Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 146(4): 1357-1372, 2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-36074901

RESUMEN

The vacuolar H+-ATPase is an enzymatic complex that functions in an ATP-dependent manner to pump protons across membranes and acidify organelles, thereby creating the proton/pH gradient required for membrane trafficking by several different types of transporters. We describe heterozygous point variants in ATP6V0C, encoding the c-subunit in the membrane bound integral domain of the vacuolar H+-ATPase, in 27 patients with neurodevelopmental abnormalities with or without epilepsy. Corpus callosum hypoplasia and cardiac abnormalities were also present in some patients. In silico modelling suggested that the patient variants interfere with the interactions between the ATP6V0C and ATP6V0A subunits during ATP hydrolysis. Consistent with decreased vacuolar H+-ATPase activity, functional analyses conducted in Saccharomyces cerevisiae revealed reduced LysoSensor fluorescence and reduced growth in media containing varying concentrations of CaCl2. Knockdown of ATP6V0C in Drosophila resulted in increased duration of seizure-like behaviour, and the expression of selected patient variants in Caenorhabditis elegans led to reduced growth, motor dysfunction and reduced lifespan. In summary, this study establishes ATP6V0C as an important disease gene, describes the clinical features of the associated neurodevelopmental disorder and provides insight into disease mechanisms.


Asunto(s)
Epilepsia , ATPasas de Translocación de Protón Vacuolares , Humanos , ATPasas de Translocación de Protón Vacuolares/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Epilepsia/genética , Adenosina Trifosfato
2.
J Med Genet ; 60(1): 65-73, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-34872991

RESUMEN

BACKGROUND: Large-scale mitochondrial DNA deletions (LMD) are a common genetic cause of mitochondrial disease and give rise to a wide range of clinical features. Lack of longitudinal data means the natural history remains unclear. This study was undertaken to describe the clinical spectrum in a large cohort of patients with paediatric disease onset. METHODS: A retrospective multicentre study was performed in patients with clinical onset <16 years of age, diagnosed and followed in seven European mitochondrial disease centres. RESULTS: A total of 80 patients were included. The average age at disease onset and at last examination was 10 and 31 years, respectively. The median time from disease onset to death was 11.5 years. Pearson syndrome was present in 21%, Kearns-Sayre syndrome spectrum disorder in 50% and progressive external ophthalmoplegia in 29% of patients. Haematological abnormalities were the hallmark of the disease in preschool children, while the most common presentations in older patients were ptosis and external ophthalmoplegia. Skeletal muscle involvement was found in 65% and exercise intolerance in 25% of the patients. Central nervous system involvement was frequent, with variable presence of ataxia (40%), cognitive involvement (36%) and stroke-like episodes (9%). Other common features were pigmentary retinopathy (46%), short stature (42%), hearing impairment (39%), cardiac disease (39%), diabetes mellitus (25%) and renal disease (19%). CONCLUSION: Our study provides new insights into the phenotypic spectrum of childhood-onset, LMD-associated syndromes. We found a wider spectrum of more prevalent multisystem involvement compared with previous studies, most likely related to a longer time of follow-up.


Asunto(s)
Síndrome de Kearns-Sayre , Enfermedades Musculares , Oftalmoplejía Externa Progresiva Crónica , Preescolar , Humanos , Niño , Anciano , ADN Mitocondrial/genética , Síndrome de Kearns-Sayre/epidemiología , Síndrome de Kearns-Sayre/genética , Oftalmoplejía Externa Progresiva Crónica/epidemiología , Oftalmoplejía Externa Progresiva Crónica/genética , Enfermedades Musculares/genética , Progresión de la Enfermedad
3.
BMC Ophthalmol ; 23(1): 394, 2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37752499

RESUMEN

BACKGROUND: We describe the case of a 47-year-old man referred to a retinal clinic and diagnosed with late-onset retinitis pigmentosa. Surprisingly, genetic testing revealed compound heterozygous pathogenic variants in GNPTG, leading to the diagnosis of the autosomal recessive lysosomal storage disorder mucolipidosis type III gamma. Mucolipidosis type III gamma is typically diagnosed during childhood due to symptoms relating to skeletal dysplasia. Retinal dystrophy is not a common phenotypic feature. CASE PRESENTATION: Ophthalmologic examination was consistent with a mild form of retinitis pigmentosa and included fundus photography, measurement of best-corrected visual acuity, optical coherence tomography, electroretinogram and visual field testing. Extraocular findings included joint restriction and pains from an early age leading to bilateral hip replacement by age 30, aortic insufficiency, and hypertension. Genetic analysis was performed by whole genome sequencing filtered for a gene panel of 325 genes associated with retinal disease. Two compound heterozygous pathogenic variants were identified in GNPTG, c.347_349del and c.607dup. The diagnosis of mucolipidosis type III gamma was confirmed biochemically by measurement of increased activities of specific lysosomal enzymes in plasma. CONCLUSION: To our knowledge this is the first description of retinitis pigmentosa caused by compound heterozygous variants in GNPTG, providing further indications that late-onset retinal dystrophy is part of the phenotypic spectrum of mucolipidosis type III gamma.


Asunto(s)
Mucolipidosis , Distrofias Retinianas , Masculino , Humanos , Adulto , Persona de Mediana Edad , Mucolipidosis/diagnóstico , Mucolipidosis/genética , Secuenciación Completa del Genoma , Electrorretinografía , Transferasas (Grupos de Otros Fosfatos Sustitutos)
4.
J Inherit Metab Dis ; 45(2): 248-263, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34873726

RESUMEN

The vast clinical and radiological spectrum of pyruvate dehydrogenase complex (PDHc) deficiency continues to pose challenges both in diagnostics and disease monitoring. Prompt diagnosis is important to enable early initiation of ketogenic diet. The patients were recruited from an ongoing population-based study in Sweden. All patients with a genetically confirmed diagnosis who had been investigated with an MRI of the brain were included. Repeated investigations were assessed to study the evolution of the MRI changes. Sixty-two MRI investigations had been performed in 34 patients (23 females). The genetic cause was mutations in PDHA1 in 29, PDHX and DLAT in 2 each, and PDHB in 1. The lesions were prenatal developmental in 16, prenatal clastic in 18, and postnatal clastic in 15 individuals. Leigh-like lesions with predominant involvement of globus pallidus were present in 12, while leukoencephalopathy was present in 6 and stroke-like lesions in 3 individuals. A combination of prenatal developmental and clastic lesions was present in 15 individuals. In addition, one male with PDHA1 also had postnatal clastic lesions. The most common lesions found in our study were agenesis or hypoplasia of corpus callosum, ventriculomegaly, or Leigh-like lesions. Furthermore, we describe a broad spectrum of other MRI changes that include leukoencephalopathy and stroke-like lesions. We argue that a novel important clue, suggesting the possibility of PDHc deficiency on MRI scans, is the simultaneous presence of multiple lesions on MRI that have occurred during different phases of brain development.


Asunto(s)
Leucoencefalopatías , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa , Accidente Cerebrovascular , Encéfalo/patología , Femenino , Humanos , Leucoencefalopatías/diagnóstico por imagen , Leucoencefalopatías/patología , Imagen por Resonancia Magnética , Masculino , Embarazo , Piruvato Deshidrogenasa (Lipoamida)/genética , Complejo Piruvato Deshidrogenasa/genética , Complejo Piruvato Deshidrogenasa/metabolismo , Accidente Cerebrovascular/patología
5.
Hum Mutat ; 42(4): 378-384, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33502047

RESUMEN

Mutations in structural subunits and assembly factors of complex I of the oxidative phosphorylation system constitute the most common cause of mitochondrial respiratory chain defects. Such mutations can present a wide range of clinical manifestations, varying from mild deficiencies to severe, lethal disorders. We describe a patient presenting intrauterine growth restriction and anemia, which displayed postpartum hypertrophic cardiomyopathy, lactic acidosis, encephalopathy, and a severe complex I defect with fatal outcome. Whole genome sequencing revealed an intronic biallelic mutation in the NDUFB7 gene (c.113-10C>G) and splicing pattern alterations in NDUFB7 messenger RNA were confirmed by RNA Sequencing. The detected variant resulted in a significant reduction of the NDUFB7 protein and reduced complex I activity. Complementation studies with expression of wild-type NDUFB7 in patient fibroblasts normalized complex I function. Here we report a case with a primary complex I defect due to a homozygous mutation in an intron region of the NDUFB7 gene.


Asunto(s)
Acidosis Láctica , Cardiomiopatía Hipertrófica , Enfermedades Mitocondriales , NADH NADPH Oxidorreductasas/genética , Acidosis Láctica/genética , Cardiomiopatía Hipertrófica/genética , Complejo I de Transporte de Electrón/genética , Humanos , Enfermedades Mitocondriales/genética , Mutación
6.
J Pediatr ; 228: 240-251.e2, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32827528

RESUMEN

OBJECTIVES: To evaluate the clinical symptoms and biochemical findings and establish the genetic etiology in a cohort of pediatric patients with combined deficiencies of the mitochondrial respiratory chain complexes. STUDY DESIGN: Clinical and biochemical data were collected from 55 children. All patients were subjected to sequence analysis of the entire mitochondrial genome, except when the causative mutations had been identified based on the clinical picture. Whole exome sequencing/whole genome sequencing (WES/WGS) was performed in 32 patients. RESULTS: Onset of disease was generally early in life (median age, 6 weeks). The most common symptoms were muscle weakness, hypotonia, and developmental delay/intellectual disability. Nonneurologic symptoms were frequent. Disease causing mutations were found in 20 different nuclear genes, and 7 patients had mutations in mitochondrial DNA. Causative variants were found in 18 of the 32 patients subjected to WES/WGS. Interestingly, many patients had low levels of coenzyme Q10 in muscle, irrespective of genetic cause. CONCLUSIONS: Children with combined enzyme defects display a diversity of clinical symptoms with varying age of presentation. We established the genetic diagnosis in 35 of the 55 patients (64%). The high diagnostic yield was achieved by the introduction of massive parallel sequencing, which also revealed novel genes and enabled elucidation of new disease mechanisms.


Asunto(s)
ADN Mitocondrial/genética , Enfermedades Metabólicas/genética , Enfermedades Mitocondriales/genética , Mutación , Ubiquinona/análogos & derivados , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Humanos , Lactante , Recién Nacido , Enfermedades Metabólicas/enzimología , Enfermedades Mitocondriales/enzimología , Ubiquinona/sangre , Secuenciación del Exoma , Adulto Joven
7.
Am J Hum Genet ; 101(6): 965-976, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29220678

RESUMEN

Zellweger spectrum disorders (ZSDs) are autosomal-recessive disorders that are caused by defects in peroxisome biogenesis due to bi-allelic mutations in any of 13 different PEX genes. Here, we identified seven unrelated individuals affected with an apparent dominant ZSD in whom a heterozygous mutant PEX6 allele (c.2578C>T [p.Arg860Trp]) was overrepresented due to allelic expression imbalance (AEI). We demonstrated that AEI of PEX6 is a common phenomenon and is correlated with heterozygosity for a frequent variant in the 3' untranslated region (UTR) of the mutant allele, which disrupts the most distal of two polyadenylation sites. Asymptomatic parents, who were heterozygous for PEX c.2578C>T, did not show AEI and were homozygous for the 3' UTR variant. Overexpression models confirmed that the overrepresentation of the pathogenic PEX6 c.2578T variant compared to wild-type PEX6 c.2578C results in a peroxisome biogenesis defect and thus constitutes the cause of disease in the affected individuals. AEI promoting the overrepresentation of a mutant allele might also play a role in other autosomal-recessive disorders, in which only one heterozygous pathogenic variant is identified.


Asunto(s)
Regiones no Traducidas 3'/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , Desequilibrio Alélico/genética , Síndrome de Zellweger/genética , Alelos , Células Cultivadas , Femenino , Regulación de la Expresión Génica/genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Peroxisomas/genética , Peroxisomas/patología , Secuenciación del Exoma
8.
J Inherit Metab Dis ; 43(4): 726-736, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32391929

RESUMEN

BACKGROUND: Variants in POLG are one of the most common causes of inherited mitochondrial disease. Phenotypic classification of POLG disease has evolved haphazardly making it complicated and difficult to implement in everyday clinical practise. The aim of our study was to simplify the classification and facilitate better clinical recognition. METHODS: A multinational, retrospective study using data from 155 patients with POLG variants recruited from seven European countries. RESULTS: We describe the spectrum of clinical features associated with POLG variants in the largest known cohort of patients. While clinical features clearly form a continuum, stratifying patients simply according to age of onset-onset prior to age 12 years; onset between 12 and 40 years and onset after the age of 40 years, permitted us to identify clear phenotypic and prognostic differences. Prior to 12 years of age, liver involvement (87%), seizures (84%), and feeding difficulties (84%) were the major features. For those with onset between 12 and 40 years, ataxia (90%), peripheral neuropathy (84%), and seizures (71%) predominated, while for those with onset over 40 years, ptosis (95%), progressive external ophthalmoplegia (89%), and ataxia (58%) were the major clinical features. The earlier the onset the worse the prognosis. Patients with epilepsy and those with compound heterozygous variants carried significantly worse prognosis. CONCLUSION: Based on our data, we propose a simplified POLG disease classification, which can be used to guide diagnostic investigations and predict disease course.


Asunto(s)
ADN Polimerasa gamma/genética , Predisposición Genética a la Enfermedad/genética , Enfermedades Mitocondriales/clasificación , Enfermedades Mitocondriales/genética , Adolescente , Adulto , Edad de Inicio , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Europa (Continente) , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Enfermedades Mitocondriales/mortalidad , Mutación , Estudios Retrospectivos , Análisis de Supervivencia , Adulto Joven
9.
Hum Mol Genet ; 26(13): 2515-2525, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28430993

RESUMEN

Mutations in the mitochondrial DNA polymerase, POLG, are associated with a variety of clinical presentations, ranging from early onset fatal brain disease in Alpers syndrome to chronic progressive external ophthalmoplegia. The majority of mutations are linked with disturbances of mitochondrial DNA (mtDNA) integrity and maintenance. On a molecular level, depending on their location within the enzyme, mutations either lead to mtDNA depletion or the accumulation of multiple mtDNA deletions, and in some cases these molecular changes can be correlated to the clinical presentation. We identified a patient with a dominant p.Y955H mutation in POLG, presenting with a severe, early-onset multi-systemic mitochondrial disease with bilateral sensorineural hearing loss, cataract, myopathy, and liver failure. Using a combination of disease models of Drosophila melanogaster and in vitro biochemistry analysis, we compare the molecular consequences of the p.Y955H mutation to the well-documented p.Y955C mutation. We demonstrate that both mutations affect mtDNA replication and display a dominant negative effect, with the p.Y955H allele resulting in a more severe polymerase dysfunction.


Asunto(s)
ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , ADN Polimerasa gamma , Replicación del ADN/genética , ADN Mitocondrial/genética , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Femenino , Humanos , Lactante , Mitocondrias/genética , Mutación/genética , Oftalmoplejía Externa Progresiva Crónica/enzimología , Linaje , Fenotipo
10.
J Med Genet ; 55(1): 21-27, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29101127

RESUMEN

BACKGROUND: Leigh syndrome is a phenotypically and genetically heterogeneous mitochondrial disorder. While some genetic defects are associated with well-described phenotypes, phenotype-genotype correlations in Leigh syndrome are not fully explored. OBJECTIVE: We aimed to identify phenotype-genotype correlations in Leigh syndrome in a large cohort of systematically evaluated patients. METHODS: We studied 96 patients with genetically confirmed Leigh syndrome diagnosed and followed in eight European centres specialising in mitochondrial diseases. RESULTS: We found that ataxia, ophthalmoplegia and cardiomyopathy were more prevalent among patients with mitochondrial DNA defects. Patients with mutations in MT-ND and NDUF genes with complex I deficiency shared common phenotypic features, such as early development of central nervous system disease, followed by high occurrence of cardiac and ocular manifestations. The cerebral cortex was affected in patients with NDUF mutations significantly more often than the rest of the cohort. Patients with the m.8993T>G mutation in MT-ATP6 gene had more severe clinical and radiological manifestations and poorer disease outcome compared with patients with the m.8993T>C mutation. CONCLUSION: Our study provides new insights into phenotype-genotype correlations in Leigh syndrome and particularly in patients with complex I deficiency and with defects in the mitochondrial ATP synthase.


Asunto(s)
Estudios de Asociación Genética , Enfermedad de Leigh/genética , Núcleo Celular/metabolismo , ADN/genética , ADN Mitocondrial/genética , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , Mutación/genética , Fenotipo
11.
Am J Hum Genet ; 97(5): 761-8, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26522469

RESUMEN

S-adenosylmethionine (SAM) is the predominant methyl group donor and has a large spectrum of target substrates. As such, it is essential for nearly all biological methylation reactions. SAM is synthesized by methionine adenosyltransferase from methionine and ATP in the cytoplasm and subsequently distributed throughout the different cellular compartments, including mitochondria, where methylation is mostly required for nucleic-acid modifications and respiratory-chain function. We report a syndrome in three families affected by reduced intra-mitochondrial methylation caused by recessive mutations in the gene encoding the only known mitochondrial SAM transporter, SLC25A26. Clinical findings ranged from neonatal mortality resulting from respiratory insufficiency and hydrops to childhood acute episodes of cardiopulmonary failure and slowly progressive muscle weakness. We show that SLC25A26 mutations cause various mitochondrial defects, including those affecting RNA stability, protein modification, mitochondrial translation, and the biosynthesis of CoQ10 and lipoic acid.


Asunto(s)
Sistemas de Transporte de Aminoácidos/genética , Proteínas de Unión al Calcio/genética , Metilación de ADN , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Debilidad Muscular/genética , Mutación/genética , S-Adenosilmetionina/metabolismo , Secuencia de Aminoácidos , Preescolar , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Debilidad Muscular/patología , Linaje , Pronóstico , Estabilidad del ARN , Homología de Secuencia de Aminoácido , Ácido Tióctico/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo
12.
Ann Neurol ; 82(6): 1004-1015, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29205472

RESUMEN

OBJECTIVE: 3-Methylglutaconic aciduria, dystonia-deafness, hepatopathy, encephalopathy, Leigh-like syndrome (MEGDHEL) syndrome is caused by biallelic variants in SERAC1. METHODS: This multicenter study addressed the course of disease for each organ system. Metabolic, neuroradiological, and genetic findings are reported. RESULTS: Sixty-seven individuals (39 previously unreported) from 59 families were included (age range = 5 days-33.4 years, median age = 9 years). A total of 41 different SERAC1 variants were identified, including 20 that have not been reported before. With the exception of 2 families with a milder phenotype, all affected individuals showed a strikingly homogeneous phenotype and time course. Severe, reversible neonatal liver dysfunction and hypoglycemia were seen in >40% of all cases. Starting at a median age of 6 months, muscular hypotonia (91%) was seen, followed by progressive spasticity (82%, median onset = 15 months) and dystonia (82%, 18 months). The majority of affected individuals never learned to walk (68%). Seventy-nine percent suffered hearing loss, 58% never learned to speak, and nearly all had significant intellectual disability (88%). Magnetic resonance imaging features were accordingly homogenous, with bilateral basal ganglia involvement (98%); the characteristic "putaminal eye" was seen in 53%. The urinary marker 3-methylglutaconic aciduria was present in virtually all patients (98%). Supportive treatment focused on spasticity and drooling, and was effective in the individuals treated; hearing aids or cochlear implants did not improve communication skills. INTERPRETATION: MEGDHEL syndrome is a progressive deafness-dystonia syndrome with frequent and reversible neonatal liver involvement and a strikingly homogenous course of disease. Ann Neurol 2017;82:1004-1015.


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Trastornos Sordoceguera/diagnóstico por imagen , Trastornos Sordoceguera/genética , Progresión de la Enfermedad , Distonía/diagnóstico por imagen , Distonía/genética , Discapacidad Intelectual/diagnóstico por imagen , Discapacidad Intelectual/genética , Mutación/genética , Atrofia Óptica/diagnóstico por imagen , Atrofia Óptica/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Niño , Preescolar , Estudios de Cohortes , Trastornos Sordoceguera/terapia , Distonía/terapia , Femenino , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/terapia , Masculino , Atrofia Óptica/terapia , Adulto Joven
13.
Epilepsia ; 59(8): 1595-1602, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29920680

RESUMEN

OBJECTIVE: Epilepsy is common in individuals with mutations in POLG, the gene encoding the catalytic subunit of the mitochondrial DNA polymerase gamma. Early recognition and aggressive seizure management are crucial for patient survival. Disruption of the blood-brain barrier (BBB) is implicated in various neurological disorders including epilepsy. The aim of this study was to assess whether POLG-related disease is associated with BBB dysfunction and what clinical implications this has for patients. METHODS: Our retrospective study used data from 83 patients with pathogenic POLG mutations from 4 countries--Norway, Sweden, Finland, and the United Kingdom. Data were collected using a structured questionnaire. We used the presence of raised cerebrospinal fluid (CSF) protein and a raised CSF/serum ratio of albumin (Q-alb) to evaluate the integrity of the blood-CSF barrier. RESULTS: Raised CSF protein was found in 70% of patients (n = 58/83) and appeared to be associated with the most severe phenotypes. In those in whom it was measured, the Q-alb ratio was markedly elevated (n = 18). The majority of those with epilepsy (n = 50/66, 76%) had raised CSF protein, and this preceded seizure debut in 75% (n = 15/20). The median survival time from symptom onset for those with raised CSF protein was decreased (13 months) compared to those with normal CSF protein (32 months). SIGNIFICANCE: Our results indicate that there is disruption of the BBB in POLG-related disease, as evidenced by a raised CSF protein and Q-alb ratio. We also find that raised CSF protein is a common finding in patients with POLG disease. Our data suggest that the presence of BBB dysfunction predicts a poorer outcome, and elevated CSF protein may therefore be an additional biomarker both for early diagnosis and to identify those at high risk of developing epilepsy.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Proteínas del Líquido Cefalorraquídeo/metabolismo , ADN Polimerasa gamma/genética , Epilepsia , Mutación/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Epilepsia/líquido cefalorraquídeo , Epilepsia/diagnóstico , Epilepsia/genética , Femenino , Humanos , Lactante , Recién Nacido , Cooperación Internacional , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
14.
Acta Paediatr ; 107(11): 1995-2003, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29683519

RESUMEN

AIM: This prospective study assessed the long-term ocular and visual outcomes of children with mucopolysaccharidoses type I Hurler syndrome (MPS IH) who were treated with haematopoietic stem cell transplants (HSCT). METHODS: Clinical ophthalmological assessments were performed on eight patients at the St Erik Eye Hospital, Huddinge, Stockholm, Sweden, from 2001-2018: The median age at diagnosis and HSCT were 12.2 (range 5.0-16.4) and 16.7 (8.0-20.4) months. The last eye examination was at a median of 13.4 (6.3-19.0) years and follow-up lasted a median of 12.0 (5.0-17.4) years. RESULTS: Poor visual acuity, poor night vision and, or, photophobia were reported by six children. The best corrected visual acuity at the last visit was a median of 0.4 and 0.5 in the right and left eye and had declined significantly in two patients. Corneal opacities had increased despite HSCT in five patients. High hyperopia, at a median of +6 Dioptres, occurred in all patients and stiff corneas in all four patients that were measured. The patients' corrected intraocular pressures were normal. Retinal degeneration was identified in two patients. CONCLUSION: Despite HSCT, the long-term follow-up of patients with MPS IH showed reduced visual acuity due to corneal opacities or retinal degeneration.


Asunto(s)
Córnea/fisiopatología , Trasplante de Células Madre Hematopoyéticas , Mucopolisacaridosis/complicaciones , Retina/fisiopatología , Trastornos de la Visión/etiología , Adolescente , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , Mucopolisacaridosis/fisiopatología , Mucopolisacaridosis/terapia , Estudios Prospectivos , Refracción Ocular , Estrabismo , Agudeza Visual , Adulto Joven
15.
Hum Mol Genet ; 24(23): 6580-7, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26374844

RESUMEN

Muscle weakness and exercise intolerance are hallmark symptoms in mitochondrial disorders. Little is known about the mechanisms leading to impaired skeletal muscle function and ultimately muscle weakness in these patients. In a mouse model of lethal mitochondrial myopathy, the muscle-specific Tfam knock-out (KO) mouse, we previously demonstrated an excessive mitochondrial Ca(2+) uptake in isolated muscle fibers that could be inhibited by the cyclophilin D (CypD) inhibitor, cyclosporine A (CsA). Here we show that the Tfam KO mice have increased CypD levels, and we demonstrate that this increase is a common feature in patients with mitochondrial myopathy. We tested the effect of CsA treatment on Tfam KO mice during the transition from a mild to terminal myopathy. CsA treatment counteracted the development of muscle weakness and improved muscle fiber Ca(2+) handling. Importantly, CsA treatment prolonged the lifespan of these muscle-specific Tfam KO mice. These results demonstrate that CsA treatment is an efficient therapeutic strategy to slow the development of severe mitochondrial myopathy.


Asunto(s)
Ciclofilinas/antagonistas & inhibidores , Ciclosporina/uso terapéutico , Mitocondrias/metabolismo , Miopatías Mitocondriales/tratamiento farmacológico , Músculo Esquelético/metabolismo , Animales , Calcio/metabolismo , Peptidil-Prolil Isomerasa F , Ciclofilinas/efectos de los fármacos , Ciclofilinas/genética , ADN Mitocondrial , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Miopatías Mitocondriales/genética , Miopatías Mitocondriales/metabolismo , Músculo Esquelético/efectos de los fármacos , Mutación
16.
J Inherit Metab Dis ; 39(2): 243-52, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26475597

RESUMEN

BACKGROUND: The encephalomyopathic mtDNA depletion syndrome with methylmalonic aciduria is associated with deficiency of succinate-CoA ligase, caused by mutations in SUCLA2 or SUCLG1. We report here 25 new patients with succinate-CoA ligase deficiency, and review the clinical and molecular findings in these and 46 previously reported patients. PATIENTS AND RESULTS: Of the 71 patients, 50 had SUCLA2 mutations and 21 had SUCLG1 mutations. In the newly-reported 20 SUCLA2 patients we found 16 different mutations, of which nine were novel: two large gene deletions, a 1 bp duplication, two 1 bp deletions, a 3 bp insertion, a nonsense mutation and two missense mutations. In the newly-reported SUCLG1 patients, five missense mutations were identified, of which two were novel. The median onset of symptoms was two months for patients with SUCLA2 mutations and at birth for SUCLG1 patients. Median survival was 20 years for SUCLA2 and 20 months for SUCLG1. Notable clinical differences between the two groups were hepatopathy, found in 38% of SUCLG1 cases but not in SUCLA2 cases, and hypertrophic cardiomyopathy which was not reported in SUCLA2 patients, but documented in 14% of cases with SUCLG1 mutations. Long survival, to age 20 years or older, was reported in 12% of SUCLA2 and in 10% of SUCLG1 patients. The most frequent abnormality on neuroimaging was basal ganglia involvement, found in 69% of SUCLA2 and 80% of SUCLG1 patients. Analysis of respiratory chain enzyme activities in muscle generally showed a combined deficiency of complexes I and IV, but normal histological and biochemical findings in muscle did not preclude a diagnosis of succinate-CoA ligase deficiency. In five patients, the urinary excretion of methylmalonic acid was only marginally elevated, whereas elevated plasma methylmalonic acid was consistently found. CONCLUSIONS: To our knowledge, this is the largest study of patients with SUCLA2 and SUCLG1 deficiency. The most important findings were a significantly longer survival in patients with SUCLA2 mutations compared to SUCLG1 mutations and a trend towards longer survival in patients with missense mutations compared to loss-of-function mutations. Hypertrophic cardiomyopathy and liver involvement was exclusively found in patients with SUCLG1 mutations, whereas epilepsy was much more frequent in patients with SUCLA2 mutations compared to patients with SUCLG1 mutations. The mutation analysis revealed a number of novel mutations, including a homozygous deletion of the entire SUCLA2 gene, and we found evidence of two founder mutations in the Scandinavian population, in addition to the known SUCLA2 founder mutation in the Faroe Islands.


Asunto(s)
Codón sin Sentido/genética , Enfermedades Mitocondriales/genética , Mutación Missense/genética , Succinato-CoA Ligasas/genética , Adolescente , Adulto , Errores Innatos del Metabolismo de los Aminoácidos/genética , Secuencia de Aminoácidos , Niño , Preescolar , Análisis Mutacional de ADN/métodos , ADN Mitocondrial/genética , Femenino , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Ácido Metilmalónico/metabolismo , Encefalomiopatías Mitocondriales/genética , Fenotipo , Adulto Joven
17.
J Med Genet ; 52(11): 779-83, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26084283

RESUMEN

BACKGROUND: Coenzyme Q is an essential mitochondrial electron carrier, redox cofactor and a potent antioxidant in the majority of cellular membranes. Coenzyme Q deficiency has been associated with a range of metabolic diseases, as well as with some drug treatments and ageing. METHODS: We used whole exome sequencing (WES) to investigate patients with inherited metabolic diseases and applied a novel ultra-pressure liquid chromatography-mass spectrometry approach to measure coenzyme Q in patient samples. RESULTS: We identified a homozygous missense mutation in the COQ7 gene in a patient with complex mitochondrial deficiency, resulting in severely reduced coenzyme Q levels We demonstrate that the coenzyme Q analogue 2,4-dihydroxybensoic acid (2,4DHB) was able to specifically bypass the COQ7 deficiency, increase cellular coenzyme Q levels and rescue the biochemical defect in patient fibroblasts. CONCLUSION: We report the first patient with primary coenzyme Q deficiency due to a homozygous COQ7 mutation and a potentially beneficial treatment using 2,4DHB.


Asunto(s)
Ataxia/genética , Hidroxibenzoatos/uso terapéutico , Enfermedades Mitocondriales/genética , Debilidad Muscular/genética , Mutación Missense , Ubiquinona/deficiencia , Secuencia de Aminoácidos , Ataxia/diagnóstico , Ataxia/tratamiento farmacológico , Niño , Preescolar , Cromatografía Liquida , Análisis Mutacional de ADN , Exoma , Homocigoto , Humanos , Recién Nacido , Masculino , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/tratamiento farmacológico , Datos de Secuencia Molecular , Debilidad Muscular/diagnóstico , Debilidad Muscular/tratamiento farmacológico , Alineación de Secuencia , Espectrometría de Masas en Tándem , Ubiquinona/genética
18.
Neuropediatrics ; 46(2): 98-103, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25642805

RESUMEN

Pediatric movement disorders are still a diagnostic challenge, as many patients remain without a (genetic) diagnosis. Magnetic resonance imaging (MRI) pattern recognition can lead to the diagnosis. MEGDEL syndrome (3-MethylGlutaconic aciduria, Deafness, Encephalopathy, Leigh-like syndrome MIM #614739) is a clinically and biochemically highly distinctive dystonia deafness syndrome accompanied by 3-methylglutaconic aciduria, severe developmental delay, and progressive spasticity. Mutations are found in SERAC1, encoding a phosphatidylglycerol remodeling enzyme essential for both mitochondrial function and intracellular cholesterol trafficking. Based on the homogenous phenotype, we hypothesized an accordingly characteristic MRI pattern. A total of 43 complete MRI studies of 30 patients were systematically reevaluated. All patients presented a distinctive brain MRI pattern with five characteristic disease stages affecting the basal ganglia, especially the putamen. In stage 1, T2 signal changes of the pallidum are present. In stage 2, swelling of the putamen and caudate nucleus is seen. The dorsal putamen contains an "eye" that shows no signal alteration and (thus) seems to be spared during this stage of the disease. It later increases, reflecting progressive putaminal involvement. This "eye" was found in all patients with MEGDEL syndrome during a specific age range, and has not been reported in other disorders, making it pathognomonic for MEDGEL and allowing diagnosis based on MRI findings.


Asunto(s)
Ganglios Basales/patología , Sordera/patología , Trastornos Distónicos/patología , Enfermedades Mitocondriales/patología , Preescolar , Sordera/complicaciones , Sordera/genética , Progresión de la Enfermedad , Trastornos Distónicos/complicaciones , Trastornos Distónicos/genética , Humanos , Lactante , Imagen por Resonancia Magnética , Enfermedades Mitocondriales/complicaciones , Putamen/patología , Síndrome
19.
BMC Genomics ; 15: 1090, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25495354

RESUMEN

BACKGROUND: Massively parallel DNA sequencing (MPS) has the potential to revolutionize diagnostics, in particular for monogenic disorders. Inborn errors of metabolism (IEM) constitute a large group of monogenic disorders with highly variable clinical presentation, often with acute, nonspecific initial symptoms. In many cases irreversible damage can be reduced by initiation of specific treatment, provided that a correct molecular diagnosis can be rapidly obtained. MPS thus has the potential to significantly improve both diagnostics and outcome for affected patients in this highly specialized area of medicine. RESULTS: We have developed a conceptually novel approach for acute MPS, by analysing pulsed whole genome sequence data in real time, using automated analysis combined with data reduction and parallelization. We applied this novel methodology to an in-house developed customized work flow enabling clinical-grade analysis of all IEM with a known genetic basis, represented by a database containing 474 disease genes which is continuously updated. As proof-of-concept, two patients were retrospectively analysed in whom diagnostics had previously been performed by conventional methods. The correct disease-causing mutations were identified and presented to the clinical team after 15 and 18 hours from start of sequencing, respectively. With this information available, correct treatment would have been possible significantly sooner, likely improving outcome. CONCLUSIONS: We have adapted MPS to fit into the dynamic, multidisciplinary work-flow of acute metabolic medicine. As the extent of irreversible damage in patients with IEM often correlates with timing and accuracy of management in early, critical disease stages, our novel methodology is predicted to improve patient outcome. All procedures have been designed such that they can be implemented in any technical setting and to any genetic disease area. The strategy conforms to international guidelines for clinical MPS, as only validated disease genes are investigated and as clinical specialists take responsibility for translation of results. As follow-up in patients without any known IEM, filters can be lifted and the full genome investigated, after genetic counselling and informed consent.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Errores Innatos del Metabolismo/diagnóstico , Biología Computacional , Bases de Datos Genéticas , Genoma Humano , Humanos , Errores Innatos del Metabolismo/genética , Piruvato Deshidrogenasa (Lipoamida)/genética , Análisis de Secuencia de ADN
20.
J Neurol ; 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38822839

RESUMEN

We aimed to provide a detailed phenotypic description of status epilepticus (SE) in a large cohort of patients with POLG disease and identify prognostic biomarkers to improve the management of this life-threatening condition. In a multinational, retrospective study with data on patients with POLG disease from seven European countries, we identified those who had SE. The age of SE onset, accompanying clinical, laboratory, imaging and genetic findings were analysed. One hundred and ninety-five patients with genetically confirmed POLG disease were recruited, of whom 67% (130/194) had epilepsy. SE was identified in 77% (97/126), with a median age of SE onset of 7 years. SE was the presenting symptom of the disease in 43% (40/93) of those with SE, while 57% (53/93) developed SE during the disease course. Convulsive SE was reported in 97% (91/94) followed by epilepsia partialis continua in 67% (56/84). Liver impairment 78% (74/95), ataxia 69% (60/87), stroke-like episodes 57% (50/88), were the major comorbidities. In the majority (66%; 57/86) with SE this became refractory or super-refractory. The presence of seizures was associated with significantly higher mortality compared to those without (P ≤ 0.001). The median time from SE debut to death was 5 months. SE is a major clinical feature of POLG disease in early and juvenile to adult-onset disease and can be the presenting feature or arise as part of a multisystem disease. It is associated with high morbidity and mortality, with the majority of patients with SE going on to develop refractory or super-refractory SE.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA