1. Academic Validation
  2. The anti-inflammatory and immunomodulatory potential of braylin: Pharmacological properties and mechanisms by in silico, in vitro and in vivo approaches

The anti-inflammatory and immunomodulatory potential of braylin: Pharmacological properties and mechanisms by in silico, in vitro and in vivo approaches

  • PLoS One. 2017 Jun 8;12(6):e0179174. doi: 10.1371/journal.pone.0179174.
Renan Fernandes Espírito-Santo 1 Cassio Santana Meira 2 Rafael Dos Santos Costa 1 Otávio Passos Souza Filho 1 Afranio Ferreira Evangelista 2 Gustavo Henrique Goulart Trossini 3 Glaucio Monteiro Ferreira 3 Eudes da Silva Velozo 1 Cristiane Flora Villarreal 1 2 Milena Botelho Pereira Soares 2 4
Affiliations

Affiliations

  • 1 Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brazil.
  • 2 Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil.
  • 3 Faculdade de Ciências Farmacêuticas, Universidade São Paulo, São Paulo, São Paulo, Brazil.
  • 4 Centro de Biotecnologia e Terapia Celular, Hospital São Rafael, Salvador, Bahia, Brazil.
Abstract

Braylin belongs to the group of natural Coumarins, a group of compounds with a wide range of pharmacological properties. Here we characterized the pharmacological properties of braylin in vitro, in silico and in vivo in models of inflammatory/immune responses. In in vitro assays, braylin exhibited concentration-dependent suppressive activity on activated macrophages. Braylin (10-40 μM) reduced the production of nitrite, IL-1β, TNF-α and IL-6 by J774 cells or peritoneal exudate macrophages stimulated with LPS and IFN-γ. Molecular docking calculations suggested that braylin present an interaction pose to act as a Glucocorticoid Receptor ligand. Corroborating this idea, the inhibitory effect of braylin on macrophages was prevented by RU486, a Glucocorticoid Receptor antagonist. Furthermore, treatment with braylin strongly reduced the NF-κB-dependent transcriptional activity on RAW 264.7 cells. Using the complete Freund's adjuvant (CFA)-induced paw inflammation model in mice, the pharmacological properties of braylin were demonstrated in vivo. Braylin (12.5-100 mg/kg) produced dose-related antinociceptive and antiedematogenic effects on CFA model. Braylin did not produce antinociception on the tail flick and hot plate tests in mice, suggesting that braylin-induced antinociception is not a centrally-mediated action. Braylin exhibited immunomodulatory properties on the CFA model, inhibiting the production of pro-inflammatory cytokines IL-1β, TNF-α and IL-6, while increased the anti-inflammatory cytokine TGF-β. Our results show, for the first time, anti-inflammatory, antinociceptive and immunomodulatory effects of braylin, which possibly act through the Glucocorticoid Receptor activation and by inhibition of the transcriptional activity of NF-κB. Because braylin is a phosphodiesterase-4 inhibitor, this coumarin could represent an ideal prototype of Glucocorticoid Receptor ligand, able to induce synergic immunomodulatory effects.

Figures
Products