Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(16): e2214997120, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37043537

RESUMEN

While somatic variants of TRAF7 (Tumor necrosis factor receptor-associated factor 7) underlie anterior skull-base meningiomas, here we report the inherited mutations of TRAF7 that cause congenital heart defects. We show that TRAF7 mutants operate in a dominant manner, inhibiting protein function via heterodimerization with wild-type protein. Further, the shared genetics of the two disparate pathologies can be traced to the common origin of forebrain meninges and cardiac outflow tract from the TRAF7-expressing neural crest. Somatic and inherited mutations disrupt TRAF7-IFT57 interactions leading to cilia degradation. TRAF7-mutant meningioma primary cultures lack cilia, and TRAF7 knockdown causes cardiac, craniofacial, and ciliary defects in Xenopus and zebrafish, suggesting a mechanistic convergence for TRAF7-driven meningiomas and developmental heart defects.


Asunto(s)
Cardiopatías Congénitas , Neoplasias Meníngeas , Meningioma , Animales , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cardiopatías Congénitas/genética , Neoplasias Meníngeas/genética , Meningioma/genética , Meningioma/patología , Mutación , Cráneo/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Humanos , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral
2.
Genet Med ; 25(1): 135-142, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36399134

RESUMEN

PURPOSE: Protein arginine methyltransferase 7 (PRMT7) is a member of a family of enzymes that catalyzes the methylation of arginine residues on several protein substrates. Biallelic pathogenic PRMT7 variants have previously been associated with a syndromic neurodevelopmental disorder characterized by short stature, brachydactyly, intellectual developmental disability, and seizures. To our knowledge, no comprehensive study describes the detailed clinical characteristics of this syndrome. Thus, we aim to delineate the phenotypic spectrum of PRMT7-related disorder. METHODS: We assembled a cohort of 51 affected individuals from 39 different families, gathering clinical information from 36 newly described affected individuals and reviewing data of 15 individuals from the literature. RESULTS: The main clinical characteristics of the PRMT7-related syndrome are short stature, mild to severe developmental delay/intellectual disability, hypotonia, brachydactyly, and distinct facial morphology, including bifrontal narrowing, prominent supraorbital ridges, sparse eyebrows, short nose with full/broad nasal tip, thin upper lip, full and everted lower lip, and a prominent or squared-off jaw. Additional variable findings include seizures, obesity, nonspecific magnetic resonance imaging abnormalities, eye abnormalities (i.e., strabismus or nystagmus), and hearing loss. CONCLUSION: This study further delineates and expands the molecular, phenotypic spectrum and natural history of PRMT7-related syndrome characterized by a neurodevelopmental disorder with skeletal, growth, and endocrine abnormalities.


Asunto(s)
Braquidactilia , Enanismo , Discapacidad Intelectual , Anomalías Musculoesqueléticas , Trastornos del Neurodesarrollo , Humanos , Trastornos del Neurodesarrollo/genética , Discapacidad Intelectual/genética , Enanismo/genética , Obesidad/genética , Fenotipo , Proteína-Arginina N-Metiltransferasas/genética
3.
Mol Genet Genomic Med ; 10(6): e1944, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35481623

RESUMEN

BACKGROUNDS: While many studies agree that consanguinity increases the rate of congenital heart disease (CHD), few genome analyses have been conducted with consanguineous CHD cohorts. METHODS: We recruited 73 CHD probands from consanguineous families in Turkey and used whole-exome sequencing (WES) to identify genetic lesions in these patients. RESULTS: On average, each patient had 6.95 rare damaging homozygous variants, 0.68 of which are loss-of-function (LoF) variants. Seven patients (9.6%) carried damaging homozygous variants in five causal CHD genes. Six of those patients exhibited laterality defects (six HTX and one D-TGA). Three additional patients (4.1%) harbored other types of CHD-associated genomic alterations, which overall explained 13.7% (10/73) of the cohort. The contribution from recessive variants in our cohort is higher than 1.8% reported from a cohort of 2871 CHD subjects where 5.6% of subjects met the criteria for consanguinity. CONCLUSIONS: Our WES screen of a Turkish consanguineous population with structural CHD revealed its unique genetic architecture. Six of seven damaging homozygous variants in CHD causal genes occur in the setting of laterality defects implies a strong contribution from consanguinity to these defects specifically. Our study thus provided valuable information about the genetic landscape of CHD in consanguineous families in Turkey.


Asunto(s)
Cardiopatías Congénitas , Consanguinidad , Cardiopatías Congénitas/genética , Humanos , Mutación , Turquía , Secuenciación del Exoma
5.
Nat Med ; 27(12): 2165-2175, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34887573

RESUMEN

Intracranial aneurysm (IA) rupture leads to subarachnoid hemorrhage, a sudden-onset disease that often causes death or severe disability. Although genome-wide association studies have identified common genetic variants that increase IA risk moderately, the contribution of variants with large effect remains poorly defined. Using whole-exome sequencing, we identified significant enrichment of rare, deleterious mutations in PPIL4, encoding peptidyl-prolyl cis-trans isomerase-like 4, in both familial and index IA cases. Ppil4 depletion in vertebrate models causes intracerebral hemorrhage, defects in cerebrovascular morphology and impaired Wnt signaling. Wild-type, but not IA-mutant, PPIL4 potentiates Wnt signaling by binding JMJD6, a known angiogenesis regulator and Wnt activator. These findings identify a novel PPIL4-dependent Wnt signaling mechanism involved in brain-specific angiogenesis and maintenance of cerebrovascular integrity and implicate PPIL4 gene mutations in the pathogenesis of IA.


Asunto(s)
Encéfalo/irrigación sanguínea , Ciclofilinas/genética , Aneurisma Intracraneal/genética , Neovascularización Patológica/genética , Proteínas de Unión al ARN/genética , Ciclofilinas/fisiología , Humanos , Mutación , Proteínas de Unión al ARN/fisiología , Secuenciación del Exoma , Vía de Señalización Wnt/fisiología
6.
Stem Cell Reports ; 16(5): 1228-1244, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33891865

RESUMEN

Emerging technologies in stem cell engineering have produced sophisticated organoid platforms by controlling stem cell fate via biomaterial instructive cues. By micropatterning and differentiating human induced pluripotent stem cells (hiPSCs), we have engineered spatially organized cardiac organoids with contracting cardiomyocytes in the center surrounded by stromal cells distributed along the pattern perimeter. We investigated how geometric confinement directed the structural morphology and contractile functions of the cardiac organoids and tailored the pattern geometry to optimize organoid production. Using modern data-mining techniques, we found that pattern sizes significantly affected contraction functions, particularly in the parameters related to contraction duration and diastolic functions. We applied cardiac organoids generated from 600 µm diameter circles as a developmental toxicity screening assay and quantified the embryotoxic potential of nine pharmaceutical compounds. These cardiac organoids have potential use as an in vitro platform for studying organoid structure-function relationships, developmental processes, and drug-induced cardiac developmental toxicity.


Asunto(s)
Desarrollo Embrionario , Corazón/embriología , Organoides/embriología , Ingeniería de Tejidos , Pruebas de Toxicidad , Señalización del Calcio , Diferenciación Celular , Corazón/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Organoides/fisiología
7.
Sci Rep ; 10(1): 20335, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33230135

RESUMEN

Brown adipose tissue (BAT) is the primary non-shivering thermogenesis organ in mammals, which plays essential roles in maintaining the body temperature of infants. Although the development of BAT during embryogenesis has been well addressed in rodents, how BAT grows after birth remains unknown. Using mouse interscapular BAT (iBAT) as an example, we studied the cellular and molecular mechanisms that regulate postnatal BAT growth. By analyzing the developmental dynamics of brown adipocytes (BAs), we found that BAs size enlargement partially accounts for iBAT growth. By investigating the BAs cell cycle activities, we confirmed the presence of proliferative BAs in the neonatal mice. Two weeks after birth, most of the BAs exit cell cycle, and the further expansion of the BAT was mainly due to lipogenesis-mediated BAs volume increase. Microscopy and fluorescence-activated cell sorting analyses suggest that most BAs are mononuclear and diploid. Based on the developmental dynamics of brown adipocytes, we propose that the murine iBAT has two different growth phases between birth and weaning: increase of BAs size and number in the first two weeks, and BAs size enlargement thereafter. In summary, our data demonstrate that both lipogenesis and proliferation of BAs contribute to postnatal iBAT growth in mice.


Asunto(s)
Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/crecimiento & desarrollo , Aumento de la Célula , Proliferación Celular/fisiología , Lipogénesis/fisiología , Animales , Animales Recién Nacidos , Tamaño de la Célula , Células Cultivadas , Metabolismo Energético/fisiología , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Obesidad Infantil/metabolismo , Termogénesis/fisiología , Triglicéridos/metabolismo
8.
Am J Med Genet A ; 176(2): 421-425, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29226631

RESUMEN

We here describe novel compound heterozygous missense variants, NM_133443:c.[400C>T] and NM_133443:[1435G>A], in the glutamic-pyruvic transaminase 2 (GPT2) gene in a large consanguineous family with two affected siblings diagnosed with microcephaly intellectual disability and developmental delay (IDD). In addition to these clinical phenotypes, the male sibling has spastic paraplegia, and the female sibling has epilepsy. Their four extended family members have IDD and microcephaly. Both of these variants, c.400C>T (p.R134C) and c.1435G>A (p.V479M), reside in the pyridoxal phosphate-dependent aminotransferase domain. The missense variants affect highly conserved amino acids and are classified to be disease-causing by meta-SVM. The candidate variants were not found in the Exome Aggregation Consortium (ExAC) dataset or in dbSNP. Both GPT2 variants have an allele frequency of 0% (0/ ∼ 600) in the whole-exome sequenced Turkish cohort. Upon Sanger sequencing, we confirmed these mutations in all affected family members and showed that the index patient and his affected sister inherited one mutant allele from each unaffected parent. To the best of our knowledge, this is the first family in which a novel compound heterozygous variant in the GPT2 gene was identified.


Asunto(s)
Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Transaminasas/genética , Adolescente , Niño , Preescolar , Consanguinidad , Discapacidades del Desarrollo/fisiopatología , Epilepsia/genética , Epilepsia/fisiopatología , Exoma/genética , Femenino , Heterocigoto , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Microcefalia/fisiopatología , Persona de Mediana Edad , Mutación , Paraplejía/genética , Paraplejía/fisiopatología , Linaje , Fenotipo , Hermanos , Secuenciación del Exoma
9.
Mol Autism ; 8: 19, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28392909

RESUMEN

BACKGROUND: Childhood disintegrative disorder (CDD) is a rare form of autism spectrum disorder (ASD) of unknown etiology. It is characterized by late-onset regression leading to significant intellectual disability (ID) and severe autism. Although there are phenotypic differences between CDD and other forms of ASD, it is unclear if there are neurobiological differences. METHODS: We pursued a multidisciplinary study of CDD (n = 17) and three comparison groups: low-functioning ASD (n = 12), high-functioning ASD (n = 50), and typically developing (n = 26) individuals. We performed whole-exome sequencing (WES), copy number variant (CNV), and gene expression analyses of CDD and, on subsets of each cohort, non-sedated functional magnetic resonance imaging (fMRI) while viewing socioemotional (faces) and non-socioemotional (houses) stimuli and eye tracking while viewing emotional faces. RESULTS: We observed potential differences between CDD and other forms of ASD. WES and CNV analyses identified one or more rare de novo, homozygous, and/or hemizygous (mother-to-son transmission on chrX) variants for most probands that were not shared by unaffected sibling controls. There were no clearly deleterious variants or highly recurrent candidate genes. Candidate genes that were found to be most conserved at variant position and most intolerant of variation, such as TRRAP, ZNF236, and KIAA2018, play a role or may be involved in transcription. Using the human BrainSpan transcriptome dataset, CDD candidate genes were found to be more highly expressed in non-neocortical regions than neocortical regions. This expression profile was similar to that of an independent cohort of ASD probands with regression. The non-neocortical regions overlapped with those identified by fMRI as abnormally hyperactive in response to viewing faces, such as the thalamus, cerebellum, caudate, and hippocampus. Eye-tracking analysis showed that, among individuals with ASD, subjects with CDD focused on eyes the most when shown pictures of faces. CONCLUSIONS: Given that cohort sizes were limited by the rarity of CDD, and the challenges of conducting non-sedated fMRI and eye tracking in subjects with ASD and significant ID, this is an exploratory study designed to investigate the neurobiological features of CDD. In addition to reporting the first multimodal analysis of CDD, a combination of fMRI and eye-tracking analyses are being presented for the first time for low-functioning individuals with ASD. Our results suggest differences between CDD and other forms of ASD on the neurobiological as well as clinical level.


Asunto(s)
Trastorno del Espectro Autista/genética , Encéfalo/fisiopatología , Cromosomas Humanos X/química , Discapacidad Intelectual/genética , Transcriptoma , Proteínas Adaptadoras Transductoras de Señales/genética , Trastorno del Espectro Autista/diagnóstico por imagen , Trastorno del Espectro Autista/fisiopatología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Mapeo Encefálico , Estudios de Casos y Controles , Niño , Preescolar , Variaciones en el Número de Copia de ADN , Progresión de la Enfermedad , Femenino , Expresión Génica , Humanos , Discapacidad Intelectual/diagnóstico por imagen , Discapacidad Intelectual/fisiopatología , Imagen por Resonancia Magnética , Masculino , Herencia Materna , Proteínas Nucleares/genética , Fenotipo , Polimorfismo Genético , Índice de Severidad de la Enfermedad , Hermanos , Factores de Transcripción/genética , Secuenciación del Exoma
10.
Nat Genet ; 48(10): 1253-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27548314

RESUMEN

RNA polymerase II mediates the transcription of all protein-coding genes in eukaryotic cells, a process that is fundamental to life. Genomic mutations altering this enzyme have not previously been linked to any pathology in humans, which is a testament to its indispensable role in cell biology. On the basis of a combination of next-generation genomic analyses of 775 meningiomas, we report that recurrent somatic p.Gln403Lys or p.Leu438_His439del mutations in POLR2A, which encodes the catalytic subunit of RNA polymerase II (ref. 1), hijack this essential enzyme and drive neoplasia. POLR2A mutant tumors show dysregulation of key meningeal identity genes, including WNT6 and ZIC1/ZIC4. In addition to mutations in POLR2A, NF2, SMARCB1, TRAF7, KLF4, AKT1, PIK3CA, and SMO, we also report somatic mutations in AKT3, PIK3R1, PRKAR1A, and SUFU in meningiomas. Our results identify a role for essential transcriptional machinery in driving tumorigenesis and define mutually exclusive meningioma subgroups with distinct clinical and pathological features.


Asunto(s)
Neoplasias Meníngeas/genética , Meningioma/genética , Mutación , ARN Polimerasa II/genética , Dominio Catalítico/genética , Cromosomas Humanos Par 22 , Estudios de Cohortes , Análisis Mutacional de ADN , Elementos de Facilitación Genéticos , Exoma , Regulación Neoplásica de la Expresión Génica , Genotipo , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Meníngeas/clasificación , Meningioma/clasificación , Neurofibromina 2/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética
11.
Am J Hum Genet ; 99(2): 501-10, 2016 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-27453578

RESUMEN

Cell division terminates with cytokinesis and cellular separation. Autosomal-recessive primary microcephaly (MCPH) is a neurodevelopmental disorder characterized by a reduction in brain and head size at birth in addition to non-progressive intellectual disability. MCPH is genetically heterogeneous, and 16 loci are known to be associated with loss-of-function mutations predominantly affecting centrosomal-associated proteins, but the multiple roles of centrosomes in cellular function has left questions about etiology. Here, we identified three families affected by homozygous missense mutations in CIT, encoding citron rho-interacting kinase (CIT), which has established roles in cytokinesis. All mutations caused substitution of conserved amino acid residues in the kinase domain and impaired kinase activity. Neural progenitors that were differentiated from induced pluripotent stem cells (iPSCs) derived from individuals with these mutations exhibited abnormal cytokinesis with delayed mitosis, multipolar spindles, and increased apoptosis, rescued by CRISPR/Cas9 genome editing. Our results highlight the importance of cytokinesis in the pathology of primary microcephaly.


Asunto(s)
Alelos , Citocinesis/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Microcefalia/genética , Microcefalia/patología , Mitosis/genética , Mutación Missense/genética , Proteínas Serina-Treonina Quinasas/genética , Apoptosis/genética , Centrosoma/metabolismo , Niño , Preescolar , Femenino , Genes Recesivos , Humanos , Recién Nacido , Masculino , Linaje
12.
Nat Genet ; 48(1): 59-66, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26618343

RESUMEN

Gliomas represent approximately 30% of all central nervous system tumors and 80% of malignant brain tumors. To understand the molecular mechanisms underlying the malignant progression of low-grade gliomas with mutations in IDH1 (encoding isocitrate dehydrogenase 1), we studied paired tumor samples from 41 patients, comparing higher-grade, progressed samples to their lower-grade counterparts. Integrated genomic analyses, including whole-exome sequencing and copy number, gene expression and DNA methylation profiling, demonstrated nonlinear clonal expansion of the original tumors and identified oncogenic pathways driving progression. These include activation of the MYC and RTK-RAS-PI3K pathways and upregulation of the FOXM1- and E2F2-mediated cell cycle transitions, as well as epigenetic silencing of developmental transcription factor genes bound by Polycomb repressive complex 2 in human embryonic stem cells. Our results not only provide mechanistic insight into the genetic and epigenetic mechanisms driving glioma progression but also identify inhibition of the bromodomain and extraterminal (BET) family as a potential therapeutic approach.


Asunto(s)
Neoplasias del Sistema Nervioso Central/genética , Glioma/genética , Isocitrato Deshidrogenasa/genética , Mutación , Neoplasias del Sistema Nervioso Central/patología , Metilación de ADN , Células Madre Embrionarias/metabolismo , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Genes myc , Glioma/patología , Humanos , Isocitrato Deshidrogenasa/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo
13.
Neuron ; 87(6): 1215-1233, 2015 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-26402605

RESUMEN

Analysis of de novo CNVs (dnCNVs) from the full Simons Simplex Collection (SSC) (N = 2,591 families) replicates prior findings of strong association with autism spectrum disorders (ASDs) and confirms six risk loci (1q21.1, 3q29, 7q11.23, 16p11.2, 15q11.2-13, and 22q11.2). The addition of published CNV data from the Autism Genome Project (AGP) and exome sequencing data from the SSC and the Autism Sequencing Consortium (ASC) shows that genes within small de novo deletions, but not within large dnCNVs, significantly overlap the high-effect risk genes identified by sequencing. Alternatively, large dnCNVs are found likely to contain multiple modest-effect risk genes. Overall, we find strong evidence that de novo mutations are associated with ASD apart from the risk for intellectual disability. Extending the transmission and de novo association test (TADA) to include small de novo deletions reveals 71 ASD risk loci, including 6 CNV regions (noted above) and 65 risk genes (FDR ≤ 0.1).


Asunto(s)
Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/genética , Sitios Genéticos/genética , Variación Genética/genética , Mapas de Interacción de Proteínas/genética , Femenino , Humanos , Masculino
14.
Eur J Hum Genet ; 23(2): 165-72, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24781755

RESUMEN

The combination of family-based linkage analysis with high-throughput sequencing is a powerful approach to identifying rare genetic variants that contribute to genetically heterogeneous syndromes. Using parametric multipoint linkage analysis and whole exome sequencing, we have identified a gene responsible for microcephaly (MCP), severe visual impairment, intellectual disability, and short stature through the mapping of a homozygous nonsense alteration in a multiply-affected consanguineous family. This gene, DIAPH1, encodes the mammalian Diaphanous-related formin (mDia1), a member of the diaphanous-related formin family of Rho effector proteins. Upon the activation of GTP-bound Rho, mDia1 generates linear actin filaments in the maintenance of polarity during adhesion, migration, and division in immune cells and neuroepithelial cells, and in driving tangential migration of cortical interneurons in the rodent. Here, we show that patients with a homozygous nonsense DIAPH1 alteration (p.Gln778*) have MCP as well as reduced height and weight. diap1 (mDia1 knockout (KO))-deficient mice have grossly normal body and brain size. However, our histological analysis of diap1 KO mouse coronal brain sections at early and postnatal stages shows unilateral ventricular enlargement, indicating that this mutant mouse shows both important similarities as well as differences with human pathology. We also found that mDia1 protein is expressed in human neuronal precursor cells during mitotic cell division and has a major impact in the regulation of spindle formation and cell division.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Portadoras/genética , Codón sin Sentido , Homocigoto , Microcefalia/genética , Adolescente , Adulto , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Encéfalo/patología , Proteínas Portadoras/metabolismo , Línea Celular , Niño , Preescolar , Femenino , Forminas , Humanos , Lactante , Masculino , Ratones , Microcefalia/diagnóstico , Linaje
15.
Cell Rep ; 9(1): 16-23, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25284784

RESUMEN

Whole-exome sequencing (WES) studies have demonstrated the contribution of de novo loss-of-function single-nucleotide variants (SNVs) to autism spectrum disorder (ASD). However, challenges in the reliable detection of de novo insertions and deletions (indels) have limited inclusion of these variants in prior analyses. By applying a robust indel detection method to WES data from 787 ASD families (2,963 individuals), we demonstrate that de novo frameshift indels contribute to ASD risk (OR = 1.6; 95% CI = 1.0-2.7; p = 0.03), are more common in female probands (p = 0.02), are enriched among genes encoding FMRP targets (p = 6 × 10(-9)), and arise predominantly on the paternal chromosome (p < 0.001). On the basis of mutation rates in probands versus unaffected siblings, we conclude that de novo frameshift indels contribute to risk in approximately 3% of individuals with ASD. Finally, by observing clustering of mutations in unrelated probands, we uncover two ASD-associated genes: KMT2E (MLL5), a chromatin regulator, and RIMS1, a regulator of synaptic vesicle release.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Mutación del Sistema de Lectura , Eliminación de Secuencia , Niño , Trastornos Generalizados del Desarrollo Infantil/sangre , Trastornos Generalizados del Desarrollo Infantil/diagnóstico , ADN/sangre , ADN/genética , Proteínas de Unión al ADN/genética , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteínas de Unión al GTP/genética , Humanos , Masculino , Proteínas del Tejido Nervioso/genética , Linaje , Fenotipo , Factores Sexuales
16.
Neuron ; 81(1): 77-90, 2014 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-24411733

RESUMEN

Tourette syndrome (TS) is characterized by tics, sensorimotor gating deficiencies, and abnormalities of cortico-basal ganglia circuits. A mutation in histidine decarboxylase (Hdc), the key enzyme for the biosynthesis of histamine (HA), has been implicated as a rare genetic cause. Hdc knockout mice exhibited potentiated tic-like stereotypies, recapitulating core phenomenology of TS; these were mitigated by the dopamine (DA) D2 antagonist haloperidol, a proven pharmacotherapy, and by HA infusion into the brain. Prepulse inhibition was impaired in both mice and humans carrying Hdc mutations. HA infusion reduced striatal DA levels; in Hdc knockout mice, striatal DA was increased and the DA-regulated immediate early gene Fos was upregulated. DA D2/D3 receptor binding was altered both in mice and in humans carrying the Hdc mutation. These data confirm histidine decarboxylase deficiency as a rare cause of TS and identify HA-DA interactions in the basal ganglia as an important locus of pathology.


Asunto(s)
Encéfalo/enzimología , Histidina Descarboxilasa/deficiencia , Mutación/genética , Síndrome de Tourette/enzimología , Síndrome de Tourette/genética , Adolescente , Adulto , Anfetamina , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Niño , Agonistas de Dopamina/uso terapéutico , Antagonistas de Dopamina/farmacocinética , Conducta Exploratoria/fisiología , Femenino , Histidina Descarboxilasa/genética , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Oxazinas , Racloprida/farmacocinética , Cintigrafía , Conducta Estereotipada/efectos de los fármacos , Conducta Estereotipada/fisiología , Factores de Tiempo , Síndrome de Tourette/diagnóstico por imagen , Síndrome de Tourette/fisiopatología , Triptófano/genética , Adulto Joven
17.
Cell ; 155(5): 997-1007, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24267886

RESUMEN

Autism spectrum disorder (ASD) is a complex developmental syndrome of unknown etiology. Recent studies employing exome- and genome-wide sequencing have identified nine high-confidence ASD (hcASD) genes. Working from the hypothesis that ASD-associated mutations in these biologically pleiotropic genes will disrupt intersecting developmental processes to contribute to a common phenotype, we have attempted to identify time periods, brain regions, and cell types in which these genes converge. We have constructed coexpression networks based on the hcASD "seed" genes, leveraging a rich expression data set encompassing multiple human brain regions across human development and into adulthood. By assessing enrichment of an independent set of probable ASD (pASD) genes, derived from the same sequencing studies, we demonstrate a key point of convergence in midfetal layer 5/6 cortical projection neurons. This approach informs when, where, and in what cell types mutations in these specific genes may be productively studied to clarify ASD pathophysiology.


Asunto(s)
Encéfalo/metabolismo , Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/fisiopatología , Animales , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Trastornos Generalizados del Desarrollo Infantil/patología , Exoma , Femenino , Feto/metabolismo , Feto/patología , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Ratones , Mutación , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Análisis de Secuencia de ADN
18.
Science ; 338(6105): 394-7, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22956686

RESUMEN

Autism spectrum disorders are a genetically heterogeneous constellation of syndromes characterized by impairments in reciprocal social interaction. Available somatic treatments have limited efficacy. We have identified inactivating mutations in the gene BCKDK (Branched Chain Ketoacid Dehydrogenase Kinase) in consanguineous families with autism, epilepsy, and intellectual disability. The encoded protein is responsible for phosphorylation-mediated inactivation of the E1α subunit of branched-chain ketoacid dehydrogenase (BCKDH). Patients with homozygous BCKDK mutations display reductions in BCKDK messenger RNA and protein, E1α phosphorylation, and plasma branched-chain amino acids. Bckdk knockout mice show abnormal brain amino acid profiles and neurobehavioral deficits that respond to dietary supplementation. Thus, autism presenting with intellectual disability and epilepsy caused by BCKDK mutations represents a potentially treatable syndrome.


Asunto(s)
3-Metil-2-Oxobutanoato Deshidrogenasa (Lipoamida)/administración & dosificación , 3-Metil-2-Oxobutanoato Deshidrogenasa (Lipoamida)/genética , Trastorno Autístico/dietoterapia , Trastorno Autístico/genética , Epilepsia/dietoterapia , Epilepsia/genética , 3-Metil-2-Oxobutanoato Deshidrogenasa (Lipoamida)/deficiencia , Adolescente , Aminoácidos de Cadena Ramificada/administración & dosificación , Aminoácidos de Cadena Ramificada/sangre , Aminoácidos de Cadena Ramificada/deficiencia , Animales , Arginina/genética , Trastorno Autístico/enzimología , Secuencia de Bases , Encéfalo/metabolismo , Niño , Preescolar , Dieta , Epilepsia/enzimología , Femenino , Homocigoto , Humanos , Discapacidad Intelectual/dietoterapia , Discapacidad Intelectual/enzimología , Discapacidad Intelectual/genética , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación , Linaje , Fosforilación , Pliegue de Proteína , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Adulto Joven
19.
Nature ; 485(7397): 237-41, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22495306

RESUMEN

Multiple studies have confirmed the contribution of rare de novo copy number variations to the risk for autism spectrum disorders. But whereas de novo single nucleotide variants have been identified in affected individuals, their contribution to risk has yet to be clarified. Specifically, the frequency and distribution of these mutations have not been well characterized in matched unaffected controls, and such data are vital to the interpretation of de novo coding mutations observed in probands. Here we show, using whole-exome sequencing of 928 individuals, including 200 phenotypically discordant sibling pairs, that highly disruptive (nonsense and splice-site) de novo mutations in brain-expressed genes are associated with autism spectrum disorders and carry large effects. On the basis of mutation rates in unaffected individuals, we demonstrate that multiple independent de novo single nucleotide variants in the same gene among unrelated probands reliably identifies risk alleles, providing a clear path forward for gene discovery. Among a total of 279 identified de novo coding mutations, there is a single instance in probands, and none in siblings, in which two independent nonsense variants disrupt the same gene, SCN2A (sodium channel, voltage-gated, type II, α subunit), a result that is highly unlikely by chance.


Asunto(s)
Trastorno Autístico/genética , Exoma/genética , Exones/genética , Predisposición Genética a la Enfermedad/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Canales de Sodio/genética , Alelos , Codón sin Sentido/genética , Heterogeneidad Genética , Humanos , Canal de Sodio Activado por Voltaje NAV1.2 , Sitios de Empalme de ARN/genética , Hermanos
20.
Eur J Med Genet ; 55(2): 128-31, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22266071

RESUMEN

We report the clinical and cytogenetic findings on a male child with developmental language disorder, no physical abnormalities, and a balanced t(10;15)(q24.1;q21.1) translocation. As the child's parents are unavailable for investigations, it is unclear whether the translocation is inherited or de novo. Fluorescence in situ hybridization (FISH) analyses were carried out using specific RP11-BAC clones mapping near 15q21.1 and 10q24.1 to refine the location of the breakpoints. The breakpoint on 15q21.1 interrupts the SEMA6D gene and the breakpoint on 10q24.1 is located between the ENTPD1 and CCNJ genes. The SEMA6D gene was further investigated in samples of individuals with developmental language disorders and controls; this investigation offered further evidence of the involvement of SEMA6D with developmental language disorders.


Asunto(s)
Trastornos del Desarrollo del Lenguaje/genética , Translocación Genética , Niño , Puntos de Rotura del Cromosoma , Cromosomas Humanos Par 10 , Cromosomas Humanos Par 15 , Humanos , Hibridación Fluorescente in Situ , Masculino , Semaforinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...