Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Mol Genet Metab ; 135(4): 350-356, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35279367

RESUMO

BACKGROUND: Seventy-five percent of patients with pyridoxine-dependent epilepsy due to α-aminoadipic semialdehyde dehydrogenase deficiency (PDE-ALDH7A1) suffer intellectual developmental disability despite pyridoxine treatment. Adjunct lysine reduction therapies (LRT), aimed at lowering putative neurotoxic metabolites, are associated with improved cognitive outcomes. However, possibly due to timing of treatment, not all patients have normal intellectual function. METHODS: This retrospective, multi-center cohort study evaluated the effect of timing of pyridoxine monotherapy and pyridoxine with adjunct LRT on neurodevelopmental outcome. Patients with confirmed PDE-ALDH7A1 with at least one sibling with PDE-ALDH7A1 and a difference in age at treatment initiation were eligible and identified via the international PDE registry, resulting in thirty-seven patients of 18 families. Treatment regimen was pyridoxine monotherapy in ten families and pyridoxine with adjunct LRT in the other eight. Primary endpoints were standardized and clinically assessed neurodevelopmental outcomes. Clinical neurodevelopmental status was subjectively assessed over seven domains: overall neurodevelopment, speech/language, cognition, fine and gross motor skills, activities of daily living and behavioral/psychiatric abnormalities. RESULTS: The majority of early treated siblings on pyridoxine monotherapy performed better than their late treated siblings on the clinically assessed domain of fine motor skills. For siblings on pyridoxine and adjunct LRT, the majority of early treated siblings performed better on clinically assessed overall neurodevelopment, cognition, and behavior/psychiatry. Fourteen percent of the total cohort was assessed as normal on all domains. CONCLUSION: Early treatment with pyridoxine and adjunct LRT may be beneficial for neurodevelopmental outcome. When evaluating a more extensive neurodevelopmental assessment, the actual impairment rate may be higher than the 75% reported in literature. TAKE- HOME MESSAGE: Early initiation of lysine reduction therapies adjunct to pyridoxine treatment in patients with PDE-ALDH7A1 may result in an improved neurodevelopmental outcome.


Assuntos
Lisina , Piridoxina , Atividades Cotidianas , Estudos de Coortes , Epilepsia , Humanos , Piridoxina/uso terapêutico , Estudos Retrospectivos
2.
Am J Med Genet A ; 185(1): 157-167, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33112498

RESUMO

Mutations in the short-chain enoyl-CoA hydratase (SCEH) gene, ECHS1, cause a rare autosomal recessive disorder of valine catabolism. Patients usually present with developmental delay, regression, dystonia, feeding difficulties, and abnormal MRI with bilateral basal ganglia involvement. We present clinical, biochemical, molecular, and functional data for four affected patients from two unrelated families of Samoan descent with identical novel compound heterozygous mutations. Family 1 has three affected boys while Family 2 has an affected daughter, all with clinical and MRI findings of Leigh syndrome and intermittent episodes of acidosis and ketosis. WES identified a single heterozygous variant in ECHS1 at position c.832G > A (p.Ala278Thr). However, western blot revealed significantly reduced ECHS1 protein for all affected family members. Decreased SCEH activity in fibroblasts and a mild increase in marker metabolites in urine further supported ECHS1 as the underlying gene defect. Additional investigations at the DNA (aCGH, WGS) and RNA (qPCR, RT-PCR, RNA-Seq, RNA-Array) level identified a silent, common variant at position c.489G > A (p.Pro163=) as the second mutation. This substitution, present at high frequency in the Samoan population, is associated with decreased levels of normally spliced mRNA. To our understanding, this is the first report of a novel, hypomorphic allele c.489G > A (p.Pro163=), associated with SCEH deficiency.


Assuntos
Enoil-CoA Hidratase/genética , Predisposição Genética para Doença , Doenças Raras/genética , Adolescente , Criança , Pré-Escolar , Feminino , Heterozigoto , Humanos , Lactente , Masculino , Mutação/genética , Doenças Raras/diagnóstico , Doenças Raras/epidemiologia , Doenças Raras/patologia , Samoa/epidemiologia
3.
J Inherit Metab Dis ; 44(1): 178-192, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33200442

RESUMO

Pyridoxine-dependent epilepsy (PDE-ALDH7A1) is an autosomal recessive condition due to a deficiency of α-aminoadipic semialdehyde dehydrogenase, which is a key enzyme in lysine oxidation. PDE-ALDH7A1 is a developmental and epileptic encephalopathy that was historically and empirically treated with pharmacologic doses of pyridoxine. Despite adequate seizure control, most patients with PDE-ALDH7A1 were reported to have developmental delay and intellectual disability. To improve outcome, a lysine-restricted diet and competitive inhibition of lysine transport through the use of pharmacologic doses of arginine have been recommended as an adjunct therapy. These lysine-reduction therapies have resulted in improved biochemical parameters and cognitive development in many but not all patients. The goal of these consensus guidelines is to re-evaluate and update the two previously published recommendations for diagnosis, treatment, and follow-up of patients with PDE-ALDH7A1. Members of the International PDE Consortium initiated evidence and consensus-based process to review previous recommendations, new research findings, and relevant clinical aspects of PDE-ALDH7A1. The guideline development group included pediatric neurologists, biochemical geneticists, clinical geneticists, laboratory scientists, and metabolic dieticians representing 29 institutions from 16 countries. Consensus guidelines for the diagnosis and management of patients with PDE-ALDH7A1 are provided.


Assuntos
Arginina/administração & dosagem , Suplementos Nutricionais , Epilepsia/dietoterapia , Epilepsia/diagnóstico , Aldeído Desidrogenase/deficiência , Consenso , Epilepsia/tratamento farmacológico , Humanos , Cooperação Internacional , Lisina/deficiência , Piridoxina/uso terapêutico
4.
Genet Med ; 22(1): 199-209, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31462754

RESUMO

PURPOSE: Mitochondrial DNA (mtDNA) depletion syndrome (MDDS) encompasses a group of genetic disorders of mtDNA maintenance. Mutation of RRM2B is an uncommon cause of infantile-onset encephalomyopathic MDDS. Here we describe the natural history of this disease. METHODS: Multinational series of new genetically confirmed cases from six pediatric centers. RESULTS: Nine new cases of infantile-onset RRM2B deficiency, and 22 previously published cases comprised a total cohort of 31 patients. Infants presented at a mean of 1.95 months with truncal hypotonia, generalized weakness, and faltering growth. Seizures evolved in 39% at a mean of 3.1 months. Non-neurological manifestations included respiratory distress/failure (58%), renal tubulopathy (55%), sensorineural hearing loss (36%), gastrointestinal disturbance (32%), eye abnormalities (13%), and anemia (13%). Laboratory features included elevated lactate (blood, cerebrospinal fluid (CSF), urine, magnetic resonance (MR), spectroscopy), ragged-red and cytochrome c oxidase-deficient fibers, lipid myopathy, and multiple oxidative phosphorylation enzyme deficiencies in skeletal muscle. Eight new RRM2B variants were identified. Patients with biallelic truncating variants had the worst survival. Overall survival was 29% at 6 months and 16% at 1 year. CONCLUSIONS: Infantile-onset MDDS due to RRM2B deficiency is a severe disorder with characteristic clinical features and extremely poor prognosis. Presently management is supportive as there is no effective treatment. Novel treatments are urgently needed.


Assuntos
Proteínas de Ciclo Celular/genética , Pseudo-Obstrução Intestinal/genética , Distrofia Muscular Oculofaríngea/genética , Mutação de Sentido Incorreto , Ribonucleotídeo Redutases/genética , Proteínas de Ciclo Celular/química , Feminino , Humanos , Lactente , Recém-Nascido , Pseudo-Obstrução Intestinal/mortalidade , Masculino , Modelos Moleculares , Distrofia Muscular Oculofaríngea/mortalidade , Oftalmoplegia/congênito , Prognóstico , Conformação Proteica , Ribonucleotídeo Redutases/química , Análise de Sobrevida
5.
Mol Genet Metab ; 126(1): 53-63, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30473481

RESUMO

Primary mitochondrial complex I deficiency is the most common defect of the mitochondrial respiratory chain. It is caused by defects in structural components and assembly factors of this large protein complex. Mutations in the assembly factor NDUFAF5 are rare, with only five families reported to date. This study provides clinical, biochemical, molecular and functional data for four unrelated additional families, and three novel pathogenic variants. Three cases presented in infancy with lactic acidosis and classic Leigh syndrome. One patient, however, has a milder phenotype, with symptoms starting at 27 months and a protracted clinical course with improvement and relapsing episodes. She is homozygous for a previously reported mutation, p.Met279Arg and alive at 19 years with mild neurological involvement, normal lactate but abnormal urine organic acids. We found the same mutation in one of our severely affected patients in compound heterozygosity with a novel p.Lys52Thr mutation. Both patients with p.Met279Arg are of Taiwanese descent and had severe hyponatremia. Our third and fourth patients, both Caucasian, shared a common, newly described, missense mutation p.Lys109Asn which we show induces skipping of exon 3. Both Caucasian patients were compound heterozygotes, one with a previously reported Ashkenazi founder mutation while the other was negative for additional exonic variants. Whole genome sequencing followed by RNA studies revealed a novel deep intronic variant at position c.223-907A>C inducing an exonic splice enhancer. Our report adds significant new information to the mutational spectrum of NDUFAF5, further delineating the phenotypic heterogeneity of this mitochondrial defect.


Assuntos
Complexo I de Transporte de Elétrons/deficiência , Doença de Leigh/genética , Metiltransferases/genética , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Mutação , Fenótipo , Adolescente , Biópsia , Criança , Pré-Escolar , Complexo I de Transporte de Elétrons/genética , Feminino , Humanos , Lactente , Masculino , Linhagem , Pele/patologia , Sequenciamento do Exoma , Sequenciamento Completo do Genoma , Adulto Jovem
6.
J Inherit Metab Dis ; 41(2): 169-180, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29238895

RESUMO

Combined D-2- and L-2-hydroxyglutaric aciduria (D/L-2-HGA) is a devastating neurometabolic disorder, usually lethal in the first years of life. Autosomal recessive mutations in the SLC25A1 gene, which encodes the mitochondrial citrate carrier (CIC), were previously detected in patients affected with combined D/L-2-HGA. We showed that transfection of deficient fibroblasts with wild-type SLC25A1 restored citrate efflux and decreased intracellular 2-hydroxyglutarate levels, confirming that deficient CIC is the cause of D/L-2-HGA. We developed and implemented a functional assay and applied it to all 17 missense variants detected in a total of 26 CIC-deficient patients, including eight novel cases, showing reduced activities of varying degrees. In addition, we analyzed the importance of residues affected by these missense variants using our existing scoring system. This allowed not only a clinical and biochemical overview of the D/L-2-HGA patients but also phenotype-genotype correlation studies.


Assuntos
Proteínas de Transporte de Ânions/metabolismo , Encefalopatias Metabólicas Congênitas/metabolismo , Ácido Cítrico/metabolismo , Glutaratos/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas de Transporte de Ânions/química , Proteínas de Transporte de Ânions/genética , Bioensaio/métodos , Encefalopatias Metabólicas Congênitas/genética , Células Cultivadas , Pré-Escolar , Análise Mutacional de DNA , Feminino , Fibroblastos , Predisposição Genética para Doença , Humanos , Lactente , Recém-Nascido , Masculino , Proteínas Mitocondriais/química , Proteínas Mitocondriais/genética , Modelos Moleculares , Mutação de Sentido Incorreto , Transportadores de Ânions Orgânicos , Fenótipo , Conformação Proteica , Relação Estrutura-Atividade
7.
PLoS Genet ; 11(3): e1005097, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25807530

RESUMO

Here we demonstrate association of variants in the mitochondrial asparaginyl-tRNA synthetase NARS2 with human hearing loss and Leigh syndrome. A homozygous missense mutation ([c.637G>T; p.Val213Phe]) is the underlying cause of nonsyndromic hearing loss (DFNB94) and compound heterozygous mutations ([c.969T>A; p.Tyr323*] + [c.1142A>G; p.Asn381Ser]) result in mitochondrial respiratory chain deficiency and Leigh syndrome, which is a neurodegenerative disease characterized by symmetric, bilateral lesions in the basal ganglia, thalamus, and brain stem. The severity of the genetic lesions and their effects on NARS2 protein structure cosegregate with the phenotype. A hypothetical truncated NARS2 protein, secondary to the Leigh syndrome mutation p.Tyr323* is not detectable and p.Asn381Ser further decreases NARS2 protein levels in patient fibroblasts. p.Asn381Ser also disrupts dimerization of NARS2, while the hearing loss p.Val213Phe variant has no effect on NARS2 oligomerization. Additionally we demonstrate decreased steady-state levels of mt-tRNAAsn in fibroblasts from the Leigh syndrome patients. In these cells we show that a decrease in oxygen consumption rates (OCR) and electron transport chain (ETC) activity can be rescued by overexpression of wild type NARS2. However, overexpression of the hearing loss associated p.Val213Phe mutant protein in these fibroblasts cannot complement the OCR and ETC defects. Our findings establish lesions in NARS2 as a new cause for nonsyndromic hearing loss and Leigh syndrome.


Assuntos
Aspartato-tRNA Ligase/genética , Doença de Leigh/genética , Aminoacil-RNA de Transferência/genética , Adulto , Sequência de Aminoácidos/genética , Animais , Aspartato-tRNA Ligase/biossíntese , Surdez/genética , Surdez/patologia , Orelha Interna/metabolismo , Orelha Interna/patologia , Feminino , Fibroblastos , Expressão Gênica/genética , Predisposição Genética para Doença , Humanos , Doença de Leigh/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Mitocôndrias/genética , Mitocôndrias/patologia , Mutação de Sentido Incorreto/genética , Consumo de Oxigênio/genética , Linhagem
8.
Hum Mutat ; 37(7): 653-60, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26931382

RESUMO

Congenital disorders of glycosylation (CDG) arise from pathogenic mutations in over 100 genes leading to impaired protein or lipid glycosylation. ALG1 encodes a ß1,4 mannosyltransferase that catalyzes the addition of the first of nine mannose moieties to form a dolichol-lipid linked oligosaccharide intermediate required for proper N-linked glycosylation. ALG1 mutations cause a rare autosomal recessive disorder termed ALG1-CDG. To date 13 mutations in 18 patients from 14 families have been described with varying degrees of clinical severity. We identified and characterized 39 previously unreported cases of ALG1-CDG from 32 families and add 26 new mutations. Pathogenicity of each mutation was confirmed based on its inability to rescue impaired growth or hypoglycosylation of a standard biomarker in an alg1-deficient yeast strain. Using this approach we could not establish a rank order comparison of biomarker glycosylation and patient phenotype, but we identified mutations with a lethal outcome in the first two years of life. The recently identified protein-linked xeno-tetrasaccharide biomarker, NeuAc-Gal-GlcNAc2 , was seen in all 27 patients tested. Our study triples the number of known patients and expands the molecular and clinical correlates of this disorder.


Assuntos
Defeitos Congênitos da Glicosilação/genética , Manosiltransferases/genética , Mutação , Polissacarídeos/metabolismo , Biomarcadores/metabolismo , Defeitos Congênitos da Glicosilação/metabolismo , Feminino , Genes Letais , Glicosilação , Humanos , Masculino , Análise de Sequência de DNA , Análise de Sobrevida
9.
Am J Hum Genet ; 93(1): 29-41, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23768512

RESUMO

Congenital muscular dystrophies with hypoglycosylation of α-dystroglycan (α-DG) are a heterogeneous group of disorders often associated with brain and eye defects in addition to muscular dystrophy. Causative variants in 14 genes thought to be involved in the glycosylation of α-DG have been identified thus far. Allelic mutations in these genes might also cause milder limb-girdle muscular dystrophy phenotypes. Using a combination of exome and Sanger sequencing in eight unrelated individuals, we present evidence that mutations in guanosine diphosphate mannose (GDP-mannose) pyrophosphorylase B (GMPPB) can result in muscular dystrophy variants with hypoglycosylated α-DG. GMPPB catalyzes the formation of GDP-mannose from GTP and mannose-1-phosphate. GDP-mannose is required for O-mannosylation of proteins, including α-DG, and it is the substrate of cytosolic mannosyltransferases. We found reduced α-DG glycosylation in the muscle biopsies of affected individuals and in available fibroblasts. Overexpression of wild-type GMPPB in fibroblasts from an affected individual partially restored glycosylation of α-DG. Whereas wild-type GMPPB localized to the cytoplasm, five of the identified missense mutations caused formation of aggregates in the cytoplasm or near membrane protrusions. Additionally, knockdown of the GMPPB ortholog in zebrafish caused structural muscle defects with decreased motility, eye abnormalities, and reduced glycosylation of α-DG. Together, these data indicate that GMPPB mutations are responsible for congenital and limb-girdle muscular dystrophies with hypoglycosylation of α-DG.


Assuntos
Distroglicanas/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação de Sentido Incorreto , Nucleotidiltransferases/metabolismo , Animais , Pré-Escolar , Análise Mutacional de DNA/métodos , Distroglicanas/genética , Anormalidades do Olho/patologia , Feminino , Fibroblastos/enzimologia , Fibroblastos/patologia , Estudos de Associação Genética/métodos , Glicosilação , Guanosina Difosfato Manose/metabolismo , Heterozigoto , Humanos , Lactente , Recém-Nascido , Masculino , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/enzimologia , Nucleotidiltransferases/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
10.
Mol Genet Metab ; 119(1-2): 91-9, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27448789

RESUMO

In humans, mitochondrial DNA (mtDNA) depletion syndromes are a group of genetically and clinically heterogeneous autosomal recessive disorders that arise as a consequence of defects in mtDNA replication or nucleotide synthesis. Clinical manifestations are variable and include myopathic, encephalomyopathic, neurogastrointestinal or hepatocerebral phenotypes. Through clinical exome sequencing, we identified a homozygous missense variant (c.533C>T; p.Pro178Leu) in mitochondrial transcription factor A (TFAM) segregating in a consanguineous kindred of Colombian-Basque descent in which two siblings presented with IUGR, elevated transaminases, conjugated hyperbilirubinemia and hypoglycemia with progression to liver failure and death in early infancy. Results of the liver biopsy in the proband revealed cirrhosis, micro- and macrovesicular steatosis, cholestasis and mitochondrial pleomorphism. Electron microscopy of muscle revealed abnormal mitochondrial morphology and distribution while enzyme histochemistry was underwhelming. Electron transport chain testing in muscle showed increased citrate synthase activity suggesting mitochondrial proliferation, while respiratory chain activities were at the lower end of normal. mtDNA content was reduced in liver and muscle (11% and 21% of normal controls respectively). While Tfam mRNA expression was upregulated in primary fibroblasts, Tfam protein level was significantly reduced. Furthermore, functional investigations of the mitochondria revealed reduced basal respiration and spare respiratory capacity, decreased mtDNA copy number and markedly reduced nucleoids. TFAM is essential for transcription, replication and packaging of mtDNA into nucleoids. Tfam knockout mice display embryonic lethality secondary to severe mtDNA depletion. In this report, for the first time, we associate a homozygous variant in TFAM with a novel mtDNA depletion syndrome.


Assuntos
DNA Mitocondrial/genética , Proteínas de Ligação a DNA/genética , Falência Hepática/genética , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Fatores de Transcrição/genética , Animais , Replicação do DNA/genética , DNA Mitocondrial/metabolismo , Feminino , Homozigoto , Humanos , Recém-Nascido , Fígado/metabolismo , Fígado/fisiopatologia , Falência Hepática/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Doenças Mitocondriais/fisiopatologia , Mutação de Sentido Incorreto , Triagem Neonatal , Sequenciamento do Exoma
11.
Am J Hum Genet ; 91(1): 202-8, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22748208

RESUMO

Neuronal ceroid lipofuscinosis (NCL) is a genetically heterogeneous group of lysosomal diseases that collectively compose the most common Mendelian form of childhood-onset neurodegeneration. It is estimated that ∼8% of individuals diagnosed with NCL by conservative clinical and histopathologic criteria have been ruled out for mutations in the nine known NCL-associated genes, suggesting that additional genes remain unidentified. To further understand the genetic underpinnings of the NCLs, we performed whole-exome sequencing on DNA samples from a Mexican family affected by a molecularly undefined form of NCL characterized by infantile-onset progressive myoclonic epilepsy (PME), vision loss, cognitive and motor regression, premature death, and prominent NCL-type storage material. Using a recessive model to filter the identified variants, we found a single homozygous variant, c.550C>T in KCTD7, that causes a p.Arg184Cys missense change in potassium channel tetramerization domain-containing protein 7 (KCTD7) in the affected individuals. The mutation was predicted to be deleterious and was absent in over 6,000 controls. The identified variant altered the localization pattern of KCTD7 and abrogated interaction with cullin-3, a ubiquitin-ligase component and known KCTD7 interactor. Intriguingly, murine cerebellar cells derived from a juvenile NCL model (CLN3) showed enrichment of endogenous KCTD7. Whereas KCTD7 mutations have previously been linked to PME without lysosomal storage, this study clearly demonstrates that KCTD7 mutations also cause a rare, infantile-onset NCL subtype designated as CLN14.


Assuntos
Mutação , Lipofuscinoses Ceroides Neuronais/genética , Canais de Potássio/genética , Animais , Pré-Escolar , Feminino , Células HEK293 , Humanos , Lactente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linhagem , Complexo de Endopeptidases do Proteassoma/genética , Ubiquitina/genética
12.
Mol Genet Metab ; 115(4): 161-7, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26026795

RESUMO

PURPOSE: 3-Hydroxyisobutryl-CoA hydrolase (HIBCH) deficiency is a rare disorder of valine metabolism. We present a family with the oldest reported subjects with HIBCH deficiency and provide support that HIBCH deficiency should be included in the differential for elevated hydroxy-C4-carnitine in newborn screening (NBS). METHODS: Whole exome sequencing (WES) was performed on one affected sibling. HIBCH enzymatic activity was measured in patient fibroblasts. Acylcarnitines were measured by electrospray ionization tandem mass spectrometry (ESI-MS/MS). Disease incidence was estimated using a cohort of 61,434 individuals. RESULTS: Two siblings presented with infantile-onset, progressive neurodegenerative disease. WES identified a novel homozygous variant in HIBCH c.196C>T; p.Arg66Trp. HIBCH enzymatic activity was significantly reduced in patients' fibroblasts. Acylcarnitine analysis showed elevated hydroxy-C4-carnitine in blood spots of both affected siblings, including in their NBS cards, while plasma acylcarnitines were normal. Estimates show HIBCH deficiency incidence as high as 1 in ~130,000 individuals. CONCLUSION: We describe a novel family with HIBCH deficiency at the biochemical, enzymatic and molecular level. Disease incidence estimates indicate HIBCH deficiency may be under-diagnosed. This together with the elevated hydroxy-C4-carnitine found in the retrospective analysis of our patient's NBS cards suggests that this disorder could be screened for by NBS programs and should be added to the differential diagnosis for elevated hydroxy-C4-carnitine which is already measured in most NBS programs using MS/MS.


Assuntos
Anormalidades Múltiplas/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Doença de Leigh/metabolismo , Triagem Neonatal , Tioléster Hidrolases/deficiência , Anormalidades Múltiplas/metabolismo , Adolescente , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Carnitina/análogos & derivados , Carnitina/metabolismo , Criança , Pré-Escolar , Estudos de Coortes , Exoma , Feminino , Fibroblastos/enzimologia , Humanos , Lactente , Recém-Nascido , Doença de Leigh/enzimologia , Masculino , Espectrometria de Massas , Prognóstico , Estudos Retrospectivos , Análise de Sequência de DNA , Irmãos , Tioléster Hidrolases/química , Tioléster Hidrolases/metabolismo
13.
J Inherit Metab Dis ; 38(5): 905-14, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25868664

RESUMO

FBXL4 deficiency is a recently described disorder of mitochondrial maintenance associated with a loss of mitochondrial DNA in cells. To date, the genetic diagnosis of FBXL4 deficiency has been established in 28 individuals. This paper retrospectively reviews proxy-reported clinical and biochemical findings and evaluates brain imaging, morphological and genetic data in 21 of those patients. Neonatal/early-onset severe lactic acidosis, muscular hypotonia, feeding problems and failure to thrive is the characteristic pattern at first presentation. Facial dysmorphic features are present in 67% of cases. Seven children died (mean age 37 months); 11 children were alive (mean age at follow-up 46 months), three children were lost to follow-up. All survivors developed severe psychomotor retardation. Brain imaging was non-specific in neonates but a later-onset, rapidly progressive brain atrophy was noted. Elevated blood lactate and metabolic acidosis were observed in all individuals; creatine kinase was elevated in 45% of measurements. Diagnostic workup in patient tissues and cells revealed a severe combined respiratory chain defect with a general decrease of enzymes associated with mitochondrial energy metabolism and a relative depletion of mitochondrial DNA content. Mutations were detected throughout the FBXL4 gene albeit with no clear delineation of a genotype-phenotype correlation. Treatment with "mitochondrial medications" did not prove effective. In conclusion, a clinical pattern of early-onset encephalopathy, persistent lactic acidosis, profound muscular hypotonia and typical facial dysmorphism should prompt initiation of molecular genetic analysis of FBXL4. Establishment of the diagnosis permits genetic counselling, prevents patients undergoing unhelpful diagnostic procedures and allows for accurate prognosis.


Assuntos
Proteínas F-Box/genética , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/genética , Doenças Mitocondriais/patologia , Mutação , Ubiquitina-Proteína Ligases/genética , Acidose Láctica/complicações , Acidose Láctica/congênito , Acidose Láctica/genética , Criança , Pré-Escolar , Progressão da Doença , Assimetria Facial/complicações , Assimetria Facial/congênito , Assimetria Facial/genética , Família , Feminino , Estudos de Associação Genética , Humanos , Lactente , Recém-Nascido , Masculino , Doenças Mitocondriais/mortalidade , Hipotonia Muscular/complicações , Hipotonia Muscular/congênito , Hipotonia Muscular/genética , Neuroimagem , Prognóstico , Estudos Retrospectivos
14.
Genet Med ; 16(6): 477-83, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24177054

RESUMO

PURPOSE: Newborn screening includes testing for many metabolic diseases. False-positive results are higher among neonatal intensive care unit infants, resulting in increased confirmatory testing and family stress. Amino acid administration as a component of total parenteral nutrition is commonly used in the neonatal intensive care unit and suggested as a factor increasing false-positive results. The purpose of this study was to investigate the impact of a new sample collection protocol on false-positive results. METHODS: This was a 2-year retrospective cohort study. Infants were grouped by birth year into pre- and postprotocol implementation and stratified by birth weight category. In 2010, newborn screening samples were collected from all infants regardless of total parenteral nutrition administration. In 2011, the protocol was changed, and total parenteral nutrition was replaced with 10% dextrose in water (D10W) for 3 h before sample collection. RESULTS: Data from 539 neonatal intensive care unit admissions were reviewed. The new protocol reduced false-positive results for each birth weight group by at least 50% and overall by 74% (P = 0.008). The odds of having a false-positive result preintervention were 3.87 times higher than postintervention. The protocol reduced estimated costs by >80%. CONCLUSION: A protocol interrupting total parenteral nutrition for 3 h before newborn screening collection resulted in a 74% reduction in false-positive results in a neonatal intensive care unit.


Assuntos
Reações Falso-Positivas , Triagem Neonatal/métodos , Peso ao Nascer , Estudos de Coortes , Custos e Análise de Custo , Feminino , Humanos , Recém-Nascido , Masculino , Triagem Neonatal/economia , Nutrição Parenteral , Estudos Retrospectivos
15.
Genet Med ; 16(11): e1, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25356975

RESUMO

PURPOSE: Glycogen storage disease type I (GSD I) is a rare disease of variable clinical severity that primarily affects the liver and kidney. It is caused by deficient activity of the glucose 6-phosphatase enzyme (GSD Ia) or a deficiency in the microsomal transport proteins for glucose 6-phosphate (GSD Ib), resulting in excessive accumulation of glycogen and fat in the liver, kidney, and intestinal mucosa. Patients with GSD I have a wide spectrum of clinical manifestations, including hepatomegaly, hypoglycemia, lactic acidemia, hyperlipidemia, hyperuricemia, and growth retardation. Individuals with GSD type Ia typically have symptoms related to hypoglycemia in infancy when the interval between feedings is extended to 3­4 hours. Other manifestations of the disease vary in age of onset, rate of disease progression, and severity. In addition, patients with type Ib have neutropenia, impaired neutrophil function, and inflammatory bowel disease. This guideline for the management of GSD I was developed as an educational resource for health-care providers to facilitate prompt, accurate diagnosis and appropriate management of patients. METHODS: A national group of experts in various aspects of GSD I met to review the evidence base from the scientific literature and provided their expert opinions. Consensus was developed in each area of diagnosis, treatment, and management. RESULTS: This management guideline specifically addresses evaluation and diagnosis across multiple organ systems (hepatic, kidney, gastrointestinal/nutrition, hematologic, cardiovascular, reproductive) involved in GSD I. Conditions to consider in the differential diagnosis stemming from presenting features and diagnostic algorithms are discussed. Aspects of diagnostic evaluation and nutritional and medical management, including care coordination, genetic counseling, hepatic and renal transplantation, and prenatal diagnosis, are also addressed. CONCLUSION: A guideline that facilitates accurate diagnosis and optimal management of patients with GSD I was developed. This guideline helps health-care providers recognize patients with all forms of GSD I, expedite diagnosis, and minimize adverse sequelae from delayed diagnosis and inappropriate management. It also helps to identify gaps in scientific knowledge that exist today and suggests future studies.


Assuntos
Doença de Depósito de Glicogênio Tipo I/diagnóstico , Doença de Depósito de Glicogênio Tipo I/terapia , Antiporters/genética , Diagnóstico Diferencial , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/patologia , Humanos , Proteínas de Transporte de Monossacarídeos/genética
17.
Mol Genet Metab ; 111(4): 484-92, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24503138

RESUMO

Very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) is a fatty acid oxidation disorder with widely varying presentations that has presented a significant challenge to newborn screening (NBS). The Western States Regional Genetics Services Collaborative developed a workgroup to study infants with NBS positive for VLCADD. We performed retrospective analysis of newborns with elevated C14:1-acylcarnitine on NBS in California, Oregon, Washington, and Hawai'i including available confirmatory testing and clinical information. Overall, from 2,802,504 children screened, there were 242 cases screen-positive for VLCADD. There were 34 symptomatic true positive cases, 18 asymptomatic true positives, 112 false positives, 55 heterozygotes, 11 lost to follow-up, and 12 other disorders. One in 11,581 newborns had an abnormal NBS for suspected VLCADD. Comparison of analytes and analyte ratios from the NBS demonstrated statistically significant differences between true positive and false positive groups for C14:1, C14, C14:1/C2, and C14:1/C16. The positive predictive value for all true positive cases was 94%, 54%, and 23% when C14:1 was ≥2.0 µM, ≥1.0 µM, and ≥0.7 µM, respectively. Sequential post-analytical analysis could reduce the referral rate in 25.8% of cases. This study is the largest reported follow-up of infants with NBS screen-positive results for suspected VLCADD and demonstrates the necessity of developing comprehensive and consistent long-term follow-up NBS systems. Application of clinical information revealed differences between symptomatic and asymptomatic children with VLCADD. Comparison of NBS analytes and analyte ratios may be valuable in developing more effective diagnostic algorithms.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa/deficiência , Erros Inatos do Metabolismo Lipídico/diagnóstico , Doenças Mitocondriais/diagnóstico , Doenças Musculares/diagnóstico , Triagem Neonatal/métodos , Carnitina/análogos & derivados , Carnitina/metabolismo , Síndrome Congênita de Insuficiência da Medula Óssea , Análise Mutacional de DNA , Demografia , Ácidos Graxos/metabolismo , Feminino , Genótipo , Humanos , Lactente , Recém-Nascido , Masculino , Fenótipo , Reprodutibilidade dos Testes
18.
J Inherit Metab Dis ; 37(5): 791-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24619150

RESUMO

OBJECTIVE: Acyl-CoA oxidase (ACOX1) deficiency is a rare disorder of peroxisomal very-long chain fatty acid oxidation. No reports detailing attempted treatment, longitudinal imaging, or neuropathology exist. We describe the natural history of clinical symptoms and brain imaging in two siblings with ACOX1 deficiency, including the younger sibling's response to allogeneic unrelated donor hematopoietic stem cell transplantation (HSCT). METHODS: We conducted retrospective chart review to obtain clinical history, neuro-imaging, and neuropathology data. ACOX1 genotyping were performed to confirm the disease. In vitro fibroblast and neural stem cell fatty acid oxidation assays were also performed. RESULTS: Both patients experienced a fatal neurodegenerative course, with late-stage cerebellar and cerebral gray matter atrophy. Serial brain magnetic resonance imaging in the younger sibling indicated demyelination began in the medulla and progressed rostrally to include the white matter of the cerebellum, pons, midbrain, and eventually subcortical white matter. The successfully engrafted younger sibling had less brain inflammation, cortical atrophy, and neuronal loss on neuro-imaging and neuropathology compared to the untreated older sister. Fibroblasts and stem cells demonstrated deficient very long chain fatty acid oxidation. INTERPRETATION: Although HSCT did not halt the course of ACOX1 deficiency, it reduced the extent of white matter inflammation in the brain. Demyelination continued because of ongoing neuronal loss, which may be due to inability of transplant to prevent progression of gray matter disease, adverse effects of chronic corticosteroid use to control graft-versus-host disease, or intervention occurring beyond a critical point for therapeutic efficacy.


Assuntos
Acil-CoA Oxidase/deficiência , Encefalopatias Metabólicas Congênitas/terapia , Transplante de Células-Tronco Hematopoéticas/métodos , Encéfalo/patologia , Encefalopatias Metabólicas Congênitas/enzimologia , Encefalopatias Metabólicas Congênitas/patologia , Pré-Escolar , Evolução Fatal , Feminino , Movimentos da Cabeça/fisiologia , Humanos , Lactente , Recém-Nascido , Masculino , Hipotonia Muscular/etiologia , Células-Tronco Neurais/transplante , Irmãos , Resultado do Tratamento
19.
Mol Genet Metab Rep ; 38: 101025, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38125072

RESUMO

Background: LBSL is a mitochondrial disorder caused by mutations in the mitochondrial aspartyl-tRNA synthetase gene DARS2, resulting in a distinctive pattern on brain magnetic resonance imaging (MRI) and spectroscopy. Clinical presentation varies from severe infantile to chronic, slowly progressive neuronal deterioration in adolescents or adults. Most individuals with LBSL are compound heterozygous for one splicing defect in an intron 2 mutational hotspot and a second defect that could be a missense, non-sense, or splice site mutation or deletion resulting in decreased expression of the full-length protein. Aim: To present a new family with two affected members with LBSL and report a novel DARS2 mutation. Results: An 8-year-old boy (Patient 1) was referred due to headaches and abnormal MRI, suggestive of LBSL. Genetic testing revealed a previously reported c.492 + 2 T > C mutation in the DARS2 gene. Sanger sequencing uncovered a novel variant c.228-17C > G in the intron 2 hotspot. Family studies found the same genetic changes in an asymptomatic 4-year-old younger brother (Patient 2), who was found on follow-up to have an abnormal MRI. mRNA extracted from patients' fibroblasts showed that the c.228-17C > G mutation caused skipping of exon 3 resulting in lower DARS2 mRNA level. Complete absence of DARS2 protein was also found in both patients. Summary: We present a new family with two children affected with LBSL and describe a novel mutation in the DARS2 intron 2 hotspot. Despite findings of extensive white matter disease in the brain and spine, the proband in this family presented only with headaches, while the younger sibling, who also had extensive white matter changes, was asymptomatic. Our in-vitro results confirmed skipping of exon 3 in patients and family members carrying the intron 2 variant, which is consistent with previous reported mutations in intron 2 hotspots. DARS2 mRNA and protein levels were also reduced in both patients, further supporting the pathogenicity of the novel variant.

20.
Hum Mutat ; 34(3): 446-52, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23281071

RESUMO

Mitochondrial complex III (CIII) deficiency is a relatively rare disease with high clinical and genetic heterogeneity. CIII comprises 11 subunits encoded by one mitochondrial and 10 nuclear genes. Abnormalities of the nuclear genes such as BCS1L and TTC19 encoding mitochondrial assembly factors are well known, but an explanation of the majority of CIII deficiency remains elusive. Here, we report three patients from a consanguineous Mexican family presenting with neonatal onset of hypoglycemia, lactic acidosis, ketosis, and hyperammonemia. We found a homozygous missense mutation in UQCRC2 that encodes mitochondrial ubiquinol-cytochrome c reductase core protein II by whole-exome sequencing combined with linkage analysis. On the basis of structural modeling, the mutation (p.Arg183Trp) was predicted to destabilize the hydrophobic core at the subunit interface of the core protein II homodimer. In vitro studies using fibroblasts from the index patient clearly indicated CIII deficiency, as well as impaired assembly of the supercomplex formed from complexes I, III, and IV. This is the first described human disease caused by a core protein abnormality in mitochondrial CIII.


Assuntos
Complexo III da Cadeia de Transporte de Elétrons/genética , Homozigoto , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/genética , Mutação de Sentido Incorreto , ATPases Associadas a Diversas Atividades Celulares , Acidose Láctica/genética , Adulto , Western Blotting , Complexo III da Cadeia de Transporte de Elétrons/deficiência , Exoma , Feminino , Ligação Genética , Humanos , Hiperamonemia/genética , Hipoglicemia/genética , Cetose/genética , Masculino , Proteínas de Membrana/genética , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Linhagem , Conformação Proteica , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa