Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Phytomedicine ; 126: 155372, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38382281

RESUMO

BACKGROUND: Renal fibrosis is a common pathway that drives the advancement of numerous kidney maladies towards end-stage kidney disease (ESKD). Suppressing renal fibrosis holds paramount clinical importance in forestalling or retarding the transition of chronic kidney diseases (CKD) to renal failure. Schisandrin A (Sch A) possesses renoprotective effect in acute kidney injury (AKI), but its effects on renal fibrosis and underlying mechanism(s) have not been studied. STUDY DESIGN: Serum biochemical analysis, histological staining, and expression levels of related proteins were used to assess the effect of PKCß knockdown on renal fibrosis progression. Untargeted metabolomics was used to assess the effect of PKCß knockdown on serum metabolites. Unilateral Ureteral Obstruction (UUO) model and TGF-ß induced HK-2 cells and NIH-3T3 cells were used to evaluate the effect of Schisandrin A (Sch A) on renal fibrosis. PKCß overexpressed NIH-3T3 cells were used to verify the possible mechanism of Sch A. RESULTS: PKCß was upregulated in the UUO model. Knockdown of PKCß mitigated the progression of renal fibrosis by ameliorating perturbations in serum metabolites and curbing oxidative stress. Sch A alleviated renal fibrosis by downregulating the expression of PKCß in kidney. Treatment with Sch A significantly attenuated the upregulated proteins levels of FN, COL-I, PKCß, Vimentin and α-SMA in UUO mice. Moreover, Sch A exhibited a beneficial impact on markers associated with oxidative stress, including MDA, SOD, and GSH-Px. Overexpression of PKCß was found to counteract the renoprotective efficacy of Sch A in vitro. CONCLUSION: Sch A alleviates renal fibrosis by inhibiting PKCß and attenuating oxidative stress.


Assuntos
Ciclo-Octanos , Nefropatias , Lignanas , Compostos Policíclicos , Obstrução Ureteral , Camundongos , Animais , Fator de Crescimento Transformador beta1/metabolismo , Nefropatias/tratamento farmacológico , Rim , Fibrose , Obstrução Ureteral/patologia , Estresse Oxidativo
3.
J Ethnopharmacol ; 316: 116729, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37277081

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Saikosaponins B2 (SSB2) is one of the main active components isolated from Radix Bupleuri (Bupleurum chinense DC.), a herb widely used of traditional Chinese medicine. It has been used for the treatment of depression for more than two thousand years. However, the molecular mechanisms remain to be determined. AIM OF THE STUDY: In this study, we evaluated the anti-inflammatory effect and elucidated underlying molecular mechanisms of SSB2 in LPS-induced primary microglia and CUMS-induced mice model of depression. METHOD: The effects of SSB2 treatment were investigated both in vitro and in vivo. The chronic unpredictable mild stimulation (CUMS) procedure was applied to establish the animal model of depression. Behavioural tests were used to evaluate the depressive-like behaviors in CUMS-exposed mice, including sucrose preference test, open field test, tail suspension test, and forced swimming test. The GPX4 gene of microglia was silenced using shRNA, and inflammatory cytokines were determined by Western Blot and immunofluorescence analysis. Endoplasmic reticulum stress and ferroptosis-related markers were detected by qPCR, flow cytometry and confocal microscopy. RESULT: SSB2 reversed depressive-like behaviours in CUMS-exposed mice and relieved central neuroinflammation and ameliorated hippocampal neural damage. SSB2 alleviated LPS-induced activation of microglia through the TLR4/NF-κB pathway. LPS-induced ferroptosis, with increased levels of ROS, intracellular Fe2+, mitochondrial membrane potential, lipid peroxidation, GSH, SLC7A11, FTH, GPX4 and Nrf2, and decreased transcription levels of ACSL4 and TFR1, was attenuated with SSB2 treatment in primary microglia cells. GPX4 knockdown activated ferroptosis, induced endoplasmic reticulum (ER) stress, and abrogated the protective effects of SSB2. Further, SSB2 attenuated ER stress, balanced calcium homeostasis, reduced lipid peroxidation and intracellular Fe2+ content by regulating the level of intracellular Ca2+. CONCLUSIONS: Our study suggested that SSB2 treatment can inhibit ferroptosis, maintain calcium homeostasis, relieve endoplasmic reticulum stress and attenuate central neuroinflammation. SSB2 exhibited anti-ferroptosis and anti-neuroinflammatory effects through the TLR4/NF-κB pathway in a GPX4-dependent manner.


Assuntos
Depressão , NF-kappa B , Camundongos , Animais , NF-kappa B/metabolismo , Depressão/tratamento farmacológico , Transdução de Sinais , Doenças Neuroinflamatórias , Microglia/metabolismo , Receptor 4 Toll-Like/metabolismo , Lipopolissacarídeos/farmacologia , Cálcio/metabolismo , Estresse do Retículo Endoplasmático , Estresse Psicológico/tratamento farmacológico , Hipocampo/metabolismo , Modelos Animais de Doenças
4.
Cell Cycle ; 22(23-24): 2584-2601, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38197217

RESUMO

The family of high mobility group box (HMGB) proteins participates in various biological processes including immunity, inflammation, as well as cancer formation and progression. However, its role in thyroid cancer remains to be clarified. We performed quantitative RT-PCR (qRT-PCR), western blot, enzyme-linked immunosorbent, immunohistochemistry, and immunofluorescence assays to evaluate the expression level and subcellular location of HMGB3. The effects of HMGB3 knockdown on malignant biological behaviors of thyroid cancer were determined by cell proliferation assays, cell cycle and apoptosis assays, and transwell chamber migration and invasion assays. Differential expression genes (DEGs) altered by HMGB3 were analyzed using the Ingenuity Pathway Analysis (IPA) and TRRUST v2 database. HMGB3 correlated pathways predicted by bioinformatic analysis were then confirmed using western blot, co-immunoprecipitation, dual-luciferase reporter assay, and flow cytometry. We found that HMGB3 is overexpressed and its downregulation inhibits cell viability, promotes cell apoptosis and cell cycle arrest, and suppresses cell migration and invasion in thyroid cancer. In PTC, both tissue and serum levels of HMGB3 are elevated and are correlated with lymph node metastasis and advanced tumor stage. Mechanistically, we observed the translocation of HMGB3 in PTC, induced at least partially by hypoxia. Cytoplasmic HMGB3 activates nucleic-acid-mediated TLR3/NF-κB signaling and extracellular HMGB3 interacts with the transmembrane TREM1 receptor in PTC. This study demonstrates the oncogenic role of HMGB3 cytoplasmic and extracellular translocation in papillary thyroid cancers; we recommend its future use as a potential circulating biomarker and therapeutic target for PTC.


Assuntos
Proteína HMGB3 , MicroRNAs , Neoplasias da Glândula Tireoide , Humanos , Câncer Papilífero da Tireoide/genética , Câncer Papilífero da Tireoide/patologia , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo , Linhagem Celular Tumoral , Receptor Gatilho 1 Expresso em Células Mieloides/genética , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo , Neoplasias da Glândula Tireoide/genética , Proteína HMGB3/genética , Proteína HMGB3/metabolismo , Proliferação de Células/genética , MicroRNAs/genética , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica
5.
Genet Test Mol Biomarkers ; 26(4): 159-173, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35481968

RESUMO

Background: Low selenium levels are associated with an increased incidence and advanced stage of thyroid cancers (THCAs). In response to changes in selenium levels, a hierarchy of selenoprotein biosynthesis allows tissue-specific fine-tuning of the 25 selenoproteins. To determine the role of individual selenoproteins on thyroid carcinogenesis, we carried out a multiomic data mining study. Methods: The expression levels of individual selenoproteins and their correlations with prognosis in THCAs were analyzed using Oncomine, GEPIA, and Kaplan-Meier plotter platforms. Co-expression analyses using the cBioportal database were carried out to identify genes that are correlated with selenoproteins. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichments were performed for genes correlated with selenoproteins that were identified as clinically significant. Results and Discussion: DIO1, GPX3, SELENOO, SELENOP, SELENOS, and SELENOV were significantly downregulated in THCAs and were associated with poor prognoses. Biological processes including negative regulation of growth and angiogenesis were enriched in DIO1-positively and DIO1-negatively correlated genes, respectively. Many biological processes including negative regulation of growth and MAPK cascade were enriched in GPX3-positively and GPX3-negatively correlated genes, respectively. The antitumor effects of SELENOS might be attributed to their protection against endoplasmic reticulum (ER) stress. SELENOO was revealed to be correlated with ER stress, mitochondrial translation, and telomere maintenance. Biological processes of SELENOV-correlated genes were enriched in redox processes and ER calcium ion homeostasis. Moreover, cell adhesion and angiogenesis were also shown to be negatively regulated by SELENOV, providing an antimetastatic effect similar as DIO1. Conclusion: This study explored the distinct roles of the 25 selenoproteins in THCA pathogenesis, providing potential oncosuppressing effects of 6 selenoproteins.


Assuntos
Selênio , Neoplasias da Glândula Tireoide , Humanos , Prognóstico , Selênio/metabolismo , Selenoproteínas/genética , Selenoproteínas/metabolismo , Neoplasias da Glândula Tireoide/genética
6.
J Ethnopharmacol ; 287: 114967, 2022 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-34995692

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Astragali Radix (AR) is a popular traditional Chinese medicine that has been used for more than 2000 years. It is a well-known tonic for weak people with chronic diseases, such as heart failure and cerebral ischemia. Previous studies have reported that AR could support the "weak heart" of cancer patients who suffered from doxorubicin (DOX)-induced cardiotoxicity (DIC). However, the underlying mechanism remains unclear. AIM OF THE STUDY: This study aimed to uncover the critical pathways and molecular determinants for AR against DIC by fully characterizing the network-based relationship. MATERIALS AND METHODS: We integrated ultra-high-performance liquid chromatography-high-resolution mass spectrometry (UHPLC-HRMS) profiling, database and literature searching, and the human protein-protein interactome to discover the specific network module associated with AR against DIC. To validate the network-based findings, a low-dose, long-term DIC mouse model and rat cardiomyoblast H9c2 cells were employed. The levels of potential key metabolites and proteins in hearts and cells were quantified by the LC-MS/MS targeted analysis and western blotting, respectively. RESULTS: We constructed one of the most comprehensive AR component-target network described to date, which included 730 interactions connecting 64 unique components and 359 unique targets. Relying on the network-based evaluation, we identified fatty acid metabolism as a putative critical pathway and peroxisome proliferator-activated receptors (PPARα and PPARγ) as potential molecular determinants. We then confirmed that DOX caused the accumulation of fatty acids in the mouse failing heart, while AR promoted fatty acid metabolism and preserved heart function. By inhibiting PPARγ in H9c2 cells, we further found that AR could alleviate DIC by activating PPARγ to maintain fatty acid homeostasis. CONCLUSIONS: Our findings imply that AR is a promising drug candidate that treats DIC by maintaining fatty acid homeostasis. More importantly, the network-based method developed here could facilitate the mechanism discovery of AR therapy and help catalyze innovation in its clinical application.


Assuntos
Cardiotoxicidade/prevenção & controle , Doxorrubicina/toxicidade , Medicamentos de Ervas Chinesas/farmacologia , Mioblastos Cardíacos/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/toxicidade , Astragalus propinquus , Cardiotoxicidade/etiologia , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Ácidos Graxos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos Cardíacos/patologia , Farmacologia em Rede , Ratos , Espectrometria de Massas em Tandem
7.
Neurotox Res ; 39(3): 872-885, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33025359

RESUMO

Inflammation plays an important role in depression pathology, making it a promising target for ameliorating depression-like behaviors. The peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) is a transcriptional coactivator being able to constrain inflammatory events through NF-κB signaling. However, the role of PGC-1α in depression is not yet clear. This study was designed to investigate the role of PGC-1α in depression and explore the underlying mechanisms. Mice modeled with chronic unpredictable mild stimulation (CUMS) were explored for the relationship between depression-like behaviors and PGC-1α. Baicalin was used to evaluate the effect regulating PGC-1α. Furthermore, the anti-neuroinflammatory effect of baicalin was investigated both in BV2-SH-SY5Y co-culture system and in mice by LPS challenge. The role of PGC-1α in neuroinflammation was explored in cell co-culture systems under gene silencing conditions targeting NF-κB signaling. We found that the expression of PGC-1α was inhibited in the hippocampus of mice exposed to CUMS or LPS, while baicalin could increase the expression of PGC-1α and alleviate the depression-like behaviors. Furthermore, baicalin attenuated neuroinflammation in the hippocampus of mice and BV2-SH-SY5Y co-culture system by LPS challenge via regulating NF-κB signaling; however, knockdown of the PGC-1α could reverse the effect of baicalin on neuroinflammation and NF-κB signaling. Our results revealed a vital role for PGC-1α in attenuating neuroinflammation in depression, indicating that PGC-1α might be a therapeutic target for depression.


Assuntos
Depressão/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/biossíntese , Regulação para Cima/fisiologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Linhagem Celular Tumoral , Depressão/tratamento farmacológico , Depressão/psicologia , Relação Dose-Resposta a Droga , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Elevação dos Membros Posteriores/efeitos adversos , Elevação dos Membros Posteriores/psicologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/psicologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Regulação para Cima/efeitos dos fármacos
8.
Clin Transl Med ; 10(8): e243, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33377633

RESUMO

Neural stem/progenitor cells (NPCs) are multipotent stem cells in the central nervous system. Damage to NPCs has been demonstrated to cause adverse effects on neurogenesis and to contribute to neurological diseases. Our previous research suggested that saikosaponin-d (SSd), a cytostatic drug belonging to the bioactive triterpenoid saponins, exhibited neurotoxicity by inhibiting hippocampal neurogenesis, but the underlying mechanism remained elusive. This study was performed to clarify the role of SSd in cognitive function and the mechanism by which SSd induced damage to hippocampal neurogenesis and NPCs. Our results indicated that SSd caused hippocampus-dependent cognitive deficits and inhibited hippocampal neurogenesis by reducing the numbers of newborn neurons in mice. RNA sequencing analysis revealed that SSd-induced neurotoxicity in the hippocampus involved neurotrophin receptor-interacting MAGE (NRAGE)/neurotrophin receptor interacting factor (NRIF)/p75NTR -associated cell death executor (NADE) cell signaling activated by the p75 neurotrophin receptor (p75NTR ). Mechanistic studies showed that a short hairpin RNA targeting p75NTR intracellular domain reversed SSd-increased NRAGE/NRIF/NADE signaling and the c-Jun N-terminal kinase/caspase apoptotic pathway, subsequently contributing to the survival of NPCs, as well as cell proliferation and differentiation. The addition of recombinant brain-derived neurotrophic factor (BDNF) ameliorated the SSd-induced inhibition of BDNF/Tyrosine kinase receptor B (TrkB) neurotrophic signaling, but did not affect SSd-activated pro-BDNF/p75NTR signaling. Moreover, the SSd-induced elevation of cytosolic Ca2+ concentration was responsible for damage to NPCs. The extracellular Ca2+ chelator ethylene glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid (EGTA), rather than the intracellular Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl ester) (BAPTA/AM), attenuated SSd-induced cytosolic Ca2+ dysfunction and SSd-disordered TrkB/p75NTR signaling. Overall, this study demonstrated a new mechanism for the neurotoxic effect of SSd, which has emerging implications for pharmacological research of SSd and provides a better understanding of neurotoxicity induced by cytostatic drugs.

9.
CNS Neurol Disord Drug Targets ; 18(7): 566-577, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31389320

RESUMO

BACKGROUND: Alzheimer's disease is one of the most common neurodegenerative diseases in many modern societies. The core pathogenesis of Alzheimer's disease includes the aggregation of hyperphosphorylated Tau and abnormal Amyloid-ß generation. In addition, previous studies have shown that neuroinflammation is one of the pathogenesis of Alzheimer's disease. Formononetin, an isoflavone compound extracted from Trifolium pratense L., has been found to have various properties including anti-obesity, anti-inflammation, and neuroprotective effects. But there are very few studies on the treatment of Alzheimer's disease with Formononetin. OBJECTIVE: The present study focused on the protective activities of Formononetin on a high-fat dietinduced cognitive decline and explored the underlying mechanisms. METHODS: Mice were fed with HFD for 10 weeks and intragastric administrated daily with metformin (300 mg/kg) and Formononetin (20 and 40 mg/kg). RESULTS: We found that Formononetin (20, 40 mg/kg) significantly attenuated the learning and memory deficits companied by weight improvement and decreased the levels of blood glucose, total cholesterol and triglyceride in high-fat diet-induced mice. Meanwhile, we observed high-fat diet significantly caused the Tau hyperphosphorylation in the hippocampus of mice, whereas Formononetin reversed this effect. Additionally, Formononetin markedly reduced the levels of inflammation cytokines IL-1ß and TNF-α in high-fat diet-induced mice. The mechanism study showed that Formononetin suppressed the pro-inflammatory NF-κB signaling and enhanced the anti-inflammatory Nrf-2/HO-1 signaling, which might be related to the regulation of PGC-1α in the hippocampus of high-fat diet -induced mice. CONCLUSION: Taken together, our results showed that Formononetin could improve the cognitive function by inhibiting neuroinflammation, which is attributed to the regulation of PGC-1α pathway in HFD-induced mice.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Dieta Hiperlipídica , Inflamação/tratamento farmacológico , Isoflavonas/farmacologia , Nootrópicos/farmacologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Dieta Hiperlipídica/efeitos adversos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Masculino , Camundongos Endogâmicos ICR , Proteínas tau/metabolismo
10.
Behav Brain Res ; 372: 112007, 2019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31238056

RESUMO

Mitochondrial dynamics regulate mitochondrial autophagy (mitophagy) and apoptosis, which are important events for the quality control of mitochondria and mitochondrial-associated diseases. Esculetin (ESC) is a natural coumarin that exhibits inspiring biological activities in a variety of animal models, but its neuroprotective effects on cerebral ischaemia have not been clearly elucidated. In this paper, we demonstrated the effects of ESC on transient cerebral ischaemia and reperfusion injury induced in a mouse model and examined the possible underlying mechanisms by investigating mitochondrial fragmentation-regulated mitochondrial autophagy and apoptosis. The experimental results showed that ESC treatment alleviated neurological defects and improved cognitive impairments in transient bilateral common carotid artery occlusion (tBCCAO)-treated mice. Further mechanism studies showed that tBCCAO induced mitochondrial oxidative stress injuries and triggered mitochondrial fragmentation, which were evident by the elevated levels of malondialdehyde and mitochondrial dynamin-related protein 1 (Drp1) and the downregulated activities of superoxide dismutase and nuclear transcription factor E2-related factor 2 (Nrf2). ESC treatment significantly alleviated tBCCAO-induced mitochondrial stress and mitochondrial fragmentation. Moreover, mitophagy and mitochondrial apoptosis were stimulated in response to the mitochondrial oxidative stress in the hippocampus of tBCCAO-treated mice, and ESC treatment regulated the expression of mitophagy-related factors, including Bnip3, Beclin1, Pink1, and parkin, the LC-3 II/I ratio, and apoptosis-related factors, including p53, Bax, and caspase 3. Taken together, our results suggest that ESC treatment regulated hippocampal mitophagy and mitochondrial apoptosis triggered by mitochondrial stress via the mediation of mitochondrial fragmentation during transient cerebral ischaemia and reperfusion injury, which provides insight into the potential of ESC for further therapeutic implications.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Ataque Isquêmico Transitório/tratamento farmacológico , Umbeliferonas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Dinaminas/análise , Ataque Isquêmico Transitório/metabolismo , Masculino , Malondialdeído/análise , Camundongos , Camundongos Endogâmicos ICR , Mitocôndrias/metabolismo , Mitofagia/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/análise , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Superóxido Dismutase/análise , Umbeliferonas/metabolismo
11.
Toxicology ; 424: 152233, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31175886

RESUMO

Saikosaponins-d (SSd) is a major bioactive compound isolated from Radix Bupleuri, an herb widely used in traditional Chinese medicine. Emerging studies demonstrate that SSd adversely affects adult neurogenesis and impairs learning and memory. However, the molecular mechanisms remain to be determined. The current study investigated the potential regulatory of GSK3ß/ß-catenin signaling on SSd-induced neurotoxicity. We demonstrated that SSd exposure inhibited the cell viability and proliferation of primary neuronal stem/progenitor cells (NPCs) from hippocampus in a concentration-dependent manner. Significantly, SSd exposure induced activation of GSK3ß and reduced the cellular ß-catenin in NPCs. Treatment with SB216763, a specific inhibitor for GSK3ß activation, we showed that inactivation GSK3ß improved the ß-catenin signaling by inhibiting degradation complex comprising Axin and APC and attenuated SSd-induced toxicity in NPCs. In addition, administration of SB216763 ameliorated SSd-induced inhibition of NPCs proliferation and enhanced radial glial cells in the hippocampus of mice. Moreover, inactivation GSK3ß promoted dendritic arborization and the survival of newborn neurons together with alleviated the impairment of SSd-induced cognitive function in mice. Overall, these data demonstrated that the significant inhibitory effects of SSd on NPCs proliferation and adult neurogenesis via GSK3ß/ß-catenin signaling pathway. Our results contribute to a better understanding of the molecular mechanisms of SSd-induced neurotoxicity.


Assuntos
Glicogênio Sintase Quinase 3 beta/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Ácido Oleanólico/análogos & derivados , Saponinas/toxicidade , Transdução de Sinais/efeitos dos fármacos , beta Catenina/efeitos dos fármacos , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dendritos/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Ácido Oleanólico/toxicidade
12.
Cell Death Dis ; 10(5): 370, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-31068571

RESUMO

Na+/H+ exchanger 1 (NHE1), an important regulator of intracellular pH (pHi) and extracellular pH (pHe), plays a crucial role in various physiological and pathological processes. However, the role of NHE1 in depression has not yet been reported. This study was designed to investigate the role of NHE1 in the animal model of depression and explore the underlying mechanisms. Our results showed that inhibition of rho-associated kinase 2 (ROCK2) by fasudil (Fas) or baicalin (BA) significantly alleviated chronic unpredictable mild stress (CUMS) paradigm-induced depression-related behaviours in mice, as shown by decreased sucrose consumption in sucrose preference test (SPT), reduced locomotor activity in the open field test (OFT), and increased immobility time in the tail suspension test (TST) and forced swimming test (FST). Furthermore, ROCK2 inhibition inhibited the activation of NHE1, calpain1, and reduced neuronal apoptosis in the CUMS animal model of depression. Next, we used the lipopolysaccharide (LPS)-challenged animal model of depression to induce NHE1 activation. Our results revealed that mice subjected to 1 µl LPS (10 mg/ml) injection intracerebroventricularly (i.c.v.) showed depressive-like behaviours and NHE1 activation. Amiloride (Ami), an NHE1 inhibitor, significantly reversed the decrease in sucrose consumption and reduction in immobility time in the TST and FST induced by LPS challenge. Furthermore, Ami decreased the expression of ROCK2, NHE1, calpain1, and caspase-3 and increased the Bcl-1/Bax ratio in the hippocampus of LPS-challenged mice. Ami treatment also led to antidepressive effects in the CUMS-induced animal model of depression. Thus ROCK2 inhibition could be proposed as a neuroprotective strategy against neuronal apoptosis, and NHE1 might be a potential therapeutic target in depression.


Assuntos
Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Trocador 1 de Sódio-Hidrogênio/metabolismo , Amilorida/farmacologia , Animais , Antidepressivos/química , Antidepressivos/metabolismo , Antidepressivos/farmacologia , Apoptose/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Sítios de Ligação , Citocinas/metabolismo , Depressão/patologia , Modelos Animais de Doenças , Flavonoides/química , Flavonoides/metabolismo , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/patologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos ICR , Simulação de Acoplamento Molecular , Transdução de Sinais/efeitos dos fármacos , Trocador 1 de Sódio-Hidrogênio/antagonistas & inibidores , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
13.
J Neuroinflammation ; 16(1): 95, 2019 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-31068207

RESUMO

BACKGROUND: Baicalin, which is isolated from Radix Scutellariae, possesses strong biological activities including an anti-inflammation property. Recent studies have shown that the anti-inflammatory effect of baicalin is linked to toll-like receptor 4 (TLR4), which participates in pathological changes of central nervous system diseases such as depression. In this study, we explored whether baicalin could produce antidepressant effects via regulation of TLR4 signaling in mice and attempted to elucidate the underlying mechanisms. METHODS: A chronic unpredictable mild stress (CUMS) mice model was performed to explore whether baicalin could produce antidepressant effects via the inhibition of neuroinflammation. To clarify the role of TLR4 in the anti-neuroinflammatory efficacy of baicalin, a lipopolysaccharide (LPS) was employed in mice to specially activate TLR4 and the behavioral changes were determined. Furthermore, we used LY294002 to examine the molecular mechanisms of baicalin in regulating the expression of TLR4 in vivo and in vitro using western blot, ELISA kits, and immunostaining. In the in vitro tests, the BV2 microglia cell lines and primary microglia cultures were pretreated with baicalin and LY292002 for 1 h and then stimulated 24 h with LPS. The primary microglial cells were transfected with the forkhead transcription factor forkhead box protein O 1 (FoxO1)-specific siRNA for 5 h and then co-stimulated with baicalin and LPS to investigate whether FoxO1 participated in the effect of baicalin on TLR4 expression. RESULTS: The administration of baicalin (especially 60 mg/kg) dramatically ameliorated CUMS-induced depressive-like symptoms; substantially decreased the levels of interleukin-1 beta (IL-1ß), interleukin-6 (IL-6), and tumor necrosis factor alpha (TNF-α) in the hippocampus; and significantly decreased the expression of TLR4. The activation of TLR4 by the LPS triggered neuroinflammation and evoked depressive-like behaviors in mice, which were also alleviated by the treatment with baicalin (60 mg/kg). Furthermore, the application of baicalin significantly increased the phosphorylation of phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT), and FoxO1. The application of baicalin also promoted FoxO1 nuclear exclusion and contributed to the inhibition of the FoxO1 transactivation potential, which led to the downregulation of the expression of TLR4 in CUMS mice or LPS-treated BV2 cells and primary microglia cells. However, prophylactic treatment of LY294002 abolished the above effects of baicalin. In addition, we found that FoxO1 played a vital role in baicalin by regulating the TLR4 and TLR4-mediating neuroinflammation triggered by the LPS via knocking down the expression of FoxO1 in the primary microglia. CONCLUSION: Collectively, these results demonstrate that baicalin ameliorated neuroinflammation-induced depressive-like behaviors through the inhibition of TLR4 expression via the PI3K/AKT/FoxO1 pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Depressão/imunologia , Flavonoides/farmacologia , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/efeitos dos fármacos , Animais , Depressão/etiologia , Proteína Forkhead Box O1/metabolismo , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Angústia Psicológica/complicações , Angústia Psicológica/imunologia , Receptor 4 Toll-Like/biossíntese
14.
Neuropharmacology ; 155: 150-161, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31145906

RESUMO

Lysophosphatidic acid (LPA) is a bioactive lipid mediator of inflammation that binds to its specific cell surface G protein coupled receptors (LPA1-6). It is reported that LPA induced cell apoptosis by targeting LPA1, while LPA1 blockade eliminated LPS-induced production of peritoneal neutrophil chemokines and cytokines. Previous studies have shown that Saikosaponin-d (SSd) mitigated depressive-like behaviors in rats exposed to chronic unpredictable mild stress (CUMS), as well as corticosterone-induced apoptosis in PC12 cells. The present study explored the role of SSd during modulating LPA1 mediated neuronal apoptosis in LPS-stimulated mice. The phenomenon that SSd alleviated LPS-induced depressive-like behaviors were observed by open field test (OPT), forced swim test (FST) and tail suspension test (TST). SSd inhibited the protein expression of LPA1 both in the CA1 and CA3 region of the hippocampus. Moreover, SSd significantly decreased the levels of RhoA, ROCK2, p-p38, p-ERK, p-p65, p-IκBα in LPS-stimulated mice as well as in LPA-stimulated SH-SY5Y cells. Additionally, SSd significantly decreased the expression of LPA1 and the degree of neuronal apoptosis in SH-SY5Y cells which were co-cultured with LPS-stimulated BV2 microglia. These results suggested that SSd improved LPS-induced depressive-like behaviors in mice and suppressed neuronal apoptosis by regulating LPA1/RhoA/ROCK2 signaling pathway.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Apoptose/fisiologia , Depressão/metabolismo , Neurônios/metabolismo , Ácido Oleanólico/análogos & derivados , Receptores de Ácidos Lisofosfatídicos/metabolismo , Saponinas/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Técnicas de Cocultura , Depressão/induzido quimicamente , Depressão/tratamento farmacológico , Relação Dose-Resposta a Droga , Humanos , Lisofosfolipídeos/metabolismo , Lisofosfolipídeos/toxicidade , Camundongos , Camundongos Endogâmicos ICR , Neurônios/efeitos dos fármacos , Ácido Oleanólico/metabolismo , Ácido Oleanólico/farmacologia , Ácido Oleanólico/uso terapêutico , Distribuição Aleatória , Saponinas/farmacologia , Saponinas/uso terapêutico
15.
Life Sci ; 221: 241-248, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30794828

RESUMO

BACKGROUND AND AIMS: Impaired neurogenesis in hippocampus may contribute to a variety of neurological diseases, such as Alzheimer's disease and depression. Baicalin (BA), which is mainly isolated from the root Scutellaria baicalensis Georgi (traditional Chinese herb), which was reported to facilitate neurogenesis, but how to play the role and the underlying molecular mechanisms are still unknown. MAIN METHODS: In this study, we adopted the chronic unpredictable mild stress (CUMS)-induced mouse model of depression, and then explore antidepressant-like effects and possible molecular mechanisms. KEY FINDINGS: We found that BA significantly increased sucrose consumption in sucrose preference test, the number of crossing in open filed test and attenuated immobility time in tail suspension test. Additionally, BA administration notably promoted neuronal differentiation and the number of DCX+ cells. Moreover, BA facilitated immature neurons develop into mature neurons and their survival. FOXG1, a transcription factor gene, which is crucial for mammalian telencephalon development, specifically stimulates dendrite elongation. BA could reverse the decrease of p-Akt, FOXG1 and FGF2 caused by CUMS-induced. Additionally, the expression of FOXG1 and FGF2 significantly decreased when the Akt pathway were inhibited by LY294002 in SH-SY5Y cells. Interestingly, BA failed to counteract the decline. SIGNIFICANCE: These results suggest that BA could promote the differentiation of neurons, which transformation into mature neurons and their survival via the Akt/FOXG1 pathway to exert antidepressant effects.


Assuntos
Flavonoides/metabolismo , Flavonoides/farmacologia , Animais , Antidepressivos/metabolismo , Antidepressivos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Proteína Duplacortina , Fator 2 de Crescimento de Fibroblastos/efeitos dos fármacos , Fatores de Transcrição Forkhead/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Hipocampo , Masculino , Medicina Tradicional Chinesa , Camundongos , Camundongos Endogâmicos ICR , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Estresse Psicológico
16.
Brain Behav Immun ; 79: 195-206, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30738183

RESUMO

Depression is highly prevalent among patients with chronic obstructive pulmonary disease (COPD). However, depression with COPD comorbidity is often underdiagnosed and undertreated, and pathogenic research is also insufficient. In the present study, we characterised pulmonary and hippocampal dysfunction by researching the interaction between inflammasome-regulated cytokines and glucocorticoid receptor (GR) signalling by investigating the role of fluoxetine (FLU), one of the most widely used antidepressants in clinical practice. Mice were exposed to cigarette smoke (CS) to induce the model of COPD with comorbid depression, and pathological alterations in serum, hippocampus, lung, and bronchoalveolar lavage fluid were determined. Our results showed that the CS procedure induced the accumulation of inflammatory cells (macrophages, neutrophils, and lymphocytes), the production of cytokines, the activation of inflammasome components (NLRP3, ASC, caspase-1), depression-related behaviours, and the stimulation of GR signalling. Intriguingly, glucocorticoid resistance occurred in CS-exposed mice, with elevated serum corticosterone and suppressed hippocampal GR levels, which suggested a novel potential regulatory mechanism underlying COPD-induced depression comorbidity. Furthermore, chronic CS exposure decreased the pGR-S211/pGR-S226 ratio, increased the active nuclear GR, and impaired cytosolic GR binding capacity and GR transcriptional activity, which might be responsible for the activation of the inflammasome-induced inflammatory cascade. These alterations were reversed by chronic FLU treatment, indicating that FLU-mediated GR signalling was involved in the COPD induced inflammasome activation. Our research explored the underlying molecular mechanism of comorbid COPD/depression and provided in vivo evidence that glucocorticoid resistance occurred during CS-induced central nervous system inflammation, a potential mechanism underlying the cross talk between the lung and brain.


Assuntos
Inflamassomos/metabolismo , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Líquido da Lavagem Broncoalveolar/química , Caspase 1/metabolismo , Fumar Cigarros/efeitos adversos , Citocinas/metabolismo , Depressão/metabolismo , Depressão/fisiopatologia , Transtorno Depressivo/metabolismo , Modelos Animais de Doenças , Fluoxetina/farmacologia , Inflamassomos/imunologia , Inflamação/metabolismo , Pulmão/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doença Pulmonar Obstrutiva Crônica/complicações , Receptores de Glucocorticoides/imunologia , Transdução de Sinais , Fumar
17.
Int Immunopharmacol ; 64: 175-182, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30195108

RESUMO

Chronic stress can provoke depressive-like behaviors through activation of inflammation and apoptosis, leading to a reduction of neurons. Antidepressant therapy may contribute to inhibiting inflammation responses and have neuroprotective effects. Baicalin (BA) has an antidepressant effect in the chronic unpredictable mild stress (CUMS) animal model and exerts anti-inflammation, anti-apoptosis, as well as neuroprotective effects in many central nervous system (CNS)-related diseases. But the effects of BA on neuroprotection, apoptosis, and neuroinflammation and the potential mechanisms in depression are unclear. Here, we focused on examining the therapeutic effects of BA in CUMS-induced depression rats and investigating the molecular mechanisms. Results showed that administration of BA improved depressive-like behaviors and significantly increased the levels of doublecortin (DCX), Neuron-specific enolase (NSE), and Brain-derived neurotrophic factor (BDNF) in hippocampus. Furthermore, administration of BA increased the cell survival by reducing the level of malondialdehyde (MDA) and increasing the level of superoxide dismutase (SOD). Finally, administration of BA significantly decreased CUMS-induced apoptosis and inflammatory cytokines (caspase-1 and IL-1ß) in hippocampus. These responses were mediated by Glycogen synthase kinase-3 (GSK3) ß/Nuclear factor-κB (NF-κB)/Nucleotide-binding domain, leucine-rich repeat, pyrin domain containing protein 3 (NLRP3) signal pathway. Taken together, these results indicate that BA could promote neuronal maturation and rescue neurons from apoptosis via inhibiting activation of GSK3ß/NF-κB/NLRP3 signal pathway that is known to be associated with inflammation, thus exerting neuroprotective effects and preventing CUMS-induced depressive-like behaviors.


Assuntos
Depressão/tratamento farmacológico , Flavonoides/farmacologia , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/efeitos dos fármacos , Estresse Psicológico/complicações , Animais , Apoptose/efeitos dos fármacos , Depressão/etiologia , Proteína Duplacortina , Flavonoides/uso terapêutico , Glicogênio Sintase Quinase 3 beta/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Masculino , Atividade Motora/efeitos dos fármacos , NF-kappa B/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Ratos , Ratos Sprague-Dawley
18.
J Mol Neurosci ; 65(3): 391-399, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29974381

RESUMO

Ischemic stroke is one of the most dangerous acute diseases which causes death or deformity. Apoptosis has been shown to play an important role in the development and pathogenesis of cerebral ischemia-reperfusion injury (I/R injury), but the related mechanism is unclear. Levo-tetrahydropalmatine (L-THP), a bioactive ingredient extracted from the Chinese herb Corydalis, can penetrate the blood-brain barrier and exert various pharmacological effects on neural tissues. The present study examined the neuroprotective effect of L-THP on neuronal apoptosis induced by cerebral I/R injury. Results showed that pretreatment with L-THP (12.5, 25, and 50 mg/kg) improved neurological outcomes and reduced infarct volume and cerebral edema in comparison with the brains of the middle cerebral artery occlusion (MCAO) group. These findings provided evidence for the neuroprotective effects of L-THP against cerebral I/R injury. Furthermore, administration of L-THP enhanced the expression of Bcl-2 and attenuated the content of Bax, cleaved caspase-3, and PARP. L-THP could improve the reduction of NeuN-positive cells induced by I/R injury. These results suggested that L-THP could inhibit neuroapoptosis in cerebral ischemic rats. c-Abl was discovered as the critical protein responsible for neurocyte apoptosis; however, few data have been published on the relation between ischemic stroke and the expression of c-Abl. We found that both c-Abl expression and neuronal apoptosis were significantly increased in the MCAO group, while pretreatment with L-THP could ameliorate this effect. Therefore, we deduced that reduced c-Abl overexpression may play a role in the anti-apoptosis effect of L-THP after cerebral I/R injury. Thus, L-THP may provide a potential therapeutic approach for the treatment of ischemic stroke. Graphical Abstract ᅟ.


Assuntos
Apoptose , Alcaloides de Berberina/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-abl/metabolismo , Ratos , Ratos Sprague-Dawley
19.
Shanghai Kou Qiang Yi Xue ; 27(5): 546-553, 2018 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-30680405

RESUMO

PURPOSE: The aim of this meta analysis was to assess the influence of platelet-rich plasma (PRP)combined with demineralized freeze-dried bone allografts(DFDBA) on regeneration of periodontal periodontal defects by means of evaluating clinical and radiographic outcomes in prospective human clinical trials. METHODS: The following databases such as PubMed, The Cochrane Library, EMbase, CNKI, Wanfang data and VIP data were searched on computer from inception to December, 2016. According to the inclusion and exclusion criteria, two reviewers independently extracted the data,assessed the methodological quality of the included studies. RevMan 5.2 was applied for meta analysis. RESULTS: Six papers were obtained reviewed which included 205 periodontal bone defect sites. Six articles showed that there was no significant difference in probing depth decrease between PRP combined with DFDBA and PRP or DFBDA group[MD=0.35, 95%CI(-0.09,0.79), P=0.12], but there was significant difference in clinical attachment loss increase between the two groups[MD= 0.68,95%CI(0.41,0.94),P<0.00001]. Three articles were included for evaluating bone filling, there was significant difference in the distance from the cemento-enamel junction(CEJ) to the vertical bone defect(BD)(CEJ-BD)[MD=0.71,95%CI(0.46,0.95),P<0.00001]between the two groups; there was also significant difference in the distance from the alveolar crest to the vertical bone defect(AC-BD)[MD=0.64,95%CI(0.41,0.87),P<0.00001]between the two groups. but there was no significant difference in the distance from the cemento-enamel junction(CEJ)to the alveolar crest (AC)(CEJ-AC)[MD=0.03,95%CI(-0.10,0.16),P=0.68] between the two groups. CONCLUSIONS: Within the limitations of this meta analysis, PRP combined with DFDBA is superior to PRP or DFDBA alone in clinical attachment loss and bone filling ,but there was no significant difference in probing depth. However, given the limited sample size and quantity of included studies, the above findings still need to be further proved by conducting more high-quality and large-scale RCTs.


Assuntos
Perda do Osso Alveolar , Regeneração Óssea , Transplante Ósseo , Regeneração Tecidual Guiada Periodontal , Plasma Rico em Plaquetas , Aloenxertos , Perda do Osso Alveolar/terapia , Humanos , Perda da Inserção Periodontal , Estudos Prospectivos , Resultado do Tratamento
20.
Eur J Pharmacol ; 815: 118-126, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28743390

RESUMO

Cerebral ischemia/reperfusion (I/R) is a lethal and disabling disease. Studies have suggested that hyperglycemia is a risk factor for cerebral I/R. Baicalin is a natural bioactive flavonoid extracted from Scutellaria baicalensis Georgi with neuroprotective activity. In the present study, we investigated the effects of baicalin on hyperglycemia-exacerbated cerebral I/R injury. Streptozotocin (STZ) injection aggravated the brain damage induced by middle cerebral artery occlusion (MCAO) surgery, while baicalin administration reduced blood glucose, relieved neurological deficit and decreased infarct volume. In vitro, Oxygen-glucose deprivation/ reperfusion (OGD/REP) induced inordinate reactive oxygen species (ROS) production and mitochondrial dynamic impairments were markedly increased under high glucose (HG) condition. Baicalin treatment in PC12 cells inhibited dynamin-related protein 1 (Drp-1) expression, decreased mitochondrial fission, promoted mitofusin-2 (MFN2) generation, increased Drp-1 Ser637 phosphorylation, and elevated mitochondrial membrane potential (Δψm) via the suppression of ROS production. However, AMPKα1 knockdown abolished the protective effects of baicalin. Baicalin also suppressed cell apoptosis and enhanced mitophagy. These results suggested that baicalin protected against hyperglycemia aggravated I/R injury by regulating mitochondrial functions in a manner dependent on AMPK.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Flavonoides/farmacologia , Hiperglicemia/complicações , Mitocôndrias/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/patologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Quinases Associadas com Morte Celular/metabolismo , Ativação Enzimática/efeitos dos fármacos , Flavonoides/uso terapêutico , GTP Fosfo-Hidrolases , Infarto da Artéria Cerebral Média/complicações , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Mitocôndrias/patologia , Dinâmica Mitocondrial/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Células PC12 , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA