Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Adicionar filtros








Intervalo de ano
1.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 30-41, 2023.
Artigo em Chinês | WPRIM | ID: wpr-998160

RESUMO

ObjectiveTo investigate the analgesic effect and mechanism of Osteoking (OK) on nerve compression in lumbar disc herniation. MethodThe rat model of chronic compression of dorsal root ganglion (CCD) was established to simulate clinical lumbar disc herniation. The CCD rats were randomly divided into model group, low, medium, and high dose OK groups (1.31, 2.63, 5.25 mL·kg-1·d-1), and pregabalin group (5 mg·kg-1), with eight rats in each group. Another eight SD rats were taken as the blank group, and the same volume of normal saline was given by gavage. Behavioral tests, side effect evaluation, network analysis, Western blot, immunofluorescence, and antagonist application were used to explore the effect. ResultCompared with the blank group, the mechanical hyperalgesia threshold, thermal hyperalgesia threshold, and the expression of inflammatory factors in the spinal dorsal horn of the model group are significantly increased (P<0.01), and the related indicators of the affected foot footprints are significantly down-regulated (P<0.01). The expression of signal transducer and activator of transcription 3 (STAT3), vascular endothelial growth factor A (VEGFA), and phosphorylated extracellular regulated protein kinase (p-ERK) in microglia in the spinal dorsal horn is significantly increased in the model group (P<0.01). Compared with the model group, low, medium, and high dose OK groups can increase the mechanical hyperalgesia and thermal hyperalgesia thresholds of CCD rats (P<0.05, P<0.01) in a dose-dependent manner, improve the gait of CCD rats (P<0.05, P<0.01), and reduce the expression of inflammatory factors in the spinal dorsal horn (P<0.05, P<0.01). The expression of STAT3, VEGFA, and p-ERK in the spinal dorsal horn microglia of CCD rats is significantly decreased (P<0.05, P<0.01), and the acetic acid-induced nociceptive response in rats is effectively reduced (P<0.05, P<0.01). In addition, there is no tolerance. The results of the body mass test, organ index, forced swimming, and rotation show that OK has no obvious toxic or side effects. Further antagonist experiments show that MRS1523 and RS127445 can reverse the transient analgesic effect of OK compared with the high dose OK group (P<0.01). ConclusionOK has a good analgesic effect on the CCD model without obvious toxic side effects, and its mechanism may be related to the activation of ADORA3 and HTR2B and the inhibition of STAT3, VEGFA, p-ERK, and other elements in microglia.

2.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 156-165, 2023.
Artigo em Chinês | WPRIM | ID: wpr-965659

RESUMO

ObjectiveTo investigate the intervention effect of Qufeng Gutong Babu ointment (QFGT) on rats with osteoarthritis (OA) with cold-dampness obstruction, and preliminarily clarify its mechanism. MethodSD male rats were divided into 6 groups, namely, the blank group, model group, positive control drug Huoxue Zhitong ointment (HXZTG) group (1.26 cm2·d-1), and low, medium, and high-dose QFGT group (75, 150, 300 mg·d-1). OA model was prepared by joint cavity injection of papain and L-cysteine. On the second day of modeling, climate factors were applied to establish an animal model of combination of disease and syndrome of OA rats with cold-dampness obstruction. Standard VonFrey fiber was used to evaluate the threshold of mechanical pain. Weight bearing difference score and joint function score of both hind limbs were recorded. Hematoxylin-eosin (HE) staining and safranine fixation green staining were used to observe the pathological changes and cartilage degeneration of rat knee joint. Immunohistochemistry (IHC) was used to detect the expression of interleukin-1β (IL-1β), interleukin-8 (IL-8), tumor necrosis factor-α (TNF-α), matrix metalloproteinase-9 (MMP-9), and cathepsin K (CTSK). Western blot was used to detect the protein expression of kinase B (Akt), phosphorylated protein kinase B (p-Akt), phosphatidylinositol 3-kinase (PI3K), nuclear factor 1 (NFATc1), MMP-9, and CTSK in T cells. ResultCompared with the normal group, the model group showed significant mechanical pain sensitivity reaction after modeling (P<0.01), and the weight bearing difference of both hind limbs and joint function score were significantly increased (P<0.05, P<0.01). Compared with the model group, both the high-dose QFGT group and the HXZTG group significantly reduced the mechanical pain sensitivity, weight difference, and joint function score of rats (P<0.05, P<0.01), and the medium-dose QFGT group also improved the joint function to a certain extent, and the degeneration of the knee joint cartilage of rats was significantly reduced (P<0.05, P<0.01). QFGT and HXZTG both inhibited the protein expression of IL-1β, IL-8, TNF-α, MMP-9, CTAK, PI3K, p-Akt, Akt, and other related proteins in articular cartilage of rats with OA to a certain extent (P<0.05, P<0.01). ConclusionQFGT can inhibit the release of inflammatory factors and matrix metalloproteinases by inhibiting the PI3K/Akt signal pathway in articular articular cartilage of rats with OA with cold-dampness obstruction, thus ultimately weakening local cartilage degeneration and improving joint function.

3.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 192-200, 2022.
Artigo em Chinês | WPRIM | ID: wpr-940812

RESUMO

ObjectiveThis study aimed to predict the pharmacodynamic material basis and core targets of Bailing capsules in the treatment of chronic obstructive pulmonary disease (COPD) based on network pharmacology and molecular docking, which were further verified by cell experiments to explore the mechanism. MethodThe main active ingredients and related targets of Bailing capsules were screened in Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and SwissTargetPrediction. The main COPD targets were searched from GeneCards, DrugBank, Online Mendelian Inheritance in Man (OMIM) and Therapeutic Target Database (TTD). The protein-protein interaction (PPI) network was constructed by STRING and Cytoscape 3.6.1. Gene Ontology (GO) function annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed by the Database for Annotation, Visualization and Integrated Discovery (DAVID). Molecular docking verification was carried out using AutoDock Vina. The cell viability was detected by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay, and the mRNA level of the targets was detected by real-time polymerase chain reaction (Real-time PCR). ResultA total of 11 active ingredients of Bailing capsules such as cerevisterol, 270 related drug targets, and 1 020 COPD target proteins were obtained, with 74 intersection targets. The visualization analysis of the PPI network showed that the core targets of Bailing capsules in the treatment of COPD were tumor protein P53 (TP53), catenin beta 1 (CTNNB1), tumor necrosis factor (TNF), interleukin-6 (IL-6) and insulin (INS). Further, 20 signaling pathways were screened by KEGG enrichment analysis as the main pathways for Bailing capsules to treat COPD, involving phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt), cyclic adenosine monophosphate (cAMP), forkhead box O (FoxO), TNF, and hypoxia inducible factor-1 (HIF-1) signaling pathways. Molecular docking validation demonstrated that four active ingredients had stable binding to IL-6, with the lowest energy. Bailing capsules could reduce the mRNA level of IL-6 in RAW264.7 cells induced by lipopolysaccharide (LPS) (P<0.01) compared with the control group. ConclusionThe pharmacological mechanism of Bailing capsules in the treatment of COPD might be that its main active ingredients improved the inflammatory response by acting on TP53, CTNNB1, TNF, IL-6 and other targets and regulating PI3K/Akt, cAMP and other signaling pathways, thereby ameliorating COPD symptoms. This study provided experimental basis for subsequent in-depth research, and provided a diagnosis and treatment direction for disease-related clinical treatment.

4.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 70-76, 2022.
Artigo em Chinês | WPRIM | ID: wpr-940209

RESUMO

ObjectiveTo investigate the therapeutic effect of Qingmei compound on acute gouty arthritis (AGA) in rats and preliminarily clarify its mechanism. MethodForty male SD rats were randomly divided into a blank group, a model group, a colchicine group (0.3 mg·kg-1), and low- and high-dose Qingmei compound groups (200 and 400 mg·kg-1), with eight rats in each group. The AGA model was induced by injecting 50 g·L-1 monosodium urate (MSU) into the ankle joint of the rats except those in the blank group. The ankle swelling index was measured before and 6, 24, and 48 h after modeling. The pathological changes in the joint tissues of AGA rats were observed by hematoxylin-eosin (HE) staining. The expression of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the joint tissues of rats was detected by immunohistochemistry. The protein expression of NOD-like receptor protein 3 (NLRP3) pathway and key proteins in the joint tissues of rats was detected by Western blot. ResultCompared with the blank group, the model group showed increased ankle swelling index, synovial hyperplasia, and inflammatory infiltration, and up-regulated expression of IL-1β, TNF-α, and NLRP3 proteins in the ankle joint and the ratio of Caspase-1 shear body to Caspase-1 precursor protein (Caspase-1 p20/Caspase-1) (P<0.01). Compared with the model group, the Qingmei compound groups showed reduced ankle swelling index of AGA rats, especially the low-dose Qingmei compound group (P<0.01). Meanwhile, Qingmei compound inhibited synovial hyperplasia and inflammatory infiltration (P<0.01) and reduced the levels of IL-1β, TNF-α, and NLRP3 proteins and Caspase-1 p20/Caspase-1 in joint tissues (P<0.01). ConclusionQingmei Compound can significantly alleviate the joint swelling and inflammatory infiltration of AGA, and its mechanism may be related to the inhibition of the NLRP3 signaling pathway.

5.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 70-76, 2022.
Artigo em Chinês | WPRIM | ID: wpr-940112

RESUMO

ObjectiveTo investigate the therapeutic effect of Qingmei compound on acute gouty arthritis (AGA) in rats and preliminarily clarify its mechanism. MethodForty male SD rats were randomly divided into a blank group, a model group, a colchicine group (0.3 mg·kg-1), and low- and high-dose Qingmei compound groups (200 and 400 mg·kg-1), with eight rats in each group. The AGA model was induced by injecting 50 g·L-1 monosodium urate (MSU) into the ankle joint of the rats except those in the blank group. The ankle swelling index was measured before and 6, 24, and 48 h after modeling. The pathological changes in the joint tissues of AGA rats were observed by hematoxylin-eosin (HE) staining. The expression of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the joint tissues of rats was detected by immunohistochemistry. The protein expression of NOD-like receptor protein 3 (NLRP3) pathway and key proteins in the joint tissues of rats was detected by Western blot. ResultCompared with the blank group, the model group showed increased ankle swelling index, synovial hyperplasia, and inflammatory infiltration, and up-regulated expression of IL-1β, TNF-α, and NLRP3 proteins in the ankle joint and the ratio of Caspase-1 shear body to Caspase-1 precursor protein (Caspase-1 p20/Caspase-1) (P<0.01). Compared with the model group, the Qingmei compound groups showed reduced ankle swelling index of AGA rats, especially the low-dose Qingmei compound group (P<0.01). Meanwhile, Qingmei compound inhibited synovial hyperplasia and inflammatory infiltration (P<0.01) and reduced the levels of IL-1β, TNF-α, and NLRP3 proteins and Caspase-1 p20/Caspase-1 in joint tissues (P<0.01). ConclusionQingmei Compound can significantly alleviate the joint swelling and inflammatory infiltration of AGA, and its mechanism may be related to the inhibition of the NLRP3 signaling pathway.

6.
Journal of Southern Medical University ; (12): 1121-1124, 2014.
Artigo em Chinês | WPRIM | ID: wpr-312625

RESUMO

<p><b>OBJECTIVE</b>To investigate the activation of the complement system in the retina in a mouse model of endotoxin-induced uveitis (EIU).</p><p><b>METHODS</b>Balb/c mice were randomly divided into control group and lipopolysaccharide (LPS)-induced EIU group. Twenty-four hours after modeling, the expressions of the major complement components of the classical pathway (CP), mannose-binding lectin (MBL) pathway, alternative pathway (AP), and terminal pathway in the retina were determined by quantitative RT-PCR. Western blotting was employed to examine the protein expressions of the key components of the complement system involved in the CP and AP pathways in the retina.</p><p><b>RESULTS</b>s Normal mouse retina expressed a variety of complement components that were involved mainly in the CP and AP pathways. The expressions of the complement components involved in the CP and AP pathways were up-regulated in the retina of mice with EIU. Although MASP1 and MASP2 were detected in the retina in both the EIU and control groups, their expressions were weak and showed no significant difference between the two groups.</p><p><b>CONCLUSION</b>The AP and CP but not the MBL pathways of the complement system are activated in the retina of mice with EIU, suggesting a role of the activated complement system in the pathological process of EIU.</p>


Assuntos
Animais , Camundongos , Western Blotting , Proteínas do Sistema Complemento , Modelos Animais de Doenças , Lipopolissacarídeos , Camundongos Endogâmicos BALB C , Retina , Uveíte
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA