Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 219
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 159(7): 1511-23, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25525873

RESUMEN

Alternative splicing (AS) generates vast transcriptomic and proteomic complexity. However, which of the myriad of detected AS events provide important biological functions is not well understood. Here, we define the largest program of functionally coordinated, neural-regulated AS described to date in mammals. Relative to all other types of AS within this program, 3-15 nucleotide "microexons" display the most striking evolutionary conservation and switch-like regulation. These microexons modulate the function of interaction domains of proteins involved in neurogenesis. Most neural microexons are regulated by the neuronal-specific splicing factor nSR100/SRRM4, through its binding to adjacent intronic enhancer motifs. Neural microexons are frequently misregulated in the brains of individuals with autism spectrum disorder, and this misregulation is associated with reduced levels of nSR100. The results thus reveal a highly conserved program of dynamic microexon regulation associated with the remodeling of protein-interaction networks during neurogenesis, the misregulation of which is linked to autism.


Asunto(s)
Empalme Alternativo , Trastornos Generalizados del Desarrollo Infantil/patología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Animales , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Humanos , Ratones , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neurogénesis , Dominios y Motivos de Interacción de Proteínas , Análisis de Secuencia de ARN , Lóbulo Temporal/patología
2.
Cell ; 155(7): 1451-63, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24315484

RESUMEN

Neurodevelopmental disorders, including autism spectrum disorder (ASD), are defined by core behavioral impairments; however, subsets of individuals display a spectrum of gastrointestinal (GI) abnormalities. We demonstrate GI barrier defects and microbiota alterations in the maternal immune activation (MIA) mouse model that is known to display features of ASD. Oral treatment of MIA offspring with the human commensal Bacteroides fragilis corrects gut permeability, alters microbial composition, and ameliorates defects in communicative, stereotypic, anxiety-like and sensorimotor behaviors. MIA offspring display an altered serum metabolomic profile, and B. fragilis modulates levels of several metabolites. Treating naive mice with a metabolite that is increased by MIA and restored by B. fragilis causes certain behavioral abnormalities, suggesting that gut bacterial effects on the host metabolome impact behavior. Taken together, these findings support a gut-microbiome-brain connection in a mouse model of ASD and identify a potential probiotic therapy for GI and particular behavioral symptoms in human neurodevelopmental disorders.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/microbiología , Tracto Gastrointestinal/microbiología , Animales , Ansiedad/metabolismo , Ansiedad/microbiología , Bacteroides fragilis , Conducta Animal , Encéfalo/fisiología , Niño , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Modelos Animales de Enfermedad , Femenino , Tracto Gastrointestinal/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Microbiota , Probióticos/administración & dosificación
3.
Cell ; 155(5): 1008-21, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24267887

RESUMEN

Genetic studies have identified dozens of autism spectrum disorder (ASD) susceptibility genes, raising two critical questions: (1) do these genetic loci converge on specific biological processes, and (2) where does the phenotypic specificity of ASD arise, given its genetic overlap with intellectual disability (ID)? To address this, we mapped ASD and ID risk genes onto coexpression networks representing developmental trajectories and transcriptional profiles representing fetal and adult cortical laminae. ASD genes tightly coalesce in modules that implicate distinct biological functions during human cortical development, including early transcriptional regulation and synaptic development. Bioinformatic analyses suggest that translational regulation by FMRP and transcriptional coregulation by common transcription factors connect these processes. At a circuit level, ASD genes are enriched in superficial cortical layers and glutamatergic projection neurons. Furthermore, we show that the patterns of ASD and ID risk genes are distinct, providing a biological framework for further investigating the pathophysiology of ASD.


Asunto(s)
Encéfalo/embriología , Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Redes Reguladoras de Genes , Encéfalo/fisiopatología , Corteza Cerebral/fisiopatología , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Neuronas/metabolismo , Transcripción Genética
4.
Cell ; 143(3): 442-55, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-21029865

RESUMEN

The mechanisms that promote excitatory synapse formation and maturation have been extensively studied. However, the molecular events that limit excitatory synapse development so that synapses form at the right time and place and in the correct numbers are less well understood. We have identified a RhoA guanine nucleotide exchange factor, Ephexin5, which negatively regulates excitatory synapse development until EphrinB binding to the EphB receptor tyrosine kinase triggers Ephexin5 phosphorylation, ubiquitination, and degradation. The degradation of Ephexin5 promotes EphB-dependent excitatory synapse development and is mediated by Ube3A, a ubiquitin ligase that is mutated in the human cognitive disorder Angelman syndrome and duplicated in some forms of Autism Spectrum Disorders (ASDs). These findings suggest that aberrant EphB/Ephexin5 signaling during the development of synapses may contribute to the abnormal cognitive function that occurs in Angelman syndrome and, possibly, ASDs.


Asunto(s)
Sinapsis/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Síndrome de Angelman/metabolismo , Animales , Niño , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Giro Dentado/citología , Giro Dentado/metabolismo , Embrión de Mamíferos/metabolismo , Técnicas de Inactivación de Genes , Humanos , Ratones , Ratas , Ratas Long-Evans , Receptores de la Familia Eph/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteína de Unión al GTP rhoA/genética
5.
Nat Rev Neurosci ; 18(3): 147-157, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28179641

RESUMEN

Several large-scale genomic studies have supported an association between cases of autism spectrum disorder and mutations in the genes SH3 and multiple ankyrin repeat domains protein 1 (SHANK1), SHANK2 and SHANK3, which encode a family of postsynaptic scaffolding proteins that are present at glutamatergic synapses in the CNS. An evaluation of human genetic data, as well as of in vitro and in vivo animal model data, may allow us to understand how disruption of SHANK scaffolding proteins affects the structure and function of neural circuits and alters behaviour.


Asunto(s)
Trastorno del Espectro Autista/metabolismo , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , Animales , Trastorno del Espectro Autista/genética , Trastornos Generalizados del Desarrollo Infantil/genética , Humanos , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo
6.
Nat Rev Neurosci ; 16(9): 551-63, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26289574

RESUMEN

Genetics studies of autism spectrum disorder (ASD) have identified several risk genes that are key regulators of synaptic plasticity. Indeed, many of the risk genes that have been linked to these disorders encode synaptic scaffolding proteins, receptors, cell adhesion molecules or proteins that are involved in chromatin remodelling, transcription, protein synthesis or degradation, or actin cytoskeleton dynamics. Changes in any of these proteins can increase or decrease synaptic strength or number and, ultimately, neuronal connectivity in the brain. In addition, when deleterious mutations occur, inefficient genetic buffering and impaired synaptic homeostasis may increase an individual's risk for ASD.


Asunto(s)
Encéfalo/fisiología , Trastornos Generalizados del Desarrollo Infantil/genética , Plasticidad Neuronal/fisiología , Sinapsis/genética , Encéfalo/patología , Trastornos Generalizados del Desarrollo Infantil/diagnóstico , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Humanos , Plasticidad Neuronal/genética , Sinapsis/metabolismo , Sinapsis/patología
7.
Nature ; 492(7429): 382-6, 2012 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-23235829

RESUMEN

Fragile X syndrome (FXS) is a multi-organ disease that leads to mental retardation, macro-orchidism in males and premature ovarian insufficiency in female carriers. FXS is also a prominent monogenic disease associated with autism spectrum disorders (ASDs). FXS is typically caused by the loss of fragile X mental retardation 1 (FMR1) expression, which codes for the RNA-binding protein FMRP. Here we report the discovery of distinct RNA-recognition elements that correspond to the two independent RNA-binding domains of FMRP, in addition to the binding sites within the messenger RNA targets for wild-type and I304N mutant FMRP isoforms and the FMRP paralogues FXR1P and FXR2P (also known as FXR1 and FXR2). RNA-recognition-element frequency, ratio and distribution determine target mRNA association with FMRP. Among highly enriched targets, we identify many genes involved in ASD and show that FMRP affects their protein levels in human cell culture, mouse ovaries and human brain. Notably, we discovered that these targets are also dysregulated in Fmr1(-/-) mouse ovaries showing signs of premature follicular overdevelopment. These results indicate that FMRP targets share signalling pathways across different cellular contexts. As the importance of signalling pathways in both FXS and ASD is becoming increasingly apparent, our results provide a ranked list of genes as basis for the pursuit of new therapeutic targets for these neurological disorders.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Regulación de la Expresión Génica/genética , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , Secuencias Reguladoras de Ácido Ribonucleico/genética , Animales , Secuencia de Bases , Sitios de Unión , Encéfalo/metabolismo , Niño , Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Reactivos de Enlaces Cruzados , Femenino , Células HEK293 , Humanos , Inmunoprecipitación , Ratones , Datos de Secuencia Molecular , Familia de Multigenes , Mutación , Ovario/metabolismo , Ovario/patología , ARN Mensajero/metabolismo , Elementos de Respuesta/genética , Transducción de Señal , Especificidad por Sustrato
8.
Development ; 141(21): 4031-41, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25336734

RESUMEN

The cerebellum is a pre-eminent model for the study of neurogenesis and circuit assembly. Increasing interest in the cerebellum as a participant in higher cognitive processes and as a locus for a range of disorders and diseases make this simple yet elusive structure an important model in a number of fields. In recent years, our understanding of some of the more familiar aspects of cerebellar growth, such as its territorial allocation and the origin of its various cell types, has undergone major recalibration. Furthermore, owing to its stereotyped circuitry across a range of species, insights from a variety of species have contributed to an increasingly rich picture of how this system develops. Here, we review these recent advances and explore three distinct aspects of cerebellar development - allocation of the cerebellar anlage, the significance of transit amplification and the generation of neuronal diversity - each defined by distinct regulatory mechanisms and each with special significance for health and disease.


Asunto(s)
Encéfalo/embriología , Cerebelo/embriología , Animales , Encéfalo/anatomía & histología , Encéfalo/citología , Cerebelo/anatomía & histología , Cerebelo/citología , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Trastornos Generalizados del Desarrollo Infantil/patología , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patología , Modelos Biológicos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Proc Natl Acad Sci U S A ; 111(42): E4468-77, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25294932

RESUMEN

Truncating mutations of chromodomain helicase DNA-binding protein 8 (CHD8), and of many other genes with diverse functions, are strong-effect risk factors for autism spectrum disorder (ASD), suggesting multiple mechanisms of pathogenesis. We explored the transcriptional networks that CHD8 regulates in neural progenitor cells (NPCs) by reducing its expression and then integrating transcriptome sequencing (RNA sequencing) with genome-wide CHD8 binding (ChIP sequencing). Suppressing CHD8 to levels comparable with the loss of a single allele caused altered expression of 1,756 genes, 64.9% of which were up-regulated. CHD8 showed widespread binding to chromatin, with 7,324 replicated sites that marked 5,658 genes. Integration of these data suggests that a limited array of direct regulatory effects of CHD8 produced a much larger network of secondary expression changes. Genes indirectly down-regulated (i.e., without CHD8-binding sites) reflect pathways involved in brain development, including synapse formation, neuron differentiation, cell adhesion, and axon guidance, whereas CHD8-bound genes are strongly associated with chromatin modification and transcriptional regulation. Genes associated with ASD were strongly enriched among indirectly down-regulated loci (P < 10(-8)) and CHD8-bound genes (P = 0.0043), which align with previously identified coexpression modules during fetal development. We also find an intriguing enrichment of cancer-related gene sets among CHD8-bound genes (P < 10(-10)). In vivo suppression of chd8 in zebrafish produced macrocephaly comparable to that of humans with inactivating mutations. These data indicate that heterozygous disruption of CHD8 precipitates a network of gene-expression changes involved in neurodevelopmental pathways in which many ASD-associated genes may converge on shared mechanisms of pathogenesis.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Axones/metabolismo , Sitios de Unión , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Cromatina/metabolismo , ADN Helicasas/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Genoma , Heterocigoto , Humanos , Megalencefalia/metabolismo , Mutación , Neoplasias/metabolismo , Neuronas/metabolismo , Unión Proteica , Factores de Riesgo , Análisis de Secuencia de ARN , Programas Informáticos , Pez Cebra , Proteínas de Pez Cebra/genética
10.
Hum Mol Genet ; 23(13): 3490-505, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24497577

RESUMEN

Accelerated head and brain growth (macrocephaly) during development is a replicated biological finding in a subset of individuals with autism spectrum disorder (ASD). However, the relationship between brain overgrowth and the behavioral and cognitive symptoms of ASD is poorly understood. The PI3K-Akt-mTOR pathway regulates cellular growth; several genes encoding negative regulators of this pathway are ASD risk factors, including PTEN. Mutations in PTEN have been reported in individuals with ASD and macrocephaly. We report that brain overgrowth is widespread in Pten germline haploinsufficient (Pten(+/-)) mice, reflecting Pten mRNA expression in the developing brain. We then ask if broad brain overgrowth translates into general or specific effects on the development of behavior and cognition by testing Pten(+/-) mice using assays relevant to ASD and comorbidities. Deficits in social behavior were observed in both sexes. Males also showed abnormalities related to repetitive behavior and mood/anxiety. Females exhibited circadian activity and emotional learning phenotypes. Widespread brain overgrowth together with selective behavioral impairments in Pten(+/-) mice raises the possibility that most brain areas and constituent cell types adapt to an altered trajectory of growth with minimal impact on the behaviors tested in our battery; however, select areas/cell types relevant to social behavior are more vulnerable or less adaptable, thus resulting in social deficits. Probing dopaminergic neurons as a candidate vulnerable cell type, we found social behavioral impairments in mice with Pten conditionally inactivated in dopaminergic neurons that are consistent with the possibility that desynchronized growth in key cell types may contribute to ASD endophenotypes.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/metabolismo , Haploinsuficiencia/genética , Fosfohidrolasa PTEN/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Trastornos Generalizados del Desarrollo Infantil/genética , Femenino , Masculino , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/genética
11.
Hum Mol Genet ; 23(1): 90-103, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23956174

RESUMEN

An increasing number of genes predisposing to autism spectrum disorders (ASDs) has been identified, many of which are implicated in synaptic function. This 'synaptic autism pathway' notably includes disruption of SYN1 that is associated with epilepsy, autism and abnormal behavior in both human and mice models. Synapsins constitute a multigene family of neuron-specific phosphoproteins (SYN1-3) present in the majority of synapses where they are implicated in the regulation of neurotransmitter release and synaptogenesis. Synapsins I and II, the major Syn isoforms in the adult brain, display partially overlapping functions and defects in both isoforms are associated with epilepsy and autistic-like behavior in mice. In this study, we show that nonsense (A94fs199X) and missense (Y236S and G464R) mutations in SYN2 are associated with ASD in humans. The phenotype is apparent in males. Female carriers of SYN2 mutations are unaffected, suggesting that SYN2 is another example of autosomal sex-limited expression in ASD. When expressed in SYN2  knockout neurons, wild-type human Syn II fully rescues the SYN2 knockout phenotype, whereas the nonsense mutant is not expressed and the missense mutants are virtually unable to modify the SYN2 knockout phenotype. These results identify for the first time SYN2  as a novel predisposing gene for ASD and strengthen the hypothesis that a disturbance of synaptic homeostasis underlies ASD.


Asunto(s)
Axones/metabolismo , Axones/patología , Trastornos Generalizados del Desarrollo Infantil/genética , Sinapsinas/genética , Vesículas Sinápticas/patología , Animales , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Codón sin Sentido , Femenino , Predisposición Genética a la Enfermedad , Células HeLa , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Missense , Neuronas/metabolismo , Vesículas Sinápticas/metabolismo
12.
Hum Mol Genet ; 23(24): 6495-511, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25027319

RESUMEN

Disturbances of lipid metabolism have been implicated in psychiatric illnesses. We previously reported an association between the gene for fatty acid binding protein 7 (FABP7) and schizophrenia. Furthermore, we identified and reported several rare non-synonymous polymorphisms of the brain-expressed genes FABP3, FABP5 and FABP7 from schizophrenia and autism spectrum disorder (ASD), diseases known to part share genetic architecture. Here, we conducted further studies to better understand the contribution these genes make to the pathogenesis of schizophrenia and ASD. In postmortem brains, we detected altered mRNA expression levels of FABP5 in schizophrenia, and of FABP7 in ASD and altered FABP5 in peripheral lymphocytes. Using a patient cohort, comprehensive mutation screening identified six missense and two frameshift variants from the three FABP genes. The two frameshift proteins, FABP3 E132fs and FABP7 N80fs, formed cellular aggregates and were unstable when expressed in cultured cells. The four missense mutants with predicted possible damaging outcomes showed no changes in intracellular localization. Examining ligand binding properties, FABP7 S86G and FABP7 V126L lost their preference for docosahexaenoic acid to linoleic acid. Finally, mice deficient in Fabp3, Fabp5 and Fabp7 were evaluated in a systematic behavioral test battery. The Fabp3 knockout (KO) mice showed decreased social memory and novelty seeking, and Fabp7 KO mice displayed hyperactive and anxiety-related phenotypes, while Fabp5 KO mice showed no apparent phenotypes. In conclusion, disturbances in brain-expressed FABPs could represent an underlying disease mechanism in a proportion of schizophrenia and ASD sufferers.


Asunto(s)
Conducta Animal , Proteínas Portadoras/genética , Trastornos Generalizados del Desarrollo Infantil/genética , Proteínas de Unión a Ácidos Grasos/genética , Esquizofrenia/genética , Proteínas Supresoras de Tumor/genética , Secuencia de Aminoácidos , Animales , Ansiedad/genética , Ansiedad/fisiopatología , Encéfalo/metabolismo , Encéfalo/patología , Proteínas Portadoras/metabolismo , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Trastornos Generalizados del Desarrollo Infantil/fisiopatología , Ácidos Docosahexaenoicos/metabolismo , Conducta Exploratoria , Proteína 3 de Unión a Ácidos Grasos , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos/metabolismo , Mutación del Sistema de Lectura , Humanos , Ácido Linoleico/metabolismo , Linfocitos/metabolismo , Ratones , Ratones Transgénicos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación Missense , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Alineación de Secuencia , Proteínas Supresoras de Tumor/metabolismo
13.
Neural Plast ; 2016: 3597209, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27840741

RESUMEN

Several genetic causes of autism spectrum disorder (ASD) have been identified. However, more recent work has highlighted that certain environmental exposures early in life may also account for some cases of autism. Environmental insults during pregnancy, such as infection or malnutrition, seem to dramatically impact brain development. Maternal viral or bacterial infections have been characterized as disruptors of brain shaping, even if their underlying mechanisms are not yet fully understood. Poor nutritional diversity, as well as nutrient deficiency, is strongly associated with neurodevelopmental disorders in children. For instance, imbalanced levels of essential fatty acids, and especially polyunsaturated fatty acids (PUFAs), are observed in patients with ASD and other neurodevelopmental disorders (e.g., attention deficit hyperactivity disorder (ADHD) and schizophrenia). Interestingly, PUFAs, and specifically n-3 PUFAs, are powerful immunomodulators that exert anti-inflammatory properties. These prenatal dietary and immunologic factors not only impact the fetal brain, but also affect the microbiota. Recent work suggests that the microbiota could be the missing link between environmental insults in prenatal life and future neurodevelopmental disorders. As both nutrition and inflammation can massively affect the microbiota, we discuss here how understanding the crosstalk between these three actors could provide a promising framework to better elucidate ASD etiology.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/metabolismo , Trastorno del Espectro Autista/metabolismo , Trastorno Autístico/metabolismo , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Ácidos Grasos Omega-3/metabolismo , Microbiota/fisiología , Animales , Trastornos Generalizados del Desarrollo Infantil/complicaciones , Humanos , Estado Nutricional/fisiología
14.
BMC Bioinformatics ; 16: 144, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25943565

RESUMEN

BACKGROUND: Modern biotechnologies often result in high-dimensional data sets with many more variables than observations (n≪p). These data sets pose new challenges to statistical analysis: Variable selection becomes one of the most important tasks in this setting. Similar challenges arise if in modern data sets from observational studies, e.g., in ecology, where flexible, non-linear models are fitted to high-dimensional data. We assess the recently proposed flexible framework for variable selection called stability selection. By the use of resampling procedures, stability selection adds a finite sample error control to high-dimensional variable selection procedures such as Lasso or boosting. We consider the combination of boosting and stability selection and present results from a detailed simulation study that provide insights into the usefulness of this combination. The interpretation of the used error bounds is elaborated and insights for practical data analysis are given. RESULTS: Stability selection with boosting was able to detect influential predictors in high-dimensional settings while controlling the given error bound in various simulation scenarios. The dependence on various parameters such as the sample size, the number of truly influential variables or tuning parameters of the algorithm was investigated. The results were applied to investigate phenotype measurements in patients with autism spectrum disorders using a log-linear interaction model which was fitted by boosting. Stability selection identified five differentially expressed amino acid pathways. CONCLUSION: Stability selection is implemented in the freely available R package stabs (http://CRAN.R-project.org/package=stabs). It proved to work well in high-dimensional settings with more predictors than observations for both, linear and additive models. The original version of stability selection, which controls the per-family error rate, is quite conservative, though, this is much less the case for its improvement, complementary pairs stability selection. Nevertheless, care should be taken to appropriately specify the error bound.


Asunto(s)
Algoritmos , Biomarcadores/análisis , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Reacciones Falso Positivas , Modelos Estadísticos , Estudios de Casos y Controles , Trastornos Generalizados del Desarrollo Infantil/genética , Simulación por Computador , Perfilación de la Expresión Génica , Humanos , Dinámicas no Lineales , Fenotipo
15.
J Neurochem ; 134(4): 783-93, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25977097

RESUMEN

Autism spectrum disorder (ASD) is a developmental brain disorder. Mutations in synaptic components including synaptic adhesion molecules have been found in ASD patients. Contactin-associated protein-like 2 (CASPR2) is one of the synaptic adhesion molecules associated with ASD. CASPR2 forms a complex with receptors via interaction with multiple PDZ domain protein 1 (MUPP1). Little is known about the relationship between impaired CASPR2-MUPP1-receptor complex and the pathogenesis of ASD. GPR37 is a receptor for survival factors. We recently identified mutations including R558Q in the G-protein-coupled receptor 37 (GPR37) gene in ASD patients. The mutated GPR37s accumulate in the endoplasmic reticulum. In this study, we show that GPR37 is a component of the CASPR2-MUPP1 receptor complex in the mouse brain. CASPR2 and GPR37 mainly interacted with the PDZ3 and PDZ11 domains of MUPP1, respectively. Compared to GPR37, GPR37(R558Q) slightly interacted with MUPP1 and caused dendritic alteration. GPR37, but not GPR37(R558Q) nor GPR37-deltaC which lacks its PDZ binding domain, was transported to the cell surface by MUPP1. In primary hippocampal neurons, GPR37 co-localized with MUPP1 and CASPR2 at the synapse, but not GPR37(R558Q). Thus, ASD-related mutation of GPR37 may cause the impaired CASPR2-MUPP1-GPR37 complex on the dendrites associated with one of the pathogenesis of ASD. In this study, we identified that GPR37 is a component of the MUPP1 and CASPR2 receptor complex. Autism deleterious mutated GPR37(R558Q) slightly interacts with MUPP1 and retains in ER, resulting in dendritic alteration. In neuron, GPR37, but not GPR37(R558Q), is transported to the dendrite and synapse by MUPP1. Thus, ASD-related mutation of GPR37 may cause the impaired CASPR2-MUPP1-GPR37 complex on the dendrites associated with one of the pathogenesis of ASD.


Asunto(s)
Proteínas Portadoras/metabolismo , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Proteínas de la Membrana/metabolismo , Mutación/fisiología , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Células COS , Proteínas Portadoras/genética , Células Cultivadas , Trastornos Generalizados del Desarrollo Infantil/genética , Chlorocebus aethiops , Hipocampo/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Unión Proteica/fisiología , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/genética
16.
Hum Mol Genet ; 22(16): 3306-14, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23615299

RESUMEN

Existence of a discrete new X-linked intellectual disability (XLID) syndrome due to KIAA2022 deficiency was questioned by disruption of KIAA2022 by an X-chromosome pericentric inversion in a XLID family we reported in 2004. Three additional families with likely pathogenic KIAA2022 mutations were discovered within the frame of systematic parallel sequencing of familial cases of XLID or in the context of routine array-CGH evaluation of sporadic intellectual deficiency (ID) cases. The c.186delC and c.3597dupA KIAA2022 truncating mutations were identified by X-chromosome exome sequencing, while array CGH discovered a 70 kb microduplication encompassing KIAA2022 exon 1 in the third family. This duplication decreased KIAA2022 mRNA level in patients' lymphocytes by 60%. Detailed clinical examination of all patients, including the two initially reported, indicated moderate-to-severe ID with autistic features, strabismus in all patients, with no specific dysmorphic features other than a round face in infancy and no structural brain abnormalities on magnetic resonance imaging (MRI). Interestingly, the patient with decreased KIAA2022 expression had only mild ID with severe language delay and repetitive behaviors falling in the range of an autism spectrum disorder (ASD). Since little is known about KIAA2022 function, we conducted morphometric studies in cultured rat hippocampal neurons. We found that siRNA-mediated KIAA2022 knockdown resulted in marked impairment in neurite outgrowth including both the dendrites and the axons, suggesting a major role for KIAA2022 in neuron development and brain function.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Genes Ligados a X , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Neuritas/fisiología , Adolescente , Adulto , Animales , Encéfalo/metabolismo , Células Cultivadas , Niño , Preescolar , Técnicas de Silenciamiento del Gen , Ligamiento Genético , Variación Genética , Humanos , Masculino , Neuritas/metabolismo , Ratas , Análisis de Secuencia de ARN , Adulto Joven
17.
Bioinformatics ; 30(12): i175-84, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24931981

RESUMEN

MOTIVATION: Discovering the transcriptional regulatory architecture of the metabolism has been an important topic to understand the implications of transcriptional fluctuations on metabolism. The reporter algorithm (RA) was proposed to determine the hot spots in metabolic networks, around which transcriptional regulation is focused owing to a disease or a genetic perturbation. Using a z-score-based scoring scheme, RA calculates the average statistical change in the expression levels of genes that are neighbors to a target metabolite in the metabolic network. The RA approach has been used in numerous studies to analyze cellular responses to the downstream genetic changes. In this article, we propose a mutual information-based multivariate reporter algorithm (MIRA) with the goal of eliminating the following problems in detecting reporter metabolites: (i) conventional statistical methods suffer from small sample sizes, (ii) as z-score ranges from minus to plus infinity, calculating average scores can lead to canceling out opposite effects and (iii) analyzing genes one by one, then aggregating results can lead to information loss. MIRA is a multivariate and combinatorial algorithm that calculates the aggregate transcriptional response around a metabolite using mutual information. We show that MIRA's results are biologically sound, empirically significant and more reliable than RA. RESULTS: We apply MIRA to gene expression analysis of six knockout strains of Escherichia coli and show that MIRA captures the underlying metabolic dynamics of the switch from aerobic to anaerobic respiration. We also apply MIRA to an Autism Spectrum Disorder gene expression dataset. Results indicate that MIRA reports metabolites that highly overlap with recently found metabolic biomarkers in the autism literature. Overall, MIRA is a promising algorithm for detecting metabolic drug targets and understanding the relation between gene expression and metabolic activity. AVAILABILITY AND IMPLEMENTATION: The code is implemented in C# language using .NET framework. Project is available upon request.


Asunto(s)
Algoritmos , Redes y Vías Metabólicas/genética , Niño , Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Transcripción Genética
18.
Mol Psychiatry ; 19(12): 1314-25, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25048006

RESUMEN

Over the last few years, awareness of autism spectrum disorder (ASD) in adults has increased. The precise etiology of ASD is still unresolved. Animal research, genetic and postmortem studies suggest that the glutamate (Glu) system has an important role, possibly related to a cybernetic imbalance between neuronal excitation and inhibition. To clarify the possible disruption of Glu metabolism in adults with high-functioning autism, we performed a magnetic resonance spectroscopy (MRS) study investigating the anterior cingulate cortex (ACC) and the cerebellum in adults with high-functioning ASD. Twenty-nine adult patients with high-functioning ASD and 29 carefully matched healthy volunteers underwent MRS scanning of the pregenual ACC and the left cerebellar hemisphere. Metabolic data were compared between groups and were correlated with psychometric measures of autistic features. We found a significant decrease in the cingulate N-acetyl-aspartate (NAA) and the combined Glu and glutamine (Glx) signals in adults with ASD, whereas we did not find other metabolic abnormalities in the ACC or the cerebellum. The Glx signal correlated significantly with psychometric measures of autism, particularly with communication deficits. Our data support the hypothesis that there is a link between disturbances of the cingulate NAA and Glx metabolism, and autism. The findings are discussed in the context of the hypothesis of excitatory/inhibitory imbalance in autism. Further research should clarify the specificity and dynamics of these findings regarding other neuropsychiatric disorders and other brain areas.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/metabolismo , Ácido Glutámico/metabolismo , Giro del Cíngulo/metabolismo , Adulto , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Mapeo Encefálico , Cerebelo/metabolismo , Trastornos Generalizados del Desarrollo Infantil/psicología , Femenino , Glutamina/metabolismo , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Psicometría
19.
Brain ; 137(Pt 1): 137-52, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24277719

RESUMEN

Proteins of the ProSAP/Shank family act as major organizing scaffolding elements within the postsynaptic density of excitatory synapses. Deletions, mutations or the downregulation of these molecules has been linked to autism spectrum disorders, the related Phelan McDermid Syndrome or Alzheimer's disease. ProSAP/Shank proteins are targeted to synapses depending on binding to zinc, which is a prerequisite for the assembly of the ProSAP/Shank scaffold. To gain insight into whether the previously reported assembly of ProSAP/Shank through zinc ions provides a crossing point between genetic forms of autism spectrum disorder and zinc deficiency as an environmental risk factor for autism spectrum disorder, we examined the interplay between zinc and ProSAP/Shank in vitro and in vivo using neurobiological approaches. Our data show that low postsynaptic zinc availability affects the activity dependent increase in ProSAP1/Shank2 and ProSAP2/Shank3 levels at the synapse in vitro and that a loss of synaptic ProSAP1/Shank2 and ProSAP2/Shank3 occurs in a mouse model for acute and prenatal zinc deficiency. Zinc-deficient animals displayed abnormalities in behaviour such as over-responsivity and hyperactivity-like behaviour (acute zinc deficiency) and autism spectrum disorder-related behaviour such as impairments in vocalization and social behaviour (prenatal zinc deficiency). Most importantly, a low zinc status seems to be associated with an increased incidence rate of seizures, hypotonia, and attention and hyperactivity issues in patients with Phelan-McDermid syndrome, which is caused by haploinsufficiency of ProSAP2/Shank3. We suggest that the molecular underpinning of prenatal zinc deficiency as a risk factor for autism spectrum disorder may unfold through the deregulation of zinc-binding ProSAP/Shank family members.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/metabolismo , Saposinas/metabolismo , Sinapsis/fisiología , Zinc/deficiencia , Animales , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Conducta Animal/fisiología , Western Blotting , Células Cultivadas , Trastornos Generalizados del Desarrollo Infantil/fisiopatología , Deleción Cromosómica , Trastornos de los Cromosomas/metabolismo , Trastornos de los Cromosomas/fisiopatología , Cromosomas Humanos Par 22/metabolismo , Femenino , Hipocampo/metabolismo , Humanos , Inmunohistoquímica , Ratones , Técnicas de Cultivo de Órganos , Embarazo , ARN Interferente Pequeño/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Fluorescencia , Transfección , Vocalización Animal/fisiología
20.
Nutr Neurosci ; 18(4): 177-85, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-24564346

RESUMEN

OBJECTIVES: Studies have suggested a link between diet and behavior in children with autism spectrum disorders (ASDs). Parental reports of behavioral changes upon exposure to gluten and/or casein are common in clinical practice. An association between diet type, intestinal permeability (IP) ('leaky gut'), and behavior has been long proposed but not substantiated. We explored this possible association in this trial. METHODS: This randomized double-blind, placebo-controlled study explored the effects of gluten and milk on IP and behavior in children with ASDs over a period of 4 weeks. IP assessed by lactulose:mannitol (L/M) sugar permeability test and behavior assessed by the Aberrant Behavior Checklist and Conners Parent Rating were measured. Gastrointestinal symptoms in both groups were also monitored. RESULTS: Neither the L/M ratio nor behavioral scores were different between groups exposed to gluten/dairy or placebo. The changes observed were noted to be small and not clinically significant. DISCUSSION: Our study although underpowered to show small differences does not support an association between dietary gluten/milk, IP, and behavioral changes in subjects with ASD.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Trastornos Generalizados del Desarrollo Infantil/dietoterapia , Productos Lácteos , Dietoterapia/métodos , Absorción Gastrointestinal/efectos de los fármacos , Glútenes/farmacología , Permeabilidad de la Membrana Celular/fisiología , Niño , Trastornos Generalizados del Desarrollo Infantil/etiología , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Preescolar , Método Doble Ciego , Femenino , Absorción Gastrointestinal/fisiología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/patología , Glútenes/administración & dosificación , Humanos , Lactulosa/metabolismo , Masculino , Manitol/metabolismo , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA