Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 108(9): 1725-1734, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34433009

RESUMEN

Copy-number variations (CNVs) are a common cause of congenital limb malformations and are interpreted primarily on the basis of their effect on gene dosage. However, recent studies show that CNVs also influence the 3D genome chromatin organization. The functional interpretation of whether a phenotype is the result of gene dosage or a regulatory position effect remains challenging. Here, we report on two unrelated families with individuals affected by bilateral hypoplasia of the femoral bones, both harboring de novo duplications on chromosome 10q24.32. The ∼0.5 Mb duplications include FGF8, a key regulator of limb development and several limb enhancer elements. To functionally characterize these variants, we analyzed the local chromatin architecture in the affected individuals' cells and re-engineered the duplications in mice by using CRISPR-Cas9 genome editing. We found that the duplications were associated with ectopic chromatin contacts and increased FGF8 expression. Transgenic mice carrying the heterozygous tandem duplication including Fgf8 exhibited proximal shortening of the limbs, resembling the human phenotype. To evaluate whether the phenotype was a result of gene dosage, we generated another transgenic mice line, carrying the duplication on one allele and a concurrent Fgf8 deletion on the other allele, as a control. Surprisingly, the same malformations were observed. Capture Hi-C experiments revealed ectopic interaction with the duplicated region and Fgf8, indicating a position effect. In summary, we show that duplications at the FGF8 locus are associated with femoral hypoplasia and that the phenotype is most likely the result of position effects altering FGF8 expression rather than gene dosage effects.


Asunto(s)
Duplicación Cromosómica , Cromosomas Humanos Par 10/química , Variaciones en el Número de Copia de ADN , Factor 8 de Crecimiento de Fibroblastos/genética , Deformidades Congénitas de las Extremidades Inferiores/genética , Adolescente , Alelos , Animales , Sistemas CRISPR-Cas , Preescolar , Cromatina/química , Cromatina/metabolismo , Cromosomas Humanos Par 10/metabolismo , Elementos de Facilitación Genéticos , Familia , Femenino , Fémur/anomalías , Fémur/diagnóstico por imagen , Fémur/metabolismo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Edición Génica , Heterocigoto , Humanos , Lactante , Deformidades Congénitas de las Extremidades Inferiores/diagnóstico por imagen , Deformidades Congénitas de las Extremidades Inferiores/metabolismo , Deformidades Congénitas de las Extremidades Inferiores/patología , Masculino , Ratones , Ratones Transgénicos , Linaje , Fenotipo
2.
Epilepsy Behav ; 150: 109535, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38118233

RESUMEN

AIM: To evaluate the effectiveness of the ketogenic diet treatment in a cohort of patients with drug-resistant epilepsy with a mutation in the DEPDC5 gene. MATERIALS AND METHODS: We followed four paediatric patients with drug resistant DEPDC5-related epilepsy through a ketogenic diet (KD) treatment course. We analyzed the following parameters of their clinical profiles: past medical history, clinical characteristics of seizure morphology, EEG records pre- and post-KD treatment, the results of MRI head and neurological and psychological examinations (pre-treatment and throughout treatment course). We evaluated the effectiveness of previous therapeutic approaches and the current treatment with ketogenic diet alongside results of neuroimaging studies. Effect of KD on co-morbid behavioural and psychiatric symptoms, as well as adverse effects from KD were also assessed. RESULTS: In three patients, the introduction of the ketogenic diet resulted in the cessation of seizures, while in 1 patient with co-morbid cortical dysplasia, epileptic seizures of lesser severity returned after an initial seizure-free period of several weeks. Further, 1 patient was able to transition to a KD-only treatment regimen. The remaining patients were able to reduce the number of antiseizure medicine (ASM) to a monotherapy. In all cases we observed improvements in EEG results. Our cohort included one patient whose MRI head showed cortical dysplasia. However, no patients demonstrated any neurological signs in neurological examination. Psychological examination showed normal intellectual development in all patients, although behavioral disorders and difficulties at school were observed. The introduction of KD treatment correlated with improvement in school performance and improved behavioral regulation. No clinically significant adverse events were observed. CONCLUSIONS: KD seems to be both effective and well tolerated in young patients with DEPDC5-related epilepsy, both as a monotherapy and as an adjunct to ASM. We recommend an early adoption of this therapeutic approach in this patient demographic. Our results demonstrate that the positive effects of KD treatment encompass improvements in general functioning, particularly in the context of school performance and behavior, in addition to the achievement of good seizure control.


Asunto(s)
Dieta Cetogénica , Epilepsia Refractaria , Epilepsia , Malformaciones del Desarrollo Cortical , Niño , Humanos , Dieta Cetogénica/métodos , Resultado del Tratamiento , Estudios Retrospectivos , Convulsiones
3.
Pediatr Nephrol ; 39(6): 1847-1858, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38196016

RESUMEN

BACKGROUND: We aimed to develop a tool for predicting HNF1B mutations in children with congenital abnormalities of the kidneys and urinary tract (CAKUT). METHODS: The clinical and laboratory data from 234 children and young adults with known HNF1B mutation status were collected and analyzed retrospectively. All subjects were randomly divided into a training (70%) and a validation set (30%). A random forest model was constructed to predict HNF1B mutations. The recursive feature elimination algorithm was used for feature selection for the model, and receiver operating characteristic curve statistics was used to verify its predictive effect. RESULTS: A total of 213 patients were analyzed, including HNF1B-positive (mut + , n = 109) and HNF1B-negative (mut - , n = 104) subjects. The majority of patients had mild chronic kidney disease. Kidney phenotype was similar between groups, but bilateral kidney anomalies were more frequent in the mut + group. Hypomagnesemia and hypermagnesuria were the most common abnormalities in mut + patients and were highly selective of HNF1B. Hypomagnesemia based on age-appropriate norms had a better discriminatory value than the age-independent cutoff of 0.7 mmol/l. Pancreatic anomalies were almost exclusively found in mut + patients. No subjects had hypokalemia; the mean serum potassium level was lower in the HNF1B cohort. The abovementioned, discriminative parameters were selected for the model, which showed a good performance (area under the curve: 0.85; sensitivity of 93.67%, specificity of 73.57%). A corresponding calculator was developed for use and validation. CONCLUSIONS: This study developed a simple tool for predicting HNF1B mutations in children and young adults with CAKUT.


Asunto(s)
Enfermedades Renales , Sistema Urinario , Anomalías Urogenitales , Reflujo Vesicoureteral , Niño , Humanos , Adulto Joven , Estudios Retrospectivos , Riñón/anomalías , Sistema Urinario/anomalías , Mutación , Enfermedades Renales/genética , Magnesio , Factor Nuclear 1-beta del Hepatocito/genética
4.
Genet Med ; 25(11): 100928, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37427568

RESUMEN

PURPOSE: HOXD13 is an important regulator of limb development. Pathogenic variants in HOXD13 cause synpolydactyly type 1 (SPD1). How different types and positions of HOXD13 variants contribute to genotype-phenotype correlations, penetrance, and expressivity of SPD1 remains elusive. Here, we present a novel cohort and a literature review to elucidate HOXD13 phenotype-genotype correlations. METHODS: Patients with limb anomalies suggestive of SPD1 were selected for analysis of HOXD13 by Sanger sequencing, repeat length analysis, and next-generation sequencing. Literature was reviewed for HOXD13 heterozygotes. Variants were annotated for phenotypic data. Severity was calculated, and cluster and decision-tree analyses were performed. RESULTS: We identified 98 affected members of 38 families featuring 11 different (likely) causative variants and 4 variants of uncertain significance. The most frequent (25/38) were alanine repeat expansions. Phenotypes ranged from unaffected heterozygotes to severe osseous synpolydactyly, with intra- and inter-familial heterogeneity and asymmetry. A literature review provided 160 evaluable affected members of 49 families with SPD1. Computer-aided analysis only corroborated a positive correlation between alanine repeat length and phenotype severity. CONCLUSION: Our findings support that HOXD13-protein condensation in addition to haploinsufficiency is the molecular pathomechanism of SPD1. Our data may, also, facilitate the interpretation of synpolydactyly radiographs by future automated tools.


Asunto(s)
Proteínas de Homeodominio , Sindactilia , Humanos , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Sindactilia/genética , Genotipo , Fenotipo , Linaje , Alanina/genética , Mutación
5.
Am J Med Genet A ; 191(1): 205-219, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36317839

RESUMEN

Many unbalanced large copy number variants reviewed in the paper are associated with syndromic orofacial clefts, including a 1.6 Mb deletion on chromosome 3q29. The current report presents a new family with this recurrent deletion identified via whole-exome sequencing and confirmed by array comparative genomic hybridization. The proband exhibited a more severe clinical phenotype than his affected mother, comprising right-sided cleft lip/alveolus and cleft palate, advanced dental caries, heart defect, hypospadias, psychomotor, and speech delay, and an intellectual disability. Data analysis from the 3q29 registry revealed that the 3q29 deletion increases the risk of clefting by nearly 30-fold. No additional rare and pathogenic nucleotide variants were identified that could explain the clefting phenotype and observed intrafamilial phenotypic heterogeneity. These data suggest that the 3q29 deletion may be the primary risk factor for clefting, with additional genomic variants located outside the coding sequences, methylation changes, or environmental exposure serving as modifiers of this risk. Additional studies, including whole-genome sequencing or methylation analyses, should be performed to identify genetic factors underlying the phenotypic variation associated with the recurrent 3q29 deletion.


Asunto(s)
Labio Leporino , Fisura del Paladar , Caries Dental , Masculino , Humanos , Labio Leporino/diagnóstico , Labio Leporino/genética , Fisura del Paladar/diagnóstico , Fisura del Paladar/genética , Secuenciación del Exoma , Hibridación Genómica Comparativa , Síndrome
6.
J Med Genet ; 59(3): 209-219, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34782442

RESUMEN

Clubfoot (talipes equinovarus) is a congenital malformation affecting muscles, bones, connective tissue and vascular or neurological structures in limbs. It has a complex aetiology, both genetic and environmental. To date, the most important findings in clubfoot genetics involve PITX1 variants, which were linked to clubfoot phenotype in mice and humans. Additionally, copy number variations encompassing TBX4 or single nucleotide variants in HOXC11, the molecular targets of the PITX1 transcription factor, were linked to the clubfoot phenotype. In general, genes of cytoskeleton and muscle contractile apparatus, as well as components of the extracellular matrix and connective tissue, are frequently linked with clubfoot aetiology. Last but not least, an equally important element, that brings us closer to a better understanding of the clubfoot genotype/phenotype correlation, are studies on the two known animal models of clubfoot-the pma or EphA4 mice. This review will summarise the current state of knowledge of the molecular basis of this congenital malformation.


Asunto(s)
Pie Equinovaro , Animales , Pie Equinovaro/genética , Variaciones en el Número de Copia de ADN , Estudios de Asociación Genética , Proteínas de Homeodominio/genética , Humanos , Ratones , Fenotipo , Factores de Transcripción/genética
7.
Mol Genet Genomics ; 297(5): 1343-1352, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35821352

RESUMEN

Herein, we report on a large Polish family presenting with a classical triphalangeal thumb-polysyndactyly syndrome (TPT-PS). This rare congenital limb anomaly is generally caused by microduplications encompassing the Sonic Hedgehog (SHH) limb enhancer, termed the zone of polarizing activity (ZPA) regulatory sequence (ZRS). Recently, a pathogenic variant in the pre-ZRS (pZRS), a conserved sequence located near the ZRS, has been described in a TPT-PS Dutch family. We performed targeted ZRS sequencing, array comparative genomic hybridization, and whole-exome sequencing. Next, we sequenced the recently described pZRS region. Finally, we performed a circular chromatin conformation capture-sequencing (4C-seq) assay on skin fibroblasts of one affected family member and control samples to examine potential alterations in the SHH regulatory domain and functionally characterize the identified variant. We found that all affected individuals shared a recently identified pathogenic point mutation in the pZRS region: NC_000007.14:g.156792782C>G (GRCh38/hg38), which is the same as in the Dutch family. The results of 4C-seq experiments revealed increased interactions within the whole SHH regulatory domain (SHH-LMBR1 TAD) in the patient compared to controls. Our study expands the number of TPT-PS families carrying a pathogenic alteration of the pZRS and underlines the importance of routine pZRS sequencing in the genetic diagnostics of patients with TPT-PS or similar phenotypes. The pathogenic mutation causative for TPT-PS in our patient gave rise to increased interactions within the SHH regulatory domain in yet unknown mechanism.


Asunto(s)
Anomalías Congénitas , Proteínas Hedgehog , Disostosis Mandibulofacial , Polidactilia , Hibridación Genómica Comparativa , Anomalías Congénitas/genética , Elementos de Facilitación Genéticos , Proteínas Hedgehog/genética , Humanos , Disostosis Mandibulofacial/genética , Mutación , Linaje , Pulgar
8.
Am J Med Genet A ; 188(2): 642-647, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34773354

RESUMEN

Aniridia is usually an autosomal dominant, rare disorder characterized by a variable degree of hypoplasia or the absence of iris tissue, with additional ocular abnormalities. Pathogenic variants in the PAX6 gene are associated with aniridia in most patients. However, in up to 30% of individuals, disease results from 11p13 chromosomal rearrangements. Here we present a patient with a clinical diagnosis of partial aniridia born to consanguineous Polish parents. The parents were asymptomatic and ophthalmologically normal. We performed PAX6 sequencing, array comparative genomic hybridization, quantitative real-time PCR, and whole genome sequencing. aCGH revealed a homozygous deletion of the DCDC1 gene fragment in the patient. The same, but heterozygous deletion, was detected in each of the patient's asymptomatic parents and brother. In the presented family, the signs and symptoms of aniridia are observed only in the homozygous proband. Whole genome sequencing analysis was performed to determine other possible causes of the disease and did not detect any additional or alternative potentially pathogenic variant. We report a novel homozygous deletion located in the 11p13 region, which does not include the PAX6 gene or any known PAX6 enhancers. To our best knowledge, this is the first reported case of a patient presented with isolated aniridia carrying a homozygous microdeletion downstream of the PAX6 gene.


Asunto(s)
Aniridia , Proteínas del Ojo , Aniridia/diagnóstico , Aniridia/genética , Hibridación Genómica Comparativa , Proteínas del Ojo/genética , Proteínas de Homeodominio/genética , Homocigoto , Humanos , Masculino , Factor de Transcripción PAX6/genética , Linaje , Eliminación de Secuencia
9.
Hum Genet ; 140(8): 1229-1239, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34159400

RESUMEN

The extensive clinical and genetic heterogeneity of congenital limb malformation calls for comprehensive genome-wide analysis of genetic variation. Genome sequencing (GS) has the potential to identify all genetic variants. Here we aim to determine the diagnostic potential of GS as a comprehensive one-test-for-all strategy in a cohort of undiagnosed patients with congenital limb malformations. We collected 69 cases (64 trios, 1 duo, 5 singletons) with congenital limb malformations with no molecular diagnosis after standard clinical genetic testing and performed genome sequencing. We also developed a framework to identify potential noncoding pathogenic variants. We identified likely pathogenic/disease-associated variants in 12 cases (17.4%) including four in known disease genes, and one repeat expansion in HOXD13. In three unrelated cases with ectrodactyly, we identified likely pathogenic variants in UBA2, establishing it as a novel disease gene. In addition, we found two complex structural variants (3%). We also identified likely causative variants in three novel high confidence candidate genes. We were not able to identify any noncoding variants. GS is a powerful strategy to identify all types of genomic variants associated with congenital limb malformation, including repeat expansions and complex structural variants missed by standard diagnostic approaches. In this cohort, no causative noncoding SNVs could be identified.


Asunto(s)
Heterogeneidad Genética , Proteínas de Homeodominio/genética , Deformidades Congénitas de las Extremidades/genética , Mutación , Factores de Transcripción/genética , Enzimas Activadoras de Ubiquitina/genética , Secuencia de Bases , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , Expresión Génica , Pruebas Genéticas , Humanos , Lactante , Deformidades Congénitas de las Extremidades/metabolismo , Deformidades Congénitas de las Extremidades/patología , Masculino , Linaje , Factores de Transcripción/deficiencia , Enzimas Activadoras de Ubiquitina/deficiencia , Secuenciación Completa del Genoma
10.
Clin Genet ; 96(5): 429-438, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31347140

RESUMEN

RUNX2 (Runt-related transcription factor 2) is a master regulator of osteoblast differentiation, cartilage and bone development. Pathogenic variants in RUNX2 have been linked to the Cleidocranial dysplasia (CCD), which is characterized by hypoplasia or aplasia of clavicles, delayed fontanelle closure, and dental anomalies. Here, we report 11 unrelated Polish patients with CCD caused by pathogenic alterations located in the Runt domain of RUNX2. In total, we identified eight different intragenic variants, including seven missense and one splicing mutation. Three of them are novel: c.407T>A p.(Leu136Gln), c.480C>G p.(Asn160Lys), c.659C>G p.(Thr220Arg), additional three were not functionally tested: c.391C>T p.(Arg131Cys), c.580+1G>T p.(Lys195_Arg229del), c.652A>G p.(Lys218Glu), and the remaining two: c.568C>T p.(Arg190Trp), c.673C>T p.(Arg225Trp) were previously reported and characterized. The performed transactivation and localization studies provide evidence of decreased transcriptional activity of RUNX2 due to mutations targeting the Runt domain and prove that impairment of nuclear localization signal (NLS) affects the subcellular localization of the protein. Presented data show that pathogenic variants discovered in our patients have a detrimental effect on RUNX2, triggering the CCD phenotype.


Asunto(s)
Displasia Cleidocraneal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/química , Predisposición Genética a la Enfermedad , Conformación Proteica , Preescolar , Displasia Cleidocraneal/epidemiología , Displasia Cleidocraneal/patología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/ultraestructura , Femenino , Humanos , Lactante , Masculino , Mutación/genética , Mutación Missense/genética , Fenotipo , Polonia/epidemiología , Isoformas de Proteínas/genética , Relación Estructura-Actividad
11.
Am J Med Genet A ; 179(12): 2447-2453, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31512363

RESUMEN

Hartsfield syndrome is a rare clinical entity characterized by holoprosencephaly and ectrodactyly with the variable feature of cleft lip/palate. In addition to these symptoms patients with Hartsfield syndrome can show developmental delay of variable severity, isolated hypogonadotropic hypogonadism, central diabetes insipidus, vertebral anomalies, eye anomalies, and cardiac malformations. Pathogenic variants in FGFR1 have been described to cause phenotypically different FGFR1-related disorders such as Hartsfield syndrome, hypogonadotropic hypogonadism with or without anosmia, Jackson-Weiss syndrome, osteoglophonic dysplasia, Pfeiffer syndrome, and trigonocephaly Type 1. Here, we report three patients with Hartsfield syndrome from two unrelated families. Exome sequencing revealed two siblings harboring a novel de novo heterozygous synonymous variant c.1029G>A, p.Ala343Ala causing a cryptic splice donor site in exon 8 of FGFR1 likely due to gonadal mosaicism in one parent. The third case was a sporadic patient with a novel de novo heterozygous missense variant c.1868A>G, p.(Asp623Gly).


Asunto(s)
Labio Leporino/diagnóstico , Labio Leporino/genética , Fisura del Paladar/diagnóstico , Fisura del Paladar/genética , Dedos/anomalías , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Deformidades Congénitas de la Mano/diagnóstico , Deformidades Congénitas de la Mano/genética , Holoprosencefalia/diagnóstico , Holoprosencefalia/genética , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Mutación Missense , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Mutación Silenciosa , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética/métodos , Humanos , Masculino , Linaje , Fenotipo
12.
Genet Med ; 20(6): 599-607, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29236091

RESUMEN

PurposeCopy-number variants (CNVs) are generally interpreted by linking the effects of gene dosage with phenotypes. The clinical interpretation of noncoding CNVs remains challenging. We investigated the percentage of disease-associated CNVs in patients with congenital limb malformations that affect noncoding cis-regulatory sequences versus genes sensitive to gene dosage effects.MethodsWe applied high-resolution copy-number analysis to 340 unrelated individuals with isolated limb malformation. To investigate novel candidate CNVs, we re-engineered human CNVs in mice using clustered regularly interspaced short palindromic repeats (CRISPR)-based genome editing.ResultsOf the individuals studied, 10% harbored CNVs segregating with the phenotype in the affected families. We identified 31 CNVs previously associated with congenital limb malformations and four novel candidate CNVs. Most of the disease-associated CNVs (57%) affected the noncoding cis-regulatory genome, while only 43% included a known disease gene and were likely to result from gene dosage effects. In transgenic mice harboring four novel candidate CNVs, we observed altered gene expression in all cases, indicating that the CNVs had a regulatory effect either by changing the enhancer dosage or altering the topological associating domain architecture of the genome.ConclusionOur findings suggest that CNVs affecting noncoding regulatory elements are a major cause of congenital limb malformations.


Asunto(s)
ADN Intergénico/genética , Deformidades Congénitas de las Extremidades/genética , Animales , Variaciones en el Número de Copia de ADN/genética , Femenino , Dosificación de Gen/genética , Genoma Humano , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Ratones , Ratones Transgénicos , Linaje , Fenotipo
13.
J Hum Genet ; 61(7): 577-83, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27030147

RESUMEN

Desbuquois dysplasia type 2 (DBQD2) is a rare recessively inherited skeletal genetic disorder characterized by severe prenatal and postnatal growth retardation, generalized joint laxity with dislocation of large joints and facial dysmorphism. The condition was recently described to result from autosomal recessive mutations in XYLT1, encoding the enzyme xylosyltransferase-1. In this paper, we report on a Polish patient with DBQD2 who presented with severe short stature of prenatal onset, joint laxity, psychomotor retardation and multiple radiological abnormalities including short metacarpals, advanced bone age and exaggerated trochanters. Endocrinological examinations revealed that sleep-induced growth hormone (GH) release and GH peak in clonidine- and glucagon-induced provocative tests as well as insulin-like growth factor 1 (IGF-1) and IGF-binding protein-3 levels were all markedly decreased, confirming deficiency of GH secretion. Bone age, unlikely to GH deficiency, was significantly advanced. To establish the diagnosis at a molecular level, we performed whole-exome sequencing and bioinformatic analysis in the index patient, which revealed compound heterozygous XYLT1 mutations: c.595C>T(p.Gln199*) and c.1651C>T(p.Arg551Cys), both of which are novel. Sanger sequencing showed that the former mutation was inherited from the healthy mother, whereas the latter one most probably occurred de novo. Our study describes the first case of DBQD2 resulting from compound heterozygous XYLT1 mutation, expands the mutational spectrum of the disease and provides evidence that the severe growth retardation and microsomia observed in DBQD2 patients may result not only from the skeletal dysplasia itself but also from GH and IGF-1 deficiency.


Asunto(s)
Anomalías Craneofaciales/diagnóstico , Anomalías Craneofaciales/genética , Enanismo Hipofisario/diagnóstico , Enanismo Hipofisario/genética , Enanismo/diagnóstico , Enanismo/genética , Heterocigoto , Inestabilidad de la Articulación/diagnóstico , Inestabilidad de la Articulación/genética , Mutación , Osificación Heterotópica/diagnóstico , Osificación Heterotópica/genética , Pentosiltransferasa/genética , Fenotipo , Polidactilia/diagnóstico , Polidactilia/genética , Adulto , Análisis Mutacional de ADN , Exoma , Femenino , Haplotipos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Repeticiones de Microsatélite , Polonia , Embarazo , Diagnóstico Prenatal , Esqueleto/diagnóstico por imagen , Esqueleto/patología , UDP Xilosa Proteína Xilosiltransferasa
14.
J Hum Genet ; 61(5): 457-61, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26763883

RESUMEN

Brachydactyly refers to shortening of digits due to hypoplasia or aplasia of bones forming the hands and/or feet. Isolated brachydactyly type E (BDE), which is characterized by shortened metacarpals and/or metatarsals, results in a small proportion of patients from HOXD13 or PTHLH mutations, although in the majority of cases molecular lesion remains unknown. BDE, like other brachydactylies, shows clinical heterogeneity with highly variable intrafamilial and interindividual expressivity. In this study, we investigated two Polish cases (one familial and one sporadic) presenting with BDE and additional symptoms due to novel PTHLH mutations. Apart from BDE, the affected family showed short stature, mild craniofacial dysmorphism and delayed bone age. Sanger sequencing of PTHLH revealed a novel heterozygous frameshift mutation c.258delC(p.N87Tfs*18) in two affected individuals and one relative manifesting mild brachydactyly. The sporadic patient, in addition to BDE, presented with craniofacial dysmorphism, normal stature and bone age, and was demonstrated to carry a de novo heterozygous c.166C>T(p.R56*) mutation. Our paper reports on the two novel truncating PTHLH variants, resulting in variable combination of BDE and other symptoms. Data shown here expand the knowledge on the phenotypic presentation of PTHLH mutations, highlighting significant clinical variability and incomplete penetrance of the PTHLH-related symptoms.


Asunto(s)
Braquidactilia/genética , Anomalías Craneofaciales/genética , Enanismo/genética , Heterocigoto , Mutación , Proteína Relacionada con la Hormona Paratiroidea/genética , Fenotipo , Adolescente , Huesos/diagnóstico por imagen , Huesos/patología , Braquidactilia/diagnóstico , Niño , Preescolar , Anomalías Craneofaciales/diagnóstico , Análisis Mutacional de ADN , Enanismo/diagnóstico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje , Síndrome
15.
Ginekol Pol ; 87(10): 706-710, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27958623

RESUMEN

OBJECTIVES: Holt-Oram syndrome manifests with defects of upper limbs, pectoral girdle and cardiovascular system. The aim of this paper was to present complex clinical picture of the syndrome and its variable expression on the example of the family diagnosed genetically on the neonatal ward, after proband's prenatal examination. MARETIAL AND METHODS: Nine family members were tested for TBX5 gene mutation. RESULTS: Four of family members were diagnosed with Holt-Oram syndrome and five had correct genetic test results. The diagnosis allowed to identify a genetic risk family and enabled to provide them with genetic counselling. CONCLUSIONS: Diagnosis of Holt-Oram syndrome is possible as early as in prenatal period and it can be verified by genetic tests.


Asunto(s)
Anomalías Múltiples/genética , Cardiopatías Congénitas/genética , Defectos del Tabique Interatrial/diagnóstico , Defectos del Tabique Interatrial/genética , Deformidades Congénitas de las Extremidades Inferiores/diagnóstico , Deformidades Congénitas de las Extremidades Inferiores/genética , Mutación , Diagnóstico Prenatal , Proteínas de Dominio T Box/genética , Deformidades Congénitas de las Extremidades Superiores/diagnóstico , Deformidades Congénitas de las Extremidades Superiores/genética , Anomalías Múltiples/sangre , Anomalías Múltiples/diagnóstico , Biomarcadores/sangre , Femenino , Asesoramiento Genético , Pruebas Genéticas , Cardiopatías Congénitas/sangre , Cardiopatías Congénitas/diagnóstico , Defectos del Tabique Interatrial/sangre , Humanos , Recién Nacido , Deformidades Congénitas de las Extremidades Inferiores/sangre , Linaje , Polimorfismo Genético , Embarazo , Proteínas de Dominio T Box/sangre , Deformidades Congénitas de las Extremidades Superiores/sangre
16.
Birth Defects Res A Clin Mol Teratol ; 100(10): 764-71, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25196357

RESUMEN

BACKGROUND: Split-hand/foot malformation (SHFM) is a clinically and genetically heterogeneous limb abnormality characterized by the absence or hypoplasia of the central rays of the autopod. SHFM1, which is one out of seven known SHFM loci, maps to 7q21.2-q21.3. SHFM1 is usually inherited as an autosomal dominant trait with reduced penetrance, although recessive inheritance has been described for a single family carrying a homozygous DLX5 missense variant. In most cases, SHFM1 results from heterozygous deletions encompassing DLX5/DLX6 genes or from inversions and translocations separating the genes from their limb specific enhancers. Recently, a single Chinese family with dominant SHFM1 was shown to result from a heterozygous DLX5 missense mutation. METHODS: In this study, we report on four male individuals from two unrelated Polish families (one sporadic and one familial case) presenting with isolated SHFM. We tested both probands for known molecular causes of SHFM, including TP63, WNT10B, DLX5 mutations and copy number changes using 1.4 M array CGH. RESULTS: Sanger sequencing of DLX5 revealed a novel heterozygous nonsense mutation c.G115T(p.E39X) in both index patients. Segregation studies demonstrated that the variant was present in all affected family members but also in three apparently healthy relatives (two females and one male). CONCLUSION: This is the first report of a heterozygous DLX5 nonsense mutation resulting in incompletely penetrant autosomal dominant isolated SHFM1. Data shown here provides further evidence for the contribution of DLX5 point mutations to the development of ectrodactyly and suggest the possibility of sex-related segregation distortion with an excess of affected males.


Asunto(s)
Proteínas de Homeodominio/genética , Deformidades Congénitas de las Extremidades/genética , Mutación Missense/genética , Factores de Transcripción/genética , Secuencia de Bases , Hibridación Genómica Comparativa , Genes Dominantes , Heterocigoto , Humanos , Masculino , Datos de Secuencia Molecular , Polonia , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN
17.
Birth Defects Res A Clin Mol Teratol ; 100(4): 314-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24706454

RESUMEN

BACKGROUND: Metacarpal 4-5 fusion (MF4; MIM#309630) is a rare congenital malformation of the hand characterized by the partial or complete fusion of the fourth and fifth metacarpals. The anomaly occurs as an isolated trait or part of a genetic syndrome. Recently, we have identified FGF16 nonsense mutations as the underlying cause of isolated X-linked recessive MF4. METHODS: In this report, we provide a detailed clinical description of a sporadic male patient showing MF4 in whom we performed Sanger sequencing of the entire coding sequence of FGF16. RESULTS: In addition to MF4 symptoms, the patient presented with generalized joint laxity and hypermobility. FGF16 sequencing detected a novel truncating mutation (c.474_477del; p.E158DfsX25) in exon 3 of the gene. A heterozygous mutation was found in a clinically and radiologically unaffected mother of the proband. CONCLUSION: Our finding confirms that truncating mutations of FGF16 are causative for X-linked recessive metacarpal 4-5 fusion. Importantly, the mutation detected in this study was located in last exon of the gene (exon 3), like the only two FGF16 disease-causing variants identified to date. Thus, all FGF16 mutations known to give rise to this rare skeletal hand malformation are C-terminal and most probably do not result in a nonsense mediated decay. Additionally, our proband showed mild symptoms of a connective tissue disorder, as some other patients previously reported to have X-linked MF4. Therefore, we suggest that impaired FGF16 function may also be responsible for connective tissue symptoms in MF4 patients.


Asunto(s)
Factores de Crecimiento de Fibroblastos/genética , Genes Recesivos , Genes Ligados a X , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Deformidades Congénitas de la Mano/genética , Mutación INDEL , Huesos del Metacarpo/anomalías , Adulto , Exones/genética , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Deformidades Congénitas de la Mano/patología , Humanos , Masculino , Huesos del Metacarpo/patología
18.
J Med Genet ; 50(9): 579-84, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23709756

RESUMEN

BACKGROUND: Metacarpal 4-5 fusion (MF4; MIM %309630) is a rare congenital malformation of the hand characterised by the partial or complete fusion of the fourth and fifth metacarpals. The anomaly occurs as an isolated trait or part of a genetic syndrome. METHODS: To search for disease-causing mutation, whole exome sequencing (WES) was performed on samples from a single trio. Before WES, molecular screening including gene sequencing and array comparative genomic hybridisation was applied. Validation of WES and segregation studies were done using routine Sanger sequencing. RESULTS: Exome sequencing detected a nonsense mutation (c.C535T; p.R179X) in exon 3 of the FGF16 gene, which maps to chromosome Xq21.1. Mutational screening of the FGF16 gene performed in an unrelated proband of different ethnicity showed another nonsense mutation in exon 3 (c.C470A; p.S157X). CONCLUSIONS: This study shows that truncating mutations of FGF16 are associated with X-linked recessive metacarpal 4-5 fusion. The study provides evidence for the involvement of FGF16 in the fine tuning of the human skeleton of the hand.


Asunto(s)
Codón sin Sentido , Exoma , Factores de Crecimiento de Fibroblastos/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Deformidades Congénitas de la Mano/genética , Huesos del Metacarpo/anomalías , Niño , Embrión de Mamíferos , Femenino , Humanos , Masculino , Especificidad de Órganos , Análisis de Secuencia de ADN
19.
Ginekol Pol ; 85(7): 541-4, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25118508

RESUMEN

Craniosynostosis (a premature fusion of the cranial sutures) occurs with a frequency of 1 in 2100-2500 births and in over 40% cases is caused by known genetic factors--either single gene mutations or chromosomal rearrangements. Cases caused by complex chromosomal abnormalities are uncommon and likely associated with compound phenotype. Saethre-Chotzen syndrome (SCS) [#101400] is caused by TWIST1 gene haploinsufficiency. Its phenotype includes uni- or bicoronal synostosis, short stature, facial dysmorphism and variable anomalies of the hands and feet. Due to its poor sonographic manifestation a prenatal diagnosis of SCS is challenging. We report a case of a prenatally detected craniosynostosis (compound Saethre-Chotzen syndrome phenotype) caused by a de novo complex chromosomal rearrangement (1; 4; 7) with a microdeletion of 7p21.3-7p15.3, including TWIST1 gene.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos Par 7 , Craneosinostosis/diagnóstico , Craneosinostosis/genética , Eliminación de Gen , Proteínas Nucleares/genética , Diagnóstico Prenatal , Proteína 1 Relacionada con Twist/genética , Cariotipo Anormal , Adulto , Femenino , Humanos , Cariotipificación , Masculino , Fenotipo , Embarazo
20.
Orphanet J Rare Dis ; 19(1): 32, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38291488

RESUMEN

Vertebral malformations (VMs) pose a significant global health problem, causing chronic pain and disability. Vertebral defects occur as isolated conditions or within the spectrum of various congenital disorders, such as Klippel-Feil syndrome, congenital scoliosis, spondylocostal dysostosis, sacral agenesis, and neural tube defects. Although both genetic abnormalities and environmental factors can contribute to abnormal vertebral development, our knowledge on molecular mechanisms of numerous VMs is still limited. Furthermore, there is a lack of resource that consolidates the current knowledge in this field. In this pioneering review, we provide a comprehensive analysis of the latest research on the molecular basis of VMs and the association of the VMs-related causative genes with bone developmental signaling pathways. Our study identifies 118 genes linked to VMs, with 98 genes involved in biological pathways crucial for the formation of the vertebral column. Overall, the review summarizes the current knowledge on VM genetics, and provides new insights into potential involvement of biological pathways in VM pathogenesis. We also present an overview of available data regarding the role of epigenetic and environmental factors in VMs. We identify areas where knowledge is lacking, such as precise molecular mechanisms in which specific genes contribute to the development of VMs. Finally, we propose future research avenues that could address knowledge gaps.


Asunto(s)
Anomalías Múltiples , Hernia Diafragmática , Síndrome de Klippel-Feil , Escoliosis , Humanos , Columna Vertebral/anomalías , Columna Vertebral/patología , Anomalías Múltiples/patología , Síndrome de Klippel-Feil/patología , Hernia Diafragmática/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA