Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
bioRxiv ; 2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37546796

RESUMEN

Dendritic spines are structural correlates of excitatory synapses maintaining stable synaptic communications. However, this strong spine-synapse relationship was mainly characterized in excitatory pyramidal neurons (PyNs), raising a possibility that inferring synaptic density from dendritic spine number may not be universally applied to all neuronal types. Here we found that the ectopic expression of H-Ras increased dendritic spine numbers regardless of cortical cell types such as layer 2/3 pyramidal neurons (PyNs), parvalbumin (PV)- and vasoactive intestinal peptide (VIP)-positive interneurons (INs) in the primary motor cortex (M1). The probability of detecting dendritic spines was positively correlated with the magnitude of H-Ras activity, suggesting elevated local H-Ras activity is involved in the process of dendritic spine formation. H-Ras overexpression caused high spine turnover rate via adding more spines rather than eliminating them. Two-photon photolysis of glutamate triggered de novo dendritic spine formation in mature neurons, suggesting H-Ras induced spine formation is not restricted to the early development. In PyNs and PV-INs, but not VIP-INs, we observed a shift in average spine neck length towards longer filopodia-like phenotypes. The portion of dendritic spines lacking key excitatory synaptic proteins were significantly increased in H-Ras transfected neurons, suggesting that these increased spines have other distinct functions. High spine density caused by H-Ras did not result in change in the frequency or the amplitude of miniature excitatory postsynaptic currents (mEPSCs). Thus, our results propose that dendritic spines possess more multifaceted functions beyond the morphological proxy of excitatory synapse.

3.
eNeuro ; 10(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37253589

RESUMEN

Synaptic modification in postnatal development is essential for the maturation of neural networks. Developmental maturation of excitatory synapses occurs at the loci of dendritic spines that are dynamically regulated by growth and pruning. Striatal spiny projection neurons (SPNs) receive excitatory input from the cerebral cortex and thalamus. SPNs of the striatonigral direct pathway (dSPNs) and SPNs of the striatopallidal indirect pathway (iSPNs) have different developmental roots and functions. The spatial and temporal dynamics of dendritic spine maturation of these two types of SPNs remain elusive. Here, we delineate the developmental trajectories of dendritic spines of dSPNs and iSPNs in the caudoputamen and nucleus accumbens (NAc). We labeled dendritic spines of SPNs by microinjecting Cre-dependent AAV-eYFP viruses into newborn Drd1-Cre or Adora2a-Cre mice, and analyzed spinogenesis at three levels, including different SPN cell types, subregions and postnatal times. In the dorsolateral striatum, spine pruning of dSPNs and iSPNs occurred at postnatal day (P)30-P50. In the dorsomedial striatum, the spine density of both dSPNs and iSPNs reached its peak between P30 and P50, and spine pruning occurred after P30 and P50, respectively, for dSPNs and iSPNs. In the NAc shell, spines of dSPNs and iSPNs were pruned after P21-P30, but no significant pruning was observed in iSPNs of lateral NAc shell. In the NAc core, the spine density of dSPNs and iSPNs reached its peak at P21 and P30, respectively, and subsequently declined. Collectively, the developmental maturation of dendritic spines in dSPNs and iSPNs follows distinct spatiotemporal trajectories in the dorsal and ventral striatum.


Asunto(s)
Espinas Dendríticas , Núcleo Accumbens , Ratones , Animales , Ratones Transgénicos , Cuerpo Estriado/metabolismo , Neuronas/fisiología
4.
J Biol Chem ; 296: 100730, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33933448

RESUMEN

Proper dendrite morphogenesis and synapse formation are essential for neuronal development and function. Dasm1, a member of the immunoglobulin superfamily, is known to promote dendrite outgrowth and excitatory synapse maturation in vitro. However, the in vivo function of Dasm1 in neuronal development and the underlying mechanisms are not well understood. To learn more, Dasm1 knockout mice were constructed and employed to confirm that Dasm1 regulates dendrite arborization and spine formation in vivo. We performed a yeast two-hybrid screen using Dasm1, revealing MRCKß as a putative partner; additional lines of evidence confirmed this interaction and identified cytoplasmic proline-rich region (823-947 aa) of Dasm1 and MRCKß self-activated kinase domain (CC1, 410-744 aa) as necessary and sufficient for binding. Using co-immunoprecipitation assay, autophosphorylation assay, and BS3 cross-linking assay, we show that Dasm1 binding triggers a change in MRCKß's conformation and subsequent dimerization, resulting in autophosphorylation and activation. Activated MRCKß in turn phosphorylates a class 2 regulatory myosin light chain, which leads to enhanced actin rearrangement, causing the dendrite outgrowth and spine formation observed before. Removal of Dasm1 in mice leads to behavioral abnormalities. Together, these results reveal a crucial molecular pathway mediating cell surface and intracellular signaling communication to regulate actin dynamics and neuronal development in the mammalian brain.


Asunto(s)
Actinas/metabolismo , Dendritas/metabolismo , Inmunoglobulinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Espinas Dendríticas/metabolismo , Inmunoglobulinas/química , Ratones , Proteínas del Tejido Nervioso/química , Unión Proteica , Dominios Proteicos
5.
J Cell Sci ; 134(4)2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33536244

RESUMEN

Morphological remodeling of dendritic spines is critically involved in memory formation and depends on adhesion molecules. Serotonin receptors are also implicated in this remodeling, though the underlying mechanisms remain enigmatic. Here, we uncovered a signaling pathway involving the adhesion molecule L1CAM (L1) and serotonin receptor 5-HT4 (5-HT4R, encoded by HTR4). Using Förster resonance energy transfer (FRET) imaging, we demonstrated a physical interaction between 5-HT4R and L1, and found that 5-HT4R-L1 heterodimerization facilitates mitogen-activated protein kinase activation in a Gs-dependent manner. We also found that 5-HT4R-L1-mediated signaling is involved in G13-dependent modulation of cofilin-1 activity. In hippocampal neurons in vitro, the 5-HT4R-L1 pathway triggers maturation of dendritic spines. Thus, the 5-HT4R-L1 signaling module represents a previously unknown molecular pathway regulating synaptic remodeling.


Asunto(s)
Molécula L1 de Adhesión de Célula Nerviosa , Hipocampo , Molécula L1 de Adhesión de Célula Nerviosa/genética , Neuronas , Serotonina , Transducción de Señal
6.
Acta Neuropathol Commun ; 8(1): 185, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33168105

RESUMEN

Abnormal synaptic formation and signaling is one of the key molecular features of autism spectrum disorders (ASD). Cortactin binding protein 2 (CTTNBP2), an ASD-linked gene, is known to regulate the subcellular distribution of synaptic proteins, such as cortactin, thereby controlling dendritic spine formation and maintenance. However, it remains unclear how ASD-linked mutations of CTTNBP2 influence its function. Here, using cultured hippocampal neurons and knockin mouse models, we screen seven ASD-linked mutations in the short form of the Cttnbp2 gene and identify that M120I, R533* and D570Y mutations impair CTTNBP2 protein-protein interactions via divergent mechanisms to reduce dendritic spine density in neurons. R533* mutation impairs CTTNBP2 interaction with cortactin due to lack of the C-terminal proline-rich domain. Through an N-C terminal interaction, M120I mutation at the N-terminal region of CTTNBP2 also negatively influences cortactin interaction. D570Y mutation increases the association of CTTNBP2 with microtubule, resulting in a dendritic localization of CTTNBP2, consequently reducing the distribution of CTTNBP2 in dendritic spines and impairing the synaptic function of CTTNBP2. Finally, we generated heterozygous M120I knockin mice to mimic the genetic variation of patients and found they exhibit reduced social interaction. Our study elucidates that different ASD-linked mutations of CTTNBP2 result in diverse molecular deficits, but all have the similar consequence of synaptic impairment.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas del Citoesqueleto/genética , Espinas Dendríticas/metabolismo , Proteínas de Microfilamentos/genética , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Conducta Social , Interacción Social , Sinapsis/metabolismo , Animales , Trastorno del Espectro Autista/patología , Trastorno del Espectro Autista/fisiopatología , Células Cultivadas , Espinas Dendríticas/patología , Técnicas de Sustitución del Gen , Hipocampo/citología , Ratones , Plasticidad Neuronal/genética , Neuronas/patología , Ratas , Sinapsis/patología
7.
Cell Rep ; 31(13): 107835, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32610136

RESUMEN

Neurofibromatosis type 1 (NF1) is a dominant genetic disorder manifesting, in part, as cognitive defects. Previous study indicated that neurofibromin (NF1 protein) interacts with valosin-containing protein (VCP)/P97 to control dendritic spine formation, but the mechanism is unknown. Here, using Nf1+/- mice and transgenic mice overexpressing wild-type Vcp/p97, we demonstrate that neurofibromin acts with VCP to control endoplasmic reticulum (ER) formation and consequent protein synthesis and regulates dendritic spine formation, thereby modulating contextual fear memory and social interaction. To validate the role of protein synthesis, we perform leucine supplementation in vitro and in vivo. Our results suggest that leucine can effectively enter the brain and increase protein synthesis and dendritic spine density of Nf1+/- neurons. Contextual memory and social behavior of Nf1+/- mice are also restored by leucine supplementation. Our study suggests that the "ER-protein synthesis" pathway downstream of neurofibromin and VCP is a critical regulator of dendritic spinogenesis and brain function.


Asunto(s)
Miedo/fisiología , Leucina/administración & dosificación , Memoria/fisiología , Neurofibromina 1/metabolismo , Biosíntesis de Proteínas , Conducta Social , Proteína que Contiene Valosina/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Células Cultivadas , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Suplementos Dietéticos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Ratones Mutantes , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Proteoma/metabolismo , Sirolimus/farmacología , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
8.
Cell Rep ; 31(9): 107700, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32492416

RESUMEN

Synaptic dysregulation is a critical feature of autism spectrum disorders (ASDs). Among various autism-associated genes, cortactin binding protein 2 (CTTNBP2) is a cytoskeleton regulator predominantly expressed in neurons and highly enriched at dendritic spines. Here, using Cttnbp2 knockout and ASD-linked mutant mice, we demonstrate that Cttnbp2 deficiency reduces zinc levels in the brain, alters synaptic protein targeting, impairs dendritic spine formation and ultrastructure of postsynaptic density, and influences neuronal activation and autism-like behaviors. A link to autism, the NMDAR-SHANK pathway, and zinc-related regulation are three features shared by CTTNBP2-regulated synaptic proteins. Zinc supplementation rescues the synaptic expression of CTTNBP2-regulated proteins. Moreover, zinc supplementation and administration of D-cycloserine, an NMDAR coagonist, improve the social behaviors of Cttnbp2-deficient mice. We suggest that CTTNBP2 controls the synaptic expression of a set of zinc-regulated autism-associated genes and influences NMDAR function and signaling, providing an example of how genetic and environmental factor crosstalk controls social behaviors.


Asunto(s)
Espinas Dendríticas/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Zinc/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Proteínas de Unión a Calmodulina/genética , Proteínas de Unión a Calmodulina/metabolismo , Cicloserina/farmacología , Espinas Dendríticas/ultraestructura , Suplementos Dietéticos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Conducta Social , Zinc/farmacología , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo
9.
Growth Horm IGF Res ; 50: 42-47, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31862540

RESUMEN

OBJECTIVE: Growth hormone (GH) is widely known for its peripheral effects during growth and development. However, numerous reports also suggest that GH exert pro-cognitive, restorative, and protective properties in the brain. In in vitro studies, the detection of dendritic spines, small protrusions extending from axons, can act as a marker for cognition-related function as spine formation is considered to be associated with learning and memory. Here we show that an acute 24-hour treatment of GH can increase dendritic spine density in primary hippocampal cell cultures. DESIGN: Primary hippocampal cells were harvested from embryonic Wistar rats and cultured for 14 days. Cells were treated with supra-physiological doses of GH (10-1000 nM) and subjected to a high-throughput screening protocol. Images were acquired and analyzed using automated image analysis and the number of spines, spines per neurite length, neurite length, and mean area of spines, was reported. RESULTS: GH treatment (1000 nM) increased the number of dendritic spines by 83% and spines per neurite length by 82% when compared to control. For comparison BDNF, a known inducer of spine densities, produced statistically non-significant increase in this setting. CONCLUSION: The results was found significant using the highest supra-physiological dose of GH, and the present study further confirms a potential role of the hormone in the treatment of cognitive dysfunction.


Asunto(s)
Espinas Dendríticas/efectos de los fármacos , Hormona del Crecimiento/farmacología , Hipocampo/citología , Neuritas/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Ensayos Analíticos de Alto Rendimiento , Técnicas In Vitro , Cultivo Primario de Células , Ratas
10.
Behav Brain Res ; 372: 112023, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31212238

RESUMEN

Epidemiological and clinical studies suggest that early-life stress (ELS) may lead to the development of mental disorders in adulthood. Maternal separation (MS) is a valid animal model of ELS that produces detrimental effects on brain and behavior of experimental animals. Positive environmental stimuli have been shown to counteract the behavioral deficits of ELS and enhance neuroplasticity. Recent data indicate that music may serve as a form of environmental enrichment in experimental animals. However, the underlying mechanisms through which musical enrichment exerts its effects are poorly understood. Male Sprague-Dawley rats were submitted to a 3 h MS protocol during postnatal days (PND) 2-14, while another group was left undisturbed. Half of the animals within each group were exposed from PND 21 to PND 76 for 12 h/day to Mozart K. 448. At approximately three months of age, elevated plus maze procedure, forced swim test and social approach task were applied to test whether music exposure can mitigate the effects of MS stress on animal emotional behaviors. Moreover, we investigated the effects of these treatments on dendritic spine density in the CA1 region of hippocampus. As expected, MS rats showed decreased sociability, increased anxiety- and depressive-like behaviors, and decreased mature dendritic spines in the CA1 region of hippocampus in adulthood. Musical enrichment reversed these effects. Our results suggest that musical enrichment can reverse the negative effects of MS on anxiety, depression, and sociability in adult rats and modulate neuronal plasticity and provide additional data for a new therapeutic intervention to rescue emotional symptoms.


Asunto(s)
Ansiedad/terapia , Depresión/terapia , Música/psicología , Animales , Animales Recién Nacidos , Trastornos de Ansiedad/terapia , Conducta Animal/fisiología , Encéfalo/efectos de los fármacos , Espinas Dendríticas , Trastorno Depresivo/terapia , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Femenino , Hipocampo/efectos de los fármacos , Masculino , Privación Materna , Neurogénesis/efectos de los fármacos , Plasticidad Neuronal/fisiología , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/metabolismo
11.
Eur J Neurosci ; 50(4): 2722-2739, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31001844

RESUMEN

Cylindromatosis tumor suppressor protein (CYLD) was initially identified as a tumor suppressor deubiquitylating protein in familial cylindromatosis patients. Proteomic analyses using rodent brain samples revealed enrichment of CYLD in purified postsynaptic density fractions. Here, we report that CYLD regulates dendritic growth and postsynaptic differentiation in mouse hippocampal neurons. CYLD showed diffuse localization in rapidly growing dendrites, but was gradually concentrated in spines. Overexpression and knockdown of CYLD in the early stage of cultured neurons demonstrated that CYLD positively regulated dendritic growth. Phenotypes in dendritic morphogenesis induced by CYLD overexpression and knockdown could be reversed by manipulation of the critical acetylation site of α-tubulin, suggesting tubulin acetylation is a downstream pathway of CYLD-dependent dendritic growth. Overexpression and knockdown of CYLD in the later stage of cultured neurons revealed that CYLD promoted formation of postsynaptic spines. Influence of CYLD on spines was not affected by co-expression of acetylation mutant forms of α-tubulin, indicating that CYLD regulates dendritic growth and spine formation through different molecular mechanisms. Analyses with the truncated and mutated forms of CYLD demonstrated that the first microtubule-binding domain of CYLD was critical for spine formation. These results suggest important roles of CYLD in sequential promotion of dendritic growth and postsynaptic spine maturation.


Asunto(s)
Dendritas/ultraestructura , Espinas Dendríticas/ultraestructura , Enzima Desubiquitinante CYLD/genética , Enzima Desubiquitinante CYLD/fisiología , Morfogénesis/genética , Acetilación , Animales , Diferenciación Celular , Células Cultivadas , Técnicas de Silenciamiento del Gen , Hipocampo/ultraestructura , Ratones , Microtúbulos/metabolismo , Cultivo Primario de Células , Unión Proteica , Tubulina (Proteína)/metabolismo
12.
Neurochem Res ; 44(5): 1243-1251, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30875016

RESUMEN

A majority of excitatory synapses in the brain are localized on the dendritic spines. Alterations of spine density and morphology are associated with many neurological diseases. Understanding the molecular mechanisms underlying spine formation is important for understanding these diseases. Kalirin7 (Kal-7) is localized to the postsynaptic side of excitatory synapses in the neurons. Overexpression of Kal-7 causes an increase in spine density whereas knockdown expression of endogenous Kal-7 results in a decrease in spine density in primary cultured cortical neurons. However, the mechanisms underlying Kal-7-mediated spine formation are not entirely clear. Cyclin-dependent kinase 5 (Cdk5) plays a vital role in the formation of spines and synaptic plasticity. Kal-7 is phosphorylated by CDK5 at Thr1590, the unique Cdk5 phosphorylation site in the Kal-7 protein. This study was to explore the role of CDK5-mediated phosphorylation of Kal-7 in spine formation and the underlying mechanisms. Our results showed expression of Kal-7T/D (mimicked phosphorylation), Kal-7T/A mutants (blocked phosphorylation) or wild-type (Wt) Kal-7 caused in a similar increase in spine density, while spine size of Wt Kal-7-expressing cortical neurons was bigger than that in Kal-7 T\A-expressing neurons, but smaller than that in Kal-7T/D-expressing neurons. The fluorescence intensity of NMDA receptor subunit NR2B (GluN2B) staining was stronger along the MAP2 positive dendrites of Kal-7T/D-expressing neurons than that in Kal-7T/A- or Wt Kal-7-expressing neurons. The fluorescence intensity of AMPA receptor subunit GluR1 (GluA1) staining showed the same trend as GluN2B staining. These findings suggest that Cdk5 affects the function of Kal-7 on spine morphology and function via GluN2B and GluA1 receptors during dendritic spine formation.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Dendritas/metabolismo , Espinas Dendríticas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Animales , Hipocampo/metabolismo , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Sinapsis/metabolismo
13.
EBioMedicine ; 30: 62-73, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29580842

RESUMEN

Abnormalities in limbic neural circuits have been implicated in the onset of anxiety disorders. However, the molecular pathogenesis underlying anxiety disorders remains poorly elucidated. Here, we demonstrate that myristoylated alanine-rich C-kinase substrate like 1 (MARCKSL1) regulates amygdala circuitry to control the activity of the hypothalamic-pituitary-adrenal (HPA) axis, as well as induces anxiety-like behaviors in mice. MARCKSL1 expression was predominantly localized in the prefrontal cortex (PFC), hypothalamus, hippocampus, and amygdala of the adult mouse brain. MARCKSL1 transgenic (Tg) mice exhibited anxiety-like behaviors dependent on corticotropin-releasing hormone. MARCKSL1 increased spine formation in the central amygdala, and downregulation of MARCKSL1 in the amygdala normalized both increased HPA axis activity and elevated anxiety-like behaviors in Tg mice. Furthermore, MARCKSL1 expression was increased in the PFC and amygdala in a brain injury model associated with anxiety-like behaviors. Our findings suggest that MARCKSL1 expression in the amygdala plays an important role in anxiety-like behaviors.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/metabolismo , Ansiedad/patología , Espinas Dendríticas/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Envejecimiento/metabolismo , Amígdala del Cerebelo/patología , Animales , Conducta Animal , Proteínas de Unión a Calmodulina , Hormona Liberadora de Corticotropina/biosíntesis , Regulación hacia Abajo , Emociones , Técnicas de Silenciamiento del Gen , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos , Núcleo Hipotalámico Paraventricular/metabolismo , Regulación hacia Arriba
14.
J Biomed Sci ; 25(1): 2, 2018 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-29310658

RESUMEN

The endoplasmic reticulum (ER) is the biggest organelle in cells and is involved in versatile cellular processes. Formation and maintenance of ER morphology are regulated by a series of proteins controlling membrane fusion and curvature. At least six different ER morphology regulators have been demonstrated to be involved in neurological disorders-including Valosin-containing protein (VCP), Atlastin-1 (ATL1), Spastin (SPAST), Reticulon 2 (RTN2), Receptor expression enhancing protein 1 (REEP1) and RAB10-suggesting a critical role of ER formation in neuronal activity and function. Among these genes, mutations in VCP gene involve in inclusion body myopathy with Paget disease of bone and frontotemporal dementia (IBMPFD), familial amyotrophic lateral sclerosis (ALS), autism spectrum disorders (ASD), and hereditary spastic paraplegia (HSP). ATL1 is also one of causative genes of HSP. RAB10 is associated with Parkinson's disease (PD). A recent study showed that VCP and ATL1 work together to regulate dendritic spine formation by controlling ER formation and consequent protein synthesis efficiency. RAB10 shares the same function with VCP and ATL1 to control ER formation and protein synthesis efficiency but acts independently. Increased protein synthesis by adding extra leucine to cultured neurons ameliorated dendritic spine deficits caused by VCP and ATL1 deficiencies, strengthening the significance of protein synthesis in VCP- and ATL1-regulated dendritic spine formation. These findings provide new insight into the roles of ER and protein synthesis in controlling dendritic spine formation and suggest a potential etiology of neurodegenerative disorders caused by mutations in VCP, ATL1 and other genes encoding proteins regulating ER formation and morphogenesis.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas de Unión al GTP/genética , Proteínas de la Membrana/genética , Enfermedades del Sistema Nervioso/genética , Proteína que Contiene Valosina/genética , Proteínas de Unión al GTP/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Enfermedades del Sistema Nervioso/fisiopatología , Biosíntesis de Proteínas , Proteína que Contiene Valosina/metabolismo
15.
Neuron ; 94(2): 304-311.e4, 2017 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-28426965

RESUMEN

Dendritic spines are the major transmitter reception compartments of glutamatergic synapses in most principal neurons of the mammalian brain and play a key role in the function of nerve cell circuits. The formation of functional spine synapses is thought to be critically dependent on presynaptic glutamatergic signaling. By analyzing CA1 pyramidal neurons in mutant hippocampal slice cultures that are essentially devoid of presynaptic transmitter release, we demonstrate that the formation and maintenance of dendrites and functional spines are independent of synaptic glutamate release.


Asunto(s)
Calcio/metabolismo , Dendritas/metabolismo , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Sinapsis/metabolismo , Animales , Espinas Dendríticas/metabolismo , Ratones , Transducción de Señal/fisiología , Sinapsis/fisiología
16.
Neurosci Bull ; 33(2): 143-152, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28258509

RESUMEN

Autism spectrum disorder (ASD) encompasses a complex set of developmental neurological disorders, characterized by deficits in social communication and excessive repetitive behaviors. In recent years, ASD is increasingly being considered as a disease of the synapse. One main type of genetic aberration leading to ASD is gene duplication, and several mouse models have been generated mimicking these mutations. Here, we studied the effects of MECP2 duplication and human chromosome 15q11-13 duplication on synaptic development and neural circuit wiring in the mouse sensory cortices. We showed that mice carrying MECP2 duplication had specific defects in spine pruning, while the 15q11-13 duplication mouse model had impaired spine formation. Our results demonstrate that spine pathology varies significantly between autism models and that distinct aspects of neural circuit development may be targeted in different ASD mutations. Our results further underscore the importance of gene dosage in normal development and function of the brain.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/patología , Espinas Dendríticas/ultraestructura , Duplicación de Gen/genética , Proteína 2 de Unión a Metil-CpG/genética , Corteza Somatosensorial/patología , Sinapsis/ultraestructura , Factores de Edad , Análisis de Varianza , Animales , Cromosomas Humanos Par 15 , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tinción con Nitrato de Plata
17.
Front Mol Neurosci ; 10: 27, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28223918

RESUMEN

Alzheimer's disease (AD) is a heterogeneous neurodegenerative disease. Recent studies employing microRNA-seq and genome-wide sequencing have identified some non-coding RNAs that are influentially involved in AD pathogenesis. Non-coding RNAs can compete with other endogenous RNAs by microRNA response elements (MREs) and manipulate biological processes, such as tumorigenesis. However, only a few non-coding RNAs have been reported in the pathogenesis of AD. In this study, we constructed the first competing endogenous RNA (ceRNA) network leveraging whole transcriptome sequencing and a previously studied microRNA-seq of APPswe/PS1ΔE9 transgenic mice. The underlying mechanisms for the involvement of ceRNA in AD were validated using the Dual Luciferase Reporter Assay, detection of transcription levels by quantitative RT-PCR and translation levels by Western blotting, and morphological examination in primary cultured neurons. In the ceRNA network, four lncRNAs (C030034L19Rik, Rpph1, A830012C17Rik, and Gm15477) and five miRNAs (miR-182-5p, miR-330-5p, miR-326-3p, miR-132-3p, and miR-484) are enriched in nine pathways and an AD-related gene pool. Among them, Ribonuclease P RNA component H1 (Rpph1) is upregulated in the cortex of APPswe/PS1ΔE9 mice compared to wild type controls. Rpph1 binds to miR326-3p/miR-330-5p and causes the release of their downstream target Cdc42, which leads to CDC42 upregulation. This effect was disrupted upon mutation of the MRE on Rpph1. Moreover, overexpression of Rpph1 increased dendritic spine density in primary cultured hippocampal pyramidal neurons, whereas knocking down of Rpph1 had the reverse effect. In conclusion, Rpph1 modulates CDC42 expression level in a ceRNA-dependent manner, which may represent a compensatory mechanism in the early stage of the AD pathogenesis.

18.
J Neurochem ; 141(2): 179-194, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28178750

RESUMEN

Maternal separation (MS) is known to affect hippocampal function such as learning and memory, yet the molecular mechanism remains unknown. We hypothesized that these impairments are attributed to abnormities of neural circuit formation by MS, and focused on brain-derived neurotrophic factor (BDNF) as key factor because BDNF signaling has an essential role in synapse formation during early brain development. Using rat offspring exposed to MS for 6 h/day during postnatal days (PD) 2-20, we estimated BDNF signaling in the hippocampus during brain development. Our results show that MS attenuated BDNF expression and activation of extracellular signal-regulated kinase (ERK) around PD 7. Moreover, plasticity-related immediate early genes, which are transcriptionally regulated by BDNF-ERK signaling, were also reduced by MS around PD 7. Interestingly, detailed analysis revealed that MS particularly reduced expression of BDNF gene and immediate early genes in the cornu ammonis 1 (CA1) of hippocampus at PD 7. Considering that BDNF-ERK signaling is involved in spine formation, we next evaluated spine formation in the hippocampus during the weaning period. Our results show that MS particularly reduced mature spine density in proximal apical dendrites of CA1 pyramidal neurons at PD 21. These results suggest that MS could attenuate BDNF-ERK signaling during primary synaptogenesis with a region-specific manner, which is likely to lead to decreased spine formation and maturation observed in the hippocampal CA1 region. It is speculated that this incomplete spine formation during early brain development has an influence on learning capabilities throughout adulthood.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Espinas Dendríticas/metabolismo , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Privación Materna , Animales , Animales Recién Nacidos , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Espinas Dendríticas/patología , Femenino , Hipocampo/patología , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
19.
EMBO Rep ; 18(1): 169-183, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27979975

RESUMEN

Viral infection during fetal or neonatal stages increases the risk of developing neuropsychiatric disorders such as schizophrenia and autism spectrum disorders. Although neurons express several key regulators of innate immunity, the role of neuronal innate immunity in psychiatric disorders is still unclear. Using cultured neurons and in vivo mouse brain studies, we show here that Toll-like receptor 3 (TLR3) acts through myeloid differentiation primary response gene 88 (MYD88) to negatively control Disrupted in schizophrenia 1 (Disc1) expression, resulting in impairment of neuronal development. Cytokines are not involved in TLR3-mediated inhibition of dendrite outgrowth. Instead, TLR3 signaling suppresses expression of several psychiatric disorder-related genes, including Disc1 The impaired dendritic arborization caused by TLR3 activation is rescued by MYD88 deficiency or DISC1 overexpression. In addition, TLR3 activation at the neonatal stage increases dendritic spine density, but narrows spine heads at postnatal day 21 (P21), suggesting a long-lasting effect of TLR3 activation on spinogenesis. Our study reveals a novel mechanism of TLR3 in regulation of dendritic morphology and provides an explanation for how environmental factors influence mental health.


Asunto(s)
Regulación de la Expresión Génica , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Esquizofrenia/genética , Esquizofrenia/metabolismo , Receptor Toll-Like 3/metabolismo , Animales , Biomarcadores , Línea Celular , Células Cultivadas , Citocinas/metabolismo , Dendritas/genética , Dendritas/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Noqueados , Morfogénesis/genética , Factor 88 de Diferenciación Mieloide/genética , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Receptor Toll-Like 3/genética
20.
J Neurosci ; 34(9): 3429-42, 2014 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-24573299

RESUMEN

We previously identified SPARC-related protein-containing immunoglobulin domains 1 (SPIG1, also known as Follistatin-like protein 4) as one of the dorsal-retina-specific molecules expressed in the developing chick retina. We here demonstrated that the knockdown of SPIG1 in the retinal ganglion cells (RGCs) of developing chick embryos induced the robust ectopic branching of dorsal RGC axons and failed to form a tight terminal zone at the proper position on the tectum. The knockdown of SPIG1 in RGCs also led to enhanced axon branching in vitro. However, this was canceled by the addition of a neutralizing antibody against brain-derived neurotrophic factor (BDNF) to the culture medium. SPIG1 and BDNF were colocalized in vesicle-like structures in cells. SPIG1 bound with the proform of BDNF (proBDNF) but very weakly with mature BDNF in vitro. The expression and secretion of mature BDNF were significantly decreased when SPIG1 was exogenously expressed with BDNF in HEK293T or PC12 cells. The amount of mature BDNF proteins as well as the tyrosine phosphorylation level of the BDNF receptor, tropomyosin-related kinase B (TrkB), in the hippocampus were significantly higher in SPIG1-knockout mice than in wild-type mice. Here the spine density of CA1 pyramidal neurons was consistently increased. Together, these results suggest that SPIG1 negatively regulated BDNF maturation by binding to proBDNF, thereby suppressing axonal branching and spine formation.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Ganglionares de la Retina/metabolismo , Aminoácidos/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Proteínas de Unión al Calcio/genética , Células Cultivadas , Embrión de Pollo , Proteínas de la Matriz Extracelular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Unión Proteica/genética , Ratas , Retina/citología , Retina/embriología , Retina/crecimiento & desarrollo , Células Ganglionares de la Retina/ultraestructura , Transducción de Señal/genética , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA