Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Am J Hum Genet ; 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39013458

ABSTRACT

The shift to a genotype-first approach in genetic diagnostics has revolutionized our understanding of neurodevelopmental disorders, expanding both their molecular and phenotypic spectra. Kleefstra syndrome (KLEFS1) is caused by EHMT1 haploinsufficiency and exhibits broad clinical manifestations. EHMT1 encodes euchromatic histone methyltransferase-1-a pivotal component of the epigenetic machinery. We have recruited 209 individuals with a rare EHMT1 variant and performed comprehensive molecular in silico and in vitro testing alongside DNA methylation (DNAm) signature analysis for the identified variants. We (re)classified the variants as likely pathogenic/pathogenic (molecularly confirming Kleefstra syndrome) in 191 individuals. We provide an updated and broader clinical and molecular spectrum of Kleefstra syndrome, including individuals with normal intelligence and familial occurrence. Analysis of the EHMT1 variants reveals a broad range of molecular effects and their associated phenotypes, including distinct genotype-phenotype associations. Notably, we showed that disruption of the "reader" function of the ankyrin repeat domain by a protein altering variant (PAV) results in a KLEFS1-specific DNAm signature and milder phenotype, while disruption of only "writer" methyltransferase activity of the SET domain does not result in KLEFS1 DNAm signature or typical KLEFS1 phenotype. Similarly, N-terminal truncating variants result in a mild phenotype without the DNAm signature. We demonstrate how comprehensive variant analysis can provide insights into pathogenesis of the disorder and DNAm signature. In summary, this study presents a comprehensive overview of KLEFS1 and EHMT1, revealing its broader spectrum and deepening our understanding of its molecular mechanisms, thereby informing accurate variant interpretation, counseling, and clinical management.

2.
HGG Adv ; 3(1): 100074, 2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35047859

ABSTRACT

Robinow syndrome (RS) is a genetically heterogeneous disorder with six genes that converge on the WNT/planar cell polarity (PCP) signaling pathway implicated (DVL1, DVL3, FZD2, NXN, ROR2, and WNT5A). RS is characterized by skeletal dysplasia and distinctive facial and physical characteristics. To further explore the genetic heterogeneity, paralog contribution, and phenotypic variability of RS, we investigated a cohort of 22 individuals clinically diagnosed with RS from 18 unrelated families. Pathogenic or likely pathogenic variants in genes associated with RS or RS phenocopies were identified in all 22 individuals, including the first variant to be reported in DVL2. We retrospectively collected medical records of 16 individuals from this cohort and extracted clinical descriptions from 52 previously published cases. We performed Human Phenotype Ontology (HPO) based quantitative phenotypic analyses to dissect allele-specific phenotypic differences. Individuals with FZD2 variants clustered into two groups with demonstrable phenotypic differences between those with missense and truncating alleles. Probands with biallelic NXN variants clustered together with the majority of probands carrying DVL1, DVL2, and DVL3 variants, demonstrating no phenotypic distinction between the NXN-autosomal recessive and dominant forms of RS. While phenotypically similar diseases on the RS differential matched through HPO analysis, clustering using phenotype similarity score placed RS-associated phenotypes in a unique cluster containing WNT5A, FZD2, and ROR2 apart from non-RS-associated paralogs. Through human phenotype analyses of this RS cohort and OMIM clinical synopses of Mendelian disease, this study begins to tease apart specific biologic roles for non-canonical WNT-pathway proteins.

4.
Am J Hum Genet ; 108(2): 346-356, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33513338

ABSTRACT

Whereas large-scale statistical analyses can robustly identify disease-gene relationships, they do not accurately capture genotype-phenotype correlations or disease mechanisms. We use multiple lines of independent evidence to show that different variant types in a single gene, SATB1, cause clinically overlapping but distinct neurodevelopmental disorders. Clinical evaluation of 42 individuals carrying SATB1 variants identified overt genotype-phenotype relationships, associated with different pathophysiological mechanisms, established by functional assays. Missense variants in the CUT1 and CUT2 DNA-binding domains result in stronger chromatin binding, increased transcriptional repression, and a severe phenotype. In contrast, variants predicted to result in haploinsufficiency are associated with a milder clinical presentation. A similarly mild phenotype is observed for individuals with premature protein truncating variants that escape nonsense-mediated decay, which are transcriptionally active but mislocalized in the cell. Our results suggest that in-depth mutation-specific genotype-phenotype studies are essential to capture full disease complexity and to explain phenotypic variability.


Subject(s)
Matrix Attachment Region Binding Proteins/genetics , Mutation , Neurodevelopmental Disorders/genetics , Chromatin/metabolism , Female , Genetic Association Studies , Haploinsufficiency , Humans , Male , Matrix Attachment Region Binding Proteins/chemistry , Matrix Attachment Region Binding Proteins/metabolism , Models, Molecular , Mutation, Missense , Protein Binding , Protein Domains , Transcription, Genetic
5.
Front Mol Neurosci ; 13: 12, 2020.
Article in English | MEDLINE | ID: mdl-32116545

ABSTRACT

Multiple TREX mRNA export complex subunits (e.g., THOC1, THOC2, THOC5, THOC6, THOC7) have now been implicated in neurodevelopmental disorders (NDDs), neurodegeneration and cancer. We previously implicated missense and splicing-defective THOC2 variants in NDDs and a broad range of other clinical features. Here we report 10 individuals from nine families with rare missense THOC2 variants including the first case of a recurrent variant (p.Arg77Cys), and an additional individual with an intragenic THOC2 microdeletion (Del-Ex37-38). Ex vivo missense variant testing and patient-derived cell line data from current and published studies show 9 of the 14 missense THOC2 variants result in reduced protein stability. The splicing-defective and deletion variants result in a loss of small regions of the C-terminal THOC2 RNA binding domain (RBD). Interestingly, reduced stability of THOC2 variant proteins has a flow-on effect on the stability of the multi-protein TREX complex; specifically on the other NDD-associated THOC subunits. Our current, expanded cohort refines the core phenotype of THOC2 NDDs to language disorder and/or ID, with a variable severity, and disorders of growth. A subset of affected individuals' has severe-profound ID, persistent hypotonia and respiratory abnormalities. Further investigations to elucidate the pathophysiological basis for this severe phenotype are warranted.

6.
Am J Hum Genet ; 103(5): 752-768, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388402

ABSTRACT

The nuclear factor I (NFI) family of transcription factors play an important role in normal development of multiple organs. Three NFI family members are highly expressed in the brain, and deletions or sequence variants in two of these, NFIA and NFIX, have been associated with intellectual disability (ID) and brain malformations. NFIB, however, has not previously been implicated in human disease. Here, we present a cohort of 18 individuals with mild ID and behavioral issues who are haploinsufficient for NFIB. Ten individuals harbored overlapping microdeletions of the chromosomal 9p23-p22.2 region, ranging in size from 225 kb to 4.3 Mb. Five additional subjects had point sequence variations creating a premature termination codon, and three subjects harbored single-nucleotide variations resulting in an inactive protein as determined using an in vitro reporter assay. All individuals presented with additional variable neurodevelopmental phenotypes, including muscular hypotonia, motor and speech delay, attention deficit disorder, autism spectrum disorder, and behavioral abnormalities. While structural brain anomalies, including dysgenesis of corpus callosum, were variable, individuals most frequently presented with macrocephaly. To determine whether macrocephaly could be a functional consequence of NFIB disruption, we analyzed a cortex-specific Nfib conditional knockout mouse model, which is postnatally viable. Utilizing magnetic resonance imaging and histology, we demonstrate that Nfib conditional knockout mice have enlargement of the cerebral cortex but preservation of overall brain structure and interhemispheric connectivity. Based on our findings, we propose that haploinsufficiency of NFIB causes ID with macrocephaly.


Subject(s)
Haploinsufficiency/genetics , Intellectual Disability/genetics , Megalencephaly/genetics , NFI Transcription Factors/genetics , Adolescent , Adult , Animals , Cerebral Cortex/pathology , Child , Child, Preschool , Codon, Nonsense/genetics , Cohort Studies , Corpus Callosum/pathology , Female , Humans , Male , Mice , Mice, Knockout , Polymorphism, Single Nucleotide/genetics , Young Adult
9.
Nat Genet ; 48(7): 777-84, 2016 07.
Article in English | MEDLINE | ID: mdl-27213289

ABSTRACT

We identified biallelic mutations in NANS, the gene encoding the synthase for N-acetylneuraminic acid (NeuNAc; sialic acid), in nine individuals with infantile-onset severe developmental delay and skeletal dysplasia. Patient body fluids showed an elevation in N-acetyl-D-mannosamine levels, and patient-derived fibroblasts had reduced NANS activity and were unable to incorporate sialic acid precursors into sialylated glycoproteins. Knockdown of nansa in zebrafish embryos resulted in abnormal skeletal development, and exogenously added sialic acid partially rescued the skeletal phenotype. Thus, NANS-mediated synthesis of sialic acid is required for early brain development and skeletal growth. Normal sialylation of plasma proteins was observed in spite of NANS deficiency. Exploration of endogenous synthesis, nutritional absorption, and rescue pathways for sialic acid in different tissues and developmental phases is warranted to design therapeutic strategies to counteract NANS deficiency and to shed light on sialic acid metabolism and its implications for human nutrition.


Subject(s)
Bone Diseases, Developmental/pathology , Brain/embryology , Developmental Disabilities/pathology , Mutation/genetics , Oxo-Acid-Lyases/genetics , Sialic Acids/metabolism , Zebrafish/embryology , Adult , Age of Onset , Animals , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/metabolism , Brain/metabolism , Brain/pathology , Child, Preschool , Developmental Disabilities/genetics , Developmental Disabilities/metabolism , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Infant , Infant, Newborn , Male , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/pathology , Zebrafish/genetics , Zebrafish/metabolism
10.
J Clin Invest ; 125(9): 3585-99, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26280580

ABSTRACT

The genetic disorder Kabuki syndrome (KS) is characterized by developmental delay and congenital anomalies. Dominant mutations in the chromatin regulators lysine (K)-specific methyltransferase 2D (KMT2D) (also known as MLL2) and lysine (K)-specific demethylase 6A (KDM6A) underlie the majority of cases. Although the functions of these chromatin-modifying proteins have been studied extensively, the physiological systems regulated by them are largely unknown. Using whole-exome sequencing, we identified a mutation in RAP1A that was converted to homozygosity as the result of uniparental isodisomy (UPD) in a patient with KS and a de novo, dominant mutation in RAP1B in a second individual with a KS-like phenotype. We elucidated a genetic and functional interaction between the respective KS-associated genes and their products in zebrafish models and patient cell lines. Specifically, we determined that dysfunction of known KS genes and the genes identified in this study results in aberrant MEK/ERK signaling as well as disruption of F-actin polymerization and cell intercalation. Moreover, these phenotypes could be rescued in zebrafish models by rebalancing MEK/ERK signaling via administration of small molecule inhibitors of MEK. Taken together, our studies suggest that the KS pathophysiology overlaps with the RASopathies and provide a potential direction for treatment design.


Subject(s)
Abnormalities, Multiple/genetics , Exome , Face/abnormalities , Hematologic Diseases/genetics , MAP Kinase Signaling System/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation , Telomere-Binding Proteins/genetics , Vestibular Diseases/genetics , Zebrafish Proteins/genetics , Abnormalities, Multiple/metabolism , Actins/genetics , Actins/metabolism , Animals , Cattle , Child , Child, Preschool , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Hematologic Diseases/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Male , Mice , Monomeric GTP-Binding Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Rats , Shelterin Complex , Telomere-Binding Proteins/metabolism , Vestibular Diseases/metabolism , Zebrafish , Zebrafish Proteins/metabolism
11.
Eur J Med Genet ; 58(9): 455-65, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26206081

ABSTRACT

INTRODUCTION: Oculo-auriculo-vertebral spectrum (OAVS OMIM 164210) is a craniofacial developmental disorder affecting the development of the structures derived from the 1st and the 2nd branchial arches during embryogenesis, with consequential maxillary, mandibular, and ear abnormalities. The phenotype in OAVS is variable and associated clinical features can involve the cardiac, renal, skeletal, and central nervous systems. Its aetiology is still poorly understood. METHODS: We have evaluated the clinical phenotypes of 51 previously unpublished patients with OAVS and their parents, and performed comparative genomic hybridization microarray studies to identify potential causative loci. RESULTS: Of all 51 patients, 16 (31%) had a family history of OAVS. Most had no relevant pre-natal history and only 5 (10%) cases had a history of environmental exposures that have previously been described as risk factors for OAVS. In 28 (55%) cases, the malformations were unilateral. When the involvement was bilateral, it was asymmetric. Ear abnormalities were present in 47 (92%) patients (unilateral in 24; and bilateral in 23). Hearing loss was common (85%), mostly conductive, but also sensorineural, or a combination of both. Hemifacial microsomia was present in 46 (90%) patients (17 also presented facial nerve palsy). Ocular anomalies were present in 15 (29%) patients. Vertebral anomalies were confirmed in 10 (20%) cases; 50% of those had additional heart, brain and/or other organ abnormalities. Brain abnormalities were present in 5 (10%) patients; developmental delay was more common among these patients. Limb abnormalities were found in 6 (12%) patients, and urogenital anomalies in 5 (10%). Array-CGH analysis identified 22q11 dosage anomalies in 10 out of 22 index cases screened. DISCUSSION: In this study we carried out in-depth phenotyping of OAVS in a large, multicentre cohort. Clinical characteristics are in line with those reported previously, however, we observed a higher incidence of hemifacial microsomia and lower incidence of ocular anomalies. Furthermore our data suggests that OAVS patients with vertebral anomalies or congenital heart defects have a higher frequency of additional brain, limb or other malformations. We had a higher rate of familial cases in our cohort in comparison with previous reports, possibly because these cases were referred preferentially to our genetic clinic where family members underwent examination. We propose that familial OAVS cases show phenotypic variability, hence, affected relatives might have been misclassified in previous reports. Moreover, in view of its phenotypic variability, OAVS is potentially a spectrum of conditions, which overlap with other conditions, such as mandibulofacial dysostosis. Array CGH in our cohort identified recurrent dosage anomalies on 22q11, which may contribute to, or increase the risk of OAVS. We hypothesize that although the 22q11 locus may harbour gene(s) or regulatory elements that play a role in the regulation of craniofacial symmetry and 1st and 2nd branchial arch development, OAVS is a heterogeneous condition and many cases have a multifactorial aetiology or are caused by mutations in as yet unidentified gene(s).


Subject(s)
Developmental Disabilities/genetics , Goldenhar Syndrome/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Brain/abnormalities , Comparative Genomic Hybridization , Ear/abnormalities , Ear/embryology , Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Female , Goldenhar Syndrome/diagnosis , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/genetics , Hernia, Diaphragmatic/diagnosis , Hernia, Diaphragmatic/genetics , Humans , Male , Spine/abnormalities
12.
Ann Rheum Dis ; 74(6): 1249-56, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24442880

ABSTRACT

OBJECTIVES: Leri's pleonosteosis (LP) is an autosomal dominant rheumatic condition characterised by flexion contractures of the interphalangeal joints, limited motion of multiple joints, and short broad metacarpals, metatarsals and phalanges. Scleroderma-like skin thickening can be seen in some individuals with LP. We undertook a study to characterise the phenotype of LP and identify its genetic basis. METHODS AND RESULTS: Whole-genome single-nucleotide polymorphism genotyping in two families with LP defined microduplications of chromosome 8q22.1 as the cause of this condition. Expression analysis of dermal fibroblasts from affected individuals showed overexpression of two genes, GDF6 and SDC2, within the duplicated region, leading to dysregulation of genes that encode proteins of the extracellular matrix and downstream players in the transforming growth factor (TGF)-ß pathway. Western blot analysis revealed markedly decreased inhibitory SMAD6 levels in patients with LP. Furthermore, in a cohort of 330 systemic sclerosis cases, we show that the minor allele of a missense SDC2 variant, p.Ser71Thr, could confer protection against disease (p<1×10(-5)). CONCLUSIONS: Our work identifies the genetic cause of LP in these two families, demonstrates the phenotypic range of the condition, implicates dysregulation of extracellular matrix homoeostasis genes in its pathogenesis, and highlights the link between TGF-ß/SMAD signalling, growth/differentiation factor 6 and syndecan-2. We propose that LP is an additional member of the growing 'TGF-ß-pathies' group of musculoskeletal disorders, which includes Myhre syndrome, acromicric dysplasia, geleophysic dysplasias, Weill-Marchesani syndromes and stiff skin syndrome. Identification of a systemic sclerosis-protective SDC2 variant lays the foundation for exploration of the role of syndecan-2 in systemic sclerosis in the future.


Subject(s)
Chromosomes, Human, Pair 8/genetics , Gene Duplication , Growth Differentiation Factor 6/genetics , Hand Deformities, Congenital/genetics , Joint Diseases/congenital , Ossification, Heterotopic/genetics , Scleroderma, Systemic/genetics , Syndecan-2/genetics , Adult , Aged , Child, Preschool , Extracellular Matrix/metabolism , Facies , Female , Fibroblasts/metabolism , Gene Expression Profiling , Growth Differentiation Factor 6/metabolism , Hand Deformities, Congenital/metabolism , Hand Deformities, Congenital/physiopathology , Humans , Infant , Joint Diseases/genetics , Joint Diseases/metabolism , Joint Diseases/physiopathology , Male , Middle Aged , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/physiopathology , Phenotype , Signal Transduction , Syndecan-2/metabolism , Transforming Growth Factor beta/metabolism , Young Adult
13.
Am J Hum Genet ; 95(5): 622-32, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25439729

ABSTRACT

Filippi syndrome is a rare, presumably autosomal-recessive disorder characterized by microcephaly, pre- and postnatal growth failure, syndactyly, and distinctive facial features, including a broad nasal bridge and underdeveloped alae nasi. Some affected individuals have intellectual disability, seizures, undescended testicles in males, and teeth and hair abnormalities. We performed homozygosity mapping and whole-exome sequencing in a Sardinian family with two affected children and identified a homozygous frameshift mutation, c.571dupA (p.Ile191Asnfs(∗)6), in CKAP2L, encoding the protein cytoskeleton-associated protein 2-like (CKAP2L). The function of this protein was unknown until it was rediscovered in mice as Radmis (radial fiber and mitotic spindle) and shown to play a pivotal role in cell division of neural progenitors. Sanger sequencing of CKAP2L in a further eight unrelated individuals with clinical features consistent with Filippi syndrome revealed biallelic mutations in four subjects. In contrast to wild-type lymphoblastoid cell lines (LCLs), dividing LCLs established from the individuals homozygous for the c.571dupA mutation did not show CKAP2L at the spindle poles. Furthermore, in cells from the affected individuals, we observed an increase in the number of disorganized spindle microtubules owing to multipolar configurations and defects in chromosome segregation. The observed cellular phenotypes are in keeping with data from in vitro and in vivo knockdown studies performed in human cells and mice, respectively. Our findings show that loss-of-function mutations in CKAP2L are a major cause of Filippi syndrome.


Subject(s)
Cytoskeletal Proteins/genetics , Growth Disorders/genetics , Intellectual Disability/genetics , Microcephaly/genetics , Syndactyly/genetics , Animals , Base Sequence , Cytogenetic Analysis , Facies , Frameshift Mutation/genetics , Gene Components , Genes, Recessive/genetics , Growth Disorders/pathology , Humans , Intellectual Disability/pathology , Italy , Male , Mice , Microcephaly/pathology , Microscopy, Confocal , Molecular Sequence Data , Sequence Analysis, DNA , Syndactyly/pathology
14.
Eur J Hum Genet ; 22(11): 1272-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24424121

ABSTRACT

Myhre syndrome is characterized by short stature, brachydactyly, facial features, pseudomuscular hypertrophy, joint limitation and hearing loss. We identified SMAD4 mutations as the cause of Myhre syndrome. SMAD4 mutations have also been identified in laryngotracheal stenosis, arthropathy, prognathism and short stature syndrome (LAPS). This study aimed to review the features of Myhre and LAPS patients to define the clinical spectrum of SMAD4 mutations. We included 17 females and 15 males ranging in age from 8 to 48 years. Thirty were diagnosed with Myhre syndrome and two with LAPS. SMAD4 coding sequence was analyzed by Sanger sequencing. Clinical and radiological features were collected from a questionnaire completed by the referring physicians. All patients displayed a typical facial gestalt, thickened skin, joint limitation and muscular pseudohypertrophy. Growth retardation was common (68.7%) and was variable in severity (from -5.5 to -2 SD), as was mild-to-moderate intellectual deficiency (87.5%) with additional behavioral problems in 56.2% of the patients. Significant health concerns like obesity, arterial hypertension, bronchopulmonary insufficiency, laryngotracheal stenosis, pericarditis and early death occurred in four. Twenty-nine patients had a de novo heterozygous SMAD4 mutation, including both patients with LAPS. In 27 cases mutation affected Ile500 and in two cases Arg496. The three patients without SMAD4 mutations had typical findings of Myhre syndrome. Myhre-LAPS syndrome is a clinically homogenous condition with life threatening complications in the course of the disease. Our identification of SMAD4 mutations in 29/32 cases confirms that SMAD4 is the major gene responsible for Myhre syndrome.


Subject(s)
Cryptorchidism/genetics , Growth Disorders/genetics , Hand Deformities, Congenital/genetics , Hypertrophy/genetics , Intellectual Disability/genetics , Joint Diseases/genetics , Smad4 Protein/genetics , Adolescent , Adult , Child , Facies , Female , Follow-Up Studies , Heterozygote , Humans , Male , Middle Aged , Mutation , Sequence Analysis, DNA , Young Adult
15.
Hum Mutat ; 34(2): 296-300, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23086778

ABSTRACT

Kohlschütter-Tönz syndrome (KTS) is a rare autosomal recessive disorder characterized by amelogenesis imperfecta, psychomotor delay or regression and seizures starting early in childhood. KTS was established as a distinct clinical entity after the first report by Kohlschütter in 1974, and to date, only a total of 20 pedigrees have been reported. The genetic etiology of KTS remained elusive until recently when mutations in ROGDI were independently identified in three unrelated families and in five likely related Druze families. Herein, we report a clinical and genetic study of 10 KTS families. By using a combination of whole exome sequencing, linkage analysis, and Sanger sequencing, we identify novel homozygous or compound heterozygous ROGDI mutations in five families, all presenting with a typical KTS phenotype. The other families, mostly presenting with additional atypical features, were negative for ROGDI mutations, suggesting genetic heterogeneity of atypical forms of the disease.


Subject(s)
Amelogenesis Imperfecta/genetics , Dementia/genetics , Epilepsy/genetics , Genetic Heterogeneity , Membrane Proteins/genetics , Nuclear Proteins/genetics , Child, Preschool , Exome , Female , Gene Deletion , Genetic Linkage , Humans , Infant , Male , Mutation , Pedigree , Phenotype , Sequence Analysis, DNA
16.
J Community Genet ; 3(3): 167-74, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22760671

ABSTRACT

We review the reasons why a couple might seek specialist genetic counselling about a possible reproductive risk and the options available to them. Most commonly, the couple will be concerned about the risk of recurrence of a medical condition that has already occurred in the family. Sometimes, the increased risk may come from their ethnicity or because of a consanguineous marriage, rather than because any problem has occurred previously. The geneticist must identify the exact nature of any problem and determine the risks in the light of the mode of inheritance, any investigations undertaken and any other relevant information. The geneticist will then review the options open to the couple, and help them arrive at their own decision in a non-directive way. Some couples may opt to do nothing and let nature take its course but others may request prenatal or pre-implantation diagnosis, or they may avoid the conception of an at-risk child by using donor gametes, adoption or even decide not to have children.

17.
Am J Hum Genet ; 89(5): 675-81, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-22077973

ABSTRACT

Say-Barber-Biesecker-Young-Simpson syndrome (SBBYSS or Ohdo syndrome) is a multiple anomaly syndrome characterized by severe intellectual disability, blepharophimosis, and a mask-like facial appearance. A number of individuals with SBBYSS also have thyroid abnormalities and cleft palate. The condition usually occurs sporadically and is therefore presumed to be due in most cases to new dominant mutations. In individuals with SBBYSS, a whole-exome sequencing approach was used to demonstrate de novo protein-truncating mutations in the highly conserved histone acetyltransferase gene KAT6B (MYST4/MORF)) in three out of four individuals sequenced. Sanger sequencing was used to confirm truncating mutations of KAT6B, clustering in the final exon of the gene in all four individuals and in a further nine persons with typical SBBYSS. Where parental samples were available, the mutations were shown to have occurred de novo. During mammalian development KAT6B is upregulated specifically in the developing central nervous system, facial structures, and limb buds. The phenotypic features seen in the Qkf mouse, a hypomorphic Kat6b mutant, include small eyes, ventrally placed ears and long first digits that mirror the human phenotype. This is a further example of how perturbation of a protein involved in chromatin modification might give rise to a multisystem developmental disorder.


Subject(s)
Codon, Nonsense/genetics , Congenital Hypothyroidism/genetics , Exome/genetics , Histone Acetyltransferases , Intellectual Disability/genetics , Abnormalities, Multiple/genetics , Adult , Animals , Blepharophimosis/genetics , Child , Chromatin/metabolism , Chromosomes, Human, Pair 10/genetics , Facies , Female , Gene Expression Regulation, Developmental , Heart Defects, Congenital , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Humans , INDEL Mutation/genetics , Joint Instability , Male , Metabolism, Inborn Errors/genetics , Mice , Mice, Transgenic , Microarray Analysis , Polymorphism, Single Nucleotide/genetics
18.
Am J Med Genet A ; 155A(9): 2203-11, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21834056

ABSTRACT

Pierpont syndrome is a multiple congenital anomaly syndrome with learning disability first described in 1998. There are only three patients with Pierpont syndrome who have previously been published in the literature. Details of a series of patients with features of this condition were therefore obtained retrospectively to better characterize its key features. These patients were noted to have distinctive shared facial characteristics, in addition to plantar fat pads and other limb abnormalities. Further individuals with equally striking hand and foot findings were identified whose facies were less characteristic, and hence we considered them unlikely to be affected with the same condition. Despite several patients with possible Pierpont syndrome having had high-resolution array CGH or SNP array, the etiology of this phenotype remains unknown. Whilst it is as yet unclear whether it is a single entity, there appears to be a group of patients in whom Pierpont syndrome may be a recognizable condition, with typical facies, particularly when smiling, and characteristic hand and foot findings.


Subject(s)
Abnormalities, Multiple/genetics , Learning Disabilities/genetics , Adult , Child , Child, Preschool , Craniofacial Abnormalities/genetics , Developmental Disabilities , Face/abnormalities , Facies , Female , Foot Deformities, Congenital/genetics , Hand Deformities, Congenital/genetics , Humans , Karyotype , Male , Middle Aged , Phenotype , Retrospective Studies , Syndrome
19.
Am J Hum Genet ; 89(1): 148-53, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21737058

ABSTRACT

3-M syndrome, a primordial growth disorder, is associated with mutations in CUL7 and OBSL1. Exome sequencing now identifies mutations in CCDC8 as a cause of 3-M syndrome. CCDC8 is a widely expressed gene that is transcriptionally associated to CUL7 and OBSL1, and coimmunoprecipitation indicates a physical interaction between CCDC8 and OBSL1 but not CUL7. We propose that CUL7, OBSL1, and CCDC8 are members of a pathway controlling mammalian growth.


Subject(s)
Cullin Proteins/genetics , Cytoskeletal Proteins/genetics , Dwarfism/genetics , Intellectual Disability/genetics , Muscle Hypotonia/genetics , Cell Line , Child, Preschool , Cullin Proteins/metabolism , Cytoskeletal Proteins/metabolism , Female , Gene Expression , Homozygote , Humans , Infant , Male , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Spine/abnormalities , Transcription Factors
20.
Am J Med Genet A ; 155A(7): 1511-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21671394

ABSTRACT

Kabuki syndrome is a rare, multiple malformation disorder characterized by a distinctive facial appearance, cardiac anomalies, skeletal abnormalities, and mild to moderate intellectual disability. Simplex cases make up the vast majority of the reported cases with Kabuki syndrome, but parent-to-child transmission in more than a half-dozen instances indicates that it is an autosomal dominant disorder. We recently reported that Kabuki syndrome is caused by mutations in MLL2, a gene that encodes a Trithorax-group histone methyltransferase, a protein important in the epigenetic control of active chromatin states. Here, we report on the screening of 110 families with Kabuki syndrome. MLL2 mutations were found in 81/110 (74%) of families. In simplex cases for which DNA was available from both parents, 25 mutations were confirmed to be de novo, while a transmitted MLL2 mutation was found in two of three familial cases. The majority of variants found to cause Kabuki syndrome were novel nonsense or frameshift mutations that are predicted to result in haploinsufficiency. The clinical characteristics of MLL2 mutation-positive cases did not differ significantly from MLL2 mutation-negative cases with the exception that renal anomalies were more common in MLL2 mutation-positive cases. These results are important for understanding the phenotypic consequences of MLL2 mutations for individuals and their families as well as for providing a basis for the identification of additional genes for Kabuki syndrome.


Subject(s)
Abnormalities, Multiple/genetics , DNA-Binding Proteins/genetics , Hematologic Diseases/genetics , Mutation/genetics , Neoplasm Proteins/genetics , Vestibular Diseases/genetics , Abnormalities, Multiple/diagnosis , Alleles , Face/abnormalities , Gene Order , Genetic Testing , Genotype , Hematologic Diseases/diagnosis , Humans , Phenotype , Prognosis , Vestibular Diseases/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL