Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cells ; 13(6)2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38534384

RESUMEN

POGZ (Pogo transposable element derived with ZNF domain) is known to function as a regulator of gene expression. While variations in the POGZ gene have been associated with intellectual disabilities and developmental delays in humans, the exact pathophysiological mechanisms remain unclear. To shed light on this, we created two lines of conditional knockout mice for Pogz, one specific to excitatory neurons (Emx1-Pogz mice) and the other to inhibitory neurons (Gad2-Pogz mice) in the brain. Emx1-Pogz mice showed a decrease in body weight, similar to total Pogz knockout mice. Although the two lines did not display significant morphological abnormalities in the telencephalon, impaired POGZ function affected the electrophysiological properties of both excitatory and inhibitory neurons differently. These findings suggest that these mouse lines could be useful tools for clarifying the precise pathophysiological mechanisms of neurodevelopmental disorders associated with POGZ gene abnormalities.


Asunto(s)
Discapacidad Intelectual , Trastornos del Neurodesarrollo , Animales , Humanos , Ratones , Encéfalo , Discapacidad Intelectual/genética , Ratones Noqueados , Trastornos del Neurodesarrollo/genética , Neuronas/metabolismo
2.
Mol Biol Rep ; 51(1): 106, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38227057

RESUMEN

BACKGROUND: ARF (ADP-ribosylation factor) GTPases are major regulators of intracellular trafficking, and classified into 3 groups (Type I - III), among which the type I group members, ARF1 and 3, are responsible genes for neurodevelopmental disorders. METHODS: In this study, we analysed the expression of Type I ARFs ARF1-3 during mouse brain development using biochemical and morphological methods. RESULTS: Western blotting analyses revealed that ARF1-3 are weakly expressed in the mouse brain at embryonic day 13 and gradually increase until postnatal day 30. ARF1-3 appear to be abundantly expressed in various telencephalon regions. Biochemical fractionation studies detected ARF1-3 in the synaptosome fraction of cortical neurons containing both pre- and post-synapses, however ARF1-3 were not observed in post-synaptic compartments. In immunohistochemical analyses, ARF1-3 appeared to be distributed in the cytoplasm and dendrites of cortical and hippocampal neurons as well as in the cerebellar molecular layer including dendrites of Purkinje cells and granule cell axons. Immunofluorescence in primary cultured hippocampal neurons revealed that ARF1-3 are diffusely distributed in the cytoplasm and dendrites with partial colocalization with a pre-synaptic marker, synaptophysin. CONCLUSIONS: Overall, our results support the notion that ARF1-3 could participate in vesicle trafficking both in the dendritic shaft (excluding spines) and axon terminals (pre-synaptic compartments).


Asunto(s)
Proteínas de Unión al GTP Monoméricas , Animales , Ratones , Factores de Ribosilacion-ADP/genética , Neuronas , Axones , Cerebelo
3.
Dev Neurosci ; 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37906993

RESUMEN

INTRODUCTION: CtBP1 (C-terminal-binding protein 1) is a multi-functional protein with well-established roles as a transcriptional co-repressor in the nucleus and a regulator of membrane fission in the cytoplasm. Although CtBP1 gene abnormalities have been reported to cause neurodevelopmental disorders, the physiological role and expression profile of CtBP1 remains to be elucidated. METHODS: In this study, we used biochemical, immunohistochemical and immunofluorescence methods to analyze the expression of CtBP1 during mouse brain development. RESULTS: Western blotting analyses revealed that CtBP1 appeared to be expressed mainly in the central nervous system throughout the developmental process. In immunohistochemical analyses, region-specific nuclear as well as weak cytoplasmic distribution of CtBP1 was observed in telencephalon at embryonic day (E)15 and E17. It is of note that CtBP1 was barely detected in axons, but observed in the nucleus of oligodendrocytes in the white matter at E17. As to cerebellum at postnatal day 30, CtBP1 appeared to be expressed in the nucleus and cytoplasm of Purkinje cells, the nucleus of granule cells and cells in the molecular layer (ML), and the ML per se where granule cell axons and Purkinje cell dendrites are enriched. In addition, CtBP1 was detected in the cerebellar nuclei. CONCLUSION: The obtained results suggest involvement of CtBP1 in brain function.

4.
J Neurochem ; 165(3): 334-347, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36798993

RESUMEN

The mediator complex comprises multiple subcellular subunits that collectively function as a molecular interface between RNA polymerase II and gene-specific transcription factors. Recently, genetic variants to one subunit of the complex, known as MED13L (mediator complex subunit 13 like), have been implicated in syndromic intellectual disability and distinct facial features, frequently accompanied by congenital heart defects. We investigated the impact of five disease-associated MED13L variants on the subcellular localization and biochemical stability of MED13L protein in vitro and in vivo. In overexpression assays using cortical neurons from embryonic mouse cerebral cortices transduced by in utero electroporation-mediated gene transfer, we found that mouse orthologues of human MED13L-p.P866L and -p.T2162M missense variants accumulated in the nucleus, while the p.S2163L and p.S2177Y variants were diffusely distributed in the cytoplasm. In contrast, we found that the p.Q1922* truncation variant was barely detectable in transduced cells, a phenotype reminiscent of this variant that results in MED13L haploinsufficiency in humans. Next, we analyzed these variants for their effects on neuronal migration, dendritic growth, spine morphology, and axon elongation of cortical neurons in vivo. There, we found that overexpression of the p.P866L variant resulted in reduced number and length of dendrites of cortical layer II/III pyramidal neurons. Furthermore, we show that mMED13L-knockdown abrogated dendritic growth in vivo, and this effect was significantly rescued by co-electroporation of an RNAi-resistant mMED13L, but weakly by the p.T2162M variant, and not at all by the p.S2163L variant. However, overexpression of the p.S2163L variant inhibited mature dendritic spine formation in vivo. Expression of each of the 5 variants did not affect neuronal cell migration and callosal axon elongation in vivo. Taken together, our results demonstrate that MED13L expression is relevant to corticogenesis and influences the dendritic branching characteristics of cortical excitatory neurons. Our study also suggests that disease-associated MED13L variants may directly cause morphological and functional defects in cortical neurons in different ways.


Asunto(s)
Discapacidad Intelectual , Complejo Mediador , Neuronas , Animales , Humanos , Ratones , Encéfalo , Corteza Cerebral , Discapacidad Intelectual/genética , Mamíferos , Complejo Mediador/metabolismo , Fenotipo , Factores de Transcripción/genética
5.
Dev Neurosci ; 44(6): 643-650, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36067731

RESUMEN

Polo-like kinase 4 (Plk4) is a ser/thr kinase, which plays a central role in centriole duplication during the cell cycle. PLK4 gene abnormalities are responsible for autosomal recessive chorioretinopathy-microcephaly syndrome and Seckel syndrome. In this study, we performed expression analyses of Plk4 by focusing on mouse brain development. Western blotting analyses revealed that Plk4 with a molecular mass of ∼100 kDa was broadly expressed in adult mouse tissues with specific subcellular distribution. As to the central nervous system, Plk4 was expressed throughout the developmental process with drastic increase after P15, suggesting an essential role of Plk4 in differentiated neurons. In immunohistochemical analyses with mouse brain at embryonic day 14, Plk4 was detected dominantly at the cell-cell contact sites of neuronal progenitors in the ventricular zone. Plk4 was then diffusely distributed in the cell body of cortical neurons at P7, while it was enriched in the neuropil as well as soma of excitatory neurons in the cerebral cortex and hippocampus and Purkinje cells in the cerebellum at P30. Notably, biochemical fractionation analysis found an enrichment of Plk4 in the postsynaptic density fraction. Then, immunofluorescent analyses showed partial co-localization of Plk4 with excitatory synaptic markers, PSD95 and synaptophysin, in differentiated primary cultured hippocampal neurons. These results suggest that Plk4 takes part in the regulation of synaptic function in differentiated neurons.


Asunto(s)
Microcefalia , Animales , Ratones , Microcefalia/genética , Ciclo Celular , División Celular , Neuronas , Encéfalo
6.
Dev Neurosci ; 44(3): 162-170, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35259752

RESUMEN

Centrosomal protein 152 (Cep152) regulates centriole duplication as a molecular scaffold during the cell cycle. Its gene abnormalities are responsible for autosomal recessive primary microcephaly 9 and Seckel syndrome. In this study, we prepared an antibody against mouse Cep152, anti-Cep152, and performed expression analyses focusing on mouse brain development. Western blotting analyses revealed that Cep152 with a molecular mass of ∼150 kDa was expressed strongly at embryonic day (E)13 and then gradually decreased during the brain development process. Instead, protein bands of ∼80 kDa and ∼60 kDa came to be recognized after postnatal day (P)15 and P30, respectively. In immunohistochemical analyses, Cep152 was enriched in the centrosome of neuronal progenitors in the ventricular zone at E14, whereas it was diffusely distributed mainly in the cytoplasm of cortical neurons at P18. In developing cerebellum at P7, Cep152 was localized at the centrosome in the external granular layer, where neurogenesis takes place. Notably, biochemical analysis revealed that Cep152 was also present in the postsynaptic density fraction. Subsequent immunofluorescent analyses showed co-localization of Cep152 with excitatory synaptic markers, PSD95 and synaptophysin, but not with an inhibitory synaptic marker gephyrin in differentiated primary cultured hippocampal neurons. The obtained results suggest that Cep152 takes part not only in neurogenesis during corticogenesis but also in the regulation of synaptic function in differentiated neurons.


Asunto(s)
Microcefalia , Animales , Hipocampo/metabolismo , Ratones , Microcefalia/genética , Microcefalia/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo
7.
Dev Neurosci ; 43(1): 43-52, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33794529

RESUMEN

MED13L (mediator complex subunit 13-like) is a component of the mediator complex, which functions as a regulator for gene transcription. Since gene abnormalities in MED13L are responsible for neurodevelopmental disorders, MED13L is presumed to play an essential role in brain development. In this study, we prepared a specific antibody against MED13L, anti-MED13L, and analyzed its expression profile in mouse tissues with focusing on the central nervous system. In Western blotting, MED13L exhibited a tissue-dependent expression profile in the adult mouse and was expressed in a developmental stage-dependent manner in brain. In immunofluorescence analyses, MED13L was at least partially colocalized with pre- and post-synaptic markers, synaptophysin, and PSD95, in primary cultured hippocampal neurons. Immunohistochemical analyses revealed that MED13L was relatively highly expressed in ventricular zone surface of cerebral cortex, and was also located both in the cytoplasm and nucleus of neurons in the cortical plate at embryonic day 14. Then, MED13L showed diffuse cytoplasmic distribution throughout the cerebral cortex at the postnatal day (P) 30. In addition, MED13L appeared to be localized in cell type- and developmental stage-specific manners in the hippocampus and cerebellum. These results suggest that MED13L is involved in the development of the central nervous system and synaptic function.


Asunto(s)
Trastornos del Neurodesarrollo , Neuronas , Animales , Encéfalo , Hipocampo , Complejo Mediador/genética , Ratones , Trastornos del Neurodesarrollo/genética
8.
J Neurochem ; 157(4): 1167-1181, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33025585

RESUMEN

Heterotrimeric G-proteins are composed of α, ß, and γ subunits, and function as signal transducers. Critical roles of the α-subunits of Gi/o family heterotrimeric G-proteins, Gαi2, and Gαo1, have so far been reported in brain development and neurodevelopmental disorders. In this study, we tried to clarify the role of Gαi1, α-subunit of another Gi/o family member Gi1, during corticogenesis, based on the recent identification of its gene abnormalities in neurodevelopmental disorders. In western blot analyses, Gαi1 was found to be expressed in mouse brain in a developmental stage-dependent manner. Morphological analyses revealed that Gαi1 was broadly distributed in cerebral cortex with relatively high expression in the ventricular zone (VZ) at embryonic day (E) 14. Meanwhile, Gαi1 was enriched in membrane area of yet unidentified early mitotic cells in the VZ and the marginal zone at E14. Acute knockdown of Gαi1 with in utero electroporation in cerebral cortex caused cell cycle elongation of the neural progenitor cells and promoted their cell cycle exit. Gαi1-deficient cortical neurons also exhibited delayed radial migration during corticogenesis, with abnormally elongated leading processes and hampered nucleokinesis. In addition, silencing of Gαi1 prevented basal dendrite development. The migration and dendritic phenotypes were at least partially rescued by an RNAi-resistant version of Gαi1. Collectively, these results strongly suggest a crucial role of Gi1 in cortical development, and disturbance of its function may cause deficits in synaptic network formation, leading to neurodevelopmental disorders.


Asunto(s)
Corteza Cerebral/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Animales , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Dendritas/metabolismo , Ratones , Ratones Endogámicos ICR , Neuronas/metabolismo
9.
Biochem Biophys Res Commun ; 529(4): 1033-1037, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32819561

RESUMEN

Takenouchi-Kosaki syndrome (TKS) is an autosomal dominant congenital syndrome, of which pathogenesis is not well understood. Recently, a heterozygous mutation c.1449T > C/p.(Tyr64Cys) in the CDC42 gene, encoding a Rho family small GTPase, has been demonstrated to contribute to the TKS clinical features, including developmental delay with intellectual disability (ID). However, specific molecular mechanisms underlying the neuronal pathophysiology of TKS remain largely unknown. In this study, biochemical analyses revealed that the mutation moderately activates Cdc42. In utero electroporation-based acute expression of Cdc42-Y64C in ventricular zone progenitor cells in embryonic mice cerebral cortex resulted in migration defects and cluster formation of excitatory neurons. Expression the mutant in primary cultured hippocampal neurons caused impaired axon elongation. These data suggest that the c.1449T > C/p.(Tyr64Cys) mutation causes altered CDC42 function and results in defects in neuronal morphology and migration during brain development, which is likely to be responsible for pathophysiology of psychomotor delay and ID in TKS.


Asunto(s)
Encéfalo/patología , Encéfalo/fisiopatología , Predisposición Genética a la Enfermedad , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Proteína de Unión al GTP cdc42/genética , Animales , Axones/metabolismo , Células COS , Agregación Celular , Movimiento Celular , Células Cultivadas , Corteza Cerebral , Chlorocebus aethiops , Hipocampo/patología , Ratones Endogámicos ICR , Proteínas Mutantes/metabolismo , Neuritas/metabolismo , Organogénesis , Síndrome
10.
Med Mol Morphol ; 53(4): 221-228, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32146512

RESUMEN

Septins are a highly conserved family of GTPases which are identified in diverse organisms ranging from yeast to humans. In mammals, nervous tissues abundantly contain septins and associations of septins with neurological disorders such as Alzheimer's disease and Parkinson's disease have been reported. However, roles of septins in the brain development have not been fully understood. In this study, we produced a specific antibody against mouse SEPT1 and carried out biochemical and morphological characterization of SEPT1. When the expression profile of SEPT1 during mouse brain development was analyzed by western blotting, we found that SEPT1 expression began to increase after birth and the increase continued until postnatal day 22. Subcellular fractionation of mouse brain and subsequent western blot analysis revealed the distribution of SEPT1 in synaptic fractions. Immunofluorescent analyses showed the localization of SEPT1 at synapses in primary cultured mouse hippocampal neurons. We also found the distribution of SEPT1 at synapses in mouse brain by immunohistochemistry. These results suggest that SEPT1 participates in various synaptic events such as the signaling, the neurotransmitter release, and the synapse formation/maintenance.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hipocampo/crecimiento & desarrollo , Septinas/metabolismo , Animales , Animales Recién Nacidos , Células COS , Chlorocebus aethiops , Embrión de Mamíferos , Perfilación de la Expresión Génica , Hipocampo/citología , Hipocampo/metabolismo , Inmunohistoquímica , Masculino , Ratones , Neuronas/metabolismo , Cultivo Primario de Células , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Septinas/análisis , Septinas/genética , Transducción de Señal/genética , Sinapsis/metabolismo
11.
Dev Neurosci ; 41(1-2): 139-148, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31430754

RESUMEN

POGZ is a heterochromatin protein 1 α-binding protein and regulates gene expression. On the other hand, accumulating pieces of evidence indicate that the POGZ gene abnormalities are involved in various neurodevelopmental disorders. In this study, we prepared a specific antibody against POGZ, anti-POGZ, and carried out biochemical and morphological characterization with mouse brain tissues. Western blotting analyses revealed that POGZ is expressed strongly at embryonic day 13 and then gradually decreased throughout the brain development process. In immunohistochemical analyses, POGZ was found to be enriched in cerebrocortical and hippocampal neurons in the early developmental stage. The nuclear expression was also detected in Purkinje cells in cerebellum at postnatal day (P)7 and P15 but disappeared at P30. In primary cultured hippocampal neurons, while POGZ was distributed mainly in the nucleus, it was also visualized in axon and dendrites with partial localization at synapses in consistency with the results obtained in biochemical fractionation analyses. The obtained results suggest that POGZ takes part in the regulation of synaptic function as well as gene expression during brain development.


Asunto(s)
Encéfalo/metabolismo , Neurogénesis/fisiología , Transposasas/metabolismo , Animales , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo
12.
Sci Rep ; 9(1): 5874, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30971765

RESUMEN

Per3 is one of the primary components of circadian clock system. While circadian dysregulation is known to be involved in the pathogenesis of several neuropsychiatric diseases. It remains largely unknown whether they participate in embryonic brain development. Here, we examined the role of clock gene Per3 in the development of mouse cerebral cortex. In situ hybridization analysis revealed that Per3 is expressed in the developing mouse cortex. Acute knockdown of Per3 with in utero electroporation caused abnormal positioning of cortical neurons, which was rescued by RNAi-resistant Per3. Per3-deficient cells showed abnormal migration phenotypes, impaired axon extension and dendritic arbor formation. Taken together, Per3 was found to play a pivotal role in corticogenesis via regulation of excitatory neuron migration and synaptic network formation.


Asunto(s)
Corteza Cerebral/metabolismo , Desarrollo Embrionario/genética , Proteínas Circadianas Period/genética , Animales , Axones/fisiología , Movimiento Celular , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/patología , Hibridación Fluorescente in Situ , Ratones , Neuronas/citología , Neuronas/metabolismo , Proteínas Circadianas Period/antagonistas & inhibidores , Proteínas Circadianas Period/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Células Madre/citología , Células Madre/metabolismo , Imagen de Lapso de Tiempo
13.
Brain ; 141(11): 3098-3114, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30256902

RESUMEN

Trio-based whole exome sequencing identified two de novo heterozygous missense mutations [c.1449T > C/p.(Leu500Pro) and c.1436A > T/p.(Asn479Ile)] in PHACTR1, encoding a molecule critical for the regulation of protein phosphatase 1 (PP1) and the actin cytoskeleton, in unrelated Japanese individuals with West syndrome (infantile spasms with intellectual disability). We then examined the role of Phactr1 in the development of mouse cerebral cortex and the pathophysiological significance of these two mutations and others [c.1561C > T/p.(Arg521Cys) and c.1553T > A/p.(Ile518Asn)], which had been reported in undiagnosed patients with intellectual disability. Immunoprecipitation analyses revealed that actin-binding activity of PHACTR1 was impaired by the p.Leu500Pro, p.Asn479Ile and p.Ile518Asn mutations while the p.Arg521Cys mutation exhibited impaired binding to PP1. Acute knockdown of mouse Phactr1 using in utero electroporation caused defects in cortical neuron migration during corticogenesis, which were rescued by an RNAi-resistant PHACTR1 but not by the four mutants. Experiments using knockdown combined with expression mutants, aimed to mimic the effects of the heterozygous mutations under conditions of haploinsufficiency, suggested a dominant negative effect of the mutant allele. As for dendritic development in vivo, only the p.Arg521Cys mutant was determined to have dominant negative effects, because the three other mutants appeared to be degraded with these experimental conditions. Electrophysiological analyses revealed abnormal synaptic properties in Phactr1-deficient excitatory cortical neurons. Our data show that the PHACTR1 mutations may cause morphological and functional defects in cortical neurons during brain development, which is likely to be related to the pathophysiology of West syndrome and other neurodevelopmental disorders.


Asunto(s)
Salud de la Familia , Proteínas de Microfilamentos/genética , Mutación/genética , Espasmos Infantiles/genética , Espasmos Infantiles/fisiopatología , Animales , Células COS , Movimiento Celular/genética , Células Cultivadas , Chlorocebus aethiops , Embrión de Mamíferos , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Humanos , Lactante , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , N-Metilaspartato/farmacología , Plasticidad Neuronal/genética , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Urea/administración & dosificación , Urea/análogos & derivados
14.
Acta Histochem Cytochem ; 51(3): 119-128, 2018 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-30083020

RESUMEN

ARHGEF9, also known as Collybistin, a guanine nucleotide exchange factor for Rho family GTPases, is thought to play an essential role in the mammalian brain. In this study, we prepared a specific polyclonal antibody against ARHGEF9, anti-ARHGEF9, and carried out expression analyses with mouse tissues especially brain. Western blotting analyses demonstrated tissue-dependent expression profiles of ARHGEF9 in the young adult mouse, and strongly suggested a role during brain development. Immunohistochemical analyses revealed developmental stage-dependent expression profiles of ARHGEF9 in cerebral cortex, hippocampus and cerebellum. ARHGEF9 exhibited partial localization at dendritic spines in cultured hippocampal neurons. From the obtained results, anti-ARHGEF9 was found to be a useful tool for biochemical and cell biological analyses of ARHGEF9.

15.
Neurosci Res ; 128: 50-57, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28803787

RESUMEN

Phactr1 (Phosphatase and actin regulator 1) is abundantly expressed in the central nervous system and considered to regulate various neuronal processes through the regulation of protein phosphorylation and actin cytoskeletal organization. In this study, we prepared a specific antibody against Phactr1, anti-Phactr1, and carried out biochemical and morphological analyses of Phactr1 with mouse brain tissues. Western blotting analyses revealed that Phactr1 was expressed in a tissue-dependent profile in the young adult mouse and in a developmental stage-dependent manner in the mouse brain. In primary cultured hippocampal neurons, while Phactr1 was diffusely distributed in the nucleus and cytoplasm, it was visualized in axon and dendrites with partial colocalization with synapses. Phactr1 was also detected in the synaptosomal and postsynaptic density fractions in biochemical fractionation. Immunohistochemical analyses clarified that Phactr1 was differentially expressed in cortical neurons during corticogenesis; the protein was frequently accumulated in the nucleus at the embryonic stage while it came to diffusely distribute in the cell body at the prepubertal stage. The obtained results suggest that Phactr1 takes part in neuronal functions regulated in a spatiotemporal manner.


Asunto(s)
Hipocampo/crecimiento & desarrollo , Proteínas de Microfilamentos/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , Animales , Axones/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Inmunohistoquímica/métodos , Ratones
16.
Med Mol Morphol ; 51(2): 111-117, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29282540

RESUMEN

Dusp22 (dual-specificity phosphatase 22) is considered to regulate various cellular processes through the regulation of protein dephosphorylation. In this study, we prepared a specific antibody against Dusp22, anti-Dusp22, and carried out expression analyses with mouse tissues and cultured cell lines. Western blotting analyses demonstrated a tissue-dependent expression profile of Dusp22 in the adult mouse, and strongly suggested the presence of isoforms with larger molecular masses. In fibroblast NIH3T3 cells, while both endogenous and Myc-tagged Dusp22 was diffusely distributed in the cytoplasm, Myc-Dusp22 was partially colocalized with actin cytoskeleton. From the obtained results, anti-Dusp22 was found to be a useful tool for biochemical and cell biological analyses of Dusp22.


Asunto(s)
Fosfatasas de Especificidad Dual/metabolismo , Animales , Anticuerpos , Western Blotting , Células COS , Línea Celular , Chlorocebus aethiops , Fosfatasas de Especificidad Dual/inmunología , Células HeLa , Humanos , Ratones , Peso Molecular , Células 3T3 NIH , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , Conejos
17.
Acta Neuropathol Commun ; 5(1): 92, 2017 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-29191246

RESUMEN

While Munc18-1 interacts with Syntaxin1 and controls the formation of soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) complex to regulate presynaptic vesicle fusion in developed neurons, this molecule is likely to be involved in brain development since its gene abnormalities cause early infantile epileptic encephalopathy with suppression-burst (Ohtahara syndrome), neonatal epileptic encephalopathy and other neurodevelopmental disorders. We thus analyzed physiological significance of Munc18-1 during cortical development. Munc18-1-knockdown impaired cortical neuron positioning during mouse corticogenesis. Time-lapse imaging revealed that the mispositioning was attributable to defects in radial migration in the intermediate zone and cortical plate. Notably, Syntaxin1A was critical for radial migration downstream of Munc18-1. As for the underlying mechanism, Munc18-1-knockdown in cortical neurons hampered post-Golgi vesicle trafficking and subsequent vesicle fusion at the plasma membrane in vivo and in vitro, respectively. Notably, Syntaxin1A-silencing did not affect the post-Golgi vesicle trafficking. Taken together, Munc18-1 was suggested to regulate radial migration by modulating not only vesicle fusion at the plasma membrane to distribute various proteins on the cell surface for interaction with radial fibers, but also preceding vesicle transport from Golgi to the plasma membrane. Although knockdown experiments suggested that Syntaxin1A does not participate in the vesicle trafficking, it was supposed to regulate subsequent vesicle fusion under the control of Munc18-1. These observations may shed light on the mechanism governing radial migration of cortical neurons. Disruption of Munc18-1 function may result in the abnormal corticogenesis, leading to neurodevelopmental disorders with MUNC18-1 gene abnormalities.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Munc18/genética , Proteínas Munc18/metabolismo , Mutación/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Animales , Animales Recién Nacidos , Células COS , Movimiento Celular/genética , Células Cultivadas , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Chlorocebus aethiops , Quinasa 5 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Ratones , Ratones Transgénicos , Neurogénesis/genética , Neuronas/fisiología , Proteína Quinasa C/metabolismo , Transporte de Proteínas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Sintaxina 1/metabolismo , Transfección
18.
Sci Rep ; 6: 30805, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27481563

RESUMEN

Gene abnormalities in RBFOX1, encoding an mRNA-splicing factor, have been shown to cause autism spectrum disorder and other neurodevelopmental disorders. Since pathophysiological significance of the dominant nuclear isoform in neurons, RBFOX1-isoform1 (iso1), remains to be elucidated, we performed comprehensive analyses of Rbfox1-iso1 during mouse corticogenesis. Knockdown of Rbfox1-iso1 by in utero electroporation caused abnormal neuronal positioning during corticogenesis, which was attributed to impaired migration. The defects were found to occur during radial migration and terminal translocation, perhaps due to impaired nucleokinesis. Axon extension and dendritic arborization were also suppressed in vivo in Rbfox1-iso1-deficient cortical neurons. In addition, electrophysiology experiments revealed significant defects in the membrane and synaptic properties of the deficient neurons. Aberrant morphology was further confirmed by in vitro analyses; Rbfox1-iso1-konckdown in hippocampal neurons resulted in the reduction of primary axon length, total length of dendrites, spine density and mature spine number. Taken together, this study shows that Rbfox1-iso1 plays an important role in neuronal migration and synapse network formation during corticogenesis. Defects in these critical processes may induce structural and functional defects in cortical neurons, and consequently contribute to the pathophysiology of neurodevelopmental disorders with RBFOX1 abnormalities.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Núcleo Celular/metabolismo , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Animales , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Axones/fisiología , Encéfalo/anomalías , Encéfalo/metabolismo , Movimiento Celular , Núcleo Celular/genética , Células Cultivadas , Técnicas de Inactivación de Genes , Humanos , Ratones , Neurogénesis , Plasticidad Neuronal , Neuronas/citología , Neuronas/metabolismo , Isoformas de Proteínas/metabolismo
19.
Med Mol Morphol ; 49(1): 28-33, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26242203

RESUMEN

The mammalian Class III phosphoinositide 3-kinase (PIK3C3, also known as mammalian vacuolar protein sorting 34 homologue, Vps34) is a regulator of vesicular trafficking, autophagy, and nutrient sensing. In this study, we generated a specific antibody against PIK3C3, and carried out expression and morphological analyses of PIK3C3 during mouse brain development. In Western blotting, PIK3C3 was detected throughout the developmental process with higher expression in the early embryonic stage. In immunohistochemical analyses with embryonic day 16 mouse brain, PIK3C3 was detected strongly in the axon of cortical neurons. While PIK3C3 was distributed at the soma, nucleus, axon, and dendrites in primary cultured mouse hippocampal neurons at 3 days in vitro (div), it was also found in a punctate distribution with partial colocalization with synaptic marker, synaptophysin, at 21 div. The obtained results indicate that PIK3C3 is expressed and may have a physiological role in central nervous system during corticogenesis.


Asunto(s)
Encéfalo/enzimología , Neuronas/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Axones/enzimología , Encéfalo/embriología , Células COS , Células Cultivadas , Chlorocebus aethiops , Fosfatidilinositol 3-Quinasas Clase III , Hipocampo/citología , Hipocampo/enzimología , Ratones Endogámicos ICR , Fosfatidilinositol 3-Quinasas/genética , Sinaptofisina/metabolismo
20.
Mol Autism ; 6: 56, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26500751

RESUMEN

BACKGROUND: RBFOX1 (also known as FOX1 or A2BP1) regulates alternative splicing of a variety of transcripts crucial for neuronal functions. Physiological significance of RBFOX1 during brain development is seemingly essential since abnormalities in the gene cause autism spectrum disorder (ASD) and other neurodevelopmental and neuropsychiatric disorders such as intellectual disability, epilepsy, attention deficit hyperactivity disorder, and schizophrenia. RBFOX1 was also shown to serve as a "hub" in ASD gene transcriptome network. However, the pathophysiological significance of RBFOX1 gene abnormalities remains to be clarified. METHODS: To elucidate the pathophysiological relevance of Rbfox1, we performed a battery of in vivo and in vitro analyses of the brain-specific cytoplasmic isoform, Rbfox1-iso2, during mouse corticogenesis. In vivo analyses were based on in utero electroporation, and the role of Rbfox1-iso2 in cortical neuron migration, neurogenesis, and morphology was investigated by morphological methods including confocal laser microscope-assisted time-lapse imaging. In vitro analyses were carried out to examine the morphology of primary cultured mouse hippocampal neurons. RESULTS: Silencing of Rbfox1-iso2 in utero caused defects in the radial migration and terminal translocation of cortical neurons during corticogenesis. Time-lapse imaging revealed that radial migration was apparently impaired by dysregulated nucleokinesis. Rbfox1-iso2 also regulated neuronal network formation in vivo since axon extension to the opposite hemisphere and dendritic arborization were hampered by the knockdown. In in vitro analyses, spine density and mature spine number were reduced in Rbfox1-iso2-deficient hippocampal neurons. CONCLUSIONS: Impaired Rbfox1-iso2 function was found to cause abnormal corticogenesis during brain development. The abnormal process may underlie the basic pathophysiology of ASD and other neurodevelopmental disorders and may contribute to the emergence of the clinical symptoms of the patients with RBFOX1 gene abnormalities.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA