Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Nature ; 590(7847): 612-617, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33361813

RESUMO

In the adult hippocampus, synapses are constantly formed and eliminated1,2. However, the exact function of synapse elimination in the adult brain, and how it is regulated, are largely unknown. Here we show that astrocytic phagocytosis3 is important for maintaining proper hippocampal synaptic connectivity and plasticity. By using fluorescent phagocytosis reporters, we find that excitatory and inhibitory synapses are eliminated by glial phagocytosis in the CA1 region of the adult mouse hippocampus. Unexpectedly, we found that astrocytes have a major role in the neuronal activity-dependent elimination of excitatory synapses. Furthermore, mice in which astrocytes lack the phagocytic receptor MEGF10 show a reduction in the elimination of excitatory synapses; as a result, excessive but functionally impaired synapses accumulate. Finally, Megf10-knockout mice show defective long-term synaptic plasticity and impaired formation of hippocampal memories. Together, our data provide strong evidence that astrocytes eliminate unnecessary excitatory synaptic connections in the adult hippocampus through MEGF10, and that this astrocytic function is crucial for maintaining circuit connectivity and thereby supporting cognitive function.


Assuntos
Envelhecimento , Astrócitos/citologia , Região CA1 Hipocampal/citologia , Homeostase , Vias Neurais , Fagocitose , Sinapses/metabolismo , Animais , Potenciais Pós-Sinápticos Excitadores , Feminino , Potenciais Pós-Sinápticos Inibidores , Masculino , Proteínas de Membrana/metabolismo , Memória/fisiologia , Camundongos , Plasticidade Neuronal/fisiologia
2.
FASEB J ; 34(5): 6965-6983, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32237183

RESUMO

Microtubule-associated protein (MAP) 2 has been perceived as a static cytoskeletal protein enriched in neuronal dendritic shafts. Emerging evidence indicates dynamic functions for various MAPs in activity-dependent synaptic plasticity. However, it is unclear how MAP2 is associated with synaptic plasticity mechanisms. Here, we demonstrate that specific silencing of high-molecular-weight MAP2 in vivo abolished induction of long-term potentiation (LTP) in the Schaffer collateral pathway of CA1 pyramidal neurons and in vitro blocked LTP-induced surface delivery of AMPA receptors and spine enlargement. In mature hippocampal neurons, we observed rapid translocation of a subpopulation of MAP2, present in dendritic shafts, to spines following LTP stimulation. Time-lapse confocal imaging showed that spine translocation of MAP2 was coupled with LTP-induced spine enlargement. Consistently, immunogold electron microscopy revealed that LTP stimulation of the Schaffer collateral pathway promoted MAP2 labeling in spine heads of CA1 neurons. This translocation depended on NMDA receptor activation and Ras-MAPK signaling. Furthermore, LTP stimulation led to an increase in surface-expressed AMPA receptors specifically in the neurons with MAP2 spine translocation. Altogether, this study indicates a novel role for MAP2 in LTP mechanisms and suggests that MAP2 participates in activity-dependent synaptic plasticity in mature hippocampal networks.


Assuntos
Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Potenciação de Longa Duração/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Células Piramidais/metabolismo , Animais , Células Cultivadas , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/ultraestrutura , Técnicas In Vitro , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos Endogâmicos C57BL , Microscopia Imunoeletrônica , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Plasticidade Neuronal/fisiologia , Transporte Proteico , Células Piramidais/ultraestrutura , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Receptores de AMPA/metabolismo
3.
Learn Mem ; 26(9): 299-306, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31416903

RESUMO

Storage of long-term memory requires not only protein synthesis but also protein degradation. In this article, we overview recent publications related to this issue, stressing that the balanced actions of protein synthesis and degradation are critical for long-term memory formation. We particularly focused on the brain-derived neurotrophic factor signaling that leads to protein synthesis; proteasome- and autophagy-dependent protein degradation that removes molecular constraints; the role of Fragile X mental retardation protein in translational suppression; and epigenetic modifications that control gene expression at the genomic level. Numerous studies suggest that an imbalance between protein synthesis and degradation leads to intellectual impairment and cognitive disorders.


Assuntos
Encéfalo/fisiologia , Memória de Longo Prazo/fisiologia , Biossíntese de Proteínas , Proteólise , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Epigênese Genética , Humanos , Plasticidade Neuronal , Transdução de Sinais
4.
J Neurochem ; 147(5): 595-608, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30125942

RESUMO

Guanine nucleotide exchange factors (GEFs) play important roles in many cellular processes, including regulation of the structural plasticity of dendritic spines. A GEF protein, adenomatous polyposis coli-stimulated GEF 1 (Asef1, ARHGEF4) is highly expressed in the nervous system. However, the function of Asef1 has not been investigated in neurons. Here, we present evidence showing that Asef1 negatively regulates the synaptic localization of postsynaptic density protein 95 (PSD-95) in the excitatory synapse by inhibiting Staufen-mediated synaptic localization of PSD-95. Accordingly, Asef1 expression impairs synaptic transmission in hippocampal cultured neurons. In addition, neuronal activity facilitates the dissociation of Asef1 from Staufen in a phosphoinositide 3 kinase (PI3K)-dependent manner. Taken together, our data reveal Asef1 functions as a negative regulator of synaptic localization of PSD-95 and synaptic transmission.


Assuntos
Adenosina Trifosfatases/fisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Fosfoproteínas/fisiologia , Sinapses/fisiologia , Adenosina Trifosfatases/genética , Animais , Dendritos/fisiologia , Dendritos/ultraestrutura , Proteína 4 Homóloga a Disks-Large/biossíntese , Proteína 4 Homóloga a Disks-Large/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Hipocampo/citologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Técnicas de Patch-Clamp , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia , Ratos , Transmissão Sináptica/fisiologia
5.
Nat Rev Neurosci ; 14(1): 7-23, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23254191

RESUMO

Brain-derived neurotrophic factor (BDNF)--a member of a small family of secreted proteins that includes nerve growth factor, neurotrophin 3 and neurotrophin 4--has emerged as a key regulator of neural circuit development and function. The expression, secretion and actions of BDNF are directly controlled by neural activity, and secreted BDNF is capable of mediating many activity-dependent processes in the mammalian brain, including neuronal differentiation and growth, synapse formation and plasticity, and higher cognitive functions. This Review summarizes some of the recent progress in understanding the cellular and molecular mechanisms underlying neurotrophin regulation of neural circuits. The focus of the article is on BDNF, as this is the most widely expressed and studied neurotrophin in the mammalian brain.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Encéfalo , Rede Nervosa/fisiologia , Neurônios/metabolismo , Animais , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Humanos , Sinapses/fisiologia
6.
Neurobiol Dis ; 62: 62-72, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24055772

RESUMO

Alzheimer's disease (AD) is among the most prevalent forms of dementia affecting the aging population, and pharmacological therapies to date have not been successful in preventing disease progression. Future therapeutic efforts may benefit from the development of models that enable basic investigation of early disease pathology. In particular, disease-relevant models based on human pluripotent stem cells (hPSCs) may be promising approaches to assess the impact of neurotoxic agents in AD on specific neuronal populations and thereby facilitate the development of novel interventions to avert early disease mechanisms. We implemented an efficient paradigm to convert hPSCs into enriched populations of cortical glutamatergic neurons emerging from dorsal forebrain neural progenitors, aided by modulating Sonic hedgehog (Shh) signaling. Since AD is generally known to be toxic to glutamatergic circuits, we exposed glutamatergic neurons derived from hESCs to an oligomeric pre-fibrillar forms of Aß known as "globulomers", which have shown strong correlation with the level of cognitive deficits in AD. Administration of such Aß oligomers yielded signs of the disease, including cell culture age-dependent binding of Aß and cell death in the glutamatergic populations. Furthermore, consistent with previous findings in postmortem human AD brain, Aß-induced toxicity was selective for glutamatergic rather than GABAeric neurons present in our cultures. This in vitro model of cortical glutamatergic neurons thus offers a system for future mechanistic investigation and therapeutic development for AD pathology using human cell types specifically affected by this disease.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Ácido Glutâmico/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Células-Tronco Pluripotentes/citologia , Fatores Etários , Peptídeos beta-Amiloides/metabolismo , Animais , Morte Celular , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Feminino , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/patologia , Neurônios GABAérgicos/fisiologia , Proteínas Hedgehog/metabolismo , Humanos , Neurônios/patologia , Ratos Endogâmicos F344
7.
Biol Psychiatry Glob Open Sci ; 4(4): 100315, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38726036

RESUMO

Background: Fear responses significantly affect daily life and shape our approach to uncertainty. However, the potential resurgence of fear in unfamiliar situations poses a significant challenge to exposure-based therapies for maladaptive fear responses. Nonetheless, how novel contextual stimuli are associated with the relapse of extinguished fear remains unknown. Methods: Using a context-dependent fear renewal model, the functional circuits and underlying mechanisms of the posterior parietal cortex (PPC) and anterior cingulate cortex (ACC) were investigated using optogenetic, histological, in vivo, and ex vivo electrophysiological and pharmacological techniques. Results: We demonstrated that the PPC-to-ACC pathway governs fear relapse in a novel context. We observed enhanced populational calcium activity in the ACC neurons that received projections from the PPC and increased synaptic activity in the basolateral amygdala-projecting PPC-to-ACC neurons upon renewal in a novel context, where excitatory postsynaptic currents amplitudes increased but inhibitory postsynaptic current amplitudes decreased. In addition, we found that parvalbumin-expressing interneurons controlled novel context-dependent fear renewal, which was blocked by the chronic administration of fluoxetine. Conclusions: Our findings highlight the PPC-to-ACC pathway in mediating the relapse of extinguished fear in novel contexts, thereby contributing significant insights into the intricate neural mechanisms that govern fear renewal.


To improve outcomes for exposure-based therapy, it is vital to understand the renewal of fear after extinction in new environments. Using optogenetics and other techniques, Joo et al. found that a brain circuit connecting the posterior parietal cortex (PPC) to the anterior cingulate cortex (ACC) is crucial for the return of fear memories in mice exposed to a novel context. Certain PPC→ACC neuron types and their connections to the amygdala became more active during fear renewal in a novel context, and inhibiting parvalbumin-expressing interneurons reduced this fear response. This study provides insights into the brain mechanisms underlying the reappearance of fear in unfamiliar situations.

8.
Sci Adv ; 10(7): eadk0639, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38354231

RESUMO

We investigate how matrix stiffness regulates chromatin reorganization and cell reprogramming and find that matrix stiffness acts as a biphasic regulator of epigenetic state and fibroblast-to-neuron conversion efficiency, maximized at an intermediate stiffness of 20 kPa. ATAC sequencing analysis shows the same trend of chromatin accessibility to neuronal genes at these stiffness levels. Concurrently, we observe peak levels of histone acetylation and histone acetyltransferase (HAT) activity in the nucleus on 20 kPa matrices, and inhibiting HAT activity abolishes matrix stiffness effects. G-actin and cofilin, the cotransporters shuttling HAT into the nucleus, rises with decreasing matrix stiffness; however, reduced importin-9 on soft matrices limits nuclear transport. These two factors result in a biphasic regulation of HAT transport into nucleus, which is directly demonstrated on matrices with dynamically tunable stiffness. Our findings unravel a mechanism of the mechano-epigenetic regulation that is valuable for cell engineering in disease modeling and regenerative medicine applications.


Assuntos
Reprogramação Celular , Cromatina , Cromatina/genética , Reprogramação Celular/genética , Fibroblastos , Epigênese Genética
9.
BMB Rep ; 56(2): 172-177, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36593105

RESUMO

BEST family is a class of Ca2+-activated Cl- channels evolutionary well conserved from bacteria to human. The human BEST paralogs (BEST1-BEST4) share significant amino acid sequence homology in the N-terminal region, which forms the transmembrane helicases and contains the direct calcium-binding site, Ca2+-clasp. But the cytosolic C-terminal region is less conserved in the paralogs. Interestingly, this domain-specific sequence conservation is also found in the BEST1 orthologs. However, the functional role of the C-terminal region in the BEST channels is still poorly understood. Thus, we aimed to understand the functional role of the C-terminal region in the human and mouse BEST1 channels by using electrophysiological recordings. We found that the calcium-dependent activation of BEST1 channels can be modulated by the C-terminal region. The C-terminal deletion hBEST1 reduced the Ca2+-dependent current activation and the hBEST1-mBEST1 chimera showed a significantly reduced calcium sensitivity to hBEST1 in the HEK293 cells. And the C-terminal domain could regulate cellular expression and plasma membrane targeting of BEST1 channels. Our results can provide a basis for understanding the C-terminal roles in the structure-function of BEST family proteins. [BMB Reports 2023; 56(3): 172-177].


Assuntos
Cálcio , Proteínas do Olho , Humanos , Animais , Camundongos , Bestrofinas/metabolismo , Cálcio/metabolismo , Proteínas do Olho/metabolismo , Células HEK293 , Membrana Celular/metabolismo
10.
Adv Sci (Weinh) ; 10(24): e2300152, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37357983

RESUMO

The role of transcription factors and biomolecules in cell type conversion has been widely studied. Yet, it remains unclear whether and how intracellular mechanotransduction through focal adhesions (FAs) and the cytoskeleton regulates the epigenetic state and cell reprogramming. Here, it is shown that cytoskeletal structures and the mechanical properties of cells are modulated during the early phase of induced neuronal (iN) reprogramming, with an increase in actin cytoskeleton assembly induced by Ascl1 transgene. The reduction of actin cytoskeletal tension or cell adhesion at the early phase of reprogramming suppresses the expression of mesenchymal genes, promotes a more open chromatin structure, and significantly enhances the efficiency of iN conversion. Specifically, reduction of intracellular tension or cell adhesion not only modulates global epigenetic marks, but also decreases DNA methylation and heterochromatin marks and increases euchromatin marks at the promoter of neuronal genes, thus enhancing the accessibility for gene activation. Finally, micro- and nano-topographic surfaces that reduce cell adhesions enhance iN reprogramming. These novel findings suggest that the actin cytoskeleton and FAs play an important role in epigenetic regulation for cell fate determination, which may lead to novel engineering approaches for cell reprogramming.


Assuntos
Reprogramação Celular , Epigênese Genética , Adesão Celular , Mecanotransdução Celular , Cromatina
11.
Proc Natl Acad Sci U S A ; 106(34): 14634-9, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19706550

RESUMO

Serotonin (5-HT) plays a critical role in modulating synaptic plasticity in the marine mollusc Aplysia and in the mammalian nervous system. In Aplysia sensory neurons, 5-HT can activate several signal cascades, including PKA and PKC, presumably via distinct types of G protein-coupled receptors. However, the molecular identities of these receptors have not yet been identified. We here report the cloning and functional characterization of a 5-HT receptor that is positively coupled to adenylyl cyclase in Aplysia neurons. The cloned receptor, 5-HT(apAC1), stimulates the production of cAMP in HEK293T cells and in Xenopus oocytes. Moreover, the knockdown of 5-HT(apAC1) expression by RNA interference blocked 5-HT-induced cAMP production in Aplysia sensory neurons and blocked synaptic facilitation in nondepressed or partially depressed sensory-to-motor neuron synapses. These data implicate 5-HT(apAC1) as a major modulator of learning related synaptic facilitation in the direct sensory to motor neuron pathway of the gill withdrawal reflex.


Assuntos
Adenilil Ciclases/metabolismo , Aplysia/fisiologia , Pareamento Cromossômico/fisiologia , Aprendizagem/fisiologia , Receptores de Serotonina/fisiologia , Adenilil Ciclases/genética , Sequência de Aminoácidos , Animais , Aplysia/citologia , Aplysia/genética , Western Blotting , Linhagem Celular , Células Cultivadas , Clonagem Molecular , AMP Cíclico/metabolismo , Feminino , Humanos , Hibridização In Situ , Potenciais da Membrana , Dados de Sequência Molecular , Oócitos/metabolismo , Oócitos/fisiologia , Filogenia , Receptores de Serotonina/classificação , Receptores de Serotonina/genética , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Serotonina/farmacologia , Transfecção , Xenopus laevis
12.
Learn Mem ; 17(9): 469-79, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20813835

RESUMO

Phosphodiesterases (PDEs) are known to play a key role in the compartmentalization of cAMP signaling; however, the molecular mechanisms underlying intracellular localization of different PDE isoforms are not understood. In this study, we have found that each of the supershort, short, and long forms of apPDE4 showed distinct localization in the cytoplasm, plasma membrane, and both plasma membrane and presynaptic terminals, respectively. The N-terminal 20 amino acids of the long form of apPDE4 were involved in presynaptic terminal targeting by binding to several lipids. In addition, the N terminus of the short form of apPDE4 bound to several lipids including phosphoinositols, thereby targeting the plasma membrane. Overexpression of the long and the short forms, but not the supershort form attenuated 5-HT-induced membrane hyperexcitability. Finally, the knockdown of apPDE4s in sensory neurons impaired both short-term and long-term facilitation. Thus, these results suggest that apPDE4s can participate in the regulation of cAMP signaling through specific subcellular localization by means of lipid binding activities.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/química , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Potenciais da Membrana/genética , Células Receptoras Sensoriais/fisiologia , Sequência de Aminoácidos , Animais , Aplysia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Cisteína/metabolismo , Gânglios dos Invertebrados/citologia , Proteínas de Fluorescência Verde/genética , Humanos , Imunoprecipitação/métodos , Lipídeos de Membrana/metabolismo , Mutação/genética , Terminações Pré-Sinápticas/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estrutura Terciária de Proteína , Interferência de RNA/fisiologia , Células Receptoras Sensoriais/efeitos dos fármacos , Serotonina/farmacologia , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Sinaptofisina/metabolismo , Transfecção/métodos
13.
Sci Rep ; 11(1): 21203, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34707216

RESUMO

Brain-derived neurotrophic factor (BDNF) regulates diverse brain functions via TrkB receptor signaling. Due to the expression of TrkB receptors, astrocytes can internalize extracellular BDNF proteins via receptor-mediated endocytosis. Endocytosed BDNF can be re-secreted upon stimulation, but the molecular mechanism underlying this phenomenon remains unrecognized. Our study reveals that vesicle-associated membrane protein 3 (Vamp3) selectively regulates the release of endocytic BDNF from astrocytes. By using quantum dot (QD)-conjugated mature BDNF (QD-BDNF) as a proxy for the extracellular BDNF protein, we monitored the uptake, transport, and secretion of BDNF from cultured cortical astrocytes. Our data showed that endocytic QD-BDNF particles were enriched in Vamp3-containing vesicles in astrocytes and that ATP treatment sufficiently triggered either the antero- or retrograde transport and exocytosis of QD-BDNF-containing vesicles. Downregulation of Vamp3 expression disrupted endocytic BDNF secretion from astrocytes but did not affect uptake or transport. Collectively, these results provide evidence of the selective ability of astrocytic Vamp3 to control endocytic BDNF secretion during BDNF recycling.


Assuntos
Astrócitos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Exocitose , Proteína 3 Associada à Membrana da Vesícula/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Endocitose , Camundongos , Camundongos Endogâmicos C57BL , Pontos Quânticos , Proteína 3 Associada à Membrana da Vesícula/genética
14.
J Neurosci ; 29(41): 13063-73, 2009 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-19828819

RESUMO

In mammalian brain, neurons and astrocytes are reported to express various chloride and anion channels, but the evidence for functional expression of Ca(2+)-activated anion channel (CAAC) and its molecular identity have been lacking. Here we report electrophysiological evidence for the CAAC expression and its molecular identity by mouse Bestrophin 1 (mBest1) in astrocytes of the mouse brain. Using Ca(2+) imaging and perforated-patch-clamp analysis, we demonstrate that astrocytes displayed an inward current at holding potential of -70 mV that was dependent on an increase in intracellular Ca(2+) after G(alphaq)-coupled receptor activation. This current was mediated mostly by anions and was sensitive to well known anion channel blockers such as niflumic acid, 5-nitro-2(3-phenylpropylamino)-benzoic acid, and flufenamic acid. To find the molecular identity of the anion channel responsible for the CAAC current, we analyzed the expression of candidate genes and found that the mRNA for mouse mBest1 is predominantly expressed in acutely dissociated or cultured astrocytes. Whole-cell patch-clamp analysis using HEK293T cells heterologously expressing full-length mBest1 showed a Ca(2+)-dependent current mediated by mBest1, with a complete impairment of the current by a putative pore mutation, W93C. Furthermore, mBest1-mediated CAAC from cultured astrocytes was significantly reduced by expression of mBest1-specific short hairpin RNA (shRNA), suggesting that the CAAC is mediated by a channel encoded by mBest1. Finally, hippocampal CA1 astrocytes in hippocampal slice also showed mBest1-mediated CAAC because it was inhibited by mBest1-specific shRNA. Collectively, these data provide molecular evidence that the mBest1 channel is responsible for CAAC function in astrocytes.


Assuntos
Astrócitos/fisiologia , Cálcio/metabolismo , Canais de Cloreto/fisiologia , Proteínas do Olho/metabolismo , Hipocampo/citologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Bestrofinas , Bradicinina/farmacologia , Células Cultivadas , Córtex Cerebral/citologia , Quelantes/farmacologia , Canais de Cloreto/genética , Dinoprostona/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Estimulação Elétrica/métodos , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Regulação da Expressão Gênica/genética , Humanos , Técnicas In Vitro , Canais Iônicos , Lisofosfolipídeos/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Oligopeptídeos/farmacologia , Técnicas de Patch-Clamp , Pirrolidinonas/farmacologia , RNA Interferente Pequeno/genética , Tapsigargina/farmacologia , Tionucleotídeos/farmacologia , Transfecção/métodos
15.
Exp Neurobiol ; 29(3): 219-229, 2020 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-32624506

RESUMO

Understanding brain function-related neural circuit connectivity is essential for investigating how cognitive functions are decoded in neural circuits. Trans-synaptic viral vectors are useful for identifying neural synaptic connectivity because of their ability to be transferred from transduced cells to synaptically connected cells. However, concurrent labeling of multisynaptic inputs to postsynaptic neurons is impossible with currently available trans-synaptic viral vectors. Here, we report a neural circuit tracing system that can simultaneously label postsynaptic neurons with two different markers, the expression of which is defined by presynaptic input connectivity. This system, called "cFork (see fork)", includes delivering serotype 1-packaged AAV vectors (AAV1s) containing Cre or flippase recombinase (FlpO) into two different presynaptic brain areas, and AAV5 with a dual gene expression cassette in postsynaptic neurons. Our in vitro and in vivo tests showed that selective expression of two different fluorescence proteins, EGFP and mScarlet, in postsynaptic neurons could be achieved by AAV1-mediated anterograde trans-synaptic transfer of Cre or FlpO constructs. When this tracing system was applied to the somatosensory barrel field cortex (S1BF) or striatum innervated by multiple presynaptic inputs, postsynaptic neurons defined by presynaptic inputs were simultaneously labeled with EGFP or mScarlet. Our new anterograde tracing tool may be useful for elucidating the complex multisynaptic connectivity of postsynaptic neurons regulating diverse brain functions.

16.
Cell Signal ; 20(4): 714-25, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18221855

RESUMO

The present study was undertaken to characterize neuronal activity-dependent expression and release of vascular endothelial growth factor (VEGF) from rat hippocampal neurons and its contribution to neuronal functions. Increased levels of VEGF164 mRNA were evident both in cultured neurons and slices, but not astrocytes, following membrane depolarization with KCl. Activity-dependent expression of VEGF, as well as its release, was dependent on the activation of the N-methyl-d-aspartate receptors or L-type voltage-activated calcium channels. A brief (10 min) application of recombinant VEGF165 to neurons elicited a slow rise in cytosolic Ca2+ in a VEGFR2 dependent manner. The VEGF-induced Ca2+ responses required Ca2+ influx, phospholipase Cgamma and Ca2+ stores. An inhibitor of transient receptor potential canonical channels reduced the VEGF-induced Ca2+ responses by 50%, suggesting the involvement of transient receptor potential canonical channels in the VEGF-mediated responses. The same brief stimulus with VEGF led to long-term synaptic enhancement dependent on protein synthesis. VEGF had prominent effects on the activation calcium/calmodulin protein kinase II and cAMP responsive element binding protein as well as extracellular signal-regulated protein kinase and mammalian target of rapamycin-all in a VEGFR2 dependent manner. Our findings suggest that VEGF released from neuronal cells plays a local role in Ca2+ influx and synaptic transmission that may influence the generation of long-term changes in synaptic efficacy.


Assuntos
Sinalização do Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteínas de Transporte/metabolismo , Hipocampo/metabolismo , Plasticidade Neuronal , Neurônios/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Quinases/metabolismo , Transmissão Sináptica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Astrócitos/metabolismo , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/embriologia , Hipocampo/enzimologia , Potenciais da Membrana , Proteínas do Tecido Nervoso/biossíntese , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Fosfolipase C gama/metabolismo , Cloreto de Potássio/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Recombinantes/metabolismo , Serina-Treonina Quinases TOR , Fatores de Tempo , Transcrição Gênica , Canais de Potencial de Receptor Transitório/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
Mol Brain ; 12(1): 97, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31753031

RESUMO

Postsynaptic density protein 95 (PSD-95) is a pivotal postsynaptic scaffolding protein in excitatory neurons. Although the transport and regulation of PSD-95 in synaptic regions is well understood, dendritic transport of PSD-95 before synaptic localization still remains to be clarified. To evaluate the role of KIF5, conventional kinesin, in the dendritic transport of PSD-95 protein, we expressed a transport defective form of KIF5A (ΔMD) that does not contain the N-terminal motor domain. Expression of ΔMD significantly decreased PSD-95 level in the dendrites. Consistently, KIF5 was associated with PSD-95 in in vitro and in vivo assays. This interaction was mediated by the C-terminal tail regions of KIF5A and the third PDZ domain of PSD-95. Additionally, the ADPDZ3 (the association domain of NMDA receptor and PDZ3 domain) expression significantly reduced the levels of PSD-95, glutamate receptor 1 (GluA1) in dendrites. The association between PSD-95 and KIF5A was dose-dependent on Staufen protein, suggesting that the Staufen plays a role as a regulatory role in the association. Taken together, our data suggest a new mechanism for dendritic transport of the AMPA receptor-PSD-95.


Assuntos
Dendritos/metabolismo , Proteína 4 Homóloga a Disks-Large/metabolismo , Cinesinas/metabolismo , Animais , Proteína 4 Homóloga a Disks-Large/química , Células HEK293 , Humanos , Cinesinas/química , Camundongos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Domínios PDZ , Ligação Proteica , Transporte Proteico , Proteínas de Ligação a RNA/metabolismo , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo
18.
Exp Neurobiol ; 28(2): 183-215, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31138989

RESUMO

In the brain, a reduction in extracellular osmolality causes water-influx and swelling, which subsequently triggers Cl-- and osmolytes-efflux via volume-regulated anion channel (VRAC). Although LRRC8 family has been recently proposed as the pore-forming VRAC which is activated by low cytoplasmic ionic strength but not by swelling, the molecular identity of the pore-forming swelling-dependent VRAC (VRACswell) remains unclear. Here we identify and characterize Tweety-homologs (TTYH1, TTYH2, TTYH3) as the major VRACswell in astrocytes. Gene-silencing of all Ttyh1/2/3 eliminated hypo-osmotic-solution-induced Cl- conductance (ICl,swell) in cultured and hippocampal astrocytes. When heterologously expressed in HEK293T or CHO-K1 cells, each TTYH isoform showed a significant ICl,swell with similar aquaporin-4 dependency, pharmacological properties and glutamate permeability as ICl,swell observed in native astrocytes. Mutagenesis-based structure-activity analysis revealed that positively charged arginine residue at 165 in TTYH1 and 164 in TTYH2 is critical for the formation of the channel-pore. Our results demonstrate that TTYH family confers the bona fide VRACswell in the brain.

19.
Mol Cells ; 26(1): 87-92, 2008 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-18547981

RESUMO

We employed dual color Fluorescence Cross Correlation Spectroscopy (FCCS) to measure the interaction between PKA regulatory (RII) and catalytic subunits (CAT) in living cells. Elevation of intracellular cAMP with forskolin decreased the cross-correlation amplitude between RFP-fused RII (RII-mRFP) and GFP-fused CAT (CAT-EGFP) by 50%, indicating that cAMP elevation leads to dissociation of RII-CAT complexes. Moreover, diffusion coefficient analysis showed that the diffusion rate of CAT-EGFP was significantly increased, suggesting that the decreased RII-CAT association caused by cAMP generated free CAT subunits. Our study demonstrates that in vivo FCCS measurements and their quantitative analysis permit one not only to directly quantify protein-protein interactions but also to estimate changes in the intracellular cAMP concentration.


Assuntos
Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Espectrometria de Fluorescência , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico/genética , Células HeLa , Humanos , Rim/citologia , Rim/metabolismo , Plasmídeos , Subunidades Proteicas
20.
Exp Neurobiol ; 27(3): 217-225, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30022873

RESUMO

Deficient BDNF signaling is known to be involved in neurodegenerative diseases such as Huntington's disease (HD). Mutant huntingtin (mhtt)-mediated disruption of either BDNF transcription or transport is thought to be a factor contributing to striatal atrophy in the HD brain. Whether and how activity-dependent BDNF secretion is affected by the mhtt remains unclear. In the present study, I provide evidence for differential effects of the mhtt on cortical BDNF secretion in the striatum during HD progression. By two-photon imaging of fluorescent BDNF sensor (BDNF-pHluorin and -EGFP) in acute striatal slices of HD knock-in model mice, I found deficient cortical BDNF secretion regardless of the HD onset, but antisense oligonucleotide (ASO)-mediated reduction of htts only rescues BDNF secretion in the early HD brain before the disease onset. Although secretion modes of individual BDNF-containing vesicle were not altered in the pre-symptomatic brain, the full-fusion and partial-fusion modes of BDNF-containing vesicles were significantly altered after the onset of HD symptoms. Thus, besides abnormal BDNF transcription and transport, our results suggest that mhtt-mediated alteration in activity-dependent BDNF secretion at corticostriatal synapses also contributes to the development of HD.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA