Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 313
Filtrar
2.
Mol Med Rep ; 29(1)2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38063180

RESUMO

Alzheimer's disease (AD) is the most common type of age­related dementia, and causes progressive memory degradation, neuronal loss and brain atrophy. The pathological hallmarks of AD consist of amyloid­ß (Aß) plaque accumulation and abnormal neurofibrillary tangles. Amyloid fibrils are constructed from Aß peptides, which are recognized to assemble into toxic oligomers and exert cytotoxicity. The fibrillar Aß­protein fragment 25­35 (Aß25­35) induces local inflammation, thereby exacerbating neuronal apoptosis. Notoginsenoside R1 (NGR1), one of the primary bioactive ingredients isolated from Panax notoginseng, exhibits effective anti­inflammatory and anti­oxidative activities. However, NGR1 pharmacotherapies targeting Aß­induced inflammation and cell injury cascade remain to be elucidated. The present study investigated the effect and mechanism of NGR1 in Aß25­35­treated PC12 cells. NGR1 doses between 250 and 1,000 µg/ml significantly increased cell viability suppressed by 20 µM Aß25­35 peptide treatment. Notably, the present study demonstrated that Aß25­35 peptide­induced sphingosine kinase 1 (SphK1) signaling activation was reduced after NGR1 treatment, further inhibiting the downstream NF­κB inflammatory signaling pathway. In addition, administration of SphK1 inhibitor II (SKI­II), a SphK1 inhibitor, also significantly reduced Aß25­35 peptide­induced apoptosis and the ratio of NF­κB p­p65/p65. Furthermore, SphK1 knockdown in PC12 cells using small interfering RNA alleviated Aß­induced cell apoptosis and inflammation, suggesting a pivotal role of SphK1 signaling in the anti­inflammatory effect of NGR1. In summary, NGR1 alleviated inflammation and apoptosis stimulated by Aß25­35 by inhibiting the SphK1/NF­κB signaling pathway and may be a promising agent for future AD treatment.


Assuntos
Doença de Alzheimer , Ginsenosídeos , Animais , Ratos , Doença de Alzheimer/metabolismo , Anti-Inflamatórios/farmacologia , Apoptose , Ginsenosídeos/farmacologia , Ginsenosídeos/uso terapêutico , Inflamação/patologia , NF-kappa B/metabolismo , Células PC12 , Transdução de Sinais , Peptídeos beta-Amiloides/efeitos adversos , Peptídeos beta-Amiloides/farmacologia
3.
Acta Cir Bras ; 38: e385523, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38055394

RESUMO

PURPOSE: Intravenous anesthetics have excellent analgesic activity without inducing the side effect in the respiratory system. The aim and objective of the current experimental study was to access the neuroprotective effect of sevoflurane against isoflurane induced cognitive dysfunction in rats. METHODS: Isoflurane was used for induction the neurodysfunction in the rats, and rats received the oral administration of sevoflurane (2.5, 5 and 10 mg/kg). Morris water test was carried out for the estimation of cognitive function. Neurochemical parameters, antioxidant parameters and pro-inflammatory cytokines were also estimated. RESULTS: Sevoflurane significantly (P < 0.001) altered the neurochemical parameters such as anti-choline acetyltransferase, acetylcholine esterase, acetylcholine, protein carbonyl, choline brain-derived neurotrophic factor, and amyloid ß; antioxidant parameters such as glutathione, superoxide dismutase, and malondialdehyde; pro-inflammatory cytokines include interleukin (IL-2, IL-10, IL-4, IL-6, IL-10, IL-1ß), and tumor necrosis factor-α. Sevoflurane significantly reduced the activity of caspase-3. CONCLUSIONS: Sevoflurane exhibited the neuroprotection against the cognitive dysfunction in rats via anti-inflammatory and antioxidant mechanism.


Assuntos
Anestésicos Inalatórios , Disfunção Cognitiva , Isoflurano , Fármacos Neuroprotetores , Ratos , Animais , Isoflurano/efeitos adversos , Sevoflurano/uso terapêutico , Antioxidantes/uso terapêutico , Interleucina-10 , Anestésicos Inalatórios/efeitos adversos , Neuroproteção , Acetilcolina/efeitos adversos , Peptídeos beta-Amiloides/efeitos adversos , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/prevenção & controle , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Citocinas/metabolismo , Fármacos Neuroprotetores/farmacologia
4.
Niger J Physiol Sci ; 38(1): 91-99, 2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38243363

RESUMO

Decline in cholinergic function and oxidative/nitrosative stress play a central role in Alzheimer's disease (AD). Previous quantitative HPLC profiling analysis has revealed the presence of Pinostrobin, formononetin, vitexin and other neuroprotective flavonoids in Cajanus cajan seed extract. This study was designed to investigate the protective action of Cajanus cajan ethanol seed extract (CC) on learning and memory functions using scopolamine mouse model of amnesia. Materials and methods: Adult mice were pretreated with CC (50, 100, or 200mg/kg, p.o) or vehicle (10ml/kg, p.o) for 16 days consecutively. Scopolamine, a competitive muscarinic cholinergic receptor antagonist (1mg/kg, i.p.) was given an hour after CC pretreatment from days 3 to 16.  The mice were subjected to behavioural tests from day 11 (open field test (OFT)/ Y-maze test (YMT) and Morris water maze task (MWM) from days 12-16. Animals were euthanized 1h after behavioral test on day 16 and discrete brain regions isolated for markers of oxidative stress and cholinergic signaling. Molecular docking analysis was undertaken to predict the possible mechanism(s) of CC-induced anti-amnesic action.  pre-administration of CC significantly reversed working memory and learning deficits caused by scopolamine in YMT and MWM tests, respectively. Moreover, CC prevented scopolamine-induced oxidative and nitrosative stress radicals in the hippocampus evidenced in significant increase in glutathione (GSH) level, superoxide dismutase (SOD) and catalase (CAT) activities with a marked decrease in malondialdehyde (MDA) production, as well as significant inhibition of hippocampal scopolamine-induced increase in acetylcholinesterase activity by CC. The molecular docking analysis showed that out of the 19 compounds, the following had the highest binding affinity; Pinostrobin (-8.7 Kcal/mol), friedeline (-7.5kCal/mol), and lupeol (-8.2 Kcal/mol), respectively, to neuronal muscarinic M1 acetylcholine receptor, α7 nicotinic acetylcholine receptor and amyloid beta peptide binding pockets, which further supports the ability of CC to enhance neuronal cholinergic signaling and possible inhibition of amyloid beta aggregation. This study showed that Cajanus cajan seeds extract improved working memory and learning through enhancement of cholinergic signaling, antioxidant capacity and reduction in amyloidogenesis.


Assuntos
Antioxidantes , Cajanus , Camundongos , Animais , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Escopolamina/farmacologia , Cajanus/metabolismo , Acetilcolinesterase/metabolismo , Acetilcolinesterase/farmacologia , Peptídeos beta-Amiloides/efeitos adversos , Peptídeos beta-Amiloides/metabolismo , Simulação de Acoplamento Molecular , Aprendizagem em Labirinto , Amnésia/induzido quimicamente , Amnésia/tratamento farmacológico , Amnésia/prevenção & controle , Estresse Oxidativo , Glutationa/metabolismo , Transmissão Sináptica , Hipocampo , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Colinérgicos/efeitos adversos , Colinérgicos/metabolismo , Mecanismos de Defesa , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/metabolismo
5.
Aging (Albany NY) ; 14(10): 4305-4325, 2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35604830

RESUMO

Myocardia-Related Transcription Factors-A (MRTF-A), which is enriched in the hippocampus and cerebral cortex, has been shown to have a protective function against ischemia hypoxia-induced neuronal apoptosis. However, the function of MRTF-A on ß-amyloid peptide (Aß)-induced neurotoxicity and autophagy dysfunction in Alzheimer's disease is still unclear. This study shows that the expression of MRTF-A in the hippocampus of Tg2576 transgenic mice is reduced, and the overexpression of MRTF-A mediated by lentiviral vectors carrying MRTF-A significantly reduces the accumulation of hippocampal ß-amyloid peptide and reduces cognition defect. Overexpression of MRTF-A inhibits neuronal apoptosis, increases the protein levels of microtubule-associated protein 1 light chain 3-II (MAP1LC3/LC3-II) and Beclin1, reduces the accumulation of SQSTM1/p62 protein, and promotes autophagosomes-Lysosomal fusion in vivo and in vitro. Microarray analysis and bioinformatics analysis show that MRTF-A reverses Aß-induced autophagy impairment by up-regulating miR-1273g-3p level leading to negative regulation of the mammalian target of rapamycin (mTOR), which is confirmed in Aß1-42-treated SH-SY5Y cells. Further, overexpression of MRTF-A reduces Aß1-42-induced neuronal apoptosis. And the effect was abolished by miR-1273g-3p inhibitor or MHY1485 (mTOR agonist), indicating that the protection of MRTF-A on neuronal damage is through targeting miR-1273g-3p/mTOR axis. Targeting this signaling may be a promising approach to protect against Aß-induced neuronal injury.


Assuntos
Peptídeos beta-Amiloides , Autofagia , Hipocampo , MicroRNAs , Transativadores , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/efeitos adversos , Peptídeos beta-Amiloides/metabolismo , Animais , Apoptose/genética , Autofagia/genética , Hipocampo/lesões , Hipocampo/metabolismo , Humanos , Mamíferos/metabolismo , Camundongos , Camundongos Transgênicos , MicroRNAs/metabolismo , Neuroblastoma , Neurônios/metabolismo , Serina-Treonina Quinases TOR , Transativadores/biossíntese , Transativadores/genética
6.
Int J Biol Macromol ; 209(Pt A): 1280-1287, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35461860

RESUMO

Chondroitin sulfate E (CS-E), which is characterized by oversulfated disaccharide units, has been shown to regulate neuronal adhesion, neurite outgrowth and exert neuroprotective effects. In view of these findings, here we investigated the anti-Alzheimer's disease (AD) activities of CSE by using transgenic Caenorhabditis elegans model of Alzheimer's disease. The behavioral experiments demonstrated that CSE at the concentration of 1 mg/mL significantly delayed the worm paralysis caused by Aß aggregation as compared with control group. Western blot analysis revealed that the level of small oligomers in the transgenic C. elegans was significantly reduced upon treatment with CSE. The number of Aß plaque deposits in transgenic worm was significantly decreased. In addition, CSE also protected the worms from oxidative stress and rescued chemotaxis dysfunction in transgenic strain CL2355. Taken together, these data suggested that CSE could protect against Aß-induced toxicity in C. elegans. These results offer valuable evidence for the future use of CSE in the development of agents for the treatment of AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/efeitos adversos , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Sulfatos de Condroitina/farmacologia , Modelos Animais de Doenças
7.
Radiat Environ Biophys ; 61(2): 205-219, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35325276

RESUMO

Alzheimer's disease is characterized by deposition of amyloid-beta plaques in the brain. Available pharmaceuticals provide temporary symptomatic relief without affecting disease progression. Use of radiation was found effective in treating extra-cranial amyloidosis, therefore, the present study was designed to investigate the neuroprotective role of fractionated X-irradiation in Aß1-42-based rodent model of Alzheimer's disease. S.D. female rats were randomly divided into four groups: sham control (Group 1), Aß1-42 injected (Group 2), cranial X-irradiated (Group 3) and Aß1-42 injected followed by cranial X-irradiation (Group 4). A single dose of 5 µL Aß1-42 peptide was administered through intracerebroventricular (icv) injection in Group 2 and 4 animals, while Group 1 animals were administered 5 µL of bi-distilled water (icv). The group 4 animals were further subjected to 10 Gy X-irradiation (fractionated dose, 2 Gy × 5 days) after 4 weeks of Aß1-42 infusion of peptide. The animals in Group 3 were subjected to same dose of cranial fractionated X-irradiation (2 Gy × 5 days) only. Significant decrease in amyloid deposits were observed in the Aß1-42 + radiation-treated animals confirmed by histopathological analysis. These finding were in concordance with neurobehavioral tests that showed a significant improvement in Aß1-42-induced memory impairment in the animals subjected to fractionated cranial X-irradiation. Restoration of alterations in neurochemical and antioxidant defense indices further supported our results. The present study highlights the underexplored role of fractionated X-irradiation in curtailing the Aß1-42-induced neurotoxicity, suggesting a novel treatment option for Alzheimer's disease-associated pathologies.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/radioterapia , Peptídeos beta-Amiloides/efeitos adversos , Animais , Cognição , Modelos Animais de Doenças , Feminino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Ratos , Roedores
8.
Int J Biol Macromol ; 206: 823-836, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35307462

RESUMO

Amyloid-ß (Aß) deposition and neurotoxicity play an important role in Alzheimer's disease (AD). Notably, the nonnegligible role of endogenous heparan sulfate (HS) in the release, uptake and misfolding of Aß sheds light on the discovery of HS as an effective drug for AD. In this work, the effects of HS from porcine mucosa (PMHS) on Aß1-42-induced neurotoxicity were investigated both in vitro and in vivo. The in vitro AD model was established in SH-SY5Y via treatment with oligomeric Aß1-42, and the in vivo AD model was established by intracerebroventricular injection of Aß1-42 to KM mice. The results showed that in vitro, PMHS could ameliorate the inflammation and apoptosis response of SH-SY5Y cells induced by Aß1-42; in vivo, PMHS could not only improve the cognitive impairment induced by Aß1-42 but also inhibit neuroinflammation and apoptosis in the brain. Furthermore, PMHS lowered the levels of Aß1-42 in the peripheral circulation and brain by improving the phagocytosis function of neutrophils. This is the first report that PMHS enhances the phagocytosis function of neutrophils to alleviate Aß-induced neurotoxicity. Moreover, our work verified the feasibility of peripheral Aß clearance for improving neurotoxicity. Conclusively, we believe that PMHS could be developed into neuroprotective drugs for AD.


Assuntos
Doença de Alzheimer , Fragmentos de Peptídeos , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/efeitos adversos , Animais , Cadáver , Heparitina Sulfato/farmacologia , Camundongos , Mucosa , Fragmentos de Peptídeos/efeitos adversos , Suínos
9.
Bioengineered ; 13(1): 1921-1930, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35001806

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder that is pathologically related to oxidative stress and cellular senescence. Safinamide is one of the clinically prescribed monoamine oxidase B (MAOB) inhibitors. It has been reported to possess therapeutic potential in neurological disorders. However, the therapeutic potential of safinamide in AD is still under investigation. In this study, we explored the effect of safinamide in amyloid (Aß)1-42 oligomers-stimulated M17 neuronal cells. We established the in vitro model with M17 cells by treating them with 1 µM Aß1-42 oligomers with or without safinamide (100 or 200 nM). The results show that safinamide ameliorated Aß1-42 oligomers-induced oxidative stress in M17 cells as revealed by the decreased reactive oxygen species (ROS) production and reduced glutathione (GSH) content. Safinamide treatment significantly ameliorated senescence-associated-ß-galactosidase (SA-ß-gal)-positive cells and telomerase activity. Further, we show that safinamide treatment resulted in decreased mRNA and protein expressions of p21 and plasminogen activator inhibitor-1 (PAI-1). Moreover, silencing of Sirtuin1 (SIRT1) abolished the effects of safinamide on the mRNA levels of p21 and PAI-1, as well as SA-ß-gal-positive cells in Aß1-42 oligomers-induced M17 cells. In conclusion, we reveal that safinamide exerted a protective function on M17 cells from Aß1-42 oligomers induction-caused oxidative stress and cellular senescence through SIRT1 signaling. These present results provide meaningful evidence that safinamide may be medically developed for the prevention and therapy of AD.


Assuntos
Alanina/análogos & derivados , Peptídeos beta-Amiloides/efeitos adversos , Benzilaminas/farmacologia , Neurônios/citologia , Fármacos Neuroprotetores/farmacologia , Alanina/química , Alanina/farmacologia , Benzilaminas/química , Linhagem Celular , Senescência Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Humanos , Modelos Biológicos , Estrutura Molecular , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo
10.
Int J Mol Sci ; 24(1)2022 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-36613533

RESUMO

This study was conducted to compare the synbiotic activity between Corni fructus (C. fructus) and Limosilactobacillus reuteri (L. reuteri) on dextran sulfate sodium (DSS)-induced colitis and cognitive dysfunction in C57BL/6 mice. C. fructus (as prebiotics, PRE), L. reuteri (as probiotics, PRO), and synbiotics (as a mixture of L. reuteri and C. fructus, SYN) were fed to mice for 3 weeks. Consumption of PRE, PRO, and SYN ameliorated colitis symptoms in body weight, large intestinal length, and serum albumin level. Moreover, SYN showed a synergistic effect on intestinal permeability and intestinal anti-inflammation response. Also, SYN significantly improved cognitive function as a result of measuring the Y-maze and passive avoidance tests in DSS-induced behavioral disorder mice. Especially, SYN also restored memory function by increasing the cholinergic system and reducing tau and amyloid ß pathology. In addition, PRE, PRO, and SYN ameliorated dysbiosis by regulating the gut microbiota and the concentration of short-chain fatty acids (SCFAs) in feces. The bioactive compounds of C. fructus were identified with quinic acid, morroniside, loganin, and cornuside, using ultra-performance liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (UPLC-Q-TOF-MS2). In conclusion, synbiotic supplementation alleviated DSS-induced colitis and cognitive dysfunction by modulating gut microbiota, proinflammatory cytokines, and SCFAs production.


Assuntos
Colite , Cornus , Limosilactobacillus reuteri , Simbióticos , Camundongos , Animais , Peptídeos beta-Amiloides/efeitos adversos , Camundongos Endogâmicos C57BL , Colite/induzido quimicamente , Colite/tratamento farmacológico , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Colo/patologia
11.
Aging Cell ; 21(1): e13533, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34939734

RESUMO

BACKGROUND: A previous study demonstrated that nearly 40%-60% of brain Aß flows out into the peripheral system for clearance. However, where and how circulating Aß is cleared in the periphery remains unclear. The spleen acts as a blood filter and an immune organ. The aim of the present study was to investigate the role of the spleen in the clearance of Aß in the periphery. METHODS: We investigated the physiological clearance of Aß by the spleen and established a mouse model of AD and spleen excision by removing the spleens of APP/PS1 mice to investigate the effect of splenectomy on AD mice. RESULTS: We found that Aß levels in the splenic artery were higher than those in the splenic vein, suggesting that circulating Aß is cleared when blood flows through the spleen. Next, we found that splenic monocytes/macrophages could take up Aß directly in vivo and in vitro. Splenectomy aggravated behaviour deficits, brain Aß burden and AD-related pathologies in AD mice. CONCLUSION: Our study reveals for the first time that the spleen exerts a physiological function of clearing circulating Aß in the periphery. Our study also suggests that splenectomy, which is a routine treatment for splenic rupture and hypersplenism, might accelerate the development of AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/efeitos adversos , Baço/patologia , Esplenectomia/métodos , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Transgênicos
12.
J Appl Toxicol ; 42(2): 285-294, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34133789

RESUMO

With the growth of the aging population, the prevalence of Alzheimer's disease (AD) has increased and influenced the work and daily life of AD patients, imposing a heavy burden on society and the patients' families. AD is a progressive disease with a long duration, and the pathogenesis is very complicated. Here, we found that alpha-lipoic acid (LA), an endogenous, naturally synthesized compound, could attenuate amyloid beta fragment (Aß25-35 )-induced PC12 cell toxicity. Aß25-35 treatment largely decreased the viability of PC12 cells, increased reactive oxygen species (ROS) levels, and increased the percentage of apoptotic cells, which were accompanied by changes in the expression of the apoptosis-related genes. Further, the Wnt pathway was inactivated, and the expression of Wnt pathway-related proteins such as Frizzled2, GSK3ß, and phosphorylated GSK3ß were dysregulated after Aß25-35 treatment. LA efficiently attenuated Aß25-35 -induced PC12 cell apoptosis and downregulated the phosphorylation-mediated degradation of ß-catenin as well as GSK3ß. Our results demonstrate that LA rescues Aß25-35 -induced neurocytotoxicity through the Wnt-ß-catenin pathway.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/efeitos adversos , Fármacos Neuroprotetores/farmacologia , Ácido Tióctico/farmacologia , Animais , Modelos Animais de Doenças , Células PC12 , Ratos
13.
Neuromolecular Med ; 24(2): 183-194, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34231190

RESUMO

Alzheimer's disease (AD) is associated with the accumulation of ß-amyloid and leads to cognitive impairment. Numerous studies have established that neuronal calcium homeostasis is perturbed in AD. Recently, transient receptor potential vanilloid 2 (TRPV2) channels, a non-selective calcium-permeable channel, have been investigated in several diseases. However, the role of the TRPV2 channel has not been investigated in AD yet. In this study, intracerebroventricular administration of ß-amyloid (10 µg) to Sprague Dawley rats resulted in cognitive impairment which was evident from the assessment of cognitive tests. Also, TRPV2 mRNA and protein expression were found to be upregulated, while the expression of Ca2+/calmodulin-dependent protein kinase II (p-CaMKII-Thr-286), glycogen synthase kinase 3ß (p-GSK-3ß-Ser-9), cAMP response element-binding protein (p-CREB-Ser-133), and postsynaptic density protein 95 (PSD-95) were downregulated in the hippocampus of ß-amyloid-treated animals. Even, ß-amyloid-treated animals showed upregulation of mRNA level of calcium buffering proteins (parvalbumin and calsequestrin) and calcineurin A (PPP3CA) in the hippocampus. Acetylcholinesterase activity was also increased in the cortex of ß-amyloid-treated animals. Three-week treatment with tranilast showed improvement in the cognitive parameters which was associated with a decrease in TRPV2 expression and AChE activity. Additionally, an increase in the protein expression of p-CaMKII, p-GSK-3ß, p-CREB and PSD-95 in the hippocampus was found. Downregulation in the mRNA level of calcium buffering proteins (parvalbumin and calsequestrin) and calcineurin A in the hippocampus was also seen. These results reveal the importance of TRPV2 channels in the ß-amyloid-induced cognitive deficits and suggest TRPV2 as a potential target for AD.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Canais de Cátion TRPV , Acetilcolinesterase/metabolismo , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/efeitos adversos , Animais , Calcineurina/metabolismo , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Calsequestrina , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Glicogênio Sintase Quinase 3 beta , Parvalbuminas , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPV/metabolismo , ortoaminobenzoatos
14.
Comb Chem High Throughput Screen ; 25(5): 919-933, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33966617

RESUMO

AIM: The aim of this study was to investigate the effect and molecular mechanism of Scutellaria baicalensis Georgi stems and leaves flavonoids (SSF) in promoting neurogenesis and improving memory impairment induced by the PI3K-AKT-CREB signaling pathway. METHODS: Alzheimer's disease (AD) was induced in the male Wistar rats by intracerebroventricular injection of amyloid beta peptide 25-35 (Aß25-35) in combination with aluminum trichloride (AlCl3) and recombinant human transforming growth factor-ß1(RHTGF-ß1) (composited Aß). The Morris water maze was used to screen the successful establishment of the memory impairment model of rats. The screened successful model rats were randomly divided into a model group and three groups of three different doses of the drug (SSF). Rats in the drug group were treated with 35, 70, and 140 mg/kg of SSF for 43 days. The Eight-arm maze was used to measure the spatial learning and memory abilities of the rat, including working memory errors (WME) and reference memory errors (RME). Immunohistochemistry was used to detect the expression of BrdU, an indicator of neuronal proliferation, in the hippocampal gyrus of rats. The mRNA and protein expressions of TRKB, PI3K, AKT, P-AKT, and IGF2 in the PI3K-AKT-CREB signaling pathway in the hippocampus and cerebral cortex of the rats were determined by quantitative real-time PCR (qPCR) and Western blotting methods. RESULTS: Compared to the sham group, the spatial memory ability of rats with composited Aß was decreased, the number of WME and RME (P < 0.01) was increased, the expression of BrdU protein (P < 0.01) in the hippocampal gyrus was reduced, the mRNA and protein expression levels of TRKB, AKT, and IGF2 (P < 0.01, P < 0.05) in the hippocampus and cerebral cortex were lowered, and the mRNA expression level of PI3K (P < 0.01) in the cerebral cortex and the protein expression level of PI3K (P < 0.01) in the hippocampus were augmented. However, compared to the model group, the three-doses of SSF improved memory disorder induced by composited Aß, reduced the number of WME and RME, increased the expression of BrdU protein in the hippocampal gyrus, and differently regulated the mRNA and protein expressions in composited Aß rats. CONCLUSION: SSF improved memory impairment and neurogenesis disorder induced by composited Aß in rats by activating the PI3K-AKT-CREB signaling pathway and up-regulating the mRNA and protein expressions of TRKB, PI3K, AKT, CREB, and IGF2.


Assuntos
Doença de Alzheimer , Scutellaria baicalensis , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/efeitos adversos , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Flavonoides/farmacologia , Masculino , Transtornos da Memória/tratamento farmacológico , Neurogênese , Fosfatidilinositol 3-Quinases/metabolismo , Folhas de Planta , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Scutellaria baicalensis/química , Scutellaria baicalensis/metabolismo , Transdução de Sinais
15.
Artigo em Inglês | MEDLINE | ID: mdl-34363866

RESUMO

The accumulation of amyloid-ß (Aß) peptides in the brain is considered to be the initial event in the Alzheimer's disease (AD). Neurotoxicity mediated by Aß has been demonstrated to damage the cognitive function. In the present study, we sought to determine the effects of O-1602, a specific G-protein coupled receptor 55 (GPR55) agonist, on the impairment of learning and memory induced by intracerebroventricular (i.c.v.) of Aß1-42 (400 pmol/mouse) in mice. Our results showed that i.c.v. injection of aggregated Aß1-42 into the brain of mice resulted in cognitive impairment and neurotoxicity. In contrast, O-1602 (2.0 or 4.0 µg/mouse, i.c.v.) can improve memory impairment induced by Aß1-42 in the Morris water maze (MWM), and novel object recognition (NOR) tests. Besides, we found that O-1602 reduced the activity of ß-secretase 1 (BACE1) and the level of soluble Aß1-42 in the hippocampus and frontal cortex. Importantly, O-1602 treatment reversed Aß1-42-induced GPR55 down-regulation, decreased pro-inflammatory cytokines, and the level of malondialdehyde (MDA), increased the levels of glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT), as well as suppressed apoptosis as indicated by decreased TUNEL-positive cells, and increased the ratio of Bcl-2/Bax. O-1602 treatment also pronouncedly ameliorated synaptic dysfunction by promoting the upregulation of PSD-95 and synaptophysin (SYN) proteins. Moreover, O-1602 concurrently down regulated the protein levels of RhoA, and ROCK2, the critical proteins in the RhoA/ROCK2 pathway. This study indicates that O-1602 may reverse Aß1-42-induced cognitive impairment and neurotoxicity in mice by inhibiting RhoA/ROCK2 pathway. Taken together, these findings suggest that GPR55 could be a novel and promising target for the treatment of AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides , Canabidiol/análogos & derivados , Disfunção Cognitiva/tratamento farmacológico , Síndromes Neurotóxicas , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/efeitos adversos , Peptídeos beta-Amiloides/metabolismo , Animais , Apoptose/efeitos dos fármacos , Encéfalo/metabolismo , Canabidiol/administração & dosagem , Modelos Animais de Doenças , Hipocampo/metabolismo , Infusões Intraventriculares , Transtornos da Memória/induzido quimicamente , Camundongos , Fragmentos de Peptídeos , Receptores de Canabinoides/genética
16.
Cells ; 10(12)2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34943956

RESUMO

The lack of an animal model or an in vitro model limits experimental options for studying temporal molecular events in pseudoexfoliation syndrome (PXF), an age related fibrillopathy causing trabecular meshwork damage and glaucoma. Our goal was to create a workable in vitro model of PXF using primary human TM (HTM) cell lines simulating human disease. Primary HTM cells harvested from healthy donors (n = 3), were exposed to various concentrations (5 ng/mL, 10 ng/mL, 15 ng/mL) of transforming growth factor-beta1 (TGF-ß1) for different time points. Morphological change of epithelial-mesenchymal transition (EMT) was analyzed by direct microscopic visualization and immunoblotting for EMT markers. Expression of pro-fibrotic markers were analyzed by quantitative RT-PCR and immunoblotting. Cell viability and death in treated cells was analyzed using FACS and MTT assay. Protein complex and amyloid aggregate formation was analyzed by Immunofluorescence of oligomer11 and amyloid beta fibrils. Effect of these changes with pharmacological inhibitors of canonical and non-canonical TGF pathway was done to analyze the pathway involved. The expression of pro-fibrotic markers was markedly upregulated at 10 ng/mL of TGF-ß1 exposure at 48-72 h of exposure with associated EMT changes at the same time point. Protein aggregates were seen maximally at these time points that were found to be localized around the nucleus and in the extracellular matrix (ECM). EMT and pro-fibrotic expression was differentially regulated by different canonical and non-canonical pathways suggesting complex regulatory mechanisms. This in vitro model using HTM cells simulated the main characteristics of human disease in PXF like pro-fibrotic gene expression, EMT, and aggregate formation.


Assuntos
Síndrome de Exfoliação/genética , Agregados Proteicos/genética , Malha Trabecular/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Peptídeos beta-Amiloides/efeitos adversos , Peptídeos beta-Amiloides/genética , Sobrevivência Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Síndrome de Exfoliação/complicações , Síndrome de Exfoliação/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glaucoma/complicações , Glaucoma/genética , Glaucoma/patologia , Humanos , Cultura Primária de Células/métodos , Agregados Proteicos/efeitos dos fármacos , Malha Trabecular/patologia , Fator de Crescimento Transformador beta1/genética
17.
Mol Biol Rep ; 48(12): 7667-7676, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34724130

RESUMO

BACKGROUND: Studies showed the protective role of Salvia in traditional medicine against neurodegenerative diseases. Salvia macilenta is one of the potent antioxidant herbs among Salvia species against oxidative stress. In the current study, the effect of oral administration of S. macilenta in the antioxidant, anti-inflammatory activities of Aß-injected male albino Wistar rats was determined. METHODS: Rats were received S. macilenta (50 mg/kg/day) orally, for ten successive days and then some of them received Aß (10 ng/µl) in their hippocampus (CA1 region). Proteins involved in antioxidant defense system and inflammatory signaling pathways in the hippocampus and prefrontal cortex were evaluated using Western blotting technique. To study apoptosis, Western blotting technique and histological staining were used. Catalase activity, glutathione peroxidase (GSH) and nitric oxide levels were measured. RESULTS: Results demonstrated that S. macilenta increased Nrf2 protein level and decreased TNFα and IL-6 protein level in Aß-injected rats compared to the Aß-injected group in the hippocampus and prefrontal cortex. Histological analysis showed pretreatment with S. macilenta decreased apoptosis levels in the hippocampus and prefrontal cortex, about 41 and 42%, compared to Aß-injected rats, respectively. This study showed that catalase activity was changed in the S. macilenta + Aß group compared to the Aß-injected rats. Also, GSH level was increased in the S. macilenta + Aß group compared to the Aß-injected rat. CONCLUSION: Orally treatment of S. macilenta extract in Aß-injected rats could ameliorate protective pathways and, so, it can be one of the proposed dietary supplements for the prevention of Alzheimer's disease and dementia.


Assuntos
Fator 2 Relacionado a NF-E2/metabolismo , Extratos Vegetais/farmacologia , Salvia/metabolismo , Peptídeos beta-Amiloides/efeitos adversos , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Suplementos Nutricionais , Inflamação/metabolismo , Interleucina-6/metabolismo , Masculino , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
18.
Int J Mol Sci ; 22(21)2021 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-34769380

RESUMO

Despite the well-accepted role of the two main neuropathological markers (ß-amyloid and tau) in the progression of Alzheimer's disease, the interaction and specific contribution of each of them is not fully elucidated. To address this question, in the present study, an adeno-associated virus (AAV9) carrying the mutant P301L form of human tau, was injected into the dorsal hippocampi of APP/PS1 transgenic mice or wild type mice (WT). Three months after injections, memory tasks, biochemical and immunohistochemical analysis were performed. We found that the overexpression of hTauP301L accelerates memory deficits in APP/PS1 mice, but it did not affect memory function of WT mice. Likewise, biochemical assays showed that only in the case of APP/PS1-hTauP301L injected mice, an important accumulation of tau was observed in the insoluble urea fraction. Similarly, electron microscopy images revealed that numerous clusters of tau immunoparticles appear at the dendrites of APP/PS1 injected mice and not in WT animals, suggesting that the presence of amyloid is necessary to induce tau aggregation. Interestingly, these tau immunoparticles accumulate in dendritic mitochondria in the APP/PS1 mice, whereas most of mitochondria in WT injected mice remain free of tau immunoparticles. Taken together, it seems that amyloid induces tau aggregation and accumulation in the dendritic mitochondria and subsequently may alter synapse function, thus, contributing to accelerate cognitive decline in APP/PS1 mice.


Assuntos
Doença de Alzheimer/complicações , Peptídeos beta-Amiloides/efeitos adversos , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Mitocôndrias/patologia , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/metabolismo , Fosforilação , Presenilina-1/fisiologia , Sinapses , Proteínas tau/genética
19.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34638977

RESUMO

The acetylcholinesterase inhibitors donepezil and rivastigmine have been used as therapeutic drugs for Alzheimer's disease (AD), but their effects on LPS- and Aß-induced neuroinflammatory responses and the underlying molecular pathways have not been studied in detail in vitro and in vivo. In the present study, we found that 10 or 50 µM donepezil significantly decreased the LPS-induced increases in the mRNA levels of a number of proinflammatory cytokines in BV2 microglial cells, whereas 50 µM rivastigmine significantly diminished only LPS-stimulated IL-6 mRNA levels. In subsequent experiments in primary astrocytes, donepezil suppressed only LPS-stimulated iNOS mRNA levels. To identify the molecular mechanisms by which donepezil regulates LPS-induced neuroinflammation, we examined whether donepezil alters LPS-stimulated proinflammatory responses by modulating LPS-induced downstream signaling and the NLRP3 inflammasome. Importantly, we found that donepezil suppressed LPS-induced AKT/MAPK signaling, the NLRP3 inflammasome, and transcription factor NF-kB/STAT3 phosphorylation to reduce neuroinflammatory responses. In LPS-treated wild-type mice, a model of neuroinflammatory disease, donepezil significantly attenuated LPS-induced microglial activation, microglial density/morphology, and proinflammatory cytokine COX-2 and IL-6 levels. In a mouse model of AD (5xFAD mice), donepezil significantly reduced Aß-induced microglial and astrocytic activation, density, and morphology. Taken together, our findings indicate that donepezil significantly downregulates LPS- and Aß-evoked neuroinflammatory responses in vitro and in vivo and may be a therapeutic agent for neuroinflammation-associated diseases such as AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/efeitos adversos , Inibidores da Colinesterase/administração & dosagem , Donepezila/administração & dosagem , Inflamassomos/metabolismo , Lipopolissacarídeos/efeitos adversos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Rivastigmina/farmacologia , Fator de Transcrição STAT3/metabolismo , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/genética , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/metabolismo
20.
Oxid Med Cell Longev ; 2021: 1020614, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34616501

RESUMO

Astragaloside IV (AS-IV) is an active component in Astragalus membranaceus with the potential to treat neurodegenerative diseases, especially Alzheimer's diseases (ADs). However, its mechanisms are still not known. Herein, we aimed to explore the systematic pharmacological mechanism of AS-IV for treating AD. Drug prediction, network pharmacology, and functional bioinformatics analyses were conducted. Molecular docking was applied to validate reliability of the interactions and binding affinities between AS-IV and related targets. Finally, experimental verification was carried out in AßO infusion produced AD-like phenotypes to investigate the molecular mechanisms. We found that AS-IV works through a multitarget synergistic mechanism, including inflammation, nervous system, cell proliferation, apoptosis, pyroptosis, calcium ion, and steroid. AS-IV highly interacted with PPARγ, caspase-1, GSK3Β, PSEN1, and TRPV1 after docking simulations. Meanwhile, PPARγ interacts with caspase-1, GSK3Β, PSEN1, and TRPV1. In vivo experiments showed that AßO infusion produced AD-like phenotypes in mice, including impairment of fear memory, neuronal loss, tau hyperphosphorylation, neuroinflammation, and synaptic deficits in the hippocampus. Especially, the expression of PPARγ, as well as BDNF, was also reduced in the hippocampus of AD-like mice. Conversely, AS-IV improved AßO infusion-induced memory impairment, inhibited neuronal loss and the phosphorylation of tau, and prevented the synaptic deficits. AS-IV prevented AßO infusion-induced reduction of PPARγ and BDNF. Moreover, the inhibition of PPARγ attenuated the effects of AS-IV on BDNF, neuroflammation, and pyroptosis in AD-like mice. Taken together, AS-IV could prevent AD-like phenotypes and reduce tau hyperphosphorylation, synaptic deficits, neuroinflammation, and pyroptosis, possibly via regulating PPARγ.


Assuntos
Doença de Alzheimer/prevenção & controle , Astragalus propinquus/química , Medicamentos de Ervas Chinesas/administração & dosagem , Doenças Neuroinflamatórias/prevenção & controle , Fenótipo , Fitoterapia/métodos , Saponinas/administração & dosagem , Triterpenos/administração & dosagem , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/efeitos adversos , Animais , Biologia Computacional/métodos , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/metabolismo , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular/métodos , Farmacologia em Rede/métodos , Doenças Neuroinflamatórias/induzido quimicamente , Doenças Neuroinflamatórias/metabolismo , PPAR gama/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/efeitos adversos , Fosforilação/efeitos dos fármacos , Piroptose/efeitos dos fármacos , Saponinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Máquina de Vetores de Suporte , Triterpenos/metabolismo , Proteínas tau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...