Skip to main content
Top
Published in: Inflammation 5/2014

01-10-2014

Anti-septic Effects of Fisetin In Vitro and In Vivo

Authors: Hayoung Yoo, Sae-Kwang Ku, Min-Su Han, Kyung-Min Kim, Jong-Sup Bae

Published in: Inflammation | Issue 5/2014

Login to get access

Abstract

Sepsis is a state of disrupted inflammatory homeostasis that is initiated by infection. High mobility group box 1 (HMGB1) protein acting as a late mediator of severe vascular inflammatory conditions, such as sepsis and endothelial cell protein C receptor (EPCR), is involved in vascular inflammation. Fisetin, an active compound from the family Fabaceae, was reported to have antiviral, neuroprotective, and anti-inflammatory activities. Here, we determined the anti-septic effects of fisetin on HMGB1-mediated inflammatory responses and on the shedding of EPCR in vitro and in vivo, for the first time. First, we monitored the effects of post-treatment fisetin on lipopolysaccharide (LPS) and cecal ligation and puncture (CLP)-mediated release of HMGB1 and HMGB1-mediated regulation of pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. Post-treatment fisetin was found to suppress LPS-mediated release of HMGB1 and HMGB1-mediated cytoskeletal rearrangements. Fisetin also inhibited HMGB1-mediated hyperpermeability and leukocyte migration in septic mice. Fisetin induced potent inhibition of phorbol-12-myristate 13-acetate (PMA) and CLP-induced EPCR. Fisetin also inhibited the expression and activity of tumor necrosis factor-α converting enzyme, induced by PMA in endothelial cells. In addition, fisetin inhibited the production of tumor necrosis factor-α and the activation of AKT, nuclear factor-κB, and extracellular regulated kinases 1/2 by HMGB1 in HUVECs. Fisetin also down-regulated CLP-induced release of HMGB1, production of interleukin 1β, and reduced septic mortality. Collectively, these results suggest that fisetin may be a candidate therapeutic agent for the treatment of vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway.
Literature
1.
go back to reference Lotze, M.T., and K.J. Tracey. 2005. High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal. Nature Reviews Immunology 5: 331–342.PubMedCrossRef Lotze, M.T., and K.J. Tracey. 2005. High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal. Nature Reviews Immunology 5: 331–342.PubMedCrossRef
2.
go back to reference Degryse, B., T. Bonaldi, P. Scaffidi, et al. 2001. The high mobility group (HMG) boxes of the nuclear protein HMG1 induce chemotaxis and cytoskeleton reorganization in rat smooth muscle cells. Journal of Cell Biology 152: 1197–1206.PubMedCrossRefPubMedCentral Degryse, B., T. Bonaldi, P. Scaffidi, et al. 2001. The high mobility group (HMG) boxes of the nuclear protein HMG1 induce chemotaxis and cytoskeleton reorganization in rat smooth muscle cells. Journal of Cell Biology 152: 1197–1206.PubMedCrossRefPubMedCentral
3.
go back to reference Ito, I., J. Fukazawa, and M. Yoshida. 2007. Post-translational methylation of high mobility group box 1 (HMGB1) causes its cytoplasmic localization in neutrophils. Journal of Biological Chemistry 282: 16336–16344.PubMedCrossRef Ito, I., J. Fukazawa, and M. Yoshida. 2007. Post-translational methylation of high mobility group box 1 (HMGB1) causes its cytoplasmic localization in neutrophils. Journal of Biological Chemistry 282: 16336–16344.PubMedCrossRef
4.
go back to reference Bae, J.S. 2012. Role of high mobility group box 1 in inflammatory disease: Focus on sepsis. Archives of Pharmacal Research 35: 1511–1523.PubMedCrossRef Bae, J.S. 2012. Role of high mobility group box 1 in inflammatory disease: Focus on sepsis. Archives of Pharmacal Research 35: 1511–1523.PubMedCrossRef
5.
go back to reference Andersson, U., and K.J. Tracey. 2011. HMGB1 is a therapeutic target for sterile inflammation and infection. Annual Review of Immunology 29: 139–162.PubMedCrossRef Andersson, U., and K.J. Tracey. 2011. HMGB1 is a therapeutic target for sterile inflammation and infection. Annual Review of Immunology 29: 139–162.PubMedCrossRef
6.
go back to reference Hori, O., J. Brett, T. Slattery, et al. 1995. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. Journal of Biological Chemistry 270: 25752–25761.PubMedCrossRef Hori, O., J. Brett, T. Slattery, et al. 1995. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. Journal of Biological Chemistry 270: 25752–25761.PubMedCrossRef
7.
go back to reference Park, J.S., D. Svetkauskaite, Q. He, et al. 2004. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. Journal of Biological Chemistry 279: 7370–7377.PubMedCrossRef Park, J.S., D. Svetkauskaite, Q. He, et al. 2004. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. Journal of Biological Chemistry 279: 7370–7377.PubMedCrossRef
8.
9.
go back to reference Yang, H., M. Ochani, J. Li, et al. 2004. Reversing established sepsis with antagonists of endogenous high-mobility group box 1. Proceedings of the National Academy of Sciences of the United States of America 101: 296–301.PubMedCrossRefPubMedCentral Yang, H., M. Ochani, J. Li, et al. 2004. Reversing established sepsis with antagonists of endogenous high-mobility group box 1. Proceedings of the National Academy of Sciences of the United States of America 101: 296–301.PubMedCrossRefPubMedCentral
10.
go back to reference Mosnier, L.O., B.V. Zlokovic, and J.H. Griffin. 2007. The cytoprotective protein C pathway. Blood 109: 3161–3172.PubMedCrossRef Mosnier, L.O., B.V. Zlokovic, and J.H. Griffin. 2007. The cytoprotective protein C pathway. Blood 109: 3161–3172.PubMedCrossRef
11.
go back to reference Xu, J., D. Qu, N.L. Esmon, and C.T. Esmon. 2000. Metalloproteolytic release of endothelial cell protein C receptor. Journal of Biological Chemistry 275: 6038–6044.PubMedCrossRef Xu, J., D. Qu, N.L. Esmon, and C.T. Esmon. 2000. Metalloproteolytic release of endothelial cell protein C receptor. Journal of Biological Chemistry 275: 6038–6044.PubMedCrossRef
12.
go back to reference Kurosawa, S., D.J. Stearns-Kurosawa, C.W. Carson, A. D'Angelo, P. Della Valle, and C.T. Esmon. 1998. Plasma levels of endothelial cell protein C receptor are elevated in patients with sepsis and systemic lupus erythematosus: lack of correlation with thrombomodulin suggests involvement of different pathological processes. Blood 91: 725–727.PubMed Kurosawa, S., D.J. Stearns-Kurosawa, C.W. Carson, A. D'Angelo, P. Della Valle, and C.T. Esmon. 1998. Plasma levels of endothelial cell protein C receptor are elevated in patients with sepsis and systemic lupus erythematosus: lack of correlation with thrombomodulin suggests involvement of different pathological processes. Blood 91: 725–727.PubMed
13.
go back to reference Park, H.J., B.T. Jeon, H.C. Kim, et al. 2012. Aged red garlic extract reduces lipopolysaccharide-induced nitric oxide production in RAW 264.7 macrophages and acute pulmonary inflammation through haeme oxygenase-1 induction. Acta Physiologica (Oxford, England) 205: 61–70.CrossRef Park, H.J., B.T. Jeon, H.C. Kim, et al. 2012. Aged red garlic extract reduces lipopolysaccharide-induced nitric oxide production in RAW 264.7 macrophages and acute pulmonary inflammation through haeme oxygenase-1 induction. Acta Physiologica (Oxford, England) 205: 61–70.CrossRef
14.
go back to reference Persson, P.B., and A.B. Persson. 2012. Age your garlic for longevity! Acta Physiologica (Oxford, England) 205: 1–2.CrossRef Persson, P.B., and A.B. Persson. 2012. Age your garlic for longevity! Acta Physiologica (Oxford, England) 205: 1–2.CrossRef
15.
go back to reference Middleton Jr., E., and G. Drzewiecki. 1984. Flavonoid inhibition of human basophil histamine release stimulated by various agents. Biochemical Pharmacology 33: 3333–3338.PubMedCrossRef Middleton Jr., E., and G. Drzewiecki. 1984. Flavonoid inhibition of human basophil histamine release stimulated by various agents. Biochemical Pharmacology 33: 3333–3338.PubMedCrossRef
16.
go back to reference Mukaida, N. 2000. Interleukin-8: An expanding universe beyond neutrophil chemotaxis and activation. International Journal of Hematology 72: 391–398.PubMed Mukaida, N. 2000. Interleukin-8: An expanding universe beyond neutrophil chemotaxis and activation. International Journal of Hematology 72: 391–398.PubMed
17.
go back to reference Hirano, T., S. Higa, J. Arimitsu, et al. 2006. Luteolin, a flavonoid, inhibits AP-1 activation by basophils. Biochemical and Biophysical Research Communications 340: 1–7.PubMedCrossRef Hirano, T., S. Higa, J. Arimitsu, et al. 2006. Luteolin, a flavonoid, inhibits AP-1 activation by basophils. Biochemical and Biophysical Research Communications 340: 1–7.PubMedCrossRef
18.
go back to reference Bakay, M., I. Mucsi, I. Beladi, and M. Gabor. 1968. Effect of flavonoids and related substances. II. Antiviral effect of quercetin, dihydroquercetin and dihydrofisetin. Acta Microbiologica Academiae Scientiarum Hungaricae 15: 223–227.PubMed Bakay, M., I. Mucsi, I. Beladi, and M. Gabor. 1968. Effect of flavonoids and related substances. II. Antiviral effect of quercetin, dihydroquercetin and dihydrofisetin. Acta Microbiologica Academiae Scientiarum Hungaricae 15: 223–227.PubMed
19.
go back to reference Sung, B., M.K. Pandey, and B.B. Aggarwal. 2007. Fisetin, an inhibitor of cyclin-dependent kinase 6, down-regulates nuclear factor-kappaB-regulated cell proliferation, antiapoptotic and metastatic gene products through the suppression of TAK-1 and receptor-interacting protein-regulated IkappaBalpha kinase activation. Molecular Pharmacology 71: 1703–1714.PubMedCrossRef Sung, B., M.K. Pandey, and B.B. Aggarwal. 2007. Fisetin, an inhibitor of cyclin-dependent kinase 6, down-regulates nuclear factor-kappaB-regulated cell proliferation, antiapoptotic and metastatic gene products through the suppression of TAK-1 and receptor-interacting protein-regulated IkappaBalpha kinase activation. Molecular Pharmacology 71: 1703–1714.PubMedCrossRef
20.
go back to reference Akaishi, T., T. Morimoto, M. Shibao, et al. 2008. Structural requirements for the flavonoid fisetin in inhibiting fibril formation of amyloid beta protein. Neuroscience Letters 444: 280–285.PubMedCrossRef Akaishi, T., T. Morimoto, M. Shibao, et al. 2008. Structural requirements for the flavonoid fisetin in inhibiting fibril formation of amyloid beta protein. Neuroscience Letters 444: 280–285.PubMedCrossRef
21.
go back to reference Bae, J.S., and A.R. Rezaie. 2008. Protease activated receptor 1 (PAR-1) activation by thrombin is protective in human pulmonary artery endothelial cells if endothelial protein C receptor is occupied by its natural ligand. Thrombosis and Haemostasis 100: 101–109.PubMedPubMedCentral Bae, J.S., and A.R. Rezaie. 2008. Protease activated receptor 1 (PAR-1) activation by thrombin is protective in human pulmonary artery endothelial cells if endothelial protein C receptor is occupied by its natural ligand. Thrombosis and Haemostasis 100: 101–109.PubMedPubMedCentral
22.
go back to reference Lee, W., E.J. Yang, S.K. Ku, K.S. Song, and J.S. Bae. 2012. Anticoagulant activities of oleanolic acid via inhibition of tissue factor expressions. BMB Reports 45: 390–395.PubMedCrossRef Lee, W., E.J. Yang, S.K. Ku, K.S. Song, and J.S. Bae. 2012. Anticoagulant activities of oleanolic acid via inhibition of tissue factor expressions. BMB Reports 45: 390–395.PubMedCrossRef
23.
go back to reference Kim, T.H., and J.S. Bae. 2010. Ecklonia cava extracts inhibit lipopolysaccharide induced inflammatory responses in human endothelial cells. Food and Chemical Toxicology 48: 1682–1687.PubMedCrossRef Kim, T.H., and J.S. Bae. 2010. Ecklonia cava extracts inhibit lipopolysaccharide induced inflammatory responses in human endothelial cells. Food and Chemical Toxicology 48: 1682–1687.PubMedCrossRef
24.
go back to reference Bae, J.S., and A.R. Rezaie. 2013. Thrombin inhibits HMGB1-mediated proinflammatory signaling responses when endothelial protein C receptor is occupied by its natural ligand. BMB Reports 46: 544–549.PubMedCrossRefPubMedCentral Bae, J.S., and A.R. Rezaie. 2013. Thrombin inhibits HMGB1-mediated proinflammatory signaling responses when endothelial protein C receptor is occupied by its natural ligand. BMB Reports 46: 544–549.PubMedCrossRefPubMedCentral
25.
go back to reference Lee, W., S.K. Ku, and J.S. Bae. 2013. Emodin-6-O-beta-D-glucoside down-regulates endothelial protein C receptor shedding. Archives of Pharmacal Research 36: 1160–1165.PubMedCrossRef Lee, W., S.K. Ku, and J.S. Bae. 2013. Emodin-6-O-beta-D-glucoside down-regulates endothelial protein C receptor shedding. Archives of Pharmacal Research 36: 1160–1165.PubMedCrossRef
26.
go back to reference Ku, S.K., E.J. Yang, K.S. Song, and J.S. Bae. 2013. Rosmarinic acid down-regulates endothelial protein C receptor shedding in vitro and in vivo. Food and Chemical Toxicology 59: 311–315.PubMedCrossRef Ku, S.K., E.J. Yang, K.S. Song, and J.S. Bae. 2013. Rosmarinic acid down-regulates endothelial protein C receptor shedding in vitro and in vivo. Food and Chemical Toxicology 59: 311–315.PubMedCrossRef
27.
go back to reference Miller, M.A., A.S. Meyer, M.T. Beste, et al. 2013. ADAM-10 and −17 regulate endometriotic cell migration via concerted ligand and receptor shedding feedback on kinase signaling. Proceedings of the National Academy of Sciences of the United States of America 110: E2074–2083.PubMedCrossRefPubMedCentral Miller, M.A., A.S. Meyer, M.T. Beste, et al. 2013. ADAM-10 and −17 regulate endometriotic cell migration via concerted ligand and receptor shedding feedback on kinase signaling. Proceedings of the National Academy of Sciences of the United States of America 110: E2074–2083.PubMedCrossRefPubMedCentral
28.
go back to reference Che, W., N. Lerner-Marmarosh, Q. Huang, et al. 2002. Insulin-like growth factor-1 enhances inflammatory responses in endothelial cells: Role of Gab1 and MEKK3 in TNF-alpha-induced c-Jun and NF-kappaB activation and adhesion molecule expression. Circulation Research 90: 1222–1230.PubMedCrossRef Che, W., N. Lerner-Marmarosh, Q. Huang, et al. 2002. Insulin-like growth factor-1 enhances inflammatory responses in endothelial cells: Role of Gab1 and MEKK3 in TNF-alpha-induced c-Jun and NF-kappaB activation and adhesion molecule expression. Circulation Research 90: 1222–1230.PubMedCrossRef
29.
go back to reference Kim, T.H., S.K. Ku, I.C. Lee, and J.S. Bae. 2012. Anti-inflammatory functions of purpurogallin in LPS-activated human endothelial cells. BMB Reports 45: 200–205.PubMedCrossRef Kim, T.H., S.K. Ku, I.C. Lee, and J.S. Bae. 2012. Anti-inflammatory functions of purpurogallin in LPS-activated human endothelial cells. BMB Reports 45: 200–205.PubMedCrossRef
30.
go back to reference Lee, W., S.K. Ku, J.A. Kim, T. Lee, and J.S. Bae. 2013. Inhibitory effects of epi-sesamin on HMGB1-induced vascular barrier disruptive responses in vitro and in vivo. Toxicology and Applied Pharmacology 267: 201–208.PubMedCrossRef Lee, W., S.K. Ku, J.A. Kim, T. Lee, and J.S. Bae. 2013. Inhibitory effects of epi-sesamin on HMGB1-induced vascular barrier disruptive responses in vitro and in vivo. Toxicology and Applied Pharmacology 267: 201–208.PubMedCrossRef
31.
go back to reference Bae, J.S., W. Lee, and A.R. Rezaie. 2012. Polyphosphate elicits proinflammatory responses that are counteracted by activated protein C in both cellular and animal models. Journal of Thrombosis and Haemostasis 10(9): 1736–44.CrossRef Bae, J.S., W. Lee, and A.R. Rezaie. 2012. Polyphosphate elicits proinflammatory responses that are counteracted by activated protein C in both cellular and animal models. Journal of Thrombosis and Haemostasis 10(9): 1736–44.CrossRef
32.
go back to reference Lee, J.D., J.E. Huh, G. Jeon, et al. 2009. Flavonol-rich RVHxR from Rhus verniciflua Stokes and its major compound fisetin inhibits inflammation-related cytokines and angiogenic factor in rheumatoid arthritic fibroblast-like synovial cells and in vivo models. International Immunopharmacology 9: 268–276.PubMedCrossRef Lee, J.D., J.E. Huh, G. Jeon, et al. 2009. Flavonol-rich RVHxR from Rhus verniciflua Stokes and its major compound fisetin inhibits inflammation-related cytokines and angiogenic factor in rheumatoid arthritic fibroblast-like synovial cells and in vivo models. International Immunopharmacology 9: 268–276.PubMedCrossRef
33.
go back to reference Valerio, D.A., T.M. Cunha, N.S. Arakawa, et al. 2007. Anti-inflammatory and analgesic effects of the sesquiterpene lactone budlein A in mice: Inhibition of cytokine production-dependent mechanism. European Journal of Pharmacology 562: 155–163.PubMedCrossRef Valerio, D.A., T.M. Cunha, N.S. Arakawa, et al. 2007. Anti-inflammatory and analgesic effects of the sesquiterpene lactone budlein A in mice: Inhibition of cytokine production-dependent mechanism. European Journal of Pharmacology 562: 155–163.PubMedCrossRef
34.
go back to reference Akeson, A.L., and C.W. Woods. 1993. A fluorometric assay for the quantitation of cell adherence to endothelial cells. Journal of Immunological Methods 163: 181–185.PubMedCrossRef Akeson, A.L., and C.W. Woods. 1993. A fluorometric assay for the quantitation of cell adherence to endothelial cells. Journal of Immunological Methods 163: 181–185.PubMedCrossRef
35.
go back to reference Wang, H., H. Liao, M. Ochani, et al. 2004. Cholinergic agonists inhibit HMGB1 release and improve survival in experimental sepsis. Nature Medicine 10: 1216–1221.PubMedCrossRef Wang, H., H. Liao, M. Ochani, et al. 2004. Cholinergic agonists inhibit HMGB1 release and improve survival in experimental sepsis. Nature Medicine 10: 1216–1221.PubMedCrossRef
36.
go back to reference Mullins, G.E., J. Sunden-Cullberg, A.S. Johansson, et al. 2004. Activation of human umbilical vein endothelial cells leads to relocation and release of high-mobility group box chromosomal protein 1. Scandinavian Journal of Immunology 60: 566–573.PubMedCrossRef Mullins, G.E., J. Sunden-Cullberg, A.S. Johansson, et al. 2004. Activation of human umbilical vein endothelial cells leads to relocation and release of high-mobility group box chromosomal protein 1. Scandinavian Journal of Immunology 60: 566–573.PubMedCrossRef
37.
go back to reference Buras, J.A., B. Holzmann, and M. Sitkovsky. 2005. Animal models of sepsis: setting the stage. Nature Reviews Drug Discovery 4: 854–865.PubMedCrossRef Buras, J.A., B. Holzmann, and M. Sitkovsky. 2005. Animal models of sepsis: setting the stage. Nature Reviews Drug Discovery 4: 854–865.PubMedCrossRef
38.
go back to reference Rittirsch, D., M.S. Huber-Lang, M.A. Flierl, and P.A. Ward. 2009. Immunodesign of experimental sepsis by cecal ligation and puncture. Nature Protocols 4: 31–36.PubMedCrossRefPubMedCentral Rittirsch, D., M.S. Huber-Lang, M.A. Flierl, and P.A. Ward. 2009. Immunodesign of experimental sepsis by cecal ligation and puncture. Nature Protocols 4: 31–36.PubMedCrossRefPubMedCentral
39.
go back to reference Sama, A.E., J. D'Amore, M.F. Ward, G. Chen, and H. Wang. 2004. Bench to bedside: HMGB1-a novel proinflammatory cytokine and potential therapeutic target for septic patients in the emergency department. Academic Emergency Medicine 11: 867–873.PubMed Sama, A.E., J. D'Amore, M.F. Ward, G. Chen, and H. Wang. 2004. Bench to bedside: HMGB1-a novel proinflammatory cytokine and potential therapeutic target for septic patients in the emergency department. Academic Emergency Medicine 11: 867–873.PubMed
40.
go back to reference Berman, R.S., J.D. Frew, and W. Martin. 1993. Endotoxin-induced arterial endothelial barrier dysfunction assessed by an in vitro model. British Journal of Pharmacology 110: 1282–1284.PubMedCrossRefPubMedCentral Berman, R.S., J.D. Frew, and W. Martin. 1993. Endotoxin-induced arterial endothelial barrier dysfunction assessed by an in vitro model. British Journal of Pharmacology 110: 1282–1284.PubMedCrossRefPubMedCentral
41.
go back to reference Goldblum, S.E., X. Ding, T.W. Brann, and J. Campbell-Washington. 1993. Bacterial lipopolysaccharide induces actin reorganization, intercellular gap formation, and endothelial barrier dysfunction in pulmonary vascular endothelial cells: Concurrent F-actin depolymerization and new actin synthesis. Journal of Cellular Physiology 157: 13–23.PubMedCrossRef Goldblum, S.E., X. Ding, T.W. Brann, and J. Campbell-Washington. 1993. Bacterial lipopolysaccharide induces actin reorganization, intercellular gap formation, and endothelial barrier dysfunction in pulmonary vascular endothelial cells: Concurrent F-actin depolymerization and new actin synthesis. Journal of Cellular Physiology 157: 13–23.PubMedCrossRef
42.
go back to reference Wolfson, R.K., E.T. Chiang, and J.G. Garcia. 2011. HMGB1 induces human lung endothelial cell cytoskeletal rearrangement and barrier disruption. Microvascular Research 81: 189–197.PubMedCrossRefPubMedCentral Wolfson, R.K., E.T. Chiang, and J.G. Garcia. 2011. HMGB1 induces human lung endothelial cell cytoskeletal rearrangement and barrier disruption. Microvascular Research 81: 189–197.PubMedCrossRefPubMedCentral
43.
go back to reference Yang, H., H. Wang, C.J. Czura, and K.J. Tracey. 2005. The cytokine activity of HMGB1. Journal of Leukocyte Biology 78: 1–8.PubMedCrossRef Yang, H., H. Wang, C.J. Czura, and K.J. Tracey. 2005. The cytokine activity of HMGB1. Journal of Leukocyte Biology 78: 1–8.PubMedCrossRef
44.
go back to reference Qin, Y.H., S.M. Dai, G.S. Tang, et al. 2009. HMGB1 enhances the proinflammatory activity of lipopolysaccharide by promoting the phosphorylation of MAPK p38 through receptor for advanced glycation end products. Journal of Immunology 183: 6244–6250.CrossRef Qin, Y.H., S.M. Dai, G.S. Tang, et al. 2009. HMGB1 enhances the proinflammatory activity of lipopolysaccharide by promoting the phosphorylation of MAPK p38 through receptor for advanced glycation end products. Journal of Immunology 183: 6244–6250.CrossRef
45.
go back to reference Sun, C., C. Liang, Y. Ren, et al. 2009. Advanced glycation end products depress function of endothelial progenitor cells via p38 and ERK 1/2 mitogen-activated protein kinase pathways. Basic Research in Cardiology 104: 42–49.PubMedCrossRef Sun, C., C. Liang, Y. Ren, et al. 2009. Advanced glycation end products depress function of endothelial progenitor cells via p38 and ERK 1/2 mitogen-activated protein kinase pathways. Basic Research in Cardiology 104: 42–49.PubMedCrossRef
46.
go back to reference Schnittler, H.J., S.W. Schneider, H. Raifer, et al. 2001. Role of actin filaments in endothelial cell–cell adhesion and membrane stability under fluid shear stress. Pflügers Archiv 442: 675–687.PubMedCrossRef Schnittler, H.J., S.W. Schneider, H. Raifer, et al. 2001. Role of actin filaments in endothelial cell–cell adhesion and membrane stability under fluid shear stress. Pflügers Archiv 442: 675–687.PubMedCrossRef
47.
go back to reference Friedl, J., M. Puhlmann, D.L. Bartlett, et al. 2002. Induction of permeability across endothelial cell monolayers by tumor necrosis factor (TNF) occurs via a tissue factor-dependent mechanism: Relationship between the procoagulant and permeability effects of TNF. Blood 100: 1334–1339.PubMed Friedl, J., M. Puhlmann, D.L. Bartlett, et al. 2002. Induction of permeability across endothelial cell monolayers by tumor necrosis factor (TNF) occurs via a tissue factor-dependent mechanism: Relationship between the procoagulant and permeability effects of TNF. Blood 100: 1334–1339.PubMed
48.
go back to reference Petrache, I., A. Birukova, S.I. Ramirez, J.G. Garcia, and A.D. Verin. 2003. The role of the microtubules in tumor necrosis factor-alpha-induced endothelial cell permeability. American Journal of Respiratory Cell and Molecular Biology 28: 574–581.PubMedCrossRef Petrache, I., A. Birukova, S.I. Ramirez, J.G. Garcia, and A.D. Verin. 2003. The role of the microtubules in tumor necrosis factor-alpha-induced endothelial cell permeability. American Journal of Respiratory Cell and Molecular Biology 28: 574–581.PubMedCrossRef
49.
go back to reference Qu, D., Y. Wang, Y. Song, N.L. Esmon, and C.T. Esmon. 2006. The Ser219– > Gly dimorphism of the endothelial protein C receptor contributes to the higher soluble protein levels observed in individuals with the A3 haplotype. Journal of Thrombosis and Haemostasis 4: 229–235.PubMedCrossRef Qu, D., Y. Wang, Y. Song, N.L. Esmon, and C.T. Esmon. 2006. The Ser219– > Gly dimorphism of the endothelial protein C receptor contributes to the higher soluble protein levels observed in individuals with the A3 haplotype. Journal of Thrombosis and Haemostasis 4: 229–235.PubMedCrossRef
50.
go back to reference Qu, D., Y. Wang, N.L. Esmon, and C.T. Esmon. 2007. Regulated endothelial protein C receptor shedding is mediated by tumor necrosis factor-alpha converting enzyme/ADAM17. Journal of Thrombosis and Haemostasis 5: 395–402.PubMedCrossRef Qu, D., Y. Wang, N.L. Esmon, and C.T. Esmon. 2007. Regulated endothelial protein C receptor shedding is mediated by tumor necrosis factor-alpha converting enzyme/ADAM17. Journal of Thrombosis and Haemostasis 5: 395–402.PubMedCrossRef
51.
go back to reference Menschikowski, M., A. Hagelgans, G. Eisenhofer, and G. Siegert. 2009. Regulation of endothelial protein C receptor shedding by cytokines is mediated through differential activation of MAP kinase signaling pathways. Experimental Cell Research 315: 2673–2682.PubMedCrossRef Menschikowski, M., A. Hagelgans, G. Eisenhofer, and G. Siegert. 2009. Regulation of endothelial protein C receptor shedding by cytokines is mediated through differential activation of MAP kinase signaling pathways. Experimental Cell Research 315: 2673–2682.PubMedCrossRef
52.
go back to reference Andersson, U., H. Wang, K. Palmblad, et al. 2000. High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. Journal of Experimental Medicine 192: 565–570.PubMedCrossRefPubMedCentral Andersson, U., H. Wang, K. Palmblad, et al. 2000. High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. Journal of Experimental Medicine 192: 565–570.PubMedCrossRefPubMedCentral
53.
go back to reference Hansson, G.K., and P. Libby. 2006. The immune response in atherosclerosis: A double-edged sword. Nature Reviews Immunology 6: 508–519.PubMedCrossRef Hansson, G.K., and P. Libby. 2006. The immune response in atherosclerosis: A double-edged sword. Nature Reviews Immunology 6: 508–519.PubMedCrossRef
54.
go back to reference Kawahara, K., T. Hashiguchi, K. Kikuchi, et al. 2008. Induction of high mobility group box 1 release from serotonin-stimulated human umbilical vein endothelial cells. International Journal of Molecular Medicine 22: 639–644.PubMed Kawahara, K., T. Hashiguchi, K. Kikuchi, et al. 2008. Induction of high mobility group box 1 release from serotonin-stimulated human umbilical vein endothelial cells. International Journal of Molecular Medicine 22: 639–644.PubMed
55.
go back to reference Dagia, N.M., G. Agarwal, D.V. Kamath, et al. 2010. A preferential p110alpha/gamma PI3K inhibitor attenuates experimental inflammation by suppressing the production of proinflammatory mediators in a NF-kappaB-dependent manner. American Journal of Physiology - Cellular Physiology 298: C929–941.CrossRef Dagia, N.M., G. Agarwal, D.V. Kamath, et al. 2010. A preferential p110alpha/gamma PI3K inhibitor attenuates experimental inflammation by suppressing the production of proinflammatory mediators in a NF-kappaB-dependent manner. American Journal of Physiology - Cellular Physiology 298: C929–941.CrossRef
56.
go back to reference Wang, F.P., L. Li, J. Li, J.Y. Wang, L.Y. Wang, and W. Jiang. 2013. High mobility group box-1 promotes the proliferation and migration of hepatic stellate cells via TLR4-dependent signal pathways of PI3K/Akt and JNK. PLoS ONE 8: e64373.PubMedCrossRefPubMedCentral Wang, F.P., L. Li, J. Li, J.Y. Wang, L.Y. Wang, and W. Jiang. 2013. High mobility group box-1 promotes the proliferation and migration of hepatic stellate cells via TLR4-dependent signal pathways of PI3K/Akt and JNK. PLoS ONE 8: e64373.PubMedCrossRefPubMedCentral
57.
go back to reference Lockyer, J.M., J.S. Colladay, W.L. Alperin-Lea, T. Hammond, and A.J. Buda. 1998. Inhibition of nuclear factor-kappaB-mediated adhesion molecule expression in human endothelial cells. Circulation Research 82: 314–320.PubMedCrossRef Lockyer, J.M., J.S. Colladay, W.L. Alperin-Lea, T. Hammond, and A.J. Buda. 1998. Inhibition of nuclear factor-kappaB-mediated adhesion molecule expression in human endothelial cells. Circulation Research 82: 314–320.PubMedCrossRef
58.
go back to reference Marui, N., M.K. Offermann, R. Swerlick, et al. 1993. Vascular cell adhesion molecule-1 (VCAM-1) gene transcription and expression are regulated through an antioxidant-sensitive mechanism in human vascular endothelial cells. Journal of Clinical Investigation 92: 1866–1874.PubMedCrossRefPubMedCentral Marui, N., M.K. Offermann, R. Swerlick, et al. 1993. Vascular cell adhesion molecule-1 (VCAM-1) gene transcription and expression are regulated through an antioxidant-sensitive mechanism in human vascular endothelial cells. Journal of Clinical Investigation 92: 1866–1874.PubMedCrossRefPubMedCentral
59.
go back to reference Rose, B.A., T. Force, and Y. Wang. 2010. Mitogen-activated protein kinase signaling in the heart: Angels versus demons in a heart-breaking tale. Physiological Reviews 90: 1507–1546.PubMedCrossRef Rose, B.A., T. Force, and Y. Wang. 2010. Mitogen-activated protein kinase signaling in the heart: Angels versus demons in a heart-breaking tale. Physiological Reviews 90: 1507–1546.PubMedCrossRef
60.
go back to reference Park, J.S., F. Gamboni-Robertson, Q. He, et al. 2006. High mobility group box 1 protein interacts with multiple Toll-like receptors. American Journal of Physiology - Cellular Physiology 290: C917–924.CrossRef Park, J.S., F. Gamboni-Robertson, Q. He, et al. 2006. High mobility group box 1 protein interacts with multiple Toll-like receptors. American Journal of Physiology - Cellular Physiology 290: C917–924.CrossRef
61.
go back to reference Yang, H., and K.J. Tracey. 2010. Targeting HMGB1 in inflammation. Biochimica et Biophysica Acta 1799: 149–156.PubMedCrossRef Yang, H., and K.J. Tracey. 2010. Targeting HMGB1 in inflammation. Biochimica et Biophysica Acta 1799: 149–156.PubMedCrossRef
62.
go back to reference Palumbo, R., B.G. Galvez, T. Pusterla, et al. 2007. Cells migrating to sites of tissue damage in response to the danger signal HMGB1 require NF-kappaB activation. Journal of Cell Biology 179: 33–40.PubMedCrossRefPubMedCentral Palumbo, R., B.G. Galvez, T. Pusterla, et al. 2007. Cells migrating to sites of tissue damage in response to the danger signal HMGB1 require NF-kappaB activation. Journal of Cell Biology 179: 33–40.PubMedCrossRefPubMedCentral
64.
go back to reference Bhatia, M., M. He, H. Zhang, and S. Moochhala. 2009. Sepsis as a model of SIRS. Frontiers in Bioscience 14: 4703–4711.CrossRef Bhatia, M., M. He, H. Zhang, and S. Moochhala. 2009. Sepsis as a model of SIRS. Frontiers in Bioscience 14: 4703–4711.CrossRef
65.
go back to reference Tracey, K.J., Y. Fong, D.G. Hesse, et al. 1987. Anti-cachectin/TNF monoclonal antibodies prevent septic shock during lethal bacteraemia. Nature 330: 662–664.PubMedCrossRef Tracey, K.J., Y. Fong, D.G. Hesse, et al. 1987. Anti-cachectin/TNF monoclonal antibodies prevent septic shock during lethal bacteraemia. Nature 330: 662–664.PubMedCrossRef
66.
go back to reference Wichterman, K.A., A.E. Baue, and I.H. Chaudry. 1980. Sepsis and septic shock—a review of laboratory models and a proposal. Journal of Surgical Research 29: 189–201.PubMedCrossRef Wichterman, K.A., A.E. Baue, and I.H. Chaudry. 1980. Sepsis and septic shock—a review of laboratory models and a proposal. Journal of Surgical Research 29: 189–201.PubMedCrossRef
67.
go back to reference Wang, H., H. Yang, C.J. Czura, A.E. Sama, and K.J. Tracey. 2001. HMGB1 as a late mediator of lethal systemic inflammation. American Journal of Respiratory and Critical Care Medicine 164: 1768–1773.PubMedCrossRef Wang, H., H. Yang, C.J. Czura, A.E. Sama, and K.J. Tracey. 2001. HMGB1 as a late mediator of lethal systemic inflammation. American Journal of Respiratory and Critical Care Medicine 164: 1768–1773.PubMedCrossRef
Metadata
Title
Anti-septic Effects of Fisetin In Vitro and In Vivo
Authors
Hayoung Yoo
Sae-Kwang Ku
Min-Su Han
Kyung-Min Kim
Jong-Sup Bae
Publication date
01-10-2014
Publisher
Springer US
Published in
Inflammation / Issue 5/2014
Print ISSN: 0360-3997
Electronic ISSN: 1573-2576
DOI
https://doi.org/10.1007/s10753-014-9883-4

Other articles of this Issue 5/2014

Inflammation 5/2014 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine