Proteolytic inactivation of nuclear alarmin high-mobility group box 1 by complement protease C1s during apoptosis

J G Yeo, J Leong, T Arkachaisri, Y Cai, B H D Teo, J H T Tan, L Das, J Lu, J G Yeo, J Leong, T Arkachaisri, Y Cai, B H D Teo, J H T Tan, L Das, J Lu

Abstract

Effective clearance of apoptotic cells by phagocytes prevents the release of intracellular alarmins and manifestation of autoimmunity. This prompt efferocytosis is complemented by intracellular proteolytic degradation that occurs within the apoptotic cells and in the efferosome of the phagocytes. Although the role of extracellular proteases in apoptotic cells clearance is unknown, the strong association of congenital C1s deficiency with Systemic Lupus Erythematosus highlights the protective nature that this extracellular protease has against autoimmunity. The archetypical role of serine protease C1s as the catalytic arm of C1 complex (C1qC1r2C1s2) involve in the propagation of the classical complement pathway could not provide the biological basis for this association. However, a recent observation of the ability of C1 complex to cleave a spectrum of intracellular cryptic targets exposed during apoptosis provides a valuable insight to the underlying protective mechanism. High-mobility group box 1 (HMGB1), an intracellular alarmin that is capable of inducing the formation of antinuclear autoantibodies and causes lupus-like conditions in mice, is identified as a novel potential target by bioinformatics analysis. This is verified experimentally with C1s, both in its purified and physiological form as C1 complex, cleaving HMGB1 into defined fragments of 19 and 12 kDa. This cleavage diminishes HMGB1 ability to enhance lipopolysaccharide mediated pro-inflammatory cytokines production from monocytes, macrophages and dendritic cells. Further mass spectrometric analysis of the C1 complex treated apoptotic cellular proteins demonstrated additional C1s substrates and revealed the complementary role of C1s in apoptotic cells clearance through the proteolytic cleavage of intracellular alarmins and autoantigens. C1 complex may have evolved as, besides the bacteriolytic arm of antibodies in which it activates the complement cascade, a tissue renewal mechanism that reduces the immunogenicity of apoptotic tissue debris and decreases the likelihood of autoimmunity.

Figures

Figure 1
Figure 1
HMGB1 is cleavable by C1s. (a) The PoPS model for predicting C1s substrate sequences. The C1s protease active site contains eight contiguous pockets or subsites each having a unique structural and chemical property and preferred specific amino acid residues (Single letter amino acid code is used). These amino acid residues were experimentally determined previously. The subsites display different importance with varying specificity except for the A4 position where an arginine, which is conserved in C4, C2 and C1-INH, is considered an essential residue. Position of the `scissile bond’ is indicated with triangle. (b) Predicted C1s cleavage sites on HMGB1. The HMGB1 sequence is presented and the A box and B-box are shaded. The C-terminal acidic tail is underlined. Where secondary structures are predicted, these are indicated under the sequence. `-’, lack of secondary structure. The 3 inverted triangles indicate predicted C1s cleavage sites with PoPS scores (Arg70, Arg97 and Arg163). (c) Purified rHMGB1 was cleaved by purified sC1s. RHMGB1 produced in mouse myeloma cells (10 μg/ml), serum C4 (10 μg/ml), and BSA (5 μg/ml) were incubated overnight at 37 °C with activated sC1s (11 μg/ml). The samples were separated by SDS-PAGE on 12.5% (w/v) gels under reducing conditions and visualized by silver staining. The three C4 subunit chains (α, β and γ) are labeled. The cleaved C4 α chain was labeled α’. After C1 digestion of rHMGB1, a fragment was generated (F1). (d) Two HMGB1 fragments were detected after C1s digestion by western blotting (F1 and F2). Western blotting was performed using a rabbit polyclonal anti-HMGB1 antibody. (e) C1 complex (C1 cplx) cleaved HMGB1 more effectively than C1s but both generated two similar fragments. In this experiment, His-HMGB1 expressed in bacteria E. coli (20 μg/ml) was incubated overnight at 37 °C with C1 complex and activated sC1s at the concentrations indicated. The reactions were separated by SDS-PAGE on 15% (w/v) gels under reducing conditions and western blotting was performed using the polyclonal rabbit anti-HMGB1 antibody. F1 and F2 marked the HMGB1 fragments after cleavage.
Figure 2
Figure 2
C1 and C1s cleavage of HMGB1 released from apoptotic cells. (a) DNA laddering of UV-irradiated Jurkat cells. Cells were, after UV irradiation, cultured for 4 h in serum-free medium (post UV). As control, cells were cultured for 4 h without prior UV irradiation (control). Extracted DNA was examined on 1.2% (w/v) agarose gels and DNA was visualized using GelGreen (Biotium, Inc., Hayward, CA, USA) (b) HMGB1 release from apoptotic cells. After UV irradiation, Jurkat cells were cultured for a series of time periods up to 48 h. After centrifugation, supernatants were harvested and subjected to SDS-PAGE on 12.5% (w/v) gels and western blotting using a rabbit polyclonal anti-HMGB1 antibody. (c) C1 degradation of HMGB1 released from apoptotic cells. In this experiment, Jurkat cells were harvested 2.5 h after UV irradiation and, after mixing, divided in 490-μl aliquots. C1 (240 μg/ml) was added to a final concentration of 5 μg/ml (10.4 μl) and incubated for 30 min at 37 °C. After centrifugation, supernatants were separated by SDS-PAGE on 12.5% gels and then analyzed by western blotting using goat anti-C1q and rabbit anti-C1s and anti-HMGB1 antibodies. (d) C1 cleavage of HMGB1 was inhibited by C1-INH. Apoptotic cells were incubated with C1 for 30 min at 37 °C in the presence or absence of C1-INH (105 μg/ml). As controls, apoptotic cells were incubated with C1-INH only. After centrifugation, supernatants were subjected to SDS-PAGE on 12.5% (w/v) gels and western blotting using the rabbit anti-HMGB1 antibody. (e) Comparison of C1 with C1s in HMGB1 cleavage during apoptosis. As in (c), apoptotic cells harvested 2.5 h after UV irradiation were incubated for 30 min at 37 °C with C1 (5 μg/ml) or sC1s (1.0, 2.5 or 5.0 μg/ml). As controls, Jurkat cells were incubated with C1 or C1s without prior UV irradiation. After centrifugation, supernatants were subjected to SDS-PAGE on 12.5% gels and western blotting using the rabbit anti-HMGB1 antibody. Band intensities were quantified by densitometry. Values for C1s represent combined signals of the heavy and light chains in each lane. Values for HMGB1 were derived from the single band in each lane. Note the reduced HMGB1 intensities at 2.5 and 5.0 μg/ml of sC1s. Apoptotic cells: AC.
Figure 3
Figure 3
Alignment of HMGB1 fragments potentially generated after C1s cleavage and HMGB1 regions that interact with LPS and CD14. Based on the sizes of the two HMGB1 fragments after C1s digestion, that is, 19 and 12 kDa (right panel), the two likely cleavage sites are Arg163 (F1ʹ) and Arg97 (F2ʹ). In this western blotting experiment, bacteria-expressed His-HMGB1 was used. SC1s was used at reducing concentrations. The corresponding LPS and CD14 interactive regions in HMGB1 were aligned below. Note that, both C1s cleavage sites fall into bioactive regions of HMGB1 (triangles) and can potentially impair HMGB1 synergy with LPS.
Figure 4
Figure 4
HMGB1 enables monocyte, macrophage and DC activation by suboptimal levels of LPS but this is inactivated with C1s digestion. Macrophages were stimulated for 20 h with LPS at 1 ng/ml and rHMGB1 was added at 12.5, 25, 50 and 100 ng/ml. As controls, macrophages were stimulated with LPS at 1 and 10 ng/ml without rHMGB1. Production of IL-6 (a) and TNF-α (b) was measured by ELISA. In another experiment, different concentrations of LPS (12.5–500 ng/ml) were pre-incubated for 20 h at 37 °C with rHMGB1 (5 μg/ml) in the presence or absence of rC1s (2.75 μg/ml). As controls, LPS was incubated with PBS or rC1s. These stimuli mixtures were used at 1/50 dilution to stimulate macrophages so that the final concentration of HMGB1 and rC1s in the culture are 100 and 55 ng/ml respectively. IL-6 (c) and TNF-α (d) production was determined by ELISA. DC (e) and monocytes (f) were similarly stimulated except that LPS concentrations were increased in the pre-incubation mixture (25–5000 ng/ml). IL-6 production was determined by ELISA. Experiments were performed in triplicates. Results were analyzed using Student’s 2-tailed unpaired t-test and presented as mean±S.D. *P<0.05, **P<0.01.
Figure 5
Figure 5
C1 cleaves multiple proteins released by apoptotic cells. (a) Visible protein profile released from apoptotic cells and their susceptibility to C1 or C1s degradation. UV-irradiated Jurkat cells were cultured for 2.5 h and then incubated with sC1s, C1 or PBS for 30 min at 37 °C. After centrifugation, supernatants were harvested and subjected to SDS-PAGE on 15% (w/v) gels. Proteins were visualized by Commassie blue staining. Band regions in the C1-treated sample that diminished compared with control sample (PBS-treated) are indicated with arrows and alphabets. Four band regions were identified: A (~100 kDa), B (~60 kDa), C (~50 kDa) and D (~40 kDa). In a separate gel, these four band regions in the control sample were excised for mass spectrometry analysis (Supplementary Table 1). (b) The separated proteins were also analyzed by western blotting where HMGB1, NPM1, β-actin and, as controls, C1s and C1q were detected. (c) C1 showed no cleavage of HSP90α and RPLP0. By mass spectrometry, HSP90α was detected in band A and RPLP0 was found in band C (Supplementary Table 1). Western blot detection of HSP90α and RPLP0 were done using rabbit polyclonal antibodies. HMGB1 was used as a control. AC: apoptotic cells.
Figure 6
Figure 6
C1 cleavage of autoantigens released by apoptotic cells. U937 cells (5×107/ml) in PBS were UV-irradiated. The supernatant was obtained after 24 h and incubated for 1 h with C1 (48 μg/ml) or, as a control, with PBS. The supernatant samples were then separated by SDS-PAGE on 12.5% (w/v) gels and analyzed by western blotting with rheumatological patient sera at 1 : 5000 dilutions. Horseradish peroxidase-conjugated goat anti-human IgG Fc was used as secondary antibody and signals were visualized using the Bio-Rad MPS ChemiDoc imaging system. (a) Sera from four patients detected autoantigens in the apoptotic U937 supernatant. A 45-kDa autoantigen which was detected by sera from both patient B and C and degraded by C1 has been indicated (arrow). A 40-kDa autoantigen detected by serum from patient D was also degraded by C1 (arrow). (b) Sera from patients E-H detected insignificant autoantigens in the apoptotic U937 supernatant. In this validating experiment, serum from patient C was used as a positive control. A single blot was cut into 5 strips and incubated separately with the patients’ sera. Apoptotic cells: AC.

References

    1. Rahman A, Isenberg DA. Systemic lupus erythematosus. N Engl J Med 2008; 358: 929–939.
    1. Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med 2003; 197: 711–723.
    1. Tan EM. Autoantibodies, autoimmune disease, and the birth of immune diagnostics. J Clin Invest 2012; 122: 3835–3836.
    1. Arbuckle MR, McClain MT, Rubertone MV, Scofield RH, Dennis GJ, James JA et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. N Engl J Med 2003; 349: 1526–1533.
    1. Doria A, Zen M, Canova M, Bettio S, Bassi N, Nalotto L et al. SLE diagnosis and treatment: when early is early. Autoimmun Rev 2010; 10: 55–60.
    1. Muller S, Dieker J, Tincani A, Meroni PL. Pathogenic anti-nucleosome antibodies. Lupus 2008; 17: 431–436.
    1. Bave U, Magnusson M, Eloranta ML, Perers A, Alm GV, Ronnblom L. Fc gamma RIIa is expressed on natural IFN-alpha-producing cells (plasmacytoid dendritic cells) and is required for the IFN-alpha production induced by apoptotic cells combined with lupus IgG. J Immunol 2003; 171: 3296–3302.
    1. Cui Y, Sheng Y, Zhang X. Genetic susceptibility to SLE: recent progress from GWAS. J Autoimmun 2013; 41: 25–33.
    1. Deng Y, Tsao BP. Advances in lupus genetics and epigenetics. Curr Opin Rheumatol 2014; 26: 482–492.
    1. Moser KL, Kelly JA, Lessard CJ, Harley JB. Recent insights into the genetic basis of systemic lupus erythematosus. Genes Immun 2009; 10: 373–379.
    1. Chowdhury D, Beresford PJ, Zhu P, Zhang D, Sung JS, Demple B et al. The exonuclease TREX1 is in the SET complex and acts in concert with NM23-H1 to degrade DNA during granzyme A-mediated cell death. Mol Cell 2006; 23: 133–142.
    1. Grieves JL, Fye JM, Harvey S, Grayson JM, Hollis T, Perrino FW. Exonuclease TREX1 degrades double-stranded DNA to prevent spontaneous lupus-like inflammatory disease. Proc Natl Acad Sci USA 2015; 112: 5117–5122.
    1. Reid KB. Activation and control of the complement system. Essays Biochem 1986; 22: 27–68.
    1. Korb LC, Ahearn JM. C1q binds directly and specifically to surface blebs of apoptotic human keratinocytes: complement deficiency and systemic lupus erythematosus revisited. J Immunol 1997; 158: 4525–4528.
    1. Ogden CA, deCathelineau A, Hoffmann PR, Bratton D, Ghebrehiwet B, Fadok VA et al. C1q and mannose binding lectin engagement of cell surface calreticulin and CD91 initiates macropinocytosis and uptake of apoptotic cells. J Exp Med 2001; 194: 781–795.
    1. Green DR, Oguin TH, Martinez J. The clearance of dying cells: table for two. Cell Death Differ 2016; 23: 915–926.
    1. Clarke EV, Weist BM, Walsh CM, Tenner AJ. Complement protein C1q bound to apoptotic cells suppresses human macrophage and dendritic cell-mediated Th17 and Th1 T cell subset proliferation. J Leukoc Biol 2015; 97: 147–160.
    1. Teh BK, Yeo JG, Chern LM, Lu J. C1q regulation of dendritic cell development from monocytes with distinct cytokine production and T cell stimulation. Mol Immunol 2011; 48: 1128–1138.
    1. Lood C, Gullstrand B, Truedsson L, Olin AI, Alm GV, Ronnblom L et al. C1q inhibits immune complex-induced interferon-alpha production in plasmacytoid dendritic cells: a novel link between C1q deficiency and systemic lupus erythematosus pathogenesis. Arthritis Rheum 2009; 60: 3081–3090.
    1. Santer DM, Hall BE, George TC, Tangsombatvisit S, Liu CL, Arkwright PD et al. C1q deficiency leads to the defective suppression of IFN-alpha in response to nucleoprotein containing immune complexes. J Immunol 2010; 185: 4738–4749.
    1. Manderson AP, Botto M, Walport MJ. The role of complement in the development of systemic lupus erythematosus. Ann Rev Immunol 2004; 22: 431–456.
    1. Pickering MC, Botto M, Taylor PR, Lachmann PJ, Walport MJ. Systemic lupus erythematosus, complement deficiency, and apoptosis. Adv Immunol 2000; 76: 227–324.
    1. Amano MT, Ferriani VP, Florido MP, Reis ES, Delcolli MI, Azzolini AE et al. Genetic analysis of complement C1s deficiency associated with systemic lupus erythematosus highlights alternative splicing of normal C1s gene. Mol Immunol 2008; 45: 1693–1702.
    1. Bienaime F, Quartier P, Dragon-Durey MA, Fremeaux-Bacchi V, Bader-Meunier B, Patey N et al. Lupus nephritis associated with complete C1s deficiency efficiently treated with rituximab: a case report. Arthritis Care Res (Hoboken) 2010; 62: 1346–1350.
    1. Dragon-Durey MA, Quartier P, Fremeaux-Bacchi V, Blouin J, de Barace C, Prieur AM et al. Molecular basis of a selective C1s deficiency associated with early onset multiple autoimmune diseases. J Immunol 2001; 166: 7612–7616.
    1. Abe K, Endo Y, Nakazawa N, Kanno K, Okubo M, Hoshino T et al. Unique phenotypes of C1s deficiency and abnormality caused by two compound heterozygosities in a Japanese family. J Immunol 2009; 182: 1681–1688.
    1. Wu YL, Brookshire BP, Verani RR, Arnett FC, Yu CY. Clinical presentations and molecular basis of complement C1r deficiency in a male African-American patient with systemic lupus erythematosus. Lupus 2011; 20: 1126–1134.
    1. Loos M, Heinz HP. Component deficiencies. 1. The first component: C1q, C1r, C1s. Prog Allergy 1986; 39: 212–231.
    1. Walport MJ. Complement. First of two parts. N Engl J Med 2001; 344: 1058–1066.
    1. Busby WH Jr., Nam TJ, Moralez A, Smith C, Jennings M, Clemmons DR. The complement component C1s is the protease that accounts for cleavage of insulin-like growth factor-binding protein-5 in fibroblast medium. J Biol Chem 2000; 275: 37638–37644.
    1. Eriksson H, Nissen MH. Proteolysis of the heavy chain of major histocompatibility complex class I antigens by complement component C1s. Biochim Biophys Acta 1990; 1037: 209–215.
    1. Naito AT, Sumida T, Nomura S, Liu ML, Higo T, Nakagawa A et al. Complement C1q activates canonical Wnt signaling and promotes aging-related phenotypes. Cell 2012; 149: 1298–1313.
    1. Kerr FK, O'Brien G, Quinsey NS, Whisstock JC, Boyd S, de la Banda MG et al. Elucidation of the substrate specificity of the C1s protease of the classical complement pathway. J Biol Chem 2005; 280: 39510–39514.
    1. Cai Y, Teo BH, Yeo JG, Lu J. C1q protein binds to the apoptotic nucleolus and causes C1 protease degradation of nucleolar proteins. J Biol Chem 2015; 290: 22570–22580.
    1. Andersson U, Tracey KJ. HMGB1 is a therapeutic target for sterile inflammation and infection. Annu Rev Immunol 2011; 29: 139–162.
    1. Harris HE, Andersson U, Pisetsky DS. HMGB1: a multifunctional alarmin driving autoimmune and inflammatory disease. Nat Rev Rheumatol 2012; 8: 195–202.
    1. Urbonaviciute V, Furnrohr BG, Meister S, Munoz L, Heyder P, De Marchis F et al. Induction of inflammatory and immune responses by HMGB1-nucleosome complexes: implications for the pathogenesis of SLE. J Exp Med 2008; 205: 3007–3018.
    1. Youn JH, Oh YJ, Kim ES, Choi JE, Shin JS. High mobility group box 1 protein binding to lipopolysaccharide facilitates transfer of lipopolysaccharide to CD14 and enhances lipopolysaccharide-mediated TNF-alpha production in human monocytes. J Immunol 2008; 180: 5067–5074.
    1. Boyd SE, Pike RN, Rudy GB, Whisstock JC, Garcia de la Banda M. PoPS: a computational tool for modeling and predicting protease specificity. J Bioinform Comput Biol 2005; 3: 551–585.
    1. Lotze MT, Tracey KJ. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol 2005; 5: 331–342.
    1. Fairlie DP, Tyndall JD, Reid RC, Wong AK, Abbenante G, Scanlon MJ et al. Conformational selection of inhibitors and substrates by proteolytic enzymes: implications for drug design and polypeptide processing. J Med Chem 2000; 43: 1271–1281.
    1. O'Brien G, Quinsey NS, Whisstock JC, Pike RN. Importance of the prime subsites of the C1s protease of the classical complement pathway for recognition of substrates. Biochemistry 2003; 42: 14939–14945.
    1. Hreggvidsdottir HS, Ostberg T, Wahamaa H, Schierbeck H, Aveberger AC, Klevenvall L et al. The alarmin HMGB1 acts in synergy with endogenous and exogenous danger signals to promote inflammation. J Leukoc Biol 2009; 86: 655–662.
    1. Youn JH, Kwak MS, Wu J, Kim ES, Ji Y, Min HJ et al. Identification of lipopolysaccharide-binding peptide regions within HMGB1 and their effects on subclinical endotoxemia in a mouse model. Eur J Immunol 2011; 41: 2753–2762.
    1. Palmer CD, Romero-Tejeda M, Sirignano M, Sharma S, Allen TM, Altfeld M et al. Naturally Occurring Subclinical Endotoxemia in Humans Alters Adaptive and Innate Immune Functions through Reduced MAPK and Increased STAT1 Phosphorylation. J Immunol 2016; 196: 668–677.
    1. Candiano G, Bruschi M, Musante L, Santucci L, Ghiggeri GM, Carnemolla B et al. Blue silver: a very sensitive colloidal Coomassie G-250 staining for proteome analysis. Electrophoresis 2004; 25: 1327–1333.
    1. Minota S, Koyasu S, Yahara I, Winfield J. Autoantibodies to the heat-shock protein hsp90 in systemic lupus erythematosus. J Clin Invest 1988; 81: 106–109.
    1. Pfeifle J, Anderer FA, Franke M. Characterisation of nucleolar proteins as autoantigens using human autoimmune sera. Ann Rheum Dis 1986; 45: 978–986.
    1. Arnett FC, Reveille JD, Moutsopoulos HM, Georgescu L, Elkon KB. Ribosomal P autoantibodies in systemic lupus erythematosus. Frequencies in different ethnic groups and clinical and immunogenetic associations. Arthritis Rheum 1996; 39: 1833–1839.
    1. Bave U, Alm GV, Ronnblom L. The combination of apoptotic U937 cells and lupus IgG is a potent IFN-alpha inducer. J Immunol 2000; 165: 3519–3526.
    1. Pettigrew HD, Teuber SS, Gershwin ME. Clinical significance of complement deficiencies. Ann NY Acad Sci 2009; 1173: 108–123.
    1. Paidassi H, Tacnet-Delorme P, Garlatti V, Darnault C, Ghebrehiwet B, Gaboriaud C et al. C1q binds phosphatidylserine and likely acts as a multiligand-bridging molecule in apoptotic cell recognition. J Immunol 2008; 180: 2329–2338.
    1. Heesters BA, Das A, Chatterjee P, Carroll MC. Do follicular dendritic cells regulate lupus-specific B cells? Mol Immunol 2014; 62: 283–288.
    1. Gnauck A, Lentle RG, Kruger MC. Chasing a ghost? - Issues with the determination of circulating levels of endotoxin in human blood. Crit Rev Clin Lab Sci 2016; 53: 197–215.
    1. Casiano CA, Ochs RL, Tan EM. Distinct cleavage products of nuclear proteins in apoptosis and necrosis revealed by autoantibody probes. Cell Death Differ 1998; 5: 183–190.
    1. Cao W, Tan P, Lee CH, Zhang H, Lu J. A transforming growth factor-beta-induced protein stimulates endocytosis and is up-regulated in immature dendritic cells. Blood 2006; 107: 2777–2785.
    1. Song J, Matthews AY, Reboul CF, Kaiserman D, Pike RN, Bird PI et al. Predicting serpin/protease interactions. Methods Enzymol 2011; 501: 237–273.
    1. Matthews DJ, Goodman LJ, Gorman CM, Wells JA. A survey of furin substrate specificity using substrate phage display. Protein Sci 1994; 3: 1197–1205.
    1. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods 2012; 9: 671–675.
    1. Chevallet M, Luche S, Rabilloud T. Silver staining of proteins in polyacrylamide gels. Nat Protoc 2006; 1: 1852–1858.

Source: PubMed

3
S'abonner