Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hum Genet ; 110(7): 1068-1085, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37352860

ABSTRACT

ERI1 is a 3'-to-5' exoribonuclease involved in RNA metabolic pathways including 5.8S rRNA processing and turnover of histone mRNAs. Its biological and medical significance remain unclear. Here, we uncover a phenotypic dichotomy associated with bi-allelic ERI1 variants by reporting eight affected individuals from seven unrelated families. A severe spondyloepimetaphyseal dysplasia (SEMD) was identified in five affected individuals with missense variants but not in those with bi-allelic null variants, who showed mild intellectual disability and digital anomalies. The ERI1 missense variants cause a loss of the exoribonuclease activity, leading to defective trimming of the 5.8S rRNA 3' end and a decreased degradation of replication-dependent histone mRNAs. Affected-individual-derived induced pluripotent stem cells (iPSCs) showed impaired in vitro chondrogenesis with downregulation of genes regulating skeletal patterning. Our study establishes an entity previously unreported in OMIM and provides a model showing a more severe effect of missense alleles than null alleles within recessive genotypes, suggesting a key role of ERI1-mediated RNA metabolism in human skeletal patterning and chondrogenesis.


Subject(s)
Exoribonucleases , Histones , Humans , Exoribonucleases/genetics , Histones/genetics , Mutation, Missense/genetics , RNA, Ribosomal, 5.8S , RNA , RNA, Messenger/genetics
2.
Am J Hum Genet ; 110(5): 774-789, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37054711

ABSTRACT

The Integrator complex is a multi-subunit protein complex that regulates the processing of nascent RNAs transcribed by RNA polymerase II (RNAPII), including small nuclear RNAs, enhancer RNAs, telomeric RNAs, viral RNAs, and protein-coding mRNAs. Integrator subunit 11 (INTS11) is the catalytic subunit that cleaves nascent RNAs, but, to date, mutations in this subunit have not been linked to human disease. Here, we describe 15 individuals from 10 unrelated families with bi-allelic variants in INTS11 who present with global developmental and language delay, intellectual disability, impaired motor development, and brain atrophy. Consistent with human observations, we find that the fly ortholog of INTS11, dIntS11, is essential and expressed in the central nervous systems in a subset of neurons and most glia in larval and adult stages. Using Drosophila as a model, we investigated the effect of seven variants. We found that two (p.Arg17Leu and p.His414Tyr) fail to rescue the lethality of null mutants, indicating that they are strong loss-of-function variants. Furthermore, we found that five variants (p.Gly55Ser, p.Leu138Phe, p.Lys396Glu, p.Val517Met, and p.Ile553Glu) rescue lethality but cause a shortened lifespan and bang sensitivity and affect locomotor activity, indicating that they are partial loss-of-function variants. Altogether, our results provide compelling evidence that integrity of the Integrator RNA endonuclease is critical for brain development.


Subject(s)
Drosophila Proteins , Nervous System Diseases , Adult , Animals , Humans , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Mutation/genetics , RNA, Messenger
3.
Hum Mol Genet ; 32(9): 1457-1465, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36458889

ABSTRACT

Mosaic variants in the PIK3CA gene, encoding the catalytic subunit of phosphoinositide 3-kinase (PI3K), produce constitutive PI3K activation, which causes PIK3CA-related overgrowth spectrum disorders. To date, fewer than 20 patients have been described with germline alterations in PIK3CA. In this study, we describe three unrelated individuals with overgrowth and germline PIK3CA variants. These variants were discovered through whole-exome sequencing and confirmed as germline by testing multiple tissue types, when available. Functional analysis using Patient 1's fibroblast cell line and two previously reported patients' cell lines showed increased phosphorylation of AKT during cellular starvation revealing constitutive activation of the phosphoinositide-3-kinase/protein kinase B/mechanistic target of rapamycin (PI3K/AKT/mTOR) pathway. Alternatively, stimulation of the cells by fetal bovine serum produced a reduced response, indicating an activated status of the PI3K complex reducing the pathway response to further external stimulation. Additional studies utilizing Biolog Phenotype Microarray technology indicated reduced energy production when cells were exposed to growth factors stimulating the PI3K/AKT/mTOR pathway, confirming the trend observed in the AKT phosphorylation test after stimulation. Furthermore, treatment with inhibitors of the PI3K/AKT/mTOR pathway rescued the normal energy response in the patients' cells. Collectively, these data demonstrate that disease-causing germline PIK3CA variants have a functional consequence, similar to mosaic variants in the PI3K/AKT/mTOR pathway.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Genetic Diseases, Inborn , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Germ Cells/metabolism , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/physiopathology , Germ-Line Mutation , Phosphorylation
4.
Clin Genet ; 103(2): 226-230, 2023 02.
Article in English | MEDLINE | ID: mdl-36189577

ABSTRACT

NSD2 dimethylates histone H3 at lysine 36 (H3K36me2) and is located in the Wolf-Hirschhorn syndrome (WHS) critical region. Recent descriptions have delineated loss-of-function (LoF) variants in NSD2 with a distinct disorder. The oncogenic missense variant p.Glu1099Lys occurs somatically in leukemia and has a gain-of-function (GoF) effect. We describe two individuals carrying p.Glu1099Lys as heterozygous de novo germline variant identified by exome sequencing (ES) of blood DNA and subsequently confirmed in two ectodermal tissues. Clinically, these individuals are characterized by intellectual disability, coarse/ square facial gestalt, abnormalities of the hands, and organomegaly. Public cell lines with NSD2 GoF variants had increased K36me2, DNA promoter methylation, and dysregulated RNA expression. NSD2 GoF caused by p.Glu1099Lys is associated with a novel phenotype different from WHS and Rauch-Steindl syndrome (RAUST).


Subject(s)
Repressor Proteins , Wolf-Hirschhorn Syndrome , Humans , Repressor Proteins/genetics , Gain of Function Mutation , Histones/genetics , Histones/metabolism , Wolf-Hirschhorn Syndrome/genetics , DNA
5.
Brain ; 145(9): 3274-3287, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35769015

ABSTRACT

Reelin, a large extracellular protein, plays several critical roles in brain development and function. It is encoded by RELN, first identified as the gene disrupted in the reeler mouse, a classic neurological mutant exhibiting ataxia, tremors and a 'reeling' gait. In humans, biallelic variants in RELN have been associated with a recessive lissencephaly variant with cerebellar hypoplasia, which matches well with the homozygous mouse mutant that has abnormal cortical structure, small hippocampi and severe cerebellar hypoplasia. Despite the large size of the gene, only 11 individuals with RELN-related lissencephaly with cerebellar hypoplasia from six families have previously been reported. Heterozygous carriers in these families were briefly reported as unaffected, although putative loss-of-function variants are practically absent in the population (probability of loss of function intolerance = 1). Here we present data on seven individuals from four families with biallelic and 13 individuals from seven families with monoallelic (heterozygous) variants of RELN and frontotemporal or temporal-predominant lissencephaly variant. Some individuals with monoallelic variants have moderate frontotemporal lissencephaly, but with normal cerebellar structure and intellectual disability with severe behavioural dysfunction. However, one adult had abnormal MRI with normal intelligence and neurological profile. Thorough literature analysis supports a causal role for monoallelic RELN variants in four seemingly distinct phenotypes including frontotemporal lissencephaly, epilepsy, autism and probably schizophrenia. Notably, we observed a significantly higher proportion of loss-of-function variants in the biallelic compared to the monoallelic cohort, where the variant spectrum included missense and splice-site variants. We assessed the impact of two canonical splice-site variants observed as biallelic or monoallelic variants in individuals with moderately affected or normal cerebellum and demonstrated exon skipping causing in-frame loss of 46 or 52 amino acids in the central RELN domain. Previously reported functional studies demonstrated severe reduction in overall RELN secretion caused by heterozygous missense variants p.Cys539Arg and p.Arg3207Cys associated with lissencephaly suggesting a dominant-negative effect. We conclude that biallelic variants resulting in complete absence of RELN expression are associated with a consistent and severe phenotype that includes cerebellar hypoplasia. However, reduced expression of RELN remains sufficient to maintain nearly normal cerebellar structure. Monoallelic variants are associated with incomplete penetrance and variable expressivity even within the same family and may have dominant-negative effects. Reduced RELN secretion in heterozygous individuals affects only cortical structure whereas the cerebellum remains intact. Our data expand the spectrum of RELN-related neurodevelopmental disorders ranging from lethal brain malformations to adult phenotypes with normal brain imaging.


Subject(s)
Lissencephaly , Reelin Protein , Adult , Cerebellum/abnormalities , Child , Developmental Disabilities/genetics , Humans , Lissencephaly/complications , Mutation , Nervous System Malformations , Reelin Protein/genetics
6.
Am J Hum Genet ; 105(4): 844-853, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31585108

ABSTRACT

Lissencephaly is a severe brain malformation in which failure of neuronal migration results in agyria or pachygyria and in which the brain surface appears unusually smooth. It is often associated with microcephaly, profound intellectual disability, epilepsy, and impaired motor abilities. Twenty-two genes are associated with lissencephaly, accounting for approximately 80% of disease. Here we report on 12 individuals with a unique form of lissencephaly; these individuals come from eight unrelated families and have bi-allelic mutations in APC2, encoding adenomatous polyposis coli protein 2. Brain imaging studies demonstrate extensive posterior predominant lissencephaly, similar to PAFAH1B1-associated lissencephaly, as well as co-occurrence of subcortical heterotopia posterior to the caudate nuclei, "ribbon-like" heterotopia in the posterior frontal region, and dysplastic in-folding of the mesial occipital cortex. The established role of APC2 in integrating the actin and microtubule cytoskeletons to mediate cellular morphological changes suggests shared function with other lissencephaly-encoded cytoskeletal proteins such as α-N-catenin (CTNNA2) and platelet-activating factor acetylhydrolase 1b regulatory subunit 1 (PAFAH1B1, also known as LIS1). Our findings identify APC2 as a radiographically distinguishable recessive form of lissencephaly.


Subject(s)
Alleles , Classical Lissencephalies and Subcortical Band Heterotopias/genetics , Cytoskeletal Proteins/genetics , Developmental Disabilities/genetics , Lissencephaly/genetics , Female , Humans , Male , Pedigree
7.
Am J Hum Genet ; 105(1): 213-220, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31230721

ABSTRACT

De novo variants represent a significant cause of neurodevelopmental delay and intellectual disability. A genetic basis can be identified in only half of individuals who have neurodevelopmental disorders (NDDs); this indicates that additional causes need to be elucidated. We compared the frequency of de novo variants in patient-parent trios with (n = 2,030) versus without (n = 2,755) NDDs. We identified de novo variants in TAOK1 (thousand and one [TAO] amino acid kinase 1), which encodes the serine/threonine-protein kinase TAO1, in three individuals with NDDs but not in persons who did not have NDDs. Through further screening and the use of GeneMatcher, five additional individuals with NDDs were found to have de novo variants. All eight variants were absent from gnomAD (Genome Aggregation Database). The variant carriers shared a non-specific phenotype of developmental delay, and six individuals had additional muscular hypotonia. We established a fibroblast line of one mutation carrier, and we demonstrated that reduced mRNA levels of TAOK1 could be increased upon cycloheximide treatment. These results indicate nonsense-mediated mRNA decay. Further, there was neither detectable phosphorylated TAO1 kinase nor phosphorylated tau in these cells, and mitochondrial morphology was altered. Knockdown of the ortholog gene Tao1 (Tao, CG14217) in Drosophila resulted in delayed early development. The majority of the Tao1-knockdown flies did not survive beyond the third instar larval stage. When compared to control flies, Tao1 knockdown flies revealed changed morphology of the ventral nerve cord and the neuromuscular junctions as well as a decreased number of endings (boutons). Furthermore, mitochondria in mutant flies showed altered distribution and decreased size in axons of motor neurons. Thus, we provide compelling evidence that de novo variants in TAOK1 cause NDDs.


Subject(s)
Drosophila melanogaster/growth & development , Exome/genetics , Mutation , Neurodevelopmental Disorders/etiology , Protein Serine-Threonine Kinases/genetics , Animals , Child , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Heterozygote , Humans , Male , Neurodevelopmental Disorders/pathology , Phenotype , Exome Sequencing
8.
Am J Hum Genet ; 104(2): 203-212, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30612693

ABSTRACT

Using exome sequencing, we have identified de novo variants in MAPK8IP3 in 13 unrelated individuals presenting with an overlapping phenotype of mild to severe intellectual disability. The de novo variants comprise six missense variants, three of which are recurrent, and three truncating variants. Brain anomalies such as perisylvian polymicrogyria, cerebral or cerebellar atrophy, and hypoplasia of the corpus callosum were consistent among individuals harboring recurrent de novo missense variants. MAPK8IP3 has been shown to be involved in the retrograde axonal-transport machinery, but many of its specific functions are yet to be elucidated. Using the CRISPR-Cas9 system to target six conserved amino acid positions in Caenorhabditis elegans, we found that two of the six investigated human alterations led to a significantly elevated density of axonal lysosomes, and five variants were associated with adverse locomotion. Reverse-engineering normalized the observed adverse effects back to wild-type levels. Combining genetic, phenotypic, and functional findings, as well as the significant enrichment of de novo variants in MAPK8IP3 within our total cohort of 27,232 individuals who underwent exome sequencing, we implicate de novo variants in MAPK8IP3 as a cause of a neurodevelopmental disorder with intellectual disability and variable brain anomalies.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Brain/abnormalities , Brain/metabolism , Intellectual Disability/genetics , Mutation , Nerve Tissue Proteins/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Animals , Brain/diagnostic imaging , CRISPR-Cas Systems , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Child , Child, Preschool , Computer Simulation , Female , Humans , Intellectual Disability/diagnostic imaging , Locomotion , Lysosomes/metabolism , Male , Models, Molecular , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Exome Sequencing , Young Adult
9.
Am J Hum Genet ; 105(5): 987-995, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31587868

ABSTRACT

NKAP is a ubiquitously expressed nucleoplasmic protein that is currently known as a transcriptional regulatory molecule via its interaction with HDAC3 and spliceosomal proteins. Here, we report a disorder of transcriptional regulation due to missense mutations in the X chromosome gene, NKAP. These mutations are clustered in the C-terminal region of NKAP where NKAP interacts with HDAC3 and post-catalytic spliceosomal complex proteins. Consistent with a role for the C-terminal region of NKAP in embryogenesis, nkap mutant zebrafish with a C-terminally truncated NKAP demonstrate severe developmental defects. The clinical features of affected individuals are highly conserved and include developmental delay, hypotonia, joint contractures, behavioral abnormalities, Marfanoid habitus, and scoliosis. In affected cases, transcriptome analysis revealed the presence of a unique transcriptome signature, which is characterized by the downregulation of long genes with higher exon numbers. These observations indicate the critical role of NKAP in transcriptional regulation and demonstrate that perturbations of the C-terminal region lead to developmental defects in both humans and zebrafish.


Subject(s)
Cognitive Dysfunction/genetics , Mutation, Missense/genetics , Repressor Proteins/genetics , Transcription, Genetic/genetics , Amino Acid Sequence , Animals , Down-Regulation/genetics , Exons/genetics , Gene Expression Regulation/genetics , Genes, X-Linked/genetics , Histone Deacetylases/genetics , Humans , Sequence Alignment , Transcriptome/genetics , Zebrafish/genetics
10.
Genet Med ; 23(4): 661-668, 2021 04.
Article in English | MEDLINE | ID: mdl-33420346

ABSTRACT

PURPOSE: To identify novel genes associated with intellectual disability (ID) in four unrelated families. METHODS: Here, through exome sequencing and international collaboration, we report eight individuals from four unrelated families of diverse geographic origin with biallelic loss-of-function variants in UBE4A. RESULTS: Eight evaluated individuals presented with syndromic intellectual disability and global developmental delay. Other clinical features included hypotonia, short stature, seizures, and behavior disorder. Characteristic features were appreciated in some individuals but not all; in some cases, features became more apparent with age. We demonstrated that UBE4A loss-of-function variants reduced RNA expression and protein levels in clinical samples. Mice generated to mimic patient-specific Ube4a loss-of-function variant exhibited muscular and neurological/behavioral abnormalities, some of which are suggestive of the clinical abnormalities seen in the affected individuals. CONCLUSION: These data indicate that biallelic loss-of-function variants in UBE4A cause a novel intellectual disability syndrome, suggesting that UBE4A enzyme activity is required for normal development and neurological function.


Subject(s)
Dwarfism , Intellectual Disability , Ubiquitin-Protein Ligases/genetics , Animals , Child , Developmental Disabilities/genetics , Humans , Intellectual Disability/genetics , Mice , Muscle Hypotonia , Phenotype , Syndrome , Exome Sequencing
11.
Am J Med Genet A ; 185(9): 2719-2738, 2021 09.
Article in English | MEDLINE | ID: mdl-34087052

ABSTRACT

Cyclin D2 (CCND2) is a critical cell cycle regulator and key member of the cyclin D2-CDK4 (DC) complex. De novo variants of CCND2 clustering in the distal part of the protein have been identified as pathogenic causes of brain overgrowth (megalencephaly, MEG) and severe cortical malformations in children including the megalencephaly-polymicrogyria-polydactyly-hydrocephalus (MPPH) syndrome. Megalencephaly-associated CCND2 variants are localized to the terminal exon and result in accumulation of degradation-resistant protein. We identified five individuals from three unrelated families with novel variants in the proximal region of CCND2 associated with microcephaly, mildly simplified cortical gyral pattern, symmetric short stature, and mild developmental delay. Identified variants include de novo frameshift variants and a dominantly inherited stop-gain variant segregating with the phenotype. This is the first reported association between proximal CCND2 variants and microcephaly, to our knowledge. This series expands the phenotypic spectrum of CCND2-related disorders and suggests that distinct classes of CCND2 variants are associated with reciprocal effects on human brain growth (microcephaly and megalencephaly due to possible loss or gain of protein function, respectively), adding to the growing paradigm of inverse phenotypes due to dysregulation of key brain growth genes.


Subject(s)
Brain/abnormalities , Cyclin D2/genetics , Hydrocephalus/pathology , Megalencephaly/pathology , Mutation , Polydactyly/pathology , Polymicrogyria/pathology , Adolescent , Adult , Child , Female , Humans , Hydrocephalus/genetics , Infant , Male , Megalencephaly/genetics , Polydactyly/genetics , Polymicrogyria/genetics
12.
J Hum Genet ; 65(11): 1003-1017, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32788638

ABSTRACT

Mutations in the cytoplasmic dynein 1 heavy chain gene (DYNC1H1) have been identified in rare neuromuscular (NMD) and neurodevelopmental (NDD) disorders such as spinal muscular atrophy with lower extremity dominance (SMALED) and autosomal dominant mental retardation syndrome 13 (MRD13). Phenotypes and genotypes of ten pediatric patients with pathogenic DYNC1H1 variants were analyzed in a multi-center study. Data mining of large-scale genomic variant databases was used to investigate domain-specific vulnerability and conservation of DYNC1H1. We identified ten patients with nine novel mutations in the DYNC1H1 gene. These patients exhibit a broad spectrum of clinical findings, suggesting an overlapping disease manifestation with intermixed phenotypes ranging from neuropathy (peripheral nervous system, PNS) to severe intellectual disability (central nervous system, CNS). Genomic profiling of healthy and patient variant datasets underlines the domain-specific effects of genetic variation in DYNC1H1, specifically on toleration towards missense variants in the linker domain. A retrospective analysis of all published mutations revealed domain-specific genotype-phenotype correlations, i.e., mutations in the dimerization domain with reductions in lower limb strength in DYNC1H1-NMD and motor domain with cerebral malformations in DYNC1H1-NDD. We highlight that the current classification into distinct disease entities does not sufficiently reflect the clinical disease manifestation that clinicians face in the diagnostic work-up of DYNC1H1-related disorders. We propose a novel clinical classification for DYNC1H1-related disorders encompassing a spectrum from DYNC1H1-NMD with an exclusive PNS phenotype to DYNC1H1-NDD with concomitant CNS involvement.


Subject(s)
Brain/diagnostic imaging , Cytoplasmic Dyneins/genetics , Genomics , Muscular Atrophy, Spinal/genetics , Brain/abnormalities , Brain/pathology , DNA Mutational Analysis , Female , Humans , Infant , Lower Extremity/diagnostic imaging , Lower Extremity/pathology , Lower Extremity Deformities, Congenital/diagnostic imaging , Lower Extremity Deformities, Congenital/genetics , Lower Extremity Deformities, Congenital/pathology , Male , Muscular Atrophy, Spinal/classification , Muscular Atrophy, Spinal/diagnostic imaging , Muscular Atrophy, Spinal/pathology , Mutation, Missense/genetics , Phenotype
13.
Am J Med Genet A ; 182(12): 3040-3047, 2020 12.
Article in English | MEDLINE | ID: mdl-33026187

ABSTRACT

Congenital hiatal hernia (HH) is a rare congenital defect and is often described on a sporadic basis, but familial cases have also been reported. The mechanism of development is not well understood, and to our knowledge no specific genetic factors have been implicated to date. We report on seven individuals from two families with 9q22 duplication, who have variably associated features including congenital HH in four individuals. One family had an 1.09 Mb 9q22 duplication, and the other family had an overlapping 2.73 Mb 9q22 duplication. We review the genes in this region and discuss BARX1 (BarH-like homeobox gene 1) as a gene of interest.


Subject(s)
Chromosome Duplication , Chromosomes, Human, Pair 9/genetics , Hernia, Hiatal/pathology , Homeodomain Proteins/genetics , Mutation , Transcription Factors/genetics , Adolescent , Child, Preschool , Female , Hernia, Hiatal/congenital , Hernia, Hiatal/genetics , Humans , Infant , Infant, Newborn , Male , Pedigree
14.
PLoS Genet ; 13(3): e1006683, 2017 03.
Article in English | MEDLINE | ID: mdl-28346496

ABSTRACT

Schinzel-Giedion syndrome (SGS) is a rare developmental disorder characterized by multiple malformations, severe neurological alterations and increased risk of malignancy. SGS is caused by de novo germline mutations clustering to a 12bp hotspot in exon 4 of SETBP1. Mutations in this hotspot disrupt a degron, a signal for the regulation of protein degradation, and lead to the accumulation of SETBP1 protein. Overlapping SETBP1 hotspot mutations have been observed recurrently as somatic events in leukemia. We collected clinical information of 47 SGS patients (including 26 novel cases) with germline SETBP1 mutations and of four individuals with a milder phenotype caused by de novo germline mutations adjacent to the SETBP1 hotspot. Different mutations within and around the SETBP1 hotspot have varying effects on SETBP1 stability and protein levels in vitro and in in silico modeling. Substitutions in SETBP1 residue I871 result in a weak increase in protein levels and mutations affecting this residue are significantly more frequent in SGS than in leukemia. On the other hand, substitutions in residue D868 lead to the largest increase in protein levels. Individuals with germline mutations affecting D868 have enhanced cell proliferation in vitro and higher incidence of cancer compared to patients with other germline SETBP1 mutations. Our findings substantiate that, despite their overlap, somatic SETBP1 mutations driving malignancy are more disruptive to the degron than germline SETBP1 mutations causing SGS. Additionally, this suggests that the functional threshold for the development of cancer driven by the disruption of the SETBP1 degron is higher than for the alteration in prenatal development in SGS. Drawing on previous studies of somatic SETBP1 mutations in leukemia, our results reveal a genotype-phenotype correlation in germline SETBP1 mutations spanning a molecular, cellular and clinical phenotype.


Subject(s)
Abnormalities, Multiple/genetics , Carrier Proteins/genetics , Craniofacial Abnormalities/genetics , Genetic Predisposition to Disease/genetics , Hand Deformities, Congenital/genetics , Hematologic Neoplasms/genetics , Intellectual Disability/genetics , Mutation , Nails, Malformed/genetics , Nuclear Proteins/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Blotting, Western , Carrier Proteins/metabolism , Cell Line , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Child , Child, Preschool , Craniofacial Abnormalities/metabolism , Craniofacial Abnormalities/pathology , Female , Gene Expression Profiling , Genetic Association Studies , Germ-Line Mutation , HEK293 Cells , Hand Deformities, Congenital/metabolism , Hand Deformities, Congenital/pathology , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Infant , Infant, Newborn , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Nails, Malformed/metabolism , Nails, Malformed/pathology , Nuclear Proteins/metabolism , Phenotype
15.
Am J Hum Genet ; 99(5): 1117-1129, 2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27773430

ABSTRACT

Lissencephaly is a malformation of cortical development typically caused by deficient neuronal migration resulting in cortical thickening and reduced gyration. Here we describe a "thin" lissencephaly (TLIS) variant characterized by megalencephaly, frontal predominant pachygyria, intellectual disability, and seizures. Trio-based whole-exome sequencing and targeted re-sequencing identified recessive mutations of CRADD in six individuals with TLIS from four unrelated families of diverse ethnic backgrounds. CRADD (also known as RAIDD) is a death-domain-containing adaptor protein that oligomerizes with PIDD and caspase-2 to initiate apoptosis. TLIS variants cluster in the CRADD death domain, a platform for interaction with other death-domain-containing proteins including PIDD. Although caspase-2 is expressed in the developing mammalian brain, little is known about its role in cortical development. CRADD/caspase-2 signaling is implicated in neurotrophic factor withdrawal- and amyloid-ß-induced dendritic spine collapse and neuronal apoptosis, suggesting a role in cortical sculpting and plasticity. TLIS-associated CRADD variants do not disrupt interactions with caspase-2 or PIDD in co-immunoprecipitation assays, but still abolish CRADD's ability to activate caspase-2, resulting in reduced neuronal apoptosis in vitro. Homozygous Cradd knockout mice display megalencephaly and seizures without obvious defects in cortical lamination, supporting a role for CRADD/caspase-2 signaling in mammalian brain development. Megalencephaly and lissencephaly associated with defective programmed cell death from loss of CRADD function in humans implicate reduced apoptosis as an important pathophysiological mechanism of cortical malformation. Our data suggest that CRADD/caspase-2 signaling is critical for normal gyration of the developing human neocortex and for normal cognitive ability.


Subject(s)
Apoptosis , CRADD Signaling Adaptor Protein/genetics , Caspase 2/metabolism , Cysteine Endopeptidases/metabolism , Lissencephaly/genetics , Megalencephaly/genetics , Neurons/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Caspase 2/genetics , Cell Survival , Cloning, Molecular , Cognition , Cysteine Endopeptidases/genetics , Dendritic Cells/metabolism , Ethnicity/genetics , Genes, Recessive , Genome-Wide Association Study , HEK293 Cells , Humans , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , PC12 Cells , Rats , Signal Transduction
16.
J Transl Med ; 17(1): 351, 2019 10 26.
Article in English | MEDLINE | ID: mdl-31655630

ABSTRACT

BACKGROUND: Biallelic PTPRQ pathogenic variants have been previously reported as causative for autosomal recessive non-syndromic hearing loss. In 2018 the first heterozygous PTPRQ variant has been implicated in the development of autosomal dominant non-syndromic hearing loss (ADNSHL) in a German family. The study presented the only, so far known, PTPRQ pathogenic variant (c.6881G>A) in ADNSHL. It is located in the last PTPRQ coding exon and introduces a premature stop codon (p.Trp2294*). METHODS: A five-generation Polish family with ADNSHL was recruited for the study (n = 14). Thorough audiological, neurotological and imaging studies were carried out to precisely define the phenotype. Genomic DNA was isolated from peripheral blood samples or buccal swabs of available family members. Clinical exome sequencing was conducted for the proband. Family segregation analysis of the identified variants was performed using Sanger sequencing. Single nucleotide polymorphism array on DNA samples from the Polish and the original German family was used for genome-wide linkage analysis. RESULTS: Combining clinical exome sequencing and family segregation analysis, we have identified the same (NM_001145026.2:c.6881G>A, NP_001138498.1:p.Trp2294*) PTPRQ alteration in the Polish ADNSHL family. Using genome-wide linkage analysis, we found that the studied family and the original German family derive from a common ancestor. Deep phenotyping of the affected individuals showed that in contrast to the recessive form, the PTPRQ-related ADNSHL is not associated with vestibular dysfunction. In both families ADNSHL was progressive, affected mainly high frequencies and had a variable age of onset. CONCLUSION: Our data provide the first confirmation of PTPRQ involvement in ADNSHL. The finding strongly reinforces the inclusion of PTPRQ to the small set of genes leading to both autosomal recessive and dominant hearing loss.


Subject(s)
Hearing Loss, Sensorineural/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Adolescent , Adult , Age of Onset , Child , Female , Genes, Dominant , Hearing Loss, Sensorineural/physiopathology , Heterozygote , Humans , Male , Middle Aged , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/physiology , Mutation , Pedigree , Peptide Chain Termination, Translational/genetics , Phenotype , Poland , Polymorphism, Single Nucleotide , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Translational Research, Biomedical , Young Adult
17.
Epilepsia ; 60(6): e63-e66, 2019 06.
Article in English | MEDLINE | ID: mdl-31077350

ABSTRACT

Severe early onset epilepsies are often caused by de novo pathogenic variants. Few studies have reported the frequency of somatic mosaicism in parents of children with severe epileptic encephalopathies. Here we aim to investigate the frequency of mosaicism in the parents of children with epilepsy caused by alleged de novo variants. We tested parental genomic DNA derived from different tissues for 75 cases using targeted next-generation sequencing. Five parents (6.6%) showed mosaicism at minor allele frequencies of 0.8%-29% for the pathogenic variant detected in their offspring. Parental mosaicism was observed in the following genes: SCN1A, SCN2A, SCN8A, and STXBP1. One of the identified parents had epilepsy himself. Our results show that de novo events can occur already in parental tissue and in some cases can be detected in peripheral blood. Consequently, parents affected by low-grade mosaicism are faced with an increased recurrence risk for transmitting the pathogenic variant, compared to the overall recurrence risk for a second affected child estimated at approximately 1%. However, testing for parental somatic mosaicism will help identifying those parents who truly are at higher risk and will significantly improve genetic counseling in the respective families.


Subject(s)
Epilepsy/complications , Epilepsy/genetics , Mosaicism , Adult , Alleles , Child , Child, Preschool , DNA/genetics , Female , Gene Frequency , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Male , Parents , Pedigree , Recurrence , Risk Factors
18.
Hum Mutat ; 39(9): 1226-1237, 2018 09.
Article in English | MEDLINE | ID: mdl-29897170

ABSTRACT

Malan syndrome is an overgrowth disorder described in a limited number of individuals. We aim to delineate the entity by studying a large group of affected individuals. We gathered data on 45 affected individuals with a molecularly confirmed diagnosis through an international collaboration and compared data to the 35 previously reported individuals. Results indicate that height is > 2 SDS in infancy and childhood but in only half of affected adults. Cardinal facial characteristics include long, triangular face, macrocephaly, prominent forehead, everted lower lip, and prominent chin. Intellectual disability is universally present, behaviorally anxiety is characteristic. Malan syndrome is caused by deletions or point mutations of NFIX clustered mostly in exon 2. There is no genotype-phenotype correlation except for an increased risk for epilepsy with 19p13.2 microdeletions. Variants arose de novo, except in one family in which mother was mosaic. Variants causing Malan and Marshall-Smith syndrome can be discerned by differences in the site of stop codon formation. We conclude that Malan syndrome has a well recognizable phenotype that usually can be discerned easily from Marshall-Smith syndrome but rarely there is some overlap. Differentiation from Sotos and Weaver syndrome can be made by clinical evaluation only.


Subject(s)
Abnormalities, Multiple/genetics , Congenital Hypothyroidism/genetics , Craniofacial Abnormalities/genetics , Hand Deformities, Congenital/genetics , Intellectual Disability/genetics , NFI Transcription Factors/genetics , Sotos Syndrome/genetics , Abnormalities, Multiple/physiopathology , Adolescent , Adult , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/physiopathology , Child , Child, Preschool , Chromosome Deletion , Congenital Hypothyroidism/physiopathology , Craniofacial Abnormalities/physiopathology , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Exons/genetics , Female , Hand Deformities, Congenital/physiopathology , Humans , Intellectual Disability/physiopathology , Male , Megalencephaly/genetics , Megalencephaly/physiopathology , Mutation, Missense/genetics , Phenotype , Septo-Optic Dysplasia/genetics , Septo-Optic Dysplasia/physiopathology , Sotos Syndrome/physiopathology , Young Adult
19.
Am J Hum Genet ; 97(2): 343-52, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26235985

ABSTRACT

Intellectual disability (ID) affects approximately 1%-3% of humans with a gender bias toward males. Previous studies have identified mutations in more than 100 genes on the X chromosome in males with ID, but there is less evidence for de novo mutations on the X chromosome causing ID in females. In this study we present 35 unique deleterious de novo mutations in DDX3X identified by whole exome sequencing in 38 females with ID and various other features including hypotonia, movement disorders, behavior problems, corpus callosum hypoplasia, and epilepsy. Based on our findings, mutations in DDX3X are one of the more common causes of ID, accounting for 1%-3% of unexplained ID in females. Although no de novo DDX3X mutations were identified in males, we present three families with segregating missense mutations in DDX3X, suggestive of an X-linked recessive inheritance pattern. In these families, all males with the DDX3X variant had ID, whereas carrier females were unaffected. To explore the pathogenic mechanisms accounting for the differences in disease transmission and phenotype between affected females and affected males with DDX3X missense variants, we used canonical Wnt defects in zebrafish as a surrogate measure of DDX3X function in vivo. We demonstrate a consistent loss-of-function effect of all tested de novo mutations on the Wnt pathway, and we further show a differential effect by gender. The differential activity possibly reflects a dose-dependent effect of DDX3X expression in the context of functional mosaic females versus one-copy males, which reflects the complex biological nature of DDX3X mutations.


Subject(s)
DEAD-box RNA Helicases/genetics , Intellectual Disability/genetics , Mutation, Missense/genetics , Phenotype , Sex Characteristics , Wnt Signaling Pathway/genetics , Amino Acid Substitution/genetics , Animals , Base Sequence , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Exome/genetics , Female , Gene Dosage/genetics , Humans , Intellectual Disability/pathology , Male , Molecular Sequence Data , Sequence Analysis, DNA , Zebrafish
20.
Genet Med ; 20(6): 614-621, 2018 06.
Article in English | MEDLINE | ID: mdl-29309402

ABSTRACT

PurposeHearing loss is genetically extremely heterogeneous, making it suitable for next-generation sequencing (NGS). We identified a four-generation family with nonsyndromic mild to severe hearing loss of the mid- to high frequencies and onset from early childhood to second decade in seven members.MethodsNGS of 66 deafness genes, Sanger sequencing, genome-wide linkage analysis, whole-exome sequencing (WES), semiquantitative reverse-transcriptase polymerase chain reaction.ResultsWe identified a heterozygous nonsense mutation, c.6881G>A (p.Trp2294*), in the last coding exon of PTPRQ. PTPRQ has been linked with recessive (DFNB84A), but not dominant deafness. NGS and Sanger sequencing of all exons (including alternatively spliced 5' and N-scan-predicted exons of a putative "extended" transcript) did not identify a second mutation. The highest logarithm of the odds score was in the PTPRQ-containing region on chromosome 12, and p.Trp2294* cosegregated with hearing loss. WES did not identify other cosegregating candidate variants from the mapped region. PTPRQ expression in patient fibroblasts indicated that the mutant allele escapes nonsense-mediated decay (NMD).ConclusionKnown PTPRQ mutations are recessive and do not affect the C-terminal exon. In contrast to recessive loss-of-function mutations, c.6881G>A transcripts may escape NMD. PTPRQTrp2294* protein would lack only six terminal residues and could exert a dominant-negative effect, a possible explanation for allelic deafness, DFNA73, clinically and genetically distinct from DFNB84A.


Subject(s)
Deafness/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Amino Acid Sequence , Codon, Nonsense/genetics , Exome/genetics , Exons/genetics , Family , Female , Genetic Linkage , Genome-Wide Association Study , Hearing Loss/genetics , Humans , Male , Mutation , Pedigree , Exome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL