos , Erros Inatos do Metabolismo/genética , Mitocôndrias/metabolismo , Modelos Genéticos , Estresse Oxidativo , Proteômica/métodos , Fatores de Risco
16.
Clin Chim Acta ; 505: 92-97, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32070725

RESUMO

INTRODUCTION: Carnitine is essential for long-chain fatty acid oxidation in muscle and heart. Tissue stores are regulated by organic cation/Cn transporter plasmalemmal Octn2. We previously demonstrated low carnitine in quadriceps/gluteus and heart of adult mdx mice. METHODS: We studied protein and mRNA expression of Octn2, mitochondrial Octn1 and peroxisomal Octn3 in adult male C57BL/10ScSn-DMD mdx/J quadriceps, heart, and diaphragm compared to C57BL/10SnJ mice. RESULTS: We demonstrated reduction in mOctn2 expression on Western blot and similar expression of mOctn1 and mOctn3 in mdx quadriceps, heart and diaphragm. There was a significant upregulation of mOctn1 and mOctn2 mRNA by qRT-PCR in mdx quadriceps and of mOctn2 and mOctn3 mRNA in mdx heart. We showed upregulation of mdx mOctn1 and mOctn3 mRNA but no increase in protein expression. DISCUSSION: Dystrophin deficiency likely disrupts Octn2 expression decreasing muscle carnitine uptake thus contributing to membranotoxic long-chain acyl-CoAs with sarcolemmal and organellar membrane oxidative injury providing a treatment rationale for early L-carnitine in DMD.


Assuntos
Carnitina/química , Carnitina/uso terapêutico , Músculo Esquelético/química , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Miocárdio/química , Proteínas de Transporte de Cátions Orgânicos/biossíntese , Proteínas de Transporte de Cátions Orgânicos/genética , Membro 5 da Família 22 de Carreadores de Soluto/biossíntese , Membro 5 da Família 22 de Carreadores de Soluto/genética , Simportadores/biossíntese , Simportadores/genética , Animais , Carnitina/metabolismo , Diafragma/metabolismo , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
17.
PLoS One ; 15(9): e0238224, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32881886

RESUMO

OBJECTIVE: We previously showed that MELAS patients have decreased cerebrovascular reactivity (CVR) (p≤ 0.002) and increased cerebral blood flow (CBF) (p<0.0026); changes correlated with disease severity and % mutant mtDNA (inversely for CVR; directly for CBF). We ran a prospective pilot in 3 MELAS sibs (m.3243A>G tRNALeu(UUR)) with variable % mutant blood mtDNA to assess effects of L-Arginine (L-Arg) (single dose and 6-wk steady-state trial) on regional CBF, arterial CVR and neurovascular coupling. METHODS: Patients were studied with 3T MRI using arterial spin labeling (ASL) to measure CBF and changes in % Blood Oxygen Level Dependent (BOLD) signal to changes in arterial partial pressure of CO2 to measure CVR. Task fMRI consisted of an alternating black and white checkerboard to evaluate visual cortex response in MELAS and controls. RESULTS: Following L-Arg, there was restoration of serum Arg (76-230 µM) in MELAS sibs and a trend towards increasing CVR in frontal and corresponding decrease in occipital cortex; CVR was unchanged globally. There was a 29-37% reduction in baseline CBF in one patient following 6 wks of L-Arg. Pre-treatment fMRI activation in response to visual cortex stimulus was markedly decreased in the same patient compared to controls in primary visual striate cortex V1 and extrastriate regions V2 to V5 with a marked increase toward control values following a single dose and 6 wks of L-Arg. CONCLUSION: Proposed "healing" effect may be due to more efficient utilization of energy substrates with increased cellular energy balances and ensuing reduction in signalling pathways that augment flow in the untreated state. CLASSIFICATION OF EVIDENCE: This prospective pilot study provides Class III evidence that oral L-Arginine (100 mg/kg single dose or 100 mg/kg three times daily po X 6 weeks) normalizes resting blood flow from elevated pre-treatment levels in patients with MELAS syndrome, selectively increases their CVR from reduced pre-treatment levels in regions most impaired at the expense of less abnormal regions, and normalizes reduced BOLD fMRI activation in response to visual cortex stimulus. CLINICAL TRIALS.GOV (NIH): NCT01603446.


Assuntos
Arginina/uso terapêutico , Circulação Cerebrovascular/fisiologia , Síndrome MELAS/tratamento farmacológico , Acoplamento Neurovascular/fisiologia , Administração Oral , Adolescente , Arginina/sangue , Arginina/farmacologia , Encéfalo/irrigação sanguínea , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico , Dióxido de Carbono/sangue , Circulação Cerebrovascular/efeitos dos fármacos , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Acoplamento Neurovascular/efeitos dos fármacos , Ornitina/sangue , Oxigênio/sangue , Projetos Piloto , Estudos Prospectivos , Resultado do Tratamento , Córtex Visual/efeitos dos fármacos , Adulto Jovem
18.
Paediatr Respir Rev ; 10(3): 83-90, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19651377

RESUMO

A complex series of metabolic pathways are present in human muscle that break down substrates from nutritional sources to produce energy for different types of muscular activity. However, depending on the activity in which an individual is engaged, the body will make use of different energy systems that have been adapted for the particular activity. More specifically, utilization of bioenergetic substrates depends on the type, intensity, and duration of the exercise. The aerobic oxidative system is used for longer duration activities of low to moderate intensity, the anaerobic glycolytic system is used for short to moderate duration activities of higher intensity, and the high energy phosphagen system is used for short duration activities of high intensity. The efficiency and effectiveness of these pathways can be enhanced through physical activity and training. It is these bioenergetic pathways that are the focus of this review.


Assuntos
Metabolismo Energético/fisiologia , Exercício Físico/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Adaptação Fisiológica , Criança , Ciclo do Ácido Cítrico , Glicólise/fisiologia , Humanos , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas/ultraestrutura , Sarcômeros/fisiologia
19.
Hum Genet ; 124(1): 43-56, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18523805

RESUMO

Short-chain acyl-CoA dehydrogenase (SCAD) deficiency is an inherited disorder of mitochondrial fatty acid oxidation associated with variations in the ACADS gene and variable clinical symptoms. In addition to rare ACADS inactivating variations, two common variations, c.511C > T (p.Arg171Trp) and c.625G > A (p.Gly209Ser), have been identified in patients, but these are also present in up to 14% of normal populations leading to questions of their clinical relevance. The common variant alleles encode proteins with nearly normal enzymatic activity at physiological conditions in vitro. SCAD enzyme function, however, is impaired at increased temperature and the tendency to misfold increases under conditions of cellular stress. The present study examines misfolding of variant SCAD proteins identified in patients with SCAD deficiency. Analysis of the ACADS gene in 114 patients revealed 29 variations, 26 missense, one start codon, and two stop codon variations. In vitro import studies of variant SCAD proteins in isolated mitochondria from SCAD deficient (SCAD-/-) mice demonstrated an increased tendency of the abnormal proteins to misfold and aggregate compared to the wild-type, a phenomenon that often leads to gain-of-function cellular phenotypes. However, no correlation was found between the clinical phenotype and the degree of SCAD dysfunction. We propose that SCAD deficiency should be considered as a disorder of protein folding that can lead to clinical disease in combination with other genetic and environmental factors.


Assuntos
Butiril-CoA Desidrogenase/genética , Erros Inatos do Metabolismo/genética , Mutação de Sentido Incorreto/fisiologia , Dobramento de Proteína , Animais , Butiril-CoA Desidrogenase/química , Butiril-CoA Desidrogenase/metabolismo , Butiril-CoA Desidrogenase/fisiologia , Dimerização , Ativação Enzimática/genética , Frequência do Gene , Humanos , Malonatos/metabolismo , Malonatos/urina , Erros Inatos do Metabolismo/enzimologia , Erros Inatos do Metabolismo/urina , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Modelos Moleculares , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Relação Estrutura-Atividade
20.
Mol Genet Metab ; 94(4): 422-427, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550408

RESUMO

Three distinct clinical manifestations of carnitine palmitoyltransferase II (CPT II) deficiency have been defined including a mild adult onset myopathy, a severe infantile disorder and a lethal neonatal form. In this study we have examined the genomic DNA of five patients, 3 with the lethal neonatal form and 2 with the severe infantile form of the disease and identified two disease-causing mutations in the CPT2 gene for each patient, three of which are novel. In addition, based on currently available structural, biochemical and clinical data, we have classified all 64 known disease-causing mutations into groups with different predicted phenotypes depending on their CPT2 allelic counterparts.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Carnitina O-Palmitoiltransferase/deficiência , Carnitina O-Palmitoiltransferase/genética , Erros Inatos do Metabolismo/genética , Mutação , Erros Inatos do Metabolismo dos Aminoácidos/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Sequência de Bases , Carnitina O-Palmitoiltransferase/química , Evolução Fatal , Feminino , Humanos , Recém-Nascido , Masculino , Erros Inatos do Metabolismo/enzimologia , Doenças Musculares/enzimologia , Doenças Musculares/genética , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA