Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Aging Neurosci ; 15: 1152562, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37255534

RESUMEN

Specific and effective treatments for autism spectrum disorder (ASD) are lacking due to a poor understanding of disease mechanisms. Here we test the idea that similarities between diverse ASD mouse models are caused by deficits in common molecular pathways at neuronal synapses. To do this, we leverage the availability of multiple genetic models of ASD that exhibit shared synaptic and behavioral deficits and use quantitative mass spectrometry with isobaric tandem mass tagging (TMT) to compare their hippocampal synaptic proteomes. Comparative analyses of mouse models for Fragile X syndrome (Fmr1 knockout), cortical dysplasia focal epilepsy syndrome (Cntnap2 knockout), PTEN hamartoma tumor syndrome (Pten haploinsufficiency), ANKS1B syndrome (Anks1b haploinsufficiency), and idiopathic autism (BTBR+) revealed several common altered cellular and molecular pathways at the synapse, including changes in oxidative phosphorylation, and Rho family small GTPase signaling. Functional validation of one of these aberrant pathways, Rac1 signaling, confirms that the ANKS1B model displays altered Rac1 activity counter to that observed in other models, as predicted by the bioinformatic analyses. Overall similarity analyses reveal clusters of synaptic profiles, which may form the basis for molecular subtypes that explain genetic heterogeneity in ASD despite a common clinical diagnosis. Our results suggest that ASD-linked susceptibility genes ultimately converge on common signaling pathways regulating synaptic function and propose that these points of convergence are key to understanding the pathogenesis of this disorder.

2.
iScience ; 25(2): 103796, 2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35198865

RESUMEN

How changes in brain scaling relate to altered behavior is an important question in neurodevelopmental disorder research. Mice with germline Pten haploinsufficiency (Pten +/-) closely mirror the abnormal brain scaling and behavioral deficits seen in humans with macrocephaly/autism syndrome, which is caused by PTEN mutations. We explored whether deviation from normal patterns of growth can predict behavioral abnormalities. Brain regions associated with sensory processing (e.g., pons and inferior colliculus) had the biggest deviations from expected volume. While Pten +/- mice showed little or no abnormal behavior on most assays, both sexes showed sensory deficits, including impaired sensorimotor gating and hyporeactivity to high-intensity stimuli. Developmental analysis of this phenotype showed sexual dimorphism for hyporeactivity. Mapping behavioral phenotypes of Pten +/- mice onto relevant brain regions suggested abnormal behavior is likely when associated with relatively enlarged brain regions, while unchanged or relatively decreased brain regions have little predictive value.

3.
Mol Brain ; 14(1): 162, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34749771

RESUMEN

Molecular and cellular mechanisms underlying the role of the prelimbic cortex in contextual fear memory remain elusive. Here we examined the kinesin family of molecular motor proteins (KIFs) in the prelimbic cortex for their role in mediating contextual fear, a form of associative memory. KIFs function as critical mediators of synaptic transmission and plasticity by their ability to modulate microtubule function and transport of gene products. However, the regulation and function of KIFs in the prelimbic cortex insofar as mediating memory consolidation is not known. We find that within one hour of contextual fear conditioning, the expression of KIF3B is upregulated in the prelimbic but not the infralimbic cortex. Importantly, lentiviral-mediated knockdown of KIF3B in the prelimbic cortex produces deficits in consolidation while reducing freezing behavior during extinction of contextual fear. We also find that the depletion of KIF3B increases spine density within prelimbic neurons. Taken together, these results illuminate a key role for KIF3B in the prelimbic cortex as far as mediating contextual fear memory.


Asunto(s)
Extinción Psicológica , Memoria , Corteza Cerebral , Extinción Psicológica/fisiología , Miedo/fisiología , Memoria/fisiología , Corteza Prefrontal/metabolismo
4.
Genes (Basel) ; 12(9)2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34573348

RESUMEN

Pten germline haploinsufficient (Pten+/-) mice, which model macrocephaly/autism syndrome, show social and repetitive behavior deficits, early brain overgrowth, and cortical-subcortical hyperconnectivity. Previous work indicated that altered neuronal connectivity may be a substrate for behavioral deficits. We hypothesized that exposing Pten+/- mice to environmental enrichment after brain overgrowth has occurred may facilitate adaptation to abnormal "hard-wired" connectivity through enhancing synaptic plasticity. Thus, we reared Pten+/- mice and their wild-type littermates from weaning under either standard (4-5 mice per standard-sized cage, containing only bedding and nestlet) or enriched (9-10 mice per large-sized cage, containing objects for exploration and a running wheel, plus bedding and nestlet) conditions. Adult mice were tested on social and non-social assays in which Pten+/- mice display deficits. Environmental enrichment rescued sex-specific deficits in social behavior in Pten+/- mice and partially rescued increased repetitive behavior in Pten+/- males. We found that Pten+/- mice show increased excitatory and decreased inhibitory pre-synaptic proteins; this phenotype was also rescued by environmental enrichment. Together, our results indicate that environmental enrichment can rescue social behavioral deficits in Pten+/- mice, possibly through normalizing the excitatory synaptic protein abundance.


Asunto(s)
Conducta Animal/fisiología , Fosfohidrolasa PTEN/genética , Conducta Social , Sinapsis/patología , Animales , Trastorno Autístico/etiología , Encéfalo/anomalías , Encéfalo/patología , Modelos Animales de Enfermedad , Facies , Femenino , Haploinsuficiencia , Masculino , Megalencefalia/etiología , Ratones Endogámicos C57BL , Ratones Mutantes
5.
Cell Rep ; 36(2): 109369, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260917

RESUMEN

Synaptic structural plasticity, key to long-term memory storage, requires translation of localized RNAs delivered by long-distance transport from the neuronal cell body. Mechanisms and regulation of this system remain elusive. Here, we explore the roles of KIF5C and KIF3A, two members of kinesin superfamily of molecular motors (Kifs), and find that loss of function of either kinesin decreases dendritic arborization and spine density whereas gain of function of KIF5C enhances it. KIF5C function is a rate-determining component of local translation and is associated with ∼650 RNAs, including EIF3G, a regulator of translation initiation, and plasticity-associated RNAs. Loss of function of KIF5C in dorsal hippocampal CA1 neurons constrains both spatial and contextual fear memory, whereas gain of function specifically enhances spatial memory and extinction of contextual fear. KIF5C-mediated long-distance transport of local translation substrates proves a key mechanism underlying structural plasticity and memory.


Asunto(s)
Cinesinas/metabolismo , Memoria a Largo Plazo , Proteínas Motoras Moleculares/metabolismo , Plasticidad Neuronal , Biosíntesis de Proteínas , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Espinas Dendríticas/metabolismo , Potenciales Postsinápticos Excitadores , Miedo , Femenino , Mutación con Ganancia de Función , Células HEK293 , Hipocampo/metabolismo , Humanos , Aprendizaje , Masculino , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Ratones Endogámicos C57BL , Transporte de ARN , Transducción de Señal , Sinapsis/metabolismo , Transmisión Sináptica
6.
Biol Psychiatry ; 90(5): 295-306, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33840455

RESUMEN

BACKGROUND: Mutations in DYRK1A are a cause of microcephaly, autism spectrum disorder, and intellectual disability; however, the underlying cellular and molecular mechanisms are not well understood. METHODS: We generated a conditional mouse model using Emx1-cre, including conditional heterozygous and homozygous knockouts, to investigate the necessity of Dyrk1a in the cortex during development. We used unbiased, high-throughput phosphoproteomics to identify dysregulated signaling mechanisms in the developing Dyrk1a mutant cortex as well as classic genetic modifier approaches and pharmacological therapeutic intervention to rescue microcephaly and neuronal undergrowth caused by Dyrk1a mutations. RESULTS: We found that cortical deletion of Dyrk1a in mice causes decreased brain mass and neuronal size, structural hypoconnectivity, and autism-relevant behaviors. Using phosphoproteomic screening, we identified growth-associated signaling cascades dysregulated upon Dyrk1a deletion, including TrkB-BDNF (tyrosine receptor kinase B-brain-derived neurotrophic factor), an important regulator of ERK/MAPK (extracellular signal-regulated kinase/mitogen-activated protein kinase) and mTOR (mammalian target of rapamycin) signaling. Genetic suppression of Pten or pharmacological treatment with IGF-1 (insulin-like growth factor-1), both of which impinge on these signaling cascades, rescued microcephaly and neuronal undergrowth in neonatal mutants. CONCLUSIONS: Altogether, these findings identify a previously unknown mechanism through which Dyrk1a mutations disrupt growth factor signaling in the developing brain, thus influencing neuronal growth and connectivity. Our results place DYRK1A as a critical regulator of a biological pathway known to be dysregulated in humans with autism spectrum disorder and intellectual disability. In addition, these data position Dyrk1a within a larger group of autism spectrum disorder/intellectual disability risk genes that impinge on growth-associated signaling cascades to regulate brain size and connectivity, suggesting a point of convergence for multiple autism etiologies.


Asunto(s)
Trastorno del Espectro Autista , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Células Piramidales/patología , Animales , Ratones , Mutación/genética , Quinasas DyrK
7.
Cell Rep ; 32(2): 107899, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32668253

RESUMEN

The prefrontal cortex and amygdala are anatomical substrates linked to both social information and emotional valence processing, but it is not known whether sub-circuits in the medial prefrontal cortex (mPFC) that project to the basolateral amygdala (BLA) are recruited and functionally contribute to social approach-avoidance behavior. Using retrograde labeling of mPFC projections to the BLA, we find that BLA-projecting neurons in the infralimbic cortex (IL) are preferentially activated in response to a social cue as compared with BLA-projecting neurons in the prelimbic cortex (PL). Chemogenetic interrogation of these sub-circuits shows that activation of PL-BLA or inhibition of IL-BLA circuits impairs social behavior. Sustained closed-loop optogenetic activation of PL-BLA circuitry induces social impairment, corresponding to a negative emotional state as revealed by real-time place preference behavioral avoidance. Reactivation of foot shock-responsive PL-BLA circuitry impairs social behavior. Altogether, these data suggest a circuit-level mechanism by which valence-encoding mPFC-BLA sub-circuits shape social approach-avoidance behavior.


Asunto(s)
Amígdala del Cerebelo/fisiología , Vías Nerviosas/fisiología , Corteza Prefrontal/fisiología , Conducta Social , Animales , Ratones Endogámicos C57BL , Neuronas/fisiología , Optogenética , Factores de Tiempo
8.
Nat Commun ; 11(1): 1797, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286273

RESUMEN

Mutations that inactivate negative translation regulators cause autism spectrum disorders (ASD), which predominantly affect males and exhibit social interaction and communication deficits and repetitive behaviors. However, the cells that cause ASD through elevated protein synthesis resulting from these mutations remain unknown. Here we employ conditional overexpression of translation initiation factor eIF4E to increase protein synthesis in specific brain cells. We show that exaggerated translation in microglia, but not neurons or astrocytes, leads to autism-like behaviors in male mice. Although microglial eIF4E overexpression elevates translation in both sexes, it only increases microglial density and size in males, accompanied by microglial shift from homeostatic to a functional state with enhanced phagocytic capacity but reduced motility and synapse engulfment. Consequently, cortical neurons in the mice have higher synapse density, neuroligins, and excitation-to-inhibition ratio compared to control mice. We propose that functional perturbation of male microglia is an important cause for sex-biased ASD.


Asunto(s)
Trastorno Autístico/metabolismo , Conducta Animal , Microglía/metabolismo , Biosíntesis de Proteínas , Animales , Proteínas de Unión al Calcio/metabolismo , Movimiento Celular , Femenino , Perfilación de la Expresión Génica , Genotipo , Homeostasis , Masculino , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Neuronas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fagocitosis , Corteza Prefrontal/metabolismo , Corteza Prefrontal/ultraestructura , Conducta Social , Sinapsis/metabolismo
9.
Artículo en Inglés | MEDLINE | ID: mdl-31871231

RESUMEN

A subset of individuals with autism spectrum disorder (ASD) and macrocephaly carry mutations in the gene PTEN. Animal models, particularly mice, have been helpful in establishing a causal role for Pten mutations in autism-relevant behavioral deficits. These models are a useful tool for investigating neurobiological mechanisms of these behavioral phenotypes and developing potential therapeutic interventions. Here we provide an overview of various genetic mouse models that have been used to characterize behavioral phenotypes caused by perturbation of Pten We discuss convergent and divergent phenotypes across models with the aim of highlighting a set of behavioral domains that are sensitive to the effects of Pten mutation and that may provide useful readouts for translational and basic neuroscience research.


Asunto(s)
Trastorno del Espectro Autista/genética , Conducta , Fosfohidrolasa PTEN/genética , Animales , Modelos Animales de Enfermedad , Genotipo , Humanos , Megalencefalia/genética , Ratones , Mutación , Fenotipo
10.
Transl Psychiatry ; 9(1): 329, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804455

RESUMEN

Haploinsufficiency for PTEN is a cause of autism spectrum disorder and brain overgrowth; however, it is not known if PTEN mutations disrupt scaling across brain areas during development. To address this question, we used magnetic resonance imaging to analyze brains of male Pten haploinsufficient (Pten+/-) mice and wild-type littermates during early postnatal development and adulthood. Adult Pten+/- mice display a consistent pattern of abnormal scaling across brain areas, with white matter (WM) areas being particularly affected. This regional and WM enlargement recapitulates structural abnormalities found in individuals with PTEN haploinsufficiency and autism. Early postnatal Pten+/- mice do not display the same pattern, instead exhibiting greater variability across mice and brain regions than controls. This suggests that Pten haploinsufficiency may desynchronize growth across brain regions during early development before stabilizing by maturity. Pten+/- cortical cultures display increased proliferation of glial cell populations, indicating a potential substrate of WM enlargement, and provide a platform for testing candidate therapeutics. Pten haploinsufficiency dysregulates coordinated growth across brain regions during development. This results in abnormally scaled brain areas and associated behavioral deficits, potentially explaining the relationship between PTEN mutations and neurodevelopmental disorders.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Fosfohidrolasa PTEN/fisiología , Sustancia Blanca/crecimiento & desarrollo , Animales , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Células Cultivadas , Corteza Cerebral/diagnóstico por imagen , Modelos Animales de Enfermedad , Haploinsuficiencia , Humanos , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones de la Cepa 129 , Fosfohidrolasa PTEN/genética , Sustancia Blanca/diagnóstico por imagen
11.
Autism Res ; 12(10): 1463-1471, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31441226

RESUMEN

Heterozygous mutations in PTEN, which encodes a negative regulator of the mTOR and ß-catenin signaling pathways, cause macrocephaly/autism syndrome. However, the neurobiological substrates of the core symptoms of this syndrome are poorly understood. Here, we investigate the relationship between cerebral cortical overgrowth and social behavior deficits in conditional Pten heterozygous female mice (Pten cHet) using Emx1-Cre, which is expressed in cortical pyramidal neurons and a subset of glia. We found that conditional heterozygous mutation of Ctnnb1 (encoding ß-catenin) suppresses Pten cHet cortical overgrowth, but not social behavioral deficits, whereas conditional heterozygous mutation of Mtor suppresses social behavioral deficits, but not cortical overgrowth. Neuronal activity in response to social cues and excitatory synapse markers are elevated in the medial prefrontal cortex (mPFC) of Pten cHet mice, and heterozygous mutation in Mtor, but not Ctnnb1, rescues these phenotypes. These findings indicate that macroscale cerebral cortical overgrowth and social behavioral phenotypes caused by Pten haploinsufficiency can be dissociated based on responsiveness to genetic suppression of Ctnnb1 or Mtor. Furthermore, neuronal connectivity appears to be one potential substrate for mTOR-mediated suppression of social behavioral deficits in Pten haploinsufficient mice. Autism Res 2019, 12: 1463-1471. © 2019 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: A subgroup of individuals with autism display overgrowth of the head and the brain during development. Using a mouse model of an autism risk gene, Pten, that displays both brain overgrowth and social behavioral deficits, we show here that that these two symptoms can be dissociated. Reversal of social behavioral deficits in this model is associated with rescue of abnormal synaptic markers and neuronal activity.


Asunto(s)
Conducta Animal , Haploinsuficiencia/genética , Fosfohidrolasa PTEN/genética , Supresión Genética/genética , Sinapsis/genética , Serina-Treonina Quinasas TOR/genética , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Transducción de Señal/genética
12.
Mol Neuropsychiatry ; 3(3): 141-150, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29594133

RESUMEN

There is a pressing need to improve approaches for drug discovery related to neuropsychiatric disorders (NSDs). Therapeutic discovery in neuropsychiatric disorders would benefit from screening assays that can measure changes in complex phenotypes linked to disease mechanisms. However, traditional assays that track complex neuronal phenotypes, such as neuronal connectivity, exhibit poor scalability and are not compatible with high-throughput screening (HTS) procedures. Therefore, we created a neuronal phenotypic assay platform that focused on improving the scalability and affordability of neuron-based assays capable of tracking disease-relevant phenotypes. First, using inexpensive laboratory-level automation, we industrialized primary neuronal culture production, which enabled the creation of scalable assays within functioning neural networks. We then developed a panel of phenotypic assays based on culturing of primary neurons from genetically modified mice expressing HTS-compatible reporters that capture disease-relevant phenotypes. We demonstrated that a library of 1,280 compounds was quickly screened against both assays using only a few litters of mice in a typical academic laboratory setting. Finally, we implemented one assay in a fully automated high-throughput academic screening facility, illustrating the scalability of assays designed using this platform. These methodological improvements simplify the creation of highly scalable neuron-based phenotypic assays designed to improve drug discovery in CNS disorders.

13.
Neurobiol Aging ; 50: 134-143, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27960107

RESUMEN

The precise molecular and cellular events responsible for age-dependent cognitive dysfunctions remain unclear. We report that Rheb (ras homolog enriched in brain) GTPase, an activator of mammalian target of rapamycin (mTOR), regulates memory functions in mice. Conditional depletion of Rheb selectively in the forebrain of mice obtained from crossing Rhebf/f and CamKIICre results in spontaneous signs of age-related memory loss, that is, spatial memory deficits (T-maze, Morris water maze) without affecting locomotor (open-field test), anxiety-like (elevated plus maze), or contextual fear conditioning functions. Partial depletion of Rheb in forebrain was sufficient to elicit memory defects with little effect on the neuronal size, cortical thickness, or mammalian target of rapamycin activity. Rheb depletion, however, increased the levels of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), a protein elevated in aging and Alzheimer's disease. Overall, our study demonstrates that forebrain Rheb promotes aging-associated cognitive defects. Thus, molecular understanding of Rheb pathway in brain may provide new therapeutic targets for aging and/or Alzheimer's disease-associated memory deficits.


Asunto(s)
Envejecimiento/psicología , Trastornos de la Memoria/etiología , Proteínas de Unión al GTP Monoméricas/deficiencia , Proteínas de Unión al GTP Monoméricas/fisiología , Neuropéptidos/deficiencia , Neuropéptidos/fisiología , Prosencéfalo/enzimología , Envejecimiento/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Disfunción Cognitiva/etiología , Ratones Mutantes , Ratones Transgénicos , Terapia Molecular Dirigida , Proteína Homóloga de Ras Enriquecida en el Cerebro , Sirolimus/metabolismo , Memoria Espacial
14.
Nat Commun ; 7: 13421, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27845329

RESUMEN

Multiple autism risk genes converge on the regulation of mTOR signalling, which is a key effector of neuronal growth and connectivity. We show that mTOR signalling is dysregulated during early postnatal development in the cerebral cortex of germ-line heterozygous Pten mutant mice (Pten+/-), which model macrocephaly/autism syndrome. The basolateral amygdala (BLA) receives input from subcortical-projecting neurons in the medial prefrontal cortex (mPFC). Analysis of mPFC to BLA axonal projections reveals that Pten+/- mice exhibit increased axonal branching and connectivity, which is accompanied by increased activity in the BLA in response to social stimuli and social behavioural deficits. The latter two phenotypes can be suppressed by pharmacological inhibition of S6K1 during early postnatal life or by reducing the activity of mPFC-BLA circuitry in adulthood. These findings identify a mechanism of altered connectivity that has potential relevance to the pathophysiology of macrocephaly/autism syndrome and autism spectrum disorders featuring dysregulated mTOR signalling.


Asunto(s)
Trastorno Autístico/fisiopatología , Complejo Nuclear Basolateral/fisiopatología , Modelos Animales de Enfermedad , Megalencefalia/fisiopatología , Red Nerviosa/fisiopatología , Corteza Prefrontal/fisiopatología , Animales , Facies , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Embarazo , Transducción de Señal , Conducta Social , Serina-Treonina Quinasas TOR/metabolismo
15.
Sci Rep ; 6: 34793, 2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27713505

RESUMEN

Zika virus (ZIKV) infection in pregnant women has been established as a cause of microcephaly in newborns. Here we test the hypothesis that neurodevelopmental stages when the brain is undergoing rapid growth are particularly vulnerable to the effects of ZIKV infection. We injected ZIKV intracranially into wild type C57BL/6 mice at two different time points: early postnatal development, when the brain is growing at its maximal rate, and at weaning, when the brain has largely reached adult size. Both time points showed widespread immunoreactivity for ZIKV and cleaved caspase 3 (CC3, a marker of apoptosis) throughout the brain. However, in early postnatal ZIKV injected mice, some brain areas and cell types display particularly large increases in apoptosis that we did not observe in older animals. Corticospinal pyramidal neurons, a cell type implicated in human microcephaly associated with ZIKV infection, are an example of one such cell type. Proliferating cells in the ventricular zone stem cell compartment are also depleted. These findings are consistent with the hypothesis that periods of rapid brain growth are especially susceptible to neurodevelopmental effects of ZIKV infection, and establish a valuable model to investigate mechanisms underlying neurodevelopmental effects of ZIKV infection and explore candidate therapeutics.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/virología , Microcefalia/virología , Neuronas/patología , Infección por el Virus Zika/fisiopatología , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Encéfalo/anomalías , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos C57BL , Neuronas/virología
16.
Autism Res ; 9(12): 1248-1262, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27220363

RESUMEN

Germline heterozygous mutations in Pten (phosphatase and tensin homolog) are associated with macrocephaly and autism spectrum disorders (ASD). Pten germline heterozygous (Pten+/- ) mice approximate these mutations, and both sexes show widespread brain overgrowth and impaired social behavior. Strikingly similar behavior phenotypes have been reported in oxytocin (Oxt) and/or oxytocin receptor (OxtR) knockout mice. Thus, we hypothesized that the behavioral phenotypes of germline Pten+/- mice may be caused by reduced Pten function in Oxt-expressing cells. To investigate this, we tested mice in which Pten was conditionally deleted using oxytocin-Cre (Oxt-Cre+ ; PtenloxP/+ , Oxt-Cre+ ; PtenloxP/loxP ) on a battery including assays of social, repetitive, depression-like, and anxiety-like behaviors. Minimal behavioral abnormalities were found; decreased anxiety-like behavior in the open field test in Oxt-Cre+ ; PtenloxP/loxP males was the only result that phenocopied germline Pten+/- mice. However, Oxt cell size was dramatically increased in Oxt-Cre+ ; PtenloxP/loxP mice in adulthood. Thus, conditional deletion of Pten using Oxt-Cre has a profound effect on Oxt cell structure, but not on ASD-relevant behavior. We interpret these results as inconsistent with our starting hypothesis that reduced Pten function in Oxt-expressing cells causes the behavioral deficits observed in germline Pten+/- mice. Autism Res 2016, 9: 1248-1262. © 2016 International Society for Autism Research, Wiley Periodicals, Inc.


Asunto(s)
Trastorno Autístico/genética , Neuronas , Oxitocina/genética , Fosfohidrolasa PTEN/genética , Animales , Trastorno Autístico/fisiopatología , Conducta Animal , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
17.
PLoS One ; 10(8): e0136494, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26308619

RESUMEN

The development of social behavior is strongly influenced by the serotonin system. Serotonin 2c receptor (5-HT2cR) is particularly interesting in this context considering that pharmacological modulation of 5-HT2cR activity alters social interaction in adult rodents. However, the role of 5-HT2cR in the development of social behavior is unexplored. Here we address this using Htr2c knockout mice, which lack 5-HT2cR. We found that these animals exhibit social behavior deficits as adults but not as juveniles. Moreover, we found that the age of onset of these deficits displays similar timing as the onset of susceptibility to spontaneous death and audiogenic-seizures, consistent with the hypothesis that imbalanced excitation and inhibition (E/I) may contribute to social behavioral deficits. Given that autism spectrum disorder (ASD) features social behavioral deficits and is often co-morbid with epilepsy, and given that 5-HT2cR physically interacts with Pten, we tested whether a second site mutation in the ASD risk gene Pten can modify these phenotypes. The age of spontaneous death is accelerated in mice double mutant for Pten and Htr2c relative to single mutants. We hypothesized that pharmacological antagonism of 5-HT2cR activity in adult animals, which does not cause seizures, might modify social behavioral deficits in Pten haploinsufficient mice. SB 242084, a 5-HT2cR selective antagonist, can reverse the social behavior deficits observed in Pten haploinsufficient mice. Together, these results elucidate a role of 5-HT2cR in the modulation of social behavior and seizure susceptibility in the context of normal development and Pten haploinsufficiency.


Asunto(s)
Conducta Animal , Encéfalo/patología , Susceptibilidad a Enfermedades , Receptor de Serotonina 5-HT2C/fisiología , Convulsiones/etiología , Convulsiones/patología , Conducta Social , Animales , Encéfalo/metabolismo , Femenino , Haploinsuficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Serotonina
18.
J Neurosci ; 35(28): 10252-67, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26180201

RESUMEN

Abnormal patterns of head and brain growth are a replicated finding in a subset of individuals with autism spectrum disorder (ASD). It is not known whether risk factors associated with ASD and abnormal brain growth (both overgrowth and undergrowth) converge on common biological pathways and cellular mechanisms in the developing brain. Heterozygous mutations in PTEN (PTEN(+/-)), which encodes a negative regulator of the PI3K-Akt-mTOR pathway, are a risk factor for ASD and macrocephaly. Here we use the developing cerebral cortex of Pten(+/-) mice to investigate the trajectory of brain overgrowth and underlying cellular mechanisms. We find that overgrowth is detectable from birth to adulthood, is driven by hyperplasia, and coincides with excess neurons at birth and excess glia in adulthood. ß-Catenin signaling is elevated in the developing Pten(+/-) cortex, and a heterozygous mutation in Ctnnb1 (encoding ß-catenin), itself a candidate gene for ASD and microcephaly, can suppress Pten(+/-) cortical overgrowth. Thus, a balance of Pten and ß-catenin signaling regulates normal brain growth trajectory by controlling cell number, and imbalance in this relationship can result in abnormal brain growth. SIGNIFICANCE STATEMENT: We report that Pten haploinsufficiency leads to a dynamic trajectory of brain overgrowth during development and altered scaling of neuronal and glial cell populations. ß-catenin signaling is elevated in the developing cerebral cortex of Pten haploinsufficient mice, and a heterozygous mutation in ß-catenin, itself a candidate gene for ASD and microcephaly, suppresses Pten(+/-) cortical overgrowth. This leads to the new insight that Pten and ß-catenin signaling act in a common pathway to regulate normal brain growth trajectory by controlling cell number, and disruption of this pathway can result in abnormal brain growth.


Asunto(s)
Encéfalo , Regulación de la Expresión Génica/genética , Haploinsuficiencia/genética , Fosfohidrolasa PTEN/genética , Transducción de Señal/fisiología , beta Catenina/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/anomalías , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Proteínas Portadoras/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Embrión de Mamíferos , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , beta Catenina/genética
19.
Neurobiol Dis ; 82: 66-77, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26048156

RESUMEN

Huntington's disease (HD) is caused by an expansion of glutamine repeats in the huntingtin protein (mHtt) that invokes early and prominent damage of the striatum, a region that controls motor behaviors. Despite its ubiquitous expression, why certain brain regions, such as the cerebellum, are relatively spared from neuronal loss by mHtt remains unclear. Previously, we implicated the striatal-enriched GTPase, Rhes (Ras homolog enriched in the striatum), which binds and SUMOylates mHtt and increases its solubility and cellular cytotoxicity, as the cause for striatal toxicity in HD. Here, we report that Rhes deletion in HD mice (N171-82Q), which express the N-terminal fragment of human Htt with 82 glutamines (Rhes(-/-)/N171-82Q), display markedly reduced HD-related behavioral deficits, and absence of lateral ventricle dilatation (secondary to striatal atrophy), compared to control HD mice (N171-82Q). To further validate the role of GTPase Rhes in HD, we tested whether ectopic Rhes expression would elicit a pathology in a brain region normally less affected in HD. Remarkably, ectopic expression of Rhes in the cerebellum of N171-82Q mice, during the asymptomatic period led to an exacerbation of motor deficits, including loss of balance and motor incoordination with ataxia-like features, not apparent in control-injected N171-82Q mice or Rhes injected wild-type mice. Pathological and biochemical analysis of Rhes-injected N171-82Q mice revealed a cerebellar lesion with marked loss of Purkinje neuron layer parvalbumin-immunoreactivity, induction of caspase 3 activation, and enhanced soluble forms of mHtt. Similarly reintroducing Rhes into the striatum of Rhes deleted Rhes(-/-)Hdh(150Q/150Q) knock-in mice, elicited a progressive HD-associated rotarod deficit. Overall, these studies establish that Rhes plays a pivotal role in vivo for the selective toxicity of mHtt in HD.


Asunto(s)
Ataxia/genética , Cerebelo/metabolismo , Proteínas de Unión al GTP/genética , Enfermedad de Huntington/genética , Degeneraciones Espinocerebelosas/genética , Animales , Ataxia/metabolismo , Ataxia/patología , Cerebelo/patología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Proteínas de Unión al GTP/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Degeneraciones Espinocerebelosas/metabolismo , Degeneraciones Espinocerebelosas/patología
20.
Biol Psychiatry ; 77(9): 805-15, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25444158

RESUMEN

BACKGROUND: Genetic haploinsufficiency of SYNGAP1/Syngap1 commonly occurs in developmental brain disorders, such as intellectual disability, epilepsy, schizophrenia, and autism spectrum disorder. Thus, studying mouse models of Syngap1 haploinsufficiency may uncover pathologic developmental processes common among distinct brain disorders. METHODS: A Syngap1 haploinsufficiency model was used to explore the relationship between critical period dendritic spine abnormalities, cortical circuit assembly, and the window for genetic rescue to understand how damaging mutations disrupt key substrates of mouse brain development. RESULTS: Syngap1 mutations broadly disrupted a developmentally sensitive period that corresponded to the period of heightened postnatal cortical synaptogenesis. Pathogenic Syngap1 mutations caused a coordinated acceleration of dendrite elongation and spine morphogenesis and pruning of these structures in neonatal cortical pyramidal neurons. These mutations also prevented a form of developmental structural plasticity associated with experience-dependent reorganization of brain circuits. Consistent with these findings, Syngap1 mutant mice displayed an altered pattern of long-distance synaptic inputs into a cortical area important for cognition. Interestingly, the ability to genetically improve the behavioral endophenotype of Syngap1 mice decreased slowly over postnatal development and mapped onto the developmental period of coordinated dendritic insults. CONCLUSIONS: Pathogenic Syngap1 mutations have a profound impact on the dynamics and structural integrity of pyramidal cell postsynaptic structures known to guide the de novo wiring of nascent cortical circuits. These findings support the idea that disrupted critical periods of dendritic growth and spine plasticity may be a common pathologic process in developmental brain disorders.


Asunto(s)
Corteza Cerebral/anomalías , Corteza Cerebral/crecimiento & desarrollo , Haploinsuficiencia , Células Piramidales/fisiología , Proteínas Activadoras de ras GTPasa/deficiencia , Proteínas Activadoras de ras GTPasa/genética , Animales , Animales Recién Nacidos , Condicionamiento Psicológico/fisiología , Espinas Dendríticas/patología , Espinas Dendríticas/fisiología , Endofenotipos , Conducta Exploratoria/fisiología , Miedo/fisiología , Hipocampo/anomalías , Hipocampo/crecimiento & desarrollo , Aprendizaje por Laberinto/fisiología , Ratones Transgénicos , Vías Nerviosas/anomalías , Vías Nerviosas/crecimiento & desarrollo , Células Piramidales/patología , Privación Sensorial/fisiología , Vibrisas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...