Low-density lipoprotein (LDL)-dependent uptake of Gram-positive lipoteichoic acid and Gram-negative lipopolysaccharide occurs through LDL receptor

Peter M Grin, Dhruva J Dwivedi, Kevin M Chathely, Bernardo L Trigatti, Annik Prat, Nabil G Seidah, Patricia C Liaw, Alison E Fox-Robichaud, Peter M Grin, Dhruva J Dwivedi, Kevin M Chathely, Bernardo L Trigatti, Annik Prat, Nabil G Seidah, Patricia C Liaw, Alison E Fox-Robichaud

Abstract

Lipoteichoic acid (LTA) and lipopolysaccharide (LPS) are bacterial lipids that stimulate pro-inflammatory cytokine production, thereby exacerbating sepsis pathophysiology. Proprotein convertase subtilisin/kexin type 9 (PCSK9) negatively regulates uptake of cholesterol by downregulating hepatic lipoprotein receptors, including low-density lipoprotein (LDL) receptor (LDLR) and possibly LDLR-related protein-1 (LRP1). PCSK9 also negatively regulates Gram-negative LPS uptake by hepatocytes, however this mechanism is not completely characterized and mechanisms of Gram-positive LTA uptake are unknown. Therefore, our objective was to elucidate the mechanisms through which PCSK9 regulates uptake of LTA and LPS by investigating the roles of lipoproteins and lipoprotein receptors. Here we show that plasma PCSK9 concentrations increase transiently over time in septic and non-septic critically ill patients, with highly similar profiles over 14 days. Using flow cytometry, we demonstrate that PCSK9 negatively regulates LDLR-mediated uptake of LTA and LPS by HepG2 hepatocytes through an LDL-dependent mechanism, whereas LRP1 and high-density lipoprotein do not contribute to this uptake pathway. Bacterial lipid uptake by hepatocytes was not associated with cytokine production or hepatocellular injury. In conclusion, our study characterizes an LDL-dependent and LDLR-mediated bacterial lipid uptake pathway regulated by PCSK9, and provides evidence in support of PCSK9 inhibition as a potential therapeutic strategy for sepsis.

Trial registration: ClinicalTrials.gov NCT01355042.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Plasma PCSK9 concentrations in septic and non-septic ICU patients over time compared to healthy controls. Plasma for healthy controls was collected on a single day (mean PCSK9 concentrations of 146.4 ± 13.4 ng/mL). Data are expressed as mean ± SEM, *p 

Figure 2

Time-course and dose-response of LTA…

Figure 2

Time-course and dose-response of LTA uptake by HepG2 cells over 24 h. Uptake…

Figure 2
Time-course and dose-response of LTA uptake by HepG2 cells over 24 h. Uptake of fluorescent LTA by cells cultured in 20% normal human serum was visualized through fluorescence microscopy in untreated cells (A) or in cells treated with 10 μg/mL of BODIPY 630/650-LTA for 2 h (B), 6 h (C), or 24 h (D), or measured via flow cytometry for cells cultured in either 20% normal serum or lipoprotein-deficient serum (prepared in laboratory) and treated with increasing concentrations of fluorescent LTA over 24 h (E). Nuclei are stained in blue with DAPI, and BODIPY 630/650-LTA appears red (200× magnification). Data are representative of 3 experiments (AD), and are expressed as mean ± SEM from 3 experiments (E). ***p < 0.001 by two-way ANOVA. MFI, mean fluorescence intensity.

Figure 3

PCSK9 regulates lipoprotein-dependent uptake of…

Figure 3

PCSK9 regulates lipoprotein-dependent uptake of LTA and LPS by HepG2 cells through LDLR,…

Figure 3
PCSK9 regulates lipoprotein-dependent uptake of LTA and LPS by HepG2 cells through LDLR, not LRP1. HepG2 cells were cultured in 20% normal human serum (A,C) or (commercially available) lipoprotein-deficient human serum (B,D) and were pre-treated with 2.5 μg/mL of recombinant human PCSK9 or vehicle control at 6 h before, as well as anti-LDLR, anti-LRP1, or control IgG antibodies at 2 h before treatment with BODIPY 630/650-LTA (10 μg/mL; A,B) or AlexaFluor 488-LPS (2.5 μg/mL; C,D) for 24 h. Data were collected from 4–5 experiments at 24 hours after LPS or LTA treatment using flow cytometry, and are expressed as geometric mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 by one-way ANOVA. MFI, mean fluorescence intensity.

Figure 4

Dose-response of LDL or HDL…

Figure 4

Dose-response of LDL or HDL add-back to normal or lipoprotein-deficient serum on the…

Figure 4
Dose-response of LDL or HDL add-back to normal or lipoprotein-deficient serum on the uptake of LTA and LPS by HepG2 cells. Cells were cultured in 20% lipoprotein-deficient serum (prepared in laboratory) and treated with 2.5 μg/mL of BODIPY 630/650-LTA (A) or 2.5 μg/mL of AlexaFluor 488-LPS (B) over 24 h in the presence of increasing concentrations of LDL or HDL. Data are shown as mean ± SEM from 3–4 experiments. **p < 0.01, ***p < 0.001 vs. 0 μg/mL of LDL by one-way ANOVA. LPDS, lipoprotein-deficient serum; MFI, mean fluorescence intensity.

Figure 5

LDL-dependent uptake of LTA and…

Figure 5

LDL-dependent uptake of LTA and LPS by HepG2 cells is mediated by LDLR.…

Figure 5
LDL-dependent uptake of LTA and LPS by HepG2 cells is mediated by LDLR. Cells were cultured in 20% normal human serum or donor-matched lipoprotein-deficient serum (prepared in laboratory), with or without 100 μg/mL of LDL or HDL, and treated with anti-LDLR antibody or control IgG for two hours prior to 24 h treatment with 2.5 μg/mL of BODIPY 630/650-LTA (A) or AlexaFluor 488-LPS (B). Data are presented as geometric mean fluorescence intensity ± SEM from 3–4 experiments, and were normalized by subtracting background auto-fluorescence of untreated cells. Different letters indicate significant differences between groups with p < 0.01 by ANOVA. MFI, mean fluorescence intensity.
Figure 2
Figure 2
Time-course and dose-response of LTA uptake by HepG2 cells over 24 h. Uptake of fluorescent LTA by cells cultured in 20% normal human serum was visualized through fluorescence microscopy in untreated cells (A) or in cells treated with 10 μg/mL of BODIPY 630/650-LTA for 2 h (B), 6 h (C), or 24 h (D), or measured via flow cytometry for cells cultured in either 20% normal serum or lipoprotein-deficient serum (prepared in laboratory) and treated with increasing concentrations of fluorescent LTA over 24 h (E). Nuclei are stained in blue with DAPI, and BODIPY 630/650-LTA appears red (200× magnification). Data are representative of 3 experiments (AD), and are expressed as mean ± SEM from 3 experiments (E). ***p < 0.001 by two-way ANOVA. MFI, mean fluorescence intensity.
Figure 3
Figure 3
PCSK9 regulates lipoprotein-dependent uptake of LTA and LPS by HepG2 cells through LDLR, not LRP1. HepG2 cells were cultured in 20% normal human serum (A,C) or (commercially available) lipoprotein-deficient human serum (B,D) and were pre-treated with 2.5 μg/mL of recombinant human PCSK9 or vehicle control at 6 h before, as well as anti-LDLR, anti-LRP1, or control IgG antibodies at 2 h before treatment with BODIPY 630/650-LTA (10 μg/mL; A,B) or AlexaFluor 488-LPS (2.5 μg/mL; C,D) for 24 h. Data were collected from 4–5 experiments at 24 hours after LPS or LTA treatment using flow cytometry, and are expressed as geometric mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 by one-way ANOVA. MFI, mean fluorescence intensity.
Figure 4
Figure 4
Dose-response of LDL or HDL add-back to normal or lipoprotein-deficient serum on the uptake of LTA and LPS by HepG2 cells. Cells were cultured in 20% lipoprotein-deficient serum (prepared in laboratory) and treated with 2.5 μg/mL of BODIPY 630/650-LTA (A) or 2.5 μg/mL of AlexaFluor 488-LPS (B) over 24 h in the presence of increasing concentrations of LDL or HDL. Data are shown as mean ± SEM from 3–4 experiments. **p < 0.01, ***p < 0.001 vs. 0 μg/mL of LDL by one-way ANOVA. LPDS, lipoprotein-deficient serum; MFI, mean fluorescence intensity.
Figure 5
Figure 5
LDL-dependent uptake of LTA and LPS by HepG2 cells is mediated by LDLR. Cells were cultured in 20% normal human serum or donor-matched lipoprotein-deficient serum (prepared in laboratory), with or without 100 μg/mL of LDL or HDL, and treated with anti-LDLR antibody or control IgG for two hours prior to 24 h treatment with 2.5 μg/mL of BODIPY 630/650-LTA (A) or AlexaFluor 488-LPS (B). Data are presented as geometric mean fluorescence intensity ± SEM from 3–4 experiments, and were normalized by subtracting background auto-fluorescence of untreated cells. Different letters indicate significant differences between groups with p < 0.01 by ANOVA. MFI, mean fluorescence intensity.

References

    1. Singer M, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3) JAMA. 2016;315:801–810. doi: 10.1001/jama.2016.0287.
    1. Dellinger RP, et al. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock, 2012. Intensive Care Med. 2013;39:165–228. doi: 10.1007/s00134-012-2769-8.
    1. Stoller J, et al. Epidemiology of severe sepsis: 2008–2012. J Crit Care. 2016;31:58–62. doi: 10.1016/j.jcrc.2015.09.034.
    1. Mayr FB, Yende S, Angus DC. Epidemiology of severe sepsis. Virulence. 2014;5:4–11. doi: 10.4161/viru.27372.
    1. Fleischmann C, et al. Assessment of Global Incidence and Mortality of Hospital-treated Sepsis. Current Estimates and Limitations. Am J Respir Crit Care Med. 2016;193:259–272. doi: 10.1164/rccm.201504-0781OC.
    1. Oviedo-Boyso J, Bravo-Patino A, Baizabal-Aguirre VM. Collaborative action of Toll-like and NOD-like receptors as modulators of the inflammatory response to pathogenic bacteria. Mediators Inflamm. 2014;2014:432785. doi: 10.1155/2014/432785.
    1. Chow JC, Young DW, Golenbock DT, Christ WJ, Gusovsky F. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. J Biol Chem. 1999;274:10689–10692. doi: 10.1074/jbc.274.16.10689.
    1. Schwandner R, Dziarski R, Wesche H, Rothe M, Kirschning CJ. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2. J Biol Chem. 1999;274:17406–17409. doi: 10.1074/jbc.274.25.17406.
    1. Nguyen TTN, et al. Phosphatidylinositol 4-phosphate 5-kinase alpha contributes to Toll-like receptor 2-mediated immune responses in microglial cells stimulated with lipoteichoic acid. Cell Signal. 2017;38:159–170. doi: 10.1016/j.cellsig.2017.07.009.
    1. Tak PP, Firestein GS. NF-kappaB: a key role in inflammatory diseases. J Clin Invest. 2001;107:7–11. doi: 10.1172/JCI11830.
    1. Fox-Robichaud A, Kubes P. Molecular mechanisms of tumor necrosis factor alpha-stimulated leukocyte recruitment into the murine hepatic circulation. Hepatology. 2000;31:1123–1127. doi: 10.1053/he.2000.6961.
    1. Kowalewska PM, Patrick AL, Fox-Robichaud AE. Innate immunity of the liver microcirculation. Cell Tissue Res. 2011;343:85–96. doi: 10.1007/s00441-010-1058-5.
    1. Ondiveeran HK, Fox-Robichaud AE. Pentastarch in a balanced solution reduces hepatic leukocyte recruitment in early sepsis. Microcirculation. 2004;11:679–687. doi: 10.1080/10739680490517712.
    1. Eipel C, Bordel R, Nickels RM, Menger MD, Vollmar B. Impact of leukocytes and platelets in mediating hepatocyte apoptosis in a rat model of systemic endotoxemia. Am J Physiol Gastrointest Liver Physiol. 2004;286:G769–776. doi: 10.1152/ajpgi.00275.2003.
    1. Wittmann I, et al. Murine bactericidal/permeability-increasing protein inhibits the endotoxic activity of lipopolysaccharide and gram-negative bacteria. J Immunol. 2008;180:7546–7552. doi: 10.4049/jimmunol.180.11.7546.
    1. Vreugdenhil AC, et al. Lipopolysaccharide (LPS)-binding protein mediates LPS detoxification by chylomicrons. J Immunol. 2003;170:1399–1405. doi: 10.4049/jimmunol.170.3.1399.
    1. Hailman E, Albers JJ, Wolfbauer G, Tu AY, Wright SD. Neutralization and transfer of lipopolysaccharide by phospholipid transfer protein. J Biol Chem. 1996;271:12172–12178. doi: 10.1074/jbc.271.21.12172.
    1. Levels JH, et al. Lipopolysaccharide is transferred from high-density to low-density lipoproteins by lipopolysaccharide-binding protein and phospholipid transfer protein. Infect Immun. 2005;73:2321–2326. doi: 10.1128/IAI.73.4.2321-2326.2005.
    1. Wurfel MM, Wright SD. Lipopolysaccharide-binding protein and soluble CD14 transfer lipopolysaccharide to phospholipid bilayers: preferential interaction with particular classes of lipid. J Immunol. 1997;158:3925–3934.
    1. Harris HW, Brady SE, Rapp JH. Hepatic endosomal trafficking of lipoprotein-bound endotoxin in rats. J Surg Res. 2002;106:188–195. doi: 10.1006/jsre.2002.6413.
    1. Levels JH, Abraham PR, van den Ende A, van Deventer SJ. Distribution and kinetics of lipoprotein-bound endotoxin. Infect Immun. 2001;69:2821–2828. doi: 10.1128/IAI.69.5.2821-2828.2001.
    1. Levels JH, Abraham PR, van Barreveld EP, Meijers JC, van Deventer SJ. Distribution and kinetics of lipoprotein-bound lipoteichoic acid. Infect Immun. 2003;71:3280–3284. doi: 10.1128/IAI.71.6.3280-3284.2003.
    1. Maxwell KN, Breslow JL. Adenoviral-mediated expression of Pcsk9 in mice results in a low-density lipoprotein receptor knockout phenotype. Proc Natl Acad Sci USA. 2004;101:7100–7105. doi: 10.1073/pnas.0402133101.
    1. Canuel M, et al. Proprotein convertase subtilisin/kexin type 9 (PCSK9) can mediate degradation of the low density lipoprotein receptor-related protein 1 (LRP-1) PLoS One. 2013;8:e64145. doi: 10.1371/journal.pone.0064145.
    1. Seidah NG, et al. The secretory proprotein convertase neural apoptosis-regulated convertase 1 (NARC-1): liver regeneration and neuronal differentiation. Proc Natl Acad Sci USA. 2003;100:928–933. doi: 10.1073/pnas.0335507100.
    1. Abifadel M, et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet. 2003;34:154–156. doi: 10.1038/ng1161.
    1. Benjannet S, et al. NARC-1/PCSK9 and its natural mutants: zymogen cleavage and effects on the low density lipoprotein (LDL) receptor and LDL cholesterol. J Biol Chem. 2004;279:48865–48875. doi: 10.1074/jbc.M409699200.
    1. Walley KR, et al. PCSK9 is a critical regulator of the innate immune response and septic shock outcome. Sci Transl Med. 2014;6:258ra143. doi: 10.1126/scitranslmed.3008782.
    1. Dwivedi DJ, et al. Differential Expression of PCSK9 Modulates Infection, Inflammation, and Coagulation in a Murine Model of Sepsis. Shock. 2016;46:672–680. doi: 10.1097/SHK.0000000000000682.
    1. Topchiy E, et al. Lipopolysaccharide Is Cleared from the Circulation by Hepatocytes via the Low Density Lipoprotein Receptor. PLoS One. 2016;11:e0155030. doi: 10.1371/journal.pone.0155030.
    1. Brown MS, Ho YK, Goldstein JL. The low-density lipoprotein pathway in human fibroblasts: relation between cell surface receptor binding and endocytosis of low-density lipoprotein. Ann N Y Acad Sci. 1976;275:244–257. doi: 10.1111/j.1749-6632.1976.tb43358.x.
    1. Klop B, et al. Differential complement activation pathways promote C3b deposition on native and acetylated LDL thereby inducing lipoprotein binding to the complement receptor 1. J Biol Chem. 2014;289:35421–35430. doi: 10.1074/jbc.M114.573840.
    1. Mikhailenko I, et al. Recognition of alpha 2-macroglobulin by the low density lipoprotein receptor-related protein requires the cooperation of two ligand binding cluster regions. J Biol Chem. 2001;276:39484–39491. doi: 10.1074/jbc.M104382200.
    1. Sankaranarayanan S, et al. Serum albumin acts as a shuttle to enhance cholesterol efflux from cells. J Lipid Res. 2013;54:671–676. doi: 10.1194/jlr.M031336.
    1. Rohrl C, Eigner K, Fruhwurth S, Stangl H. Bile acids reduce endocytosis of high-density lipoprotein (HDL) in HepG2 cells. PLoS One. 2014;9:e102026. doi: 10.1371/journal.pone.0102026.
    1. Brown SA, Via DP, Gotto AM, Jr., Bradley WA, Gianturco SH. Apolipoprotein E-mediated binding of hypertriglyceridemic very low density lipoproteins to isolated low density lipoprotein receptors detected by ligand blotting. Biochem Biophys Res Commun. 1986;139:333–340. doi: 10.1016/S0006-291X(86)80118-8.
    1. Sasamoto K, et al. Changes in apolipoprotein E-containing high-density lipoprotein (HDL) have little impact on HDL-cholesterol measurements using homogeneous assays in normolipidemic and dyslipidemic subjects. Clin Chim Acta. 2017;470:56–63. doi: 10.1016/j.cca.2017.04.018.
    1. Guo L, Zheng Z, Ai J, Huang B, Li XA. Hepatic scavenger receptor BI protects against polymicrobial-induced sepsis through promoting LPS clearance in mice. J Biol Chem. 2014;289:14666–14673. doi: 10.1074/jbc.M113.537258.
    1. Berger JM, Loza Valdes A, Gromada J, Anderson N, Horton JD. Inhibition of PCSK9 does not improve lipopolysaccharide-induced mortality in mice. J Lipid Res. 2017;58:1661–1669. doi: 10.1194/jlr.M076844.
    1. Boyd JH, et al. Increased Plasma PCSK9 Levels Are Associated with Reduced Endotoxin Clearance and the Development of Acute Organ Failures during Sepsis. J Innate Immun. 2016;8:211–220. doi: 10.1159/000442976.
    1. Hsu KH, Ghanta VK, Hiramoto RN. Immunosuppressive effect of mouse serum lipoproteins. I. In vitro studies. J Immunol. 1981;126:1909–1913.
    1. Camus MC, Chapman MJ, Forgez P, Laplaud PM. Distribution and characterization of the serum lipoproteins and apoproteins in the mouse, Mus musculus. J Lipid Res. 1983;24:1210–1228.
    1. Yang D, et al. Kukoamine B promotes TLR4-independent lipopolysaccharide uptake in murine hepatocytes. Oncotarget. 2016;7:57498–57513.
    1. Gutierrez-Ruiz MC, et al. Cytokine response and oxidative stress produced by ethanol, acetaldehyde and endotoxin treatment in HepG2 cells. Isr Med Assoc J. 2001;3:131–136.
    1. Gomez-Quiroz L, et al. Interleukin 8 response and oxidative stress in HepG2 cells treated with ethanol, acetaldehyde or lipopolysaccharide. Hepatol Res. 2003;26:134–141. doi: 10.1016/S1386-6346(03)00010-X.
    1. Agarwal S, Piesco NP, Johns LP, Riccelli AE. Differential expression of IL-1 beta, TNF-alpha, IL-6, and IL-8 in human monocytes in response to lipopolysaccharides from different microbes. J Dent Res. 1995;74:1057–1065. doi: 10.1177/00220345950740040501.
    1. Guerville M, Boudry G. Gastrointestinal and hepatic mechanisms limiting entry and dissemination of lipopolysaccharide into the systemic circulation. Am J Physiol Gastrointest Liver Physiol. 2016;311:G1–G15. doi: 10.1152/ajpgi.00098.2016.
    1. Herias MV, Midtvedt T, Hanson LA, Wold AE. Role of Escherichia coli P fimbriae in intestinal colonization in gnotobiotic rats. Infect Immun. 1995;63:4781–4789.
    1. Callery MP, Mangino MJ, Kamei T, Flye MW. Interleukin-6 production by endotoxin-stimulated Kupffer cells is regulated by prostaglandin E2. J Surg Res. 1990;48:523–527. doi: 10.1016/0022-4804(90)90224-P.
    1. Overland G, et al. Lipoteichoic acid is a potent inducer of cytokine production in rat and human Kupffer cells in vitro. Surg Infect (Larchmt) 2003;4:181–191. doi: 10.1089/109629603766956979.
    1. Koo DJ, Chaudry IH, Wang P. Kupffer cells are responsible for producing inflammatory cytokines and hepatocellular dysfunction during early sepsis. J Surg Res. 1999;83:151–157. doi: 10.1006/jsre.1999.5584.
    1. Strnad P, Tacke F, Koch A, Trautwein C. Liver - guardian, modifier and target of sepsis. Nat Rev Gastroenterol Hepatol. 2017;14:55–66. doi: 10.1038/nrgastro.2016.168.
    1. West MA, Keller GA, Hyland BJ, Cerra FB, Simmons RL. Hepatocyte function in sepsis: Kupffer cells mediate a biphasic protein synthesis response in hepatocytes after exposure to endotoxin or killed Escherichia coli. Surgery. 1985;98:388–395.
    1. Treon SP, Thomas P, Broitman SA. Lipopolysaccharide (LPS) processing by Kupffer cells releases a modified LPS with increased hepatocyte binding and decreased tumor necrosis factor-alpha stimulatory capacity. Proc Soc Exp Biol Med. 1993;202:153–158. doi: 10.3181/00379727-202-43521.
    1. Barlage S, et al. Changes in HDL-associated apolipoproteins relate to mortality in human sepsis and correlate to monocyte and platelet activation. Intensive Care Med. 2009;35:1877–1885. doi: 10.1007/s00134-009-1609-y.
    1. Guirgis FW, et al. Cholesterol levels and long-term rates of community-acquired sepsis. Crit Care. 2016;20:408. doi: 10.1186/s13054-016-1579-8.
    1. Dwivedi DJ, et al. Prognostic utility and characterization of cell-free DNA in patients with severe sepsis. Crit Care. 2012;16:R151. doi: 10.1186/cc11466.
    1. Dubuc G, et al. A new method for measurement of total plasma PCSK9: clinical applications. J Lipid Res. 2010;51:140–149. doi: 10.1194/jlr.M900273-JLR200.
    1. Krieger M, Brown MS, Goldstein JL. Isolation of Chinese hamster cell mutants defective in the receptor-mediated endocytosis of low density lipoprotein. J Mol Biol. 1981;150:167–184. doi: 10.1016/0022-2836(81)90447-2.

Source: PubMed

3
Tilaa