Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(16): e2315958121, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38588427

RESUMO

The ability of neurons to rapidly remodel their synaptic structure and strength in response to neuronal activity is highly conserved across species and crucial for complex brain functions. However, mechanisms required to elicit and coordinate the acute, activity-dependent structural changes across synapses are not well understood, as neurodevelopment and structural plasticity are tightly linked. Here, using an RNAi screen in Drosophila against genes affecting nervous system functions in humans, we uncouple cellular processes important for synaptic plasticity and synapse development. We find mutations associated with neurodegenerative and mental health disorders are 2-times more likely to affect activity-induced synaptic remodeling than synapse development. We report that while both synapse development and activity-induced synaptic remodeling at the fly NMJ require macroautophagy (hereafter referred to as autophagy), bifurcation in the autophagy pathway differentially impacts development and synaptic plasticity. We demonstrate that neuronal activity enhances autophagy activation but diminishes degradative autophagy, thereby driving the pathway towards autophagy-based secretion. Presynaptic knockdown of Snap29, Sec22, or Rab8, proteins implicated in the secretory autophagy pathway, is sufficient to abolish activity-induced synaptic remodeling. This study uncovers secretory autophagy as a transsynaptic signaling mechanism modulating synaptic plasticity.


Assuntos
Proteínas de Drosophila , Junção Neuromuscular , Animais , Humanos , Junção Neuromuscular/metabolismo , Sinapses/metabolismo , Drosophila/fisiologia , Neurônios/metabolismo , Autofagia/genética , Plasticidade Neuronal/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Transmissão Sináptica/fisiologia , GTP Fosfo-Hidrolases/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(46)2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34772802

RESUMO

Change is ubiquitous in living beings. In particular, the connectome and neural representations can change. Nevertheless, behaviors and memories often persist over long times. In a standard model, associative memories are represented by assemblies of strongly interconnected neurons. For faithful storage these assemblies are assumed to consist of the same neurons over time. Here we propose a contrasting memory model with complete temporal remodeling of assemblies, based on experimentally observed changes of synapses and neural representations. The assemblies drift freely as noisy autonomous network activity and spontaneous synaptic turnover induce neuron exchange. The gradual exchange allows activity-dependent and homeostatic plasticity to conserve the representational structure and keep inputs, outputs, and assemblies consistent. This leads to persistent memory. Our findings explain recent experimental results on temporal evolution of fear memory representations and suggest that memory systems need to be understood in their completeness as individual parts may constantly change.


Assuntos
Memória/fisiologia , Neurônios/fisiologia , Animais , Homeostase/fisiologia , Modelos Neurológicos , Redes Neurais de Computação , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia
3.
Zhongguo Zhong Yao Za Zhi ; 49(9): 2489-2500, 2024 May.
Artigo em Chinês | MEDLINE | ID: mdl-38812153

RESUMO

This study aims to reveal the molecular mechanism of Chaijin Jieyu Anshen Tablets(CJJYAS) in regulating the abnormal anterior cingulate cortex(ACC)-ventral hippocampus(vHPC) glutaminergic neural circuit to alleviate synaptic remodeling of ventral hippocampal neurons in depressed rats. Firstly, the study used chemogenetics to localize glutaminergic adeno-associated virus(AAV) into the ACC brain region of rats. The model of depressed rats was established by chronic unpredictable mild stress(CUMS) combined with independent feeding. The rats were randomly divided into control group, model group, AAV empty group, AAV group, AAV+ glucocorticoid receptors(GR) blocker group, AAV+chemokine receptor 1(CX3CR1) blocker group, and AAV+CJJYAS group. Depressive-like behaviors of rats were evaluated by open-field, forced-swimming, and Morris water maze tests, combined with an animal behavior analysis system. The morphological and structural changes of ACC and vHPC neurons in rats were observed by hematoxylin-eosin(HE) staining. Immunofluorescence and nuclear phosphoprotein(c-Fos) were used to detect glutaminergic neural circuit activation of ACC-vHPC in rats. The changes in dendrites, synaptic spines, and synaptic submicrostructure of vHPC neurons were observed by Golgi staining and transmission electron microscopy, respectively. The expressions of synaptic remodeling-related proteins N-methyl-D-asprtate receptor 2A(GRIN2A), N-methyl-D-asprtate receptor 2B(GRIN2B), Ca~(2+)/calmodulin-dependent protein kinase Ⅱ(CaMKⅡ), mitogen-activated protein kinase-activated protein kinase 2(MK2), and a ubiquitous actin-binding protein(cofilin) in vHPC glutaminergic neurons of rats were detected by immunofluorescence and Western blot, respectively. The results indicated that the activated glutaminergic AAV aggravated the depressive-like behaviors phenotype of rats in the model group and deteriorated the damage of morphology and structure of ACC and vHPC neurons and synaptic ultrastructure. However, both GR and CX3CR1 bloc-kers could reverse the abnormal changes to varying degrees, suggesting that the abnormal activation of ACC-vHPC glutaminergic neural circuit mediated by GR/CX3CR1 signals in gliocytes in the ACC brain region may be closely related to the occurrence and development of depression. Interestingly, CJJYAS significantly inhibited the activation of the ACC-vHPC glutaminergic neural circuit induced by AAV and the elevated Glu level. Furthermore, CJJYAS could also effectively reverse the aggravation of depressive-like behaviors and synaptic remodeling of vHPC neurons of rats in the model group induced by the activated AAV. Additionally, the findings suggested that the molecular mechanism of CJJYAS in improving synaptic damage of vHPC neurons might be related to the regulation of synaptic remodeling-related signals such as NR/CaMKⅡ and MK2/cofilin. In conclusion, this research confirms that CJJYAS effectively regulates the abnormal ACC-vHPC glutaminergic neural circuit and alleviates the synaptic remodeling of vHPC glutaminergic neurons in depressed rats, and the molecular mechanism might be associated with the regulation of synapse-related NR/CaMKⅡ and MK2/cofilin signaling pathways, which may be the crucial mechanism of its antidepressant effect.


Assuntos
Depressão , Medicamentos de Ervas Chinesas , Giro do Cíngulo , Hipocampo , Neurônios , Ratos Sprague-Dawley , Animais , Ratos , Masculino , Neurônios/metabolismo , Hipocampo/metabolismo , Depressão/metabolismo , Depressão/fisiopatologia , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/administração & dosagem , Giro do Cíngulo/metabolismo , Giro do Cíngulo/fisiopatologia , Sinapses/metabolismo , Plasticidade Neuronal , Humanos
4.
Brain Behav Immun ; 110: 245-259, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36906076

RESUMO

Remodeling of synapses by microglia is essential for synaptic plasticity in the brain. However, during neuroinflammation and neurodegenerative diseases, microglia can induce excessive synaptic loss, although the precise underlying mechanisms are unknown. To directly observe microglia-synapse interactions under inflammatory conditions, we performed in vivo two-photon time-lapse imaging of microglia-synapse interactions after bacterial lipopolysaccharide administration to model systemic inflammation, or after inoculation of Alzheimer's disease (AD) brain extracts to model disease-associated neuroinflammatory microglial response. Both treatments prolonged microglia-neuron contacts, decreased basal surveillance of synapses and promoted synaptic remodeling in response to synaptic stress induced by focal single-synapse photodamage. Spine elimination correlated with the expression of microglial complement system/phagocytic proteins and the occurrence of synaptic filopodia. Microglia were observed contacting spines, then stretching and phagocytosing spine head filopodia. Thus, in response to inflammatory stimuli microglia exacerbated spine remodeling through prolonged microglial contact and elimination of spines 'tagged' by synaptic filopodia.


Assuntos
Doença de Alzheimer , Tauopatias , Humanos , Microglia/metabolismo , Tauopatias/metabolismo , Doença de Alzheimer/metabolismo , Sinapses/metabolismo , Inflamação/metabolismo
5.
Cell Mol Neurobiol ; 42(8): 2505-2525, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34460037

RESUMO

Microglial activation is considered as the critical pathogenic event in diverse central nervous system disorders including cerebral ischemia. Proinflammatory responses of activated microglia have been well reported in the ischemic brain and neuroinflammatory responses of activated microglia have been believed to be the potential therapeutic strategy. However, despite having proinflammatory roles, microglia can have significant anti-inflammatory roles and they are associated with the production of growth factors which are responsible for neuroprotection and recovery after ischemic injury. Microglia can directly promote neuroprotection by preventing ischemic infarct expansion and promoting functional outcomes. Indirectly, microglia are involved in promoting anti-inflammatory responses, neurogenesis, and angiogenesis in the ischemic brain which are crucial pathophysiological events for ischemic recovery. In fact, anti-inflammatory cytokines and growth factors produced by microglia can promote neuroprotection and attenuate neurobehavioral deficits. In addition, microglia regulate phagocytosis, axonal regeneration, blood-brain barrier protection, white matter integrity, and synaptic remodeling, which are essential for ischemic recovery. Microglia can also regulate crosstalk with neurons and other cell types to promote neuroprotection and ischemic recovery. This review mainly focuses on the roles of microglia in neuroprotection and recovery following ischemic injury. Furthermore, this review also sheds the light on the therapeutic potential of microglia in stroke patients.


Assuntos
Isquemia Encefálica , Microglia , Anti-Inflamatórios/farmacologia , Isquemia Encefálica/metabolismo , Citocinas/metabolismo , Humanos , Microglia/metabolismo , Neuroproteção
6.
J Neurosci ; 40(14): 2828-2848, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32127494

RESUMO

The extraordinary diversity of excitatory synapse sizes is commonly attributed to activity-dependent processes that drive synaptic growth and diminution. Recent studies also point to activity-independent size fluctuations, possibly driven by innate synaptic molecule dynamics, as important generators of size diversity. To examine the contributions of activity-dependent and independent processes to excitatory synapse size diversity, we studied glutamatergic synapse size dynamics and diversification in cultured rat cortical neurons (both sexes), silenced from plating. We found that in networks with no history of activity whatsoever, synaptic size diversity was no less extensive than that observed in spontaneously active networks. Synapses in silenced networks were larger, size distributions were broader, yet these were rightward-skewed and similar in shape when scaled by mean synaptic size. Silencing reduced the magnitude of size fluctuations and weakened constraints on size distributions, yet these were sufficient to explain synaptic size diversity in silenced networks. Model-based exploration followed by experimental testing indicated that silencing-associated changes in innate molecular dynamics and fluctuation characteristics might negatively impact synaptic persistence, resulting in reduced synaptic numbers. This, in turn, would increase synaptic molecule availability, promote synaptic enlargement, and ultimately alter fluctuation characteristics. These findings suggest that activity-independent size fluctuations are sufficient to fully diversify glutamatergic synaptic sizes, with activity-dependent processes primarily setting the scale rather than the shape of size distributions. Moreover, they point to reciprocal relationships between synaptic size fluctuations, size distributions, and synaptic numbers mediated by the innate dynamics of synaptic molecules as they move in, out, and between synapses.SIGNIFICANCE STATEMENT Sizes of glutamatergic synapses vary tremendously, even when formed on the same neuron. This diversity is commonly thought to reflect the outcome of activity-dependent forms of synaptic plasticity, yet activity-independent processes might also play some part. Here we show that in neurons with no history of activity whatsoever, synaptic sizes are no less diverse. We show that this diversity is the product of activity-independent size fluctuations, which are sufficient to generate a full repertoire of synaptic sizes at correct proportions. By combining modeling and experimentation we expose reciprocal relationships between size fluctuations, synaptic sizes and synaptic counts, and show how these phenomena might be connected through the dynamics of synaptic molecules as they move in, out, and between synapses.


Assuntos
Modelos Neurológicos , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Sinapses/fisiologia , Animais , Células Cultivadas , Córtex Cerebral/fisiologia , Feminino , Masculino , Ratos
7.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G1081-G1092, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33949202

RESUMO

Stress can trigger symptoms in patients with irritable bowel syndrome (IBS). Previously we demonstrated that chronic psychological stress induced microglial remodeling in the central nucleus of amygdala (CeA) and contributed to the development of visceral hypersensitivity via synaptic engulfment. However, the specific signaling mechanisms that microglia depend upon to recognize target neurons to facilitate visceral pain remain unknown. Here, we test the hypothesis that the microglia in the CeA contribute to chronic stress-induced visceral hypersensitivity via complement C1q/C3-CR3 signaling-mediated synaptic remodeling. In male and female Fischer-344 rats, micropellets of corticosterone (CORT) or cholesterol (control) were stereotaxically implanted bilaterally onto the CeA. After 7 days, microglial C1q, complement receptor 3 (CR3) expression, and microglia-mediated synaptic engulfment were assessed via RNAscope, quantitative PCR, and immunofluorescence. The microglial inhibitor minocycline, CR3 antagonist neutrophil inhibitory factor (NIF), or vehicle were daily infused into the CeA following CORT implantations. Visceral sensitivity was assessed via a visceromotor response (VMR) to graded pressures of isobaric colorectal distension (CRD). Our results suggest that chronic exposure to elevated CORT in the CeA induced visceral hypersensitivity and amygdala microglial morphological remodeling. CORT increased microglial C1q and CR3 expression and increased microglia-mediated synaptic engulfment. Both groups of animals with minocycline or NIF infusions reversed microglia-mediated synaptic remodeling and attenuated CORT-induced visceral hypersensitivity. Our findings demonstrate that C1q/C3-CR3 signaling is critical for microglia-mediated synaptic remodeling in the CeA and contributes to CORT-induced visceral hypersensitivity.NEW & NOTEWORTHY Patients with irritable bowel syndrome (IBS) show altered amygdala activity. We showed previously that stress induces visceral hypersensitivity partially through microglia-modulated synaptic plasticity in the central nucleus of the amygdala (CeA). Our current data suggest that the C1q/C3-CR3 cascade initiates microglia-mediated synaptic remodeling in the CeA. Blocking C3-CR3 interaction attenuates stress-induced visceral hypersensitivity. These findings uncover a role of microglia-synapse signaling in the brain-gut regulation and support a future therapeutic target to treat visceral pain.


Assuntos
Tonsila do Cerebelo/metabolismo , Complemento C1q/metabolismo , Complemento C3/metabolismo , Microglia/metabolismo , Plasticidade Neuronal/fisiologia , Dor Visceral/metabolismo , Animais , Colo/metabolismo , Corticosterona/sangue , Modelos Animais de Doenças , Feminino , Síndrome do Intestino Irritável/metabolismo , Masculino , Percepção da Dor/fisiologia , Ratos , Ratos Endogâmicos F344
8.
Cell Mol Neurobiol ; 41(8): 1665-1682, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32770528

RESUMO

With the development of neural prostheses, neural plasticity including synaptic remodeling under electrical stimulation is drawing more and more attention. Indeed, intracochlear electrical stimulation used to restore hearing in deaf can induce the loss of residual hearing and synapses of the inner hair cells (IHCs). However, the mechanism under this process is largely unknown. Considering that the guinea pig is always a suitable and convenient choice for the animal model of cochlea implant (CI), in the present study, normal-hearing guinea pigs were implanted with CIs. Four-hour electrical stimulation with the intensity of 6 dB above electrically evoked compound action potential (ECAP) threshold (which can decrease the quantity of IHC synapses and the excitability of the auditory nerve) resulted in the upregulation of Bdnf (p < 0.0001) and downregulation of Nt-3 (p < 0.05). Intracochlear perfusion of exogenous NT-3 or TrkC/Fc (which blocks NT-3) can, respectively, resist or aggravate the synaptic loss induced by electrical stimulation. In contrast, local delivery of exogenous BDNF or TrkB/Fc (which blocks BDNF) to the cochlea, respectively, exacerbated or protected against the synaptic loss caused by electrical stimulation. Notably, the synaptic changes were only observed in the basal and middle halves of the cochlea. All the findings above suggested that NT-3 and BDNF may play opposite roles in the remodeling of IHC synapses induced by intracochlear electrical stimulation, i.e. NT-3 and BDNF promoted the regeneration and degeneration of IHC synapses, respectively.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Orelha Interna/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Neurotrofina 3/biossíntese , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/administração & dosagem , Orelha Interna/efeitos dos fármacos , Estimulação Elétrica/métodos , Cobaias , Masculino , Plasticidade Neuronal/fisiologia , Neurotrofina 3/administração & dosagem
9.
J Neurogenet ; 34(3-4): 307-322, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32808848

RESUMO

Sydney Brenner's choice of Caenorhabditis elegans as a model organism for understanding the nervous system has accelerated discoveries of gene function in neural circuit development and behavior. In this review, we discuss a striking example of synaptic remodeling in the C. elegans motor circuit in which DD class motor neurons effectively reverse polarity as presynaptic and postsynaptic domains at opposite ends of the DD neurite switch locations. Originally revealed by EM reconstruction conducted over 40 years ago, DD remodeling has since been investigated by live cell imaging methods that exploit the power of C. elegans genetics to reveal key effectors of synaptic plasticity. Although synapses are also extensively rewired in developing mammalian circuits, the underlying remodeling mechanisms are largely unknown. Here, we highlight the possibility that studies in C. elegans can reveal pathways that orchestrate synaptic remodeling in more complex organisms. Specifically, we describe (1) transcription factors that regulate DD remodeling, (2) the cellular and molecular cascades that drive synaptic remodeling and (3) examples of circuit modifications in vertebrate neurons that share some similarities with synaptic remodeling in C. elegans DD neurons.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/genética , AMP Cíclico/fisiologia , Espinhas Dendríticas/fisiologia , Espinhas Dendríticas/ultraestrutura , Regulação da Expressão Gênica , Genes Reporter , Microscopia Intravital , Canais Iônicos/genética , Canais Iônicos/fisiologia , Larva , Mamíferos/fisiologia , Microscopia Eletrônica , Microtúbulos/ultraestrutura , Neurônios Motores/fisiologia , Neurônios Motores/ultraestrutura , Proteínas do Tecido Nervoso/fisiologia , Neurônios/ultraestrutura , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/fisiologia , Proteínas Recombinantes/metabolismo , Estrigiformes/fisiologia , Fatores de Transcrição/fisiologia , Ácido gama-Aminobutírico/fisiologia
10.
Hippocampus ; 28(11): 783-795, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30067287

RESUMO

Brain-derived neurotrophic factor (BDNF), a neurotrophin highly expressed in the hippocampus, plays crucial roles in cognition, neuroplasticity, synaptic function, and dendritic remodeling. The common human Val66Met polymorphism of BDNF has been implicated in the pathophysiology of neuropsychiatric and neurodegenerative disorders, and in the outcome of pro-adaptive and therapeutic treatments. Altered gene-expression profile has been previously shown in BDNF Val66Met knock-in mice, which recapitulate the phenotypic hallmarks of individuals carrying the BDNF Met allele. The aim of this study was to investigate the impact of the BDNF Val66Met polymorphism in the knock-in mouse model on two hippocampal epigenetic marks for transcriptional repression and activation, respectively: trimethylation of lysine 27 on histone H3 (H3K27me3) and acetylation of histone H3 (AcH3), using a genome-wide approach. Chromatin immunoprecipitation followed by deep sequencing of immunoprecipitated DNA (ChIP-Seq) was carried out with specific antibodies for H3K27me3 and AcH3. Our results revealed broad alteration of H3K27me3 and AcH3 marks association profiles in BDNFMet/Met , compared to BDNFVal/Val mice. Bioinformatics analysis showed changes in several biological functions and related pathways, affected by the presence of the polymorphism. In particular, a number of networks of functional interaction contained BDNF as central node. Quantitative PCR analysis confirmed epigenetically related significant changes in the expression of five genes: Dvl1, Nos3, Reln, Lypd6, and Sh3gl2. The first three are involved in dendrite and spine remodeling, morphological features altered in BDNFMet/Met mice. This work in homozygous knock-in mice shows that the human BDNF Val66Met polymorphism induces an array of histone H3 epigenetic modifications, in turn altering the expression of select genes crucial for structural and functional neuronal features.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Dendritos/metabolismo , Epigênese Genética , Hipocampo/metabolismo , Polimorfismo Genético , Animais , Biologia Computacional , Técnicas de Introdução de Genes , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Camundongos Transgênicos , Proteína Reelina , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
11.
Cell Physiol Biochem ; 48(3): 919-933, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30036869

RESUMO

BACKGROUND/AIMS: Chronic compression of the spinal cord causes the loss of motor neurons in the anterior horn, but the precise and extensive mechanism for the loss is not completely determined. Therefore, this study aims to explore the role of microRNA-494 (miR-494) in the proliferation of astrocytes and in the synaptic remodeling in the spinal cord of a rat model of chronic spinal cord injury (SCI) by regulating the Nogo/NgR signaling pathway. METHODS: A rat model of chronic, compressive SCI was established, and the spinal cord state, blood supply changes, and astrocyte apoptosis were observed. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blotting were used to detect expression of miR-494 and the Nogo/NgR signaling pathway-related genes. Fluorescence in situ hybridization (FISH) was used for detecting miR-494 expression and distribution. RESULTS: Higher miR-494 expression was accompanied by the inhibition of astrocyte proliferation and synaptic remodeling. In addition, CDK6 could be regulated by miR-494 and was shown to be one of the target genes of miR-494. Positive expression of miR-494 detected by FISH was consistent with the results from RT-qPCR that miR-494 could downregulate CDK6 gene expression. Moreover, the direct miR-494 target CDK6 plays important inhibitory roles in chronic SCI by suppressing the Nogo/ NgR signaling pathway. CONCLUSIONS: The results demonstrated that miR-494 inhibition can promote astrocyte proliferation and synaptic remodeling by suppressing the Nogo/NgR signaling pathway in a rat model of chronic SCI.


Assuntos
MicroRNAs/metabolismo , Animais , Antagomirs/metabolismo , Astrócitos/citologia , Astrócitos/metabolismo , Proliferação de Células , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Proteínas da Mielina/metabolismo , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Oligopeptídeos/antagonistas & inibidores , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Medula Espinal/metabolismo , Medula Espinal/patologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Sinaptofisina/metabolismo
12.
Epilepsia ; 57(7): 1015-26, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27207608

RESUMO

Patients who have sustained brain injury or had developmental brain lesions present a non-negligible risk for developing delayed epilepsy. Finding therapeutic strategies to prevent development of epilepsy in at-risk patients represents a crucial medical challenge. Noncoding microRNA molecules (miRNAs) are promising candidates in this area. Indeed, deregulation of diverse brain-specific miRNAs has been observed in animal models of epilepsy as well as in patients with epilepsy, mostly in temporal lobe epilepsy (TLE). Herein we review deregulated miRNAs reported in epilepsy with potential roles in key molecular and cellular processes underlying epileptogenesis, namely neuroinflammation, cell proliferation and differentiation, migration, apoptosis, and synaptic remodeling. We provide an up-to-date listing of miRNAs altered in epileptogenesis and assess recent functional studies that have interrogated their role in epilepsy. Last, we discuss potential applications of these findings for the future development of disease-modifying therapeutic strategies for antiepileptogenesis.


Assuntos
Encéfalo/metabolismo , Epilepsia , MicroRNAs/genética , Anticonvulsivantes/uso terapêutico , Epilepsia/etiologia , Epilepsia/genética , Epilepsia/patologia , Humanos , MicroRNAs/efeitos dos fármacos
13.
Autophagy ; 20(10): 2354-2356, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38899624

RESUMO

When exposed to new experiences or changes in the environment, neurons rapidly remodel their synaptic structure and function in a process called activity-induced synaptic remodeling. This process is necessary for transforming transient experiences into stable, lasting memories. The molecular mechanisms underlying acute, activity-dependent synaptic changes are not well understood, partly because processes regulating synaptic plasticity and neurodevelopment are intricately linked. By using an RNAi screen in Drosophila targeting genes associated with human nervous system function, we found that while macroautophagy (referred to as autophagy) is fundamental for both synapse development and synaptic plasticity, activity-induced synaptic remodeling does not rely on genes associated with lysosomal degradation. These findings suggest a requirement for the unconventional secretory autophagy pathway in regulating synaptic plasticity, wherein autophagosomes, instead of fusing with lysosomes for degradation, fuse with the plasma membrane to release their contents extracellularly. To test this hypothesis, we knocked down Sec22, Snap29, and Rab8, molecular components required for secretory autophagy, all of which disrupted structural and functional plasticity. Additionally, by monitoring autophagy, we demonstrated that neuronal activity suppresses degradative autophagy to shift the pathway toward secretory autophagy release. Our work unveils secretory autophagy as a novel trans-synaptic signaling mechanism crucial for activity-induced synaptic remodeling.


Assuntos
Autofagia , Plasticidade Neuronal , Animais , Plasticidade Neuronal/fisiologia , Autofagia/fisiologia , Autofagia/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Sinapses/metabolismo , Sinapses/fisiologia , Neurônios/metabolismo , Lisossomos/metabolismo , Autofagossomos/metabolismo , Humanos
14.
J Agric Food Chem ; 72(21): 11854-11870, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38743017

RESUMO

The critical role of oxidative stress in Alzheimer's disease (AD) has been recognized by researchers recently, and natural antioxidants have been demonstrated to have anti-AD activity in animal models, such as Ginkgo biloba extract, soy isoflavones, lycopene, and so on. This paper summarized these natural antioxidants and points out that natural antioxidants always have multiple advantages which are help to deal with AD, such as clearing free radicals, regulating signal transduction, protecting mitochondrial function, and synaptic plasticity. Based on the available data, we have created a relatively complete pathway map of reactive oxygen species (ROS) and AD-related targets and concluded that oxidative stress caused by ROS is the core of AD pathogenesis. In the prospect, we introduced the concept of a combined therapeutic strategy, termed "Antioxidant-Promoting Synaptic Remodeling," highlighting the integration of antioxidant interventions with synaptic remodeling approaches as a novel avenue for therapeutic exploration.


Assuntos
Doença de Alzheimer , Antioxidantes , Ginkgo biloba , Estresse Oxidativo , Extratos Vegetais , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Antioxidantes/farmacologia , Humanos , Animais , Estresse Oxidativo/efeitos dos fármacos , Ginkgo biloba/química , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Espécies Reativas de Oxigênio/metabolismo
15.
Mol Ther Nucleic Acids ; 35(2): 102200, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38831898

RESUMO

Cancer-induced bone pain (CIBP) significantly impacts the quality of life and survival of patients with advanced cancer. Despite the established role of neurexins in synaptic structure and function, their involvement in sensory processing during injury has not been extensively studied. In this study using a rat model of CIBP, we observed increased neurexin 2 expression in spinal cord neurons. Knockdown of neurexin 2 in the spinal cord reversed CIBP-related behaviors, sensitization of spinal c-Fos neurons, and pain-related negative emotional behaviors. Additionally, increased acetylation of neurexin 2 mRNA was identified in the spinal dorsal horn of CIBP rats. Decreasing the expression of N-acetyltransferase 10 (NAT10) reduced neurexin 2 mRNA acetylation and neurexin 2 expression. In PC12 cells, we confirmed that neurexin 2 mRNA acetylation enhanced its stability, and neurexin 2 expression was regulated by NAT10. Finally, we discovered that the NAT10/ac4C-neurexin 2 axis modulated neuronal synaptogenesis. This study demonstrated that the NAT10/ac4C-mediated posttranscriptional modulation of neurexin 2 expression led to the remodeling of spinal synapses and the development of conscious hypersensitivity in CIBP rats. Therefore, targeting the epigenetic modification of neurexin 2 mRNA ac4C may offer a new therapeutic approach for the treatment of nociceptive hypersensitivity in CIBP.

16.
Front Neurol ; 14: 1107538, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37181554

RESUMO

Background: Experimental complex febrile seizures induce a persistent hippocampal hyperexcitability and an enhanced seizure susceptibility in adulthood. The rearrangement of filamentous actin (F-actin) enhances the excitability of hippocampus and contributes to epileptogenesis in epileptic models. However, the remodeling of F-actin after prolonged febrile seizures is to be determined. Methods: Prolonged experimental febrile seizures were induced by hyperthermia on P10 and P14 rat pups. Changes of actin cytoskeleton in hippocampal subregions were examined at P60 and the neuronal cells and pre- /postsynaptic components were labeled. Results: F-actin was increased significantly in the stratum lucidum of CA3 region in both HT + 10D and HT + 14D groups and further comparison between the two groups showed no significant difference. The abundance of ZNT3, the presynaptic marker of mossy fiber (MF)-CA3 synapses, increased significantly whereas the postsynaptic marker PSD95 did not change significantly. Overlapping area of F-actin and ZNT3 showed a significant increase in both HT+ groups. The results of cell counts showed no significant increase or decrease in the number of neurons in each area of hippocampus. Conclusion: F-actin was significantly up-regulated in the stratum lucidum of CA3, corresponding to the increase of the presynaptic marker of MF-CA3 synapses after prolonged febrile seizures, which may enhance the excitatory output from the dentate gyrus to CA3 and contribute to the hippocampal hyperexcitability.

17.
Genes Genet Syst ; 97(6): 297-309, 2023 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-36878557

RESUMO

Neural activity-dependent synaptic plasticity is an important physiological phenomenon underlying environmental adaptation, memory and learning. However, its molecular basis, especially in presynaptic neurons, is not well understood. Previous studies have shown that the number of presynaptic active zones in the Drosophila melanogaster photoreceptor R8 is reversibly changed in an activity-dependent manner. During reversible synaptic changes, both synaptic disassembly and assembly processes were observed. Although we have established a paradigm for screening molecules involved in synaptic stability and several genes have been identified, genes involved in stimulus-dependent synaptic assembly are still elusive. Therefore, the aim of this study was to identify genes regulating stimulus-dependent synaptic assembly in Drosophila using an automated synapse quantification system. To this end, we performed RNAi screening against 300 memory-defective, synapse-related or transmembrane molecules in photoreceptor R8 neurons. Candidate genes were narrowed down to 27 genes in the first screen using presynaptic protein aggregation as a sign of synaptic disassembly. In the second screen, we directly quantified the decreasing synapse number using a GFP-tagged presynaptic protein marker. We utilized custom-made image analysis software, which automatically locates synapses and counts their number along individual R8 axons, and identified cirl as a candidate gene responsible for synaptic assembly. Finally, we present a new model of stimulus-dependent synaptic assembly through the interaction of cirl and its possible ligand, ten-a. This study demonstrates the feasibility of using the automated synapse quantification system to explore activity-dependent synaptic plasticity in Drosophila R8 photoreceptors in order to identify molecules involved in stimulus-dependent synaptic assembly.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/genética , Drosophila melanogaster/genética , Sinapses/metabolismo , Axônios/metabolismo , Neurônios/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Receptores de Peptídeos/metabolismo
18.
Folia Med (Plovdiv) ; 65(5): 707-712, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-38351751

RESUMO

The causes of schizophrenia remain obscure and complex to identify. Alterations in dopaminergic and serotonergic neurotransmission are, to date, the primary pharmacological targets in treatment. Underlying abnormalities in neural networks have been identified as cell adhesion molecules (CAMs) involved in synaptic remodeling and interplay between neurons-neurons and neurons-glial cells. Among the CAMs, several families have been identified, such as integrins, selectins, cadherins, immunoglobulins, nectins, and the neuroligin-neurexin complex. In this paper, cell adhesion molecules involved in the pathogenesis of schizophrenia will be described.


Assuntos
Esquizofrenia , Humanos , Esquizofrenia/etiologia , Moléculas de Adesão Celular , Caderinas/metabolismo , Nectinas/metabolismo , Neurônios , Adesão Celular/fisiologia
19.
Cell Rep ; 42(11): 113327, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37906594

RESUMO

Circuit refinement involves the formation of new presynaptic boutons as others are dismantled. Nascent presynaptic sites can incorporate material from recently eliminated synapses, but the recycling mechanisms remain elusive. In early-stage C. elegans larvae, the presynaptic boutons of GABAergic DD neurons are removed and new outputs established at alternative sites. Here, we show that developmentally regulated expression of the epithelial Na+ channel (ENaC) UNC-8 in remodeling DD neurons promotes a Ca2+ and actin-dependent mechanism, involving activity-dependent bulk endocytosis (ADBE), that recycles presynaptic material for reassembly at nascent DD synapses. ADBE normally functions in highly active neurons to accelerate local recycling of synaptic vesicles. In contrast, we find that an ADBE-like mechanism results in the distal recycling of synaptic material from old to new synapses. Thus, our findings suggest that a native mechanism (ADBE) can be repurposed to dismantle presynaptic terminals for reassembly at new, distant locations.


Assuntos
Caenorhabditis elegans , Terminações Pré-Sinápticas , Animais , Neurônios GABAérgicos/fisiologia , Terminações Pré-Sinápticas/metabolismo , Sinapses/metabolismo , Vesículas Sinápticas/metabolismo
20.
J Chem Neuroanat ; 132: 102309, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37423468

RESUMO

BACKGROUND AND PURPOSE: Mesenchymal stem cells (MSC) have been demonstrated to improve cardiac function via the secretion of paracrine factors rather than direct differentiation. We, therefore, investigated whether bone marrow-derived MSC (BMSC)-released exosomes (BMSC-exo) enhance neurological recovery in spontaneously hypertensive rats (SHR) with ischemic stroke. METHODS: Markers of BMSC and BMSC-exo were detected to characterize BMSC and BMSC-exo. A green fluorescent PKH-67-labeled assay was conducted to ensure BMSC-exo internalization. Rat neuronal cells (RNC) were induced with Ang II and oxygen-glucose deprivation. The protective effects of BMSC-exo on RNC were studied by CCK-8, LDH, and immunofluorescence assays. SHR were subjected to middle cerebral artery occlusion, and the systolic and diastolic blood pressure changes in the modeled rats were measured. The effects of BMSC-exo on SHR were investigated by mNSS scoring, foot-fault tests, immunohistochemistry, Western blot, TTC staining, TUNEL, and HE staining. The hub genes related to SHR and proteins shuttled by BMSC-exo were intersected to obtain a possible candidate, followed by rescue experiments. RESULTS: BMSC-exo significantly promoted RNC viability and repressed cell apoptosis and cytotoxicity. Moreover, SHR administrated with BMSC-exo exhibited significant improvement in functional recovery and narrowed infarct size. BMSC-exo shuttled the MYCBPAP protein. Knockdown of MYCBPAP inhibited the protective effects of BMSC-exo on RNC and exacerbated synaptic damage in SHR. CONCLUSIONS: MYCBPAP shuttled by BMSC-exo facilitates synaptic remodeling in SHR, which may contribute to a therapeutic strategy for ischemic stroke treatment.


Assuntos
Exossomos , AVC Isquêmico , Células-Tronco Mesenquimais , Ratos , Animais , AVC Isquêmico/metabolismo , Exossomos/metabolismo , Diferenciação Celular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA