Protective Effect of Casperome®, an Orally Bioavailable Frankincense Extract, on Lipopolysaccharide- Induced Systemic Inflammation in Mice

Konstantin Loeser, Semjon Seemann, Stefanie König, Isabell Lenhardt, Mona Abdel-Tawab, Andreas Koeberle, Oliver Werz, Amelie Lupp, Konstantin Loeser, Semjon Seemann, Stefanie König, Isabell Lenhardt, Mona Abdel-Tawab, Andreas Koeberle, Oliver Werz, Amelie Lupp

Abstract

Introduction: Despite recent advances in critical care, sepsis remains a crucial cause of morbidity and mortality in intensive care units. Therefore, the identification of new therapeutic strategies is of great importance. Since ancient times, frankincense is used in traditional medicine for the treatment of chronic inflammatory disorders such as rheumatoid arthritis. Thus, the present study intends to evaluate if Casperome® (Casp), an orally bioavailable soy lecithin-based formulation of standardized frankincense extract, is able to ameliorate systemic effects and organ damages induced by severe systemic inflammation using a murine model of sepsis, i.e., intraperitoneal administration of lipopolysaccharides (LPS). Methods: Male 60-day-old mice were assigned to six treatment groups: (1) control, (2) LPS, (3) soy lecithin (blank lecithin without frankincense extract), (4) Casp, (5) soy lecithin plus LPS, or (6) Casp plus LPS. Soy lecithin and Casp were given 3 h prior to LPS treatment; 24 h after LPS administration, animals were sacrificed and health status and serum cytokine levels were evaluated. Additionally, parameters representing liver damage or liver function and indicating oxidative stress in different organs were determined. Furthermore, markers for apoptosis and immune cell redistribution were assessed by immunohistochemistry in liver and spleen. Results: LPS treatment caused a decrease in body temperature, blood glucose levels, liver glycogen content, and biotransformation capacity along with an increase in serum cytokine levels and oxidative stress in various organs. Additionally, apoptotic processes were increased in spleen besides a pronounced immune cell infiltration in both liver and spleen. Pretreatment with Casp significantly improved health status, blood glucose values, and body temperature of the animals, while serum levels of pro-inflammatory cytokines and oxidative stress in all organs tested were significantly diminished. Finally, apoptotic processes in spleen, liver glycogen loss, and immune cell infiltration in liver and spleen were distinctly reduced. Casp also appears to induce various cytochromeP450 isoforms, thus causing re-establishment of liver biotransformation capacity in LPS-treated mice. Conclusion: Casp displayed anti-inflammatory, anti-oxidative, and hepatoprotective effects. Thus, orally bioavailable frankincense extracts may serve as a new supportive treatment option in acute systemic inflammation and accompanied liver dysfunction.

Keywords: cytokines; frankincense; lipopolysaccharides; liver function; oxidative stress; systemic inflammation.

Figures

FIGURE 1
FIGURE 1
Clinical severity score (A), blood glucose values (B), changes in body temperatures (C), changes in b.wt. (D), spleen (E), and adrenal weights (F). Mice were administered either vehicle (control), soy lecithin (Phyto), Casp, LPS (control + LPS), soy lecithin + LPS, or Casp plus LPS (Casp + LPS). Data are given as means ± SEM, n = 8 for each group. ∗, significantly different from controls; +, significantly different from LPS-treated animals; o, significantly different from soy lecithin plus LPS-treated animals (p ≤ 0.05; Mann–Whitney U-test followed by Holm–Bonferroni correction).
FIGURE 2
FIGURE 2
Serum concentrations of TNF-α (A), IL-6 (B), IL-10 (C), and ALAT (D). Mice were administered either vehicle (control), soy lecithin (Phyto), Casp, LPS (control + LPS), soy lecithin + LPS, or Casp plus LPS (Casp + LPS). Data are given as means ± SEM, n = 8 for each group. ∗, significantly different from controls; +, significantly different from LPS-treated animals; o, significantly different from soy lecithin plus LPS-treated animals (p ≤ 0.05; Mann–Whitney U-test followed by Holm–Bonferroni correction).
FIGURE 3
FIGURE 3
Tissue content of LPO in hearts (A), lungs (B), kidneys (C), livers (D), and spleens (E), and concentration of GSH in spleen tissue (F) as a measure of oxidative stress in the organs. Mice were administered either vehicle (control), soy lecithin (Phyto), Casp, LPS (control + LPS), soy lecithin + LPS, or Casp plus LPS (Casp + LPS). Data are given as means ± SEM, n = 8 for each group. ∗, significantly different from controls; +, significantly different from LPS-treated animals; o, significantly different from soy lecithin plus LPS-treated animals (p ≤ 0.05; Mann–Whitney U-test followed by Holm–Bonferroni correction).
FIGURE 4
FIGURE 4
Cleaved caspase-3 (A–C), CD3 (D–F), and CD68 (G–I) expression in the spleens of control (A,D,G), LPS (B,E,H), or Casp plus LPS (C,F,I)-treated mice. Mice were administered either vehicle (control), soy lecithin, Casp, LPS (control + LPS), soy lecithin + LPS, or Casp plus LPS (Casp + LPS). Representative photomicrographs from one of eight different tissue samples from control mice, LPS-treated, and LPS plus Casp-treated animals are shown. Photomicrographs from soy lecithin- or Casp-only-treated mice and from soy lecithin plus LPS-treated mice are not depicted separately since they did not differ from controls or LPS-only-treated mice, respectively. Immunohistochemistry (red–brown color), counterstaining with hematoxylin; original magnification: ×400. Arrows in B and C mark cleaved caspase-3-positive apoptotic cells, arrowheads indicate tingible body macrophages; arrows in G, H, and I mark CD68-positive monocytes/macrophages.
FIGURE 5
FIGURE 5
TNF-α (A–C) and iNOS (D–F) expression in the spleens of control (A,D), LPS (B,E), or Casp plus LPS (C,F)-treated mice. Mice were administered either vehicle (control), soy lecithin, Casp, LPS (control + LPS), soy lecithin + LPS, or Casp plus LPS (Casp + LPS). Representative photomicrographs from one of eight different tissue samples from control mice, LPS-treated, and LPS plus Casp-treated animals are shown. Photomicrographs from soy lecithin- or Casp-only-treated mice and soy lecithin plus LPS-treated mice are not depicted separately since they did not differ from controls or LPS-only-treated mice, respectively. Immunohistochemistry (red–brown color), counterstaining with hematoxylin; original magnification: ×400. Arrows in A–C mark TNF-α-positive monocytes/macrophages, arrows in E and F indicate iNOS-positive neutrophils, respectively.
FIGURE 6
FIGURE 6
CD3 (A–C) and F4/80 (D–F) expression in the spleens of control (A,D), LPS (B,E), or Casp plus LPS (C,F)-treated mice. Mice were administered either vehicle (control), soy lecithin, Casp, LPS (control + LPS), soy lecithin + LPS, or Casp plus LPS (Casp + LPS). Representative photomicrographs from one of eight different tissue samples from control mice, LPS-treated, and LPS plus Casp-treated animals are shown. Photomicrographs from soy lecithin- or Casp-only-treated mice and soy lecithin plus LPS-treated mice are not depicted separately since they did not differ from controls or LPS-only-treated mice, respectively. Immunohistochemistry (red–brown color), counterstaining with hematoxylin; original magnification: ×400. Arrows in A–C mark CD3-positive T-lymphocytes, arrows in D–F indicate F4/80-positive macrophages, respectively.
FIGURE 7
FIGURE 7
iNOS (A–C), TNF-α expression (D–F), and glycogen content (G–I) in the livers of control (A,D,G), LPS (B,E,H), or Casp plus LPS (C,F,I)-treated mice. Mice were administered either vehicle (control), soy lecithin, Casp, LPS (control + LPS), soy lecithin + LPS, or Casp plus LPS (Casp + LPS). Representative photomicrographs from one of eight different tissue samples from control mice, LPS-treated, and LPS plus Casp-treated animals are shown. Photomicrographs from soy lecithin- or Casp-only-treated mice and soy lecithin plus LPS-treated mice are not depicted separately since they did not differ from controls or LPS-only-treated mice, respectively. A–F: Immunohistochemistry (red–brown color), counterstaining with hematoxylin; G–I: periodic acid-Schiff staining; original magnification: ×400. Arrows in B mark iNOS-positive neutrophils, arrows in E and F indicate TNF-α-positive monocytes/macrophages.
FIGURE 8
FIGURE 8
CYP 1A1 (A–C), CYP2B1 (D–F), and CYP3A2 (G–I) expression in the livers of control (A,D,G), LPS (B,E,H), or Casp plus LPS (C,F,I)-treated mice. Mice were administered either vehicle (control), soy lecithin, Casp, LPS (control + LPS), soy lecithin + LPS, or Casp plus LPS (Casp + LPS). Representative photomicrographs from one of eight different tissue samples from control mice, LPS-treated, and LPS plus Casp-treated animals are shown. Photomicrographs from soy lecithin- or Casp-only-treated mice and soy lecithin plus LPS-treated mice are not depicted separately since they did not differ from controls or LPS-only-treated mice, respectively. Immunohistochemistry (red–brown color), counterstaining with hematoxylin; original magnification: ×400. CYP expression was mainly confined to the hepatocytes around the central veins (asterisks).
FIGURE 9
FIGURE 9
Biotransformation capacity as determined by a panel of model reactions for different CYP isoforms. Mice were administered either vehicle (control), soy lecithin (Phyto), Casp, LPS (control + LPS), soy lecithin + LPS, or Casp plus LPS (Casp + LPS). ECOD (A), BROD (B), EROD (C), MROD (D), PROD (E), and EMND (F) were determined in the 9000 × g supernatants of the livers. Data are given as means ± SEM, n = 8 for each group. ∗, significantly different from controls; +, significantly different from LPS-treated animals; o, significantly different from soy lecithin plus LPS-treated animals (p ≤ 0.05; Mann–Whitney U-test followed by Holm–Bonferroni correction).
FIGURE 10
FIGURE 10
Effect of Casp on cytokine release and cytotoxicity in human monocytes. (A) Human primary monocytes were pre-incubated with vehicle or compounds (Casp; dexamethasone, dex) for 30 min prior to stimulation with LPS for 4 (TNF-α, IL-8) or 18 h (IL-6, IL-10). Formed cytokines were measured by ELISA. Data are means ± SEM; n = 3; ∗∗∗p < 0.001; ∗∗p < 0.01; ∗p < 0.05; inhibitor vs. stimulated control (100%), one-way ANOVA plus Bonferroni test. (B) Effect of Casp on cell viability. Intact human monocytes were incubated with compounds (Casp; staurosporine, stsp) or vehicle for 24 h and cell viability was analyzed by MTT assay. Data are means ± SEM; n = 3; ∗p < 0.05; inhibitor vs. vehicle control (100%), one-way ANOVA plus Bonferroni test.

References

    1. Abdel-Tawab M., Werz O., Schubert-Zsilavecz M. (2011). Boswellia serrata: an overall assessment of in vitro, preclinical, pharmacokinetic and clinical data. Clin. Pharmacokinet. 50 349–369. 10.2165/11586800-000000000-00000
    1. Afsar V., Reddy Y. M., Saritha K. V. (2012). In vitro antioxidant activity and antiinflammatory activity of methanolic leaf extract of Boswellia serrata. Int. J. Life Sci. Biotechnol. Pharma Res. 1 15–23.
    1. Aitio A. (1978). A simple and sensitive assay of 7-ethoxycoumarin deethylation. Anal. Biochem. 85 488–491. 10.1016/0003-2697(78)90245-2
    1. Aitken A. E., Richardson T. A., Morgan E. T. (2006). Regulation of drug-metabolizing enzymes and transporters in inflammation. Annu. Rev. Pharmacol. Toxicol. 46 123–149. 10.1146/annurev.pharmtox.46.120604.141059
    1. Al-Harrasi A., Ali L., Ceniviva E., Al-Rawahi A., Hussain J., Hussain H., et al. (2013). Antiglycation and antioxidant activities and HPTLC analysis of Boswellia sacra oleogum resin: the sacred frankincense. Trop. J. Pharm. Res. 12 597–602. 10.4314/tjpr.v12i4.23
    1. Ammon H. P. T. (2006). Boswellic acids in chronic inflammatory diseases. Planta Med. 72 1100–1116. 10.1055/s-2006-947227
    1. Ammon H. P. T. (2010). Modulation of the immune system by Boswellia serrata extracts and boswellic acids. Phytomedicine 17 862–867. 10.1016/j.phymed.2010.03.003
    1. Arismendi-Morillo G. J., Briceño-García A. E., Romero-Amaro Z. R., Fernández-Abreu M. C., Girón-Piña H. E. (2004). Acute non-specific splenitis as indicator of systemic infection. Assessment of 71 autopsy cases. Invest. Clin. 45 131–135.
    1. Ayala A., Muñoz M. F., Argüelles S. (2014). Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid. Med. Cell. Longev. 2014:360438. 10.1155/2014/360438
    1. Carcillo J. A., Doughty L., Kofos D., Frye R. F., Kaplan S., Sasser H., et al. (2003). Cytochrome P450 mediated-drug metabolism is reduced in children with sepsis-induced multiple organ failure. Intensive Care Med. 29 980–984. 10.1007/s00134-003-1758-3
    1. Casteleijn E., Kuiper J., Van Rooij H. C. J., Kamps J. A. A. M., Koster J. F., Van Berkel T. J. C. (1988). Endotoxin stimulates glycogenolysis in the liver by means of intercellular communication. J. Biol. Chem. 263 6953–6955.
    1. Cuaz-Pérolin C., Billiet L., Baugé E., Copin C., Scott-Algara D., Genze F., et al. (2008). Antiinflammatory and antiatherogenic effects of the NF-κ B inhibitor acetyl-11-keto-β-boswellic acid in LPS-challenged ApoE-/- mice. Arterioscler. Thromb. Vasc. Biol. 28 272–277. 10.1161/ATVBAHA.107.155606
    1. Du Z., Liu Z., Ning Z., Liu Y., Song Z., Wang C., et al. (2015). Prospects of boswellic acids as potential pharmaceutics. Planta Med. 81 259–271. 10.1055/s-0034-1396313
    1. Ellman G. L. (1959). Tissue sulfhydryl groups. Arch. Biochem. Biophys. 82 70–77. 10.1016/0003-9861(59)90090-6
    1. Engel C., Brunkhorst F. M., Bone H. G., Brunkhorst R., Gerlach H., Grond S., et al. (2007). Epidemiology of sepsis in Germany: results from a national prospective multicenter study. Intensive Care Med. 33 606–618. 10.1007/s00134-006-0517-7
    1. Farley K. S., Wang L. F., Razavi H. M., Law C., Rohan M., McCormack D. G., et al. (2006). Effects of macrophage inducible nitric oxide synthase in murine septic lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 290 L1164–L1172. 10.1152/ajplung.00248.2005
    1. Fischer A. H., Jacobson K. A., Rose J., Zeller R. (2008). Hematoxylin and eosin staining of tissue and cell sections. CSH Protoc. 2008:pdb.rot4986. 10.1101/pdb.prot4986
    1. Flierl M. A., Rittirsch D., Huber-Lang M., Sarma J. V., Ward P. A. (2008). Catecholamines – Crafty weapons in the inflammatory arsenal of immune/inflammatory cells or opening Pandora’s box? Mol. Med. 14 195–204.
    1. Frank A., Unger M. (2006). Analysis of frankincense from various Boswellia species with inhibitory activity on human drug metabolizing cytochrome P450 enzymes using liquid chromatography mass spectrometry after automated on-line extraction. J. Chromatogr. A 1112 255–262. 10.1016/j.chroma.2005.11.116
    1. Gaieski D. F., Edwards J. M., Kallan M. J., Carr B. G. (2013). Benchmarking the incidence and mortality of severe sepsis in the United States. Crit. Care Med. 41 1167–1174. 10.1097/CCM.0b013e31827c09f8
    1. Galley H. F. (2011). Oxidative stress and mitochondrial dysfunction in sepsis. Br. J. Anaesth. 107 57–64. 10.1093/bja/aer093
    1. Gayathri B., Manjula N., Vinaykumar K. S., Lakshmi B. S., Balakrishnan A. (2007). Pure compound from Boswellia serrata extract exhibits anti-inflammatory property in human PBMCs and mouse macrophages through inhibition of TNF-α, IL-1 β, NO and MAP kinases. Int. Immunopharmacol. 7 473–482. 10.1016/j.intimp.2006.12.003
    1. Goldfarb R. D., Parker T. S., Levine D. M., Glock D., Akhter I., Alkhudari A., et al. (2003). Protein-free phospholipid emulsion treatment improved cardiopulmonary function and survival in porcine sepsis. Am. J. Physiol. Regul. Integr. Comp. Physiol. 284 R550–R557. 10.1152/ajpregu.00285.2002
    1. Gonnert F. A., Recknagel P., Seidel M., Jbeily N., Dahlke K., Bockmeyer C. L., et al. (2011). Characteristics of clinical sepsis reflected in a reliable and reproducible rodent sepsis model. J. Surg. Res. 170 e123–e134. 10.1016/j.jss.2011.05.019
    1. Gordon B. R., Parker T. S., Levine D. M., Feuerbach F., Saal S. D., Sloan B. J., et al. (2005). Neutralization of endotoxin by a phospholipid emulsion in healthy volunteers. J. Infect. Dis. 191 1515–1522. 10.1086/428908
    1. Goto M., Yoshioka T., Battelino T., Ravindranath T., Zeller P. (2001). TNF decreases gluconeogenesis in hepatocytes isolated from 10-day-old rats. Pediatr. Res. 49 552–557. 10.1203/00006450-200104000-00018
    1. Hayes J. D., McLellan L. I. (1999). Glutathione and glutathione-dependent enzymes represent a co-ordinately regulated defence against oxidative stress. Free Radic. Res. 31 273–300. 10.1080/10715769900300851
    1. Henkel A., Kather N., Mönch B., Northoff H., Jauch J., Werz O. (2012). Boswellic acids from frankincense inhibit lipopolysaccharides functionally through direct molecular interference. Biochem. Pharmacol. 82 115–121. 10.1016/j.bcp.2011.09.026
    1. Hiramatsu M., Hotchkiss R. S., Karl I. E., Buchman T. G. (1997). Cecal ligation and puncture (CLP) induces apoptosis in thymus, spleen, lung, and gut by an endotoxin and TNF-independent pathway. Shock 7 247–253. 10.1097/00024382-199704000-00002
    1. Hissin P. J., Hilf R. (1976). A fluorometric method for determination of oxidized and reduced glutathione in tissues. Anal. Biochem. 74 214–226. 10.1016/0003-2697(76)90326-2
    1. Horvatits T., Trauner M., Fuhrmann V. (2013). Hypoxic liver injury and cholestasis in critically ill patients. Curr. Opin. Crit. Care 19 128–132. 10.1097/MCC.0b013e32835ec9e6
    1. Hüsch J., Bohnet J., Fricker G., Skarke C., Artaria C., Appendino G., et al. (2013). Enhanced absorption of boswellic acids by a lecithin delivery form (Phytosome(®)) of Boswellia extract. Fitoterapia 84 89–98. 10.1016/j.fitote.2012.10.002
    1. Jacob A., Zhou M., Wu R., Wang P. (2009). The role of hepatic cytochrome P-450 in sepsis. Int. J. Clin. Exp. Med. 2 203–211.
    1. Kaemmerer D., Sänger J., Arsenic R., D’Haese J. G., Neumann J., Schmitt-Graeff A., et al. (2017). Evaluation of somatostatin, CXCR4 chemokine and endothelin A receptor expression in a large set of paragangliomas. Oncotarget 8 89958–89969. 10.18632/oncotarget.21194
    1. Kim Y. S., Morgan M. J., Choksi S., Liu Z. G. (2007). TNF-induced activation of the Nox1 NADPH oxidase and its role in the induction of necrotic cell death. Mol. Cell 26 675–687. 10.1016/j.molcel.2007.04.021
    1. Kirkeboen K. A., Strand O. A. (1999). The role of nitric oxide in sepsis – an overview. Acta Anaesthesiol. Scand. 43 275–288. 10.1034/j.1399-6576.1999.430307.x
    1. Kleeberg U., Klinger W. (1982). Sensitive formaldehyde determination with Nash’s reagent and a ‘tryptophan reaction’. J. Pharmacol. Methods 8 19–31. 10.1016/0160-5402(82)90004-3
    1. Klinger W., Müller D. (1974). The influence of age on the protein concentration in serum, liver and kidney of rats determined by various methods. Z. Versuchstierkd. 16 149–153.
    1. Koch A., Horn A., Dückers H., Yagmur E., Sanson E., Bruensing J., et al. (2011). Increased liver stiffness denotes hepatic dysfunction and mortality risk in critically ill non-cirrhotic patients at a medical ICU. Crit. Care 15:R266. 10.1186/cc10543
    1. Kramer L., Jordan B., Druml W., Bauer P., Metnitz P. G. H. Austrian Epidemiologic Study on Intensive Care et al. (2007). Incidence and prognosis of early hepatic dysfunction in critically ill patients – A prospective multicenter study. Crit. Care Med. 35 1099–1104. 10.1097/01.CCM.0000259462.97164.A0
    1. Lubet R. A., Mayer R. T., Cameron J. W., Nims R. W., Burke M. D., Wolff T., et al. (1985). Dealkylation of pentoxyresorufin: a rapid and sensitive assay for measuring induction of cytochrome(s) P-450 by phenobarbital and other xenobiotics in the rat. Arch. Biochem. Biophys. 238 43–48. 10.1016/0003-9861(85)90138-9
    1. Martin G. S. (2012). Sepsis, severe sepsis and septic shock: changes in incidence, pathogens and outcomes. Expert Rev. Anti Infect. Ther. 10 701–706. 10.1586/eri.12.50
    1. McManus J. F. (1948). Histological and histochemical uses of periodic acid. Stain Technol. 23 99–108. 10.3109/10520294809106232
    1. Morgan E. T. (2001). Regulation of cytochrome p450 by inflammatory mediators: why and how? Drug Metab. Dispos. 29 207–212.
    1. Moussaieff A., Mechoulam R. (2009). Boswellia resin: from religious ceremonies to medical uses; a review of in-vitro, in-vivo and clinical trials. J. Pharm. Pharmacol. 61 1281–1293. 10.1211/jpp/61.10.0003
    1. Nebert D. W., Russell D. W. (2002). Clinical importance of the cytochrome P450. Lancet 360 1155–1162. 10.1016/S0140-6736(02)11203-7
    1. Nesseler N., Launey Y., Aninat C., Morel F., Mallédant Y., Seguin P. (2012). Clinical review: the liver in sepsis. Crit. Care 16 235–243. 10.1186/cc11381
    1. Novotny A. R., Emmanuel K., Maier S., Westerholt A., Weighardt H., Stadler J., et al. (2007). Cytochrome P450 activity mirrors nitric oxide levels in postoperative sepsis: predictive indicators of lethal outcome. Surgery 141 376–384. 10.1016/j.surg.2006.08.011
    1. Pandey R. S., Singh B. K., Tripathi Y. B. (2005). Extract of gum resins of Boswellia serrata L. inhibits lipopolysaccharide induced nitric oxide production in rat macrophages along with hypolipidemic property. Ind. J. Exp. Biol. 43 509–516.
    1. Poeckel D., Werz O. (2006). Boswellic acids: biological actions and molecular targets. Curr. Med. Chem. 13 3359–3369. 10.2174/092986706779010333
    1. Pohl R. J., Fouts J. R. (1980). A rapid method for assaying the metabolism of 7-ethoxyresorufin by microsomal subcellular fractions. Anal. Biochem. 107 150–155. 10.1016/0003-2697(80)90505-9
    1. Prigent H., Maxime V., Annane D. (2004). Science review: mechanisms of impaired adrenal function in sepsis and molecular actions of glucocorticoids. Crit. Care 8 243–252. 10.1186/cc2878
    1. Raetzsch C. F., Brooks N. L., McKee Alderman J., Moore K. S., Hosick P. A., Klebanov S., et al. (2009). LPS inhibition of glucose production through the TLR4, MYD88, NF(B pathway. Hepatology 50 592–600. 10.1002/hep.22999
    1. Read T. E., Grunfeld C., Zindaba I., Kumwenda L., Calhoun M. C., Kane J. P., et al. (1995). Triglyceride-rich lipoproteins prevent septic death in rats. J. Exp. Med. 182 267–272. 10.1084/jem.182.1.267
    1. Riva A., Morazzoni P., Artaria C., Allegrini P., Meins J., Savio D., et al. (2016). A single-dose, randomized, cross-over, two-way, open-label study for comparing the absorption of boswellic acids and its lecithin formulation. Phytomedicine 23 1375–1382. 10.1016/j.phymed.2016.07.009
    1. Santos G. A., Moura R. F., Vitorino D. C., Roman E. A., Torsoni A. S., Velloso L. A., et al. (2013). Hypothalamic AMPK activation blocks lipopolysaccharide inhibition of glucose production in mice liver. Mol. Cell. Endocrinol. 381 88–96. 10.1016/j.mce.2013.07.018
    1. Saraiva M., O’Garra A. (2010). The regulation of IL-10 production by immune cells. Nat. Rev. Immunol. 10 170–181. 10.1038/nri2711
    1. Seemann S., Lupp A. (2015). Administration of a CXCL12 analog in endotoxemia is associated with anti-inflammatory, anti-oxidative and cytoprotective effects in vivo. PLoS One 10:e0138389. 10.1371/journal.pone.0138389
    1. Seemann S., Lupp A. (2016). Administration of AMD3100 in endotoxemia is associated with pro-inflammatory, pro-oxidative, and pro-apoptotic effects in vivo. J. Biomed. Sci. 23:68. 10.1186/s12929-016-0286-8
    1. Seemann S., Zohles F., Lupp A. (2017). Comprehensive comparison of three different animal models for systemic inflammation. J. Biomed. Sci. 24:60. 10.1186/s12929-017-0370-8
    1. Sharma S., Gupta S., Khajuria V., Bhagat A., Ahmed Z., Shah B. A. (2016). Analogues of boswellic acids as inhibitors of pro-inflammatory cytokines TNF-α and IL-6. Bioorg. Med. Chem. Lett. 26 695–698. 10.1016/j.bmcl.2015.11.035
    1. Shoji Y., Uedono Y., Ishikura H., Takeyama N., Tanaka T. (1995). DNA damage induced by tumour necrosis factor-α in L929 cells is mediated by mitochondrial oxygen radical formation. Immunology 84 543–548.
    1. Siemoneit U., Koeberle A., Rossi A., Dehm F., Verhoff M., Reckel S., et al. (2011). Inhibition of microsomal prostaglandin E2 synthase-1 as a molecular basis for the anti-inflammatory actions of boswellic acids from frankincense. Br. J. Pharmacol. 162 147–162. 10.1111/j.1476-5381.2010.01020.x
    1. Siemoneit U., Pergola C., Jazzar B., Northoff H., Skarke C., Jauch J., et al. (2009). On the interference of boswellic acids with 5-lipoxygenase: mechanistic studies in vitro and pharmacological relevance. Eur. J. Pharmacol. 606 246–254. 10.1016/j.ejphar.2009.01.044
    1. Stortz J. A., Raymond S. L., Mira J. C., Moldawer L. L., Mohr A. M., Efron P. A. (2017). Murine models of sepsis and trauma: can we bridge the gap? ILAR J. 58 90–105. 10.1093/ilar/ilx007
    1. Stürner K. H., Stellmann J. P., Dörr J., Paul F., Friede T., Schammler S., et al. (2017). A standardized frankincense extract reduces disease activity in relapsing-remitting multiple sclerosis (the SABA phase IIa trial). J. Neurol. Neurosurg. Psychiatry 89 1–9. 10.1136/jnnp-2017-317101
    1. Syrovets T., Büchele B., Gedig E., Slupsky J. R., Simmet T. (2000). Acetyl-boswellic acids are novel catalytic inhibitors of human topoisomerases I and IIα. Mol. Pharmacol. 58 71–81. 10.1124/mol.58.1.71
    1. Syrovets T., Gschwend J. E., Büchele B., Laumonnier Y., Zugmaier W., Genze F., et al. (2005). Inhibition of IκB kinase activity by acetyl-boswellic acids promotes apoptosis in androgen-independent PC-3 prostate cancer cells in vitro and in vivo. J. Biol. Chem. 280 6170–6180. 10.1074/jbc.M409477200
    1. Takada Y., Ichikawa H., Badmaev V., Aggarwal B. B. (2006). Acetyl-11-keto-β-boswellic acid potentiates apoptosis, inhibits invasion, and abolishes osteoclastogenesis by suppressing NF-κ B and NF-κ B-regulated gene expression. J. Immunol. 176 3127–3140. 10.4049/jimmunol.176.5.3127
    1. Tsukahara Y., Morisaki T., Kojima M., Uchiyama A., Tanaka M. (2001). iNOS expression by activated neutrophils from patients with sepsis. ANZ J. Surg. 71 15–20. 10.1046/j.1440-1622.2001.02025.x
    1. Valdés-Ferrer S. I., Rosas-Ballina M., Olofsson P. S., Lu B., Dancho M. E., Ochani M., et al. (2013). HMGB1 mediates splenomegaly and expansion of splenic CD11b+ Ly-6Chigh inflammatory monocytes in murine sepsis survivors. J. Intern. Med. 274 381–390. 10.1111/joim.12104
    1. Wang L., Taneja R., Razavi H. M., Law C., Gillis C., Mehta S. (2012). Specific role of neutrophil inducible nitric oxide synthase in murine sepsis-induced lung injury in vivo. Shock 37 539–547. 10.1097/SHK.0b013e31824dcb5a
    1. Yagi K. (1987). Lipid peroxides and human diseases. Chem. Phys. Lipids 45 337–351. 10.1016/0009-3084(87)90071-5
    1. Yan J., Li S., Li S. (2014). The role of the liver in sepsis. Int. Rev. Immunol. 33 498–510. 10.3109/08830185.2014.889129
    1. Yang Y., Liu L., Zhao B., Li M. Q., Wu B., Yan Z., et al. (2007). Relationship between adrenal function and prognosis in patients with severe sepsis. Chin. Med. J. 120 1578–1582.
    1. Zhang G., Dong G., Zhao X., Wang M., Li C. S. (2014). Prognostic significance of hypothalamic-pituitary-adrenal axis hormones in early sepsis: a study performed in the emergency department. Intensive Care Med. 40 1499–1508. 10.1007/s00134-014-3468-4

Source: PubMed

3
Se inscrever