Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Am J Hum Genet ; 105(5): 1048-1056, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31668703

ABSTRACT

NTNG2 encodes netrin-G2, a membrane-anchored protein implicated in the molecular organization of neuronal circuitry and synaptic organization and diversification in vertebrates. In this study, through a combination of exome sequencing and autozygosity mapping, we have identified 16 individuals (from seven unrelated families) with ultra-rare homozygous missense variants in NTNG2; these individuals present with shared features of a neurodevelopmental disorder consisting of global developmental delay, severe to profound intellectual disability, muscle weakness and abnormal tone, autistic features, behavioral abnormalities, and variable dysmorphisms. The variants disrupt highly conserved residues across the protein. Functional experiments, including in silico analysis of the protein structure, in vitro assessment of cell surface expression, and in vitro knockdown, revealed potential mechanisms of pathogenicity of the variants, including loss of protein function and decreased neurite outgrowth. Our data indicate that appropriate expression of NTNG2 plays an important role in neurotypical development.


Subject(s)
GPI-Linked Proteins/genetics , Mutation, Missense/genetics , Netrins/genetics , Neurodevelopmental Disorders/genetics , Adolescent , Adult , Child , Child, Preschool , Exome/genetics , Female , Homozygote , Humans , Intellectual Disability/genetics , Male , Pedigree , Exome Sequencing/methods , Young Adult
2.
Nature ; 539(7628): 242-247, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27830782

ABSTRACT

Sensory stimuli drive the maturation and function of the mammalian nervous system in part through the activation of gene expression networks that regulate synapse development and plasticity. These networks have primarily been studied in mice, and it is not known whether there are species- or clade-specific activity-regulated genes that control features of brain development and function. Here we use transcriptional profiling of human fetal brain cultures to identify an activity-dependent secreted factor, Osteocrin (OSTN), that is induced by membrane depolarization of human but not mouse neurons. We find that OSTN has been repurposed in primates through the evolutionary acquisition of DNA regulatory elements that bind the activity-regulated transcription factor MEF2. In addition, we demonstrate that OSTN is expressed in primate neocortex and restricts activity-dependent dendritic growth in human neurons. These findings suggest that, in response to sensory input, OSTN regulates features of neuronal structure and function that are unique to primates.


Subject(s)
Evolution, Molecular , Muscle Proteins/metabolism , Neocortex/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Transcriptome , Animals , Base Sequence , Bone and Bones/metabolism , Dendrites/metabolism , Enhancer Elements, Genetic/genetics , Female , Humans , MEF2 Transcription Factors/metabolism , Macaca mulatta , Male , Mice , Molecular Sequence Data , Muscle Proteins/genetics , Muscles/metabolism , Neocortex/cytology , Neurons/cytology , Organ Specificity , Species Specificity , Transcription Factors/genetics
3.
Am J Hum Genet ; 100(3): 537-545, 2017 Mar 02.
Article in English | MEDLINE | ID: mdl-28190459

ABSTRACT

Congenital muscular dystrophies display a wide phenotypic and genetic heterogeneity. The combination of clinical, biochemical, and molecular genetic findings must be considered to obtain the precise diagnosis and provide appropriate genetic counselling. Here we report five individuals from four families presenting with variable clinical features including muscular dystrophy with a reduction in dystroglycan glycosylation, short stature, intellectual disability, and cataracts, overlapping both the dystroglycanopathies and Marinesco-Sjögren syndrome. Whole-exome sequencing revealed homozygous missense and compound heterozygous mutations in INPP5K in the affected members of each family. INPP5K encodes the inositol polyphosphate-5-phosphatase K, also known as SKIP (skeletal muscle and kidney enriched inositol phosphatase), which is highly expressed in the brain and muscle. INPP5K localizes to both the endoplasmic reticulum and to actin ruffles in the cytoplasm. It has been shown to regulate myoblast differentiation and has also been implicated in protein processing through its interaction with the ER chaperone HSPA5/BiP. We show that morpholino-mediated inpp5k loss of function in the zebrafish results in shortened body axis, microphthalmia with disorganized lens, microcephaly, reduced touch-evoked motility, and highly disorganized myofibers. Altogether these data demonstrate that mutations in INPP5K cause a congenital muscular dystrophy syndrome with short stature, cataracts, and intellectual disability.


Subject(s)
Muscular Dystrophies, Limb-Girdle/genetics , Phosphoric Monoester Hydrolases/genetics , Spinocerebellar Degenerations/genetics , Adolescent , Adult , Amino Acid Sequence , Animals , Brain/metabolism , Child , Disease Models, Animal , Dystroglycans/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Female , Genome-Wide Association Study , Glycosylation , Growth Disorders/genetics , Humans , Intellectual Disability/genetics , Male , Microcephaly/genetics , Muscle, Skeletal/metabolism , Mutation , Pedigree , Young Adult , Zebrafish/genetics
4.
Genome Res ; 27(8): 1323-1335, 2017 08.
Article in English | MEDLINE | ID: mdl-28630177

ABSTRACT

While next-generation sequencing has accelerated the discovery of human disease genes, progress has been largely limited to the "low hanging fruit" of mutations with obvious exonic coding or canonical splice site impact. In contrast, the lack of high-throughput, unbiased approaches for functional assessment of most noncoding variants has bottlenecked gene discovery. We report the integration of transcriptome sequencing (RNA-seq), which surveys all mRNAs to reveal functional impacts of variants at the transcription level, into the gene discovery framework for a unique human disease, microcephaly-micromelia syndrome (MMS). MMS is an autosomal recessive condition described thus far in only a single First Nations population and causes intrauterine growth restriction, severe microcephaly, craniofacial anomalies, skeletal dysplasia, and neonatal lethality. Linkage analysis of affected families, including a very large pedigree, identified a single locus on Chromosome 21 linked to the disease (LOD > 9). Comprehensive genome sequencing did not reveal any pathogenic coding or canonical splicing mutations within the linkage region but identified several nonconserved noncoding variants. RNA-seq analysis detected aberrant splicing in DONSON due to one of these noncoding variants, showing a causative role for DONSON disruption in MMS. We show that DONSON is expressed in progenitor cells of embryonic human brain and other proliferating tissues, is co-expressed with components of the DNA replication machinery, and that Donson is essential for early embryonic development in mice as well, suggesting an essential conserved role for DONSON in the cell cycle. Our results demonstrate the utility of integrating transcriptomics into the study of human genetic disease when DNA sequencing alone is not sufficient to reveal the underlying pathogenic mutation.


Subject(s)
Cell Cycle Proteins/genetics , DNA Replication , Microcephaly/genetics , Microcephaly/pathology , Mutation , Nuclear Proteins/genetics , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Transcriptome , Animals , Chromosome Mapping , Female , Genetic Linkage , Genomic Instability , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Mice, Knockout , Microcephaly/etiology , Osteochondrodysplasias/etiology , Pedigree , Pregnancy , RNA Splicing , Sequence Analysis, RNA , Whole Genome Sequencing
5.
Genet Med ; 22(6): 1040-1050, 2020 06.
Article in English | MEDLINE | ID: mdl-32103185

ABSTRACT

PURPOSE: The exocyst complex is a conserved protein complex that mediates fusion of intracellular vesicles to the plasma membrane and is implicated in processes including cell polarity, cell migration, ciliogenesis, cytokinesis, autophagy, and fusion of secretory vesicles. The essential role of these genes in human genetic disorders, however, is unknown. METHODS: We performed homozygosity mapping and exome sequencing of consanguineous families with recessively inherited brain development disorders. We modeled an EXOC7 splice variant in vitro and examined EXOC7 messenger RNA (mRNA) expression in developing mouse and human cortex. We modeled exoc7 loss-of-function in a zebrafish knockout. RESULTS: We report variants in exocyst complex members, EXOC7 and EXOC8, in a novel disorder of cerebral cortex development. In EXOC7, we identified four independent partial loss-of-function (LOF) variants in a recessively inherited disorder characterized by brain atrophy, seizures, and developmental delay, and in severe cases, microcephaly and infantile death. In EXOC8, we found a homozygous truncating variant in a family with a similar clinical disorder. We modeled exoc7 deficiency in zebrafish and found the absence of exoc7 causes microcephaly. CONCLUSION: Our results highlight the essential role of the exocyst pathway in normal cortical development and how its perturbation causes complex brain disorders.


Subject(s)
Brain Diseases , Microcephaly , Animals , Cell Proliferation/genetics , Homozygote , Humans , Mice , Microcephaly/genetics , Zebrafish/genetics
6.
Genet Med ; 21(3): 545-552, 2019 03.
Article in English | MEDLINE | ID: mdl-30214071

ABSTRACT

PURPOSE: Congenital microcephaly (CM) is an important birth defect with long term neurological sequelae. We aimed to perform detailed phenotypic and genomic analysis of patients with Mendelian forms of CM. METHODS: Clinical phenotyping, targeted or exome sequencing, and autozygome analysis. RESULTS: We describe 150 patients (104 families) with 56 Mendelian forms of CM. Our data show little overlap with the genetic causes of postnatal microcephaly. We also show that a broad definition of primary microcephaly -as an autosomal recessive form of nonsyndromic CM with severe postnatal deceleration of occipitofrontal circumference-is highly sensitive but has a limited specificity. In addition, we expand the overlap between primary microcephaly and microcephalic primordial dwarfism both clinically (short stature in >52% of patients with primary microcephaly) and molecularly (e.g., we report the first instance of CEP135-related microcephalic primordial dwarfism). We expand the allelic and locus heterogeneity of CM by reporting 37 novel likely disease-causing variants in 27 disease genes, confirming the candidacy of ANKLE2, YARS, FRMD4A, and THG1L, and proposing the candidacy of BPTF, MAP1B, CCNH, and PPFIBP1. CONCLUSION: Our study refines the phenotype of CM, expands its genetics heterogeneity, and informs the workup of children born with this developmental brain defect.


Subject(s)
Microcephaly/genetics , Microcephaly/physiopathology , Adult , Child , Child, Preschool , Dwarfism/genetics , Female , Genomics/methods , Genotype , Humans , Infant , Infant, Newborn , Male , Mutation/genetics , Pedigree , Phenotype , Exome Sequencing/methods
7.
Proc Natl Acad Sci U S A ; 113(38): E5598-607, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27601654

ABSTRACT

Mutations that cause neurological phenotypes are highly informative with regard to mechanisms governing human brain function and disease. We report autosomal recessive mutations in the enzyme glutamate pyruvate transaminase 2 (GPT2) in large kindreds initially ascertained for intellectual and developmental disability (IDD). GPT2 [also known as alanine transaminase 2 (ALT2)] is one of two related transaminases that catalyze the reversible addition of an amino group from glutamate to pyruvate, yielding alanine and α-ketoglutarate. In addition to IDD, all affected individuals show postnatal microcephaly and ∼80% of those followed over time show progressive motor symptoms, a spastic paraplegia. Homozygous nonsense p.Arg404* and missense p.Pro272Leu mutations are shown biochemically to be loss of function. The GPT2 gene demonstrates increasing expression in brain in the early postnatal period, and GPT2 protein localizes to mitochondria. Akin to the human phenotype, Gpt2-null mice exhibit reduced brain growth. Through metabolomics and direct isotope tracing experiments, we find a number of metabolic abnormalities associated with loss of Gpt2. These include defects in amino acid metabolism such as low alanine levels and elevated essential amino acids. Also, we find defects in anaplerosis, the metabolic process involved in replenishing TCA cycle intermediates. Finally, mutant brains demonstrate misregulated metabolites in pathways implicated in neuroprotective mechanisms previously associated with neurodegenerative disorders. Overall, our data reveal an important role for the GPT2 enzyme in mitochondrial metabolism with relevance to developmental as well as potentially to neurodegenerative mechanisms.


Subject(s)
Brain/growth & development , Mitochondria/enzymology , Nervous System Diseases/genetics , Transaminases/genetics , Amino Acid Sequence/genetics , Animals , Brain/metabolism , Brain/pathology , Citric Acid Cycle/genetics , Homozygote , Humans , Ketoglutaric Acids/metabolism , Mice , Mitochondria/pathology , Mutation, Missense , Nervous System Diseases/pathology , Phenotype , Pyruvic Acid/metabolism , Transaminases/metabolism
8.
Am J Hum Genet ; 96(5): 709-19, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25865492

ABSTRACT

Despite recent advances in understanding the genetic bases of microcephaly, a large number of cases of microcephaly remain unexplained, suggesting that many microcephaly syndromes and associated genes have yet to be identified. Here, we report mutations in PYCR2, which encodes an enzyme in the proline biosynthesis pathway, as the cause of a unique syndrome characterized by postnatal microcephaly, hypomyelination, and reduced cerebral white-matter volume. Linkage mapping and whole-exome sequencing identified homozygous mutations (c.355C>T [p.Arg119Cys] and c.751C>T [p.Arg251Cys]) in PYCR2 in the affected individuals of two consanguineous families. A lymphoblastoid cell line from one affected individual showed a strong reduction in the amount of PYCR2. When mutant cDNAs were transfected into HEK293FT cells, both variant proteins retained normal mitochondrial localization but had lower amounts than the wild-type protein, suggesting that the variant proteins were less stable. A PYCR2-deficient HEK293FT cell line generated by genome editing with the clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9 system showed that PYCR2 loss of function led to decreased mitochondrial membrane potential and increased susceptibility to apoptosis under oxidative stress. Morpholino-based knockdown of a zebrafish PYCR2 ortholog, pycr1b, recapitulated the human microcephaly phenotype, which was rescued by wild-type human PYCR2 mRNA, but not by mutant mRNAs, further supporting the pathogenicity of the identified variants. Hypomyelination and the absence of lax, wrinkly skin distinguishes this condition from that caused by previously reported mutations in the gene encoding PYCR2's isozyme, PYCR1, suggesting a unique and indispensable role for PYCR2 in the human CNS during development.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Hereditary Central Nervous System Demyelinating Diseases/genetics , Microcephaly/genetics , Mitochondrial Diseases/genetics , Psychomotor Disorders/genetics , Pyrroline Carboxylate Reductases/genetics , Amino Acid Transport Systems, Acidic/genetics , Antiporters/genetics , Female , Genotype , Hereditary Central Nervous System Demyelinating Diseases/pathology , Homozygote , Humans , Male , Microcephaly/pathology , Mitochondrial Diseases/pathology , Mutation , Phenotype , Psychomotor Disorders/pathology , delta-1-Pyrroline-5-Carboxylate Reductase
9.
Am J Med Genet B Neuropsychiatr Genet ; 177(8): 736-745, 2018 12.
Article in English | MEDLINE | ID: mdl-30421579

ABSTRACT

Protein homeostasis is tightly regulated by the ubiquitin proteasome pathway. Disruption of this pathway gives rise to a host of neurological disorders. Through whole exome sequencing (WES) in families with neurodevelopmental disorders, we identified mutations in PSMD12, a core component of the proteasome, underlying a neurodevelopmental disorder with intellectual disability (ID) and features of autism spectrum disorder (ASD). We performed WES on six affected siblings from a multiplex family with ID and autistic features, the affected father, and two unaffected mothers, and a trio from a simplex family with one affected child with ID and periventricular nodular heterotopia. We identified an inherited heterozygous nonsense mutation in PSMD12 (NM_002816: c.367C>T: p.R123X) in the multiplex family and a de novo nonsense mutation in the same gene (NM_002816: c.601C>T: p.R201X) in the simplex family. PSMD12 encodes a non-ATPase regulatory subunit of the 26S proteasome. We confirm the association of PSMD12 with ID, present the first cases of inherited PSMD12 mutation, and demonstrate the heterogeneity of phenotypes associated with PSMD12 mutations.


Subject(s)
Intellectual Disability/genetics , Proteasome Endopeptidase Complex/genetics , Adolescent , Adult , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Child , Child, Preschool , Family , Female , Genetic Predisposition to Disease , Haploinsufficiency/genetics , Humans , Male , Mutation , Neurodevelopmental Disorders/genetics , Pedigree , Proteasome Endopeptidase Complex/metabolism , Siblings , Exome Sequencing
10.
Hum Mutat ; 38(10): 1348-1354, 2017 10.
Article in English | MEDLINE | ID: mdl-28493438

ABSTRACT

Aminoacyl-transfer RNA (tRNA) synthetases ligate amino acids to specific tRNAs and are essential for protein synthesis. Although alanyl-tRNA synthetase (AARS) is a synthetase implicated in a wide range of neurological disorders from Charcot-Marie-Tooth disease to infantile epileptic encephalopathy, there have been limited data on their pathogenesis. Here, we report loss-of-function mutations in AARS in two siblings with progressive microcephaly with hypomyelination, intractable epilepsy, and spasticity. Whole-exome sequencing identified that the affected individuals were compound heterozygous for mutations in AARS gene, c.2067dupC (p.Tyr690Leufs*3) and c.2738G>A (p.Gly913Asp). A lymphoblastoid cell line developed from one of the affected individuals showed a strong reduction in AARS abundance. The mutations decrease aminoacylation efficiency by 70%-90%. The p.Tyr690Leufs*3 mutation also abolished editing activity required for hydrolyzing misacylated tRNAs, thereby increasing errors during aminoacylation. Our study has extended potential mechanisms underlying AARS-related disorders to include destabilization of the protein, aminoacylation dysfunction, and defective editing activity.


Subject(s)
Alanine-tRNA Ligase/genetics , Lennox Gastaut Syndrome/genetics , Microcephaly/genetics , Spasms, Infantile/genetics , Spastic Paraplegia, Hereditary/genetics , Amino Acid Sequence/genetics , Aminoacylation/genetics , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Child, Preschool , Electroencephalography , Female , Humans , Infant , Lennox Gastaut Syndrome/complications , Lennox Gastaut Syndrome/diagnosis , Lennox Gastaut Syndrome/pathology , Microcephaly/diagnostic imaging , Microcephaly/pathology , Mutation/genetics , Protein Biosynthesis/genetics , Siblings , Spasms, Infantile/complications , Spasms, Infantile/diagnostic imaging , Spasms, Infantile/pathology , Spastic Paraplegia, Hereditary/complications , Spastic Paraplegia, Hereditary/pathology , Exome Sequencing
11.
Am J Hum Genet ; 94(4): 547-58, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24656866

ABSTRACT

Progressive microcephaly is a heterogeneous condition with causes including mutations in genes encoding regulators of neuronal survival. Here, we report the identification of mutations in QARS (encoding glutaminyl-tRNA synthetase [QARS]) as the causative variants in two unrelated families affected by progressive microcephaly, severe seizures in infancy, atrophy of the cerebral cortex and cerebellar vermis, and mild atrophy of the cerebellar hemispheres. Whole-exome sequencing of individuals from each family independently identified compound-heterozygous mutations in QARS as the only candidate causative variants. QARS was highly expressed in the developing fetal human cerebral cortex in many cell types. The four QARS mutations altered highly conserved amino acids, and the aminoacylation activity of QARS was significantly impaired in mutant cell lines. Variants p.Gly45Val and p.Tyr57His were located in the N-terminal domain required for QARS interaction with proteins in the multisynthetase complex and potentially with glutamine tRNA, and recombinant QARS proteins bearing either substitution showed an over 10-fold reduction in aminoacylation activity. Conversely, variants p.Arg403Trp and p.Arg515Trp, each occurring in a different family, were located in the catalytic core and completely disrupted QARS aminoacylation activity in vitro. Furthermore, p.Arg403Trp and p.Arg515Trp rendered QARS less soluble, and p.Arg403Trp disrupted QARS-RARS (arginyl-tRNA synthetase 1) interaction. In zebrafish, homozygous qars loss of function caused decreased brain and eye size and extensive cell death in the brain. Our results highlight the importance of QARS during brain development and that epilepsy due to impairment of QARS activity is unusually severe in comparison to other aminoacyl-tRNA synthetase disorders.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Brain Diseases/genetics , Genetic Predisposition to Disease , Microcephaly/genetics , Mutation , Seizures/genetics , Aminoacylation , Animals , Child, Preschool , Female , Humans , Magnetic Resonance Imaging , Male , Microcephaly/pathology , Pedigree , Zebrafish
12.
N Engl J Med ; 371(8): 733-43, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25140959

ABSTRACT

BACKGROUND: Although there is increasing recognition of the role of somatic mutations in genetic disorders, the prevalence of somatic mutations in neurodevelopmental disease and the optimal techniques to detect somatic mosaicism have not been systematically evaluated. METHODS: Using a customized panel of known and candidate genes associated with brain malformations, we applied targeted high-coverage sequencing (depth, ≥200×) to leukocyte-derived DNA samples from 158 persons with brain malformations, including the double-cortex syndrome (subcortical band heterotopia, 30 persons), polymicrogyria with megalencephaly (20), periventricular nodular heterotopia (61), and pachygyria (47). We validated candidate mutations with the use of Sanger sequencing and, for variants present at unequal read depths, subcloning followed by colony sequencing. RESULTS: Validated, causal mutations were found in 27 persons (17%; range, 10 to 30% for each phenotype). Mutations were somatic in 8 of the 27 (30%), predominantly in persons with the double-cortex syndrome (in whom we found mutations in DCX and LIS1), persons with periventricular nodular heterotopia (FLNA), and persons with pachygyria (TUBB2B). Of the somatic mutations we detected, 5 (63%) were undetectable with the use of traditional Sanger sequencing but were validated through subcloning and subsequent sequencing of the subcloned DNA. We found potentially causal mutations in the candidate genes DYNC1H1, KIF5C, and other kinesin genes in persons with pachygyria. CONCLUSIONS: Targeted sequencing was found to be useful for detecting somatic mutations in patients with brain malformations. High-coverage sequencing panels provide an important complement to whole-exome and whole-genome sequencing in the evaluation of somatic mutations in neuropsychiatric disease. (Funded by the National Institute of Neurological Disorders and Stroke and others.).


Subject(s)
Cerebral Cortex/abnormalities , DNA Mutational Analysis/methods , Malformations of Cortical Development/genetics , Mutation , Classical Lissencephalies and Subcortical Band Heterotopias/genetics , Humans , Lissencephaly/genetics , Magnetic Resonance Imaging , Malformations of Cortical Development/pathology , Periventricular Nodular Heterotopia/genetics
13.
Hum Mol Genet ; 23(13): 3456-66, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24501276

ABSTRACT

Whereas many genes associated with intellectual disability (ID) encode synaptic proteins, transcriptional defects leading to ID are less well understood. We studied a large, consanguineous pedigree of Arab origin with seven members affected with ID and mild dysmorphic features. Homozygosity mapping and linkage analysis identified a candidate region on chromosome 17 with a maximum multipoint logarithm of odds score of 6.01. Targeted high-throughput sequencing of the exons in the candidate region identified a homozygous 4-bp deletion (c.169_172delCACT) in the METTL23 (methyltransferase like 23) gene, which is predicted to result in a frameshift and premature truncation (p.His57Valfs*11). Overexpressed METTL23 protein localized to both nucleus and cytoplasm, and physically interacted with GABPA (GA-binding protein transcription factor, alpha subunit). GABP, of which GABPA is a component, is known to regulate the expression of genes such as THPO (thrombopoietin) and ATP5B (ATP synthase, H+ transporting, mitochondrial F1 complex, beta polypeptide) and is implicated in a wide variety of important cellular functions. Overexpression of METTL23 resulted in increased transcriptional activity at the THPO promoter, whereas knockdown of METTL23 with siRNA resulted in decreased expression of ATP5B, thus revealing the importance of METTL23 as a regulator of GABPA function. The METTL23 mutation highlights a new transcriptional pathway underlying human intellectual function.


Subject(s)
DNA Modification Methylases/metabolism , GA-Binding Protein Transcription Factor/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Modification Methylases/genetics , Female , GA-Binding Protein Transcription Factor/genetics , Genotype , Humans , Immunoprecipitation , Male , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Polymorphism, Single Nucleotide/genetics , Protein Binding , RNA, Small Interfering , Thrombopoietin/genetics , Thrombopoietin/metabolism , Two-Hybrid System Techniques
14.
Am J Hum Genet ; 91(3): 541-7, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22958903

ABSTRACT

Whole-exome sequencing (WES), which analyzes the coding sequence of most annotated genes in the human genome, is an ideal approach to studying fully penetrant autosomal-recessive diseases, and it has been very powerful in identifying disease-causing mutations even when enrollment of affected individuals is limited by reduced survival. In this study, we combined WES with homozygosity analysis of consanguineous pedigrees, which are informative even when a single affected individual is available, to identify genetic mutations responsible for Walker-Warburg syndrome (WWS), a genetically heterogeneous autosomal-recessive disorder that severely affects the development of the brain, eyes, and muscle. Mutations in seven genes are known to cause WWS and explain 50%-60% of cases, but multiple additional genes are expected to be mutated because unexplained cases show suggestive linkage to diverse loci. Using WES in consanguineous WWS-affected families, we found multiple deleterious mutations in GTDC2 (also known as AGO61). GTDC2's predicted role as an uncharacterized glycosyltransferase is consistent with the function of other genes that are known to be mutated in WWS and that are involved in the glycosylation of the transmembrane receptor dystroglycan. Therefore, to explore the role of GTDC2 loss of function during development, we used morpholino-mediated knockdown of its zebrafish ortholog, gtdc2. We found that gtdc2 knockdown in zebrafish replicates all WWS features (hydrocephalus, ocular defects, and muscular dystrophy), strongly suggesting that GTDC2 mutations cause WWS.


Subject(s)
Glycosyltransferases/genetics , Walker-Warburg Syndrome/genetics , Exome , Humans , Mutation
15.
Am J Hum Genet ; 88(5): 536-47, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21529751

ABSTRACT

Genes disrupted in human microcephaly (meaning "small brain") define key regulators of neural progenitor proliferation and cell-fate specification. In comparison, genes mutated in human lissencephaly (lissos means smooth and cephalos means brain) highlight critical regulators of neuronal migration. Here, we report two families with extreme microcephaly and grossly simplified cortical gyral structure, a condition referred to as microlissencephaly, and show that they carry homozygous frameshift mutations in NDE1, which encodes a multidomain protein that localizes to the centrosome and mitotic spindle poles. Both human mutations in NDE1 truncate the C-terminal NDE1domains, which are essential for interactions with cytoplasmic dynein and thus for regulation of cytoskeletal dynamics in mitosis and for cell-cycle-dependent phosphorylation of NDE1 by Cdk1. We show that the patient NDE1 proteins are unstable, cannot bind cytoplasmic dynein, and do not localize properly to the centrosome. Additionally, we show that CDK1 phosphorylation at T246, which is within the C-terminal region disrupted by the mutations, is required for cell-cycle progression from the G2 to the M phase. The role of NDE1 in cell-cycle progression probably contributes to the profound neuronal proliferation defects evident in Nde1-null mice and patients with NDE1 mutations, demonstrating the essential role of NDE1 in human cerebral cortical neurogenesis.


Subject(s)
Frameshift Mutation , Lissencephaly/genetics , Microcephaly/genetics , Microtubule-Associated Proteins/genetics , Animals , CDC2 Protein Kinase/metabolism , Cell Differentiation , Cell Line , Cell Movement , Centrosome/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Child , Child, Preschool , Female , Genetic Linkage , Homozygote , Humans , Infant , Male , Mice , Mice, Knockout , Neurons/cytology , Phosphorylation , Protein Stability , Spindle Apparatus/metabolism , Transfection
16.
Ann Neurol ; 74(6): 873-82, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24596948

ABSTRACT

OBJECTIVE: To identify a genetic cause for migrating partial seizures in infancy (MPSI). METHODS: We characterized a consanguineous pedigree with MPSI and obtained DNA from affected and unaffected family members. We analyzed single nucleotide polymorphism 500K data to identify regions with evidence of linkage. We performed whole exome sequencing and analyzed homozygous variants in regions of linkage to identify a candidate gene and performed functional studies of the candidate gene SLC25A22. RESULTS: In a consanguineous pedigree with 2 individuals with MPSI, we identified 2 regions of linkage, chromosome 4p16.1-p16.3 and chromosome 11p15.4-pter. Using whole exome sequencing, we identified 8 novel homozygous variants in genes in these regions. Only 1 variant, SLC25A22 c.G328C, results in a change of a highly conserved amino acid (p.G110R) and was not present in control samples. SLC25A22 encodes a glutamate transporter with strong expression in the developing brain. We show that the specific G110R mutation, located in a transmembrane domain of the protein, disrupts mitochondrial glutamate transport. INTERPRETATION: We have shown that MPSI can be inherited and have identified a novel homozygous mutation in SLC25A22 in the affected individuals. Our data strongly suggest that SLC25A22 is responsible for MPSI, a severe condition with few known etiologies. We have demonstrated that a combination of linkage analysis and whole exome sequencing can be used for disease gene discovery. Finally, as SLC25A22 had been implicated in the distinct syndrome of neonatal epilepsy with suppression bursts on electroencephalogram, we have expanded the phenotypic spectrum associated with SLC25A22.


Subject(s)
Epilepsy, Benign Neonatal/genetics , Exome/genetics , Mitochondrial Membrane Transport Proteins/genetics , Adult , Consanguinity , Epilepsy, Benign Neonatal/physiopathology , Female , Genetic Linkage/genetics , Humans , Infant, Newborn , Male , Pedigree
17.
Am J Hum Genet ; 87(6): 882-9, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21109224

ABSTRACT

The tight junction, or zonula occludens, is a specialized cell-cell junction that regulates epithelial and endothelial permeability, and it is an essential component of the blood-brain barrier in the cerebrovascular endothelium. In addition to functioning as a diffusion barrier, tight junctions are also involved in signal transduction. In this study, we identified a homozygous mutation in the tight-junction protein gene JAM3 in a large consanguineous family from the United Arab Emirates. Some members of this family had a rare autosomal-recessive syndrome characterized by severe hemorrhagic destruction of the brain, subependymal calcification, and congenital cataracts. Their clinical presentation overlaps with some reported cases of pseudo-TORCH syndrome as well as with cases involving mutations in occludin, another component of the tight-junction complex. However, massive intracranial hemorrhage distinguishes these patients from others. Homozygosity mapping identified the disease locus in this family on chromosome 11q25 with a maximum multipoint LOD score of 6.15. Sequence analysis of genes in the candidate interval uncovered a mutation in the canonical splice-donor site of intron 5 of JAM3. RT-PCR analysis of a patient lymphoblast cell line confirmed abnormal splicing, leading to a frameshift mutation with early termination. JAM3 is known to be present in vascular endothelium, although its roles in cerebral vasculature have not been implicated. Our results suggest that JAM3 is essential for maintaining the integrity of the cerebrovascular endothelium as well as for normal lens development in humans.


Subject(s)
Calcinosis/genetics , Cataract/congenital , Cell Adhesion Molecules/genetics , Cerebral Hemorrhage/genetics , Ependyma/pathology , Homozygote , Mutation , Tight Junctions/metabolism , Cataract/genetics , Child , Female , Humans , Infant , Infant, Newborn , Male , Pedigree
18.
Ann Neurol ; 67(4): 516-25, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20437587

ABSTRACT

OBJECTIVE: We sought to explore the genetic and molecular causes of Troyer syndrome, one of several complicated hereditary spastic paraplegias (HSPs). Troyer syndrome had been thought to be restricted to the Amish; however, we identified 2 Omani families with HSP, short stature, dysarthria and developmental delay-core features of Troyer syndrome-and a novel mutation in the SPG20 gene, which is also mutated in the Amish. In addition, we analyzed SPG20 expression throughout development to infer how disruption of this gene might generate the constellation of developmental and degenerative Troyer syndrome phenotypes. METHODS: Clinical characterization of 2 non-Amish families with Troyer syndrome was followed by linkage and sequencing analysis. Quantitative polymerase chain reaction and in situ hybridization analysis of SPG20 expression were carried out in embryonic and adult human and mouse tissue. RESULTS: Two Omani families carrying a novel SPG20 mutation displayed clinical features remarkably similar to the Amish patients with Troyer syndrome. SPG20 mRNA is expressed broadly but at low relative levels in the adult brain; however, it is robustly and specifically expressed in the limbs, face, and brain during early morphogenesis. INTERPRETATION: Null mutations in SPG20 cause Troyer syndrome, a specific clinical entity with developmental and degenerative features. Maximal expression of SPG20 in the limb buds and forebrain during embryogenesis may explain the developmental origin of the skeletal and cognitive defects observed in this disorder.


Subject(s)
Genetic Predisposition to Disease/genetics , Neurodegenerative Diseases/etiology , Paraplegia/complications , Paraplegia/genetics , Polymorphism, Single Nucleotide/genetics , Proteins/genetics , Adolescent , Adult , Cell Cycle Proteins , Child, Preschool , Chromosome Mapping , DNA Mutational Analysis/methods , Family Health , Female , Humans , Magnetic Resonance Imaging , Male , Oman , Paraplegia/pathology , Proteins/metabolism , RNA, Messenger/genetics , Young Adult
19.
Am J Med Genet A ; 152A(11): 2736-42, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20949537

ABSTRACT

Schizencephaly is a malformation of cortical development characterized by gray matter-lined clefts in the cerebral cortex and a range of neurological presentations. In some cases, there are features of septo-optic dysplasia concurrently with schizencephaly. The etiologies of both schizencephaly and septo-optic dysplasia are thought to be heterogeneous, but there is evidence that at least some cases have genetic origin. We hypothesized that these disorders may be caused by mutations in three candidate genes: LHX2, a gene with an important cortical patterning role, and HESX1 and SOX2, genes that have been associated with septo-optic dysplasia. We sequenced a large cohort of patients with schizencephaly, some with features of septo-optic dysplasia, for mutations in these genes. No pathogenic mutations were observed, suggesting that other genes or non-genetic factors influencing genes critical to brain development must be responsible for schizencephaly.


Subject(s)
Homeodomain Proteins/genetics , Malformations of Cortical Development/genetics , SOXB1 Transcription Factors/genetics , Sequence Analysis, DNA , Transcription Factors/genetics , Adult , Base Sequence , Child, Preschool , Cohort Studies , Female , Humans , Infant , Infant, Newborn , LIM-Homeodomain Proteins , Magnetic Resonance Imaging , Male , Malformations of Cortical Development/complications , Septo-Optic Dysplasia/complications , Septo-Optic Dysplasia/genetics , Young Adult
20.
Neuron ; 106(2): 246-255.e6, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32097629

ABSTRACT

Genes mutated in human neuronal migration disorders encode tubulin proteins and a variety of tubulin-binding and -regulating proteins, but it is very poorly understood how these proteins function together to coordinate migration. Additionally, the way in which regional differences in neocortical migration are controlled is completely unknown. Here we describe a new syndrome with remarkably region-specific effects on neuronal migration in the posterior cortex, reflecting de novo variants in CEP85L. We show that CEP85L is required cell autonomously in vivo and in vitro for migration, that it localizes to the maternal centriole, and that it forms a complex with many other proteins required for migration, including CDK5, LIS1, NDE1, KIF2A, and DYNC1H1. Loss of CEP85L disrupts CDK5 localization and activation, leading to centrosome disorganization and disrupted microtubule cytoskeleton organization. Together, our findings suggest that CEP85L highlights a complex that controls CDK5 activity to promote neuronal migration.


Subject(s)
Cell Movement , Cyclin-Dependent Kinase 5/genetics , Cytoskeletal Proteins/genetics , Lissencephaly/genetics , Lissencephaly/pathology , Neocortex/pathology , Neurons/pathology , Oncogene Proteins, Fusion/genetics , Centrioles/genetics , Child , Child, Preschool , Female , Humans , Male , Microtubules/genetics , Microtubules/ultrastructure , Nerve Tissue Proteins/physiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL