Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Front Pediatr ; 12: 1278047, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38445077

RESUMEN

Background/purpose: MEGDHEL syndrome is a rare autosomal recessive metabolic disorder, which is characterized by 3-methylglutaconic aciduria with deafness-dystonia, hepatopathy, encephalopathy and Leigh-like syndrome. It is caused by biallelic pathogenic variants in the SERAC1 gene. Due to the unspecific symptoms and the diverse manifestations of the clinical phenotype, the diagnosis is challenging. Infantile MEGDHEL syndrome often has a severe disease course with acute liver failure. Differentiation from other metabolic disorders is difficult and requires a multidisciplinary approach. Case presentation: A two-day-old small for gestational age neonate was admitted to our pediatric intensive care unit (PICU) due to severe liver failure with distinct hyperammonemia and hypoglycemia without elevation of transaminases or cholestasis. Due to high ammonia level, continuous hemodialysis was established immediately after admission. In addition, protein intake was stopped, and the patient anabolized with intravenous glucose. Temporary stabilization could be achieved after four days. In the further course, severe neurological and cardiocirculatory complications occurred, which ultimately led to the infant's death. In the metabolic diagnostics, a pronounced lactate acidosis and in urine an increased excretion of 3-methylglutaconic acid as well as other metabolites of mitochondrial energy metabolism has been the leading findings besides the hyperammonemia. Post-mortem trio whole genome analysis detected a homozygous pathogenic variant in SERAC1 with evidence of SERAC1 deficiency leading to the diagnosis of infantile MEGDHEL syndrome. Conclusion: When pediatricians are faced with hepatopathy or even acute liver failure without elevation of transaminases or cholestasis in newborns, SERAC1 deficiency should be considered as a potential differential diagnosis. The initial treatment is based on the recommended management of suspected metabolic disorders. Even while no cure is available yet, patients should be offered proper supportive management through a multidisciplinary team. In addition, genetic confirmation of the diagnosis is important for the families, especially regarding further family planning.If a newborn presents with hyperammonemia, hypoglycemia and impaired liver synthesis function without elevation of transaminases or cholestasis, the possible presence of MEGDHEL syndrome due to a SERAC1 mutation should be considered.

2.
J Pediatr ; 258: 113399, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37019330

RESUMEN

We describe 16 infants born preterm with birth weights <1500 g and transient hyperinsulinism. The onset of hyperinsulinism was delayed and often coincident with clinical stabilization. We hypothesize that postnatal stress caused by prematurity and associated problems may contribute to development of delayed-onset transient hyperinsulinism.


Asunto(s)
Hiperinsulinismo , Hipoglucemia , Enfermedades Pancreáticas , Recién Nacido , Humanos , Lactante , Hipoglucemia/complicaciones , Hipoglucemia/diagnóstico , Estudios de Cohortes , Recien Nacido con Peso al Nacer Extremadamente Bajo , Hiperinsulinismo/complicaciones , Recien Nacido Prematuro , Enfermedades Pancreáticas/complicaciones
3.
Stem Cell Res Ther ; 13(1): 256, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35715829

RESUMEN

The most frequent biochemical defect of inherited mitochondrial disease is isolated complex I deficiency. There is no cure for this disorder, and treatment is mainly supportive. In this study, we investigated the effects of human mesenchymal stem cells (MSCs) on skin fibroblast derived from three individuals with complex I deficiency carrying different pathogenic variants in mitochondrial DNA-encoded subunits (MT-ND3, MT-ND6). Complex I-deficient fibroblasts were transiently co-cultured with bone marrow-derived MSCs. Mitochondrial transfer was analysed by fluorescence labelling and validated by Sanger sequencing. Levels of reactive oxygen species (ROS) were measured using MitoSOX Red. Moreover, mitochondrial respiration was analysed by Seahorse XFe96 Extracellular Flux Analyzer. Levels of antioxidant proteins were investigated via immunoblotting. Co-culturing of complex I-deficient fibroblast with MSCs lowered cellular ROS levels. The effect on ROS production was more sustained compared to treatment of patient fibroblasts with culture medium derived from MSC cultures. Investigation of cellular antioxidant defence systems revealed an upregulation of SOD2 (superoxide dismutase 2, mitochondrial) and HO-1 (heme oxygenase 1) in patient-derived cell lines. This adaptive response was normalised upon MSC treatment. Moreover, Seahorse experiments revealed a significant improvement of mitochondrial respiration, indicating a mitigation of the oxidative phosphorylation defect. Experiments with repetitive MSC co-culture at two consecutive time points enhanced this effect. Our study indicates that MSC-based treatment approaches might constitute an interesting option for patients with mitochondrial DNA-encoded mitochondrial diseases. We suggest that this strategy may prove more promising for defects caused by mitochondrial DNA variants compared to nuclear-encoded defects.


Asunto(s)
Antioxidantes , Células Madre Mesenquimatosas , Antioxidantes/metabolismo , Línea Celular , ADN Mitocondrial/genética , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Fibroblastos/metabolismo , Homeostasis , Humanos , Células Madre Mesenquimatosas/metabolismo , Enfermedades Mitocondriales , NADH Deshidrogenasa/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Respiración
4.
J Med Genet ; 59(9): 878-887, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34656997

RESUMEN

BACKGROUND: Human coenzyme Q4 (COQ4) is essential for coenzyme Q10 (CoQ10) biosynthesis. Pathogenic variants in COQ4 cause childhood-onset neurodegeneration. We aimed to delineate the clinical spectrum and the cellular consequences of COQ4 deficiency. METHODS: Clinical course and neuroradiological findings in a large cohort of paediatric patients with COQ4 deficiency were analysed. Functional studies in patient-derived cell lines were performed. RESULTS: We characterised 44 individuals from 36 families with COQ4 deficiency (16 newly described). A total of 23 different variants were identified, including four novel variants in COQ4. Correlation analyses of clinical and neuroimaging findings revealed three disease patterns: type 1: early-onset phenotype with neonatal brain anomalies and epileptic encephalopathy; type 2: intermediate phenotype with distinct stroke-like lesions; and type 3: moderate phenotype with non-specific brain pathology and a stable disease course. The functional relevance of COQ4 variants was supported by in vitro studies using patient-derived fibroblast lines. Experiments revealed significantly decreased COQ4 protein levels, reduced levels of cellular CoQ10 and elevated levels of the metabolic intermediate 6-demethoxyubiquinone. CONCLUSION: Our study describes the heterogeneous clinical presentation of COQ4 deficiency and identifies phenotypic subtypes. Cell-based studies support the pathogenic characteristics of COQ4 variants. Due to the insufficient clinical response to oral CoQ10 supplementation, alternative treatment strategies are warranted.


Asunto(s)
Proteínas Mitocondriales , Ubiquinona , Línea Celular , Niño , Humanos , Recién Nacido , Proteínas Mitocondriales/genética , Neuroimagen , Fenotipo , Ubiquinona/genética , Ubiquinona/metabolismo
5.
Biochim Biophys Acta Bioenerg ; 1861(8): 148202, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32275929

RESUMEN

Protein complexes from the oxidative phosphorylation (OXPHOS) system are assembled with the help of proteins called assembly factors. We here delineate the function of the inner mitochondrial membrane protein TMEM70, in which mutations have been linked to OXPHOS deficiencies, using a combination of BioID, complexome profiling and coevolution analyses. TMEM70 interacts with complex I and V and for both complexes the loss of TMEM70 results in the accumulation of an assembly intermediate followed by a reduction of the next assembly intermediate in the pathway. This indicates that TMEM70 has a role in the stability of membrane-bound subassemblies or in the membrane recruitment of subunits into the forming complex. Independent evidence for a role of TMEM70 in OXPHOS assembly comes from evolutionary analyses. The TMEM70/TMEM186/TMEM223 protein family, of which we show that TMEM186 and TMEM223 are mitochondrial in human as well, only occurs in species with OXPHOS complexes. Our results validate the use of combining complexome profiling with BioID and evolutionary analyses in elucidating congenital defects in protein complex assembly.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Biotinilación , Evolución Molecular , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Fosforilación Oxidativa , Unión Proteica
7.
J Pediatr ; 196: 309-313.e3, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29395179

RESUMEN

We demonstrate that a heterozygous nuclear variant in the gene encoding mitochondrial complex I subunit NDUFV1 aggravates the cellular phenotype in the presence of a mitochondrial DNA variant in complex I subunit ND1. Our findings suggest that heterozygous variants could be more significant in inherited mitochondrial diseases than hitherto assumed.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Enfermedades Mitocondriales/genética , NADH Deshidrogenasa/genética , Niño , ADN Mitocondrial/genética , Complejo I de Transporte de Electrón/genética , Femenino , Pruebas Genéticas/métodos , Heterocigoto , Humanos , Recién Nacido , Masculino , Enfermedades Mitocondriales/diagnóstico , Mutación , Fenotipo
8.
Eur J Hum Genet ; 25(11): 1273-1277, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28853723

RESUMEN

Mitochondrial respiratory chain complex I consists of 44 different subunits and can be subgrouped into three functional modules: the Q-, the P- and the N-module. NDUFAF4 (C6ORF66) is an assembly factor of complex I that associates with assembly intermediates of the Q-module. Via exome sequencing, we identified a homozygous missense variant in a complex I-deficient patient with Leigh syndrome. Supercomplex analysis in patient fibroblasts revealed specifically altered stoichiometry. Detailed assembly analysis of complex I, indicative of all of its assembly routes, showed an accumulation of parts of the P- and the N-module but not the Q-module. Lentiviral complementation of patient fibroblasts with wild-type NDUFAF4 rescued complex I deficiency and the assembly defect, confirming the causal role of the variant. Our report on the second family affected by an NDUFAF4 variant further characterizes the phenotypic spectrum and sheds light into the role of NDUFAF4 in mitochondrial complex I biogenesis.


Asunto(s)
Proteínas de Unión a Calmodulina/genética , Enfermedad de Leigh/genética , Mutación Missense , Proteínas de Unión a Calmodulina/metabolismo , Células Cultivadas , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Fibroblastos/metabolismo , Homocigoto , Humanos , Lactante , Enfermedad de Leigh/patología , Masculino , Multimerización de Proteína
9.
Stem Cell Res Ther ; 8(1): 150, 2017 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-28646906

RESUMEN

BACKGROUND: Disorders of the oxidative phosphorylation (OXPHOS) system represent a large group among the inborn errors of metabolism. The most frequently observed biochemical defect is isolated deficiency of mitochondrial complex I (CI). No effective treatment strategies for CI deficiency are so far available. The purpose of this study was to investigate whether and how mesenchymal stem cells (MSCs) are able to modulate metabolic function in fibroblast cell models of CI deficiency. METHODS: We used human and murine fibroblasts with a defect in the nuclear DNA encoded NDUFS4 subunit of CI. Fibroblasts were co-cultured with MSCs under different stress conditions and intercellular mitochondrial transfer was assessed by flow cytometry and fluorescence microscopy. Reactive oxygen species (ROS) levels were measured using MitoSOX-Red. Protein levels of CI were analysed by blue native polyacrylamide gel electrophoresis (BN-PAGE). RESULTS: Direct cellular interactions and mitochondrial transfer between MSCs and human as well as mouse fibroblast cell lines were demonstrated. Mitochondrial transfer was visible in 13.2% and 6% of fibroblasts (e.g. fibroblasts containing MSC mitochondria) for human and mouse cell lines, respectively. The transfer rate could be further stimulated via treatment of cells with TNF-α. MSCs effectively lowered cellular ROS production in NDUFS4-deficient fibroblast cell lines (either directly via co-culture or indirectly via incubation of cell lines with cell-free MSC supernatant). However, CI protein expression and activity were not rescued by MSC treatment. CONCLUSION: This study demonstrates the interplay between MSCs and fibroblast cell models of isolated CI deficiency including transfer of mitochondria as well as modulation of cellular ROS levels. Further exploration of these cellular interactions might help to develop MSC-based treatment strategies for human CI deficiency.


Asunto(s)
Complejo I de Transporte de Electrón , Fibroblastos/enzimología , Células Madre Mesenquimatosas/enzimología , Mitocondrias/enzimología , NADH Deshidrogenasa , Fosforilación Oxidativa , Animales , Técnicas de Cocultivo , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/metabolismo , Fibroblastos/citología , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Mitocondrias/genética , NADH Deshidrogenasa/deficiencia , NADH Deshidrogenasa/metabolismo
10.
Hum Mutat ; 38(6): 692-703, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28247525

RESUMEN

COX5A is a nuclear-encoded subunit of mitochondrial respiratory chain complex IV (cytochrome c oxidase). We present patients with a homozygous pathogenic variant in the COX5A gene. Clinical details of two affected siblings suffering from early-onset pulmonary arterial hypertension, lactic acidemia, failure to thrive, and isolated complex IV deficiency are presented. We show that the variant lies within the evolutionarily conserved COX5A/COX4 interface domain, suggesting that it alters the interaction between these two subunits during complex IV biogenesis. In patient skin fibroblasts, the enzymatic activity and protein levels of complex IV and several of its subunits are reduced. Lentiviral complementation rescues complex IV deficiency. The monomeric COX1 assembly intermediate accumulates demonstrating a function of COX5A in complex IV biogenesis. A potential therapeutic lead is demonstrated by showing that copper supplementation leads to partial rescue of complex IV deficiency in patient fibroblasts.


Asunto(s)
Acidosis Láctica/genética , Ciclooxigenasa 1/genética , Grupo Citocromo c/genética , Insuficiencia de Crecimiento/genética , Hipertensión Pulmonar/genética , Acidosis Láctica/patología , Núcleo Celular/genética , Ciclooxigenasa 1/química , Grupo Citocromo c/química , Deficiencia de Citocromo-c Oxidasa , Complejo IV de Transporte de Electrones , Insuficiencia de Crecimiento/patología , Fibroblastos , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Hipertensión Pulmonar/patología , Mitocondrias/genética , Mutación , Subunidades de Proteína/genética
11.
Mol Genet Metab ; 120(3): 243-246, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27986404

RESUMEN

NDUFAF3 is an assembly factor of mitochondrial respiratory chain complex I. Variants in NDUFAF3 have been identified as a cause of severe multisystem mitochondrial disease. In a patient presenting with Leigh syndrome, which has hitherto not been described as a clinical feature of NDUFAF3 deficiency, we identified a novel homozygous variant and confirmed its pathogenicity in patient fibroblasts studies. Furthermore, we present an analysis of complex I assembly routes representative of each functional module and, thereby, link NDUFAF3 to a specific step in complex I assembly. Therefore, our report expands the phenotype of NDUFAF3 deficiency and further characterizes the role of NDUFAF3 in complex I biogenesis.


Asunto(s)
Enfermedad de Leigh/genética , Proteínas Mitocondriales/genética , Mutación , Análisis de Secuencia de ADN/métodos , Células Cultivadas , Exoma , Resultado Fatal , Femenino , Fibroblastos/citología , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Lactante , Enfermedad de Leigh/patología , Fenotipo
12.
Cell Metab ; 25(1): 128-139, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-27720676

RESUMEN

Mitochondrial complex I is the largest integral membrane enzyme of the respiratory chain and consists of 44 different subunits encoded in the mitochondrial and nuclear genome. Its biosynthesis is a highly complicated and multifaceted process involving at least 14 additional assembly factors. How these subunits assemble into a functional complex I and where the assembly factors come into play is largely unknown. Here, we applied a dynamic complexome profiling approach to elucidate the assembly of human mitochondrial complex I and its further incorporation into respiratory chain supercomplexes. We delineate the stepwise incorporation of all but one subunit into a series of distinct assembly intermediates and their association with known and putative assembly factors, which had not been implicated in this process before. The resulting detailed and comprehensive model of complex I assembly is fully consistent with recent structural data and the remarkable modular architecture of this multiprotein complex.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Mitocondrias/metabolismo , Línea Celular Tumoral , Cloranfenicol/farmacología , Humanos , Modelos Biológicos , Complejos Multiproteicos/metabolismo , Subunidades de Proteína/metabolismo , Proteoma/metabolismo , Factores de Tiempo
13.
Metab Brain Dis ; 32(1): 267-270, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27502409

RESUMEN

VARS2 encodes a mitochondrial aminoacyl-tRNA-synthetase. Mutations in VARS2 have recently been identified as a cause of mitochondrial encephalomyopathy in three individuals. However, clinical information remained scarce. Exome sequencing lead us to identify compound heterozygous pathogenic VARS2 variants in a boy presenting with severe lactic acidosis, hypertrophic cardiomyopathy, epilepsy, and abnormalities on brain imaging including hypoplasia of corpus callosum and cerebellum as well as a massive lactate peak on MR-spectroscopy. Studies in patient-derived fibroblasts confirmed the functional relevance of the identified VARS2 variants. Our report expands the phenotypic spectrum associated with this rare mitochondrial defect, in that VARS2 deficiency may also cause severe neonatal presentations with cardiac involvement and structural brain abnormalities.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Epilepsia/genética , Antígenos HLA/genética , Encefalomiopatías Mitocondriales/genética , Mutación , Valina-ARNt Ligasa/genética , Análisis Mutacional de ADN , Exoma , Humanos , Masculino
14.
Childs Nerv Syst ; 32(11): 2077-2083, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27449766

RESUMEN

The knowledge about the genetic spectrum underlying paediatric mitochondrial diseases is rapidly growing. As a consequence, the range of neuroimaging findings associated with mitochondrial diseases became extremely broad. This has important implications for radiologists and clinicians involved in the care of these patients. Here, we provide a condensed overview of brain magnetic resonance imaging (MRI) findings in children with genetically confirmed mitochondrial diseases. The neuroimaging spectrum ranges from classical Leigh syndrome with symmetrical lesions in basal ganglia and/or brain stem to structural abnormalities including cerebellar hypoplasia and corpus callosum dysgenesis. We highlight that, although some imaging patterns can be suggestive of a genetically defined mitochondrial syndrome, brain MRI-based candidate gene prioritization is only successful in a subset of patients.


Asunto(s)
Encéfalo/patología , Enfermedades Mitocondriales/patología , Niño , Humanos , Imagen por Resonancia Magnética , Neuroimagen
15.
Brain ; 139(Pt 2): 346-54, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26626369

RESUMEN

Thioredoxin 2 (TXN2; also known as Trx2) is a small mitochondrial redox protein essential for the control of mitochondrial reactive oxygen species homeostasis, apoptosis regulation and cell viability. Exome sequencing in a 16-year-old adolescent suffering from an infantile-onset neurodegenerative disorder with severe cerebellar atrophy, epilepsy, dystonia, optic atrophy, and peripheral neuropathy, uncovered a homozygous stop mutation in TXN2. Analysis of patient-derived fibroblasts demonstrated absence of TXN2 protein, increased reactive oxygen species levels, impaired oxidative stress defence and oxidative phosphorylation dysfunction. Reconstitution of TXN2 expression restored all these parameters, indicating the causal role of TXN2 mutation in disease development. Supplementation with antioxidants effectively suppressed cellular reactive oxygen species production, improved cell viability and mitigated clinical symptoms during short-term follow-up. In conclusion, our report on a patient with TXN2 deficiency suggests an important role of reactive oxygen species homeostasis for human neuronal maintenance and energy metabolism.


Asunto(s)
Homeostasis/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales/deficiencia , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/metabolismo , Niño , Humanos , Masculino , Mitocondrias/genética , Proteínas Mitocondriales/genética , Enfermedades Neurodegenerativas/genética , Oxidación-Reducción , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/genética
16.
Neurogenetics ; 16(4): 319-23, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25797485

RESUMEN

Defects in mitochondrial translation may lead to combined respiratory chain deficiency and typically cause childhood-onset multisystem disease. Only recently, a homozygous missense mutation (c.467T > G, p.Leu156Arg) in MRPL44, encoding a protein of the large subunit of the mitochondrial ribosome, has been identified in two siblings with hypertrophic cardiomyopathy. Using exome sequencing, we identified two further unrelated patients harboring the previously reported mutation c.467T > G, p.Leu156Arg in MRPL44 in the homozygous state and compound heterozygous with a novel missense mutation c.233G > A, p.Arg78Gln, respectively. Both patients presented with childhood-onset hypertrophic cardiomyopathy, which seems to be the core clinical feature associated with MRPL44 deficiency. However, we observed several additional clinical signs and symptoms including pigmentary retinopathy, hemiplegic migraine, Leigh-like lesions on brain MRI, renal insufficiency, and hepatopathy. Our findings expand the clinical spectrum associated with MRPL44 mutations and indicate that MRPL44-associated mitochondrial dysfunction can also manifest as a progressive multisystem disease with central nervous system involvement. Of note, neurological and neuro-ophthalmological impairment seems to be a disease feature of the second and third decades of life, which should be taken into account in patient management and counseling.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Progresión de la Enfermedad , Proteínas Mitocondriales/genética , Mutación Missense , Proteínas Ribosómicas/genética , Adulto , Encéfalo/patología , Cardiomiopatía Hipertrófica/complicaciones , Cardiomiopatía Hipertrófica/patología , Niño , Exoma , Femenino , Humanos , Linaje
17.
Neurogenetics ; 16(3): 237-40, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25663021

RESUMEN

The mitochondrial ribosomes are required for the synthesis of mitochondrial DNA-encoded subunits of the oxidative phosphorylation (OXPHOS) system. Here, we present a neonate with fatal lactic acidosis and combined OXPHOS deficiency caused by a homozygous mutation in MRPS22, a gene encoding a mitochondrial ribosomal small subunit protein. Brain imaging revealed several structural abnormalities, including agenesis of the corpus callosum, multiple periventricular cysts, and suspected intracerebral calcifications. Moreover, echocardiography demonstrated atrial and ventricular septal defects as well as a coronary artery fistula. Our report expands the clinical spectrum of this rare mitochondrial disorder and confirms the severe clinical phenotype associated with this defect.


Asunto(s)
Acidosis Láctica/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Proteínas Ribosómicas/genética , Acidosis Láctica/complicaciones , Encéfalo/patología , Resultado Fatal , Fibroblastos/metabolismo , Mutación del Sistema de Lectura , Humanos , Recién Nacido , Masculino , Enfermedades Mitocondriales/complicaciones , Proteínas Mitocondriales/metabolismo , Miocardio/patología , Proteínas Ribosómicas/metabolismo
18.
Hum Mutat ; 36(1): 34-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25339201

RESUMEN

COA6/C1ORF31 is involved in cytochrome c oxidase (complex IV) biogenesis. We present a new pathogenic COA6 variant detected in a patient with neonatal hypertrophic cardiomyopathy and isolated complex IV deficiency. For the first time, clinical details about a COA6-deficient patient are given and patient fibroblasts are functionally characterized: COA6 protein is undetectable and steady-state levels of complex IV and several of its subunits are reduced. The monomeric COX1 assembly intermediate accumulates. Using pulse-chase experiments, we demonstrate an increased turnover of mitochondrial encoded complex IV subunits. Although monomeric complex IV is decreased in patient fibroblasts, the CI/CIII2 /CIVn -supercomplexes remain unaffected. Copper supplementation shows a partial rescue of complex IV deficiency in patient fibroblasts. We conclude that COA6 is required for complex IV subunit stability. Furthermore, the proposed role in the copper delivery pathway to complex IV subunits is substantiated and a therapeutic lead for COA6-deficient patients is provided.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Deficiencia de Citocromo-c Oxidasa/genética , Complejo IV de Transporte de Electrones/genética , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/patología , Cobre/administración & dosificación , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Células HEK293 , Humanos , Recién Nacido , Mitocondrias/metabolismo
19.
Eur J Pediatr ; 173(12): 1719-22, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25233985

RESUMEN

UNLABELLED: Malonic aciduria is an extremely rare autosomal recessive inborn error of metabolism. We present clinical, biochemical and genetic information for several years of follow-up of new malonic aciduria patients who were diagnosed by newborn screening. These data are discussed with regard to treatment options and possible diagnostic pitfalls. The cases presented here show that the course of malonic aciduria is unpredictable and can even significantly differ in two siblings harbouring identical mutations. Early treatment can lead to the rapid improvement of cardiomyopathy in the course of malonic aciduria. Biochemical parameters seem to be variable and can intermittently be undetectable in the blood or urine samples of affected patients. Therefore, confirmatory tests following a positive newborn screening should be taken with caution and include both malonyl carnitine detection in dried blood spots and urinary organic acid analysis as initial measures. CONCLUSION: Patients with a suspected or confirmed diagnosis of malonic aciduria should undergo thorough diagnostic procedures and be regularly screened for complications such as cardiomyopathy even when they are asymptomatic in order to ensure early therapy of treatable complications.


Asunto(s)
Carboxiliasas/deficiencia , Errores Innatos del Metabolismo/diagnóstico , Tamizaje Neonatal/métodos , Hermanos , Niño , Diagnóstico Diferencial , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Masculino , Malonil Coenzima A , Ácido Metilmalónico , Factores de Tiempo
20.
J Neurol Neurosurg Psychiatry ; 85(3): 257-65, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23772060

RESUMEN

Leigh syndrome is a devastating neurodegenerative disease, typically manifesting in infancy or early childhood. However, also late-onset cases have been reported. Since its first description by Denis Archibald Leigh in 1951, it has evolved from a postmortem diagnosis, strictly defined by histopathological observations, to a clinical entity with indicative laboratory and radiological findings. Hallmarks of the disease are symmetrical lesions in the basal ganglia or brain stem on MRI, and a clinical course with rapid deterioration of cognitive and motor functions. Examinations of fresh muscle tissue or cultured fibroblasts are important tools to establish a biochemical and genetic diagnosis. Numerous causative mutations in mitochondrial and nuclear genes, encoding components of the oxidative phosphorylation system have been described in the past years. Moreover, dysfunctions in pyruvate dehydrogenase complex or coenzyme Q10 metabolism may be associated with Leigh syndrome. To date, there is no cure for affected patients, and treatment options are mostly unsatisfactory. Here, we review the most important clinical aspects of Leigh syndrome, and discuss diagnostic steps as well as treatment options.


Asunto(s)
Enfermedad de Leigh/diagnóstico , Adolescente , Adulto , Edad de Inicio , Biopsia , Encéfalo/patología , Niño , Preescolar , ADN Mitocondrial/genética , Diagnóstico Diferencial , Asesoramiento Genético , Humanos , Enfermedad de Leigh/genética , Enfermedad de Leigh/patología , Enfermedad de Leigh/terapia , Imagen por Resonancia Magnética , Músculo Esquelético/patología , Neuroimagen , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...