Skip to main content
Top
Published in: Journal of Natural Medicines 4/2017

01-10-2017 | Original Paper

Shikonin changes the lipopolysaccharide-induced expression of inflammation-related genes in macrophages

Authors: Lucia Satiko Yoshida, Tomohito Kakegawa, Yasukatsu Yuda, Hiromi Takano-Ohmuro

Published in: Journal of Natural Medicines | Issue 4/2017

Login to get access

Abstract

We aimed to find candidate molecules possibly involved in the anti-inflammatory activity of shikonin (active compound of “Shikon”) by analyzing its effects on gene expression of lipopolysaccharide (LPS)-treated THP-1 macrophages. Polysome-associated mRNAs (those expected to be under translation: translatome) from cells treated with LPS alone (LPS: 5 µg/mL), shikonin alone (S: 100 nM), or LPS plus shikonin (LPS&S) for 3 h were analyzed by DNA microarray followed by detection of enriched pathways/gene ontologies using the tools of the STRING database. Candidate genes in enriched pathways in the comparison of LPS&S cells vs. LPS cells were analyzed by reverse-transcription quantitative real-time PCR (RT-qPCR; 1, 2, and 3 h). DNA microarray showed shikonin significantly influences gene expression. Gene expression changes between LPS&S cells and LPS cells were compared to detect relevant proteins and/or mRNAs underlying its anti-inflammatory effects: shikonin downregulated pathways which were upregulated in LPS cells, for example, ‘innate immune response’. Within changed pathways, three genes were selected for RT-qPCR analyses as key candidates influencing inflammatory responses: CYBA (component of the superoxide-generating Nox2 enzyme), GSK3B (controller of cell responses after toll-like receptor stimulation), and EIF4E (a key factor of the eukaryotic translation initiation factor 4F complex that regulates abundance of other proteins involved in immune functions). All three mRNAs were decreased at 2 h, and CYBA continued low at 3 h relative to LPS cells. Given that shikonin decreased the expression of CYBA gene of Nox2, in addition to the direct inhibition of the Nox2 activity that we have previously shown, it is suggested that one of its anti-inflammatory mechanisms could be attenuation of oxidative stress.
Appendix
Available only for authorised users
Literature
1.
go back to reference Chen X, Yang L, Oppenheim JJ, Howard MZ (2002) Cellular pharmacology studies of shikonin derivatives. Phytother Res 16:199–209CrossRef Chen X, Yang L, Oppenheim JJ, Howard MZ (2002) Cellular pharmacology studies of shikonin derivatives. Phytother Res 16:199–209CrossRef
2.
go back to reference Papageorgiou VP, Assimopoulou AN, Ballis AC (2008) Alkannins and shikonins: a new class of wound healing agents. Curr Med Chem 15:3248–3267CrossRef Papageorgiou VP, Assimopoulou AN, Ballis AC (2008) Alkannins and shikonins: a new class of wound healing agents. Curr Med Chem 15:3248–3267CrossRef
3.
go back to reference Andujar I, Rios JL, Giner RM, Recio MC (2013) Pharmacological properties of shikonin—a review of literature since 2002. Planta Med 79:1685–1697CrossRef Andujar I, Rios JL, Giner RM, Recio MC (2013) Pharmacological properties of shikonin—a review of literature since 2002. Planta Med 79:1685–1697CrossRef
4.
go back to reference Sekine T, Masamizu T, Maitani Y, Nagai T (1998) Evaluation of superoxide anion radical scavenging activity of shikonin by electron spin resonance. Int J Pharm 174:133–139CrossRef Sekine T, Masamizu T, Maitani Y, Nagai T (1998) Evaluation of superoxide anion radical scavenging activity of shikonin by electron spin resonance. Int J Pharm 174:133–139CrossRef
5.
go back to reference Gao D, Kakuma M, Oka S, Sugino K, Sakurai H (2000) Reaction of β-alkannin (shikonin) with reactive oxygen species: detection of β-alkannin free radicals. Bioorg Med Chem 8:2561–2569CrossRef Gao D, Kakuma M, Oka S, Sugino K, Sakurai H (2000) Reaction of β-alkannin (shikonin) with reactive oxygen species: detection of β-alkannin free radicals. Bioorg Med Chem 8:2561–2569CrossRef
6.
go back to reference Yoshida LS, Kohri S, Tsunawaki S, Kakegawa T, Taniguchi T, Takano-Ohmuro H, Fujii H (2014) Evaluation of radical scavenging properties of shikonin. J Clin Biochem Nutr 55:90–96CrossRef Yoshida LS, Kohri S, Tsunawaki S, Kakegawa T, Taniguchi T, Takano-Ohmuro H, Fujii H (2014) Evaluation of radical scavenging properties of shikonin. J Clin Biochem Nutr 55:90–96CrossRef
7.
go back to reference Sekine T, Masumizu T, Maitani Y, Takayama K, Kohno M, Nagai T (1998) Effect of shikonin and alkannin on hydroxyl radical generation system concerned with iron ion. Yakugaku Zasshi 118:609–615CrossRef Sekine T, Masumizu T, Maitani Y, Takayama K, Kohno M, Nagai T (1998) Effect of shikonin and alkannin on hydroxyl radical generation system concerned with iron ion. Yakugaku Zasshi 118:609–615CrossRef
8.
go back to reference Kawakami N, Koyama Y, Tanaka J, Ohara A, Hayakawa T, Fujimoto S (1996) Inhibitory effect of acetylshikonin on the activation of NADPH oxidase in polymorphonuclear leukocytes in both whole cell and cell-free systems. Biol Pharm Bull 19:1266–1270CrossRef Kawakami N, Koyama Y, Tanaka J, Ohara A, Hayakawa T, Fujimoto S (1996) Inhibitory effect of acetylshikonin on the activation of NADPH oxidase in polymorphonuclear leukocytes in both whole cell and cell-free systems. Biol Pharm Bull 19:1266–1270CrossRef
9.
go back to reference Wang JP, Tsao LT, Raung SL, Hsu MF, Kuo SC (1997) Investigation of the inhibition by acetylshikonin of the respiratory burst in rat neutrophils. Br J Pharmacol 121:409–416CrossRef Wang JP, Tsao LT, Raung SL, Hsu MF, Kuo SC (1997) Investigation of the inhibition by acetylshikonin of the respiratory burst in rat neutrophils. Br J Pharmacol 121:409–416CrossRef
10.
go back to reference Yoshida LS, Kawada T, Irie K, Yuda Y, Himi T, Ikemoto F, Takano-Ohmuro H (2010) Shikonin directly inhibits nitric oxide synthases: possible targets that affect thoracic aorta relaxation response and nitric oxide release from RAW 264.7 macrophages. J Pharmacol Sci 112:343–351CrossRef Yoshida LS, Kawada T, Irie K, Yuda Y, Himi T, Ikemoto F, Takano-Ohmuro H (2010) Shikonin directly inhibits nitric oxide synthases: possible targets that affect thoracic aorta relaxation response and nitric oxide release from RAW 264.7 macrophages. J Pharmacol Sci 112:343–351CrossRef
11.
go back to reference Takano-Ohmuro H, Yoshida LS, Yuda Y, Morioka K, Kitani S (2008) Shikonin inhibits IgE-mediated histamine release by human basophils and Syk kinase activity. Inflamm Res 57:484–488CrossRef Takano-Ohmuro H, Yoshida LS, Yuda Y, Morioka K, Kitani S (2008) Shikonin inhibits IgE-mediated histamine release by human basophils and Syk kinase activity. Inflamm Res 57:484–488CrossRef
12.
go back to reference Wiench B, Chen YR, Paulsen M, Hamm R, Schroder S, Yang NS, Efferth T (2013) Integration of different “-omics” technologies identifies inhibition of the IGF1R-Akt-mTOR signaling cascade involved in the cytotoxic effect of shikonin against leukemia cells. Evid Based Complement Alternat Med. doi:10.1155/2013/818709 CrossRefPubMedPubMedCentral Wiench B, Chen YR, Paulsen M, Hamm R, Schroder S, Yang NS, Efferth T (2013) Integration of different “-omics” technologies identifies inhibition of the IGF1R-Akt-mTOR signaling cascade involved in the cytotoxic effect of shikonin against leukemia cells. Evid Based Complement Alternat Med. doi:10.​1155/​2013/​818709 CrossRefPubMedPubMedCentral
13.
go back to reference Fujii N, Yamashita Y, Arima Y, Nagashima M, Nakano H (1992) Induction of topoisomerase II-mediated DNA cleavage by the plant naphthoquinones plumbagin and shikonin. Antimicrob Agents Chemother 36:2589–2594CrossRef Fujii N, Yamashita Y, Arima Y, Nagashima M, Nakano H (1992) Induction of topoisomerase II-mediated DNA cleavage by the plant naphthoquinones plumbagin and shikonin. Antimicrob Agents Chemother 36:2589–2594CrossRef
14.
go back to reference Yang F, Chen Y, Duan W, Zhang C, Zhu H, Ding J (2006) SH-7, a new synthesized shikonin derivative, exerting its potent antitumor activities as a topoisomerase inhibitor. Int J Cancer 119:1184–1193CrossRef Yang F, Chen Y, Duan W, Zhang C, Zhu H, Ding J (2006) SH-7, a new synthesized shikonin derivative, exerting its potent antitumor activities as a topoisomerase inhibitor. Int J Cancer 119:1184–1193CrossRef
15.
go back to reference Plyta ZF, Li T, Papageorgiou VP, Mellidis AS, Assimopoulou AN, Pitsinos EN, Couladouros EA (1998) Inhibition of topoisomerase I by naphthoquinone derivatives. Bioorg Med Chem Lett 8:3385–3390CrossRef Plyta ZF, Li T, Papageorgiou VP, Mellidis AS, Assimopoulou AN, Pitsinos EN, Couladouros EA (1998) Inhibition of topoisomerase I by naphthoquinone derivatives. Bioorg Med Chem Lett 8:3385–3390CrossRef
16.
go back to reference Wang JP, Kuo SC (1997) Impairment of phosphatidylinositol signaling in acetylshikonin-treated neutrophils. Biochem Pharmacol 53:1173–1177CrossRef Wang JP, Kuo SC (1997) Impairment of phosphatidylinositol signaling in acetylshikonin-treated neutrophils. Biochem Pharmacol 53:1173–1177CrossRef
17.
go back to reference Nigorikawa K, Yoshikawa K, Sasaki T, Iida E, Tsukamoto M, Murakami H, Maehama T, Hazeki K, Hazeki O (2006) A naphthoquinone derivative, shikonin, has insulin-like actions by inhibiting both phosphatase and tensin homolog deleted on chromosome 10 and tyrosine phosphatases. Mol Pharmacol 70:1143–1149CrossRef Nigorikawa K, Yoshikawa K, Sasaki T, Iida E, Tsukamoto M, Murakami H, Maehama T, Hazeki K, Hazeki O (2006) A naphthoquinone derivative, shikonin, has insulin-like actions by inhibiting both phosphatase and tensin homolog deleted on chromosome 10 and tyrosine phosphatases. Mol Pharmacol 70:1143–1149CrossRef
18.
go back to reference Hsu MF, Chang LC, Huang LJ, Kuo SC, Lee HY, Lu MC, Wang JP (2009) The influence of acetylshikonin, a natural naphthoquinone, on the production of leukotriene B4 and thromboxane A2 in rat neutrophils. Eur J Pharmacol 607:234–243CrossRef Hsu MF, Chang LC, Huang LJ, Kuo SC, Lee HY, Lu MC, Wang JP (2009) The influence of acetylshikonin, a natural naphthoquinone, on the production of leukotriene B4 and thromboxane A2 in rat neutrophils. Eur J Pharmacol 607:234–243CrossRef
19.
go back to reference Andujar I, Recio MC, Bacelli T, Giner RM, Rios JL (2010) Shikonin reduces oedema induced by phorbol ester by interfering with IκBα degradation thus inhibiting translocation of NF-κB to the nucleus. Br J Pharmacol 160:376–388CrossRef Andujar I, Recio MC, Bacelli T, Giner RM, Rios JL (2010) Shikonin reduces oedema induced by phorbol ester by interfering with IκBα degradation thus inhibiting translocation of NF-κB to the nucleus. Br J Pharmacol 160:376–388CrossRef
20.
go back to reference Chen J, Xie J, Jiang Z, Wang B, Wang Y, Hu X (2011) Shikonin and its analogs inhibit cancer cell glycolysis by targeting tumor pyruvate kinase-M2. Oncogene 30:4297–4306CrossRef Chen J, Xie J, Jiang Z, Wang B, Wang Y, Hu X (2011) Shikonin and its analogs inhibit cancer cell glycolysis by targeting tumor pyruvate kinase-M2. Oncogene 30:4297–4306CrossRef
21.
go back to reference Kazumura K, Yoshida LS, Hara A, Tsuchiya H, Morishita N, Kawagishi H, Kakegawa T, Yuda Y, Takano-Ohmuro H (2016) Inhibition of neutrophil superoxide generation by shikonin is associated with suppression of cellular Ca2+ fluxes. J Clin Biochem Nutr 59:1–9CrossRef Kazumura K, Yoshida LS, Hara A, Tsuchiya H, Morishita N, Kawagishi H, Kakegawa T, Yuda Y, Takano-Ohmuro H (2016) Inhibition of neutrophil superoxide generation by shikonin is associated with suppression of cellular Ca2+ fluxes. J Clin Biochem Nutr 59:1–9CrossRef
22.
go back to reference Cheng YWCC, Lin KL, Hu CM, Lin CH, Kang JJ (2008) Shikonin derivatives inhibited LPS-induced NOS in RAW 264.7 cells via downregulation of MAPK/NF-κB signaling. J Ethnopharmacol 120:264–271CrossRef Cheng YWCC, Lin KL, Hu CM, Lin CH, Kang JJ (2008) Shikonin derivatives inhibited LPS-induced NOS in RAW 264.7 cells via downregulation of MAPK/NF-κB signaling. J Ethnopharmacol 120:264–271CrossRef
23.
go back to reference Nam KN, Son MS, Park JH, Lee EH (2008) Shikonins attenuate microglial inflammatory responses by inhibition of ERK, Akt, and NF-κB: neuroprotective implications. Neuropharmacology 55:819–825CrossRef Nam KN, Son MS, Park JH, Lee EH (2008) Shikonins attenuate microglial inflammatory responses by inhibition of ERK, Akt, and NF-κB: neuroprotective implications. Neuropharmacology 55:819–825CrossRef
24.
go back to reference Staniforth V, Wang SY, Shyur LF, Yang NS (2004) Shikonins, phytocompounds from Lithospermum erythrorhizon, inhibit the transcriptional activation of human tumor necrosis factor α promoter in vivo. J Biol Chem 279:5877–5885CrossRef Staniforth V, Wang SY, Shyur LF, Yang NS (2004) Shikonins, phytocompounds from Lithospermum erythrorhizon, inhibit the transcriptional activation of human tumor necrosis factor α promoter in vivo. J Biol Chem 279:5877–5885CrossRef
25.
go back to reference Chiu SC, Tsao SW, Hwang PI, Vanisree S, Chen YA, Yang NS (2010) Differential functional genomic effects of anti-inflammatory phytocompounds on immune signaling. BMC Genom 11:513. doi:10.1186/1471-2164-11-513 CrossRef Chiu SC, Tsao SW, Hwang PI, Vanisree S, Chen YA, Yang NS (2010) Differential functional genomic effects of anti-inflammatory phytocompounds on immune signaling. BMC Genom 11:513. doi:10.​1186/​1471-2164-11-513 CrossRef
26.
go back to reference Dai Q, Fang J, Zhang FS (2009) Dual role of shikonin in early and late stages of collagen type II arthritis. Mol Biol Rep 36:1597–1604CrossRef Dai Q, Fang J, Zhang FS (2009) Dual role of shikonin in early and late stages of collagen type II arthritis. Mol Biol Rep 36:1597–1604CrossRef
27.
go back to reference Yang Y, Wang J, Yang Q, Wu S, Yang Z, Zhu H, Zheng M, Liu W, Wu W, He J et al (2014) Shikonin inhibits the lipopolysaccharide-induced release of HMGB1 in RAW264.7 cells via IFN and NF-κB signaling pathways. Int Immunopharmacol 19:81–87CrossRef Yang Y, Wang J, Yang Q, Wu S, Yang Z, Zhu H, Zheng M, Liu W, Wu W, He J et al (2014) Shikonin inhibits the lipopolysaccharide-induced release of HMGB1 in RAW264.7 cells via IFN and NF-κB signaling pathways. Int Immunopharmacol 19:81–87CrossRef
28.
go back to reference Ahmed K, Furusawa Y, Tabuchi Y, Emam HF, Piao JL, Hassan MA, Yamamoto T, Kondo T, Kadowaki M (2012) Chemical inducers of heat shock proteins derived from medicinal plants and cytoprotective genes response. Int J Hyperthermia 28:1–8CrossRef Ahmed K, Furusawa Y, Tabuchi Y, Emam HF, Piao JL, Hassan MA, Yamamoto T, Kondo T, Kadowaki M (2012) Chemical inducers of heat shock proteins derived from medicinal plants and cytoprotective genes response. Int J Hyperthermia 28:1–8CrossRef
29.
go back to reference Wang X, Hayashi S, Umezaki M, Yamamoto T, Kageyama-Yahara N, Kondo T, Kadowaki M (2014) Shikonin, a constituent of Lithospermum erythrorhizon exhibits anti-allergic effects by suppressing orphan nuclear receptor Nr4a family gene expression as a new prototype of calcineurin inhibitors in mast cells. Chem Biol Interact 224:117–127CrossRef Wang X, Hayashi S, Umezaki M, Yamamoto T, Kageyama-Yahara N, Kondo T, Kadowaki M (2014) Shikonin, a constituent of Lithospermum erythrorhizon exhibits anti-allergic effects by suppressing orphan nuclear receptor Nr4a family gene expression as a new prototype of calcineurin inhibitors in mast cells. Chem Biol Interact 224:117–127CrossRef
30.
go back to reference Kakegawa T, Miyazaki A, Yasukawa K (2016) Anti-inflammatory effects of alpinone 3-acetate from Alpinia japonica seeds. J Nat Med 70:653–660CrossRef Kakegawa T, Miyazaki A, Yasukawa K (2016) Anti-inflammatory effects of alpinone 3-acetate from Alpinia japonica seeds. J Nat Med 70:653–660CrossRef
32.
go back to reference Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP et al (2015) STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43(Database issue):D447–D452CrossRef Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP et al (2015) STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43(Database issue):D447–D452CrossRef
33.
go back to reference Vignais PV (2002) The superoxide-generating NADPH oxidase: structural aspects and activation mechanism. Cell Mol Life Sci 59:1428–1459CrossRef Vignais PV (2002) The superoxide-generating NADPH oxidase: structural aspects and activation mechanism. Cell Mol Life Sci 59:1428–1459CrossRef
34.
go back to reference Jinnouchi A, Aida Y, Nozoe K, Maeda K, Pabst MJ (2005) Local anesthetics inhibit priming of neutrophils by lipopolysaccharide for enhanced release of superoxide: suppression of cytochrome b 558 expression by disparate mechanisms. J Leukoc Biol 78:1356–1365CrossRef Jinnouchi A, Aida Y, Nozoe K, Maeda K, Pabst MJ (2005) Local anesthetics inhibit priming of neutrophils by lipopolysaccharide for enhanced release of superoxide: suppression of cytochrome b 558 expression by disparate mechanisms. J Leukoc Biol 78:1356–1365CrossRef
35.
go back to reference Piccirillo CA, Bjur E, Topisirovic I, Sonenberg N, Larsson O (2014) Translational control of immune responses: from transcripts to translatomes. Nat Immunol 15:503–511CrossRef Piccirillo CA, Bjur E, Topisirovic I, Sonenberg N, Larsson O (2014) Translational control of immune responses: from transcripts to translatomes. Nat Immunol 15:503–511CrossRef
36.
go back to reference Morris MC, Gilliam EA, Button J, Li L (2014) Dynamic modulation of innate immune response by varying dosages of lipopolysaccharide (LPS) in human monocytic cells. J Biol Chem 289:21584–21590CrossRef Morris MC, Gilliam EA, Button J, Li L (2014) Dynamic modulation of innate immune response by varying dosages of lipopolysaccharide (LPS) in human monocytic cells. J Biol Chem 289:21584–21590CrossRef
37.
go back to reference Yoshida LS, Kawada T, Irie K, Yuda Y, Himi T, Ikemoto F, Takano-Ohmuro H (2010) New targets of shikonin that impair RAW 264.7 macrophages and vascular smooth muscle responses. J Pharmacol Sci 112(Suppl. 1):149P Yoshida LS, Kawada T, Irie K, Yuda Y, Himi T, Ikemoto F, Takano-Ohmuro H (2010) New targets of shikonin that impair RAW 264.7 macrophages and vascular smooth muscle responses. J Pharmacol Sci 112(Suppl. 1):149P
38.
go back to reference McCubrey JA, Steelman LS, Bertrand FE, Davis NM, Abrams SL, Montalto G, D’Assoro AB, Libra M, Nicoletti F, Maestro R, Basecke J, Cocco L, Cervello M, Martelli AM (2014) Multifaceted roles of GSK-3 and Wnt/β-catenin in hematopoiesis and leukemogenesis: opportunities for therapeutic intervention. Leukemia 28:15–33CrossRef McCubrey JA, Steelman LS, Bertrand FE, Davis NM, Abrams SL, Montalto G, D’Assoro AB, Libra M, Nicoletti F, Maestro R, Basecke J, Cocco L, Cervello M, Martelli AM (2014) Multifaceted roles of GSK-3 and Wnt/β-catenin in hematopoiesis and leukemogenesis: opportunities for therapeutic intervention. Leukemia 28:15–33CrossRef
39.
go back to reference Shin S, Wolgamott L, Tcherkezian J, Vallabhapurapu S, Yu Y, Roux PP, Yoon SO (2014) Glycogen synthase kinase-3β positively regulates protein synthesis and cell proliferation through the regulation of translation initiation factor 4E-binding protein 1. Oncogene 33:1690–1699CrossRef Shin S, Wolgamott L, Tcherkezian J, Vallabhapurapu S, Yu Y, Roux PP, Yoon SO (2014) Glycogen synthase kinase-3β positively regulates protein synthesis and cell proliferation through the regulation of translation initiation factor 4E-binding protein 1. Oncogene 33:1690–1699CrossRef
41.
go back to reference Zhao Q, Assimopoulou AN, Klauck SM, Damianakos H, Chinou I, Kretschmer N, Rios JL, Papageorgiou VP, Bauer R, Efferth T (2015) Inhibition of c-MYC with involvement of ERK/JNK/MAPK and AKT pathways as a novel mechanism for shikonin and its derivatives in killing leukemia cells. Oncotarget 6:38934–38951PubMedPubMedCentral Zhao Q, Assimopoulou AN, Klauck SM, Damianakos H, Chinou I, Kretschmer N, Rios JL, Papageorgiou VP, Bauer R, Efferth T (2015) Inhibition of c-MYC with involvement of ERK/JNK/MAPK and AKT pathways as a novel mechanism for shikonin and its derivatives in killing leukemia cells. Oncotarget 6:38934–38951PubMedPubMedCentral
42.
go back to reference Ignatchenko V, Ignatchenko A, Sinha A, Boutros PC, Kislinger T (2015) VennDIS: a JavaFX-based Venn and Euler diagram software to generate publication quality figures. Proteomics 15:1239–1244CrossRef Ignatchenko V, Ignatchenko A, Sinha A, Boutros PC, Kislinger T (2015) VennDIS: a JavaFX-based Venn and Euler diagram software to generate publication quality figures. Proteomics 15:1239–1244CrossRef
Metadata
Title
Shikonin changes the lipopolysaccharide-induced expression of inflammation-related genes in macrophages
Authors
Lucia Satiko Yoshida
Tomohito Kakegawa
Yasukatsu Yuda
Hiromi Takano-Ohmuro
Publication date
01-10-2017
Publisher
Springer Singapore
Published in
Journal of Natural Medicines / Issue 4/2017
Print ISSN: 1340-3443
Electronic ISSN: 1861-0293
DOI
https://doi.org/10.1007/s11418-017-1106-5

Other articles of this Issue 4/2017

Journal of Natural Medicines 4/2017 Go to the issue