Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Mol Psychiatry ; 28(2): 668-697, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36385166

RESUMO

Missense and truncating variants in the X-chromosome-linked CLCN4 gene, resulting in reduced or complete loss-of-function (LOF) of the encoded chloride/proton exchanger ClC-4, were recently demonstrated to cause a neurocognitive phenotype in both males and females. Through international clinical matchmaking and interrogation of public variant databases we assembled a database of 90 rare CLCN4 missense variants in 90 families: 41 unique and 18 recurrent variants in 49 families. For 43 families, including 22 males and 33 females, we collated detailed clinical and segregation data. To confirm causality of variants and to obtain insight into disease mechanisms, we investigated the effect on electrophysiological properties of 59 of the variants in Xenopus oocytes using extended voltage and pH ranges. Detailed analyses revealed new pathophysiological mechanisms: 25% (15/59) of variants demonstrated LOF, characterized by a "shift" of the voltage-dependent activation to more positive voltages, and nine variants resulted in a toxic gain-of-function, associated with a disrupted gate allowing inward transport at negative voltages. Functional results were not always in line with in silico pathogenicity scores, highlighting the complexity of pathogenicity assessment for accurate genetic counselling. The complex neurocognitive and psychiatric manifestations of this condition, and hitherto under-recognized impacts on growth, gastrointestinal function, and motor control are discussed. Including published cases, we summarize features in 122 individuals from 67 families with CLCN4-related neurodevelopmental condition and suggest future research directions with the aim of improving the integrated care for individuals with this diagnosis.


Assuntos
Transtornos do Neurodesenvolvimento , Masculino , Feminino , Humanos , Transtornos do Neurodesenvolvimento/genética , Mutação de Sentido Incorreto , Genes Ligados ao Cromossomo X , Fenótipo , Canais de Cloreto/genética
2.
J Assist Reprod Genet ; 39(3): 609-618, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35064435

RESUMO

PURPOSE: Providing additional insights on the efficacy of human nuclear transfer (NT). Here, and earlier, NT has been applied to minimize transmission risk of mitochondrial DNA (mtDNA) diseases. NT has also been proposed for treating infertility, but it is still unclear which infertility indications would benefit. In this work, we therefore additionally assess the applicability of NT to overcome failed fertilization. METHODS: Patient 1 carries a homoplasmic mtDNA mutation (m.11778G > A). Seventeen metaphase II (MII) oocytes underwent pre-implantation genetic testing (PGT), while five MII oocytes were used for spindle transfer (ST), and one in vitro matured (IVM) metaphase I oocyte underwent early pronuclear transfer (ePNT). Patients 2-3 experienced multiple failed intracytoplasmic sperm injection (ICSI) and ICSI-assisted oocyte activation (AOA) cycles. For these patients, the obtained MII oocytes underwent an additional ICSI-AOA cycle, while the IVM oocytes were subjected to ST. RESULTS: For patient 1, PGT-M confirmed mutation loads close to 100%. All ST-reconstructed oocytes fertilized and cleaved, of which one progressed to the blastocyst stage. The reconstructed ePNT-zygote reached the morula stage. These samples showed an average mtDNA carry-over rate of 2.9% ± 0.8%, confirming the feasibility of NT to reduce mtDNA transmission. For patient 2-3 displaying fertilization failure, ST resulted in, respectively, 4/5 and 6/6 fertilized oocytes, providing evidence, for the first time, that NT can enable successful fertilization in this patient population. CONCLUSION: Our study showcases the repertoire of disorders for which NT can be beneficial, to overcome either mitochondrial disease transmission or failed fertilization after ICSI-AOA.


Assuntos
Infertilidade , Doenças Mitocondriais , DNA Mitocondrial/genética , Fertilização , Fertilização in vitro/métodos , Humanos , Infertilidade/genética , Infertilidade/terapia , Oócitos , Injeções de Esperma Intracitoplásmicas
3.
Clin Genet ; 99(2): 259-268, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33131045

RESUMO

The CAMTA1-associated phenotype was initially defined in patients with intragenic deletions and duplications who showed nonprogressive congenital ataxia, with or without intellectual disability. Here, we describe 10 individuals with CAMTA1 variants: nine previously unreported (likely) pathogenic variants comprising one missense, four frameshift and four nonsense variants, and one missense variant of unknown significance. Six patients were diagnosed following whole exome sequencing and four individuals with exome-based targeted panel analysis. Most of them present with developmental delay, manifesting in speech and motor delay. Other frequent findings are hypotonia, cognitive impairment, cerebellar dysfunction, oculomotor abnormalities, and behavioral problems. Feeding problems occur more frequently than previously observed. In addition, we present a systematic review of 19 previously published individuals with causal variants, including copy number, truncating, and missense variants. We note a tendency of more severe cognitive impairment and recurrent dysmorphic features in individuals with a copy number variant. Pathogenic variants are predominantly observed in and near the N- and C- terminal functional domains. Clinical heterogeneity is observed, but 3'-terminal variants seem to associate with less pronounced cerebellar dysfunction.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Doenças do Sistema Nervoso/genética , Transativadores/genética , Adolescente , Criança , Pré-Escolar , Transtornos Cognitivos/genética , Análise Mutacional de DNA , Deficiências do Desenvolvimento/genética , Feminino , Humanos , Masculino , Fenótipo
4.
Muscle Nerve ; 61(2): 173-181, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31749205

RESUMO

INTRODUCTION: Small fiber neuropathies (SFN) are associated with a reduction in quality of life. In adults, epidermal nerve fiber density (END) analysis is recommended for the diagnosis of SFN. In children, END assessment is not often performed. We analyzed small nerve fiber innervation to elucidate the potential diagnostic role of skin biopsies in young patients with pain. METHODS: Epidermal nerve fiber density and sudomotor neurite density (SND) were assessed in skin biopsies from 26 patients aged 7 to 20 years (15 female patients) with unexplained chronic pain. The results were compared with clinical data. RESULTS: Epidermal nerve fiber density was abnormal in 50% and borderline in 35% of patients. An underlying medical condition was found in 42% of patients, including metabolic, autoimmune, and genetic disorders. DISCUSSION: Reduction of epidermal nerve fibers can be associated with treatable conditions. Therefore, the analysis of END in children with pain may help to uncover a possible cause and guide potential treatment options.


Assuntos
Dor Crônica/diagnóstico , Dor Crônica/patologia , Fibras Nervosas/patologia , Pele/patologia , Neuropatia de Pequenas Fibras/patologia , Adolescente , Biópsia , Criança , Epiderme/inervação , Epiderme/patologia , Feminino , Humanos , Masculino , Neuralgia/diagnóstico , Neuritos/patologia , Medição da Dor , Glândulas Sudoríparas/inervação , Glândulas Sudoríparas/patologia , Adulto Jovem
5.
Am J Hum Genet ; 99(1): 217-27, 2016 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-27374774

RESUMO

Complex I deficiency is the most common biochemical phenotype observed in individuals with mitochondrial disease. With 44 structural subunits and over 10 assembly factors, it is unsurprising that complex I deficiency is associated with clinical and genetic heterogeneity. Massively parallel sequencing (MPS) technologies including custom, targeted gene panels or unbiased whole-exome sequencing (WES) are hugely powerful in identifying the underlying genetic defect in a clinical diagnostic setting, yet many individuals remain without a genetic diagnosis. These individuals might harbor mutations in poorly understood or uncharacterized genes, and their diagnosis relies upon characterization of these orphan genes. Complexome profiling recently identified TMEM126B as a component of the mitochondrial complex I assembly complex alongside proteins ACAD9, ECSIT, NDUFAF1, and TIMMDC1. Here, we describe the clinical, biochemical, and molecular findings in six cases of mitochondrial disease from four unrelated families affected by biallelic (c.635G>T [p.Gly212Val] and/or c.401delA [p.Asn134Ilefs(∗)2]) TMEM126B variants. We provide functional evidence to support the pathogenicity of these TMEM126B variants, including evidence of founder effects for both variants, and establish defects within this gene as a cause of complex I deficiency in association with either pure myopathy in adulthood or, in one individual, a severe multisystem presentation (chronic renal failure and cardiomyopathy) in infancy. Functional experimentation including viral rescue and complexome profiling of subject cell lines has confirmed TMEM126B as the tenth complex I assembly factor associated with human disease and validates the importance of both genome-wide sequencing and proteomic approaches in characterizing disease-associated genes whose physiological roles have been previously undetermined.


Assuntos
Alelos , Complexo I de Transporte de Elétrons/deficiência , Proteínas de Membrana/genética , Doenças Mitocondriais/genética , Mutação/genética , Fenótipo , Adolescente , Adulto , Idade de Início , Sequência de Aminoácidos , Criança , Complexo I de Transporte de Elétrons/genética , Feminino , Humanos , Lactente , Masculino , Proteínas de Membrana/química , Pessoa de Meia-Idade , Linhagem , Adulto Jovem
6.
Genet Med ; 21(4): 1021-1026, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30293988

RESUMO

PURPOSE: RAC3 is an underexamined member of the Rho GTPase gene family that is expressed in the developing brain and linked to key cellular functions. De novo missense variants in the homolog RAC1 were recently associated with developmental disorders. In the RAC subfamily, transforming missense changes at certain shared residues have been observed in human cancers and previously characterized in experimental studies. The purpose of this study was to determine whether constitutional dysregulation of RAC3 is associated with human disease. METHODS: We discovered a RAC3 variant in the index case using genome sequencing, and searched for additional variants using international data-sharing initiatives. Functional effects of the variants were assessed using a multifaceted approach generalizable to most clinical laboratory settings. RESULTS: We rapidly identified five individuals with de novo monoallelic missense variants in RAC3, including one recurrent change. Every participant had severe intellectual disability and brain malformations. In silico protein modeling, and prior in vivo and in situ experiments, supported a transforming effect for each of the three different RAC3 variants. All variants were observed in databases of somatic variation in cancer. CONCLUSIONS: Missense variants in RAC3 cause a novel brain disorder, likely through a mechanism of constitutive protein activation.


Assuntos
Predisposição Genética para Doença , Deficiência Intelectual/genética , Transtornos do Neurodesenvolvimento/genética , Proteínas rac de Ligação ao GTP/genética , Adulto , Pré-Escolar , GTP Fosfo-Hidrolases/genética , Humanos , Recém-Nascido , Deficiência Intelectual/diagnóstico por imagem , Deficiência Intelectual/fisiopatologia , Mutação de Sentido Incorreto , Transtornos do Neurodesenvolvimento/diagnóstico por imagem , Transtornos do Neurodesenvolvimento/fisiopatologia , Fenótipo , Sequenciamento Completo do Genoma
7.
J Inherit Metab Dis ; 42(5): 898-908, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31276219

RESUMO

Exome sequencing has recently identified mutations in the gene TANGO2 (transport and Golgi organization 2) as a cause of developmental delay associated with recurrent crises involving rhabdomyolysis, cardiac arrhythmias, and metabolic derangements. The disease is not well understood, in part as the cellular function and subcellular localization of the TANGO2 protein remain unknown. Furthermore, the clinical syndrome with its heterogeneity of symptoms, signs, and laboratory findings is still being defined. Here, we describe 11 new cases of TANGO2-related disease, confirming and further expanding the previously described clinical phenotype. Patients were homozygous or compound heterozygous for previously described exonic deletions or new frameshift, splice site, and missense mutations. All patients showed developmental delay with ataxia, dysarthria, intellectual disability, or signs of spastic diplegia. Of importance, we identify two subjects (aged 12 and 17 years) who have never experienced any overt episode of the catabolism-induced metabolic crises typical for the disease. Mitochondrial complex II activity was mildly reduced in patients investigated in association with crises but normal in other patients. In one deceased patient, post-mortem autopsy revealed heterotopic neurons in the cerebral white matter, indicating a possible role for TANGO2 in neuronal migration. Furthermore, we have addressed the subcellular localization of several alternative isoforms of TANGO2, none of which were mitochondrial but instead appeared to have a primarily cytoplasmic localization. Previously described aberrations in Golgi morphology were not observed in cultured skin fibroblasts.


Assuntos
Translocador Nuclear Receptor Aril Hidrocarboneto/deficiência , Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Deficiências do Desenvolvimento/genética , Metabolismo Energético/genética , Deficiência Intelectual/genética , Mitocôndrias/genética , Adolescente , Translocador Nuclear Receptor Aril Hidrocarboneto/fisiologia , Ataxia/genética , Paralisia Cerebral/genética , Criança , Pré-Escolar , Disartria/genética , Exoma , Éxons , Feminino , Humanos , Masculino , Mutação , Linhagem , Fenótipo , Sequenciamento do Exoma
9.
Am J Hum Genet ; 95(6): 708-20, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25434004

RESUMO

Respiratory chain deficiencies exhibit a wide variety of clinical phenotypes resulting from defective mitochondrial energy production through oxidative phosphorylation. These defects can be caused by either mutations in the mtDNA or mutations in nuclear genes coding for mitochondrial proteins. The underlying pathomechanisms can affect numerous pathways involved in mitochondrial physiology. By whole-exome and candidate gene sequencing, we identified 11 individuals from 9 families carrying compound heterozygous or homozygous mutations in GTPBP3, encoding the mitochondrial GTP-binding protein 3. Affected individuals from eight out of nine families presented with combined respiratory chain complex deficiencies in skeletal muscle. Mutations in GTPBP3 are associated with a severe mitochondrial translation defect, consistent with the predicted function of the protein in catalyzing the formation of 5-taurinomethyluridine (τm(5)U) in the anticodon wobble position of five mitochondrial tRNAs. All case subjects presented with lactic acidosis and nine developed hypertrophic cardiomyopathy. In contrast to individuals with mutations in MTO1, the protein product of which is predicted to participate in the generation of the same modification, most individuals with GTPBP3 mutations developed neurological symptoms and MRI involvement of thalamus, putamen, and brainstem resembling Leigh syndrome. Our study of a mitochondrial translation disorder points toward the importance of posttranscriptional modification of mitochondrial tRNAs for proper mitochondrial function.


Assuntos
Acidose Láctica/genética , Encefalopatias/genética , Cardiomiopatia Hipertrófica/genética , Proteínas de Ligação ao GTP/genética , Processamento de Proteína Pós-Traducional , Acidose Láctica/fisiopatologia , Sequência de Aminoácidos , Encéfalo/patologia , Encefalopatias/fisiopatologia , Cardiomiopatia Hipertrófica/fisiopatologia , Linhagem Celular , Criança , Pré-Escolar , Consanguinidade , Feminino , Fibroblastos , Proteínas de Ligação ao GTP/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Dados de Sequência Molecular , Mutação , Linhagem , Biossíntese de Proteínas , Interferência de RNA , RNA de Transferência/genética , RNA de Transferência/metabolismo , Alinhamento de Sequência
10.
Mol Genet Metab ; 122(4): 172-181, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29126765

RESUMO

Mutations in FARS2 are known to cause dysfunction of mitochondrial translation due to deficient aminoacylation of the mitochondrial phenylalanine tRNA. Here, we report three novel mutations in FARS2 found in two patients in a compound heterozygous state. The missense mutation c.1082C>T (p.Pro361Leu) was detected in both patients. The mutations c.461C>T (p.Ala154Val) and c.521_523delTGG (p.Val174del) were each detected in one patient. We report abnormal in vitro aminoacylation assays as a functional validation of the molecular genetic findings. Based on the phenotypic data of previously reported subjects and the two subjects reported here, we conclude that FARS2 deficiency can be associated with two phenotypes: (i) an epileptic phenotype, and (ii) a spastic paraplegia phenotype.


Assuntos
Epilepsia/genética , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Fenótipo , Fenilalanina-tRNA Ligase/deficiência , Fenilalanina-tRNA Ligase/genética , Paraplegia Espástica Hereditária/genética , Adolescente , Aminoacil-tRNA Sintetases/metabolismo , Aminoacilação , Encéfalo/diagnóstico por imagem , Células Cultivadas , Exoma , Feminino , Fibroblastos/metabolismo , Heterozigoto , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Músculo Esquelético/patologia , Mutação de Sentido Incorreto/genética , Consumo de Oxigênio , RNA de Transferência/metabolismo , Análise de Sequência de DNA
12.
Biochim Biophys Acta ; 1853(2): 285-98, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25450972

RESUMO

The tight interrelationship between peroxisomes and mitochondria is illustrated by their cooperation in lipid metabolism, antiviral innate immunity and shared use of proteins executing organellar fission. In addition, we previously reported that disruption of peroxisome biogenesis in hepatocytes severely impacts on mitochondrial integrity, primarily damaging the inner membrane. Here we investigated the molecular impairments of the dysfunctional mitochondria in hepatocyte selective Pex5 knockout mice. First, by using blue native electrophoresis and in-gel activity stainings we showed that the respiratory complexes were differentially affected with reduction of complexes I and III and incomplete assembly of complex V, whereas complexes II and IV were normally active. This resulted in impaired oxygen consumption in cultured Pex5(-/-) hepatocytes. Second, mitochondrial DNA was depleted causing an imbalance in the expression of mitochondrial- and nuclear-encoded subunits of the respiratory chain complexes. Third, mitochondrial membranes showed increased permeability and fluidity despite reduced content of the polyunsaturated fatty acid docosahexaenoic acid. Fourth, the affected mitochondria in peroxisome deficient hepatocytes displayed increased oxidative stress. Acute deletion of PEX5 in vivo using adeno-Cre virus phenocopied these effects, indicating that mitochondrial perturbations closely follow the loss of functional peroxisomes in time. Likely to compensate for the functional impairments, the volume of the mitochondrial compartment was increased several folds. This was not driven by PGC-1α but mediated by activation of PPARα, possibly through c-myc overexpression. In conclusion, loss of peroxisomal metabolism in hepatocytes perturbs the mitochondrial inner membrane, depletes mitochondrial DNA and causes mitochondrial biogenesis independent of PGC-1α.


Assuntos
DNA Mitocondrial/metabolismo , Hepatócitos/metabolismo , Mitocôndrias/metabolismo , Peroxissomos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Compartimento Celular , Proliferação de Células , Respiração Celular , Transporte de Elétrons , Deleção de Genes , Hepatócitos/ultraestrutura , Lipídeos/química , Fluidez de Membrana , Camundongos Knockout , Mitocôndrias/ultraestrutura , Oxirredução , Fosforilação Oxidativa , Estresse Oxidativo , PPAR alfa/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Receptor 1 de Sinal de Orientação para Peroxissomos , Subunidades Proteicas/metabolismo , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/metabolismo
13.
J Med Genet ; 52(8): 532-40, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25787132

RESUMO

BACKGROUND: Mitochondrial disease is often suspected in cases of severe epileptic encephalopathy especially when a complex movement disorder, liver involvement and progressive developmental regression are present. Although mutations in either mitochondrial DNA or POLG are often present, other nuclear defects in mitochondrial DNA replication and protein translation have been associated with a severe epileptic encephalopathy. METHODS AND RESULTS: We identified a proband with an epileptic encephalopathy, complex movement disorder and a combined mitochondrial respiratory chain enzyme deficiency. The child presented with neurological regression, complex movement disorder and intractable seizures. A combined deficiency of mitochondrial complexes I, III and IV was noted in liver tissue, along with increased mitochondrial DNA content in skeletal muscle. Incomplete assembly of complex V, using blue native polyacrylamide gel electrophoretic analysis and complex I, using western blotting, suggested a disorder of mitochondrial transcription or translation. Exome sequencing identified compound heterozygous mutations in CARS2, a mitochondrial aminoacyl-tRNA synthetase. Both mutations affect highly conserved amino acids located within the functional ligase domain of the cysteinyl-tRNA synthase. A specific decrease in the amount of charged mt-tRNA(Cys) was detected in patient fibroblasts compared with controls. Retroviral transfection of the wild-type CARS2 into patient skin fibroblasts led to the correction of the incomplete assembly of complex V, providing functional evidence for the role of CARS2 mutations in disease aetiology. CONCLUSIONS: Our findings indicate that mutations in CARS2 result in a mitochondrial translational defect as seen in individuals with mitochondrial epileptic encephalopathy.


Assuntos
Aminoacil-tRNA Sintetases/genética , Encefalopatias/genética , Epilepsia/genética , Sequência de Aminoácidos , Aminoacilação , Criança , Análise Mutacional de DNA , Exoma , Humanos , Masculino , Dados de Sequência Molecular , RNA de Transferência/metabolismo , Alinhamento de Sequência
14.
Hum Mutat ; 36(2): 222-31, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25385316

RESUMO

A homozygous missense mutation (c.822G>C) was found in the gene encoding the mitochondrial asparaginyl-tRNA synthetase (NARS2) in two siblings born to consanguineous parents. These siblings presented with different phenotypes: one had mild intellectual disability and epilepsy in childhood, whereas the other had severe myopathy. Biochemical analysis of the oxidative phosphorylation (OXPHOS) complexes in both siblings revealed a combined complex I and IV deficiency in skeletal muscle. In-gel activity staining after blue native-polyacrylamide gel electrophoresis confirmed the decreased activity of complex I and IV, and, in addition, showed the presence of complex V subcomplexes. Considering the consanguineous descent, homozygosity mapping and whole-exome sequencing were combined revealing the presence of one single missense mutation in the shared homozygous region. The c.822G>C variant affects the 3' splice site of exon 7, leading to skipping of the whole exon 7 and a part of exon 8 in the NARS2 mRNA. In EBV-transformed lymphoblasts, a specific decrease in the amount of charged mt-tRNA(Asn) was demonstrated as compared with controls. This confirmed the pathogenic nature of the variant. To conclude, the reported variant in NARS2 results in a combined OXPHOS complex deficiency involving complex I and IV, making NARS2 a new member of disease-associated aaRS2.


Assuntos
Aspartato-tRNA Ligase/genética , Mutação de Sentido Incorreto , Adulto , Aspartato-tRNA Ligase/metabolismo , Sequência de Bases , Células Cultivadas , Consanguinidade , Análise Mutacional de DNA , Feminino , Estudos de Associação Genética , Homozigoto , Humanos , Masculino , Doenças Musculares/genética , Biossíntese de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sítios de Splice de RNA
15.
Hum Mol Genet ; 22(13): 2590-602, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23462291

RESUMO

Two siblings from consanguineous parents died perinatally with a condition characterized by generalized hypotonia, respiratory insufficiency, arthrogryposis, microcephaly, congenital brain malformations and hyperglycinemia. Catalytic activities of the mitochondrial respiratory complexes I and II were deficient in skeletal muscle, a finding suggestive of an inborn error in mitochondrial biogenesis. Homozygosity mapping identified IBA57 located in the largest homozygous region on chromosome 1 as a culprit candidate gene. IBA57 is known to be involved in the biosynthesis of mitochondrial [4Fe-4S] proteins. Sequence analysis of IBA57 revealed the homozygous mutation c.941A > C, p.Gln314Pro. Severely decreased amounts of IBA57 protein were observed in skeletal muscle and cultured skin fibroblasts from the affected subjects. HeLa cells depleted of IBA57 showed biochemical defects resembling the ones found in patient-derived cells, including a decrease in various mitochondrial [4Fe-4S] proteins and in proteins covalently linked to lipoic acid (LA), a cofactor produced by the [4Fe-4S] protein LA synthase. The defects could be complemented by wild-type IBA57 and partially by mutant IBA57. As a result of the mutation, IBA57 protein was excessively degraded, an effect ameliorated by protease inhibitors. Hence, we propose that the mutation leads to partial functional impairment of IBA57, yet the major pathogenic impact is due to its proteolytic degradation below physiologically critical levels. In conclusion, the ensuing lethal complex biochemical phenotype of a novel metabolic syndrome results from multiple Fe/S protein defects caused by a deficiency in the Fe/S cluster assembly protein IBA57.


Assuntos
Encefalopatias/genética , Proteínas de Transporte/genética , Doenças Musculares/genética , Mutação , Encéfalo/patologia , Encefalopatias/diagnóstico , Proteínas de Transporte/metabolismo , Consanguinidade , Análise Mutacional de DNA , Transporte de Elétrons/genética , Feminino , Fibroblastos/metabolismo , Humanos , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Músculo Esquelético/metabolismo , Doenças Musculares/diagnóstico , Linhagem , Fenótipo , Irmãos , Pele/metabolismo
16.
Am J Med Genet A ; 167A(12): 3214-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26358756

RESUMO

VPS45 mutations cause severe congenital neutropenia (SCN). We report on a girl with SCN and neurological impairment harboring a homozygous p.E238K mutation in VPS45 (vacuolar sorting protein 45). She successfully underwent hematopoietic stem cell transplantation. Our findings delineate the phenotype and indicate a possible genotype-phenotype correlation for neurological involvement.


Assuntos
Homozigoto , Mutação/genética , Doenças do Sistema Nervoso/etiologia , Neutropenia/congênito , Proteínas de Transporte Vesicular/genética , Pré-Escolar , Síndrome Congênita de Insuficiência da Medula Óssea , Feminino , Genótipo , Humanos , Doenças do Sistema Nervoso/patologia , Neutropenia/complicações , Neutropenia/genética , Neutropenia/patologia , Fenótipo , Prognóstico
17.
Am J Med Genet A ; 167A(12): 3038-45, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26420380

RESUMO

The ability to identify the clinical nature of the recurrent duplication of chromosome 17q12 has been limited by its rarity and the diverse range of phenotypes associated with this genomic change. In order to further define the clinical features of affected patients, detailed clinical information was collected in the largest series to date (30 patients and 2 of their siblings) through a multi-institutional collaborative effort. The majority of patients presented with developmental delays varying from mild to severe. Though dysmorphic features were commonly reported, patients do not have consistent and recognizable features. Cardiac, ophthalmologic, growth, behavioral, and other abnormalities were each present in a subset of patients. The newly associated features potentially resulting from 17q12 duplication include height and weight above the 95th percentile, cataracts, microphthalmia, coloboma, astigmatism, tracheomalacia, cutaneous mosaicism, pectus excavatum, scoliosis, hypermobility, hypospadias, diverticulum of Kommerell, pyloric stenosis, and pseudohypoparathryoidism. The majority of duplications were inherited with some carrier parents reporting learning disabilities or microcephaly. We identified additional, potentially contributory copy number changes in a subset of patients, including one patient each with 16p11.2 deletion and 15q13.3 deletion. Our data further define and expand the clinical spectrum associated with duplications of 17q12 and provide support for the role of genomic modifiers contributing to phenotypic variability.


Assuntos
Anormalidades Múltiplas/genética , Duplicação Cromossômica , Adolescente , Criança , Pré-Escolar , Variações do Número de Cópias de DNA , Deficiências do Desenvolvimento/genética , Face/anormalidades , Feminino , Humanos , Lactente , Masculino , Microcefalia/genética , Fenótipo , Adulto Jovem
18.
Anesthesiology ; 122(2): 343-52, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25296107

RESUMO

BACKGROUND: Propofol is a short-acting intravenous anesthetic agent. In rare conditions, a life-threatening complication known as propofol infusion syndrome can occur. The pathophysiologic mechanism is still unknown. Some studies suggested that propofol acts as uncoupling agent, others suggested that it inhibits complex I or complex IV, or causes increased oxidation of cytochrome c and cytochrome aa3, or inhibits mitochondrial fatty acid metabolism. Although the exact site of interaction is not known, most hypotheses point to the direction of the mitochondria. METHODS: Eight rats were ventilated and sedated with propofol up to 20 h. Sequential biopsy specimens were taken from liver and skeletal muscle and used for determination of respiratory chain activities and propofol concentration. Activities were also measured in skeletal muscle from a patient who died of propofol infusion syndrome. RESULTS: In rats, authors detected a decrease in complex II+III activity starting at low tissue concentration of propofol (20 to 25 µM), further declining at higher concentrations. Before starting anesthesia, the complex II+III/citrate synthase activity ratio in liver was 0.46 (0.25) and in skeletal muscle 0.23 (0.05) (mean [SD]). After 20 h of anesthesia, the ratios declined to 0.17 (0.03) and 0.12 (0.02), respectively. When measured individually, the activities of complexes II and III remained normal. Skeletal muscle from one patient taken in the acute phase of propofol infusion syndrome also shows a selective decrease in complex II+III activity (z-score: -2.96). CONCLUSION: Propofol impedes the electron flow through the respiratory chain and coenzyme Q is the main site of interaction with propofol.


Assuntos
Anestésicos Intravenosos/toxicidade , Propofol/toxicidade , Ubiquinona/metabolismo , Animais , Ciclo do Ácido Cítrico/efeitos dos fármacos , Transporte de Elétrons/efeitos dos fármacos , Masculino , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Ratos , Ratos Wistar , Respiração Artificial , Síndrome
19.
J Inherit Metab Dis ; 38(6): 1147-53, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25971455

RESUMO

Leukodystrophies are a heterogeneous group of severe genetic neurodegenerative disorders. A multiple mitochondrial dysfunctions syndrome was found in an infant presenting with a progressive leukoencephalopathy. Homozygosity mapping, whole exome sequencing, and functional studies were used to define the underlying molecular defect. Respiratory chain studies in skeletal muscle isolated from the proband revealed a combined deficiency of complexes I and II. In addition, western blotting indicated lack of protein lipoylation. The combination of these findings was suggestive for a defect in the iron-sulfur (Fe/S) protein assembly pathway. SNP array identified loss of heterozygosity in large chromosomal regions, covering the NFU1 and BOLA3, and the IBA57 and ABCB10 candidate genes, in 2p15-p11.2 and 1q31.1-q42.13, respectively. A homozygous c.436C > T (p.Arg146Trp) variant was detected in IBA57 using whole exome sequencing. Complementation studies in a HeLa cell line depleted for IBA57 showed that the mutant protein with the semi-conservative amino acid exchange was unable to restore the biochemical phenotype indicating a loss-of-function mutation of IBA57. In conclusion, defects in the Fe/S protein assembly gene IBA57 can cause autosomal recessive neurodegeneration associated with progressive leukodystrophy and fatal outcome at young age. In the affected patient, the biochemical phenotype was characterized by a defect in the respiratory chain complexes I and II and a decrease in mitochondrial protein lipoylation, both resulting from impaired assembly of Fe/S clusters.


Assuntos
Proteínas de Transporte/genética , Proteínas Ferro-Enxofre/genética , Leucoencefalopatias/diagnóstico , Leucoencefalopatias/genética , Doenças Mitocondriais/diagnóstico , Complexo I de Transporte de Elétrons/genética , Evolução Fatal , Heterozigoto , Homozigoto , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino , Mitocôndrias/genética , Mutação , Fenótipo
20.
Eur J Pediatr ; 174(2): 267-70, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25009317

RESUMO

UNLABELLED: The propositus presented with hypotonia, respiratory failure, and seizures in the newborn period and was found to have severe hyperlactacidemia and a hypertrophic heart. He carried a de novo pathogenic mutation (m.8993 T>G) in the gene encoding subunit 6 of the mitochondrial ATP synthase (MTATP6). Although the lactate concentration in serum normalized and the proband recovered after a short period at the neonatal intensive care unit, his ultimate motor and cognitive development was poor. Brain MRI at the age of 6 months showed bilaterally signal abnormalities in the caudate nucleus, putamen, thalamus, and mesencephalon. He died at the age of 9 months. The difficulty in genetic counseling in families with a maternal mitochondrial mutation disorder is emphasized. CONCLUSION: Here, we report on a neonate with the m.8993 T>G mutation and emphasize implications of mtDNA disorders on family planning decisions.


Assuntos
Acidose Láctica/genética , DNA Mitocondrial/genética , Doença de Leigh/genética , Mitocôndrias/genética , ATPases Mitocondriais Próton-Translocadoras/genética , Mutação Puntual/genética , Evolução Fatal , Aconselhamento Genético , Humanos , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA