Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 266
Filtrar
1.
Dis Markers ; 2022: 8339759, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35186168

RESUMO

BACKGROUND: There is still a lack of knowledge regarding the association between hypertension and ferroptosis. A single-cell approach was used to study the changes in neuropeptide expression as they might contribute to the mechanisms leading to ferroptosis in a hypertensive microenvironment. METHODS: We analyzed 11798 cells from the SHR group and 12589 cells from the WKY group of mouse arterial cells. CellPhoneDB was used for cell communication analysis, and the SCENIC method was used to identify key transcription factors in neurons. The correlation between Ntrk2 and ferroptosis-related genes was further analyzed and validated via quantitative polymerase chain reaction. RESULTS: The arterial cells were clustered into six cell types. Ligand-receptor analysis suggested that Ngf, Ntf3, Cxcr4, and Ntrk2 were key neuropeptide-related genes involved in the communication between vascular smooth muscle cells and neural cells. In the hypertensive microenvironment, the neuronal transcription factor Creb3l1 appears to play a key role in the upregulation of Ntrk2 to promote the interaction between neurons and vascular smooth muscle cells. An association between Ntrk2 and the ferroptosis death inhibitor Gpx4 was suggested. RT-qPCR experiments confirmed that Ntrk2 downregulation in neural cells was followed by downregulated expression of Gpx4. CONCLUSIONS: Creb3l1, a key transcription factor in vascular neurons, may upregulate Ntrk2 to promote vascular smooth muscle cell-neuron interaction and thereby potentially prevent ferroptosis in neurons.


Assuntos
Biologia Computacional , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Ferroptose , Hipertensão/genética , Músculo Liso Vascular/citologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Receptor trkB/fisiologia , Regulação para Cima , Animais , Sequência de Bases , Comunicação Celular , Camundongos , Análise de Célula Única
2.
Sleep Breath ; 26(1): 287-295, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-33993395

RESUMO

PURPOSE: Chronic intermittent hypoxia (CIH) plays a key role in the complications of obstructive sleep apnea (OSA), which is strongly associated with retinal and optic nerve diseases. Additionally, the brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signaling pathway plays an important protective role in neuronal injury. In the present study, we investigated the role of 7,8-dihydroxyflavone (7,8-DHF) in regulating CIH-induced injury in mice retinas and rat primary retinal ganglion cells (RGCs). METHODS: C57BL/6 mice and in vitro primary RGCs were exposed to CIH or normoxia and treated with or without 7,8-DHF. The mice eyeballs or cultured cells were then taken for histochemistry, immunofluorescence or biochemistry, and the protein expression of the BDNF/TrkB signaling pathway analysis. RESULTS: Our results showed that CIH induced oxidative stress (OS) in in vivo and in vitro models and inhibited the conversion of BDNF precursor (pro-BDNF) to a mature form of BDNF, which increased neuronal cell apoptosis. 7,8-DHF reduced the production of reactive oxygen species (ROS) caused by CIH and effectively activated TrkB signals and downstream protein kinase B (Akt) and extracellular signal-regulated kinase (Erk) survival signaling pathways, which upregulated the expression of mature BDNF. ANA-12 (a TrkB specific inhibitor) blocked the protective effect of 7,8-DHF. CONCLUSION: In short, the activation of the BDNF/TrkB signaling pathway alleviated CIH-induced oxidative stress damage of the optic nerve and retinal ganglion cells. 7,8-DHF may serve as a promising agent for OSA related neuropathy.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Hipóxia Celular/efeitos dos fármacos , Flavonas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Receptor trkB/efeitos dos fármacos , Receptor trkB/fisiologia , Células Ganglionares da Retina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
3.
Pediatr Neurol ; 118: 35-39, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33773288

RESUMO

Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, has an extensively studied classical role in neuronal growth, differentiation, survival, and plasticity. Neurotrophic, from the Greek neuro and trophos, roughly translates as "vital nutrition for the brain." During development, BDNF and its associated receptor tyrosine receptor kinase B are tightly regulated as they influence the formation and maturation of neuronal synapses. Preclinical research investigating the role of BDNF in neurological disorders has focused on the effects of decreased BDNF expression on the development and maintenance of neuronal synapses. In contrast, heightened BDNF-tyrosine receptor kinase B activity has received less scrutiny for its role in neurological disorders. Recent studies suggest that excessive BDNF-tyrosine receptor kinase B signaling in the developing brain may promote the hyperexcitability that underlies refractory neonatal seizures. This review will critically examine BDNF-tyrosine receptor kinase B signaling in the immature brain, its role in the emergence of refractory neonatal seizures, and the potential of targeting BDNF-TrkB signaling as a novel antiseizure strategy.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Convulsões/etiologia , Convulsões/terapia , Humanos , Recém-Nascido , Glicoproteínas de Membrana/fisiologia , Receptor trkB/fisiologia , Convulsões/metabolismo , Transdução de Sinais/fisiologia
4.
Nutr Neurosci ; 24(12): 940-950, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31793392

RESUMO

Objectives: Although mulberry fruit possesses some biological activities, it is not known how it protects neuronal cells in neurodegenerative diseases. Here, we examined whether mulberry fruit extract (MFE) protected neuronal cells against oxidative stress-induced neurodegeneration.Methods: In this experiments, glutamate challenged hippocampal neuronal HT-22 cell lines as an in vitro model and scopolamine-induced memoty-impairment mice model were used.Results: MFE improved cell viability and glutathione level as well as reducing reactive oxygen species level in glutamate-treated HT-22 cells. Additionally, MFE suppressed apoptotic bodies and mitochondrial depolarization through regulating expression of apoptosis-related proteins. Furthermore, MFE elevated expression of p-TrkB, p-Akt, p-CREB, BDNF, and antioxidant enzymes as well as nuclear translocation of Nrf2. In contrast, the inclusion of K252a, a TrkB inhibitor, or MK-2206, an Akt selective inhibitor, neutralized the neuroprotective actions of MFE. Separately, MFE attenuated scopolamine-induced amnesia via regulating the activities of enzymes related with cholinergic function and the antioxidant system in mice. Additionally, MFE protected neuronal cells in the hippocampal CA1 and CA3 regions in brain of mice.Conclusions: MFE protects neuronal cells against oxidative stress-induced apoptosis through upregulating the expression of BDNF and antioxidant enzymes by stabilizing the activation of the TrkB/Akt pathway. Such an effect of MFE, which includes rich polyphenols, may provide information for its application as a food supplement for the prevention and treatment of neurodegenerative diseases.


Assuntos
Antioxidantes , Colinérgicos , Transtornos da Memória/tratamento farmacológico , Morus , Extratos Vegetais/administração & dosagem , Receptor trkB/fisiologia , Animais , Apoptose/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/genética , Linhagem Celular , Frutas/química , Ácido Glutâmico/farmacologia , Hipocampo/citologia , Masculino , Transtornos da Memória/induzido quimicamente , Camundongos , Camundongos Endogâmicos ICR , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fármacos Neuroprotetores , Estresse Oxidativo/efeitos dos fármacos , Fitoterapia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptor trkB/antagonistas & inibidores , Escopolamina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima
5.
Naunyn Schmiedebergs Arch Pharmacol ; 394(1): 151-164, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32444989

RESUMO

Psychiatric diseases and metabolic disorders frequently cooccur, yet the mechanisms underlying this interaction remain unknown. The aim of this study was to determine the role of glucocorticoid receptor (GR) phosphorylation in the comorbidity of metabolic and psychiatric disorders. Neonatal Sprague-Dawley rats were subcutaneously injected with monosodium glutamate (MSG) every 2 days for 10 days after birth. Metabolic and behavioral tests were performed 12 weeks later. Golgi staining and transmission electron microscopy (TEM) were performed to evaluate synaptic structural plasticity. Changes in GR phosphorylation and the BDNF/TrkB pathway were evaluated by western blotting and immunofluorescence. We found that MSG-treated rats displayed significant metabolic abnormalities accompanied by anxiogenic and depressive behaviors, an altered synaptic ultrastructure and the loss of dendritic spines. The expression of phosphorylated GR was reduced in the brain. Furthermore, a specific agonist of BDNF/TrkB significantly reversed the reduction in GR phosphorylation, as well as the metabolic and behavioral outcomes. These findings indicate that a decrease in BDNF/TrkB pathway-dependent GR phosphorylation is a long-term effect of MSG treatment that may contribute to metabolic and behavioral disturbances.


Assuntos
Ansiedade/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Depressão/fisiopatologia , Plasticidade Neuronal , Receptor trkB/fisiologia , Receptores de Glucocorticoides/fisiologia , Animais , Ansiedade/induzido quimicamente , Comportamento Animal , Encéfalo/fisiologia , Encéfalo/ultraestrutura , Corticosterona/sangue , Espinhas Dendríticas/fisiologia , Espinhas Dendríticas/ultraestrutura , Depressão/induzido quimicamente , Masculino , Fosforilação , Ratos Sprague-Dawley , Glutamato de Sódio
6.
Cell Biol Int ; 45(2): 432-446, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33200854

RESUMO

Neural stem cells (NSCs) or neuronal progenitor cells are cells capable of differentiating into oligodendrocytes, myelin-forming cells that have the potential of remyelination. Brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) are two neurotrophic factors that have been studied to stimulate NSC differentiation thus playing a role in multiple sclerosis pathogenesis and several other demyelinating disorders. While several studies have demonstrated the proliferative and protective capabilities of these neurotrophic factors, their cellular and molecular functions are still not well understood. Thus, in the present study, we focus on understanding the role of these neurotrophins (BDNF and NGF) in oligodendrogenesis from NSCs. Both neurotrophic factors have been shown to promote NSC proliferation and NSC differentiation particularly into oligodendroglial lineage in a dose-dependent fashion. Further, to establish the role of these neurotrophins in NSC differentiation, we have employed pharmacological inhibitors for TrkA and TrkB receptors in NSCs. The use of these inhibitors suppressed NSC differentiation into oligodendrocytes along with the downregulation of phosphorylated ERK suggesting active involvement of ERK in the functioning of these neurotrophins. The morphometric analysis also revealed the important role of both neurotrophins in oligodendrocytes development. These findings highlight the importance of neurotrophic factors in stimulating NSC differentiation and may pave a role for future studies to develop neurotrophic factor replacement therapies to achieve remyelination.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Fator de Crescimento Neural/fisiologia , Células-Tronco Neurais , Receptor trkB/fisiologia , Receptores de Fator de Crescimento Neural/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Oligodendroglia/citologia
7.
J Neurosci ; 40(42): 8070-8087, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32948677

RESUMO

Brain-derived neurotropic factor (BDNF) has been shown to play critical roles in neural development, plasticity, and neurodegenerative diseases. The main function of BDNF in the brain is widely accepted to be synaptic regulation. However, how BDNF modulates synaptic transmission, especially the underlying signaling cascades between presynaptic and postsynaptic neurons, remains controversial. In the present study, we investigated the actions of BDNF at rat calyx-type synapses of either sex by measuring the excitatory postsynaptic current (EPSC) and presynaptic calcium current and capacitance changes. We found that BDNF inhibits the EPSC, presynaptic calcium influx, and exocytosis/endocytosis via activation of the presynaptic cannabinoid Type 1 receptors (CB1Rs). Inhibition of the CB1Rs abolished the BDNF-induced presynaptic inhibition, whereas CB1R agonist mimicked the effect of BDNF. Exploring the underlying signaling cascade, we found that BDNF specifically activates the postsynaptic TrkB receptors, inducing the release of endocannabinoids via the PLCγ/DGL pathway and retrogradely activating presynaptic CB1Rs. We also reported the involvement of AC/PKA in modulating vesicle endocytosis, which may account for the BDNF-induced calcium-dependent and -independent regulation of endocytosis. Thus, our study provides new insights into the BDNF/endocannabinoid-associated modulation of neurotransmission in physiological and pathologic processes.SIGNIFICANCE STATEMENT BDNF plays critical roles in the modulation of synaptic strength. However, how BDNF regulates synaptic transmission and its underlying signaling cascade(s) remains elusive. By measuring EPSC and the presynaptic calcium current and capacitance changes at rat calyces, we found that BDNF inhibits synaptic transmission via BDNF-TrkB-eCB signaling pathway. Activation of postsynaptic TrkB receptors induces endocannabinoid release via the PLCγ/DGL pathway, retrogradely activating the presynaptic CB1Rs, inhibiting the AC/PKA, and suppressing calcium influx. Our findings provide a comprehensive understanding of BDNF/endocannabinoid-associated modulation of neuronal activities.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Sinalização do Cálcio/fisiologia , Endocanabinoides/metabolismo , Receptor trkB/fisiologia , Receptores Pré-Sinápticos/fisiologia , Sinapses/fisiologia , Animais , Endocitose/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Exocitose/fisiologia , Feminino , Técnicas In Vitro , Masculino , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/metabolismo , Transmissão Sináptica
8.
Behav Brain Res ; 386: 112596, 2020 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-32194188

RESUMO

Cocaine-cue extinction training combined with brief interventions of environmental enrichment (EE) was shown previously to facilitate extinction and attenuate reacquisition of cocaine self-administration in rats. It is unknown whether or not the usefulness of this approach would be undermined if extinction training took place in a novel rather than familiar context. Drawing on previous studies involving pharmacological interventions, we hypothesized that the facilitative effects of EE for cocaine relapse prevention would be independent of the context used for extinction training. Rats trained to self-administer cocaine underwent cocaine-cue extinction training in either the familiar self-administration context or a novel context, with or without EE. Rats then were tested for reacquisition of cocaine self-administration in the familiar context. Target brain regions were lysed and probed for memory-related changes in receptors for glutamate and BDNF by western blotting. Contrary to our hypothesis, the facilitative effects of EE for cocaine relapse prevention were dependent on the context used for extinction training. While EE facilitated extinction regardless of context used, it inhibited cocaine relapse only after extinction training in the familiar context. EE was associated with increased GluA2 in nucleus accumbens, TrkB in dorsal hippocampus and activated TrkB in ventromedial prefrontal cortex. Of these, the changes in dorsal hippocampus and ventromedial prefrontal cortex mirrored outcomes of the cocaine relapse tests in that these changes were specific to rats receiving EE plus extinction training in the familiar context. These findings support a role for hippocampal-prefrontal BDNF-TrkB signaling in extinction-based relapse prevention strategies involving EE.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Receptor trkB/metabolismo , Prevenção Secundária/métodos , Animais , Comportamento Aditivo/psicologia , Encéfalo/metabolismo , Cocaína/farmacologia , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Condicionamento Operante/efeitos dos fármacos , Sinais (Psicologia) , Inibidores da Captação de Dopamina/farmacologia , Extinção Psicológica/fisiologia , Hipocampo/metabolismo , Masculino , Memória/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Wistar , Receptor trkB/fisiologia , Recidiva , Autoadministração
9.
J Neurosci ; 40(7): 1405-1426, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-31915257

RESUMO

BDNF signaling via its transmembrane receptor TrkB has an important role in neuronal survival, differentiation, and synaptic plasticity. Remarkably, BDNF is capable of modulating its own expression levels in neurons, forming a transcriptional positive feedback loop. In the current study, we have investigated this phenomenon in primary cultures of rat cortical neurons using overexpression of dominant-negative forms of several transcription factors, including CREB, ATF2, C/EBP, USF, and NFAT. We show that CREB family transcription factors, together with the coactivator CBP/p300, but not the CRTC family, are the main regulators of rat BDNF gene expression after TrkB signaling. CREB family transcription factors are required for the early induction of all the major BDNF transcripts, whereas CREB itself directly binds only to BDNF promoter IV, is phosphorylated in response to BDNF-TrkB signaling, and activates transcription from BDNF promoter IV by recruiting CBP. Our complementary reporter assays with BDNF promoter constructs indicate that the regulation of BDNF by CREB family after BDNF-TrkB signaling is generally conserved between rat and human. However, we demonstrate that a nonconserved functional cAMP-responsive element in BDNF promoter IXa in humans renders the human promoter responsive to BDNF-TrkB-CREB signaling, whereas the rat ortholog is unresponsive. Finally, we show that extensive BDNF transcriptional autoregulation, encompassing all major BDNF transcripts, occurs also in vivo in the adult rat hippocampus during BDNF-induced LTP. Collectively, these results improve the understanding of the intricate mechanism of BDNF transcriptional autoregulation.SIGNIFICANCE STATEMENT Deeper understanding of stimulus-specific regulation of BDNF gene expression is essential to precisely adjust BDNF levels that are dysregulated in various neurological disorders. Here, we have elucidated the molecular mechanisms behind TrkB signaling-dependent BDNF mRNA induction and show that CREB family transcription factors are the main regulators of BDNF gene expression after TrkB signaling. Our results suggest that BDNF-TrkB signaling may induce BDNF gene expression in a distinct manner compared with neuronal activity. Moreover, our data suggest the existence of a stimulus-specific distal enhancer modulating BDNF gene expression.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Fator Neurotrófico Derivado do Encéfalo/genética , Córtex Cerebral/citologia , Regulação da Expressão Gênica/genética , Hipocampo/citologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Transcrição Gênica/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células Cultivadas , Córtex Cerebral/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/genética , Retroalimentação Fisiológica , Feminino , Genes Dominantes , Genes Reporter , Genes Sintéticos , Hipocampo/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor trkB/fisiologia , Proteínas Recombinantes/farmacologia , Elementos de Resposta , Especificidade da Espécie , Transdução Genética
10.
Cereb Cortex ; 30(1): 197-214, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31329835

RESUMO

The dorsal striatum exhibits bidirectional corticostriatal synaptic plasticity, NMDAR and endocannabinoids (eCB) mediated, necessary for the encoding of procedural learning. Therefore, characterizing factors controlling corticostriatal plasticity is of crucial importance. Brain-derived neurotrophic factor (BDNF) and its receptor, the tropomyosine receptor kinase-B (TrkB), shape striatal functions, and their dysfunction deeply affects basal ganglia. BDNF/TrkB signaling controls NMDAR plasticity in various brain structures including the striatum. However, despite cross-talk between BDNF and eCBs, the role of BDNF in eCB plasticity remains unknown. Here, we show that BDNF/TrkB signaling promotes eCB-plasticity (LTD and LTP) induced by rate-based (low-frequency stimulation) or spike-timing-based (spike-timing-dependent plasticity, STDP) paradigm in striatum. We show that TrkB activation is required for the expression and the scaling of both eCB-LTD and eCB-LTP. Using 2-photon imaging of dendritic spines combined with patch-clamp recordings, we show that TrkB activation prolongs intracellular calcium transients, thus increasing eCB synthesis and release. We provide a mathematical model for the dynamics of the signaling pathways involved in corticostriatal plasticity. Finally, we show that TrkB activation enlarges the domain of expression of eCB-STDP. Our results reveal a novel role for BDNF/TrkB signaling in governing eCB-plasticity expression in striatum and thus the engram of procedural learning.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Endocanabinoides/fisiologia , Neostriado/fisiologia , Plasticidade Neuronal , Receptor trkB/fisiologia , Córtex Somatossensorial/fisiologia , Animais , Modelos Neurológicos , Vias Neurais/fisiologia , Ratos
11.
J Mol Neurosci ; 69(3): 360-370, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31286407

RESUMO

Intracerebral hemorrhage (ICH) leads to widespread pathological lesions in the brain, especially impacting neuronal survival and axonal regeneration. This study aimed to elucidate whether the Nogo-A (a myelin-related protein)/paired immunoglobulin-like receptor B (Pir-B)/tropomyosin receptor kinase B (TrkB) pathway could exert a regulatory effect in ICH. An ICH model was first established in Sprague Dawley rats, followed by different administrations of vehicle, k252a, or NSC 87877. The Morris water maze test was performed to observe ICH-induced cognitive dysfunction in rats. Rats in the ICH + NSC 87877 group showed better cognitive performance compared with those injected with vehicle or k252a. Neurobehavioral scores were identical. By harvesting brain tissues at different time points after ICH, we detected the expression levels of Nogo-A and PirB with western blot and immunofluorescence and found that they were markedly upregulated at 48 h after ICH. TUNEL and Fluoro-Jade B staining showed that NSC 87877 treatment attenuated ICH-induced apoptosis and neuronal death, whereas k252a treatment aggravated these pathological changes. The expression levels of growth-associated protein 43 (GAP43) and neurofilament 200 (NF200) were higher in the ICH + NSC 87877 group compared with the ICH + vehicle group, but were lower in the ICH + k252a group. Finally, we confirmed the protective role of p-TrkB/TrkB in ICH by western blot. To sum up, our study identified the inhibitory role of the Nogo-A/PirB/TrkB pathway in ICH; however, p-TrkB/TrkB may serve as a potential target for secondary brain injury post-ICH.


Assuntos
Hemorragia Cerebral/fisiopatologia , Crescimento Neuronal/fisiologia , Neurônios/fisiologia , Proteínas Nogo/fisiologia , Receptor trkB/fisiologia , Receptores Imunológicos/fisiologia , Transdução de Sinais , Animais , Apoptose , Encéfalo/patologia , Carbazóis/toxicidade , Morte Celular , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/patologia , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/prevenção & controle , Alcaloides Indólicos/toxicidade , Masculino , Aprendizagem em Labirinto , Atividade Motora , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Proteínas Nogo/biossíntese , Quinolinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Receptores Imunológicos/biossíntese , Regeneração , Regulação para Cima
12.
Behav Brain Res ; 373: 112080, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31301412

RESUMO

Vagus nerve stimulation (VNS) is being explored as a potential therapeutic for Parkinson's disease (PD). VNS is less invasive than other surgical treatments and has beneficial effects on behavior and brain pathology. It has been suggested that VNS exerts these effects by increasing brain-derived neurotrophic factor (BDNF) to enhance pro-survival mechanisms of its receptor, tropomyosin receptor kinase-B (TrkB). We have previously shown that striatal BDNF is increased after VNS in a lesion model of PD. By chronically administering ANA-12, a TrkB-specific antagonist, we aimed to determine TrkB's role in beneficial VNS effects for a PD model. In this study, we administered a noradrenergic neurotoxin, DSP-4, intraperitoneally and one week later administered a bilateral intrastriatal dopaminergic neurotoxin, 6-OHDA. At this time, the left vagus nerve was cuffed for stimulation. Eleven days later, rats received VNS twice per day for ten days, with daily locomotor assessment. Daily ANA-12 injections were given one hour prior to the afternoon stimulation and concurrent locomotor session. Following the final VNS session, rats were euthanized, and left striatum, bilateral substantia nigra and locus coeruleus were sectioned for immunohistochemical detection of neurons, α-synuclein, astrocytes, and microglia. While ANA-12 did not avert behavioral improvements of VNS, and only partially prevented VNS-induced attenuation of neuronal loss in the locus coeruleus, it did stop neuronal and anti-inflammatory effects of VNS in the nigrostriatal system, indicating a role for TrkB in mediating VNS efficacy. However, our data also suggest that BDNF-TrkB is not the sole mechanism of action for VNS in PD.


Assuntos
Doença de Parkinson/metabolismo , Receptor trkB/metabolismo , Nervo Vago/metabolismo , Animais , Azepinas/farmacologia , Benzamidas/farmacologia , Encéfalo/metabolismo , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Dopamina/farmacologia , Locus Cerúleo/metabolismo , Masculino , Neostriado/metabolismo , Norepinefrina/farmacologia , Oxidopamina/farmacologia , Doença de Parkinson/patologia , Ratos , Ratos Long-Evans , Receptor trkB/fisiologia , Substância Negra/metabolismo , Estimulação do Nervo Vago/métodos
13.
Neurochem Int ; 128: 32-38, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30986502

RESUMO

In most mature neurons, low levels of intracellular Cl- concentrations ([Cl-]i) are maintained by channels and transporters, particularly the K+-Cl- cotransporter 2 (KCC2), which is the only Cl- extruder in most neurons. Recent studies have implicated KCC2 expression in the molecular mechanisms underlying neuronal disorders, such as spasticity, epilepsy and neuropathic pain. Alterations in KCC2 expression have been associated with brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin-related kinase B (TrkB). The present review summarizes recent progress regarding the roles of Cl- regulators in immature and mature neurons. Moreover, we focus on the role of KCC2 regulation via the BDNF-TrkB pathway in spinal cord injury and rehabilitation, as prior studies have shown that the BDNF-TrkB pathway can affect both the pathological development and functional amelioration of spinal cord injuries. Evidence suggests that rehabilitation using active exercise and mechanical stimulation can attenuate spasticity and neuropathic pain in animal models, likely due to the upregulation of KCC2 expression via the BDNF-TrkB pathway. Moreover, research suggests that such rehabilitation efforts may recover KCC2 expression without the use of exogenous BDNF.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Glicoproteínas de Membrana/fisiologia , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/reabilitação , Reabilitação Neurológica/tendências , Receptor trkB/fisiologia , Simportadores/fisiologia , Animais , Transdução de Sinais/fisiologia
14.
Surg Today ; 49(9): 721-727, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30848386

RESUMO

Neuroblastoma is one of the most frequent, yet distinctive and challenging childhood tumors. The uniqueness of this tumor depends on its biological markers, which classify neuroblastomas into favorable and unfavorable, with 5-year survival rates ranging from almost 100-30%. In this review, we focus on some biological factors that play major roles in neuroblastoma: MYCN, Trk, and ALK. The MYCN and Trk family genes have been studied for decades and are known to be crucial for the tumorigenesis and progression of neuroblastoma. ALK gene mutations have been recognized recently to be responsible for familial neuroblastomas. Each factor plays an important role in normal neural development, regulating cell proliferation or differentiation by activating several signaling pathways, and interacting with each other. These factors have been studied not only as prognostic factors, but also as targets of neuroblastoma therapy, and some clinical trials are ongoing. We review the basic aspects of MYCN, Trk, and ALK in both neural development and in neuroblastoma.


Assuntos
Quinase do Linfoma Anaplásico/fisiologia , Carcinogênese/genética , Glicoproteínas de Membrana/fisiologia , Proteína Proto-Oncogênica N-Myc/fisiologia , Sistema Nervoso/crescimento & desenvolvimento , Neuroblastoma/genética , Receptor trkA/fisiologia , Receptor trkB/fisiologia , Diferenciação Celular/genética , Proliferação de Células/genética , Criança , Progressão da Doença , Humanos , Mutação , Neuroblastoma/patologia , Transdução de Sinais
15.
Cereb Cortex ; 29(9): 3666-3675, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30272136

RESUMO

We have previously reported that the blockage of TrkB and TrkC signaling in primary culture of opossum neocortical cells affects neurogenesis that involves a range of processes including cell proliferation, differentiation, and survival. Here, we studied whether TrkB and TrkC activity specifically affects various types of progenitor cell populations during neocortex formation in the Monodelphis opossum in vivo. We found that the inhibition of TrkB and TrkC activities affects the same proliferative cellular phenotype, but TrkC causes more pronounced changes in the rate of cell divisions. Additionally, inhibition of TrkB and TrkC does not affect apoptosis in vivo, which was found in cell culture experiments. The lack of TrkB and TrkC receptor activity caused the arrest of newly generated neurons; therefore, they could not penetrate the subplate zone. We suggest that at this time point in development, migration consists of 2 steps. During the initial step, neurons migrate and reach the base of the subplate, whereas during the next step the migration of neurons to their final position is regulated by TrkB or TrkC signaling.


Assuntos
Monodelphis/fisiologia , Neocórtex/fisiologia , Neurogênese , Neurônios/fisiologia , Receptor trkB/fisiologia , Receptor trkC/fisiologia , Animais , Diferenciação Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Feminino , Masculino , Transdução de Sinais
16.
Behav Neurosci ; 133(1): 32-38, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30489133

RESUMO

The expression of brain derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B (TrkB) are regulated by gonadal hormone signaling and are expressed in brain areas that are important for sexual behaviors. Accordingly, BDNF and TrkB signaling have been shown to be important for the expression of consummatory sexual behaviors. However, the role of TrkB in sexually motivated behaviors remains to be fully elucidated. To this end, male rats were administered either the TrkB antagonist, ANA-12, or a vehicle control prior to sexual motivation testing, which took place on a noncontact version of a partner preference task. Vehicle treated rats, but not rats treated with ANA-12, exhibited a preference for the sexually receptive stimulus female rat relative to the sexually active stimulus male rat, as indicated by the percentage of time and entries in the vicinity of the female throughout the entire 20-min test. In addition, when compared directly with vehicle treated rats, rats treated with ANA-12 exhibited a significant decrease in levels of sexual motivation as indicated by the magnitude of each group's preference for the female during the early stages of testing. Collectively, these results suggest that TrkB plays a role in the sexual preferences and corresponding initial levels of sexual motivation in male rats. (PsycINFO Database Record (c) 2019 APA, all rights reserved).


Assuntos
Motivação/fisiologia , Receptor trkB/fisiologia , Comportamento Sexual Animal , Animais , Azepinas/administração & dosagem , Benzamidas/administração & dosagem , Comportamento de Escolha , Feminino , Masculino , Ratos Long-Evans , Receptor trkB/antagonistas & inibidores
17.
PLoS One ; 13(10): e0205648, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30308049

RESUMO

INTRODUCTION: Suicide Attempts (SA) are the main complications of Major Depressive Episodes (MDE) and are difficult to predict. Suicide is associated with the expression of Receptor Tyrosin-Kinase B (TRKB), the receptor of the Brain Derived Neurotrophic Factor (BDNF) involved in MDE. However, the impact of its genetic polymorphisms as predictive factors of SA should be clarified. Our main aim is to assess the association of 8 TRKB genetic polymorphisms and SA in depressed patients. MATERIAL AND METHODS: In 624 patients currently experiencing an MDE in the context of Major Depressive Disorder (MDD) (METADAP study), we assessed the association between 8 TRKB genetic polymorphisms (rs1778933, rs1187352, rs2289658, rs2289657, rs2289656, rs3824519, rs56142442 and rs1439050) and acute (previous month) or past (older than one month) SA. Bonferroni corrections and multivariate analysis adjusted for age, sex, level of education, marital status, Hamilton Depression Rating Scale score and previous MDE were used. RESULTS: The rs2289656 was associated with acute SA (CC = 28.5%, CT = 15.0% and TT = 11.5%, p = 0.0008). However, the other SNPs were not. Patients with the CC genotype had a higher rate of acute SA (28.5%) as compared to T carriers (14.6%) (adjusted OR = 2.2, CI95% [1.4; 3.5], p<0.0001). CONCLUSION: The TRKB rs2289656 CC genotype is associated with a 2.2 fold higher risk of acute SA in depressed patients. If this result could be confirmed, this TRKB SNP may be assessed to contribute to the prediction of SA in depressed patients.


Assuntos
Transtorno Depressivo Maior/genética , Glicoproteínas de Membrana/genética , Polimorfismo de Nucleotídeo Único/genética , Receptor trkB/genética , Tentativa de Suicídio , Adolescente , Adulto , Idoso , Estudos de Casos e Controles , Feminino , Predisposição Genética para Doença/genética , Haplótipos/genética , Humanos , Masculino , Glicoproteínas de Membrana/fisiologia , Pessoa de Meia-Idade , Receptor trkB/fisiologia , Adulto Jovem
18.
Psychiatry Clin Neurosci ; 72(9): 633-653, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29802758

RESUMO

The network hypothesis of depression proposes that mood disorders reflect problems in information processing within particular neural networks. Antidepressants (AD), including selective serotonin reuptake inhibitors (SSRI), function by gradually improving information processing within these networks. AD have been shown to induce a state of juvenile-like plasticity comparable to that observed during developmental critical periods: Such critical-period-like plasticity allows brain networks to better adapt to extrinsic and intrinsic signals. We have coined this drug-induced state of juvenile-like plasticity 'iPlasticity.' A combination of iPlasticity induced by chronic SSRI treatment together with training, rehabilitation, or psychotherapy improves symptoms of neuropsychiatric disorders and issues underlying the developmentally or genetically malfunctioning networks. We have proposed that iPlasticity might be a critical component of AD action. We have demonstrated that iPlasticity occurs in the visual cortex, fear erasure network, extinction of aggression caused by social isolation, and spatial reversal memory in rodent models. Chronic SSRI treatment is known to promote neurogenesis and to cause dematuration of granule cells in the dentate gyrus and of interneurons, especially parvalbumin interneurons enwrapped by perineuronal nets in the prefrontal cortex, visual cortex, and amygdala. Brain-derived neurotrophic factor (BDNF), via its receptor tropomyosin kinase receptor B, is involved in the processes of synaptic plasticity, including neurogenesis, neuronal differentiation, weight of synapses, and gene regulation of synaptic formation. BDNF can be activated by both chronic SSRI treatment and neuronal activity. Accordingly, the BDNF/tropomyosin kinase receptor B pathway is critical for iPlasticity, but further analyses will be needed to provide mechanical insight into the processes of iPlasticity.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Humanos , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Receptor trkB/fisiologia
19.
J Neurophysiol ; 120(2): 633-643, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29694277

RESUMO

Brain-derived neurotrophic factor (BDNF) expression increases in the paraventricular nucleus of the hypothalamus (PVN) during stress, and our recent studies indicate that BDNF induces sympathoexcitatory and hypertensive responses when injected acutely or overexpressed chronically in the PVN. However, it remained to be investigated whether BDNF is involved in the mediation of stress-induced cardiovascular responses. Here we tested the hypothesis that inhibition of the high-affinity BDNF receptor TrkB in the PVN diminishes acute stress-induced cardiovascular responses. Male Sprague-Dawley rats were equipped with radiotelemetric transmitters for blood pressure measurement. BDNF-TrkB signaling was selectively inhibited by viral vector-mediated bilateral PVN overexpression of a dominant-negative truncated TrkB receptor (TrkB.T1, n = 7), while control animals ( n = 7) received green fluorescent protein (GFP)-expressing vector injections. Rats were subjected to acute water and restraint stress 3-4 wk after vector injections. We found that body weight, food intake, baseline mean arterial pressure (MAP), and heart rate were unaffected by TrkB.T1 overexpression. However, peak MAP increases were significantly reduced in the TrkB.T1 group compared with GFP both during water stress (GFP: 39 ± 2 mmHg, TrkB.T1: 27 ± 4 mmHg; P < 0.05) and restraint stress (GFP: 41 ± 3 mmHg, TrkB.T1: 34 ± 2 mmHg; P < 0.05). Average MAP elevations during the poststress period were also significantly reduced after both water and restraint stress in the TrkB.T1 group compared with GFP. In contrast, heart rate elevations to both stressors remained unaffected by TrkB.T1 overexpression. Our results demonstrate that activation of BDNF high-affinity TrkB receptors within the PVN is a major contributor to acute stress-induced blood pressure elevations. NEW & NOTEWORTHY We have shown that inhibition of the high-affinity brain-derived neurotrophic factor receptor TrkB in the paraventricular nucleus of the hypothalamus significantly reduces blood pressure elevations to acute stress without having a significant impact on resting blood pressure, body weight, and food intake.


Assuntos
Pressão Arterial , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Receptor trkB/fisiologia , Estresse Psicológico/fisiopatologia , Animais , Ingestão de Alimentos , Frequência Cardíaca , Masculino , Ratos Sprague-Dawley , Ratos Transgênicos , Transdução de Sinais
20.
eNeuro ; 5(1)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29662939

RESUMO

In cognitive memory, long-term potentiation (LTP) has been shown to occur when presynaptic and postsynaptic activities are highly correlated and glucocorticoid concentrations are in an optimal (i.e., low normal) range. In all other conditions, LTP is attenuated or even long-term depression (LTD) occurs. In this paper, we focus on NMDA receptor (NMDA-R)-dependent LTP and LTD, two processes involving various molecular mechanisms. To understand which of these mechanisms are indispensable for explaining the experimental evidence reported in the literature, we here propose a parsimonious model of NMDA-R-dependent synaptic plasticity. Central to this model are two processes. First, AMPA receptor-subunit trafficking; and second, glucocorticoid-dependent modifications of the brain-derived neurotrophic factor (BDNF)-receptor system. In 2008, we have published a core model, which contained the first process, while in the current paper we present an extended model, which also includes the second process. Using the extended model, we could show that stress attenuates LTP, while it enhances LTD. These simulation results are in agreement with experimental findings from other labs. In 2013, surprising experimental evidence showed that the GluA1 C-tail is unnecessary for LTP. When using our core model in its original form, our simulations already predicted that there would be no requirement for the GluA1 C-tail for LTP, allowing to eliminate a redundant mechanism from our model. In summary, we present a mathematical model that displays reduced complexity and is useful for explaining when and how LTP or LTD occurs at synapses during cognitive memory formation.


Assuntos
Potenciação de Longa Duração , Depressão Sináptica de Longo Prazo , Modelos Neurológicos , Neurônios/fisiologia , Estresse Psicológico , Animais , Simulação por Computador , Humanos , Receptor trkB/fisiologia , Receptores de AMPA/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...