Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cereb Cortex ; 32(7): 1494-1507, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-34467373

RESUMEN

Homozygous mutations in the gene encoding the scavenger mRNA-decapping enzyme, DcpS, have been shown to underlie developmental delay and intellectual disability. Intellectual disability is associated with both abnormal neocortical development and mRNA metabolism. However, the role of DcpS and its scavenger decapping activity in neuronal development is unknown. Here, we show that human neurons derived from patients with a DcpS mutation have compromised differentiation and neurite outgrowth. Moreover, in the developing mouse neocortex, DcpS is required for the radial migration, polarity, neurite outgrowth, and identity of developing glutamatergic neurons. Collectively, these findings demonstrate that the scavenger mRNA decapping activity contributes to multiple pivotal roles in neural development and further corroborate that mRNA metabolism and neocortical pathologies are associated with intellectual disability.


Asunto(s)
Endorribonucleasas , Neurogénesis , Animales , Humanos , Ratones , Proyección Neuronal , ARN Mensajero
2.
Mol Cell Neurosci ; 92: 149-163, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30144504

RESUMEN

Tuberous Sclerosis Complex (TSC) is a disease caused by autosomal dominant mutations in the TSC1 or TSC2 genes, and is characterized by tumor susceptibility, brain lesions, seizures and behavioral impairments. The TSC1 and TSC2 genes encode proteins forming a complex (TSC), which is a major regulator and suppressor of mammalian target of rapamycin complex 1 (mTORC1), a signaling complex that promotes cell growth and proliferation. TSC1/2 loss of heterozygosity (LOH) and the subsequent complete loss of TSC regulatory activity in null cells causes mTORC1 dysregulation and TSC-associated brain lesions or other tissue tumors. However, it is not clear whether TSC1/2 heterozygous brain cells are abnormal and contribute to TSC neuropathology. To investigate this issue, we generated induced pluripotent stem cells (iPSCs) from TSC patients and unaffected controls, and utilized these to obtain neural progenitor cells (NPCs) and differentiated neurons in vitro. These patient-derived TSC2 heterozygous NPCs were delayed in their ability to differentiate into neurons. Patient-derived progenitor cells also exhibited a modest activation of mTORC1 signaling downstream of TSC, and a marked attenuation of upstream PI3K/AKT signaling. We further show that pharmacologic PI3K or AKT inhibition, but not mTORC1 inhibition, causes a neuronal differentiation delay, mimicking the patient phenotype. Together these data suggest that heterozygous TSC2 mutations disrupt neuronal development, potentially contributing to the disease neuropathology, and that this defect may result from dysregulated PI3K/AKT signaling in neural progenitor cells.


Asunto(s)
Células-Madre Neurales/metabolismo , Neurogénesis , Transducción de Señal , Esclerosis Tuberosa/metabolismo , Adolescente , Adulto , Línea Celular , Células Cultivadas , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , Células-Madre Neurales/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esclerosis Tuberosa/patología
3.
J Biol Chem ; 289(29): 20307-17, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24876378

RESUMEN

Reelin is an extracellular protein that controls many aspects of pre- and postnatal brain development and function. The molecular mechanisms that mediate postnatal activities of Reelin are not well understood. Here, we first set out to express and purify the full length Reelin protein and a biologically active central fragment. Second, we investigated in detail the signal transduction mechanisms elicited by these purified Reelin proteins in cortical neurons. Unexpectedly, we discovered that the full-length Reelin moiety, but not the central fragment, is capable of activating Erk1/2 signaling, leading to increased p90RSK phosphorylation and the induction of immediate-early gene expression. Remarkably, Erk1/2 activation is not mediated by the canonical signal transduction pathway, involving ApoER2/VLDLR and Dab1, that mediates other functions of Reelin in early brain development. The activation of Erk1/2 signaling likely contributes to the modulation of neuronal maturation and synaptic plasticity by Reelin in the postnatal and adult brain.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Corteza Cerebral/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/química , Moléculas de Adhesión Celular Neuronal/genética , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Expresión Génica , Genes Inmediatos-Precoces , Heterocigoto , Proteínas Relacionadas con Receptor de LDL/metabolismo , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Ratones Mutantes Neurológicos , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de LDL/metabolismo , Proteína Reelina , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/genética
4.
Epilepsia ; 56(4): 636-46, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25752454

RESUMEN

OBJECTIVE: Hyperactivation of the mechanistic target of rapamycin (mTOR; also known as mammalian target of rapamycin) pathway has been demonstrated in human cortical dysplasia (CD) as well as in animal models of epilepsy. Although inhibition of mTOR signaling early in epileptogenesis suppressed epileptiform activity in the neuron subset-specific Pten knockout (NS-Pten KO) mouse model of CD, the effects of mTOR inhibition after epilepsy is fully established were not previously examined in this model. Here, we investigated whether mTOR inhibition suppresses epileptiform activity and other neuropathological correlates in adult NS-Pten KO mice with severe and well-established epilepsy. METHODS: The progression of epileptiform activity, mTOR pathway dysregulation, and associated neuropathology with age in NS-Pten KO mice were evaluated using video-electroencephalography (EEG) recordings, Western blotting, and immunohistochemistry. A cohort of NS-Pten KO mice was treated with the mTOR inhibitor rapamycin (10 mg/kg i.p., 5 days/week) starting at postnatal week 9 and video-EEG monitored for epileptiform activity. Western blotting and immunohistochemistry were performed to evaluate the effects of rapamycin on the associated pathology. RESULTS: Epileptiform activity worsened with age in NS-Pten KO mice, with parallel increases in the extent of hippocampal mTOR complex 1 and 2 (mTORC1 and mTORC2, respectively) dysregulation and progressive astrogliosis and microgliosis. Rapamycin treatment suppressed epileptiform activity, improved baseline EEG activity, and increased survival in severely epileptic NS-Pten KO mice. At the molecular level, rapamycin treatment was associated with a reduction in both mTORC1 and mTORC2 signaling and decreased astrogliosis and microgliosis. SIGNIFICANCE: These findings reveal a wide temporal window for successful therapeutic intervention with rapamycin in the NS-Pten KO mouse model, and they support mTOR inhibition as a candidate therapy for established, late-stage epilepsy associated with CD and genetic dysregulation of the mTOR pathway.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia/metabolismo , Malformaciones del Desarrollo Cortical/metabolismo , Fosfohidrolasa PTEN/deficiencia , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Animales , Epilepsia/tratamiento farmacológico , Femenino , Masculino , Malformaciones del Desarrollo Cortical/tratamiento farmacológico , Ratones , Ratones Noqueados , Sirolimus/farmacología , Sirolimus/uso terapéutico
5.
J Neurosci ; 33(39): 15652-68, 2013 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-24068831

RESUMEN

Disabled-1 (Dab1) is an adaptor protein that is an obligate effector of the Reelin signaling pathway, and is critical for neuronal migration and dendrite outgrowth during development. Components of the Reelin pathway are highly expressed during development, but also continue to be expressed in the adult brain. Here we investigated in detail the expression pattern of Dab1 in the postnatal and adult forebrain, and determined that it is expressed in excitatory as well as inhibitory neurons. Dab1 was found to be localized in different cellular compartments, including the soma, dendrites, presynaptic and postsynaptic structures. Mice that are deficient in Dab1, Reelin, or the Reelin receptors ApoER2 and VLDLR exhibit severely perturbed brain cytoarchitecture, limiting the utility of these mice for investigating the role of this signaling pathway in the adult brain. In this study, we developed an adult forebrain-specific and excitatory neuron-specific conditional knock-out mouse line, and demonstrated that Dab1 is a critical regulator of synaptic function and hippocampal-dependent associative and spatial learning. These dramatic abnormalities were accompanied by a reduction in dendritic spine size, and defects in basal and plasticity-induced Akt and ERK1/2 signaling. Deletion of Dab1 led to no obvious changes in neuronal positioning, dendrite morphology, spine density, or synaptic composition. Collectively, these data conclusively demonstrate an important role for Reelin-Dab1 signaling in the adult forebrain, and underscore the importance of this pathway in learning and memory.


Asunto(s)
Aprendizaje , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal , Animales , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Dendritas/metabolismo , Dendritas/fisiología , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiología , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Prosencéfalo/citología , Prosencéfalo/metabolismo , Prosencéfalo/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Proteína Reelina , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Sinapsis/metabolismo , Sinapsis/fisiología
6.
Nat Genet ; 35(3): 270-6, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14578885

RESUMEN

Loss-of-function mutations in RELN (encoding reelin) or PAFAH1B1 (encoding LIS1) cause lissencephaly, a human neuronal migration disorder. In the mouse, homozygous mutations in Reln result in the reeler phenotype, characterized by ataxia and disrupted cortical layers. Pafah1b1(+/-) mice have hippocampal layering defects, whereas homozygous mutants are embryonic lethal. Reln encodes an extracellular protein that regulates layer formation by interacting with VLDLR and ApoER2 (Lrp8) receptors, thereby phosphorylating the Dab1 signaling molecule. Lis1 associates with microtubules and modulates neuronal migration. We investigated interactions between the reelin signaling pathway and Lis1 in brain development. Compound mutant mice with disruptions in the Reln pathway and heterozygous Pafah1b1 mutations had a higher incidence of hydrocephalus and enhanced cortical and hippocampal layering defects. Dab1 and Lis1 bound in a reelin-induced phosphorylation-dependent manner. These data indicate genetic and biochemical interaction between the reelin signaling pathway and Lis1.


Asunto(s)
Encéfalo/embriología , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Transducción de Señal , 1-Alquil-2-acetilglicerofosfocolina Esterasa , Animales , Humanos , Ratones , Ratones Mutantes Neurológicos , Proteínas del Tejido Nervioso , Proteína Reelina , Serina Endopeptidasas
7.
J Adv Res ; 51: 73-90, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36402285

RESUMEN

INTRODUCTION: Spontaneous recovery after CNS injury is often very limited and incomplete, leaving most stroke patients with permanent disability. Maf1 is known as a key growth suppressor in proliferating cells. However, its role in neuronal cells after stroke remains unclear. OBJECTIVE: We aimed to investigate the mechanistic role of Maf1 in spontaneous neural repair and evaluated the therapeutic effect of targeting Maf1 on stroke recovery. METHODS: We used mouse primary neurons to determine the signaling mechanism of Maf1, and the cleavage-under-targets-and-tagmentation-sequencing to map the whole-genome promoter binding sites of Maf1 in isolated mature cortical neurons. Photothrombotic stroke model was used to determine the therapeutic effect on neural repair and functional recovery by AAV-mediated Maf1 knockdown. RESULTS: We found that Maf1 mediates mTOR signaling to regulate RNA polymerase III (Pol III)-dependent rRNA and tRNA transcription in mouse cortical neurons. mTOR regulates neuronal Maf1 phosphorylation and subcellular localization. Maf1 knockdown significantly increases Pol III transcription, neurite outgrowth and dendritic spine formation in neurons. Conversely, Maf1 overexpression suppresses such activities. In response to photothrombotic stroke in mice, Maf1 expression is increased and accumulates in the nucleus of neurons in the peripheral region of infarcted cortex, which is the key region for neural remodeling and repair during spontaneous recovery. Intriguingly, Maf1 knockdown in the peri-infarct cortex significantly enhances neural plasticity and functional recovery. Mechanistically, Maf1 not only interacts with the promoters and represses Pol III-transcribed genes, but also those of CREB-associated genes that are critical for promoting plasticity during neurodevelopment and neural repair. CONCLUSION: These findings indicate Maf1 as an intrinsic neural repair suppressor against regenerative capability of mature CNS neurons, and suggest that Maf1 is a potential therapeutic target for enhancing functional recovery after ischemic stroke and other CNS injuries.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Animales , Ratones , Transcripción Genética , Serina-Treonina Quinasas TOR/genética , Fosforilación , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
8.
Dev Neurosci ; 34(2-3): 198-209, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22572802

RESUMEN

The phosphatase and tensin homolog located on chromosome 10 (PTEN) suppresses the activity of the phosphoinositide-3-kinase/Akt/mammalian target of rapamycin (mTOR) pathway, a signaling cascade critically involved in the regulation of cell proliferation and growth. Human patients carrying germ line PTEN mutations have an increased predisposition to tumors, and also display a variety of neurological symptoms and increased risk of epilepsy and autism, implicating PTEN in neuronal development and function. Consistently, loss of Pten in mouse neural cells results in ataxia, seizures, cognitive abnormalities, increased soma size and synaptic abnormalities. To better understand how Pten regulates the excitability of principal forebrain neurons, a factor that is likely to be altered in cognitive disorders, epilepsy and autism, we generated a novel conditional knockout mouse line (NEX-Pten) in which Cre, under the control of the NEX promoter, drives the deletion of Pten specifically in early postmitotic, excitatory neurons of the developing forebrain. Homozygous mutant mice exhibited a massive enlargement of the forebrain, and died shortly after birth due to excessive mTOR activation. Analysis of the neonatal cerebral cortex further identified molecular defects resulting from Pten deletion that likely affect several aspects of neuronal development and excitability.


Asunto(s)
Modelos Animales de Enfermedad , Neuronas/metabolismo , Fosfohidrolasa PTEN/genética , Prosencéfalo/metabolismo , Animales , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/metabolismo
9.
J Neurosci ; 30(11): 4004-14, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20237271

RESUMEN

Dyrk1A is a member of the mammalian Dyrk [dual-specificity tyrosine-(Y)-phosphorylation regulated kinase] family of protein kinases that is expressed at high levels in the brain, but its role in the development and function of this organ is not well understood. The human DYRK1A gene is located on trisomic chromosome 21 in Down syndrome (DS) patients, leading to its overexpression. Dyrk1A is also overexpressed in animal models of DS and in gene-specific transgenic mice that consistently exhibit cognitive impairment. To elucidate the cellular and molecular mechanisms that are affected by increased levels of Dyrk1A in the developing brain, we overexpressed this kinase in the embryonic mouse neocortex using the in utero electroporation technique. We found that Dyrk1A overexpression inhibits neural cell proliferation and promotes premature neuronal differentiation in the developing cerebral cortex without affecting cell fate and layer positioning. These effects are dependent on the Dyrk1A kinase activity and are mediated by the nuclear export and degradation of cyclin D1. This study identifies specific Dyrk1A-induced mechanisms that disrupt the normal process of corticogenesis and possibly contribute to cognitive impairment observed in DS patients and animal models.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/genética , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Células Madre/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Inhibidores de Crecimiento/fisiología , Humanos , Ratones , Ratones Transgénicos , Neuronas/citología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Tirosina Quinasas/fisiología , Células Madre/citología , Quinasas DyrK
10.
J Neurosci ; 30(15): 5219-28, 2010 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-20392944

RESUMEN

Neurons that reenter a cell cycle after maturation are at increased risk for death, yet the mechanisms by which a normal neuron suppresses the cycle remain mostly unknown. Our laboratory has shown that cyclin-dependent kinase 5 (Cdk5) is a potent cell cycle suppressor, and we report here on the molecular basis of this activity. Cell cycle suppression by Cdk5 requires its binding to the p35 activator protein. The related p39 and p25 proteins cannot serve as substitutes. Unexpectedly, Cdk5 enzymatic activity is not required to perform this function. Rather, the link to cell cycle regulation is made through the formation of a previously unknown complex consisting of the p35-Cdk5 dimer and E2F1. Formation of this complex excludes the E2F1 cofactor, DP1, thus inhibiting E2F1 binding to the promoters of various cell cycle genes. This anti-cell cycle activity is most likely a neuroprotective function of Cdk5.


Asunto(s)
Ciclo Celular/fisiología , Quinasa 5 Dependiente de la Ciclina/metabolismo , Factor de Transcripción E2F1/metabolismo , Neuronas/fisiología , Fosfotransferasas/metabolismo , Factor de Transcripción DP1/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/fisiología , Línea Celular Tumoral , Núcleo Celular/fisiología , Células Cultivadas , Quinasa 5 Dependiente de la Ciclina/deficiencia , Quinasa 5 Dependiente de la Ciclina/genética , Citoplasma/fisiología , ADN/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Neurológicos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/enzimología
11.
Epilepsia ; 52(11): 2065-75, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21973019

RESUMEN

PURPOSE: Increased activity of mTOR Complex 1 (mTORC1) has been demonstrated in cortical dysplasia and tuberous sclerosis complex, as well as in animal models of epilepsy. Recent studies in such models revealed that inhibiting mTORC1 with rapamycin effectively suppressed seizure activity. However, seizures can recur after treatment cessation, and continuous rapamycin exposure can adversely affect animal growth and health. Here, we evaluated the efficacy of an intermittent rapamycin treatment protocol on epilepsy progression using neuron subset-specific-Pten (NS-Pten) conditional knockout mice. METHODS: NS-Pten knockouts were treated with a single course of rapamycin during postnatal weeks 4 and 5, or intermittently over a period of 5 months. Epileptiform activity was monitored using video-electroencephalography (EEG) recordings, and mossy fiber sprouting was evaluated using Timm staining. Survival and body weight were assessed in parallel. KEY FINDINGS: NS-Pten knockouts treated with a single course of rapamycin had recurrence of epilepsy 4-7 weeks after treatment ended. In contrast, epileptiform activity remained suppressed, and survival increased if knockout mice received additional rapamycin during weeks 10-11 and 16-17. Aberrant mossy fiber sprouting, present by 4 weeks of age and progressing in parallel with epileptiform activity, was also blocked by rapamycin. SIGNIFICANCE: These findings demonstrate that a single course of rapamycin treatment suppresses epileptiform activity and mossy fiber sprouting for several weeks before epilepsy recurs. However, additional intermittent treatments with rapamycin prevented this recurrence and enhanced survival without compromising growth. Therefore, these studies add to the growing body of evidence implicating an important role for mTORC1 signaling in epilepsy.


Asunto(s)
Epilepsia/fisiopatología , Fosfohidrolasa PTEN/fisiología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Giro Dentado/efectos de los fármacos , Giro Dentado/fisiopatología , Progresión de la Enfermedad , Electroencefalografía , Epilepsia/tratamiento farmacológico , Femenino , Masculino , Ratones , Fibras Nerviosas/efectos de los fármacos , Fosfohidrolasa PTEN/genética , Sirolimus/administración & dosificación , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/fisiología
12.
Structure ; 29(10): 1156-1170.e6, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34089653

RESUMEN

Reelin operates through canonical and non-canonical pathways that mediate several aspects of brain development and function. Reelin's dimeric central fragment (CF), generated through proteolytic cleavage, is required for the lipoprotein-receptor-dependent canonical pathway activation. Here, we analyze the signaling properties of a variety of Reelin fragments and measure the differential binding affinities of monomeric and dimeric CF fragments to lipoprotein receptors to investigate the mode of canonical signal activation. We also present the cryoelectron tomography-solved dimeric structure of Reelin CF and support it using several other biophysical techniques. Our findings suggest that Reelin CF forms a covalent parallel dimer with some degree of flexibility between the two protein chains. As a result of this conformation, Reelin binds to lipoprotein receptors in a manner inaccessible to its monomeric form and is capable of stimulating canonical pathway signaling.


Asunto(s)
Proteína Reelina/química , Microscopía por Crioelectrón , Células HEK293 , Humanos , Dominios Proteicos , Multimerización de Proteína , Receptores de LDL/metabolismo , Proteína Reelina/metabolismo , Transducción de Señal
13.
Biomolecules ; 10(7)2020 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-32610618

RESUMEN

Traumatic brain injury (TBI) is a relatively common occurrence following accidents or violence, and often results in long-term cognitive or motor disability. Despite the high health cost associated with this type of injury, presently there are no effective treatments for many neurological symptoms resulting from TBI. This is due in part to our limited understanding of the mechanisms underlying brain dysfunction after injury. In this study, we used the mouse controlled cortical impact (CCI) model to investigate the effects of TBI, and focused on Reelin, an extracellular protein that critically regulates brain development and modulates synaptic activity in the adult brain. We found that Reelin expression decreases in forebrain regions after TBI, and that the number of Reelin-expressing cells decrease specifically in the hippocampus, an area of the brain that plays an important role in learning and memory. We also conducted in vitro experiments using mouse neuronal cultures and discovered that Reelin protects hippocampal neuronal cells from glutamate-induced neurotoxicity, a well-known secondary effect of TBI. Together our findings suggest that the loss of Reelin expression may contribute to neuronal death in the hippocampus after TBI, and raise the possibility that increasing Reelin levels or signaling activity may promote functional recovery.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Regulación hacia Abajo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/etiología , Lesiones Traumáticas del Encéfalo/genética , Células Cultivadas , Modelos Animales de Enfermedad , Ácido Glutámico/efectos adversos , Masculino , Ratones , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteína Reelina , Transducción de Señal
14.
Neuroreport ; 31(10): 762-769, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32453020

RESUMEN

The mammalian neocortex is composed of six major layers of neurons. Each group of neurons in the cortical layers has distinct characteristics based on the expression of specific genes and connectivity patterns of neural circuits. Neuronal subtype transition and regional identity acquisition are established by temporal cues and interaction between several transcription factors during neurogenesis. The impairment of cortical lamination or neural circuits results in a wide range of neurodevelopmental disorders such as autism, schizophrenia, and certain forms of childhood epilepsy. Despite continuous efforts to classify neurons with the aid of genetic and epigenetic analyses, the neuron-specific properties associated with post-transcriptional modification remain unclear. In the present study, the distribution of phosphorylated S6-positive layers across the neocortex was examined using several layer markers. The development of pS6 S235/236 layers in layer V and the subplate was spatiotemporally regulated in the mouse brain. In addition, enhanced phosphorylation of ribosomal protein S6 in Ctip2-positive layer V neurons in vivo was sustained under in-vitro conditions using a culture of primary cortical neurons.


Asunto(s)
Neocórtex/crecimiento & desarrollo , Neuronas/fisiología , Proteínas Quinasas S6 Ribosómicas/metabolismo , Animales , Ratones , Neocórtex/metabolismo , Neuronas/metabolismo , Fosforilación
15.
Neuron ; 47(4): 471-3, 2005 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-16102527

RESUMEN

The extracellular protein Reelin is crucial for neuronal positioning during brain development, but its expression persists long after cell migration is completed. In this issue of Neuron, Beffert et al. demonstrate that Reelin exerts an additional function in the mature brain, to modulate synaptic plasticity and to favor memory formation. This activity is carried out exquisitely by the Apoer2 receptor and critically requires the presence of an alternatively spliced exon. This exon encodes an intracellular domain that interacts with postsynaptic proteins and promotes binding and phosphorylation of NMDA receptors.


Asunto(s)
Encéfalo/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Lipoproteína/metabolismo , Serina Endopeptidasas/metabolismo , Sinapsis/metabolismo , Empalme Alternativo/fisiología , Animales , Humanos , Proteínas Relacionadas con Receptor de LDL , Plasticidad Neuronal/fisiología , Fosforilación , Unión Proteica/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína Reelina
16.
J Neurosci ; 28(41): 10339-48, 2008 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-18842893

RESUMEN

The development of distinct cellular layers and precise synaptic circuits is essential for the formation of well functioning cortical structures in the mammalian brain. The extracellular protein Reelin, through the activation of a core signaling pathway, including the receptors ApoER2 and VLDLR (very low density lipoprotein receptor) and the adapter protein Dab1 (Disabled-1), controls the positioning of radially migrating principal neurons, promotes the extension of dendritic processes in immature forebrain neurons, and affects synaptic transmission. Here we report for the first time that the Reelin signaling pathway promotes the development of postsynaptic structures such as dendritic spines in hippocampal pyramidal neurons. Our data underscore the importance of Reelin as a factor that promotes the maturation of target neuronal populations and the development of excitatory circuits in the postnatal hippocampus. These findings may have implications for understanding the origin of cognitive disorders associated with Reelin deficiency.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Espinas Dendríticas/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Hipocampo/fisiología , Proteínas del Tejido Nervioso/metabolismo , Células Piramidales/fisiología , Serina Endopeptidasas/metabolismo , Transducción de Señal/fisiología , Animales , Moléculas de Adhesión Celular Neuronal/deficiencia , Células Cultivadas , Espinas Dendríticas/ultraestructura , Proteínas de la Matriz Extracelular/deficiencia , Hipocampo/citología , Hipocampo/ultraestructura , Proteínas Relacionadas con Receptor de LDL , Ratones , Proteínas del Tejido Nervioso/deficiencia , Células Piramidales/ultraestructura , Receptores de LDL/metabolismo , Receptores de Lipoproteína/metabolismo , Proteínas Recombinantes/metabolismo , Proteína Reelina , Serina Endopeptidasas/deficiencia , Familia-src Quinasas/metabolismo
17.
Epilepsia ; 50 Suppl 9: 28-33, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19761451

RESUMEN

Cortical dysplasia, a malformation of the cerebral cortex of developmental origin, accounts for most cases of drug-resistant childhood epilepsy. Despite its prevalence, the genetic and molecular bases for the disease are not known for most cases. With the exception of tuberous sclerosis complex, a type of cortical malformation with known genetic etiology, no mutated genes have been identified that can lead to the neuroanatomic abnormalities observed in the patients. Furthermore, there are currently no animal models that completely recapitulate the features of the disease, hindering progress in developing new treatments for this devastating condition. In this article, I review some of the approaches that researchers in the field are undertaking to utilize the biopsy tissue obtained from patients with cortical dysplasia who are undergoing epilepsy surgery. These multifaceted approaches are providing a plethora of new information that is then utilized to generate and evaluate animal models and to further our understanding of the disease. These studies will enable the development of novel antiepileptic treatments for patients with cortical dysplasia and similar disorders.


Asunto(s)
Corteza Cerebral/patología , Epilepsia/patología , Malformaciones del Desarrollo Cortical/patología , Animales , Biopsia/estadística & datos numéricos , Corteza Cerebral/cirugía , Modelos Animales de Enfermedad , Epilepsia/genética , Epilepsia/cirugía , Genes Supresores de Tumor/fisiología , Humanos , Lactante , Malformaciones del Desarrollo Cortical/genética , Malformaciones del Desarrollo Cortical/cirugía , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos , Proteínas , Transducción de Señal/genética , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR , Factores de Transcripción/genética , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/patología , Esclerosis Tuberosa/cirugía , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología
18.
Neuron ; 41(1): 71-84, 2004 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-14715136

RESUMEN

Reelin is a secreted glycoprotein that regulates neuronal positioning in cortical brain structures through the VLDLR and ApoER2 receptors and the adaptor protein Dab1. In addition to cellular disorganization, dendrite abnormalities are present in the brain of reeler mice lacking Reelin. It is unclear whether these defects are due primarily to cellular ectopia or the absence of Reelin. Here we examined dendrite development in the hippocampus of normal and mutant mice and in dissociated cultures. We found that dendrite complexity is severely reduced in homozygous mice deficient in Reelin signaling both in vivo and in vitro, and it is also reduced in heterozygous mice in the absence of cellular ectopia. Addition of Reelin interfering antibodies, receptor antagonists, and Dab1 phosphorylation inhibitors prevented dendrite outgrowth from normal neurons, whereas addition of recombinant Reelin rescued the deficit in reeler cultures. Thus, the same signaling pathway controls both neuronal migration and dendrite maturation.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Dendritas/fisiología , Proteínas de la Matriz Extracelular/fisiología , Hipocampo/crecimiento & desarrollo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/fisiología , Receptores de LDL/metabolismo , Receptores de Lipoproteína/metabolismo , Serina Endopeptidasas/fisiología , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Células Cultivadas , Proteínas de la Matriz Extracelular/metabolismo , Proteínas Relacionadas con Receptor de LDL , Ratones , Ratones Noqueados , Ratones Mutantes Neurológicos , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/fisiología , Proteína Reelina , Serina Endopeptidasas/metabolismo , Transducción de Señal/fisiología
19.
Neurosci Lett ; 439(1): 100-5, 2008 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-18514414

RESUMEN

Reelin, an extracellular protein that signals through the Dab1 adapter protein, and Lis1 regulate neuronal migration and cellular layer formation in the brain. Loss of Reelin and reduction in Lis1 activity in mice or humans results in the disorganization of cortical structures. Lis1, the product of the Pafah1b1 gene associates with Alpha1 (the product of the Pafah1b3 gene) and Alpha2 (the product of the Pafah1b2 gene) to form the Pafah1b heterotrimeric complex. This complex interacts biochemically and genetically with the Reelin pathway, however, the role of Alpha1 and Alpha2 in brain development is poorly understood. We previously demonstrated that compound mutations of Pafah1b1 with genes in Reelin pathway result in layering defects and the appearance of hydrocephalus in double mutant mice. Here, we generate triple mouse mutants to investigate the effect of individual Pafah1b Alpha subunits on cellular layer formation and hydrocephalus. We found that Pafah1b3 mutations exacerbate the layering defects, whereas Pafah1b2 mutations strongly suppress the hydrocephalus phenotype of compound mutant mice. The data indicate that the two Pafah1b Alpha subunits have profoundly different effects on brain development and interact in a significantly different manner with the Reelin signaling pathway.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/genética , 1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Hidrocefalia/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Animales Recién Nacidos , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Genotipo , Hipocampo/patología , Hidrocefalia/patología , Ratones , Ratones Mutantes , Proteínas Asociadas a Microtúbulos/genética , Proteína Reelina
20.
Brain Res ; 1140: 75-83, 2007 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-16996039

RESUMEN

The majority of cortical and hippocampal interneurons originate in the subcortical telencephalon and migrate tangentially into pallial regions before settling in various cortical layers. The molecular cues that regulate final positioning of specific interneurons in cortical structures have not yet been identified. The positioning of radially migrating principal neurons of the cortex and hippocampus depends upon Reelin, an extracellular protein expressed near the pial surface during embryonic development that is absent in reeler mutant mice. To determine whether the layer specification of interneurons, like that of principal neurons, requires Reelin, we crossed reeler with transgenic mice that contain Green Fluorescent Protein (GFP)-expressing Inhibitory Neurons (GINs). These neurons express basal forebrain markers Dlx1/2 in normal and reeler mice. In normal mice, GINs express Reelin and are localized to specific layers of the cortex and hippocampus. In reeler mutant mice, we show that GINs migrate normally into the pallium, but fail to acquire proper layer position. Double labeling experiments indicate that the neurochemical profile of these interneurons is not generally altered in reeler mice. However, the extension of their cellular processes is abnormal. Quantitative analysis of GINs in the cortex revealed that they are hypertrophic, bearing longer neuritic branches than normal. Thus, the lack of Reelin signaling results in abnormal positioning and altered morphology of forebrain interneurons.


Asunto(s)
Dendritas/fisiología , Interneuronas/citología , Ratones Mutantes Neurológicos/anatomía & histología , Prosencéfalo/anomalías , Prosencéfalo/citología , Animales , Animales Recién Nacidos , Tipificación del Cuerpo/fisiología , Recuento de Células , Movimiento Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , Hipocampo/fisiología , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Proteína Reelina , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA