Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Commun Signal ; 18(2): e12023, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38946727

RESUMO

Microglia are resident immune cells in the central nervous system that are rapidly activated to mediate neuroinflammation and apoptosis, thereby aggravating brain tissue damage after ischemic stroke (IS). Although scutellarin has a specific therapeutic effect on IS, the potential target mechanism of its treatment has not been fully elucidated. In this study, we explored the potential mechanism of scutellarin in treating IS using network pharmacology. Lipopolysaccharide (LPS) was used to induce an in vitro BV-2 microglial cell model, while middle cerebral artery occlusion (MCAO) was used to induce an in vivo animal model. Our findings indicated that scutellarin promoted the recovery of cerebral blood flow in MCAO rats at 3 days, significantly different from that in the MCAO group. Western blotting and immunofluorescence revealed that scutellarin treatment of BV-2 microglial cells resulted in a significant reduction in the protein expression levels and incidence of cells immunopositive for p-NF-κB, TNF-α, IL-1ß, Bax, and C-caspase-3. In contrast, the expression levels of p-PI3K, p-AKT, p-GSK3ß, and Bcl-2 were further increased, significantly different from those in the LPS group. The PI3K inhibitor LY294002 had similar effects to scutellarin by inhibiting neuroinflammation and apoptosis in activated microglia. The results of the PI3K/AKT/GSK3ß signaling pathway and NF-κB pathway in vivo in MCAO models induced microglia at 3 days were consistent with those obtained from in vitro cells. These findings indicate that scutellarin plays a neuroprotective role by reducing microglial neuroinflammation and apoptosis mediated by the activated PI3K/AKT/GSK3ß/NF-κB signaling pathway.

3.
Adv Biol (Weinh) ; 8(7): e2400123, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38684459

RESUMO

Scutellarin is an herbal agent which can exert anti-neuroinflammatory effects in activated microglia. However, it remains uncertain if it can inhibit microglia-mediated neuroinflammation by regulating miRNAs. This study sought to elucidate the upstream regulatory mechanisms by endogenous microRNAs and its target gene in activated microglia in lipopolysaccharide (LPS)-induced BV-2 microglia. Results show that scutellarin suppressed the expression of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and inducible nitric oxide synthase (iNOS) significantly in LPS-stimulated BV-2 microglia. As with the results of miRNAs function classification in vitro, the expression levels of mir-7036a-5p are upregulated in LPS-activated BV-2 microglia, but are downregulated by scutellarin. Rescue experiments indicated that mir-7036a-5p is a pro-inflammatory factor in activated BV-2 microglia. mir-7036a-5p agomir promoted the expression of phosphorylated tau proteins (p-tau), protein kinase C gamma type (PRKCG), extracellular regulated protein kinases (ERK1/2), but the is reversed by mir-7036a-5p antagomir in vitro. It is shown here that mir-7036a-5p is involved in microglia-mediated inflammation in LPS-induced BV-2 microglia. More important is the novel finding that scutellarin mitigated microglia inflammation by down-regulating the mir-7036a-5p/MAPT/PRKCG/ERK signaling pathway.


Assuntos
Apigenina , Glucuronatos , Lipopolissacarídeos , MicroRNAs , Microglia , Apigenina/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Glucuronatos/farmacologia , Lipopolissacarídeos/farmacologia , MicroRNAs/metabolismo , MicroRNAs/genética , Animais , Camundongos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Linhagem Celular , Proteína Quinase C/metabolismo
4.
FASEB J ; 38(2): e23387, 2024 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-38193649

RESUMO

Human brain microvascular endothelial cells (HBMVECs) and microglia play critical roles in regulating cerebral homeostasis during ischemic stroke. However, the role of HBMVECs-derived exosomes in microglia polarization after stroke remains unknown. We isolated exosomes (Exos) from oxygen glucose deprivation (OGD)-exposed HBMVECs, before added them into microglia. Microglia polarization markers were tested using RT-qPCR or flow cytometry. Inflammatory cytokines were measured with ELISA. Endothelial cell damage was assessed by cell viability, apoptosis, apoptosis-related proteins, oxidative stress, and angiogenic activity using CCK-8, flow cytometry, western blot, ELISA, and endothelial tube formation assay, respectively. We also established middle cerebral artery occlusion (MCAO) mice model to examine the function of circ_0000495 on stroke in vivo. Our study found that HBMVECs-Exos reduced M2 markers (IL-10, CD163, and CD206), increased M1 markers (TNF-α, IL-1ß, and IL-12), CD86-positive cells, and inflammatory cytokines (TNF-α and IL-1ß), indicating the promotion of microglial M1-polarization. Microglial M1-polarization induced by HBMVECs-Exos reduced viability and promoted apoptosis and oxidative stress, revealing the aggravation of endothelial cell damage. However, circ_0000495 silencing inhibited HBMVECs-Exos-induced alterations. Mechanistically, circ_0000495 adsorbed miR-579-3p to upregulate toll-like receptor 4 (TLR4) in microglia; miR-579-3p suppressed HBMVECs-Exos-induced alterations via declining TLR4; furthermore, Yin Yang 1 (YY1) transcriptionally activated circ_0000495 in HBMVECs. Importantly, circ_0000495 aggravated ischemic brain injury in vivo via activating TLR4/nuclear factor-κB (NF-κB) pathway. Collectively, OGD-treated HBMVECs-Exos transmitted circ_0000495 to regulate miR-579-3p/TLR4/NF-κB axis in microglia, thereby facilitating microglial M1-polarization and endothelial cell damage.


Assuntos
Exossomos , MicroRNAs , Acidente Vascular Cerebral , Animais , Camundongos , Humanos , Células Endoteliais , Microglia , Receptor 4 Toll-Like/genética , NF-kappa B , Fator de Necrose Tumoral alfa , Encéfalo , Hipóxia , Oxigênio , Citocinas , MicroRNAs/genética
7.
Biomed Pharmacother ; 126: 109786, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32113052

RESUMO

Ischemic stroke is a serious threat to human life and health, which is often accompanied by cerebral ischemia-reperfusion (I/R) injury in clinic. Ischemic postconditioning (IPostC) is a short period of mild non-fatal ischemia in the early stage of cerebral I/R injury. However, there are few reports about the protective effect of IPostC. In the present study, we investigated the neuroprotective effect of IPostC in a mice model of ischemia induced by the middle cerebral artery occlusion (MCAO). MicroRNA-124(miR-124) is a small RNA highly expressed in the brain. Several studies have shown that miR-124 is significantly decreased in IPostC. Therefore, we hypothesize that IPostC may play an important role by downregulating the expression of miR-124. Mice were treated with cerebral I/R and IPostC treatment on the basis of MCAO. The results showed that IPostC significantly reduced neurobehavioral deficits and decreased brain infarct volume. Moreover, we also found that inhibiting miR-124 effectively reduced neurons/cells apoptosis in vivo and vitro. In addition, western blot analysis of apoptosis-related proteins and PI3K/Akt2 signaling pathway proteins showed that downregulation of miR-124 significantly decreased the expression of Caspase-3 and BAX, and increased the expression of anti-apoptotic protein Bcl-2. Inhibition of miR-124 also increase PI3K/Akt/mTOR signaling pathway, thus inhibiting cell apoptosis and autophagy. However, overexpression of miR-124 weakens the protective effect of IPostC. These observations suggest that IPostC exerts its neuroprotective effect through negatively regulating PI3K/Akt2 signaling pathway by miR-124.


Assuntos
Infarto Cerebral/patologia , Pós-Condicionamento Isquêmico , MicroRNAs/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose , Regulação para Baixo , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/agonistas , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Neurônios , Células PC12 , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Transdução de Sinais
8.
Neuromolecular Med ; 22(2): 264-277, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31792810

RESUMO

BACKGROUND: Scutellarin, an herbal compound, can effectively suppress the inflammatory response in activated microglia/brain macrophage(AM/BM) in experimentally induced cerebral ischemia; however, the underlying mechanism for this has not been fully clarified. We sought to elucidate if scutellarin would exert its anti-inflammatory effects on AM/BM through the MAPKs pathway. MATERIALS AND METHODS: Western blot and immunofluorescence labeling were used to determine the expression of the MAPKs pathway in AM/BM in rats subjected to middle cerebral artery occlusion (MCAO) also in lipopolysaccharide (LPS)-activated BV-2 microglia in vitro. Furthermore, expression of p-p38 along with that of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta(IL-1ß), and inducible nitric oxide synthase (iNOS) in LPS-activated microglia subjected to pretreatment with p38 inhibitor SB203580, p38 activator sc-201214, scutellarin, or a combination of them was evaluated. FINDINGS: Scutellarin markedly attenuated the expression of p-p38, p-JNK in AM/BM in MCAO rats and in vitro. Conversely, p-ERK1/2 expression level was significantly increased by scutellarin. Meanwhile, scutellarin suppressed the expression of proinflammatory mediators including iNOS, TNF-α, and IL-1ß in AM/BM. More importantly, SB203580 suppressed p-p38 protein expression level in LPS-activated BV-2 microglia that was coupled with decreased expression of proinflammatory mediators (TNF-α, iNOS) in LPS-activated BV-2 microglia. However, p38 activator sc-201214 increased expression of proinflammatory mediators TNF-α, iNOS, and IL-1ß. Interestingly, the decreased expression of both proinflammatory markers by p38 MAPK inhibitor and increased expression of proinflammatory markers by p38 MAPK activator were compatible with that in BV-2-activated microglia pretreated with scutellarin. CONCLUSIONS: The results suggest that scutellarin down-regulates the expression of proinflammatory mediators in AM/BM through suppressing the p-JNK and p-p38 MAPKs. Of note, the anti-inflammatory effect of p38 MAPK inhibitor and scutellarin is comparable. Besides, p38 MAPKs activator reverses the effect of scutellarin. Additionally, scutellarin increases p-ERK1/2 expression that may be neuroprotective.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Apigenina/farmacologia , Glucuronatos/farmacologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Apigenina/uso terapêutico , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucuronatos/uso terapêutico , Imidazóis/farmacologia , Infarto da Artéria Cerebral Média/patologia , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Proteínas Quinases/biossíntese , Proteínas Quinases/genética , Piridinas/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
9.
Histol Histopathol ; 34(9): 965-983, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30900737

RESUMO

Cerebral ischemia affects many especially with the ageing population. The ensuing ischemic reactions include oxidative stress, inflammation, and excitotoxicity among others. In the search for effective therapeutic strategies for cerebral ischemia, activated microglia which are the key player in neuroinflammation are now recognized as a potential therapeutic target. Microglia possess both neurotoxic and neuroprotective roles. They are protective by continuously surveilling the microenvironment, phagocytosing dead cells, secreting trophic factors and sculpting the neuronal connections by removing axons and pruning excess synapses. On the other hand, hyperactivated microglia may impair cerebral oxidative metabolism, and produce excessive proinflammatory mediators that may exacerbate the brain damage. In view of this, suppression of microglial activation has been considered a therapeutic strategy to mitigate microglia-based neuroinflammation in cerebral ischemia. However, balancing the neuroprotective and neurotoxic roles of activated microglia remains a challenging issue. Many traditional Chinese herbal agents have been used in clinic for treatment of cerebral ischemia. Here, we provide an overview of five common Chinese herbs targeting specifically microglia-mediated neuroinflammation in cerebral ischemia. It is hoped that a common parallel may be drawn from their beneficial effects especially in the latter pathological conditions for their better and effective use in the future.


Assuntos
Isquemia Encefálica/patologia , Medicamentos de Ervas Chinesas/farmacologia , Microglia/efeitos dos fármacos , Degeneração Neural/patologia , Fármacos Neuroprotetores/farmacologia , Animais , Humanos
10.
J Cell Biochem ; 120(6): 9768-9780, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30582210

RESUMO

OBJECTIVE: Osteoarthritis (OA) is a progressive and disabling disorder, characterized by synovial inflammation and joint effusion. This study aimed to explore the role of stanniocalcin-1 (STC1) in the development of OA by regulating the survival and inflammation of fibroblast-like synovial (FLS) cells. METHODS: Microarray analyses were adopted to screen differentially expressed genes (DEGs) related to OA, and regulatory microRNA (miR) was also identified. Synovial tissue samples from patients with OA and healthy individuals were obtained to determine the expression levels of miR-454, STC1, IL-6, IL-8, and MMP3/13. The targeted relationship between miR-454 and STC1 was verified by dual-luciferase reporter gene assay. With the treatment of miR-454 mimic and STC1 overexpression vector, the effect of miR-454 and STC1 on FLS cell viability and apoptosis as well as production of inflammatory cytokines were tested. RESULTS: STC1 with aberrant low expression was screened from GSE1919 profile in OA. STC1 was found to be downregulated in OA-FLS tissues and cells. STC1 overexpression inhibited OA-FLS cell viability but induced apoptosis of OA-FLS cells. Moreover, STC1 overexpression decreased levels of IL-6, IL-8, and MMP3/13, suggesting that STC1 overexpression suppressed inflammatory reactions. In addition, miR-454 blocked the inhibitory effects of STC1 overexpression on OA-FLS cell viability and inflammatory reaction and exerted a promotion effect of STC1 overexpression on apoptosis of OA-FLS cells. CONCLUSIONS: Taken together, the results revealed that upregulation of STC1 could repress proliferation of OA-FLS cells and inflammatory reaction, and enhance apoptosis of OA-FLS cells, which was negatively regulated by miR-454.


Assuntos
Fibroblastos/metabolismo , Glicoproteínas/biossíntese , Osteoartrite/metabolismo , Membrana Sinovial/metabolismo , Regulação para Cima , Adulto , Sobrevivência Celular , Feminino , Fibroblastos/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Masculino , Osteoartrite/patologia , Membrana Sinovial/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...