3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Securidaca inappendiculata-Derived Xanthones Protected Joints from Degradation in Male Rats with Collagen-Induced Arthritis by Regulating PPAR-γ Signaling

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          The bark of Securidaca inappendiculata Hassk. is traditionally used for treating inflammatory diseases and bone fractures in China. We have previously validated the xanthone-enriched fraction (XRF) of S. inappendiculata with anti-rheumatic potentials, but mechanism underlying the joints protective effects is still largely unknown.

          Materials and Methods

          The male rats with collagen-induced arthritis (CIA) were treated with XRF. The therapeutic efficacy of XRF was evaluated by arthritis score changes, morphological observation of paws, histological examinations and serological analyses. Protein expression in tissues and cells was investigated by either immunohistochemical or immunoblotting methods, while levels of mRNA expression were investigated by RT-qPCR. Metabolites in serum were detected by LC-MS approach. The joints homogenates were used for analyzing possible targeted genes by genome microarray analyses.

          Results

          Treatment with XRF and methotrexate (MTX) led to significant decrease in arthritis scores, and alleviated deformation of paws in CIA rats. In addition, XRF and MTX reduced circulating TNF-α, IL-1β and IL-17α in the serum and down-regulated TLR4/NF-κB and JNK pathways in joints of CIA rats. Compared to MTX, XRF-loading microemulsion significantly protected joints, which was accompanied by dramatic decrease in MMP3. Differential genes-based KEGG enrichment and metabolomics analysis suggested that XRF reduced fatty acids biosynthesis by regulating PPAR-γ signaling. S inappendiculata-derived 1,7-dihydroxy-3,4-dimethoxyxanthone (XAN) up-regulated PPAR-γ expression in macrophages, but suppressed it in pre-adipocytes in vitro, which was synchronized with SIRT1 changes. Adiponectin production and SCD-1 expression in pre-adipocytes were also decreased. Aside the direct inhibition on MMP3 expression in synovioblast, the presence of XAN in macrophages-pre-adipocytes co-culture system further reinforced this effect.

          Conclusion

          This study revealed the joint protective  advantages of the bioactive fraction from S. inappendiculata in CIA rats over MTX, and demonstrated that S. inappendiculata-derived xanthones suppressed the erosive nature of synovioblast acquired under inflammatory circumstances by regulating PPAR-γ signaling-controlled metabolism-immunity feedback.

          Related collections

          Most cited references34

          • Record: found
          • Abstract: found
          • Article: not found

          Inflammation, metaflammation and immunometabolic disorders

          Proper regulation and management of energy, substrate diversity and quantity, as well as macromolecular synthesis and breakdown processes, are fundamental to cellular and organismal survival and are paramount to health. Cellular and multicellular organization are defended by the immune response, a robust and critical system
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties.

            Th1 cytokines promote monocyte differentiation into proatherogenic M1 macrophages, while Th2 cytokines lead to an "alternative" anti-inflammatory M2 macrophage phenotype. Here we show that in human atherosclerotic lesions, the expression of M2 markers and PPARgamma, a nuclear receptor controlling macrophage inflammation, correlate positively. Moreover, PPARgamma activation primes primary human monocytes into M2 differentiation, resulting in a more pronounced anti-inflammatory activity in M1 macrophages. However, PPARgamma activation does not influence M2 marker expression in resting or M1 macrophages, nor does PPARgamma agonist treatment influence the expression of M2 markers in atherosclerotic lesions, indicating that only native monocytes can be primed by PPARgamma activation to an enhanced M2 phenotype. Furthermore, PPARgamma activation significantly increases expression of the M2 marker MR in circulating peripheral blood mononuclear cells. These data demonstrate that PPARgamma activation skews human monocytes toward an anti-inflammatory M2 phenotype.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Saturated fatty acids trigger TLR4-mediated inflammatory response.

              Toll-like receptors (TLR) mediate infection-induced inflammation and sterile inflammation by endogenous molecules. Among the TLR family, TLR4 is the best understood. However, while its downstream signaling pathways have been well defined, not all ligands of TLR4 are currently known. Current evidence suggests that saturated fatty acids (SFA) act as non-microbial TLR4 agonists, and trigger its inflammatory response. Thus, our present review provides a new perspective on the potential mechanism by which SFAs could modulate TLR4-induced inflammatory responses: (1) SFAs can be recognized by CD14-TLR4-MD2 complex and trigger inflammatory pathways, similar to lipopolysaccharide (LPS). (2) SFAs lead to modification of gut microbiota with an overproduction of LPS after a high-fat intake, enhancing this natural TLR4 ligand. (3) In addition, this metabolic endotoxemia leads to an oxidative stress thereby producing atherogenic lipids - oxLDL and oxidized phospholipids - which trigger CD36-TLR4-TLR6 inflammatory response. (4) Also, the high SFA consumption increases the lipemia and the mmLDL and oxLDL formation through oxidative modifications of LDL. The mmLDL, unlike oxLDL, is involved in activation of the CD14-TLR4-MD2 inflammatory pathway. Those molecules can induce TLR4 inflammatory response by MyD88-dependent and/or MyD88-independent pathways that, in turn, promotes the expression of proinflammatory transcript factors such as factor nuclear kappa B (NF-κB), which plays a crucial role in the induction of inflammatory mediators (cytokines, chemokines, or costimulatory molecules) implicated in the development and progression of many chronic diseases.
                Bookmark

                Author and article information

                Journal
                J Inflamm Res
                J Inflamm Res
                jir
                jinres
                Journal of Inflammation Research
                Dove
                1178-7031
                16 February 2021
                2021
                : 14
                : 395-411
                Affiliations
                [1 ]Department of Traditional Chinese Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital) , Wuhu, 241000, People’s Republic of China
                [2 ]Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College , Wuhu, 241000, People’s Republic of China
                [3 ]Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College , Wuhu, 241000, People’s Republic of China
                [4 ]Department of Electronic Science, Xiamen University , Xiamen, 361005, People’s Republic of China
                [5 ]Faculty of Traditional Thai Medicine, Prince of Songkla University , Hat Yai, 90112, Thailand
                [6 ]Drug Research and Development Center, School of Pharmacy, Wannan Medical College , Wuhu, 241000, Anhui, People’s Republic of China
                [7 ]School of Pharmacy, Anhui College of Traditional Chinese Medicine , Wuhu, 241000, Anhui, People’s Republic of China
                Author notes
                Correspondence: Jun Han Drug Research and Development Center, School of Pharmacy, Wannan Medical College , Wuhu, 241000, Anhui, People’s Republic of China Email hanjun@wnmc.edu.cn
                Cong-Lan Ji School of Pharmacy, Anhui College of Traditional Chinese Medicine , Wuhu, 241000, Anhui, People’s Republic of China Email 37709103@qq.com
                [*]

                These authors contributed equally to this work

                Author information
                http://orcid.org/0000-0002-1683-9391
                http://orcid.org/0000-0002-6800-4919
                http://orcid.org/0000-0002-1064-6548
                Article
                295957
                10.2147/JIR.S295957
                7896782
                ddf85f36-cd73-4bf9-9ecd-f2f984881ac1
                © 2021 Zuo et al.

                This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License ( http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms ( https://www.dovepress.com/terms.php).

                History
                : 08 December 2020
                : 21 January 2021
                Page count
                Figures: 6, References: 34, Pages: 17
                Categories
                Original Research

                Immunology
                peroxisome proliferators-activated receptor gamma,ppar-γ,energy metabolism,rheumatoid arthritis,metabolomics,macrophage,inflammation

                Comments

                Comment on this article