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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 210(1): 63-76, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38626355

RESUMEN

Rationale: Bronchiectasis is a pathological dilatation of the bronchi in the respiratory airways associated with environmental or genetic causes (e.g., cystic fibrosis, primary ciliary dyskinesia, and primary immunodeficiency disorders), but most cases remain idiopathic. Objectives: To identify novel genetic defects in unsolved cases of bronchiectasis presenting with severe rhinosinusitis, nasal polyposis, and pulmonary Pseudomonas aeruginosa infection. Methods: DNA was analyzed by next-generation or targeted Sanger sequencing. RNA was analyzed by quantitative PCR and single-cell RNA sequencing. Patient-derived cells, cell cultures, and secretions (mucus, saliva, seminal fluid) were analyzed by Western blotting and immunofluorescence microscopy, and mucociliary activity was measured. Blood serum was analyzed by electrochemiluminescence immunoassay. Protein structure and proteomic analyses were used to assess the impact of a disease-causing founder variant. Measurements and Main Results: We identified biallelic pathogenic variants in WAP four-disulfide core domain 2 (WFDC2) in 11 individuals from 10 unrelated families originating from the United States, Europe, Asia, and Africa. Expression of WFDC2 was detected predominantly in secretory cells of control airway epithelium and also in submucosal glands. We demonstrate that WFDC2 is below the limit of detection in blood serum and hardly detectable in samples of saliva, seminal fluid, and airway surface liquid from WFDC2-deficient individuals. Computer simulations and deglycosylation assays indicate that the disease-causing founder variant p.Cys49Arg structurally hampers glycosylation and, thus, secretion of mature WFDC2. Conclusions: WFDC2 dysfunction defines a novel molecular etiology of bronchiectasis characterized by the deficiency of a secreted component of the airways. A commercially available blood test combined with genetic testing allows its diagnosis.


Asunto(s)
Bronquiectasia , Pólipos Nasales , Humanos , Bronquiectasia/genética , Bronquiectasia/fisiopatología , Masculino , Femenino , Pólipos Nasales/genética , Adulto , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP , Adolescente , Niño , Persona de Mediana Edad , Adulto Joven
2.
Hum Genet ; 143(6): 761-773, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38787418

RESUMEN

Chung-Jansen syndrome is a neurodevelopmental disorder characterized by intellectual disability, behavioral problems, obesity and dysmorphic features. It is caused by pathogenic variants in the PHIP gene that encodes for the Pleckstrin homology domain-interacting protein, which is part of an epigenetic modifier protein complex. Therefore, we hypothesized that PHIP haploinsufficiency may impact genome-wide DNA methylation (DNAm). We assessed the DNAm profiles of affected individuals with pathogenic and likely pathogenic PHIP variants with Infinium Methylation EPIC arrays and report a specific and sensitive DNAm episignature biomarker for Chung-Jansen syndrome. In addition, we observed similarities between the methylation profile of Chung-Jansen syndrome and that of functionally related and clinically partially overlapping genetic disorders, White-Kernohan syndrome (caused by variants in DDB1 gene) and Börjeson-Forssman-Lehmann syndrome (caused by variants in PHF6 gene). Based on these observations we also proceeded to develop a common episignature biomarker for these disorders. These newly defined episignatures can be used as part of a multiclass episignature classifier for screening of affected individuals with rare disorders and interpretation of genetic variants of unknown clinical significance, and provide further insights into the common molecular pathophysiology of the clinically-related Chung-Jansen, Börjeson-Forssman-Lehmann and White-Kernohan syndromes.


Asunto(s)
Metilación de ADN , Discapacidad Intelectual , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/diagnóstico , Masculino , Femenino , Haploinsuficiencia/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/diagnóstico , Niño
3.
Am J Hum Genet ; 108(5): 951-961, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33894126

RESUMEN

The collapsin response mediator protein (CRMP) family proteins are intracellular mediators of neurotrophic factors regulating neurite structure/spine formation and are essential for dendrite patterning and directional axonal pathfinding during brain developmental processes. Among this family, CRMP5/DPYSL5 plays a significant role in neuronal migration, axonal guidance, dendrite outgrowth, and synapse formation by interacting with microtubules. Here, we report the identification of missense mutations in DPYSL5 in nine individuals with brain malformations, including corpus callosum agenesis and/or posterior fossa abnormalities, associated with variable degrees of intellectual disability. A recurrent de novo p.Glu41Lys variant was found in eight unrelated patients, and a p.Gly47Arg variant was identified in one individual from the first family reported with Ritscher-Schinzel syndrome. Functional analyses of the two missense mutations revealed impaired dendritic outgrowth processes in young developing hippocampal primary neuronal cultures. We further demonstrated that these mutations, both located in the same loop on the surface of DPYSL5 monomers and oligomers, reduced the interaction of DPYSL5 with neuronal cytoskeleton-associated proteins MAP2 and ßIII-tubulin. Our findings collectively indicate that the p.Glu41Lys and p.Gly47Arg variants impair DPYSL5 function on dendritic outgrowth regulation by preventing the formation of the ternary complex with MAP2 and ßIII-tubulin, ultimately leading to abnormal brain development. This study adds DPYSL5 to the list of genes implicated in brain malformation and in neurodevelopmental disorders.


Asunto(s)
Agenesia del Cuerpo Calloso/genética , Cerebelo/anomalías , Mutación Missense/genética , Trastornos del Neurodesarrollo/genética , Adulto , Agenesia del Cuerpo Calloso/diagnóstico por imagen , Cerebelo/diagnóstico por imagen , Niño , Preescolar , Femenino , Humanos , Hidrolasas/química , Hidrolasas/genética , Discapacidad Intelectual/diagnóstico por imagen , Discapacidad Intelectual/genética , Masculino , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Moleculares , Trastornos del Neurodesarrollo/diagnóstico por imagen , Tubulina (Proteína)/metabolismo , Adulto Joven
4.
Am J Hum Genet ; 107(6): 1170-1177, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33232677

RESUMEN

KDM4B is a lysine-specific demethylase with a preferential activity on H3K9 tri/di-methylation (H3K9me3/2)-modified histones. H3K9 tri/di-demethylation is an important epigenetic mechanism responsible for silencing of gene expression in animal development and cancer. However, the role of KDM4B on human development is still poorly characterized. Through international data sharing, we gathered a cohort of nine individuals with mono-allelic de novo or inherited variants in KDM4B. All individuals presented with dysmorphic features and global developmental delay (GDD) with language and motor skills most affected. Three individuals had a history of seizures, and four had anomalies on brain imaging ranging from agenesis of the corpus callosum with hydrocephalus to cystic formations, abnormal hippocampi, and polymicrogyria. In mice, lysine demethylase 4B is expressed during brain development with high levels in the hippocampus, a region important for learning and memory. To understand how KDM4B variants can lead to GDD in humans, we assessed the effect of KDM4B disruption on brain anatomy and behavior through an in vivo heterozygous mouse model (Kdm4b+/-), focusing on neuroanatomical changes. In mutant mice, the total brain volume was significantly reduced with decreased size of the hippocampal dentate gyrus, partial agenesis of the corpus callosum, and ventriculomegaly. This report demonstrates that variants in KDM4B are associated with GDD/ intellectual disability and neuroanatomical defects. Our findings suggest that KDM4B variation leads to a chromatinopathy, broadening the spectrum of this group of Mendelian disorders caused by alterations in epigenetic machinery.


Asunto(s)
Discapacidades del Desarrollo/genética , Variación Genética , Histona Demetilasas con Dominio de Jumonji/genética , Malformaciones del Sistema Nervioso/genética , Animales , Encéfalo/diagnóstico por imagen , Epigénesis Genética , Femenino , Heterocigoto , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Histonas/metabolismo , Humanos , Imagen por Resonancia Magnética , Masculino , Metilación , Ratones , Procesamiento Proteico-Postraduccional , Convulsiones/genética , Transducción de Señal
5.
J Allergy Clin Immunol ; 149(3): 1120-1127.e8, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34416217

RESUMEN

BACKGROUND: Proteasome-associated autoinflammatory syndromes (PRAASs) form a family of recently described rare autosomal recessive disorders of disturbed proteasome assembly and proteolytic activity caused by mutations in genes coding for proteasome subunits. The treatment options for these proteasome disorders consist of lifelong immunosuppressive drugs or Janus kinase inhibitors, which may have partial efficacy and noticeable side effects. Because proteasomes are ubiquitously expressed, it is unknown whether hematopoietic stem cell transplantation (HSCT) may be a sufficient treatment option. OBJECTIVE: Our aim was to report the case of a young boy with a treatment-resistant cutaneous vasculitis that was initially suspected to be associated with a gene variant in SH2D1A. METHODS: Whole-exome sequencing was performed to identify the genetic defect. Molecular and functional analyses were performed to assess the impact of variants on proteasomal function. The immune characterization led to the decision to perform HSCT on our patient and conduct follow-up over the 7-year period after the transplant. Because loss of myeloid chimerism after the first HSCT was associated with relapse of autoinflammation, a second HSCT was performed. RESULTS: After the successful second HSCT, the patient developed mild symptoms of lipodystrophy, which raised the suspicion of a PRAAS. Genetic analysis revealed 2 novel heterozygous variants in PSMB4 (encoding proteasomal subunit ß7). Retrospective analysis of patient cells stored before the first HSCT and patient cells obtained after the second HSCT demonstrated that HSCT successfully rescued proteasome function, restored protein homeostasis, and resolved the interferon-stimulated gene signature. Furthermore, successful HSCT alleviated the autoinflammatory manifestations in our patient. CONCLUSION: Patients with treatment-resistant PRAAS can be cured by HSCT.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Lipodistrofia , Niño , Humanos , Lipodistrofia/genética , Masculino , Complejo de la Endopetidasa Proteasomal/genética , Estudios Retrospectivos , Síndrome
6.
Cleft Palate Craniofac J ; 60(9): 1118-1127, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-35469463

RESUMEN

To (1) appraise current international classification and clinical management strategies for craniofacial microsomia (CFM) and microtia, and (2) to assess agreement with the European Reference Network "European Guideline Craniofacial Microsomia" recommendations on screening and monitoring.This was a cross-sectional online survey study. The survey consisted of 44 questions on demographics, diagnostics and classification, obstructive sleep apnea, feeding difficulties, speech and language development, hearing, ocular abnormalities, visual development, orthodontic screening, genetic counselling, psychological wellbeing, and extracraniofacial anomalies.Respondents were participants of 3 international cleft and craniofacial conferences, members of the American Cleft Palate and Craniofacial Association and members of the International Society for Auricular Reconstruction. Respondents were requested to complete 1 questionnaire per multidisciplinary team.Fifty-seven responses were received from 30 countries (response rate ∼3%).The International Consortium for Health Outcomes Measurement diagnostic criteria were used by 86% of respondents, though 65% considered isolated microtia a mild form of CFM. The Orbit, Mandible, Ear, Facial Nerve and Soft Tissue classification system was used by 74% of respondents. Agreement with standardized screening and monitoring recommendations was between 61% and 97%. A majority of respondents agreed with screening for extracraniofacial anomalies (63%-68%) and with genetic counselling (81%).This survey did not reveal consistent agreement on the diagnostic criteria for CFM. Respondents mostly supported management recommendations, but frequently disagreed with the standardization of care. Future studies could focus on working towards international consensus on diagnostic criteria, and exploring internationally feasible management strategies.


Asunto(s)
Microtia Congénita , Síndrome de Goldenhar , Humanos , Síndrome de Goldenhar/psicología , Microtia Congénita/diagnóstico , Estudios Transversales , Mandíbula , Encuestas y Cuestionarios
7.
Am J Hum Genet ; 104(6): 1210-1222, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31079897

RESUMEN

We delineate a KMT2E-related neurodevelopmental disorder on the basis of 38 individuals in 36 families. This study includes 31 distinct heterozygous variants in KMT2E (28 ascertained from Matchmaker Exchange and three previously reported), and four individuals with chromosome 7q22.2-22.23 microdeletions encompassing KMT2E (one previously reported). Almost all variants occurred de novo, and most were truncating. Most affected individuals with protein-truncating variants presented with mild intellectual disability. One-quarter of individuals met criteria for autism. Additional common features include macrocephaly, hypotonia, functional gastrointestinal abnormalities, and a subtle facial gestalt. Epilepsy was present in about one-fifth of individuals with truncating variants and was responsive to treatment with anti-epileptic medications in almost all. More than 70% of the individuals were male, and expressivity was variable by sex; epilepsy was more common in females and autism more common in males. The four individuals with microdeletions encompassing KMT2E generally presented similarly to those with truncating variants, but the degree of developmental delay was greater. The group of four individuals with missense variants in KMT2E presented with the most severe developmental delays. Epilepsy was present in all individuals with missense variants, often manifesting as treatment-resistant infantile epileptic encephalopathy. Microcephaly was also common in this group. Haploinsufficiency versus gain-of-function or dominant-negative effects specific to these missense variants in KMT2E might explain this divergence in phenotype, but requires independent validation. Disruptive variants in KMT2E are an under-recognized cause of neurodevelopmental abnormalities.


Asunto(s)
Proteínas de Unión al ADN/genética , Epilepsia/etiología , Variación Genética , Heterocigoto , Trastornos del Neurodesarrollo/etiología , Adolescente , Adulto , Niño , Preescolar , Epilepsia/patología , Femenino , Haploinsuficiencia , Humanos , Lactante , Masculino , Trastornos del Neurodesarrollo/patología , Linaje , Fenotipo , Adulto Joven
8.
Int J Mol Sci ; 23(14)2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35887345

RESUMEN

JARID2 (Jumonji, AT Rich Interactive Domain 2) pathogenic variants cause a neurodevelopmental syndrome, that is characterized by developmental delay, cognitive impairment, hypotonia, autistic features, behavior abnormalities and dysmorphic facial features. JARID2 encodes a transcriptional repressor protein that regulates the activity of various histone methyltransferase complexes. However, the molecular etiology is not fully understood, and JARID2-neurodevelopmental syndrome may vary in its typical clinical phenotype. In addition, the detection of variants of uncertain significance (VUSs) often results in a delay of final diagnosis which could hamper the appropriate care. In this study we aim to detect a specific and sensitive DNA methylation signature for JARID2-neurodevelopmental syndrome. Peripheral blood DNA methylation profiles from 56 control subjects, 8 patients with (likely) pathogenic JARID2 variants and 3 patients with JARID2 VUSs were analyzed. DNA methylation analysis indicated a clear and robust separation between patients with (likely) pathogenic variants and controls. A binary model capable of classifying patients with the JARID2-neurodevelopmental syndrome was constructed on the basis of the identified episignature. Patients carrying VUSs clustered with the control group. We identified a distinct DNA methylation signature associated with JARID2-neurodevelopmental syndrome, establishing its utility as a biomarker for this syndrome and expanding the EpiSign diagnostic test.


Asunto(s)
Metilación de ADN , Complejo Represivo Polycomb 2 , Humanos , Motivos de Nucleótidos , Fenotipo , Complejo Represivo Polycomb 2/genética , Procesamiento Proteico-Postraduccional , Síndrome
9.
Am J Med Genet A ; 185(12): 3814-3820, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34254723

RESUMEN

Terminal osseous dysplasia with pigmentary defects (TODPD), also known as digitocutaneous dysplasia, is one of the X-linked filaminopathies caused by a variety of FLNA-variants. TODPD is characterized by skeletal defects, skin fibromata and dysmorphic facial features. So far, only a single recurrent variant (c.5217G>A;p.Val1724_Thr1739del) in FLNA has found to be responsible for TODPD. We identified a novel c.5217+5G>C variant in FLNA in a female proband with skeletal defects, skin fibromata, interstitial lung disease, epilepsy, and restrictive cardiomyopathy. This variant causes mis-splicing of exon 31 predicting the production of a FLNA-protein with an in-frame-deletion of 16 residues identical to the miss-splicing-effect of the recurrent TODPD c.5217G>A variant. This mis-spliced transcript was explicitly detected in heart tissue, but was absent from blood, skin, and lung. X-inactivation analyses showed extreme skewing with almost complete inactivation of the mutated allele (>90%) in these tissues, except for heart. The mother of the proband, who also has fibromata and skeletal abnormalities, is also carrier of the FLNA-variant and was diagnosed with noncompaction cardiomyopathy after cardiac screening. No other relevant variants in cardiomyopathy-related genes were found. Here we describe a novel variant in FLNA (c.5217+5G>C) as the second pathogenic variant responsible for TODPD. Cardiomyopathy has not been described as a phenotypic feature of TODPD before.


Asunto(s)
Cardiomiopatías/genética , Filaminas/genética , Dedos/anomalías , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Predisposición Genética a la Enfermedad , Deformidades Congénitas de las Extremidades/genética , Osteocondrodisplasias/genética , Trastornos de la Pigmentación/genética , Dedos del Pie/anomalías , Cardiomiopatías/complicaciones , Cardiomiopatías/patología , Preescolar , Femenino , Dedos/patología , Genes Ligados a X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/complicaciones , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Lactante , Deformidades Congénitas de las Extremidades/complicaciones , Deformidades Congénitas de las Extremidades/patología , Mutación/genética , Osteocondrodisplasias/complicaciones , Osteocondrodisplasias/patología , Fenotipo , Trastornos de la Pigmentación/complicaciones , Trastornos de la Pigmentación/patología , Eliminación de Secuencia/genética , Dedos del Pie/patología , Inactivación del Cromosoma X/genética
10.
Hum Genet ; 139(5): 575-592, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32193685

RESUMEN

RAD21 encodes a key component of the cohesin complex, and variants in RAD21 have been associated with Cornelia de Lange Syndrome (CdLS). Limited information on phenotypes attributable to RAD21 variants and genotype-phenotype relationships is currently published. We gathered a series of 49 individuals from 33 families with RAD21 alterations [24 different intragenic sequence variants (2 recurrent), 7 unique microdeletions], including 24 hitherto unpublished cases. We evaluated consequences of 12 intragenic variants by protein modelling and molecular dynamic studies. Full clinical information was available for 29 individuals. Their phenotype is an attenuated CdLS phenotype compared to that caused by variants in NIPBL or SMC1A for facial morphology, limb anomalies, and especially for cognition and behavior. In the 20 individuals with limited clinical information, additional phenotypes include Mungan syndrome (in patients with biallelic variants) and holoprosencephaly, with or without CdLS characteristics. We describe several additional cases with phenotypes including sclerocornea, in which involvement of the RAD21 variant is uncertain. Variants were frequently familial, and genotype-phenotype analyses demonstrated striking interfamilial and intrafamilial variability. Careful phenotyping is essential in interpreting consequences of RAD21 variants, and protein modeling and dynamics can be helpful in determining pathogenicity. The current study should be helpful when counseling families with a RAD21 variation.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Deleción Cromosómica , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Síndrome de Cornelia de Lange/genética , Síndrome de Cornelia de Lange/patología , Mutación , Adolescente , Adulto , Proteínas de Ciclo Celular/química , Niño , Preescolar , Proteínas de Unión al ADN/química , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Simulación de Dinámica Molecular , Fenotipo , Conformación Proteica , Adulto Joven
11.
Hum Mutat ; 40(12): 2270-2285, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31206972

RESUMEN

Pathogenic variants in the X-linked gene ZC4H2, which encodes a zinc-finger protein, cause an infrequently described syndromic form of arthrogryposis multiplex congenita (AMC) with central and peripheral nervous system involvement. We present genetic and detailed phenotypic information on 23 newly identified families and simplex cases that include 19 affected females from 18 families and 14 affected males from nine families. Of note, the 15 females with deleterious de novo ZC4H2 variants presented with phenotypes ranging from mild to severe, and their clinical features overlapped with those seen in affected males. By contrast, of the nine carrier females with inherited ZC4H2 missense variants that were deleterious in affected male relatives, four were symptomatic. We also compared clinical phenotypes with previously published cases of both sexes and provide an overview on 48 males and 57 females from 42 families. The spectrum of ZC4H2 defects comprises novel and recurrent mostly inherited missense variants in affected males, and de novo splicing, frameshift, nonsense, and partial ZC4H2 deletions in affected females. Pathogenicity of two newly identified missense variants was further supported by studies in zebrafish. We propose ZC4H2 as a good candidate for early genetic testing of males and females with a clinical suspicion of fetal hypo-/akinesia and/or (neurogenic) AMC.


Asunto(s)
Artrogriposis/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación , Proteínas Nucleares/genética , Animales , Codón sin Sentido , Modelos Animales de Enfermedad , Femenino , Mutación del Sistema de Lectura , Genes Ligados a X , Predisposición Genética a la Enfermedad , Humanos , Masculino , Mutación Missense , Linaje , Fenotipo , Eliminación de Secuencia , Caracteres Sexuales , Pez Cebra
12.
Hum Genet ; 138(1): 61-72, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30535804

RESUMEN

ATP2B2 encodes the PMCA2 Ca2+ pump that plays an important role in maintaining ion homeostasis in hair cells among others by extrusion of Ca2+ from the stereocilia to the endolymph. Several mouse models have been described for this gene; mice heterozygous for loss-of-function defects display a rapidly progressive high-frequency hearing impairment. Up to now ATP2B2 has only been reported as a modifier, or in a digenic mechanism with CDH23 for hearing impairment in humans. Whole exome sequencing in hearing impaired index cases of Dutch and Polish origins revealed five novel heterozygous (predicted to be) loss-of-function variants of ATP2B2. Two variants, c.1963G>T (p.Glu655*) and c.955delG (p.Ala319fs), occurred de novo. Three variants c.397+1G>A (p.?), c.1998C>A (p.Cys666*), and c.2329C>T (p.Arg777*), were identified in families with an autosomal dominant inheritance pattern of hearing impairment. After normal newborn hearing screening, a rapidly progressive high-frequency hearing impairment was diagnosed at the age of about 3-6 years. Subjects had no balance complaints and vestibular testing did not yield abnormalities. There was no evidence for retrocochlear pathology or structural inner ear abnormalities. Although a digenic inheritance pattern of hearing impairment has been reported for heterozygous missense variants of ATP2B2 and CDH23, our findings indicate a monogenic cause of hearing impairment in cases with loss-of-function variants of ATP2B2.


Asunto(s)
Biomarcadores/análisis , Predisposición Genética a la Enfermedad , Pérdida Auditiva/genética , Mutación , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Estudios de Seguimiento , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Linaje , Pronóstico , Adulto Joven
13.
Gastroenterology ; 155(1): 118-129.e6, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29601828

RESUMEN

BACKGROUND & AIMS: Hirschsprung disease (HSCR) is an inherited congenital disorder characterized by absence of enteric ganglia in the distal part of the gut. Variants in ret proto-oncogene (RET) have been associated with up to 50% of familial and 35% of sporadic cases. We searched for variants that affect disease risk in a large, multigenerational family with history of HSCR in a linkage region previously associated with the disease (4q31.3-q32.3) and exome wide. METHODS: We performed exome sequencing analyses of a family in the Netherlands with 5 members diagnosed with HSCR and 2 members diagnosed with functional constipation. We initially focused on variants in genes located in 4q31.3-q32.3; however, we also performed an exome-wide analysis in which known HSCR or HSCR-associated gene variants predicted to be deleterious were prioritized for further analysis. Candidate genes were expressed in HEK293, COS-7, and Neuro-2a cells and analyzed by luciferase and immunoblot assays. Morpholinos were designed to target exons of candidate genes and injected into 1-cell stage zebrafish embryos. Embryos were allowed to develop and stained for enteric neurons. RESULTS: Within the linkage region, we identified 1 putative splice variant in the lipopolysaccharide responsive beige-like anchor protein gene (LRBA). Functional assays could not confirm its predicted effect on messenger RNA splicing or on expression of the mab-21 like 2 gene (MAB21L2), which is embedded in LRBA. Zebrafish that developed following injection of the lrba morpholino had a shortened body axis and subtle gut morphological defects, but no significant reduction in number of enteric neurons compared with controls. Outside the linkage region, members of 1 branch of the family carried a previously unidentified RET variant or an in-frame deletion in the glial cell line derived neurotrophic factor gene (GDNF), which encodes a ligand of RET. This deletion was located 6 base pairs before the last codon. We also found variants in the Indian hedgehog gene (IHH) and its mediator, the transcription factor GLI family zinc finger 3 (GLI3). When expressed in cells, the RET-P399L variant disrupted protein glycosylation and had altered phosphorylation following activation by GDNF. The deletion in GDNF prevented secretion of its gene product, reducing RET activation, and the IHH-Q51K variant reduced expression of the transcription factor GLI1. Injection of morpholinos that target ihh reduced the number of enteric neurons to 13% ± 1.4% of control zebrafish. CONCLUSIONS: In a study of a large family with history of HSCR, we identified variants in LRBA, RET, the gene encoding the RET ligand (GDNF), IHH, and a gene encoding a mediator of IHH signaling (GLI3). These variants altered functions of the gene products when expressed in cells and knockout of ihh reduced the number of enteric neurons in the zebrafish gut.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Proteínas Hedgehog/genética , Enfermedad de Hirschsprung/genética , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas c-ret/genética , Proteína Gli3 con Dedos de Zinc/genética , Animales , Células COS , Chlorocebus aethiops , Familia , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Células HEK293 , Humanos , Masculino , Morfolinos , Países Bajos , Linaje , Isoformas de Proteínas , Proto-Oncogenes Mas , Análisis de Secuencia de ADN , Transducción de Señal , Pez Cebra
14.
Genet Med ; 21(8): 1808-1820, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30635621

RESUMEN

PURPOSE: Beckwith-Wiedemann syndrome (BWS) is a developmental disorder caused by dysregulation of the imprinted gene cluster of chromosome 11p15.5 and often associated with loss of methylation (LOM) of the imprinting center 2 (IC2) located in KCNQ1 intron 10. To unravel the etiological mechanisms underlying these epimutations, we searched for genetic variants associated with IC2 LOM. METHODS: We looked for cases showing the clinical features of both BWS and long QT syndrome (LQTS), which is often associated with KCNQ1 variants. Pathogenic variants were identified by genomic analysis and targeted sequencing. Functional experiments were performed to link these pathogenic variants to the imprinting defect. RESULTS: We found three rare cases in which complete IC2 LOM is associated with maternal transmission of KCNQ1 variants, two of which were demonstrated to affect KCNQ1 transcription upstream of IC2. As a consequence of KCNQ1 haploinsufficiency, these variants also cause LQTS on both maternal and paternal transmission. CONCLUSION: These results are consistent with the hypothesis that, similar to what has been demonstrated in mouse, lack of transcription across IC2 results in failure of methylation establishment in the female germline and BWS later in development, and also suggest a new link between LQTS and BWS that is important for genetic counseling.


Asunto(s)
Síndrome de Beckwith-Wiedemann/genética , Metilación de ADN/genética , Canal de Potasio KCNQ1/genética , Adolescente , Adulto , Animales , Síndrome de Beckwith-Wiedemann/epidemiología , Síndrome de Beckwith-Wiedemann/patología , Niño , Preescolar , Cromosomas Humanos Par 11/genética , Femenino , Impresión Genómica/genética , Humanos , Lactante , Intrones/genética , Masculino , Herencia Materna/genética , Ratones , Linaje , Adulto Joven
15.
Hum Mutat ; 39(9): 1226-1237, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29897170

RESUMEN

Malan syndrome is an overgrowth disorder described in a limited number of individuals. We aim to delineate the entity by studying a large group of affected individuals. We gathered data on 45 affected individuals with a molecularly confirmed diagnosis through an international collaboration and compared data to the 35 previously reported individuals. Results indicate that height is > 2 SDS in infancy and childhood but in only half of affected adults. Cardinal facial characteristics include long, triangular face, macrocephaly, prominent forehead, everted lower lip, and prominent chin. Intellectual disability is universally present, behaviorally anxiety is characteristic. Malan syndrome is caused by deletions or point mutations of NFIX clustered mostly in exon 2. There is no genotype-phenotype correlation except for an increased risk for epilepsy with 19p13.2 microdeletions. Variants arose de novo, except in one family in which mother was mosaic. Variants causing Malan and Marshall-Smith syndrome can be discerned by differences in the site of stop codon formation. We conclude that Malan syndrome has a well recognizable phenotype that usually can be discerned easily from Marshall-Smith syndrome but rarely there is some overlap. Differentiation from Sotos and Weaver syndrome can be made by clinical evaluation only.


Asunto(s)
Anomalías Múltiples/genética , Hipotiroidismo Congénito/genética , Anomalías Craneofaciales/genética , Deformidades Congénitas de la Mano/genética , Discapacidad Intelectual/genética , Factores de Transcripción NFI/genética , Síndrome de Sotos/genética , Anomalías Múltiples/fisiopatología , Adolescente , Adulto , Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/fisiopatología , Niño , Preescolar , Deleción Cromosómica , Hipotiroidismo Congénito/fisiopatología , Anomalías Craneofaciales/fisiopatología , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/fisiopatología , Exones/genética , Femenino , Deformidades Congénitas de la Mano/fisiopatología , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Megalencefalia/genética , Megalencefalia/fisiopatología , Mutación Missense/genética , Fenotipo , Displasia Septo-Óptica/genética , Displasia Septo-Óptica/fisiopatología , Síndrome de Sotos/fisiopatología , Adulto Joven
16.
Prenat Diagn ; 37(2): 162-167, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27943390

RESUMEN

OBJECTIVES: Polyhydramnios is suggested to be associated with oral clefts (OCs) due to swallowing problems. This study assessed incidence and outcome of idiopathic polyhydramnios in isolated OC pregnancies. METHODS: This was a retrospective cohort study of prenatally diagnosed OC. The incidence of idiopathic polyhydramnios in isolated OC pregnancies was determined. Pregnancy outcome, neonatal and paediatric follow-up were compared between cases with polyhydramnios and those with normal amniotic fluid. Subgroup analysis was conducted to evaluate whether an association exists between polyhydramnios and presence of associated anomalies diagnosed after birth. RESULTS: In 230 cases of isolated OC, 15 developed polyhydramnios (6.5%). Involvement of the palate was significantly more common in the presence than in the absence of polyhydramnios (13/15 or 87% vs 125/215 or 58%, p = 0.03, odds ratio 4.7, 95% confidence interval 1.0-30.8). No significant differences were seen in pregnancy outcome or neonatal and paediatric follow-up between the two groups. In subgroup analysis, rate of polyhydramnios was not significantly different in associated cases that appeared isolated prenatally (1/27; 3.7%) compared with that in the isolated cases (15/230; 6.5%). CONCLUSIONS: The incidence of idiopathic polyhydramnios in isolated OC pregnancies is 6.5%. Polyhydramnios in isolated OC increases the risk of palate involvement. The presence of polyhydramnios is not associated with adverse perinatal or long-term outcome. If isolated at prenatal assessment, polyhydramnios does not increase the risk of associated anomalies postpartum. © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Labio Leporino/epidemiología , Fisura del Paladar/epidemiología , Polihidramnios/epidemiología , Resultado del Embarazo/epidemiología , Adolescente , Adulto , Labio Leporino/complicaciones , Labio Leporino/diagnóstico , Fisura del Paladar/complicaciones , Fisura del Paladar/diagnóstico , Femenino , Humanos , Incidencia , Polihidramnios/diagnóstico , Polihidramnios/etiología , Embarazo , Segundo Trimestre del Embarazo , Tercer Trimestre del Embarazo , Diagnóstico Prenatal/estadística & datos numéricos , Estudios Retrospectivos , Adulto Joven
17.
Am J Med Genet A ; 170(9): 2248-60, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27419809

RESUMEN

Patients with Beckwith-Wiedemann syndrome (BWS) have an increased risk to develop cancer in childhood, especially Wilms tumor and hepatoblastoma. The risk varies depending on the cause of BWS. We obtained clinical and molecular data in our cohort of children with BWS, including tumor occurrences, and correlated phenotype and genotype. We obtained similar data from larger cohorts reported in the literature. Phenotype, genotype and tumor occurrence were available in 229 of our own patients. Minor differences in phenotype existed depending on genotype/epigenotype, similar to earlier studies. By adding patients from the literature, we obtained data on genotype and tumor occurrence of in total 1,971 BWS patients. Tumor risks were highest in the IC1 (H19/IGF2:IG-DMR) hypermethylation subgroup (28%) and pUPD subgroup (16%) and were lower in the KCNQ1OT1:TSS-DMR (IC2) subgroup (2.6%), CDKN1C (6.9%) subgroup, and the group in whom no molecular defect was detectable (6.7%). Wilms tumors (median age 24 months) were frequent in the IC1 (24%) and pUPD (7.9%) subgroups. Hepatoblastoma occurred mostly in the pUPD (3.5%) and IC2 (0.7%) subgroups, never in the IC1 and CDKN1C subgroups, and always before 30 months of age. In the CDKN1C subgroup 2.8% of patients developed neuroblastoma. We conclude tumor risks in BWS differ markedly depending on molecular background. We propose a differentiated surveillance protocol, based on tumor risks in the various molecular subgroups causing BWS. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/genética , Estudios de Asociación Genética , Neoplasias/etiología , Fenotipo , Vigilancia de la Población , Adolescente , Síndrome de Beckwith-Wiedemann/epidemiología , Niño , Estudios de Cohortes , Metilación de ADN , Femenino , Impresión Genómica , Hepatoblastoma/epidemiología , Hepatoblastoma/etiología , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Masculino , Repeticiones de Minisatélite , Neoplasias/epidemiología , Canales de Potasio con Entrada de Voltaje/genética , ARN Largo no Codificante/genética , Riesgo , Tumor de Wilms/epidemiología , Tumor de Wilms/etiología , Adulto Joven
18.
Am J Med Genet A ; 170(10): 2681-93, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27311832

RESUMEN

Mutations in CREBBP cause Rubinstein-Taybi syndrome. By using exome sequencing, and by using Sanger in one patient, CREBBP mutations were detected in 11 patients who did not, or only in a very limited manner, resemble Rubinstein-Taybi syndrome. The combined facial signs typical for Rubinstein-Taybi syndrome were absent, none had broad thumbs, and three had only somewhat broad halluces. All had apparent developmental delay (being the reason for molecular analysis); five had short stature and seven had microcephaly. The facial characteristics were variable; main characteristics were short palpebral fissures, telecanthi, depressed nasal ridge, short nose, anteverted nares, short columella, and long philtrum. Six patients had autistic behavior, and two had self-injurious behavior. Other symptoms were recurrent upper airway infections (n = 5), feeding problems (n = 7) and impaired hearing (n = 7). Major malformations occurred infrequently. All patients had a de novo missense mutation in the last part of exon 30 or beginning of exon 31 of CREBBP, between base pairs 5,128 and 5,614 (codons 1,710 and 1,872). No missense or truncating mutations in this region have been described to be associated with the classical Rubinstein-Taybi syndrome phenotype. No functional studies have (yet) been performed, but we hypothesize that the mutations disturb protein-protein interactions by altering zinc finger function. We conclude that patients with missense mutations in this specific CREBBP region show a phenotype that differs substantially from that in patients with Rubinstein-Taybi syndrome, and may prove to constitute one (or more) separate entities. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteína de Unión a CREB/genética , Estudios de Asociación Genética , Mutación , Fenotipo , Síndrome de Rubinstein-Taybi/diagnóstico , Síndrome de Rubinstein-Taybi/genética , Adolescente , Adulto , Alelos , Secuencia de Aminoácidos , Niño , Preescolar , Exoma , Exones , Facies , Femenino , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Mutación Missense , Adulto Joven
19.
Am J Med Genet A ; 167(6): 1215-22, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25899317

RESUMEN

Frontometaphyseal dysplasia (FMD) is a distinctive sclerosing skeletal dysplasia associated with a number of non-skeletal manifestations including hearing loss, cardiac malformations, and stenosis, particularly of the upper airway and urinary tract. Some, but not all, patients have mutations in FLNA causing the condition. Consonant with the X chromosomal location of FLNA males are generally more severely affected than females. FLNA mutations can be detected in 82% of affected males. We describe seven patients (one male, six females) all of whom have the major clinical and radiological features of FMD, but without detectable mutations in FLNA. The females in our cohort are affected to a similar degree as is usually found in males. In addition, all patients have marked keloid formation at various body sites, including the eye, from an early age. Other features that may indicate a different etiology in these patients are the increased frequency of cleft palate, Robin sequence, tracheal stenosis, and mild intellectual disability, which all occur in three of more patients in the present group. All patients are isolated. We hypothesize that the presently reported patients represent further evidence that phenotypes strongly resembling FMD exist that are not accounted for by mutations in FLNA. Since the frequency of several of the manifestations, their sporadic presentations, and the presence of keloid formation differ from the X-linked form of this condition we propose de novo autosomal dominant acting mutations in a gene functionally related to FLNA, underpin this disorder.


Asunto(s)
Fisura del Paladar/genética , Frente/anomalías , Discapacidad Intelectual/genética , Queloide/genética , Osteocondrodisplasias/genética , Síndrome de Pierre Robin/genética , Estenosis Traqueal/genética , Adolescente , Niño , Duplicación Cromosómica , Cromosomas Humanos Par 22 , Fisura del Paladar/patología , Femenino , Filaminas/genética , Frente/patología , Expresión Génica , Humanos , Discapacidad Intelectual/patología , Queloide/patología , Masculino , Mutación , Osteocondrodisplasias/patología , Síndrome de Pierre Robin/patología , Factores Sexuales , Estenosis Traqueal/patología
20.
Am J Med Genet A ; 167A(9): 1983-92, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26033782

RESUMEN

Robin sequence (RS) can be defined as the combination of micrognathia and upper airway obstruction/glossoptosis causing neonatal respiratory problems, with or without a cleft palate and either isolated or non-isolated. Pathogenesis varies widely. We hypothesize that optimal treatment depends on pathogenesis and therefore patients should be stratified according to diagnosis. Here, we evaluate diagnoses and (presumed) pathogeneses in an RS cohort. Medical records of all RS patients presenting between 1995-2013 in three academic hospitals were evaluated. Four clinical geneticists re-evaluated all information, including initial diagnosis. Diagnoses were either confirmed, considered uncertain, or rejected. If uncertain or rejected, patients were re-evaluated. Subsequent results were re-discussed and a final conclusion was drawn. We included 191 RS patients. After re-evaluation and changing initial diagnoses in 48 of the 191 patients (25.1%), 37.7% of the cohort had isolated RS, 8.9% a chromosome anomaly, 29.3% a Mendelian disorder, and 24.1% no detectable cause. Twenty-two different Mendelian disorders were diagnosed, of which Stickler syndrome was most frequent. Stratification of diagnoses according to (presumed) pathogenic mechanism in 73 non-isolated patients with reliable diagnoses showed 43.9% to have a connective tissue dysplasia, 5.5% a neuromuscular disorder, 47.9% a multisystem disorder, and 2.7% an unknown mechanism. We diagnosed more non-isolated RS patients compared to other studies. Re-evaluation changed initial diagnosis in a quarter of patients. We suggest standardized re-evaluation of all RS patients. Despite the relatively high diagnostic yield pathogenesis could be determined in only 59.7% (71/119), due to limited insight in pathogenesis in diagnosed entities. Further studies into pathogenesis of entities causing RS are indicated.


Asunto(s)
Síndrome de Pierre Robin/etiología , Síndrome de Pierre Robin/patología , Obstrucción de las Vías Aéreas/etiología , Obstrucción de las Vías Aéreas/patología , Artritis/etiología , Artritis/patología , Fisura del Paladar/patología , Enfermedades del Tejido Conjuntivo/etiología , Enfermedades del Tejido Conjuntivo/patología , Femenino , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/patología , Humanos , Masculino , Micrognatismo/etiología , Micrognatismo/patología , Desprendimiento de Retina/etiología , Desprendimiento de Retina/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA