Effect of Propofol on the Production of Inflammatory Cytokines by Human Polarized Macrophages

Tsukasa Kochiyama, Xiaojia Li, Hitoshi Nakayama, Madoka Kage, Yui Yamane, Kenji Takamori, Kazuhisa Iwabuchi, Eiichi Inada, Tsukasa Kochiyama, Xiaojia Li, Hitoshi Nakayama, Madoka Kage, Yui Yamane, Kenji Takamori, Kazuhisa Iwabuchi, Eiichi Inada

Abstract

Macrophages are key immune system cells involved in inflammatory processes. Classically activated (M1) macrophages are characterized by strong antimicrobicidal properties, whereas alternatively activated (M2) macrophages are involved in wound healing. Severe inflammation can induce postoperative complications during the perioperative period. Invasive surgical procedures induce polarization to M1 macrophages and associated complications. As perioperative management, it is an important strategy to regulate polarization and functions of macrophages during inflammatory processes. Although propofol has been found to exhibit anti-inflammatory activities in monocytes and macrophages, it is unclear whether propofol regulates the functions of M1 and M2 macrophages during inflammatory processes. This study therefore investigated the effects of propofol on human macrophage polarization. During M1 polarization, propofol suppressed the production of IL-6 and IL-1β but did not affect TNF-α production. In contrast, propofol did not affect the gene expression of M2 markers, such as IL-10, TGF-β, and CD206, during M2 polarization. Propofol was similar to the GABAA agonist muscimol in inducing nuclear translocation of nuclear factor-E2-related factor 2 (Nrf2) and inhibiting IL-6 and IL-1β, but not TNF-α, production. Knockdown of Nrf2 using siRNA significantly reduced the effect of propofol on IL-6 and IL-1β production. These results suggest that propofol prevents inflammatory responses during polarization of human M1 macrophages by suppressing the expression of IL-6 and IL-1β through the GABAA receptor and the Nrf2-mediated signal transduction pathway.

Figures

Figure 1
Figure 1
Propofol had no effect on CD86 mRNA and cell surface expression in M1 macrophages. M0 macrophages were polarized to M1 macrophages in the presence of 0.05% DMSO (solvent control) or propofol (1–5 μM). (a) qRT-PCR assays of CD86 mRNA levels. Data were normalized relative to β-actin mRNA (internal control) and presented as mean ± SD (n = 3 per group). (b) Flow cytometric analysis of CD86 surface expression on M1 macrophages treated with propofol (thick line) or DMSO (thin line) and on M1 macrophages incubated with isotype-matched control IgG and propofol (gray-filled line) or DMSO (dashed line).
Figure 2
Figure 2
Propofol reduced IL-6 and IL-1β gene expression and protein production in M1 macrophages. M0 macrophages were polarized to M1 macrophages in the presence of 0.05% DMSO (solvent control) or propofol (1–5 μM). (a–c) qRT-PCR assays of IL-6, IL-1β, and TNF-α mRNA levels in M1 macrophages. Data were normalized relative to β-actin mRNA (internal control) and presented as mean ± SD (n = 3 per group). (d–f) ELISA measurements of IL-6 (d), IL-1β (e), and TNF-α (f) secreted by M1 macrophages. Data are presented as mean ± SD (n = 3 per group). ∗P < 0.05, ∗∗P < 0.01, and ∗∗∗P < 0.001 compared with control cells by one-way ANOVA with Bonferroni's post hoc test.
Figure 3
Figure 3
Muscimol reduced IL-6 and IL-1β gene expression and protein production in M1 macrophages. M0 macrophages were polarized to M1 macrophages in the absence or presence of muscimol (100 μM). (a–d) qRT-PCR assays of IL-6, IL-1β, TNF-α, and CD86 mRNA. Data were normalized relative to β-actin mRNA (internal control) and presented as mean ± SD (n = 4 per group). (e–g) ELISA measurements of IL-6 (e), IL-1β (f), and TNF-α (g) secreted by M1 macrophages. Data are presented as mean ± SD (n = 4 per group). ∗P < 0.05 compared with control cells by Wilcoxon-Mann-Whitney test.
Figure 4
Figure 4
Propofol had no effect on IL-10, TGF-β1, and CD206 gene expression and protein production in M2 macrophages. M0 macrophages were polarized to M2 macrophages in the presence of 0.05% DMSO (solvent control) or propofol (1–5 μM). (a–c) qRT-PCR assays of IL-10, TGF-β1, and CD206 mRNA. Data were normalized relative to β-actin mRNA (internal control) and presented as mean ± SD (n = 3 per group). ELISA measurements of IL-10 (d) and TGF-β1 (e), secreted by M2 macrophages. Data are presented as mean ± SD (n = 3 per group). Statistical comparisons were analyzed using one-way ANOVA with Bonferroni's post hoc test.
Figure 5
Figure 5
Propofol and muscimol enhanced nuclear translocation of Nrf2 in M1 THP-1 cells. (a) Immunoblotting analysis of the effects of propofol (50 μM) on nuclear translocation of Nrf2. (b) Densitometric analysis of bands in (a). (c) Immunoblotting analysis of the effects of muscimol (100 μM) on nuclear translocation of Nrf2. (d) Densitometric analysis of bands in (c). Data were normalized relative to lamin C (internal control for nuclear proteins) and presented as mean ± SD of four independent experiments. ∗P < 0.05 compared with control cells by one-way ANOVA with Bonferroni's post hoc test or by the Wilcoxon-Mann-Whitney test.
Figure 6
Figure 6
siRNA knockdown of Nrf2 significantly reduced the anti-inflammatory effects of propofol in M1 THP-1 cells. (a) Immunoblotting analysis of Nrf2 expression. THP-1 cells were transfected with control nontarget or Nrf2 siRNA and treated with PMA for 72 hr. (b) Densitometric analysis of bands in (a). Data were normalized relative to β-actin (internal control) and presented as mean ± SD of four independent experiments. ∗P < 0.05 compared with control cells by the Wilcoxon-Mann-Whitney test. (c–e) Effects of Nrf2 siRNA on IL-6, IL-1β, and TNF-α production by M1 THP-1 cells in the absence or presence of propofol. PMA-differentiated siRNA-transfected cells were further polarized into M1 macrophages in the presence of 0.05% DMSO (solvent control) or propofol (50 μM). IL-6, IL-1β, and TNF-α concentrations in supernatants were measured by ELISA. ∗P < 0.05 compared with cells transfected with nontarget siRNA by one-way ANOVA with Bonferroni's post hoc test. NS = not significant.

References

    1. Mosser D. M., Edwards J. P. Exploring the full spectrum of macrophage activation. Nature Reviews Immunology. 2008;8(12):958–969. doi: 10.1038/nri2448.
    1. Lawrence T., Natoli G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nature Reviews Immunology. 2011;11(11):750–761. doi: 10.1038/nri3088.
    1. Genin M., Clement F., Fattaccioli A., Raes M., Michiels C. M1 and M2 macrophages derived from THP-1 cells differentially modulate the response of cancer cells to etoposide. BMC Cancer. 2015;15(1):p. 577. doi: 10.1186/s12885-015-1546-9.
    1. Muraille E., Leo O., Moser M. TH1/TH2 paradigm extended: macrophage polarization as an unappreciated pathogen-driven escape mechanism? Frontiers in Immunology. 2014;5:p. 603. doi: 10.3389/fimmu.2014.00603.
    1. Yuan P. Q., Tache Y. Abdominal surgery induced gastric ileus and activation of M1-like macrophages in the gastric myenteric plexus: prevention by central vagal activation in rats. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2017;313(4):G320–g329. doi: 10.1152/ajpgi.00121.2017.
    1. Murray P. J., Wynn T. A. Protective and pathogenic functions of macrophage subsets. Nature Reviews Immunology. 2011;11(11):723–737. doi: 10.1038/nri3073.
    1. Desborough J. P. The stress response to trauma and surgery. British Journal of Anaesthesia. 2000;85(1):109–117. doi: 10.1093/bja/85.1.109.
    1. Tang J., Chen X., Tu W., et al. Propofol inhibits the activation of p38 through up-regulating the expression of annexin A1 to exert its anti-inflammation effect. PLoS One. 2011;6(12, article e27890) doi: 10.1371/journal.pone.0027890.
    1. Song X. M., Wang Y. L., Li J. G., et al. Effects of propofol on pro-inflammatory cytokines and nuclear factor kappaB during polymicrobial sepsis in rats. Molecular Biology Reports. 2009;36(8):2345–2351. doi: 10.1007/s11033-009-9456-z.
    1. Meng T., Yu J., Lei Z., et al. Propofol reduces lipopolysaccharide-induced, NADPH oxidase (NOX2) mediated TNF-α and IL-6 production in macrophages. Clinical and Developmental Immunology. 2013;2013, article 325481:9. doi: 10.1155/2013/325481.
    1. Hsing C. H., Lin M. C., Choi P. C., et al. Anesthetic propofol reduces endotoxic inflammation by inhibiting reactive oxygen species-regulated Akt/IKKβ/NF-κB signaling. PLoS One. 2011;6(3, article e17598) doi: 10.1371/journal.pone.0017598.
    1. Chen R. M., Chen T. G., Chen T. L., et al. Anti-inflammatory and antioxidative effects of propofol on lipopolysaccharide-activated macrophages. Annals of the New York Academy of Sciences. 2005;1042(1):262–271. doi: 10.1196/annals.1338.030.
    1. Sanna E., Mascia M. P., Klein R. L., Whiting P. J., Biggio G., Harris R. A. Actions of the general anesthetic propofol on recombinant human GABAA receptors: influence of receptor subunits. The Journal of Pharmacology and Experimental Therapeutics. 1995;274(1):353–360.
    1. Mendu S. K., Bhandage A., Jin Z., Birnir B. Different subtypes of GABA-A receptors are expressed in human, mouse and rat T lymphocytes. PLoS One. 2012;7(8, article e42959) doi: 10.1371/journal.pone.0042959.
    1. Sanders R. D., Grover V., Goulding J., et al. Immune cell expression of GABAA receptors and the effects of diazepam on influenza infection. Journal of Neuroimmunology. 2015;282:97–103. doi: 10.1016/j.jneuroim.2015.04.001.
    1. Reyes-Garcia M. G., Hernandez-Hernandez F., Hernandez-Tellez B., Garcia-Tamayo F. GABA (A) receptor subunits RNA expression in mice peritoneal macrophages modulate their IL-6/IL-12 production. Journal of Neuroimmunology. 2007;188(1-2):64–68. doi: 10.1016/j.jneuroim.2007.05.013.
    1. Wheeler D. W., Thompson A. J., Corletto F., et al. Anaesthetic impairment of immune function is mediated via GABAA receptors. PLoS One. 2011;6(2, article e17152) doi: 10.1371/journal.pone.0017152.
    1. Jin Z., Mendu S. K., Birnir B. GABA is an effective immunomodulatory molecule. Amino Acids. 2013;45(1):87–94. doi: 10.1007/s00726-011-1193-7.
    1. Jia J., Sun Y., Hu Z., Li Y., Ruan X. Propofol inhibits the release of interleukin-6, 8 and tumor necrosis factor-α correlating with high-mobility group box 1 expression in lipopolysaccharides-stimulated RAW 264.7 cells. BMC Anesthesiology. 2017;17(1):p. 148. doi: 10.1186/s12871-017-0441-0.
    1. Martinez F. O., Gordon S., Locati M., Mantovani A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. The Journal of Immunology. 2006;177(10):7303–7311. doi: 10.4049/jimmunol.177.10.7303.
    1. Hirose K., Iwabuchi K., Shimada K., et al. Different responses to oxidized low-density lipoproteins in human polarized macrophages. Lipids in Health and Disease. 2011;10(1):p. 1. doi: 10.1186/1476-511X-10-1.
    1. Gordon S. Alternative activation of macrophages. Nature Reviews Immunology. 2003;3(1):23–35. doi: 10.1038/nri978.
    1. Daigneault M., Preston J. A., Marriott H. M., Whyte M. K. B., Dockrell D. H. The identification of markers of macrophage differentiation in PMA-stimulated THP-1 cells and monocyte-derived macrophages. PLoS One. 2010;5(1, article e8668) doi: 10.1371/journal.pone.0008668.
    1. Matsuki Y., Ichinohe T., Kaneko Y. Amnesia for electric dental pulp stimulation and picture recall test under different levels of propofol or midazolam sedation. Acta Anaesthesiologica Scandinavica. 2007;51(1):16–21. doi: 10.1111/j.1399-6576.2006.01170.x.
    1. Bhat R., Axtell R., Mitra A., et al. Inhibitory role for GABA in autoimmune inflammation. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(6):2580–2585. doi: 10.1073/pnas.0915139107.
    1. Baird L., Dinkova-Kostova A. T. The cytoprotective role of the Keap1–Nrf2 pathway. Archives of Toxicology. 2011;85(4):241–272. doi: 10.1007/s00204-011-0674-5.
    1. Yao W., Luo G., Zhu G., et al. Propofol activation of the Nrf2 pathway is associated with amelioration of acute lung injury in a rat liver transplantation model. Oxidative Medicine and Cellular Longevity. 2014;2014:9. doi: 10.1155/2014/258567.258567
    1. Shinjo T., Tanaka T., Okuda H., et al. Propofol induces nuclear localization of Nrf2 under conditions of oxidative stress in cardiac H9c2 cells. PLoS One. 2018;13(4, article e0196191) doi: 10.1371/journal.pone.0196191.
    1. Kobayashi E. H., Suzuki T., Funayama R., et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nature Communications. 2016;7(1, article 11624) doi: 10.1038/ncomms11624.
    1. Suzuki T., Motohashi H., Yamamoto M. Toward clinical application of the Keap1–Nrf2 pathway. Trends in Pharmacological Sciences. 2013;34(6):340–346. doi: 10.1016/j.tips.2013.04.005.
    1. Schildberger A., Rossmanith E., Eichhorn T., Strassl K., Weber V. Monocytes, peripheral blood mononuclear cells, and THP-1 cells exhibit different cytokine expression patterns following stimulation with lipopolysaccharide. Mediators of Inflammation. 2013;2013:10. doi: 10.1155/2013/697972.697972
    1. Moreira-Tabaka H., Peluso J., Vonesch J. L., et al. Unlike for human monocytes after LPS activation, release of TNF-α by THP-1 cells is produced by a TACE catalytically different from constitutive TACE. PLoS One. 2012;7(3, article e34184) doi: 10.1371/journal.pone.0034184.
    1. Hu J., Chen C., Ou G., et al. Nrf2 regulates the inflammatory response, including heme oxygenase-1 induction, by Mycoplasma pneumoniae lipid-associated membrane proteins in THP-1 cells. Pathogens and Disease. 2017;75(4) doi: 10.1093/femspd/ftx044.
    1. Kobayashi A., Kang M. I., Okawa H., et al. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Molecular and Cellular Biology. 2004;24(16):7130–7139. doi: 10.1128/MCB.24.16.7130-7139.2004.
    1. Wu G. J., Chen T. L., Chang C. C., Chen R. M. Propofol suppresses tumor necrosis factor-α biosynthesis in lipopolysaccharide-stimulated macrophages possibly through downregulation of nuclear factor-kappa B-mediated toll-like receptor 4 gene expression. Chemico-Biological Interactions. 2009;180(3):465–471. doi: 10.1016/j.cbi.2009.05.003.
    1. Pei Z., Wang J. Propofol attenuates LPS-induced tumor necrosis factor-α, interleukin-6 and nitric oxide expression in canine peripheral blood mononuclear cells possibly through down-regulation of nuclear factor (NF)-κB activation. Journal of Veterinary Medical Science. 2015;77(2):139–145. doi: 10.1292/jvms.14-0212.
    1. Chorley B. N., Campbell M. R., Wang X., et al. Identification of novel NRF2-regulated genes by ChIP-Seq: influence on retinoid X receptor alpha. Nucleic Acids Research. 2012;40(15):7416–7429. doi: 10.1093/nar/gks409.
    1. Helmy S. A. K., Wahby M. A. M., el-Nawaway M. The effect of anaesthesia and surgery on plasma cytokine production. Anaesthesia. 1999;54(8):733–738. doi: 10.1046/j.1365-2044.1999.00947.x.
    1. Quinton L. J., Jones M. R., Robson B. E., Mizgerd J. P. Mechanisms of the hepatic acute-phase response during bacterial pneumonia. Infection and Immunity. 2009;77(6):2417–2426. doi: 10.1128/IAI.01300-08.
    1. Sheeran P., Hall G. M. Cytokines in anaesthesia. British Journal of Anaesthesia. 1997;78(2):201–219. doi: 10.1093/bja/78.2.201.
    1. Vernooy J. H. J., Reynaert N., Wolfs T. G. A. M., et al. Rapid pulmonary expression of acute-phase reactants after local lipopolysaccharide exposure in mice is followed by an interleukin-6 mediated systemic acute-phase response. Experimental Lung Research. 2005;31(9-10):855–871. doi: 10.1080/01902140600611645.
    1. Oka Y., Murata A., Nishijima J., et al. Circulating interleukin 6 as a useful marker for predicting postoperative complications. Cytokine. 1992;4(4):298–304. doi: 10.1016/1043-4666(92)90070-8.
    1. Ohzato H., Yoshizaki K., Nishimoto N., et al. Interleukin-6 as a new indicator of inflammatory status: detection of serum levels of interleukin-6 and C-reactive protein after surgery. Surgery. 1992;111(2):201–209.

Source: PubMed

3
Sottoscrivi