Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Gastrointest Endosc ; 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38360118

ABSTRACT

BACKGROUND AND AIMS: Obtaining adequate tissue samples in subepithelial lesions (SELs) remains challenging. Several biopsy techniques are available, but a systematic review including all available techniques to obtain a histological diagnosis of SEL is lacking. The aim of this study is to evaluate the diagnostic yield and complication rates of endoscopic biopsies, endoscopic ultrasound guided-FNA and -FNB (EUS-FNA/B), and mucosal incision assisted biopsy (MIAB) for SELs in the upper gastro-intestinal tract. METHODS: A search strategy in multiple databases was performed. The primary outcome was diagnostic yield, defined as the percentage of procedures where histology was obtained and resulted in a definitive histopathological diagnosis. Secondary outcome measures included reported procedure-related adverse events which were classified according to the AGREE classification. RESULTS: A total of 94 original papers were included. Studies were classified per endoscopic technique to obtain histopathology. This resulted in 8 included studies for endoscopic biopsy methods, 55 studies for EUS-FNA, 33 studies for EUS-FNB and 26 studies for MIAB. Pooled rates for diagnostic yield were 40.6% (95% CI 30.8-51.2%) for endoscopic biopsy, 74.6% (95% CI 69.9-78.7%) for EUS-FNA, 84.2% (95% CI 80.7-87.2%) for EUS-FNB, 88.2% (95% CI 84.7-91.1%) for MIAB. Reported procedure-related complications graded AGREE II or higher were 2.8-3.9% for endoscopic biopsies, 1.0-4.5% for EUS-FNA, 0.9-7.7 % for EUS-FNB, 1.9-7.9% for MIAB. CONCLUSION: Based on the available evidence, MIAB and EUS-FNB seem to be most effective in terms of achieving a high diagnostic yield, with similar rates of adverse events.

3.
Genet Med ; 24(10): 2051-2064, 2022 10.
Article in English | MEDLINE | ID: mdl-35833929

ABSTRACT

PURPOSE: Although haploinsufficiency of ANKRD11 is among the most common genetic causes of neurodevelopmental disorders, the role of rare ANKRD11 missense variation remains unclear. We characterized clinical, molecular, and functional spectra of ANKRD11 missense variants. METHODS: We collected clinical information of individuals with ANKRD11 missense variants and evaluated phenotypic fit to KBG syndrome. We assessed pathogenicity of variants through in silico analyses and cell-based experiments. RESULTS: We identified 20 unique, mostly de novo, ANKRD11 missense variants in 29 individuals, presenting with syndromic neurodevelopmental disorders similar to KBG syndrome caused by ANKRD11 protein truncating variants or 16q24.3 microdeletions. Missense variants significantly clustered in repression domain 2 at the ANKRD11 C-terminus. Of the 10 functionally studied missense variants, 6 reduced ANKRD11 stability. One variant caused decreased proteasome degradation and loss of ANKRD11 transcriptional activity. CONCLUSION: Our study indicates that pathogenic heterozygous ANKRD11 missense variants cause the clinically recognizable KBG syndrome. Disrupted transrepression capacity and reduced protein stability each independently lead to ANKRD11 loss-of-function, consistent with haploinsufficiency. This highlights the diagnostic relevance of ANKRD11 missense variants, but also poses diagnostic challenges because the KBG-associated phenotype may be mild and inherited pathogenic ANKRD11 (missense) variants are increasingly observed, warranting stringent variant classification and careful phenotyping.


Subject(s)
Abnormalities, Multiple , Bone Diseases, Developmental , Intellectual Disability , Repressor Proteins , Tooth Abnormalities , Abnormalities, Multiple/genetics , Bone Diseases, Developmental/etiology , Bone Diseases, Developmental/genetics , Chromosome Deletion , Facies , Humans , Intellectual Disability/genetics , Mutation, Missense , Phenotype , Proteasome Endopeptidase Complex/genetics , Repressor Proteins/genetics , Tooth Abnormalities/diagnosis , Transcription Factors/genetics
4.
Eur J Hum Genet ; 30(3): 282-290, 2022 03.
Article in English | MEDLINE | ID: mdl-34719684

ABSTRACT

Craniosynostosis is a birth defect occurring in approximately one in 2000 live births, where premature fusion of the cranial bones inhibits growth of the skull during critical periods of brain development. The resulting changes in skull shape can lead to compression of the brain, causing severe complications. While we have some understanding of the molecular pathology of craniosynostosis, a large proportion of cases are of unknown genetic aetiology. Based on studies in mouse, we previously proposed that the ciliopathy gene Fuz should be considered a candidate craniosynostosis gene. Here, we report a novel variant of FUZ (c.851 G > C, p.(Arg284Pro)) found in monozygotic twins presenting with craniosynostosis. To investigate whether Fuz has a direct role in regulating osteogenic fate and mineralisation, we cultured primary osteoblasts and mouse embryonic fibroblasts (MEFs) from Fuz mutant mice. Loss of Fuz resulted in increased osteoblastic mineralisation. This suggests that FUZ protein normally acts as a negative regulator of osteogenesis. We then used Fuz mutant MEFs, which lose functional primary cilia, to test whether the FUZ p.(Arg284Pro) variant could restore FUZ function during ciliogenesis. We found that expression of the FUZ p.(Arg284Pro) variant was sufficient to partially restore cilia numbers, but did not mediate a comparable response to Hedgehog pathway activation. Together, this suggests the osteogenic effects of FUZ p.(Arg284Pro) do not depend upon initiation of ciliogenesis.


Subject(s)
Craniosynostoses , Cytoskeletal Proteins/genetics , Hedgehog Proteins , Intracellular Signaling Peptides and Proteins/genetics , Animals , Craniosynostoses/diagnosis , Craniosynostoses/genetics , Fibroblasts/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Mice
5.
Eur J Orthod ; 44(3): 287-293, 2022 05 24.
Article in English | MEDLINE | ID: mdl-34424951

ABSTRACT

OBJECTIVES: To determine whether dental maturity (dental development) was delayed in patients with Muenke syndrome, Saethre-Chotzen syndrome, and TCF12-related craniosynostosis, compared with a Dutch control group without syndromes. MATERIALS AND METHODS: This study included 60 patients (38 patients with Muenke syndrome, 17 patients with Saethre-Chotzen syndrome, and 5 with TCF12-related craniosynostosis), aged 5.8-16.8 years that were treated at the Department of Oral Maxillofacial Surgery, Special Dental Care, and Orthodontics, in Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, the Netherlands. Dental age was calculated according to Demirjian's index of dental maturity. The control group included 451 children without a syndrome. RESULTS: Compared with the control group, dental development was delayed by an average of one year in 5- to 8-year-old patients with Muenke syndrome (P = 0.007) and in 8- to 10-year-old patients with Saethre-Chotzen syndrome (P = 0.044), but not in patients with TCF12-related craniosynostosis. CONCLUSIONS: Our results indicated that dental development was delayed by one year, on average, in patients with Muenke syndrome and Saethre-Chotzen syndrome, compared with a Dutch control group without syndromes. IMPLICATIONS: Our findings have improved the understanding of dental development in patients with Muenke and Saethre-Chotzen syndrome. These results can provide guidance on whether the orthodontist needs to consider growth disturbances related to dental development.


Subject(s)
Acrocephalosyndactylia , Craniosynostoses , Acrocephalosyndactylia/diagnostic imaging , Acrocephalosyndactylia/surgery , Basic Helix-Loop-Helix Transcription Factors , Child , Child, Preschool , Craniosynostoses/complications , Craniosynostoses/diagnostic imaging , Craniosynostoses/surgery , Humans , Netherlands , Syndrome
6.
Front Immunol ; 11: 614, 2020.
Article in English | MEDLINE | ID: mdl-32373116

ABSTRACT

Background: Genetic tests for primary immunodeficiency disorders (PIDs) are expensive, time-consuming, and not easily accessible in developing countries. Therefore, we studied the feasibility of a customized single nucleotide variant (SNV) microarray that we developed to detect disease-causing variants and copy number variation (CNV) in patients with PIDs for only 40 Euros. Methods: Probes were custom-designed to genotype 9,415 variants of 277 PID-related genes, and were added to the genome-wide Illumina Global Screening Array (GSA). Data analysis of GSA was performed using Illumina GenomeStudio 2.0, Biodiscovery Nexus 10.0, and R-3.4.4 software. Validation of genotype calling was performed by comparing the GSA with whole-genome sequencing (WGS) data of 56 non-PID controls. DNA samples of 95 clinically diagnosed PID patients, of which 60 patients (63%) had a genetically established diagnosis (by Next-Generation Sequencing (NGS) PID panels or Sanger sequencing), were analyzed to test the performance of the GSA. The additional SNVs detected by GSA were validated by Sanger sequencing. Results: Genotype calling of the customized array had an accuracy rate of 99.7%. The sensitivity for detecting rare PID variants was high (87%). The single sample replication in two runs was high (94.9%). The customized GSA was able to generate a genetic diagnosis in 37 out of 95 patients (39%). These 37 patients included 29 patients in whom the genetic variants were confirmed by conventional methods (26 patients by SNV and 3 by CNV analysis), while in 8 patients a new genetic diagnosis was established (6 patients by SNV and 2 patients suspected for leukemia by CNV analysis). Twenty-eight patients could not be detected due to the limited coverage of the custom probes. However, the diagnostic yield can potentially be increased when newly updated variants are added. Conclusion: Our robust customized GSA seems to be a promising first-line rapid screening tool for PIDs at an affordable price, which opens opportunities for low-cost genetic testing in developing countries. The technique is scalable, allows numerous new genetic variants to be added, and offers the potential for genetic testing not only in PIDs, but also in many other genetic diseases.


Subject(s)
Genetic Testing/methods , Genotyping Techniques/methods , High-Throughput Nucleotide Sequencing/methods , Primary Immunodeficiency Diseases/genetics , Costs and Cost Analysis , DNA Copy Number Variations , Genotyping Techniques/economics , Humans , Polymorphism, Single Nucleotide , Reproducibility of Results
7.
Hum Mol Genet ; 28(15): 2501-2513, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31067316

ABSTRACT

Craniosynostosis, the premature ossification of cranial sutures, is a developmental disorder of the skull vault, occurring in approximately 1 in 2250 births. The causes are heterogeneous, with a monogenic basis identified in ~25% of patients. Using whole-genome sequencing, we identified a novel, de novo variant in BCL11B, c.7C>A, encoding an R3S substitution (p.R3S), in a male patient with coronal suture synostosis. BCL11B is a transcription factor that interacts directly with the nucleosome remodelling and deacetylation complex (NuRD) and polycomb-related complex 2 (PRC2) through the invariant proteins RBBP4 and RBBP7. The p.R3S substitution occurs within a conserved amino-terminal motif (RRKQxxP) of BCL11B and reduces interaction with both transcriptional complexes. Equilibrium binding studies and molecular dynamics simulations show that the p.R3S substitution disrupts ionic coordination between BCL11B and the RBBP4-MTA1 complex, a subassembly of the NuRD complex, and increases the conformational flexibility of Arg-4, Lys-5 and Gln-6 of BCL11B. These alterations collectively reduce the affinity of BCL11B p.R3S for the RBBP4-MTA1 complex by nearly an order of magnitude. We generated a mouse model of the BCL11B p.R3S substitution using a CRISPR-Cas9-based approach, and we report herein that these mice exhibit craniosynostosis of the coronal suture, as well as other cranial sutures. This finding provides strong evidence that the BCL11B p.R3S substitution is causally associated with craniosynostosis and confirms an important role for BCL11B in the maintenance of cranial suture patency.


Subject(s)
Chromatin Assembly and Disassembly , Cranial Sutures/growth & development , Craniosynostoses/metabolism , Mutation, Missense , Nucleosomes/metabolism , Osteogenesis , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Cranial Sutures/metabolism , Craniosynostoses/genetics , Craniosynostoses/physiopathology , DNA Mutational Analysis , Disease Models, Animal , Humans , Infant , Male , Mice , Protein Binding , Protein Conformation , Repressor Proteins/metabolism , Repressor Proteins/physiology , Retinoblastoma-Binding Protein 4/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology , White People , Whole Genome Sequencing
8.
Mol Syndromol ; 10(1-2): 6-23, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30976276

ABSTRACT

In 1993, Jabs et al. were the first to describe a genetic origin of craniosynostosis. Since this discovery, the genetic causes of the most common syndromes have been described. In 2015, a total of 57 human genes were reported for which there had been evidence that mutations were causally related to craniosynostosis. Facilitated by rapid technological developments, many others have been identified since then. Reviewing the literature, we characterize the most common craniosynostosis syndromes followed by a description of the novel causes that were identified between January 2015 and December 2017.

9.
Am J Hum Genet ; 104(4): 709-720, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30905399

ABSTRACT

The Mediator is an evolutionarily conserved, multi-subunit complex that regulates multiple steps of transcription. Mediator activity is regulated by the reversible association of a four-subunit module comprising CDK8 or CDK19 kinases, together with cyclin C, MED12 or MED12L, and MED13 or MED13L. Mutations in MED12, MED13, and MED13L were previously identified in syndromic developmental disorders with overlapping phenotypes. Here, we report CDK8 mutations (located at 13q12.13) that cause a phenotypically related disorder. Using whole-exome or whole-genome sequencing, and by international collaboration, we identified eight different heterozygous missense CDK8 substitutions, including 10 shown to have arisen de novo, in 12 unrelated subjects; a recurrent mutation, c.185C>T (p.Ser62Leu), was present in five individuals. All predicted substitutions localize to the ATP-binding pocket of the kinase domain. Affected individuals have overlapping phenotypes characterized by hypotonia, mild to moderate intellectual disability, behavioral disorders, and variable facial dysmorphism. Congenital heart disease occurred in six subjects; additional features present in multiple individuals included agenesis of the corpus callosum, ano-rectal malformations, seizures, and hearing or visual impairments. To evaluate the functional impact of the mutations, we measured phosphorylation at STAT1-Ser727, a known CDK8 substrate, in a CDK8 and CDK19 CRISPR double-knockout cell line transfected with wild-type (WT) or mutant CDK8 constructs. These experiments demonstrated a reduction in STAT1 phosphorylation by all mutants, in most cases to a similar extent as in a kinase-dead control. We conclude that missense mutations in CDK8 cause a developmental disorder that has phenotypic similarity to syndromes associated with mutations in other subunits of the Mediator kinase module, indicating probable overlap in pathogenic mechanisms.


Subject(s)
Cyclin-Dependent Kinase 8/genetics , Developmental Disabilities/genetics , Mediator Complex/genetics , Mutation, Missense , Brain/abnormalities , Child , Child, Preschool , Cyclin C/genetics , Cyclin-Dependent Kinases/genetics , Exome , Female , Heart Defects, Congenital/genetics , Heterozygote , Humans , Infant , Intellectual Disability/genetics , Male , Mutation , Phenotype , Phosphorylation , Syndrome
10.
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
11.
Eur J Hum Genet ; 25(10): 1126-1133, 2017 10.
Article in English | MEDLINE | ID: mdl-28905882

ABSTRACT

Burn-McKeown syndrome (BMKS) is a rare syndrome characterized by choanal atresia, prominent ears, abnormalities of the outer third of the lower eyelid, structural cardiac abnormalities, conductive and sensorineural hearing loss, and cleft lip. Recently, causative compound heterozygous variants were identified in TXNL4A. We analyzed an individual with clinical features of BMKS and her parents by whole-genome sequencing and identified compound heterozygous variants in TXNL4A (a novel splice site variant (c.258-2A>G, (p.?)) and a 34 bp promoter deletion (hg19 chr18:g.77748581_77748614del (type 1Δ) in the proband). Subsequently, we tested a cohort of 19 individuals with (mild) features of BMKS and 17 individuals with isolated choanal atresia for causative variants in TXNL4A by dideoxy-sequence analysis. In one individual with BMKS unrelated to the first family, we identified the identical compound heterozygous variants. In an individual with isolated choanal atresia, we found homozygosity for the same type 1Δ promoter deletion, whilst in two cousins from a family with choanal atresia and other minor anomalies we found homozygosity for a different deletion within the promoter (hg19 chr18: g.77748604_77748637del (type 2Δ)). Hence, we identified causative recessive variants in TXNL4A in two individuals with BMKS as well as in three individuals (from two families) with isolated choanal atresia.


Subject(s)
Choanal Atresia/genetics , Deafness/congenital , Gene Deletion , Heart Defects, Congenital/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Choanal Atresia/diagnosis , Deafness/diagnosis , Deafness/genetics , Facies , Female , Heart Defects, Congenital/diagnosis , Heterozygote , Homozygote , Humans , Male , Pedigree , Promoter Regions, Genetic
12.
J Med Genet ; 54(4): 260-268, 2017 04.
Article in English | MEDLINE | ID: mdl-27884935

ABSTRACT

BACKGROUND: Craniosynostosis, the premature fusion of one or more cranial sutures, occurs in ∼1 in 2250 births, either in isolation or as part of a syndrome. Mutations in at least 57 genes have been associated with craniosynostosis, but only a minority of these are included in routine laboratory genetic testing. METHODS: We used exome or whole genome sequencing to seek a genetic cause in a cohort of 40 subjects with craniosynostosis, selected by clinical or molecular geneticists as being high-priority cases, and in whom prior clinically driven genetic testing had been negative. RESULTS: We identified likely associated mutations in 15 patients (37.5%), involving 14 different genes. All genes were mutated in single families, except for IL11RA (two families). We classified the other positive diagnoses as follows: commonly mutated craniosynostosis genes with atypical presentation (EFNB1, TWIST1); other core craniosynostosis genes (CDC45, MSX2, ZIC1); genes for which mutations are only rarely associated with craniosynostosis (FBN1, HUWE1, KRAS, STAT3); and known disease genes for which a causal relationship with craniosynostosis is currently unknown (AHDC1, NTRK2). In two further families, likely novel disease genes are currently undergoing functional validation. In 5 of the 15 positive cases, the (previously unanticipated) molecular diagnosis had immediate, actionable consequences for either genetic or medical management (mutations in EFNB1, FBN1, KRAS, NTRK2, STAT3). CONCLUSIONS: This substantial genetic heterogeneity, and the multiple actionable mutations identified, emphasises the benefits of exome/whole genome sequencing to identify causal mutations in craniosynostosis cases for which routine clinical testing has yielded negative results.


Subject(s)
Craniosynostoses/genetics , Genome, Human , High-Throughput Nucleotide Sequencing , Neoplasm Proteins/genetics , Craniosynostoses/diagnosis , Craniosynostoses/pathology , Exome/genetics , Genetic Testing , Humans , Mutation , Predictive Value of Tests
13.
Hum Mutat ; 37(8): 732-6, 2016 08.
Article in English | MEDLINE | ID: mdl-27158814

ABSTRACT

TCF12-related craniosynostosis can be caused by small heterozygous loss-of-function mutations in TCF12. Large intragenic rearrangements, however, have not been described yet. Here, we present the identification of four large rearrangements in TCF12 causing TCF12-related craniosynostosis. Whole-genome sequencing was applied on the DNA of 18 index cases with coronal synostosis and their family members (43 samples in total). The data were analyzed using an autosomal-dominant disease model. Structural variant analysis reported intragenic exon deletions (of sizes 84.9, 8.6, and 5.4 kb) in TCF12 in three different families. The results were confirmed by deletion-specific PCR and dideoxy-sequence analysis. Separately, targeted sequencing of the TCF12 genomic region in a patient with coronal synostosis identified a tandem duplication of 11.3 kb. The pathogenic effect of this duplication was confirmed by cDNA analysis. These findings indicate the importance of screening for larger rearrangements in patients suspected to have TCF12-related craniosynostosis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Craniosynostoses/genetics , Sequence Analysis, DNA/methods , Sequence Deletion , Tandem Repeat Sequences , Base Sequence , Exons , Female , Genetic Predisposition to Disease , Humans , Male , Mutation , Pedigree
14.
Am J Hum Genet ; 97(3): 378-88, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26340333

ABSTRACT

Human ZIC1 (zinc finger protein of cerebellum 1), one of five homologs of the Drosophila pair-rule gene odd-paired, encodes a transcription factor previously implicated in vertebrate brain development. Heterozygous deletions of ZIC1 and its nearby paralog ZIC4 on chromosome 3q25.1 are associated with Dandy-Walker malformation of the cerebellum, and loss of the orthologous Zic1 gene in the mouse causes cerebellar hypoplasia and vertebral defects. We describe individuals from five families with heterozygous mutations located in the final (third) exon of ZIC1 (encoding four nonsense and one missense change) who have a distinct phenotype in which severe craniosynostosis, specifically involving the coronal sutures, and variable learning disability are the most characteristic features. The location of the nonsense mutations predicts escape of mutant ZIC1 transcripts from nonsense-mediated decay, which was confirmed in a cell line from an affected individual. Both nonsense and missense mutations are associated with altered and/or enhanced expression of a target gene, engrailed-2, in a Xenopus embryo assay. Analysis of mouse embryos revealed a localized domain of Zic1 expression at embryonic days 11.5-12.5 in a region overlapping the supraorbital regulatory center, which patterns the coronal suture. We conclude that the human mutations uncover a previously unsuspected role for Zic1 in early cranial suture development, potentially by regulating engrailed 1, which was previously shown to be critical for positioning of the murine coronal suture. The diagnosis of a ZIC1 mutation has significant implications for prognosis and we recommend genetic testing when common causes of coronal synostosis have been excluded.


Subject(s)
Codon, Nonsense/genetics , Craniosynostoses/genetics , Gene Expression Regulation, Developmental/genetics , Learning Disabilities/genetics , Phenotype , Transcription Factors/genetics , Animals , Base Sequence , Cloning, Molecular , Female , Homeodomain Proteins/metabolism , Humans , In Situ Hybridization , Karyotyping , Male , Mice , Molecular Sequence Data , Mutation, Missense/genetics , Nerve Tissue Proteins/metabolism , Pedigree , Sequence Analysis, DNA , Xenopus laevis
15.
Am J Med Genet A ; 167A(1): 123-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25425289

ABSTRACT

Craniosynostosis is a congenital anomaly that can occur as an isolated condition or as part of a syndrome. Although several genes are known to cause syndromic craniosynostosis, only 24% can be attributed to known genes. Therefore, it is likely that more mutations and other genes are involved. We present the identification of a novel point mutation in fibroblast growth factor receptor 2 (FGFR2), c.812G>T, p.(Gly271Val) or c.1851G>C, p.(Leu617Phe). Furthermore, we describe a mutation that has been identified just recently, c.812G>T, (p.Gly271Val) or c.1851G>C, (p.Leu617Phe). In addition, we describe findings from a sequence analysis of all coding exons and exon/intron boundaries of FGFR2 performed on 124 patients with syndromic craniosynostosis.


Subject(s)
Mutation/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Adult , Child , Child, Preschool , Exons/genetics , Facies , Female , Humans , Infant , Infant, Newborn , Introns/genetics , Male , Sequence Analysis, DNA
16.
BMC Med Genet ; 15: 95, 2014 Aug 31.
Article in English | MEDLINE | ID: mdl-25174698

ABSTRACT

BACKGROUND: Mutations of fibroblast growth factor receptor 2 (FGFR2) account for a higher proportion of genetic cases of craniosynostosis than any other gene, and are associated with a wide spectrum of severity of clinical problems. Many of these mutations are highly recurrent and their associated features well documented. Crouzon syndrome is typically caused by heterozygous missense mutations in the third immunoglobulin domain of FGFR2. CASE PRESENTATION: Here we describe two families, each segregating a different, previously unreported FGFR2 mutation of the same nucleotide, c.1083A>G and c.1083A>T, both of which encode an apparently synonymous change at the Pro361 codon. We provide experimental evidence that these mutations affect normal FGFR2 splicing and document the clinical consequences, which include a mild Crouzon syndrome phenotype and reduced penetrance of craniosynostosis. CONCLUSIONS: These observations add to a growing list of FGFR2 mutations that affect splicing and provide important clinical information for genetic counselling of families affected by these specific mutations.


Subject(s)
Amino Acid Substitution , Craniofacial Dysostosis/genetics , Craniofacial Dysostosis/pathology , Craniosynostoses/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Craniosynostoses/pathology , Exons , Female , Heterozygote , Humans , Male , Mutation, Missense , Pedigree , RNA Splicing
17.
Nat Genet ; 45(3): 304-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23354436

ABSTRACT

Craniosynostosis, the premature fusion of the cranial sutures, is a heterogeneous disorder with a prevalence of ∼1 in 2,200 (refs. 1,2). A specific genetic etiology can be identified in ∼21% of cases, including mutations of TWIST1, which encodes a class II basic helix-loop-helix (bHLH) transcription factor, and causes Saethre-Chotzen syndrome, typically associated with coronal synostosis. Using exome sequencing, we identified 38 heterozygous TCF12 mutations in 347 samples from unrelated individuals with craniosynostosis. The mutations predominantly occurred in individuals with coronal synostosis and accounted for 32% and 10% of subjects with bilateral and unilateral pathology, respectively. TCF12 encodes one of three class I E proteins that heterodimerize with class II bHLH proteins such as TWIST1. We show that TCF12 and TWIST1 act synergistically in a transactivation assay and that mice doubly heterozygous for loss-of-function mutations in Tcf12 and Twist1 have severe coronal synostosis. Hence, the dosage of TCF12-TWIST1 heterodimers is critical for normal coronal suture development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Craniosynostoses , Nuclear Proteins/genetics , Twist-Related Protein 1/genetics , Acrocephalosyndactylia/complications , Acrocephalosyndactylia/genetics , Acrocephalosyndactylia/pathology , Animals , Cranial Sutures/growth & development , Cranial Sutures/pathology , Craniosynostoses/complications , Craniosynostoses/genetics , Craniosynostoses/pathology , Dimerization , Exome , Gene Expression Regulation, Developmental , Heterozygote , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Sequence Analysis, DNA , Transcriptional Activation
18.
PLoS One ; 7(4): e35293, 2012.
Article in English | MEDLINE | ID: mdl-22558137

ABSTRACT

The ventromedial medulla (VM), subdivided in a rostral (RVM) and a caudal (CVM) part, has a powerful influence on the spinal cord. In this study, we have identified the distribution of glycine and GABA containing neurons in the VM with projections to the cervical spinal cord, the lumbar dorsal horn, and the lumbar ventral horn. For this purpose, we have combined retrograde tracing using fluorescent microspheres with fluorescent in situ hybridization (FISH) for glycine transporter 2 (GlyT2) and GAD67 mRNAs to identify glycinergic and/or GABAergic (Gly/GABA) neurons. Since the results obtained with FISH for GlyT2, GAD67, or GlyT2 + GAD67 mRNAs were not significantly different, we concluded that glycine and GABA coexisted in the various projection neurons. After injections in the cervical cord, we found that 29% ± 1 (SEM) of the retrogradely labeled neurons in the VM were Gly/GABA (RVM: 43%; CVM: 21%). After lumbar dorsal horn injections 31% ± 3 of the VM neurons were Gly/GABA (RVM: 45%; CVM: 12%), and after lumbar ventral horn injections 25% ± 2 were Gly/GABA (RVM: 35%; CVM: 17%). In addition, we have identified a novel ascending Gly/GABA pathway originating from neurons in the area around the central canal (CC) throughout the spinal cord and projecting to the RVM, emphasizing the interaction between the ventromedial medulla and the spinal cord. The present study has now firmly established that GABA and glycine are present in many VM neurons that project to the spinal cord. These neurons strongly influence spinal processing, most notably the inhibition of nociceptive transmission.


Subject(s)
GABAergic Neurons/metabolism , Glycine/metabolism , Medulla Oblongata/cytology , Nociception/physiology , Spinal Cord/cytology , Animals , Fluorescence , Glutamate Decarboxylase/metabolism , Glycine Plasma Membrane Transport Proteins/metabolism , In Situ Hybridization, Fluorescence , Microspheres , Neural Pathways/physiology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...