Li Min, Yin Zhu-Jun, Li Li, Quan Yun-Yun, Wang Ting, Zhu Xin, Tan Rui-Rong, Zeng Jin, Hua Hua, Wu Qin-Xuan, Zhao Jun-Ning
Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China.
Chin J Integr Med. 2025 May 8. doi: 10.1007/s11655-025-4204-3.
To investigate the anti-inflammatory effect of rutaecarpine (RUT) on monosodium urate crystal (MSU)-induced murine peritonitis in mice and further explored the underlying mechanism of RUT in lipopolysaccharide (LPS)/MSU-induced gout model in vitro.
In MSU-induced mice, 36 male C57BL/6 mice were randomly divided into 6 groups of 8 mice each group, including the control group, model group, RUT low-, medium-, and high-doses groups, and prednisone acetate group. The mice in each group were orally administered the corresponding drugs or vehicle once a day for 7 consecutive days. The gout inflammation model was established by intraperitoneal injection of MSU to evaluate the anti-gout inflammatory effects of RUT. Then the proinflammatory cytokines were measured by enzyme-linked immunosorbent assay (ELISA) and the proportions of infiltrating neutrophils cytokines were detected by flow cytometry. In LPS/MSU-treated or untreated THP-1 macrophages, cell viability was observed by cell counting kit 8 and proinflammatory cytokines were measured by ELISA. The percentage of pyroptotic cells were detected by flow cytometry. Respectively, the mRNA and protein levels were measured by real-time quantitative polymerase chain reaction (qRT-PCR) and Western blot, the nuclear translocation of nuclear factor κB (NF-κB) p65 was observed by laser confocal imaging. Additionally, surface plasmon resonance (SPR) and molecular docking were applied to validate the binding ability of RUT components to tumor necrosis factor α (TNF-α) targets.
RUT reduced the levels of infiltrating neutrophils and monocytes and decreased the levels of the proinflammatory cytokines interleukin 1β (IL-1β) and interleukin 6 (IL-6, all P<0.01). In vitro, RUT reduced the production of IL-1β, IL-6 and TNF-α. In addition, RT-PCR revealed the inhibitory effects of RUT on the mRNA levels of IL-1β, IL-6, cyclooxygenase-2 and TNF-α (P<0.05 or P<0.01). Mechanistically, RUT markedly reduced protein expressions of tumor necrosis factor receptor (TNFR), phospho-mitogen-activated protein kinase (p-MAPK), phospho-extracellular signal-regulated kinase, phospho-c-Jun N-terminal kinase, phospho-NF-κB, phospho-kinase α/β, NOD-like receptor thermal protein domain associated protein 3 (NLRPS), cleaved-cysteinyl aspartate specific proteinase-1 and cleaved-gasdermin D in macrophages (P<0.05 or P<0.01). Molecularly, SPR revealed that RUT bound to TNF-α with a calculated equilibrium dissociation constant of 31.7 µmol/L. Molecular docking further confirmed that RUT could interact directly with the TNF-α protein via hydrogen bonding, van der Waals interactions, and carbon-hydrogen bonding.
RUT alleviated MSU-induced peritonitis and inhibited the TNFR1-MAPK/NF-κB and NLRP3 inflammasome signaling pathway to attenuate gouty inflammation induced by LPS/MSU in THP-1 macrophages, suggesting that RUT could be a potential therapeutic candidate for gout.
研究吴茱萸次碱(RUT)对尿酸钠晶体(MSU)诱导的小鼠腹膜炎的抗炎作用,并进一步探讨RUT在体外脂多糖(LPS)/MSU诱导的痛风模型中的潜在作用机制。
在MSU诱导的小鼠中,将36只雄性C57BL/6小鼠随机分为6组,每组8只,包括对照组、模型组、RUT低、中、高剂量组和醋酸泼尼松组。每组小鼠每天口服相应药物或赋形剂1次,连续7天。通过腹腔注射MSU建立痛风炎症模型,以评估RUT的抗痛风炎症作用。然后通过酶联免疫吸附测定(ELISA)检测促炎细胞因子,并通过流式细胞术检测浸润中性粒细胞细胞因子的比例。在LPS/MSU处理或未处理的THP-1巨噬细胞中,通过细胞计数试剂盒8观察细胞活力,并通过ELISA检测促炎细胞因子。通过流式细胞术检测焦亡细胞的百分比。分别通过实时定量聚合酶链反应(qRT-PCR)和蛋白质印迹法检测mRNA和蛋白质水平,通过激光共聚焦成像观察核因子κB(NF-κB)p65的核转位。此外,应用表面等离子体共振(SPR)和分子对接来验证RUT成分与肿瘤坏死因子α(TNF-α)靶点的结合能力。
RUT降低了浸润中性粒细胞和单核细胞的水平,并降低了促炎细胞因子白细胞介素1β(IL-1β)和白细胞介素6(IL-6)的水平(均P<0.01)。在体外,RUT降低了IL-1β、IL-6和TNF-α的产生。此外,RT-PCR显示RUT对IL-1β、IL-6、环氧合酶-2和TNF-α的mRNA水平具有抑制作用(P<0.05或P<0.01)。机制上,RUT显著降低了巨噬细胞中肿瘤坏死因子受体(TNFR)、磷酸化丝裂原活化蛋白激酶(p-MAPK)、磷酸化细胞外信号调节激酶、磷酸化c-Jun氨基末端激酶、磷酸化NF-κB、磷酸化激酶α/β、NOD样受体热蛋白结构域相关蛋白3(NLRPS)、裂解的半胱天冬酶-1和裂解的gasdermin D的蛋白表达(P<0.05或P<0.01)。分子水平上,SPR显示RUT与TNF-α结合,计算得到的平衡解离常数为31.7 μmol/L。分子对接进一步证实RUT可通过氢键、范德华相互作用和碳氢键与TNF-α蛋白直接相互作用。
RUT减轻了MSU诱导的腹膜炎,并抑制了TNFR1-MAPK/NF-κB和NLRP3炎性小体信号通路,以减轻LPS/MSU在THP-1巨噬细胞中诱导的痛风性炎症,表明RUT可能是痛风的潜在治疗候选药物。