Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Med Genet ; 60(6): 620-626, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36368868

RESUMEN

BACKGROUND: Oculo-auriculo-vertebral spectrum (OAVS) is the second most common cause of head and neck malformations in children after orofacial clefts. OAVS is clinically heterogeneous and characterised by a broad range of clinical features including ear anomalies with or without hearing loss, hemifacial microsomia, orofacial clefts, ocular defects and vertebral abnormalities. Various genetic causes were associated with OAVS and copy number variations represent a recurrent cause of OAVS, but the responsible gene often remains elusive. METHODS: We described an international cohort of 17 patients, including 10 probands and 7 affected relatives, presenting with OAVS and carrying a 14q22.3 microduplication detected using chromosomal microarray analysis. For each patient, clinical data were collected using a detailed questionnaire addressed to the referring clinicians. We subsequently studied the effects of OTX2 overexpression in a zebrafish model. RESULTS: We defined a 272 kb minimal common region that only overlaps with the OTX2 gene. Head and face defects with a predominance of ear malformations were present in 100% of patients. The variability in expressivity was significant, ranging from simple chondromas to severe microtia, even between intrafamilial cases. Heterologous overexpression of OTX2 in zebrafish embryos showed significant effects on early development with alterations in craniofacial development. CONCLUSIONS: Our results indicate that proper OTX2 dosage seems to be critical for the normal development of the first and second branchial arches. Overall, we demonstrated that OTX2 genomic duplications are a recurrent cause of OAVS marked by auricular malformations of variable severity.


Asunto(s)
Labio Leporino , Fisura del Paladar , Síndrome de Goldenhar , Humanos , Animales , Síndrome de Goldenhar/genética , Pez Cebra/genética , Variaciones en el Número de Copia de ADN/genética , Factores de Transcripción Otx/genética
2.
Genet Med ; 23(6): 1028-1040, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33658631

RESUMEN

PURPOSE: We describe a novel neurobehavioral phenotype of autism spectrum disorder (ASD), intellectual disability, and/or attention-deficit/hyperactivity disorder (ADHD) associated with de novo or inherited deleterious variants in members of the RFX family of genes. RFX genes are evolutionarily conserved transcription factors that act as master regulators of central nervous system development and ciliogenesis. METHODS: We assembled a cohort of 38 individuals (from 33 unrelated families) with de novo variants in RFX3, RFX4, and RFX7. We describe their common clinical phenotypes and present bioinformatic analyses of expression patterns and downstream targets of these genes as they relate to other neurodevelopmental risk genes. RESULTS: These individuals share neurobehavioral features including ASD, intellectual disability, and/or ADHD; other frequent features include hypersensitivity to sensory stimuli and sleep problems. RFX3, RFX4, and RFX7 are strongly expressed in developing and adult human brain, and X-box binding motifs as well as RFX ChIP-seq peaks are enriched in the cis-regulatory regions of known ASD risk genes. CONCLUSION: These results establish a likely role of deleterious variation in RFX3, RFX4, and RFX7 in cases of monogenic intellectual disability, ADHD and ASD, and position these genes as potentially critical transcriptional regulators of neurobiological pathways associated with neurodevelopmental disease pathogenesis.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Trastorno del Espectro Autista , Trastorno Autístico , Discapacidad Intelectual , Adulto , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno del Espectro Autista/genética , Trastorno Autístico/genética , Humanos , Discapacidad Intelectual/genética , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética
3.
Hum Mutat ; 41(10): 1738-1744, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32643838

RESUMEN

Congenital heart defects and skeletal malformations syndrome (CHDSKM) is a rare autosomal dominant disorder characterized by congenital heart disease, skeletal abnormalities, and failure to thrive. CHDSKM is caused by germline mutations in ABL1. To date, three variants have been in association with CHDSKM. In this study, we describe three de novo missense variants, c.407C>T (p.Thr136Met), c.746C>T (p.Pro249Leu), and c.1573G>A (p.Val525Met), and one recurrent variant, c.1066G>A (p.Ala356Thr), in six patients, thereby expanding the phenotypic spectrum of CHDSKM to include hearing impairment, lipodystrophy-like features, renal hypoplasia, and distinct ocular abnormalities. Functional investigation of the three novel variants showed an increased ABL1 kinase activity. The cardiac findings in additional patients with p.Ala356Thr contribute to the accumulating evidence that patients carrying either one of the recurrent variants, p.Tyr245Cys and p.Ala356Thr, have a high incidence of cardiac abnormalities. The phenotypic expansion has implications for the clinical diagnosis of CHDSKM in patients with germline ABL1 variants.


Asunto(s)
Anomalías Múltiples , Cardiopatías Congénitas , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Células Germinativas , Cardiopatías Congénitas/genética , Humanos , Fenotipo , Síndrome
4.
BMC Med Genet ; 21(1): 96, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32381069

RESUMEN

BACKGROUND: Joubert syndrome (JBTS) is a genetically heterogeneous group of neurodevelopmental syndromes caused by primary cilia dysfunction. Usually the neurological presentation starts with abnormal neonatal breathing followed by muscular hypotonia, psychomotor delay, and cerebellar ataxia. Cerebral MRI shows mid- and hindbrain anomalies including the molar tooth sign. We report a male patient with atypical presentation of Joubert syndrome type 23, thus expanding the phenotype. CASE PRESENTATION: Clinical features were consistent with JBTS already from infancy, yet the syndrome was not suspected before cerebral MRI later in childhood showed the characteristic molar tooth sign and ectopic neurohypophysis. From age 11 years seizures developed and after few years became increasingly difficult to treat, also related to inadequate compliance to therapy. He died at 23 years of sudden unexpected death in epilepsy (SUDEP). The genetic diagnosis remained elusive for many years, despite extensive genetic testing. We reached the genetic diagnosis by performing whole genome sequencing of the family trio and analyzing the data with the combination of one analysis pipeline for single nucleotide variants (SNVs)/indels and one for structural variants (SVs). This lead to the identification of the most common variant detected in patients with JBTS23 (OMIM# 616490), rs534542684, in compound heterozygosity with a 8.3 kb deletion in KIAA0586, not previously reported. CONCLUSIONS: We describe for the first time ectopic neurohypophysis and SUDEP in JBTS23, expanding the phenotype of this condition and raising the attention on the possible severity of the epilepsy in this disease. We also highlight the diagnostic power of WGS, which efficiently detects SNVs/indels and in addition allows the identification of SVs.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Ciclo Celular/genética , Cerebelo/anomalías , Muerte Súbita/patología , Epilepsia/genética , Anomalías del Ojo/genética , Enfermedades Renales Quísticas/genética , Retina/anomalías , Anomalías Múltiples/mortalidad , Anomalías Múltiples/patología , Adulto , Cerebelo/patología , Niño , Muerte Súbita/epidemiología , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/mortalidad , Discapacidades del Desarrollo/patología , Epilepsia/mortalidad , Epilepsia/patología , Anomalías del Ojo/mortalidad , Anomalías del Ojo/patología , Femenino , Heterocigoto , Humanos , Mutación INDEL , Enfermedades Renales Quísticas/mortalidad , Enfermedades Renales Quísticas/patología , Masculino , Neurohipófisis/metabolismo , Neurohipófisis/patología , Retina/patología , Secuenciación Completa del Genoma , Adulto Joven
5.
Genet Med ; 19(8): 900-908, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28151491

RESUMEN

PURPOSE: To characterize features associated with de novo mutations affecting SATB2 function in individuals ascertained on the basis of intellectual disability. METHODS: Twenty previously unreported individuals with 19 different SATB2 mutations (11 loss-of-function and 8 missense variants) were studied. Fibroblasts were used to measure mutant protein production. Subcellular localization and mobility of wild-type and mutant SATB2 were assessed using fluorescently tagged protein. RESULTS: Recurrent clinical features included neurodevelopmental impairment (19/19), absent/near absent speech (16/19), normal somatic growth (17/19), cleft palate (9/19), drooling (12/19), and dental anomalies (8/19). Six of eight missense variants clustered in the first CUT domain. Sibling recurrence due to gonadal mosaicism was seen in one family. A nonsense mutation in the last exon resulted in production of a truncated protein retaining all three DNA-binding domains. SATB2 nuclear mobility was mutation-dependent; p.Arg389Cys in CUT1 increased mobility and both p.Gly515Ser in CUT2 and p.Gln566Lys between CUT2 and HOX reduced mobility. The clinical features in individuals with missense variants were indistinguishable from those with loss of function. CONCLUSION: SATB2 haploinsufficiency is a common cause of syndromic intellectual disability. When mutant SATB2 protein is produced, the protein appears functionally inactive with a disrupted pattern of chromatin or matrix association.Genet Med advance online publication 02 February 2017.


Asunto(s)
Discapacidad Intelectual/genética , Mutación con Pérdida de Función , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Mutación Missense , Factores de Transcripción/genética , Línea Celular , Estudios de Cohortes , Estudios de Asociación Genética , Haploinsuficiencia/genética , Células HeLa , Humanos , Discapacidad Intelectual/fisiopatología , Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Unión Proteica/genética , Factores de Transcripción/fisiología , Secuenciación Completa del Genoma
6.
Am J Med Genet A ; 167A(8): 1890-6, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25846056

RESUMEN

We report on a 10-year-old-boy presenting with moderate intellectual disability (ID), impaired motor skills, hypotonia, growth delay, minor anomalies, misaligned teeth, pectus excavatum, small hands and feet, widely spaced nipples, and a 1.13 Mb de novo deletion on HSA12q12 (chr12:44,830,147-45,964,945 bp, hg19), deleting ANO6, NELL2, and DBX2 and the pseudogenes PLEKHA8P1 and RACGAP1P. We suggest DBX2 and NELL2 as disease-causing genes and their haploinsufficiency to be involved in the psychomotor delay in the patient. DBX2 encodes a homeobox protein, highly expressed during neuronal development and regulating differentiation of interneurons in brain and spinal cord. NELL2 is expressed in most of the central and peripheral nervous system, with highest expression in hippocampus and cerebellum, maximizing during neuronal differentiation. The deletion in our patient is the smallest in HSA12q12 reported to date, and it is included in the deletion carried by four previously reported patients. The clinical presentation of these patients points to the recurrence of the following manifestation, possibly delineating a 12q12 deletion syndrome phenotype: moderate to severe developmental/intellectual delay, hypotonia, postnatal growth retardation, skeletal and dental anomalies, minor facial anomalies including strabismus, down slanting palpebral fissures, and large/low-set ears.


Asunto(s)
Discapacidades del Desarrollo/genética , Haploinsuficiencia , Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/genética , Proteínas de Transferencia de Fosfolípidos/genética , Anoctaminas , Niño , Humanos , Masculino
7.
Am J Med Genet A ; 164A(7): 1622-6, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24678003

RESUMEN

MEIS2 is a homeodomain-containing transcription factor of the TALE superfamily that has been proven important for development. We confirm and extend a recent single clinical report stating that deletions in MEIS2 can cause cleft palate [Crowley et al. (2010); Am J Med Genet 152A:1326-1327]. Here we report on five additional patients with 15q14 deletions of sizes 0.6, 0.6, 1.0, 1.9, and 4.8 Mb, respectively, all involving MEIS2. In addition, we present a family with four affected individuals and an intragenic 58 kb direct duplication disrupting MEIS2. In total, 7/9 cases had clefting, from mild (submucous cleft palate) to severe (cleft lip and palate), and 3/9 cases had ventricular septal defects. All cases had delayed motor development and most had learning disability, at worst in the mild intellectual disability range. The cases had overlapping facial features (broad forehead, finely arched eyebrows, mildly shortened philtrum, and tented upper lip) but individually they were not considered to be dysmorphic. Our results show that MEIS2 is a gene needed for palate closure. In syndromic cases of cleft palate, MEIS2 should be considered among the candidate genes, for example, in cases without 22q11.2 deletions.


Asunto(s)
Labio Leporino/genética , Fisura del Paladar/genética , Estudios de Asociación Genética , Haploinsuficiencia , Proteínas de Homeodominio/genética , Discapacidades para el Aprendizaje/genética , Factores de Transcripción/genética , Adolescente , Adulto , Niño , Preescolar , Aberraciones Cromosómicas , Labio Leporino/diagnóstico , Fisura del Paladar/diagnóstico , Hibridación Genómica Comparativa , Facies , Femenino , Humanos , Discapacidades para el Aprendizaje/diagnóstico , Masculino , Fenotipo , Análisis de Secuencia de ADN , Adulto Joven
8.
Am J Med Genet A ; 161A(5): 1137-42, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23463539

RESUMEN

The transcription factor SOX3 is widely expressed in early vertebrate brain development. In humans, duplication of SOX3 and polyalanine expansions at its C-terminus may cause intellectual disability and hypopituitarism. Sox3 knock-out mice show a variable phenotype including structural and functional anomalies affecting the branchial arches and midline cerebral structures such as the optic chiasm and the hypothalamo-pituitary axis. SOX3 is claimed to be required in normal brain development and function in mice and humans, as well as in pituitary and craniofacial development. We report on an 8-year-old boy with a 2.1 Mb deletion in Xq27.1q27.2, which was found to be inherited from his healthy mother. To our knowledge, this is the smallest deletion including the entire SOX3 gene in a male reported to date. He is mildly intellectually disabled with language delay, dysarthria, behavior problems, minor facial anomalies, and hyperphagia. Hormone levels including growth, adrenocorticotropic and thyroid stimulating hormones are normal. Magnetic resonance imaging (MRI) at age 6 years showed no obvious brain anomalies. Genetic redundancy between the three members of the B1 subfamily of SOX proteins during early human brain development likely explains the apparently normal development of brain structures in our patient who is nullisomic for SOX3.


Asunto(s)
Encéfalo/anomalías , Discapacidades del Desarrollo/genética , Hiperfagia/genética , Discapacidad Intelectual/genética , Factores de Transcripción SOXB1/genética , Niño , Análisis Citogenético , Humanos , Masculino , Reacción en Cadena de la Polimerasa , Eliminación de Secuencia
9.
Eur J Hum Genet ; 25(8): 946-951, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28513610

RESUMEN

The introduction of whole-exome sequencing into the Pediatric Genetics clinic has increased the identification of novel genes associated with neurodevelopmental disorders and congenital anomalies. This agnostic approach has shed light on multiple proteins and pathways not previously known to be associated with disease. Here we report eight subjects from six families with predicted loss of function variants in ZNF462, a zinc-finger protein of unknown function. These individuals have overlapping phenotypes that include ptosis, metopic ridging, craniosynostosis, dysgenesis of the corpus callosum, and developmental delay. We propose that ZNF462 plays an important role in embryonic development, and is associated with craniofacial and neurodevelopmental abnormalities.


Asunto(s)
Agenesia del Cuerpo Calloso/genética , Blefaroptosis/genética , Anomalías Craneofaciales/genética , Proteínas de Unión al ADN/genética , Discapacidades del Desarrollo/genética , Haploinsuficiencia , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética , Adulto , Agenesia del Cuerpo Calloso/diagnóstico , Blefaroptosis/diagnóstico , Niño , Preescolar , Anomalías Craneofaciales/diagnóstico , Discapacidades del Desarrollo/diagnóstico , Femenino , Humanos , Lactante , Masculino , Síndrome
10.
Neurology ; 86(10): 954-62, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26865513

RESUMEN

OBJECTIVE: To give a comprehensive overview of the phenotypic and genetic spectrum of STXBP1 encephalopathy (STXBP1-E) by systematically reviewing newly diagnosed and previously reported patients. METHODS: We recruited newly diagnosed patients with STXBP1 mutations through an international network of clinicians and geneticists. Furthermore, we performed a systematic literature search to review the phenotypes of all previously reported patients. RESULTS: We describe the phenotypic features of 147 patients with STXBP1-E including 45 previously unreported patients with 33 novel STXBP1 mutations. All patients have intellectual disability (ID), which is mostly severe to profound (88%). Ninety-five percent of patients have epilepsy. While one-third of patients presented with Ohtahara syndrome (21%) or West syndrome (9.5%), the majority has a nonsyndromic early-onset epilepsy and encephalopathy (53%) with epileptic spasms or tonic seizures as main seizure type. We found no correlation between severity of seizures and severity of ID or between mutation type and seizure characteristics or cognitive outcome. Neurologic comorbidities including autistic features and movement disorders are frequent. We also report 2 previously unreported adult patients with prominent extrapyramidal features. CONCLUSION: De novo STXBP1 mutations are among the most frequent causes of epilepsy and encephalopathy. Most patients have severe to profound ID with little correlation among seizure onset, seizure severity, and the degree of ID. Accordingly, we hypothesize that seizure severity and ID present 2 independent dimensions of the STXBP1-E phenotype. STXBP1-E may be conceptualized as a complex neurodevelopmental disorder rather than a primary epileptic encephalopathy.


Asunto(s)
Encefalopatías/genética , Epilepsia/genética , Proteínas Munc18/genética , Trastornos del Neurodesarrollo/genética , Adolescente , Adulto , Encefalopatías/diagnóstico , Encefalopatías/epidemiología , Niño , Preescolar , Epilepsia/diagnóstico , Epilepsia/epidemiología , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación/genética , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/epidemiología , Adulto Joven
11.
Mol Cytogenet ; 8: 57, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26236398

RESUMEN

We report two unrelated patients with overlapping chromosome 2q13 deletions (patient 1 in chr2:111415137-113194067 bp and patient 2 in chr2:110980342-113007823 bp, hg 19). Patient 1 presents with developmental delay, microcephaly and mild dysmorphic facial features, and patient 2 with autism spectrum disorder, borderline cognitive abilities, deficits in attention and executive functions and mild dysmorphic facial features. The mother and maternal grandmother of patient 1 were healthy carriers of the deletion. Previously, 2q13 deletions were reported in 27 patients, and the interpretation of its clinical significance varied. Our findings support that the 2q13 deletion is associated with a developmental delay syndrome manifesting with variable expressivity and reduced penetrance which poses a challenge for genetic counselling as well as the clinical recognition of 2q13 deletion patients.

12.
Eur J Med Genet ; 57(9): 513-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24911659

RESUMEN

2p15p16.1-deletion syndrome was first described in 2007 based on the clinical presentation of two patients. The syndrome is characterized by intellectual disability, autism spectrum disorders, microcephaly, dysmorphic facial features and a variety of congenital organ defects. The precise genotype-phenotype correlation in 2p15-deletion syndrome is not understood. However, greater insight can be obtained by thorough clinical investigation of patients carrying deletions, especially those of small size. We report a 21-year-old male patient with features overlapping the clinical spectrum of the 2p15p16.1-deletion syndrome, such as intellectual disability, dysmorphic facial features, and congenital defects. He carried a 230 kb de novo deletion (chr2:61500346-61733075 bp, hg19), which affects the genes USP34, SNORA70B and XPO1. While there is a lack of functional data on SNORA70B, the involvement of USP34 and XPO1 in the regulation of fundamental developmental processes is well known. We suggest that haploinsufficiency of one or both of these genes is likely to be responsible for the disease in our patient.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 2 , Anomalías Craneofaciales/genética , Haploinsuficiencia , Discapacidad Intelectual/genética , Carioferinas/genética , Receptores Citoplasmáticos y Nucleares/genética , Proteasas Ubiquitina-Específicas/genética , Adulto , Hibridación Genómica Comparativa , Facies , Heterogeneidad Genética , Humanos , Cariotipificación , Masculino , Fenotipo , Adulto Joven , Proteína Exportina 1
13.
Eur J Med Genet ; 56(7): 361-4, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23664928

RESUMEN

We report a 14 year old male patient ascertained for developmental delay, carrying a de novo pericentric inversion on chr(7)(p14.3q22.3). Sequencing revealed that the breakpoints overlap a LTR sequence on 7q22.3 and a LINE on 7p14.3. A TTTAAA motif was found in proximity of the breakpoints on both arms. In addition the sequencing detected several small micro-rearrangements, deletion, duplication, insertion, at the breakpoints. No significant sequence identity exists between the 7p14.3 and 7q22.3 breakpoints. These features at the breakpoint junctions suggest that the inversion was triggered by the TTTAAA motif, LTR and LINE and healed by a Non Homologous End Joining (NHEJ) mechanism. The genes ATXN7L1 and PDE1C are disrupted by the inversion. PDE1C is responsible for the hydrolysis of the second messenger molecules cAMP and cGMP and is highly expressed in the human heart and certain brain regions. In mice, Pde1c is expressed in migrating neuronal cells within the central nervous system during early embryo development. Although neuronal migration disorder was not seen in our patient, this is the first patient described with haploinsufficiency of PDE1C possibly causing developmental delay.


Asunto(s)
Inversión Cromosómica , Cromosomas Humanos Par 7/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/genética , Discapacidades del Desarrollo/genética , Proteínas del Tejido Nervioso/genética , Adolescente , Ataxina-7 , Puntos de Rotura del Cromosoma , Reparación del ADN por Unión de Extremidades , Discapacidades del Desarrollo/diagnóstico , Humanos , Elementos de Nucleótido Esparcido Largo , Masculino , Secuencias Repetidas Terminales
14.
Orphanet J Rare Dis ; 8: 3, 2013 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-23294540

RESUMEN

BACKGROUND: Nineteen patients with deletions in chromosome 6p22-p24 have been published so far. The syndromic phenotype is varied, and includes intellectual disability, behavioural abnormalities, dysmorphic features and structural organ defects. Heterogeneous deletion breakpoints and sizes (1-17 Mb) and overlapping phenotypes have made the identification of the disease causing genes challenging. We suggest JARID2 and ATXN1, both harbored in 6p22.3, as disease causing genes. METHODS AND RESULTS: We describe five unrelated patients with de novo deletions (0.1-4.8 Mb in size) in chromosome 6p22.3-p24.1 detected by aCGH in a cohort of approximately 3600 patients ascertained for neurodevelopmental disorders. Two patients (Patients 4 and 5) carried non-overlapping deletions that were encompassed by the deletions of the remaining three patients (Patients 1-3), indicating the existence of two distinct dosage sensitive genes responsible for impaired cognitive function in 6p22.3 deletion-patients. The smallest region of overlap (SRO I) in Patients 1-4 (189 kb) included the genes JARID2 and DTNBP1, while SRO II in Patients 1-3 and 5 (116 kb) contained GMPR and ATXN1. Patients with deletion of SRO I manifested variable degrees of cognitive impairment, gait disturbance and distinct, similar facial dysmorphic features (prominent supraorbital ridges, deep set eyes, dark infraorbital circles and midface hypoplasia) that might be ascribed to the haploinsufficiency of JARID2. Patients with deletion of SRO II showed intellectual disability and behavioural abnormalities, likely to be caused by the deletion of ATXN1. Patients 1-3 presented with lower cognitive function than Patients 4 and 5, possibly due to the concomitant haploinsufficiency of both ATXN1 and JARID2. The chromatin modifier genes ATXN1 and JARID2 are likely candidates contributing to the clinical phenotype in 6p22-p24 deletion-patients. Both genes exert their effect on the Notch signalling pathway, which plays an important role in several developmental processes. CONCLUSIONS: Patients carrying JARID2 deletion manifested with cognitive impairment, gait disturbance and a characteristic facial appearance, whereas patients with deletion of ATXN1 seemed to be characterized by intellectual disability and behavioural abnormalities. Due to the characteristic facial appearance, JARID2 haploinsufficiency might represent a clinically recognizable neurodevelopmental syndrome.


Asunto(s)
Cromosomas Humanos Par 6 , Haploinsuficiencia , Histonas/metabolismo , Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Complejo Represivo Polycomb 2/genética , Adolescente , Ataxina-1 , Ataxinas , Niño , Preescolar , Hibridación Genómica Comparativa , Femenino , Marcha , Humanos , Cariotipificación , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Eliminación de Secuencia
15.
Eur J Med Genet ; 55(12): 715-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22975012

RESUMEN

The 1q44 deletion syndrome has shown to be a recognizable phenotype with developmental delay, short stature and corpus callosum abnormalities as relatively consistent features. However, the disorder is still clinically heterogeneous and a genotype-phenotype correlation has been challenging to establish. In particular, a delineation of a critical region for the corpus callosum development has turned out to be difficult, and many candidate genes have been proposed. We present here a patient boy with a clinical picture of the 1q44 deletion syndrome, including a thin corpus callosum, and a small de novo 1q44 deletion. The deletion spans a maximum of 163 kb, a region which only contains the two genes FAM36A and HNRNPU. This finding supports the previously suggested hypothesis that the HNRNPU is an essential gene to the development of corpus callosum. However, as patients with deletions outside this interval also have been reported to have corpus callosum abnormalities, other mechanisms are probably also involved. We also identified two conserved non-coding regions in the deleted region of the patient, and speculate that also other elements interfere with the complex interplay and spatiotemporal gene expression during embryonic development.


Asunto(s)
Agenesia del Cuerpo Calloso/genética , Deleción Cromosómica , Cromosomas Humanos Par 1 , Discapacidades del Desarrollo/genética , Convulsiones/genética , Agenesia del Cuerpo Calloso/diagnóstico , Encéfalo/patología , Bandeo Cromosómico , Hibridación Genómica Comparativa , Discapacidades del Desarrollo/diagnóstico , Facies , Estudios de Asociación Genética , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Convulsiones/diagnóstico
16.
Eur J Med Genet ; 55(12): 695-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22986108

RESUMEN

We report a 11 year old male patient ascertained for mild intellectual disability and minor dysmorphic features, carrying a 1 Mb de novo deletion on chromosome 11q13.1q13.2 detected by aCGH. This is the first report of a deletion in this region in a patient presenting with intellectual impairment and mild dysmorphic traits. The 1 Mb deleted area encompasses 47 RefSeq genes, including Cornichon homologue 2 (CNIH2), Cofilin-1 (CFL1) and neuronal PAS domain-containing protein 4 (NPAS4), which are highly expressed in the central nervous system. Knockout of the CNIH2 and CFL1 orthologues in animals results in migration disturbances, while low or no expression of Npas4 in mice results in impairment of memory and learning. These three genes have previously been suggested as candidate genes for neurological disorders.


Asunto(s)
Anomalías Múltiples/genética , Deleción Cromosómica , Cromosomas Humanos Par 11 , Discapacidad Intelectual/genética , Anomalías Múltiples/diagnóstico , Niño , Facies , Deformidades Congénitas de la Mano/genética , Humanos , Discapacidad Intelectual/diagnóstico , Masculino , Fenotipo
17.
Eur J Med Genet ; 53(4): 221-4, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20382277

RESUMEN

We report on a 11-year-old boy investigated for a clinical suspicion of Angelman syndrome (AS) (OMIM 105830) who was found to carry a de novo interstitial deletion of chromosome 15q13.2q13.3. The deletion overlaps the critical region for the newly recognized recurrent 15q13.3 deletion syndrome. This is the first report of a patient with 15q13.3 deletion syndrome with clinical features similar to that of AS, thus broadening the phenotypic spectrum associated with the 15q13.3 microdeletion syndrome.


Asunto(s)
Síndrome de Angelman/genética , Deleción Cromosómica , Cromosomas Humanos Par 15/genética , Adulto , Síndrome de Angelman/patología , Niño , Hibridación Genómica Comparativa , Femenino , Humanos , Hibridación Fluorescente in Situ , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA