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

Renan Fernandes Espírito-Santo, Cassio Santana Meira, Rafael Dos Santos Costa, Otávio Passos Souza Filho, Afranio Ferreira Evangelista, Gustavo Henrique Goulart Trossini, Glaucio Monteiro Ferreira, Eudes da Silva Velozo, Cristiane Flora Villarreal, Milena Botelho Pereira Soares, Renan Fernandes Espírito-Santo, Cassio Santana Meira, Rafael Dos Santos Costa, Otávio Passos Souza Filho, Afranio Ferreira Evangelista, Gustavo Henrique Goulart Trossini, Glaucio Monteiro Ferreira, Eudes da Silva Velozo, Cristiane Flora Villarreal, Milena Botelho Pereira Soares

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.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Chemical structure of braylin.
Fig 1. Chemical structure of braylin.
Fig 2. Cytotoxic effect of braylin and…
Fig 2. Cytotoxic effect of braylin and its modulation of nitric oxide production on macrophages.
Panels A and C: J774 cells (A) or peritoneal exudate macrophages (C) were incubated with vehicle (50% propylene glycol in saline, Ct, control group) or different concentrations of braylin (BRA; 10, 20, 40 or 80 μM) for 72 hours and cell viability was determined by Alamar Blue assay. Gentian violet (GV) was used as positive control. Data are expressed as means ± SEM; n = 9 determinations per group. *Significantly different from the vehicle treated cultures (p < 0.05). ANOVA followed by Tukey´s multiple comparison test. Panels B and D: Concentrations of nitrite were determined in J774 macrophages (B) or peritoneal exudate macrophages (D) treated with vehicle (50% propylene glycol in saline, Ct+, control group), braylin (BRA; 10, 20 or 40 μM) or dexamethasone (Dexa; 40 μM) in the presence of LPS (500 ng/mL) + IFN-γ (5 ng/mL). Cell-free supernatants were collected 24 hours after treatments for nitrite quantification by the Griess method. Ct- shows concentrations of nitrite in unstimulated cells. Data are expressed as means ± SEM; n = 9 determinations per group. *Significantly different from the vehicle treated cultures stimulated with LPS + IFN-γ (p< 0.05). ANOVA followed by Tukey´s multiple comparison test.
Fig 3. Effect of braylin on cytokine…
Fig 3. Effect of braylin on cytokine production by activated macrophages.
Concentrations of TNF-α, IL-1β and IL-6 were determined in cultures of J774 macrophages (panels A, C and E) or peritoneal exudate macrophages (panels B, D and F) treated with vehicle (50% propylene glycol in saline, Ct+, control group), braylin (BRA; 10, 20 or 40 μM) or dexamethasone (Dexa; 40 μM) in the presence of LPS (500 ng/mL) plus IFN-γ (5 ng/mL). Cell-free supernatants were collected 4 hours (for TNF-α measurement) and 24 hours (for IL-1β and IL-6) after treatments for ELISA assay. Ct- shows cytokine concentrations in unstimulated cells. Data are expressed as means ± SEM; n = 10 determinations per group. *Significantly different from the vehicle treated cultures stimulated with LPS + IFN-γ (p < 0.05). ANOVA followed by Tukey´s multiple comparison test.
Fig 4. Best poses of the docking…
Fig 4. Best poses of the docking results to RU486 (pink), dexamethasone (orange) and braylin (cyan) superimposed in GR (pdb 1NHZ).
Fig 5
Fig 5
Docking solutions showing the main interactions for (A) RU486 (pink), (B) dexamethasone (orange) and (C) braylin (cyan) superimposed in GR (pdb 1NHZ).
Fig 6. Involvement of glucocorticoid receptors and…
Fig 6. Involvement of glucocorticoid receptors and NF-κB dependent transcriptional activity in the immunomodulatory effect of braylin.
Panel A shows data from glucocorticoid receptor antagonism assay. Concentrations of TNF-α were determined in J774 macrophages treated with vehicle (50% propylene glycol in saline, Ct+, control group), braylin (BRA, 40 μM), RU486 (GR antagonist, 10 μM) + braylin 40 μM, dexamethasone (Dexa; 40 μM) or RU486 (10 μM) + dexamethasone (40 μM) in the presence of LPS (500 ng/mL) and IFN-γ (5 ng/mL). Cell-free supernatants were collected 4 hours after treatments for TNF-α measurement by ELISA. Ct- and RU-show concentrations of TNF-α in unstimulated cells, treated with vehicle and RU486, respectively. Data are expressed as means ± SEM; n = 10 determinations per group. $Significantly different from the vehicle treated cultures unstimulated (p < 0.05); *Significantly different from the vehicle treated cultures stimulated with LPS + IFN-γ (p < 0.05). +Significantly different from the group untreated with antagonist (p < 0.05). Panel B shows the effect of braylin on the activation of NF-κB on RAW 264.7 Luc macrophages. Cells were pretreated with vehicle (50% propylene glycol in saline, Ct+, control group), braylin (BRA; 10, 20 or 40 μM) or dexamethasone (Dexa; 40 μM) for 1 hour prior to stimulated with LPS (500 ng/mL) and IFN-γ (5 ng/mL) for 3 hours. Ct- shows luciferase activity in unstimulated cells. Luciferase activity was measured in a luminometer. $Significantly different from the vehicle treated cultures unstimulated (p < 0.05); *Significantly different from the vehicle treated cultures stimulated with LPS + IFN-γ (p < 0.05). #Significantly different from the Dexa group (p < 0.05). ANOVA followed by Tukey´s multiple comparison test.
Fig 7. Effects of braylin on complete…
Fig 7. Effects of braylin on complete Freund’s adjuvant (CFA)-induced paw inflammation.
Mice were injected with braylin (BRA; 12.5–100 mg/kg), vehicle (50% propylene glycol in saline; control group) or dexamethasone (Dexa; 2 mg/kg; reference drug) by ip route 40 minutes before CFA (injected at time zero). (A) Inflammatory hyperalgesia measured at 2, 4, 8 and 24 hours after the CFA stimulus. The mechanical nociceptive threshold (axis of ordinates) is represented as the filament weight (g) in which the animal responds in 50% of presentations. (B) Paw edema measured at 2, 4, 8 and 24 hours after CFA, represented as paw volume variation. Data are expressed as means ± SEM; n = 6 mice per group. * Significantly different from the control group (p < 0.05). Two-way ANOVA followed by the Bonferroni’s test.
Fig 8. Effects of braylin on tail…
Fig 8. Effects of braylin on tail flick and hot plate tests in mice.
Panels representing the latency in seconds in the tail flick (panel A) and hot plate (panel B) tests, after ip injection of braylin (BRA; 100 mg/kg), vehicle (50% propylene glycol in saline; control group) or morphine (5 mg/kg; reference drug). Data are reported as means ± SEM; n = 6 mice per group. * Significantly different from the control group (p < 0.05). Two-way ANOVA followed by the Bonferroni’s test.
Fig 9. Effects of braylin on cytokines…
Fig 9. Effects of braylin on cytokines paw levels during CFA-induced inflammation.
Mice were injected with braylin (BRA; 50 mg/kg), vehicle (50% propylene glycol in saline; control group) or dexamethasone (Dexa; 2 mg/kg; reference drug) by ip route 40 minutes before CFA (injected at time zero). The naïve group consists of mice that did not receive any experimental manipulation. Panels shows the paw levels of (A) interleukin-1β (IL-1β), (B) tumor necrosis factor-α (TNF-α), (C) interleukin-6 (IL-6), (D) interleukin-13 (IL-13), (E) interleukin-10 (IL-10) and (F) transforming growth factor-β (TGF-β), determined in skin tissues samples by ELISA, 3 hours after the CFA injection. The results are expressed as picograms of cytokine per milligram of protein. Data are expressed as means ± SEM; n = 6 mice per group. * Significantly different from the vehicle group in the same time (p < 0.05); # significantly different from the naive group (p < 0.05). ANOVA followed by Tukey´s multiple comparison test.

References

    1. Williams JP, Meyers JA. Immune-mediated inflammatory disorders (I.M.I.D.s): the economic and clinical costs. Am J Manag Care 2002;21: S664–681.
    1. Davidson A, Diamond B. Autoimmune diseases. N Engl J Med. 2001;345: 340–350. doi:
    1. Kuek A, Hazleman BL, Ostor AJK. Immune-mediated inflammatory diseases (IMIDs) and biologic therapy: a medical revolution. Postgrad Med J. 2007;83: 251–260. doi:
    1. Verpoorte R, Vander Heijden R, Memelink J. Engineering the plant cell factory for secondary metabolite production. Transgenic Res. 2000;9: 323–343.
    1. Venugopala KN, Rashmi V, Odhav B. Review on natural coumarin lead compounds for their pharmacological activity. Biomed Res Int. 2013. doi:
    1. Peng XM, Damu GL, Zhou C. Current developments of coumarin compounds in medicinal chemistry. Curr Pharm Des. 2013;19: 3884–3930.
    1. Fylaktakidou KC, Hadjipavlou-Litina DJ, Litinas KE, Nicolaides DN. Natural and synthetic coumarin derivatives with anti-inflammatory/antioxidant activities. Current Pharm Design 2004;10: 3813–3833.
    1. Lima FO, Nonato FR, Couto RD, Barbosa-Filho JM, Nunes XP, Ribeiro dos Santos R, et al. Mechanisms involved in the antinociceptive effects of 7-hydroxycoumarin. J Nat Prod. 2011;74: 596–602. doi:
    1. Li ZP, Hu JF, Sun MN, Ji HJ, Zhao M, Wu DH, et al. Effect of compound IMMLG5521, a novel coumarin derivative, on carrageenan-induced pleurisy in rats. Eur J Pharmacol. 2011;661: 118–123. doi:
    1. Huang GJ, Deng JS, Liao JC, Hou WC, Wang SY, Sung PJ, et al. Inducible nitric oxide synthase and cyclooxygenase-2 participate in anti-inflammatory activity of imperatorin from Glehnia littoralis. J Agric Food Chem. 2012;60: 1673–1681. doi:
    1. Aneja R, Mukerjee SK, Seshadri TR. Synthesis of benzo-furan derivatives-I: karanj ketone, karanjin and pongapin. Tetrahedron 1958;2: 203–210.
    1. Lin TT, Huang YY, Tang GH, Cheng ZB, Liu X, Luo HB, et al. Prenylated coumarins: natural phosphodiesterase-4 inhibitors from Toddalia asiatica. J Nat Prod. 2014;77: 955–962. doi:
    1. Bender AT, Beavo JA. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev. 2006;58: 488–520. doi:
    1. Serezani CH, Ballinger MN, Aronoff DM, Peters-Golden M. Cyclic AMP: master regulator of innate immune cell function. Am J Respir Cell Mol Biol. 2008;39: 127–132. doi:
    1. Salari P, Abdollahi M. Phosphodiesterase inhibitors in inflammatory bowel disease. Expert Opin Investig Drugs 2012;21: 261–264. doi:
    1. Houslay MD, Schafer P, Zhang KY. Keynote review: phosphodiesterase-4 as a therapeutic target. Drug Discov Today 2005;10: 1503–1519. doi:
    1. Baumer W, Hoppmann J, Rundfeldt C, Kietzmann M. Highly selective phosphodiesterase 4 inhibitors for the treatment of allergic skin diseases and psoriasis. Inflamm Allergy Drug Targets 2007;6: 17–26.
    1. Strand V, Fiorentino D, Hu C, Day RM, Stevens RM, Papp KA. Improvements in patient-reported outcomes with apremilast, an oral phosphodiesterase 4 inhibitor, in the treatment of moderate to severe psoriasis: results from a phase IIb randomized, controlled study. Health Qual Life Outcomes 2013. doi:
    1. Randrianarivelojosia M, Mulholland DA, Farland KMC. Prenylated coumarins from Cedrelopsis longibracteata (Ptaeroxylaceae). Biochem Syst Ecol. 2005;33: 301–304.
    1. Green LC, Wagne DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR. Analysis of nitrate, nitrite, and [15N] nitrate in biological fluids. Anal Biochem. 1982;126: 131–138.
    1. Zhong H, SuYang H, Erdjument-Bromage H, Tempst P, Ghosh S. The transcriptional activity of NF-kappaB is regulated by the IkappaB-associated PKAc subunit through a cyclic AMP-independent mechanism. Cell 1997;89(3): 413–424.
    1. Verdonk ML, Cole JC, Hartshorn MJ, Murray CW, Taylor RD. Improved protein-ligand docking using GOLD. Proteins 2003;52: 609–623. doi:
    1. Lima MS, Quintans LJ jr, Santana WA, Kaneto CM, Soares MBP, Villarreal CF. Anti-inflammatory effects of carvacrol: evidence for a key role of interleukin-10. Eur J Pharmacol. 2013; 699: 112–117. doi:
    1. Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Meth. 1994;53: 55–63.
    1. Oliveira CM, Nonato FR, Lima FO, Couto RD, David JP, David JM, et al. Antinociceptive properties of bergenin. J Nat Prod. 2011;74: 2062–2068. doi:
    1. Gama KB, Quintans JS, Antoniolli AR, Quintans LJ Jr, Santana WA, Branco A, et al. Evidence for the involvement of descending pain-inhibitory mechanisms in the antinociceptive effect of hecogenin acetate. J Nat Prod. 2013;76: 559–563. doi:
    1. Woolfe G, MacDonald A. The evaluation of the analgesic action of pethidine hydrochloride (demerol). J Pharmacol Exp Ther. 1944;80: 300–307.
    1. Fujiwara N, Kobayashi K. Macrophages in inflammation. Curr Drug Targets Inflamm Allergy 2005;4: 281–286.
    1. Korhonen R, Lahti A, Kankaanranta H, Moilanen E. Nitric oxide production and signaling in inflammation. Curr Drug Targets Inflamm Allergy 2005;4: 471–479.
    1. Kauppi B, Jakob C, Färnegårdh M, Yang J, Ahola H, Alarcon M, et al. The three-dimensional structures of antagonistic and agonistic forms of the glucocorticoid receptor ligand-binding domain: RU-486 induces a transconformation that leads to active antagonism. J Biol Chem. 2003;278(25): 22748–22754. doi:
    1. Edman K, Hosseini A, Bjursell MK, Aagaard A, Wissler L, Gunnarsson A, et al. Ligand Binding Mechanism in Steroid Receptors: From Conserved Plasticity to Differential Evolutionary Constraints. Structure 2015;23(12): 2280–2290. doi:
    1. Matsuo Y, Yamada A, Tsukamoto K, Tamura H, Ikezawa H, Nakamura H, et al. A distant evolutionary relationship between bacterial sphingomyelinase and mammalian DNase I. Protein Sci. 1996;5(12): 2459–2467. doi:
    1. Barnes PJ, Adcock IM. Glucocorticoid resistance in inflammatory diseases. Lancet 2009;373: 1905–1917. doi:
    1. Hapgood JP, Avenant C, Moliki JM. Glucocorticoid-independent modulation of GR activity: Implications for immunotherapy. Pharmacol Ther. 2016;165: 93–113. doi:
    1. Giembycz MA, Newton R. Potential mechanisms to explain how LABAs and PDE4 inhibitors enhance the clinical efficacy of glucocorticoids in inflammatory lung diseases. F1000Prime Rep. 2015. doi:
    1. Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther. 2002;96: 23–43.
    1. Pagès L, Gavaldà A, Lehner MD. PDE4 inhibitors: a review of current developments (2005–2009). Expert Opin Ther Pat. 2009;19: 1501–1519. doi:
    1. Woolf CJ, Allchorne A, Safieh-Garabedian B, Poole S. Cytokines, nerve growth factor and inflammatory hyperalgesia: the contribution of tumour necrosis factor alpha. Br J Pharmacol. 1997;121: 417–424. doi:
    1. Dinarello CA. Proinflammatory cytokines. Chest 2000;118: 503–508.
    1. Cunha FQ, Ferreira SH. Peripheral hyperalgesic cytokines. Adv Exp Med Biol. 2003;521: 22–39.
    1. Conti B, Tabarean I, Andrei C, Bartfai T. Cytokines and fever. Front Biosci. 2004;9: 1433–1449.
    1. Fehrenbacher JC, Vasko MR, Duarte DB. Models of inflammation: Carrageenan- or complete Freund's Adjuvant (CFA)-induced edema and hypersensitivity in the rat. Curr Protoc Pharmacol. 2012. doi:
    1. Le Bars D, Gozariu M, Cadden SW. Animal models of nociception. Pharmacol Rev. 2001;53: 597–652.
    1. Safieh-Garabedian B, Poole S, Allchorne A, Winter J, Woolf CJ. Contribution of interleukin-1 beta to the inflammation-induced increase in nerve growth factor levels and inflammatory hyperalgesia. Br J Pharmacol. 1995;115: 1265–1275.
    1. Faccioli LH, Souza GE, Cunha FQ, Poole S Ferreira SH. Recombinant interleukin-1 and tumor necrosis factor induce neutrophil migration "in vivo" by indirect mechanisms. Agents Actions 1990;30: 344–349.
    1. Verri WA Jr, Cunha TM, Parada CA, Poole S, Cunha FQ, Ferreira SH. Hypernociceptive role of cytokines and chemokines: targets for analgesic drug development? Pharmacol Ther. 2006;112: 116–138. doi:
    1. Cunha FQ, Poole S, Lorenzetti BB, Ferreira SH. The pivotal role of tumour necrosis factor alpha in the development of inflammatory hyperalgesia. Br J Pharmacol. 1992;107: 660–664.
    1. Cunha TM, Verri WA Jr, Silva JS, Poole S, Cunha FQ, Ferreira SH. A cascade of cytokines mediates mechanical inflammatory hypernociception in mice. Proc Natl Acad Sci. USA 2005;102: 1755–1760. doi:
    1. Zucali JR, Dinarello CA, Oblon DJ, Gross MA, Anderson L, Weiner RS. Interleukin 1 stimulates fibroblasts to produce granulocyte-macrophage colony-stimulating activity and prostaglandin E2. J Clin Invest. 1986;77: 1857–1863. doi:
    1. Gitter BD, Labus JM, Lees SL, Scheetz ME. Characteristics of human synovial fibroblast activation by IL-1 beta and TNF alpha. Immunology 1989;66: 196–200.
    1. Hohjoh H, Inazumi T, Tsuchiya S, Sugimoto Y. Prostanoid receptors and acute inflammation in skin. Biochimie 2014. doi:
    1. Letterio JJ, Roberts AB. TGF-beta: A critical modulator of immune cell function. Clin Immunol Immunopathol. 1997;84: 244–250.
    1. Grande JP. Role of transforming growth factor-beta in tissue injury and repair. Proc Soc Exp Biol Med. 1997;214: 27–40.
    1. Dobolyi A, Vincze C, Pal G, Lovas G. The neuroprotective functions of transforming growth factor beta proteins. Int J Mol Sci. 2012;13: 8219–8258. doi:
    1. Chen W, Jin W, Cook M, Weiner HL, Wahl SM. Oral delivery of group a streptococcal cell walls augments circulating TGF-beta and suppresses streptococcal cell wall arthritis. J Immunol. 1998;161: 6297–6304.
    1. Jin YX, Xu LY, Guo H, Ishikawa M, Link H, Xiao BG. TGF-beta1 inhibits protracted-relapsing experimental autoimmune encephalomyelitis by activating dendritic cells. J Autoimmun. 2000;14: 213–220. doi:
    1. Bottner M, Krieglstein K, Unsicker K. Transforming growth factor-beta1 impairs neuropathic pain through pleiotropic effects. J Neurochem. 2000;75: 2227–2240.
    1. Echeverry S, Shi XQ, Haw A, Liu H, Zhang ZW, Zhang J. Transforming growth factor-beta1 impairs neuropathic pain through pleiotropic effects. Mol Pain 2009. doi:
    1. Benveniste EN, Tang LP, Law RM. Differential regulation of astrocyte TNF-alpha expression by the cytokines TGF-beta, IL-6 and IL-10. Int J Dev Neurosci. 1995;13: 341–349.
    1. Chen NF, Huang SY, Chen WF, Chen CH, Lu CH, Chen CL, et al. TGF-beta1 attenuates spinal neuroinflammation and the excitatory amino acid system in rats with neuropathic pain. J Pain 2013;14: 1671–1685. doi:

Source: PubMed

3
Předplatit