Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Brain ; 147(5): 1887-1898, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38193360

RESUMO

RFC1 disease, caused by biallelic repeat expansion in RFC1, is clinically heterogeneous in terms of age of onset, disease progression and phenotype. We investigated the role of the repeat size in influencing clinical variables in RFC1 disease. We also assessed the presence and role of meiotic and somatic instability of the repeat. In this study, we identified 553 patients carrying biallelic RFC1 expansions and measured the repeat expansion size in 392 cases. Pearson's coefficient was calculated to assess the correlation between the repeat size and age at disease onset. A Cox model with robust cluster standard errors was adopted to describe the effect of repeat size on age at disease onset, on age at onset of each individual symptoms, and on disease progression. A quasi-Poisson regression model was used to analyse the relationship between phenotype and repeat size. We performed multivariate linear regression to assess the association of the repeat size with the degree of cerebellar atrophy. Meiotic stability was assessed by Southern blotting on first-degree relatives of 27 probands. Finally, somatic instability was investigated by optical genome mapping on cerebellar and frontal cortex and unaffected peripheral tissue from four post-mortem cases. A larger repeat size of both smaller and larger allele was associated with an earlier age at neurological onset [smaller allele hazard ratio (HR) = 2.06, P < 0.001; larger allele HR = 1.53, P < 0.001] and with a higher hazard of developing disabling symptoms, such as dysarthria or dysphagia (smaller allele HR = 3.40, P < 0.001; larger allele HR = 1.71, P = 0.002) or loss of independent walking (smaller allele HR = 2.78, P < 0.001; larger allele HR = 1.60; P < 0.001) earlier in disease course. Patients with more complex phenotypes carried larger expansions [smaller allele: complex neuropathy rate ratio (RR) = 1.30, P = 0.003; cerebellar ataxia, neuropathy and vestibular areflexia syndrome (CANVAS) RR = 1.34, P < 0.001; larger allele: complex neuropathy RR = 1.33, P = 0.008; CANVAS RR = 1.31, P = 0.009]. Furthermore, larger repeat expansions in the smaller allele were associated with more pronounced cerebellar vermis atrophy (lobules I-V ß = -1.06, P < 0.001; lobules VI-VII ß = -0.34, P = 0.005). The repeat did not show significant instability during vertical transmission and across different tissues and brain regions. RFC1 repeat size, particularly of the smaller allele, is one of the determinants of variability in RFC1 disease and represents a key prognostic factor to predict disease onset, phenotype and severity. Assessing the repeat size is warranted as part of the diagnostic test for RFC1 expansion.


Assuntos
Idade de Início , Proteína de Replicação C , Humanos , Masculino , Feminino , Proteína de Replicação C/genética , Adulto , Expansão das Repetições de DNA/genética , Pessoa de Meia-Idade , Adulto Jovem , Adolescente , Criança , Fenótipo , Índice de Gravidade de Doença , Pré-Escolar , Progressão da Doença
2.
Neuromuscul Disord ; 34: 27-31, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38052666

RESUMO

Genetic variation at HNRNPA2B1 is associated with inclusion body myopathy, Paget's disease and paediatric onset oculopharyngeal muscular dystrophy. We present a pedigree where a mother and two daughters presented with adolescent to early-adulthood onset of symptoms reminiscent of oculopharyngeal muscular dystrophy or chronic progressive external ophthalmoplegia, with a later limb-girdle pattern of weakness. Creatine Kinase was ∼1000 U/L. Myoimaging identified fatty replacement of sartorius, adductors longus and magnus, biceps femoris, semitendinosus and gastrocnemii. Muscle biopsies showed a variation of fibre size, occasional rimmed vacuoles and increased internalised myonuclei. Cases were heterozygous for a frameshift variant at HNRNPA2B1, consistent with a dominant and fully-penetrant mode of inheritance. Genetic variation at HNRNPA2B1 should be considered in adults with an oculopharyngeal muscular dystrophy-like or chronic progressive external ophthalmoplegia-like myopathy where initial testing fails to identify a cause.


Assuntos
Doenças Musculares , Distrofia Muscular Oculofaríngea , Oftalmoplegia Externa Progressiva Crônica , Adolescente , Adulto , Criança , Humanos , Músculo Esquelético/patologia , Doenças Musculares/genética , Distrofia Muscular Oculofaríngea/diagnóstico , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/patologia , Oftalmoplegia Externa Progressiva Crônica/patologia , Linhagem , Fenótipo
3.
Nat Commun ; 13(1): 2306, 2022 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-35484142

RESUMO

Missense variants in RNA-binding proteins (RBPs) underlie a spectrum of disease phenotypes, including amyotrophic lateral sclerosis, frontotemporal dementia, and inclusion body myopathy. Here, we present ten independent families with a severe, progressive muscular dystrophy, reminiscent of oculopharyngeal muscular dystrophy (OPMD) but of much earlier onset, caused by heterozygous frameshift variants in the RBP hnRNPA2/B1. All disease-causing frameshift mutations abolish the native stop codon and extend the reading frame, creating novel transcripts that escape nonsense-mediated decay and are translated to produce hnRNPA2/B1 protein with the same neomorphic C-terminal sequence. In contrast to previously reported disease-causing missense variants in HNRNPA2B1, these frameshift variants do not increase the propensity of hnRNPA2 protein to fibrillize. Rather, the frameshift variants have reduced affinity for the nuclear import receptor karyopherin ß2, resulting in cytoplasmic accumulation of hnRNPA2 protein in cells and in animal models that recapitulate the human pathology. Thus, we expand the phenotypes associated with HNRNPA2B1 to include an early-onset form of OPMD caused by frameshift variants that alter its nucleocytoplasmic transport dynamics.


Assuntos
Esclerose Lateral Amiotrófica , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Distrofia Muscular Oculofaríngea , Esclerose Lateral Amiotrófica/genética , Animais , Mutação da Fase de Leitura , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Heterozigoto , Humanos , Distrofia Muscular Oculofaríngea/genética
4.
Neuromuscul Disord ; 31(3): 249-252, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33546848

RESUMO

A 63 year old male presented with a 20 year history of facial weakness and several years of nasal regurgitation and dysphonia. Examination revealed bilateral facial weakness with nasal speech. Serum creatine kinase was 918 U/L. Neurophysiological studies suggested a myopathy and biopsy of the left vastus lateralis showed serpentine basophilic inclusions in the sarcoplasm and strong oxidative enzyme activity suggesting mitochondria accumulation. The muscle MRI showed selective fatty replacement within semitendinosus, gastrocnemius and soleus indicative of a desminopathy. A heterozygous missense variant c.17C>G (p.Ser6Trp) was identified within DES, predicted to be pathogenic in silico and previously described in a family with distal limb weakness. There are no previous case reports of desminopathy presenting with facial weakness, to our knowledge. Diagnosis was suggested following myoimaging of clinically unaffected muscles. Our study highlights the importance of muscle MRI in the diagnostic evaluation of muscle disease and further expands the known phenotypic heterogeneity of desminopathies.


Assuntos
Cardiomiopatias/diagnóstico por imagem , Músculos Faciais/diagnóstico por imagem , Extremidade Inferior/diagnóstico por imagem , Imageamento por Ressonância Magnética , Debilidade Muscular/diagnóstico por imagem , Distrofias Musculares/diagnóstico por imagem , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto
5.
Pract Neurol ; 2020 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-32980811

RESUMO

Mitochondrial neurogastrointestinal encephalopathy (MNGIE) is a rare autosomal recessive condition. Deficiency of thymidine phosphorylase disrupts the nucleoside pool, with progressive secondary mitochondrial DNA damage. MNGIE is clinically diagnosable because of a distinctive tetrad of gastrointestinal dysmotility, progressive external ophthalmoplegia, demyelinating neuropathy and asymptomatic leucoencephalopathy. The diagnosis may be confirmed genetically or biochemically. Misdiagnosis is frequent, but early and accurate recognition allows the possibility of novel transplant therapies capable of rectifying the biochemical defects. Its management remains difficult in the face of progressive disability and the risks of treatment.

6.
J Neurol Neurosurg Psychiatry ; 89(7): 762-768, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29437916

RESUMO

BACKGROUND: Defects in glycosylation of alpha-dystroglycan (α-DG) cause autosomal-recessive disorders with wide clinical and genetic heterogeneity, with phenotypes ranging from congenital muscular dystrophies to milder limb girdle muscular dystrophies. Patients show variable reduction of immunoreactivity to antibodies specific for glycoepitopes of α-DG on a muscle biopsy. Recessive mutations in 18 genes, including guanosine diphosphate mannose pyrophosphorylase B (GMPPB), have been reported to date. With no specific clinical and pathological handles, diagnosis requires parallel or sequential analysis of all known genes. METHODS: We describe clinical, genetic and biochemical findings of 21 patients with GMPPB-associated dystroglycanopathy. RESULTS: We report eight novel mutations and further expand current knowledge on clinical and muscle MRI features of this condition. In addition, we report a consistent shift in the mobility of beta-dystroglycan (ß-DG) on Western blot analysis of all patients analysed by this mean. This was only observed in patients with GMPPB in our large dystroglycanopathy cohort. We further demonstrate that this mobility shift in patients with GMPPB was due to abnormal N-linked glycosylation of ß-DG. CONCLUSIONS: Our data demonstrate that a change in ß-DG electrophoretic mobility in patients with dystroglycanopathy is a distinctive marker of the molecular defect in GMPPB.


Assuntos
Distroglicanas/metabolismo , Guanosina Difosfato Manose/genética , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Mutação/genética , Nucleotidiltransferases/genética , Adolescente , Idoso , Biomarcadores/metabolismo , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Distrofias Musculares/patologia
7.
Muscle Nerve ; 57(3): 380-387, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29053898

RESUMO

INTRODUCTION: Earlier small case series and clinical observations reported on chronic pain playing an important role in facioscapulohumeral dystrophy (FSHD). The aim of this study was to determine the characteristics and impact of pain on quality of life (QoL) in patients with FSHD. METHODS: We analyzed patient reported outcome measures collected through the U.K. FSHD Patient Registry. RESULTS: Of 398 patients, 88.6% reported pain at the time of study. The most frequent locations were shoulders and lower back. A total of 203 participants reported chronic pain, 30.4% of them as severe. The overall disease impact on QoL was significantly higher in patients with early onset and long disease duration. Chronic pain had a negative impact on all Individualised Neuromuscular Quality of Life Questionnaire domains and overall disease score. DISCUSSION: Our study shows that pain in FSHD type 1 (FSHD1) is frequent and strongly impacts on QoL, similar to other chronic, painful disorders. Management of pain should be considered when treating FSHD1 patients. Muscle Nerve 57: 380-387, 2018.


Assuntos
Dor Crônica/psicologia , Distrofia Muscular Facioescapuloumeral/psicologia , Qualidade de Vida/psicologia , Adulto , Idoso , Dor Crônica/complicações , Dor Crônica/diagnóstico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Distrofia Muscular Facioescapuloumeral/complicações , Distrofia Muscular Facioescapuloumeral/diagnóstico , Medição da Dor , Índice de Gravidade de Doença , Adulto Jovem
8.
J Neurol ; 264(5): 979-988, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28397002

RESUMO

Myotonic dystrophy type 1 (DM1) is the most frequent muscular dystrophy worldwide with complex, multi-systemic, and progressively worsening symptoms. There is currently no treatment for this inherited disorder and research can be challenging due to the rarity and variability of the disease. The UK Myotonic Dystrophy Patient Registry is a patient self-enrolling online database collecting clinical and genetic information. For this cross-sectional "snapshot" analysis, 556 patients with a confirmed diagnosis of DM1 registered between May 2012 and July 2016 were included. An almost even distribution was seen between genders and a broad range of ages was present from 8 months to 78 years, with the largest proportion between 30 and 59 years. The two most frequent symptoms were fatigue and myotonia, reported by 79 and 78% of patients, respectively. The severity of myotonia correlated with the severity of fatigue as well as mobility impairment, and dysphagia occurred mostly in patients also reporting myotonia. Men reported significantly more frequent severe myotonia, whereas severe fatigue was more frequently reported by women. Cardiac abnormalities were diagnosed in 48% of patients and more than one-third of them needed a cardiac implant. Fifteen percent of patients used a non-invasive ventilation and cataracts were removed in 26% of patients, 65% of which before the age of 50 years. The registry's primary aim was to facilitate and accelerate clinical research. However, these data also allow us to formulate questions for hypothesis-driven research that may lead to improvements in care and treatment.


Assuntos
Fadiga/etiologia , Distrofia Miotônica/epidemiologia , Sistema de Registros , Adolescente , Adulto , Distribuição por Idade , Idoso , Arritmias Cardíacas/epidemiologia , Catarata/epidemiologia , Criança , Pré-Escolar , Estudos Transversais , Eletrocardiografia , Fadiga/epidemiologia , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Transtornos dos Movimentos/epidemiologia , Transtornos dos Movimentos/etiologia , Distrofia Miotônica/diagnóstico , Distrofia Miotônica/genética , Distrofia Miotônica/terapia , Miotonina Proteína Quinase/genética , Fatores Sexuais , Expansão das Repetições de Trinucleotídeos/genética , Reino Unido/epidemiologia , Adulto Jovem
9.
Neurogenetics ; 18(2): 111-117, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28229249

RESUMO

We describe a family with an autosomal dominant familial dyskinesia resembling myoclonus-dystonia associated with a novel missense mutation in ADCY5, found through whole-exome sequencing. A tiered analytical approach was used to analyse whole-exome sequencing data from an affected grandmother-granddaughter pair. Whole-exome sequencing identified 18,000 shared variants, of which 46 were non-synonymous changes not present in a local cohort of control exomes (n = 422). Further filtering based on predicted splicing effect, minor allele frequency in the 1000 Genomes Project and on phylogenetic conservation yielded 13 candidate variants, of which the heterozygous missense mutation c.3086T>G, p. M1029R in ADCY5 most closely matched the observed phenotype. This report illustrates the utility of whole-exome sequencing in cases of undiagnosed movement disorders with clear autosomal dominant inheritance. Moreover, ADCY5 mutations should be considered in cases with apparent myoclonus-dystonia, particularly where SCGE mutations have been excluded. ADCY5-related dyskinesia may manifest variable expressivity within a single family, and affected individuals may be initially diagnosed with differing neurological phenotypes.


Assuntos
Adenilil Ciclases/genética , Discinesias/genética , Distúrbios Distônicos/genética , Adolescente , Adulto , Pré-Escolar , Discinesias/complicações , Distúrbios Distônicos/complicações , Família , Feminino , Humanos , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Linhagem , Fenótipo
10.
JAMA Neurol ; 73(12): 1433-1439, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27749956

RESUMO

IMPORTANCE: Adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) is a frequent cause of adult-onset leukodystrophy known to be caused by autosomal dominant mutations in the CSF1R (colony-stimulating factor 1) gene. The discovery that CSF1R mutations cause ALSP led to more accurate prognosis and genetic counseling for these patients in addition to increased interest in microglia as a target in neurodegeneration. However, it has been known since the discovery of the CSF1R gene that there are patients with typical clinical and radiologic evidence of ALSP who do not carry pathogenic CSF1R mutations. These patients include those in whom the pathognomonic features of axonal spheroids and pigmented microglia have been found. Achieving a genetic diagnosis in these patients is important to our understanding of this disorder. OBJECTIVE: To genetically characterize a group of patients with typical features of ALSP who do not carry CSF1R mutations. DESIGN, SETTINGS, AND PARTICIPANTS: In this case series study, 5 patients from 4 families were identified with clinical, radiologic, or pathologic features of ALSP in whom CSF1R mutations had been excluded previously by sequencing. Data were collected between May 2014 and September 2015 and analyzed between September 2015 and February 2016. MAIN OUTCOMES AND MEASURES: Focused exome sequencing was used to identify candidate variants. Family studies, long-range polymerase chain reaction with cloning, and complementary DNA sequencing were used to confirm pathogenicity. RESULTS: Of these 5 patients, 4 were men (80%); mean age at onset of ALSP was 29 years (range, 15-44 years). Biallelic mutations in the alanyl-transfer (t)RNA synthetase 2 (AARS2) gene were found in all 5 patients. Frameshifting and splice site mutations were common, found in 4 of 5 patients, and sequencing of complementary DNA from affected patients confirmed that the variants were loss of function. All patients presented in adulthood with prominent cognitive, neuropsychiatric, and upper motor neuron signs. Magnetic resonance imaging in all patients demonstrated a symmetric leukoencephalopathy with punctate regions of restricted diffusion, typical of ALSP. In 1 patient, brain biopsy demonstrated axonal spheroids and pigmented microglia, which are the pathognomonic signs of ALSP. CONCLUSIONS AND RELEVANCE: This work indicates that mutations in the tRNA synthetase AARS2 gene cause a recessive form of ALSP. The CSF1R and AARS2 proteins have different cellular functions but overlap in a final common pathway of neurodegeneration. This work points to novel targets for research and will lead to improved diagnostic rates in patients with adult-onset leukoencephalopathy.


Assuntos
Alanina-tRNA Ligase/genética , Leucoencefalopatias/diagnóstico por imagem , Leucoencefalopatias/genética , Leucoencefalopatias/fisiopatologia , Microglia/patologia , Adolescente , Adulto , Exoma , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Mutação , Linhagem , Análise de Sequência de DNA , Adulto Jovem
11.
Br J Hosp Med (Lond) ; 76(10): 576-82, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26457938

RESUMO

Patients with muscle disease present not only to neurologists, but also to rheumatologists and general physicians. This article provides a framework of how to approach patients with suspected muscle disease, and reviews the clinical features of the most frequently encountered acquired and genetic conditions in adult practice.


Assuntos
Distrofias Musculares/diagnóstico , Miosite/diagnóstico , Adulto , Humanos , Doenças Musculares/diagnóstico , Doenças Musculares/terapia , Distrofias Musculares/terapia , Miosite/terapia
12.
Brain ; 138(Pt 10): 2847-58, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26264513

RESUMO

Haematopoietic stem cell transplantation has been proposed as treatment for mitochondrial neurogastrointestinal encephalomyopathy, a rare fatal autosomal recessive disease due to TYMP mutations that result in thymidine phosphorylase deficiency. We conducted a retrospective analysis of all known patients suffering from mitochondrial neurogastrointestinal encephalomyopathy who underwent allogeneic haematopoietic stem cell transplantation between 2005 and 2011. Twenty-four patients, 11 males and 13 females, median age 25 years (range 10-41 years) treated with haematopoietic stem cell transplantation from related (n = 9) or unrelated donors (n = 15) in 15 institutions worldwide were analysed for outcome and its associated factors. Overall, 9 of 24 patients (37.5%) were alive at last follow-up with a median follow-up of these surviving patients of 1430 days. Deaths were attributed to transplant in nine (including two after a second transplant due to graft failure), and to mitochondrial neurogastrointestinal encephalomyopathy in six patients. Thymidine phosphorylase activity rose from undetectable to normal levels (median 697 nmol/h/mg protein, range 262-1285) in all survivors. Seven patients (29%) who were engrafted and living more than 2 years after transplantation, showed improvement of body mass index, gastrointestinal manifestations, and peripheral neuropathy. Univariate statistical analysis demonstrated that survival was associated with two defined pre-transplant characteristics: human leukocyte antigen match (10/10 versus <10/10) and disease characteristics (liver disease, history of gastrointestinal pseudo-obstruction or both). Allogeneic haematopoietic stem cell transplantation can restore thymidine phosphorylase enzyme function in patients with mitochondrial neurogastrointestinal encephalomyopathy and improve clinical manifestations of mitochondrial neurogastrointestinal encephalomyopathy in the long term. Allogeneic haematopoietic stem cell transplantation should be considered for selected patients with an optimal donor.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Pseudo-Obstrução Intestinal/cirurgia , Encefalomiopatias Mitocondriais/cirurgia , Resultado do Tratamento , Adolescente , Adulto , Peso Corporal , Encéfalo/patologia , Criança , Feminino , Seguimentos , Humanos , Imageamento por Ressonância Magnética , Masculino , Distrofia Muscular Oculofaríngea , Condução Nervosa/fisiologia , Exame Neurológico , Neutrófilos , Oftalmoplegia/congênito , Estudos Retrospectivos , Análise de Sobrevida , Timidina Fosforilase/metabolismo , Transplante Homólogo/métodos , Adulto Jovem
13.
Sci Rep ; 5: 10042, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25975230

RESUMO

We report a new family with autosomal dominant inheritance of a late onset rapidly progressive leukodystrophy in which exome sequencing has revealed a novel mutation p.R782G in the Colony-Stimulating Factor 1 Receptor gene (CSF1R). Neuropathology of two affected family members showed cerebral white matter degeneration with axonal swellings and pigmented macrophages. The few recently reported families with CSF1R mutations had been previously labelled "hereditary diffuse leukencephalopathy with axonal spheroids" (HDLS) and "pigmentary orthochromatic leukodystrophy" (POLD), disorders which now appear to form a disease continuum. The term "adult-onset leukoencephalopathy with axonal spheroids and pigmented glia" (ALSP) has been proposed to encompass this spectrum. As CSF1R regulates microglia this mutation implies that dysregulation of microglia is the primary cause of the disease.


Assuntos
Axônios/patologia , Neuroglia/patologia , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Substância Branca/patologia , Adulto , Feminino , Humanos , Leucoencefalopatias/genética , Macrófagos/citologia , Masculino , Mutação/genética , Neuroglia/citologia
14.
Eur J Hum Genet ; 22(6): 801-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24193343

RESUMO

Myosin myopathies comprise a group of inherited diseases caused by mutations in myosin heavy chain (MyHC) genes. Homozygous or compound heterozygous truncating MYH2 mutations have been demonstrated to cause recessive myopathy with ophthalmoplegia, mild-to-moderate muscle weakness and complete lack of type 2A muscle fibers. In this study, we describe for the first time the clinical and morphological characteristics of recessive myosin IIa myopathy associated with MYH2 missense mutations. Seven patients of five different families with a myopathy characterized by ophthalmoplegia and mild-to-moderate muscle weakness were investigated. Muscle biopsy was performed to study morphological changes and MyHC isoform expression. Five of the patients were homozygous for MYH2 missense mutations, one patient was compound heterozygous for a missense and a nonsense mutation and one patient was homozygous for a frame-shift MYH2 mutation. Muscle biopsy demonstrated small or absent type 2A muscle fibers and reduced or absent expression of the corresponding MyHC IIa transcript and protein. We conclude that mild muscle weakness and ophthalmoplegia in combination with muscle biopsy demonstrating small or absent type 2A muscle fibers are the hallmark of recessive myopathy associated with MYH2 mutations.


Assuntos
Predisposição Genética para Doença/genética , Doenças Musculares/genética , Mutação de Sentido Incorreto , Cadeias Pesadas de Miosina/genética , Oftalmoplegia/genética , Adulto , Biópsia , Criança , Códon sem Sentido , Análise Mutacional de DNA , Saúde da Família , Feminino , Expressão Gênica , Genes Recessivos , Humanos , Masculino , Pessoa de Meia-Idade , Fibras Musculares de Contração Rápida/patologia , Debilidade Muscular/patologia , Doenças Musculares/patologia , Oftalmoplegia/patologia , Linhagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Hum Mutat ; 34(9): 1260-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23696415

RESUMO

Mutations in the mitochondrial genome, and in particular the mt-tRNAs, are an important cause of human disease. Accurate classification of the pathogenicity of novel variants is vital to allow accurate genetic counseling for patients and their families. The use of weighted criteria based on functional studies-outlined in a validated pathogenicity scoring system--is therefore invaluable in determining whether novel or rare mt-tRNA variants are pathogenic. Here, we describe the identification of nine novel mt--tRNA variants in nine families, in which the probands presented with a diverse range of clinical phenotypes including mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes, isolated progressive external ophthalmoplegia, epilepsy, deafness and diabetes. Each of the variants identified (m.4289T>C, MT-TI; m.5541C>T, MT-TW; m.5690A>G, MT-TN; m.7451A>T, MT-TS1; m.7554G>A, MT-TD; m.8304G>A, MT-TK; m.12206C>T, MT-TH; m.12317T>C, MT-TL2; m.16023G>A, MT-TP) was present in a different tRNA, with evidence in support of pathogenicity, and where possible, details of mutation transmission documented. Through the application of the pathogenicity scoring system, we have classified six of these variants as "definitely pathogenic" mutations (m.5541C>T, m.5690A>G, m.7451A>T, m.12206C>T, m.12317T>C, and m.16023G>A), whereas the remaining three currently lack sufficient evidence and are therefore classed as 'possibly pathogenic' (m.4289T>C, m.7554G>A, and m.8304G>A).


Assuntos
Doenças Mitocondriais/genética , Mutação Puntual , RNA de Transferência/genética , RNA/genética , Adolescente , Adulto , Criança , DNA Mitocondrial/genética , Feminino , Variação Genética , Humanos , Síndrome MELAS/genética , Masculino , Pessoa de Meia-Idade , Mitocôndrias/genética , Doenças Mitocondriais/patologia , Encefalomiopatias Mitocondriais/genética , RNA/metabolismo , RNA Mitocondrial , RNA de Transferência/metabolismo , Análise de Sequência de DNA , Adulto Jovem
16.
Hum Mutat ; 34(8): 1111-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23606453

RESUMO

Limb girdle muscular dystrophy type 2L or anoctaminopathy is a condition mainly characterized by adult onset proximal lower limb muscular weakness and raised CK values, due to recessive ANO5 gene mutations. An exon 5 founder mutation (c.191dupA) has been identified in most of the British and German LGMD2L patients so far reported. We aimed to further investigate the prevalence and spectrum of ANO5 gene mutations and related clinical phenotypes, by screening 205 undiagnosed patients referred to our molecular service with a clinical suspicion of anoctaminopathy. A total of 42 unrelated patients had two ANO5 mutations (21%), whereas 14 carried a single change. We identified 34 pathogenic changes, 15 of which are novel. The c.191dupA mutation represents 61% of mutated alleles and appears to be less prevalent in non-Northern European populations. Retrospective clinical analysis corroborates the prevalently proximal lower limb phenotype, the male predominance and absence of major cardiac or respiratory involvement. Identification of cases with isolated hyperCKaemia and very late symptomatic male and female subjects confirms the extension of the phenotypic spectrum of the disease. Anoctaminopathy appears to be one of the most common adult muscular dystrophies in Northern Europe, with a prevalence of about 20%-25% in unselected undiagnosed cases.


Assuntos
Canais de Cloreto/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , Adulto , Idoso , Anoctaminas , Canais de Cloreto/metabolismo , Europa (Continente)/epidemiologia , Feminino , Variação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Distrofia Muscular do Cíngulo dos Membros/epidemiologia , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Fenótipo , Prevalência , Estudos Retrospectivos , Fatores Sexuais
17.
Neurology ; 80(3): 268-75, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23269600

RESUMO

OBJECTIVE: To assess the frequency of mutations in C19orf12 in the greater neurodegeneration with brain iron accumulation (NBIA) population and further characterize the associated phenotype. METHODS: Samples from 161 individuals with idiopathic NBIA were screened, and C19orf12 mutations were identified in 23 subjects. Direct examinations were completed on 8 of these individuals, and medical records were reviewed on all 23. Histochemical and immunohistochemical studies were performed on brain tissue from one deceased subject. RESULTS: A variety of mutations were detected in this cohort, in addition to the Eastern European founder mutation described previously. The characteristic clinical features of mitochondrial membrane protein-associated neurodegeneration (MPAN) across all age groups include cognitive decline progressing to dementia, prominent neuropsychiatric abnormalities, and a motor neuronopathy. A distinctive pattern of brain iron accumulation is universal. Neuropathologic studies revealed neuronal loss, widespread iron deposits, and eosinophilic spheroidal structures in the basal ganglia. Lewy neurites were present in the globus pallidus, and Lewy bodies and neurites were widespread in other areas of the corpus striatum and midbrain structures. CONCLUSIONS: MPAN is caused by mutations in C19orf12 leading to NBIA and prominent, widespread Lewy body pathology. The clinical phenotype is recognizable and distinctive, and joins pantothenate kinase-associated neurodegeneration and PLA2G6-associated neurodegeneration as one of the major forms of NBIA.


Assuntos
Sobrecarga de Ferro/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Adolescente , Adulto , Química Encefálica/genética , Criança , Pré-Escolar , Estudos de Coortes , DNA/genética , Distonia/etiologia , Eletroencefalografia , Eletromiografia , Incontinência Fecal/etiologia , Feminino , Transtornos Neurológicos da Marcha/etiologia , Humanos , Imuno-Histoquímica , Sobrecarga de Ferro/diagnóstico por imagem , Sobrecarga de Ferro/patologia , Doença por Corpos de Lewy/patologia , Masculino , Proteínas Mitocondriais/genética , Mutação , Doenças Neurodegenerativas/complicações , Doenças Neurodegenerativas/diagnóstico por imagem , Exame Neurológico , Fenótipo , Radiografia , Incontinência Urinária/etiologia , Adulto Jovem
18.
Nat Genet ; 44(3): 338-42, 2012 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-22267198

RESUMO

Coats plus is a highly pleiotropic disorder particularly affecting the eye, brain, bone and gastrointestinal tract. Here, we show that Coats plus results from mutations in CTC1, encoding conserved telomere maintenance component 1, a member of the mammalian homolog of the yeast heterotrimeric CST telomeric capping complex. Consistent with the observation of shortened telomeres in an Arabidopsis CTC1 mutant and the phenotypic overlap of Coats plus with the telomeric maintenance disorders comprising dyskeratosis congenita, we observed shortened telomeres in three individuals with Coats plus and an increase in spontaneous γH2AX-positive cells in cell lines derived from two affected individuals. CTC1 is also a subunit of the α-accessory factor (AAF) complex, stimulating the activity of DNA polymerase-α primase, the only enzyme known to initiate DNA replication in eukaryotic cells. Thus, CTC1 may have a function in DNA metabolism that is necessary for but not specific to telomeric integrity.


Assuntos
Anormalidades Múltiplas/genética , Predisposição Genética para Doença/genética , Telangiectasia Retiniana/genética , Proteínas de Ligação a Telômeros/genética , Telômero/patologia , Sequência de Bases , Citometria de Fluxo , Histonas/metabolismo , Dados de Sequência Molecular , Telangiectasia Retiniana/patologia , Análise de Sequência de DNA/métodos
19.
Nat Genet ; 43(6): 595-600, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21532572

RESUMO

DNA methyltransferase 1 (DNMT1) is crucial for maintenance of methylation, gene regulation and chromatin stability. DNA mismatch repair, cell cycle regulation in post-mitotic neurons and neurogenesis are influenced by DNA methylation. Here we show that mutations in DNMT1 cause both central and peripheral neurodegeneration in one form of hereditary sensory and autonomic neuropathy with dementia and hearing loss. Exome sequencing led to the identification of DNMT1 mutation c.1484A>G (p.Tyr495Cys) in two American kindreds and one Japanese kindred and a triple nucleotide change, c.1470-1472TCC>ATA (p.Asp490Glu-Pro491Tyr), in one European kindred. All mutations are within the targeting-sequence domain of DNMT1. These mutations cause premature degradation of mutant proteins, reduced methyltransferase activity and impaired heterochromatin binding during the G2 cell cycle phase leading to global hypomethylation and site-specific hypermethylation. Our study shows that DNMT1 mutations cause the aberrant methylation implicated in complex pathogenesis. The discovered DNMT1 mutations provide a new framework for the study of neurodegenerative diseases.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Adolescente , Adulto , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA , Demência/genética , Feminino , Fase G2 , Perda Auditiva/genética , Neuropatias Hereditárias Sensoriais e Autônomas/genética , Heterocromatina/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Mutação
20.
Acta Neuropathol ; 121(3): 421-7, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20721566

RESUMO

Unverricht-Lundborg disease (EPM1A), also known as Baltic myoclonus, is the most common form of progressive myoclonic epilepsy. It is inherited as an autosomal recessive trait, due to mutations in the Cystatin-B gene promoter region. Although there is much work on rodent models of this disease, there is very little published neuropathology in patients with EPM1A. Here, we present the neuropathology of a patient with genetically confirmed EPM1A, who died at the age of 76. There was atrophy and gliosis affecting predominantly the cerebellum, frontotemporal cortex, hippocampus and thalamus. We have identified neuronal cytoplasmic inclusions containing the lysosomal proteins, Cathepsin-B and CD68. These inclusions also showed immunopositivity to both TDP-43 and FUS, in some cases associated with an absence of normal neuronal nuclear TDP-43 staining. There were also occasional ubiquitinylated neuronal intranuclear inclusions, some of which were FUS immunopositive. This finding is consistent with neurodegeneration in EPM1A as at least a partial consequence of lysosomal damage to neurons, which have reduced Cystatin-B-related neuroprotection. It also reveals a genetically defined neurodegenerative disease with both FUS and TDP-43 related pathology.


Assuntos
Corpos de Inclusão/patologia , Corpos de Inclusão Intranuclear/patologia , Neurônios/patologia , Síndrome de Unverricht-Lundborg/patologia , Idoso , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Atrofia/patologia , Cistatina B/genética , Cistatina B/metabolismo , Proteínas de Ligação a DNA/metabolismo , Evolução Fatal , Feminino , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , Mutação/genética , Neurônios/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA