Abstract

Studies have discovered that different extracts of Evodia rutaecarpa and its phytochemicals show a variety of biological activities associated with inflammation. Although rutaecarpine, an alkaloid isolated from the unripe fruit of E. rutaecarpa, has been exposed to have anti-inflammatory properties, the mechanism of action has not been well studied. Thus, this study investigated the molecular mechanisms of rutaecarpine (RUT) in lipopolysaccharide (LPS)-induced RAW 264.7 macrophages. RUT reserved the production of nitric oxide (NO) and the expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor (TNF-α), and interleukin (IL)-1β in the LPS-induced macrophages. RUT showed an inhibitory effect on the mitogen-activated protein kinases (MAPKs), and it also inhibited nuclear transcription factor kappa-B (NF-κB) by hindering IκBα and NF-κB p65 phosphorylation and p65 nuclear translocation. The phospho-PI3K and Akt was concentration-dependently suppressed by RUT. However, RUT not only suggestively reduced the migratory ability of macrophages and their numbers induced by LPS but also inhibited the phospho-Src, and FAK. Taken together, these results indicate that RUT participates a vital role in the inhibition of LPS-induced inflammatory processes in RAW 264.7 macrophages and that the mechanisms involve PI3K/Akt and MAPK-mediated downregulation of NF-κB signaling pathways. Notably, reducing the migration and number of cells induced by LPS via inhibiting of Src/FAK pathway was also included to the anti-inflammatory mechanism of RUT. Therefore, RUT may have potential benefits as a therapeutic agent against chronic inflammatory diseases.

Highlights

  • Inflammation is a physiological event of an organism which protects from chemical, physical, infectious agents, environmental toxins, ischemia, or an antigen-antibody interaction

  • Compared with the LPS group, 10 and 20 μM of RUT momentously suppressed the level of nitric oxide (NO) (p < 0.01) and the expression of inducible nitric oxide synthase (iNOS), COX-2 (p < 0.05 and 0.01), TNF-α (p < 0.01), and IL-1β (p < 0.05 and p < 0.01) in a concentration-dependent manner (Figure 2A–F)

  • RUT pretreatment significantly reduced this green fluorescence staining in the nuclear fraction (Figure 6A,B). These results indicate that the antiinflammatory activity of RUT appears to be mediated by the inhibition of the inflammatory mediators, including NO, iNOS, TNF-α, IL-1β, and COX2, as well as the regulation of mitogen-activated protein kinases (MAPKs), NF-κB p65, and Phosphoinositide 3-Kinase (PI3K)/Akt signaling pathways

Read more

Summary

Introduction

Inflammation is a physiological event of an organism which protects from chemical, physical, infectious agents, environmental toxins, ischemia, or an antigen-antibody interaction. In the United States, every year, more than 500,000 patients suffer from sepsis activated by severe systemic inflammation [1]. A variety of external or internal stimulators, such as calcium homeostasis, platelet-activating factor, cytokines, interleukins (IL), chemotaxis, cyclooxygenase (COX), adhesion molecules, reactive oxygen species (ROS), and nitric oxide (NO), are involved in inflammation. Ibuprofen and aspirin have developed among NSAIDs as potential anti-inflammatory drugs that could effectively suppress the role of COX enzyme and inhibit prostaglandin synthesis [4,5]. On the other hand, prolonged intake of NSAIDs is associated with a variety of side effects, including bleeding, coagulopathy, interstitial nephritis, gastrointestinal mucosa damage, and allergic reactions, because of their very low prostaglandin synthesis [6,7]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call