Turn off MathJax
Article Contents
Junping Zhu, Ye Lin, Gejing Li, Yini He, Zhaoli Su, Yuanyuan Tang, Ye Zhang, Qian Xu, Zhongliu Yao, Hua Zhou, Bin Liu, Xiong Cai. Dual-targeted halofuginone hydrobromide nanocomplexes for promotion of macrophage repolarization and apoptosis of rheumatoid arthritis fibroblast-like synoviocytes in adjuvant-induced arthritis in rats[J]. Journal of Pharmaceutical Analysis. doi: 10.1016/j.jpha.2024.100981
Citation: Junping Zhu, Ye Lin, Gejing Li, Yini He, Zhaoli Su, Yuanyuan Tang, Ye Zhang, Qian Xu, Zhongliu Yao, Hua Zhou, Bin Liu, Xiong Cai. Dual-targeted halofuginone hydrobromide nanocomplexes for promotion of macrophage repolarization and apoptosis of rheumatoid arthritis fibroblast-like synoviocytes in adjuvant-induced arthritis in rats[J]. Journal of Pharmaceutical Analysis. doi: 10.1016/j.jpha.2024.100981

Dual-targeted halofuginone hydrobromide nanocomplexes for promotion of macrophage repolarization and apoptosis of rheumatoid arthritis fibroblast-like synoviocytes in adjuvant-induced arthritis in rats

doi: 10.1016/j.jpha.2024.100981
Funds:

This work was funded by grants from the National Natural Science Foundation of China (Grant No.:82274506), the China Postdoctoral Science Foundation (Grant No.:2022M721128), the Science and Technology Innovation Program of Hunan, China (Grant No.:2021RC4035), the Natural Science Foundation of Hunan, China (Grant No.:2023JJ40477), and the Open-competing Disciple Construction Project of Hunan University of Chinese Medicine (HNUCM), China (Grant No.:22JBZ003), and was financially supported by the Furong Distinguished Scholar Program of Hunan, China (Program No.:XJT[2020]58), the 121 Training Project for Innovative Talents of Hunan, China (Project No.:XRSH[2019]192), the Chinese Academy of Engineering Academician Liang Liu's Workstation Project, China (Project No.:KH[2023]3-23YS001), and the World First-class Discipline Incubation Project of HNUCM, China (Project No.:XJF[2022]57). We acknowledged the technical support from the Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan, School of Pharmacy, HNUCM, China.

  • Received Date: Jan. 09, 2024
  • Accepted Date: Apr. 18, 2024
  • Rev Recd Date: Apr. 16, 2024
  • Available Online: Apr. 23, 2024
  • Rheumatoid arthritis (RA) is a prevalent autoimmune disease characterized by chronic inflammation and excessive proliferation of the synovium. Currently, treatment options focus on either reducing inflammation or inhibiting synovial hyperplasia. However, these modalities are unsatisfactory in achieving the desired therapeutic outcomes. Halofuginone hydrobromide (HF), an herbal active ingredient, has demonstrated pharmacological effects of both anti-inflammation and inhibition of synovial hyperplasia proliferation. However, HF's medical efficacy is limited due to its poor water solubility, short half-life, and non-target toxicity. In the current study, by using the advantages of nanotechnology, we presented a novel dual-targeted nanocomplex, termed HA-M@P@HF NPs, which consisted of a hyaluronic acid (HA)-modified hybrid membrane (M)-camouflaged poly lactic-co-glycolic acid (PLGA) nanosystem for HF delivery. These nanocomplexes not only overcame the limitations of HF but also achieved simultaneous targeting of inflammatory macrophages and human fibroblast-like synoviocytes-rheumatoid arthritis (HFLS-RA). In vivo experiments demonstrated that these nanocomplexes effectively suppressed immune-mediated inflammation and synovial hyperplasia, safeguarding against bone destruction in rats with adjuvant-induced arthritis (AIA). Remarkable anti-arthritic effects of these nanocomplexes were accomplished through promoting repolarization of M1-to-M2 macrophages and apoptosis of HFLS-RA, thereby offering a promising therapeutic strategy for RA.
  • loading
  • [1]
    A. Di Matteo, J.M. Bathon, P. Emery, Rheumatoid arthritis, Lancet. 402(2023)2019-2033.
    [2]
    Y. Ma, Z. Lu, B. Jia, et al., DNA origami as a nanomedicine for targeted rheumatoid arthritis therapy through reactive oxygen species and nitric oxide scavenging, ACS Nano. 16(2022)12520-12531.
    [3]
    E.M.L. Philippon, L.J.E. van Rooijen, F. Khodadust, et al., A novel 3D spheroid model of rheumatoid arthritis synovial tissue incorporating fibroblasts, endothelial cells, and macrophages, Front. Immunol. 14(2023), 1188835.
    [4]
    J. Tu, X. Wang, X. Gong, et al., Synovial macrophages in rheumatoid arthritis:The past, present, and future, Mediators Inflamm. 2020(2020), 1583647.
    [5]
    J. Zhu, J. Wei, Y. Lin, et al., Inhibition of IL-17 signaling in macrophages underlies the anti-arthritic effects of halofuginone hydrobromide:Network pharmacology, molecular docking, and experimental validation, BMC Complementary Med. Ther. 24(2024), 105.
    [6]
    J. Smolen, R.B. Landewé, P. Mease, et al., Efficacy and safety of certolizumab pegol plus methotrexate in active rheumatoid arthritis:The RAPID 2 study. A randomised controlled trial, Ann. Rheum. Dis. 68(2009)797-804.
    [7]
    E. Berthet, M.De Rosa, P. Vorilhon, et al., AB1073 Exploration of the difficulties encountered by general practitioners with methotrexate and tnf-inhibitor treatment in common practice, Ann. Rheum. Dis. 75(2016), 1268.
    [8]
    R.A. Moura, J.E. Fonseca, JAK inhibitors and modulation of B cell Immune responses in rheumatoid arthritis, Front. Med. 7(2021), 607725.
    [9]
    M.K. Park, J.S. Park, E.M. Park, et al., Halofuginone ameliorates autoimmune arthritis in mice by regulating the balance between Th17 and Treg cells and inhibiting osteoclastogenesis, Arthritis Rheumatol. 66(2014)1195-1207.
    [10]
    S. Zeng, K. Wang, M. Huang, et al., Halofuginone inhibits TNF-α-induced the migration and proliferation of fibroblast-like synoviocytes from rheumatoid arthritis patients, Int. Immunopharmacol. 43(2017)187-194.
    [11]
    J. Gill, A. Sharma, Prospects of halofuginone as an antiprotozoal drug scaffold, Drug Discov. Today. 27(2022)2586-2592.
    [12]
    K. Park, S. Skidmore, J. Hadar, et al., Injectable, long-acting PLGA formulations:Analyzing PLGA and understanding microparticle formation, J. Control. Release. 304(2019)125-134.
    [13]
    J. Fan, Y. Qin, C. Xiao, et al., Biomimetic PLGA-based nanocomplexes for improved tumor penetration to enhance chemo-photodynamic therapy against metastasis of TNBC, Mater. Today Adv. 16(2022), 100289.
    [14]
    J. Lu, X. Gao, S. Wang, et al., Advanced strategies to evade the mononuclear phagocyte system clearance of nanomaterials, Explor. Beijing China. 3(2023), 20220045.
    [15]
    Z. Lei, J. Fan, X. Li, et al., Biomimetic graphene oxide quantum dots nanoparticles targeted photothermal-chemotherapy for gastric cancer, J. Drug Target. 31(2023)320-333.
    [16]
    P. You, A. Mayier, H. Zhou, et al., Targeting and promoting atherosclerosis regression using hybrid membrane coated nanomaterials via alleviated inflammation and enhanced autophagy, Appl. Mater. Today. 26(2022), 101386.
    [17]
    Y. Lin, O. Yi, M. Hu, et al., Multifunctional nanoparticles of sinomenine hydrochloride for treat-to-target therapy of rheumatoid arthritis via modulation of proinflammatory cytokines, J. Control. Release. 348(2022)42-56.
    [18]
    M. Ge, C. Du, Preparation process of shear thickening gel based on constrained uniform mixture design and orthogonal experimental design, Polym. Test. 129(2023), 108267.
    [19]
    B. Liu, W. Wang, J. Fan, et al., RBC membrane camouflaged prussian blue nanoparticles for gamabutolin loading and combined chemo/photothermal therapy of breast cancer, Biomaterials. 217(2019), 119301.
    [20]
    C. He, W. Quan, Y. Zeng, et al., Construction of nicotinic acid curcumin nanoparticles and its Anti-atherosclerosis effect via PCSK9/LDL-R, ABCA1/Caveolin-1/LXR pathway, Mater. Des. 229(2023), 111931.
    [21]
    L. Concha, A.L. Resende Pires, A.M. Moraes, et al., Cost function analysis applied to different kinetic release models of Arrabidaea chica verlot extract from chitosan/alginate membranes, Polymers. 14(2022), 1109.
    [22]
    P. You, A. Yang, Y. Sun, et al., Pro-efferocytosis biomimetic nanocomplexes for targeted atherosclerosis therapy through promoting macrophage re-polarization and inhibiting senescence, Mater. Des. 234(2023), 112316.
    [23]
    S. Hu, Y. Lin, C. Tong, et al., A pH-Driven indomethacin-loaded nanomedicine for effective rheumatoid arthritis therapy by combining with photothermal therapy, J. Drug Target. 30(2022)737-752.
    [24]
    S. Alivernini, L. MacDonald, A. Elmesmari, et al., Distinct synovial tissue macrophage subsets regulate inflammation and remission in rheumatoid arthritis, Nat. Med. 26(2020)1295-1306.
    [25]
    S. Tardito, G. Martinelli, S. Soldano, et al., Macrophage M1/M2 polarization and rheumatoid arthritis:A systematic review, Autoimmun. Rev. 18(2019), 102397.
    [26]
    M. Hu, Z. Yao, L. Xu, et al., M2 macrophage polarization in systemic sclerosis fibrosis:Pathogenic mechanisms and therapeutic effects, Heliyon. 9(2023), e16206.
    [27]
    H. Li, Y. Feng, X. Zheng, et al., M2-type exosomes nanoparticles for rheumatoid arthritis therapy via macrophage re-polarization, J. Control. Release. 341(2022)16-30.
    [28]
    M. Noack, P. Miossec, Selected cytokine pathways in rheumatoid arthritis, Semin. Immunopathol. 39(2017)365-383.
    [29]
    T. Huang, N. Mu, J. Gu, et al., DDR2-CYR61-MMP1 signaling pathway promotes bone erosion in rheumatoid arthritis through regulating migration and invasion of fibroblast-like synoviocytes, J. Bone Miner. Res. 34(2019)779-780.
    [30]
    P.D. Katsikis, C.Q. Chu, F.M. Brennan, et al., Immunoregulatory role of interleukin 10 in rheumatoid arthritis, J. Exp. Med. 179(1994)1517-1527.
    [31]
    H. Matsumoto, Y. Fujita, T. Asano, et al., Association between inflammatory cytokines and immune-checkpoint molecule in rheumatoid arthritis, PLoS One. 16(2021), e0260254.
    [32]
    K.S. Nandakumar, Q. Fang, I. Wingbro Ågren, et al., Aberrant activation of immune and non-immune cells contributes to joint inflammation and bone degradation in rheumatoid arthritis, Int. J. Mol. Sci. 24(2023), 15883.
    [33]
    J.G. Navashenaq, A.G. Shabgah, M. Hedayati-Moghadam, et al., The role of myeloid-derived suppressor cells in rheumatoid arthritis:An update, Life Sci. 269(2021), 119083.
    [34]
    H.J. Kim, B. Ohk, H.J. Yoon, et al., Docosahexaenoic acid signaling attenuates the proliferation and differentiation of bone marrow-derived osteoclast precursors and promotes apoptosis in mature osteoclasts, Cell. Signal. 29(2017)226-232.
    [35]
    I.P. Perpétuo, J. Caetano-Lopes, A.M. Rodrigues, et al., Effect of tumor necrosis factor inhibitor therapy on osteoclasts precursors in rheumatoid arthritis, BioMed Res. Int. 2017(2017), 2690402.
    [36]
    K.A. Lee, K.W. Kim, B.M. Kim, et al., Promotion of osteoclastogenesis by IL-26 in rheumatoid arthritis, Arthritis Res. Ther. 21(2019), 283.
    [37]
    J.S. Smolen, R.B.M. Landewé, J.W.J. Bijlsma, et al., EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs:2019 update, Ann. Rheum. Dis. 79(2020)685-699.
    [38]
    X. Zhang, Y. Liu, W. Liu, et al., Macrophage-hitchhiking interleukin-10 plasmid DNA delivery system modulates rheumatoid arthritis microenvironment via the re-polarization of macrophages, Nano Today. 54(2024), 102068.
    [39]
    S. Li, J. Li, Y. Zhao, et al., Supramolecular integration of multifunctional nanomaterial by mannose-decorated azocalixarene with ginsenoside Rb1 for synergistic therapy of rheumatoid arthritis, ACS Nano. 17(2023)25468-25482.
    [40]
    Z. Tang, S. Meng, X. Yang, et al., Neutrophil-mimetic, ROS responsive, and oxygen generating nanovesicles for targeted interventions of refractory rheumatoid arthritis, Small.(2023), e2307379.
    [41]
    C. Tao, F. Li, Z. Ma, et al., Highly efficient oral iguratimod/polyvinyl alcohol nanodrugs fabricated by high-gravity nanoprecipitation technique for treatment of rheumatoid arthritis, Small.(2023), e2304150.
    [42]
    D. Kim, J. Park, Y. Kang, et al., Drug repositioning of metformin encapsulated in PLGA combined with photothermal therapy ameliorates rheumatoid arthritis, Int. J. Nanomedicine. 18(2023)7267-7285.
    [43]
    T. Puidokas, M. Kubilius, A. Stumbras, et al., Effect of leukocytes included in platelet concentrates on cell behaviour, Platelets. 30(2019)937-945.
    [44]
    A. Nakashima, K. Suzuki, H. Fujii, et al., pos0527 acute kidney injury (aki) in patients with rheumatoid arthritis (RA), Ann. Rheum. Dis. 80(2021), 497.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(1)

    Article Metrics

    Article views (24) PDF downloads(1) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return