Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Transl Neurodegener ; 10(1): 34, 2021 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-34496956

RESUMO

BACKGROUND: ß Amyloid (Aß)-mediated neuronal hyperactivity, a key feature of the early stage of Alzheimer's disease (AD), is recently proposed to be initiated by the suppression of glutamate reuptake. Nevertheless, the underlying mechanism by which the impaired glutamate reuptake causes neuronal hyperactivity remains unclear. Chronic suppression of the glutamate reuptake causes accumulation of ambient glutamate that could diffuse from synaptic sites at the dendrites to the soma to elevate the tonic activation of somatic N-methyl-D-aspartate receptors (NMDARs). However, less attention has been paid to the potential role of tonic activity change in extrasynaptic glutamate receptors (GluRs) located at the neuronal soma on generation of neuronal hyperactivity. METHODS: Whole-cell patch-clamp recordings were performed on CA1 pyramidal neurons in acute hippocampal slices exposed to TFB-threo-ß-benzyloxyaspartic acid (TBOA) or human Aß1-42 peptide oligomer. A series of dendritic patch-clamp recordings were made at different distances from the soma to identify the location of the changes in synaptic inputs. Moreover, single-channel recording in the cell-attached mode was performed to investigate the activity changes of single NMDARs at the soma. RESULTS: Blocking glutamate uptake with either TBOA or the human Aß1-42 peptide oligomer elicited potentiation of synaptic inputs in CA1 hippocampal neurons. Strikingly, this potentiation  specifically occurred at the soma, depending on the activation of somatic GluN2B-containing NMDARs (GluN2B-NMDARs) and accompanied by a substantial and persistent increment in the open probability of somatic NMDARs. Blocking the activity of GluN2B-NMDARs at the soma completely reversed both the TBOA-induced or the Aß1-42-induced somatic potentiation and neuronal hyperactivity. CONCLUSIONS: The somatic potentiation of synaptic inputs may represent a novel amplification mechanism that elevates cell excitability and thus contributes to neuronal hyperactivity initiated by impaired glutamate reuptake in AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Corpo Celular/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Neurônios/fisiologia , Fragmentos de Peptídeos/toxicidade , Receptores de N-Metil-D-Aspartato/fisiologia , Sinapses/fisiologia , Animais , Ácido Aspártico/toxicidade , Corpo Celular/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Humanos , Masculino , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Sinapses/efeitos dos fármacos
2.
Neuropharmacology ; 158: 107731, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31376424

RESUMO

Disruption of the hypothalamic-pituitary-adrenal axis is an established finding in patients with anxiety and/or depression. Chronic corticosterone administration in animals has been proposed as a model for the study of these stress-related disorders and the antidepressant action. Alterations of the central noradrenergic system and specifically of inhibitory α2-adrenoceptors seem to be part of the pathophysiology of depression and contribute to the antidepressant activity. The present study evaluates in male rats the effect of chronic corticosterone treatment during 35 days (16-20 mg kg-1 day-1) on the sensitivity of α2-adrenoceptors expressed in the somatodendritic and terminal noradrenergic areas locus coeruleus (LC) and prefrontal cortex (PFC), respectively. Further, the effect of chronic fluoxetine treatment (5 mg kg-1, i.p., since the 15th day) on the sensitivity of α2-adrenoceptors was examined under control conditions and in corticosterone-treated rats. The α2-adrenoceptor functionality was analysed in vitro by agonist-mediated [35S]GTPγS binding stimulation and in vivo through the modulation of noradrenaline (NA) release evaluated by dual-probe microdialysis. The concentration-effect curves of the [35S]GTPγS binding stimulation by the agonist UK14304 (5-bromo-N-(4,5-dihydro-1H-imidazol-2-yl)-6-quinoxalinamine) demonstrated a desensitization of cortical α2-adrenoceptors induced by corticosterone (-logEC50 = 6.7 ±â€¯0.2 vs 8.2 ±â€¯0.3 in controls) that was reverted by fluoxetine treatment (-logEC50 = 7.5 ±â€¯0.3). Local administration of the α2-adrenoceptor antagonist RS79948 ((8aR,12aS,13aS)-5,8,8a,9,10,11,12,12a,13,13a-decahydro-3-methoxy-12-(ethylsulfonyl)-6H-isoquino[2,1-g][1,6]naphthyridine) (0.1-100 µmol L-1) into the LC induced a concentration-dependent NA increase in the PFC of the control group (Emax = 191 ±â€¯30%) but non-significant effect was observed in corticosterone-treated rats (Emax = 133 ±â€¯46%), reflecting a desensitization of α2-adrenoceptors that control the firing of noradrenergic neurons. Fluoxetine treatment did not alter the corticosterone-induced desensitization in this area (Emax = 136 ±â€¯19%). No effect of fluoxetine on α2-adrenoceptor functionality was observed in control animals (Emax = 223 ±â€¯30%). In PFC, the local administration of RS79948 increased NA in controls (Emax = 226 ±â€¯27%) without effect in the corticosterone group (Emax = 115 ±â€¯26%), suggesting a corticosterone-induced desensitization of terminal α2-adrenoceptors. Fluoxetine administration prevented the desensitization induced by corticosterone in the PFC (Emax = 233 ±â€¯33%) whereas desensitized α2-adrenoceptors in control animals (Emax = -24 ±â€¯10%). These data indicate that chronic corticosterone increases noradrenergic activity by acting at different α2-adrenoceptor subpopulations. Treatment with the antidepressant fluoxetine seems to counteract these changes by acting mainly on presynaptic α2-adrenoceptors expressed in terminal areas.


Assuntos
Neurônios Adrenérgicos/efeitos dos fármacos , Antidepressivos de Segunda Geração/farmacologia , Corticosterona/farmacologia , Fluoxetina/farmacologia , Locus Cerúleo/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Neurônios Adrenérgicos/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Tartarato de Brimonidina/farmacologia , Corpo Celular/efeitos dos fármacos , Corpo Celular/metabolismo , Dendritos/efeitos dos fármacos , Dendritos/metabolismo , Modelos Animais de Doenças , Guanosina 5'-O-(3-Tiotrifosfato) , Sistema Hipotálamo-Hipofisário/metabolismo , Técnicas In Vitro , Isoquinolinas/farmacologia , Locus Cerúleo/metabolismo , Masculino , Microdiálise , Naftiridinas/farmacologia , Norepinefrina/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Córtex Pré-Frontal/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Ratos , Receptores Adrenérgicos alfa 2/metabolismo , Estresse Psicológico/metabolismo , Radioisótopos de Enxofre
3.
ACS Chem Neurosci ; 10(3): 1497-1505, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30412381

RESUMO

Substance abuse disorders are devastating, costly, and difficult to treat. Identifying the neurochemical mechanisms underlying reinforcement promises to provide critical information in the development of effective treatments. Several lines of evidence suggest that striatal dopamine (DA) release serves as a teaching signal in reinforcement learning, and that shifts in DA release from the primary reward to reward-predicting stimuli play a critical role in the self-administration of both natural and non-natural rewards. However, far less is known about the reinforcing effects of motivationally neutral sensory stimuli, or how these signals can facilitate self-administration behavior. Thus, we trained rats ( n = 7) to perform a visual stimulus-induced instrumental task, which involved lever pressing for activation of a stimulus light. We then microinfused vehicle (phosphate buffered saline), carbachol (acetylcholine receptor agonist), or carbachol in the presence of an N-methyl-d-aspartate (NMDA) receptor-specific drug (NMDA itself, or the antagonist, AP5) into the ventral tegmental area (VTA). This enabled us to directly evaluate how chemical modulation of dopamine cell bodies affects the instrumental behavior, as well as the nature of extracellular dopamine transients recorded in the nucleus accumbens shell (NAc shell) using fast-scan cyclic voltammetry (FSCV). Intra-VTA infusion of carbachol enhanced the magnitude and frequency of dopamine transients in the NAc shell and potentiated active lever responding without altering inactive lever responding, as compared to infusion of vehicle. Coinfusion of carbachol with AP5 abolished dopamine transients recorded in the NAc and attenuated active lever responding without altering inactive lever responding. Finally, coadministration of carbachol and NMDA into the VTA restored both lever pressing and dopaminergic signals recorded in the striatum. Together, these results suggest that acetylcholine and glutamate synergistically act at dopamine cells in the VTA to modulate VTA-NAc shell dopaminergic output, and this underlies motivation to lever press for a motivationally neutral visual stimulus.


Assuntos
Corpo Celular/metabolismo , Dopamina/metabolismo , Estimulação Luminosa/métodos , Desempenho Psicomotor/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Corpo Celular/efeitos dos fármacos , Agonistas Colinérgicos/administração & dosagem , Masculino , Microinjeções/métodos , N-Metilaspartato/administração & dosagem , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Área Tegmentar Ventral/efeitos dos fármacos
4.
Dev Cell ; 42(6): 626-639.e5, 2017 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-28919207

RESUMO

Axonal targeting of signaling receptors is essential for neuronal responses to extracellular cues. Here, we report that retrograde signaling by target-derived nerve growth factor (NGF) is necessary for soma-to-axon transcytosis of TrkA receptors in sympathetic neurons, and we define the molecular underpinnings of this positive feedback regulation that enhances neuronal sensitivity to trophic factors. Activated TrkA receptors are retrogradely transported in signaling endosomes from distal axons to cell bodies, where they are inserted on soma surfaces and promote phosphorylation of resident naive receptors, resulting in their internalization. Endocytosed TrkA receptors are then dephosphorylated by PTP1B, an ER-resident protein tyrosine phosphatase, prior to axonal transport. PTP1B inactivation prevents TrkA exit from soma and causes receptor degradation, suggesting a "gatekeeper" mechanism that ensures targeting of inactive receptors to axons to engage with ligand. In mice, PTP1B deletion reduces axonal TrkA levels and attenuates neuron survival and target innervation under limiting NGF (NGF+/-) conditions.


Assuntos
Axônios/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Transcitose , Animais , Axônios/efeitos dos fármacos , Corpo Celular/efeitos dos fármacos , Corpo Celular/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Deleção de Genes , Células HEK293 , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos , Fatores de Crescimento Neural/farmacologia , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Proteólise/efeitos dos fármacos , Ratos Sprague-Dawley , Transcitose/efeitos dos fármacos
5.
Microb Pathog ; 112: 1-4, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28923601

RESUMO

Studies show that highly diluted medications demonstrate benefits in treating infections, constituting an alternative for their treatment. The present study evaluated the effects of Lycopodium clavatum, dynamization 13c, in Wistar rats infected with T. cruzi. In this study 42 male rats were intraperitoneally inoculated with T. cruzi - Y strain and allocated into groups: IC (infected control group) and Ly (treated with L. clavatum 13c). The cytokines dosage (IFN-γ, IL-12, IL-10, IL-4), quantification and morphometry of myenteric neurons were evaluated. The treatment with L. clavatum modifies the immune response, with increase of IFN-γ on day 10 a.i. and IL-12 on day 24 a.i., decrease of IL-10 concentration on day 10 a.i. and subsequent increase of this cytokine and IL-4 on day 24 a.i., affording a bigger number of myenteric neurons compared to IC group. Thus, L. clavatum 13c promoted on rats infected with T. cruzi a beneficial immunomodulatory action reducing the pathogenic progression of digestive Chagas disease.


Assuntos
Doença de Chagas/imunologia , Imunomodulação , Lycopodium/química , Neurônios/imunologia , Extratos Vegetais/farmacologia , Trypanosoma cruzi/efeitos dos fármacos , Animais , Corpo Celular/efeitos dos fármacos , Corpo Celular/imunologia , Corpo Celular/parasitologia , Corpo Celular/patologia , Doença de Chagas/tratamento farmacológico , Colo/inervação , Colo/parasitologia , Colo/patologia , Citocinas/metabolismo , Digestão , Modelos Animais de Doenças , Homeopatia , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Interleucina-4/metabolismo , Masculino , Neurônios/efeitos dos fármacos , Neurônios/parasitologia , Neurônios/patologia , Ratos , Ratos Wistar , Trypanosoma cruzi/imunologia , Trypanosoma cruzi/patogenicidade
6.
Learn Mem ; 24(8): 341-357, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28716954

RESUMO

High-frequency stimulation of the medial perforant path triggers robust phosphorylation of ribosomal protein S6 (rpS6) in activated dendritic domains and granule cell bodies. Here we dissect the signaling pathways responsible for synaptically driven rpS6 phosphorylation in the dentate gyrus using pharmacological agents to inhibit PI3-kinase/mTOR and MAPK/ERK-dependent kinases. Using phospho-specific antibodies for rpS6 at different sites (ser235/236 versus ser240/244), we show that delivery of the PI3-kinase inhibitor, wortmannin, decreased rpS6 phosphorylation throughout the somatodendritic compartment (granule cell layer, inner molecular layer, outer molecular layer), especially in granule cell bodies while sparing phosphorylation at activated synapses (middle molecular layer). In contrast, delivery of U0126, an MEK inhibitor, attenuated rpS6 phosphorylation specifically in the dendritic laminae leaving phosphorylation in the granule cell bodies intact. Delivery of the mTOR inhibitor, rapamycin, abolished activation of rpS6 phosphorylation in granule cell bodies and dendrites, whereas delivery of a selective S6K1 inhibitor, PF4708671, or RSK inhibitor, SL0101-1, attenuated rpS6 phosphorylation throughout the postsynaptic cell. These results reveal that MAPK/ERK-dependent signaling is predominately responsible for the selective induction of rpS6 phosphorylation at active synapses. In contrast, PI3-kinase/mTOR-dependent signaling induces rpS6 phosphorylation throughout the somatodendritic compartment but plays a minimal role at active synapses. Collectively, these results suggest a potential mechanism by which PI3-kinase/mTOR and MAPK/ERK pathways regulate translation at specific subcellular compartments in response to synaptic activity.


Assuntos
Corpo Celular/metabolismo , Dendritos/metabolismo , Sistema de Sinalização das MAP Quinases , Fosfatidilinositol 3-Quinases/metabolismo , Proteína S6 Ribossômica/metabolismo , Sinapses/metabolismo , Androstadienos/farmacologia , Animais , Benzopiranos/farmacologia , Butadienos/farmacologia , Corpo Celular/efeitos dos fármacos , Cromonas/farmacologia , Dendritos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Monossacarídeos/farmacologia , Morfolinas/farmacologia , Nitrilas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Piperazinas/farmacologia , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Quinases S6 Ribossômicas/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas/metabolismo , Sirolimo/farmacologia , Sinapses/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Wortmanina
7.
Neuropharmacology ; 123: 385-394, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28603026

RESUMO

Although MDMA (3,4-methylendioxymethamphetamine, ecstasy) neurotoxicity in serotonin neurons is largely recognized in a wide variety of species including man, neurotoxicity in dopamine (DA) neurons is thought to be species-specific. MDMA is mainly consumed by adolescents, often in conjunction with caffeine (Energy Drinks) and this association has been reported to exacerbate MDMA toxic effects. In order to model these aspects of MDMA use, vis-à-vis their impact on DA neurons, we investigated the effects of adolescent exposure to low doses of MDMA (5 mg/kg for 10 days), alone or in combination with caffeine (10 mg/kg) on neuronal and functional DA indices and on recognition memory in adult rats. MDMA reduced density of tyrosine hydroxylase (TH) positive neurons in the ventral tegmental area and in the substantia nigra pars compacta, and immunoreactivity of TH and DA transporter in the nucleus accumbens (NAc) shell and core, and caudate-putamen. This same treatment caused a reduction of basal dialysate DA in the NAc core. MDMA-pretreated rats also showed behavioral sensitization to a MDMA challenge at adulthood and potentiation of MDMA-induced increase of dialysate DA in the NAc core, but not in the NAc shell. In addition, MDMA-treated rats displayed a deficit in recognition memory. Caffeine co-administration did not affect the above outcomes. Our results show that adolescent exposure of rats to low doses of MDMA induces long-lasting and widespread reduction of DA neurons indicative of a neurotoxic effect on DA neurons and suggestive of a degeneration of the same neurons.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Transtornos da Memória/induzido quimicamente , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Cafeína/toxicidade , Corpo Celular/efeitos dos fármacos , Corpo Celular/patologia , Contagem de Células , Neurônios Dopaminérgicos/metabolismo , Interações Medicamentosas , Imunofluorescência , Masculino , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Ratos Sprague-Dawley , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Tirosina 3-Mono-Oxigenase/metabolismo
8.
Toxicol Sci ; 156(1): 275-288, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28115644

RESUMO

Chemotherapy-induced peripheral neuropathy (CIPN) is a major, dose-limiting adverse effect experienced by cancer patients. Advancements in mechanism-based risk mitigation and effective treatments for CIPN can be aided by suitable in vitro assays. To this end, we developed a multiparametric morphology-centered rat dorsal root ganglion (DRG) assay. Morphologic alterations in subcellular structures of neurons and non-neurons were analyzed with an automated microscopy system. Stains for NeuN (a neuron-specific nuclear protein) and Tuj-1 (ß-III tubulin) were used to identify neuronal cell nuclei and neuronal cell bodies/neurites, respectively. Vimentin staining (a component of Schwann cell intermediate filaments) was used to label non-neuronal supporting cells. Nuclei that stained with DAPI, but lacked NeuN represented non-neuronal cells. Images were analyzed following 24 h of continuous exposure to CIPN-inducing agents and 72 h after drug removal to provide a dynamic measure of recovery from initial drug effects. Treatment with bortezomib, cisplatin, eribulin, paclitaxel or vincristine induced a dose-dependent loss of neurite/process areas, mimicking the 'dying back' degeneration of axons, a histopathological hallmark of clinical CIPN in vivo. The IC50 for neurite loss was within 3-fold of the maximal clinical exposure (Cmax) for all five CIPN-inducing drugs, but was >4- or ≥ 28-fold of the Cmax for 2 non-CIPN-inducing agents. Compound-specific effects, eg, neurite fragmentation by cisplatin or bortezomib and enlarged neuronal cell bodies by paclitaxel, were also observed. Collectively, these results support the use of a quantitative, morphologic evaluation and a DRG cell culture model to inform risk and examine mechanisms of CIPN.


Assuntos
Antineoplásicos/efeitos adversos , Gânglios Espinais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Corpo Celular/efeitos dos fármacos , Corpo Celular/metabolismo , Corpo Celular/patologia , Forma do Núcleo Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/métodos , Eletroforese Capilar , Imunofluorescência , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Processamento de Imagem Assistida por Computador , Cinética , Peso Molecular , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neuritos/patologia , Neurônios/metabolismo , Neurônios/patologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Forma das Organelas/efeitos dos fármacos , Tamanho das Organelas/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/etiologia , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Ratos
9.
Cell Rep ; 17(5): 1238-1246, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27783939

RESUMO

The second messenger cyclic AMP (cAMP) plays an important role in synaptic plasticity. Although there is evidence for local control of synaptic transmission and plasticity, it is less clear whether a similar spatial confinement of cAMP signaling exists. Here, we suggest a possible biophysical basis for the site-specific regulation of synaptic plasticity by cAMP, a highly diffusible small molecule that transforms the physiology of synapses in a local and specific manner. By exploiting the octopaminergic system of Drosophila, which mediates structural synaptic plasticity via a cAMP-dependent pathway, we demonstrate the existence of local cAMP signaling compartments of micrometer dimensions within single motor neurons. In addition, we provide evidence that heterogeneous octopamine receptor localization, coupled with local differences in phosphodiesterase activity, underlies the observed differences in cAMP signaling in the axon, cell body, and boutons.


Assuntos
AMP Cíclico/metabolismo , Drosophila melanogaster/metabolismo , Neurônios Motores/metabolismo , Terminações Pré-Sinápticas/metabolismo , Transdução de Sinais , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Corpo Celular/efeitos dos fármacos , Corpo Celular/metabolismo , Drosophila melanogaster/efeitos dos fármacos , Transferência Ressonante de Energia de Fluorescência , Iontoforese , Neurônios Motores/efeitos dos fármacos , Octopamina/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
10.
J Nutr Biochem ; 38: 41-49, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27721115

RESUMO

Folic acid (FA) deficiency is not only associated with an increased risk of ischemic stroke, but also with increased oxidative DNA damage and brain injury after cerebral ischemia-reperfusion. However, the cellular and molecular mechanisms underlying FA deficiency-associated neuropathogenesis are not completely understood. In the present study, we tested the hypothesis that neuronal autophagy in focal cerebral ischemia rats may be involved in the mechanisms of FA deficiency-induced injury to neuronal cells. The results demonstrated that, accompanied by obvious neuron damage, the expression of the autophagic markers LC3 and Beclin-1, and the formation of 8-OHdG (a marker of oxidative stress to DNA) and autophagosomes were significantly increased in the brain cortex after ischemia-reperfusion. FA deficiency further induced neuronal cell death, and significantly increased the formation of autophagosomes and the expression of LC3 and Beclin-1 in NeuN-positive cell bodies after ischemia-reperfusion. The elevated level of 8-OHdG was also observed in the ischemic cortex of FA deficiency-treated animals. Conversely, the neuronal cell injury, autophagosome accumulation and the effects of LC3 and Beclin1 overexpression caused by FA deficiency were partially blocked by an autophagic inhibitor 3-methyladenine. These results suggest that FA deficiency progresses autophagic activation and aggravates the damage in rat brain cortex following focal cerebral ischemia-reperfusion. The oxidative injury may be involved in cell morphological damage and autophagy alteration caused by FA deficiency.


Assuntos
Autofagia , Isquemia Encefálica/complicações , Córtex Cerebral/patologia , Deficiência de Ácido Fólico/complicações , Neurônios/patologia , Estresse Oxidativo , Regulação para Cima , Adenina/análogos & derivados , Adenina/uso terapêutico , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagossomos/patologia , Autofagossomos/ultraestrutura , Autofagia/efeitos dos fármacos , Proteína Beclina-1/metabolismo , Biomarcadores/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Corpo Celular/efeitos dos fármacos , Corpo Celular/metabolismo , Corpo Celular/patologia , Corpo Celular/ultraestrutura , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/ultraestrutura , Dano ao DNA/efeitos dos fármacos , Deficiência de Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/patologia , Masculino , Microscopia Eletrônica de Transmissão , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/ultraestrutura , Estresse Oxidativo/efeitos dos fármacos , Distribuição Aleatória , Ratos Sprague-Dawley , Traumatismo por Reperfusão/prevenção & controle , Organismos Livres de Patógenos Específicos , Regulação para Cima/efeitos dos fármacos
11.
An Acad Bras Cienc ; 88 Suppl 1: 609-22, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27142540

RESUMO

The objective of this study was to investigate the effects of 2% L-glutamine supplementation on myenteric innervation in the ileum of diabetic rats, grouped as follows: normoglycemic (N); normoglycemic supplemented with L-glutamine (NG); diabetic (D); and diabetic supplemented with L-glutamine (DG). The ileums were subjected to immunohistochemical techniques to localize neurons immunoreactive to HuC/D protein (HuC/D-IR) and neuronal nitric oxide synthase enzyme (nNOS-IR) and to analyze varicosities immunoreactive to vasoactive intestinal polypeptide (VIP-IR) and calcitonin gene-related peptide (CGRP-IR). L-Glutamine in the DG group (i) prevented the increase in the cell body area of nNOS-IR neurons, (ii) prevented the increase in the area of VIP-IR varicosities, (iii) did not prevent the loss of HuC/D-IR and nNOS-IR neurons per ganglion, and (iv) reduced the size of CGRP-IR varicosities. L-Glutamine in the NG group reduced (i) the number of HuC/D-IR and nNOS-IR neurons per ganglion, (ii) the cell body area of nNOS-IR neurons, and (iii) the size of VIP-IR and CGRP-IR varicosities. 2% L-glutamine supplementation exerted differential neuroprotective effects in experimental diabetes neuropathy that depended on the type of neurotransmitter analyzed. However, the effects of this dose of L-glutamine on normoglycemic animals suggests there are additional actions of this beyond its antioxidant capacity.


Assuntos
Diabetes Mellitus Experimental , Glutamina/farmacologia , Íleo/inervação , Plexo Mientérico/efeitos dos fármacos , Animais , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Corpo Celular/efeitos dos fármacos , Glutamina/administração & dosagem , Imuno-Histoquímica , Neurônios/efeitos dos fármacos , Neurônios Nitrérgicos , Óxido Nítrico Sintase Tipo I/farmacologia , Ratos , Ratos Wistar , Peptídeo Intestinal Vasoativo/farmacologia
12.
PLoS One ; 11(5): e0155317, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27171274

RESUMO

In oligodendrocytes (OLGs), an indirect, transcytotic pathway is mediating transport of de novo synthesized PLP, a major myelin specific protein, from the apical-like plasma membrane to the specialized basolateral-like myelin membrane to prevent its premature compaction. MAL is a well-known regulator of polarized trafficking in epithelial cells, and given its presence in OLGs it was therefore of interest to investigate whether MAL played a similar role in PLP transport in OLGs, taking into account its timely expression in these cells. Our data revealed that premature expression of mCherry-MAL in oligodendrocyte progenitor cells interfered with terminal OLG differentiation, although myelin membrane formation per se was not impaired. In fact, also PLP transport to myelin membranes via the cell body plasma membrane was unaffected. However, the typical shift of PLP from TX-100-insoluble membrane domains to CHAPS-resistant, but TX-100-soluble membrane domains, seen in the absence of MAL expression, is substantially reduced upon expression of the MAL protein. Interestingly, not only in vitro, but also in developing brain a strongly diminished shift from TX-100 resistant to TX-100 soluble domains was observed. Consistently, the MAL-expression mediated annihilation of the typical membrane microdomain shift of PLP is also reflected by a loss of the characteristic surface expression profile of conformation-sensitive anti-PLP antibodies. Hence, these findings suggest that MAL is not involved in vesicular PLP trafficking to either the plasma membrane and/or the myelin membrane as such. Rather, we propose that MAL may regulate PLP's distribution into distinct membrane microdomains that allow for lateral diffusion of PLP, directly from the plasma membrane to the myelin membrane once the myelin sheath has been assembled.


Assuntos
Microdomínios da Membrana/metabolismo , Proteína Proteolipídica de Mielina/metabolismo , Proteínas Proteolipídicas Associadas a Linfócitos e Mielina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Corpo Celular/efeitos dos fármacos , Corpo Celular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Detergentes/farmacologia , Feminino , Células Hep G2 , Humanos , Microdomínios da Membrana/efeitos dos fármacos , Modelos Biológicos , Octoxinol/farmacologia , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos Wistar , Solubilidade , Células-Tronco/citologia , Células-Tronco/metabolismo
13.
Asian Pac J Trop Med ; 7(8): 655-658, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25149381

RESUMO

OBJECTIVE: To explore mechanism of nduction of fibroblast to corneal endothelial cell. METHODS: Rabbit conjunctiva fibroblasts were used as feeder cells, rabbit oral mucosa epithelial cells were used as seed cells, and human denuded amniotic membrane was used as carrier to establish tissue engineering corneal endothelium. The transformation effect was observed. RESULTS: As concentration of mitomycin C increased, cell survival rate gradually decreased, cell proliferation was obviously inhibited when concentration≥25 µg/mL; 5 days after being treated by 5 µg/mL mitomycin C, cell body was enlarged and extended without cell fusion, however after being treated by 0.5 µg/mL mitomycin C, cell body was significantly proliferated and gradually fused; after 3 weeks of culture, stratified epithelium appeared on rabbit oral mucosa epithelial cells, differentiation layers were 4-5 and were well differentiated, the morphology was similar to corneal endothelial cells; Under electron microscope, surface layer of cells were polygonal, tightly connected to another with microvilli on the border, there was hemidesmosome between basal cells and human denuded amniotic membrane. CONCLUSIONS: Fibroblast cells have the potential of multi-directional differentiation, effective induction can promote emergence of intercellular desmosomes between seed cells and emergence of epithelial surface microvilli, and differentiate to the corneal endothelial cell. However, clinical application still needs more research and safety evaluation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Endotélio Corneano/citologia , Células Epiteliais/citologia , Fibroblastos/citologia , Mitomicina/farmacologia , Âmnio/citologia , Animais , Corpo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Humanos , Mucosa Bucal/citologia , Coelhos
14.
Biomed Res Int ; 2014: 191767, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25050325

RESUMO

A persistent inflammatory and oxidative stress is a hallmark of most chronic CNS pathologies (Alzheimer's (ALS)) as well as the aging CNS orchestrated by the proinflammatory cytokines tumor necrosis factor alpha (TNFα) and interleukin-1 beta (IL-1ß). Loss of the integrity and plasticity of neuronal morphology and connectivity comprises an early step in neuronal degeneration and ultimate decline of cognitive function. We examined in vitro whether TNFα or IL-1ß impaired morphology and motility of growth cones in spinal cord neuron cultures. TNFα and IL-1ß paralyzed growth cone motility and induced growth cone collapse in a dose-dependent manner reflected by complete attenuation of neurite outgrowth. Scavenging reactive oxygen species (ROS) or inhibiting NADPH oxidase activity rescued loss of neuronal motility and morphology. TNFα and IL-1ß provoked rapid, NOX-mediated generation of ROS in advancing growth cones, which preceded paralysis of motility and collapse of morphology. Increases in ROS intermediates were accompanied by an aberrant, nonproductive reorganization of actin filaments. These findings suggest that NADPH oxidase serves as a pivotal source of oxidative stress in neurons and together with disruption of actin filament reorganization contributes to the progressive degeneration of neuronal morphology in the diseased or aging CNS.


Assuntos
Cones de Crescimento/patologia , Mediadores da Inflamação/toxicidade , Interleucina-1beta/toxicidade , Paralisia/patologia , Espécies Reativas de Oxigênio/toxicidade , Medula Espinal/patologia , Fator de Necrose Tumoral alfa/toxicidade , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Animais , Corpo Celular/efeitos dos fármacos , Corpo Celular/metabolismo , Embrião de Galinha , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/enzimologia , Humanos , Modelos Biológicos , NADPH Oxidases/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Oxirredução/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
15.
Mol Neurobiol ; 50(1): 49-59, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24515839

RESUMO

Inhibitors of the mevalonate pathway, including the highly prescribed statins, reduce the production of cholesterol and isoprenoids such as geranylgeranyl pyrophosphates. The Rho family of small guanine triphosphatases (GTPases) requires isoprenylation, specifically geranylgeranylation, for activation. Because Rho GTPases are primary regulators of actin filament rearrangements required for process extension, neurite arborization, and synaptic plasticity, statins may affect cognition or recovery from nervous system injury. Here, we assessed how manipulating geranylgeranylation affects neurite initiation, elongation, and branching in neuroblastoma growth cones. Treatment with the statin, lovastatin (20 µM), decreased measures of neurite initiation by 17.0 to 19.0 % when a source of cholesterol was present and increased neurite branching by 4.03- to 9.54-fold (regardless of exogenous cholesterol). Neurite elongation was increased by treatment with lovastatin only in cholesterol-free culture conditions. Treatment with lovastatin decreased growth cone actin filament content by up to 24.3 %. In all cases, co-treatment with the prenylation precursor, geranylgeraniol (10 µM), reversed the effect of lovastatin. In a prior work, statin effects on outgrowth were linked to modulating the actin depolymerizing factor, cofilin. In our assays, treatment with lovastatin or geranylgeraniol decreased cofilin phosphorylation in whole cell lysates. However, lovastatin increased cofilin phosphorylation in cell bodies and decreased it in growth cones, indicating differential regulation in specific cell regions. Together, we interpret these data to suggest that protein geranylgeranylation likely regulates growth cone actin filament content and subsequent neurite outgrowth through mechanisms that also affect actin nucleation and polymerization.


Assuntos
Corpo Celular/metabolismo , Cofilina 1/metabolismo , Cones de Crescimento/metabolismo , Neuritos/metabolismo , Prenilação de Proteína/fisiologia , Animais , Corpo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Diterpenos/farmacologia , Cones de Crescimento/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Neuritos/efeitos dos fármacos , Prenilação de Proteína/efeitos dos fármacos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA