Auraptene Acts as an Anti-Inflammatory Agent in the Mouse Brain

Satoshi Okuyama, Mayu Morita, Miki Kaji, Yoshiaki Amakura, Morio Yoshimura, Koji Shimamoto, Yu Ookido, Mitsunari Nakajima, Yoshiko Furukawa, Satoshi Okuyama, Mayu Morita, Miki Kaji, Yoshiaki Amakura, Morio Yoshimura, Koji Shimamoto, Yu Ookido, Mitsunari Nakajima, Yoshiko Furukawa

Abstract

The anti-inflammatory activity of auraptene (AUR), a citrus coumarin, in peripheral tissues is well-known, and we previously demonstrated that AUR exerts anti-inflammatory effects in the ischemic brain; the treatment of mice with AUR for eight days immediately after ischemic surgery suppressed demise and neuronal cell death in the hippocampus, possibly through its anti-inflammatory effects in the brain. We suggested that these effects were at least partly mediated by the suppression of inflammatory mediators derived from astrocytes. The present study showed that (1) AUR, as a pretreatment for five days before and another three days after ischemic surgery, suppressed microglial activation, cyclooxygenase (COX)-2 expression in astrocytes, and COX-2 mRNA expression in the hippocampus; (2) AUR suppressed the lipopolysaccharide-induced expression of COX-2 mRNA and the mRNA of pro-inflammatory cytokines in cultured astrocytes; (3) AUR was still detectable in the brain 60 min after its intraperitoneal administration. These results support our previous suggestion that AUR directly exerts anti-inflammatory effects on the brain.

Keywords: COX-2; anti-inflammation; astrocytes; auraptene; cerebral ischemia; cyclooxygenase-2; hippocampus.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Effects of AUR on the activation of microglia in the hippocampus. (A) Region within the hippocampal region for observations of IBA-1-positive cells; (B) Representative photographs of IBA-1-positive cells in the indicated groups. The scale bar indicates 100 µm; (C) Number of IBA-1-positive cells in the four groups. Values are the means ± SEM (15~18 sections in each group). *** p < 0.001; and ##p < 0.01, as indicated by the brackets.
Figure 2
Figure 2
Effects of AUR on the expression of COX-2 and GFAP in the hippocampus. (A) Representative photographs of COX-2-positive cells (green; a, d, g, j), GFAP-positive cells (red; b, e, h, k), and cells co-expressing COX-2 and GFAP (yellow; c, f, i, l). The scale bar indicates 100 µm; (B) The numbers of COX-2-positive cells; and (C) GFAP-positive cells are shown. Values are the means ± SEM (6 sections in each group). ** p < 0.01; *** p < 0.001; #p < 0.05; and ###p < 0.001, as indicated by the brackets.
Figure 2
Figure 2
Effects of AUR on the expression of COX-2 and GFAP in the hippocampus. (A) Representative photographs of COX-2-positive cells (green; a, d, g, j), GFAP-positive cells (red; b, e, h, k), and cells co-expressing COX-2 and GFAP (yellow; c, f, i, l). The scale bar indicates 100 µm; (B) The numbers of COX-2-positive cells; and (C) GFAP-positive cells are shown. Values are the means ± SEM (6 sections in each group). ** p < 0.01; *** p < 0.001; #p < 0.05; and ###p < 0.001, as indicated by the brackets.
Figure 3
Figure 3
Effects of AUR on the expression of COX-2 mRNA in the hippocampus. (A) Densitometric patterns of COX-2 mRNA and β-actin mRNA bands; (B) Densitometric quantification of COX-2 mRNA band densities normalized by the density of the β-actin mRNA band. Values are the means ± SEM (5~6 for each group). * p < 0.05 and #p < 0.05, as indicated by the brackets.
Figure 4
Figure 4
Effects of AUR on the expression of COX-2, IL-1β, and TNF-α mRNAs in cultured astrocytes. Cells were not treated or were treated with AUR (25 μM) alone for 30 min, with LPS (10 ng/mL) alone for 4 h, or with AUR (25 μM) for 30 min and then with LPS (10 ng/mL) for 4 h. Representative densitometric patterns of COX-2, IL-1β, TNF-α and GAPDH mRNAs bands were shown. Similar results were obtained from at least three independent experiments.
Figure 5
Figure 5
HPLC chromatograms of vehicle-treated brains (A); and brains from mice treated i.p. with AUR (B).
Figure 6
Figure 6
Time course of brain AUR concentration profiles following its i.p. administration (n = 3 at each time point).

References

    1. Murakami A., Kuki W., Takahashi Y., Yonei H., Nakamura Y., Ohto Y., Ohigashi H., Koshimizu K. Auraptene, a citrus coumarin, inhibits 12-O-tetradecanoylphorbol-13-acetate-induced tumor promotion in ICR mouse skin, possibly through suppression of superoxide generation in leukocytes. Jpn. J. Cancer Res. 1997;88:443–452. doi: 10.1111/j.1349-7006.1997.tb00402.x.
    1. Tanaka T., Kawabata K., Kakumoto M., Hara A., Murakami A., Kuki W., Takahashi Y., Yonei H., Maeda M., Ota T., et al. Citrus auraptene exerts dose-dependent chemopreventive activity in rat large bowel tumorigenesis; the inhibition correlates with suppression of cell proliferation and lipid peroxidation and with induction of phase II drug-metabolizing enzymes. Cancer Res. 1998;58:2550–2556.
    1. Murakami A., Nakamura Y., Tanaka T., Kawabata K., Takahashi D., Koshimizu K., Ohigashi H. Suppression by citrus auraptene of phorbol ester- and endotoxin-induced inflammatory responses: Role of attenuation of leukocyte activation. Carcinogenesis. 2000;21:1843–1850. doi: 10.1093/carcin/21.10.1843.
    1. Tanaka T., Yasui Y., Ishigamori-Suzuki R., Oyama T. Citrus compounds inhibit inflammation- and obesity-related colon carcinogenesis in mice. Nutr. Cancer. 2008;60(Suppl. S1):70–80. doi: 10.1080/01635580802381253.
    1. Lin S., Hirai S., Goto T., Sakamoto T., Takahashi N., Yano M., Sasaki T., Yu R., Kawada T. Auraptene suppresses inflammatory responses in activated RAW264 macrophages by inhibiting p38 mitogen-activated protein kinase activation. Mol. Nutr. Food Res. 2013;57:1136–1144. doi: 10.1002/mnfr.201200611.
    1. Yan H., Ma Z., Peng S., Deng X. Anti-inflammatory effect of auraptene extracted from Trifoliate orange (Poncitrus Trifoliate) on LPS-stimulated RAW 264.4 cells. Inflammation. 2013;36:1525–1532. doi: 10.1007/s10753-013-9695-y.
    1. Murakami A., Shigemori T., Ohigashi H. Zingiberaceous and citrus constituents, 1′-acetoxychavicol acetate, zerumbone, auraptene, and nobiletin, suppress lipopolysaccharide-induced cyclooxygenase-2 expression in RAW264.7 murine macrophages through different modes of action. J. Nutr. 2005;135:2987S–2992S.
    1. Okuyama S., Minami S., Shimada N., Makihata N., Nakajima M., Furukawa Y. Anti-inflammatory and neuroprotective effects of auraptene, a citrus coumarin, following cerebral global ischemia in mice. Eur. J. Pharmacol. 2013;699:118–123. doi: 10.1016/j.ejphar.2012.11.043.
    1. Okuyama S., Yamamoto K., Mori H., Toyoda N., Yoshimura M., Amakura Y., Yoshida T., Sugawara S., Sudo M., Nakajima M., et al. Auraptene in the peels of Citrus kawachiensis (kawachi bankan) ameliorates lipopolysaccharide (LPS)-induced inflammation in the mouse brain. Evid. Based Complement. Altern. Med. 2014;2014 doi: 10.1155/2014/408503.
    1. Cao C., Matsumura K., Yamagata K., Watanabe Y. Induction by lipopolysaccharide of cyclooxygenase-2 mRNA in rat brain; its possible role in the febrile response. Brain Res. 1995;697:187–196. doi: 10.1016/0006-8993(95)00839-I.
    1. Weinstein J.R., Koerner I.P., Möller T. Microglia in ischemic brain injury. Future Neurol. 2010;5:227–246. doi: 10.2217/fnl.10.1.
    1. Ito D., Tanaka K., Suzuki S., Dembo T., Fukuuchi Y. Enhanced expression of iba1, ionized calcium-binding adapter molecules 1, after transient focal cerebral ischemia in rat brain. Stroke. 2001;32:1208–1215. doi: 10.1161/01.STR.32.5.1208.
    1. Koistinaho J., Koponen S., Chan P.H. Expression of cyclooxygenase-2 mRNA after global ischemia is regulated by AMP receptors and glucocorticoids. Stroke. 1999;30:1900–1906. doi: 10.1161/01.STR.30.9.1900.
    1. Wu T., Wu H., Wang J., Wang J. Expression and cellular localization of cyclooxygenases and prostaglandin E synthases in the hemorrhagic brain. J. Neuroinflamm. 2011;8:22–29. doi: 10.1186/1742-2094-8-22.
    1. Ouyang Y.-B., Voloboueva L.A., Xu L.-J., Giffard R.G. Selective dysfunction of hippocampal CA1 astrocytes contributes to delayed neuronal damage after transient forebrain ischemia. J. Neurosci. 2007;27:4253–4260. doi: 10.1523/JNEUROSCI.0211-07.2007.
    1. Katsuta K., Umemura K., Ueyama N., Matsuoka N. Pharmacological evidence for a correlation between hippocampal CA1 cell damage and hyperlocomotion following global cerebral ischemia in gerbils. Eur. J. Pharmacol. 2003;467:103–109. doi: 10.1016/S0014-2999(03)01573-5.
    1. Okuyama S., Shimada N., Kaji M., Morita M., Miyoshi K., Minami S., Amakura Y., Yoshimura M., Yoshida T., Watanabe S., et al. Heptamethoxyflavone, a citrus flavonoid, enhances brain-derived neurotrophic factor production and neurogenesis in the hippocampus following cerebral global ischemia in mice. Neurosci. Lett. 2012;528:190–195. doi: 10.1016/j.neulet.2012.08.079.
    1. Soromou L.W., Zhang Z., Li R., Chen N., Guo W., Huo M., Guan S., Lu J., Deng X. Regulation of inflammatory cytokines in lipopolysaccharide-stimulated RAW 264.7 murine macrophage by 7-O-methyl-naringenin. Molecules. 2012;17:3574–3585. doi: 10.3390/molecules17033574.
    1. Bramlett H.M., Dietrich W.D. Pathophysiology of cerebral ischemia and brain trauma: Similarities and differences. J. Cereb. Blood Flow Metab. 2004;24:133–150. doi: 10.1097/01.WCB.0000111614.19196.04.
    1. Ladecola D., Anrather J. The immunology of stroke: From mechanisms to translation. Nat. Med. 2012;17:796–808. doi: 10.1038/nm.2399.
    1. Wang Q., Tang X.N., Yenari M.A. The inflammatory response in stroke. J. Neuroimmunol. 2007;184:53–68. doi: 10.1016/j.jneuroim.2006.11.014.
    1. Townsend K.P., Praticò D. Novel therapeutic opportunities for Alzheimer’s disease: Focus on nonsteroidal anti-inflammatory drugs. FASEB J. 2005;19:1592–1601. doi: 10.1096/fj.04-3620rev.
    1. Nitta A., Ito M., Fukumitsu H., Ohmiya M., Ito H., Sometani A., Nomoto H., Furukawa Y., Furukawa S. 4-Methylcatechol increases brain-derived neurotrophic factor content and mRNA expression in cultured brain cells and in rat brain in vivo. J. Pharmcol. Exp. Ther. 1999;291:1276–1283.
    1. Furukawa Y., Watanabe S., Okuyama S., Nakajima M. Neurotrophic effect of citrus auraptene: Neuritogenic activity in PC12 cells. Int. J. Mol. Sci. 2012;13:5338–5347. doi: 10.3390/ijms13055338.
    1. Okuyama S., Makihata N., Yoshimura M., Amakura Y., Yoshida T., Nakajima M., Furukawa Y. Oenothein B suppresses lipopolysaccharide (LPS)-induced inflammation in the mouse brain. Int. J. Mol. Sci. 2013;14:9767–9778. doi: 10.3390/ijms14059767.

Source: PubMed

3
Subscribe