Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Publication year range
1.
Cell ; 176(3): 505-519.e22, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30612738

ABSTRACT

Genomic instability can be a hallmark of both human genetic disease and cancer. We identify a deleterious UBQLN4 mutation in families with an autosomal recessive syndrome reminiscent of genome instability disorders. UBQLN4 deficiency leads to increased sensitivity to genotoxic stress and delayed DNA double-strand break (DSB) repair. The proteasomal shuttle factor UBQLN4 is phosphorylated by ATM and interacts with ubiquitylated MRE11 to mediate early steps of homologous recombination-mediated DSB repair (HRR). Loss of UBQLN4 leads to chromatin retention of MRE11, promoting non-physiological HRR activity in vitro and in vivo. Conversely, UBQLN4 overexpression represses HRR and favors non-homologous end joining. Moreover, we find UBQLN4 overexpressed in aggressive tumors. In line with an HRR defect in these tumors, UBQLN4 overexpression is associated with PARP1 inhibitor sensitivity. UBQLN4 therefore curtails HRR activity through removal of MRE11 from damaged chromatin and thus offers a therapeutic window for PARP1 inhibitor treatment in UBQLN4-overexpressing tumors.


Subject(s)
Carrier Proteins/genetics , Nuclear Proteins/genetics , Carrier Proteins/metabolism , Chromatin/metabolism , DNA , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Female , Genomic Instability , Germ-Line Mutation , Homologous Recombination , Humans , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Male , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/metabolism , Primary Cell Culture , Recombinational DNA Repair
2.
Hum Genet ; 143(1): 71-84, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38117302

ABSTRACT

Coffin-Siris syndrome (CSS) is a rare multisystemic autosomal dominant disorder. Since 2012, alterations in genes of the SWI/SNF complex were identified as the molecular basis of CSS, studying largely pediatric cohorts. Therefore, there is a lack of information on the phenotype in adulthood, particularly on the clinical outcome in adulthood and associated risks. In an international collaborative effort, data from 35 individuals ≥ 18 years with a molecularly ascertained CSS diagnosis (variants in ARID1B, ARID2, SMARCA4, SMARCB1, SMARCC2, SMARCE1, SOX11, BICRA) using a comprehensive questionnaire was collected. Our results indicate that overweight and obesity are frequent in adults with CSS. Visual impairment, scoliosis, and behavioral anomalies are more prevalent than in published pediatric or mixed cohorts. Cognitive outcomes range from profound intellectual disability (ID) to low normal IQ, with most individuals having moderate ID. The present study describes the first exclusively adult cohort of CSS individuals. We were able to delineate some features of CSS that develop over time and have therefore been underrepresented in previously reported largely pediatric cohorts, and provide recommendations for follow-up.


Subject(s)
Abnormalities, Multiple , Face/abnormalities , Hand Deformities, Congenital , Intellectual Disability , Micrognathism , Adult , Humans , Child , Intellectual Disability/genetics , Intellectual Disability/diagnosis , Abnormalities, Multiple/genetics , Abnormalities, Multiple/diagnosis , Micrognathism/genetics , Micrognathism/diagnosis , Hand Deformities, Congenital/genetics , Neck/abnormalities , Phenotype , DNA Helicases/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics
3.
Am J Hum Genet ; 106(6): 830-845, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32442410

ABSTRACT

SOX6 belongs to a family of 20 SRY-related HMG-box-containing (SOX) genes that encode transcription factors controlling cell fate and differentiation in many developmental and adult processes. For SOX6, these processes include, but are not limited to, neurogenesis and skeletogenesis. Variants in half of the SOX genes have been shown to cause severe developmental and adult syndromes, referred to as SOXopathies. We here provide evidence that SOX6 variants also cause a SOXopathy. Using clinical and genetic data, we identify 19 individuals harboring various types of SOX6 alterations and exhibiting developmental delay and/or intellectual disability; the individuals are from 17 unrelated families. Additional, inconstant features include attention-deficit/hyperactivity disorder (ADHD), autism, mild facial dysmorphism, craniosynostosis, and multiple osteochondromas. All variants are heterozygous. Fourteen are de novo, one is inherited from a mosaic father, and four offspring from two families have a paternally inherited variant. Intragenic microdeletions, balanced structural rearrangements, frameshifts, and nonsense variants are predicted to inactivate the SOX6 variant allele. Four missense variants occur in residues and protein regions highly conserved evolutionarily. These variants are not detected in the gnomAD control cohort, and the amino acid substitutions are predicted to be damaging. Two of these variants are located in the HMG domain and abolish SOX6 transcriptional activity in vitro. No clear genotype-phenotype correlations are found. Taken together, these findings concur that SOX6 haploinsufficiency leads to a neurodevelopmental SOXopathy that often includes ADHD and abnormal skeletal and other features.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Craniosynostoses/genetics , Neurodevelopmental Disorders/genetics , Osteochondroma/genetics , SOXD Transcription Factors/genetics , Active Transport, Cell Nucleus , Adolescent , Amino Acid Sequence , Base Sequence , Brain/embryology , Brain/growth & development , Brain/metabolism , Child , Child, Preschool , Computer Simulation , Female , Genomic Structural Variation/genetics , Humans , Infant , Male , Mutation, Missense , Neurodevelopmental Disorders/diagnosis , RNA-Seq , SOXD Transcription Factors/chemistry , SOXD Transcription Factors/metabolism , Syndrome , Transcription, Genetic , Transcriptome , Translocation, Genetic/genetics
4.
Clin Genet ; 103(4): 484-491, 2023 04.
Article in English | MEDLINE | ID: mdl-36576126

ABSTRACT

Protein translation is an essential cellular process and dysfunctional protein translation causes various neurodevelopmental disorders. The eukaryotic translation elongation factor 1A (eEF1A) delivers aminoacyl-tRNA to the ribosome, while the eEF1B complex acts as a guanine exchange factor (GEF) of GTP for GDP indirectly catalyzing the release of eEF1A from the ribosome. The gene EEF1D encodes the eEF1Bδ subunit of the eEF1B complex. EEF1D is alternatively spliced giving rise to one long and three short isoforms. Two different homozygous, truncating variants in EEF1D had been associated with severe intellectual disability and microcephaly in two families. The published variants only affect the long isoform of EEF1D that acts as a transcription factor of heat shock element proteins. By exome sequencing, we identified two different homozygous variants in EEF1D in two families with severe developmental delay, severe microcephaly, spasticity, and failure to thrive with optic atrophy, poor feeding, and recurrent aspiration pneumonia. The EEF1D variants reported in this study are localized in the C-terminal GEF domain, suggesting that a disturbed protein translation machinery might contribute to the neurodevelopmental phenotype. Pathogenic variants localized in both the alternatively spliced domain or the GEF domain of EEF1D cause a severe neurodevelopmental disorder with microcephaly and spasticity.


Subject(s)
Microcephaly , Neurodevelopmental Disorders , Humans , Guanine , Guanine Nucleotide Exchange Factors/genetics , Neurodevelopmental Disorders/genetics , Protein Isoforms/genetics , Peptide Elongation Factor 1
5.
Am J Hum Genet ; 102(3): 468-479, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29429572

ABSTRACT

Variants affecting the function of different subunits of the BAF chromatin-remodelling complex lead to various neurodevelopmental syndromes, including Coffin-Siris syndrome. Furthermore, variants in proteins containing PHD fingers, motifs recognizing specific histone tail modifications, have been associated with several neurological and developmental-delay disorders. Here, we report eight heterozygous de novo variants (one frameshift, two splice site, and five missense) in the gene encoding the BAF complex subunit double plant homeodomain finger 2 (DPF2). Affected individuals share common clinical features described in individuals with Coffin-Siris syndrome, including coarse facial features, global developmental delay, intellectual disability, speech impairment, and hypoplasia of fingernails and toenails. All variants occur within the highly conserved PHD1 and PHD2 motifs. Moreover, missense variants are situated close to zinc binding sites and are predicted to disrupt these sites. Pull-down assays of recombinant proteins and histone peptides revealed that a subset of the identified missense variants abolish or impaire DPF2 binding to unmodified and modified H3 histone tails. These results suggest an impairment of PHD finger structural integrity and cohesion and most likely an aberrant recognition of histone modifications. Furthermore, the overexpression of these variants in HEK293 and COS7 cell lines was associated with the formation of nuclear aggregates and the recruitment of both wild-type DPF2 and BRG1 to these aggregates. Expression analysis of truncating variants found in the affected individuals indicated that the aberrant transcripts escape nonsense-mediated decay. Altogether, we provide compelling evidence that de novo variants in DPF2 cause Coffin-Siris syndrome and propose a dominant-negative mechanism of pathogenicity.


Subject(s)
Abnormalities, Multiple/genetics , DNA-Binding Proteins/genetics , Face/abnormalities , Hand Deformities, Congenital/genetics , Intellectual Disability/genetics , Micrognathism/genetics , Mutation/genetics , Neck/abnormalities , Protein Subunits/genetics , Adolescent , Amino Acid Sequence , Animals , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , DNA-Binding Proteins/chemistry , Facies , Female , HEK293 Cells , Histones/metabolism , Humans , Male , Phenotype , Transcription Factors
6.
Am J Hum Genet ; 102(6): 1195-1203, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29861108

ABSTRACT

Next-generation sequencing is a powerful tool for the discovery of genes related to neurodevelopmental disorders (NDDs). Here, we report the identification of a distinct syndrome due to de novo or inherited heterozygous mutations in Tousled-like kinase 2 (TLK2) in 38 unrelated individuals and two affected mothers, using whole-exome and whole-genome sequencing technologies, matchmaker databases, and international collaborations. Affected individuals had a consistent phenotype, characterized by mild-borderline neurodevelopmental delay (86%), behavioral disorders (68%), severe gastro-intestinal problems (63%), and facial dysmorphism including blepharophimosis (82%), telecanthus (74%), prominent nasal bridge (68%), broad nasal tip (66%), thin vermilion of the upper lip (62%), and upslanting palpebral fissures (55%). Analysis of cell lines from three affected individuals showed that mutations act through a loss-of-function mechanism in at least two case subjects. Genotype-phenotype analysis and comparison of computationally modeled faces showed that phenotypes of these and other individuals with loss-of-function variants significantly overlapped with phenotypes of individuals with other variant types (missense and C-terminal truncating). This suggests that haploinsufficiency of TLK2 is the most likely underlying disease mechanism, leading to a consistent neurodevelopmental phenotype. This work illustrates the power of international data sharing, by the identification of 40 individuals from 26 different centers in 7 different countries, allowing the identification, clinical delineation, and genotype-phenotype evaluation of a distinct NDD caused by mutations in TLK2.


Subject(s)
Genetic Association Studies , Inheritance Patterns/genetics , Loss of Function Mutation/genetics , Neurodevelopmental Disorders/genetics , Protein Kinases/genetics , Adolescent , Adult , Base Sequence , Cell Line , Child , Child, Preschool , Facies , Female , Humans , Infant , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Translocation, Genetic , Young Adult
7.
Genet Med ; 22(3): 547-556, 2020 03.
Article in English | MEDLINE | ID: mdl-31649276

ABSTRACT

PURPOSE: Treacher Collins syndrome (TCS) is a rare autosomal dominant mandibulofacial dysostosis, with a prevalence of 0.2-1/10,000. Features include bilateral and symmetrical malar and mandibular hypoplasia and facial abnormalities due to abnormal neural crest cell (NCC) migration and differentiation. To date, three genes have been identified: TCOF1, POLR1C, and POLR1D. Despite a large number of patients with a molecular diagnosis, some remain without a known genetic anomaly. METHODS: We performed exome sequencing for four individuals with TCS but who were negative for pathogenic variants in the known causative genes. The effect of the pathogenic variants was investigated in zebrafish. RESULTS: We identified three novel pathogenic variants in POLR1B. Knockdown of polr1b in zebrafish induced an abnormal craniofacial phenotype mimicking TCS that was associated with altered ribosomal gene expression, massive p53-associated cellular apoptosis in the neuroepithelium, and reduced number of NCC derivatives. CONCLUSION: Pathogenic variants in the RNA polymerase I subunit POLR1B might induce massive p53-dependent apoptosis in a restricted neuroepithelium area, altering NCC migration and causing cranioskeletal malformations. We identify POLR1B as a new causative gene responsible for a novel TCS syndrome (TCS4) and establish a novel experimental model in zebrafish to study POLR1B-related TCS.


Subject(s)
Craniofacial Abnormalities/genetics , DNA-Directed RNA Polymerases/genetics , Mandibulofacial Dysostosis/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Craniofacial Abnormalities/pathology , Genetic Predisposition to Disease , Humans , Mandibulofacial Dysostosis/pathology , Mutation , Neural Crest/abnormalities , Neural Crest/pathology , Tumor Suppressor Protein p53/genetics , Exome Sequencing , Zebrafish/genetics
8.
Clin Genet ; 96(3): 246-253, 2019 09.
Article in English | MEDLINE | ID: mdl-31090057

ABSTRACT

Two distinct genomic disorders have been linked to Xq28-gains, namely Xq28-duplications including MECP2 and Int22h1/Int22h2-mediated duplications involving RAB39B. Here, we describe six unrelated patients, five males and one female, with Xq28-gains distal to MECP2 and proximal to the Int22h1/Int22h2 low copy repeats. Comparison with patients carrying overlapping duplications in the literature defined the MidXq28-duplication syndrome featuring intellectual disability, language impairment, structural brain malformations, microcephaly, seizures and minor craniofacial features. The duplications overlapped for 108 kb including FLNA, RPL10 and GDI1 genes, highly expressed in brain and candidates for the neurologic phenotype.


Subject(s)
Chromosome Duplication , Chromosomes, Human, X , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/genetics , Methyl-CpG-Binding Protein 2/genetics , rab GTP-Binding Proteins/genetics , Adolescent , Adult , Brain/abnormalities , Brain/diagnostic imaging , Child , Facies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Magnetic Resonance Imaging , Male , Pedigree , Phenotype , Young Adult
9.
Brain ; 141(8): 2299-2311, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29985992

ABSTRACT

The transcription factor BCL11B is essential for development of the nervous and the immune system, and Bcl11b deficiency results in structural brain defects, reduced learning capacity, and impaired immune cell development in mice. However, the precise role of BCL11B in humans is largely unexplored, except for a single patient with a BCL11B missense mutation, affected by multisystem anomalies and profound immune deficiency. Using massively parallel sequencing we identified 13 patients bearing heterozygous germline alterations in BCL11B. Notably, all of them are affected by global developmental delay with speech impairment and intellectual disability; however, none displayed overt clinical signs of immune deficiency. Six frameshift mutations, two nonsense mutations, one missense mutation, and two chromosomal rearrangements resulting in diminished BCL11B expression, arose de novo. A further frameshift mutation was transmitted from a similarly affected mother. Interestingly, the most severely affected patient harbours a missense mutation within a zinc-finger domain of BCL11B, probably affecting the DNA-binding structural interface, similar to the recently published patient. Furthermore, the most C-terminally located premature termination codon mutation fails to rescue the progenitor cell proliferation defect in hippocampal slice cultures from Bcl11b-deficient mice. Concerning the role of BCL11B in the immune system, extensive immune phenotyping of our patients revealed alterations in the T cell compartment and lack of peripheral type 2 innate lymphoid cells (ILC2s), consistent with the findings described in Bcl11b-deficient mice. Unsupervised analysis of 102 T lymphocyte subpopulations showed that the patients clearly cluster apart from healthy children, further supporting the common aetiology of the disorder. Taken together, we show here that mutations leading either to BCL11B haploinsufficiency or to a truncated BCL11B protein clinically cause a non-syndromic neurodevelopmental delay. In addition, we suggest that missense mutations affecting specific sites within zinc-finger domains might result in distinct and more severe clinical outcomes.


Subject(s)
Neurodevelopmental Disorders/genetics , Repressor Proteins/genetics , Repressor Proteins/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Adolescent , Animals , Child , Child, Preschool , Female , Gene Expression Regulation/genetics , Germ-Line Mutation , Haploinsufficiency , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Infant , Lymphocytes/pathology , Lymphocytes/physiology , Male , Mice , Mutation , Repressor Proteins/metabolism , T-Lymphocytes/physiology , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism
10.
Hum Mutat ; 39(8): 1126-1138, 2018 08.
Article in English | MEDLINE | ID: mdl-29851191

ABSTRACT

Highly conserved TREX-mediated mRNA export is emerging as a key pathway in neuronal development and differentiation. TREX subunit variants cause neurodevelopmental disorders (NDDs) by interfering with mRNA export from the cell nucleus to the cytoplasm. Previously we implicated four missense variants in the X-linked THOC2 gene in intellectual disability (ID). We now report an additional six affected individuals from five unrelated families with two de novo and three maternally inherited pathogenic or likely pathogenic variants in THOC2 extending the genotypic and phenotypic spectrum. These comprise three rare missense THOC2 variants that affect evolutionarily conserved amino acid residues and reduce protein stability and two with canonical splice-site THOC2 variants that result in C-terminally truncated THOC2 proteins. We present detailed clinical assessment and functional studies on a de novo variant in a female with an epileptic encephalopathy and discuss an additional four families with rare variants in THOC2 with supportive evidence for pathogenicity. Severe neurocognitive features, including movement and seizure disorders, were observed in this cohort. Taken together our data show that even subtle alterations to the canonical molecular pathways such as mRNA export, otherwise essential for cellular life, can be compatible with life, but lead to NDDs in humans.


Subject(s)
Epilepsy/metabolism , Exons/genetics , Growth Disorders/metabolism , Intellectual Disability/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Child , Child, Preschool , Epilepsy/genetics , Female , Growth Disorders/genetics , HEK293 Cells , HeLa Cells , Humans , Intellectual Disability/genetics , Male , Mutation, Missense/genetics , Protein Isoforms/genetics , RNA Transport/genetics , RNA Transport/physiology , RNA-Binding Proteins/genetics
11.
Hum Genet ; 137(9): 753-768, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30167850

ABSTRACT

NALCN is a conserved cation channel, which conducts a permanent sodium leak current and regulates resting membrane potential and neuronal excitability. It is part of a large ion channel complex, the "NALCN channelosome", consisting of multiple proteins including UNC80 and UNC79. The predominant neuronal expression pattern and its function suggest an important role in neuronal function and disease. So far, biallelic NALCN and UNC80 variants have been described in a small number of individuals leading to infantile hypotonia, psychomotor retardation, and characteristic facies 1 (IHPRF1, OMIM 615419) and 2 (IHPRF2, OMIM 616801), respectively. Heterozygous de novo NALCN missense variants in the S5/S6 pore-forming segments lead to congenital contractures of the limbs and face, hypotonia, and developmental delay (CLIFAHDD, OMIM 616266) with some clinical overlap. In this study, we present detailed clinical information of 16 novel individuals with biallelic NALCN variants, 1 individual with a heterozygous de novo NALCN missense variant and an interesting clinical phenotype without contractures, and 12 individuals with biallelic UNC80 variants. We report for the first time a missense NALCN variant located in the predicted S6 pore-forming unit inherited in an autosomal-recessive manner leading to mild IHPRF1. We show evidence of clinical variability, especially among IHPRF1-affected individuals, and discuss differences between the IHPRF1- and IHPRF2 phenotypes. In summary, we provide a comprehensive overview of IHPRF1 and IHPRF2 phenotypes based on the largest cohort of individuals reported so far and provide additional insights into the clinical phenotypes of these neurodevelopmental diseases to help improve counseling of affected families.


Subject(s)
Carrier Proteins/genetics , Channelopathies/genetics , Developmental Disabilities/genetics , Genetic Markers , Genetic Variation , Membrane Proteins/genetics , Sodium Channels/genetics , Adolescent , Adult , Channelopathies/pathology , Child , Child, Preschool , Developmental Disabilities/pathology , Female , Humans , Infant , Infant, Newborn , Ion Channels , Male , Phenotype , Young Adult
12.
Hum Genet ; 136(7): 821-834, 2017 07.
Article in English | MEDLINE | ID: mdl-28393272

ABSTRACT

Pathogenic variants in genes encoding subunits of the spliceosome are the cause of several human diseases, such as neurodegenerative diseases. The RNA splicing process is facilitated by the spliceosome, a large RNA-protein complex consisting of small nuclear ribonucleoproteins (snRNPs), and many other proteins, such as heterogeneous nuclear ribonucleoproteins (hnRNPs). The HNRNPU gene (OMIM *602869) encodes the heterogeneous nuclear ribonucleoprotein U, which plays a crucial role in mammalian development. HNRNPU is expressed in the fetal brain and adult heart, kidney, liver, brain, and cerebellum. Microdeletions in the 1q44 region encompassing HNRNPU have been described in patients with intellectual disability (ID) and other clinical features, such as seizures, corpus callosum abnormalities (CCA), and microcephaly. Recently, pathogenic HNRNPU variants were identified in large ID and epileptic encephalopathy cohorts. In this study, we provide detailed clinical information of five novels and review two of the previously published individuals with (likely) pathogenic de novo variants in the HNRNPU gene including three non-sense and two missense variants, one small intragenic deletion, and one duplication. The phenotype in individuals with variants in HNRNPU is characterized by early onset seizures (6/7), severe ID (6/6), severe speech impairment (6/6), hypotonia (6/7), and central nervous system (CNS) (5/6), cardiac (4/6), and renal abnormalities (3/4). In this study, we broaden the clinical and mutational HNRNPU-associated spectrum, and demonstrate that heterozygous HNRNPU variants cause epilepsy, severe ID with striking speech impairment and variable CNS, cardiac, and renal anomalies.


Subject(s)
Epilepsy/genetics , Heterogeneous-Nuclear Ribonucleoprotein U/genetics , Heterozygote , Intellectual Disability/genetics , Age of Onset , Agenesis of Corpus Callosum/genetics , Central Nervous System/abnormalities , Central Nervous System/pathology , Chromosome Deletion , Chromosomes, Human, Pair 1 , Epilepsy/diagnosis , Female , Genetic Variation , Humans , Infant , Intellectual Disability/diagnosis , Kidney/abnormalities , Male , Microcephaly/diagnosis , Microcephaly/genetics , Muscle Hypotonia/diagnosis , Muscle Hypotonia/genetics , Phenotype , RNA Splicing , Ribonucleoproteins, Small Nuclear/genetics , Seizures/diagnosis , Seizures/genetics
13.
Am J Hum Genet ; 95(6): 698-707, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25434003

ABSTRACT

Mutations in components of the major spliceosome have been described in disorders with craniofacial anomalies, e.g., Nager syndrome and mandibulofacial dysostosis type Guion-Almeida. The U5 spliceosomal complex of eight highly conserved proteins is critical for pre-mRNA splicing. We identified biallelic mutations in TXNL4A, a member of this complex, in individuals with Burn-McKeown syndrome (BMKS). This rare condition is characterized by bilateral choanal atresia, hearing loss, cleft lip and/or palate, and other craniofacial dysmorphisms. Mutations were found in 9 of 11 affected families. In 8 families, affected individuals carried a rare loss-of-function mutation (nonsense, frameshift, or microdeletion) on one allele and a low-frequency 34 bp deletion (allele frequency 0.76%) in the core promoter region on the other allele. In a single highly consanguineous family, formerly diagnosed as oculo-oto-facial dysplasia, the four affected individuals were homozygous for a 34 bp promoter deletion, which differed from the promoter deletion in the other families. Reporter gene and in vivo assays showed that the promoter deletions led to reduced expression of TXNL4A. Depletion of TXNL4A (Dib1) in yeast demonstrated reduced assembly of the tri-snRNP complex. Our results indicate that BMKS is an autosomal-recessive condition, which is frequently caused by compound heterozygosity of low-frequency promoter deletions in combination with very rare loss-of-function mutations.


Subject(s)
Choanal Atresia/genetics , Deafness/congenital , Gene Deletion , Heart Defects, Congenital/genetics , Promoter Regions, Genetic/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Spliceosomes/genetics , Alleles , Child, Preschool , Choanal Atresia/diagnosis , Deafness/diagnosis , Deafness/genetics , Exosomes/genetics , Facies , Female , Gene Expression Profiling , Gene Frequency , Genes, Reporter , Heart Defects, Congenital/diagnosis , Heterozygote , Homozygote , Humans , Male , Mutation , Oligonucleotide Array Sequence Analysis , Pedigree , Phenotype , Ribonucleoprotein, U5 Small Nuclear/metabolism , Sequence Analysis, DNA , Spliceosomes/metabolism
14.
Am J Med Genet A ; 173(2): 435-443, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27862890

ABSTRACT

Loss-of-function mutations and deletions of the SOX2 gene are known to cause uni- and bilateral anophthalmia and microphthalmia as well as related disorders such as anophthalmia-esophageal-genital syndrome. Thus, anophthalmia/microphthalmia is the primary indication for targeted, "phenotype first" analyses of SOX2. However, SOX2 mutations are also associated with a wide range of non-ocular abnormalities, such as postnatal growth retardation, structural brain anomalies, hypogenitalism, and developmental delay. The present report describes three patients without anophthalmia/microphthalmia and loss-of-function mutations or microdeletions of SOX2 who had been investigated in a "genotype first" manner due to intellectual disability/developmental delay using whole exome sequencing or chromosomal microarray analyses. This result prompted us to perform SOX2 Sanger sequencing in 192 developmental delay/intellectual disability patients without anophthalmia or microphthalmia. No additional SOX2 loss-of-function mutations were detected in this cohort, showing that SOX2 is clearly not a major cause of intellectual disability without anophthalmia/microphthalmia. In our three patients and four further, reported "genotype first" SOX2 microdeletion patients, anophthalmia/microphthalmia was present in less than half of the patients. Thus, SOX2 is another example of a gene whose clinical spectrum is broadened by the generation of "genotype first" findings using hypothesis-free, genome-wide methods. © 2016 Wiley Periodicals, Inc.


Subject(s)
Genetic Association Studies , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Phenotype , Point Mutation , SOXB1 Transcription Factors/genetics , Sequence Deletion , Brain/abnormalities , Child, Preschool , Comparative Genomic Hybridization , Exome , Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Facies , Female , High-Throughput Nucleotide Sequencing , Humans , Infant, Newborn , Magnetic Resonance Imaging/methods , Male , Polymorphism, Single Nucleotide , Registries
15.
Hum Genet ; 134(6): 553-68, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25724810

ABSTRACT

Coffin-Siris syndrome (CSS) and Nicolaides-Baraitser syndrome (NCBRS) are rare intellectual disability/congenital malformation syndromes that represent distinct entities but show considerable clinical overlap. They are caused by mutations in genes encoding members of the BRG1- and BRM-associated factor (BAF) complex. However, there are a number of patients with the clinical diagnosis of CSS or NCBRS in whom the causative mutation has not been identified. In this study, we performed trio-based whole-exome sequencing (WES) in ten previously described but unsolved individuals with the tentative diagnosis of CSS or NCBRS and found causative mutations in nine out of ten individuals. Interestingly, our WES analysis disclosed overlapping differential diagnoses including Wiedemann-Steiner, Kabuki, and Adams-Oliver syndromes. In addition, most likely causative de novo mutations were identified in GRIN2A and SHANK3. Moreover, trio-based WES detected SMARCA2 and SMARCA4 deletions, which had not been annotated in a previous Haloplex target enrichment and next-generation sequencing of known CSS/NCBRS genes emphasizing the advantages of WES as a diagnostic tool. In summary, we discuss the phenotypic and diagnostic challenges in clinical genetics, establish important differential diagnoses, and emphasize the cardinal features and the broad clinical spectrum of BAF complex disorders and other disorders caused by mutations in epigenetic landscapers.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Exome , Face/abnormalities , Foot Deformities, Congenital/diagnosis , Foot Deformities, Congenital/genetics , Hand Deformities, Congenital/diagnosis , Hand Deformities, Congenital/genetics , Hypotrichosis/diagnosis , Hypotrichosis/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Micrognathism/diagnosis , Micrognathism/genetics , Mutation , Neck/abnormalities , Adult , Aged, 80 and over , Child , DNA Helicases/genetics , Diagnosis, Differential , Facies , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Middle Aged , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Transcription Factors/genetics
16.
Hum Genet ; 134(1): 97-109, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25326669

ABSTRACT

Recently, de novo heterozygous loss-of-function mutations in beta-catenin (CTNNB1) were described for the first time in four individuals with intellectual disability (ID), microcephaly, limited speech and (progressive) spasticity, and functional consequences of CTNNB1 deficiency were characterized in a mouse model. Beta-catenin is a key downstream component of the canonical Wnt signaling pathway. Somatic gain-of-function mutations have already been found in various tumor types, whereas germline loss-of-function mutations in animal models have been shown to influence neuronal development and maturation. We report on 16 additional individuals from 15 families in whom we newly identified de novo loss-of-function CTNNB1 mutations (six nonsense, five frameshift, one missense, two splice mutation, and one whole gene deletion). All patients have ID, motor delay and speech impairment (both mostly severe) and abnormal muscle tone (truncal hypotonia and distal hypertonia/spasticity). The craniofacial phenotype comprised microcephaly (typically -2 to -4 SD) in 12 of 16 and some overlapping facial features in all individuals (broad nasal tip, small alae nasi, long and/or flat philtrum, thin upper lip vermillion). With this detailed phenotypic characterization of 16 additional individuals, we expand and further establish the clinical and mutational spectrum of inactivating CTNNB1 mutations and thereby clinically delineate this new CTNNB1 haploinsufficiency syndrome.


Subject(s)
Intellectual Disability/genetics , Microcephaly/genetics , Mutation/genetics , beta Catenin/genetics , Child , Child, Preschool , Female , Follow-Up Studies , Haploinsufficiency , Humans , Infant , Intellectual Disability/pathology , Male , Microcephaly/pathology , Phenotype , Syndrome
17.
Am J Hum Genet ; 90(6): 1014-27, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22633399

ABSTRACT

The evolutionarily conserved cohesin complex was originally described for its role in regulating sister-chromatid cohesion during mitosis and meiosis. Cohesin and its regulatory proteins have been implicated in several human developmental disorders, including Cornelia de Lange (CdLS) and Roberts syndromes. Here we show that human mutations in the integral cohesin structural protein RAD21 result in a congenital phenotype consistent with a "cohesinopathy." Children with RAD21 mutations display growth retardation, minor skeletal anomalies, and facial features that overlap findings in individuals with CdLS. Notably, unlike children with mutations in NIPBL, SMC1A, or SMC3, these individuals have much milder cognitive impairment than those with classical CdLS. Mechanistically, these mutations act at the RAD21 interface with the other cohesin proteins STAG2 and SMC1A, impair cellular DNA damage response, and disrupt transcription in a zebrafish model. Our data suggest that, compared to loss-of-function mutations, dominant missense mutations result in more severe functional defects and cause worse structural and cognitive clinical findings. These results underscore the essential role of RAD21 in eukaryotes and emphasize the need for further understanding of the role of cohesin in human development.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Mutation , Nuclear Proteins/genetics , Phosphoproteins/genetics , Animals , Cell Line , Cell Survival , Cognition Disorders/genetics , Comet Assay/methods , Craniofacial Abnormalities/genetics , DNA Damage , DNA-Binding Proteins , De Lange Syndrome/genetics , Ectromelia/genetics , Gene Dosage , Genome, Human , Humans , Hypertelorism/genetics , Micronucleus Tests , Mutation, Missense , Sister Chromatid Exchange , Two-Hybrid System Techniques , Zebrafish , Cohesins
18.
BMC Med Genet ; 16: 30, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25943194

ABSTRACT

BACKGROUND: Beckwith-Wiedemann syndrome (BWS) is a rare pediatric overgrowth disorder with a variable clinical phenotype caused by deregulation affecting imprinted genes in the chromosomal region 11p15. Alterations of the imprinting control region 1 (ICR1) at the IGF2/H19 locus resulting in biallelic expression of IGF2 and biallelic silencing of H19 account for approximately 10% of patients with BWS. The majority of these patients have epimutations of the ICR1 without detectable DNA sequence changes. Only a few patients were found to have deletions. Most of these deletions are small affecting different parts of the ICR1 differentially methylated region (ICR1-DMR) removing target sequences for CTCF. Only a very few deletions reported so far include the H19 gene in addition to the CTCF binding sites. None of these deletions include IGF2. CASE PRESENTATION: A male patient was born with hypotonia, facial dysmorphisms and hypoglycemia suggestive of Beckwith-Wiedemann syndrome. Using methylation-specific (MS)-MLPA (Multiplex ligation-dependent probe amplification) we have identified a maternally inherited large deletion of the ICR1 region in a patient and his mother. The deletion results in a variable clinical expression with a classical BWS in the mother and a more severe presentation of BWS in her son. By genome-wide SNP array analysis the deletion was found to span ~100 kb genomic DNA including the ICR1DMR, H19, two adjacent non-imprinted genes and two of three predicted enhancer elements downstream to H19. Methylation analysis by deep bisulfite next generation sequencing revealed hypermethylation of the maternal allele at the IGF2 locus in both, mother and child, although IGF2 is not affected by the deletion. CONCLUSIONS: We here report on a novel large familial deletion of the ICR1 region in a BWS family. Due to the deletion of the ICR1-DMR CTCF binding cannot take place and the residual enhancer elements have access to the IGF2 promoters. The aberrant methylation (hypermethylation) of the maternal IGF2 allele in both affected family members may reflect the active state of the normally silenced maternal IGF2 copy and can be a consequence of the deletion. The deletion results in a variable clinical phenotype and expression.


Subject(s)
Beckwith-Wiedemann Syndrome/genetics , Enhancer Elements, Genetic/genetics , Genetic Loci/genetics , Sequence Deletion , Adult , Cytogenetic Analysis , DNA Methylation , Female , Humans , Infant, Newborn , Insulin-Like Growth Factor II/genetics , Male , Phenotype , Pregnancy
19.
J Med Genet ; 50(12): 838-47, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24092917

ABSTRACT

BACKGROUND: Borjeson-Forssman-Lehmann syndrome (BFLS) is an X-linked recessive intellectual disability (ID) disorder caused by mutations in the PHF6 gene and characterised by variable cognitive impairment, a distinct facial gestalt, obesity, and hypogonadism. Female carriers are usually not affected or only mildly affected, and so far only two females with de novo mutations or deletions in PHF6 have been reported. METHODS AND RESULTS: We performed PHF6 mutational analysis and screening for intragenic deletions and duplications by quantitative real-time PCR and multiplex ligation dependent probe amplification (MLPA) in female patients with variable ID and a distinct appearance of sparse hair, remarkable facial features, hypoplastic nails, and teeth anomalies. We detected two truncating mutations and two duplications of exons 4 and 5. Furthermore, two female patients with PHF6 deletions and a similar phenotype were identified by routine molecular karyotyping. Recently, two patients with a clinical diagnosis of Coffin-Siris syndrome in early infancy had been found to harbour mutations in PHF6, and their phenotype in advanced ages is now described. Further studies revealed skewed X-inactivation in blood lymphocytes, while it was normal in fibroblasts, thus indicating functional mosaicism. CONCLUSIONS: Our findings indicate that de novo defects in PHF6 in females result in a recognisable phenotype which might have been under-recognised so far and which comprises variable ID, a characteristic facial gestalt, hypoplastic nails, brachydactyly, clinodactyly mainly of fingers IV and V, dental anomalies, and linear skin hyperpigmentation. It shows overlap with BFLS but also additional distinct features, thus adding a new facet to this disorder.


Subject(s)
Carrier Proteins/genetics , Epilepsy/genetics , Face/abnormalities , Fingers/abnormalities , Growth Disorders/genetics , Hypogonadism/genetics , Mental Retardation, X-Linked/genetics , Mutation/genetics , Obesity/genetics , Adolescent , Adult , Child , DNA Mutational Analysis , Epilepsy/pathology , Face/pathology , Female , Fingers/pathology , Foot/pathology , Growth Disorders/pathology , Hand/pathology , Humans , Hypogonadism/pathology , Mental Retardation, X-Linked/pathology , Obesity/pathology , Phenotype , Repressor Proteins , Young Adult
20.
Ren Fail ; 36(4): 619-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24502542

ABSTRACT

INTRODUCTION: The tricho-rhino-phalangeal syndrome type III (TRPS III) is a rare autosomal dominantly inherited condition. The main clinical features are sparse and slow-growing hair and nails, a pear-shaped nose with a bulbous tip, elongated and flat philtrum, thin upper lip, cone-shaped epiphyses of the phalanges, and short stature. All patients have a point mutation in the TRPS1 gene. CASE REPORT: In this paper, we present a 13-year-old female with the typical clinical features of TRPS III, extreme growth retardation, severe deformities of both proximal radii resulting in limited extension of the elbows, and chronic renal failure (CRF) in addition. Molecular diagnostics revealed a missense mutation in exon 6 of TRPS1 that she inherited from her father who is also affected with TRPS III, but does not have CRF. In the index patient, the CRF was found to be due to bilateral renal hypodysplasia (RHD). CONCLUSION: Beside the renal dysplasia, the girl had severe deformities of the proximal radii - findings which have not been reported so far in TRPS III.


Subject(s)
Fingers/abnormalities , Growth Disorders/genetics , Hair Diseases/genetics , Kidney Failure, Chronic/genetics , Langer-Giedion Syndrome/genetics , Nose/abnormalities , Abnormalities, Multiple/genetics , Adolescent , DNA-Binding Proteins/genetics , Female , Humans , Kidney/abnormalities , Point Mutation , Radius/abnormalities , Repressor Proteins , Syndrome , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL