Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Am J Hum Genet ; 110(2): 251-272, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36669495

ABSTRACT

For neurodevelopmental disorders (NDDs), a molecular diagnosis is key for management, predicting outcome, and counseling. Often, routine DNA-based tests fail to establish a genetic diagnosis in NDDs. Transcriptome analysis (RNA sequencing [RNA-seq]) promises to improve the diagnostic yield but has not been applied to NDDs in routine diagnostics. Here, we explored the diagnostic potential of RNA-seq in 96 individuals including 67 undiagnosed subjects with NDDs. We performed RNA-seq on single individuals' cultured skin fibroblasts, with and without cycloheximide treatment, and used modified OUTRIDER Z scores to detect gene expression outliers and mis-splicing by exonic and intronic outliers. Analysis was performed by a user-friendly web application, and candidate pathogenic transcriptional events were confirmed by secondary assays. We identified intragenic deletions, monoallelic expression, and pseudoexonic insertions but also synonymous and non-synonymous variants with deleterious effects on transcription, increasing the diagnostic yield for NDDs by 13%. We found that cycloheximide treatment and exonic/intronic Z score analysis increased detection and resolution of aberrant splicing. Importantly, in one individual mis-splicing was found in a candidate gene nearly matching the individual's specific phenotype. However, pathogenic splicing occurred in another neuronal-expressed gene and provided a molecular diagnosis, stressing the need to customize RNA-seq. Lastly, our web browser application allowed custom analysis settings that facilitate diagnostic application and ranked pathogenic transcripts as top candidates. Our results demonstrate that RNA-seq is a complementary method in the genomic diagnosis of NDDs and, by providing accessible analysis with improved sensitivity, our transcriptome analysis approach facilitates wider implementation of RNA-seq in routine genome diagnostics.


Subject(s)
Gene Expression Profiling , Neurodevelopmental Disorders , Humans , RNA-Seq , Cycloheximide , Sequence Analysis, RNA/methods , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics
2.
Am J Hum Genet ; 109(2): 361-372, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35051358

ABSTRACT

Nuclear deubiquitinase BAP1 (BRCA1-associated protein 1) is a core component of multiprotein complexes that promote transcription by reversing the ubiquitination of histone 2A (H2A). BAP1 is a tumor suppressor whose germline loss-of-function variants predispose to cancer. To our knowledge, there are very rare examples of different germline variants in the same gene causing either a neurodevelopmental disorder (NDD) or a tumor predisposition syndrome. Here, we report a series of 11 de novo germline heterozygous missense BAP1 variants associated with a rare syndromic NDD. Functional analysis showed that most of the variants cannot rescue the consequences of BAP1 inactivation, suggesting a loss-of-function mechanism. In T cells isolated from two affected children, H2A deubiquitination was impaired. In matching peripheral blood mononuclear cells, histone H3 K27 acetylation ChIP-seq indicated that these BAP1 variants induced genome-wide chromatin state alterations, with enrichment for regulatory regions surrounding genes of the ubiquitin-proteasome system (UPS). Altogether, these results define a clinical syndrome caused by rare germline missense BAP1 variants that alter chromatin remodeling through abnormal histone ubiquitination and lead to transcriptional dysregulation of developmental genes.


Subject(s)
BRCA1 Protein/genetics , Germ-Line Mutation , Loss of Function Mutation , Mutation, Missense , Neurodevelopmental Disorders/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Adolescent , BRCA1 Protein/immunology , Child , Child, Preschool , Chromatin/chemistry , Chromatin/immunology , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/immunology , Family , Female , Gene Expression Regulation , Heterozygote , Histones/genetics , Histones/immunology , Host Cell Factor C1/genetics , Host Cell Factor C1/immunology , Humans , Infant , Male , Neurodevelopmental Disorders/immunology , Neurodevelopmental Disorders/pathology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/immunology , Ubiquitin/genetics , Ubiquitin/immunology , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology , Ubiquitination
3.
Eur J Paediatr Neurol ; 35: 27-34, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34592643

ABSTRACT

The increasing pace of gene discovery in the last decade has brought a major change in the way the genetic causes of brain malformations are being diagnosed. Unbiased genomic screening has gained the first place in the diagnostic protocol of a child with congenital (brain) anomalies and the detected variants are matched with the phenotypic presentation afterwards. This process is defined as "reverse phenotyping". Screening of DNA, through copy number variant analysis of microarrays and analysis of exome data on different platforms, obtained from the index patient and both parents has become a routine approach in many centers worldwide. Clinicians are used to multidisciplinary team interaction in patient care and disease management and this explains why the majority of research that has led to the discovery of new genetic disorders nowadays proceeds from clinical observations to genomic analysis and to data exchange facilitated by open access sharing databases. However, the relevance of multidisciplinary team interaction has not been object of systematic research in the field of brain malformations. This review will illustrate some examples of how diagnostically driven questions through multidisciplinary interaction, among clinical and preclinical disciplines, can be successful in the discovery of new genes related to brain malformations. The first example illustrates the setting of interaction among neurologists, geneticists and neuro-radiologists. The second illustrates the importance of interaction among clinical dysmorphologists for pattern recognition of syndromes with multiple congenital anomalies. The third example shows how fruitful it can be to step out of the "clinical comfort zone", and interact with basic scientists in applying emerging technologies to solve the diagnostic puzzles.


Subject(s)
Abnormalities, Multiple , DNA Copy Number Variations , Abnormalities, Multiple/genetics , Child , Exome , Genetic Association Studies , Humans , Exome Sequencing
4.
Eur J Hum Genet ; 29(3): 434-446, 2021 03.
Article in English | MEDLINE | ID: mdl-33162552

ABSTRACT

Pompe disease is a lysosomal and neuromuscular disorder caused by deficiency of acid alpha-glucosidase (GAA), and causes classic infantile, childhood onset, or adulthood onset phenotypes. The biochemical diagnosis is based on GAA activity assays in dried blood spots, leukocytes, or fibroblasts. Diagnosis can be complicated by the existence of pseudodeficiencies, i.e., GAA variants that lower GAA activity but do not cause Pompe disease. A large-scale comparison between these assays for patient samples, including exceptions and borderline cases, along with clinical diagnoses has not been reported so far. Here we analyzed GAA activity in a total of 1709 diagnostic cases over the past 28 years using a total of 2591 analyses and we confirmed the clinical diagnosis in 174 patients. We compared the following assays: leukocytes using glycogen or 4MUG as substrate, fibroblasts using 4MUG as substrate, and dried blood spots using 4MUG as substrate. In 794 individuals, two or more assays were performed. We found that phenotypes could only be distinguished using fibroblasts with 4MUG as substrate. Pseudodeficiencies caused by the GAA2 allele could be ruled out using 4MUG rather than glycogen as substrate in leukocytes or fibroblasts. The Asian pseudodeficiency could only be ruled out in fibroblasts using 4MUG as substrate. We conclude that fibroblasts using 4MUG as substrate provides the most reliable assay for biochemical diagnosis and can serve to validate results from leukocytes or dried blood spots.


Subject(s)
Clinical Enzyme Tests/methods , Dried Blood Spot Testing/methods , Genetic Testing/methods , Glycogen Storage Disease Type II/genetics , Cells, Cultured , Clinical Enzyme Tests/statistics & numerical data , Dried Blood Spot Testing/statistics & numerical data , Fibroblasts/enzymology , Fibroblasts/metabolism , Genetic Testing/statistics & numerical data , Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/metabolism , Humans , Hymecromone/analogs & derivatives , Hymecromone/metabolism , Leukocytes/enzymology , Leukocytes/metabolism , Mutation , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
5.
Am J Hum Genet ; 106(6): 779-792, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32413283

ABSTRACT

The evolutionarily conserved hedgehog (Hh) pathway is essential for organogenesis and plays critical roles in postnatal tissue maintenance and renewal. A unique feature of the vertebrate Hh pathway is that signal transduction requires the primary cilium (PC) where major pathway components are dynamically enriched. These factors include smoothened (SMO) and patched, which constitute the core reception system for sonic hedgehog (SHH) as well as GLI transcription factors, the key mediators of the pathway. Here, we report bi-allelic loss-of-function variations in SMO in seven individuals from five independent families; these variations cause a wide phenotypic spectrum of developmental anomalies affecting the brain (hypothalamic hamartoma and microcephaly), heart (atrioventricular septal defect), skeleton (postaxial polydactyly, narrow chest, and shortening of long bones), and enteric nervous system (aganglionosis). Cells derived from affected individuals showed normal ciliogenesis but severely altered Hh-signal transduction as a result of either altered PC trafficking or abnormal activation of the pathway downstream of SMO. In addition, Hh-independent GLI2 accumulation at the PC tip in cells from the affected individuals suggests a potential function of SMO in regulating basal ciliary trafficking of GLI2 when the pathway is off. Thus, loss of SMO function results in abnormal PC dynamics of key components of the Hh signaling pathway and leads to a large continuum of malformations in humans.


Subject(s)
Alleles , Developmental Disabilities/genetics , Hedgehog Proteins/metabolism , Signal Transduction , Smoothened Receptor/genetics , Base Sequence , Child , Child, Preschool , Cilia/physiology , Female , Humans , Infant , Male , Models, Molecular , Neoplasms/genetics , Nerve Tissue Proteins , Nuclear Proteins , Pedigree , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3
6.
Am J Med Genet A ; 182(6): 1378-1386, 2020 06.
Article in English | MEDLINE | ID: mdl-32212377

ABSTRACT

DNA double-strand breaks (DSBs) are highly toxic DNA lesions that can lead to chromosomal instability, loss of genes and cancer. The MRE11/RAD50/NBN (MRN) complex is keystone involved in signaling processes inducing the repair of DSB by, for example, in activating pathways leading to homologous recombination repair and nonhomologous end joining. Additionally, the MRN complex also plays an important role in the maintenance of telomeres and can act as a stabilizer at replication forks. Mutations in NBN and MRE11 are associated with Nijmegen breakage syndrome (NBS) and ataxia telangiectasia (AT)-like disorder, respectively. So far, only one single patient with biallelic loss of function variants in RAD50 has been reported presenting with features classified as NBS-like disorder. Here, we report a long-term follow-up of an unrelated patient with facial dysmorphisms, microcephaly, skeletal features, and short stature who is homozygous for a novel variant in RAD50. We could show that this variant, c.2524G > A in exon 15 of the RAD50 gene, induces aberrant splicing of RAD50 mRNA mainly leading to premature protein truncation and thereby, most likely, to loss of RAD50 function. Using patient-derived primary fibroblasts, we could show abnormal radioresistant DNA synthesis confirming pathogenicity of the identified variant. Immunoblotting experiments showed strongly reduced protein levels of RAD50 in the patient-derived fibroblasts and provided evidence for a markedly reduced radiation-induced AT-mutated signaling. Comparison with the previously reported case and with patients presenting with NBS confirms that RAD50 mutations lead to a similar, but distinctive phenotype.


Subject(s)
Acid Anhydride Hydrolases/genetics , Ataxia Telangiectasia/genetics , DNA Repair-Deficiency Disorders/genetics , DNA-Binding Proteins/genetics , Growth Disorders/genetics , Microcephaly/genetics , Nijmegen Breakage Syndrome/genetics , Alleles , Ataxia Telangiectasia/complications , Ataxia Telangiectasia/pathology , Cell Cycle Proteins/genetics , Child , Child, Preschool , DNA Breaks, Double-Stranded , DNA Repair-Deficiency Disorders/complications , DNA Repair-Deficiency Disorders/pathology , Female , Growth Disorders/complications , Growth Disorders/pathology , Humans , Infant , Infant, Newborn , MRE11 Homologue Protein/genetics , Microcephaly/complications , Microcephaly/pathology , Nijmegen Breakage Syndrome/complications , Nijmegen Breakage Syndrome/pathology , Nuclear Proteins/genetics , Pedigree
7.
Am J Hum Genet ; 105(4): 689-705, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31495489

ABSTRACT

Sphingomyelinases generate ceramide from sphingomyelin as a second messenger in intracellular signaling pathways involved in cell proliferation, differentiation, or apoptosis. Children from 12 unrelated families presented with microcephaly, simplified gyral pattern of the cortex, hypomyelination, cerebellar hypoplasia, congenital arthrogryposis, and early fetal/postnatal demise. Genomic analysis revealed bi-allelic loss-of-function variants in SMPD4, coding for the neutral sphingomyelinase-3 (nSMase-3/SMPD4). Overexpression of human Myc-tagged SMPD4 showed localization both to the outer nuclear envelope and the ER and additionally revealed interactions with several nuclear pore complex proteins by proteomics analysis. Fibroblasts from affected individuals showed ER cisternae abnormalities, suspected for increased autophagy, and were more susceptible to apoptosis under stress conditions, while treatment with siSMPD4 caused delayed cell cycle progression. Our data show that SMPD4 links homeostasis of membrane sphingolipids to cell fate by regulating the cross-talk between the ER and the outer nuclear envelope, while its loss reveals a pathogenic mechanism in microcephaly.


Subject(s)
Arthrogryposis/genetics , Microcephaly/genetics , Neurodevelopmental Disorders/genetics , Sphingomyelin Phosphodiesterase/genetics , Arthrogryposis/pathology , Cell Lineage , Child , Endoplasmic Reticulum/metabolism , Female , Gene Expression Profiling , HEK293 Cells , Humans , Male , Microcephaly/pathology , Mitosis , Neurodevelopmental Disorders/pathology , Pedigree , RNA Splicing
8.
Circ Genom Precis Med ; 12(9): 397-406, 2019 09.
Article in English | MEDLINE | ID: mdl-31461301

ABSTRACT

BACKGROUND: Pediatric cardiomyopathies are a clinically and genetically heterogeneous group of heart muscle disorders associated with high morbidity and mortality. Although knowledge of the genetic basis of pediatric cardiomyopathy has improved considerably, the underlying cause remains elusive in a substantial proportion of cases. METHODS: Exome sequencing was used to screen for the causative genetic defect in a pair of siblings with rapidly progressive dilated cardiomyopathy and death in early infancy. Protein expression was assessed in patient samples, followed by an in vitro tail-anchored protein insertion assay and functional analyses in zebrafish. RESULTS: We identified compound heterozygous variants in the highly conserved ASNA1 gene (arsA arsenite transporter, ATP-binding, homolog), which encodes an ATPase required for post-translational membrane insertion of tail-anchored proteins. The c.913C>T variant on the paternal allele is predicted to result in a premature stop codon p.(Gln305*), and likely explains the decreased protein expression observed in myocardial tissue and skin fibroblasts. The c.488T>C variant on the maternal allele results in a valine to alanine substitution at residue 163 (p.Val163Ala). Functional studies showed that this variant leads to protein misfolding as well as less effective tail-anchored protein insertion. Loss of asna1 in zebrafish resulted in reduced cardiac contractility and early lethality. In contrast to wild-type mRNA, injection of either mutant mRNA failed to rescue this phenotype. CONCLUSIONS: Biallelic variants in ASNA1 cause severe pediatric cardiomyopathy and early death. Our findings point toward a critical role of the tail-anchored membrane protein insertion pathway in vertebrate cardiac function and disease.


Subject(s)
Arsenite Transporting ATPases/genetics , Cardiomyopathies/genetics , Cytosol/enzymology , Point Mutation , Zebrafish Proteins/genetics , Alleles , Amino Acid Sequence , Animals , Arsenite Transporting ATPases/chemistry , Arsenite Transporting ATPases/metabolism , Cardiomyopathies/enzymology , Child, Preschool , Disease Models, Animal , Exome , Female , Genetic Variation , Humans , Protein Transport , Sequence Alignment , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
9.
Am J Hum Genet ; 104(3): 520-529, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30824121

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are essential enzymes responsible for charging tRNA molecules with cognate amino acids. Consistent with the essential function and ubiquitous expression of ARSs, mutations in 32 of the 37 ARS-encoding loci cause severe, early-onset recessive phenotypes. Previous genetic and functional data suggest a loss-of-function mechanism; however, our understanding of the allelic and locus heterogeneity of ARS-related disease is incomplete. Cysteinyl-tRNA synthetase (CARS) encodes the enzyme that charges tRNACys with cysteine in the cytoplasm. To date, CARS variants have not been implicated in any human disease phenotype. Here, we report on four subjects from three families with complex syndromes that include microcephaly, developmental delay, and brittle hair and nails. Each affected person carries bi-allelic CARS variants: one individual is compound heterozygous for c.1138C>T (p.Gln380∗) and c.1022G>A (p.Arg341His), two related individuals are compound heterozygous for c.1076C>T (p.Ser359Leu) and c.1199T>A (p.Leu400Gln), and one individual is homozygous for c.2061dup (p.Ser688Glnfs∗2). Measurement of protein abundance, yeast complementation assays, and assessments of tRNA charging indicate that each CARS variant causes a loss-of-function effect. Compared to subjects with previously reported ARS-related diseases, individuals with bi-allelic CARS variants are unique in presenting with a brittle-hair-and-nail phenotype, which most likely reflects the high cysteine content in human keratins. In sum, our efforts implicate CARS variants in human inherited disease, expand the locus and clinical heterogeneity of ARS-related clinical phenotypes, and further support impaired tRNA charging as the primary mechanism of recessive ARS-related disease.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Charcot-Marie-Tooth Disease/etiology , Developmental Disabilities/etiology , Hair Diseases/etiology , Microcephaly/etiology , Mutation , Nail Diseases/etiology , Adult , Amino Acid Sequence , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/pathology , Developmental Disabilities/enzymology , Developmental Disabilities/pathology , Female , Genes, Recessive , Genetic Predisposition to Disease , Hair Diseases/enzymology , Hair Diseases/pathology , Humans , Male , Microcephaly/enzymology , Microcephaly/pathology , Nail Diseases/enzymology , Nail Diseases/pathology , Pedigree , Phenotype , Prognosis , Sequence Homology , Young Adult
10.
Brain ; 142(4): 867-884, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30879067

ABSTRACT

Recessive mutations in RTTN, encoding the protein rotatin, were originally identified as cause of polymicrogyria, a cortical malformation. With time, a wide variety of other brain malformations has been ascribed to RTTN mutations, including primary microcephaly. Rotatin is a centrosomal protein possibly involved in centriolar elongation and ciliogenesis. However, the function of rotatin in brain development is largely unknown and the molecular disease mechanism underlying cortical malformations has not yet been elucidated. We performed both clinical and cell biological studies, aimed at clarifying rotatin function and pathogenesis. Review of the 23 published and five unpublished clinical cases and genomic mutations, including the effect of novel deep intronic pathogenic mutations on RTTN transcripts, allowed us to extrapolate the core phenotype, consisting of intellectual disability, short stature, microcephaly, lissencephaly, periventricular heterotopia, polymicrogyria and other malformations. We show that the severity of the phenotype is related to residual function of the protein, not only the level of mRNA expression. Skin fibroblasts from eight affected individuals were studied by high resolution immunomicroscopy and flow cytometry, in parallel with in vitro expression of RTTN in HEK293T cells. We demonstrate that rotatin regulates different phases of the cell cycle and is mislocalized in affected individuals. Mutant cells showed consistent and severe mitotic failure with centrosome amplification and multipolar spindle formation, leading to aneuploidy and apoptosis, which could relate to depletion of neuronal progenitors often observed in microcephaly. We confirmed the role of rotatin in functional and structural maintenance of primary cilia and determined that the protein localized not only to the basal body, but also to the axoneme, proving the functional interconnectivity between ciliogenesis and cell cycle progression. Proteomics analysis of both native and exogenous rotatin uncovered that rotatin interacts with the neuronal (non-muscle) myosin heavy chain subunits, motors of nucleokinesis during neuronal migration, and in human induced pluripotent stem cell-derived bipolar mature neurons rotatin localizes at the centrosome in the leading edge. This illustrates the role of rotatin in neuronal migration. These different functions of rotatin explain why RTTN mutations can lead to heterogeneous cerebral malformations, both related to proliferation and migration defects.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Adult , Brain/pathology , Carrier Proteins/genetics , Cell Cycle/physiology , Cilia/metabolism , Female , Genetic Association Studies/methods , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Infant , Infant, Newborn , Male , Malformations of Cortical Development/genetics , Malformations of Cortical Development/metabolism , Microcephaly/genetics , Mutation , Nervous System Malformations/genetics , Polymicrogyria/etiology , Polymicrogyria/pathology
11.
Eur J Hum Genet ; 27(6): 919-927, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30737479

ABSTRACT

Analyses in our diagnostic DNA laboratory include genes involved in autosomal recessive (AR) lysosomal storage disorders such as glycogenosis type II (Pompe disease) and mucopolysaccharidosis type I (MPSI, Hurler disease). We encountered 4 cases with apparent homozygosity for a disease-causing sequence variant that could be traced to one parent only. In addition, in a young child with cardiomyopathy, in the absence of other symptoms, a diagnosis of Pompe disease was considered. Remarkably, he presented with different enzymatic and genotypic features between leukocytes and skin fibroblasts. All cases were examined with microsatellite markers and SNP genotyping arrays. We identified one case of total uniparental disomy (UPD) of chromosome 17 leading to Pompe disease and three cases of segmental uniparental isodisomy (UPiD) causing Hurler-(4p) or Pompe disease (17q). One Pompe patient with unusual combinations of features was shown to have a mosaic segmental UPiD of chromosome 17q. The chromosome 17 UPD cases amount to 11% of our diagnostic cohort of homozygous Pompe patients (plus one case of pseudoheterozygosity) where segregation analysis was possible. We conclude that inclusion of parental DNA is mandatory for reliable DNA diagnostics. Mild or unusual phenotypes of AR diseases should alert physicians to the possibility of mosaic segmental UPiD. SNP genotyping arrays are used in diagnostic workup of patients with developmental delay. Our results show that even small Regions of Homozygosity that include telomeric areas are worth reporting, regardless of the imprinting status of the chromosome, as they might indicate segmental UPiD.


Subject(s)
Glycogen Storage Disease Type II/genetics , Mucopolysaccharidosis I/genetics , Polymorphism, Single Nucleotide , Uniparental Disomy , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male
12.
Mol Psychiatry ; 24(5): 757-771, 2019 05.
Article in English | MEDLINE | ID: mdl-29302076

ABSTRACT

Schizophrenia is highly heritable, yet its underlying pathophysiology remains largely unknown. Among the most well-replicated findings in neurobiological studies of schizophrenia are deficits in myelination and white matter integrity; however, direct etiological genetic and cellular evidence has thus far been lacking. Here, we implement a family-based approach for genetic discovery in schizophrenia combined with functional analysis using induced pluripotent stem cells (iPSCs). We observed familial segregation of two rare missense mutations in Chondroitin Sulfate Proteoglycan 4 (CSPG4) (c.391G > A [p.A131T], MAF 7.79 × 10-5 and c.2702T > G [p.V901G], MAF 2.51 × 10-3). The CSPG4A131T mutation was absent from the Swedish Schizophrenia Exome Sequencing Study (2536 cases, 2543 controls), while the CSPG4V901G mutation was nominally enriched in cases (11 cases vs. 3 controls, P = 0.026, OR 3.77, 95% CI 1.05-13.52). CSPG4/NG2 is a hallmark protein of oligodendrocyte progenitor cells (OPCs). iPSC-derived OPCs from CSPG4A131T mutation carriers exhibited abnormal post-translational processing (P = 0.029), subcellular localization of mutant NG2 (P = 0.007), as well as aberrant cellular morphology (P = 3.0 × 10-8), viability (P = 8.9 × 10-7), and myelination potential (P = 0.038). Moreover, transfection of healthy non-carrier sibling OPCs confirmed a pathogenic effect on cell survival of both the CSPG4A131T (P = 0.006) and CSPG4V901G (P = 3.4 × 10-4) mutations. Finally, in vivo diffusion tensor imaging of CSPG4A131T mutation carriers demonstrated a reduction of brain white matter integrity compared to unaffected sibling and matched general population controls (P = 2.2 × 10-5). Together, our findings provide a convergence of genetic and functional evidence to implicate OPC dysfunction as a candidate pathophysiological mechanism of familial schizophrenia.


Subject(s)
Chondroitin Sulfate Proteoglycans/genetics , Membrane Proteins/genetics , Oligodendrocyte Precursor Cells/metabolism , Schizophrenia/genetics , Adult , Antigens/genetics , Cell Differentiation/physiology , Chondroitin Sulfate Proteoglycans/metabolism , Diffusion Tensor Imaging , Family , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Membrane Proteins/metabolism , Mutation/genetics , Oligodendrocyte Precursor Cells/physiology , Oligodendroglia/metabolism , Pedigree , Proteoglycans/genetics , Schizophrenia/metabolism , White Matter/metabolism
13.
Am J Hum Genet ; 103(6): 1009-1021, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30471716

ABSTRACT

To date, mutations in 15 actin- or microtubule-associated genes have been associated with the cortical malformation lissencephaly and variable brainstem hypoplasia. During a multicenter review, we recognized a rare lissencephaly variant with a complex brainstem malformation in three unrelated children. We searched our large brain-malformation databases and found another five children with this malformation (as well as one with a less severe variant), analyzed available whole-exome or -genome sequencing data, and tested ciliogenesis in two affected individuals. The brain malformation comprised posterior predominant lissencephaly and midline crossing defects consisting of absent anterior commissure and a striking W-shaped brainstem malformation caused by small or absent pontine crossing fibers. We discovered heterozygous de novo missense variants or an in-frame deletion involving highly conserved zinc-binding residues within the GAR domain of MACF1 in the first eight subjects. We studied cilium formation and found a higher proportion of mutant cells with short cilia than of control cells with short cilia. A ninth child had similar lissencephaly but only subtle brainstem dysplasia associated with a heterozygous de novo missense variant in the spectrin repeat domain of MACF1. Thus, we report variants of the microtubule-binding GAR domain of MACF1 as the cause of a distinctive and most likely pathognomonic brain malformation. A gain-of-function or dominant-negative mechanism appears likely given that many heterozygous mutations leading to protein truncation are included in the ExAC Browser. However, three de novo variants in MACF1 have been observed in large schizophrenia cohorts.


Subject(s)
Axon Guidance/genetics , Cell Movement/genetics , Conserved Sequence/genetics , Microfilament Proteins/genetics , Mutation/genetics , Neurons/pathology , Zinc/metabolism , Adolescent , Brain Stem/pathology , Child , Child, Preschool , Cilia/genetics , Female , Humans , Lissencephaly/genetics , Male , Microtubules/genetics , Nervous System Malformations/genetics
15.
Am J Med Genet A ; 173(6): 1601-1606, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28544736

ABSTRACT

We describe the clinical presentation and 17 years follow up of a boy, born to consanguineous parents and presenting with intellectual disability (ID), autism, "marfanoid" dysmorphic features, and moderate abnormalities of sulfite metabolism compatible with molybdenum cofactor deficiency, but normal sulfite oxidase activity in cultured skin fibroblasts. Genomic exome analysis revealed a homozygous MOCS3 missense mutation, leading to a p.Ala257Thr substitution in the highly conserved ubiquitin-like-domain of the protein. MOCS3 is the third protein, besides MOCS1 and MOCS2, involved in the biosynthesis of the molybdenum cofactor and has a dual ubiquitin-like function in tRNA thiolation. It is plausible that the phenotype results from deficiency of this dual function, not only from defective synthesis of molybdenum cofactor, which would explain similarities and differences from the MOCS1 and MOCS2-related disorders. This observation should encourage testing of additional ID patients with mild abnormalities of sulfite metabolism for MOCS3 mutations.


Subject(s)
Autistic Disorder/genetics , Intellectual Disability/genetics , Metal Metabolism, Inborn Errors/genetics , Nucleotidyltransferases/genetics , Sulfurtransferases/genetics , Adolescent , Autistic Disorder/complications , Autistic Disorder/physiopathology , Gene Expression , Homozygote , Humans , Intellectual Disability/complications , Intellectual Disability/physiopathology , Male , Metal Metabolism, Inborn Errors/complications , Metal Metabolism, Inborn Errors/physiopathology , Mutation, Missense , Phenotype
16.
PLoS Genet ; 13(5): e1006809, 2017 May.
Article in English | MEDLINE | ID: mdl-28542170

ABSTRACT

Integrator is an RNA polymerase II (RNAPII)-associated complex that was recently identified to have a broad role in both RNA processing and transcription regulation. Importantly, its role in human development and disease is so far largely unexplored. Here, we provide evidence that biallelic Integrator Complex Subunit 1 (INTS1) and Subunit 8 (INTS8) gene mutations are associated with rare recessive human neurodevelopmental syndromes. Three unrelated individuals of Dutch ancestry showed the same homozygous truncating INTS1 mutation. Three siblings harboured compound heterozygous INTS8 mutations. Shared features by these six individuals are severe neurodevelopmental delay and a distinctive appearance. The INTS8 family in addition presented with neuronal migration defects (periventricular nodular heterotopia). We show that the first INTS8 mutation, a nine base-pair deletion, leads to a protein that disrupts INT complex stability, while the second missense mutation introduces an alternative splice site leading to an unstable messenger. Cells from patients with INTS8 mutations show increased levels of unprocessed UsnRNA, compatible with the INT function in the 3'-end maturation of UsnRNA, and display significant disruptions in gene expression and RNA processing. Finally, the introduction of the INTS8 deletion mutation in P19 cells using genome editing alters gene expression throughout the course of retinoic acid-induced neural differentiation. Altogether, our results confirm the essential role of Integrator to transcriptome integrity and point to the requirement of the Integrator complex in human brain development.


Subject(s)
Developmental Disabilities/genetics , Gene Deletion , Mutation, Missense , Protein Subunits/genetics , RNA, Messenger/metabolism , Adult , Alternative Splicing , Brain/growth & development , Brain/metabolism , Brain/pathology , Cells, Cultured , Child , Developmental Disabilities/diagnosis , Female , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Heterozygote , Humans , Male , Mutation , Pedigree , Protein Subunits/metabolism , RNA, Messenger/genetics , Syndrome , Transcriptome , Wnt1 Protein
17.
Exp Neurol ; 291: 106-119, 2017 05.
Article in English | MEDLINE | ID: mdl-28189729

ABSTRACT

Slc17a5-/- mice represent an animal model for the infantile form of sialic acid storage disease (SASD). We analyzed genetic and histological time-course expression of myelin and oligodendrocyte (OL) lineage markers in different parts of the CNS, and related this to postnatal neurobehavioral development in these mice. Sialin-deficient mice display a distinct spatiotemporal pattern of sialic acid storage, CNS hypomyelination and leukoencephalopathy. Whereas few genes are differentially expressed in the perinatal stage (p0), microarray analysis revealed increased differential gene expression in later postnatal stages (p10-p18). This included progressive upregulation of neuroinflammatory genes, as well as continuous down-regulation of genes that encode myelin constituents and typical OL lineage markers. Age-related histopathological analysis indicates that initial myelination occurs normally in hindbrain regions, but progression to more frontal areas is affected in Slc17a5-/- mice. This course of progressive leukoencephalopathy and CNS hypomyelination delays neurobehavioral development in sialin-deficient mice. Slc17a5-/- mice successfully achieve early neurobehavioral milestones, but exhibit progressive delay of later-stage sensory and motor milestones. The present findings may contribute to further understanding of the processes of CNS myelination as well as help to develop therapeutic strategies for SASD and other myelination disorders.


Subject(s)
Brain/pathology , Gene Expression Regulation, Developmental/genetics , Leukoencephalopathies , Mental Disorders/etiology , Organic Anion Transporters/deficiency , Sialic Acid Storage Disease , Symporters/deficiency , Age Factors , Animals , Animals, Newborn , Brain/metabolism , Developmental Disabilities/etiology , Developmental Disabilities/genetics , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Intermediate Filaments/metabolism , Leukoencephalopathies/complications , Leukoencephalopathies/etiology , Leukoencephalopathies/genetics , Lysosomal-Associated Membrane Protein 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organic Anion Transporters/genetics , Sialic Acid Storage Disease/complications , Sialic Acid Storage Disease/genetics , Sialic Acid Storage Disease/pathology , Symporters/genetics
18.
Mol Ther Nucleic Acids ; 5(9): e361, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27623443

ABSTRACT

While 9% of human pathogenic variants have an established effect on pre-mRNA splicing, it is suspected that an additional 20% of otherwise classified variants also affect splicing. Aberrant splicing includes disruption of splice sites or regulatory elements, or creation or strengthening of cryptic splice sites. For the majority of variants, it is poorly understood to what extent and how these may affect splicing. We have identified cryptic splicing in an unbiased manner. Three types of cryptic splicing were analyzed in the context of pathogenic variants in the acid α-glucosidase gene causing Pompe disease. These involved newly formed deep intronic or exonic cryptic splice sites, and a natural cryptic splice that was utilized due to weakening of a canonical splice site. Antisense oligonucleotides that targeted the identified cryptic splice sites repressed cryptic splicing at the expense of canonical splicing in all three cases, as shown by reverse-transcriptase-quantitative polymerase chain reaction analysis and by enhancement of acid α-glucosidase enzymatic activity. This argues for a competition model for available splice sites, including intact or weakened canonical sites and natural or newly formed cryptic sites. The pipeline described here can detect cryptic splicing and correct canonical splicing using antisense oligonucleotides to restore the gene defect.

19.
Eur J Hum Genet ; 24(10): 1424-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27329734

ABSTRACT

Biotinidase deficiency is a rare inherited metabolic disorder that can cause severe neurological symptoms. To prevent severe clinical presentations, it was included in the Dutch neonatal screening programme in 2007. Since then the number of cases detected has been high. This study set out to describe the incidence of the disease, the clinical and demographic characteristics of the neonates identified and the type of mutations found. In the south-western Netherlands, 304 982 neonates were screened between 2007 and 2012; and 92 were identified for further testing. Confirmatory testing revealed 6 (7%) with a profound biotinidase deficiency (<10% enzyme activity), 44 (48%) with a partial deficiency (10-30%) and 42 (46%) with normal activity (>30%). All six patients whose profound deficiency was confirmed had enzyme activities below 15% on neonatal screening. Mutation analysis was performed in 61 neonates: 5 'profound', 35 'partial' and 21 'normal'. All five 'profound' cases had two severe mutations. Comparison with the northern Netherlands showed that the frequency and types of mutation were representative for the Netherlands as a whole. The most common mutation detected was c.[1330G>C] (p.(Asp444His); 34%), which is considered to be mild, followed by three severe mutations c.[1368A>C], c.[1595C>T] and c.[1330G>C;511G>A]. Seven new mutations were identified. We conclude that neonatal screening for profound biotinidase produces a high number of false positives. Biotinidase deficiency was profound in less than 10% of cases identified. As biotinidase activity lay below 15% on neonatal screening in all such cases, the screening threshold might be reduced to 15%.


Subject(s)
Biotinidase Deficiency/genetics , Genetic Testing/standards , Biotinidase Deficiency/diagnosis , False Positive Reactions , Female , Genetic Testing/statistics & numerical data , Humans , Infant, Newborn , Male , Netherlands , Sensitivity and Specificity
20.
J Exp Med ; 213(7): 1163-74, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27325888

ABSTRACT

Pseudo-TORCH syndrome (PTS) is characterized by microcephaly, enlarged ventricles, cerebral calcification, and, occasionally, by systemic features at birth resembling the sequelae of congenital infection but in the absence of an infectious agent. Genetic defects resulting in activation of type 1 interferon (IFN) responses have been documented to cause Aicardi-Goutières syndrome, which is a cause of PTS. Ubiquitin-specific peptidase 18 (USP18) is a key negative regulator of type I IFN signaling. In this study, we identified loss-of-function recessive mutations of USP18 in five PTS patients from two unrelated families. Ex vivo brain autopsy material demonstrated innate immune inflammation with calcification and polymicrogyria. In vitro, patient fibroblasts displayed severely enhanced IFN-induced inflammation, which was completely rescued by lentiviral transduction of USP18. These findings add USP18 deficiency to the list of genetic disorders collectively termed type I interferonopathies. Moreover, USP18 deficiency represents the first genetic disorder of PTS caused by dysregulation of the response to type I IFNs. Therapeutically, this places USP18 as a promising target not only for genetic but also acquired IFN-mediated CNS disorders.


Subject(s)
Autoimmune Diseases of the Nervous System , Brain/immunology , Calcinosis , Endopeptidases/deficiency , Immunity, Innate , Interferon Type I/immunology , Microglia/immunology , Nervous System Malformations , Signal Transduction , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/pathology , Brain/pathology , Calcinosis/genetics , Calcinosis/immunology , Calcinosis/pathology , Endopeptidases/immunology , Female , Humans , Interferon Type I/genetics , Male , Microglia/pathology , Nervous System Malformations/genetics , Nervous System Malformations/immunology , Nervous System Malformations/pathology , Signal Transduction/genetics , Signal Transduction/immunology , Ubiquitin Thiolesterase
SELECTION OF CITATIONS
SEARCH DETAIL
...