Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Am J Hum Genet ; 110(1): 105-119, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36493768

ABSTRACT

Adult-onset cerebellar ataxias are a group of neurodegenerative conditions that challenge both genetic discovery and molecular diagnosis. In this study, we identified an intronic (GAA) repeat expansion in fibroblast growth factor 14 (FGF14). Genetic analysis of 95 Australian individuals with adult-onset ataxia identified four (4.2%) with (GAA)>300 and a further nine individuals with (GAA)>250. PCR and long-read sequence analysis revealed these were pure (GAA) repeats. In comparison, no control subjects had (GAA)>300 and only 2/311 control individuals (0.6%) had a pure (GAA)>250. In a German validation cohort, 9/104 (8.7%) of affected individuals had (GAA)>335 and a further six had (GAA)>250, whereas 10/190 (5.3%) control subjects had (GAA)>250 but none were (GAA)>335. The combined data suggest (GAA)>335 are disease causing and fully penetrant (p = 6.0 × 10-8, OR = 72 [95% CI = 4.3-1,227]), while (GAA)>250 is likely pathogenic with reduced penetrance. Affected individuals had an adult-onset, slowly progressive cerebellar ataxia with variable features including vestibular impairment, hyper-reflexia, and autonomic dysfunction. A negative correlation between age at onset and repeat length was observed (R2 = 0.44, p = 0.00045, slope = -0.12) and identification of a shared haplotype in a minority of individuals suggests that the expansion can be inherited or generated de novo during meiotic division. This study demonstrates the power of genome sequencing and advanced bioinformatic tools to identify novel repeat expansions via model-free, genome-wide analysis and identifies SCA50/ATX-FGF14 as a frequent cause of adult-onset ataxia.


Subject(s)
Cerebellar Ataxia , Fibroblast Growth Factors , Friedreich Ataxia , Trinucleotide Repeat Expansion , Adult , Humans , Ataxia/genetics , Australia , Cerebellar Ataxia/genetics , Friedreich Ataxia/genetics , Trinucleotide Repeat Expansion/genetics
2.
Hum Mutat ; 43(1): 16-29, 2022 01.
Article in English | MEDLINE | ID: mdl-34633740

ABSTRACT

Autism spectrum disorders (ASD) are neurodevelopmental disorders with an estimated heritability of >60%. Family-based genetic studies of ASD have generally focused on multiple small kindreds, searching for de novo variants of major effect. We hypothesized that molecular genetic analysis of large multiplex families would enable the identification of variants of milder effects. We studied a large multigenerational family of European ancestry with multiple family members affected with ASD or the broader autism phenotype (BAP). We identified a rare heterozygous variant in the gene encoding 1,4-ɑ-glucan branching enzyme 1 (GBE1) that was present in seven of seven individuals with ASD, nine of ten individuals with the BAP, and none of four tested unaffected individuals. We genotyped a community-acquired cohort of 389 individuals with ASD and identified three additional probands. Cascade analysis demonstrated that the variant was present in 11 of 13 individuals with familial ASD/BAP and neither of the two tested unaffected individuals in these three families, also of European ancestry. The variant was not enriched in the combined UK10K ASD cohorts of European ancestry but heterozygous GBE1 deletion was overrepresented in large ASD cohorts, collectively suggesting an association between GBE1 and ASD.


Subject(s)
1,4-alpha-Glucan Branching Enzyme , Autism Spectrum Disorder , Glycogen Debranching Enzyme System , 1,4-alpha-Glucan Branching Enzyme/genetics , Autism Spectrum Disorder/genetics , Exome , Genetic Predisposition to Disease , Glucans , Glycogen Debranching Enzyme System/genetics , Humans
3.
Am J Hum Genet ; 105(1): 151-165, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31230722

ABSTRACT

Genomic technologies such as next-generation sequencing (NGS) are revolutionizing molecular diagnostics and clinical medicine. However, these approaches have proven inefficient at identifying pathogenic repeat expansions. Here, we apply a collection of bioinformatics tools that can be utilized to identify either known or novel expanded repeat sequences in NGS data. We performed genetic studies of a cohort of 35 individuals from 22 families with a clinical diagnosis of cerebellar ataxia with neuropathy and bilateral vestibular areflexia syndrome (CANVAS). Analysis of whole-genome sequence (WGS) data with five independent algorithms identified a recessively inherited intronic repeat expansion [(AAGGG)exp] in the gene encoding Replication Factor C1 (RFC1). This motif, not reported in the reference sequence, localized to an Alu element and replaced the reference (AAAAG)11 short tandem repeat. Genetic analyses confirmed the pathogenic expansion in 18 of 22 CANVAS-affected families and identified a core ancestral haplotype, estimated to have arisen in Europe more than twenty-five thousand years ago. WGS of the four RFC1-negative CANVAS-affected families identified plausible variants in three, with genomic re-diagnosis of SCA3, spastic ataxia of the Charlevoix-Saguenay type, and SCA45. This study identified the genetic basis of CANVAS and demonstrated that these improved bioinformatics tools increase the diagnostic utility of WGS to determine the genetic basis of a heterogeneous group of clinically overlapping neurogenetic disorders.


Subject(s)
Cerebellar Ataxia/etiology , Computational Biology/methods , Introns , Microsatellite Repeats , Polyneuropathies/etiology , Replication Protein C/genetics , Sensation Disorders/etiology , Vestibular Diseases/etiology , Algorithms , Cerebellar Ataxia/pathology , Cohort Studies , Family , Female , Genomics , Humans , Male , Middle Aged , Polyneuropathies/pathology , Sensation Disorders/pathology , Syndrome , Vestibular Diseases/pathology , Whole Genome Sequencing
5.
J Hum Genet ; 64(12): 1173-1186, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31530938

ABSTRACT

Coffin-Siris syndrome (CSS, MIM#135900) is a congenital disorder characterized by coarse facial features, intellectual disability, and hypoplasia of the fifth digit and nails. Pathogenic variants for CSS have been found in genes encoding proteins in the BAF (BRG1-associated factor) chromatin-remodeling complex. To date, more than 150 CSS patients with pathogenic variants in nine BAF-related genes have been reported. We previously reported 71 patients of whom 39 had pathogenic variants. Since then, we have recruited an additional 182 CSS-suspected patients. We performed comprehensive genetic analysis on these 182 patients and on the previously unresolved 32 patients, targeting pathogenic single nucleotide variants, short insertions/deletions and copy number variations (CNVs). We confirmed 78 pathogenic variations in 78 patients. Pathogenic variations in ARID1B, SMARCB1, SMARCA4, ARID1A, SOX11, SMARCE1, and PHF6 were identified in 48, 8, 7, 6, 4, 1, and 1 patients, respectively. In addition, we found three CNVs including SMARCA2. Of particular note, we found a partial deletion of SMARCB1 in one CSS patient and we thoroughly investigated the resulting abnormal transcripts.


Subject(s)
Abnormalities, Multiple/genetics , Face/abnormalities , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Hand Deformities, Congenital/genetics , Intellectual Disability/genetics , Micrognathism/genetics , Neck/abnormalities , Cohort Studies , Genetic Association Studies/methods , Humans
6.
Am J Med Genet A ; 179(2): 150-158, 2019 02.
Article in English | MEDLINE | ID: mdl-30614194

ABSTRACT

Cornelia de Lange syndrome (CdLS) is a dominant multisystemic malformation syndrome due to mutations in five genes-NIPBL, SMC1A, HDAC8, SMC3, and RAD21. The characteristic facial dysmorphisms include microcephaly, arched eyebrows, synophrys, short nose with depressed bridge and anteverted nares, long philtrum, thin lips, micrognathia, and hypertrichosis. Most affected individuals have intellectual disability, growth deficiency, and upper limb anomalies. This study looked at individuals from diverse populations with both clinical and molecularly confirmed diagnoses of CdLS by facial analysis technology. Clinical data and images from 246 individuals with CdLS were obtained from 15 countries. This cohort included 49% female patients and ages ranged from infancy to 37 years. Individuals were grouped into ancestry categories of African descent, Asian, Latin American, Middle Eastern, and Caucasian. Across these populations, 14 features showed a statistically significant difference. The most common facial features found in all ancestry groups included synophrys, short nose with anteverted nares, and a long philtrum with thin vermillion of the upper lip. Using facial analysis technology we compared 246 individuals with CdLS to 246 gender/age matched controls and found that sensitivity was equal or greater than 95% for all groups. Specificity was equal or greater than 91%. In conclusion, we present consistent clinical findings from global populations with CdLS while demonstrating how facial analysis technology can be a tool to support accurate diagnoses in the clinical setting. This work, along with prior studies in this arena, will assist in earlier detection, recognition, and treatment of CdLS worldwide.


Subject(s)
Abnormalities, Multiple/genetics , Cell Cycle Proteins/genetics , De Lange Syndrome/genetics , Intellectual Disability/genetics , Abnormalities, Multiple/epidemiology , Abnormalities, Multiple/physiopathology , Adolescent , Adult , Child , Child, Preschool , Chondroitin Sulfate Proteoglycans/genetics , Chromosomal Proteins, Non-Histone/genetics , De Lange Syndrome/epidemiology , De Lange Syndrome/physiopathology , Face/physiopathology , Female , Humans , Image Processing, Computer-Assisted , Infant , Infant, Newborn , Intellectual Disability/epidemiology , Intellectual Disability/physiopathology , Male , Mutation , Phenotype , Racial Groups/genetics , Young Adult
7.
Am J Hum Genet ; 95(6): 729-35, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25434005

ABSTRACT

Advances in understanding the etiology of Parkinson disease have been driven by the identification of causative mutations in families. Genetic analysis of an Australian family with three males displaying clinical features of early-onset parkinsonism and intellectual disability identified a ∼45 kb deletion resulting in the complete loss of RAB39B. We subsequently identified a missense mutation (c.503C>A [p.Thr168Lys]) in RAB39B in an unrelated Wisconsin kindred affected by a similar clinical phenotype. In silico and in vitro studies demonstrated that the mutation destabilized the protein, consistent with loss of function. In vitro small-hairpin-RNA-mediated knockdown of Rab39b resulted in a reduction in the density of α-synuclein immunoreactive puncta in dendritic processes of cultured neurons. In addition, in multiple cell models, we demonstrated that knockdown of Rab39b was associated with reduced steady-state levels of α-synuclein. Post mortem studies demonstrated that loss of RAB39B resulted in pathologically confirmed Parkinson disease. There was extensive dopaminergic neuron loss in the substantia nigra and widespread classic Lewy body pathology. Additional pathological features included cortical Lewy bodies, brain iron accumulation, tau immunoreactivity, and axonal spheroids. Overall, we have shown that loss-of-function mutations in RAB39B cause intellectual disability and pathologically confirmed early-onset Parkinson disease. The loss of RAB39B results in dysregulation of α-synuclein homeostasis and a spectrum of neuropathological features that implicate RAB39B in the pathogenesis of Parkinson disease and potentially other neurodegenerative disorders.


Subject(s)
Genes, X-Linked , Intellectual Disability/genetics , Nerve Degeneration/genetics , Parkinson Disease/genetics , alpha-Synuclein/metabolism , rab GTP-Binding Proteins/genetics , Amino Acid Substitution , Australia , Base Sequence , Dopamine/metabolism , Female , Gene Expression Regulation , Humans , Intellectual Disability/physiopathology , Lewy Bodies/metabolism , Male , Middle Aged , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Nerve Degeneration/physiopathology , Parkinson Disease/physiopathology , Pedigree , Sequence Analysis, DNA , Sequence Deletion , Substantia Nigra/physiopathology , rab GTP-Binding Proteins/metabolism
8.
Ann Neurol ; 79(1): 132-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26285051

ABSTRACT

We describe first cousin sibling pairs with focal epilepsy, one of each pair having focal cortical dysplasia (FCD) IIa. Linkage analysis and whole-exome sequencing identified a heterozygous germline frameshift mutation in the gene encoding nitrogen permease regulator-like 3 (NPRL3). NPRL3 is a component of GAP Activity Towards Rags 1, a negative regulator of the mammalian target of rapamycin complex 1 signaling pathway. Immunostaining of resected brain tissue demonstrated mammalian target of rapamycin activation. Screening of 52 unrelated individuals with FCD identified 2 additional patients with FCDIIa and germline NPRL3 mutations. Similar to DEPDC5, NPRL3 mutations may be considered as causal variants in patients with FCD or magnetic resonance imaging-negative focal epilepsy.


Subject(s)
Epilepsies, Partial/genetics , Epilepsy/genetics , GTPase-Activating Proteins/genetics , Malformations of Cortical Development, Group I/genetics , Child , Child, Preschool , Female , Humans , Male , Mutation , Pedigree , Signal Transduction , TOR Serine-Threonine Kinases
9.
Eur J Hum Genet ; 31(1): 122-124, 2023 01.
Article in English | MEDLINE | ID: mdl-35945246

ABSTRACT

Several neurological disorders, such as myotonic dystrophy are caused by expansions of short tandem repeats (STRs) which can be difficult to detect by molecular tools. Methodological advances have made repeat expansion (RE) detection with whole genome sequencing (WGS) feasible. We recruited a multi-generational family (family A) ascertained for genetic studies of autism spectrum disorder. WGS was performed on seven children from four nuclear families from family A and analyzed for REs of STRs known to cause neurological disorders. We detected an expansion of a heterozygous intronic CCTG STR in CNBP in two siblings. This STR causes myotonic dystrophy type 2 (DM2). The expansion did not segregate with the ASD phenotype. Repeat-primed PCR showed that the DM2 CCTG motif was expanded above the pathogenic threshold in both children and their mother. On subsequent examination, the mother had mild features of DM2. We show that screening of STRs in WGS datasets has diagnostic utility, both in the clinical and research domain, with potential management and genetic counseling implications.


Subject(s)
Autism Spectrum Disorder , Myotonic Dystrophy , Humans , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Autism Spectrum Disorder/genetics , Chromosome Mapping , Microsatellite Repeats , Introns
11.
Brain Commun ; 3(1): fcaa235, 2021.
Article in English | MEDLINE | ID: mdl-33738444

ABSTRACT

Brain somatic mutations are an increasingly recognized cause of epilepsy, brain malformations and autism spectrum disorders and may be a hidden cause of other neurodevelopmental and neurodegenerative disorders. At present, brain mosaicism can be detected only in the rare situations of autopsy or brain biopsy. Liquid biopsy using cell-free DNA derived from cerebrospinal fluid has detected somatic mutations in malignant brain tumours. Here, we asked if cerebrospinal fluid liquid biopsy can be used to detect somatic mosaicism in non-malignant brain diseases. First, we reliably quantified cerebrospinal fluid cell-free DNA in 28 patients with focal epilepsy and 28 controls using droplet digital PCR. Then, in three patients we identified somatic mutations in cerebrospinal fluid: in one patient with subcortical band heterotopia the LIS1 p. Lys64* variant at 9.4% frequency; in a second patient with focal cortical dysplasia the TSC1 p. Phe581His*6 variant at 7.8% frequency; and in a third patient with ganglioglioma the BRAF p. Val600Glu variant at 3.2% frequency. To determine if cerebrospinal fluid cell-free DNA was brain-derived, whole-genome bisulphite sequencing was performed and brain-specific DNA methylation patterns were found to be significantly enriched (P = 0.03). Our proof of principle study shows that cerebrospinal fluid liquid biopsy is valuable in investigating mosaic neurological disorders where brain tissue is unavailable.

12.
Neurology ; 95(18): e2542-e2551, 2020 11 03.
Article in English | MEDLINE | ID: mdl-32847954

ABSTRACT

OBJECTIVE: To determine the genetic basis of bottom-of-sulcus dysplasia (BOSD), which is a highly focal and epileptogenic cortical malformation in which the imaging, electrophysiologic, and pathologic abnormalities are maximal at the bottom of sulcus, tapering to a normal gyral crown. METHODS: Targeted panel deep sequencing (>500×) was performed on paired blood and brain-derived genomic DNA from 20 operated patients with drug-resistant focal epilepsy and BOSD. Histopathology was assessed using immunohistochemistry. RESULTS: Brain-specific pathogenic somatic variants were found in 6 patients and heterozygous pathogenic germline variants were found in 2. Somatic variants were identified in MTOR and germline variants were identified in DEPDC5 and NPRL3. Two patients with somatic MTOR variants showed a mutation gradient, with higher mutation load at the bottom of sulcus compared to the gyral crown. Immunohistochemistry revealed an abundance of dysmorphic neurons and balloon cells in the bottom of sulcus but not in the gyral crown or adjacent gyri. CONCLUSIONS: BOSD is associated with mTOR pathway dysregulation and shares common genetic etiologies and pathogenic mechanisms with other forms of focal and hemispheric cortical dysplasia, suggesting these disorders are on a genetic continuum.


Subject(s)
Malformations of Cortical Development/genetics , TOR Serine-Threonine Kinases/genetics , Adolescent , Child , Child, Preschool , Drug Resistant Epilepsy/complications , Drug Resistant Epilepsy/genetics , Epilepsies, Partial/complications , Epilepsies, Partial/genetics , Female , Humans , Infant , Male , Malformations of Cortical Development/complications , Malformations of Cortical Development/pathology , Malformations of Cortical Development/surgery , Mutation
13.
Parkinsonism Relat Disord ; 64: 308-311, 2019 07.
Article in English | MEDLINE | ID: mdl-30928208

ABSTRACT

BACKGROUND: Bi-allelic mutations in PARK7 are a rare cause of autosomal recessive early onset Parkinson's disease (EO-PD). To date, 30 individuals harbouring 20 unique causative variants have been described. Understanding of the spectrum of clinical features and natural history of PARK7 mediated EO-PD remain limited. METHODS: We studied a family with three offspring, two of whom were affected with EO-PD. Family members underwent detailed clinical examination and DNA samples from both affected individuals and parents were analysed by exome sequencing. RESULTS: Two brothers of Iranian descent presented at age 29 years with Parkinsonism associated with high-pitched voice and hypomimia. The brothers were followed over a six and fifteen-year period and displayed typical levodopa responsive slowly-progressive Parkinsonism. A novel homozygous frameshift mutation in PARK7 [NM_007262.4:c.90dupG, p(Ile31Aspfs*2)] was identified. CONCLUSIONS: Here we report the clinical presentation and progression of EO-PD in brothers with a novel pathogenic PARK7 variant. We expand the clinical phenotype and provide an update of clinical and pathological features of the disorder.


Subject(s)
Parkinson Disease/genetics , Parkinson Disease/physiopathology , Protein Deglycase DJ-1/genetics , Adult , Facial Expression , Frameshift Mutation , Humans , Iran , Male , Parkinson Disease/complications , Pedigree , Siblings , Voice Disorders/etiology , Voice Disorders/physiopathology
14.
Ann Clin Transl Neurol ; 6(7): 1338-1344, 2019 07.
Article in English | MEDLINE | ID: mdl-31353856

ABSTRACT

Focal cortical dysplasia (FCD) causes drug-resistant epilepsy and is associated with pathogenic variants in mTOR pathway genes. How germline variants cause these focal lesions is unclear, however a germline + somatic "2-hit" model is hypothesized. In a boy with drug-resistant epilepsy, FCD, and a germline DEPDC5 pathogenic variant, we show that a second-hit DEPDC5 variant is limited to dysmorphic neurons, and the somatic mutation load correlates with both dysmorphic neuron density and the epileptogenic zone. These findings provide new insights into the molecular and cellular correlates of FCD determining drug-resistant epilepsy and refine conceptualization of the epileptogenic zone.


Subject(s)
GTPase-Activating Proteins/genetics , Malformations of Cortical Development/genetics , Neurons/pathology , Brain/diagnostic imaging , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Child , Drug Resistant Epilepsy/genetics , Humans , Male , Malformations of Cortical Development/diagnostic imaging , Malformations of Cortical Development/pathology , Malformations of Cortical Development/physiopathology , Mutation , TOR Serine-Threonine Kinases/genetics
15.
Neurol Genet ; 4(3): e236, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29725622

ABSTRACT

OBJECTIVE: To determine whether the GNAQ R183Q mutation is present in the forme fruste cases of Sturge-Weber syndrome (SWS) to establish a definitive molecular diagnosis. METHODS: We used sensitive droplet digital PCR (ddPCR) to detect and quantify the GNAQ mutation in tissues from epilepsy surgery in 4 patients with leptomeningeal angiomatosis; none had ocular or cutaneous manifestations. RESULTS: Low levels of the GNAQ mutation were detected in the brain tissue of all 4 cases-ranging from 0.42% to 7.1% frequency-but not in blood-derived DNA. Molecular evaluation confirmed the diagnosis in 1 case in which the radiologic and pathologic data were equivocal. CONCLUSIONS: We detected the mutation at low levels, consistent with mosaicism in the brain or skin (1.0%-18.1%) of classic cases. Our data confirm that the forme fruste is part of the spectrum of SWS, with the same molecular mechanism as the classic disease and that ddPCR is helpful where conventional diagnosis is uncertain.

16.
Nat Genet ; 49(4): 511-514, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28250454

ABSTRACT

Brain malformations involving the corpus callosum are common in children with developmental disabilities. We identified DCC mutations in four families and five sporadic individuals with isolated agenesis of the corpus callosum (ACC) without intellectual disability. DCC mutations result in variable dominant phenotypes with decreased penetrance, including mirror movements and ACC associated with a favorable developmental prognosis. Possible phenotypic modifiers include the type and location of mutation and the sex of the individual.


Subject(s)
Agenesis of Corpus Callosum/genetics , Developmental Disabilities/genetics , Mutation/genetics , Receptors, Cell Surface/genetics , Tumor Suppressor Proteins/genetics , Abnormalities, Multiple/genetics , Brain/pathology , Corpus Callosum/pathology , DCC Receptor , Family , Female , Humans , Male , Nervous System Malformations/genetics , Neural Stem Cells/pathology , Penetrance , Phenotype
17.
Neurol Genet ; 2(6): e114, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27790638

ABSTRACT

OBJECTIVE: To determine the genetic cause of slowly progressive cerebellar ataxia, sensorineural deafness, and hypergonadotropic hypogonadism in 5 patients from 3 different families. METHODS: The patients comprised 2 sib pairs and 1 sporadic patient. Clinical assessment included history, physical examination, and brain MRI. Linkage analysis was performed separately on the 2 sets of sib pairs using single nucleotide polymorphism microarrays, followed by analysis of the intersection of the regions. Exome sequencing was performed on 1 affected patient with variant filtering and prioritization undertaken using these intersected regions. RESULTS: Using a combination of sequencing technologies, we identified compound heterozygous mutations in HSD17B4 in all 5 affected patients. In all 3 families, peroxisomal D-bifunctional protein (DBP) deficiency was caused by compound heterozygosity for 1 nonsense/deletion mutation and 1 missense mutation. CONCLUSIONS: We describe 5 patients with juvenile DBP deficiency from 3 different families, bringing the total number of reported patients to 14, from 8 families. This report broadens and consolidates the phenotype associated with juvenile DBP deficiency.

18.
Eur J Hum Genet ; 13(1): 26-33, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15292920

ABSTRACT

Mitochondrial mutations have previously been reported anecdotally in families with maternally inherited, nonsyndromic hearing impairment. To ascertain the contribution of mitochondrial mutations to postlingual but early-onset, nonsyndromic hearing impairment, we screened patients collected from within two different populations (southern Italy and UK) for previously reported mtDNA mutations associated with hearing disorders. Primer extension (SNP analysis) was used to screen for specific mutations, revealing cases of heteroplasmy and its extent. The most frequently implicated tRNA genes, Leu(UUR) and Ser(UCN), were also sequenced in all Italian patients. All tRNA genes were sequenced in those UK patients showing the clearest likelihood of maternal inheritance. Causative mtDNA mutations were found in approximately 5% of patients in both populations, representing almost 10% of cases that were clearly familial. Age of onset, where known, was generally before adulthood, and hearing loss was typically progressive. Haplogroup analysis revealed a possible excess of haplogroup cluster HV in the patients, compared with population controls, but of borderline statistical significance. In contrast, we did not find any of the previously reported mtDNA mutations, nor a significant deviation from haplogroup cluster frequencies typical of the control population, in patients with late adult-onset hearing loss (age-related hearing impairment) from the UK or Finland.


Subject(s)
DNA, Mitochondrial/genetics , Hearing Loss/genetics , Language Disorders/genetics , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Age of Onset , Aged , Aged, 80 and over , DNA Mutational Analysis , Female , Finland , Haplotypes/genetics , Hearing Loss/epidemiology , Humans , Italy , Language Disorders/epidemiology , Male , Middle Aged , United Kingdom
19.
Neurology ; 84(20): 2029-32, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25878179

ABSTRACT

OBJECTIVE: To define causative somatic mutations in resected brain tissue from an infant with intractable epilepsy secondary to hemispheric cortical dysplasia. METHODS: Whole-exome sequencing was conducted on genomic DNA derived from both resected brain tissue and peripheral blood leukocytes. Comparison of the brain vs blood sequencing results was performed using bioinformatic methods designed to detect low-frequency genetic variation between tissue pairs. RESULTS: Histopathology of the resected tissue showed dyslamination and dysmorphic neurons, but no balloon cells, consistent with focal cortical dysplasia type IIa. mTOR activation was observed by immunohistochemistry in the dysplasia. A missense mutation (c.4487T>G; p.W1456G) was detected in the FAT domain of MTOR in DNA from the dysplasia but not in lymphocytes. The mutation is predicted damaging (i.e., leading to mTOR activation) and was observed as a low-level mosaic with 8% of cells being heterozygous for the variant. CONCLUSIONS: We report the novel finding of an MTOR mutation associated with nonsyndromic cortical dysplasia. Somatic-specific mutations in MTOR and related genes should be considered in a broader spectrum of patients with hemispheric malformations and more restricted forms of cortical dysplasia.


Subject(s)
Malformations of Cortical Development/genetics , Malformations of Cortical Development/pathology , Mutation, Missense , TOR Serine-Threonine Kinases/genetics , Brain/pathology , Epilepsy/etiology , Humans , Infant , Magnetic Resonance Imaging , Male , Malformations of Cortical Development/complications , Neurons/pathology , Positron-Emission Tomography
20.
Ann Clin Transl Neurol ; 2(5): 575-80, 2015 May.
Article in English | MEDLINE | ID: mdl-26000329

ABSTRACT

Whole-exome sequencing of two brothers with drug-resistant, early-onset, focal epilepsy secondary to extensive type IIA focal cortical dysplasia identified a paternally inherited, nonsense variant of DEPDC5 (c.C1663T, p.Arg555*). This variant has previously been reported to cause familial focal epilepsy with variable foci in patients with normal brain imaging. Immunostaining of resected brain tissue from both brothers demonstrated mammalian target of rapamycin (mTOR) activation. This report shows the histopathological features of cortical dysplasia associated with a DEPDC5 mutation, confirms mTOR dysregulation in the malformed tissue and expands the spectrum of neurological manifestations of DEPDC5 mutations to include severe phenotypes with large areas of cortical malformation.

SELECTION OF CITATIONS
SEARCH DETAIL