Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 109(8): 1500-1519, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35931052

RESUMEN

Identifying causative gene(s) within disease-associated large genomic regions of copy-number variants (CNVs) is challenging. Here, by targeted sequencing of genes within schizophrenia (SZ)-associated CNVs in 1,779 SZ cases and 1,418 controls, we identified three rare putative loss-of-function (LoF) mutations in OTU deubiquitinase 7A (OTUD7A) within the 15q13.3 deletion in cases but none in controls. To tie OTUD7A LoF with any SZ-relevant cellular phenotypes, we modeled the OTUD7A LoF mutation, rs757148409, in human induced pluripotent stem cell (hiPSC)-derived induced excitatory neurons (iNs) by CRISPR-Cas9 engineering. The mutant iNs showed a ∼50% decrease in OTUD7A expression without undergoing nonsense-mediated mRNA decay. The mutant iNs also exhibited marked reduction of dendritic complexity, density of synaptic proteins GluA1 and PSD-95, and neuronal network activity. Congruent with the neuronal phenotypes in mutant iNs, our transcriptomic analysis showed that the set of OTUD7A LoF-downregulated genes was enriched for those relating to synapse development and function and was associated with SZ and other neuropsychiatric disorders. These results suggest that OTUD7A LoF impairs synapse development and neuronal function in human neurons, providing mechanistic insight into the possible role of OTUD7A in driving neuropsychiatric phenotypes associated with the 15q13.3 deletion.


Asunto(s)
Células Madre Pluripotentes Inducidas , Esquizofrenia , Variaciones en el Número de Copia de ADN , Humanos , Neuronas , Esquizofrenia/metabolismo , Sinapsis/metabolismo
2.
Mol Biol Rep ; 51(1): 804, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39001960

RESUMEN

Schizophrenia is a neuropsychiatric disorder characterized by various symptoms such as hallucinations, delusions, and disordered thinking. The etiology of this disease is unknown; however, it has been linked to many microdeletion syndromes that are likely to contribute to the pathology of schizophrenia. In this review we have comprehensively analyzed the role of various microdeletion syndromes, like 3q29, 15q13.3, and 22q11.2, which are known to be involved with schizophrenia. A variety of factors lead to schizophrenia phenotypes, but copy number variants that disrupt gene regulation and impair brain function and cognition are one of the causes that have been identified. Multiple case studies have shown that loss of one or more genes in the microdeletion regions lead to brain activity defects. In this article, we present a coherent paradigm that connects copy number variations (CNVs) to numerous neurological and behavioral abnormalities associated with schizophrenia. It would be helpful in understanding the different aspects of the microdeletions and how they contribute in the pathophysiology of schizophrenia.


Asunto(s)
Deleción Cromosómica , Variaciones en el Número de Copia de ADN , Esquizofrenia , Humanos , Esquizofrenia/genética , Variaciones en el Número de Copia de ADN/genética , Fenotipo , Cromosomas Humanos Par 15/genética , Predisposición Genética a la Enfermedad , Discapacidad Intelectual/genética , Trastornos de los Cromosomas/genética , Discapacidades del Desarrollo , Cromosomas Humanos Par 3 , Convulsiones
3.
Cereb Cortex ; 31(3): 1609-1621, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33123721

RESUMEN

The 15q13.3 deletion is associated with multiple neurodevelopmental disorders including epilepsy, schizophrenia, and autism. The Df(h15q13)/+ mouse model was recently generated that recapitulates several phenotypic features of the human 15q13.3 deletion syndrome (DS). However, the biological substrates underlying these phenotypes in Df(h15q13)/+ mice have not yet been fully characterized. RNA sequencing followed by real-time quantitative PCR, western blotting, liquid chromatography-mass spectrometry, and stereological analysis were employed to dissect the molecular, structural, and neurochemical phenotypes of the medial prefrontal cortex (mPFC) circuits in Df(h15q13)/+ mouse model. Transcriptomic profiling revealed enrichment for astrocyte-specific genes among differentially expressed genes, translated by a decrease in the number of glial fibrillary acidic protein positive cells in mPFC of Df(h15q13)/+ mice compared with wild-type mice. mPFC in Df(h15q13)/+ mice also showed a deficit of the inhibitory presynaptic marker GAD65, in addition to a reduction in dendritic arborization and spine density of pyramidal neurons from layers II/III. mPFC levels of GABA and glutamate neurotransmitters were not different between genotypes. Our results suggest that the 15q13.3 deletion modulates nonneuronal circuits in mPFC and confers molecular and morphometric alterations in the inhibitory and excitatory neurocircuits, respectively. These alterations potentially contribute to the phenotypes accompanied with the 15q13.3DS.


Asunto(s)
Astrocitos/patología , Trastornos de los Cromosomas/patología , Trastornos de los Cromosomas/fisiopatología , Discapacidad Intelectual/patología , Discapacidad Intelectual/fisiopatología , Corteza Prefrontal/patología , Corteza Prefrontal/fisiopatología , Convulsiones/patología , Convulsiones/fisiopatología , Sinapsis/patología , Animales , Deleción Cromosómica , Cromosomas Humanos Par 15 , Modelos Animales de Enfermedad , Masculino , Ratones
4.
BMC Biol ; 19(1): 147, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34320968

RESUMEN

BACKGROUND: Copy number variants (CNVs) linked to genes involved in nervous system development or function are often associated with neuropsychiatric disease. While CNVs involving deletions generally cause severe and highly penetrant patient phenotypes, CNVs leading to duplications tend instead to exhibit widely variable and less penetrant phenotypic expressivity among affected individuals. CNVs located on chromosome 15q13.3 affecting the alpha-7 nicotinic acetylcholine receptor subunit (CHRNA7) gene contribute to multiple neuropsychiatric disorders with highly variable penetrance. However, the basis of such differential penetrance remains uncharacterized. Here, we generated induced pluripotent stem cell (iPSC) models from first-degree relatives with a 15q13.3 duplication and analyzed their cellular phenotypes to uncover a basis for the dissimilar phenotypic expressivity. RESULTS: The first-degree relatives studied included a boy with autism and emotional dysregulation (the affected proband-AP) and his clinically unaffected mother (UM), with comparison to unrelated control models lacking this duplication. Potential contributors to neuropsychiatric impairment were modeled in iPSC-derived cortical excitatory and inhibitory neurons. The AP-derived model uniquely exhibited disruptions of cellular physiology and neurodevelopment not observed in either the UM or unrelated controls. These included enhanced neural progenitor proliferation but impaired neuronal differentiation, maturation, and migration, and increased endoplasmic reticulum (ER) stress. Both the neuronal migration deficit and elevated ER stress could be selectively rescued by different pharmacologic agents. Neuronal gene expression was also dysregulated in the AP, including reduced expression of genes related to behavior, psychological disorders, neuritogenesis, neuronal migration, and Wnt, axonal guidance, and GABA receptor signaling. The UM model instead exhibited upregulated expression of genes in many of these same pathways, suggesting that molecular compensation could have contributed to the lack of neurodevelopmental phenotypes in this model. However, both AP- and UM-derived neurons exhibited shared alterations of neuronal function, including increased action potential firing and elevated cholinergic activity, consistent with increased homomeric CHRNA7 channel activity. CONCLUSIONS: These data define both diagnosis-associated cellular phenotypes and shared functional anomalies related to CHRNA7 duplication that may contribute to variable phenotypic penetrance in individuals with 15q13.3 duplication. The capacity for pharmacological agents to rescue some neurodevelopmental anomalies associated with diagnosis suggests avenues for intervention for carriers of this duplication and other CNVs that cause related disorders.


Asunto(s)
Cromosomas Humanos Par 15 , Variaciones en el Número de Copia de ADN , Receptor Nicotínico de Acetilcolina alfa 7/genética , Cromosomas Humanos Par 15/genética , Humanos , Masculino , Neuronas , Fenotipo
5.
Curr Issues Mol Biol ; 43(2): 1090-1113, 2021 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-34563047

RESUMEN

BACKGROUND: a migraine is a neurological disease. Copy number variation (CNV) is a phenomenon in which parts of the genome are repeated. We investigated the effects of the CNV and gene expression at the location 15q13.3 in the Cholinergic Receptor Nicotinic Alpha 7 Subunit (CHRNA7) gene, which we believe to be effective in the migraine clinic. METHODS: we evaluated changes in CHRNA7 gene expression levels and CNV of 15q13.3 in patients with migraine (n = 102, with aura, n = 43; without aura, n = 59) according to healthy controls (n = 120) by q-PCR. The data obtained were analyzed against the reference telomerase reverse transcriptase (TERT) gene with the double copy number by standard curve analysis. Copy numbers were graded as a normal copy (2), gain (2>), and loss (<2). RESULTS: we analyzed using the 2-ΔΔCT calculation method. The CHRNA7 gene was significantly downregulated in patients (p < 0.05). The analysis of CNV in the CHRNA7 gene was statistically significant in the patient group, according to healthy controls (p < 0.05). A decreased copy number indicates a dosage loss. However, no significant difference was observed among gain, normal, and loss copy numbers and expression values in patients (p > 0.05). The change in CNV was not associated with the downregulation of the CHRNA7 gene. CONCLUSION: Downregulation of the CHRNA7 gene may contribute to the formation of migraine by inactivation of the alpha-7 nicotinic receptor (α7nAChR). The association of CNV gains and losses with migraines will lead to better understanding of the molecular mechanisms and pathogenesis, to better define the disease, to be used as a treatment target.


Asunto(s)
Colinérgicos/farmacología , Variaciones en el Número de Copia de ADN/genética , Trastornos Migrañosos/genética , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Regulación hacia Abajo , Humanos , Receptor Nicotínico de Acetilcolina alfa 7/genética
6.
Am J Hum Genet ; 102(2): 296-308, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29395075

RESUMEN

15q13.3 microdeletion syndrome is characterized by a wide spectrum of neurodevelopmental disorders, including developmental delay, intellectual disability, epilepsy, language impairment, abnormal behaviors, neuropsychiatric disorders, and hypotonia. This syndrome is caused by a deletion on chromosome 15q, which typically encompasses six genes. Here, through studies on OTU deubiquitinase 7A (Otud7a) knockout mice, we identify OTUD7A as a critical gene responsible for many of the cardinal phenotypes associated with 15q13.3 microdeletion syndrome. Otud7a-null mice show reduced body weight, developmental delay, abnormal electroencephalography patterns and seizures, reduced ultrasonic vocalizations, decreased grip strength, impaired motor learning/motor coordination, and reduced acoustic startle. We show that OTUD7A localizes to dendritic spines and that Otud7a-null mice have decreased dendritic spine density compared to their wild-type littermates. Furthermore, frequency of miniature excitatory postsynaptic currents (mEPSCs) is reduced in the frontal cortex of Otud7a-null mice, suggesting a role of Otud7a in regulation of dendritic spine density and glutamatergic synaptic transmission. Taken together, our results suggest decreased OTUD7A dosage as a major contributor to the neurodevelopmental phenotypes associated with 15q13.3 microdeletion syndrome, through the misregulation of dendritic spine density and activity.


Asunto(s)
Trastornos de los Cromosomas/enzimología , Trastornos de los Cromosomas/genética , Enzimas Desubicuitinizantes/genética , Endopeptidasas/genética , Discapacidad Intelectual/enzimología , Discapacidad Intelectual/genética , Convulsiones/enzimología , Convulsiones/genética , Potenciales de Acción , Animales , Secuencia de Bases , Conducta Animal , Deleción Cromosómica , Cromosomas Humanos Par 15/enzimología , Cromosomas Humanos Par 15/genética , Espinas Dendríticas/metabolismo , Modelos Animales de Enfermedad , Electroencefalografía , Endopeptidasas/deficiencia , Epilepsia/enzimología , Epilepsia/genética , Epilepsia/fisiopatología , Femenino , Homocigoto , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Sinapsis/metabolismo
7.
Am J Hum Genet ; 102(2): 278-295, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29395074

RESUMEN

Copy-number variations (CNVs) are strong risk factors for neurodevelopmental and psychiatric disorders. The 15q13.3 microdeletion syndrome region contains up to ten genes and is associated with numerous conditions, including autism spectrum disorder (ASD), epilepsy, schizophrenia, and intellectual disability; however, the mechanisms underlying the pathogenesis of 15q13.3 microdeletion syndrome remain unknown. We combined whole-genome sequencing, human brain gene expression (proteome and transcriptome), and a mouse model with a syntenic heterozygous deletion (Df(h15q13)/+ mice) and determined that the microdeletion results in abnormal development of cortical dendritic spines and dendrite outgrowth. Analysis of large-scale genomic, transcriptomic, and proteomic data identified OTUD7A as a critical gene for brain function. OTUD7A was found to localize to dendritic and spine compartments in cortical neurons, and its reduced levels in Df(h15q13)/+ cortical neurons contributed to the dendritic spine and dendrite outgrowth deficits. Our results reveal OTUD7A as a major regulatory gene for 15q13.3 microdeletion syndrome phenotypes that contribute to the disease mechanism through abnormal cortical neuron morphological development.


Asunto(s)
Trastornos de los Cromosomas/enzimología , Trastornos de los Cromosomas/genética , Enzimas Desubicuitinizantes/fisiología , Endopeptidasas/genética , Discapacidad Intelectual/enzimología , Discapacidad Intelectual/genética , Trastornos del Neurodesarrollo/enzimología , Trastornos del Neurodesarrollo/genética , Convulsiones/enzimología , Convulsiones/genética , Animales , Trastorno del Espectro Autista/genética , Deleción Cromosómica , Cromosomas Humanos Par 15/enzimología , Cromosomas Humanos Par 15/genética , Espinas Dendríticas/metabolismo , Enzimas Desubicuitinizantes/genética , Endopeptidasas/metabolismo , Femenino , Eliminación de Gen , Estudios de Asociación Genética , Humanos , Masculino , Ratones , Fenotipo , Prosencéfalo/patología
8.
Am J Med Genet A ; 185(4): 1182-1186, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33381903

RESUMEN

The heterozygous deletion of 15q13.3 is a recurrently observed microdeletion syndrome associated with a relatively mild phenotype including learning disability and language impairment. In contrast, the homozygous deletion of 15q13.3 is extremely rare and is associated with a much severer phenotype that includes epileptic encephalopathy, profound intellectual disability, and hypotonia. Which of the genes within the deleted interval is responsible for the more severe features when biallelically deleted is currently unknown. Here, we report a patient with profound hypotonia, severe intellectual disability, and seizures who had biallelic loss-of-function variants in OTUD7A: a 15q13.3 deletion including the OTUD7A locus, and a frameshift OTUD7A variant c.1125del, p.(Glu375Aspfs*11). Unexpectedly, both aberrations occurred de novo. Our experiment using Caenorhabditis elegans showed that worms carrying a corresponding homozygous variant in the homolog OTUB-2 exhibited weakened muscle contraction suggestive of aberrant neuromuscular transmission. We concluded that the biallelic complete loss of OTUD7A in humans represents a presumably new autosomal recessive disorder characterized by profound hypotonia, severe intellectual disability, and seizures.


Asunto(s)
Enzimas Desubicuitinizantes/genética , Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Enfermedades de la Unión Neuromuscular/embriología , Animales , Caenorhabditis elegans/genética , Preescolar , Mutación del Sistema de Lectura/genética , Homocigoto , Humanos , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/fisiopatología , Pérdida de Heterocigocidad/genética , Masculino , Contracción Muscular/genética , Contracción Muscular/fisiología , Hipotonía Muscular/fisiopatología , Enfermedades de la Unión Neuromuscular/complicaciones , Enfermedades de la Unión Neuromuscular/genética , Enfermedades de la Unión Neuromuscular/fisiopatología , Convulsiones/complicaciones , Convulsiones/genética , Convulsiones/fisiopatología , Tioléster Hidrolasas/genética
9.
Am J Hum Genet ; 101(6): 874-887, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29129316

RESUMEN

Copy-number variants (CNVs) of chromosome 15q13.3 manifest clinically as neuropsychiatric disorders with variable expressivity. CHRNA7, encoding for the α7 nicotinic acetylcholine receptor (nAChR), has been suggested as a candidate gene for the phenotypes observed. Here, we used induced pluripotent stem cells (iPSCs) and neural progenitor cells (NPCs) derived from individuals with heterozygous 15q13.3 deletions and heterozygous 15q13.3 duplications to investigate the CHRNA7-dependent molecular consequences of the respective CNVs. Unexpectedly, both deletions and duplications lead to decreased α7 nAChR-associated calcium flux. For deletions, this decrease in α7 nAChR-dependent calcium flux is expected due to haploinsufficiency of CHRNA7. For duplications, we found that increased expression of CHRNA7 mRNA is associated with higher expression of nAChR-specific and resident ER chaperones, indicating increased ER stress. This is likely a consequence of inefficient chaperoning and accumulation of α7 subunits in the ER, as opposed to being incorporated into functional α7 nAChRs at the cell membrane. Here, we showed that α7 nAChR-dependent calcium signal cascades are downregulated in both 15q13.3 deletion and duplication NPCs. While it may seem surprising that genomic changes in opposite direction have consequences on downstream pathways that are in similar direction, it aligns with clinical data, which suggest that both individuals with deletions and duplications of 15q13.3 manifest neuropsychiatric disease and cognitive deficits.


Asunto(s)
Señalización del Calcio/genética , Trastornos de los Cromosomas/genética , Variaciones en el Número de Copia de ADN/genética , Estrés del Retículo Endoplásmico/genética , Dosificación de Gen/genética , Células Madre Pluripotentes Inducidas/citología , Discapacidad Intelectual/genética , Células-Madre Neurales/citología , Convulsiones/genética , Receptor Nicotínico de Acetilcolina alfa 7/genética , Adolescente , Niño , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 15/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Receptor Nicotínico de Acetilcolina alfa 7/biosíntesis
10.
Clin Genet ; 97(4): 567-575, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31997314

RESUMEN

Heterozygous microdeletions of chromosome 15q13.3 (MIM: 612001) show incomplete penetrance and are associated with a highly variable phenotype that may include intellectual disability, epilepsy, facial dysmorphism and digit anomalies. Rare patients carrying homozygous deletions show more severe phenotypes including epileptic encephalopathy, hypotonia and poor growth. For years, CHRNA7 (MIM: 118511), was considered the candidate gene that could account for this syndrome. However, recent studies in mouse models have shown that OTUD7A/CEZANNE2 (MIM: 612024), which encodes for an ovarian tumor (OTU) deubiquitinase, should be considered the critical gene responsible for brain dysfunction. In this study, a patient presenting with severe global developmental delay, language impairment and epileptic encephalopathy was referred to our genetics center. Trio exome sequencing (tES) analysis identified a homozygous OTUD7A missense variant (NM_130901.2:c.697C>T), predicted to alter an ultraconserved amino acid, p.(Leu233Phe), lying within the OTU catalytic domain. Its subsequent segregation analysis revealed that the parents, presenting with learning disability, and brother were heterozygous carriers. Biochemical assays demonstrated that proteasome complex formation and function were significantly reduced in patient-derived fibroblasts and in OTUD7A knockout HAP1 cell line. We provide evidence that biallelic pathogenic OTUD7A variation is linked to early-onset epileptic encephalopathy and proteasome dysfunction.


Asunto(s)
Trastornos de los Cromosomas/genética , Enzimas Desubicuitinizantes/genética , Epilepsia/genética , Discapacidad Intelectual/genética , Convulsiones/genética , Animales , Deleción Cromosómica , Trastornos de los Cromosomas/fisiopatología , Cromosomas Humanos Par 15/genética , Epilepsia/fisiopatología , Femenino , Heterocigoto , Homocigoto , Humanos , Discapacidad Intelectual/patología , Discapacidad Intelectual/fisiopatología , Masculino , Ratones , Mutación Missense/genética , Fenotipo , Convulsiones/fisiopatología , Secuenciación del Exoma , Receptor Nicotínico de Acetilcolina alfa 7/genética
11.
Cytogenet Genome Res ; 158(4): 192-198, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31394532

RESUMEN

Prader-Willi syndrome (PWS) and recurrent 15q13.3 microdeletion syndrome can be caused by genomic rearrangements in the complex 15q11q13 chromosomal region. Here, we describe the first female child with PWS and 15q13.3 microdeletion syndrome resulting from an unusual 10.7-Mb deletion from 15pter to 15q13.3 due to an unbalanced de novo 15;19 translocation. The patient presents with hypotonia, microcephaly, developmental delay with lack of speech, intellectual disability, happy demeanor, clinodactyly of the 4th and 5th fingers, and dysmorphic facial features discordant for PWS and consistent with an atypical phenotype.


Asunto(s)
Trastornos de los Cromosomas/complicaciones , Trastornos de los Cromosomas/genética , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/genética , Síndrome de Prader-Willi/complicaciones , Síndrome de Prader-Willi/genética , Convulsiones/complicaciones , Convulsiones/genética , Translocación Genética/genética , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 15/genética , Femenino , Humanos , Recién Nacido
12.
Medicina (Kaunas) ; 55(7)2019 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-31284637

RESUMEN

Congenital sensorineural hearing loss may occur in association with inborn pigmentary defects of the iris, hair, and skin. These conditions, named auditory-pigmentary disorders (APDs), represent extremely heterogeneous hereditary diseases, including Waardenburg syndromes, oculocutaneous albinism, Tietz syndrome, and piebaldism. APDs are part of the neurocristopathies, a group of congenital multisystem disorders caused by an altered development of the neural crest cells, multipotent progenitors of a wide variety of different lineages, including those differentiating into peripheral nervous system glial cells and melanocytes. We report on clinical and genetic findings of two monozygotic twins from a large Albanian family who showed a complex phenotype featured by sensorineural congenital deafness, severe neuropsychiatric impairment, and inborn pigmentary defects of hair and skin. The genetic analyzes identified, in both probands, an unreported co-occurrence of a new heterozygous germline pathogenic variant (c.2484 + 5G > T splicing mutation) in the KIT gene, consistent with the diagnosis of piebaldism, and a heterozygous deletion at chromosome 15q13.3, responsible for the neuropsychiatric impairment. This case represents the first worldwide report of dual locus inherited syndrome in piebald patients affected by a complex auditory-pigmentary multisystem phenotype. Here we also synthesize the clinical and genetic findings of all known neurocristopathies characterized by a hypopigmentary congenital disorder.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Piebaldismo/genética , Femenino , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Piebaldismo/complicaciones , Piebaldismo/fisiopatología , Reacción en Cadena de la Polimerasa/métodos , Gemelos/genética , Adulto Joven
13.
Int J Mol Sci ; 19(11)2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30463371

RESUMEN

Moyamoya angiopathy (MA) is a cerebrovascular disease determining a progressive stenosis of the terminal part of the internal carotid arteries (ICAs) and their proximal branches and the compensatory development of abnormal "moyamoya" vessels. MA occurs as an isolated cerebral angiopathy (so-called moyamoya disease) or in association with various conditions (moyamoya syndromes) including several heritable conditions such as Down syndrome, neurofibromatosis type 1 and other genomic defects. Although the mechanism that links MA to these genetic syndromes is still unclear, it is believed that the involved genes may contribute to the disease susceptibility. Herein, we describe the case of a 43 years old woman with bilateral MA and peculiar facial characteristics, having a 484-kb microduplication of the chromosomal region 15q13.3 and a previously unreported 786 kb microdeletion in 18q21.32. This patient may have a newly-recognized genetic syndrome associated with MA. Although the relationship between these genetic variants and MA is unclear, our report would contribute to widening the genetic scenario of MA, in which not only genic mutation, but also genome unbalances are possible candidate susceptibility factors.


Asunto(s)
Deleción Cromosómica , Duplicación Cromosómica , Cromosomas Humanos Par 15/genética , Cromosomas Humanos Par 18/genética , Enfermedad de Moyamoya/genética , Adulto , Femenino , Humanos , Imagen por Resonancia Magnética , Enfermedad de Moyamoya/diagnóstico por imagen
14.
Am J Med Genet A ; 173(5): 1251-1256, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28345786

RESUMEN

ZBTB18 has been proposed as candidate gene for microcephaly and abnormalities of the corpus callosum based on overlapping microdeletions of 1q43q44. More recently, de novo mutations of ZBTB18 have been identified in patients with syndromic and non-syndromic intellectual disability. Heterozygous microdeletions of 15q13.3 encompassing the candidate gene CHRNA7 are associated with developmental delay or intellectual disability with speech problems, hypotonia, and seizures. They are characterized by significant variability and reduced penetrance. We report on a patient with a de novo ZBTB18 nonsense mutation and a de novo 15q13.3 microdeletion, both in a heterozygous state, identified by next generation sequencing and array-CGH. The 6-year-old girl showed global developmental delay, absent speech, therapy-refractory seizures, ataxia, muscular hypotonia, and discrete facial dysmorphisms. Almost all of these features have been reported for both genetic aberrations, but the severity could hardly been explained by the microdeletion 15q13.3 alone. We assume an additive effect of haploinsufficiency of ZBTB18 and CHRNA7 in our patient. Assembling the features of our patient and the published patients, we noted that only one of them showed mild anomalies of the corpus callosum. Moreover, we hypothesize that nonsense mutations of ZBTB18 are associated with a more severe phenotype than missense mutations. This report indicates that haploinsufficiency of additional genes beside ZBTB18 causes the high frequency of corpus callosum anomalies in patients with microdeletions of 1q43q44 and underlines the importance of an NGS-based molecular diagnostic in complex phenotypes.


Asunto(s)
Agenesia del Cuerpo Calloso/genética , Trastornos de los Cromosomas/genética , Discapacidad Intelectual/genética , Proteínas Represoras/genética , Convulsiones/genética , Receptor Nicotínico de Acetilcolina alfa 7/genética , Agenesia del Cuerpo Calloso/fisiopatología , Deleción Cromosómica , Trastornos de los Cromosomas/patología , Cromosomas Humanos Par 15/genética , Codón sin Sentido , Cuerpo Calloso/fisiopatología , Femenino , Haploinsuficiencia/genética , Humanos , Discapacidad Intelectual/patología , Discapacidad Intelectual/fisiopatología , Convulsiones/patología
15.
J Neurosci ; 35(49): 16282-94, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26658876

RESUMEN

The chromosome 15q13.3 microdeletion is a pathogenic copy number variation conferring epilepsy, intellectual disability, schizophrenia, and autism spectrum disorder (ASD). We generated mice carrying a deletion of 1.2 Mb homologous to the 15q13.3 microdeletion in human patients. Here, we report that mice with a heterozygous deletion on a C57BL/6 background (D/+ mice) demonstrated phenotypes including enlarged/heavier brains (macrocephaly) with enlarged lateral ventricles, decreased social interactions, increased repetitive grooming behavior, reduced ultrasonic vocalizations, decreased auditory-evoked gamma band EEG, and reduced event-related potentials. D/+ mice had normal body weight, activity levels, sensory gating, and cognitive abilities and no signs of epilepsy/seizures. Our results demonstrate that D/+ mice represent ASD-related phenotypes associated with 15q13.3 microdeletion syndrome. Further investigations using this chromosome-engineered mouse model may uncover the common mechanism(s) underlying ASD and other neurodevelopmental/psychiatric disorders representing the 15q13.3 microdeletion syndrome, including epilepsy, intellectual disability, and schizophrenia. SIGNIFICANCE STATEMENT: Recently discovered pathologic copy number variations (CNVs) from patients with neurodevelopmental/psychiatric disorders show very strong penetrance and thus are excellent candidates for mouse models of disease that can mirror the human genetic conditions with high fidelity. A 15q13.3 microdeletion in humans results in a range of neurodevelopmental/psychiatric disorders, including epilepsy, intellectual disability, schizophrenia, and autism spectrum disorder (ASD). The disorders conferred by a 15q13.3 microdeletion also have overlapping genetic architectures and comorbidity in other patient populations such as those with epilepsy and schizophrenia/psychosis, as well as schizophrenia and ASD. We generated mice carrying a deletion of 1.2 Mb homologous to the 15q13.3 microdeletion in human patients, which allowed us to investigate the potential causes of neurodevelopmental/psychiatric disorders associated with the CNV.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Encéfalo/patología , Trastornos de los Cromosomas/fisiopatología , Discapacidad Intelectual/fisiopatología , Convulsiones/fisiopatología , Animales , Ansiedad/etiología , Aprendizaje por Asociación/fisiología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/patología , Cromosomas Humanos Par 15/genética , Discriminación en Psicología/efectos de los fármacos , Discriminación en Psicología/fisiología , Potenciales Evocados/fisiología , Femenino , Expresión Génica/fisiología , Aseo Animal/fisiología , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Relaciones Interpersonales , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pilocarpina/farmacología , Convulsiones/genética , Convulsiones/patología , Olfato/fisiología , Vocalización Animal/fisiología
16.
Am J Med Genet A ; 170(11): 2895-2904, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27411073

RESUMEN

Chromosomal microarray is an increasingly utilized diagnostic test, particularly in the pediatric setting. However, the clinical significance of copy number variants detected by this technology is not always understood, creating uncertainties in interpreting and communicating results. The aim of this study was to explore parents' experiences of an uncertain microarray result for their child. This research utilized a qualitative approach with a phenomenological methodology. Semi-structured interviews were conducted with nine parents of eight children who received an uncertain microarray result for their child, either a 16p11.2 microdeletion or 15q13.3 microdeletion. Interviews were transcribed verbatim and thematic analysis was used to identify themes within the data. Participants were unprepared for the abnormal test result. They had a complex perception of the extent of their child's condition and a mixed understanding of the clinical relevance of the result, but were accepting of the limitations of medical knowledge, and appeared to have adapted to the result. The test result was empowering for parents in terms of access to medical and educational services; however, they articulated significant unmet support needs. Participants expressed hope for the future, in particular that more information would become available over time. This research has demonstrated that parents of children who have an uncertain microarray result appeared to adapt to uncertainty and limited availability of information and valued honesty and empathic ongoing support from health professionals. Genetic health professionals are well positioned to provide such support and aid patients' and families' adaptation to their situation as well as promote empowerment. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Aberraciones Cromosómicas , Pruebas Genéticas , Análisis de Secuencia por Matrices de Oligonucleótidos , Padres/psicología , Incertidumbre , Adulto , Anciano , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Encuestas y Cuestionarios
17.
Am J Med Genet B Neuropsychiatr Genet ; 171(6): 777-83, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26968334

RESUMEN

We report two cases of paternally inherited 15q13.3 duplications in carriers diagnosed with childhood-onset schizophrenia (COS), a rare neurodevelopmental disorder of proposed polygenic origin with onset in children before age 13. This study documents that the 15q13.3 deletion and duplication exhibit pathogenicity for COS, with both copy number variants (CNVs) sharing a disrupted CHRNA7 gene. CHRNA7 encodes the neuronal alpha7 nicotinic acetylcholine receptor (α7nAChR) and is a candidate gene that has been suggested as a pathophysiological process mediating adult-onset schizophrenia (AOS) and other neurodevelopmental disorders. These results support the incomplete penetrance and variable expressivity of this CNV and represent the first report of 15q13.3 duplication carriers exhibiting COS. Published 2016. This article is a U.S. Government work and is in the public domain in the USA. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics published by Wiley Periodicals, Inc.


Asunto(s)
Esquizofrenia Infantil/genética , Esquizofrenia Infantil/psicología , Niño , Deleción Cromosómica , Cromosomas Humanos Par 15/genética , Variaciones en el Número de Copia de ADN , Femenino , Humanos , Discapacidad Intelectual/genética , Masculino , Linaje , Esquizofrenia/genética
18.
Am J Med Genet A ; 167A(4): 715-23, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25655306

RESUMEN

Chromosome 15q13.3 recurrent microdeletions are causally associated with a wide range of phenotypes, including autism spectrum disorder (ASD), seizures, intellectual disability, and other psychiatric conditions. Whether the reciprocal microduplication is pathogenic is less certain. CHRNA7, encoding for the alpha7 subunit of the neuronal nicotinic acetylcholine receptor, is considered the likely culprit gene in mediating neurological phenotypes in 15q13.3 deletion cases. To assess if CHRNA7 rare variants confer risk to ASD, we performed copy number variant analysis and Sanger sequencing of the CHRNA7 coding sequence in a sample of 135 ASD cases. Sequence variation in this gene remains largely unexplored, given the existence of a fusion gene, CHRFAM7A, which includes a nearly identical partial duplication of CHRNA7. Hence, attempts to sequence coding exons must distinguish between CHRNA7 and CHRFAM7A, making next-generation sequencing approaches unreliable for this purpose. A CHRNA7 microduplication was detected in a patient with autism and moderate cognitive impairment; while no rare damaging variants were identified in the coding region, we detected rare variants in the promoter region, previously described to functionally reduce transcription. This study represents the first sequence variant analysis of CHRNA7 in a sample of idiopathic autism.


Asunto(s)
Trastorno del Espectro Autista/genética , Receptor Nicotínico de Acetilcolina alfa 7/genética , Adolescente , Estudios de Casos y Controles , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Polimorfismo de Nucleótido Simple
19.
Am J Med Genet A ; 164A(6): 1537-44, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24668847

RESUMEN

The 15q13.3 heterozygous microdeletion is a fairly common microdeletion syndrome with marked clinical variability and incomplete penetrance. The average size of the deletion, which comprises six genes including CHRNA7, is 1.5 Mb. CHRNA7 has been identified as the gene responsible for the neurological phenotype in this microdeletion syndrome. Only seven patients with a homozygous microdeletion that includes at least CHRNA7, and is inherited from both parents have been described in the literature. The aim of this study was to further describe the distinctive eye manifestations from the analysis in the three French patients diagnosed with the classical 1.5 Mb homozygous microdeletion. Patients' ages ranged from 30 months to 9 years, and included one sib pair. They all displayed a remarkably severe identifiable clinical phenotype that included congenital blindness and convulsive encephalopathy with inconstant abnormal movements. The ophthalmological examination revealed a lack of eye tracking, optic nerve pallor, an immature response with increased latencies with no response to the checkerboard stimulations at the visual evoked potential examination, and a distinctive retina dystrophy with a negative electroretinogram in which the "b" wave was smaller than the "a" wave after a dark adapted pupil and bright flash in all patients. Clear genotype-phenotype correlations emerged, showing that this eye phenotype was secondary to homozygous deletion of TRPM1, the gene responsible for autosomal recessive congenital stationary night blindness. The main differential diagnosis is ceroid lipofuscinosis.


Asunto(s)
Ceguera/genética , Trastornos de los Cromosomas/genética , Discapacidad Intelectual/genética , Lipofuscinosis Ceroideas Neuronales/genética , Convulsiones/genética , Canales Catiónicos TRPM/genética , Receptor Nicotínico de Acetilcolina alfa 7/genética , Niño , Preescolar , Deleción Cromosómica , Trastornos de los Cromosomas/patología , Cromosomas Humanos Par 15/genética , Electrorretinografía , Ojo/patología , Anomalías del Ojo/genética , Enfermedades Hereditarias del Ojo/genética , Femenino , Estudios de Asociación Genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Humanos , Discapacidad Intelectual/patología , Masculino , Miopía/genética , Lipofuscinosis Ceroideas Neuronales/patología , Ceguera Nocturna/genética , Nervio Óptico/anomalías , Distrofias Retinianas/genética , Convulsiones/patología
20.
Am J Med Genet A ; 164A(7): 1815-20, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24700535

RESUMEN

Homozygous or compound heterozygous microdeletion of 15q13.3 region is a rare but clinically recognizable syndrome manifested by profound intellectual disability, muscular hypotonia, intractable seizures, and visual impairment. We identified a compound heterozygous 15q13.3 microdeletion in a 23-month-old girl with global developmental delay, generalized muscular hypotonia, and visual dysfunction. The larger deletion was approximately 1.28 Mb in size and contained seven genes including the TRPM1 and CHRNA7, while the smaller deletion was estimated to be 410 Kb in size and contained only CHRNA7. Compound heterozygous 15q13.3 microdeletion is extremely rare and to the best of our knowledge only two such patients have been reported in literature thus far. The findings in our patient suggest that the pathogenesis of visual dysfunction, which is a consistent finding in homozygous/compound heterozygous 15q13.3 microdeletion depends upon the size of microdeletion. Homozygous loss of TRPM1 likely causes retinal dysfunction while homozygous loss of CHRNA7 alone may lead to visual impairment by cortical mechanisms.


Asunto(s)
Trastornos de los Cromosomas/diagnóstico , Trastornos de los Cromosomas/genética , Estudios de Asociación Genética , Heterocigoto , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Fenotipo , Convulsiones/diagnóstico , Convulsiones/genética , Deleción Cromosómica , Cromosomas Humanos Par 15/genética , Hibridación Genómica Comparativa , Discapacidades del Desarrollo , Humanos , Hibridación Fluorescente in Situ , Lactante , Masculino , Hipotonía Muscular , Trastornos de la Visión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA