A Xanthohumol-Rich Hop Extract Diminishes Endotoxin-Induced Activation of TLR4 Signaling in Human Peripheral Blood Mononuclear Cells: A Study in Healthy Women

Finn Jung, Raphaela Staltner, Anja Baumann, Katharina Burger, Emina Halilbasic, Claus Hellerbrand, Ina Bergheim, Finn Jung, Raphaela Staltner, Anja Baumann, Katharina Burger, Emina Halilbasic, Claus Hellerbrand, Ina Bergheim

Abstract

Infections with Gram-negative bacteria are still among the leading causes of infection-related deaths. Several studies suggest that the chalcone xanthohumol (XN) found in hop (Humulus lupulus) possesses anti-inflammatory effects. In a single-blinded, placebo controlled randomized cross-over design study we assessed if the oral intake of a single low dose of 0.125 mg of a XN derived through a XN-rich hop extract (75% XN) affects lipopolysaccharide (LPS)-induced immune responses in peripheral blood mononuclear cells (PBMCs) ex vivo in normal weight healthy women (n = 9) (clinicaltrials.gov: NCT04847193) and determined associated molecular mechanisms. LPS-stimulation of PBMCs isolated from participants 1 h after the intake of the placebo for 2 h resulted in a significant induction of pro-inflammatory cytokine release which was significantly attenuated when participants had consumed XN. The XN-dependent attenuation of proinflammatory cytokine release was less pronounced 6 h after the LPS stimulation while the release of sCD14 was significantly reduced at this timepoint. The LPS-dependent activation of hTLR4 transfected HEK293 cells was significantly and dose-dependently suppressed by the XN-rich hop extract which was attenuated when cells were co-challenged with sCD14. Taken together, our results suggest even a one-time intake of low doses of XN consumed in a XN-rich hop extract can suppress LPS-dependent stimulation of PBMCs and that this is related to the interaction of the hop compound with the CD14/TLR4 signaling cascade.

Keywords: CD14; LPS; TLR4; hop; inflammation.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Fluorescence imaging and XN determination of PBMCs isolated from buffy coat of healthy donors co-cultured with Caco-2 cells incubated with XN. (a) Graphical illustration of the experimental co-culture setup using Caco-2 cells and PBMCs. (b) Representative pictures of fluorescence of XN in PBMCs cells after incubation of Caco-2 cells ± XN (0.125 mg/mL derived through a XN-rich hop extract) for 1 h (magnification 400×) in a co-culture model. White arrows indicate autofluorescence of XN. PBMC, peripheral blood mononuclear cell, XN, xanthohumol. Data are expressed as means ± SEM.
Figure 2
Figure 2
Graphical visualization of the study design. (a) Study design and (b) the procedure performed on each day of the study. XN, xanthohumol derived though a xanthohumol-rich hop extract.
Figure 3
Figure 3
Cytokine concentrations in supernatant of LPS−stimulated PBMCs obtained from healthy study participants. Protein concentrations of IL−−1β (a), IL−−6 (b) and TNF−−α (c) in cell culture supernatant of PBMCs stimulated with 0 or 100 ng/mL LPS for 2 h isolated from healthy study participants receiving either a placebo or the study drink containing XN derived through a XN−rich hop extract. IL, interleukin; LPS, lipopolysaccharide; PBMC, peripheral blood mononuclear cell; XN, xanthohumol. Data are expressed as means ± SEM. * = p < 0.005.
Figure 4
Figure 4
Protein concentration of MD−2, TLR4 and CD14 in LPS−stimulated PBMCs obtained from healthy study participants. Representative blots (a) and densitometric analysis of MD−2 western blot (b), TLR4 protein concentration in total protein lysate (c) and sCD14 protein concentration in cell culture supernatant (d) of PBMCs obtained from study participants either receiving a placebo or XN stimulated with LPS (100 ng/mL) for 6 h. LPS, lipopolysaccharide; MD−2, myeloid differentiation factor 2; PBMC, peripheral blood mononuclear cell; sCD14, soluble cluster of differentiation 14; TLR, toll−like receptor; XN, xanthohumol derived through a XN−rich hop extract. Data are expressed as means ± SEM. * = p < 0.05.
Figure 5
Figure 5
Receptor activities of HEK293 cells co-stimulated with LPS and XN for 12h. HEK293 cells were stimulated with LPS (100 ng/mL) and increasing concentrations of XN (0–8 μg/mL) for 12 h. LPS, lipopolysaccharide; XN, xanthohumol derived through a XN-rich hop extract. Data are expressed as means ± SEM. a = p < 0.05 compared to 0 XN + LPS, b = p < 0.05 compared to 0.5 XN + LPS, c = p < 0.05 compared to 1 XN + LPS, d = p < 0.05 compared to 2 XN, e = p < 0.05 compared to 4 XN + LPS.
Figure 6
Figure 6
Inhibitory effect of XN on LPS−binding to MD−2, TLR4 and CD14. Effect of increasing concentrations of XN (0–8 μg/mL) on LPS−binding to MD−2 (a), TLR4 (b) and CD14 (c) as well as receptor activity of hTLR4 HEK293 cells co−stimulated with LPS (100 ng/mL), XN (4 μg/mL) and sCD14 (1000 ng/mL) for 12 h (d). LPS, lipopolysaccharide; MD−2, myeloid differentiation factor 2; sCD14, soluble cluster of differentiation 14; TLR, toll−like receptor; XN, xanthohumol derived through a XN−rich hop extract. Data are expressed as means ± SEM. # = p < 0.05 compared to unstimulated control (0 ng/mL LPS, 0 μg/mL XN). * = p < 0.05 compared to LPS−stimulated cells.

References

    1. Antimicrobial Resistance Collaborators Global burden of bacterial antimicrobial resistance in 2019: A systematic analysis. Lancet. 2022;399:629–655. doi: 10.1016/S0140-6736(21)02724-0.
    1. Massier L., Blüher M., Kovacs P., Chakaroun R.M. Impaired Intestinal Barrier and Tissue Bacteria: Pathomechanisms for Metabolic Diseases. Front. Endocrinol. 2021;12:616506. doi: 10.3389/fendo.2021.616506.
    1. Hritz I., Mandrekar P., Velayudham A., Catalano D., Dolganiuc A., Kodys K., Kurt-Jones E., Szabo G. The critical role of toll-like receptor (TLR) 4 in alcoholic liver disease is independent of the common TLR adapter MyD88. Hepatology. 2008;48:1224–1231. doi: 10.1002/hep.22470.
    1. Lu Z., Zhang X., Li Y., Lopes-Virella M.F., Huanga Y. TLR4 antagonist attenuates atherogenesis in LDL receptor-deficient mice with diet-induced type 2 diabetes. Immunobiology. 2015;220:1246–1254. doi: 10.1016/j.imbio.2015.06.016.
    1. Spruss A., Kanuri G., Wagnerberger S., Haub S., Bischoff S.C., Bergheim I. Toll-like receptor 4 is involved in the development of fructose-induced hepatic steatosis in mice. Hepatology. 2009;50:1094–1104. doi: 10.1002/hep.23122.
    1. Lu Y.C., Yeh W.C., Ohashi P.S. LPS/TLR4 signal transduction pathway. Cytokine. 2008;42:145–151. doi: 10.1016/j.cyto.2008.01.006.
    1. Hege M., Jung F., Sellmann C., Jin C., Ziegenhardt D., Hellerbrand C., Bergheim I. An iso-alpha-acid-rich extract from hops (Humulus lupulus) attenuates acute alcohol-induced liver steatosis in mice. Nutrition. 2018;45:68–75. doi: 10.1016/j.nut.2017.07.010.
    1. Hsu H.-Y., Hua K.-F., Lin C.-C., Lin C.-H., Hsu J., Wong C.-H. Extract of Reishi polysaccharides induces cytokine expression via TLR4-modulated protein kinase signaling pathways. J. Immunol. 2004;173:5989–5999. doi: 10.4049/jimmunol.173.10.5989.
    1. Iniguez A.B., Zhu M.J. Hop bioactive compounds in prevention of nutrition-related noncommunicable diseases. Crit. Rev. Food Sci. Nutr. 2020;61:1900–1913. doi: 10.1080/10408398.2020.1767537.
    1. Legette L., Karnpracha C., Reed R.L., Choi J., Bobe G., Christensen J.M., Rodriguez-Proteau R., Purnell J.Q., Stevens J.F. Human pharmacokinetics of xanthohumol, an antihyperglycemic flavonoid from hops. Mol. Nutr. Food Res. 2014;58:248–255. doi: 10.1002/mnfr.201300333.
    1. Dorn C., Kraus B., Motyl M., Weiss T.S., Gehrig M., Schölmerich J., Heilmann J., Hellerbrand C. Xanthohumol, a chalcon derived from hops, inhibits hepatic inflammation and fibrosis. Mol. Nutr. Food Res. 2010;54((Suppl. S2)):S205–S213. doi: 10.1002/mnfr.200900314.
    1. Miranda C.L., Elias V.D., Hay J.J., Choi J., Reed R.L., Stevens J. Xanthohumol improves dysfunctional glucose and lipid metabolism in diet-induced obese C57BL/6J mice. Arch. Biochem. Biophys. 2016;599:22–30. doi: 10.1016/j.abb.2016.03.008.
    1. Cho Y.-C., Kim H.J., Kim Y.-J., Lee K.Y., Choi H.J., Lee I.-S., Kang B.Y. Differential anti-inflammatory pathway by xanthohumol in IFN-gamma and LPS-activated macrophages. Int. Immunopharmacol. 2008;8:567–573. doi: 10.1016/j.intimp.2007.12.017.
    1. Jung F., Staltner R., Tahir A., Baumann A., Burger K., Halilbasic E., Hellerbrand C., Bergheim I. Oral intake of xanthohumol attenuates lipoteichoic acid-induced inflammatory response in human PBMCs. Eur. J. Nutr. 2022:61. doi: 10.1007/s00394-022-02964-2.
    1. Peluso M.R., Miranda C.L., Hobbs D.J., Proteau R.R., Stevens J.F. Xanthohumol and related prenylated flavonoids inhibit inflammatory cytokine production in LPS-activated THP-1 monocytes: Structure-activity relationships and in silico binding to myeloid differentiation protein-2 (MD-2) Planta Med. 2010;76:1536–1543. doi: 10.1055/s-0029-1241013.
    1. Chen G., Xiao B., Chen L., Bai B., Zhang Y., Xu Z., Fu L., Liu Z., Li X., Zhao Y., et al. Discovery of new MD2-targeted anti-inflammatory compounds for the treatment of sepsis and acute lung injury. Eur. J. Med. Chem. 2017;139:726–740. doi: 10.1016/j.ejmech.2017.08.036.
    1. Langley B.O., Ryan J.J., Hanes D., Phipps J., Stack E., Metz T.O., Stevens J.F., Bradley R. Xanthohumol Microbiome and Signature in Healthy Adults (the XMaS Trial): Safety and Tolerability Results of a Phase I Triple-Masked, Placebo-Controlled Clinical Trial. Mol. Nutr. Food Res. 2021;65:e2001170. doi: 10.1002/mnfr.202001170.
    1. Pichler C., Ferk F., Al-Serori H., Huber W., Jäger W., Waldherr M., Mišík M., Kundi M., Nersesyan A., Herbacek I., et al. Xanthohumol Prevents DNA Damage by Dietary Carcinogens: Results of a Human Intervention Trial. Cancer Prev. Res. 2017;10:153–160. doi: 10.1158/1940-6207.CAPR-15-0378.
    1. Stevens J.F., Page J.E. Xanthohumol and related prenylflavonoids from hops and beer: To your good health! Phytochemistry. 2004;65:1317–1330. doi: 10.1016/j.phytochem.2004.04.025.
    1. Stevens J.F., Taylor A.W., Clawson J.E., Deinzer M.L. Fate of xanthohumol and related prenylflavonoids from hops to beer. J. Agric. Food Chem. 1999;4:2421–2428. doi: 10.1021/jf990101k.
    1. Chen H., Zhang Y., Zhang W., Liu H., Sun C., Zhang B., Bai B., Wu D., Xiao Z., Lum H., et al. Inhibition of myeloid differentiation factor 2 by baicalein protects against acute lung injury. Phytomedicine. 2019;63:152997. doi: 10.1016/j.phymed.2019.152997.
    1. Ventola C.L. The antibiotic resistance crisis: Part 1: Causes and threats. Pharmacol. Ther. 2015;40:277–283.
    1. Ferk F., Mišík M., Nersesyan A., Pichler C., Jäger W., Szekeres T., Marculescu R., Poulsen H.E., Henriksen T., Bono R., et al. Impact of xanthohumol (a prenylated flavonoid from hops) on DNA stability and other health-related biochemical parameters: Results of human intervention trials. Mol. Nutr. Food Res. 2016;60:773–786. doi: 10.1002/mnfr.201500355.
    1. Chen X., Li Z., Hong H., Wang N., Chen J., Lu S., Zhang H., Zhang X., Bei C. Xanthohumol suppresses inflammation in chondrocytes and ameliorates osteoarthritis in mice. Biomed. Pharmacother. 2021;137:111238. doi: 10.1016/j.biopha.2021.111238.
    1. Zhang M., Zhang R., Zheng T., Chen Z., Ji G., Peng F., Wang W. Xanthohumol Attenuated Inflammation and ECM Degradation by Mediating HO-1/C/EBPbeta Pathway in Osteoarthritis Chondrocytes. Front. Pharmacol. 2021;12:680585. doi: 10.3389/fphar.2021.680585.
    1. Bouchard-Boivin F., Désy O., Béland S., Houde I., De Serres S.A. TNF-alpha Production by Monocytes Stimulated with Epstein-Barr Virus-Peptides as a Marker of Immunosuppression-Related Adverse Events in Kidney Transplant Recipients. Kidney Int. Rep. 2019;4:1446–1453. doi: 10.1016/j.ekir.2019.07.007.
    1. Ciesielska A., Matyjek M., Kwiatkowska K. TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling. Cell Mol. Life Sci. 2021;78:1233–1261. doi: 10.1007/s00018-020-03656-y.
    1. Da Silva Correia J., Soldau K., Christen U., Tobias P.S., Ulevitch R.J. Lipopolysaccharide is in close proximity to each of the proteins in its membrane receptor complex. transfer from CD14 to TLR4 and MD-2. J. Biol. Chem. 2001;276:21129–21135. doi: 10.1074/jbc.M009164200.
    1. Da Silva Correia J., Ulevitch R.J. MD-2 and TLR4 N-linked glycosylations are important for a functional lipopolysaccharide receptor. J. Biol. Chem. 2002;277:1845–1854. doi: 10.1074/jbc.M109910200.
    1. Bazil V., Strominger J.L. Shedding as a mechanism of down-modulation of CD14 on stimulated human monocytes. J. Immunol. 1991;147:1567–1574.
    1. Rokita E., Menzel E.J. Characteristics of CD14 shedding from human monocytes. Evidence for the competition of soluble CD14 (sCD14) with CD14 receptors for lipopolysaccharide (LPS) binding. APMIS. 1997;105:510–518. doi: 10.1111/j.1699-0463.1997.tb05048.x.
    1. Dresel M., Dunkel A., Hofmann T. Sensomics analysis of key bitter compounds in the hard resin of hops (Humulus lupulus L.) and their contribution to the bitter profile of Pilsner-type beer. J. Agric. Food Chem. 2015;63:3402–3418. doi: 10.1021/acs.jafc.5b00239.
    1. Menck K., Behme D., Pantke M., Reiling N., Binder C., Pukrop T., Klemm F. Isolation of human monocytes by double gradient centrifugation and their differentiation to macrophages in teflon-coated cell culture bags. J. Vis. Exp. 2014;91:e51554. doi: 10.3791/51554.
    1. Motyl M., Kraus B., Heilmann J. Pitfalls in cell culture work with xanthohumol. Pharmazie. 2012;67:91–94.
    1. Wolff H., Motyl M., Hellerbrand C., Heilmann J., Kraus B. Xanthohumol uptake and intracellular kinetics in hepatocytes, hepatic stellate cells, and intestinal cells. J. Agric. Food Chem. 2011;59:12893–12901. doi: 10.1021/jf203689z.
    1. Capó X., Martorell M., Sureda A., Batle J.M., Tur J.A., Pons A. Docosahexaenoic diet supplementation, exercise and temperature affect cytokine production by lipopolysaccharide-stimulated mononuclear cells. J. Physiol. Biochem. 2016;72:421–434. doi: 10.1007/s13105-016-0490-8.
    1. Brandt A., Jin C.J., Nolte K., Sellmann C., Engstler A.J., Bergheim I. Short-Term Intake of a Fructose-, Fat- and Cholesterol-Rich Diet Causes Hepatic Steatosis in Mice: Effect of Antibiotic Treatment. Nutrients. 2017;9:1013. doi: 10.3390/nu9091013.
    1. Zhang Y., Wu J., Ying S., Chen G., Wu B., Xu T., Liang G. Discovery of new MD2 inhibitor from chalcone derivatives with anti-inflammatory effects in LPS-induced acute lung injury. Sci. Rep. 2016;6:25130. doi: 10.1038/srep25130.

Source: PubMed

3
Prenumerera