Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Genet Med ; 22(3): 598-609, 2020 03.
Article in English | MEDLINE | ID: mdl-31700164

ABSTRACT

PURPOSE: Most classical aniridia is caused by PAX6 haploinsufficiency. PAX6 missense variants can be hypomorphic or mimic haploinsufficiency. We hypothesized that missense variants also cause previously undescribed disease by altering the affinity and/or specificity of PAX6 genomic interactions. METHODS: We screened PAX6 in 372 individuals with bilateral microphthalmia, anophthalmia, or coloboma (MAC) from the Medical Research Council Human Genetics Unit eye malformation cohort (HGUeye) and reviewed data from the Deciphering Developmental Disorders study. We performed cluster analysis on PAX6-associated ocular phenotypes by variant type and molecular modeling of the structural impact of 86 different PAX6 causative missense variants. RESULTS: Eight different PAX6 missense variants were identified in 17 individuals (15 families) with MAC, accounting for 4% (15/372) of our cohort. Seven altered the paired domain (p.[Arg26Gln]x1, p.[Gly36Val]x1, p.[Arg38Trp]x2, p.[Arg38Gln]x1, p.[Gly51Arg]x2, p.[Ser54Arg]x2, p.[Asn124Lys]x5) and one the homeodomain (p.[Asn260Tyr]x1). p.Ser54Arg and p.Asn124Lys were exclusively associated with severe bilateral microphthalmia. MAC-associated variants were predicted to alter but not ablate DNA interaction, consistent with the electrophoretic mobility shifts observed using mutant paired domains with well-characterized PAX6-binding sites. We found no strong evidence for novel PAX6-associated extraocular disease. CONCLUSION: Altering the affinity and specificity of PAX6-binding genome-wide provides a plausible mechanism for the worse-than-null effects of MAC-associated missense variants.


Subject(s)
Eye Abnormalities/genetics , Genetic Predisposition to Disease , Microphthalmos/genetics , PAX6 Transcription Factor/genetics , Adolescent , Adult , Binding Sites/genetics , Child , Child, Preschool , Cohort Studies , DNA-Binding Proteins/genetics , Eye Abnormalities/pathology , Female , Heterozygote , Humans , Infant , Male , Microphthalmos/pathology , Mutation, Missense/genetics , Pedigree , Young Adult
2.
Genet Med ; 22(3): 547-556, 2020 03.
Article in English | MEDLINE | ID: mdl-31649276

ABSTRACT

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


Subject(s)
Craniofacial Abnormalities/genetics , DNA-Directed RNA Polymerases/genetics , Mandibulofacial Dysostosis/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Craniofacial Abnormalities/pathology , Genetic Predisposition to Disease , Humans , Mandibulofacial Dysostosis/pathology , Mutation , Neural Crest/abnormalities , Neural Crest/pathology , Tumor Suppressor Protein p53/genetics , Exome Sequencing , Zebrafish/genetics
3.
Prenat Diagn ; 40(10): 1300-1309, 2020 09.
Article in English | MEDLINE | ID: mdl-32627857

ABSTRACT

OBJECTIVE: Conventional genetic tests (quantitative fluorescent-PCR [QF-PCR] and single nucleotide polymorphism-array) only diagnose ~40% of fetuses showing ultrasound abnormalities. Rapid exome sequencing (rES) may improve this diagnostic yield, but includes challenges such as uncertainties in fetal phenotyping, variant interpretation, incidental unsolicited findings, and rapid turnaround times. In this study, we implemented rES in prenatal care to increase diagnostic yield. METHODS: We prospectively studied 55 fetuses. Inclusion criteria were: (a) two or more independent major fetal anomalies, (b) hydrops fetalis or bilateral renal cysts alone, or (c) one major fetal anomaly and a first-degree relative with the same anomaly. In addition to conventional genetic tests, we performed trio rES analysis using a custom virtual gene panel of ~3850 Online Mendelian Inheritance in Man (OMIM) genes. RESULTS: We established a genetic rES-based diagnosis in 8 out of 23 fetuses (35%) without QF-PCR or array abnormalities. Diagnoses included MIRAGE (SAMD9), Zellweger (PEX1), Walker-Warburg (POMGNT1), Noonan (PTNP11), Kabuki (KMT2D), and CHARGE (CHD7) syndrome and two cases of Osteogenesis Imperfecta type 2 (COL1A1). In six cases, rES diagnosis aided perinatal management. The median turnaround time was 14 (range 8-20) days. CONCLUSION: Implementing rES as a routine test in the prenatal setting is challenging but technically feasible, with a promising diagnostic yield and significant clinical relevance.


Subject(s)
Abnormalities, Multiple/diagnosis , Exome Sequencing , Prenatal Diagnosis/methods , Abnormalities, Multiple/epidemiology , Abnormalities, Multiple/genetics , Adult , Diagnostic Tests, Routine/statistics & numerical data , Feasibility Studies , Female , Fetus/diagnostic imaging , Genetic Testing/methods , Genetic Testing/statistics & numerical data , Humans , Infant, Newborn , Male , Netherlands/epidemiology , Polymerase Chain Reaction/methods , Polymorphism, Single Nucleotide , Predictive Value of Tests , Pregnancy , Pregnancy Outcome/epidemiology , Prenatal Diagnosis/statistics & numerical data , Prospective Studies , Ultrasonography, Prenatal
4.
Proc Natl Acad Sci U S A ; 114(13): E2739-E2747, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28292896

ABSTRACT

Megacystis microcolon intestinal hypoperistalsis syndrome (MMIHS) is a congenital visceral myopathy characterized by severe dilation of the urinary bladder and defective intestinal motility. The genetic basis of MMIHS has been ascribed to spontaneous and autosomal dominant mutations in actin gamma 2 (ACTG2), a smooth muscle contractile gene. However, evidence suggesting a recessive origin of the disease also exists. Using combined homozygosity mapping and whole exome sequencing, a genetically isolated family was found to carry a premature termination codon in Leiomodin1 (LMOD1), a gene preferentially expressed in vascular and visceral smooth muscle cells. Parents heterozygous for the mutation exhibited no abnormalities, but a child homozygous for the premature termination codon displayed symptoms consistent with MMIHS. We used CRISPR-Cas9 (CRISPR-associated protein) genome editing of Lmod1 to generate a similar premature termination codon. Mice homozygous for the mutation showed loss of LMOD1 protein and pathology consistent with MMIHS, including late gestation expansion of the bladder, hydronephrosis, and rapid demise after parturition. Loss of LMOD1 resulted in a reduction of filamentous actin, elongated cytoskeletal dense bodies, and impaired intestinal smooth muscle contractility. These results define LMOD1 as a disease gene for MMIHS and suggest its role in establishing normal smooth muscle cytoskeletal-contractile coupling.


Subject(s)
Abnormalities, Multiple/genetics , Autoantigens/physiology , Colon/abnormalities , Cytoskeletal Proteins/physiology , Intestinal Pseudo-Obstruction/genetics , Muscle Proteins/physiology , Urinary Bladder/abnormalities , Animals , Autoantigens/genetics , Autoantigens/metabolism , Codon, Nonsense , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Humans , Infant, Newborn , Mice , Muscle Contraction/genetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth/physiology
5.
Gastroenterology ; 155(1): 130-143.e15, 2018 07.
Article in English | MEDLINE | ID: mdl-29604290

ABSTRACT

BACKGROUND & AIMS: Congenital diarrheal disorders are rare inherited intestinal disorders characterized by intractable, sometimes life-threatening, diarrhea and nutrient malabsorption; some have been associated with mutations in diacylglycerol-acyltransferase 1 (DGAT1), which catalyzes formation of triacylglycerol from diacylglycerol and acyl-CoA. We investigated the mechanisms by which DGAT1 deficiency contributes to intestinal failure using patient-derived organoids. METHODS: We collected blood samples from 10 patients, from 6 unrelated pedigrees, who presented with early-onset severe diarrhea and/or vomiting, hypoalbuminemia, and/or (fatal) protein-losing enteropathy with intestinal failure; we performed next-generation sequencing analysis of DNA from 8 patients. Organoids were generated from duodenal biopsies from 3 patients and 3 healthy individuals (controls). Caco-2 cells and patient-derived dermal fibroblasts were transfected or transduced with vectors that express full-length or mutant forms of DGAT1 or full-length DGAT2. We performed CRISPR/Cas9-guided disruption of DGAT1 in control intestinal organoids. Cells and organoids were analyzed by immunoblot, immunofluorescence, flow cytometry, chromatography, quantitative real-time polymerase chain reaction, and for the activity of caspases 3 and 7. RESULTS: In the 10 patients, we identified 5 bi-allelic loss-of-function mutations in DGAT1. In patient-derived fibroblasts and organoids, the mutations reduced expression of DGAT1 protein and altered triacylglycerol metabolism, resulting in decreased lipid droplet formation after oleic acid addition. Expression of full-length DGAT2 in patient-derived fibroblasts restored formation of lipid droplets. Organoids derived from patients with DGAT1 mutations were more susceptible to lipid-induced cell death than control organoids. CONCLUSIONS: We identified a large cohort of patients with congenital diarrheal disorders with mutations in DGAT1 that reduced expression of its product; dermal fibroblasts and intestinal organoids derived from these patients had altered lipid metabolism and were susceptible to lipid-induced cell death. Expression of full-length wildtype DGAT1 or DGAT2 restored normal lipid metabolism in these cells. These findings indicate the importance of DGAT1 in fat metabolism and lipotoxicity in the intestinal epithelium. A fat-free diet might serve as the first line of therapy for patients with reduced DGAT1 expression. It is important to identify genetic variants associated with congenital diarrheal disorders for proper diagnosis and selection of treatment strategies.


Subject(s)
Diacylglycerol O-Acyltransferase/genetics , Duodenum/metabolism , Fibroblasts/metabolism , Hypoalbuminemia/genetics , Lipid Metabolism Disorders/genetics , Organoids/metabolism , Protein-Losing Enteropathies/genetics , Caco-2 Cells , Case-Control Studies , Caspase 3/metabolism , Caspase 7/metabolism , Child , Child, Preschool , Consanguinity , Dermis/cytology , Diacylglycerol O-Acyltransferase/deficiency , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Netherlands , Phorbols , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Turkey
6.
Genet Med ; 21(8): 1751-1760, 2019 08.
Article in English | MEDLINE | ID: mdl-30643219

ABSTRACT

PURPOSE: Using exome sequencing, the underlying variants in many persons with autosomal recessive diseases remain undetected. We explored autosomal recessive Stargardt disease (STGD1) as a model to identify the missing heritability. METHODS: Sequencing of ABCA4 was performed in 8 STGD1 cases with one variant and p.Asn1868Ile in trans, 25 cases with one variant, and 3 cases with no ABCA4 variant. The effect of intronic variants was analyzed using in vitro splice assays in HEK293T cells and patient-derived fibroblasts. Antisense oligonucleotides were used to correct splice defects. RESULTS: In 24 of the probands (67%), one known and five novel deep-intronic variants were found. The five novel variants resulted in messenger RNA pseudoexon inclusions, due to strengthening of cryptic splice sites or by disrupting a splicing silencer motif. Variant c.769-784C>T showed partial insertion of a pseudoexon and was found in cis with c.5603A>T (p.Asn1868Ile), so its causal role could not be fully established. Variant c.4253+43G>A resulted in partial skipping of exon 28. Remarkably, antisense oligonucleotides targeting the aberrant splice processes resulted in (partial) correction of all splicing defects. CONCLUSION: Our data demonstrate the importance of assessing noncoding variants in genetic diseases, and show the great potential of splice modulation therapy for deep-intronic variants.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Oligonucleotides, Antisense/genetics , Protein Isoforms/genetics , Stargardt Disease/genetics , Adolescent , Adult , Aged , Child , Exons/genetics , HEK293 Cells , Humans , Introns/genetics , Middle Aged , Mutation/genetics , Oligonucleotides, Antisense/pharmacology , Pedigree , Polymorphism, Single Nucleotide/genetics , RNA Splicing/genetics , Stargardt Disease/pathology , Young Adult
7.
Hum Mol Genet ; 25(3): 571-83, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26647307

ABSTRACT

Megacystis Microcolon Intestinal Hypoperistalsis Syndrome (MMIHS) is a rare congenital disorder, in which heterozygous missense variants in the Enteric Smooth Muscle actin γ-2 (ACTG2) gene have been recently identified. To investigate the mechanism by which ACTG2 variants lead to MMIHS, we screened a cohort of eleven MMIHS patients, eight sporadic and three familial cases, and performed immunohistochemistry, molecular modeling and molecular dynamics (MD) simulations, and in vitro assays. In all sporadic cases, a heterozygous missense variant in ACTG2 was identified. ACTG2 expression was detected in all intestinal layers where smooth muscle cells are present in different stages of human development. No histopathological abnormalities were found in the patients. Using molecular modeling and MD simulations, we predicted that ACTG2 variants lead to significant changes to the protein function. This was confirmed by in vitro studies, which showed that the identified variants not only impair ACTG2 polymerization, but also contribute to reduced cell contractility. Taken together, our results confirm the involvement of ACTG2 in sporadic MMIHS, and bring new insights to MMIHS pathogenesis.


Subject(s)
Abnormalities, Multiple/genetics , Actins/genetics , Colon/abnormalities , Intestinal Mucosa/metabolism , Intestinal Pseudo-Obstruction/genetics , Muscle Contraction/genetics , Muscle, Smooth/metabolism , Mutation, Missense , Urinary Bladder/abnormalities , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Actins/chemistry , Actins/metabolism , Colon/metabolism , Colon/pathology , Fatal Outcome , Female , Gene Expression , Heterozygote , Humans , Infant, Newborn , Intestinal Pseudo-Obstruction/metabolism , Intestinal Pseudo-Obstruction/pathology , Intestines/pathology , Male , Molecular Dynamics Simulation , Muscle, Smooth/pathology , Pedigree , Protein Multimerization , Urinary Bladder/metabolism , Urinary Bladder/pathology , Young Adult
8.
Hum Mol Genet ; 25(23): 5265-5275, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27702942

ABSTRACT

Hirschsprung disease (HSCR) is the most common cause of neonatal intestinal obstruction. It is characterized by the absence of ganglia in the nerve plexuses of the lower gastrointestinal tract. So far, three common disease-susceptibility variants at the RET, SEMA3 and NRG1 loci have been detected through genome-wide association studies (GWAS) in Europeans and Asians to understand its genetic etiologies. Here we present a trans-ethnic meta-analysis of 507 HSCR cases and 1191 controls, combining all published GWAS results on HSCR to fine-map these loci and narrow down the putatively causal variants to 99% credible sets. We also demonstrate that the effects of RET and NRG1 are universal across European and Asian ancestries. In contrast, we detected a European-specific association of a low-frequency variant, rs80227144, in SEMA3 [odds ratio (OR) = 5.2, P = 4.7 × 10-10]. Conditional analyses on the lead SNPs revealed a secondary association signal, corresponding to an Asian-specific, low-frequency missense variant encoding RET p.Asp489Asn (rs9282834, conditional OR = 20.3, conditional P = 4.1 × 10-14). When in trans with the RET intron 1 enhancer risk allele, rs9282834 increases the risk of HSCR from 1.1 to 26.7. Overall, our study provides further insights into the genetic architecture of HSCR and has profound implications for future study designs.


Subject(s)
Genetic Predisposition to Disease , Hirschsprung Disease/genetics , Neuregulin-1/genetics , Proto-Oncogene Proteins c-ret/genetics , Semaphorin-3A/genetics , Alleles , Asian People/genetics , Ethnicity/genetics , Female , Genome-Wide Association Study , Genotype , Hirschsprung Disease/pathology , Humans , Introns/genetics , Male , Polymorphism, Single Nucleotide , White People/genetics
9.
Am J Hum Genet ; 96(4): 581-96, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25839327

ABSTRACT

Innervation of the gut is segmentally lost in Hirschsprung disease (HSCR), a consequence of cell-autonomous and non-autonomous defects in enteric neuronal cell differentiation, proliferation, migration, or survival. Rare, high-penetrance coding variants and common, low-penetrance non-coding variants in 13 genes are known to underlie HSCR risk, with the most frequent variants in the ret proto-oncogene (RET). We used a genome-wide association (220 trios) and replication (429 trios) study to reveal a second non-coding variant distal to RET and a non-coding allele on chromosome 7 within the class 3 Semaphorin gene cluster. Analysis in Ret wild-type and Ret-null mice demonstrates specific expression of Sema3a, Sema3c, and Sema3d in the enteric nervous system (ENS). In zebrafish embryos, sema3 knockdowns show reduction of migratory ENS precursors with complete ablation under conjoint ret loss of function. Seven candidate receptors of Sema3 proteins are also expressed within the mouse ENS and their expression is also lost in the ENS of Ret-null embryos. Sequencing of SEMA3A, SEMA3C, and SEMA3D in 254 HSCR-affected subjects followed by in silico protein structure modeling and functional analyses identified five disease-associated alleles with loss-of-function defects in semaphorin dimerization and binding to their cognate neuropilin and plexin receptors. Thus, semaphorin 3C/3D signaling is an evolutionarily conserved regulator of ENS development whose dys-regulation is a cause of enteric aganglionosis.


Subject(s)
Epistasis, Genetic/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation , Hirschsprung Disease/genetics , Proto-Oncogene Proteins c-ret/genetics , Semaphorins/genetics , Animals , Base Sequence , Genome-Wide Association Study , Mice , Molecular Sequence Data , Semaphorins/deficiency , Semaphorins/metabolism , Sequence Analysis, DNA
10.
Am J Hum Genet ; 97(3): 475-82, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26299364

ABSTRACT

Adams-Oliver syndrome (AOS) is a rare developmental disorder characterized by the presence of aplasia cutis congenita (ACC) of the scalp vertex and terminal limb-reduction defects. Cardiovascular anomalies are also frequently observed. Mutations in five genes have been identified as a cause for AOS prior to this report. Mutations in EOGT and DOCK6 cause autosomal-recessive AOS, whereas mutations in ARHGAP31, RBPJ, and NOTCH1 lead to autosomal-dominant AOS. Because RBPJ, NOTCH1, and EOGT are involved in NOTCH signaling, we hypothesized that mutations in other genes involved in this pathway might also be implicated in AOS pathogenesis. Using a candidate-gene-based approach, we prioritized DLL4, a critical NOTCH ligand, due to its essential role in vascular development in the context of cardiovascular features in AOS-affected individuals. Targeted resequencing of the DLL4 gene with a custom enrichment panel in 89 independent families resulted in the identification of seven mutations. A defect in DLL4 was also detected in two families via whole-exome or genome sequencing. In total, nine heterozygous mutations in DLL4 were identified, including two nonsense and seven missense variants, the latter encompassing four mutations that replace or create cysteine residues, which are most likely critical for maintaining structural integrity of the protein. Affected individuals with DLL4 mutations present with variable clinical expression with no emerging genotype-phenotype correlations. Our findings demonstrate that DLL4 mutations are an additional cause of autosomal-dominant AOS or isolated ACC and provide further evidence for a key role of NOTCH signaling in the etiology of this disorder.


Subject(s)
Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/pathology , Intercellular Signaling Peptides and Proteins/genetics , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/pathology , Mutation/genetics , Scalp Dermatoses/congenital , Signal Transduction/genetics , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Base Sequence , Calcium-Binding Proteins , Heterozygote , Humans , Molecular Sequence Data , Pedigree , Receptors, Notch/genetics , Scalp Dermatoses/genetics , Scalp Dermatoses/pathology , Sequence Analysis, DNA
11.
Biochim Biophys Acta ; 1852(11): 2352-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26282049

ABSTRACT

Congenital Short Bowel Syndrome (CSBS) is a rare gastrointestinal disorder in which the mean length of the small intestine is substantially reduced when compared to its normal counterpart. Families with several affected members have been described and CSBS has been suggested to have a genetic basis. Recently, our group found mutations in CLMP as the cause of the recessive form of CSBS, and mutations in FLNA as the cause of the X-linked form of the disease. These findings have improved the quality of genetic counselling for CSBS patients and made prenatal diagnostics possible. Moreover, they provided a reliable starting point to further investigate the pathogenesis of CSBS, and to better understand the development of the small intestine. In this review, we present our current knowledge on CSBS and discuss hypotheses on how the recent genetic findings can help understand the cause of CSBS.

12.
Am J Hum Genet ; 87(1): 60-74, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20598273

ABSTRACT

The major gene for Hirschsprung disease (HSCR) encodes the receptor tyrosine kinase RET. In a study of 690 European- and 192 Chinese-descent probands and their parents or controls, we demonstrate the ubiquity of a >4-fold susceptibility from a C-->T allele (rs2435357: p = 3.9 x 10(-43) in European ancestry; p = 1.1 x 10(-21) in Chinese samples) that probably arose once within the intronic RET enhancer MCS+9.7. With in vitro assays, we now show that the T variant disrupts a SOX10 binding site within MCS+9.7 that compromises RET transactivation. The T allele, with a control frequency of 20%-30%/47% and case frequency of 54%-62%/88% in European/Chinese-ancestry individuals, is involved in all forms of HSCR. It is marginally associated with proband gender (p = 0.13) and significantly so with length of aganglionosis (p = 7.6 x 10(-5)) and familiality (p = 6.2 x 10(-4)). The enhancer variant is more frequent in the common forms of male, short-segment, and simplex families whereas multiple, rare, coding mutations are the norm in the less common and more severe forms of female, long-segment, and multiplex families. The T variant also increases penetrance in patients with rare RET coding mutations. Thus, both rare and common mutations, individually and together, make contributions to the risk of HSCR. The distribution of RET variants in diverse HSCR patients suggests a "cellular-recessive" genetic model where both RET alleles' function is compromised. The RET allelic series, and its genotype-phenotype correlations, shows that success in variant identification in complex disorders may strongly depend on which patients are studied.


Subject(s)
Hirschsprung Disease/genetics , Proto-Oncogene Proteins c-ret/genetics , Asian People , Base Sequence , Case-Control Studies , Enhancer Elements, Genetic , Female , Gene Frequency , Genome-Wide Association Study , Haplotypes , Hirschsprung Disease/ethnology , Hirschsprung Disease/physiopathology , Humans , Male , Mutation , Penetrance , Polymorphism, Single Nucleotide , Protein Binding , Proto-Oncogene Proteins c-ret/metabolism , SOXE Transcription Factors/metabolism , Sex Factors , Transcriptional Activation , White People
13.
Gastroenterology ; 142(3): 453-462.e3, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22155368

ABSTRACT

BACKGROUND & AIMS: Short-bowel syndrome usually results from surgical resection of the small intestine for diseases such as intestinal atresias, volvulus, and necrotizing enterocolitis. Patients with congenital short-bowel syndrome (CSBS) are born with a substantial shortening of the small intestine, to a mean length of 50 cm, compared with a normal length at birth of 190-280 cm. They also are born with intestinal malrotation. Because CSBS occurs in many consanguineous families, it is considered to be an autosomal-recessive disorder. We aimed to identify and characterize the genetic factor causing CSBS. METHODS: We performed homozygosity mapping using 610,000 K single-nucleotide polymorphism arrays to analyze the genomes of 5 patients with CSBS. After identifying a gene causing the disease, we determined its expression pattern in human embryos. We also overexpressed forms of the gene product that were and were not associated with CSBS in Chinese Hamster Ovary and T84 cells and generated a zebrafish model of the disease. RESULTS: We identified loss-of-function mutations in Coxsackie- and adenovirus receptor-like membrane protein (CLMP) in CSBS patients. CLMP is a tight-junction-associated protein that is expressed in the intestine of human embryos throughout development. Mutations in CLMP prevented its normal localization to the cell membrane. Knock-down experiments in zebrafish resulted in general developmental defects, including shortening of the intestine and the absence of goblet cells. Because goblet cells are characteristic for the midintestine in zebrafish, which resembles the small intestine in human beings, the zebrafish model mimics CSBS. CONCLUSIONS: Loss-of-function mutations in CLMP cause CSBS in human beings, likely by interfering with tight-junction formation, which disrupts intestinal development. Furthermore, we developed a zebrafish model of CSBS.


Subject(s)
Intestine, Small/abnormalities , Mutation, Missense , Receptors, Virus/genetics , Short Bowel Syndrome/genetics , Adolescent , Adult , Animals , CHO Cells , Child , Child, Preschool , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Cricetinae , Cricetulus , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Intestine, Small/metabolism , Male , Morphogenesis , Phenotype , Polymorphism, Single Nucleotide , Receptors, Virus/metabolism , Short Bowel Syndrome/embryology , Short Bowel Syndrome/metabolism , Short Bowel Syndrome/pathology , Transfection , Young Adult , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
Genet Med ; 15(4): 310-3, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23037936

ABSTRACT

PURPOSE: Autosomal recessive congenital short bowel syndrome is caused by mutations in CLMP. No mutations were found in the affected males of a family with presumed X-linked congenital short bowel syndrome or in an isolated male patient. Our aim was to identify the disease-causing mutation in these patients. METHODS: We performed mutation analysis of the second exon of FLNA in the two surviving affected males of the presumed X-linked family and in the isolated patient. RESULTS: We identified a novel 2-base-pair deletion in the second exon of FLNA in all these male patients. The deletion is located between two nearby methionines at the N-terminus of filamin A. Previous studies showed that translation of FLNA occurs from both methionines, resulting in two isoforms of the protein. We hypothesized that the longer isoform is no longer translated due to the mutation and that this mutation is therefore not lethal for males in utero. CONCLUSION: Our findings emphasize that congenital short bowel syndrome can be the presenting symptom in male patients with mutations in FLNA.


Subject(s)
Filamins/genetics , Mutation , Short Bowel Syndrome/diagnosis , Short Bowel Syndrome/genetics , Adolescent , Adult , Base Sequence , Exons , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/genetics , Humans , Male , Pedigree , Phenotype , Sequence Deletion , Young Adult
15.
Genet Med ; 15(3): 195-202, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22995989

ABSTRACT

PURPOSE: Radial ray deficiencies are characterized by unilateral or bilateral absence of varying portions of the radius and thumb. Both isolated and syndromic forms have been described, and although for some of the syndromes the causal gene has been identified, many patients remain without a genetic diagnosis. METHODS: In this study, a cohort of 54 patients with radial ray deficiencies was screened for genomic aberrations by molecular karyotyping. RESULTS: In 8 of 54 cases, an aberration was detected. Two unrelated patients inherited a 1q21.1 microduplication from a healthy parent, whereas in a third patient, a 16p13.11 microduplication was identified. Two other interesting microdeletions were detected: a 10q24.3 deletion at the split hand-foot malformation (SHFM3) locus and a 7p22.1 deletion including the RAC1 gene. CONCLUSION: The finding of these microduplications may just be coincidental or, alternatively, they may illustrate the broad phenotypic spectrum of these microduplications. Duplications in the 10q24.3 region result in split hand-foot malformations, and our observation indicates that deletions may cause radial ray defects. Finally, a candidate gene for radial ray deficiencies was detected in the 7p22.1 deletion. RAC1 plays an important role in the canonical Wnt pathway and conditional RAC1 knockout mice exhibit truncated-limb defects.


Subject(s)
Upper Extremity Deformities, Congenital/genetics , Chromosome Duplication , Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 7 , Comparative Genomic Hybridization , F-Box Proteins/genetics , Female , Humans , Male , Radiography , Upper Extremity Deformities, Congenital/diagnosis , Upper Extremity Deformities, Congenital/diagnostic imaging , rac1 GTP-Binding Protein/genetics
16.
HGG Adv ; 4(2): 100181, 2023 04 13.
Article in English | MEDLINE | ID: mdl-36785559

ABSTRACT

A significant number of individuals with a rare disorder such as Usher syndrome (USH) and (non-)syndromic autosomal recessive retinitis pigmentosa (arRP) remain genetically unexplained. Therefore, we assessed subjects suspected of USH2A-associated disease and no or mono-allelic USH2A variants using whole genome sequencing (WGS) followed by an improved pipeline for variant interpretation to provide a conclusive diagnosis. One hundred subjects were screened using WGS to identify causative variants in USH2A or other USH/arRP-associated genes. In addition to the existing variant interpretation pipeline, a particular focus was put on assessing splice-affecting properties of variants, both in silico and in vitro. Also structural variants were extensively addressed. For variants resulting in pseudoexon inclusion, we designed and evaluated antisense oligonucleotides (AONs) using minigene splice assays and patient-derived photoreceptor precursor cells. Biallelic variants were identified in 49 of 100 subjects, including novel splice-affecting variants and structural variants, in USH2A or arRP/USH-associated genes. Thirteen variants were shown to affect USH2A pre-mRNA splicing, including four deep-intronic USH2A variants resulting in pseudoexon inclusion, which could be corrected upon AON treatment. We have shown that WGS, combined with a thorough variant interpretation pipeline focused on assessing pre-mRNA splicing defects and structural variants, is a powerful method to provide subjects with a rare genetic condition, a (likely) conclusive genetic diagnosis. This is essential for the development of future personalized treatments and for patients to be eligible for such treatments.


Subject(s)
Retinitis Pigmentosa , Usher Syndromes , Humans , Usher Syndromes/diagnosis , RNA Precursors , Mutation , Pedigree , Retinitis Pigmentosa/diagnosis , Whole Genome Sequencing , Extracellular Matrix Proteins/genetics
17.
Invest Ophthalmol Vis Sci ; 60(13): 4249-4256, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31618761

ABSTRACT

Purpose: To investigate the role of two deep-intronic ABCA4 variants, that showed a mild splice defect in vitro and can occur on the same allele as the low penetrant c.5603A>T, in Stargardt disease (STGD1). Methods: Ophthalmic data were assessed of 18 STGD1 patients who harbored c.769-784C>T or c.4253+43G>A in combination with a severe ABCA4 variant. Subjects carrying c.[769-784C>T; 5603A>T] were clinically compared with a STGD1 cohort previously published carrying c.5603A>T noncomplex. We calculated the penetrances of the intronic variants using ABCA4 allele frequency data of the general population and investigated the effect of c.769-784C>T on splicing in photoreceptor progenitor cells (PPCs). Results: Mostly, late-onset, foveal-sparing STGD1 was observed among subjects harboring c.769-784C>T or c.4253+43G>A (median age of onset, 54.5 and 52.0 years, respectively). However, ages of onset, phenotypes in fundo, and visual acuity courses varied widely. No significant clinical differences were observed between the c.[769-784C>T; 5603A>T] cohort and the c.4253+43G>A or the c.5603A>T cohort. The penetrances of c.769-784C>T (20.5%-39.6%) and c.4253+43G>A (35.8%-43.1%) were reduced, when not considering the effect of yet unidentified or known factors in cis, such as c.5603A>T (identified in 7/7 probands with c.769-784C>T; 1/8 probands with c.4253+43G>A). Variant c.769-784C>T resulted in a pseudo-exon insertion in 15% of the total mRNA (i.e., ∼30% of the c.769-784C>T allele alone). Conclusions: Two mild intronic ABCA4 variants could further explain missing heritability in late-onset STGD1, distinguishing it from AMD. The observed clinical variability and calculated reduced penetrance urge research into modifiers within and outside of the ABCA4 gene.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Stargardt Disease/genetics , Aged , Alleles , Female , Gene Frequency , Genetic Variation , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Visual Acuity
18.
Eur J Hum Genet ; 16(6): 673-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18231121

ABSTRACT

Hypohidrotic ectodermal dysplasia (HED) can be caused by mutations in the X-linked ectodysplasin A (ED1) gene or the autosomal ectodysplasin A-receptor (EDAR) and EDAR-associated death domain (EDARADD) genes. X-linked and autosomal forms are sometimes clinically indistinguishable. For genetic counseling in families, it is therefore important to know the gene involved. In 24 of 42 unrelated patients with features of HED, we found a mutation in ED1. ED1-negative patients were screened for mutations in EDAR and EDARADD. We found mutations in EDAR in 5 of these 18 patients. One mutation, p.Glu354X, is novel. In EDARADD, a novel variant p.Ser93Phe, probably a neutral polymorphism, was also found. Clinically, there was a difference between autosomal dominant and autosomal recessive HED patients. The phenotype in patients with mutations in both EDAR alleles was comparable to males with X-linked HED. Patients with autosomal dominant HED had features comparable to those of female carriers of X-linked HED. The teeth of these patients were quite severely affected. Hypohidrosis and sparse hair were also evident, but less severe. This study confirms Chassaing et al's earlier finding that mutations in EDAR account for approximately 25% of non-ED1-related HED. Mutations leading to a premature stop codon have a recessive effect except when the stop codon is in the last exon. Heterozygous missense mutations in the functional domains of the gene may have a dominant-negative effect with much variation in expression. Patients with homozygous or compound heterozygous mutations in the EDAR gene have a more severe phenotype than those with a heterozygous missense, nonsense or frame-shift mutation.


Subject(s)
Ectodermal Dysplasia/genetics , Edar Receptor/genetics , Mutation , Child, Preschool , Female , Genotype , Humans , Infant , Male , Phenotype
19.
Am J Med Genet A ; 146A(1): 43-50, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18074359

ABSTRACT

CHARGE syndrome is an autosomal dominant condition that is caused by mutations in the CHD7 gene. Few familial cases of this syndrome have been reported and these were characterized by a wide clinical variability. We here report on five CHD7 mutation positive families and comment on their clinical features. We observed somatic and germline mosaicism as well as parent-to-child transmission of non-mosaic CHD7 mutations as causes of familial CHARGE syndrome. In one family with two affected sibs a somatic mutation was identified in lymphocytes of a clinically unaffected parent (2520G > A in exon 8). This is the second report of somatic CHD7 mosaicism in an unaffected parent. In two further families with affected siblings, we could not detect the mutation in parental lymphocytes suggesting germline mosaicism. The previously reported clinical variability was strikingly present in all five families. We find that alterations in CHD7 can result in a very mild phenotype, characterized by only a few minor symptoms of the CHARGE syndrome clinical spectrum. Such a mild phenotype was present in two families that shared the same 6322G > A missense mutation. These two families showed parent-to-child transmission. Phenotypically milder forms of CHARGE syndrome have a higher risk of transmission to multiple family members.


Subject(s)
Abnormalities, Multiple/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Genetic Variation , Mutation, Missense , Pedigree , Amino Acid Sequence , Amino Acid Substitution , Arginine/metabolism , Case-Control Studies , Conserved Sequence , Diseases in Twins , Female , Genes, Dominant , Humans , Male , Molecular Sequence Data , Mosaicism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Siblings , Syndrome , Twins, Monozygotic
20.
Invest Ophthalmol Vis Sci ; 59(8): 3220-3231, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29971439

ABSTRACT

Purpose: To assess the occurrence and the disease expression of the common p.Asn1868Ile variant in patients with Stargardt disease (STGD1) harboring known, monoallelic causal ABCA4 variants. Methods: The coding and noncoding regions of ABCA4 were sequenced in 67 and 63 STGD1 probands respectively, harboring monoallelic ABCA4 variants. In case p.Asn1868Ile was detected, segregation analysis was performed whenever possible. Probands and affected siblings harboring p.Asn1868Ile without additional variants in cis were clinically evaluated retrospectively. Two asymptomatic siblings carrying the same ABCA4 variants as their probands were clinically examined. The penetrance of p.Asn1868Ile was calculated using allele frequency data of ABCA4 variants in non-Finnish European individuals. Results: The p.Asn1868Ile variant was found in cis with known variants in 14/67 probands. In 27/67 probands, we identified p.Asn1868Ile without additional variants in cis, in combination with known, mainly severe ABCA4 variants. In 23/27 probands, the trans configuration was established. Among 27 probands and 6/7 STGD1 siblings carrying p.Asn1868Ile, 42% manifested late-onset disease (>44 years). We additionally identified four asymptomatic relatives carrying a combination of a severe variant and p.Asn1868Ile; ophthalmologic examination in two persons did not reveal STGD1. Based on ABCA4 allele frequency data, we conservatively estimated the penetrance of p.Asn1868Ile, when present in trans with a severe variant, to be below 5%. Conclusions: A significant fraction of genetically unexplained STGD1 cases carries p.Asn1868Ile as a second variant. Our findings suggest exceptional differences in disease expression or even nonpenetrance of this ABCA4 variant, pointing toward an important role for genetic or environmental modifiers in STGD1.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Macular Degeneration/congenital , Mutation , Polymorphism, Single Nucleotide , Adult , Age of Onset , Aged , Electroretinography , Female , Fluorescein Angiography , Gene Frequency , Genetic Complementation Test , Humans , Macular Degeneration/diagnosis , Macular Degeneration/genetics , Macular Degeneration/physiopathology , Male , Middle Aged , Pedigree , Penetrance , Retrospective Studies , Sequence Analysis, DNA , Siblings , Stargardt Disease , Tomography, Optical Coherence , Visual Acuity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL