Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Genes Dev ; 34(21-22): 1520-1533, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33060134

ABSTRACT

DNA replication is fundamental for cell proliferation in all organisms. Nonetheless, components of the replisome have been implicated in human disease, and here we report PRIM1 encoding the catalytic subunit of DNA primase as a novel disease gene. Using a variant classification agnostic approach, biallelic mutations in PRIM1 were identified in five individuals. PRIM1 protein levels were markedly reduced in patient cells, accompanied by replication fork asymmetry, increased interorigin distances, replication stress, and prolonged S-phase duration. Consequently, cell proliferation was markedly impaired, explaining the patients' extreme growth failure. Notably, phenotypic features distinct from those previously reported with DNA polymerase genes were evident, highlighting differing developmental requirements for this core replisome component that warrant future investigation.


Subject(s)
DNA Primase/genetics , Dwarfism/genetics , Fetal Growth Retardation/genetics , DNA Primase/chemistry , DNA Primase/deficiency , Dwarfism/diagnostic imaging , Dwarfism/pathology , Female , Fetal Growth Retardation/diagnostic imaging , Fetal Growth Retardation/pathology , Genetic Variation , Humans , Infant , Male , Pedigree , Syndrome
2.
J Med Genet ; 61(2): 132-141, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-37580113

ABSTRACT

BACKGROUND: Pathogenic variants in the zinc finger protein coding genes are rare causes of intellectual disability and congenital malformations. Mutations in the ZNF148 gene causing GDACCF syndrome (global developmental delay, absent or hypoplastic corpus callosum, dysmorphic facies; MIM #617260) have been reported in five individuals so far. METHODS: As a result of an international collaboration using GeneMatcher Phenome Central Repository and personal communications, here we describe the clinical and molecular genetic characteristics of 22 previously unreported individuals. RESULTS: The core clinical phenotype is characterised by developmental delay particularly in the domain of speech development, postnatal growth retardation, microcephaly and facial dysmorphism. Corpus callosum abnormalities appear less frequently than suggested by previous observations. The identified mutations concerned nonsense or frameshift variants that were mainly located in the last exon of the ZNF148 gene. Heterozygous deletion including the entire ZNF148 gene was found in only one case. Most mutations occurred de novo, but were inherited from an affected parent in two families. CONCLUSION: The GDACCF syndrome is clinically diverse, and a genotype-first approach, that is, exome sequencing is recommended for establishing a genetic diagnosis rather than a phenotype-first approach. However, the syndrome may be suspected based on some recurrent, recognisable features. Corpus callosum anomalies were not as constant as previously suggested, we therefore recommend to replace the term 'GDACCF syndrome' with 'ZNF148-related neurodevelopmental disorder'.


Subject(s)
Intellectual Disability , Leukoencephalopathies , Humans , Child , Corpus Callosum , Facies , Mutation/genetics , Phenotype , Genotype , Intellectual Disability/genetics , Intellectual Disability/diagnosis , Syndrome , Developmental Disabilities/pathology , DNA-Binding Proteins/genetics , Transcription Factors/genetics
3.
J Med Genet ; 61(6): 503-519, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38471765

ABSTRACT

Rubinstein-Taybi syndrome (RTS) is an archetypical genetic syndrome that is characterised by intellectual disability, well-defined facial features, distal limb anomalies and atypical growth, among numerous other signs and symptoms. It is caused by variants in either of two genes (CREBBP, EP300) which encode for the proteins CBP and p300, which both have a function in transcription regulation and histone acetylation. As a group of international experts and national support groups dedicated to the syndrome, we realised that marked heterogeneity currently exists in clinical and molecular diagnostic approaches and care practices in various parts of the world. Here, we outline a series of recommendations that document the consensus of a group of international experts on clinical diagnostic criteria for types of RTS (RTS1: CREBBP; RTS2: EP300), molecular investigations, long-term management of various particular physical and behavioural issues and care planning. The recommendations as presented here will need to be evaluated for improvements to allow for continued optimisation of diagnostics and care.


Subject(s)
CREB-Binding Protein , E1A-Associated p300 Protein , Rubinstein-Taybi Syndrome , Rubinstein-Taybi Syndrome/genetics , Rubinstein-Taybi Syndrome/diagnosis , Rubinstein-Taybi Syndrome/therapy , Humans , CREB-Binding Protein/genetics , E1A-Associated p300 Protein/genetics , Consensus , Disease Management , Mutation
4.
Eur J Public Health ; 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905592

ABSTRACT

BACKGROUND: Somatic and germline genetic alterations are significant drivers of cancer. Increasing integration of new technologies which profile these alterations requires timely, equitable and high-quality genetic counselling to facilitate accurate diagnoses and informed decision-making by patients and their families in preventive and clinical settings. This article aims to provide an overview of genetic counselling legislation and practice across European Union (EU) Member States to serve as a foundation for future European recommendations and action. METHODS: National legislative databases of all 27 Member States were searched using terms relevant to genetic counselling, translated as appropriate. Interviews with relevant experts from each Member State were conducted to validate legislative search results and provide detailed insights into genetic counselling practice in each country. RESULTS: Genetic counselling is included in national legislative documents of 22 of 27 Member States, with substantial variation in legal mechanisms and prescribed details (i.e. the 'who, what, when and where' of counselling). Practice is similarly varied. Workforce capacity (25 of 27 Member States) and genetic literacy (all Member States) were common reported barriers. Recognition and/or better integration of genetic counsellors and updated legislation and were most commonly noted as the 'most important change' which would improve practice. CONCLUSIONS: This review highlights substantial variability in genetic counselling across EU Member States, as well as common barriers notwithstanding this variation. Future recommendations and action should focus on addressing literacy and capacity challenges through legislative, regulatory and/or strategic approaches at EU, national, regional and/or local levels.

5.
Eur J Haematol ; 111(2): 254-262, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37194391

ABSTRACT

BACKGROUND: FA patients are hypersensitive to preconditioning of bone marrow transplantation. OBJECTIVE: Assessment of the power of mitomycin C (MMC) test to assign FA patients. METHODS: We analysed 195 patients with hematological disorders using spontaneous and two types of chromosomal breakage tests (MMC and bleomycin). In case of presumed Ataxia telangiectasia (AT), patients' blood was irradiated in vitro to determine the radiosensitivity of the patients. RESULTS: Seven patients were diagnosed as having FA. The number of spontaneous chromosomal aberrations was significantly higher in FA patients than in aplastic anemia (AA) patients including chromatid breaks, exchanges, total aberrations, aberrant cells. MMC-induced ≥10 break/cell was 83.9 ± 11.4% in FA patients and 1.94 ± 0.41% in AA patients (p < .0001). The difference in bleomycin-induced breaks/cell was also significant: 2.01 ± 0.25 (FA) versus 1.30 ± 0.10 (AA) (p = .019). Seven patients showed increased radiation sensitivity. Both dicentric + ring, and total aberrations were significantly higher at 3 and 6 Gy compared to controls. CONCLUSIONS: MMC and Bleomycin tests together proved to be more informative than MMC test alone for the diagnostic classification of AA patients, while in vitro irradiation tests could help detect radiosensitive-as such, individuals with AT.


Subject(s)
Anemia, Aplastic , Fanconi Anemia , Humans , Anemia, Aplastic/etiology , Anemia, Aplastic/genetics , Fanconi Anemia/complications , Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Chromosome Breakage , Diagnosis, Differential , Mitomycin , Bleomycin
6.
BMC Med Genet ; 21(1): 61, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32216767

ABSTRACT

BACKGROUND: Wolcott-Rallison Syndrome (WRS) is a rare autosomal recessive disease that is the most common cause of neonatal diabetes in consanguineous families. WRS is caused by various genetic alterations of the Eukaryotic Translation Initiation Factor 2-Alpha Kinase 3 (EIF2AK3) gene. METHODS: Genetic analysis of a consanguineous family where two children were diagnosed with WRS was performed by Sanger sequencing. The altered protein was investigated by in vitro cloning, expression and immunohistochemistry. RESULTS: The first cases in Hungary, - two patients in one family, where the parents were fourth-degree cousins - showed the typical clinical features of WRS: early onset diabetes mellitus with hyperglycemia, growth retardation, infection-induced multiple organ failure. The genetic background of the disease was a novel alteration in the EIF2AK3 gene involving the splice site of exon 11- intron 11-12 boundary: g.53051_53062delinsTG. According to cDNA sequencing this created a new splice site and resulted in a frameshift and the development of an early termination codon at amino acid position 633 (p.Pro627AspfsTer7). Based on in vitro cloning and expression studies, the truncated protein was functionally inactive. Immunohistochemistry revealed that the intact protein was absent in the islets of pancreas, furthermore insulin expressing cells were also dramatically diminished. Elevated GRP78 and reduced CHOP protein expression were observed in the liver. CONCLUSIONS: The novel genetic alteration causing the absence of the EIF2AK3 protein resulted in insufficient handling of severe endoplasmic reticulum stress, leading to liver failure and demise of the patients.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Epiphyses/abnormalities , INDEL Mutation , Osteochondrodysplasias/genetics , RNA Splice Sites/genetics , eIF-2 Kinase/genetics , Child, Preschool , Consanguinity , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/pathology , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , Epiphyses/pathology , Fatal Outcome , Female , Frameshift Mutation , Humans , Hungary , Infant , Liver Failure/complications , Liver Failure/genetics , Liver Failure/pathology , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/pathology , Pedigree , Siblings , Virus Diseases/complications , Virus Diseases/pathology
7.
Hum Genet ; 137(11-12): 921-939, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30450527

ABSTRACT

Juvenile segmental progeroid syndromes are rare, heterogeneous disorders characterized by signs of premature aging affecting more than one tissue or organ starting in childhood. Hutchinson-Gilford progeria syndrome (HGPS), caused by a recurrent de novo synonymous LMNA mutation resulting in aberrant splicing and generation of a mutant product called progerin, is a prototypical example of such disorders. Here, we performed a joint collaborative study using massively parallel sequencing and targeted Sanger sequencing, aimed at delineating the underlying genetic cause of 14 previously undiagnosed, clinically heterogeneous, non-LMNA-associated juvenile progeroid patients. The molecular diagnosis was achieved in 11 of 14 cases (~ 79%). Furthermore, we firmly establish biallelic mutations in POLR3A as the genetic cause of a recognizable, neonatal, Wiedemann-Rautenstrauch-like progeroid syndrome. Thus, we suggest that POLR3A mutations are causal for a portion of under-diagnosed early-onset segmental progeroid syndromes. We additionally expand the clinical spectrum associated with PYCR1 mutations by showing that they can somewhat resemble HGPS in the first year of life. Moreover, our results lead to clinical reclassification in one single case. Our data emphasize the complex genetic and clinical heterogeneity underlying progeroid disorders.


Subject(s)
Fetal Growth Retardation/genetics , Progeria/genetics , Pyrroline Carboxylate Reductases/genetics , RNA Polymerase III/genetics , Adolescent , Alternative Splicing/genetics , Child , Female , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/pathology , Genetic Predisposition to Disease , Humans , Infant , Lamin Type A/genetics , Male , Mutation , Phenotype , Progeria/diagnosis , Progeria/pathology , Progeria/physiopathology , delta-1-Pyrroline-5-Carboxylate Reductase
8.
Am J Med Genet A ; 176(1): 187-193, 2018 01.
Article in English | MEDLINE | ID: mdl-29160006

ABSTRACT

The "blepharophimosis-mental retardation" syndromes (BMRS) consist of a group of clinically and genetically heterogeneous congenital malformation syndromes, where short palpebral fissures and intellectual disability associate with a distinct set of other morphological features. Kaufman oculocerebrofacial syndrome represents a rare and recently reevaluated entity within the BMR syndromes and is caused by biallelic mutations of UBE3B. Affected individuals typically show microcephaly, impaired somatic growth, gastrointestinal and genitourinary problems, ectodermal anomalies and a characteristic face with short, upslanted palpebral fissures, depressed nasal bridge. and anteverted nares. Here we present four patients with five novel UBE3B mutations and propose the inclusion of clinical features to the characteristics of Kaufman oculocerebrofacial syndrome, including prominence of the cheeks and limb anomalies.


Subject(s)
Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Genetic Association Studies , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Limb Deformities, Congenital/diagnosis , Limb Deformities, Congenital/genetics , Microcephaly/diagnosis , Microcephaly/genetics , Mutation , Phenotype , Ubiquitin-Protein Ligases/genetics , Biomarkers , Child , DNA Mutational Analysis , Diagnostic Imaging , Eye Abnormalities/therapy , Facies , Female , Genetic Heterogeneity , High-Throughput Nucleotide Sequencing , Humans , Infant , Intellectual Disability/therapy , Limb Deformities, Congenital/therapy , Microcephaly/therapy , Sequence Analysis, DNA
9.
Hum Mutat ; 36(11): 1070-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26172944

ABSTRACT

Segmental progeroid syndromes are rare, heterogeneous disorders characterized by signs of premature aging affecting more than one tissue or organ. A prototypic example is the Werner syndrome (WS), caused by biallelic germline mutations in the Werner helicase gene (WRN). While heterozygous lamin A/C (LMNA) mutations are found in a few nonclassical cases of WS, another 10%-15% of patients initially diagnosed with WS do not have mutations in WRN or LMNA. Germline POLD1 mutations were recently reported in five patients with another segmental progeroid disorder: mandibular hypoplasia, deafness, progeroid features syndrome. Here, we describe eight additional patients with heterozygous POLD1 mutations, thereby substantially expanding the characterization of this new example of segmental progeroid disorders. First, we identified POLD1 mutations in patients initially diagnosed with WS. Second, we describe POLD1 mutation carriers without clinically relevant hearing impairment or mandibular underdevelopment, both previously thought to represent obligate diagnostic features. These patients also exhibit a lower incidence of metabolic abnormalities and joint contractures. Third, we document postnatal short stature and premature greying/loss of hair in POLD1 mutation carriers. We conclude that POLD1 germline mutations can result in a variably expressed and probably underdiagnosed segmental progeroid syndrome.


Subject(s)
Cockayne Syndrome/diagnosis , Cockayne Syndrome/genetics , DNA Polymerase III/genetics , Germ-Line Mutation , Werner Syndrome/diagnosis , Adolescent , Adult , Alleles , Amino Acid Substitution , Cell Line, Transformed , Child , Chromosomal Instability , Chromosome Aberrations , DNA Mutational Analysis , DNA Polymerase III/chemistry , Diagnosis, Differential , Facies , Female , Genotype , Humans , Male , Middle Aged , Models, Molecular , Phenotype , Protein Conformation , Registries , Young Adult
10.
Am J Med Genet A ; 167A(12): 3006-10, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26334766

ABSTRACT

Mutations of the histone acetyltransferase-encoding KAT6B gene cause the Say-Barber-Biesecker/Young-Simpson (SBBYS) type of blepharophimosis-"mental retardation" syndromes and the more severe genitopatellar syndrome. The SBBYS syndrome-causing mutations are clustered in the large exon 18 of KAT6B and almost exclusively lead to predicted protein truncation. An atypical KAT6B mutation, a de novo synonymous variant located in exon 16 (c.3147G>A, p.(Pro1049Pro)) was previously identified in three unrelated patients. This exonic mutation was predicted in silico to cause protein truncation through aberrant splicing. Here, we report three additional unrelated children with typical SBBYS syndrome and the KAT6B c.3147G>A mutation. We show on RNA derived from patient blood that the mutation indeed induces aberrant splicing through the use of a cryptic exonic splice acceptor site created by the sequence variant. Our results thus identify the synonymous variant c.3147G>A as a splice site mutation and a mutational hot spot in SBBYS syndrome.


Subject(s)
Blepharophimosis/genetics , Congenital Hypothyroidism/genetics , Heart Defects, Congenital/genetics , Histone Acetyltransferases/genetics , Intellectual Disability/genetics , Joint Instability/genetics , Mutation , Blepharophimosis/etiology , Child , Child, Preschool , Congenital Hypothyroidism/etiology , Facies , Female , Heart Defects, Congenital/etiology , Humans , Intellectual Disability/etiology , Joint Instability/etiology , Male , Pregnancy , RNA Splice Sites , RNA Splicing
11.
Am J Med Genet A ; 167A(12): 2985-91, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26289989

ABSTRACT

Craniosynostosis, the premature closure of cranial sutures, is a common craniofacial disorder with heterogeneous etiology and appearance. The purpose of this study was to investigate the clinical and molecular characteristics of craniosynostoses in Hungary, including the classification of patients and the genetic analysis of the syndromic forms. Between 2006 and 2012, 200 patients with craniosynostosis were studied. Classification was based on the suture(s) involved and the associated clinical features. In syndromic cases, genetic analyses, including mutational screening of the hotspot regions of the FGFR1, FGFR2, FGFR3, and TWIST1 genes, karyotyping and FISH study of TWIST1, were performed. The majority (88%) of all patients with craniosynostosis were nonsyndromic. The sagittal suture was most commonly involved, followed by the coronal, metopic, and lambdoid sutures. Male, twin gestation, and very low birth weight were risk factors for craniosynostosis. Syndromic craniosynostosis was detected in 24 patients. In 17 of these patients, Apert, Crouzon, Pfeiffer, Muenke, or Saethre-Chotzen syndromes were identified. In one patient, multiple-suture craniosynostosis was associated with achondroplasia. Clinical signs were not typical for any particular syndrome in six patients. Genetic abnormalities were detected in 18 syndromic patients and in 8 relatives. In addition to 10 different, known mutations in FGFR1,FGFR2 or FGFR3, one novel missense mutation, c.528C>G(p.Ser176Arg), was detected in the TWIST1 gene of a patient with Saethre-Chotzen syndrome. Our results indicate that detailed clinical assessment is of paramount importance in the classification of patients and allows indication of targeted molecular testing with the highest possible diagnostic yield.


Subject(s)
Craniosynostoses/etiology , Mutation , Acrocephalosyndactylia/genetics , Adolescent , Adult , Child , Child, Preschool , Craniosynostoses/genetics , Female , Humans , Hungary , Infant , Male , Nuclear Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Risk Factors , Twist-Related Protein 1/genetics
12.
Hum Mutat ; 35(4): 462-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24415674

ABSTRACT

Guanidinoacetate methyltransferase deficiency (GAMT-D) is an autosomal recessively inherited disorder of creatine biosynthesis. Creatine deficiency on cranial proton magnetic resonance spectroscopy, and elevated guanidinoacetate levels in body fluids are the biomarkers of GAMT-D. In 74 patients, 50 different mutations in the GAMT gene have been identified with missense variants being the most common. Clinical and biochemical features of the patients with missense variants were obtained from their physicians using a questionnaire. In 20 patients, 17 missense variants, 25% had a severe, 55% a moderate, and 20% a mild phenotype. The effect of these variants on GAMT enzyme activity was overexpressed using primary GAMT-D fibroblasts: 17 variants retained no significant activity and are therefore considered pathogenic. Two additional variants, c.22C>A (p.Pro8Thr) and c.79T>C (p.Tyr27His) (the latter detected in control cohorts) are in fact not pathogenic as these alleles restored GAMT enzyme activity, although both were predicted to be possibly damaging by in silico analysis. We report 13 new patients with GAMT-D, six novel mutations and functional analysis of 19 missense variants, all being included in our public LOVD database. Our functional assay is important for the confirmation of the pathogenicity of identified missense variants in the GAMT gene.


Subject(s)
Guanidinoacetate N-Methyltransferase/deficiency , Language Development Disorders/genetics , Language Development Disorders/pathology , Movement Disorders/congenital , Adolescent , Adult , Child , Child, Preschool , Female , Fibroblasts/enzymology , Genetic Predisposition to Disease , Genetic Variation , Guanidinoacetate N-Methyltransferase/genetics , Guanidinoacetate N-Methyltransferase/metabolism , Humans , Male , Movement Disorders/genetics , Movement Disorders/pathology , Mutation, Missense , Surveys and Questionnaires , Young Adult
13.
Am J Med Genet A ; 164A(12): 3176-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25251565

ABSTRACT

Pfeiffer syndrome is an autosomal dominant disorder classically characterized by craniosynostosis, facial dysmorphism and limb anomalies. The majority of cases are caused by mutations in the fibroblast growth factor receptor 2 (FGFR2) gene. A specific, rare mutation p.Pro252Arg, located between the second and third extracellular immunoglobulin-like domain of FGFR1, is associated with mild clinical signs. We report on a three-generation family with five members having a heterozygous FGFR1 p.Pro252Arg mutation. Phenotypic features within the family showed high variability from the apparently normal skull and limbs to the characteristic brachycephaly and digital anomalies. The typical features of Pfeiffer syndrome appeared only in the third generation allowing us to unveil the syndrome in several further family members in two previous generations. Variable expressivity can complicate the recognition of Pfeiffer syndrome, principally the mild type 1, requiring careful phenotyping and genetic counseling.


Subject(s)
Acrocephalosyndactylia/genetics , Acrocephalosyndactylia/pathology , Mutation, Missense/genetics , Phenotype , Receptor, Fibroblast Growth Factor, Type 1/genetics , Female , Genes, Dominant/genetics , Humans , Hungary , Pedigree
14.
Orv Hetil ; 155(9): 348-57, 2014 Mar 02.
Article in Hungarian | MEDLINE | ID: mdl-24566699

ABSTRACT

INTRODUCTION: 80% of rare diseases have a genetic origin, and 50% manifest themselves as congenital anomalies. Their adequate health care includes early recognition of genetic anomalies and prevention of recurrence. AIM: The aims of the authors were to provide correct diagnoses to patients with multiple congenital anomalies with or without mental retardation attending to the outpatient clinic of the Clinical Genetics Center at the University of Debrecen in the time interval between August 1, 2007 and March 31, 2013, establish the possibility of prenatal diagnosis, assess the distribution of different genetic mechanisms in the background of rare genetic diseases, compare them with international data, and develop an algorithm for the diagnostic approach of rare genetic diseases applicable in Hungary. METHOD: Clinical data and genetic results of patients were evaluated, and patients were categorized into one of the ten proposed etiological groups, based on which the distribution of genetic causes was defined. RESULTS: Clinical diagnosis was achieved in 64.3% of patients, confirmed genetic diagnosis in 37.8%, while 35.7% of patients remained undiagnosed. Several dysmorphic syndromes and metabolic disorders were first diagnosed in Hungary, two of which unique in the literature. CONCLUSIONS: In the centre of the authors the diagnostic effectiveness of chromosome aberrations exceeds the international standards, that of known microdeletions and dysmorphic syndromes meets international data, and the genetic diagnosis of mendelian disorders and submicroscopic copy number changes remain below international figures.


Subject(s)
Chromosome Aberrations , Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Prenatal Diagnosis , Rare Diseases/diagnosis , Rare Diseases/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Adult , Aged , Chromosome Disorders/epidemiology , Female , Humans , Hungary/epidemiology , Male , Middle Aged , Rare Diseases/epidemiology
15.
Orv Hetil ; 155(9): 362-4, 2014 Mar 02.
Article in Hungarian | MEDLINE | ID: mdl-24566701

ABSTRACT

The association of short stature, microcephaly, congenital cardiac anomaly and intellectual deficit should always raise the suspicion of chromosomal etiology. If G-banded karyotyping fails to detect large chromosomal aberrations, array comparative genomic hybridization (array CGH) should be performed to screen for submicroscopic pathological copy number changes. The authors present a six-year-old girl whose symptoms arose from a 4.1 Mb loss in the 15q26.2-26.3 telomeric region. The syndrome is characterized by a resistance to the insulin-like growth factor 1 - in our case the increased level of the insulin-like growth factor 1 together with the persistent longitudinal growth failure was an important finding and differential diagnostic feature. A brief overview of the literature is provided.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 15/genetics , Gene Deletion , Growth Disorders/genetics , Heart Defects, Congenital/genetics , Insulin-Like Growth Factor I/metabolism , Microcephaly/genetics , Child , Comparative Genomic Hybridization , Craniofacial Abnormalities/genetics , Diagnosis, Differential , Dwarfism/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Magnetic Resonance Imaging , Syndrome
16.
HGG Adv ; 5(3): 100289, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38571311

ABSTRACT

Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder caused by pathogenic variants in TCF4, leading to intellectual disability, specific morphological features, and autonomic nervous system dysfunction. Epigenetic dysregulation has been implicated in PTHS, prompting the investigation of a DNA methylation (DNAm) "episignature" specific to PTHS for diagnostic purposes and variant reclassification and functional insights into the molecular pathophysiology of this disorder. A cohort of 67 individuals with genetically confirmed PTHS and three individuals with intellectual disability and a variant of uncertain significance (VUS) in TCF4 were studied. The DNAm episignature was developed with an Infinium Methylation EPIC BeadChip array analysis using peripheral blood cells. Support vector machine (SVM) modeling and clustering methods were employed to generate a DNAm classifier for PTHS. Validation was extended to an additional cohort of 11 individuals with PTHS. The episignature was assessed in relation to other neurodevelopmental disorders and its specificity was examined. A specific DNAm episignature for PTHS was established. The classifier exhibited high sensitivity for TCF4 haploinsufficiency and missense variants in the basic-helix-loop-helix domain. Notably, seven individuals with TCF4 variants exhibited negative episignatures, suggesting complexities related to mosaicism, genetic factors, and environmental influences. The episignature displayed degrees of overlap with other related disorders and biological pathways. This study defines a DNAm episignature for TCF4-related PTHS, enabling improved diagnostic accuracy and VUS reclassification. The finding that some cases scored negatively underscores the potential for multiple or nested episignatures and emphasizes the need for continued investigation to enhance specificity and coverage across PTHS-related variants.

17.
Am J Med Genet A ; 161A(4): 884-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23436491

ABSTRACT

The Say-Barber/Biesecker/Young-Simpson (SBBYS) type of the blepharophimosis-mental retardation syndrome group (Ohdo-like syndromes) is a multiple congenital malformation syndrome characterized by vertical narrowing and shortening of the palpebral fissures, ptosis, intellectual disability, hypothyroidism, hearing impairment, and dental anomalies. Mutations of the gene encoding the histone-acetyltransferase KAT6B have been recently identified in individuals affected by SBBYS syndrome. SBBYS syndrome-causing KAT6B mutations cluster in a ~1,700 basepair region in the 3' part of the large exon 18, while mutations located in the 5' region of the same exon have recently been identified to cause the genitopatellar syndrome (GPS), a clinically distinct although partially overlapping malformation-intellectual disability syndrome. Here, we present two children with clinical features of SBBYS syndrome and de novo truncating KAT6B mutations, including a boy who was diagnosed at the age of 4 months. Our results confirm the implication of KAT6B mutations in typical SBBYS syndrome and emphasize the importance of genotype-phenotype correlations at the KAT6B locus where mutations truncating the KAT6B protein at the amino-acid positions ~1,350-1,920 cause SBBYS syndrome.


Subject(s)
Blepharophimosis/genetics , Congenital Hypothyroidism/genetics , Heart Defects, Congenital/genetics , Histone Acetyltransferases/genetics , Intellectual Disability/genetics , Joint Instability/genetics , Mutation , Abnormalities, Multiple , Base Sequence , Blepharophimosis/diagnosis , Child, Preschool , Comparative Genomic Hybridization , Congenital Hypothyroidism/diagnosis , Exons , Facies , Female , Genetic Association Studies , Heart Defects, Congenital/diagnosis , Heterozygote , Humans , Infant , Intellectual Disability/diagnosis , Joint Instability/diagnosis , Karyotype , Male , Phenotype
18.
Article in English | MEDLINE | ID: mdl-34654706

ABSTRACT

The MED13L-related intellectual disability or MRFACD syndrome (Mental retardation and distinctive facial features with or without cardiac defects; MIM # 616789) is one of the most common forms of syndromic intellectual disability with about a hundred cases reported so far. Affected individuals share overlapping features comprising intellectual disability, hypotonia, motor delay, remarkable speech delay, and a recognizable facial gestalt. De novo disruption of the MED13L gene by deletions, duplications, or sequence variants has been identified as deleterious. Siblings affected by intragenic deletion transmitted from a mosaic parent have been reported once in the literature. We now present the first case of paternal germinal mosaicism for a missense MED13L variant causing MRFACD syndrome in one of the father's children and being the likely cause of intellectual disability and facial dysmorphism in the other. As part of the Mediator complex, the MED proteins have an essential role in regulating transcription. Thirty-two subunits of the Mediator complex genes have been linked to congenital malformations that are now acknowledged as transcriptomopathies. The MRFACD syndrome has been suggested to represent a recognizable phenotype.


Subject(s)
Intellectual Disability , Mediator Complex , Mosaicism , Fathers , Humans , Intellectual Disability/genetics , Male , Mediator Complex/genetics , Mutation, Missense , Paternal Inheritance , Phenotype
19.
Genes (Basel) ; 13(12)2022 12 15.
Article in English | MEDLINE | ID: mdl-36553633

ABSTRACT

The ADNP-gene-related neurodevelopmental disorder Helsmoortel-Van der Aa syndrome is a rare syndromic-intellectual disability-an autism spectrum disorder first described by Helsmoortel and Van der Aa in 2014. Recently, a large cohort including 78 patients and their detailed phenotypes were presented by Van Dijck et al., 2019, who reported developmental delay, speech delay and autism spectrum disorder as nearly constant findings with or without variable cardiological, gastroenterological, urogenital, endocrine and neurological manifestations. Among cardiac malformations, atrial septal defect, patent ductus arteriosus, patent foramen ovale and mitral valve prolapse were the most common findings, but other unspecified defects, such as mild pulmonary valve stenosis, were also described. We present two patients with pathogenic ADNP variants and unusual cardiothoracic manifestations-Bland-White-Garland syndrome, pectus carinatum superiorly along the costochondral junctions and pectus excavatum inferiorly in one patient, and Kawasaki syndrome with pericardiac effusion, coronary artery dilatation and aneurysm in the other-who were successfully treated with intravenous immunoglobulin, corticosteroid and aspirin. Both patients had ectodermal and/or skeletal features overlapping those seen in RASopathies, supporting the observations of Alkhunaizi et al. 2018. on the clinical overlap between Helsmoortel-Van der Aa syndrome and Noonan syndrome. We observed a morphological overlap with the Noonan-like disorder with anagen hair in our patients.


Subject(s)
Abnormalities, Multiple , Autism Spectrum Disorder , Intellectual Disability , Humans , Intellectual Disability/genetics , Autism Spectrum Disorder/genetics , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Mutation , Abnormalities, Multiple/genetics , Phenotype
20.
Neurogenetics ; 12(4): 273-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21965147

ABSTRACT

Ataxia telangiectasia (AT) is an autosomal recessive disorder characterized by cerebellar degeneration, immunodeficiency, oculocutaneous telangiectasias, chromosomal instability, radiosensitivity, and cancer predisposition. The gene mutated in the patients, ATM, encodes a member of the phosphatidylinositol 3-kinase family proteins. The ATM protein has a key role in the cellular response to DNA damage. Truncating and splice site mutations in ATM have been found in most patients with the classical AT phenotype. Here we report of our extensive ATM mutation screening on 25 AT patients from 19 families of different ethnic origin. Previously unknown mutations were identified in six patients including a new homozygous missense mutation, c.8110T>C (p.Cys2704Arg), in a severely affected patient. Comprehensive clinical data are presented for all patients described here along with data on ATM function generated by analysis of cell lines established from a subset of the patients.


Subject(s)
Ataxia Telangiectasia/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Child , Child, Preschool , DNA Mutational Analysis , DNA-Binding Proteins/metabolism , Female , Haplotypes , Humans , Male , Phenotype , Protein Serine-Threonine Kinases/metabolism , RNA Splicing , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL