Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
EMBO J ; 37(7)2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29282205

RESUMO

Neural development is accomplished by differentiation events leading to metabolic reprogramming. Glycosphingolipid metabolism is reprogrammed during neural development with a switch from globo- to ganglio-series glycosphingolipid production. Failure to execute this glycosphingolipid switch leads to neurodevelopmental disorders in humans, indicating that glycosphingolipids are key players in this process. Nevertheless, both the molecular mechanisms that control the glycosphingolipid switch and its function in neurodevelopment are poorly understood. Here, we describe a self-contained circuit that controls glycosphingolipid reprogramming and neural differentiation. We find that globo-series glycosphingolipids repress the epigenetic regulator of neuronal gene expression AUTS2. AUTS2 in turn binds and activates the promoter of the first and rate-limiting ganglioside-producing enzyme GM3 synthase, thus fostering the synthesis of gangliosides. By this mechanism, the globo-AUTS2 axis controls glycosphingolipid reprogramming and neural gene expression during neural differentiation, which involves this circuit in neurodevelopment and its defects in neuropathology.


Assuntos
Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Glicoesfingolipídeos/metabolismo , Neurogênese/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Reprogramação Celular/efeitos dos fármacos , Proteínas do Citoesqueleto , Epigenômica , Gangliosídeos/metabolismo , Expressão Gênica , Inativação Gênica , Glicoesfingolipídeos/farmacologia , Células HeLa , Histonas/metabolismo , Humanos , Transtornos do Neurodesenvolvimento , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Neurônios/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas/genética , Proteínas/metabolismo , Sialiltransferases/genética , Sialiltransferases/metabolismo , Fatores de Transcrição
2.
Genes Cells ; 26(3): 136-151, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33501714

RESUMO

The molecular mechanisms underlying neurodevelopmental disorders (NDDs) remain unclear. We previously identified Down syndrome cell adhesion molecule like 1 (Dscaml1) as a responsible gene for Ihara epileptic rat (IER), a rat model for human NDDs with epilepsy. However, the relationship between NDDs and DSCAML1 in humans is still elusive. In this study, we screened databases of autism spectrum disorders (ASD), intellectual disability (ID)/developmental disorders (DD) and schizophrenia for genomic mutations in human DSCAML1. We then performed in silico analyses to estimate the potential damage to the mutated DSCAML1 proteins and chose three representative mutations (DSCAML1C729R , DSCAML1R1685* and DSCAML1K2108Nfs*37 ), which lacked a cysteine residue in the seventh Ig domain, the intracellular region and the C-terminal PDZ-binding motif, respectively. In overexpression experiments in a cell line, DSCAML1C729R lost its mature N-glycosylation, whereas DSCAML1K2108Nfs*37 was abnormally degraded via proteasome-dependent protein degradation. Furthermore, in primary hippocampal neurons, the ability of the wild-type DSCAML1 to regulate the number of synapses was lost with all mutant proteins. These results provide insight into understanding the roles of the domains in the DSCAML1 protein and further suggest that these mutations cause functional changes, albeit through different mechanisms, that likely affect the pathophysiology of NDDs.


Assuntos
Moléculas de Adesão Celular/genética , Mutação/genética , Transtornos do Neurodesenvolvimento/genética , Animais , Transtorno do Espectro Autista/genética , Adesão Celular , Membrana Celular/metabolismo , Espinhas Dendríticas/metabolismo , Feminino , Glicosilação , Hipocampo/patologia , Humanos , Células L , Masculino , Camundongos , Anotação de Sequência Molecular , Proteínas Mutantes/metabolismo , Proteólise , Ratos Wistar , Esquizofrenia/genética , Sinapses/metabolismo
4.
Am J Kidney Dis ; 65(2): 312-21, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25218680

RESUMO

BACKGROUND: Brain atrophy has been reported in patients with end-stage renal disease receiving hemodialysis, although its mechanism is unknown. However, little is known regarding brain atrophy in patients receiving peritoneal dialysis (PD). Therefore, we examined brain volume and its annual change over 2 years in PD patients compared with patients with non-dialysis-dependent chronic kidney disease (NDD-CKD). STUDY DESIGN: Cross-sectional and longitudinal cohort. SETTING & PARTICIPANTS: 62 PD patients and 69 patients with NDD-CKD with no history of cerebrovascular disease who underwent brain magnetic resonance imaging (MRI) were recruited in a cross-sectional study. Among them, 34 PD patients and 61 patients with NDD-CKD, who underwent a second brain MRI after 2 years, were recruited in a longitudinal study. PREDICTOR: PD therapy versus NDD-CKD. OUTCOMES & MEASUREMENTS: T1-weighted magnetic resonance images were analyzed. Total gray matter volume (GMV), total white matter volume (WMV), and cerebrospinal fluid space volume were segmented, and each volume was quantified using statistical parametric mapping software. Normalized GMV and WMV values were calculated by division of GMV and WMV by intracranial volume to adjust for variations in head size. We compared normalized GMV and normalized WMV between PD patients and patients with NDD-CKD in the cross-sectional study and the annual change in normalized GMV in the longitudinal study. RESULTS: In the cross-sectional study, normalized GMV, which was correlated inversely with age, was lower in PD patients than in patients with NDD-CKD. However, normalized WMV, which was not correlated with age, was comparable between the groups. Annual change in normalized GMV was significantly higher in PD patients than in patients with NDD-CKD. These differences remained significant even after adjustment for potential confounding factors. LIMITATIONS: A short observation period and high dropout rate in the longitudinal study. CONCLUSIONS: Decline in normalized GMV is faster in PD patients than in patients with NDD-CKD.


Assuntos
Encéfalo/patologia , Diálise Peritoneal/efeitos adversos , Insuficiência Renal Crônica/epidemiologia , Insuficiência Renal Crônica/terapia , Idoso , Atrofia/diagnóstico , Atrofia/epidemiologia , Estudos de Coortes , Estudos Transversais , Feminino , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Diálise Peritoneal/tendências
5.
Nat Commun ; 15(1): 458, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38302444

RESUMO

In the central nervous system, astrocytes enable appropriate synapse function through glutamate clearance from the synaptic cleft; however, it remains unclear how astrocytic glutamate transporters function at peri-synaptic contact. Here, we report that Down syndrome cell adhesion molecule (DSCAM) in Purkinje cells controls synapse formation and function in the developing cerebellum. Dscam-mutant mice show defects in CF synapse translocation as is observed in loss of function mutations in the astrocytic glutamate transporter GLAST expressed in Bergmann glia. These mice show impaired glutamate clearance and the delocalization of GLAST away from the cleft of parallel fibre (PF) synapse. GLAST complexes with the extracellular domain of DSCAM. Riluzole, as an activator of GLAST-mediated uptake, rescues the proximal impairment in CF synapse formation in Purkinje cell-selective Dscam-deficient mice. DSCAM is required for motor learning, but not gross motor coordination. In conclusion, the intercellular association of synaptic and astrocyte proteins is important for synapse formation and function in neural transmission.


Assuntos
Neuroglia , Neurônios , Animais , Camundongos , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Cerebelo/metabolismo , Ácido Glutâmico/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Células de Purkinje/metabolismo , Sinapses/metabolismo
6.
Elife ; 122024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38529532

RESUMO

Increased levels of lactate, an end-product of glycolysis, have been proposed as a potential surrogate marker for metabolic changes during neuronal excitation. These changes in lactate levels can result in decreased brain pH, which has been implicated in patients with various neuropsychiatric disorders. We previously demonstrated that such alterations are commonly observed in five mouse models of schizophrenia, bipolar disorder, and autism, suggesting a shared endophenotype among these disorders rather than mere artifacts due to medications or agonal state. However, there is still limited research on this phenomenon in animal models, leaving its generality across other disease animal models uncertain. Moreover, the association between changes in brain lactate levels and specific behavioral abnormalities remains unclear. To address these gaps, the International Brain pH Project Consortium investigated brain pH and lactate levels in 109 strains/conditions of 2294 animals with genetic and other experimental manipulations relevant to neuropsychiatric disorders. Systematic analysis revealed that decreased brain pH and increased lactate levels were common features observed in multiple models of depression, epilepsy, Alzheimer's disease, and some additional schizophrenia models. While certain autism models also exhibited decreased pH and increased lactate levels, others showed the opposite pattern, potentially reflecting subpopulations within the autism spectrum. Furthermore, utilizing large-scale behavioral test battery, a multivariate cross-validated prediction analysis demonstrated that poor working memory performance was predominantly associated with increased brain lactate levels. Importantly, this association was confirmed in an independent cohort of animal models. Collectively, these findings suggest that altered brain pH and lactate levels, which could be attributed to dysregulated excitation/inhibition balance, may serve as transdiagnostic endophenotypes of debilitating neuropsychiatric disorders characterized by cognitive impairment, irrespective of their beneficial or detrimental nature.


Assuntos
Disfunção Cognitiva , Endofenótipos , Animais , Camundongos , Humanos , Encéfalo/metabolismo , Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Lactatos/metabolismo , Concentração de Íons de Hidrogênio
7.
Development ; 136(17): 2945-54, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19641016

RESUMO

PTF1-J is a trimeric transcription factor complex essential for generating the correct balance of GABAergic and glutamatergic interneurons in multiple regions of the nervous system, including the dorsal horn of the spinal cord and the cerebellum. Although the components of PTF1-J have been identified as the basic helix-loop-helix (bHLH) factor Ptf1a, its heterodimeric E-protein partner, and Rbpj, no neural targets are known for this transcription factor complex. Here we identify the neuronal differentiation gene Neurog2 (Ngn2, Math4A, neurogenin 2) as a direct target of PTF1-J. A Neurog2 dorsal neural tube enhancer localized 3' of the Neurog2 coding sequence was identified that requires a PTF1-J binding site for dorsal activity in mouse and chick neural tube. Gain and loss of Ptf1a function in vivo demonstrate its role in Neurog2 enhancer activity. Furthermore, chromatin immunoprecipitation from neural tube tissue demonstrates that Ptf1a is bound to the Neurog2 enhancer. Thus, Neurog2 expression is directly regulated by the PTF1-J complex, identifying Neurog2 as the first neural target of Ptf1a and revealing a bHLH transcription factor cascade functioning in the specification of GABAergic neurons in the dorsal spinal cord and cerebellum.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Medula Espinal/embriologia , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Sítios de Ligação , Diferenciação Celular/fisiologia , Embrião de Galinha , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/genética , Transdução de Sinais/fisiologia , Medula Espinal/citologia , Medula Espinal/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica
8.
Neural Plast ; 2012: 921732, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22830054

RESUMO

In the nervous system, there are a wide variety of neuronal cell types that have morphologically, physiologically, and histochemically different characteristics. These various types of neurons can be classified into two groups: excitatory and inhibitory neurons. The elaborate balance of the activities of the two types is very important to elicit higher brain function, because its imbalance may cause neurological disorders, such as epilepsy and hyperalgesia. In the central nervous system, inhibitory neurons are mainly represented by GABAergic ones with some exceptions such as glycinergic. Although the machinery to specify GABAergic neurons was first studied in the telencephalon, identification of key molecules, such as pancreatic transcription factor 1a (Ptf1a), as well as recently developed genetic lineage-tracing methods led to the better understanding of GABAergic specification in other brain regions, such as the spinal cord, the cerebellum, and the cochlear nucleus.


Assuntos
Cerebelo/citologia , Cerebelo/fisiologia , Núcleo Coclear/citologia , Núcleo Coclear/fisiologia , Neurônios/fisiologia , Medula Espinal/citologia , Medula Espinal/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Humanos , Interneurônios/fisiologia
9.
eNeuro ; 9(1)2022.
Artigo em Inglês | MEDLINE | ID: mdl-35082173

RESUMO

The neuropeptide oxytocin (Oxt) plays important roles in modulating social behaviors. Oxt receptor (Oxtr) is abundantly expressed in the brain and its relationship to socio-behavioral controls has been extensively studied using mouse brains. Several genetic tools to visualize and/or manipulate Oxtr-expressing cells, such as fluorescent reporters and Cre recombinase drivers, have been generated by ES-cell based gene targeting or bacterial artificial chromosome (BAC) transgenesis. However, these mouse lines displayed some differences in their Oxtr expression profiles probably because of the complex context and integrity of their genomic configurations in each line. Here, we apply our sophisticated genome-editing techniques to the Oxtr locus, systematically generating a series of knock-in mouse lines, in which its endogenous transcriptional regulations are intactly preserved and evaluate their expression profiles to ensure the reliability of our new tools. We employ the epitope tagging strategy, with which C-terminally fused tags can be detected by highly specific antibodies, to successfully visualize the Oxtr protein distribution on the neural membrane with super-resolution imaging for the first time. By using T2A self-cleaving peptide sequences, we also induce proper expressions of tdTomato reporter, codon-improved Cre recombinase (iCre), and spatiotemporally inducible Cre-ERT2 in Oxtr-expressing neurons. Electrophysiological recordings from tdTomato-positive cells in the reporter mice support the validity of our tool design. Retro-orbital injections of AAV-PHP.eB vector into the Cre line further enabled visualization of recombinase activities in the appropriate brain regions. Moreover, the first-time Cre-ERT2 line drives Cre-mediated recombination in a spatiotemporally controlled manner on tamoxifen (TMX) administration. These tools thus provide an excellent resource for future functional studies in Oxt-responsive neurons and should prove of broad interest in the field.


Assuntos
Neurônios , Receptores de Ocitocina , Animais , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Ocitocina/metabolismo , Receptores de Ocitocina/genética , Receptores de Ocitocina/metabolismo , Reprodutibilidade dos Testes , Comportamento Social
10.
Prog Neurobiol ; 216: 102288, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35654209

RESUMO

Duchenne muscular dystrophy (DMD) is a muscle disorder caused by DMD mutations and is characterized by neurobehavioural comorbidities due to dystrophin deficiency in the brain. The lack of Dp140, a dystrophin short isoform, is clinically associated with intellectual disability and autism spectrum disorders (ASDs), but its postnatal functional role is not well understood. To investigate synaptic function in the presence or absence of brain Dp140, we utilized two DMD mouse models, mdx23 and mdx52 mice, in which Dp140 is preserved or lacking, respectively. ASD-like behaviours were observed in pups and 8-week-old mdx52 mice lacking Dp140. Paired-pulse ratio of excitatory postsynaptic currents, glutamatergic vesicle number in basolateral amygdala neurons, and glutamatergic transmission in medial prefrontal cortex-basolateral amygdala projections were significantly reduced in mdx52 mice compared to those in wild-type and mdx23 mice. ASD-like behaviour and electrophysiological findings in mdx52 mice were ameliorated by restoration of Dp140 following intra-cerebroventricular injection of antisense oligonucleotide drug-induced exon 53 skipping or intra-basolateral amygdala administration of Dp140 mRNA-based drug. Our results implicate Dp140 in ASD-like behaviour via altered glutamatergic transmission in the basolateral amygdala of mdx52 mice.


Assuntos
Distrofina , Distrofia Muscular de Duchenne , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Distrofina/genética , Distrofina/metabolismo , Éxons , Camundongos , Distrofia Muscular de Duchenne/genética , Comportamento Social
11.
Cells ; 11(1)2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-35011572

RESUMO

Neurodevelopmental disorders (NDDs), including autism spectrum disorders (ASD) and intellectual disability (ID), are a large group of neuropsychiatric illnesses that occur during early brain development, resulting in a broad spectrum of syndromes affecting cognition, sociability, and sensory and motor functions. Despite progress in the discovery of various genetic risk factors thanks to the development of novel genomics technologies, the precise pathological mechanisms underlying the onset of NDDs remain elusive owing to the profound genetic and phenotypic heterogeneity of these conditions. Autism susceptibility candidate 2 (AUTS2) has emerged as a crucial gene associated with a wide range of neuropsychological disorders, such as ASD, ID, schizophrenia, and epilepsy. AUTS2 has been shown to be involved in multiple neurodevelopmental processes; in cell nuclei, it acts as a key transcriptional regulator in neurodevelopment, whereas in the cytoplasm, it participates in cerebral corticogenesis, including neuronal migration and neuritogenesis, through the control of cytoskeletal rearrangements. Postnatally, AUTS2 regulates the number of excitatory synapses to maintain the balance between excitation and inhibition in neural circuits. In this review, we summarize the knowledge regarding AUTS2, including its molecular and cellular functions in neurodevelopment, its genetics, and its role in behaviors.


Assuntos
Transtornos do Neurodesenvolvimento/genética , Fatores de Transcrição/genética , Animais , Citoesqueleto/metabolismo , Regulação da Expressão Gênica , Estudos de Associação Genética , Humanos , Neurogênese/genética , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
12.
PLoS One ; 16(7): e0254169, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34237104

RESUMO

BACKGROUND: Lower blood pressure (BP) levels are linked to a slower decline of kidney function in patients with chronic kidney disease (CKD) without kidney replacement therapy. However, there are limited data on this relation in peritoneal dialysis (PD) patients. Here we evaluated the association of BP levels with the decline of residual kidney function (RKF) in a retrospective cohort study. METHODS: We enrolled 228 patients whose PD was initiated between 1998 and 2014. RKF was measured as the average of creatinine and urea clearance in 24-hr urine collections. We calculated the annual decline rate of RKF by determining the regression line for individual patients. RKF is thought to decline exponentially, and thus we also calculated the annual decline rate of logarithmic scale of RKF (log RKF). We categorized the patients' BP levels at 3 months after PD initiation (BP3M) into four groups (Optimal, Normal & High normal, Grade 1 hypertension, Grade 2 & 3 hypertension) according to the 2018 European Society of Cardiology and European Society of Hypertension Guidelines for the management of arterial hypertension. RESULTS: The unadjusted, age- and sex-adjusted, and multivariable-adjusted decline rate of RKF and log RKF decreased significantly with higher BP3M levels (P for trend <0.01). Compared to those of the Optimal group, the multivariable-adjusted odds ratios (95% confidence interval) for the faster side of the median decline rate of RKF and log RKF were 4.04 (1.24-13.2) and 5.50 (1.58-19.2) in the Grade 2 and 3 hypertension group, respectively (p<0.05). CONCLUSIONS: Higher BP levels after PD initiation are associated with a faster decline in RKF among PD patients.


Assuntos
Pressão Sanguínea/fisiologia , Rim/fisiopatologia , Idoso , Creatinina/metabolismo , Progressão da Doença , Feminino , Taxa de Filtração Glomerular/fisiologia , Humanos , Testes de Função Renal/métodos , Masculino , Pessoa de Meia-Idade , Diálise Peritoneal/métodos , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/fisiopatologia , Estudos Retrospectivos
13.
Cells ; 10(5)2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33946570

RESUMO

Fluorescent reporter mouse lines and Cre/Flp recombinase driver lines play essential roles in investigating various molecular functions in vivo. Now that applications of the CRISPR/Cas9 genome-editing system to mouse fertilized eggs have drastically accelerated these knock-in mouse generations, the next need is to establish easier, quicker, and cheaper methods for knock-in donor preparation. Here, we reverify and optimize the phospho-PCR method to obtain highly pure long single-stranded DNAs (ssDNAs) suitable for knock-in mouse generation via genome editing. The sophisticated sequential use of two exonucleases, in which double-stranded DNAs (dsDNAs) amplified by a pair of 5'-phosphorylated primer and normal primer are digested by Lambda exonuclease to yield ssDNA and the following Exonuclease III treatment degrades the remaining dsDNAs, enables much easier long ssDNA productions without laborious gel extraction steps. By microinjecting these donor DNAs along with CRISPR/Cas9 components into mouse zygotes, we have effectively generated fluorescent reporter lines and recombinase drivers. To further broaden the applicability, we have prepared long ssDNA donors in higher concentrations and electroporated them into mouse eggs to successfully obtain knock-in embryos. This classical yet improved method, which is regaining attention on the progress of CRISPR/Cas9 development, shall be the first choice for long donor DNA preparation, and the resulting knock-in lines could accelerate life science research.


Assuntos
DNA de Cadeia Simples/normas , Técnicas de Introdução de Genes/métodos , Animais , Sistemas CRISPR-Cas , DNA de Cadeia Simples/química , DNA de Cadeia Simples/genética , Eletroporação/métodos , Edição de Genes/métodos , Camundongos , Camundongos Transgênicos , Microinjeções/métodos , Reação em Cadeia da Polimerase/métodos , Zigoto/metabolismo
14.
iScience ; 23(12): 101820, 2020 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-33305180

RESUMO

Autism susceptibility candidate 2 (AUTS2), a risk gene for autism spectrum disorders (ASDs), is implicated in telencephalon development. Because AUTS2 is also expressed in the cerebellum where defects have been linked to ASDs, we investigated AUTS2 functions in the cerebellum. AUTS2 is specifically localized in Purkinje cells (PCs) and Golgi cells during postnatal development. Auts2 conditional knockout (cKO) mice exhibited smaller and deformed cerebella containing immature-shaped PCs with reduced expression of Cacna1a. Auts2 cKO and knock-down experiments implicated AUTS2 participation in elimination and translocation of climbing fiber synapses and restriction of parallel fiber synapse numbers. Auts2 cKO mice exhibited behavioral impairments in motor learning and vocal communications. Because Cacna1a is known to regulate synapse development in PCs, it suggests that AUTS2 is required for PC maturation to elicit normal development of PC synapses and thus the impairment of AUTS2 may cause cerebellar dysfunction related to psychiatric illnesses such as ASDs.

15.
iScience ; 23(6): 101183, 2020 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-32498016

RESUMO

Impairments in synapse development are thought to cause numerous psychiatric disorders. Autism susceptibility candidate 2 (AUTS2) gene has been associated with various psychiatric disorders, such as autism and intellectual disabilities. Although roles for AUTS2 in neuronal migration and neuritogenesis have been reported, its involvement in synapse regulation remains unclear. In this study, we found that excitatory synapses were specifically increased in the Auts2-deficient primary cultured neurons as well as Auts2 mutant forebrains. Electrophysiological recordings and immunostaining showed increases in excitatory synaptic inputs as well as c-fos expression in Auts2 mutant brains, suggesting that an altered balance of excitatory and inhibitory inputs enhances brain excitability. Auts2 mutant mice exhibited autistic-like behaviors including impairments in social interaction and altered vocal communication. Together, these findings suggest that AUTS2 regulates excitatory synapse number to coordinate E/I balance in the brain, whose impairment may underlie the pathology of psychiatric disorders in individuals with AUTS2 mutations.

16.
Dev Biol ; 322(2): 381-93, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18721803

RESUMO

Dorsal horn neurons express many different neuropeptides that modulate sensory perception like the sensation of pain. Inhibitory neurons of the dorsal horn derive from postmitotic neurons that express Pax2, Lbx1 and Lhx1/5, and diversify during maturation. In particular, fractions of maturing inhibitory neurons express various neuropeptides. We demonstrate here that a coordinate molecular mechanism determines inhibitory and peptidergic fate in the developing dorsal horn. A bHLH factor complex that contains Ptf1a acts as upstream regulator and initiates the expression of several downstream transcription factors in the future inhibitory neurons, of which Pax2 is known to determine the neurotransmitter phenotype. We demonstrate here that dynorphin, galanin, NPY, nociceptin and enkephalin expression depends on Ptf1a, indicating that these neuropeptides are expressed in inhibitory neurons. Furthermore, we show that Neurod1/2/6 and Lhx1/5, which act downstream of Ptf1a, control distinct aspects of peptidergic differentiation. In particular, the Neurod1/2/6 factors are essential for dynorphin and galanin expression, whereas the Lhx1/5 factors are essential for NPY expression. We conclude that a transcriptional network operates in maturing dorsal horn neurons that coordinately determines transmitter and peptidergic fate.


Assuntos
Neuropeptídeos/biossíntese , Células do Corno Posterior/embriologia , Medula Espinal/embriologia , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Gânglios Espinais/citologia , Gânglios Espinais/embriologia , Gânglios Espinais/metabolismo , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Proteínas com Homeodomínio LIM , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Células do Corno Posterior/metabolismo , Medula Espinal/citologia , Medula Espinal/metabolismo
17.
Neuron ; 100(5): 1097-1115.e15, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-30392800

RESUMO

The establishment of axon-dendrite polarity is fundamental for radial migration of neurons during cortex development of mammals. We demonstrate that the E3 ubiquitin ligases WW-Containing Proteins 1 and 2 (Wwp1 and Wwp2) are indispensable for proper polarization of developing neurons. We show that knockout of Wwp1 and Wwp2 results in defects in axon-dendrite polarity in pyramidal neurons, and their aberrant laminar cortical distribution. Knockout of miR-140, encoded in Wwp2 intron, engenders phenotypic changes analogous to those upon Wwp1 and Wwp2 deletion. Intriguingly, transcription of the Wwp1 and Wwp2/miR-140 loci in neurons is induced by the transcription factor Sox9. Finally, we provide evidence that miR-140 supervises the establishment of axon-dendrite polarity through repression of Fyn kinase mRNA. Our data delineate a novel regulatory pathway that involves Sox9-[Wwp1/Wwp2/miR-140]-Fyn required for axon specification, acquisition of pyramidal morphology, and proper laminar distribution of cortical neurons.


Assuntos
Polaridade Celular , Córtex Cerebral/crescimento & desenvolvimento , MicroRNAs/fisiologia , Neurônios/fisiologia , Fatores de Transcrição SOX9/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Axônios/fisiologia , Córtex Cerebral/citologia , Dendritos/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos Knockout , MicroRNAs/genética , Neurônios/citologia , Fatores de Transcrição SOX9/genética , Ubiquitina-Proteína Ligases/genética
18.
Brain Sci ; 7(5)2017 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-28505103

RESUMO

Neuronal migration is one of the pivotal steps to form a functional brain, and disorganization of this process is believed to underlie the pathology of psychiatric disorders including schizophrenia, autism spectrum disorders (ASD) and epilepsy. However, it is not clear how abnormal neuronal migration causes mental dysfunction. Recently, a key gene for various psychiatric diseases, the Autism susceptibility candidate 2 (AUTS2), has been shown to regulate neuronal migration, which gives new insight into understanding this question. Interestingly, the AUTS2 protein has dual functions: Cytoplasmic AUTS2 regulates actin cytoskeleton to control neuronal migration and neurite extension, while nuclear AUTS2 controls transcription of various genes as a component of the polycomb complex 1 (PRC1). In this review, we discuss AUTS2 from the viewpoint of human genetics, molecular function, brain development, and behavior in animal models, focusing on its role in neuronal migration.

19.
J Neurosci ; 25(6): 1421-30, 2005 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-15703396

RESUMO

Recent studies have reported on the molecular mechanisms underlying dendritic spine (spine) dynamics. Because most of these studies investigated spine dynamics by overexpressing constitutively active or dominant-negative PSD (postsynaptic density) proteins in cultured mature neurons, the results represent the enlargement of mature spines or their return to an immature state. Here, we developed the technique of in utero electroporation to investigate spine dynamics. Using this technique, we demonstrated the suppression of spine maturation by the C-terminal variants of PSD-Zip70 in vitro and in vivo. Transient overexpression of the C terminus of PSD-Zip70 and knock-down of PSD-Zip70 also displayed the destabilization of mature spines. We further found the PSD-Zip70 and SPAR (spine-associated RapGAP) interaction via the short C-terminal region of PSD-Zip70 and the GK-binding domain of SPAR. In association with immature spines induced by overexpression of the PSD-Zip70 C terminus or knock-down of PSD-Zip70, SPAR lost its spine localization. Overexpression of the GK-binding domain of SPAR also induced to form immature spines without affecting the localization of PSD-Zip70 in the small heads of filopodial spines. Our results suggest that PSD-Zip70 in collaboration with SPAR is critically involved in spine maturity, especially in the mature spine formation and the maintenance of spine maturity.


Assuntos
Dendritos/ultraestrutura , Eletroporação/métodos , Proteínas Ativadoras de GTPase/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Animais , Encéfalo/citologia , Encéfalo/embriologia , Dendritos/metabolismo , Feminino , Proteínas Fetais/química , Proteínas Fetais/fisiologia , Proteínas Ativadoras de GTPase/química , Marcação de Genes , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Idade Gestacional , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos ICR , Microinjeções , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Gravidez , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Pseudópodes/metabolismo , Pseudópodes/ultraestrutura , Transfecção
20.
J Neurosci ; 25(10): 2670-81, 2005 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-15758177

RESUMO

The activity-dependent remodeling of postsynaptic structure is a fundamental process underlying learning and memory. Insulin receptor substrate p53 (IRSp53), a key player in cytoskeletal dynamics, is enriched in the postsynaptic density (PSD) fraction, but its significance in synaptic functions remains unclear. We report here that IRSp53 is accumulated rapidly at the postsynaptic sites of cultured hippocampal neurons after glutamate or NMDA stimulation in an actin cytoskeleton-dependent manner. Pharmacological profiles showed that a PKC inhibitor, but not other kinase inhibitors, specifically suppressed the synaptic translocation of IRSp53 in response to NMDA, and the selective activation of PKC with phorbol ester markedly induced the synaptic translocation. Reverse transcriptase-PCR and Western blotting showed that IRSp53-S is the major isoform expressed in cultured hippocampal neurons. The synaptic targeting of IRSp53-S was found to be mediated through N-terminal coiled-coil domain and the PDZ (PSD-95/Discs large/zona occludens-1)-binding sequence at its C-terminal end and regulated by the PKC phosphorylation of its N terminus. In electrophysiological experiments, overexpression of IRSp53-S wild type and IRSp53-S mutant that is spontaneously accumulated at the postsynaptic sites enhanced the postsynaptic function as detected by an increased miniature EPSC amplitude. These data suggest that IRSp53 is involved in NMDA receptor-linked synaptic plasticity via PKC signaling.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteína Quinase C/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Humanos , Insulina/farmacologia , N-Metilaspartato/farmacologia , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Proteína Quinase C/genética , Transporte Proteico/fisiologia , Ratos , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/genética , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa