Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.243
Filtrar
1.
Eur J Pharmacol ; 970: 176510, 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38493917

RESUMO

Activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathway protects against N-methyl-D-aspartic acid (NMDA)-induced excitotoxic retinal injury. AMPK activation enhances fatty acid metabolism and ketone body synthesis. Ketone bodies are transported into neurons by monocarboxylate transporters (MCTs) and exert neuroprotective effects. In this study, we examined the distribution and expression levels of MCT1 and MCT2 in the retina and analyzed the effects of pharmacological inhibition of MCTs on the protective effects of metformin and 5-aminoimidazole-4-carboxamide (AICAR), activators of AMPK, against NMDA-induced retinal injury in rats. MCT1 was expressed in the blood vessels, processes of astrocytes and Müller cells, and inner segments of photoreceptors in the rat retina, whereas MCT2 was expressed in neuronal cells in the ganglion cell layer (GCL) and in astrocyte processes. The expression levels of MCT2, but not MCT1, decreased one day after intravitreal injection of NMDA (200 nmol). Intravitreal injection of NMDA decreased the number of cells in the GCL compared to the vehicle seven days after injection. Simultaneous injection of metformin (20 nmol) or AICAR (50 nmol) with NMDA attenuated NMDA-induced cell loss in the GCL, and these protective effects were attenuated by AR-C155858 (1 pmol), an inhibitor of MCTs. AR-C155858 alone had no significant effect on the retinal structure. These results suggest that AMPK-activating compounds protect against NMDA-induced excitotoxic retinal injury via mechanisms involving MCTs in rats. NMDA-induced neurotoxicity may be associated with retinal neurodegenerative changes in glaucoma and diabetic retinopathy. Therefore, AMPK-activating compounds may be effective in managing these retinal diseases.


Assuntos
Metformina , Doenças Retinianas , Tiofenos , Uracila/análogos & derivados , Ratos , Animais , Proteínas Quinases Ativadas por AMP/metabolismo , N-Metilaspartato/toxicidade , Ratos Sprague-Dawley , Retina/metabolismo , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/prevenção & controle , Doenças Retinianas/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Metformina/efeitos adversos
2.
Acta Neuropathol Commun ; 12(1): 19, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38303097

RESUMO

Excitotoxicity from the impairment of glutamate uptake constitutes an important mechanism in neurodegenerative diseases such as Alzheimer's, multiple sclerosis, and Parkinson's disease. Within the eye, excitotoxicity is thought to play a critical role in retinal ganglion cell death in glaucoma, diabetic retinopathy, retinal ischemia, and optic nerve injury, yet how excitotoxic injury impacts different retinal layers is not well understood. Here, we investigated the longitudinal effects of N-methyl-D-aspartate (NMDA)-induced excitotoxic retinal injury in a rat model using deep learning-assisted retinal layer thickness estimation. Before and after unilateral intravitreal NMDA injection in nine adult Long Evans rats, spectral-domain optical coherence tomography (OCT) was used to acquire volumetric retinal images in both eyes over 4 weeks. Ten retinal layers were automatically segmented from the OCT data using our deep learning-based algorithm. Retinal degeneration was evaluated using layer-specific retinal thickness changes at each time point (before, and at 3, 7, and 28 days after NMDA injection). Within the inner retina, our OCT results showed that retinal thinning occurred first in the inner plexiform layer at 3 days after NMDA injection, followed by the inner nuclear layer at 7 days post-injury. In contrast, the retinal nerve fiber layer exhibited an initial thickening 3 days after NMDA injection, followed by normalization and thinning up to 4 weeks post-injury. Our results demonstrated the pathological cascades of NMDA-induced neurotoxicity across different layers of the retina. The early inner plexiform layer thinning suggests early dendritic shrinkage, whereas the initial retinal nerve fiber layer thickening before subsequent normalization and thinning indicates early inflammation before axonal loss and cell death. These findings implicate the inner plexiform layer as an early imaging biomarker of excitotoxic retinal degeneration, whereas caution is warranted when interpreting the ganglion cell complex combining retinal nerve fiber layer, ganglion cell layer, and inner plexiform layer thicknesses in conventional OCT measures. Deep learning-assisted retinal layer segmentation and longitudinal OCT monitoring can help evaluate the different phases of retinal layer damage upon excitotoxicity.


Assuntos
Aprendizado Profundo , Degeneração Retiniana , Ratos , Animais , Degeneração Retiniana/induzido quimicamente , Degeneração Retiniana/diagnóstico por imagem , Degeneração Retiniana/patologia , Tomografia de Coerência Óptica/métodos , N-Metilaspartato/toxicidade , Ratos Long-Evans , Retina/patologia , Células Ganglionares da Retina/patologia , Fibras Nervosas/patologia
3.
Exp Eye Res ; 238: 109740, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38056553

RESUMO

Glutamate induced damage to retinal ganglion cells (RGCs) requires tight physiological regulation of the N-methyl-D-aspartate (NMDA) receptors. Previously, studies have demonstrated the neuroprotective abilities of antioxidants like coenzyme Q10 (CoQ10) and vitamin E analogs like α-tocopherol against neuropathies resulting from NMDA insult, but have failed to shed light on the effect of CoQ10 and trolox, a hydrophilic analog of vitamin E, on glaucomatous neurodegeneration. In the current study, we wanted to investigate whether the combined effect of trolox with CoQ10 could alleviate NMDA-induced death of retinal cells while also trying to elucidate the underlying mechanism in relation to the yet unexplained role of vascular endothelial growth factor (VEGF) in NMDA-mediated excitotoxicity. After successful NMDA-induced degeneration, we followed it up with the treatment of combination of Trolox and CoQ10. The structural damage by NMDA was repaired significantly and retina retained structural integrity comparable to levels of control in the treatment group of Trolox and CoQ10. Detection of ROS generation after NMDA insult showed that together, Trolox and CoQ10 could significantly bring down the high levels of free radicals while also rescuing mitochondrial membrane potential (MMP). A significant increase in NMDA receptor Grin2A by CoQ10 alone as well as by CoQ10 and trolox was accompanied by a lowered Grin2B receptor expression, suggesting neuroprotective action of Trolox and CoQ10. Subsequently, lowered VEGFR1 and VEGFR2 receptor expression by NMDA treatment also recovered when subjected to combined treatment of Trolox and CoQ10. Western blot analyses also indicated the same whereby Trolox and CoQ10 could increase the diminished levels of phosphorylated VEGFR2. Immunofluorescence studies also indicated a positive correlation between recovered VEGFR2 and NMDAR2A levels and diminished levels of NMDAR2D, confirming the results obtained by RT-PCR analysis. This is the first report in our knowledge that demonstrates the efficacy of trolox in combination with CoQ10 highlighting the importance of maintaining VEGF levels that are lowered in ocular diseases due to NMDA-related toxicities.


Assuntos
Ubiquinona , Fator A de Crescimento do Endotélio Vascular , Ratos , Animais , Ubiquinona/farmacologia , Ubiquinona/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , N-Metilaspartato/toxicidade , Ácido Glutâmico/toxicidade , Ácido Glutâmico/metabolismo , Neuroproteção , Regulação para Cima , Vitamina E
4.
J Vasc Res ; 60(3): 137-147, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37285812

RESUMO

N-methyl-D-aspartate (NMDA) receptors were found to be dysfunctional in hypertensive rats. Methyl palmitate (MP) has been shown to diminish the nicotine-induced increase in blood flow in the brainstem. The aim of this study was to determine how MP modulated NMDA-induced increased regional cerebral blood flow (rCBF) in normotensive (WKY), spontaneously hypertensive (SHR), and renovascular hypertensive (RHR) rats. The increase in rCBF after the topical application of experimental drugs was measured using laser Doppler flowmetry. Topical NMDA application induced an MK-801-sensitive increase in rCBF in anesthetized WKY rats, which was inhibited by MP pretreatments. This inhibition was prevented by pretreatment with chelerythrine (a PKC inhibitor). The NMDA-induced increase in rCBF was also inhibited by the PKC activator in a concentration-dependent manner. Neither MP nor MK-801 affected the increase in rCBF induced by the topical application of acetylcholine or sodium nitroprusside. Topical application of MP to the parietal cortex of SHRs, on the other hand, increased basal rCBF slightly but significantly. MP enhanced the NMDA-induced increase in rCBF in SHRs and RHRs. These results suggested that MP had a dual effect on the modulation of rCBF. MP appears to play a significant physiological role in CBF regulation.


Assuntos
Hiperemia , Hipertensão , Ratos , Animais , N-Metilaspartato/toxicidade , Ratos Endogâmicos WKY , Ratos Endogâmicos SHR , Maleato de Dizocilpina/farmacologia , Hipertensão/induzido quimicamente , Receptores de N-Metil-D-Aspartato/fisiologia
5.
Int Immunopharmacol ; 118: 109976, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37098655

RESUMO

Glaucoma, one of the most common ocular neurodegenerative diseases worldwide, is characterized by retinal ganglion cell (RGC) loss. There is a large body of literature that describes the neuroprotective role of melatonin against neurodegenerative diseases by regulating neuroinflammation, although the exact mechanism through which melatonin acts on RGC is still uncertain. This study assessed the protective effects of melatonin using a NMDA-induced RGC injury model, and studied the possible mechanisms involved in this process. Melatonin promoted RGC survival, improved retinal function, and inhibited the apoptosis and necrosis of retinal cells. To understand the mechanism of the neuroprotective effects of melatonin on RGC, microglia and inflammation-related pathways were assessed after melatonin administration and microglia ablation. Melatonin promoted RGC survival by suppressing microglia-derived proinflammatory cytokines, in particular TNFα, which in turn inhibited the activation of p38 MAPK pathway. Inhibiting TNFα or manipulating p38 MAPK pathway protected damaged RGC. Our results suggest that melatonin protects against NMDA-induced RGC injury by inhibiting the microglial TNFα-RGC p38 MAPK pathway. It should be considered a candidate neuroprotective therapy against retinal neurodegenerative diseases.


Assuntos
Melatonina , Células Ganglionares da Retina , Microglia , N-Metilaspartato/toxicidade , N-Metilaspartato/metabolismo , Melatonina/farmacologia , Melatonina/uso terapêutico , Melatonina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Apoptose
6.
Mol Brain ; 16(1): 23, 2023 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-36774487

RESUMO

Jacob is a synapto-nuclear messenger protein that encodes and transduces the origin of synaptic and extrasynaptic NMDA receptor signals to the nucleus. The protein assembles a signalosome that differs in case of synaptic or extrasynaptic NMDAR activation. Following nuclear import Jacob docks these signalosomes to the transcription factor CREB. We have recently shown that amyloid-ß and extrasynaptic NMDAR activation triggers the translocation of a Jacob signalosome that results in inactivation of the transcription factor CREB, a phenomenon termed Jacob-induced CREB shut-off (JaCS). JaCS contributes to early Alzheimer's disease pathology and the absence of Jacob protects against amyloid pathology. Given that extrasynaptic activity is also involved in acute excitotoxicity, like in stroke, we asked whether nsmf gene knockout will also protect against acute insults, like oxygen and glucose deprivation and excitotoxic NMDA stimulation. nsmf is the gene that encodes for the Jacob protein. Here we show that organotypic hippocampal slices from wild-type and nsmf-/- mice display similar degrees of degeneration when exposed to either oxygen glucose deprivation or 50 µM NMDAto induce excitotoxicity. This lack of neuroprotection indicates that JaCS is mainly relevant in conditions of low level chronic extrasynaptic NMDAR activation that results in cellular degeneration induced by alterations in gene transcription.


Assuntos
Morte Celular , Hipóxia , N-Metilaspartato , Proteínas do Tecido Nervoso , Neurônios , Animais , Camundongos , Técnicas de Inativação de Genes , Glucose , Hipóxia/metabolismo , N-Metilaspartato/toxicidade , Neurônios/metabolismo , Oxigênio , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Fatores de Transcrição/metabolismo , Proteínas do Tecido Nervoso/genética
7.
Neurosci Res ; 193: 1-12, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36796452

RESUMO

Adenosine A1 receptors (AA1R) have been shown to counteract N-methyl-D-aspartate (NMDA)-mediated glutamatergic excitotoxicity. In the present study, we investigated the role of AA1R in neuroprotection by trans-resveratrol (TR) against NMDA-induced retinal injury. In total, 48 rats were divided into the following four groups: normal rats pretreated with vehicle; rats that received NMDA (NMDA group); rats that received NMDA after pretreatment with TR; and rats that received NMDA after pretreatment with TR and 1,3-dipropyl-8-cyclopentylxanthine (DPCPX), an AA1R antagonist. Assessment of general and visual behaviour was performed using the open field test and two-chamber mirror test, respectively, on Days 5 and 6 post NMDA injection. Seven days after NMDA injection, animals were euthanized, and eyeballs and optic nerves were harvested for histological parameters, whereas retinae were isolated to determine the redox status and expression of pro- and anti-apoptotic proteins. In the present study, the retinal and optic nerve morphology in the TR group was protected from NMDA-induced excitotoxic damage. These effects were correlated with the lower retinal expression of proapoptotic markers, lipid peroxidation, and markers of nitrosative/oxidative stress. The general and visual behavioural parameters in the TR group showed less anxiety-related behaviour and better visual function than those in the NMDA group. All the findings observed in the TR group were abolished by administration of DPCPX.


Assuntos
N-Metilaspartato , Receptor A1 de Adenosina , Ratos , Animais , N-Metilaspartato/toxicidade , Resveratrol , Ratos Sprague-Dawley , Neuroproteção , Receptores de N-Metil-D-Aspartato
8.
Pharmacol Rep ; 75(1): 177-188, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36422805

RESUMO

BACKGROUND: Neurosteroids are investigated as effective antidotes for the poisoning induced by tetramethylenedisulfotetramine (TMDT) as well as treatments for epileptic spasms during infancy. Both these conditions are quite resistant to pharmacotherapy; thus, a search for new treatments is warranted. METHODS: In this study, we determined the efficacy of two novel neurosteroids, pregnanolone glutamate (PAG) and pregnanolone pyroglutamate (PPG), and tested these drugs in doses of 1-10 mg/kg (ip) against the TMDT syndrome and in our rodent model of infantile spasms. RESULTS: Only PPG in doses 5 and 10 mg/kg suppressed the severity of the TMDT syndrome and TMDT-induced lethality, while the 1 mg/kg dose was without an effect. Interestingly, the 1 mg/kg dose of PPG in combination with 1 mg/kg of diazepam was also effective against TMDT poisoning. Neither PAG nor PPG were effective against experimental spasms in the N-methyl-D-aspartate (NMDA)-triggered model of infantile spasms. CONCLUSIONS: While evidence suggests that PAG can act through multiple actions which include allosteric inhibition of NMDA-induced and glycine receptor-evoked currents as well as augmentation of É£-aminobutyric acid subtype A (GABAA) receptor-induced currents, the agent appears to neither have the appropriate mechanistic signature for activity in the infantile spasm model, nor the adequate potency, relative to PPG, for ameliorating the TMDT syndrome. The full mechanisms of action of PPG, which may become a potent TMDT antidote either alone or in combination with diazepam are yet unknown and thus require further investigation.


Assuntos
Neuroesteroides , Síndromes Neurotóxicas , Espasmos Infantis , Animais , Espasmos Infantis/induzido quimicamente , Espasmos Infantis/tratamento farmacológico , Pregnanolona/efeitos adversos , Ácido Pirrolidonocarboxílico , N-Metilaspartato/toxicidade , N-Metilaspartato/uso terapêutico , Roedores , Diazepam/farmacologia , Ácido Glutâmico , Espasmo
9.
Front Endocrinol (Lausanne) ; 13: 986131, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36313740

RESUMO

Glaucoma is the leading cause of irreversible blindness. The progressive degeneration of retinal ganglion cells (RGCs) is the major characteristic of glaucoma. Even though the control of intraocular pressure could delay the loss of RGCs, current clinical treatments cannot protect them directly. The overactivation of N-methyl-D-aspartic acid (NMDA) receptors by excess glutamate (Glu) is among the important mechanisms of RGC death in glaucoma progression. Melatonin (MT) is an indole neuroendocrine hormone mainly secreted by the pineal gland. This study aimed to investigate the therapeutic effect of MT on glutamate excitotoxicity of mouse RGCs and R28 cells. The Glu-induced R28 cell excitotoxicity model and NMDA-induced retinal injury model were established. MT was applied to R28 cells and the vitreous cavity of mice by intravitreal injection. Cell counting kit-8 assay and propidium iodide/Hoechst were performed to evaluate cell viability. Reactive oxygen species and glutathione synthesis assays were used to detect the oxidative stress state of R28 cells. Retina immunofluorescence and hematoxylin and eosin staining were applied to assess RGC counts and retinal structure. Flash visual-evoked potential was performed to evaluate visual function in mice. RNA sequencing of the retina was performed to explore the underlying mechanisms of MT protection. Our results found that MT treatment could successfully protect R28 cells from Glu excitotoxicity and decrease reactive oxygen species. Also, MT rescued RGCs from NMDA-induced injury and protected visual function in mice. This study enriches the indications of MT in the treatment of glaucoma, providing practical research ideas for its comprehensive prevention and treatment.


Assuntos
Glaucoma , Melatonina , Fármacos Neuroprotetores , Animais , Camundongos , Células Ganglionares da Retina , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , N-Metilaspartato/toxicidade , N-Metilaspartato/uso terapêutico , Ácido Glutâmico/toxicidade , Ácido Glutâmico/uso terapêutico , Melatonina/farmacologia , Melatonina/uso terapêutico , Espécies Reativas de Oxigênio , Glaucoma/tratamento farmacológico , Receptores de N-Metil-D-Aspartato/uso terapêutico
10.
Toxicol In Vitro ; 84: 105453, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35944748

RESUMO

Memantine is a non-competitive antagonist with a moderate affinity to the N-methyl-d-Aspartate (NMDA) receptor. The present study assessed memantine's neuroprotective activity using electrophysiology of ex-vivo hippocampal slices. Interestingly, a nicotinic component was necessary for memantine's neuroprotection (NP). Memantine demonstrated a bell-shaped dose-response curve of NP against NMDA. Memantine was neuroprotective at concentrations below 3 µM, but the NP declined at higher concentrations (>3 µM) when memantine inhibits the NMDA receptor. Additional evidence that memantine NP is mediated by an alternate mechanism independent of the inhibition of the NMDA receptor is supported by its ability to protect neurons when applied before or after the NMDA insult and in the presence of D(-)-2-Amino-5-phosphonopentanoic acid (APV), the standard NMDA receptor inhibitor. We found several similarities between the memantine NP mechanism and the neuroprotective nicotinic drug, the 4R cembranoid. Memantine's NP requires the release of acetylcholine, the activation of α4ß2, and is independent of MEK/MAPK signaling. Both 4R and memantine require the activation of PI3K/AKT for NP against NMDA-mediated excitotoxicity, although at different concentrations. In conclusion, our studies show memantine is neuroprotective through a nicotinic pathway, similar to the nicotinic drug 4R. This information leads to a better understanding of memantine's mechanisms of action and explains its dose-dependent effectiveness in Alzheimer's and other neurological disorders.


Assuntos
Memantina , Fármacos Neuroprotetores , Hipocampo/metabolismo , Memantina/metabolismo , Memantina/farmacologia , N-Metilaspartato/toxicidade , Neuroproteção , Fármacos Neuroprotetores/farmacologia , Nicotina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
11.
Toxicology ; 470: 153139, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-35257817

RESUMO

N-methyl-D-aspartic acid (NMDA), a glutamate analog, can activate N-Methyl-D-Aspartate receptor (NMDAR) to induce vascular endothelial cell injury but the mechanisms are not fully understood. The present study intended to evaluate the role of caveolin-1 (Cav-1) in NMDA-induced dysfunction of human brain microvascular endothelial cells (HBEC-5i), and verify that endothelial NMDAR activation mediates the disruption of tight junction barrier integrity via extracellular signal-regulated kinase (ERK)1/2 pathway. The expression of NMDAR NR1 were confirmed firstly in HBEC-5i and compared with that in mouse brain by Western blot. To study the role of Cav-1 in NMDA mediated reduction of tight junction protein zonula occludens- (ZO) 1 expression, HBEC-5i were transduced with Cav-1 shRNA or Control shRNA, and the Cav-1 knockdown rate tested with qRT-PCR and Western blot is 99.98% or 87.5%, respectively. NMDA exposure decreased mRNA and protein levels of tight junction protein ZO-1 and suppressed transendothelial electrical resistance (TEER) values in HBEC-5i but blocked by NMDAR antagonist MK801. In addition, NMDA induced Cav-1 and ERK1/2 sequential phosphorylation,but these effects were attenuated by silencing the Cav-1 gene with shRNA and ERK1/2 inhibitor U0126, respectively. These results show that the functional presence of NMDAR NR1 in HBEC-5i. Endothelial NMDAR NR1 activation regulate the maintenance of HBEC-5i-constructed tight junction barrier integrity via the caveolin-1-associated ERK1/2 signaling pathway.


Assuntos
Caveolina 1 , N-Metilaspartato , Animais , Encéfalo/metabolismo , Caveolina 1/genética , Caveolina 1/metabolismo , Células Endoteliais , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , N-Metilaspartato/toxicidade , RNA Interferente Pequeno/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Proteínas de Junções Íntimas , Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
12.
Curr Eye Res ; 47(6): 866-873, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35188034

RESUMO

PURPOSE: Retinal and optic nerve damage in glaucoma involves excitotoxicity via N-methyl-D-aspartate (NMDA) receptors. Since, trans-resveratrol (TR) is known to provide neuroprotection, we investigated its protective effects against NMDA-induced retinal and optic nerve injury. METHODS: Sprague Dawley rats were divided into four groups which received vehicle (PBS), NMDA, and TR 0.4 or TR 4 nmol 24 h prior to NMDA, unilaterally and intravitreally. Seven days post-injection, rats were euthanized; eyeballs were enucleated and subjected to hematoxylin and eosin and terminal transferase dUTP nick end labeling staining while optic nerves were isolated for toluidine blue staining. RESULTS: Retinal morphometry showed that ganglion cell layer (GCL) layer thickness within inner retina (IR), retinal cell count (RCC) per 100-µm length of GCL, RCC per 100-µm2 area of GCL, and RCC per 100 µm2 of IR were significantly higher in both TR-treated groups compared to the NMDA group. No differences were observed between the two dose groups. Optic nerve morphology was in accordance with the retinal morphology whereby TR-treated groups showed significantly lesser degenerative changes compared to NMDA-treated group. CONCLUSIONS: TR protects against NMDA-induced changes in retinal and optic nerve morphology by preventing retinal cell apoptosis.


Assuntos
Carcinoma de Células Renais , Traumatismos Oculares , Neoplasias Renais , Traumatismos do Nervo Óptico , Doenças Retinianas , Animais , Apoptose , N-Metilaspartato/toxicidade , Nervo Óptico , Traumatismos do Nervo Óptico/induzido quimicamente , Traumatismos do Nervo Óptico/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Resveratrol/farmacologia , Retina , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/tratamento farmacológico , Doenças Retinianas/prevenção & controle
13.
Bioorg Med Chem ; 59: 116675, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35202968

RESUMO

A range of novel 1-phenyl-benzopyrrolizidin-3-one derivatives were synthesized and evaluated for neuroprotective effects against N-methyl-ᴅ-aspartate (NMDA)-induced injury in PC12 cells. Interestingly, derivatives that 1-phenyl moiety bearing electron-donating group, especially benzyloxy, and the trans-forms exhibited better protective activity against NMDA-induced neurotoxicity. Compound 11 m demonstrated the best neuroprotective potency and shown a dose-dependent prevention. The increased intracellular calcium (Ca2+) influx caused by NMDA in PC12 cells was reversed in the case of compound 11 m pretreatment at 15 µM. These results suggested that the synthesized 1-phenyl-benzopyrrolizidin-3-one derivatives exerted neuroprotective effect on NMDA-induced excitotoxicity in PC12 cells associated with inhibition of Ca2+ overload and can be further optimized for the development of neuroprotective agents.


Assuntos
N-Metilaspartato , Fármacos Neuroprotetores , Animais , Cálcio/metabolismo , N-Metilaspartato/toxicidade , Fármacos Neuroprotetores/farmacologia , Células PC12 , Ratos , Receptores de N-Metil-D-Aspartato
14.
Cell Mol Life Sci ; 79(1): 15, 2021 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-34967918

RESUMO

Excessive activation of the ionotropic N-methyl-D-aspartate (NMDA) receptor has been shown to cause abnormally high levels of Ca2+ influx, thereby leading to excitotoxic neuronal death. In this study, exposure of mouse primary cortical neurons to NMDA resulted in the cleavage and activation of mammalian sterile 20-like kinase-1 (MST1), both of which were mediated by calpain 1. In vitro cleavage assay data indicated that calpain 1 cleaves out the autoinhibitory domain of MST1 to generate an active form of the kinase. Furthermore, calpain 1 mediated the cleavage and activation of wild-type MST1, but not of MST1 (G339A). Intriguingly, NMDA/calpain-induced MST1 activation promoted the nuclear translocation of the kinase and the phosphorylation of histone H2B in mouse cortical neurons, leading to excitotoxicity. Thus, we propose a previously unrecognized mechanism of MST1 activation associated with NMDA-induced excitotoxic neuronal death.


Assuntos
Córtex Cerebral/patologia , N-Metilaspartato/toxicidade , Neurônios/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Calpaína/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Camundongos Endogâmicos C57BL , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurotoxinas/toxicidade , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico/efeitos dos fármacos , Especificidade por Substrato/efeitos dos fármacos
15.
Exp Eye Res ; 213: 108826, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34752818

RESUMO

Glaucoma is a leading cause of blindness worldwide and is characterized by degeneration associated with the death of retinal ganglion cells (RGCs). It is believed that glaucoma is a group of heterogeneous diseases with multifactorial pathomechanisms. Here, we investigate whether anti-inflammation treatment with an ER stress blockade can selectively promote neuroprotection against NMDA injury in the RGCs. Retinal excitotoxicity was induced with an intravitreal NMDA injection. Microglial activation and neuroinflammation were evaluated with Iba1 immunostaining and cytokine gene expression. A stable HT22 cell line transfected with an NF-kB reporter was used to assess NF-kB activity after hesperidin treatment. CHOP-deficient mice were used as a model of ER stress blockade. Retinal cell death was evaluated with a TUNEL assay. As results, in the NMDA injury group, Iba1-positive microglia increased 6 h after NMDA injection. Also at 6 h, pro-inflammatory cytokines and chemokine increased, including TNFα, IL-1b, IL-6 and MCP-1. In addition, the MCP-1 promoter-driven EGFP signal, which we previously identified as a stress signal in injured RGCs, also increased; hesperidin treatment suppressed this inflammatory response and reduced stressed RGCs. In CHOP-deficient mice that received an NMDA injection, the gene expression of pro-inflammatory cytokines, chemokines, markers of active microglia, and inflammatory regulators was greater than in WT mice. In WT mice, hesperidin treatment partially prevented retinal cell death after NMDA injury; this neuroprotective effect was enhanced in CHOP-deficient mice. These findings demonstrate that ER stress blockade is not enough by itself to prevent RGC loss due to neuroinflammation in the retina, but it has a synergistic neuroprotective effect after NMDA injury when combined with an anti-inflammatory treatment based on hesperidin.


Assuntos
Hesperidina/uso terapêutico , N-Metilaspartato/toxicidade , Doenças Retinianas/tratamento farmacológico , Células Ganglionares da Retina/efeitos dos fármacos , Fator de Transcrição CHOP/deficiência , Animais , Western Blotting , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Sinergismo Farmacológico , Deleção de Genes , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Microglia/metabolismo , NF-kappa B/metabolismo , Neuroproteção , Estresse Oxidativo/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/metabolismo , Células Ganglionares da Retina/metabolismo
16.
STAR Protoc ; 2(4): 100932, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34806045

RESUMO

The degeneration of retinal ganglion cells (RGCs) leads to irreversible vision loss in a variety of pathological states. Here, we describe a protocol to evaluate the role of a gene in protecting mouse RGCs when they sustain injuries from excitotoxicity or axonal damage. This protocol includes the procedures for gene transfer through AAV intravitreal injection, induction of RGC injuries by NMDA-induced excitotoxicity or optic nerve crush, and retina immunohistochemistry to assess RGC survival. For complete details on the use and execution of this protocol, please refer to Guo et al. (2021).


Assuntos
Sobrevivência Celular/genética , Imuno-Histoquímica/métodos , Microscopia Confocal/métodos , Células Ganglionares da Retina , Animais , Células Cultivadas , Camundongos , N-Metilaspartato/toxicidade , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia
17.
Invest Ophthalmol Vis Sci ; 62(13): 23, 2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34698771

RESUMO

Purpose: Optic nerve damage leads to impairment of visual functions. We previously demonstrated that apolipoprotein E-containing lipoproteins (E-LPs) protect retinal ganglion cells (RGCs) from degeneration in a glaucoma model of glutamate/aspartate transporter-deficient mice. This study aimed to determine whether E-LPs protect RGCs from N-methyl-D-aspartate (NMDA)-induced excitotoxicity, and to investigate the details of an indirect neuroprotective mechanism of E-LPs by reducing α2-macroglobulin, which interferes with the neuroprotective effect of E-LPs, in Müller glia. Methods: Excitotoxicity was caused by intravitreal injection of NMDA, and then retinae were subjected to immunoblotting or quantitative reverse transcription-PCR. Primary cultures of mouse mixed retinal cells and mouse Müller glia were used for evaluating the effects of E-LPs on the expression of α2-macroglobulin. Results: Intravitreal injection of E-LPs protected the optic nerve from degeneration and attenuated the increase in α2-macroglobulin in aqueous humor and retina of rats. E-LPs directly decreased the expression and secretion of α2-macroglobulin in primary cultures of Müller glia; this decrease in production of α2-macroglobulin was blocked by knockdown of the low-density lipoprotein receptor-related protein 1 (LRP1) with small interfering RNA. E-LPs promoted the phosphorylation of STAT3, whereas Stattic, an inhibitor of STAT3, restored the expression of α2-macroglobulin decreased by E-LPs. Conclusions: In addition to our previous findings of the protection of RGCs by E-LPs, the new observations in Müller glia indicate that a reduction of the intraocular α2-macroglobulin, regulated by the E-LP-LRP1-STAT3 pathway, might be an additional protective mechanism against excitotoxicity in the retina.


Assuntos
Apolipoproteínas E/metabolismo , Células Ependimogliais/metabolismo , Regulação da Expressão Gênica , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , alfa 2-Macroglobulinas Associadas à Gravidez/genética , Degeneração Retiniana/genética , Células Ganglionares da Retina/patologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Células Ependimogliais/efeitos dos fármacos , Células Ependimogliais/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , N-Metilaspartato/toxicidade , Fármacos Neuroprotetores/farmacologia , alfa 2-Macroglobulinas Associadas à Gravidez/biossíntese , RNA/genética , Ratos , Ratos Sprague-Dawley , Degeneração Retiniana/tratamento farmacológico , Degeneração Retiniana/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo
18.
Exp Eye Res ; 212: 108785, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34600894

RESUMO

Epigenetic gene enhancer of zeste homolog-2 (Ezh2) is reported to be associated with ocular neurodegenerative diseases; however, its underlying mechanism is poorly understood. The present study aimed to determine the role of 3-deazaneplanocin A (DZNep), which inhibits the transcription of Ezh2 by reducing the trimethylation of histone 3 lysine 27 (H3K27me3), in a retinal ganglion cell (RGC) degeneration model. Retinal damage was caused by intravitreal injection of N-methyl-D-aspartate (NMDA). DZNep and the vehicle control were intravitreally applied immediately post-NMDA injection. The severity of retinal damage was evaluated by immunofluorescence and terminal deoxyribonucleotide transferase (TdT)-mediated dUTP nick end labeling (TUNEL) staining, and retinal function was determined by electroretinogram (ERG). The transcriptome was examined by RNA sequencing and quantitative PCR (qPCR). Microglial cells were detected by immunohistochemistry. DZNep significantly prevented the cell death in the ganglion cell layer (GCL) and inner nuclear layer (INL) induced by NMDA. DZNep preserved the ERG b- and a-wave amplitudes and the b/a ratio in NMDA-treated mice. Moreover, RNA sequencing and qPCR revealed that neuroprotective genes were upregulated and played an important role in preserving retinal cells. In addition, DZNep inhibited the NMDA-induced activation of microglial cells. Our results suggest that H3K27me3 controls RGC survival at the transcriptional and epigenetic levels. The absence of H3K27me3 deposition upregulates neuroprotective genes to protect RGCs. Therefore, DZNep, which inhibits Ezh2 activity, could be a novel therapeutic treatment for ocular neurodegenerative diseases.


Assuntos
Adenosina/análogos & derivados , Degeneração Retiniana/tratamento farmacológico , Células Ganglionares da Retina/efeitos dos fármacos , Adenosina/administração & dosagem , Animais , Contagem de Células , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Eletrorretinografia , Injeções Intravítreas , Masculino , Camundongos , N-Metilaspartato/toxicidade , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia
19.
J Neurosci ; 41(35): 7461-7478, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34315810

RESUMO

Temporal costs influence reward-based decisions. This is commonly studied in temporal discounting tasks that involve choosing between cues signaling an imminent reward option or a delayed reward option. However, it is unclear whether the temporal delay before a reward can alter the value of that option. To address this, we identified the relative preference between different flavored rewards during a free-feeding test using male and female rats. Animals underwent training where either the initial preferred or the initial less preferred reward was delivered noncontingently. By manipulating the intertrial interval during training sessions, we could determine whether temporal delays impact reward preference in a subsequent free-feeding test. Rats maintained their initial preference if the same delays were used across all training sessions. When the initial less preferred option was delivered after short delays (high reward rate) and the initial preferred option was delivered after long delays (low reward rate), rats expectedly increased their preference for the initial less desirable option. However, rats also increased their preference for the initial less desirable option under the opposite training contingencies: delivering the initial less preferred reward after long delays and the initial preferred reward after short delays. These data suggest that sunk temporal costs enhance the preference for a less desirable reward option. Pharmacological and lesion experiments were performed to identify the neural systems responsible for this behavioral phenomenon. Our findings demonstrate the basolateral amygdala and retrosplenial cortex are required for temporal delays to enhance the preference for an initially less desirable reward.SIGNIFICANCE STATEMENT The goal of this study was to determine how temporal delays influence reward preference. We demonstrate that delivering an initially less desirable reward after long delays subsequently increases the consumption and preference for that reward. Furthermore, we identified the basolateral amygdala and the retrosplenial cortex as essential nuclei for mediating the change in reward preference elicited by sunk temporal costs.


Assuntos
Complexo Nuclear Basolateral da Amígdala/fisiologia , Comportamento de Escolha/fisiologia , Giro do Cíngulo/fisiologia , Recompensa , Fatores de Tempo , Animais , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Feminino , Flupentixol/farmacologia , Preferências Alimentares , Giro do Cíngulo/efeitos dos fármacos , Masculino , N-Metilaspartato/toxicidade , Ratos , Ratos Sprague-Dawley
20.
J Neurosci ; 41(33): 7148-7159, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34210784

RESUMO

Following stroke, the survival of neurons and their ability to reestablish connections is critical to functional recovery. This is strongly influenced by the balance between neuronal excitation and inhibition. In the acute phase of experimental stroke, lethal hyperexcitability can be attenuated by positive allosteric modulation of GABAA receptors (GABAARs). Conversely, in the late phase, negative allosteric modulation of GABAAR can correct the suboptimal excitability and improves both sensory and motor recovery. Here, we hypothesized that octadecaneuropeptide (ODN), an endogenous allosteric modulator of the GABAAR synthesized by astrocytes, influences the outcome of ischemic brain tissue and subsequent functional recovery. We show that ODN boosts the excitability of cortical neurons, which makes it deleterious in the acute phase of stroke. However, if delivered after day 3, ODN is safe and improves motor recovery over the following month in two different paradigms of experimental stroke in mice. Furthermore, we bring evidence that, during the subacute period after stroke, the repairing cortex can be treated with ODN by means of a single hydrogel deposit into the stroke cavity.SIGNIFICANCE STATEMENT Stroke remains a devastating clinical challenge because there is no efficient therapy to either minimize neuronal death with neuroprotective drugs or to enhance spontaneous recovery with neurorepair drugs. Around the brain damage, the peri-infarct cortex can be viewed as a reservoir of plasticity. However, the potential of wiring new circuits in these areas is restrained by a chronic excess of GABAergic inhibition. Here we show that an astrocyte-derived peptide, can be used as a delayed treatment, to safely correct cortical excitability and facilitate sensorimotor recovery after stroke.


Assuntos
Inibidor da Ligação a Diazepam/uso terapêutico , Agonistas de Receptores de GABA-A/uso terapêutico , Neurônios/efeitos dos fármacos , Neuropeptídeos/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Receptores de GABA-A/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Adulto , Animais , Astrócitos/metabolismo , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Inibidor da Ligação a Diazepam/deficiência , Inibidor da Ligação a Diazepam/fisiologia , Implantes de Medicamento , Potenciais Somatossensoriais Evocados , Feminino , Agonistas de Receptores de GABA-A/farmacologia , Humanos , Hidrogéis , Infarto da Artéria Cerebral Média/tratamento farmacológico , Trombose Intracraniana/tratamento farmacológico , Trombose Intracraniana/etiologia , Luz , Camundongos , Camundongos Endogâmicos C57BL , N-Metilaspartato/toxicidade , Neurônios/fisiologia , Neuropeptídeos/deficiência , Neuropeptídeos/fisiologia , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/deficiência , Fragmentos de Peptídeos/fisiologia , Ratos , Rosa Bengala/efeitos da radiação , Rosa Bengala/toxicidade , Método Simples-Cego , Acidente Vascular Cerebral/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...