Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Article in English | MEDLINE | ID: mdl-38330234

ABSTRACT

CONTEXT: Low birth weight, as seen in Silver-Russell syndrome (SRS), is associated with later cardiometabolic disease. Data on long term outcomes and adult body composition in SRS are limited. OBJECTIVE: To evaluate body composition and metabolic health in adults with SRS. DESIGN: This was an observational study. Body composition and metabolic health were assessed at a single appointment. Individuals with SRS were compared with unaffected men and women (from the Southampton Women's Survey (SWS)). SETTING: Clinical research facilities across the UK. PARTICIPANTS: 25 individuals with molecularly-confirmed SRS aged ≥18 years. MAIN OUTCOME MEASURES: Fat mass, lean mass, bone mineral density (BMD), blood pressure, lipids, and blood glucose were measured. RESULTS: 25 adults with SRS were included (52% female). The median age was 32.9 years (range 22.0-69.7). Fat percentage was greater in the SRS group than the SWS cohort (44.1% vs 30.3%, p<0.001). Fat mass index was similar (9.6 vs 7.8, p=0.3). Lean mass percentage (51.8% vs 66.2%, p<0.001) and lean mass index (13.5 kg/m2 vs 17.3 kg/m2, p<0.001) were lower in the SRS group than the SWS cohort. BMD was lower in the SRS group than the SWS cohort (1.08 vs 1.24, p<0.001) (all median values). Total cholesterol was ≥5mmol/L in 52.0%. Triglyceride levels were ≥1.7mmol/L in 20.8%. Fasting blood glucose levels were ≥6.1mmol/L in 25.0%. Hypertension was present in 33.3%. CONCLUSIONS: Adults with SRS have an unfavourable body composition and predisposition to cardiometabolic disease. These results support the need for a health surveillance strategy to mitigate adverse outcomes.

3.
Genet Med ; 25(10): 100927, 2023 10.
Article in English | MEDLINE | ID: mdl-37422718

ABSTRACT

PURPOSE: The SF3B splicing complex is composed of SF3B1-6 and PHF5A. We report a developmental disorder caused by de novo variants in PHF5A. METHODS: Clinical, genomic, and functional studies using subject-derived fibroblasts and a heterologous cellular system were performed. RESULTS: We studied 9 subjects with congenital malformations, including preauricular tags and hypospadias, growth abnormalities, and developmental delay who had de novo heterozygous PHF5A variants, including 4 loss-of-function (LOF), 3 missense, 1 splice, and 1 start-loss variant. In subject-derived fibroblasts with PHF5A LOF variants, wild-type and variant PHF5A mRNAs had a 1:1 ratio, and PHF5A mRNA levels were normal. Transcriptome sequencing revealed alternative promoter use and downregulated genes involved in cell-cycle regulation. Subject and control fibroblasts had similar amounts of PHF5A with the predicted wild-type molecular weight and of SF3B1-3 and SF3B6. SF3B complex formation was unaffected in 2 subject cell lines. CONCLUSION: Our data suggest the existence of feedback mechanisms in fibroblasts with PHF5A LOF variants to maintain normal levels of SF3B components. These compensatory mechanisms in subject fibroblasts with PHF5A or SF3B4 LOF variants suggest disturbed autoregulation of mutated splicing factor genes in specific cell types, that is, neural crest cells, during embryonic development rather than haploinsufficiency as pathomechanism.


Subject(s)
Craniofacial Abnormalities , Hypospadias , Male , Humans , Hypospadias/genetics , RNA Splicing Factors/genetics , RNA Splicing , Transcription Factors/genetics , Transcription Factors/metabolism , Trans-Activators/genetics , RNA-Binding Proteins/genetics
4.
J Hum Genet ; 68(7): 445-453, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36864284

ABSTRACT

BACKGROUND: Neurodevelopmental disorders (NDDs) are heterogeneous, debilitating conditions that include motor and cognitive disability and social deficits. The genetic factors underlying the complex phenotype of NDDs remain to be elucidated. Accumulating evidence suggest that the Elongator complex plays a role in NDDs, given that patient-derived mutations in its ELP2, ELP3, ELP4 and ELP6 subunits have been associated with these disorders. Pathogenic variants in its largest subunit ELP1 have been previously found in familial dysautonomia and medulloblastoma, with no link to NDDs affecting primarily the central nervous system. METHODS: Clinical investigation included patient history and physical, neurological and magnetic resonance imaging (MRI) examination. A novel homozygous likely pathogenic ELP1 variant was identified by whole-genome sequencing. Functional studies included in silico analysis of the mutated ELP1 in the context of the holo-complex, production and purification of the ELP1 harbouring the identified mutation and in vitro analyses using microscale thermophoresis for tRNA binding assay and acetyl-CoA hydrolysis assay. Patient fibroblasts were harvested for tRNA modification analysis using HPLC coupled to mass spectrometry. RESULTS: We report a novel missense mutation in the ELP1 identified in two siblings with intellectual disability and global developmental delay. We show that the mutation perturbs the ability of ELP123 to bind tRNAs and compromises the function of the Elongator in vitro and in human cells. CONCLUSION: Our study expands the mutational spectrum of ELP1 and its association with different neurodevelopmental conditions and provides a specific target for genetic counselling.


Subject(s)
Mutation, Missense , Neurodevelopmental Disorders , Transcriptional Elongation Factors , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Phenotype , RNA, Transfer/metabolism , Transcriptional Elongation Factors/genetics , Neurodevelopmental Disorders/genetics
5.
J Med Genet ; 60(7): 712-716, 2023 07.
Article in English | MEDLINE | ID: mdl-36543535

ABSTRACT

INTRODUCTION: SPRY1 encodes protein sprouty homolog 1 (Spry-1), a negative regulator of receptor tyrosine kinase signalling. Null mutant mice display kidney/urinary tract abnormalities and altered size of the skull; complete loss-of-function of Spry-1 in humans has not been reported. METHODS: Analysis of whole-genome sequencing data from individuals with craniosynostosis enrolled in the 100,000 Genomes Project identified a likely pathogenic variant within SPRY1. Reverse-transcriptase PCR and western blot analysis were used to investigate the effect of the variant on SPRY1 mRNA and protein, in lymphoblastoid cell lines from the patient and both parents. RESULTS: A nonsense variant in SPRY1, encoding p.(Leu27*), was confirmed to be heterozygous in the unaffected parents and homozygous in the child. The child's phenotype, which included sagittal craniosynostosis, subcutaneous cystic lesions overlying the lambdoid sutures, hearing loss associated with bilateral cochlear and vestibular dysplasia and a unilateral renal cyst, overlapped the features reported in Spry1-/- null mice. Functional studies supported escape from nonsense-mediated decay, but western blot analysis demonstrated complete absence of full-length protein in the affected child and a marked reduction in both parents. CONCLUSION: This is the first report of complete loss of Spry-1 function in humans, associated with abnormalities of the cranial sutures, inner ear, and kidneys.


Subject(s)
Craniosynostoses , Ear, Inner , Urinary Tract , Mice , Animals , Child , Humans , Adaptor Proteins, Signal Transducing/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphoproteins/genetics , Mice, Knockout , Craniosynostoses/genetics
6.
Clin Endocrinol (Oxf) ; 97(3): 284-292, 2022 09.
Article in English | MEDLINE | ID: mdl-35261046

ABSTRACT

OBJECTIVE: Silver-Russell syndrome (SRS) causes short stature. Growth hormone (GH) treatment aims to increase adult height. However, data are limited on the long-term outcomes of GH in patients with molecularly confirmed SRS. This study evaluated height, body mass index (BMI) and GH treatment in molecularly confirmed SRS. DESIGN: An observational study with retrospective data collection. PATIENTS: Individuals with molecularly confirmed SRS aged ≥13 years. MEASUREMENTS: Data were collected on height, height gain (change in height standard deviation score [SDS] from childhood to final or near-final height), BMI and gain in BMI (from childhood to adulthood) and previous GH treatment. RESULTS: Seventy-one individuals (40 female) were included. The median age was 22.0 years (range 13.2-69.7). The molecular diagnoses: H19/IGF2:IG-DMR LOM in 80.3% (57/71); upd(7)mat in 16.9% (12/71) and IGF2 mutation in 2.8% (2/71). GH treatment occurred in 77.5% (55/71). Total height gain was greater in GH-treated individuals (median 1.53 SDS vs. 0.53 SDS, p = .007), who were shorter at treatment initiation (-3.46 SDS vs. -2.91 SDS, p = .04) but reached comparable heights to GH-untreated individuals (-2.22 SDS vs. -2.74 SDS, p = .7). In GH-treated individuals, BMI SDS was lower at the most recent assessment (median -1.10 vs. 1.66, p = .002) with lower BMI gain (2.01 vs. 3.58, p = .006) despite similar early BMI SDS to GH-untreated individuals (median -2.65 vs. -2.78, p = .3). CONCLUSIONS: These results support the use of GH in SRS for increasing height SDS. GH treatment was associated with lower adult BMI which may reflect improved metabolic health even following discontinuation of therapy.


Subject(s)
Body Height , Body Mass Index , Human Growth Hormone , Silver-Russell Syndrome , Adolescent , Adult , Aged , Female , Human Growth Hormone/therapeutic use , Humans , Male , Middle Aged , Retrospective Studies , Silver-Russell Syndrome/drug therapy , Young Adult
7.
Arch Dis Child ; 106(12): 1195-1201, 2021 12.
Article in English | MEDLINE | ID: mdl-33741574

ABSTRACT

OBJECTIVE: The psychosocial impact of growing up with Silver-Russell syndrome (SRS), characterised by growth failure and short stature in adulthood, has been explored in adults; however, there are no accounts of contemporary lived experience in adolescents. Such data could inform current healthcare guidance and transition to adult services. We aimed to explore the lived experience of adolescents with SRS. DESIGN/SETTING/PATIENTS: In-depth, semi-structured interviews were conducted between January 2015 and October 2016 with a sample of eight adolescents aged 13-18 (five girls) with genetically confirmed SRS from the UK. Qualitative interviews were transcribed and coded to identify similarities and differences using thematic analysis; codes were then grouped to form overarching themes. RESULTS: We identified four themes from the interview data: (1) the psychosocial challenges of feeling and looking different; (2) pain, disability and fatigue; (3) anticipated stigma; and (4) building resilience and acceptance. Despite adolescents accepting SRS in their lives, they described ongoing psychosocial challenges and anticipated greater problems to come, such as stigma from prospective employers. CONCLUSIONS: Adolescents with SRS may experience psychosocial difficulties from as young as 10 years old related to feeling and looking different; pain, disability and fatigue; anticipated stigma; and future challenges around employment. We discuss these findings in relation to recommendations for the care of adolescents with SRS to prepare them for adult life.


Subject(s)
Adaptation, Psychological , Fatigue , Pain , Silver-Russell Syndrome/psychology , Social Stigma , Adolescent , Disability Evaluation , Dwarfism/psychology , Emotions , Fatigue/diagnosis , Fatigue/etiology , Female , Humans , Interviews as Topic , Male , Pain/diagnosis , Pain/etiology , Qualitative Research , Resilience, Psychological , Silver-Russell Syndrome/diagnosis
8.
J Med Genet ; 57(10): 683-691, 2020 10.
Article in English | MEDLINE | ID: mdl-32054688

ABSTRACT

BACKGROUND: Silver-Russell syndrome is an imprinting disorder that restricts growth, resulting in short adult stature that may be ameliorated by treatment. Approximately 50% of patients have loss of methylation of the imprinting control region (H19/IGF2:IG-DMR) on 11p15.5 and 5%-10% have maternal uniparental disomy of chromosome 7. Most published research focuses on the childhood phenotype. Our aim was to describe the phenotypic characteristics of older patients with SRS. METHODS: A retrospective cohort of 33 individuals with a confirmed molecular diagnosis of SRS aged 13 years or above were carefully phenotyped. RESULTS: The median age of the cohort was 29.6 years; 60.6% had a height SD score (SDS) ≤-2 SDS despite 70% having received growth hormone treatment. Relative macrocephaly, feeding difficulties and a facial appearance typical of children with SRS were no longer discriminatory diagnostic features. In those aged ≥18 years, impaired glucose tolerance in 25%, hypertension in 33% and hypercholesterolaemia in 52% were noted. While 9/33 accessed special education support, university degrees were completed in 40.0% (>21 years). There was no significant correlation between quality of life and height SDS. 9/25 were parents and none of the 17 offsprings had SRS. CONCLUSION: Historical treatment regimens for SRS were not sufficient for normal adult growth and further research to optimise treatment is justified. Clinical childhood diagnostic scoring systems are not applicable to patients presenting in adulthood and SRS diagnosis requires molecular confirmation. Metabolic ill-health warrants further investigation but SRS is compatible with a normal quality of life including normal fertility in many cases.


Subject(s)
Insulin-Like Growth Factor II/genetics , RNA, Long Noncoding/genetics , Silver-Russell Syndrome/genetics , Uniparental Disomy/genetics , Adolescent , Adult , Aged , DNA Methylation/genetics , Epigenesis, Genetic , Female , Genomic Imprinting/genetics , Humans , Male , Middle Aged , Phenotype , Quality of Life , Silver-Russell Syndrome/pathology , Uniparental Disomy/pathology , Young Adult
9.
Arch Dis Child ; 104(1): 76-82, 2019 01.
Article in English | MEDLINE | ID: mdl-29954740

ABSTRACT

OBJECTIVE: There is limited information on the psychosocial impact of growing up with Silver-Russell syndrome (SRS), characterised by slow growth in utero leading to short stature in adulthood. Such information could aid families in making difficult treatment decisions and guide management strategies for health professionals. We aimed to explore the lived experience of people with SRS across the lifespan. DESIGN/SETTING/PATIENTS: In-depth, semi-structured interviews were conducted between January 2015 and October 2016 with a sample of 15 adults (six women) with genetically confirmed SRS from the UK. Qualitative interviews were transcribed and coded to identify similarities and differences: codes were then grouped to form overarching themes. RESULTS: Four themes were identified from participant accounts: (1) appearance-related concerns extending beyond height; (2) strategies to deal with real and perceived threats; (3) women's experiences of pain, disability and feeling older than their years; and (4) feeling overlooked in romantic relationships. These themes show that other factors, beyond short stature, affect patient well-being and indicate a mismatch between patient need and healthcare provision. CONCLUSIONS: Challenges in SRS during childhood and adolescence were central to the psychosocial impact of SRS, and were not limited to height. These challenges, as well as symptoms such as pain and fatigue for women, have not previously been documented. To help individuals with SRS develop strategies to manage psychosocial issues, we recommend clinicians incorporate psychological services as an integral part of multidisciplinary teams managing individuals with SRS during childhood, adolescence and adulthood.


Subject(s)
Adaptation, Psychological/physiology , Body Height , Dwarfism , Pain , Silver-Russell Syndrome , Adult , Child , Disability Evaluation , Dwarfism/etiology , Dwarfism/physiopathology , Dwarfism/psychology , Fatigue/diagnosis , Fatigue/etiology , Female , Humans , Male , Needs Assessment , Pain/diagnosis , Pain/etiology , Psychology , Sex Factors , Silver-Russell Syndrome/diagnosis , Silver-Russell Syndrome/epidemiology , Silver-Russell Syndrome/physiopathology , Silver-Russell Syndrome/psychology , United Kingdom/epidemiology
10.
J Med Genet ; 55(12): 803-813, 2018 12.
Article in English | MEDLINE | ID: mdl-30287594

ABSTRACT

BACKGROUND: Progressive encephalopathy, hypsarrhythmia and optic atrophy (PEHO) has been described as a clinically distinct syndrome. It has been postulated that it is an autosomal recessive condition. However, the aetiology is poorly understood, and the genetic basis of the condition has not been fully elucidated. Our objective was to discover if PEHO syndrome is a single gene disorder. METHOD: Children with PEHO and PEHO-like syndrome were recruited. Clinical, neurological and dysmorphic features were recorded; EEG reports and MRI scans were reviewed. Where possible, exome sequencing was carried out first to seek mutations in known early infantile developmental and epileptic encephalopathy (DEE) genes and then to use an agnostic approach to seek novel candidate genes. We sought intra-interfamilial phenotypic correlations and genotype-phenotype correlations when pathological mutations were identified. RESULTS: Twenty-three children were recruited from a diverse ethnic background, 19 of which were suitable for inclusion. They were similar in many of the core and the supporting features of PEHO, but there was significant variation in MRI and ophthalmological findings, even between siblings with the same mutation. A pathogenic genetic variant was identified in 15 of the 19 children. One further girl's DNA failed analysis, but her two affected sisters shared confirmed variants. Pathogenic variants were identified in seven different genes. CONCLUSIONS: We found significant clinical and genetic heterogeneity. Given the intrafamily variation demonstrated, we question whether the diagnostic criteria for MRI and ophthalmic findings should be altered. We also question whether PEHO and PEHO-like syndrome represent differing points on a clinical spectrum of the DEE. We conclude that PEHO and PEHO-like syndrome are clinically and genetically diverse entities-and are phenotypic endpoints of many severe genetic encephalopathies.


Subject(s)
Brain Edema/diagnosis , Brain Edema/etiology , Epilepsy/diagnosis , Epilepsy/genetics , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/etiology , Optic Atrophy/diagnosis , Optic Atrophy/etiology , Spasms, Infantile/diagnosis , Spasms, Infantile/etiology , Age Factors , Alleles , Biomarkers , Child, Preschool , Electroencephalography , Facies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Testing , Genotype , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Mutation , Pedigree , Phenotype
11.
Eur J Hum Genet ; 26(9): 1288-1293, 2018 09.
Article in English | MEDLINE | ID: mdl-29891876

ABSTRACT

Metaphyseal dysplasia with maxillary hypoplasia and brachydactyly (MDMHB) is an autosomal-dominant skeletal dysplasia characterised by metaphyseal flaring of the long bones, enlargement of the medial halves of the clavicles, maxillary hypoplasia, brachydactyly, dental anomalies and mild osteoporosis. To date, only one large French Canadian family and a Finnish woman have been reported with the condition. In both, intragenic duplication encompassing exons 3-5 of the RUNX2 gene was identified. We describe a new, three-generation family with clinical features of MDMHB and an intragenic tandem duplication of RUNX2 exons 3-6. Dental problems were the primary presenting feature in all four affected individuals. We compare the features in our family to those previously reported in MDMHB, review the natural history of this condition and highlight the importance of considering an underlying skeletal dysplasia in patients presenting with significant dental problems and other suggestive features, including disproportionate short stature and/or digital anomalies.


Subject(s)
Brachydactyly/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Jaw Abnormalities/genetics , Osteochondrodysplasias/genetics , Tooth Abnormalities/genetics , Adult , Aged , Brachydactyly/pathology , Female , Gene Duplication , Humans , Jaw Abnormalities/pathology , Male , Middle Aged , Osteochondrodysplasias/pathology , Pedigree , Syndrome , Tooth Abnormalities/pathology
12.
J Med Genet ; 55(7): 497-504, 2018 07.
Article in English | MEDLINE | ID: mdl-29574422

ABSTRACT

BACKGROUND: Genomic imprinting results from the resistance of germline epigenetic marks to reprogramming in the early embryo for a small number of mammalian genes. Genetic, epigenetic or environmental insults that prevent imprints from evading reprogramming may result in imprinting disorders, which impact growth, development, behaviour and metabolism. We aimed to identify genetic defects causing imprinting disorders by whole-exome sequencing in families with one or more members affected by multilocus imprinting disturbance. METHODS: Whole-exome sequencing was performed in 38 pedigrees where probands had multilocus imprinting disturbance, in five of whom maternal variants in NLRP5 have previously been found. RESULTS: We now report 15 further pedigrees in which offspring had disturbance of imprinting, while their mothers had rare, predicted-deleterious variants in maternal effect genes, including NLRP2, NLRP7 and PADI6. As well as clinical features of well-recognised imprinting disorders, some offspring had additional features including developmental delay, behavioural problems and discordant monozygotic twinning, while some mothers had reproductive problems including pregnancy loss. CONCLUSION: The identification of 20 putative maternal effect variants in 38 families affected by multilocus imprinting disorders adds to the evidence that maternal genetic factors affect oocyte fitness and thus offspring development. Testing for maternal-effect genetic variants should be considered in families affected by atypical imprinting disorders.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Beckwith-Wiedemann Syndrome/genetics , Protein-Arginine Deiminases/genetics , Silver-Russell Syndrome/genetics , Apoptosis Regulatory Proteins , Beckwith-Wiedemann Syndrome/pathology , Chromosomes, Human, Pair 11/genetics , DNA Methylation/genetics , Female , Genomic Imprinting/genetics , Germ-Line Mutation/genetics , Humans , Infant, Newborn , Infant, Newborn, Diseases/genetics , Infant, Newborn, Diseases/physiopathology , Maternal Inheritance , Pedigree , Pregnancy , Protein-Arginine Deiminase Type 6 , Silver-Russell Syndrome/physiopathology
13.
Nat Rev Endocrinol ; 13(2): 105-124, 2017 02.
Article in English | MEDLINE | ID: mdl-27585961

ABSTRACT

This Consensus Statement summarizes recommendations for clinical diagnosis, investigation and management of patients with Silver-Russell syndrome (SRS), an imprinting disorder that causes prenatal and postnatal growth retardation. Considerable overlap exists between the care of individuals born small for gestational age and those with SRS. However, many specific management issues exist and evidence from controlled trials remains limited. SRS is primarily a clinical diagnosis; however, molecular testing enables confirmation of the clinical diagnosis and defines the subtype. A 'normal' result from a molecular test does not exclude the diagnosis of SRS. The management of children with SRS requires an experienced, multidisciplinary approach. Specific issues include growth failure, severe feeding difficulties, gastrointestinal problems, hypoglycaemia, body asymmetry, scoliosis, motor and speech delay and psychosocial challenges. An early emphasis on adequate nutritional status is important, with awareness that rapid postnatal weight gain might lead to subsequent increased risk of metabolic disorders. The benefits of treating patients with SRS with growth hormone include improved body composition, motor development and appetite, reduced risk of hypoglycaemia and increased height. Clinicians should be aware of possible premature adrenarche, fairly early and rapid central puberty and insulin resistance. Treatment with gonadotropin-releasing hormone analogues can delay progression of central puberty and preserve adult height potential. Long-term follow up is essential to determine the natural history and optimal management in adulthood.


Subject(s)
Disease Management , Internationality , Silver-Russell Syndrome/diagnosis , Silver-Russell Syndrome/therapy , Gonadotropin-Releasing Hormone/therapeutic use , Human Growth Hormone/therapeutic use , Humans , Silver-Russell Syndrome/metabolism
14.
Nat Genet ; 44(11): 1243-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23001123

ABSTRACT

Adenosine deaminases acting on RNA (ADARs) catalyze the hydrolytic deamination of adenosine to inosine in double-stranded RNA (dsRNA) and thereby potentially alter the information content and structure of cellular RNAs. Notably, although the overwhelming majority of such editing events occur in transcripts derived from Alu repeat elements, the biological function of non-coding RNA editing remains uncertain. Here, we show that mutations in ADAR1 (also known as ADAR) cause the autoimmune disorder Aicardi-Goutières syndrome (AGS). As in Adar1-null mice, the human disease state is associated with upregulation of interferon-stimulated genes, indicating a possible role for ADAR1 as a suppressor of type I interferon signaling. Considering recent insights derived from the study of other AGS-related proteins, we speculate that ADAR1 may limit the cytoplasmic accumulation of the dsRNA generated from genomic repetitive elements.


Subject(s)
Adenosine Deaminase/genetics , Autoimmune Diseases of the Nervous System/genetics , Interferon Type I , Nervous System Malformations/genetics , RNA, Double-Stranded/metabolism , Alu Elements/genetics , Animals , Exome , Gene Expression , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Mice , Mutation , Protein Conformation , RNA, Double-Stranded/genetics , RNA-Binding Proteins , Sequence Analysis, DNA , Signal Transduction , Structure-Activity Relationship
15.
J Dermatol Sci ; 64(3): 159-62, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21945601

ABSTRACT

BACKGROUND: X-linked ichthyosis (XLI) is a relatively common, recessive condition caused by mutations in the steroid sulfatase (STS) gene. Common loss-of-function mutations in the filaggrin gene (FLG) cause ichthyosis vulgaris and predispose individuals to atopic eczema. OBJECTIVE: To test the hypothesis that co-inheritance of FLG mutations can act as a genetic modifier in XLI. METHODS: An unusually severe XLI phenotype in addition to eczema and mild childhood asthma was investigated in a female Indian patient by fluorescent in situ hybridization (FISH) for the common STS gene deletion. Direct sequencing of the entire FLG gene was also performed. RESULTS: FISH analysis revealed that the proband was homozygous for the common STS genomic deletion mutation. Further investigation revealed a frame-shift mutation 3672del4 in the gene encoding filaggrin (FLG), leading to premature termination of profilaggrin translation. Interestingly, her father, who had a very typical mild presentation of XLI, did not carry this FLG mutation in addition to his STS deletion. Her mother was a heterozygous carrier of the FLG mutation and consistent with this, had mild symptoms of ichthyosis vulgaris; she was also a heterozygous carrier of the STS deletion. CONCLUSION: This is the second reported case of the modifying effects of FLG null alleles on XLI and strengthens the hypothesis that filaggrin defects can synergize with STS deficiency to exacerbate the ichthyosis phenotype.


Subject(s)
Gene Deletion , Ichthyosis, X-Linked/genetics , Intermediate Filament Proteins/genetics , Mutation, Missense , Skin/pathology , Steryl-Sulfatase/genetics , Administration, Cutaneous , Child , DNA Mutational Analysis , Dermatologic Agents/administration & dosage , Female , Filaggrin Proteins , Genetic Predisposition to Disease , Heredity , Heterozygote , Homozygote , Humans , Ichthyosis, X-Linked/drug therapy , Ichthyosis, X-Linked/enzymology , Ichthyosis, X-Linked/pathology , In Situ Hybridization, Fluorescence , Pedigree , Phenotype , Severity of Illness Index , Skin/drug effects
16.
Arch Dis Child ; 96(12): 1156-61, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21349887

ABSTRACT

Silver-Russell syndrome (SRS) is characterised by intrauterine growth restriction, poor postnatal growth, relative macrocephaly, triangular face, asymmetry and feeding difficulties. As many of these features are non-specific, clinical diagnosis of SRS remains difficult. Hypomethylation of the imprinting control region (ICR) 1 on chromosome 11p15 and maternal uniparental disomy (mUPD) for chromosome 7 are found in up to 60% and around 5-10% of patients with SRS, respectively. Patients with ICR1 hypomethylation are more likely to have classical features of SRS, including asymmetry; patients with mUPD7 are more likely to have learning difficulties, particularly speech problems, although these are usually mild. As features vary widely in severity, clinicians should have a low threshold for genetic investigation of patients with features suggestive of SRS.


Subject(s)
Silver-Russell Syndrome/diagnosis , Developmental Disabilities/diagnosis , Facies , Feeding Behavior , Genomic Imprinting , Genotype , Growth Disorders/diagnosis , Humans , Infant, Newborn , Phenotype , Prognosis , Silver-Russell Syndrome/genetics , Silver-Russell Syndrome/therapy , Uniparental Disomy
17.
Am J Hum Genet ; 87(6): 757-67, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21129728

ABSTRACT

Altered Bone Morphogenetic Protein (BMP) signaling leads to multiple developmental defects, including brachydactyly and deafness. Here we identify chondroitin synthase 1 (CHSY1) as a potential mediator of BMP effects. We show that loss of human CHSY1 function causes autosomal-recessive Temtamy preaxial brachydactyly syndrome (TPBS), mainly characterized by limb malformations, short stature, and hearing loss. After mapping the TPBS locus to chromosome 15q26-qterm, we identified causative mutations in five consanguineous TPBS families. In zebrafish, antisense-mediated chsy1 knockdown causes defects in multiple developmental processes, some of which are likely to also be causative in the etiology of TPBS. In the inner ears of zebrafish larvae, chsy1 is expressed similarly to the BMP inhibitor dan and in a complementary fashion to bmp2b. Furthermore, unrestricted Bmp2b signaling or loss of Dan activity leads to reduced chsy1 expression and, during epithelial morphogenesis, defects similar to those that occur upon Chsy1 inactivation, indicating that Bmp signaling affects inner-ear development by repressing chsy1. In addition, we obtained strikingly similar zebrafish phenotypes after chsy1 overexpression, which might explain why, in humans, brachydactyly can be caused by mutations leading either to loss or to gain of BMP signaling.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Mutation , N-Acetylgalactosaminyltransferases/genetics , Signal Transduction , Animals , Brachydactyly , Chromosome Mapping , Chromosomes, Human, Pair 15 , Foot Deformities, Congenital/genetics , Hand Deformities, Congenital/genetics , Humans , N-Acetylgalactosaminyltransferases/metabolism , Syndrome , Zebrafish
19.
Eur J Hum Genet ; 18(6): 648-55, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20104244

ABSTRACT

This study was an investigation of 79 patients referred to the Wessex Regional Genetics Laboratory with suspected Russell-Silver Syndrome or unexplained short stature/intra uterine growth restriction, warranting genetic investigation. Methylation status was analysed at target sequences within eleven imprinted loci (PLAGL1, IGF2R, PEG10, MEST1, GRB10, KCNQ1OT1, H19, IGF2P0, DLK1, PEG3, NESPAS). Thirty seven percent (37%) (29 of 79) of samples were shown to have a methylation abnormality. The commonest finding was a loss of methylation at H19 (23 of 29), as previously reported in Russell-Silver Syndrome. In addition, four of these patients had methylation anomalies at other loci, of whom two showed hypomethylation of multiple imprinted loci, and two showed a complete gain of methylation at IGF2R. This latter finding was also present in five other patients who did not have demonstrable changes at H19. In total, 7 of 79 patients showed a gain of methylation at IGF2R and this was significantly different from a normal control population of 267 individuals (P=0.002). This study in patients with growth restriction shows the importance of widening the epigenetic investigation to include multiple imprinted loci and highlights potential involvement of the IGF2R locus.


Subject(s)
DNA Methylation/genetics , Fetal Growth Retardation/genetics , Genetic Loci , Genomic Imprinting , Growth Disorders/genetics , Child , Child, Preschool , Cohort Studies , Developmental Disabilities/genetics , Epigenesis, Genetic , Female , Genetic Loci/genetics , Genomic Imprinting/physiology , Humans , Infant, Newborn , Pregnancy , RNA, Long Noncoding , RNA, Untranslated/genetics , Receptor, IGF Type 2/genetics , Sequence Analysis, DNA , Silver-Russell Syndrome/genetics
20.
Clin Dysmorphol ; 19(1): 23-27, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19952732

ABSTRACT

We describe four children from two consanguineous families with distinctive hand and foot anomalies including preaxial brachydactyly, together with phalangeal duplication, symphalangism and hyperphalangism of fingers I-III. These anomalies are remarkably similar to those described in a previous case report. Additional features were noted both in this case and, to variable degrees, in the four children reported here. These included sensorineural deafness, optic atrophy, mild facial dysmorphism, orodental anomalies and developmental delay. Autosomal recessive inheritance was previously suggested as the patient had a similarly affected brother and his parents were consanguineous. These four cases provide additional evidence for a novel, autosomal recessive disorder involving limb and other associated anomalies.


Subject(s)
Fingers/pathology , Foot Deformities, Congenital/genetics , Hand Deformities, Congenital/genetics , Optic Atrophy/genetics , Child , Child, Preschool , Consanguinity , Deafness/genetics , Deafness/pathology , Face/abnormalities , Female , Foot Deformities, Congenital/pathology , Genes, Recessive , Genetic Linkage , Hand Deformities, Congenital/pathology , Humans , Male , Optic Atrophy/pathology , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...