Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Eur J Med Genet ; 58(5): 324-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25858702

ABSTRACT

Intellectual disability (ID) is a very heterogeneous disorder with over 100 ID genes located on the X chromosome alone. Of these, KDM5C and IQSEC2 are located adjacent to each other at the Xp11.22 locus. While mutations in either of these genes are associated with severe ID in males, female carriers are mostly unaffected. Here, we report on a female patient with severe ID and autistic features carrying a de novo 0.4 Mb deletion containing six coding genes including KDM5C and IQSEC2. X-inactivation analysis revealed skewing in a lymphocyte-derived cell line from this patient with preferential inactivation of the mutant X chromosome. As the brain-expressed KDM5C and IQSEC2 genes escape X-inactivation, deletion of these alleles could still be detrimental despite skewing of X-inactivation. Indeed, mutations in either of both genes have been reported in a few female ID patients. Expression analysis in the patients' cell line revealed decreased KDM5C mRNA levels compared to female controls. IQSEC2 levels could not be compared due to very low expression in blood. Overall, our data suggest that heterozygous loss-of-function of the escape genes KDM5C and/or IQSEC2 can contribute to severe ID in female patients and should be taken into account in diagnostics.


Subject(s)
Autistic Disorder/diagnosis , Chromosome Deletion , Guanine Nucleotide Exchange Factors/genetics , Histone Demethylases/genetics , Intellectual Disability/diagnosis , Autistic Disorder/genetics , Female , Gene Expression , Guanine Nucleotide Exchange Factors/metabolism , Histone Demethylases/metabolism , Humans , Intellectual Disability/genetics , Young Adult
2.
J Hum Genet ; 60(4): 207-11, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25652354

ABSTRACT

Recently, we defined a minimal overlapping region for causal Xp11.22 copy number gains in males with intellectual disability (ID), and identified HECT, UBA and WWE domain-containing protein-1 (HUWE1) as the primary dosage-sensitive gene, whose overexpression leads to ID. In the present study, we used this minimal interval to search for HUWE1 copy number variations by quantitative polymerase chain reaction in a large cohort of Brazilian males with idiopathic ID. We detected two unrelated sporadic individuals with syndromic ID carrying unique overlapping duplications encompassing HUWE1. Breakpoint junction analysis showed a simple tandem duplication in the first patient, which has probably arisen by microhomology-mediated break-induced repair mechanism. In the second patient, the rearrangement is complex having an insertion of an intrachromosomal sequence at its junction. This kind of rearrangement has not been reported in Xp11.22 duplications and might have emerged by a replication- or recombination-based mechanism. Furthermore, the presence of infantile seizures in the second family suggests a potential role of increased KDM5C expression on epilepsy. Our findings highlight the importance of microduplications at Xp11.22 to ID, even in sporadic cases, and reveal new clinical and molecular insight into HUWE1 copy number gains.


Subject(s)
Chromosome Duplication , Chromosomes, Human, X , Intellectual Disability/genetics , Ubiquitin-Protein Ligases/genetics , Adolescent , Child , DNA Copy Number Variations , Facies , Female , Genetic Association Studies , Humans , Intellectual Disability/diagnosis , Male , Pedigree , Tumor Suppressor Proteins
3.
Hum Genet ; 133(11): 1359-67, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25037250

ABSTRACT

Xq28 microduplications of MECP2 are a prominent cause of a severe syndromic form of intellectual disability (ID) in males. Females are usually unaffected through near to complete X-inactivation of the aberrant X chromosome (skewing). In rare cases, affected females have been described due to random X-inactivation. Here, we report on two female patients carrying de novo MECP2 microduplications on their fully active X chromosomes. Both patients present with ID and additional clinical features. Mono-allelic expression confirmed complete skewing of X-inactivation. Consequently, significantly enhanced MECP2 mRNA levels were observed. We hypothesize that the cause for the complete skewing is due to a more harmful mutation on the other X chromosome, thereby forcing the MECP2 duplication to become active. However, we could not unequivocally identify such a second mutation by array-CGH or exome sequencing. Our data underline that, like in males, increased MECP2 dosage in females can contribute to ID too, which should be taken into account in diagnostics.


Subject(s)
Gene Expression Regulation , Mental Retardation, X-Linked/genetics , Methyl-CpG-Binding Protein 2/genetics , X Chromosome Inactivation/genetics , Adolescent , Child , Comparative Genomic Hybridization , Exome/genetics , Female , Gene Duplication , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Intellectual Disability/genetics , Microsatellite Repeats/genetics , Oligonucleotide Array Sequence Analysis , Pedigree , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA
4.
Nucleic Acids Res ; 42(9): 5728-41, 2014 May.
Article in English | MEDLINE | ID: mdl-24682812

ABSTRACT

Tandem repeats are short DNA sequences that are repeated head-to-tail with a propensity to be variable. They constitute a significant proportion of the human genome, also occurring within coding and regulatory regions. Variation in these repeats can alter the function and/or expression of genes allowing organisms to swiftly adapt to novel environments. Importantly, some repeat expansions have also been linked to certain neurodegenerative diseases. Therefore, accurate sequencing of tandem repeats could contribute to our understanding of common phenotypic variability and might uncover missing genetic factors in idiopathic clinical conditions. However, despite long-standing evidence for the functional role of repeats, they are largely ignored because of technical limitations in sequencing, mapping and typing. Here, we report on a novel capture technique and data filtering protocol that allowed simultaneous sequencing of thousands of tandem repeats in the human genomes of a three generation family using GS-FLX-plus Titanium technology. Our results demonstrated that up to 7.6% of tandem repeats in this family (4% in coding sequences) differ from the reference sequence, and identified a de novo variation in the family tree. The method opens new routes to look at this underappreciated type of genetic variability, including the identification of novel disease-related repeats.


Subject(s)
Genome, Human , Polymorphism, Genetic , Tandem Repeat Sequences , Base Sequence , Female , Gene Components , Humans , Male , Molecular Sequence Data , Pedigree , Sequence Analysis, DNA
5.
Eur J Hum Genet ; 22(5): 644-51, 2014 May.
Article in English | MEDLINE | ID: mdl-24105372

ABSTRACT

Oligophrenin-1 (OPHN1) is one of at least seven genes located on chromosome X that take part in Rho GTPase-dependent signaling pathways involved in X-linked intellectual disability (XLID). Mutations in OPHN1 were primarily described as an exclusive cause of non-syndromic XLID, but the re-evaluation of the affected individuals using brain imaging displayed fronto-temporal atrophy and cerebellar hypoplasia as neuroanatomical marks. In this study, we describe clinical, genetic and neuroimaging data of a three generation Brazilian XLID family co-segregating a novel intragenic deletion in OPHN1. This deletion results in an in-frame loss of exon 7 at transcription level (c.781_891del; r.487_597del), which is predicted to abolish 37 amino acids from the highly conserved N-terminal BAR domain of OPHN1. cDNA expression analysis demonstrated that the mutant OPHN1 transcript is stable and no abnormal splicing was observed. Features shared by the affected males of this family include neonatal hypotonia, strabismus, prominent root of the nose, deep set eyes, hyperactivity and instability/intolerance to frustration. Cranial MRI scans showed large lateral ventricles, vermis hypoplasia and cystic dilatation of the cisterna magna in all affected males. Interestingly, hippocampal alterations that have not been reported in patients with loss-of-function OPHN1 mutations were found in three affected individuals, suggesting an important function for the BAR domain in the hippocampus. This is the first description of an in-frame deletion within the BAR domain of OPHN1 and could provide new insights into the role of this domain in relation to brain and cognitive development or function.


Subject(s)
Cytoskeletal Proteins/genetics , GTPase-Activating Proteins/genetics , Hippocampus/metabolism , Intellectual Disability/genetics , Nuclear Proteins/genetics , Protein Interaction Domains and Motifs/genetics , Reading Frames , Sequence Deletion , Adolescent , Adult , Brain/metabolism , Child , Comparative Genomic Hybridization , Cytoskeletal Proteins/chemistry , DNA Mutational Analysis , Facies , Female , GTPase-Activating Proteins/chemistry , Hippocampus/pathology , Humans , Intellectual Disability/diagnosis , Magnetic Resonance Imaging , Male , Middle Aged , Nuclear Proteins/chemistry , Pedigree , Phenotype , X Chromosome Inactivation , Young Adult
6.
Hum Mutat ; 35(3): 377-83, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24357492

ABSTRACT

Copy number gains at Xq28 are a frequent cause of X-linked intellectual disability (XLID). Here, we report on a recurrent 0.5 Mb tandem copy number gain at distal Xq28 not including MECP2, in four male patients with nonsyndromic mild ID and behavioral problems. The genomic region is duplicated in two families and triplicated in a third reflected by more distinctive clinical features. The X-inactivation patterns in carrier females correspond well with their clinical symptoms. Our mapping data confirm that this recurrent gain is likely mediated by nonallelic homologous recombination between two directly oriented Int22h repeats. The affected region harbors eight genes of which RAB39B encoding a small GTPase, was the prime candidate since loss-of-function mutations had been linked to ID. RAB39B is expressed at stable levels in lymphocytes from control individuals, suggesting a tight regulation. mRNA levels in our patients were almost two-fold increased. Overexpression of Rab39b in mouse primary hippocampal neurons demonstrated a significant decrease in neuronal branching as well as in the number of synapses when compared with the control neurons. Taken together, we provide evidence that the increased dosage of RAB39B causes a disturbed neuronal development leading to cognitive impairment in patients with this recurrent copy number gain.


Subject(s)
Chromosomes, Human, X/genetics , DNA Copy Number Variations , Intellectual Disability/genetics , rab GTP-Binding Proteins/genetics , Animals , Belgium , Cell Differentiation , Child , Chromosome Mapping , Estonia , Gene Duplication , Gene Expression Regulation , Humans , Male , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Neurons/cytology , Neurons/metabolism , White People , X Chromosome Inactivation
7.
Hum Mutat ; 35(3): 350-5, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24357517

ABSTRACT

The phosphatidylinositol glycan class A (PIGA) protein is a member of the glycosylphosphatidylinositol anchor pathway. Germline mutations in PIGA located at Xp22.2 are thought to be lethal in males. However, a nonsense mutation in the last coding exon was recently described in two brothers with multiple congenital anomalies-hypotonia-seizures syndrome 2 (MCAHS2) who survived through birth likely because of the hypomorphic nature of the truncated protein, but died in their first weeks of life. Here, we report on a frameshift mutation early in the PIGA cDNA (c.76dupT; p.Y26Lfs*3) that cosegregates with the disease in a large family diagnosed with a severe syndromic form of X-linked intellectual disability. Unexpectedly, CD59 surface expression suggested the production of a shorter PIGA protein with residual functionality. We provide evidence that the second methionine at position 37 may be used for the translation of a 36 amino acids shorter PIGA. Complementation assays confirmed that this shorter PIGA cDNA was able to partially rescue the surface expression of CD59 in a PIGA-null cell line. Taken together, our data strongly suggest that the early frameshift mutation in PIGA produces a truncated hypomorph, which is sufficient to rescue the lethality in males but not the MCAHS2-like phenotype.


Subject(s)
Frameshift Mutation , Genes, X-Linked , Intellectual Disability/genetics , Membrane Proteins/genetics , Chromosomes, Human, X/genetics , Exome , Exons , Female , Germ-Line Mutation , Humans , Intellectual Disability/mortality , Male , Pedigree , Phenotype , Sequence Analysis, DNA
8.
Hum Genet ; 132(10): 1177-85, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23783460

ABSTRACT

Loss-of-function mutations in several different neuronal pathways have been related to intellectual disability (ID). Such mutations often are found on the X chromosome in males since they result in functional null alleles. So far, microdeletions at Xq24 reported in males always have been associated with a syndromic form of ID due to the loss of UBE2A. Here, we report on overlapping microdeletions at Xq24 that do not include UBE2A or affect its expression, in patients with non-syndromic ID plus some additional features from three unrelated families. The smallest region of overlap, confirmed by junction sequencing, harbors two members of the mitochondrial solute carrier family 25, SLC25A5 and SLC25A43. However, identification of an intragenic microdeletion including SLC25A43 but not SLC25A5 in a healthy boy excluded a role for SLC25A43 in cognition. Therefore, our findings point to SLC25A5 as a novel gene for non-syndromic ID. This highly conserved gene is expressed ubiquitously with high levels in cortex and hippocampus, and a presumed role in mitochondrial exchange of ADP/ATP. Our data indicate that SLC25A5 is involved in memory formation or establishment, which could add mitochondrial processes to the wide array of pathways that regulate normal cognitive functions.


Subject(s)
Adenine Nucleotide Translocator 2/metabolism , Chromosome Deletion , Chromosomes, Human, X/genetics , Intellectual Disability/genetics , Mitochondria/metabolism , Adenine Nucleotide Translocator 2/genetics , Alu Elements , Base Sequence , Brain/metabolism , Brain/pathology , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Female , Heterozygote , Humans , Infant , Intellectual Disability/pathology , Male , Mitochondria/genetics , Molecular Sequence Data , Pedigree , X Chromosome Inactivation
9.
PLoS One ; 8(5): e64144, 2013.
Article in English | MEDLINE | ID: mdl-23675524

ABSTRACT

Members of the Nuclear eXport Factor (NXF) family are involved in the export of mRNA from the nucleus to the cytoplasm, or hypothesized to play a role in transport of cytoplasmic mRNA. We previously reported on the loss of NXF5 in a male patient with a syndromic form of intellectual disability. To study the functional role of NXF5 we identified the mouse counterpart. Based on synteny, mouse Nxf2 is the ortholog of human NXF5. However, we provide several lines of evidence that mouse Nxf7 is the actual functional equivalent of NXF5. Both Nxf7 and NXF5 are predominantly expressed in the brain, show cytoplasmic localization, and present as granules in neuronal dendrites suggesting a role in cytoplasmic mRNA metabolism in neurons. Nxf7 was primarily detected in the pyramidal cells of the hippocampus and in layer V of the cortex. Similar to human NXF2, mouse Nxf2 is highly expressed in testis and shows a nuclear localization. Interestingly, these findings point to a different evolutionary path for both NXF genes in human and mouse. We thus generated and validated Nxf7 knockout mice, which were fertile and did not present any gross anatomical or morphological abnormalities. Expression profiling in the hippocampus and the cortex did not reveal significant changes between wild-type and Nxf7 knockout mice. However, impaired spatial memory was observed in these KO mice when evaluated in the Morris water maze test. In conclusion, our findings provide strong evidence that mouse Nxf7 is the functional counterpart of human NXF5, which might play a critical role in mRNA metabolism in the brain.


Subject(s)
Cerebral Cortex/metabolism , Founder Effect , Hippocampus/metabolism , Intellectual Disability/genetics , Neurons/metabolism , Nucleocytoplasmic Transport Proteins/genetics , RNA-Binding Proteins/genetics , Active Transport, Cell Nucleus/genetics , Animals , Cerebral Cortex/pathology , Cerebral Cortex/ultrastructure , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Disease Models, Animal , Gene Expression , Hippocampus/pathology , Hippocampus/ultrastructure , Humans , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Maze Learning , Memory , Mice , Mice, Knockout , Neurons/pathology , Neurons/ultrastructure , Nucleocytoplasmic Transport Proteins/metabolism , Organ Specificity , RNA-Binding Proteins/metabolism , Testis/metabolism
10.
Am J Hum Genet ; 91(2): 252-64, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22840365

ABSTRACT

We previously reported on nonrecurrent overlapping duplications at Xp11.22 in individuals with nonsyndromic intellectual disability (ID) harboring HSD17B10, HUWE1, and the microRNAs miR-98 and let-7f-2 in the smallest region of overlap. Here, we describe six additional individuals with nonsyndromic ID and overlapping microduplications that segregate in the families. High-resolution mapping of the 12 copy-number gains reduced the minimal duplicated region to the HUWE1 locus only. Consequently, increased mRNA levels were detected for HUWE1, but not HSD17B10. Marker and SNP analysis, together with identification of two de novo events, suggested a paternally derived intrachromosomal duplication event. In four independent families, we report on a polymorphic 70 kb recurrent copy-number gain, which harbors part of HUWE1 (exon 28 to 3' untranslated region), including miR-98 and let-7f-2. Our findings thus demonstrate that HUWE1 is the only remaining dosage-sensitive gene associated with the ID phenotype. Junction and in silico analysis of breakpoint regions demonstrated simple microhomology-mediated rearrangements suggestive of replication-based duplication events. Intriguingly, in a single family, the duplication was generated through nonallelic homologous recombination (NAHR) with the use of HUWE1-flanking imperfect low-copy repeats, which drive this infrequent NAHR event. The recurrent partial HUWE1 copy-number gain was also generated through NAHR, but here, the homologous sequences used were identified as TcMAR-Tigger DNA elements, a template that has not yet been reported for NAHR. In summary, we showed that an increased dosage of HUWE1 causes nonsyndromic ID and demonstrated that the Xp11.22 region is prone to recombination- and replication-based rearrangements.


Subject(s)
Chromosomes, Human, X/genetics , DNA Copy Number Variations/genetics , Gene Rearrangement/genetics , Intellectual Disability/genetics , Ubiquitin-Protein Ligases/genetics , Chromosome Mapping , Chromosomes, Artificial, Bacterial/genetics , Comparative Genomic Hybridization , Computational Biology , DNA Replication/genetics , Gene Duplication/genetics , Humans , Pedigree , Polymerase Chain Reaction , Polymorphism, Single Nucleotide/genetics , Recombination, Genetic/genetics , Tumor Suppressor Proteins
11.
Pediatrics ; 128(4): e1029-33, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21930553

ABSTRACT

Multiplex ligation-dependent probe amplification (MLPA) and array- comparative genomic hybridization analysis have been proven to be useful in the identification of submicroscopic copy-number imbalances in families with nonsyndromic X-linked intellectual disability (NS-XLID). Here we report the first description of a child with mild intellectual disability and a submicroscopic duplication at Xp22.12 identified by MLPA with a P106 MRX kit (MRC-Holland, Amsterdam, Netherlands) and further confirmed and characterized with a custom 244-k oligo-array, fluorescence in situ hybridization, quantitative polymerase chain reaction (qPCR), and immunoblotting. This 1.05-megabase duplication encompasses 7 genes, RPS6KA3 being the only of these genes known to be related to ID. The proband was an 8-year-old boy referred to the genetics unit for psychomotor retardation and learning disabilities. Both maternal brothers also showed learning difficulties and delayed language during childhood in a similar way to the proband. These boys also carried the duplication, as did the healthy mother and grandmother of the proband. The same duplication was also observed in the 5-year-old younger brother who presented with features of developmental delay and learning disabilities during the previous year. Increased RPS6KA3/RSK2 levels were demonstrated in the proband by qPCR and immunoblotting. To our knowledge, this is the first family identified with a submicroscopic duplication including the entire RPS6KA3/RSK2 gene, and our findings suggest that an increased dose of this gene is responsible for a mild form of NS-XLID.


Subject(s)
Chromosomes, Human, X/genetics , Gene Duplication , Mental Retardation, X-Linked/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Sex Chromosome Aberrations , Child , Humans , Immunoblotting , In Situ Hybridization, Fluorescence , Male , Mental Retardation, X-Linked/diagnosis , Oligonucleotide Array Sequence Analysis , Pedigree , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...