Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Clin Invest ; 133(10)2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36951944

RESUMO

Sphingolipids function as membrane constituents and signaling molecules, with crucial roles in human diseases, from neurodevelopmental disorders to cancer, best exemplified in the inborn errors of sphingolipid metabolism in lysosomes. The dihydroceramide desaturase Δ4-dihydroceramide desaturase 1 (DEGS1) acts in the last step of a sector of the sphingolipid pathway, de novo ceramide biosynthesis. Defects in DEGS1 cause the recently described hypomyelinating leukodystrophy-18 (HLD18) (OMIM #618404). Here, we reveal that DEGS1 is a mitochondria-associated endoplasmic reticulum membrane-resident (MAM-resident) enzyme, refining previous reports locating DEGS1 at the endoplasmic reticulum only. Using patient fibroblasts, multiomics, and enzymatic assays, we show that DEGS1 deficiency disrupts the main core functions of the MAM: (a) mitochondrial dynamics, with a hyperfused mitochondrial network associated with decreased activation of dynamin-related protein 1; (b) cholesterol metabolism, with impaired sterol O-acyltransferase activity and decreased cholesteryl esters; (c) phospholipid metabolism, with increased phosphatidic acid and phosphatidylserine and decreased phosphatidylethanolamine; and (d) biogenesis of lipid droplets, with increased size and numbers. Moreover, we detected increased mitochondrial superoxide species production in fibroblasts and mitochondrial respiration impairment in patient muscle biopsy tissues. Our findings shed light on the pathophysiology of HLD18 and broaden our understanding of the role of sphingolipid metabolism in MAM function.


Assuntos
Oxirredutases , Esfingolipídeos , Humanos , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Oxirredutases/metabolismo , Esfingolipídeos/metabolismo
2.
Pediatr Neurol ; 119: 40-44, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33894639

RESUMO

BACKGROUND: The ATP7A gene encodes a copper transporter whose mutations cause Menkes disease, occipital horn syndrome (OHS), and, less frequently, ATP7A-related distal hereditary motor neuropathy (dHMN). Here we describe a family with OHS caused by a novel mutation in the ATP7A gene, including a patient with a comorbid dHMN that worsened markedly after being treated with copper histidinate. METHODS: We studied in detail the clinical features of the patients and performed a genomic analysis by using TruSight One Expanded Sequencing Panel. Subsequently, we determined the ATP7A and ATP7B expression levels, mitochondrial membrane potential, and redox balance in cultured fibroblasts of Patient 1. RESULTS: We found a novel ATP7A late truncated mutation p.Lys1412AsnfsX15 in the two affected members of this family. The co-occurrence of OHS and dHMN in Patient 1 reveals the variable phenotypic expressivity of the variant. A severe clinical and neurophysiologic worsening was observed in the dHMN of Patient 1 when he was treated with copper replacement therapy, with a subsequent fast recovery after the copper histidinate was withdrawn. Functional studies revealed that the patient had low levels of both ATP7A and ATP7B, the other copper transporter, and high levels of superoxide ion in the mitochondria. CONCLUSIONS: Our findings broaden the clinical spectrum of ATP7A-related disorders and demonstrate that two clinical phenotypes can occur in the same patient. The copper-induced toxicity and low levels of both ATP7A and ATP7B in our patient suggest that copper accumulation in motor neurons is the pathogenic mechanism in ATP7A-related dHMN.


Assuntos
ATPases Transportadoras de Cobre/genética , Cobre/toxicidade , Cútis Laxa/genética , Síndrome de Ehlers-Danlos/genética , Adulto , Criança , Cobre/sangue , Cútis Laxa/sangue , Cútis Laxa/diagnóstico , Cútis Laxa/fisiopatologia , Síndrome de Ehlers-Danlos/sangue , Síndrome de Ehlers-Danlos/diagnóstico , Síndrome de Ehlers-Danlos/fisiopatologia , Humanos , Masculino , Linhagem , Adulto Jovem
4.
Neurobiol Dis ; 145: 105043, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32798727

RESUMO

Rett syndrome (RTT; OMIM#312750) is mainly caused by mutations in the X-linked MECP2 gene (methyl-CpG-binding protein 2 gene; OMIM*300005), which leads to impairments in the brain-derived neurotrophic factor (BDNF) signalling. The boost of BDNF mediated effects would be a significant breakthrough but it has been hampered by the difficulty to administer BDNF to the central nervous system. Adenosine, an endogenous neuromodulator, may accomplish that role since through A2AR it potentiates BDNF synaptic actions in healthy animals. We thus characterized several hallmarks of the adenosinergic and BDNF signalling in RTT and explored whether A2AR activation could boost BDNF actions. For this study, the RTT animal model, the Mecp2 knockout (Mecp2-/y) (B6.129P2 (C)-Mecp2tm1.1Bird/J) mouse was used. Whenever possible, parallel data was also obtained from post-mortem brain samples from one RTT patient. Ex vivo extracellular recordings of field excitatory post-synaptic potentials in CA1 hippocampal area were performed to evaluate synaptic transmission and long-term potentiation (LTP). RT-PCR was used to assess mRNA levels and Western Blot or radioligand binding assays were performed to evaluate protein levels. Changes in cortical and hippocampal adenosine content were assessed by liquid chromatography with diode array detection (LC/DAD). Hippocampal ex vivo experiments revealed that the facilitatory actions of BDNF upon LTP is absent in Mecp2-/y mice and that TrkB full-length (TrkB-FL) receptor levels are significantly decreased. Extracts of the hippocampus and cortex of Mecp2-/y mice revealed less adenosine amount as well as less A2AR protein levels when compared to WT littermates, which may partially explain the deficits in adenosinergic tonus in these animals. Remarkably, the lack of BDNF effect on hippocampal LTP in Mecp2-/y mice was overcome by selective activation of A2AR with CGS21680. Overall, in Mecp2-/y mice there is an impairment on adenosinergic system and BDNF signalling. These findings set the stage for adenosine-based pharmacological therapeutic strategies for RTT, highlighting A2AR as a therapeutic target in this devastating pathology.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Síndrome de Rett/metabolismo , Transdução de Sinais/fisiologia , Animais , Hipocampo/metabolismo , Proteína 2 de Ligação a Metil-CpG , Camundongos , Camundongos Knockout , Receptor trkB/metabolismo , Síndrome de Rett/genética
5.
Orphanet J Rare Dis ; 14(1): 66, 2019 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-30871635

RESUMO

BACKGROUND: The Registry of Adult and Paediatric Patients Treated with Cystadane® - Homocystinuria (RoCH) is a non-interventional, observational, multi-centre, post-authorization safety study that aimed to identify safety of betaine anhydrous (Cystadane®) in the treatment of patients with inborn errors of homocysteine metabolism (homocystinuria) in order to minimise the treatment associated risks and establish better knowledge on its clinical use. The registry included patients of all ages with homocystinuria who were treated with betaine anhydrous in conjunction with other therapies. Clinical data were collected retrospectively from 2007 to 2013, then prospectively up to February 2014. All adverse events (AEs) reported during the study were recorded. The clinical and biological status of patients was monitored at least once a year. RESULTS: A total of 125 patients with homocystinuria (adults [> 18 years]: 50; paediatric [≤18 years]: 75) were enrolled at 29 centres in France and Spain. Patients were treated with betaine anhydrous for a mean duration of 7.4 ± 4.3 years. The median total daily dose of betaine anhydrous at the first and last study visits was 6 g/day for cystathionine ß-synthase (CBS)-deficient vitamin B6 responders and 9 g/day for methylenetetrahydrofolate reductase-deficient patients, while the median daily dose increased in CBS-deficient B6 non-responders (from 6 to 9 g/day) and cobalamin metabolism-defective patients (from 3 to 6 g/day) between the first and last visits. Treatment caused a mean overall reduction of 29% in plasma homocysteine levels in the study population. A total of 277 AEs were reported during the study, of which two non-serious AEs (bad taste and headache) and one serious AE (interstitial lung disease) were considered to be drug related. Overall, betaine anhydrous was well tolerated with no major safety concerns. CONCLUSIONS: Data from the RoCH registry provided real-world evidence on the clinical safety and efficacy of betaine anhydrous in the management of homocystinuria in paediatric and adult patients.


Assuntos
Betaína/administração & dosagem , Homocistinúria/tratamento farmacológico , Sistema de Registros , Adolescente , Adulto , Betaína/efeitos adversos , Criança , Pré-Escolar , Feminino , França , Homocisteína/sangue , Humanos , Lactente , Masculino , Estudos Retrospectivos , Espanha , Resultado do Tratamento , Adulto Jovem
6.
Neurogenetics ; 20(2): 73-82, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30903322

RESUMO

Celia's encephalopathy (progressive encephalopathy with/without lipodystrophy (PELD)) is a childhood neurodegenerative disorder with a fatal prognosis before the age of 10, due to the variant c.985C>T in the BSCL2 gene that causes a cryptic splicing site leading to skipping of exon 7. For years, different authors have reported cases of congenital generalized lipodystrophy due to the variant c.974dupG in BSCL2 associated with neurological manifestations of variable severity, although some of them clearly superimposable to PELD. To identify the molecular mechanisms responsible for these neurological alterations in two patients with c.974dupG. Clinical characterization, biochemistry, and neuroimaging studies of two girls carrying this variant. In silico analysis, PCR amplification, and BSCL2 cDNA sequencing. BSCL2-201 transcript expression, which lacks exon 7, by qPCR in fibroblasts from the index case, from a healthy child as a control and from two patients with PELD, and in leukocytes from the index case and her parents. One with a severe encephalopathy including a picture of intellectual deficiency, severe language impairment, myoclonic epilepsy, and lipodystrophy as described in PELD, dying at 9 years and 9 months of age. The other 2-year-old patient showed incipient signs of neurological involvement. In silico and cDNA sequencing studies showed that variant c.974dupG gives rise to skipping of exon 7. The expression of BSCL2-201 in fibroblasts was significantly higher in the index case than in the healthy child, although less than in the case with homozygous PELD due to c.985C>T variant. The expression of this transcript was approximately half in the healthy carrier parents of this patient. The c.974dupG variant leads to the skipping of exon 7 of the BSCL2 gene and is responsible for a variant of Celia's encephalopathy, with variable phenotypic expression.


Assuntos
Encefalopatias/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Lipodistrofia Generalizada Congênita/genética , Doenças Neurodegenerativas/genética , Processamento Alternativo , Criança , Pré-Escolar , DNA Complementar/genética , Éxons , Evolução Fatal , Feminino , Fibroblastos/metabolismo , Variação Genética , Homozigoto , Humanos , Fenótipo , Análise de Sequência de DNA
7.
Front Genet ; 10: 1300, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31969900

RESUMO

Encephalomyopathic mitochondrial DNA (mtDNA) depletion syndrome 13 (MTDPS13) is a rare genetic disorder caused by defects in F-box leucine-rich repeat protein 4 (FBXL4). Although FBXL4 is essential for the bioenergetic homeostasis of the cell, the precise role of the protein remains unknown. In this study, we report two cases of unrelated patients presenting in the neonatal period with hyperlactacidemia and generalized hypotonia. Severe mtDNA depletion was detected in muscle biopsy in both patients. Genetic analysis showed one patient as having in compound heterozygosis a splice site variant c.858+5G>C and a missense variant c.1510T>C (p.Cys504Arg) in FBXL4. The second patient harbored a frameshift novel variant c.851delC (p.Pro284LeufsTer7) in homozygosis. To validate the pathogenicity of these variants, molecular and biochemical analyses were performed using skin-derived fibroblasts. We observed that the mtDNA depletion was less severe in fibroblasts than in muscle. Interestingly, the cells harboring a nonsense variant in homozygosis showed normal mtDNA copy number. Both patient fibroblasts, however, demonstrated reduced mitochondrial transcript quantity leading to diminished steady state levels of respiratory complex subunits, decreased respiratory complex IV (CIV) activity, and finally, low mitochondrial ATP levels. Both patients also revealed citrate synthase deficiency. Genetic complementation assays established that the deficient phenotype was rescued by the canonical version of FBXL4, confirming the pathological nature of the variants. Further analysis of fibroblasts allowed to establish that increased mitochondrial mass, mitochondrial fragmentation, and augmented autophagy are associated with FBXL4 deficiency in cells, but are probably secondary to a primary metabolic defect affecting oxidative phosphorylation.

8.
Mitochondrion ; 30: 51-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27374853

RESUMO

We evaluated the coenzyme Q10 (CoQ) levels in patients who were diagnosed with mitochondrial oxidative phosphorylation (OXPHOS) and non-OXPHOS disorders (n=72). Data from the 72 cases in this study revealed that 44.4% of patients showed low CoQ concentrations in either their skeletal muscle or skin fibroblasts. Our findings suggest that secondary CoQ deficiency is a common finding in OXPHOS and non-OXPHOS disorders. We hypothesize that cases of CoQ deficiency associated with OXPHOS defects could be an adaptive mechanism to maintain a balanced OXPHOS, although the mechanisms explaining these deficiencies and the pathophysiological role of secondary CoQ deficiency deserves further investigation.


Assuntos
Doenças Mitocondriais/patologia , Fosforilação Oxidativa , Ubiquinona/análogos & derivados , Adolescente , Adulto , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Músculos/patologia , Prevalência , Pele/patologia , Ubiquinona/deficiência , Adulto Jovem
9.
Semin Pediatr Neurol ; 23(4): 290-305, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-28284391

RESUMO

Mitochondrial diseases exhibit significant clinical and genetic heterogeneity. Mitochondria are highly dynamic organelles that are the major contributor of adenosine triphosphate, through oxidative phosphorylation. These disorders may be developed at any age, with isolated or multiple system involvement, and in any pattern of inheritance. Defects in the mitochondrial respiratory chain impair energy production and almost invariably involve skeletal muscle and peripheral nerves, causing exercise intolerance, cramps, recurrent myoglobinuria, or fixed weakness, which often affects extraocular muscles and results in droopy eyelids (ptosis), progressive external ophthalmoplegia, peripheral ataxia, and peripheral polyneuropathy. This review describes the main neuromuscular symptomatology through different syndromes reported in the literature and from our experience. We want to highlight the importance of searching for the "clue clinical signs" associated with inheritance pattern as key elements to guide the complex diagnosis process and genetic studies in mitochondrial diseases.


Assuntos
Doenças Mitocondriais/complicações , Doenças Neuromusculares/fisiopatologia , Criança , Humanos , Doenças Neuromusculares/etiologia
10.
J Inherit Metab Dis ; 38(1): 19-40, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25413954

RESUMO

Over one hundred diseases related to inherited defects of complex lipids synthesis and remodeling are now reported. Most of them were described within the last 5 years. New descriptions and phenotypes are expanding rapidly. While the associated clinical phenotype is currently difficult to outline, with only a few patients identified, it appears that all organs and systems may be affected. The main clinical presentations can be divided into (1) Diseases affecting the central and peripheral nervous system. Complex lipid synthesis disorders produce prominent motor manifestations due to upper and/or lower motoneuron degeneration. Motor signs are often complex, associated with other neurological and extra-neurological signs. Three neurological phenotypes, spastic paraparesis, neurodegeneration with brain iron accumulation and peripheral neuropathies, deserve special attention. Many apparently well clinically defined syndromes are not distinct entities, but rather clusters on a continuous spectrum, like for the PNPLA6-associated diseases, extending from Boucher-Neuhauser syndrome via Gordon Holmes syndrome to spastic ataxia and pure hereditary spastic paraplegia; (2) Muscular/cardiac presentations; (3) Skin symptoms mostly represented by syndromic (neurocutaneous) and non syndromic ichthyosis; (4) Retinal dystrophies with syndromic and non syndromic retinitis pigmentosa, Leber congenital amaurosis, cone rod dystrophy, Stargardt disease; (5) Congenital bone dysplasia and segmental overgrowth disorders with congenital lipomatosis; (6) Liver presentations characterized mainly by transient neonatal cholestatic jaundice and non alcoholic liver steatosis with hypertriglyceridemia; and (7) Renal and immune presentations. Lipidomics and molecular functional studies could help to elucidate the mechanism(s) of dominant versus recessive inheritance observed for the same gene in a growing number of these disorders.


Assuntos
Erros Inatos do Metabolismo Lipídico/diagnóstico , Lipídeos/química , Ataxia/diagnóstico , Doenças do Desenvolvimento Ósseo/diagnóstico , Fígado Gorduroso/diagnóstico , Humanos , Hipertrigliceridemia/diagnóstico , Ictiose/diagnóstico , Icterícia/diagnóstico , Amaurose Congênita de Leber/diagnóstico , Erros Inatos do Metabolismo Lipídico/genética , Lipomatose/diagnóstico , Hepatopatias/diagnóstico , Degeneração Macular/diagnóstico , Doenças Neurodegenerativas/diagnóstico , Paraparesia Espástica/diagnóstico , Doenças do Sistema Nervoso Periférico/diagnóstico , Fenótipo , Distrofias Retinianas/diagnóstico , Retinose Pigmentar/diagnóstico , Paraplegia Espástica Hereditária/diagnóstico , Doença de Stargardt
11.
Rev Neurol ; 59(4): 158-63, 2014 Aug 16.
Artigo em Espanhol | MEDLINE | ID: mdl-25059266

RESUMO

INTRODUCTION: Diploid/triploid mosaicism is a rare chromosomal abnormality. It is caused by a failure in the postzygotic division during embryonic development. It results in the coexistence of two genetically heterogeneous cell lines (46,XX and 69,XXX) in one individual. His clinical phenotype is characteristic. Pigmentary changes with a distribution pattern following Blaschko's lines abnormalities in other ectoderm-derived tissues are the main diagnostic signs. CASE REPORTS: Three cases of diploid/triploid mosaicism are described, and compared to the previously reported cases. The most frequently observed symptoms were mental retardation, truncal obesity, short stature, hemihypertrophy, small and narrow hands with clino and camptodactyly. Phenotypic characteristics of our three patients were similar to those of previously reported cases. Although there is no single and specific phenotype associated with mosaicism diploid/triploid, there are some dysmorphic features that shape a recognizable malformative syndrome. Peripheral blood lymphocytes karyotype was normal in our patients. Diagnosis was reached performing a fibroblast karyotype from hypopigmented skin. CONCLUSIONS: Intellectual disability associated with truncal obesity, short stature, hemihypertrophy or clino/camptodactyly should suggest to clinicians the possible existence of a diploid/triploid mosaicism. In most cases, karyotype from fibroblasts is needed to reach the diagnosis.


TITLE: Mosaicismo diploide/triploide: un fenotipo variable, pero caracteristico.Introduccion. El mosaicismo diploide/triploide es una alteracion cromosomica poco frecuente. La produce un fallo en la division poscigotica durante el desarrollo embrionario. Da lugar a la coexistencia de dos lineas celulares con diferente constitucion cromosomica (46,XX y 69,XXX) en un mismo individuo. Su fenotipo clinico es caracteristico. Las alteraciones pigmentarias con un patron de distribucion que sigue las lineas de Blaschko son el principal signo guia, asi como las alteraciones de otros tejidos derivados del ectodermo. Casos clinicos. Describimos las caracteristicas clinicas de tres pacientes afectos de mosaicismo diploide/triploide y realizamos una comparacion de su fenotipo clinico con el de los casos publicados previamente en la bibliografia. Las alteraciones observadas con mayor frecuencia fueron alteraciones cutaneas, discapacidad intelectual, obesidad troncular, talla baja, hemihipertrofia, y manos pequeñas y estrechas con clino y camptodactilia. Las caracteristicas fenotipicas de nuestros pacientes fueron similares a las de los casos comunicados previamente. Aunque no existe un fenotipo unico y especifico asociado al mosaicismo diploide/triploide, existen malformaciones caracteristicas que conforman un sindrome malformativo bien definido. El cariotipo realizado en linfocitos de sangre periferica en las tres pacientes fue normal, y se logro el diagnostico mediante cariotipo en fibroblastos cultivados tras biopsia de piel hipopigmentada. Conclusiones. La presencia de discapacidad intelectual asociada a obesidad troncular, talla baja, hemihipertrofia o clino y camptodactilia, ademas de las alteraciones cutaneas, debe hacer pensar en la posible existencia de un mosaicismo diploide/triploide. En la mayoria de los casos, es necesario el estudio del cariotipo en los fibroblastos para llegar al diagnostico.


Assuntos
Anormalidades Múltiplas/genética , Cariótipo Anormal , Aborto Habitual/genética , Epilepsia Tipo Ausência/genética , Face/anormalidades , Feminino , Retardo do Crescimento Fetal/etiologia , Fibroblastos/ultraestrutura , Cardiopatias Congênitas/genética , Humanos , Hipopigmentação/genética , Recém-Nascido , Recém-Nascido Pequeno para a Idade Gestacional , Deficiência Intelectual/genética , Linfócitos/ultraestrutura , Mosaicismo , Obesidade Abdominal/genética , Fenótipo , Estudos Retrospectivos , Sindactilia/genética , Triploidia
12.
Nat Genet ; 44(11): 1243-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23001123

RESUMO

Adenosine deaminases acting on RNA (ADARs) catalyze the hydrolytic deamination of adenosine to inosine in double-stranded RNA (dsRNA) and thereby potentially alter the information content and structure of cellular RNAs. Notably, although the overwhelming majority of such editing events occur in transcripts derived from Alu repeat elements, the biological function of non-coding RNA editing remains uncertain. Here, we show that mutations in ADAR1 (also known as ADAR) cause the autoimmune disorder Aicardi-Goutières syndrome (AGS). As in Adar1-null mice, the human disease state is associated with upregulation of interferon-stimulated genes, indicating a possible role for ADAR1 as a suppressor of type I interferon signaling. Considering recent insights derived from the study of other AGS-related proteins, we speculate that ADAR1 may limit the cytoplasmic accumulation of the dsRNA generated from genomic repetitive elements.


Assuntos
Adenosina Desaminase/genética , Doenças Autoimunes do Sistema Nervoso/genética , Interferon Tipo I , Malformações do Sistema Nervoso/genética , RNA de Cadeia Dupla/metabolismo , Elementos Alu/genética , Animais , Exoma , Expressão Gênica , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Mutação , Conformação Proteica , RNA de Cadeia Dupla/genética , Proteínas de Ligação a RNA , Análise de Sequência de DNA , Transdução de Sinais , Relação Estrutura-Atividade
13.
Arch Neurol ; 68(5): 615-21, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21555636

RESUMO

BACKGROUND: Cerebral folate deficiency may be amenable to therapeutic supplementation. Diverse metabolic pathways and unrelated processes can lead to cerebrospinal fluid 5-methyltetrahydrofolate (5-MTHF) depletion, the hallmark of cerebral folate deficiency. OBJECTIVE: To analyze cerebral folate abundance in a large prospective series of children diagnosed with any neurologic disorder for which a diagnostic lumbar puncture was indicated. DESIGN: We studied the spectrum and frequency of disorders associated with cerebral folate deficiency by measuring cerebrospinal fluid 5-MTHF, biogenic amines, and pterins. Direct sequencing of the FOLR1 transporter gene was also performed in some patients. SETTING: Academic pediatric medical center. PARTICIPANTS: We studied 134 individuals free of neurometabolic disease and 584 patients with any of several diseases of the central nervous system. RESULTS: Of 584 patients, 71 (12%) exhibited 5-MTHF deficiency. Mild to moderate deficiency (n = 63; range, 19-63 nmol/L) was associated with perinatal asphyxia, central nervous system infection, or diseases of probable genetic origin (inborn errors of metabolism, white matter disorders, Rett syndrome, or epileptic encephalopathies). Severe 5-MTHF depletion (n = 8; range, 0.6-13 nmol/L) was detected in severe MTHF reductase deficiency, Kearns-Sayre syndrome, biotin-responsive striatal necrosis, acute necrotizing encephalitis of Hurst, and FOLR1 defect. A strong correlation was observed between cerebrospinal fluid and plasma folate levels in cerebral folate deficiency. CONCLUSIONS: Of the 2 main forms of cerebral folate deficiency identified, mild to moderate 5-MTHF deficiency was most commonly associated with disorders bearing no primary relation to folate metabolism, whereas profound 5-MTHF depletion was associated with specific mitochondrial disorders, metabolic and transporter defects, or cerebral degenerations. The results suggest that 5-MTHF can serve either as the hallmark of inborn disorders of folate transport and metabolism or, more frequently, as an indicator of neurologic dysfunction.


Assuntos
Encéfalo/metabolismo , Deficiência de Ácido Fólico/líquido cefalorraquidiano , Deficiência de Ácido Fólico/diagnóstico , Ácido Fólico/uso terapêutico , Tetra-Hidrofolatos/líquido cefalorraquidiano , Tetra-Hidrofolatos/deficiência , Adolescente , Aminas Biogênicas/líquido cefalorraquidiano , Criança , Pré-Escolar , Feminino , Receptor 1 de Folato/genética , Ácido Fólico/administração & dosagem , Deficiência de Ácido Fólico/tratamento farmacológico , Deficiência de Ácido Fólico/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Estudos Prospectivos , Pterinas/líquido cefalorraquidiano , Análise de Sequência de DNA , Punção Espinal
14.
J Inherit Metab Dis ; 34(3): 677-94, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21431622

RESUMO

Glutaric aciduria type I (synonym, glutaric acidemia type I) is a rare organic aciduria. Untreated patients characteristically develop dystonia during infancy resulting in a high morbidity and mortality. The neuropathological correlate is striatal injury which results from encephalopathic crises precipitated by infectious diseases, immunizations and surgery during a finite period of brain development, or develops insidiously without clinically apparent crises. Glutaric aciduria type I is caused by inherited deficiency of glutaryl-CoA dehydrogenase which is involved in the catabolic pathways of L-lysine, L-hydroxylysine and L-tryptophan. This defect gives rise to elevated glutaric acid, 3-hydroxyglutaric acid, glutaconic acid, and glutarylcarnitine which can be detected by gas chromatography/mass spectrometry (organic acids) or tandem mass spectrometry (acylcarnitines). Glutaric aciduria type I is included in the panel of diseases that are identified by expanded newborn screening in some countries. It has been shown that in the majority of neonatally diagnosed patients striatal injury can be prevented by combined metabolic treatment. Metabolic treatment that includes a low lysine diet, carnitine supplementation and intensified emergency treatment during acute episodes of intercurrent illness should be introduced and monitored by an experienced interdisciplinary team. However, initiation of treatment after the onset of symptoms is generally not effective in preventing permanent damage. Secondary dystonia is often difficult to treat, and the efficacy of available drugs cannot be predicted precisely in individual patients. The major aim of this revision is to re-evaluate the previous diagnostic and therapeutic recommendations for patients with this disease and incorporate new research findings into the guideline.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/terapia , Encefalopatias Metabólicas/diagnóstico , Encefalopatias Metabólicas/terapia , Guias de Prática Clínica como Assunto , Algoritmos , Erros Inatos do Metabolismo dos Aminoácidos/complicações , Encefalopatias Metabólicas/complicações , Serviços Médicos de Emergência/métodos , Glutaril-CoA Desidrogenase/deficiência , Humanos , Recém-Nascido , Programas de Rastreamento/métodos , Monitorização Fisiológica/métodos , Triagem Neonatal/métodos , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/terapia
15.
J Inherit Metab Dis ; 33(6): 795-802, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20857335

RESUMO

Several unrelated disorders can lead to 5-methyltetrahydrofolate (5MTHF) depletion in the cerobrospinal fluid (CSF), including primary genetic disorders in folate-related pathways or those causing defective transport across the blood-CSF barrier. We report a case of cerebral folate transport deficiency due to a novel homozygous mutation in the FOLR1 gene, in an effort to clarify phenotype-genotype correlation in this newly identified neurometabolic disorder. A previously healthy infant developed an ataxic syndrome in the second year of life, followed by choreic movements and progressive myoclonic epilepsy. At the age of 26 months, we analyzed CSF 5MTHF by HPLC with fluorescence detection and conducted magnetic resonance (MR) imaging and spectroscopy studies. Finally, we performed mutational screening in the coding region of the FOLR1 gene. MR showed a diffuse abnormal signal of the cerebral white matter, cerebellar atrophy and a reduced peak of choline in spectroscopy. A profound deficiency of CSF 5MTHF (2 nmol/L; NV 48-127) with reduced CSF/plasma folate ratio (0.4; NV 1.5-3.5) was highly suggestive of defective brain folate-specific transport across the blood-CSF/brain barrier. Mutation screening of FOLR1 revealed a new homozygous missense mutation (p.Cys105Arg) that is predicted to abolish a disulfide bond, probably necessary for the correct folding of the protein. Both parents were heterozygous carriers of the same variant. Mutation screening in the FOLR1 gene is advisable in children with profound 5MTHF deficiency and decreased CSF/serum folate ratio. Progressive ataxia and myoclonic epilepsy, together with impaired brain myelination, are clinical hallmarks of the disease.


Assuntos
Ataxia/genética , Epilepsias Mioclônicas/genética , Receptor 1 de Folato/genética , Mutação de Sentido Incorreto , Ataxia/sangue , Ataxia/líquido cefalorraquidiano , Ataxia/complicações , Criança , Consanguinidade , Progressão da Doença , Eletromiografia , Epilepsias Mioclônicas/sangue , Epilepsias Mioclônicas/líquido cefalorraquidiano , Epilepsias Mioclônicas/complicações , Ácido Fólico/sangue , Ácido Fólico/líquido cefalorraquidiano , Homozigoto , Humanos , Masculino , Mutação de Sentido Incorreto/fisiologia , Linhagem
16.
Pediatr Neurol ; 41(6): 448-50, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19931168

RESUMO

Cryopyrin-associated periodic syndrome is a category of autoinflammatory disorders caused by mutations of the NLRP3 gene, with chronic infantile neurologic cutaneous and articular syndrome being the severest clinical phenotype. Various pterins have been reported as mediating immunologic functions in the central nervous system, but to date studies of pterin cerebrospinal fluid (CSF) values and cryopyrin-associated periodic syndrome have been lacking. A 2-year-old child was affected with a severe atypical form of cryopyrin-associated periodic syndrome, suspected based on the analysis of neopterin in CSF. He initially presented isolated neurologic manifestations mimicking a neuroregressive disorder. Blood and CSF analyses did not present any routine inflammatory markers, but CSF neopterin was elevated. Later, the patient developed arthritis and recurrent episodes of fever, and the cryopyrin-associated periodic syndrome diagnosis was confirmed by genetic studies. Neopterin was the most altered indicator over the time. Child neurologists should be on the alert when unexplained neurologic signs appear, giving consideration to the possibility of inflammatory or immune-mediated diseases. The present case demonstrates the clinical utility of measurement of CSF neopterin levels in screening for these immune-mediated diseases, especially when neurologic symptoms are associated with normal results on routine CSF tests.


Assuntos
Síndromes Periódicas Associadas à Criopirina/líquido cefalorraquidiano , Neopterina/líquido cefalorraquidiano , Proteínas de Transporte/genética , Síndromes Periódicas Associadas à Criopirina/sangue , Síndromes Periódicas Associadas à Criopirina/genética , Diagnóstico Diferencial , Progressão da Doença , Seguimentos , Humanos , Lactente , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR
17.
Clin Biochem ; 42(7-8): 742-5, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19094978

RESUMO

OBJECTIVES: To report on a case with a mitochondrial DNA (mtDNA) depletion syndrome. DESIGN AND METHODS: Laboratory studies were done in muscle biopsy and fibroblasts to evaluate coenzyme Q(10) (CoQ(10)) status and quantify mitochondrial DNA. RESULTS: Decreased CoQ(10) values and a 78% of mtDNA depletion were detected in muscle. Mutational studies failed to reveal any pathogenic mutation in nuclear genes related with mtDNA maintenance. CONCLUSIONS: mtDNA depletion syndrome was associated with CoQ(10) deficiency in our patient.


Assuntos
DNA Mitocondrial/genética , Doenças Mitocondriais/etiologia , Doenças Mitocondriais/genética , Ubiquinona/análogos & derivados , Feminino , Humanos , Recém-Nascido , Mitocôndrias Musculares/genética , Doenças Mitocondriais/metabolismo , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Ubiquinona/deficiência
18.
Am J Hum Genet ; 81(4): 713-25, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17846997

RESUMO

Aicardi-Goutieres syndrome (AGS) is a genetic encephalopathy whose clinical features mimic those of acquired in utero viral infection. AGS exhibits locus heterogeneity, with mutations identified in genes encoding the 3'-->5' exonuclease TREX1 and the three subunits of the RNASEH2 endonuclease complex. To define the molecular spectrum of AGS, we performed mutation screening in patients, from 127 pedigrees, with a clinical diagnosis of the disease. Biallelic mutations in TREX1, RNASEH2A, RNASEH2B, and RNASEH2C were observed in 31, 3, 47, and 18 families, respectively. In five families, we identified an RNASEH2A or RNASEH2B mutation on one allele only. In one child, the disease occurred because of a de novo heterozygous TREX1 mutation. In 22 families, no mutations were found. Null mutations were common in TREX1, although a specific missense mutation was observed frequently in patients from northern Europe. Almost all mutations in RNASEH2A, RNASEH2B, and RNASEH2C were missense. We identified an RNASEH2C founder mutation in 13 Pakistani families. We also collected clinical data from 123 mutation-positive patients. Two clinical presentations could be delineated: an early-onset neonatal form, highly reminiscent of congenital infection seen particularly with TREX1 mutations, and a later-onset presentation, sometimes occurring after several months of normal development and occasionally associated with remarkably preserved neurological function, most frequently due to RNASEH2B mutations. Mortality was correlated with genotype; 34.3% of patients with TREX1, RNASEH2A, and RNASEH2C mutations versus 8.0% RNASEH2B mutation-positive patients were known to have died (P=.001). Our analysis defines the phenotypic spectrum of AGS and suggests a coherent mutation-screening strategy in this heterogeneous disorder. Additionally, our data indicate that at least one further AGS-causing gene remains to be identified.


Assuntos
Doenças dos Gânglios da Base/genética , Adolescente , Adulto , Doenças dos Gânglios da Base/líquido cefalorraquidiano , Doenças dos Gânglios da Base/patologia , Encéfalo/patologia , Calcinose/genética , Calcinose/patologia , Pérnio/genética , Pérnio/patologia , Criança , Pré-Escolar , Análise Mutacional de DNA , Exodesoxirribonucleases/genética , Feminino , Humanos , Lactente , Recém-Nascido , Linfocitose/líquido cefalorraquidiano , Linfocitose/genética , Masculino , Dados de Sequência Molecular , Mutação , Fenótipo , Fosfoproteínas/genética , Ribonuclease H/genética , Síndrome
19.
Pediatr Radiol ; 37(10): 1043-6, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17704913

RESUMO

Molybdenum cofactor is essential for the function of three human enzymes: sulphite oxidase, xanthine dehydrogenase, and aldehyde oxidase. Molybdenum cofactor deficiency is a rare autosomal recessively inherited disease. Disturbed development and damage to the brain may occur as a result of accumulation of toxic levels of sulphite. The CT and MRI findings include severe early brain abnormalities and have been widely reported, but the cranial US imaging findings have seldom been reported. We report a chronological series of cranial US images obtained from an affected infant that show the rapid development of cerebral atrophy, calcifications and white matter cysts. Our report supports the utility of cranial US, a noninvasive bed-side technique, in the detection and follow-up of these rapidly changing lesions.


Assuntos
Encefalopatias/diagnóstico , Calcinose/diagnóstico , Coenzimas/deficiência , Ecoencefalografia/métodos , Erros Inatos do Metabolismo/diagnóstico , Metaloproteínas/deficiência , Atrofia/diagnóstico , Humanos , Recém-Nascido , Masculino , Cofatores de Molibdênio , Pteridinas , Síndrome
20.
Mol Genet Metab ; 92(3): 274-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17698383

RESUMO

We report a recessive mutation in the tyrosine hydroxylase gene (TH) promoter (c.1-71C>T), present at homozygosity in a patient with dopa-responsive encephalopathy. The change lies in a cAMP response element (CRE) and alters a binding site for the CREM transcription factor. Previous studies support that the CRE in the TH gene is essential for its transcription, suggesting that mutations within this consensus motif may cause an impairment of catecholamine biosynthesis and lead to a pathogenic phenotype.


Assuntos
Dano Encefálico Crônico/tratamento farmacológico , Di-Hidroxifenilalanina/uso terapêutico , Dopaminérgicos/uso terapêutico , Regulação Enzimológica da Expressão Gênica , Mutação Puntual/genética , Regiões Promotoras Genéticas/genética , Elementos de Resposta/genética , Tirosina 3-Mono-Oxigenase/genética , Criança , Pré-Escolar , AMP Cíclico/farmacologia , Feminino , Homozigoto , Humanos , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA