Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters

Country/Region as subject
Publication year range
1.
N Engl J Med ; 390(21): 1985-1997, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38838312

ABSTRACT

BACKGROUND: Genetic variants that cause rare disorders may remain elusive even after expansive testing, such as exome sequencing. The diagnostic yield of genome sequencing, particularly after a negative evaluation, remains poorly defined. METHODS: We sequenced and analyzed the genomes of families with diverse phenotypes who were suspected to have a rare monogenic disease and for whom genetic testing had not revealed a diagnosis, as well as the genomes of a replication cohort at an independent clinical center. RESULTS: We sequenced the genomes of 822 families (744 in the initial cohort and 78 in the replication cohort) and made a molecular diagnosis in 218 of 744 families (29.3%). Of the 218 families, 61 (28.0%) - 8.2% of families in the initial cohort - had variants that required genome sequencing for identification, including coding variants, intronic variants, small structural variants, copy-neutral inversions, complex rearrangements, and tandem repeat expansions. Most families in which a molecular diagnosis was made after previous nondiagnostic exome sequencing (63.5%) had variants that could be detected by reanalysis of the exome-sequence data (53.4%) or by additional analytic methods, such as copy-number variant calling, to exome-sequence data (10.8%). We obtained similar results in the replication cohort: in 33% of the families in which a molecular diagnosis was made, or 8% of the cohort, genome sequencing was required, which showed the applicability of these findings to both research and clinical environments. CONCLUSIONS: The diagnostic yield of genome sequencing in a large, diverse research cohort and in a small clinical cohort of persons who had previously undergone genetic testing was approximately 8% and included several types of pathogenic variation that had not previously been detected by means of exome sequencing or other techniques. (Funded by the National Human Genome Research Institute and others.).


Subject(s)
Genetic Variation , Rare Diseases , Whole Genome Sequencing , Female , Humans , Male , Cohort Studies , Exome , Exome Sequencing , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/ethnology , Genetic Diseases, Inborn/genetics , Genetic Testing , Genome, Human , Phenotype , Rare Diseases/diagnosis , Rare Diseases/ethnology , Rare Diseases/genetics , Sequence Analysis, DNA , Child , Adolescent , Young Adult , Adult
2.
Am J Hum Genet ; 110(12): 2112-2119, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37963460

ABSTRACT

Over two dozen spliceosome proteins are involved in human diseases, also referred to as spliceosomopathies. WW domain-binding protein 4 (WBP4) is part of the early spliceosomal complex and has not been previously associated with human pathologies in the Online Mendelian Inheritance in Man (OMIM) database. Through GeneMatcher, we identified ten individuals from eight families with a severe neurodevelopmental syndrome featuring variable manifestations. Clinical manifestations included hypotonia, global developmental delay, severe intellectual disability, brain abnormalities, musculoskeletal, and gastrointestinal abnormalities. Genetic analysis revealed five different homozygous loss-of-function variants in WBP4. Immunoblotting on fibroblasts from two affected individuals with different genetic variants demonstrated a complete loss of protein, and RNA sequencing analysis uncovered shared abnormal splicing patterns, including in genes associated with abnormalities of the nervous system, potentially underlying the phenotypes of the probands. We conclude that bi-allelic variants in WBP4 cause a developmental disorder with variable presentations, adding to the growing list of human spliceosomopathies.


Subject(s)
Intellectual Disability , Nervous System Malformations , Neurodevelopmental Disorders , Humans , Spliceosomes/genetics , Neurodevelopmental Disorders/genetics , Intellectual Disability/genetics , Intellectual Disability/complications , Syndrome , Nervous System Malformations/genetics , Loss of Heterozygosity , Phenotype
3.
N Engl J Med ; 389(13): 1203-1210, 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37754285

ABSTRACT

We treated a 27-year-old patient with Duchenne's muscular dystrophy (DMD) with recombinant adeno-associated virus (rAAV) serotype 9 containing dSaCas9 (i.e., "dead" Staphylococcus aureus Cas9, in which the Cas9 nuclease activity has been inactivated) fused to VP64; this transgene was designed to up-regulate cortical dystrophin as a custom CRISPR-transactivator therapy. The dose of rAAV used was 1×1014 vector genomes per kilogram of body weight. Mild cardiac dysfunction and pericardial effusion developed, followed by acute respiratory distress syndrome (ARDS) and cardiac arrest 6 days after transgene treatment; the patient died 2 days later. A postmortem examination showed severe diffuse alveolar damage. Expression of transgene in the liver was minimal, and there was no evidence of AAV serotype 9 antibodies or effector T-cell reactivity in the organs. These findings indicate that an innate immune reaction caused ARDS in a patient with advanced DMD treated with high-dose rAAV gene therapy. (Funded by Cure Rare Disease.).


Subject(s)
Dystrophin , Genetic Therapy , Muscular Dystrophy, Duchenne , Respiratory Distress Syndrome , Transgenes , Adult , Humans , Antibodies , Dystrophin/genetics , Genetic Therapy/adverse effects , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/immunology , Transgenes/genetics , Transgenes/immunology , Fatal Outcome , Immunity, Innate/genetics , Immunity, Innate/immunology
4.
Am J Hum Genet ; 108(4): 749-756, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33743206

ABSTRACT

The DNA damage-binding protein 1 (DDB1) is part of the CUL4-DDB1 ubiquitin E3 ligase complex (CRL4), which is essential for DNA repair, chromatin remodeling, DNA replication, and signal transduction. Loss-of-function variants in genes encoding the complex components CUL4 and PHIP have been reported to cause syndromic intellectual disability with hypotonia and obesity, but no phenotype has been reported in association with DDB1 variants. Here, we report eight unrelated individuals, identified through Matchmaker Exchange, with de novo monoallelic variants in DDB1, including one recurrent variant in four individuals. The affected individuals have a consistent phenotype of hypotonia, mild to moderate intellectual disability, and similar facies, including horizontal or slightly bowed eyebrows, deep-set eyes, full cheeks, a short nose, and large, fleshy and forward-facing earlobes, demonstrated in the composite face generated from the cohort. Digital anomalies, including brachydactyly and syndactyly, were common. Three older individuals have obesity. We show that cells derived from affected individuals have altered DDB1 function resulting in abnormal DNA damage signatures and histone methylation following UV-induced DNA damage. Overall, our study adds to the growing family of neurodevelopmental phenotypes mediated by disruption of the CRL4 ubiquitin ligase pathway and begins to delineate the phenotypic and molecular effects of DDB1 misregulation.


Subject(s)
Alleles , DNA Repair/genetics , DNA-Binding Proteins/genetics , Mutation , Neurodevelopmental Disorders/genetics , Adolescent , Child , Child, Preschool , Female , Humans , Male , Phenotype , Syndrome
5.
Am J Hum Genet ; 108(5): 840-856, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33861953

ABSTRACT

JAG2 encodes the Notch ligand Jagged2. The conserved Notch signaling pathway contributes to the development and homeostasis of multiple tissues, including skeletal muscle. We studied an international cohort of 23 individuals with genetically unsolved muscular dystrophy from 13 unrelated families. Whole-exome sequencing identified rare homozygous or compound heterozygous JAG2 variants in all 13 families. The identified bi-allelic variants include 10 missense variants that disrupt highly conserved amino acids, a nonsense variant, two frameshift variants, an in-frame deletion, and a microdeletion encompassing JAG2. Onset of muscle weakness occurred from infancy to young adulthood. Serum creatine kinase (CK) levels were normal or mildly elevated. Muscle histology was primarily dystrophic. MRI of the lower extremities revealed a distinct, slightly asymmetric pattern of muscle involvement with cores of preserved and affected muscles in quadriceps and tibialis anterior, in some cases resembling patterns seen in POGLUT1-associated muscular dystrophy. Transcriptome analysis of muscle tissue from two participants suggested misregulation of genes involved in myogenesis, including PAX7. In complementary studies, Jag2 downregulation in murine myoblasts led to downregulation of multiple components of the Notch pathway, including Megf10. Investigations in Drosophila suggested an interaction between Serrate and Drpr, the fly orthologs of JAG1/JAG2 and MEGF10, respectively. In silico analysis predicted that many Jagged2 missense variants are associated with structural changes and protein misfolding. In summary, we describe a muscular dystrophy associated with pathogenic variants in JAG2 and evidence suggests a disease mechanism related to Notch pathway dysfunction.


Subject(s)
Jagged-2 Protein/genetics , Muscular Dystrophies/genetics , Adolescent , Adult , Amino Acid Sequence , Animals , Cell Line , Child , Child, Preschool , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , Glucosyltransferases/genetics , Haplotypes/genetics , Humans , Jagged-1 Protein/genetics , Jagged-2 Protein/chemistry , Jagged-2 Protein/deficiency , Jagged-2 Protein/metabolism , Male , Membrane Proteins/genetics , Mice , Middle Aged , Models, Molecular , Muscles/metabolism , Muscles/pathology , Muscular Dystrophies/pathology , Myoblasts/metabolism , Myoblasts/pathology , Pedigree , Phenotype , Receptors, Notch/metabolism , Signal Transduction , Exome Sequencing , Young Adult
6.
Am J Hum Genet ; 108(9): 1692-1709, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34375587

ABSTRACT

Kainate receptors (KARs) are glutamate-gated cation channels with diverse roles in the central nervous system. Bi-allelic loss of function of the KAR-encoding gene GRIK2 causes a nonsyndromic neurodevelopmental disorder (NDD) with intellectual disability and developmental delay as core features. The extent to which mono-allelic variants in GRIK2 also underlie NDDs is less understood because only a single individual has been reported previously. Here, we describe an additional eleven individuals with heterozygous de novo variants in GRIK2 causative for neurodevelopmental deficits that include intellectual disability. Five children harbored recurrent de novo variants (three encoding p.Thr660Lys and two p.Thr660Arg), and four children and one adult were homozygous for a previously reported variant (c.1969G>A [p.Ala657Thr]). Individuals with shared variants had some overlapping behavioral and neurological dysfunction, suggesting that the GRIK2 variants are likely pathogenic. Analogous mutations introduced into recombinant GluK2 KAR subunits at sites within the M3 transmembrane domain (encoding p.Ala657Thr, p.Thr660Lys, and p.Thr660Arg) and the M3-S2 linker domain (encoding p.Ile668Thr) had complex effects on functional properties and membrane localization of homomeric and heteromeric KARs. Both p.Thr660Lys and p.Thr660Arg mutant KARs exhibited markedly slowed gating kinetics, similar to p.Ala657Thr-containing receptors. Moreover, we observed emerging genotype-phenotype correlations, including the presence of severe epilepsy in individuals with the p.Thr660Lys variant and hypomyelination in individuals with either the p.Thr660Lys or p.Thr660Arg variant. Collectively, these results demonstrate that human GRIK2 variants predicted to alter channel function are causative for early childhood development disorders and further emphasize the importance of clarifying the role of KARs in early nervous system development.


Subject(s)
Brain/metabolism , Developmental Disabilities/genetics , Epilepsy/genetics , Intellectual Disability/genetics , Mutation , Receptors, Kainic Acid/genetics , Adolescent , Adult , Alleles , Brain/diagnostic imaging , Brain/pathology , Child , Child, Preschool , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/metabolism , Developmental Disabilities/pathology , Epilepsy/diagnostic imaging , Epilepsy/metabolism , Epilepsy/pathology , Evoked Potentials/physiology , Gene Expression Regulation, Developmental , Genetic Association Studies , Heterozygote , Homozygote , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/metabolism , Intellectual Disability/pathology , Ion Channel Gating , Male , Models, Molecular , Neurons/metabolism , Neurons/pathology , Protein Conformation , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/metabolism , GluK2 Kainate Receptor
7.
Am J Hum Genet ; 106(2): 234-245, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31928709

ABSTRACT

Germline pathogenic variants in chromatin-modifying enzymes are a common cause of pediatric developmental disorders. These enzymes catalyze reactions that regulate epigenetic inheritance via histone post-translational modifications and DNA methylation. Cytosine methylation (5-methylcytosine [5mC]) of DNA is the quintessential epigenetic mark, yet no human Mendelian disorder of DNA demethylation has yet been delineated. Here, we describe in detail a Mendelian disorder caused by the disruption of DNA demethylation. TET3 is a methylcytosine dioxygenase that initiates DNA demethylation during early zygote formation, embryogenesis, and neuronal differentiation and is intolerant to haploinsufficiency in mice and humans. We identify and characterize 11 cases of human TET3 deficiency in eight families with the common phenotypic features of intellectual disability and/or global developmental delay; hypotonia; autistic traits; movement disorders; growth abnormalities; and facial dysmorphism. Mono-allelic frameshift and nonsense variants in TET3 occur throughout the coding region. Mono-allelic and bi-allelic missense variants localize to conserved residues; all but one such variant occur within the catalytic domain, and most display hypomorphic function in an assay of catalytic activity. TET3 deficiency and other Mendelian disorders of the epigenetic machinery show substantial phenotypic overlap, including features of intellectual disability and abnormal growth, underscoring shared disease mechanisms.


Subject(s)
DNA Demethylation , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Dioxygenases/deficiency , Adult , Amino Acid Sequence , Autistic Disorder/genetics , Autistic Disorder/pathology , Child , Child, Preschool , Dioxygenases/chemistry , Dioxygenases/genetics , Embryonic Development , Female , Gene Expression Regulation, Developmental , Growth Disorders/genetics , Growth Disorders/pathology , Humans , Infant , Male , Middle Aged , Movement Disorders/genetics , Movement Disorders/pathology , Pedigree , Protein Conformation , Sequence Homology , Young Adult
8.
Hum Mutat ; 43(10): 1347-1353, 2022 10.
Article in English | MEDLINE | ID: mdl-35731190

ABSTRACT

The investigated intronic CAPN3 variant NM_000070.3:c.1746-20C>G occurs in the Central and Eastern Europe with a frequency of >1% and there are conflicting interpretations on its pathogenicity. We collected data on 14 patients carrying the CAPN3 c.1746-20C>G variant in trans position with another CAPN3 pathogenic/likely pathogenic variant. The patients compound heterozygous for the CAPN3 c.1746-20C>G variant presented a phenotype consistent with calpainopathy of mild/medium severity. This variant is most frequent in the North/West regions of Russia and may originate from that area. Molecular studies revealed that different splicing isoforms are produced in the muscle. We hypothesize that c.1746-20C>G is a hypomorphic variant with a reduction of RNA and protein expression and only individuals having a higher ratio of abnormal isoforms are affected. Reclassification of the CAPN3 variant c.1746-20C>G from variant with a conflicting interpretation of pathogenicity to hypomorphic variant explains many unidentified cases of limb girdle muscular dystrophy R1 calpain 3-related in Eastern and Central Europe.


Subject(s)
Calpain , Muscle Proteins , Muscular Dystrophies, Limb-Girdle , Calpain/genetics , Humans , Muscle Proteins/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Mutation , RNA Splicing
9.
Hum Mutat ; 43(12): 1844-1851, 2022 12.
Article in English | MEDLINE | ID: mdl-35904126

ABSTRACT

TATA-binding protein associated factor 4 (TAF4) is a subunit of the Transcription Factor IID (TFIID) complex, a central player in transcription initiation. Other members of this multimeric complex have been implicated previously as monogenic disease genes in human developmental disorders. TAF4 has not been described to date as a monogenic disease gene. We here present a cohort of eight individuals, each carrying de novo putative loss-of-function (pLoF) variants in TAF4 and expressing phenotypes consistent with a neuro-developmental disorder (NDD). Common features include intellectual disability, abnormal behavior, and facial dysmorphisms. We propose TAF4 as a novel dominant disease gene for NDD, and coin this novel disorder "TAF4-related NDD" (T4NDD). We place T4NDD in the context of other disorders related to TFIID subunits, revealing shared features of T4NDD with other TAF-opathies.


Subject(s)
Neurodevelopmental Disorders , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Child , Humans , Developmental Disabilities/genetics , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , Phenotype , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism
10.
Am J Hum Genet ; 105(3): 493-508, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31447100

ABSTRACT

Histones mediate dynamic packaging of nuclear DNA in chromatin, a process that is precisely controlled to guarantee efficient compaction of the genome and proper chromosomal segregation during cell division and to accomplish DNA replication, transcription, and repair. Due to the important structural and regulatory roles played by histones, it is not surprising that histone functional dysregulation or aberrant levels of histones can have severe consequences for multiple cellular processes and ultimately might affect development or contribute to cell transformation. Recently, germline frameshift mutations involving the C-terminal tail of HIST1H1E, which is a widely expressed member of the linker histone family and facilitates higher-order chromatin folding, have been causally linked to an as-yet poorly defined syndrome that includes intellectual disability. We report that these mutations result in stable proteins that reside in the nucleus, bind to chromatin, disrupt proper compaction of DNA, and are associated with a specific methylation pattern. Cells expressing these mutant proteins have a dramatically reduced proliferation rate and competence, hardly enter into the S phase, and undergo accelerated senescence. Remarkably, clinical assessment of a relatively large cohort of subjects sharing these mutations revealed a premature aging phenotype as a previously unrecognized feature of the disorder. Our findings identify a direct link between aberrant chromatin remodeling, cellular senescence, and accelerated aging.


Subject(s)
Cellular Senescence/physiology , Histones/physiology , Aneuploidy , Cell Nucleolus/metabolism , Child , Chromatin/metabolism , DNA Methylation , Female , Histones/chemistry , Humans , Infant , Male , Middle Aged
11.
Am J Hum Genet ; 102(5): 744-759, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29656859

ABSTRACT

RORα, the RAR-related orphan nuclear receptor alpha, is essential for cerebellar development. The spontaneous mutant mouse staggerer, with an ataxic gait caused by neurodegeneration of cerebellar Purkinje cells, was discovered two decades ago to result from homozygous intragenic Rora deletions. However, RORA mutations were hitherto undocumented in humans. Through a multi-centric collaboration, we identified three copy-number variant deletions (two de novo and one dominantly inherited in three generations), one de novo disrupting duplication, and nine de novo point mutations (three truncating, one canonical splice site, and five missense mutations) involving RORA in 16 individuals from 13 families with variable neurodevelopmental delay and intellectual disability (ID)-associated autistic features, cerebellar ataxia, and epilepsy. Consistent with the human and mouse data, disruption of the D. rerio ortholog, roraa, causes significant reduction in the size of the developing cerebellum. Systematic in vivo complementation studies showed that, whereas wild-type human RORA mRNA could complement the cerebellar pathology, missense variants had two distinct pathogenic mechanisms of either haploinsufficiency or a dominant toxic effect according to their localization in the ligand-binding or DNA-binding domains, respectively. This dichotomous direction of effect is likely relevant to the phenotype in humans: individuals with loss-of-function variants leading to haploinsufficiency show ID with autistic features, while individuals with de novo dominant toxic variants present with ID, ataxia, and cerebellar atrophy. Our combined genetic and functional data highlight the complex mutational landscape at the human RORA locus and suggest that dual mutational effects likely determine phenotypic outcome.


Subject(s)
Autistic Disorder/genetics , Cerebellar Ataxia/genetics , Genes, Dominant , Intellectual Disability/genetics , Mutation, Missense/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Adolescent , Adult , Aged, 80 and over , Alleles , Animals , Autistic Disorder/complications , Brain/pathology , Cerebellar Ataxia/complications , Child , Child, Preschool , DNA Copy Number Variations/genetics , Disease Models, Animal , Female , Genetic Complementation Test , Humans , Intellectual Disability/complications , Larva/genetics , Magnetic Resonance Imaging , Male , Middle Aged , Purkinje Cells/metabolism , Purkinje Cells/pathology , Syndrome , Zebrafish/genetics
12.
Ann Neurol ; 86(1): 129-142, 2019 07.
Article in English | MEDLINE | ID: mdl-31025394

ABSTRACT

OBJECTIVE: To define a distinct, dominantly inherited, mild skeletal myopathy associated with prominent and consistent tremor in two unrelated, three-generation families. METHODS: Clinical evaluations as well as exome and panel sequencing analyses were performed in affected and nonaffected members of two families to identify genetic variants segregating with the phenotype. Histological assessment of a muscle biopsy specimen was performed in 1 patient, and quantitative tremor analysis was carried out in 2 patients. Molecular modeling studies and biochemical assays were performed for both mutations. RESULTS: Two novel missense mutations in MYBPC1 (p.E248K in family 1 and p.Y247H in family 2) were identified and shown to segregate perfectly with the myopathy/tremor phenotype in the respective families. MYBPC1 encodes slow myosin binding protein-C (sMyBP-C), a modular sarcomeric protein playing structural and regulatory roles through its dynamic interaction with actin and myosin filaments. The Y247H and E248K mutations are located in the NH2 -terminal M-motif of sMyBP-C. Both mutations result in markedly increased binding of the NH2 terminus to myosin, possibly interfering with normal cross-bridge cycling as the first muscle-based step in tremor genesis. The clinical tremor features observed in all mutation carriers, together with the tremor physiology studies performed in family 2, suggest amplification by an additional central loop modulating the clinical tremor phenomenology. INTERPRETATION: Here, we link two novel missense mutations in MYBPC1 with a dominant, mild skeletal myopathy invariably associated with a distinctive tremor. The molecular, genetic, and clinical studies are consistent with a unique sarcomeric origin of the tremor, which we classify as "myogenic tremor." ANN NEUROL 2019.


Subject(s)
Carrier Proteins/genetics , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Mutation, Missense/genetics , Tremor/diagnosis , Tremor/genetics , Adult , Carrier Proteins/chemistry , Female , Humans , Male , Middle Aged , Pedigree , Protein Structure, Secondary , Protein Structure, Tertiary
15.
Genet Med ; 21(11): 2512-2520, 2019 11.
Article in English | MEDLINE | ID: mdl-31105274

ABSTRACT

PURPOSE: Limb-girdle muscular dystrophies (LGMD) are a genetically heterogeneous category of autosomal inherited muscle diseases. Many genes causing LGMD have been identified, and clinical trials are beginning for treatment of some genetic subtypes. However, even with the gene-level mechanisms known, it is still difficult to get a robust and generalizable prevalence estimation for each subtype due to the limited amount of epidemiology data and the low incidence of LGMDs. METHODS: Taking advantage of recently published exome and genome sequencing data from the general population, we used a Bayesian method to develop a robust disease prevalence estimator. RESULTS: This method was applied to nine recessive LGMD subtypes. The estimated disease prevalence calculated by this method was largely comparable with published estimates from epidemiological studies; however, it highlighted instances of possible underdiagnosis for LGMD2B and 2L. CONCLUSION: The increasing size of aggregated population variant databases will allow for robust and reproducible prevalence estimates of recessive disease, which is critical for the strategic design and prioritization of clinical trials.


Subject(s)
Muscular Dystrophies, Limb-Girdle/epidemiology , Muscular Dystrophies, Limb-Girdle/genetics , Bayes Theorem , Chromosome Mapping , Databases, Genetic , Exome , Female , Humans , Male , Mutation , Prevalence
16.
Genet Med ; 21(12): 2723-2733, 2019 12.
Article in English | MEDLINE | ID: mdl-31239556

ABSTRACT

PURPOSE: Pathogenic variants in the chromatin organizer CTCF were previously reported in seven individuals with a neurodevelopmental disorder (NDD). METHODS: Through international collaboration we collected data from 39 subjects with variants in CTCF. We performed transcriptome analysis on RNA from blood samples and utilized Drosophila melanogaster to investigate the impact of Ctcf dosage alteration on nervous system development and function. RESULTS: The individuals in our cohort carried 2 deletions, 8 likely gene-disruptive, 2 splice-site, and 20 different missense variants, most of them de novo. Two cases were familial. The associated phenotype was of variable severity extending from mild developmental delay or normal IQ to severe intellectual disability. Feeding difficulties and behavioral abnormalities were common, and variable other findings including growth restriction and cardiac defects were observed. RNA-sequencing in five individuals identified 3828 deregulated genes enriched for known NDD genes and biological processes such as transcriptional regulation. Ctcf dosage alteration in Drosophila resulted in impaired gross neurological functioning and learning and memory deficits. CONCLUSION: We significantly broaden the mutational and clinical spectrum ofCTCF-associated NDDs. Our data shed light onto the functional role of CTCF by identifying deregulated genes and show that Ctcf alterations result in nervous system defects in Drosophila.


Subject(s)
CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Neurodevelopmental Disorders/genetics , Animals , Child , Chromatin/genetics , Chromatin/metabolism , Developmental Disabilities/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Humans , Intellectual Disability/genetics , Male , Mutation/genetics , Mutation, Missense/genetics , Neurodevelopmental Disorders/metabolism , Transcription Factors/genetics , Exome Sequencing/methods , Young Adult
17.
J Inherit Metab Dis ; 42(3): 553-564, 2019 05.
Article in English | MEDLINE | ID: mdl-30746764

ABSTRACT

SLC35A2-CDG is caused by mutations in the X-linked SLC35A2 gene encoding the UDP-galactose transporter. SLC35A2 mutations lead to hypogalactosylation of N-glycans. SLC35A2-CDG is characterized by severe neurological symptoms and, in many patients, early-onset epileptic encephalopathy. In view of the diagnostic challenges, we studied the clinical, neuroradiological, and biochemical features of 15 patients (11 females and 4 males) with SLC35A2-CDG from various centers. We describe nine novel pathogenic variations in SLC35A2. All affected individuals presented with a global developmental delay, and hypotonia, while 70% were nonambulatory. Epilepsy was present in 80% of the patients, and in EEG hypsarrhythmia and findings consistent with epileptic encephalopathy were frequently seen. The most common brain MRI abnormality was cerebral atrophy with delayed myelination and multifocal inhomogeneous abnormal patchy white matter hyperintensities, which seemed to be nonprogressive. Thin corpus callosum was also common, and all the patients had a corpus callosum shorter than normal for their age. Variable dysmorphic features and growth deficiency were noted. Biochemically, normal mucin type O-glycosylation and lipid glycosylation were found, while transferrin mass spectrometry was found to be more specific in the identification of SLC35A2-CDG, as compared to routine screening tests. Although normal glycosylation studies together with clinical variability and genetic results complicate the diagnosis of SLC35A2-CDG, our data indicate that the combination of these three elements can support the pathogenicity of mutations in SLC35A2.


Subject(s)
Brain Diseases/pathology , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/pathology , Monosaccharide Transport Proteins/genetics , Spasms, Infantile/pathology , Adolescent , Atrophy , Child , Child, Preschool , Female , Glycosylation , Humans , Infant , Internationality , Magnetic Resonance Imaging , Male , Mass Spectrometry , Mutation , Young Adult
18.
Am J Med Genet A ; 173(6): 1620-1624, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28425223

ABSTRACT

Congenital disorders of glycosylation (CDG) are caused by defective glycosylation of proteins and lipids. PMM2-CDG is the most common subtype among the CDG. The severity of PMM2-CDG is variable. Patients often have a recognizable phenotype with neurological and multisystem symptoms that might cause early death. We report six patients from three families who are diagnosed with a clinically mild PMM2-CDG and have normal cognitive development. All these patients had delayed gross motor skills with mild-to-moderate neurological findings. Cerebellar hypoplasia was detected in all siblings for whom brain MRI was performed. In 5/6 children the Wechsler Intelligence Scale for Children (WISC) showed normal cognitive development with full scale IQ scores ranging from borderline to average. Four patients were diagnosed with PMM2-CDG at the age of 8 years or later as their neurological symptoms were quite mild and they had been able to participate in regular school programs. We report patients with p.Val231Met/p.Arg239Trp and p.Ile120Thr/p.Gly228Cys genotypes which may cause milder variants of PMM2-CDG.


Subject(s)
Cerebellum/abnormalities , Cognition/physiology , Congenital Disorders of Glycosylation/genetics , Nervous System Malformations/genetics , Phosphotransferases (Phosphomutases)/genetics , Adolescent , Cerebellum/diagnostic imaging , Cerebellum/physiopathology , Child , Child, Preschool , Congenital Disorders of Glycosylation/diagnostic imaging , Congenital Disorders of Glycosylation/physiopathology , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Female , Genotype , Humans , Magnetic Resonance Imaging , Male , Mutation , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/physiopathology , Phenotype
19.
Am J Med Genet A ; 170(8): 2173-6, 2016 08.
Article in English | MEDLINE | ID: mdl-27250579

ABSTRACT

The CACNA1A gene encodes the transmembrane pore-forming alpha-1A subunit of the Cav 2.1 P/Q-type voltage-gated calcium channel. Several heterozygous mutations within this gene, including nonsense mutations, missense mutations, and expansion of cytosine-adenine-guanine repeats, are known to cause three allelic autosomal dominant conditions-episodic ataxia type 2, familial hemiplegic migraine type 1, and spinocerebellar ataxia type 6. An association with epilepsy and CACNA1A mutations has also been described. However, the link with epileptic encephalopathies has emerged only recently. Here we describe two patients, sister and brother, with compound heterozygous mutations in CACNA1A. Exome sequencing detected biallelic mutations in CACNA1A: A missense mutation c.4315T>A (p.Trp1439Arg) in exon 27, and a seven base pair deletion c.472_478delGCCTTCC (p.Ala158Thrfs*6) in exon 3. Both patients were normal at birth, but developed daily recurrent seizures in early infancy with concomitant extreme muscular hypotonia, hypokinesia, and global developmental delay. The brain MRI images showed progressive cerebral, cerebellar, and optic nerve atrophy. At the age of 5, both patients were blind and bedridden with a profound developmental delay. The elder sister died at that age. Their parents and two siblings were heterozygotes for one of those pathogenic mutations and expressed a milder phenotype. Both of them have intellectual disability and in addition the mother has adult onset cerebellar ataxia with a slowly progressive cerebellar atrophy. Compound heterozygous mutations in the CACNA1A gene presumably cause early onset epileptic encephalopathy, and progressive cerebral, cerebellar and optic nerve atrophy with reduced lifespan. © 2016 Wiley Periodicals, Inc.


Subject(s)
Alleles , Brain Diseases/genetics , Calcium Channels/genetics , Cerebellum/abnormalities , Epilepsy/genetics , Malformations of Cortical Development/genetics , Mutation , Optic Atrophy/genetics , Brain Diseases/diagnosis , Electrocardiography , Exome , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Magnetic Resonance Imaging , Male , Malformations of Cortical Development/diagnosis , Optic Atrophy/diagnosis , Pedigree , Siblings
20.
Am J Med Genet A ; 167A(8): 1913-5, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25847626

ABSTRACT

Recently, a novel autosomal recessive developmental delay-macrocephaly syndrome was described caused by homozygous or compound heterozygous mutations in the KPTN gene. All reported patients belonged to one large Amish kindred. We report on the second case of KPTN-related syndrome in two Estonian adult sibs. The brother and sister both have macrocephaly and moderate intellectual disability, and their verbal abilities are more affected than motor development. No notable minor anomalies are present. Behavioral problems and a few episodes of seizures were reported in the brother. Whole exome sequencing carried out from the brother's DNA sample identified homozygous one-nucleotide frameshift duplication c.665dupA (p.Q222fs) in the KPTN gene. Homozygosity of both affected sibs and heterozygosity of parents were confirmed by Sanger sequencing. Thus, we confirm the pathogenicity of KPTN mutations and further delineate the novel developmental delay-macrocephaly syndrome. We also support the hypothesis that KPTN-related syndrome is not restricted to the Amish population.


Subject(s)
Intellectual Disability/genetics , Megalencephaly/genetics , Microfilament Proteins/genetics , Mutation , Adult , Female , Humans , Male , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL