Lipopolysaccharide Is Cleared from the Circulation by Hepatocytes via the Low Density Lipoprotein Receptor

Elena Topchiy, Mihai Cirstea, HyeJin Julia Kong, John H Boyd, Yingjin Wang, James A Russell, Keith R Walley, Elena Topchiy, Mihai Cirstea, HyeJin Julia Kong, John H Boyd, Yingjin Wang, James A Russell, Keith R Walley

Abstract

Sepsis is the leading cause of death in critically ill patients. While decreased Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9) function improves clinical outcomes in murine and human sepsis, the mechanisms involved have not been fully elucidated. We tested the hypothesis that lipopolysaccharide (LPS), the major Gram-negative bacteria endotoxin, is cleared from the circulation by hepatocyte Low Density Lipoprotein Receptors (LDLR)-receptors downregulated by PCSK9. We directly visualized LPS uptake and found that LPS is rapidly taken up by hepatocytes into the cell periphery. Over the course of 4 hours LPS is transported towards the cell center. We next found that clearance of injected LPS from the blood was reduced substantially in Ldlr knockout (Ldlr-/-) mice compared to wild type controls and, simultaneously, hepatic uptake of LPS was also reduced in Ldlr-/- mice. Specifically examining the role of hepatocytes, we further found that primary hepatocytes isolated from Ldlr-/- mice had greatly decreased LPS uptake. In the HepG2 immortalized human hepatocyte cell line, LDLR silencing similarly resulted in decreased LPS uptake. PCSK9 treatment reduces LDLR density on hepatocytes and, therefore, was another independent strategy to test our hypothesis. Incubation with PCSK9 reduced LPS uptake by hepatocytes. Taken together, these findings demonstrate that hepatocytes clear LPS from the circulation via the LDLR and PCSK9 regulates LPS clearance from the circulation during sepsis by downregulation of hepatic LDLR.

Conflict of interest statement

Competing Interests: The University of British Columbia has filed a provisional patent application covering aspects of this manuscript. KRW, JHB, and JAR are listed as inventors (U.S. Application Serial No. 14/399,157, Methods and Uses for Proprotein Convertase Subtilisin Kexin 9 (PCSK9) Inhibitors). KRW, JHB, and JAR have founded Cyon Therapeutics, which has licensed this intellectual property. KRW, JHB, and JAR are share holders and board members of Cyon Therapeutics. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials as there was no restriction on sharing this data.

Figures

Fig 1. Time course of LPS uptake…
Fig 1. Time course of LPS uptake by hepatocytes.
(A) For a live-imaging experiment fresh isolated primary hepatocytes were plated on 35 mm glass bottom dish precoated with collagen I (MatTek) and incubated overnight. The cells were stained with Hoechst to identify nuclei (blue) prior Alexa Fluor 488-conjugated E.coli LPS (green) addition and then immediately imaged constantly with the 63x objective using a Leica Inverted Fluorescence microscope, at 37°C and 5% CO2. (B) Time course of LPS uptake by HepG2 cells. HepG2 cells were treated with Alexa-488 FluorTM E.coli LPS (2.5 μg/ml) for 15, 30 min, 1, 2, 4 and 6 hours in media containing 20% human plasma. LPS uptake was measured by flow cytometry. Data presented as mean fluorescence intensity, mean±SEM (n = 3).
Fig 2. Ldlr knockout (Ldlr-/-) reduces plasma…
Fig 2. Ldlr knockout (Ldlr-/-) reduces plasma clearance of LPS and hepatic LPS uptake.
(A) LPS levels in plasma were measured by microplate reader 5 min, 1 and 6 hours post injection of a non-lethal dose of FITC-conjugated E.coli LPS (5 mg/kg) into tail vein of control (wild type) and Ldlr-/- mice. (B) Data are presented as mean fluorescence intensity of homogenized liver tissue detected by the microplate reader and normalized to mice injected with non-fluorescent LPS and tissue weight, mean±SEM, (n = 5). Analyzed by two-way ANOVA.
Fig 3. LPS uptake into hepatocytes is…
Fig 3. LPS uptake into hepatocytes is LDLR-dependent.
(A) Primary hepatocytes were isolated from wild type and Ldlr-/- mice, plated on 12 mm coverslips precoated with Collagen I in DMEM medium with 20% Bovine Serum, 100 units/mL penicillin, and 100 units/mL streptomycin. Cells were allowed to attach to coverslips for 6–12 hours prior the treatment with 2.5 ug/mL Alexa Fluor 488–conjugated LPS was added for 1, 6 and 24 hours total. Coverslips were fixed with 4% paraformaldehyde for 20 min and stained with DAPI to identify nuclei (blue). Images are representative of three independent experiments. (B) 2D image of primary hepatocytes isolated form WT mice and treated with 2.5 μg/mL Alexa Fluor 488–conjugated LPS for 24 hours. LPS uptake was visualized using Leica Inverted Fluorescence microscope (x63 magnification) and analyzed using VOLOCITY software (PerkinElmer Inc.).
Fig 4. HepG2 cells clear LPS in…
Fig 4. HepG2 cells clear LPS in LDLR-dependent manner.
(A) The LDLR specific and scrambled siRNAs were reversely transfected into HepG2 cells. After 24, 48 and 72 hrs, the cells were detached and stained with LDLR-PE conjugated antibody. The LDLR levels were analyzed by flow cytometry. Data presented as percent of mean fluorescence intensity from scrambled control, mean±SEM (n = 3). Highest knockdown efficiency (73%) was achieved 48 hours after transfection. (B) Effect of LDL receptor knockdown on LPS uptake. HepG2 cells were reversely transfected with siRNA targeting LDLR and control scrambled siRNA. Cells were treated with Alexa Fluor 488-conjugated LPS (2.5 μg/ml) 48 hours after transfection for 24 hours. LPS uptake was measured by flow cytometry. Data presented as mean fluorescence intensity, mean±SEM (n = 3). (C) PCSK9 incubation decreased LPS uptake by HepG2 cells. Scrambled siRNA was reversely transfected into HepG2 cells, than treated similarly as described above with PCSK9 as a complementary approach to decreasing LDLR. Data presented as mean fluorescence intensity, mean±SEM (n = 3). (D) HepG2 cells uptake LDL-labeled LPS complex via LDLR. Freshly isolated LDL form healthy donors was labeled with Alexa-488 Fluor LPS, and separated with the Sephadex column. HepG2 cells were pretreated with PCSK9 (3 μg/mL) or LDLR AB (3 μg/mL) to block the LDLR levels before adding LDL-LPS complex (50 μg/mL). The LDL-LPS uptake was analyzed by flow cytometry. Data presented as mean fluorescence intensity, mean±SEM (n = 3).
Fig 5. LPS uptake is mediated by…
Fig 5. LPS uptake is mediated by the LDLR in human hepatocytes.
(A) PCSK9 further reduced LPS uptake in Ldlr-/- cells. Primary hepatocytes were isolated from Ldlr-/- mice. Cells were allowed to attach to coverslips for 6–12 hours than treated with 2.5 ug/mL Alexa Fluor 488–conjugated LPS for 6 hours total. Coverslips were fixed with 4% paraformaldehyde for 20 min and stained with DAPI to identify nuclei (blue). LPS uptake was visualized using Leica Inverted Fluorescence microscope (x63 magnification) and analyzed with VELOCITY software. Images are representative of three independent experiments. (B) Ldlr-/- mice have increased expression of VLDLR in a liver. VLDR receptor protein expression by Western blot in wild type and Ldlr-/- mouse liver at time points 1 hour and 6 hours after injection of LPS. Data presented as mean±SEM, (***P = 0.0007, compared with control 1h; **P = 0.0026, compared with control 6h, n = 3). (C) PCSK9 further altered uptake of LPS in HepG2 cells with LDLR knockdown. HepG2 cells were reversely transfected with siRNA targeting LDLR and control scrambled siRNA. Recombinant human PCSK9 (3 μg/ml) was added 4 hours before and 4 and 19 hours after LPS treatment. Cells were treated with Alexa Fluor 488-conjugated LPS (2.5 μg/ml) 48 hours after transfection for 24 hours. LPS uptake was measured by flow cytometry. Data presented as mean fluorescence intensity, mean±SEM (n = 3).

References

    1. Russell JA. Management of sepsis. The New England journal of medicine. 2006;355(16):1699–713. .
    1. Azzam KM, Fessler MB. Crosstalk between reverse cholesterol transport and innate immunity. Trends in endocrinology and metabolism: TEM. 2012;23(4):169–78. Epub 2012/03/13. 10.1016/j.tem.2012.02.001
    1. Walley KR, Thain KR, Russell JA, Reilly MP, Meyer NJ, Ferguson JF, et al. PCSK9 is a critical regulator of the innate immune response and septic shock outcome. Science translational medicine. 2014;6(258):258ra143 Epub 2014/10/17. 10.1126/scitranslmed.3008782 .
    1. Maxwell KN, Breslow JL. Adenoviral-mediated expression of Pcsk9 in mice results in a low-density lipoprotein receptor knockout phenotype. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(18):7100–5. Epub 2004/05/01. 10.1073/pnas.0402133101
    1. Lagace TA, Curtis DE, Garuti R, McNutt MC, Park SW, Prather HB, et al. Secreted PCSK9 decreases the number of LDL receptors in hepatocytes and in livers of parabiotic mice. The Journal of clinical investigation. 2006;116(11):2995–3005. Epub 2006/11/03. 10.1172/JCI29383
    1. Walley KR, Francis GA, Opal SM, Stein EA, Russell JA, Boyd JH. The Central Role of PCSK9 in Septic Pathogen Lipid Transport and Clearance. American journal of respiratory and critical care medicine. 2015. Epub 2015/08/08. 10.1164/rccm.201505-0876CI .
    1. Levels JH, Marquart JA, Abraham PR, van den Ende AE, Molhuizen HO, van Deventer SJ, et al. Lipopolysaccharide is transferred from high-density to low-density lipoproteins by lipopolysaccharide-binding protein and phospholipid transfer protein. Infection and immunity. 2005;73(4):2321–6. Epub 2005/03/24. 10.1128/IAI.73.4.2321-2326.2005
    1. Scott MJ, Liu S, Shapiro RA, Vodovotz Y, Billiar TR. Endotoxin uptake in mouse liver is blocked by endotoxin pretreatment through a suppressor of cytokine signaling-1-dependent mechanism. Hepatology. 2009;49(5):1695–708. Epub 2009/03/20. 10.1002/hep.22839
    1. Scott MJ, Liu S, Su GL, Vodovotz Y, Billiar TR. Hepatocytes enhance effects of lipopolysaccharide on liver nonparenchymal cells through close cell interactions. Shock. 2005;23(5):453–8. Epub 2005/04/19. .
    1. Deng M, Scott MJ, Loughran P, Gibson G, Sodhi C, Watkins S, et al. Lipopolysaccharide clearance, bacterial clearance, and systemic inflammatory responses are regulated by cell type-specific functions of TLR4 during sepsis. Journal of immunology. 2013;190(10):5152–60. Epub 2013/04/09. 10.4049/jimmunol.1300496
    1. Rashid S, Curtis DE, Garuti R, Anderson NN, Bashmakov Y, Ho YK, et al. Decreased plasma cholesterol and hypersensitivity to statins in mice lacking Pcsk9. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(15):5374–9. Epub 2005/04/05. 0501652102 [pii] 10.1073/pnas.0501652102
    1. .
    1. Dinh LN, Schildbach MA, Maxwell RS, Siekhaus WJ, Balazs B, McLean W 2nd. H2O outgassing from silica-filled polysiloxane TR55. Journal of colloid and interface science. 2004;274(1):25–32. Epub 2004/05/04. 10.1016/j.jcis.2003.12.052 .
    1. Choi S, Korstanje R. Proprotein convertases in high-density lipoprotein metabolism. Biomarker research. 2013;1(1):27 Epub 2013/11/21. 10.1186/2050-7771-1-27
    1. Degrace P, Moindrot B, Mohamed I, Gresti J, Du ZY, Chardigny JM, et al. Upregulation of liver VLDL receptor and FAT/CD36 expression in LDLR-/- apoB100/100 mice fed trans-10,cis-12 conjugated linoleic acid. Journal of lipid research. 2006;47(12):2647–55. Epub 2006/09/08. 10.1194/jlr.M600140-JLR200 .
    1. Mimura Y, Sakisaka S, Harada M, Sata M, Tanikawa K. Role of hepatocytes in direct clearance of lipopolysaccharide in rats. Gastroenterology. 1995;109(6):1969–76. Epub 1995/12/01. .
    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. Proceedings of the Society for Experimental Biology and Medicine Society for Experimental Biology and Medicine. 1993;202(2):153–8. Epub 1993/02/01. .
    1. Wright SD, Ramos RA, Tobias PS, Ulevitch RJ, Mathison JC. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science. 1990;249(4975):1431–3. Epub 1990/09/21. .
    1. American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference: definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Critical care medicine. 1992;20(6):864–74. Epub 1992/06/01. .
    1. Abazov VM, Abbott B, Abolins M, Acharya BS, Adams M, Adams T, et al. Search for associated higgs boson production WH—>WWW*—>l+/-nul'+/-nu'+X in pp collisions at square root s = 1.96 Te V. Physical review letters. 2006;97(15):151804 Epub 2006/12/13. .
    1. van Oosten M, van de Bilt E, van Berkel TJ, Kuiper J. New scavenger receptor-like receptors for the binding of lipopolysaccharide to liver endothelial and Kupffer cells. Infection and immunity. 1998;66(11):5107–12. Epub 1998/10/24.
    1. Li XY, Zhang C, Wang H, Ji YL, Wang SF, Zhao L, et al. Tumor necrosis factor alpha partially contributes to lipopolysaccharide-induced downregulation of CYP3A in fetal liver: its repression by a low dose LPS pretreatment. Toxicology letters. 2008;179(2):71–7. Epub 2008/05/27. 10.1016/j.toxlet.2008.04.005 .
    1. Liu S, Salyapongse AN, Geller DA, Vodovotz Y, Billiar TR. Hepatocyte toll-like receptor 2 expression in vivo and in vitro: role of cytokines in induction of rat TLR2 gene expression by lipopolysaccharide. Shock. 2000;14(3):361–5.
    1. Gautier T, Lagrost L. Plasma PLTP (phospholipid-transfer protein): an emerging role in 'reverse lipopolysaccharide transport' and innate immunity. Biochemical Society transactions. 2011;39(4):984–8. Epub 2011/07/27. 10.1042/BST0390984 .
    1. Khovidhunkit W, Kim MS, Memon RA, Shigenaga JK, Moser AH, Feingold KR, et al. Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. Journal of lipid research. 2004;45(7):1169–96.
    1. Levels JH, Abraham PR, van Barreveld EP, Meijers JC, van Deventer SJ. Distribution and kinetics of lipoprotein-bound lipoteichoic acid. Infection and immunity. 2003;71(6):3280–4. Epub 2003/05/23.
    1. Victorov AV, Medvedeva NV, Gladkaya EM, Morozkin AD, Podrez EA, Kosykh VA, et al. Composition and structure of lipopolysaccharide-human plasma low density lipoprotein complex. Analytical ultracentrifugation,31P-NMR, ESR and fluorescence spectroscopy studies. Biochimica et Biophysica Acta (BBA)—Biomembranes. 1989;984(1):119–27. 10.1016/0005-2736(89)90351-9
    1. Murch O, Collin M, Hinds CJ, Thiemermann C. Lipoproteins in inflammation and sepsis. I. Basic science. Intensive care medicine. 2007;33(1):13–24. Epub 2006/11/10. 10.1007/s00134-006-0432-y .
    1. Vreugdenhil AC, Rousseau CH, Hartung T, Greve JW, van 't Veer C, Buurman WA. Lipopolysaccharide (LPS)-binding protein mediates LPS detoxification by chylomicrons. Journal of immunology. 2003;170(3):1399–405. Epub 2003/01/23. .
    1. Liao W, Rudling M, Angelin B. Endotoxin suppresses mouse hepatic low-density lipoprotein-receptor expression via a pathway independent of the toll-like receptor 4. Hepatology. 1999;30(5):1252–6. Epub 1999/10/26. 10.1002/hep.510300524 .
    1. van Leeuwen HJ, Heezius EC, Dallinga GM, van Strijp JA, Verhoef J, van Kessel KP. Lipoprotein metabolism in patients with severe sepsis. Critical care medicine. 2003;31(5):1359–66.
    1. Bray N. Sepsis: PCSK9 blockade helps clear pathogenic lipids. Nat Rev Drug Discov. 2014;13(12):886–7. 10.1038/nrd4492
    1. Maxwell KN, Fisher EA, Breslow JL. Overexpression of PCSK9 accelerates the degradation of the LDLR in a post-endoplasmic reticulum compartment. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(6):2069–74. Epub 2005/01/29. 10.1073/pnas.0409736102
    1. Liao W, Rudling M, Angelin B. Endotoxin suppresses rat hepatic low-density lipoprotein receptor expression. Biochem J. 1996;313(Pt 3):873–8.
    1. Li XY, Zhang C, Wang SF, Ji YL, Wang H, Zhao L, et al. Maternally administered lipopolysaccharide (LPS) upregulates the expression of heme oxygenase-1 in fetal liver: the role of reactive oxygen species. Toxicology letters. 2008;176(3):169–77. Epub 2008/02/05. 10.1016/j.toxlet.2007.10.010 .
    1. Laugerette F, Furet JP, Debard C, Daira P, Loizon E, Geloen A, et al. Oil composition of high-fat diet affects metabolic inflammation differently in connection with endotoxin receptors in mice. American journal of physiology Endocrinology and metabolism. 2012;302(3):E374–86. Epub 2011/11/19. 10.1152/ajpendo.00314.2011 .
    1. Clark RW, Cunningham D, Cong Y, Subashi TA, Tkalcevic GT, Lloyd DB, et al. Assessment of cholesteryl ester transfer protein inhibitors for interaction with proteins involved in the immune response to infection. Journal of lipid research. 2010;51(5):967–74. Epub 2009/12/08. 10.1194/jlr.M002295
    1. Poirier S, Mayer G, Benjannet S, Bergeron E, Marcinkiewicz J, Nassoury N, et al. The proprotein convertase PCSK9 induces the degradation of low density lipoprotein receptor (LDLR) and its closest family members VLDLR and ApoER2. The Journal of biological chemistry. 2008;283(4):2363–72. Epub 2007/11/28. M708098200 [pii] 10.1074/jbc.M708098200 .
    1. Roubtsova A, Munkonda MN, Awan Z, Marcinkiewicz J, Chamberland A, Lazure C, et al. Circulating proprotein convertase subtilisin/kexin 9 (PCSK9) regulates VLDLR protein and triglyceride accumulation in visceral adipose tissue. Arterioscler Thromb Vasc Biol. 2011;31(4):785–91. 10.1161/ATVBAHA.110.220988
    1. Tiebel O, Oka K, Robinson K, Sullivan M, Martinez J, Nakamuta M, et al. Mouse very low-density lipoprotein receptor (VLDLR): gene structure, tissue-specific expression and dietary and developmental regulation. Atherosclerosis. 1999;145(2):239–51.
    1. Abaza R, Rashid MG, Sferra JJ. Obstructive uropathy from giant inguinal bladder and ureteral herniation. Journal of the American College of Surgeons. 2005;201(2):314 Epub 2005/07/26. 10.1016/j.jamcollsurg.2004.12.026 .
    1. Abazov VM, Abbott B, Abolins M, Acharya BS, Adams M, Adams T, et al. Measurement of the Bs(0) lifetime using semileptonic decays. Physical review letters. 2006;97(24):241801 Epub 2007/02/07. .
    1. Nesseler N, Launey Y, Aninat C, Morel F, Malledant Y, Seguin P. Clinical review: The liver in sepsis. Critical care. 2012;16(5):235 Epub 2012/11/09. 10.1186/cc11381

Source: PubMed

3
구독하다