Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell ; 149(4): 899-911, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22579290

RESUMO

Fragile X syndrome (FXS), the leading monogenic cause of intellectual disability and autism, results from loss of function of the RNA-binding protein FMRP. Here, we show that FMRP regulates translation of neuronal nitric oxide synthase 1 (NOS1) in the developing human neocortex. Whereas NOS1 mRNA is widely expressed, NOS1 protein is transiently coexpressed with FMRP during early synaptogenesis in layer- and region-specific pyramidal neurons. These include midfetal layer 5 subcortically projecting neurons arranged into alternating columns in the prospective Broca's area and orofacial motor cortex. Human NOS1 translation is activated by FMRP via interactions with coding region binding motifs absent from mouse Nos1 mRNA, which is expressed in mouse pyramidal neurons, but not efficiently translated. Correspondingly, neocortical NOS1 protein levels are severely reduced in developing human FXS cases, but not FMRP-deficient mice. Thus, alterations in FMRP posttranscriptional regulation of NOS1 in developing neocortical circuits may contribute to cognitive dysfunction in FXS.


Assuntos
Córtex Cerebral/embriologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/embriologia , Óxido Nítrico Sintase Tipo I/metabolismo , Animais , Córtex Cerebral/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/fisiopatologia , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Neurogênese , Células Piramidais/metabolismo , Processamento Pós-Transcricional do RNA , Especificidade da Espécie
2.
Exp Eye Res ; 245: 109988, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38964496

RESUMO

Autism spectrum disorder (ASD) is a group of neurodevelopment disorders characterized by deficits in social interaction and communication, and repetitive or stereotyped behavior. Autistic children are more likely to have vision problems, and ASD is unusually common among blind people. However, the mechanisms behind the vision disorders in autism are unclear. Stabilizing WNT-targeted scaffold protein Axin2 by XAV939 during embryonic development causes overproduction of cortical neurons and leads to autistic-like behaviors in mice. In this study, we investigated the relationship between vision abnormality and autism using an XAV939-induced mouse model of autism. We found that the mice receiving XAV939 had decreased amplitude of bright light-adaptive ERG. The amplitudes and latency of flash visual evoked potential recorded from XAV939-treated mice were lower and longer, respectively than in the control mice, suggesting that XAV939 inhibits visual signal processing and conductance. Anatomically, the diameters of RGC axons were reduced when Axin2 was stabilized during the development, and the optic fibers had defective myelin sheaths and reduced oligodendrocytes. The results suggest that the WNT signaling pathway is crucial for optic nerve development. This study provides experimental evidence that conditions interfering with brain development may also lead to visual problems, which in turn might exaggerate the autistic features in humans.


Assuntos
Proteína Axina , Modelos Animais de Doenças , Potenciais Evocados Visuais , Nervo Óptico , Animais , Proteína Axina/metabolismo , Camundongos , Potenciais Evocados Visuais/fisiologia , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Eletrorretinografia , Camundongos Endogâmicos C57BL , Axônios/patologia , Células Ganglionares da Retina/patologia , Células Ganglionares da Retina/metabolismo , Masculino , Via de Sinalização Wnt/fisiologia , Transtorno do Espectro Autista/fisiopatologia , Transtorno do Espectro Autista/metabolismo , Transtorno Autístico/fisiopatologia , Transtorno Autístico/metabolismo
3.
Biochem Biophys Res Commun ; 647: 9-15, 2023 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-36708662

RESUMO

Neuronal migration and morphogenesis are fundamental processes for cortical development. Their defects may cause abnormities in neural circuit formation and even neuropsychiatric disorders. Many proteins, especially layer-specific transcription factors and adhesion molecules, have been reported to regulate the processes. However, the involvement of non-coding RNAs in cortical development has not been extensively studied. Here, we identified microRNA-218 (miR-218) as a layer V-specific microRNA in mouse brains. Expression of miR-218 was elevated in patients with autism spectrum disorder (ASD) and schizophrenia. We found in this study that miR-218 overexpression in developing mouse cortex led to severe defects in radial migration, morphogenesis, and spatial distribution of the cortical neurons. Moreover, we identified Satb2, an upper-layer marker, as a molecular target repressed by miR-218. These results suggest an underlying mechanism of miR-218 involvement in neuropsychiatric disorders, and the interactions of layer-specific non-coding RNAs and proteins in regulating cortical development.


Assuntos
Transtorno do Espectro Autista , Proteínas de Ligação à Região de Interação com a Matriz , MicroRNAs , Neocórtex , Fatores de Transcrição , Animais , Camundongos , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Movimento Celular/genética , Proteínas de Ligação à Região de Interação com a Matriz/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Neocórtex/metabolismo , Neurogênese/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Humanos
4.
Cereb Cortex ; 30(7): 3960-3976, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32008040

RESUMO

De novo microdeletion of chromosome 2p15-16.1 presents clinically recognizable phenotypes that include mental retardation, autism, and microcephaly. Chromosomal maintenance 1 (CRM1) is a gene commonly missing in patients with 2p15-16.1 microdeletion and one of two genes found in the smallest deletion case. In this study, we investigate the role and mechanism of Crm1 in the developing mouse brain by inhibiting the protein or knocking down the gene in vivo. Inhibition of Crm1 reduces the proliferation and increases p53-dependent apoptosis of the cortical neural progenitors, thereby impeding the growth of embryonic cerebral cortex. Live imaging of mitosis in ex vivo embryonic brain slices reveals that inhibition of CRM1 arrests the cortical progenitors at metaphase. The arrested cells eventually slip into a pseudo-G1 phase without chromosome segregation. The mitotic slippage cells are marked by persistent expression of the spindle assembly checkpoint (SAC), repressing of which rescues the cells from apoptosis. Our study reveals that activating the SAC and inducing the mitotic slippage may lead to apoptosis of the cortical neural progenitors. The resulting cell death may well contribute to microcephaly associated with microdeletion of chromosome 2p15-16.1 involving CRM1.


Assuntos
Apoptose/genética , Transtorno Autístico/genética , Proliferação de Células/genética , Córtex Cerebral/metabolismo , Carioferinas/genética , Mitose/genética , Células-Tronco Neurais/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Animais , Córtex Cerebral/citologia , Cromossomos Humanos Par 2/genética , Deleção de Genes , Técnicas de Silenciamento de Genes , Humanos , Carioferinas/antagonistas & inibidores , Pontos de Checagem da Fase M do Ciclo Celular/genética , Camundongos , Células-Tronco Neurais/citologia , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Proteína Exportina 1
5.
Cereb Cortex ; 29(7): 3193-3208, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-30124790

RESUMO

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that has a strong genetic component. Disruptions of FOXP1, a transcription factor expressed in the developing cerebral cortex, were associated with ASD. FOXP1(R525X) is a de novo heterozygous mutation found in patients with autism and severe mental retardation. To explore the neuronal basis of FOXP1(R525X) in ASD, we created Foxp1(R521X), a mouse homolog of the human variant. Ectopic expression of Foxp1(R521X) led to cytoplasmic aggregates and activated macroautophagy in neuroblastoma N2a cells and the developing neuronal cells. Cortical neurons expressing Foxp1(R521X) exhibited delayed migration and altered dendritic morphology. As a control, mutant Y435X that was expressed diffusively in the cytoplasm did not induce autophagy and migration delay in the cortex. The embryonic cortical cells had a minimal activity of nonsense-mediated mRNA decay (NMD) as assayed by a splicing-dependent NMD reporter. We hypothesize that the developing neuronal cells use autophagy but not NMD as a safeguard mechanism against nonsense mutant aggregates, resulting in impairment of the cortical development. This study suggests a novel mechanism other than heterozygous loss of FOXP1 for the development of ASD and may advance our understanding of the complex relationships between gene mutation and the related psychiatric disorders.


Assuntos
Transtorno do Espectro Autista/genética , Autofagia/genética , Córtex Cerebral/embriologia , Fatores de Transcrição Forkhead/genética , Neurogênese/genética , Proteínas Repressoras/genética , Animais , Movimento Celular/genética , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos ICR , Mutação , Neurônios/metabolismo
6.
J Biol Chem ; 293(6): 2137-2148, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29273633

RESUMO

The progenitor cells in the cerebral cortex coordinate proliferation and mitotic exit to generate the correct number of neurons and glial cells during development. However, mechanisms for regulating the mitotic cycle of cortical progenitors are not fully understood. Otx1 is one of the homeobox-containing transcription factors frequently implicated in the development of the central nervous system. Mice bearing a targeted deletion of Otx1 exhibit brain hypoplasia and a decrease in the number of cortical neurons. We hypothesized that Otx1 might be crucial to the proliferation and differentiation of cortical progenitors. Otx1 knockdown by in utero electroporation in the mouse brain reduced the proportion of the G1 phase while increasing the S and M phases of progenitor cells. The knockdown diminished Tbr1+ neurons but increased GFAP+ astrocytes in the early postnatal cortex as revealed by lineage tracing study. Tbr2+ basal progenitors lacking Otx1 were held at the transit-amplifying stage. In contrast, overexpression of wildtype Otx1 but not an astrocytoma-related mutant Y320C inhibited proliferation of the progenitor cells in embryonic cortex. This study demonstrates that Otx1 is one of the key elements regulating cortical neurogenesis, and a loss-of-function in Otx1 may contribute to the overproduction of astrocytes in vivo.


Assuntos
Ciclo Celular , Córtex Cerebral/citologia , Células-Tronco Neurais/citologia , Fatores de Transcrição Otx/fisiologia , Animais , Astrócitos/citologia , Contagem de Células , Diferenciação Celular , Proliferação de Células , Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Camundongos , Neurogênese , Neurônios/citologia
7.
J Biol Chem ; 291(14): 7661-8, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26861874

RESUMO

Retinal ganglion cells (RGCs) are projection neurons in the neural retina that relay visual information from the environment to the central nervous system. The early expression of MATH5 endows the post-mitotic precursors with RGC competence and leads to the activation ofBrn3bthat marks committed RGCs. Nevertheless, this fate commitment process and, specifically, regulation ofBrn3bremain elusive. To explore the molecular mechanisms underlying RGC generation in the mouse retina, we analyzed the expression and function of Fez family zinc finger 2 (FEZF2), a transcription factor critical for the development of projection neurons in the cerebral cortex.Fezf2mRNA and protein were transiently expressed at embryonic day 16.5 in the inner neuroblast layer and the prospective ganglion cell layer of the retina, respectively. Knockout ofFezf2in the developing retina reduced BRN3B+ cells and increased apoptotic cell markers.Fezf2knockdown by retinalin uteroelectroporation diminished BRN3B but not the coexpressed ISLET1 and BRN3A, indicating that the BRN3B decrease was the cause, not the result, of the overall reduction of BRN3B+ RGCs in theFezf2knockout retina. Moreover, the mRNA and promoter activity ofBrn3bwere increasedin vitroby FEZF2, which bound to a 5' regulatory fragment in theBrn3bgenomic locus. These results indicate that transient expression ofFezf2in the retina modulates the transcription ofBrn3band the survival of RGCs. This study improves our understanding of the transcriptional cascade required for the specification of RGCs and provides novel insights into the molecular basis of retinal development.


Assuntos
Apoptose/fisiologia , Proteínas de Ligação a DNA/biossíntese , Regulação da Expressão Gênica/fisiologia , Proteínas de Homeodomínio/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Células Ganglionares da Retina/metabolismo , Fator de Transcrição Brn-3B/biossíntese , Transcrição Gênica/fisiologia , Animais , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Células Ganglionares da Retina/citologia , Fator de Transcrição Brn-3B/genética
8.
Neurochem Res ; 42(4): 1151-1164, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28150227

RESUMO

Retinal Müller glial cells in mammals acquire stem and progenitor cell properties after neurotoxic treatment. However, the molecular mechanisms underlying proliferation and dedifferentiation of adult Müller cells in the mammalian retina were unclear. In this study, treatments with N-methyl-D-aspartate (NMDA) plus epidermal growth factor (EGF) led to the proliferation of Müller cells and expression of stem cell markers including Nanog and Nestin in the retina. The increased mRNA for Nanog and Nestin were coincident with reduced methylation of a Nanog promoter and a Nestin enhancer specific in the neural stem cells, respectively. We found that Apolipoprotein B mRNA editing catalytic subunit 1 (Apobec1) was upregulated early in the retina treated with NMDA and EGF. Moreover, overexpression of Apobec1 in primary Müller cells increased expression of Nestin and reduced methylation of the Nestin enhancer. The data suggest that neurotoxicity-induced Apobec1 may promote expression of Nestin and help cell cycle reentry of retinal Müller cells via DNA demethylation. This study provides novel insights into the molecular mechanisms underlying dedifferentiation and proliferation of Müller cells in the mammalian retina.


Assuntos
Desaminase APOBEC-1/biossíntese , Desdiferenciação Celular/fisiologia , Células Ependimogliais/metabolismo , Fator de Crescimento Epidérmico/toxicidade , N-Metilaspartato/toxicidade , Animais , Desdiferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Ependimogliais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL
9.
Neurosci Bull ; 39(7): 1131-1145, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36646976

RESUMO

Heterozygous loss-of-function variants of FOXP4 are associated with neurodevelopmental disorders (NDDs) that exhibit delayed speech development, intellectual disability, and congenital abnormalities. The etiology of NDDs is unclear. Here we found that FOXP4 and N-cadherin are expressed in the nuclei and apical end-feet of radial glial cells (RGCs), respectively, in the mouse neocortex during early gestation. Knockdown or dominant-negative inhibition of Foxp4 abolishes the apical condensation of N-cadherin in RGCs and the integrity of neuroepithelium in the ventricular zone (VZ). Inhibition of Foxp4 leads to impeded radial migration of cortical neurons and ectopic neurogenesis from the proliferating VZ. The ectopic differentiation and deficient migration disappear when N-cadherin is over-expressed in RGCs. The data indicate that Foxp4 is essential for N-cadherin-based adherens junctions, the loss of which leads to periventricular heterotopias. We hypothesize that FOXP4 variant-associated NDDs may be caused by disruption of the adherens junctions and malformation of the cerebral cortex.


Assuntos
Caderinas , Células Ependimogliais , Camundongos , Animais , Células Ependimogliais/fisiologia , Neurônios/metabolismo , Córtex Cerebral/metabolismo , Diferenciação Celular , Movimento Celular
10.
Mol Autism ; 14(1): 34, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37691105

RESUMO

BACKGROUND: Many children and young people with autism spectrum disorder (ASD) display touch defensiveness or avoidance (hypersensitivity), or engage in sensory seeking by touching people or objects (hyposensitivity). Abnormal sensory responses have also been noticed in mice lacking ASD-associated genes. Tactile sensory information is normally processed by the somatosensory system that travels along the thalamus to the primary somatosensory cortex. The neurobiology behind tactile sensory abnormalities, however, is not fully understood. METHODS: We employed cortex-specific Foxp1 knockout (Foxp1-cKO) mice as a model of autism in this study. Tactile sensory deficits were measured by the adhesive removal test. The mice's behavior and neural activity were further evaluated by the whisker nuisance test and c-Fos immunofluorescence, respectively. We also studied the dendritic spines and barrel formation in the primary somatosensory cortex by Golgi staining and immunofluorescence. RESULTS: Foxp1-cKO mice had a deferred response to the tactile environment. However, the mice exhibited avoidance behavior and hyper-reaction following repeated whisker stimulation, similar to a fight-or-flight response. In contrast to the wild-type, c-Fos was activated in the basolateral amygdala but not in layer IV of the primary somatosensory cortex of the cKO mice. Moreover, Foxp1 deficiency in cortical neurons altered the dendrite development, reduced the number of dendritic spines, and disrupted barrel formation in the somatosensory cortex, suggesting impaired somatosensory processing may underlie the aberrant tactile responses. LIMITATIONS: It is still unclear how the defective thalamocortical connection gives rise to the hyper-reactive response. Future experiments with electrophysiological recording are needed to analyze the role of thalamo-cortical-amygdala circuits in the disinhibiting amygdala and enhanced fearful responses in the mouse model of autism. CONCLUSIONS: Foxp1-cKO mice have tactile sensory deficits while exhibit hyper-reactivity, which may represent fearful and emotional responses controlled by the amygdala. This study presents anatomical evidence for reduced thalamocortical connectivity in a genetic mouse model of ASD and demonstrates that the cerebral cortex can be the origin of atypical sensory behaviors.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Animais , Camundongos , Transtorno Autístico/genética , Tato , Córtex Cerebral , Modelos Animais de Doenças , Camundongos Knockout , Proteínas Repressoras , Fatores de Transcrição Forkhead/genética
11.
Proc Natl Acad Sci U S A ; 106(21): 8671-6, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19458043

RESUMO

Microtubule-stabilizing (MTS) agents, such as taxanes, are important chemotherapeutics with a poorly understood mechanism of action. We identified a set of genes repressed in multiple cell lines in response to MTS agents and observed that these genes are overexpressed in tumors exhibiting chromosomal instability (CIN). Silencing 22/50 of these genes, many of which are involved in DNA repair, caused cancer cell death, suggesting that these genes are involved in the survival of aneuploid cells. Overexpression of these "CIN-survival" genes is associated with poor outcome in estrogen receptor-positive breast cancer and occurs frequently in basal-like and Her2-positive cases. In diploid cells, but not in chromosomally unstable cells, paclitaxel causes repression of CIN-survival genes, followed by cell death. In the OV01 ovarian cancer clinical trial, a high level of CIN was associated with taxane resistance but carboplatin sensitivity, indicating that CIN may determine MTS response in vivo. Thus, pretherapeutic assessment of CIN may optimize treatment stratification and clinical trial design using these agents.


Assuntos
Hidrocarbonetos Aromáticos com Pontes/farmacologia , Instabilidade Cromossômica/efeitos dos fármacos , Instabilidade Cromossômica/genética , Taxoides/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Microtúbulos/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Paclitaxel/toxicidade , Reação em Cadeia da Polimerase , Prognóstico
12.
J Integr Neurosci ; 11(2): 193-200, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22744825

RESUMO

A reduction in blood flow to the brain causes stroke and damage to neuronal networks. Cerebral ischemia is frequently associated with loss of visual functions. Because retinal and small cerebral vessels are vulnerable to similar risk factors, the loss of vision could result from concurrent retinal ischemia, and it is not clear if visual functions may be inhibited by cerebral ischemia directly. In this study, the distal middle cerebral artery in the right hemisphere of mice was occluded to produce unilateral cerebral ischemia and subsequent infarction. The layer V neurons expressing YFP in transgenic yellow fluorescent protein in transgenic B6.Cg-Tg(Thy1-YFPH)2Jrs/J mice disappeared in the motor and somatosensory cortex, but not in the visual area. The latencies of flash visual evoked potential recorded from two hemispheres were imbalanced, but did not differ markedly from the latencies recorded in controls. However, the optomotor responses of the ipsilateral eye were significantly reduced by 48 h after occlusion. Our results suggest that focused cerebral ischemia may inhibit ipsilateral eye movement in the absence of damage to the visual cortex. This study may provide a platform for further investigation of the relationship between cortical ischemia and visual function.


Assuntos
Potenciais Evocados Visuais/fisiologia , Olho/fisiopatologia , Lateralidade Funcional/fisiologia , Infarto da Artéria Cerebral Média/fisiopatologia , Campos Visuais/fisiologia , Vias Visuais/fisiologia , Animais , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/patologia , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/patologia , Optometria , Estimulação Luminosa , Córtex Visual/metabolismo
13.
Eye Vis (Lond) ; 9(1): 10, 2022 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-35248156

RESUMO

BACKGROUND: The exudative form of age-related macular degeneration (AMD) is characterized by abnormal blood vessel growth, which is stimulated by vascular endothelial growth factor (VEGF) released from retinal pigment epithelium (RPE). The angiogenic behaviors of vascular endothelial cells in vitro depend on forkhead box protein P1 (Foxp1), a transcription repressor widely expressed in human and murine tissues during development. In this study, we aimed to determine whether loss of Foxp1 affects laser-induced choroidal neovascularization (CNV) in mouse. METHODS: Eye-selective deletion of Foxp1 was obtained by crossing Foxp1flox/flox with Six3-Cre mice. Laser photocoagulation was delivered to six- to eight-week-old mice to induce CNV. The expression of Foxp1 and Cre was determined by immunofluorescence in cryostat sections of the eyes. Fundus fluorescein angiography (FFA), optical coherence tomography (OCT), and B4 isolectin staining were applied to analyze the leakage, bulge height, and area of CNV lesions, respectively. RPE-choroid tissues were isolated for the determination of VEGF and pigment epithelium derived factor (PEDF) by Western blotting. RESULTS: Foxp1 was expressed in retinal ganglion cells, RPE, and the choroidal endothelial cells. Laser photocoagulation increased the number of Foxp1+-endothelial cells and induced CNV. Six3-Cre reduced Foxp1 expression in RPE but not the endothelium, leading to a lower level of VEGF in the RPE-choroid. Foxp1 knockout inhibited pathological angiogenesis and vascular leakage of the laser-induced CNV lesions. CONCLUSIONS: Foxp1 regulates the expression of VEGF in the RPE, and inhibition of Foxp1 could potentially be a novel strategy for the prevention and therapy of neovascularization related to AMD.

14.
Cereb Cortex ; 19(9): 2196-207, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19234067

RESUMO

Cortical excitatory glutamatergic projection neurons and inhibitory GABAergic interneurons follow substantially different developmental programs. In rodents, projection neurons originate from progenitors within the dorsal forebrain, whereas interneurons arise from progenitors in the ventral forebrain. In contrast, it has been proposed that in humans, the majority of cortical interneurons arise from progenitors within the dorsal forebrain, suggesting that their origin and migration is complex and evolutionarily divergent. However, whether molecularly defined human cortical interneuron subtypes originate from distinct progenitors, including those in the ventral forebrain, remains unknown. Furthermore, abnormalities in cortical interneurons have been linked to human disorders, yet no distinct cell population selective loss has been reported. Here we show that cortical interneurons expressing nitric oxide synthase 1, neuropeptide Y, and somatostatin, are either absent or substantially reduced in fetal and infant cases of human holoprosencephaly (HPE) with severe ventral forebrain hypoplasia. Notably, another interneuron subtype normally abundant from the early fetal period, marked by calretinin expression, and different subtypes of projection neuron were present in the cortex of control and HPE brains. These findings have important implications for the understanding of neuronal pathogenesis underlying the clinical manifestations associated with HPE and the developmental origins of human cortical interneuron diversity.


Assuntos
Córtex Cerebral/anormalidades , Córtex Cerebral/metabolismo , Corpo Estriado/anormalidades , Corpo Estriado/metabolismo , Holoprosencefalia/metabolismo , Interneurônios/metabolismo , Neurotransmissores/metabolismo , Corpo Estriado/patologia , Humanos , Recém-Nascido , Interneurônios/patologia
15.
Neuroscience ; 449: 228-240, 2020 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-33002559

RESUMO

Chylomicron Retention Disease (CMRD) is a rare inherited lipid malabsorption syndrome that exhibits a recessive hypocholesterolemia in infants. CMRD has been associated with genetic mutations of SAR1B-a member of the Arf GTPase family involved in the secretory pathway from the endoplasmic reticulum to the Golgi. CMRD patients suffer from multiple neurological deficits, the etiologies of which remain unclear. In this study, we found that Sar1b protein is expressed in developing mouse neocortex. The knockdown of Sar1b does not affect the proliferation and mitotic exit of the neural progenitors but inhibits the radial migration of the newborn cortical neurons. At postnatal day 3, the neurons stalled in the white matter fail to develop axons across the midline of the corpus callosum, resulting in the loss of the neurons later on. hSAR1B(D137N), a CMRD-associated mutant of SAR1B, also impairs the positioning of the cortical neurons in the mouse brain, suggesting a dominant-negative effect by the human heterozygous mutant. The results indicate that SAR1B is crucial to radial migration and axon morphogenesis of the cortical neurons. Our study reveals a cell-autonomous action of Sar1b, which is unrelated to lipid absorption from the gut, on the development of the cerebral cortex.


Assuntos
Síndromes de Malabsorção , Proteínas Monoméricas de Ligação ao GTP , Neurônios/fisiologia , Animais , Movimento Celular/genética , Córtex Cerebral/citologia , Humanos , Hipobetalipoproteinemias , Camundongos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Morfogênese
16.
Cancer Res ; 62(7): 1935-8, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11929805

RESUMO

Although microtubule interacting agents inhibit spindle dynamics, thereby leading to a block in mitosis, we report that low concentrations of these drugs result in differential mitotic effects. Microtubule-stabilizing agents including Taxol, epothilone B, and discodermolide produce aneuploid populations of A549 cells in the absence of a mitotic block. Such aneuploid populations are diminished in an epothilone B-resistant cell line. In contrast, microtubule-destabilizing agents like colchicine, nocodazole, and vinblastine are unable to initiate aneuploidy. The aneuploid cells result from aberrant mitosis as multipolar spindles are induced by the stabilizing drugs, but not by destabilizing agents. The results suggest that the mechanism underlying aberrant mitosis may not be the same as that responsible for mitotic block, and that the former determines the sensitivity of cells to Taxol-like drugs.


Assuntos
Alcanos , Antineoplásicos/farmacologia , Carbamatos , Carcinoma Pulmonar de Células não Pequenas/patologia , Epotilonas , Neoplasias Pulmonares/patologia , Microtúbulos/efeitos dos fármacos , Mitose/efeitos dos fármacos , Aneuploidia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Humanos , Lactonas/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Macrolídeos/farmacologia , Paclitaxel/farmacologia , Pironas , Fuso Acromático/efeitos dos fármacos , Células Tumorais Cultivadas
17.
Cancer Res ; 63(22): 7891-9, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14633718

RESUMO

To explore the molecular mechanisms underlying the actions of Taxol and the functionally related molecule epothilone B (EpoB), we have analyzed the gene expression profiles in A549 cells in response to increasing concentrations of these microtubule-stabilizing drugs. An almost identical expression pattern was observed in cells treated with either Taxol or EpoB. Low concentrations of the drugs induced aberrant mitosis including asymmetric and multipolar cell divisions. At drug concentrations that triggered G(2)-M arrest, cells escaped from a prolonged mitotic arrest without cell division, resulting in tetraploid G(1) cells. This mitotic slippage is correlated with diminished expression of cdc2 kinase, topoisomerase IIalpha, BUB3, and BUB2-like protein 1, as well as with an increased expression of 14-3-3-sigma. Poly(ADP-ribose) polymerase cleavage, an early indicator of apoptosis, occurred in cells undergoing mitotic slippage and in aneuploid cells resulting from aberrant mitosis. In contrast, cells arrested in mitosis demonstrated no signal for apoptosis but had an increased expression of survivin, an inhibitor of apoptosis. Induction of aneuploid or tetraploid G(1) cells was accompanied by increased expression of CD95, p21, and BTG2 that may contribute to cell death because their expression was diminished in an EpoB-resistant cell line. In contrast, expression of GADD45 and PTGF-beta could promote cell survival. We conclude that abnormal mitotic exit is required for apoptotic cell death induced by microtubule-stabilizing drugs.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Epotilonas/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Paclitaxel/farmacologia , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Microtúbulos/efeitos dos fármacos , Mitose/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Poliploidia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
18.
PLoS One ; 10(5): e0127671, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26010426

RESUMO

FOXP1 is a member of FOXP subfamily transcription factors. Mutations in FOXP1 gene have been found in various development-related cognitive disorders. However, little is known about the etiology of these symptoms, and specifically the function of FOXP1 in neuronal development. Here, we report that suppression of Foxp1 expression in mouse cerebral cortex led to a neuronal migration defect, which was rescued by overexpression of Foxp1. Mice with Foxp1 knockdown exhibited ectopic neurons in deep layers of the cortex postnatally. The neuronal differentiation of Foxp1-downregulated cells was normal. However, morphological analysis showed that the neurons with Foxp1 deficiency had an inhibited axonal growth in vitro and a weakened transition from multipolar to bipolar in vivo. Moreover, we found that the expression of Foxp1 modulated the dendritic maturation of neurons at a late postnatal date. Our results demonstrate critical roles of Foxp1 in the radial migration and morphogenesis of cortical neurons during development. This study may shed light on the complex relationship between neuronal development and the related cognitive disorders.


Assuntos
Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Córtex Cerebral/embriologia , Dendritos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Organogênese/fisiologia , Proteínas Repressoras/metabolismo , Animais , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Proteínas Repressoras/genética
19.
Neurosci Res ; 77(3): 137-42, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23999326

RESUMO

Hereditary spastic paraplegias (HSPs) are human genetic disorders characterized by lower extremity spasticity and weakness. Mutations in atlastin-1 (ATL1) have been identified in patients with HSP SPG3A. However, the function of ATL1 in the mammalian brain remains unclear. Here, we found that expression of ATL1 mRNA was restricted in the deep layer of mouse cerebral cortex during the early development. We examined ATL1 functions by delivering its plasmids to the upper layer cortical neurons using in utero electroporation. The effects of ectopic expression in the pyramidal neurons were determined both in culture and in situ at postnatal stages of neocortical development. In cultured cortical neurons, overexpressing ATL1 increased dendrite growth and arborization, whereas HSP-associated mutant R217Q, which is devoid of GTPase activity, had no such effects. Consistent with this, in vivo expression of wild type ATL1, but not of the mutant R217Q, increased dendritic growth of the cortical neurons. This suggests that the role of ATL1 on dendritic morphogenesis depends on its GTPase activity. The expression of ATL1 and R217Q did not affect the migration of cortical neurons. These results indicate that ATL1 regulates dendritic morphogenesis, which may provide new insights into the neuropathogenic mechanism of hereditary spastic paraplegia SPG3A.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Dendritos/metabolismo , Proteínas de Membrana/metabolismo , Morfogênese , Neuritos/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Dendritos/ultraestrutura , Camundongos , Camundongos Endogâmicos ICR , Neuritos/ultraestrutura
20.
Gene Expr Patterns ; 13(1-2): 51-6, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23195472

RESUMO

The mammalian cerebral cortex plays a central role in higher cognitive functions and in the complex task of motor control. Maternally expressed gene 3 (Meg3) appears to play a role in cortical development and neurodegeneration, but the expression and regulation of Meg3 in the cortex is not clear. In this study, we examined the expression of transcript variants of Meg3 in the developing mouse cerebral cortex. By in situ hybridization, we found that a novel transcript variant of Meg3 with 8 small exons was expressed in the developing cortex, whereas the long isoforms of Meg3 (~11 kb) were enriched in corticospinal neurons (CSNs) in layer V of the cortex. No transcript variants of Meg3 were found in the neural progenitors at E12.5, when the intergenic differential methylation region (IG-DMR) near Meg3 was highly methylated. IG-DMR became demethylated at E15.5 and remained hypomethylated in early CSNs isolated from Fezf2-EGFP transgenic mice. The expression of Meg3 transcript variant 1 was inversely correlated with the IG-DMR methylation level during development. Moreover, expression of paternally expressed gene Peg11 was limited to the upper layers, consistent with the idea that the maternally expressed gene may be preferentially transcribed in the lower layers of the cortex. The spatiotemporal expression pattern of Meg3 suggests that it may participate in the early development of CSNs and contribute to cortical malfunctions related to aberrant imprinting in Meg3.


Assuntos
Metilação de DNA , DNA Intergênico/metabolismo , Neurônios/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica , Camundongos , Tratos Piramidais/citologia , Tratos Piramidais/embriologia , Tratos Piramidais/metabolismo , RNA Longo não Codificante/genética , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA