Rutaecarpine Attenuates Monosodium Urate Crystal-Induced Gouty Inflammation via Inhibition of TNFR-MAPK/NF-κB and NLRP3 Inflammasome Signaling Pathways*
Original Article|Updated:2025-07-11
|
Rutaecarpine Attenuates Monosodium Urate Crystal-Induced Gouty Inflammation via Inhibition of TNFR-MAPK/NF-κB and NLRP3 Inflammasome Signaling Pathways*
Chinese Journal of Integrative MedicineVol. 31, Issue 7, Pages: 590-599(2025)
Affiliations:
1.Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu (610075), China
2.Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu (610000), China
3.The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha (410219), China
LI Min, YIN Zhu-jun, LI Li, et al. Rutaecarpine Attenuates Monosodium Urate Crystal-Induced Gouty Inflammation via Inhibition of TNFR-MAPK/NF-κB and NLRP3 Inflammasome Signaling Pathways*[J]. Chinese journal of integrative medicine, 2025, 31(7): 590-599.
DOI:
LI Min, YIN Zhu-jun, LI Li, et al. Rutaecarpine Attenuates Monosodium Urate Crystal-Induced Gouty Inflammation via Inhibition of TNFR-MAPK/NF-κB and NLRP3 Inflammasome Signaling Pathways*[J]. Chinese journal of integrative medicine, 2025, 31(7): 590-599. DOI: 10.1007/s11655-025-4204-3.
Rutaecarpine Attenuates Monosodium Urate Crystal-Induced Gouty Inflammation via Inhibition of TNFR-MAPK/NF-κB and NLRP3 Inflammasome Signaling Pathways*
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
.
Methods:
2
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.
Results:
2
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.
Conclusion:
2
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.
Advance on Chinese Medicine for Hypertensive Renal Damage:Focus on the Complex Molecular Mechanisms
Protective Effect of Silibinin on Lipopolysaccharide-Induced Endotoxemia by Inhibiting Caspase-11-Dependent Cell Pyroptosis
From Perspective of Hippocampal Plasticity: Function of Antidepressant Chinese Medicine Xiaoyaosan
Advancements of Macrophages Involvement in Pathological Progression of Colitis-Associated Colorectal Cancer and Associated Pharmacological Interventions
Effect of Guanxin Danshen Dripping Pills on Coronary Heart Disease Comorbid with Depression or Anxiety: The ADECODE-Real World Study
Related Author
LU Yan
XIE Xue-na
XIN Qi-qi
YUAN Rong
MIAO Yu
CONG Wei-hong
CHEN Ke-ji
OU Jin-ying
Related Institution
National Clinical Research Center for Chinese Medicine Cardiology
School of Pharmacy, Macau University of Science and Technology, Taipa
Laboratory of Cardiovascular Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences
School of Traditional Chinese Medicine, Southern Medical University
Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics