Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Am J Med Genet A ; : e63656, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760879

ABSTRACT

KIF1A-related disorders (KRDs) encompass recessive and dominant variants with wide clinical variability. Recent genetic investigations have expanded the clinical phenotypes of heterozygous KIF1A variants. However, there have been a few long-term observational studies of patients with heterozygous KIF1A variants. A retrospective chart review of consecutive patients diagnosed with spastic paraplegia at Miyagi Children's Hospital from 2016 to 2020 identified six patients with heterozygous KIF1A variants. To understand the long-term changes in clinical symptoms, we examined these patients in terms of their characteristics, clinical symptoms, results of electrophysiological and neuroimaging studies, and genetic testing. The median follow-up period was 30 years (4-44 years). This long-term observational study showed that early developmental delay and equinus gait, or unsteady gait, are the first signs of disease onset, appearing with the commencement of independent walking. In addition, later age-related progression was observed in spastic paraplegia, and the appearance of axonal neuropathy and reduced visual acuity were characteristic features of the late disease phenotype. Brain imaging showed age-related progression of cerebellar atrophy and the appearance of hyperintensity of optic radiation on T2WI and FLAIR imaging. Long-term follow-up revealed a pattern of steady progression and a variety of clinical symptoms, including spastic paraplegia, peripheral neuropathy, reduced visual acuity, and some degree of cerebellar ataxia. Clinical variability between patients was observed to some extent, and therefore, further studies are required to determine the phenotype-genotype correlation.

2.
Nat Commun ; 12(1): 2107, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833240

ABSTRACT

Vacuolar H+-ATPases (V-ATPases) transport protons across cellular membranes to acidify various organelles. ATP6V0A1 encodes the a1-subunit of the V0 domain of V-ATPases, which is strongly expressed in neurons. However, its role in brain development is unknown. Here we report four individuals with developmental and epileptic encephalopathy with ATP6V0A1 variants: two individuals with a de novo missense variant (R741Q) and the other two individuals with biallelic variants comprising one almost complete loss-of-function variant and one missense variant (A512P and N534D). Lysosomal acidification is significantly impaired in cell lines expressing three missense ATP6V0A1 mutants. Homozygous mutant mice harboring human R741Q (Atp6v0a1R741Q) and A512P (Atp6v0a1A512P) variants show embryonic lethality and early postnatal mortality, respectively, suggesting that R741Q affects V-ATPase function more severely. Lysosomal dysfunction resulting in cell death, accumulated autophagosomes and lysosomes, reduced mTORC1 signaling and synaptic connectivity, and lowered neurotransmitter contents of synaptic vesicles are observed in the brains of Atp6v0a1A512P/A512P mice. These findings demonstrate the essential roles of ATP6V0A1/Atp6v0a1 in neuronal development in terms of integrity and connectivity of neurons in both humans and mice.


Subject(s)
Brain Diseases/genetics , Brain/growth & development , Neurons/physiology , Neurotransmitter Agents/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Animals , Autophagosomes/pathology , Brain Mapping/methods , Cathepsin D/metabolism , Cell Line , HEK293 Cells , Humans , Loss of Function Mutation/genetics , Lysosomes/pathology , Magnetic Resonance Imaging/methods , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mutation, Missense/genetics , Neurons/cytology , Synaptic Vesicles/pathology
3.
Nat Commun ; 10(1): 2506, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31175295

ABSTRACT

Although there are many known Mendelian genes linked to epileptic or developmental and epileptic encephalopathy (EE/DEE), its genetic architecture is not fully explained. Here, we address this incompleteness by analyzing exomes of 743 EE/DEE cases and 2366 controls. We observe that damaging ultra-rare variants (dURVs) unique to an individual are significantly overrepresented in EE/DEE, both in known EE/DEE genes and the other non-EE/DEE genes. Importantly, enrichment of dURVs in non-EE/DEE genes is significant, even in the subset of cases with diagnostic dURVs (P = 0.000215), suggesting oligogenic contribution of non-EE/DEE gene dURVs. Gene-based analysis identifies exome-wide significant (P = 2.04 × 10-6) enrichment of damaging de novo mutations in NF1, a gene primarily linked to neurofibromatosis, in infantile spasm. Together with accumulating evidence for roles of oligogenic or modifier variants in severe neurodevelopmental disorders, our results highlight genetic complexity in EE/DEE, and indicate that EE/DEE is not an aggregate of simple Mendelian disorders.


Subject(s)
Genetic Variation , Spasms, Infantile/genetics , Adaptor Proteins, Vesicular Transport/genetics , Asian People/genetics , Case-Control Studies , DNA (Cytosine-5-)-Methyltransferases/genetics , Epilepsies, Myoclonic/genetics , Guanine Nucleotide Exchange Factors/genetics , Humans , Infant , Japan , Lennox Gastaut Syndrome/genetics , Logistic Models , Mutation , Neurofibromin 1/genetics , Polymorphism, Single Nucleotide , Principal Component Analysis , TRPM Cation Channels/genetics , Exome Sequencing
4.
Mol Genet Genomic Med ; 7(7): e00698, 2019 07.
Article in English | MEDLINE | ID: mdl-31106992

ABSTRACT

BACKGROUND: The tricarboxylic acid (TCA) cycle is a sequence of catabolic reactions within the mitochondrial matrix, and is a central pathway for cellular energy metabolism. Genetic defects affecting the TCA cycle are known to cause severe multisystem disorders. METHODS: We performed whole exome sequencing of genomic DNA of a patient with progressive cerebellar and cerebral atrophy, hypotonia, ataxia, seizure disorder, developmental delay, ophthalmological abnormalities and hearing loss. We also performed biochemical studies using patient fibroblasts. RESULTS: We identified new compound heterozygous mutations (c.1534G > A, p.Asp512Asn and c.1997G > C, p.Gly666Ala) in ACO2, which encodes aconitase 2, a component of the TCA cycle. In patient fibroblasts, the aconitase activity was reduced to 15% of that of the control, and the aconitase 2 level decreased to 36% of that of the control. As such a decrease in aconitase 2 in patient fibroblasts was partially restored by proteasome inhibition, mutant aconitase 2 was suggested to be relatively unstable and rapidly degraded after being synthesized. In addition, the activity of the father-derived variant of aconitase 2 (p.Gly666Ala), which had a mutation near the active center, was 55% of that of wild-type. CONCLUSION: The marked reduction of aconitase activity in patient fibroblasts was due to the combination of decreased aconitase 2 amount and activity due to mutations. Reduced aconitase activity directly suppresses the TCA cycle, resulting in mitochondrial dysfunction, which may lead to symptoms similar to those observed in mitochondrial diseases.


Subject(s)
Aconitate Hydratase/genetics , Brain Diseases/genetics , Cerebellum/pathology , Cerebrum/pathology , Mutation , Aconitate Hydratase/metabolism , Atrophy/genetics , Atrophy/pathology , Brain Diseases/pathology , Cells, Cultured , Cerebellum/metabolism , Cerebrum/metabolism , Child, Preschool , Female , Fibroblasts/metabolism , HEK293 Cells , Heterozygote , Humans
5.
J Med Genet ; 56(6): 396-407, 2019 06.
Article in English | MEDLINE | ID: mdl-30842224

ABSTRACT

BACKGROUND: Rett syndrome (RTT) is a characteristic neurological disease presenting with regressive loss of neurodevelopmental milestones. Typical RTT is generally caused by abnormality of methyl-CpG binding protein 2 (MECP2). Our objective to investigate the genetic landscape of MECP2-negative typical/atypical RTT and RTT-like phenotypes using whole exome sequencing (WES). METHODS: We performed WES on 77 MECP2-negative patients either with typical RTT (n=11), atypical RTT (n=22) or RTT-like phenotypes (n=44) incompatible with the RTT criteria. RESULTS: Pathogenic or likely pathogenic single-nucleotide variants in 28 known genes were found in 39 of 77 (50.6%) patients. WES-based CNV analysis revealed pathogenic deletions involving six known genes (including MECP2) in 8 of 77 (10.4%) patients. Overall, diagnostic yield was 47 of 77 (61.0 %). Furthermore, strong candidate variants were found in four novel genes: a de novo variant in each of ATPase H+ transporting V0 subunit A1 (ATP6V0A1), ubiquitin-specific peptidase 8 (USP8) and microtubule-associated serine/threonine kinase 3 (MAST3), as well as biallelic variants in nuclear receptor corepressor 2 (NCOR2). CONCLUSIONS: Our study provides a new landscape including additional genetic variants contributing to RTT-like phenotypes, highlighting the importance of comprehensive genetic analysis.


Subject(s)
Exome Sequencing , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Phenotype , Rett Syndrome/diagnosis , Rett Syndrome/genetics , Computational Biology/methods , DNA Copy Number Variations , Gene Ontology , Gene Regulatory Networks , Genetic Association Studies/methods , Humans , Methyl-CpG-Binding Protein 2/genetics , Polymorphism, Single Nucleotide
6.
Brain ; 141(6): 1703-1718, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29668857

ABSTRACT

V-type proton (H+) ATPase (v-ATPase) is a multi-subunit proton pump that regulates pH homeostasis in all eukaryotic cells; in neurons, v-ATPase plays additional and unique roles in synapse function. Through whole exome sequencing, we identified de novo heterozygous mutations (p.Pro27Arg, p.Asp100Tyr, p.Asp349Asn, p.Asp371Gly) in ATP6V1A, encoding the A subunit of v-ATPase, in four patients with developmental encephalopathy with epilepsy. Early manifestations, observed in all patients, were developmental delay and febrile seizures, evolving to encephalopathy with profound delay, hypotonic/dyskinetic quadriparesis and intractable multiple seizure types in two patients (p.Pro27Arg, p.Asp100Tyr), and to moderate delay with milder epilepsy in the other two (p.Asp349Asn, p.Asp371Gly). Modelling performed on the available prokaryotic and eukaryotic structures of v-ATPase predicted p.Pro27Arg to perturb subunit interaction, p.Asp100Tyr to cause steric hindrance and destabilize protein folding, p.Asp349Asn to affect the catalytic function and p.Asp371Gly to impair the rotation process, necessary for proton transport. We addressed the impact of p.Asp349Asn and p.Asp100Tyr mutations on ATP6V1A expression and function by analysing ATP6V1A-overexpressing HEK293T cells and patients' lymphoblasts. The p.Asp100Tyr mutant was characterized by reduced expression due to increased degradation. Conversely, no decrease in expression and clearance was observed for p.Asp349Asn. In HEK293T cells overexpressing either pathogenic or control variants, p.Asp349Asn significantly increased LysoTracker® fluorescence with no effects on EEA1 and LAMP1 expression. Conversely, p.Asp100Tyr decreased both LysoTracker® fluorescence and LAMP1 levels, leaving EEA1 expression unaffected. Both mutations decreased v-ATPase recruitment to autophagosomes, with no major impact on autophagy. Experiments performed on patients' lymphoblasts using the LysoSensor™ probe revealed lower pH of endocytic organelles for p.Asp349Asn and a reduced expression of LAMP1 with no effect on the pH for p.Asp100Tyr. These data demonstrate gain of function for p.Asp349Asn characterized by an increased proton pumping in intracellular organelles, and loss of function for p.Asp100Tyr with decreased expression of ATP6V1A and reduced levels of lysosomal markers. We expressed p.Asp349Asn and p.Asp100Tyr in rat hippocampal neurons and confirmed significant and opposite effects in lysosomal labelling. However, both mutations caused a similar defect in neurite elongation accompanied by loss of excitatory inputs, revealing that altered lysosomal homeostasis markedly affects neurite development and synaptic connectivity. This study provides evidence that de novo heterozygous ATP6V1A mutations cause a developmental encephalopathy with a pathomechanism that involves perturbations of lysosomal homeostasis and neuronal connectivity, uncovering a novel role for v-ATPase in neuronal development.


Subject(s)
Brain Diseases/genetics , Epilepsy/genetics , Mutation/genetics , Vacuolar Proton-Translocating ATPases/genetics , Adolescent , Animals , Brain/diagnostic imaging , Brain Diseases/complications , Brain Diseases/pathology , Cells, Cultured , Child , Cohort Studies , Epilepsy/complications , Epilepsy/pathology , Female , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomes/metabolism , Lysosomes/pathology , Male , Models, Molecular , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Rats , Synapses/metabolism , Synapses/pathology , Vacuolar Proton-Translocating ATPases/metabolism , Vesicular Transport Proteins/metabolism , Exome Sequencing
7.
Ann Clin Transl Neurol ; 5(3): 280-296, 2018 03.
Article in English | MEDLINE | ID: mdl-29560374

ABSTRACT

Objective: α (CAMK2A) and ß (CAMK2B) isoforms of Calcium/calmodulin-dependent protein kinase II (CaMKII) play a pivotal role in neuronal plasticity and in learning and memory processes in the brain. Here, we explore the possible involvement of α- and ß-CaMKII variants in neurodevelopmental disorders. Methods: Whole-exome sequencing was performed for 976 individuals with intellectual disability, developmental delay, and epilepsy. The effect of CAMK2A and CAMK2B variants on CaMKII structure and firing of neurons was evaluated by computational structural analysis, immunoblotting, and electrophysiological analysis. Results: We identified a total of five de novo CAMK2A and CAMK2B variants in three and two individuals, respectively. Seizures were common to three individuals with CAMK2A variants. Using a minigene splicing assay, we demonstrated that a splice site variant caused skipping of exon 11 leading to an in-frame deletion of the regulatory segment of CaMKII α. By structural analysis, four missense variants are predicted to impair the interaction between the kinase domain and the regulatory segment responsible for the autoinhibition of its kinase activity. The Thr286/Thr287 phosphorylation as a result of release from autoinhibition was increased in three mutants when the mutants were stably expressed in Neuro-2a neuroblastoma cells. Expression of a CaMKII α mutant in primary hippocampal neurons significantly increased A-type K+ currents, which facilitated spike repolarization of single action potentials. Interpretation: Our data highlight the importance of CaMKII α and CaMKII ß and their autoinhibitory regulation in human brain function, and suggest the enhancement of A-type K+ currents as a possible pathophysiological basis.

8.
Brain Dev ; 40(5): 406-409, 2018 May.
Article in English | MEDLINE | ID: mdl-29519750

ABSTRACT

We present a unique 11-year-old girl showing clinical features of Rett-related disorder with distinctive facial features and multiple congenital anomalies including ocular hypertelorism, arched eyebrows, a broad nose, dental anomalies, congenital heart disease, truncal obesity, and epilepsy. A novel de novo mutation in histone deacetylase 8 (HDAC8) (c.652G > T, p.Gly218Cys) was confirmed by whole exome sequencing and Sanger sequencing. X-chromosome inactivation analysis on DNA isolated from peripheral blood lymphocytes revealed a completely skewed pattern associated with an inactive maternal allele. Late clinical loss of acquired purposeful hand movements and psychomotor deterioration may be a feature of Rett-related disorder, while distinctive facial features and multiple congenital anomalies are reminiscent of Cornelia de Lange syndrome.


Subject(s)
Histone Deacetylases/genetics , Histone Deacetylases/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Rett Syndrome/genetics , Abnormalities, Multiple/genetics , Alleles , Child , De Lange Syndrome/genetics , Female , Heterozygote , Humans , Japan , Mutation , Pedigree , Phenotype , Exome Sequencing/methods
9.
Am J Hum Genet ; 102(2): 321-329, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29394991

ABSTRACT

Early-onset epileptic encephalopathies, including West syndrome (WS), are a group of neurological disorders characterized by developmental impairments and intractable seizures from early infancy. We have now identified biallelic CNPY3 variants in three individuals with WS; these include compound-heterozygous missense and frameshift variants in a family with two affected siblings (individuals 1 and 2) and a homozygous splicing variant in a consanguineous family (individual 3). All three individuals showed hippocampal malrotation. In individuals 1 and 2, electroencephalography (EEG) revealed characteristic fast waves and diffuse sharp- and slow-wave complexes. The fast waves were clinically associated with seizures. CNPY3 encodes a co-chaperone in the endoplasmic reticulum and regulates the subcellular distribution and responses of multiple Toll-like receptors. The amount of CNPY3 in lymphoblastoid cells derived from individuals 1 and 2 was severely lower than that in control cells. Cnpy3-knockout mice exhibited spastic or dystonic features under resting conditions and hyperactivity and anxiolytic behavior during the open field test. Also, their resting EEG showed enhanced activity in the fast beta frequency band (20-35 Hz), which could mimic the fast waves in individuals 1 and 2. These data suggest that CNPY3 and Cnpy3 perform essential roles in brain function in addition to known Toll-like receptor-dependent immune responses.


Subject(s)
Molecular Chaperones/genetics , Mutation , Seizures/genetics , Spasms, Infantile/genetics , Adolescent , Amino Acid Sequence , Animals , Child , Consanguinity , Electroencephalography , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Family , Female , Gene Expression , Heterozygote , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Infant , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Seizures/diagnostic imaging , Seizures/physiopathology , Sequence Alignment , Sequence Homology, Amino Acid , Siblings , Spasms, Infantile/diagnostic imaging , Spasms, Infantile/physiopathology
10.
J Hum Genet ; 63(3): 263-270, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29339779

ABSTRACT

Misato 1, mitochondrial distribution and morphology regulator (encoded by the MSTO1 gene), is involved in mitochondrial distribution and morphology. Recently, MSTO1 mutations have been shown to cause clinical manifestations suggestive of mitochondrial dysfunction, such as muscle weakness, short stature, motor developmental delay, and cerebellar atrophy. Both autosomal dominant and recessive modes of inheritance have been suggested. We performed whole-exome sequencing in two unrelated patients showing cerebellar atrophy, intellectual disability, and pigmentary retinopathy. Three novel mutations were identified: c.836 G > A (p.Arg279His), c.1099-1 G > A (p.Val367Trpfs*2), and c.79 C > T (p.Gln27*). Both patients had compound heterozygous mutations with a combination of protein-truncation mutation and missense mutation, the latter shared by them both. This survey of two patients with recessive and novel MSTO1 mutations provides additional clinical and genetic information on the pathogenicity of MSTO1 in humans.


Subject(s)
Cell Cycle Proteins/genetics , Cerebellar Diseases/diagnosis , Cerebellar Diseases/genetics , Cytoskeletal Proteins/genetics , Genes, Recessive , Mutation , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Adolescent , Age of Onset , Alleles , Atrophy , Cell Line , Child , Child, Preschool , DNA Mutational Analysis , Female , Genotype , Humans , Infant , Magnetic Resonance Imaging , Male , Pedigree , Phenotype , Whole Genome Sequencing
11.
Brain Dev ; 40(1): 53-57, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28728837

ABSTRACT

We report an 11-month-old boy with acetazolamide-responsive epileptic apnea and inherited glycosylphosphatidylinositol (GPI)-anchor deficiency who presented with decreased serum alkaline phosphatase associated with compound PIGT mutations. The patient exhibited congenital anomalies, severe intellectual disability, and seizures, including epileptic apnea with epileptiform discharges from bilateral temporal areas. Brain magnetic resonance imaging revealed delayed myelination and progressive atrophy of the brainstem, cerebellum, and cerebrum. Whole-exome sequencing revealed compound heterozygous mutations in PIGT (c.250G>T, p.Glu84X and c.1096G>T, p.Gly366Trp), which encodes a subunit of the GPI transamidase complex. Flow cytometry revealed decreased expression of CD16 (a GPI anchor protein) on granulocytes, supporting the putative pathogenicity of the mutations. Phenobarbital, clonazepam, and potassium bromide decreased the frequency of tonic seizure and acetazolamide decreased epileptic apnea. To our knowledge, this is the first reported case of intractable seizures accompanied by epileptic apnea associated with GPI anchor deficiency and a compound PIGT mutation.


Subject(s)
Apnea/genetics , Epilepsy/genetics , Glycosylphosphatidylinositols/deficiency , Abnormalities, Multiple/genetics , Acyltransferases/genetics , Acyltransferases/metabolism , Apnea/metabolism , Atrophy , Developmental Disabilities/genetics , Epilepsy/metabolism , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Humans , Infant , Intellectual Disability/genetics , Male , Muscle Hypotonia/genetics , Mutation , Seizures/genetics
12.
J Neurol Sci ; 376: 7-12, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28431631

ABSTRACT

PURPOSE: Little is known regarding neuroimaging-genotype correlations in Joubert syndrome (JBTS). To elucidate one of these correlations, we investigated the neuroimaging findings of JBTS patients with C5orf42 mutations. MATERIALS AND METHODS: Neuroimaging findings in five JBTS patients with C5orf42 mutations were retrospectively assessed with regard to the infratentorial and supratentorial structures on T1-magnetization prepared rapid gradient echo (MPRAGE), T2-weighted images, and color-coded fractional anisotropy (FA) maps; the findings were compared to those in four JBTS patients with mutations in other genes (including three with AHI1 and one with TMEM67 mutations). RESULTS: In C5orf42-mutant patients, the infratentorial magnetic resonance (MR) images showed normal or minimally thickened and minimally elongated superior cerebellar peduncles (SCP), normal or minimally deepened interpeduncular fossa (IF), and mild vermian hypoplasia (VH). However, in other patients, all had severe abnormalities in the SCP and IF, and moderate to marked VH. Supratentorial abnormalities were found in one individual in other JBTS. In JBTS with all mutations, color-coded FA maps showed the absence of decussation of the SCP (DSCP). CONCLUSION: The morphological neuroimaging findings in C5orf42-mutant JBTS were distinctly mild and made diagnosis difficult. However, the absence of DSCP on color-coded FA maps may facilitate the diagnosis of JBTS.


Subject(s)
Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/genetics , Brain/diagnostic imaging , Cerebellum/abnormalities , Eye Abnormalities/diagnostic imaging , Eye Abnormalities/genetics , Kidney Diseases, Cystic/diagnostic imaging , Kidney Diseases, Cystic/genetics , Magnetic Resonance Imaging , Membrane Proteins/genetics , Mutation , Retina/abnormalities , Cerebellum/diagnostic imaging , Child , Child, Preschool , Diagnosis, Differential , Diffusion Tensor Imaging , Female , Humans , Infant , Male , Organ Size , Retina/diagnostic imaging , Retrospective Studies
13.
J Hum Genet ; 62(6): 653-655, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28148925

ABSTRACT

Cerebral, ocular, dental, auricular, skeletal (CODAS) syndrome is a rare autosomal recessive multisystem disorder caused by mutations in LONP1. It is characterized by intellectual disability, cataracts, delayed tooth eruption, malformed auricles and skeletal abnormalities. We performed whole-exome sequencing on a 12-year-old Japanese male with severe intellectual disability, congenital bilateral cataracts, spasticity, hypotonia with motor regression and progressive cerebellar atrophy with hyperintensity of the cerebellar cortex on T2-weighted images. We detected compound heterozygous mutation in LONP1. One allele contained a paternally inherited frameshift mutation (p.Ser100Glnfs*46). The other allele contained a maternally inherited missense mutation (p.Arg786Trp), which was predicted to be pathogenic by web-based prediction tools. The two mutations were not found in Exome Variant Server or our 575 in-house control exomes. Some features were not consistent with CODAS syndrome but overlapped with Marinesco-Sjögren syndrome, a multisystem disorder caused by a mutation in SIL1. An atypical mutation site may result in atypical presentation of the LONP1 mutation.


Subject(s)
ATP-Dependent Proteases/genetics , Craniofacial Abnormalities/genetics , Eye Abnormalities/genetics , Growth Disorders/genetics , Hip Dislocation, Congenital/genetics , Intellectual Disability/genetics , Mitochondrial Proteins/genetics , Osteochondrodysplasias/genetics , Spinocerebellar Degenerations/genetics , Tooth Abnormalities/genetics , Child , Craniofacial Abnormalities/physiopathology , Exome/genetics , Eye Abnormalities/physiopathology , Frameshift Mutation/genetics , Genetic Predisposition to Disease , Growth Disorders/physiopathology , Hip Dislocation, Congenital/physiopathology , Humans , Intellectual Disability/physiopathology , Male , Osteochondrodysplasias/physiopathology , Protein Domains/genetics , Spinocerebellar Degenerations/physiopathology , Tooth Abnormalities/physiopathology
14.
Am J Hum Genet ; 99(4): 950-961, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27666374

ABSTRACT

We describe four families with affected siblings showing unique clinical features: early-onset (before 1 year of age) progressive diffuse brain atrophy with regression, postnatal microcephaly, postnatal growth retardation, muscle weakness/atrophy, and respiratory failure. By whole-exome sequencing, we identified biallelic TBCD mutations in eight affected individuals from the four families. TBCD encodes TBCD (tubulin folding co-factor D), which is one of five tubulin-specific chaperones playing a pivotal role in microtubule assembly in all cells. A total of seven mutations were found: five missense mutations, one nonsense, and one splice site mutation resulting in a frameshift. In vitro cell experiments revealed the impaired binding between most mutant TBCD proteins and ARL2, TBCE, and ß-tubulin. The in vivo experiments using olfactory projection neurons in Drosophila melanogaster indicated that the TBCD mutations caused loss of function. The wide range of clinical severity seen in this neurodegenerative encephalopathy may result from the residual function of mutant TBCD proteins. Furthermore, the autopsied brain from one deceased individual showed characteristic neurodegenerative findings: cactus and somatic sprout formations in the residual Purkinje cells in the cerebellum, which are also seen in some diseases associated with mitochondrial impairment. Defects of microtubule formation caused by TBCD mutations may underlie the pathomechanism of this neurodegenerative encephalopathy.


Subject(s)
Alleles , Brain Diseases/genetics , Microtubule-Associated Proteins/genetics , Mutation/genetics , Neurodegenerative Diseases/genetics , Adolescent , Age of Onset , Amino Acid Sequence , Animals , Brain Diseases/pathology , Brain Diseases/physiopathology , Child , Child, Preschool , Drosophila melanogaster/genetics , Exome , Female , Frameshift Mutation/genetics , GTP-Binding Proteins/metabolism , Humans , Infant , Infant, Newborn , Male , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Pedigree , RNA Splice Sites/genetics , Tubulin/metabolism , Young Adult
15.
Sci Rep ; 6: 30072, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27436767

ABSTRACT

Epilepsy of infancy with migrating focal seizures (EIMFS) is one of the early-onset epileptic syndromes characterized by migrating polymorphous focal seizures. Whole exome sequencing (WES) in ten sporadic and one familial case of EIMFS revealed compound heterozygous SLC12A5 (encoding the neuronal K(+)-Cl(-) co-transporter KCC2) mutations in two families: c.279 + 1G > C causing skipping of exon 3 in the transcript (p.E50_Q93del) and c.572 C >T (p.A191V) in individuals 1 and 2, and c.967T > C (p.S323P) and c.1243 A > G (p.M415V) in individual 3. Another patient (individual 4) with migrating multifocal seizures and compound heterozygous mutations [c.953G > C (p.W318S) and c.2242_2244del (p.S748del)] was identified by searching WES data from 526 patients and SLC12A5-targeted resequencing data from 141 patients with infantile epilepsy. Gramicidin-perforated patch-clamp analysis demonstrated strongly suppressed Cl(-) extrusion function of E50_Q93del and M415V mutants, with mildly impaired function of A191V and S323P mutants. Cell surface expression levels of these KCC2 mutants were similar to wildtype KCC2. Heterologous expression of two KCC2 mutants, mimicking the patient status, produced a significantly greater intracellular Cl(-) level than with wildtype KCC2, but less than without KCC2. These data clearly demonstrated that partially disrupted neuronal Cl(-) extrusion, mediated by two types of differentially impaired KCC2 mutant in an individual, causes EIMFS.


Subject(s)
Mutation , Seizures/pathology , Seizures/physiopathology , Symporters/genetics , Symporters/metabolism , Adult , Biological Transport , Child, Preschool , Chlorides/metabolism , Female , Humans , Infant , Male , Polymorphism, Single Nucleotide , Sequence Deletion , Young Adult
16.
Brain Dev ; 38(10): 959-963, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27371992

ABSTRACT

BACKGROUND: Aromatic l-amino acid decarboxylase (AADC) deficiency is an autosomal recessive disorder, caused by defects in the DDC gene. AADC catalyzes the synthesis of the neurotransmitters dopamine and serotonin from l-dopa and 5-HT respectively. Most patients are bed ridden for life, with little response to treatment. We now report one female patient who improved her motor and cognitive function after being prescribed a MAO-B inhibitor. CASE: A five years old female presented with the typical clinical features of AADC deficiency. She was floppy, with no head control, had intermittent limb dystonia, and an upward deviation of the eyes (oculogyric crisis). This patient possessed compound heterozygous mutations in DDC (p.Trp105Cys, p.Pro129Ser), with a CSF draw indicating abnormal patterns of biogenic amine metabolites, compatible with AADC deficiency. RESULTS: After her diagnosis at 3years of age, medication with levodopa and vitamin B6 failed to show any efficacy. Subsequent administration with a MAO-B inhibitor improved her psychomotor functions to the extent that at 5years of age she could walk several meters with support. CONCLUSION: Our analyses of chemical findings, together with in silico structure predictions, lead us to hypothesize that this patient retained some AADC activity. In these cases, accurate diagnosis and early treatment should improve patient outcome.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Aromatic-L-Amino-Acid Decarboxylases/deficiency , Monoamine Oxidase Inhibitors/therapeutic use , Neuromuscular Agents/therapeutic use , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/physiopathology , Aromatic-L-Amino-Acid Decarboxylases/genetics , Child, Preschool , DNA Mutational Analysis , Female , Humans , Motor Activity/drug effects , Motor Activity/genetics , Motor Activity/physiology , Nootropic Agents/therapeutic use , Pedigree , Protein Conformation , Treatment Outcome
17.
Clin Case Rep ; 4(5): 509-12, 2016 May.
Article in English | MEDLINE | ID: mdl-27190617

ABSTRACT

Mucolipidosis type IV (MLIV) is a rare neurodegenerative disorder characterized by severe psychomotor delay and visual impairment. We report the brain pathology in the first Japanese patient of MLIV with a novel homozygous missense mutation in MCOLN1. We detected the localized increase in p62-reactive astrocytes in the basal ganglia.

18.
Epilepsia ; 57(4): 566-73, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26918889

ABSTRACT

OBJECTIVE: GABRA1 mutations have been identified in patients with familial juvenile myoclonic epilepsy, sporadic childhood absence epilepsy, and idiopathic familial generalized epilepsy. In addition, de novo GABRA1 mutations were recently reported in a patient with infantile spasms and four patients with Dravet syndrome. Those reports suggest that GABRA1 mutations are associated with infantile epilepsy including early onset epileptic encephalopathies. In this study, we searched for GABRA1 mutations in patients with infantile epilepsy to investigate the phenotypic spectrum of GABRA1 mutations. METHODS: In total, 526 and 145 patients with infantile epilepsy were analyzed by whole-exome sequencing and GABRA1-targeted resequencing, respectively. RESULTS: We identified five de novo missense GABRA1 mutations in six unrelated patients. A p.R112Q mutation in the long extracellular N-terminus was identified in a patient with infantile epilepsy; p.P260L, p.M263T, and p.M263I in transmembrane spanning domain 1 (TM1) were identified in three unrelated patients with West syndrome and a patient with Ohtahara syndrome, respectively; and p.V287L in TM2 was identified in a patient with unclassified early onset epileptic encephalopathy. Four of these mutations have not been observed previously. SIGNIFICANCE: Our study suggests that de novo GABRA1 mutations can cause early onset epileptic encephalopathies, including Ohtahara syndrome and West syndrome.


Subject(s)
Mutation, Missense/genetics , Receptors, GABA-A/genetics , Spasms, Infantile/diagnosis , Spasms, Infantile/genetics , Amino Acid Sequence , Child , Child, Preschool , Electroencephalography , Female , Humans , Infant , Male , Molecular Sequence Data , Spasms, Infantile/physiopathology
19.
J Hum Genet ; 61(6): 527-31, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26888482

ABSTRACT

Cerebellar atrophy is recognized in various types of childhood neurological disorders with clinical and genetic heterogeneity. Genetic analyses such as whole exome sequencing are useful for elucidating the genetic basis of these conditions. Pathological recessive mutations in Sep (O-phosphoserine) tRNA:Sec (selenocysteine) tRNA synthase (SEPSECS) have been reported in a total of 11 patients with pontocerebellar hypoplasia type 2, progressive cerebellocerebral atrophy or progressive encephalopathy, yet detailed clinical features are limited to only four patients. We identified two new families with progressive cerebellar atrophy, and by whole exome sequencing detected biallelic SEPSECS mutations: c.356A>G (p.Asn119Ser) and c.77delG (p.Arg26Profs*42) in family 1, and c.356A>G (p.Asn119Ser) and c.467G>A (p.Arg156Gln) in family 2. Their development was slightly delayed regardless of normal brain magnetic resonance imaging (MRI) in infancy. The progression of clinical symptoms in these families is evidently slower than in previously reported cases, and the cerebellar atrophy milder by brain MRI, indicating that SEPSECS mutations are also involved in milder late-onset cerebellar atrophy.


Subject(s)
Alleles , Amino Acyl-tRNA Synthetases/genetics , Mutation , Olivopontocerebellar Atrophies/diagnosis , Olivopontocerebellar Atrophies/genetics , Adolescent , Amino Acid Substitution , Brain/cytology , Child , Child, Preschool , Exome , Female , Gene Frequency , Genotype , High-Throughput Nucleotide Sequencing , Humans , Infant , Magnetic Resonance Imaging , Male , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
20.
Sci Rep ; 5: 15199, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26477325

ABSTRACT

The voltage-gated Kv2.1 potassium channel encoded by KCNB1 produces the major delayed rectifier potassium current in pyramidal neurons. Recently, de novo heterozygous missense KCNB1 mutations have been identified in three patients with epileptic encephalopathy and a patient with neurodevelopmental disorder. However, the frequency of KCNB1 mutations in infantile epileptic patients and their effects on neuronal activity are yet unknown. We searched whole exome sequencing data of a total of 437 patients with infantile epilepsy, and found novel de novo heterozygous missense KCNB1 mutations in two patients showing psychomotor developmental delay and severe infantile generalized seizures with high-amplitude spike-and-wave electroencephalogram discharges. The mutation located in the channel voltage sensor (p.R306C) disrupted sensitivity and cooperativity of the sensor, while the mutation in the channel pore domain (p.G401R) selectively abolished endogenous Kv2 currents in transfected pyramidal neurons, indicating a dominant-negative effect. Both mutants inhibited repetitive neuronal firing through preventing production of deep interspike voltages. Thus KCNB1 mutations can be a rare genetic cause of infantile epilepsy, and insufficient firing of pyramidal neurons would disturb both development and stability of neuronal circuits, leading to the disease phenotypes.


Subject(s)
Epilepsy/genetics , Epilepsy/physiopathology , Membrane Potentials/genetics , Mutation , Neurons/metabolism , Shab Potassium Channels/genetics , Shab Potassium Channels/metabolism , Cell Line , Child , Child, Preschool , Electroencephalography , Epilepsy/diagnosis , Epilepsy/therapy , Exome , Female , Gene Expression , Genome-Wide Association Study , Genotype , High-Throughput Nucleotide Sequencing , Humans , Magnetic Resonance Imaging , Male , Phenotype , Potassium/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...