Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Clin Invest ; 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38980724

RESUMEN

Reelin (RELN) is a secreted glycoprotein essential for cerebral cortex development. In humans, recessive RELN variants cause cortical and cerebellar malformations, while heterozygous variants were associated to epilepsy, autism and mild cortical abnormalities. However, their functional effects remain unknown. We identified inherited and de novo RELN missense variants in heterozygous patients with neuronal migration disorders (NMDs) as diverse as pachygyria and polymicrogyria. We investigated in culture and in the developing mouse cerebral cortex how different variants impacted RELN function. Polymicrogyria-associated variants behaved as gain-of-function showing an enhanced ability to induce neuronal aggregation, while those linked to pachygyria as loss-of-function leading to defective neuronal aggregation/migration. The pachygyria-associated de novo heterozygous RELN variants acted as dominant-negative by preventing wild-type RELN secretion in culture, animal models and patients, thereby causing dominant NMDs. We demonstrated how mutant RELN proteins in vitro and in vivo predict cortical malformation phenotypes, providing valuable insights into the pathogenesis of such disorders.

2.
Nat Commun ; 13(1): 6571, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36323680

RESUMEN

Astrocytes are one of the most abundant cell types in the mammalian brain. They play essential roles in synapse formation, maturation, and elimination. However, how astrocytes migrate into the gray matter to accomplish these processes is poorly understood. Here, we show that, by combinational analyses of in vitro and in vivo time-lapse observations and lineage traces, astrocyte progenitors move rapidly and irregularly within the developing cortex, which we call erratic migration. Astrocyte progenitors also adopt blood vessel-guided migration. These highly motile progenitors are generated in the restricted prenatal stages and differentiate into protoplasmic astrocytes in the gray matter, whereas postnatally generated progenitors do not move extensively and differentiate into fibrous astrocytes in the white matter. We found Cxcr4/7, and integrin ß1 regulate the blood vessel-guided migration, and their functional blocking disrupts their positioning. This study provides insight into astrocyte development and may contribute to understanding the pathogenesis caused by their defects.


Asunto(s)
Astrocitos , Corteza Cerebral , Animales , Astrocitos/metabolismo , Corteza Cerebral/metabolismo , Encéfalo/metabolismo , Integrina beta1/metabolismo , Transducción de Señal , Mamíferos/metabolismo
3.
Commun Biol ; 5(1): 1065, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36207396

RESUMEN

N-cadherin (NCad) is a classical cadherin that mediates cell-cell interactions in a Ca2+-dependent manner. NCad participates in various biological processes, from ontogenesis to higher brain functions, though the visualization of NCad interactions in living cells remains limited. Here, we present intensiometric NCad interaction indicators, named INCIDERs, that utilize dimerization-dependent fluorescent proteins. INCIDERs successfully visualize reversible NCad interactions across cells. Compared to FRET-based indicators, INCIDERs have a ~70-fold higher signal contrast, enabling clear identification of NCad interactions. In primary neuronal cells, NCad interactions are visualized between closely apposed processes. Furthermore, visualization of NCad interaction at cell adhesion sites in dense cell populations is achieved by two-photon microscopy. INCIDERs are useful tools in the spatiotemporal investigation of NCad interactions across cells; future research should evaluate the potential of INCIDERs in mapping complex three-dimensional architectures in multi-cellular systems.


Asunto(s)
Cadherinas , Neuronas , Cadherinas/metabolismo , Adhesión Celular , Neuronas/metabolismo
4.
Mol Brain ; 14(1): 90, 2021 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-34118975

RESUMEN

Homer is a postsynaptic scaffold protein, which has long and short isoforms. The long form of Homer consists of an N-terminal target-binding domain and a C-terminal multimerization domain, linking multiple proteins within a complex. The short form of Homer only has the N-terminal domain and likely acts as a dominant negative regulator. Homer2a, one of the long form isoforms of the Homer family, expresses with a transient peak in the early postnatal stage of mouse cerebellar granule cells (CGCs); however, the functions of Homer2a in CGCs are not fully understood yet. In this study, we investigated the physiological roles of Homer2a in CGCs using recombinant adenovirus vectors. Overexpression of the Homer2a N-terminal domain construct, which was made structurally reminiscent with Homer1a, altered NMDAR1 localization, decreased NMDA currents, and promoted the survival of CGCs. These results suggest that the Homer2a N-terminal domain acts as a dominant negative protein to attenuate NMDAR-mediated excitotoxicity. Moreover, we identified a novel short form N-terminal domain-containing Homer2, named Homer2e, which was induced by apoptotic stimulation such as ischemic brain injury. Our study suggests that the long and short forms of Homer2 are involved in apoptosis of CGCs.


Asunto(s)
Apoptosis , Cerebelo/citología , Proteínas de Andamiaje Homer/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Isquemia Encefálica/patología , Proteínas de Andamiaje Homer/química , Proteínas de Andamiaje Homer/genética , Ratones Endogámicos ICR , Modelos Biológicos , N-Metilaspartato/metabolismo , Dominios Proteicos , Isoformas de Proteínas/metabolismo
5.
J Comp Neurol ; 527(10): 1577-1597, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30636008

RESUMEN

During development of the mammalian cerebral neocortex, postmitotic excitatory neurons migrate toward the outermost region of the neocortex. We previously reported that this outermost region is composed of densely packed relatively immature neurons; we named this region, which is observed during the late stage of mouse neocortical development, the "primitive cortical zone (PCZ)." Here, we report that postmigratory immature neurons spend about 1-1.5 days in the PCZ. An electron microscopic analysis showed that the neurons in the PCZ tend to be in direct contact with each other, mostly in a radial direction, forming "primitive neuronal clusters" with a height of 3-7 cells and a width of 1-2 cells. A time-course analysis of fluorescently labeled neurons revealed that the neurons took their positions within the primitive clusters in an inside-out manner. The neurons initially participated in the superficial part of the clusters, gradually shifted their relative positions downward, and then left the clusters at the bottom of this structure. GABAergic inhibitory interneurons were also found within the primitive clusters in the developing mouse neocortex, suggesting that some clusters are composed of both excitatory neurons and inhibitory interneurons. Similar clusters were also observed in the outermost region of embryonic day (E) 78 cynomolgus monkey occipital cortex and 23 gestational week (GW) human neocortices. In the primate neocortices, including human, the presumptive primitive clusters seemed to expand in the radial direction more than that observed in mice, which might contribute to the functional integrity of the primate neocortex.


Asunto(s)
Movimiento Celular/fisiología , Neocórtex/embriología , Neurogénesis/fisiología , Neuronas/fisiología , Animales , Humanos , Macaca fascicularis , Ratones
6.
J Neurosci ; 39(4): 678-691, 2019 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-30504273

RESUMEN

The actin cytoskeleton is crucial for neuronal migration in the mammalian developing cerebral cortex. The adaptor protein Drebrin-like (Dbnl) plays important roles in reorganization of the actin cytoskeleton, dendrite formation, and endocytosis by interacting with F-actin, cobl, and dynamin. Although Dbnl is known to be expressed in the brain, the functions of this molecule during brain development are largely unknown. In this study, to examine the roles of Dbnl in the developing cerebral cortex, we conducted experiments using mice of both sexes with knockdown of Dbnl, effected by in utero electroporation, in the migrating neurons of the embryonic cortex. Time-lapse imaging of the Dbnl-knockdown neurons revealed that the presence of Dbnl is a prerequisite for appropriate formation of processes in the multipolar neurons in the multipolar cell accumulation zone or the deep part of the subventricular zone, and for neuronal polarization and entry into the cortical plate. We found that Dbnl knockdown decreased the amount of N-cadherin protein expressed on the plasma membrane of the cortical neurons. The defect in neuronal migration caused by Dbnl knockdown was rescued by moderate overexpression of N-cadherin and αN-catenin or by transfection of the phospho-mimic form (Y337E, Y347E), but not the phospho-resistant form (Y337F, Y347F), of Dbnl. These results suggest that Dbnl controls neuronal migration, neuronal multipolar morphology, and cell polarity in the developing cerebral cortex via regulating N-cadherin expression.SIGNIFICANCE STATEMENT Disruption of neuronal migration can cause neuronal disorders, such as lissencephaly and subcortical band heterotopia. During cerebral cortical development, the actin cytoskeleton plays a key role in neuronal migration; however, the mechanisms of regulation of neuronal migration by the actin cytoskeleton still remain unclear. Herein, we report that the novel protein Dbnl, an actin-binding protein, controls multiple events during neuronal migration in the developing mouse cerebral cortex. We also showed that this regulation is mediated by phosphorylation of Dbnl at tyrosine residues 337 and 347 and αN-catenin/N-cadherin, suggesting that the Dbnl-αN-catenin/N-cadherin pathway is important for neuronal migration in the developing cortex.


Asunto(s)
Cadherinas/biosíntesis , Movimiento Celular/fisiología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/fisiología , Proteínas de Microfilamentos/fisiología , Neuronas/fisiología , Dominios Homologos src/fisiología , Animales , Cadherinas/genética , Membrana Celular/metabolismo , Corteza Cerebral/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ventrículos Laterales/citología , Ventrículos Laterales/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/genética , Neuronas/ultraestructura , Embarazo , Dominios Homologos src/genética
7.
Elife ; 62017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29179815

RESUMEN

CHARGE syndrome is caused by heterozygous mutations in the chromatin remodeler, CHD7, and is characterized by a set of malformations that, on clinical grounds, were historically postulated to arise from defects in neural crest formation during embryogenesis. To better delineate neural crest defects in CHARGE syndrome, we generated induced pluripotent stem cells (iPSCs) from two patients with typical syndrome manifestations, and characterized neural crest cells differentiated in vitro from these iPSCs (iPSC-NCCs). We found that expression of genes associated with cell migration was altered in CHARGE iPSC-NCCs compared to control iPSC-NCCs. Consistently, CHARGE iPSC-NCCs showed defective delamination, migration and motility in vitro, and their transplantation in ovo revealed overall defective migratory activity in the chick embryo. These results support the historical inference that CHARGE syndrome patients exhibit defects in neural crest migration, and provide the first successful application of patient-derived iPSCs in modeling craniofacial disorders.


Asunto(s)
Síndrome CHARGE/fisiopatología , Movimiento Celular , Cresta Neural/fisiología , Animales , Síndrome CHARGE/genética , Diferenciación Celular , Células Cultivadas , Embrión de Pollo , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Proteínas Mutantes/genética , Mutación
8.
EBioMedicine ; 24: 102-115, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28928015

RESUMEN

Cytoplasmic aggregation of fused in sarcoma (FUS) is detected in brain regions affected by amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), which compose the disease spectrum, FUS proteinopathy. To understand the pathomechanism of ALS-FTD-associated FUS, we examined the behavior and cellular properties of an ALS mouse model overexpressing FUS with nuclear localization signal deletion. Mutant FUS transgenic mice showed hyperactivity, social interactional deficits, and impaired fear memory retrieval, all of which are compatible with FTD phenotypes. Histological analyses showed decreased dendritic spine and synaptic density in the frontal cortex before neuronal loss. Examination of cultured cells confirmed that mutant but not wild-type FUS was associated with decreased dendritic growth, mRNA levels, and protein synthesis in dendrites. These data suggest that cytoplasmic FUS aggregates impair dendritic mRNA trafficking and translation, in turn leading to dendritic homeostasis disruption and the development of FTD phenotypes.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Demencia Frontotemporal/metabolismo , ARN Mensajero/metabolismo , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Espinas Dendríticas/metabolismo , Modelos Animales de Enfermedad , Demencia Frontotemporal/genética , Humanos , Ratones , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Biosíntesis de Proteínas
9.
Brain Struct Funct ; 222(9): 4283-4291, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28836069

RESUMEN

Mutations of PAFAH1B1 cause classical lissencephaly in humans. In addition, duplications and triplications of PAFAH1B1 are found in individuals with intellectual disability and other neurological disorders suggesting that proper brain development is highly sensitive to the PAFAH1B1 dosage. To examine the effect of PAFAH1B1 over-dosage in neural development, especially in migration of neurons and layer formation during cerebral cortical development, we overexpressed Pafah1b1 in migrating neurons in the mouse embryonic cortex using in utero electroporation. Enhanced expression of Pafah1b1 in radially-migrating neurons resulted in their over-migration into the marginal zone. Neurons that invaded the marginal zone were oriented abnormally. Layer distribution of Pafaha1b1-overexpressing neurons shifted more superficially than control neurons. Some of the Pafaha1b1-overexpressing future layer 4 neurons changed their positions to layers 2/3. Furthermore, they also changed their layer marker expression from layer 4 to layers 2/3. These results suggest that overexpression of Pafah1b1 affects the migration of neurons and disrupts layer formation in the developing cerebral cortex, and further support the idea that appropriate dosage of Pafah1b1 is crucial for the proper development of the brain.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Movimiento Celular/fisiología , Corteza Cerebral/citología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/fisiología , 1-Alquil-2-acetilglicerofosfocolina Esterasa/genética , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/genética , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteínas de Dominio T Box , Factores de Transcripción/metabolismo
10.
JCI Insight ; 2(10)2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28515367

RESUMEN

Many extremely preterm infants (born before 28 gestational weeks [GWs]) develop cognitive impairment in later life, although the underlying pathogenesis is not yet completely understood. Our examinations of the developing human neocortex confirmed that neuronal migration continues beyond 23 GWs, the gestational week at which extremely preterm infants have live births. We observed larger numbers of ectopic neurons in the white matter of the neocortex in human extremely preterm infants with brain injury and hypothesized that altered neuronal migration may be associated with cognitive impairment in later life. To confirm whether preterm brain injury affects neuronal migration, we produced brain damage in mouse embryos by occluding the maternal uterine arteries. The mice showed delayed neuronal migration, ectopic neurons in the white matter, altered neuronal alignment, and abnormal corticocortical axonal wiring. Similar to human extremely preterm infants with brain injury, the surviving mice exhibited cognitive deficits. Activation of the affected medial prefrontal cortices of the surviving mice improved working memory deficits, indicating that decreased neuronal activity caused the cognitive deficits. These findings suggest that altered neuronal migration altered by brain injury might contribute to the subsequent development of cognitive impairment in extremely preterm infants.

11.
PLoS One ; 12(3): e0173175, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28264072

RESUMEN

Very-KIND/Kndc1/KIAA1768 (v-KIND) is a brain-specific Ras guanine nucleotide exchange factor carrying two sets of the kinase non-catalytic C-lobe domain (KIND), and is predominantly expressed in cerebellar granule cells. Here, we report the impact of v-KIND deficiency on dendritic and synaptic growth in cerebellar granule cells in v-KIND knockout (KO) mice. Furthermore, we evaluate motor function in these animals. The gross anatomy of the cerebellum, including the cerebellar lobules, layered cerebellar cortex and densely-packed granule cell layer, in KO mice appeared normal, and was similar to wild-type (WT) mice. However, KO mice displayed an overgrowth of cerebellar granule cell dendrites, compared with WT mice, resulting in an increased number of dendrites, dendritic branches and terminals. Immunoreactivity for vGluT2 (a marker for excitatory presynapses of mossy fiber terminals) was increased in the cerebellar glomeruli of KO mice, compared with WT mice. The postsynaptic density around the terminals of mossy fibers was also increased in KO mice. Although there were no significant differences in locomotor ability between KO and WT animals in their home cages or in the open field, young adult KO mice had an increased grip strength and a tendency to exhibit better motor performance in balance-related tests compared with WT animals. Taken together, our results suggest that v-KIND is required for compact dendritic growth and proper excitatory synaptic connections in cerebellar granule cells, which are necessary for normal motor coordination and balance.


Asunto(s)
Encéfalo/metabolismo , Cerebelo/citología , Cerebelo/metabolismo , Dendritas/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Proteínas del Tejido Nervioso/genética , Desempeño Psicomotor , Animales , Axones/metabolismo , Biomarcadores , Potenciales Postsinápticos Excitadores , Factores de Intercambio de Guanina Nucleótido/química , Inmunohistoquímica , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/química , Especificidad de Órganos/genética , Sinapsis/metabolismo , Sinapsis/ultraestructura
12.
Proc Natl Acad Sci U S A ; 114(8): 2048-2053, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28174271

RESUMEN

Reelin is an essential glycoprotein for the establishment of the highly organized six-layered structure of neurons of the mammalian neocortex. Although the role of Reelin in the control of neuronal migration has been extensively studied at the molecular level, the mechanisms underlying Reelin-dependent neuronal layer organization are not yet fully understood. In this study, we directly showed that Reelin promotes adhesion among dissociated neocortical neurons in culture. The Reelin-mediated neuronal aggregation occurs in an N-cadherin-dependent manner, both in vivo and in vitro. Unexpectedly, however, in a rotation culture of dissociated neocortical cells that gradually reaggregated over time, we found that it was the neural progenitor cells [radial glial cells (RGCs)], rather than the neurons, that tended to form clusters in the presence of Reelin. Mathematical modeling suggested that this clustering of RGCs could be recapitulated if the Reelin-dependent promotion of neuronal adhesion were to occur only transiently. Thus, we directly measured the adhesive force between neurons and N-cadherin by atomic force microscopy, and found that Reelin indeed enhanced the adhesiveness of neurons to N-cadherin; this enhanced adhesiveness began to be observed at 30 min after Reelin stimulation, but declined by 3 h. These results suggest that Reelin transiently (and not persistently) promotes N-cadherin-mediated neuronal aggregation. When N-cadherin and stabilized ß-catenin were overexpressed in the migrating neurons, the transfected neurons were abnormally distributed in the superficial region of the neocortex, suggesting that appropriate regulation of N-cadherin-mediated adhesion is important for correct positioning of the neurons during neocortical development.


Asunto(s)
Cadherinas/metabolismo , Moléculas de Adhesión Celular Neuronal/fisiología , Adhesión Celular/fisiología , Proteínas de la Matriz Extracelular/fisiología , Neocórtex/embriología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Serina Endopeptidasas/fisiología , beta Catenina/metabolismo , Animales , Cadherinas/genética , Moléculas de Adhesión Celular Neuronal/genética , Movimiento Celular , Células Cultivadas , Células Ependimogliales , Proteínas de la Matriz Extracelular/genética , Femenino , Técnicas de Silenciamiento del Gen , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Microscopía de Fuerza Atómica , Proteínas del Tejido Nervioso/genética , Neurogénesis , Neuronas/ultraestructura , Proteína Reelina , Serina Endopeptidasas/genética , Imagen Individual de Molécula
13.
eNeuro ; 3(4)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27622210

RESUMEN

During neural development, endosomal trafficking controls cell shape and motility through the polarized transport of membrane proteins related to cell-cell and cell-extracellular matrix interactions. ADP ribosylation factor 6 (Arf6) is a critical small GTPase that regulates membrane trafficking between the plasma membrane and endosomes. We herein demonstrated that the knockdown of endogenous Arf6 in mouse cerebral cortices led to impaired neuronal migration in the intermediate zone and cytoplasmic retention of N-cadherin and syntaxin12 in migrating neurons. Rescue experiments with separation-of-function Arf6 mutants identified Rab11 family-interacting protein 3 (FIP3)/Arfophilin-1, a dual effector for Arf6 and Rab11, as a downstream effector of Arf6 in migrating neurons. The knockdown of FIP3 led to impaired neuronal migration in the intermediate zone and cytoplasmic retention of N-cadherin in migrating neurons, similar to that of Arf6, which could be rescued by the coexpression of wild-type FIP3 but not FIP3 mutants lacking the binding site for Arf6 or Rab11. These results suggest that Arf6 regulates cortical neuronal migration in the intermediate zone through the FIP3-dependent endosomal trafficking.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Portadoras/metabolismo , Movimiento Celular/fisiología , Corteza Cerebral/crecimiento & desarrollo , Neuronas/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Animales , Sitios de Unión/genética , Transporte Biológico Activo/fisiología , Cadherinas/metabolismo , Corteza Cerebral/metabolismo , Citoplasma/metabolismo , Endosomas/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Ratones Endogámicos ICR , Ratones Transgénicos , Mutación , Proteínas Qa-SNARE/metabolismo
14.
Proc Natl Acad Sci U S A ; 112(36): E4985-94, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26305926

RESUMEN

The preoptic area (POa) of the rostral diencephalon supplies the neocortex and the amygdala with GABAergic neurons in the developing mouse brain. However, the molecular mechanisms that determine the pathway and destinations of POa-derived neurons have not yet been identified. Here we show that Chicken ovalbumin upstream promoter transcription factor II (COUP-TFII)-induced expression of Neuropilin-2 (Nrp2) and its down-regulation control the destination of POa-derived GABAergic neurons. Initially, a majority of the POa-derived migrating neurons express COUP-TFII and form a caudal migratory stream toward the caudal subpallium. When a subpopulation of cells steers toward the neocortex, they exhibit decreased expression of COUP-TFII and Nrp2. The present findings show that suppression of COUP-TFII/Nrp2 changed the destination of the cells into the neocortex, whereas overexpression of COUP-TFII/Nrp2 caused cells to end up in the medial part of the amygdala. Taken together, these results reveal that COUP-TFII/Nrp2 is a molecular switch determining the pathway and destination of migrating GABAergic neurons born in the POa.


Asunto(s)
Encéfalo/metabolismo , Factor de Transcripción COUP II/metabolismo , Diencéfalo/metabolismo , Neuronas GABAérgicas/metabolismo , Neuropilina-2/metabolismo , Amígdala del Cerebelo/embriología , Amígdala del Cerebelo/metabolismo , Animales , Western Blotting , Encéfalo/embriología , Factor de Transcripción COUP II/genética , Movimiento Celular/genética , Diencéfalo/embriología , Neuronas GABAérgicas/citología , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Ratones Endogámicos ICR , Ratones Noqueados , Microscopía Confocal , Neocórtex/embriología , Neocórtex/metabolismo , Neuropilina-2/genética , Área Preóptica/embriología , Área Preóptica/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Técnicas de Cultivo de Tejidos
15.
Nat Neurosci ; 18(8): 1094-100, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26192746

RESUMEN

The cAMP and cAMP-dependent protein kinase A (PKA) signaling cascade is a ubiquitous pathway acting downstream of multiple neuromodulators. We found that the phosphorylation of phosphodiesterase-4 (PDE4) by cyclin-dependent protein kinase 5 (Cdk5) facilitated cAMP degradation and homeostasis of cAMP/PKA signaling. In mice, loss of Cdk5 throughout the forebrain elevated cAMP levels and increased PKA activity in striatal neurons, and altered behavioral responses to acute or chronic stressors. Ventral striatum- or D1 dopamine receptor-specific conditional knockout of Cdk5, or ventral striatum infusion of a small interfering peptide that selectively targeted the regulation of PDE4 by Cdk5, produced analogous effects on stress-induced behavioral responses. Together, our results demonstrate that altering cAMP signaling in medium spiny neurons of the ventral striatum can effectively modulate stress-induced behavioral states. We propose that targeting the Cdk5 regulation of PDE4 could be a new therapeutic approach for clinical conditions associated with stress, such as depression.


Asunto(s)
Conducta Animal/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Transducción de Señal/fisiología , Estrés Psicológico/metabolismo , Estriado Ventral/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Front Neurosci ; 9: 135, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25964735

RESUMEN

A fine structure of the hippocampus is required for proper functions, and disruption of this formation by neuronal migration defects during development may play a role in some psychiatric illnesses. During hippocampal development in rodents, pyramidal neurons in the Ammon's horn are mostly generated in the ventricular zone (VZ), spent as multipolar cells just above the VZ, and then migrate radially toward the pial surface, ultimately settling into the hippocampal plate. Although this process is similar to that of neocortical projection neurons, these are not identical. In addition to numerous histological studies, the development of novel techniques gives a clear picture of the cellular dynamics of hippocampal neurons, as well as neocortical neurons. In this article, we provide an overview of the cellular mechanisms of rodent hippocampal neuronal migration including those of dentate granule cells, especially focusing on the differences of migration modes between hippocampal neurons and neocortical neurons. The unique migration mode of hippocampal pyramidal neurons might enable clonally related cells in the Ammon's horn to distribute in a horizontal fashion.

17.
J Neurosci ; 34(4): 1115-26, 2014 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-24453304

RESUMEN

The hippocampus plays important roles in brain functions. Despite the importance of hippocampal functions, recent analyses of neuronal migration have mainly been performed on the cerebral neocortex, and the cellular mechanisms responsible for the formation of the hippocampus are not yet completely understood. Moreover, why a prolonged time is required for hippocampal neurons to complete their migration has been unexplainable for several decades. We analyzed the migratory profile of neurons in the developing mouse hippocampal CA1 region and found that the hippocampal pyramidal neurons generated near the ventricle became postmitotic multipolar cells and accumulated in the multipolar cell accumulation zone (MAZ) in the late stage of development. The hippocampal neurons passed through the pyramidal layer by a unique mode of migration. Their leading processes were highly branched and made contact with many radial fibers. Time-lapse imaging revealed that the migrating cells changed their scaffolds from the original radial fibers to other radial fibers, and as a result they proceed in a zigzag manner, with long intervals. The migrating cells in the hippocampus reminded us of "rock climbers" that instead of using their hands to pull up their bodies were using their leading processes to pull up their cell bodies. Because this mode of migration had never been described, we called it the "climbing" mode. The change from the "climbing" mode in the hippocampus to the "locomotion" mode in the neocortex may have contributed to the brain expansion during evolution.


Asunto(s)
Movimiento Celular/fisiología , Neurogénesis/fisiología , Células Piramidales/citología , Células Piramidales/embriología , Animales , Hipocampo/citología , Hipocampo/embriología , Ratones , Imagen de Lapso de Tiempo
18.
Amino Acids ; 42(6): 2293-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21735222

RESUMEN

Humanin (HN), a peptide of 24 amino acid residues, suppresses the neuronal cell death that is induced by the gene products of Alzheimer's disease. HN contains two Ser residues at positions 7 and 14. Because the proportion of D-Ser isomerized from L-Ser in proteins appears to increase as cellular organs age, we explored the structural effects of the isomerization of each Ser residue in HN. By using a thioflavin-T assay to detect fibril formation, we found that an HN derivative that contained two isomerized D-Ser residues had a greater tendency to form fibrils than did wild-type HN or HNs containing single D-Ser residues. A previous report showed that HN containing two D-Ser residues exerts neuroprotective activity. Our data, therefore, suggest that the fibril formation by HN that contains two D-Ser residues may promote HN neuroprotective activity.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/química , Fármacos Neuroprotectores/química , Enfermedad de Alzheimer/metabolismo , Secuencia de Aminoácidos , Benzotiazoles , Dicroismo Circular , Rojo Congo , Humanos , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Soluciones , Estereoisomerismo , Relación Estructura-Actividad , Tiazoles
19.
FEBS J ; 278(10): 1651-61, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21385318

RESUMEN

The kinase noncatalytic C-lobe domain (KIND) is a putative protein-protein interaction module. Four KIND-containing proteins, Spir-2 (actin-nuclear factor), PTPN13 (protein tyrosine phosphatase), FRMPD2 (scaffold protein) and very-KIND (v-KIND) (brain-specific Ras guanine nucleotide exchange factor), have been identified to date. Uniquely, v-KIND has two KINDs (i.e. KIND1 and KIND2), whereas the other three proteins have only one. The functional role of KIND, however, remains unclear. We previously demonstrated that v-KIND interacts with the high-molecular weight microtubule-associated protein 2 (MAP2), a dendritic microtubule-associated protein, leading to negative regulation of neuronal dendrite growth. In the present study, we analyzed the structure-function relationships of the v-KIND-MAP2 interaction by generating a series of mutant constructs. The interaction with endogenous MAP2 in mouse cerebellar granule cells was specific to v-KIND KIND2, but not KIND1, and was not observed for the KINDs from other KIND-containing proteins. The binding core modules critical for the v-KIND-MAP2 interaction were defined within 32 residues of the mouse v-KIND KIND2 and 43 residues of the mouse MAP2 central domain. Three Leu residues at amino acid positions 461, 474 and 477 in the MAP2-binding core module of KIND2 contributed to the interaction. The MAP2-binding core module itself promoted dendrite branching as a dominant-negative regulator of v-KIND in hippocampal neurons. The results reported in the present study demonstrate the structural and functional determinant underlying the v-KIND-MAP2 interaction that controls dendrite arborization patterns.


Asunto(s)
Dendritas/fisiología , Hipocampo/citología , Proteínas Asociadas a Microtúbulos/metabolismo , Estructura Terciaria de Proteína , Factores de Intercambio de Guanina Nucleótido ras/metabolismo , Secuencia de Aminoácidos , Animales , Pollos , Humanos , Leucina/química , Ratones , Proteínas Asociadas a Microtúbulos/química , Neuronas/fisiología , Mapeo de Interacción de Proteínas , Proteína Tirosina Fosfatasa no Receptora Tipo 13/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/química
20.
Neurochem Res ; 36(7): 1241-52, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21243430

RESUMEN

The cerebellar cortical circuit of mammals develops via a series of magnificent cellular events in the postnatal stage of development to accomplish the formation of functional circuit architectures. The contribution of genetic factors is thought to be crucial to cerebellar development. Therefore, it is essential to analyze the underlying transcriptome during development to understand the genetic blueprint of the cerebellar cortical circuit. In this review, we introduce the profiling of large numbers of spatiotemporal gene expression data obtained by developmental time-series microarray analyses and in situ hybridization cellular mRNA mapping, and the creation of a neuroinformatics database called the Cerebellar Development Transcriptome Database. Using this database, we have identified thousands of genes that are classified into various functional categories and are expressed coincidently with related cellular developmental stages. We have also suggested the molecular mechanisms of cerebellar development by functional characterization of several identified genes (Cupidin, p130Cas, very-KIND, CAPS2) responsible for distinct cellular events of developing cerebellar granule cells. Taken together, the gene expression profiling during the cerebellar development demonstrates that the development of cerebellar cortical circuit is attributed to the complex but orchestrated transcriptome.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Animales , Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/fisiología , Proteínas Portadoras/genética , Clonación Molecular , Proteína Sustrato Asociada a CrK/genética , Proteína Sustrato Asociada a CrK/fisiología , Bases de Datos Genéticas , Exonucleasas , Perfilación de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas de Andamiaje Homer , Glicoproteínas de Membrana/fisiología , Ratones , Proteínas de la Mielina/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Tirosina Quinasas/fisiología , Células de Purkinje/fisiología , Sinapsis/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA