Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brain Behav Immun ; 122: 301-312, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39128572

RESUMEN

Recent research has unveiled conflicting evidence regarding the link between aggression and the gut microbiome. Here, we compared behavior profiles of control, germ-free (GF), and antibiotic-treated mice, as well as re-colonized GF mice to understand the impact of the gut microbiome on aggression using the resident-intruder paradigm. Our findings revealed a link between gut microbiome depletion and higher aggression, accompanied by notable changes in urine metabolite profiles and brain gene expression. This study extends beyond classical murine models to humanized mice to reveal the clinical relevance of early-life antibiotic use on aggression. Fecal microbiome transplant from infants exposed to antibiotics in early life (and sampled one month later) into mice led to increased aggression compared to mice receiving transplants from unexposed infants. This study sheds light on the role of the gut microbiome in modulating aggression and highlights its potential avenues of action, offering insights for development of therapeutic strategies for aggression-related disorders.


Asunto(s)
Agresión , Encéfalo , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Agresión/fisiología , Animales , Microbioma Gastrointestinal/fisiología , Ratones , Trasplante de Microbiota Fecal/métodos , Masculino , Encéfalo/metabolismo , Antibacterianos/farmacología , Conducta Animal/fisiología , Vida Libre de Gérmenes , Ratones Endogámicos C57BL , Humanos
2.
Int J Mol Sci ; 23(16)2022 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-36012331

RESUMEN

The non-receptor focal adhesion kinase (FAK) is highly expressed in the central nervous system during development, where it regulates neurite outgrowth and axon guidance, but its role in the adult healthy and diseased brain, specifically in Alzheimer's disease (AD), is largely unknown. Using the 3xTg-AD mouse model, which carries three mutations associated with familial Alzheimer's disease (APP KM670/671NL Swedish, PSEN1 M146V, MAPT P301L) and develops age-related progressive neuropathology including amyloid plaques and Tau tangles, we describe here, for the first time, the in vivo role of FAK in AD pathology. Our data demonstrate that while site-specific knockdown in the hippocampi of 3xTg-AD mice has no effect on learning and memory, hippocampal overexpression of the protein leads to a significant decrease in learning and memory capabilities, which is accompanied by a significant increase in amyloid ß (Aß) load. Furthermore, neuronal morphology is altered following hippocampal overexpression of FAK in these mice. High-throughput proteomics analysis of total and phosphorylated proteins in the hippocampi of FAK overexpressing mice indicates that FAK controls AD-like phenotypes by inhibiting cytoskeletal remodeling in neurons which results in morphological changes, by increasing Tau hyperphosphorylation, and by blocking astrocyte differentiation. FAK activates cell cycle re-entry and consequent cell death while downregulating insulin signaling, thereby increasing insulin resistance and leading to oxidative stress. Our data provide an overview of the signaling networks by which FAK regulates AD pathology and identify FAK as a novel therapeutic target for treating AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Proteína-Tirosina Quinasas de Adhesión Focal , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Trastornos de la Memoria/metabolismo , Ratones , Ratones Transgénicos , Placa Amiloide/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
3.
J Strength Cond Res ; 34(2): 422-429, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29927893

RESUMEN

Martins, RS, Girouard, P, Elliott, E, and Mekary, S. Physiological responses of a jaw repositioning custom-made mouthguard on airway and their effects on athletic performance. J Strength Cond Res 34(2): 422-429, 2020-Advanced dental techniques such as jaw-repositioning have shown to increase lower body muscular power such as vertical jump, but its effects on acceleration and speed have not been studied. Similarly, jaw repositioning is commonly used to increase airways volume and ventilation in a special population (i.e., obstructive sleep apnea); however, its ergogenic effects on aerobic performance have yet not been studied. The purpose of the cross-over study was to investigate the effects of a jaw-repositioning custom-made mouthguard (JCM) on volumetric changes in airway and jaw position and determine the effects this may have on aerobic and anaerobic performance. Results indicated that jaw-repositioning custom-made mouthguard may have an ergogenic effect on performance. The JCM condition showed an increase of 13% in upper airway volume (p = 0.04), 10% in upper airway width (p = 0.004), 7% in ventilation (p = 0.006), 5% in maximal aerobic power (p = 0.003), 4% in time to exhaustion (p = 0.03), 3% in vertical jump (p = 0.03), 2% in broad jump (p = 0.009), and a decrease of 4% in 20-m (p = 0.04) and 2% in 40-m (p = 0.001) sprint times. This is the first study to demonstrate a significant link between jaw repositioning, airway volumetric change, and performance enhancement in both aerobic and anaerobic performances. The results of this study may lead to a change in culture for the use of mouthguards in different sports applications, from high orofacial injury risk sports to other sports, specifically for ergogenic enhancement.


Asunto(s)
Rendimiento Atlético/fisiología , Maxilares/fisiología , Protectores Bucales , Mecánica Respiratoria/fisiología , Estudios Cruzados , Humanos , Masculino , Respiración , Pruebas de Función Respiratoria , Adulto Joven
4.
Cell Mol Life Sci ; 75(7): 1205-1214, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29110030

RESUMEN

Recent genetic and technological advances have determined a role for chromatin structure in neurodevelopment. In particular, compounding evidence has established roles for CTCF and cohesin, two elements that are central in the establishment of chromatin structure, in proper neurodevelopment and in regulation of behavior. Genetic aberrations in CTCF, and in subunits of the cohesin complex, have been associated with neurodevelopmental disorders in human genetic studies, and subsequent animal studies have established definitive, although sometime opposing roles, for these factors in neurodevelopment and behavior. Considering the centrality of these factors in cellular processes in general, the mechanisms through which dysregulation of CTCF and cohesin leads specifically to neurological phenotypes is intriguing, although poorly understood. The connection between CTCF, cohesin, chromatin structure, and behavior is likely to be one of the next frontiers in our understanding of the development of behavior in general, and neurodevelopmental disorders in particular.


Asunto(s)
Conducta , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Sistema Nervioso/metabolismo , Animales , Humanos , Trastornos Mentales/metabolismo , Sistema Nervioso/embriología , Sistema Nervioso/crecimiento & desarrollo , Trastornos del Neurodesarrollo/metabolismo , Cohesinas
5.
Brain Behav Immun ; 73: 310-319, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29787855

RESUMEN

Recent studies have determined that the microbiome has direct effects on behavior, and may be dysregulated in neurodevelopmental conditions. Considering that neurodevelopmental conditions, such as autism, have a strong genetic etiology, it is necessary to understand if genes associated with neurodevelopmental disorders, such as Shank3, can influence the gut microbiome, and if probiotics can be a therapeutic tool. In this study, we have identified dysregulation of several genera and species of bacteria in the gut and colon of both male and female Shank3 KO mice. L. reuteri, a species with decreased relative abundance in the Shank3 KO mice, positively correlated with the expression of gamma-Aminobutyric acid (GABA) receptor subunits in the brain. Treatment of Shank3 KO mice with L. reuteri induced an attenuation of unsocial behavior specifically in male Shank3 mice, and a decrease in repetitive behaviors in both male and female Shank3 KO mice. In addition, L. reuteri treatment affected GABA receptor gene expression and protein levels in multiple brain regions. This study identifies bacterial species that are sensitive to an autism-related mutation, and further suggests a therapeutic potential for probiotic treatment.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/microbiología , Microbioma Gastrointestinal/genética , Animales , Trastorno del Espectro Autista/metabolismo , Conducta Animal/fisiología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Disbiosis/microbiología , Femenino , Microbioma Gastrointestinal/fisiología , Limosilactobacillus reuteri/genética , Masculino , Ratones , Ratones Noqueados , Proteínas de Microfilamentos , Modelos Genéticos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Probióticos/metabolismo , Probióticos/farmacología , Probióticos/uso terapéutico , Receptores de GABA/metabolismo
6.
Cereb Cortex ; 27(12): 5739-5754, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29028941

RESUMEN

Autism Spectrum Disorder (ASD) is a complex neuropsychiatric syndrome whose etiology includes genetic and environmental components. Since epigenetic marks are sensitive to environmental insult, they may be involved in the development of ASD. Initial brain studies have suggested a dysregulation of epigenetic marks in ASD. However, due to cellular heterogeneity in the brain, these studies have not determined if there is a true change in the neuronal epigenetic signature. Here, we report a genome-wide methylation study on fluorescence-activated cell sorting-sorted neuronal nuclei from the frontal cortex of 16 male ASD and 15 male control subjects. Using the 450 K BeadArray, we identified 58 differentially methylated regions (DMRs) that included loci associated to GABAergic system genes, particularly ABAT and GABBR1, and brain-specific MicroRNAs. Selected DMRs were validated by targeted Next Generation Bisulfite Sequencing. Weighted gene correlation network analysis detected 3 co-methylation modules which are significantly correlated to ASD that were enriched for genomic regions underlying neuronal, GABAergic, and immune system genes. Finally, we determined an overlap of the 58 ASD-related DMRs with neurodevelopment associated DMRs. This investigation identifies alterations in the DNA methylation pattern in ASD cortical neurons, providing further evidence that epigenetic alterations in disorder-relevant tissues may be involved in the biology of ASD.


Asunto(s)
Trastorno del Espectro Autista/metabolismo , Núcleo Celular/metabolismo , Metilación de ADN , Lóbulo Frontal/metabolismo , Neuronas/metabolismo , Trastorno del Espectro Autista/genética , Epigénesis Genética , Citometría de Flujo , Estudio de Asociación del Genoma Completo , Humanos , Masculino
7.
J Neurosci ; 36(3): 730-40, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26791204

RESUMEN

Recently, it has been suggested that alterations in DNA methylation mediate the molecular changes and psychopathologies that can occur following trauma. Despite the abundance of DNA methyltransferases (Dnmts) in the brain, which are responsible for catalyzing DNA methylation, their roles in behavioral regulation and in response to stressful challenges remain poorly understood. Here, we demonstrate that adult mice which underwent chronic social defeat stress (CSDS) displayed elevated anxiety-like behavior that was accompanied by a reduction in medial prefrontal cortex (mPFC)-DNA methyltransferase 3a (Dnmt3a) mRNA levels and a subsequent decrease in mPFC-global DNA methylation. To explore the role of mPFC-Dnmt3a in mediating the behavioral responses to stressful challenges we established lentiviral-based mouse models that express lower (knockdown) or higher (overexpression) levels of Dnmt3a specifically within the mPFC. Nonstressed mice injected with knockdown Dnmt3a lentiviruses specifically into the mPFC displayed the same anxiogenic phenotype as the CSDS mice, whereas overexpression of Dnmt3a induced an opposite, anxiolytic, effect in wild-type mice. In addition, overexpression of Dnmt3a in the mPFC of CSDS mice attenuated stress-induced anxiety. Our results indicate a central role for mPFC-Dnmt3a as a mediator of stress-induced anxiety. Significance statement: DNA methylation is suggested to mediate the molecular mechanisms linking environmental challenges, such as chronic stress or trauma, to increased susceptibility to psychopathologies. Here, we show that chronic stress-induced increase in anxiety-like behavior is accompanied by a reduction in DNA methyltransferase 3a (Dnmt3a) mRNA levels and global DNA methylation in the medial prefrontal cortex (mPFC). Overexpression or knockdown of mPFC-Dnmt3a levels induces decrease or increase in anxiety-like behavior, respectively. In addition, overexpression of Dnmt3a in the mPFC of chronic stressed mice attenuated stress-induced anxiety. We suggest that mPFC-Dnmt3a levels mediates anxiety-like behavior, which may be a primary molecular link between chronic stress and the development of anxiety disorders, including post-traumatic stress disorder.


Asunto(s)
Ansiedad/metabolismo , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , Corteza Prefrontal/metabolismo , Factores de Edad , Animales , Ansiedad/etiología , Ansiedad/psicología , ADN Metiltransferasa 3A , Técnicas de Silenciamiento del Gen/métodos , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Psicológico/complicaciones , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología
8.
Int J Mol Sci ; 18(4)2017 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-28420080

RESUMEN

In recent decades it has become clear that Autism Spectrum Disorder (ASD) possesses a diverse and heterogeneous genetic etiology. Aberrations in hundreds of genes have been associated with ASD so far, which include both rare and common variations. While one may expect that these genes converge on specific common molecular pathways, which drive the development of the core ASD characteristics, the task of elucidating these common molecular pathways has been proven to be challenging. Several studies have combined genetic analysis with bioinformatical techniques to uncover molecular mechanisms that are specifically targeted by autism-associated genetic aberrations. Recently, several analysis have suggested that particular signaling mechanisms, including the Wnt and Ca2+/Calmodulin-signaling pathways are often targeted by autism-associated mutations. In this review, we discuss several studies that determine specific molecular pathways affected by autism-associated mutations, and then discuss more in-depth into the biological roles of a few of these pathways, and how they may be involved in the development of ASD. Considering that these pathways may be targeted by specific pharmacological intervention, they may prove to be important therapeutic targets for the treatment of ASD.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Predisposición Genética a la Enfermedad , Transducción de Señal , Animales , Señalización del Calcio , Calmodulina/metabolismo , Biología Computacional/métodos , Perfilación de la Expresión Génica , Humanos , Mutación , Unión Proteica , Mapeo de Interacción de Proteínas , Vía de Señalización Wnt
9.
iScience ; 27(5): 109686, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38660396

RESUMEN

Psilocybin, and its metabolite psilocin, induces psychedelic effects through activation of the 5-HT2A receptor. Psilocybin has been proposed as a treatment for depression and anxiety but sometimes induces anxiety in humans. An understanding of mechanisms underlying the anxiety response will help to better develop therapeutic prospects of psychedelics. In the current study, psilocybin induced an acute increase in anxiety in behavioral paradigms in mice. Importantly, pharmacological blocking of the 5-HT2A receptor attenuates psilocybin-induced head twitch response, a behavioral proxy for the psychedelic response, but does not rescue psilocybin's effect on anxiety-related behavior. Phosphopeptide analysis in the amygdala uncovered signal transduction pathways that are dependent or independent of the 5-HT2A receptor. Furthermore, presynaptic proteins are specifically involved in psilocybin-induced acute anxiety. These insights into how psilocybin may induce short-term anxiety are important for understanding how psilocybin may best be used in the clinical framework.

10.
Transl Psychiatry ; 13(1): 305, 2023 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-37783686

RESUMEN

Autism is a neurodevelopmental disorder characterized by early-onset social behavioral deficits and repetitive behaviors. Chromodomain helicase DNA-binding protein (CHD8) is among the genes most strongly associated with autism. In addition to the core behavioral symptoms of autism, affected individuals frequently present with gastrointestinal symptoms that are also common among individuals harboring mutations in the gene encoding CHD8. However, little is known regarding the mechanisms whereby CHD8 affects gut function. In addition, it remains unknown whether gastrointestinal manifestations contribute to the behavioral phenotypes of autism. The current study found that mice haploinsufficient for the large isoform of Chd8 (Chd8L) exhibited increased intestinal permeability, transcriptomic dysregulation in gut epithelial cells, reduced tuft cell and goblet cell counts in the gut, and an overall increase in microbial load. Gut epithelial cell-specific Chd8 haploinsufficiency was associated with increased anxiety-related behaviors together with a decrease in tuft cell numbers. Antibiotic treatment of Chd8L haploinsufficient mice attenuated social behavioral deficits. Together, these results suggest Chd8 as a key determinant of autism-related gastrointestinal deficits, while also laying the ground for future studies on the link between GI deficits and autism-related behaviors.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Ratones , Animales , Trastorno Autístico/genética , Eje Cerebro-Intestino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Unión al ADN/genética , Trastorno del Espectro Autista/genética , Células Epiteliales
11.
NPJ Biofilms Microbiomes ; 9(1): 103, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110423

RESUMEN

Autism Spectrum Disorder (ASD) is a neurodevelopmental condition which is defined by decreased social communication and the presence of repetitive or stereotypic behaviors. Recent evidence has suggested that the gut-brain axis may be important in neurodevelopment in general and may play a role in ASD in particular. Here, we present a study of the gut microbiome in 96 individuals diagnosed with ASD in Israel, compared to 42 neurotypical individuals. We determined differences in alpha and beta diversity in the microbiome of individuals with ASD and demonstrated that the phylum Bacteroidetes and genus Bacteroides were the most significantly over-represented in individuals with ASD. To understand the possible functional significance of these changes, we treated newborn mice with Bacteroides fragilis at birth. B. fragilis-treated male mice displayed social behavior dysfunction, increased repetitive behaviors, and gene expression dysregulation in the prefrontal cortex, while female mice did not display behavioral deficits. These findings suggest that overabundance of Bacteroides, particularly in early life, may have functional consequences for individuals with ASD.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Humanos , Masculino , Ratones , Femenino , Animales , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/genética , Bacteroides/genética , Modelos Animales de Enfermedad , Conducta Social
12.
Nat Neurosci ; 26(7): 1208-1217, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37365313

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by heterogeneous cognitive, behavioral and communication impairments. Disruption of the gut-brain axis (GBA) has been implicated in ASD although with limited reproducibility across studies. In this study, we developed a Bayesian differential ranking algorithm to identify ASD-associated molecular and taxa profiles across 10 cross-sectional microbiome datasets and 15 other datasets, including dietary patterns, metabolomics, cytokine profiles and human brain gene expression profiles. We found a functional architecture along the GBA that correlates with heterogeneity of ASD phenotypes, and it is characterized by ASD-associated amino acid, carbohydrate and lipid profiles predominantly encoded by microbial species in the genera Prevotella, Bifidobacterium, Desulfovibrio and Bacteroides and correlates with brain gene expression changes, restrictive dietary patterns and pro-inflammatory cytokine profiles. The functional architecture revealed in age-matched and sex-matched cohorts is not present in sibling-matched cohorts. We also show a strong association between temporal changes in microbiome composition and ASD phenotypes. In summary, we propose a framework to leverage multi-omic datasets from well-defined cohorts and investigate how the GBA influences ASD.


Asunto(s)
Trastorno del Espectro Autista , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/genética , Eje Cerebro-Intestino , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Estudios Transversales , Teorema de Bayes , Reproducibilidad de los Resultados , Citocinas
13.
J Biol Chem ; 286(11): 8839-45, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21220432

RESUMEN

NAD(P)H:quinone oxidoreductase 1 (NQO1) is a flavoenzyme that is important in maintaining the cellular redox state and regulating protein degradation. The NQO1 polymorphism C609T has been associated with increased susceptibility to various age-related pathologies. We show here that NQO1 protein level is regulated by the E3 ligase STUB1/CHIP (C terminus of Hsc70-interacting protein). NQO1 binds STUB1 via the Hsc70-interacting domain (tetratricopeptide repeat domain) and undergoes ubiquitination and degradation. We demonstrate here that the product of the C609T polymorphism (P187S) is a stronger STUB1 interactor with increased susceptibility to ubiquitination by the E3 ligase STUB1. Furthermore, age-dependent decrease of STUB1 correlates with increased NQO1 accumulation. Remarkably, examination of hippocampi from Alzheimer disease patients revealed that in half of the cases examined the NQO1 protein level was undetectable due to C609T polymorphism, suggesting that the age-dependent accumulation of NQO1 is impaired in certain Alzheimer disease patients.


Asunto(s)
Envejecimiento/metabolismo , Enfermedad de Alzheimer/enzimología , Hipocampo/enzimología , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Envejecimiento/genética , Enfermedad de Alzheimer/genética , Células HEK293 , Proteínas del Choque Térmico HSC70/genética , Proteínas del Choque Térmico HSC70/metabolismo , Humanos , NAD(P)H Deshidrogenasa (Quinona)/genética , Proteínas del Tejido Nervioso/genética , Polimorfismo Genético , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/genética
14.
Biol Rev Camb Philos Soc ; 97(2): 582-599, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34734461

RESUMEN

The microbiome influences the emotional and cognitive phenotype of its host, as well as the neurodevelopment and pathophysiology of various brain processes and disorders, via the well-established microbiome-gut-brain axis. Rapidly accumulating data link the microbiome to severe neuropsychiatric disorders in humans, including schizophrenia, Alzheimer's and Parkinson's. Moreover, preclinical work has shown that perturbation of the microbiome is closely associated with social, cognitive and behavioural deficits. The potential of the microbiome as a diagnostic and therapeutic tool is currently undercut by a lack of clear mechanistic understanding of the microbiome-gut-brain axis. This review establishes the hypothesis that the mechanism by which this influence is carried out is synaptic plasticity - long-term changes to the physical and functional neuronal structures that enable the brain to undertake learning, memory formation, emotional regulation and more. By examining the different constituents of the microbiome-gut-brain axis through the lens of synaptic plasticity, this review explores the diverse aspects by which the microbiome shapes the behaviour and mental wellbeing of the host. Key elements of this complex bi-directional relationship include neurotransmitters, neuronal electrophysiology, immune mediators that engage with both the central and enteric nervous systems and signalling cascades that trigger long-term potentiation of synapses. The importance of establishing mechanistic correlations along the microbiome-gut-brain axis cannot be overstated as they hold the potential for furthering current understanding regarding the vast fields of neuroscience and neuropsychiatry. This review strives to elucidate the promising theory of microbiome-driven synaptic plasticity in the hope of enlightening current researchers and inspiring future ones.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Encéfalo/metabolismo , Microbioma Gastrointestinal/fisiología , Plasticidad Neuronal , Neuronas/fisiología
15.
Mol Brain ; 15(1): 30, 2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35379308

RESUMEN

CCCTC-binding factor (CTCF) is a regulator of chromatin organization and has direct effects on gene transcription. Mutations in CTCF have been identified in individuals with neurodevelopmental conditions. There are wide range of behaviors associated with these mutations, including intellectual disabilities, changes in temperament, and autism. Previous mice-model studies have identified roles for CTCF in excitatory neurons in specific behaviors, particularly in regards to learning and memory. However, the role of CTCF in inhibitory neurons is less well defined. In the current study, specific knockout of CTCF in parvalbumin-expressing neurons, a subset of inhibitory neurons, induced a specific behavioral phenotype, including locomotor abnormalities, anxiolytic behavior, and a decrease in social behavior. The anxiolytic and social abnormalities are detected before the onset of locomotor abnormalities. Immunohistochemical analysis revealed a disbalance in parvalbumin-expressing and somatostatin-expressing cells in these mice. Single nuclei RNA sequencing identified changes in gene expression in parvalbumin-expressing neurons that are specific to inhibitory neuronal identity and function. Electrophysiology analysis revealed an enhanced inhibitory tone in the hippocampal pyramidal neurons in knockout mice. These findings indicate that CTCF in parvalbumin-expressing neurons has a significant role in the overall phenotype of CTCF-associated neurodevelopmental deficits.


Asunto(s)
Neuronas , Parvalbúminas , Animales , Ansiedad , Factor de Unión a CCCTC , Ratones , Ratones Noqueados , Neuronas/metabolismo , Parvalbúminas/metabolismo , Conducta Social
16.
Genes (Basel) ; 12(8)2021 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-34440307

RESUMEN

Chromodomain-helicase-DNA-binding protein 8 (CHD8) has been identified as one of the genes with the strongest association with autism. The CHD8 protein is a transcriptional regulator that is expressed in nearly all cell types and has been implicated in multiple cellular processes, including cell cycle, cell adhesion, neuronal development, myelination, and synaptogenesis. Considering the central role of CHD8 in the genetics of autism, a deeper understanding of the physiological functions of CHD8 is important to understand the development of the autism phenotype and potential therapeutic targets. Different CHD8 mutant mouse models were developed to determine autism-like phenotypes and to fully understand their mechanisms. Here, we review the current knowledge on CHD8, with an emphasis on mechanistic lessons gained from animal models that have been studied.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Proteínas de Unión al ADN/fisiología , Trastornos del Neurodesarrollo/fisiopatología , Factores de Transcripción/fisiología , Animales , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Fenotipo , Factores de Transcripción/genética
17.
Mol Neurobiol ; 58(5): 2322-2341, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33417228

RESUMEN

Alzheimer's disease (AD) is the most common cause of senile dementia and one of the greatest medical, social, and economic challenges. According to a dominant theory, amyloid-ß (Aß) peptide is a key AD pathogenic factor. Aß-soluble species interfere with synaptic functions, aggregate gradually, form plaques, and trigger neurodegeneration. The AD-associated pathology affects numerous systems, though the substantial loss of cholinergic neurons and α7 nicotinic receptors (α7AChR) is critical for the gradual cognitive decline. Aß binds to α7AChR under various experimental settings; nevertheless, the functional significance of this interaction is ambiguous. Whereas the capability of low Aß concentrations to activate α7AChR is functionally beneficial, extensive brain exposure to high Aß concentrations diminishes α7AChR activity, contributes to the cholinergic deficits that characterize AD. Aß and snake α-neurotoxins competitively bind to α7AChR. Accordingly, we designed a chemically modified α-cobratoxin (mToxin) to inhibit the interaction between Aß and α7AChR. Subsequently, we examined mToxin in a set of original in silico, in vitro, ex vivo experiments, and in a murine AD model. We report that mToxin reversibly inhibits α7AChR, though it attenuates Aß-induced synaptic transmission abnormalities, and upregulates pathways supporting long-term potentiation and reducing apoptosis. Remarkably, mToxin demonstrates no toxicity in brain slices and mice. Moreover, its chronic intracerebroventricular administration improves memory in AD-model animals. Our results point to unique mToxin neuroprotective properties, which might be tailored for the treatment of AD. Our methodology bridges the gaps in understanding Aß-α7AChR interaction and represents a promising direction for further investigations and clinical development.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Neurotoxinas/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neuronas Colinérgicas/efectos de los fármacos , Neuronas Colinérgicas/metabolismo , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Modelos Teóricos , Neurotoxinas/uso terapéutico , Unión Proteica/efectos de los fármacos
18.
Curr Opin Neurobiol ; 62: 76-82, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31972462

RESUMEN

There is growing evidence for the involvement of the gut-microbiota in the regulation of emotions, behavior, and higher cognitive functions through the 'microbiome-gut-brain axis'. This relationship between the gut microbiota and the brain is pivotal for the development of the newborn, which receives its commensal microbiota at birth; dysbiosis may result in altered neurodevelopment. The hypothalamus-pituitary-adrenocortical (HPA) axis is actively involved in the stress response but is undeveloped in the newborn. Here, we describe how changes in the commensal microbiota influence the normal development of the HPA axis and review recent findings describing the essential crosstalk between the gut microbiota and the HPA axis and suggesting a role for the maternal and commensal microbiota in the development of the HPA axis and of the stress response.


Asunto(s)
Microbiota , Ansiedad , Encéfalo , Humanos , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Estrés Psicológico
19.
Transl Psychiatry ; 10(1): 412, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33239620

RESUMEN

The etiology of Autism Spectrum Disorders (ASD) includes a strong genetic component and a complicated environmental component. Recent evidence indicates that maternal diabetes, including gestational diabetes, is associated with an increased prevalence of ASD. While previous studies have looked into possible roles for maternal diabetes in neurodevelopment, there are few studies into how gestational diabetes, with no previous diabetic or metabolic phenotype, may affect neurodevelopment. In this study, we have specifically induced gestational diabetes in mice, followed by behavioral and molecular phenotyping of the mice offspring. Pregnant mice were injected with STZ a day after initiation of pregnancy. Glucose levels increased to diabetic levels between E7 and E14 in pregnancy in a subset of the pregnant animals. Male offspring of Gestational Diabetic mothers displayed increased repetitive behaviors with no dysregulation in the three-chambered social interaction test. RNA-seq analysis revealed a dysregulation in genes related to forebrain development in the frontal cortex and a dysregulation of a network of neurodevelopment and immune related genes in the striatum. Together, these results give evidence that gestational diabetes can induce changes in adulthood behavior and gene transcription in the brain.


Asunto(s)
Trastorno del Espectro Autista , Diabetes Gestacional , Efectos Tardíos de la Exposición Prenatal , Animales , Cognición , Diabetes Gestacional/genética , Femenino , Lóbulo Frontal , Masculino , Ratones , Embarazo , Transcripción Genética
20.
J Neurochem ; 109(4): 1168-78, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19317853

RESUMEN

The accumulation of tau and amyloid beta proteins is the major molecular pathology of Alzheimer's disease (AD). The mechanisms leading to the accumulation of these proteins are not completely clear. Hsc-70/Hsp-70, a chaperone protein, has been shown to bind both these proteins and regulate their degradation. We have previously shown that the co-chaperone protein BAG-1 can inhibit the degradation of tau by forming a complex with Hsc-70 and tau. In this current work, we show that there is an increase in the BAG-1M isoform in the hippocampus of AD patients. In addition, BAG-1 binds to both tau and amyloid precursor protein physically, and is found highly expressed in the same neurons that contain intracellular tau or amyloid in hippocampal sections from AD patients. Over-expression of BAG-1M in cell culture also induced an increase in both tau and amyloid precursor protein levels. In conclusion, we report a specific increase of BAG-1M in human AD patients, which is both physically and functionally associated to the two major molecular markers of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Unión al ADN/biosíntesis , Hipocampo/metabolismo , Factores de Transcripción/biosíntesis , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Western Blotting , Células Cultivadas , Proteínas de Unión al ADN/genética , Electroforesis en Gel de Poliacrilamida , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Proteínas HSP70 de Choque Térmico/biosíntesis , Humanos , Inmunohistoquímica , Ratones , Adhesión en Parafina , Plásmidos/genética , Factores de Transcripción/genética , Regulación hacia Arriba/fisiología , Replicación Viral , Proteínas tau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA