Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Cerebellum ; 22(6): 1182-1191, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36269527

RESUMEN

Assessment of ataxic movements is usually based on clinical judgment. Technical devices can be employed in the quantification of ataxic movements in addition to clinical evaluation. The effect of maximal speed in upper limb movements in ataxia patients has not been quantified. The aim was to quantify upper limb movements in patients with hereditary or idiopathic ataxia and to find features of movement that are characteristic for ataxia. We examined 19 patients with degenerative ataxia and 21 healthy controls. An ad hoc system comprising a touch screen, an accelerometer, and a gyroscope was used to measure speed, angular acceleration, consistency, and accuracy of upper limb movements. The movements were quantified during finger-to-nose test that the patients were asked to perform at their own pace and as fast as possible. Disease severity was estimated by using the Scale for the Assessment and Rating of Ataxia (SARA). The mean SARA score of the patients was 13.5. Compared to the controls the performance of the patients was slow (p < 0.001) and arrhythmic (p < 0.001), but end-point accuracy on the touch screen was intact. The SARA score correlated with the standard deviation of amplitude of angular acceleration in Z-axis (F(1,17) = 15.00, p < 0.001 with R2 = 0.47). Upper limb movements of the patients with degenerative ataxia were slower and more arrhythmic than those in the controls. The patients retained spatial end-point accuracy.


Asunto(s)
Ataxia Cerebelosa , Humanos , Ataxia/diagnóstico , Extremidad Superior , Movimiento , Dedos
3.
Eur J Neurol ; 30(5): 1256-1261, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36705320

RESUMEN

BACKGROUND AND PURPOSE: The biallelic repeat expansion (AAGGG)exp in the replication factor C subunit 1 gene (RFC1) is a frequent cause of cerebellar ataxia, neuropathy and vestibular areflexia syndrome (CANVAS) as well as late-onset ataxia. The clinical spectrum of RFC1 disease has expanded since the first identification of biallelic (AAGGG)exp and includes now various nonclassical phenotypes. Biallelic (AAGGG)exp in RFC1 in patients with clinically confirmed Parkinson's disease (PD) has recently been found. METHODS: A nationwide cohort of 273 Finnish patients with early-onset PD was examined for the biallelic intronic expansion in RFC1. The expansion (AAGGG)exp was first screened using extra long polymerase chain reactions (Extra Large-PCRs) and flanking multiplex PCR. The presence of biallelic (AAGGG)exp was then confirmed by repeat-primed PCR and, finally, the repeat length was determined by long-read sequencing. RESULTS: Three patients were found with the biallelic (AAGGG)exp in RFC1 giving a frequency of 1.10% (0.23%-3.18%; 95% confidence interval). The three patients fulfilled the diagnostic criteria of PD, none of them had ataxia or neuropathy, and only one patient had a mild vestibular dysfunction. The age at onset of PD symptoms was 40-48 years and their disease course had been unremarkable apart from the early onset. CONCLUSIONS: Our results suggest that (AAGGG)exp in RFC1 is a rare cause of early-onset PD. Other populations should be examined in order to determine whether our findings are specific to the Finnish population.


Asunto(s)
Ataxia Cerebelosa , Enfermedad de Parkinson , Enfermedades del Sistema Nervioso Periférico , Humanos , Ataxia , Ataxia Cerebelosa/genética , Enfermedad de Parkinson/genética , Fenotipo
4.
Ann Neurol ; 90(1): 35-42, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33901317

RESUMEN

OBJECTIVE: Parkinson's disease (PD) is a complex neurodegenerative disorder. Men are on average ~ 1.5 times more likely to develop PD compared to women with European ancestry. Over the years, genomewide association studies (GWAS) have identified numerous genetic risk factors for PD, however, it is unclear whether genetics contribute to disease etiology in a sex-specific manner. METHODS: In an effort to study sex-specific genetic factors associated with PD, we explored 2 large genetic datasets from the International Parkinson's Disease Genomics Consortium and the UK Biobank consisting of 13,020 male PD cases, 7,936 paternal proxy cases, 89,660 male controls, 7,947 female PD cases, 5,473 maternal proxy cases, and 90,662 female controls. We performed GWAS meta-analyses to identify distinct patterns of genetic risk contributing to disease in male versus female PD cases. RESULTS: In total, 19 genomewide significant regions were identified and no sex-specific effects were observed. A high genetic correlation between the male and female PD GWAS were identified (rg = 0.877) and heritability estimates were identical between male and female PD cases (~ 20%). INTERPRETATION: We did not detect any significant genetic differences between male or female PD cases. Our study does not support the notion that common genetic variation on the autosomes could explain the difference in prevalence of PD between males and females cases at least when considering the current sample size under study. Further studies are warranted to investigate the genetic architecture of PD explained by X and Y chromosomes and further evaluate environmental effects that could potentially contribute to PD etiology in male versus female patients. ANN NEUROL 2021;90:41-48.


Asunto(s)
Predisposición Genética a la Enfermedad , Genotipo , Enfermedad de Parkinson/genética , Caracteres Sexuales , Anciano , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad
5.
BMC Neurol ; 22(1): 487, 2022 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36522697

RESUMEN

BACKGROUND: Leber's hereditary optic neuropathy (LHON) is a common form of mitochondrial disease. The typical clinical presentation of LHON is subacute, painless loss of vision resulting from bilateral optic nerve atrophy. Moreover, extra-ocular manifestations such as cardiac conduction abnormalities and neurological manifestations such as multiple sclerosis (MS) like disease or parkinsonism are encountered in some patients. Abnormal findings in spinal cord MR imaging or in the cerebrospinal fluid (CSF) have been observed in previous cases of LHON-associated myelopathy. CASE PRESENTATION: We report a male patient with LHON who developed symptoms of myelopathy including gait unsteadiness, enhanced deep tendon reflexes and sensory loss of the lower extremities. Imaging of the brain and spinal cord, CSF analysis, as well as neurography and electromyography did not disclose any abnormalities. The somatosensory evoked potential (SEP) findings were suggestive of dorsal column dysfunction. CONCLUSIONS: The patient case demonstrates that myelopathy associated with LHON can present without abnormal findings in central nervous system MR imaging or in the CSF, and without evidence suggestive of multiple sclerosis or MS-like disease. The dorsal column seems to be particularly vulnerable to myelopathy changes in LHON. Evoked potential investigations may assist in confirming the diagnosis, when clinical features are in line with myelopathy but findings in CSF analysis and central nervous system imaging are normal.


Asunto(s)
Esclerosis Múltiple , Atrofia Óptica Hereditaria de Leber , Enfermedades de la Médula Espinal , Humanos , Masculino , Atrofia Óptica Hereditaria de Leber/diagnóstico , Atrofia Óptica Hereditaria de Leber/diagnóstico por imagen , Imagen por Resonancia Magnética , Enfermedades de la Médula Espinal/diagnóstico por imagen , Esclerosis Múltiple/complicaciones , ADN Mitocondrial
6.
BMC Genomics ; 22(1): 75, 2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33482721

RESUMEN

BACKGROUND: We have previously suggested that some of the mutations defining mitochondrial DNA (mtDNA) haplogroups J and K produce an uncoupling effect on oxidative phosphorylation and thus are detrimental for elite endurance performance. Here, the association between haplogroups J and K and physical performance was determined in a population-based cohort of 1036 Finnish military conscripts. RESULTS: Following a standard-dose training period, excellence in endurance performance was less frequent among subjects with haplogroups J or K than among subjects with non-JK haplogroups (p = 0.041), and this finding was more apparent among the best-performing subjects (p < 0.001). CONCLUSIONS: These results suggest that mtDNA haplogroups are one of the genetic determinants explaining individual variability in the adaptive response to endurance training, and mtDNA haplogroups J and K are markers of low-responders in exercise training.


Asunto(s)
Personal Militar , ADN Mitocondrial/genética , Ejercicio Físico , Finlandia , Haplotipos , Humanos
7.
BMC Neurol ; 21(1): 382, 2021 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-34600502

RESUMEN

BACKGROUND: The genetics of cerebellar ataxia is complex. Hundreds of causative genes have been identified, but only a few cause more than single cases. The spectrum of ataxia-causing genes differs considerably between populations. The aim of the study was to investigate the molecular epidemiology of ataxia in the Finnish population. PATIENTS AND METHODS: All patients in hospital database were reviewed for the diagnosis of unspecified ataxia. Acquired ataxias and nongenetic ataxias such as those related to infection, trauma or stroke were excluded. Sixty patients with sporadic ataxia with unknown etiology and 36 patients with familial ataxia of unknown etiology were recruited in the study. Repeat expansions in the SCA genes (ATXN1, 2, 3, 7, 8/OS, CACNA1A, TBP), FXN, and RFC1 were determined. Point mutations in POLG, SPG7 and in mitochondrial DNA (mtDNA) were investigated. In addition, DNA from 8 patients was exome sequenced. RESULTS: A genetic cause of ataxia was found in 33 patients (34.4%). Seven patients had a dominantly inherited repeat expansion in ATXN8/OS. Ten patients had mitochondrial ataxia resulting from mutations in nuclear mitochondrial genes POLG or RARS2, or from a point mutation m.8561C > G or a single deletion in mtDNA. Interestingly, five patients were biallelic for the recently identified pathogenic repeat expansion in RFC1. All the five patients presented with the phenotype of cerebellar ataxia, neuropathy, and vestibular areflexia (CANVAS). Moreover, screening of 54 patients with Charcot-Marie-Tooth neuropathy revealed four additional patients with biallelic repeat expansion in RFC1, but none of them had cerebellar symptoms. CONCLUSIONS: Expansion in ATXN8/OS results in the majority of dominant ataxias in Finland, while mutations in RFC1 and POLG are the most common cause of recessive ataxias. Our results suggest that analysis of RFC1 should be included in the routine diagnostics of idiopathic ataxia and Charcot-Marie-Tooth polyneuropathy.


Asunto(s)
Ataxia Cerebelosa , Degeneraciones Espinocerebelosas , Finlandia/epidemiología , Humanos , Epidemiología Molecular , Proteína de Replicación C/genética
8.
Brain ; 143(1): 234-248, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31755958

RESUMEN

Parkinson's disease is a genetically complex disorder. Multiple genes have been shown to contribute to the risk of Parkinson's disease, and currently 90 independent risk variants have been identified by genome-wide association studies. Thus far, a number of genes (including SNCA, LRRK2, and GBA) have been shown to contain variability across a spectrum of frequency and effect, from rare, highly penetrant variants to common risk alleles with small effect sizes. Variants in GBA, encoding the enzyme glucocerebrosidase, are associated with Lewy body diseases such as Parkinson's disease and Lewy body dementia. These variants, which reduce or abolish enzymatic activity, confer a spectrum of disease risk, from 1.4- to >10-fold. An outstanding question in the field is what other genetic factors that influence GBA-associated risk for disease, and whether these overlap with known Parkinson's disease risk variants. Using multiple, large case-control datasets, totalling 217 165 individuals (22 757 Parkinson's disease cases, 13 431 Parkinson's disease proxy cases, 622 Lewy body dementia cases and 180 355 controls), we identified 1691 Parkinson's disease cases, 81 Lewy body dementia cases, 711 proxy cases and 7624 controls with a GBA variant (p.E326K, p.T369M or p.N370S). We performed a genome-wide association study and analysed the most recent Parkinson's disease-associated genetic risk score to detect genetic influences on GBA risk and age at onset. We attempted to replicate our findings in two independent datasets, including the personal genetics company 23andMe, Inc. and whole-genome sequencing data. Our analysis showed that the overall Parkinson's disease genetic risk score modifies risk for disease and decreases age at onset in carriers of GBA variants. Notably, this effect was consistent across all tested GBA risk variants. Dissecting this signal demonstrated that variants in close proximity to SNCA and CTSB (encoding cathepsin B) are the most significant contributors. Risk variants in the CTSB locus were identified to decrease mRNA expression of CTSB. Additional analyses suggest a possible genetic interaction between GBA and CTSB and GBA p.N370S induced pluripotent cell-derived neurons were shown to have decreased cathepsin B expression compared to controls. These data provide a genetic basis for modification of GBA-associated Parkinson's disease risk and age at onset, although the total contribution of common genetics variants is not large. We further demonstrate that common variability at genes implicated in lysosomal function exerts the largest effect on GBA associated risk for disease. Further, these results have implications for selection of GBA carriers for therapeutic interventions.


Asunto(s)
Catepsina B/genética , Glucosilceramidasa/genética , Enfermedad por Cuerpos de Lewy/genética , Enfermedad de Parkinson/genética , Penetrancia , alfa-Sinucleína/genética , Edad de Inicio , Estudios de Casos y Controles , Catepsina B/metabolismo , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genotipo , Glucosilceramidasa/metabolismo , Humanos , Células Madre Pluripotentes Inducidas , Enfermedad por Cuerpos de Lewy/metabolismo , Neurogénesis/genética , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Polimorfismo de Nucleótido Simple , ARN Mensajero/metabolismo , Factores de Riesgo , Secuenciación Completa del Genoma , alfa-Sinucleína/metabolismo
9.
Int J Legal Med ; 134(1): 39-44, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31152278

RESUMEN

Cardiomyopathy and cardiac conduction defects are common manifestations of mitochondrial disease. Previous studies suggest that clinically asymptomatic individuals harbouring pathogenic mitochondrial DNA (mtDNA) mutations in the cardiac muscle may have sudden cardiac death (SCD) as the first manifestation of mitochondrial disease. We investigated the contribution of pathogenic mtDNA point mutations and mtDNA haplogroups in cardiac muscle in a cohort of 280 Finnish subjects that had died from non-ischaemic SCD with the median age of death at 59 years and in 537 population controls. We did not find any common or novel pathogenic mutations, but the frequency of haplogroup H1 was higher in the SCD subjects than that in 537 population controls (odds ratio: 1.76, confidence interval 95%: 1.02-3.04). We conclude that, at the population level, pathogenic point mutations in mtDNA do not contribute to non-ischaemic SCD, but natural variation may modify the risk.


Asunto(s)
ADN Mitocondrial/genética , Muerte Súbita Cardíaca/epidemiología , Haplotipos , Mutación Puntual , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Estudios de Cohortes , Finlandia/epidemiología , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Lactante , Persona de Mediana Edad , Factores de Riesgo , Adulto Joven
10.
BMC Genomics ; 20(1): 784, 2019 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-31664900

RESUMEN

BACKGROUND: We have previously reported on paucity of mitochondrial DNA (mtDNA) haplogroups J and K among Finnish endurance athletes. Here we aimed to further explore differences in mtDNA variants between elite endurance and sprint athletes. For this purpose, we determined the rate of functional variants and the mutational load in mtDNA of Finnish athletes (n = 141) and controls (n = 77) and determined the sequence variation in haplogroups. RESULTS: The distribution of rare and common functional variants differed between endurance athletes, sprint athletes and the controls (p = 0.04) so that rare variants occurred at a higher frequency among endurance athletes. Furthermore, the ratio between rare and common functional variants in haplogroups J and K was 0.42 of that in the remaining haplogroups (p = 0.0005). The subjects with haplogroup J and K also showed a higher mean level of nonsynonymous mutational load attributed to common variants than subjects with the other haplogroups. Interestingly, two of the rare variants detected in the sprint athletes were the disease-causing mutations m.3243A > G in MT-TL1 and m.1555A > G in MT-RNR1. CONCLUSIONS: We propose that endurance athletes harbor an excess of rare mtDNA variants that may be beneficial for oxidative phosphorylation, while sprint athletes may tolerate deleterious mtDNA variants that have detrimental effect on oxidative phosphorylation system. Some of the nonsynonymous mutations defining haplogroup J and K may produce an uncoupling effect on oxidative phosphorylation thus favoring sprint rather than endurance performance.


Asunto(s)
Atletas , ADN Mitocondrial/química , Mutación , Finlandia , Fosforilación Oxidativa
11.
Mov Disord ; 34(6): 866-875, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30957308

RESUMEN

BACKGROUND: Increasing evidence supports an extensive and complex genetic contribution to PD. Previous genome-wide association studies (GWAS) have shed light on the genetic basis of risk for this disease. However, the genetic determinants of PD age at onset are largely unknown. OBJECTIVES: To identify the genetic determinants of PD age at onset. METHODS: Using genetic data of 28,568 PD cases, we performed a genome-wide association study based on PD age at onset. RESULTS: We estimated that the heritability of PD age at onset attributed to common genetic variation was ∼0.11, lower than the overall heritability of risk for PD (∼0.27), likely, in part, because of the subjective nature of this measure. We found two genome-wide significant association signals, one at SNCA and the other a protein-coding variant in TMEM175, both of which are known PD risk loci and a Bonferroni-corrected significant effect at other known PD risk loci, GBA, INPP5F/BAG3, FAM47E/SCARB2, and MCCC1. Notably, SNCA, TMEM175, SCARB2, BAG3, and GBA have all been shown to be implicated in α-synuclein aggregation pathways. Remarkably, other well-established PD risk loci, such as GCH1 and MAPT, did not show a significant effect on age at onset of PD. CONCLUSIONS: Overall, we have performed the largest age at onset of PD genome-wide association studies to date, and our results show that not all PD risk loci influence age at onset with significant differences between risk alleles for age at onset. This provides a compelling picture, both within the context of functional characterization of disease-linked genetic variability and in defining differences between risk alleles for age at onset, or frank risk for disease. © 2019 International Parkinson and Movement Disorder Society.


Asunto(s)
Edad de Inicio , Sitios Genéticos , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Bases de Datos Genéticas , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Glucosilceramidasa/genética , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Adulto Joven
12.
Brain ; 141(1): 55-62, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29182774

RESUMEN

The m.1555A>G mtDNA variant causes maternally inherited deafness, but the reasons for the highly variable clinical penetrance are not known. Exome sequencing identified a heterozygous start loss mutation in SSBP1, encoding the single stranded binding protein 1 (SSBP1), segregating with hearing loss in a multi-generational family transmitting m.1555A>G, associated with mtDNA depletion and multiple deletions in skeletal muscle. The SSBP1 mutation reduced steady state SSBP1 levels leading to a perturbation of mtDNA metabolism, likely compounding the intra-mitochondrial translation defect due to m.1555A>G in a tissue-specific manner. This family demonstrates the importance of rare trans-acting genetic nuclear modifiers in the clinical expression of mtDNA disease.


Asunto(s)
Proteínas de Unión al ADN/genética , Salud de la Familia , Pérdida Auditiva/genética , Proteínas Mitocondriales/genética , Mutación/genética , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN , Complejo II de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Pérdida Auditiva/complicaciones , Heterocigoto , Humanos , Lactante , Masculino , Enfermedades Mitocondriales/complicaciones , Enfermedades Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Músculo Esquelético/ultraestructura , Adulto Joven
13.
Epilepsia ; 59(11): 2125-2136, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30255931

RESUMEN

OBJECTIVE: Previous studies have suggested that heterozygous variants p.Q1236H and p.E1143G in mitochondrial DNA polymerase gamma (POLG1) increase the risk for liver injury for patients on valproate (VPA) therapy. We assessed the prevalence of these common variants and seven other pathogenic mutations in POLG1 and determined the occurrence of VPA-induced hepatotoxicity (VHT) or pancreatic toxicity in a cohort of patients with epilepsy. METHODS: Patients with epilepsy (N = 367) were retrospectively identified from medical record files and screened for mutations in POLG1. Patients who had received VPA monotherapy and carried either of the two variants, p.Q1236H or p.E1143G, without other pathogenic mutations in POLG1 (n = 33, variant group) and patients without these variants (n = 28, nonvariant group) were included in the study. Clinical data on epilepsy, characteristics of VPA treatment, risk factors for VHT, laboratory data on liver and pancreas functions, and adverse effects were collected. RESULTS: A total of 122 patients had either the POLG1 p.Q1236H (n = 99) or p.E1143G (n = 24) variant in the heterozygous or homozygous state. Transient liver dysfunction was identified in three (n = 33, 9.1%) variant group patients and in one (n = 28, 3.6%) nonvariant group patient (P = 0.62). Mild to moderate elevations in liver enzymes were encountered in both groups. Furthermore, two patients on VPA polytherapy developed acute pancreatitis, and two pediatric patients with heterozygous p.Q1236H variants and mutations in IQSEC2 and GLDC, respectively, had elevated levels of VPA metabolites in urine, elevated plasma glycine, and/or increased acylglycine excretion. SIGNIFICANCE: POLG1 p.Q1236H and p.E1143G variants could not be identified as statistically significant risk factors for VHT or pancreatic toxicity. We suggest that VPA treatment could be suitable for patients who harbor these common variants in the absence of other pathogenic mutations in POLG1.


Asunto(s)
Anticonvulsivantes/efectos adversos , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , ADN Polimerasa gamma/genética , Epilepsia/genética , Mutación/genética , Enfermedades Pancreáticas/inducido químicamente , Ácido Valproico/efectos adversos , Adolescente , Adulto , Alanina Transaminasa/metabolismo , Aspartato Aminotransferasas/metabolismo , Niño , Estudios de Cohortes , Epilepsia/tratamiento farmacológico , Femenino , Humanos , Masculino , Estadísticas no Paramétricas , Adulto Joven , gamma-Glutamiltransferasa/metabolismo
14.
BMC Neurol ; 18(1): 149, 2018 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-30236074

RESUMEN

BACKGROUND: Only five patients have previously been reported to harbor mutations in the MT-TT gene encoding mitochondrial tRNA threonine. The m.15923A > G mutation has been found in three severely affected children. One of these patients died within days after birth and two had a phenotype of myoclonic epilepsy with ragged red fibers (MERRF) in early childhood. We have now found the mutation in an adult patient with mild myopathy. CASE PRESENTATION: The patient is a 64-year-old Finnish man, who developed bilateral ptosis, diplopia and exercise intolerance in his fifties. Family history was unremarkable. Muscle histology showed cytochrome c-oxidase (COX) negative and ragged red fibres. The m.15923A > G mutation heteroplasmy was 33% in the skeletal muscle and 2% in buccal epithelial cells. The mutation was undetectable in the blood. Single-fibre analysis was performed and COX-negative fibres had a substantially higher heteroplasmy of 92%, than the normal fibres in which it was 43%. CONCLUSIONS: We report the fourth patient with m. 15923A > G and with a remarkably milder phenotype than the previous three patients. Our findings and recent biochemical studies suggest that the mutation m.15923A > G is a definite disease-causing mutation. Our results also suggest that heteroplasmy of the m.15923A > G mutation correlates with the severity of the phenotype. This study expands the catalog of the phenotypes caused by mutations in mtDNA.


Asunto(s)
ADN Mitocondrial/genética , Enfermedades Musculares/genética , ARN de Transferencia/genética , Treonina/genética , Adulto , Edad de Inicio , Humanos , Masculino , Músculo Esquelético/patología , Enfermedades Musculares/patología , Mutación , Fenotipo
15.
BMC Med Genet ; 18(1): 14, 2017 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-28187756

RESUMEN

BACKGROUND: Mitochondrial diseases present with variable multi-organ symptoms. Common disease-causing mutations in mitochondrial DNA (mtDNA) are regularly screened in diagnostic work-up, but novel mutations may remain unnoticed. METHODS: Patients (N = 66) with a clinical suspicion of mitochondrial disease were screened for their mtDNA coding region using conformation sensitive gel electrophoresis and sequencing. Long-PCR was used to detect deletions followed by POLG1 sequencing in patients with multiple deletions. RESULTS: We discovered three novel mtDNA variants that included m.8743G > C, m.11322A > G and m.15933G > A. The novel MTTT variant m.15933G > A is suggested to be pathogenic. Analysis revealed also multiple mtDNA deletions in two patients and five nonsynonymous variants that were putatively pathogenic according to in-silico prediction algorithms. In addition, a rare haplogroup H associated m.7585_7586insT variant was discovered. CONCLUSIONS: Among patients with a suspected mitochondrial disease, a novel MTTT variant m.15933G > A was discovered and is suggested to be pathogenic. In addition, several putatively pathogenic nonsynonymous variants and rare variants were found. These findings highlight the importance of coding region mtDNA screening among patients with clinical features suggesting a mitochondrial disease, but who lack the common mitochondrial disease mutations.


Asunto(s)
ADN Mitocondrial/genética , Enfermedades Mitocondriales/genética , Secuencia de Bases , ADN Polimerasa gamma , ADN Mitocondrial/clasificación , ADN Mitocondrial/metabolismo , ADN Polimerasa Dirigida por ADN/genética , Electroforesis en Gel de Poliacrilamida , Femenino , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Mitocondriales/patología , Conformación de Ácido Nucleico , Sistemas de Lectura Abierta/genética , Filogenia , Eliminación de Secuencia
16.
Neuroepidemiology ; 49(1-2): 34-39, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28810241

RESUMEN

BACKGROUND: Charcot-Marie-Tooth (CMT) disease is the most common hereditary neuromuscular disorder with a population prevalence of 9.7-82.3/100,000. In this study, we have estimated the prevalence of CMT and its subtypes in Finland and examined the frequency of molecular etiologies. METHODS: A population-based survey included adult patients with peripheral neuropathy from the province of Northern Ostrobothnia, Finland. Secondary causes of peripheral polyneuropathy were excluded and patients with clinical and neurophysiological features pertinent with CMT were included. Molecular diagnostics was carried out when DNA was available. RESULTS: We found 107 subjects with CMT yielding a prevalence 34.6/100,000 in Northern Ostrobothnia. The heterozygous point mutation p.His123Arg in ganglioside induced differentiation associated protein 1 (GDAP1) was found in 31.5% and peripheral myelin protein 22 (PMP22) duplication in 16.9% of the affected. Point mutations in myelin protein zero, mitofusin 2, and gap junction protein beta 1 accounted for 6.7% of the cases. In addition, 18 persons had hereditary neuropathy with liability to pressure palsies and 15 of them carried the PMP22 deletion. CONCLUSIONS: The prevalence of CMT in Northern Ostrobothnia, Finland, seems to be slightly higher than those in previous studies in European populations. Founder mutation in the GDAP1 gene accounts for a large part of the genetically defined CMT2 in Finland.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/epidemiología , Enfermedad de Charcot-Marie-Tooth/genética , Femenino , Finlandia , Eliminación de Gen , Encuestas Epidemiológicas , Humanos , Masculino , Epidemiología Molecular , Proteínas de la Mielina/genética , Fenotipo , Mutación Puntual
17.
BMC Neurol ; 17(1): 96, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28521807

RESUMEN

BACKGROUND: Mitochondrial cytochrome c oxidase 2, MT-CO2, encodes one of the three subunits, which form the catalytic core of cytochrome c oxidase (COX), complex IV. Mutations in MT-CO2 are rare and the associated phenotypes are variable including nonsyndromic and syndromic forms of mitochondrial diseases. CASE PRESENTATION: We describe a 30-year-old man with cognitive decline, epilepsy, psychosis, exercise intolerance, sensorineural hearing impairment, retinitis pigmentosa, cataract and lactic acidosis. COX-deficient fibers and ragged red fibers were abundant in the muscle. Sequencing of mitochondrial DNA (mtDNA) revealed a novel frameshift mutation m.8156delG that was predicted to cause altered C-terminal amino acid sequence and to lead to truncation of the COX subunit 2. The deletion was heteroplasmic being present in 26% of the mtDNA in blood, 33% in buccal mucosa and 95% in muscle. Deletion heteroplasmy correlated with COX-deficiency in muscle histochemistry. The mother and the siblings of the proband did not harbor the deletion. CONCLUSIONS: The clinical features and muscle histology of the proband suggested a mitochondrial disorder. The m.8156delG deletion is a new addition to the short list of pathogenic mutations in the mtDNA-encoded subunits of COX. This case illustrates the importance of mtDNA sequence analysis in patients with an evident mitochondrial disorder.


Asunto(s)
ADN Mitocondrial/genética , Complejo IV de Transporte de Electrones/genética , Enfermedades Mitocondriales/genética , Adulto , Deficiencia de Citocromo-c Oxidasa , Complejo IV de Transporte de Electrones/metabolismo , Mutación del Sistema de Lectura , Humanos , Masculino , Mitocondrias/enzimología , Enfermedades Mitocondriales/patología , Músculos/patología , Mutación , Eliminación de Secuencia
18.
J Inherit Metab Dis ; 39(1): 67-74, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26112752

RESUMEN

Mitochondrial mutations impair glucose oxidation and increase glucose uptake in cell cultures and lead to cardiomyopathy in patients. Here we characterize cardiac glucose uptake in 14 patients with the m.3243A > G mutation in mitochondrial DNA. The 14 patients with m.3243A > G and 13 controls were similar in age, physical activity and body mass index. Ten patients had diabetes. Left ventricular glucose uptake per tissue mass (LVGU) was measured with 2-[(18) F]fluoro-2-deoxyglucose positron emission tomography during euglycemic hyperinsulinemia. Cardiac morphology and function were assessed with magnetic resonance imaging. We found that the LVGU was 25% lower in the patients than that in the controls (P = 0.029). LVGU was inversely correlated with mutation heteroplasmy, glycated haemoglobin and fasting lactate in patients. The seven patients with mutation heteroplasmy ≥ 49% had 44% lower LVGU than the seven patients with heteroplasmy < 49%. This difference remained significant after adjustment for concurrent free fatty acid concentration or glycated haemoglobin or glucose uptake in skeletal muscle or all (p < 0.048 [All]). Patients with m.3243A > G had a lower stroke volume and a higher heart rate than the controls, whereas cardiac output and work were similar. Myocardial glucose uptake is not increased but decreased with a threshold effect pattern in patients with the m.3243A > G mutation. The glucose hypometabolism adds to the impaired cardiac energetics and likely contributes to the progression of the mitochondrial cardiomyopathy.


Asunto(s)
ADN Mitocondrial/genética , Glucosa/metabolismo , Mitocondrias/genética , Mutación/genética , Miocardio/metabolismo , Glucemia/metabolismo , Estudios de Casos y Controles , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Epitelio/metabolismo , Femenino , Prueba de Tolerancia a la Glucosa/métodos , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/metabolismo
19.
BMC Neurol ; 16: 24, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26860086

RESUMEN

BACKGROUND: Seizures are common in juvenile Huntington's disease (HD), but considered to be rare in adult-onset HD. We studied the occurrence of epilepsy and seizures in a nationwide cohort of Finnish patients with adult-onset HD. METHODS: Patients with HD and their diagnoses of epilepsy or seizures were identified by a search into a nationwide registry. Cases were verified in a subsequent review of patient charts. RESULTS: Three out of 114 HD patients alive on prevalence date had been diagnosed with epilepsy giving a prevalence of 2.6% (95% CI, 0.6-7.5). In addition, one patient with a single unprovoked seizure, one patient with a medication-induced seizure and two patients with transient nonspecific attacks were identified. Epilepsy was not associated with clinical severity of HD and seizures were controlled with antiepileptic medications (AEDs). Generalized tonic-clonic seizures (GTCs) were the most common seizure type. CONCLUSIONS: Prevalence of epilepsy is similar in patients with adult-onset HD compared to general population. Seizures are easily controlled with AEDs.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/epidemiología , Enfermedad de Huntington/epidemiología , Adulto , Anciano , Anciano de 80 o más Años , Epilepsia/tratamiento farmacológico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sistema de Registros , Convulsiones/tratamiento farmacológico , Convulsiones/epidemiología , Adulto Joven
20.
Acta Neurochir (Wien) ; 158(8): 1487-90, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27272892

RESUMEN

BACKGROUND: Many features of Huntington's disease (HD) make these patients susceptible to subdural hematomas (SDH), but there are no previous reports on the epidemiology of SDH in this population. We investigated the incidence and risk factors of chronic SDH. METHODS: A national cohort of 192 Finnish patients with HD was investigated. Information was gathered from medical records and administrative registries. RESULTS: The incidence rate of SDH was 68.3/100,000 person-years among the 192 patients. Seven patients were identified with a chronic SDH at or after the diagnosis of HD. Their age was 58.5 ± 15.0 years (mean ± SD) at the time of diagnosis of HD and 60.9 ± 14.1 years at the time of diagnosis of SDH. The incidence rate of chronic SDH after the diagnosis of HD was 4.7/1000 person-years and by 8.3 years of follow-up the cumulative risk was 5.4 %. Review of the patient charts revealed only a few of the common risk factors for chronic SDH, but the rate of reoperations was high. CONCLUSIONS: The incidence of chronic SDH was higher in patients with HD than that in the general population. Incidence of chronic SDH began to increase at the time of expected motor onset of HD. Common risk factors of SDH were scarce aside from fall-related head injuries.


Asunto(s)
Hematoma Subdural Crónico/epidemiología , Enfermedad de Huntington/complicaciones , Anciano , Anciano de 80 o más Años , Femenino , Finlandia , Hematoma Subdural Crónico/etiología , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA