a. College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China;
b. Innovative Research Center for Basic Medicine on Autoimmune Diseases of Ministry of Education of China, Hangzhou, 310053, China;
c. The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China;
d. Department of Traditional Chinese Medicine Gynecology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, China
Funds:
This research was funded by the National Natural Science Foundation of China (Grant No.: 82405298), China Postdoctoral Science Foundation (Grant No.: 2023M733193), Zhejiang Provincial Natural Science Foundation of China (Grant No.: LQ24H270006), Zhejiang Traditional Medicine and Technology Program, China (Grant No.: 2024ZF063), the National Key R&D Program of China (Grant No.: 2022YFC3501204), and Science and Technology Co-constructed Plan (STD-ZJ) Project, China (Project No.: GZY-ZJ-KJ-23009).
Chronic joint pain in rheumatoid arthritis (RA) represents a persistent therapeutic challenge, and although luteolin (LUT) exhibits established anti-inflammatory properties, its precise mechanism for alleviating RA-associated chronic pain remains undefined. Through systematic investigation in collagen-induced arthritis (CIA) mice, we demonstrated that LUT administration effectively attenuated chronic pain by modulating spinal cluster of differentiation 4 positive T (CD4+ T) cell dynamics and suppressing microglial activation. Integrated multi-omics profiling (cleavage under targets and tagmentation, RNA sequencing (RNA-seq), and metabolomics) coupled with functional validation revealed nuclear factor of activated T cells 2 (NFATC2) as the central transcriptional regulator governing T helper 17 (Th17) cell differentiation and spinal infiltration through protein kinase C epsilon (PRKCE)-signal transducer and activator of transcription 3 (STAT3) signaling transduction. Significantly, our mechanistic studies uncovered a previously unrecognized epigenetic cascade: LUT-mediated suppression of lactate dehydrogenase A (LDHA) activity disrupts glycolysis-fueled histone h3 lysine 9 lactylation (H3K9la), thereby epigenetically silencing NFATC2 transcription. Translational studies using RA patient-derived CD4+ T cells confirmed LUT's capacity to normalize pathological hyperactivity of the LDHA/H3K9la/NFATC2 axis, concomitantly regulating CD4+ T dynamics. Biophysical validation through molecular docking, surface plasmon resonance, and molecular dynamics simulations established LUT's direct binding to LDHA with high affinity. Collectively, these findings delineate a novel therapeutic paradigm wherein LUT alleviates RA-associated chronic pain by orchestrating Th17 differentiation and migratory capacity through coordinated blockade of the LDHA-H3K9la-NFATC2 signaling network, highlighting its potential as a disease-modifying agent for chronic pain management in RA.