C5a and C5aR1 are key drivers of microvascular platelet aggregation in clinical entities spanning from aHUS to COVID-19

Sistiana Aiello, Sara Gastoldi, Miriam Galbusera, Piero Ruggenenti, Valentina Portalupi, Stefano Rota, Nadia Rubis, Lucia Liguori, Sara Conti, Matteo Tironi, Sara Gamba, Donata Santarsiero, Ariela Benigni, Giuseppe Remuzzi, Marina Noris, Sistiana Aiello, Sara Gastoldi, Miriam Galbusera, Piero Ruggenenti, Valentina Portalupi, Stefano Rota, Nadia Rubis, Lucia Liguori, Sara Conti, Matteo Tironi, Sara Gamba, Donata Santarsiero, Ariela Benigni, Giuseppe Remuzzi, Marina Noris

Abstract

Unrestrained activation of the complement system till the terminal products, C5a and C5b-9, plays a pathogenetic role in acute and chronic inflammatory diseases. In endothelial cells, complement hyperactivation may translate into cell dysfunction, favoring thrombus formation. The aim of this study was to investigate the role of the C5a/C5aR1 axis as opposed to C5b-9 in inducing endothelial dysfunction and loss of antithrombogenic properties. In vitro and ex vivo assays with serum from patients with atypical hemolytic uremic syndrome (aHUS), a prototype rare disease of complement-mediated microvascular thrombosis due to genetically determined alternative pathway dysregulation, and cultured microvascular endothelial cells, demonstrated that the C5a/C5aR1 axis is a key player in endothelial thromboresistance loss. C5a added to normal human serum fully recapitulated the prothrombotic effects of aHUS serum. Mechanistic studies showed that C5a caused RalA-mediated exocytosis of von Willebrand factor (vWF) and P-selectin from Weibel-Palade bodies, which favored further vWF binding on the endothelium and platelet adhesion and aggregation. In patients with severe COVID-19 who suffered from acute activation of complement triggered by severe acute respiratory syndrome coronavirus 2 infection, we found the same C5a-dependent pathogenic mechanisms. These results highlight C5a/C5aR1 as a common prothrombogenic effector spanning from genetic rare diseases to viral infections, and it may have clinical implications. Selective C5a/C5aR1 blockade could have advantages over C5 inhibition because the former preserves the formation of C5b-9, which is critical for controlling bacterial infections that often develop as comorbidities in severely ill patients. The ACCESS trial registered at www.clinicaltrials.gov as #NCT02464891 accounts for the results related to aHUS patients treated with CCX168.

© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Serum from patients with aHUS induces platelet adhesion and aggregation on microvascular endothelial cells through C5a/C5aR1 signaling. (A, upper panel) Experimental design. HMEC-1 were activated with ADP, exposed for 3 hours to ctr serum or serum from patients with aHUS in remission, and then perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. aHUS serum was added or not with the pan complement inhibitor sCR1 (150 μg/mL). Formation of platelet aggregates was evaluated at the end of blood perfusion (through confocal microscopy). (A, lower panel) Endothelial surface area with positive staining for platelet aggregates. Results are shown as fold increase of stained surface area after incubation with ctr serum run in parallel. Data are mean ± SD; n = 9 independent experiments. Red points represent fold increase values of single experiments. Purple lines are the upper and lower limits of normal range. (B) Representative images of the ultrastructure of aggregates of platelets adhered on HMEC-1 preexposed to aHUS serum, evaluated with scanning electron microscopy at low magnification (left panels), with the corresponding high magnification insets (right panels). Insets show high-power view of same platelet aggregate. Scale bars represent 10 µm (n = 2 experiments). (C, upper panel) Experimental design. HMEC-1 were activated with ADP, exposed for 3 hours to ctr or aHUS serum, and then perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. aHUS serum was added or not, with an anti-C5 antibody (135 μg/mL), or the C5aR1 antagonist CCX168 (200 ng/mL), or an anti-C7 antibody (350 μg/mL). Formation of platelet aggregates was evaluated at the end of the blood perfusion. (C, lower panel) Endothelial surface area covered by platelet aggregates. Results are shown as fold increase of stained surface area after incubation with ctr serum run in parallel. Data are mean ± SD; n = 4 independent experiments for +anti-C5; n = 3 independent experiments for +C5aR1a and +anti-C7. Red points represent fold increase values of single experiments. Purple lines are the upper and lower limits of normal range. (D, upper panel) Experimental design. HMEC-1 were activated with ADP and then exposed for 3 hours to ctr or aHUS serum. aHUS serum was added or not with sCR1 (150 μg/mL) or the anti-C5 antibody (135 μg/mL). Serum C5a levels were measured before and after incubation. (D, lower panel) C5a concentration in ctr or aHUS serum before and after incubation with ADP-activated HMEC-1. Data are mean ± SD; n = 3 independent experiments. (E) Scheme of treatment with the C5a receptor antagonist CCX168 and serum sample collection in patients with aHUS enrolled in the ACCESS study. (F) Experimental design. HMEC-1 were activated with ADP, exposed for 3 hours to ctr serum or serum from patients with aHUS (taken before treatment with CCX168, day 0, and after 14 days of treatment), and then analyzed for C5b-9 deposits or perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. Formation of platelet aggregates was evaluated at the end of blood perfusion. (G-H) Endothelial surface area with positive staining for platelet aggregates (G) or C5b-9 (H) after incubation of ADP-activated HMEC-1 with ctr serum or serum from patients with aHUS treated with CCX168 collected at days 0 and 14 of the study. Data are mean ± SD of cumulative data from 5 patients. (I) C5a levels in ctr serum or in serum from patients with aHUS taken before treatment with CCX168, day 0, and after 14 days of treatment (n = 5). *P < .05 vs the groups indicated by horizontal bar or, if not indicated, vs all groups. **P < .05 vs +C5aR1a. #P < .05 vs ctr serum.
Figure 2.
Figure 2.
C5a and normal human serum (ctr) induces platelet adhesion and aggregation on HMEC-1. (A) Experimental design. HMEC-1, ADP-activated or resting, were exposed for 3 hours to ctr serum with or without the addition of C5a (200 ng/mL) or to aHUS serum, and then perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. C5a-additioned serum was tested in the presence or absence of sCR1 (150 μg/mL) or the C5aR1 antagonist CCX168 (200 ng/mL). Formation of platelet aggregates was evaluated at the end of the blood perfusion. (B-C) Effect of C5a-additioned ctr serum on formation of platelet aggregates on HMEC-1 (B, ADP-activated; C, resting). Results are shown as fold increase of stained surface area after incubation with ctr serum alone run in parallel. Data are mean ± SD; n = 3 independent experiments. Red points represent fold increase values of single experiments. Purple lines are the upper and lower limits of normal range. *P < .05 vs the groups indicated by horizontal bar. #P < .05 vs ctr serum. (D, upper panel) Experimental design. HMEC-1 were activated or not with ADP for 10 minutes and then stimulated or not with C5a for 30 minutes, exposed for 3 hours to ctr serum, and perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. On selected slides, ADP-activated cells were exposed to aHUS serum, taken as the positive control. Formation of platelet aggregates was evaluated at the end of the blood perfusion. (D, lower panel) Endothelial surface area covered by platelet aggregates after incubation with ctr serum of resting (no ADP, no C5a), ADP-activated, C5a-stimulated or ADP-activated/C5a-stimulated HMEC-1; or after incubation with aHUS serum. Data are mean ± SD; n = 4 independent experiments. *P < .05 vs the groups indicated by horizontal bar. #P < .05 vs resting and ADP-activated HMEC-1 + ctr serum. (E) Representative images of immunostaining for C5aR1 (red staining) on resting or ADP-activated HMEC-1. Blue: DAPI staining. Original magnification: ×400. Scale bar represents 50 μm. (F, upper panel) Experimental design. HMEC-1 were activated or not with ADP for 10 minutes and then stimulated or not with C5a for 30 minutes, exposed for 3 hours to ctr serum, and perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. In 3 of 4 experiments, additional samples were run in which either sCR1 (150 μg/mL) or the C5aR1 antagonist CCX168 (200 ng/mL) were added during the C5a stimulation and the exposure to serum. Formation of platelet aggregates was evaluated at the end of blood perfusion. (F, lower panel) Endothelial surface area covered by platelet aggregates after incubation with ctr serum of resting, ADP-activated, C5a-stimulated or ADP-activated/C5a-stimulated HMEC-1. Data are mean ± SD; n = 3 to 4 independent experiments. *P < .05 vs the groups indicated by horizontal bar. #P < .05 vs resting. (G, upper panel) Experimental design. HMEC-1 were activated or not with ADP for 10 minutes and then stimulated or not with C5a for 30 minutes and perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. Formation of platelet aggregates was evaluated at the end of blood perfusion. (G, lower panel) Endothelial surface area covered by platelet aggregates on resting, ADP-activated, C5a-stimulated or ADP-activated/C5a-stimulated HMEC-1. Data are mean ± SD; n = 3 independent experiments.
Figure 3.
Figure 3.
Role of vWF and P-selectin in C5a-induced platelet adhesion and aggregation on HMEC-1. (A) Endothelial surface area stained for vWF or P-selectin on resting or C5a-stimulated HMEC-1. Results are shown as fold increase of stained area on resting HMEC-1. Data are mean ± SD; n = 4 (vWF) or 3 (P-selectin) independent experiments. *P < .05 vs no stimulation. (B, upper panel) Experimental design. Before the experiment, HMEC-1 were left for 2 hours or 16 hours with medium added or not with the RalA inhibitor BQU57 (10 µM). Thereafter, HMEC-1 were activated or not for 10 minutes with ADP and then stimulated or not for 30 minutes with C5a (200 ng/mL). vWF staining was performed at the end of C5a stimulation. (B, lower panel) Endothelial surface area stained for vWF. Data are mean ± SD of 13 fields; n = 1 to 2 experiments. *P < .05 vs the groups indicated by horizontal bar. (C, upper panel) Experimental design. HMEC-1 were activated or not with ADP and then stimulated or not with C5a and exposed or not for 3 hours to ctr serum. vWF staining was performed at the end of C5a stimulation or exposure to serum. (C, lower panel) Endothelial surface area stained for vWF. Mean ± SD of 13 fields. *P < .05. #P < .05 vs the corresponding condition with serum. (D, upper panel) Experimental design. HMEC-1 were activated or not with ADP and then stimulated or not with C5a, exposed for 3 hours to ctr serum and then perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. In 3 of 4 experiments, either an anti-vWF (10 μg/mL) or an anti-P-selectin (20 μg/mL) antibody was added during the C5a stimulation and the exposure to serum. Formation of platelet aggregates was evaluated at the end of blood perfusion. (D, lower panel) Endothelial surface area covered by platelet aggregates on resting (no ADP, no C5a), C5a-stimulated, or ADP-activated/C5a-stimulated HMEC-1 incubated with ctr serum. Results are shown as fold increase of stained surface area on resting HMEC-1 incubated with ctr serum alone run in parallel. Data are mean ± SD; n = 3 to 4 independent experiments. Red points represent fold increase values of single experiments. Purple lines are the upper and lower limits of normal range. *P < .05 vs the groups indicated by horizontal bar.
Figure 4.
Figure 4.
Role of vWF and exocytosis of WPBs in aHUS serum-induced platelet adhesion and aggregation on HMEC-1. (A, upper panel) Experimental design. HMEC-1 were activated with ADP and then exposed for 3 hours to ctr or aHUS serum. aHUS serum was added or not with sCR1 (150 μg/mL) or the C5aR1 antagonist CCX168 (200 ng/mL). vWF staining was performed at the end of exposure to serum. (A, lower panel) Endothelial surface area stained for vWF. Results are shown as fold increase of stained area after incubation, with aHUS serum (n = 4) vs ctr serum run in parallel. Data are mean ± SD. *P < .05 vs all groups. (B, upper panel) Experimental design. ADP-activated HMEC-1 were exposed for 3 hours to ctr or aHUS serum and then perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. aHUS serum was added or not with an anti-vWF antibody (10 μg/mL). Formation of platelet aggregates was evaluated at the end of blood perfusion. (B, lower panel) Endothelial surface area covered by platelet aggregates. Results are shown as fold increase of surface area covered by platelet aggregates after incubation with aHUS serum (n = 3) vs ctr serum run in parallel. Data are mean ± SD. Red points represent fold increase values of single patients. Purple lines are the upper and lower limits of normal range. *P < .05 vs all groups. (C, upper panel) Experimental design. Before the experiment, HMEC-1 were left for 16 hours in medium with or without the RalA inhibitor BQU57 (10 µM). Thereafter, HMEC-1 were activated for 10 minutes with ADP, exposed for 3 hours to ctr or aHUS serum, and then perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. Formation of platelet aggregates was evaluated at the end of blood perfusion. (C, lower panel) Endothelial surface area covered by platelet aggregates. Results are shown as fold increase of stained surface area after incubation with aHUS serum (n = 3) vs ctr serum run in parallel. Data are mean ± SD. Red points represent fold increase values of single patients. Purple lines are the upper and lower limits of normal range. *P < .05 vs all groups. (D) Representative images of experiments (green staining) relative to Figure 4C. Original magnification ×200. Scale bar represents 50 μm.
Figure 5.
Figure 5.
Terminal complement activation, and role of C5a/C5aR1 signaling in COVID-19 serum-induced platelet adhesion and aggregation on HMEC-1. (A) Endothelial surface area covered by staining for C5b-9 after incubation of ADP-activated HMEC-1 for 2 hours with ctr serum, or serum from patients with COVID-19 or aHUS, taken as positive controls. Results are shown as fold increase of stained surface area after incubation with ctr serum run in parallel. Data are mean ± SD n = 5 independent experiments. Red points represent fold increase values of single experiments. Purple lines are the upper and lower limits of normal range (calculated as shown in supplemental Figure 2). *P < .05 vs control. (B) C5a levels in plasma from healthy subjects (n = 10) or patients with COVID-19 (n = 4). Data are mean ± SD. *P < .05 vs healthy subjects. (C, upper panel) Experimental design. ADP-activated HMEC-1 were exposed for 2 hours to ctr serum or serum from patients with COVID-19 or aHUS taken as positive control, and then perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. COVID-19 serum was added or not with the C5aR1 antagonist CCX168 (200 ng/mL). Formation of platelet aggregates was evaluated at the end of blood perfusion. (C, lower panel) Endothelial surface area covered by platelet aggregates. Results are shown as fold increase of stained surface area after incubation with COVID-19 serum (n = 6) or aHUS serum (n = 4) vs ctr serum run in parallel. Data are mean ± SD. Red points represent fold increase values of single patients. Purple lines are the upper and lower limits of normal range. *P < .05 vs the groups indicated by horizontal bar. #P < .05 vs ctr serum. (D) Representative images of experiments (green staining) relative to Figure 5C. Original magnification ×200. Scale bar represents 50 μm. (E, upper panel) Experimental design. ADP-activated HMEC-1 were exposed for 2 hours to ctr serum or serum from patients with COVID-19 and then perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. COVID-19 serum was added or not with an anti-vWF antibody (10 μg/mL). Formation of platelet aggregates was evaluated at the end of blood perfusion. (E, lower panel) Endothelial surface area covered by platelet aggregates. Results are shown as fold increase of stained surface area after incubation with COVID-19 serum (n = 3) vs ctr serum run in parallel. Data are mean ± SD. Red points represent fold increase values of single patients. Purple lines are the upper and lower limits of normal range. *P < .05 vs all groups. (F, upper panel) Experimental design. Before the experiment, HMEC-1 were left for 16 hours in medium with or without the RalA inhibitor BQU57 (10 µM). Thereafter, HMEC-1 were activated for 10 minutes with ADP, exposed for 2 hours to ctr serum or COVID-19 serum, and then perfused for 3 minutes with heparinized whole blood, added with mepacrine, from healthy subjects. Formation of platelet aggregates was evaluated at the end of blood perfusion. (F, lower panel) Endothelial surface area covered by platelet aggregates. Results are shown as fold increase of stained surface area after incubation with COVID-19 serum (n = 3) vs ctr serum run in parallel. Data are mean ± SD. Red points represent fold increase values of single patients. Purple lines are the upper and lower limits of normal range. *P < .05 vs all groups.
Figure 6.
Figure 6.
Proposed mechanism underlying the pro-thrombotic action of C5a/C5aR1 signaling. Upon engagement with its receptor C5aR1, C5a induces the activation of RalA, which leads to the exocytosis of Weibel-Palade bodies (WPB). Then, vWF and P-selectin are exocytosed from WPBs, providing an adhesive scaffold on which circulating vWF is in turn recruited. As a consequence for the above events, massive platelet adhesion and aggregation occur on the endothelial cell surface.

References

    1. Roumenina LT, Rayes J, Frimat M, Fremeaux-Bacchi V. Endothelial cells: source, barrier, and target of defensive mediators. Immunol Rev. 2016;274(1):307-329.
    1. Lindemann S, Krämer B, Seizer P, Gawaz M. Platelets, inflammation and atherosclerosis. J Thromb Haemost. 2007;5(suppl 1):203-211.
    1. Noris M, Remuzzi G. Overview of complement activation and regulation. Semin Nephrol. 2013;33(6):479-492.
    1. Walport MJ. Complement. First of two parts. N Engl J Med. 2001;344(14):1058-1066.
    1. Tegla CA, Cudrici C, Patel S, et al. . Membrane attack by complement: the assembly and biology of terminal complement complexes. Immunol Res. 2011;51(1):45-60.
    1. Nester CM, Barbour T, de Cordoba SR, et al. . Atypical aHUS: state of the art. Mol Immunol. 2015;67(1):31-42.
    1. Noris M, Galbusera M, Gastoldi S, et al. . Dynamics of complement activation in aHUS and how to monitor eculizumab therapy. Blood. 2014; 124(11):1715-1726.
    1. Galbusera M, Noris M, Gastoldi S, et al. . An ex vivo test of complement activation on endothelium for individualized eculizumab therapy in hemolytic uremic syndrome. Am J Kidney Dis. 2019;74(1):56-72.
    1. Yuan X, Yu J, Gerber G, et al. . Ex vivo assays to detect complement activation in complementopathies. Clin Immunol. 2020;221:108616.
    1. Legendre CM, Licht C, Muus P, et al. . Terminal complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med. 2013; 368(23):2169-2181.
    1. Noris M, Mescia F, Remuzzi G. STEC-HUS, atypical HUS and TTP are all diseases of complement activation. Nat Rev Nephrol. 2012;8(11):622-633.
    1. Brodsky RA. Paroxysmal nocturnal hemoglobinuria. Blood. 2014;124(18):2804-2811.
    1. Noris M, Benigni A, Remuzzi G. The case of complement activation in COVID-19 multiorgan impact. Kidney Int. 2020;98(2):314-322.
    1. Ueda Y, Miwa T, Ito D, et al. . Differential contribution of C5aR and C5b-9 pathways to renal thrombic microangiopathy and macrovascular thrombosis in mice carrying an atypical hemolytic syndrome-related factor H mutation. Kidney Int. 2019;96(1):67-79.
    1. World Health Organization . Coronavirus disease (COVID-19) outbreak. Interim guidance. Available at: . Accessed 19 March 2020.
    1. Bekker P, Dairaghi D, Seitz L, et al. . Characterization of pharmacologic and pharmacokinetic properties of CCX168, a potent and selective orally administered complement 5a Receptor inhibitor, based on preclinical evaluation and randomized phase 1 clinical study [published correction appears in PLOS One. 2019;14(1):e0210593]. PLoS One. 2016;11(10):e0164646.
    1. Turner NA, Moake J. Assembly and activation of alternative complement components on endothelial cell-anchored ultra-large von Willebrand factor links complement and hemostasis-thrombosis. PLoS One. 2013;8(3):e59372.
    1. Bettoni S, Galbusera M, Gastoldi S, et al. . Interaction between multimeric von Willebrand factor and complement: a fresh look to the pathophysiology of microvascular thrombosis. J Immunol. 2017;199(3):1021-1040.
    1. Shirakawa R, Horiuchi H. Ral GTPases: crucial mediators of exocytosis and tumourigenesis. J Biochem. 2015;157(5):285-299.
    1. McCormack JJ, Lopes da Silva M, Ferraro F, Patella F, Cutler DF. Weibel-Palade bodies at a glance. J Cell Sci. 2017;130(21):3611-3617.
    1. Schillemans M, Karampini E, Kat M, Bierings R. Exocytosis of Weibel-Palade bodies: how to unpack a vascular emergency kit. J Thromb Haemost. 2019;17(1):6-18.
    1. Ikeda K, Nagasawa K, Horiuchi T, Tsuru T, Nishizaka H, Niho Y. C5a induces tissue factor activity on endothelial cells. Thromb Haemost. 1997; 77(2):394-398.
    1. Morigi M, Galbusera M, Gastoldi S, et al. . Alternative pathway activation of complement by Shiga toxin promotes exuberant C3a formation that triggers microvascular thrombosis. J Immunol. 2011;187(1):172-180.
    1. Skeie JM, Fingert JH, Russell SR, Stone EM, Mullins RF. Complement component C5a activates ICAM-1 expression on human choroidal endothelial cells. Invest Ophthalmol Vis Sci. 2010;51(10):5336-5342.
    1. Monsinjon T, Gasque P, Chan P, Ischenko A, Brady JJ, Fontaine MC. Regulation by complement C3a and C5a anaphylatoxins of cytokine production in human umbilical vein endothelial cells. FASEB J. 2003;17(9):1003-1014.
    1. Foreman KE, Vaporciyan AA, Bonish BK, et al. . C5a-induced expression of P-selectin in endothelial cells. J Clin Invest. 1994;94(3):1147-1155.
    1. Albrecht EA, Chinnaiyan AM, Varambally S, et al. . C5a-induced gene expression in human umbilical vein endothelial cells. Am J Pathol. 2004; 164(3):849-859.
    1. Yamakuchi M, Kirkiles-Smith NC, Ferlito M, et al. . Antibody to human leukocyte antigen triggers endothelial exocytosis. Proc Natl Acad Sci USA. 2007;104(4):1301-1306.
    1. Woodruff TM, Nandakumar KS, Tedesco F. Inhibiting the C5-C5a receptor axis. Mol Immunol. 2011;48(14):1631-1642.
    1. Huber-Lang M, Lambris JD, Ward PA. Innate immune responses to trauma. Nat Immunol. 2018;19(4):327-341.
    1. Bokisch VA, Top FH Jr, Russell PK, Dixon FJ, Müller-Eberhard HJ. The potential pathogenic role of complement in dengue hemorrhagic shock syndrome. N Engl J Med. 1973;289(19):996-1000.
    1. Matsumoto ML, Narzinski K, Nikiforovich GV, Baranski TJ. A comprehensive structure-function map of the intracellular surface of the human C5a receptor. II. Elucidation of G protein specificity determinants. J Biol Chem. 2007;282(5):3122-3133.
    1. Monk PN, Scola A-M, Madala P, Fairlie DP. Function, structure and therapeutic potential of complement C5a receptors. Br J Pharmacol. 2007;152(4):429-448.
    1. Jacob A, Hack B, Chiang E, Garcia JG, Quigg RJ, Alexander JJ. C5a alters blood-brain barrier integrity in experimental lupus. FASEB J. 2010; 24(6):1682-1688.
    1. Ali H. Regulation of human mast cell and basophil function by anaphylatoxins C3a and C5a. Immunol Lett. 2010;128(1):36-45.
    1. Parkos CA, Cochrane CG, Schmitt M, Jesaitis AJ. Regulation of the oxidative response of human granulocytes to chemoattractants. No evidence for stimulated traffic of redox enzymes between endo and plasma membranes. J Biol Chem. 1985;260(11):6541-6547.
    1. Michaux G, Pullen TJ, Haberichter SL, Cutler DF. P-selectin binds to the D’-D3 domains of von Willebrand factor in Weibel-Palade bodies. Blood. 2006;107(10):3922-3924.
    1. Ulrichts H, Vanhoorelbeke K, Girma JP, Lenting PJ, Vauterin S, Deckmyn H. The von Willebrand factor self-association is modulated by a multiple domain interaction. J Thromb Haemost. 2005;3(3):552-561.
    1. Ruggeri ZM. The role of von Willebrand factor in thrombus formation. Thromb Res. 2007;120(suppl 1):S5-S9.
    1. Perico L, Benigni A, Casiraghi F, Ng LFP, Renia L, Remuzzi G. Immunity, endothelial injury and complement-induced coagulopathy in COVID-19. Nat Rev Nephrol. 2021;17(1):46-64.
    1. Eriksson O, Hultström M, Persson B, et al. . Mannose-binding lectin is associated with thrombosis and coagulopathy in critically ill COVID-19 patients. Thromb Haemost. 2020;120(12):1720-1724.
    1. Yu J, Yuan X, Chen H, Chaturvedi S, Braunstein EM, Brodsky RA. Direct activation of the alternative complement pathway by SARS-CoV-2 spike proteins is blocked by factor D inhibition. Blood. 2020;136(18):2080-2089.
    1. Latour RP de, Bergeron A, Lengline E, et al. . Complement C5 inhibition in patients with COVID-19 – a promising target? Haematologica. 2020;105(12):2847-2850.
    1. Carvelli J, Demaria O, Vély F, et al. ; Explore COVID-19 Marseille Immunopole group . Association of COVID-19 inflammation with activation of the C5a-C5aR1 axis. Nature. 2020;588(7836):146-150.
    1. Alosaimi B, Mubarak A, Hamed ME, et al. . Complement anaphylatoxins and inflammatory cytokines as prognostic markers for COVID-19 severity and in-hospital mortality. Front Immunol. 2021;12:668725.
    1. O’Sullivan JM, Gonagle DM, Ward SE, Preston RJS, O’Donnell JS. Endothelial cells orchestrate COVID-19 coagulopathy. Lancet Haematol. 2020;7(8):e553-e555.
    1. Varga Z, Flammer AJ, Steiger P, et al. . Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395(10234):1417-1418.
    1. Ackermann M, Verleden SE, Kuehnel M, et al. . Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in COVID-19. N Engl J Med. 2020;383(2):120-128.
    1. Magro C, Mulvey JJ, Berlin D, et al. . Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases. Transl Res. 2020;220:1-13.
    1. Llitjos JF, Leclerc M, Chochois C, et al. . High incidence of venous thromboembolic events in anticoagulated severe COVID-19 patients. J Thromb Haemost. 2020;18(7):1743-1746.
    1. Goshua G, Pine AB, Meizlish ML, et al. . Endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study. Lancet Haematol. 2020;7(8):e575-e582.
    1. Jones SA, Hunter CA. Is IL-6 a key cytokine target for therapy in COVID-19? Nat Rev Immunol. 2021;21(6):337-339.
    1. Laudes IJ, Chu JC, Huber-Lang M, et al. . Expression and function of C5a receptor in mouse microvascular endothelial cells. J Immunol. 2002;169(10):5962-5970.
    1. Paul-Ehrlich-Institut . The Paul-Ehrlich-Institut informs – temporary suspension of vaccination with COVID-19 vaccine AstraZeneca. . Accessed 16 March 2021.
    1. Diurno F, Numis FG, Porta G, et al. . Eculizumab treatment in patients with COVID-19: preliminary results from real life ASL Napoli 2 Nord experience. Eur Rev Med Pharmacol Sci. 2020;24(7):4040-4047.
    1. Annane D, Heming N, Grimaldi-Bensouda L, et al. ; Garches COVID 19 Collaborative Group . Eculizumab as an emergency treatment for adult patients with severe COVID-19 in the intensive care unit: a proof-of-concept study. eClinicalMedicine. 2020;28:100590.
    1. Laurence J, Mulvey JJ, Seshadri M, et al. . Anti-complement C5 therapy with eculizumab in three cases of critical COVID-19. Clin Immunol. 2020;219:108555.
    1. Giudice V, Pagliano P, Vatrella A, et al. . Combination of ruxolitinib and eculizumab for treatment of severe SARS-CoV-2-related acute respiratory distress syndrome: a controlled study. Front Pharmacol. 2020;11:857.
    1. Declercq J, Bosteels C, Van Damme K, et al. . Zilucoplan in patients with acute hypoxic respiratory failure due to COVID-19 (ZILU-COV): a structured summary of a study protocol for a andomizedd controlled trial. Trials. 2020;21(1):934.
    1. Woodruff TM, Shukla AK. The complement C5a-C5aR1 GPCR axis in COVID-19 therapeutics. Trends Immunol. 2020;41(11):965-967.
    1. Huber-Lang M, Sarma VJ, Lu KT, et al. . Role of C5a in multiorgan failure during sepsis. J Immunol. 2001;166(2):1193-1199.
    1. Skendros P, Mitsios A, Chrysanthopoulou A, et al. . Complement and tissue factor-enriched neutrophil extracellular traps are key drivers in COVID-19 immunothrombosis. J Clin Invest. 2020;130(11):6151-6157.
    1. Riedl M, Noone DG, Khan MA, et al. . Complement activation induces neutrophil adhesion and neutrophil-platelet aggregate formation on vascular endothelial cells. Kidney Int Rep. 2016;2(1):66-75.
    1. Vlaar APJ, de Bruin S, Busch M, et al. . Anti-C5a antibody IFX-1 (vilobelimab) treatment versus best supportive care for patients with severe COVID-19 (PANAMO): an exploratory, open-label, phase 2 randomised controlled trial. Lancet Rheumatol. 2020;2(12):e764-e773.
    1. Naik A, Sharma S, Quigg RJ. Complement regulation in renal disease models. Semin Nephrol. 2013;33(6):575-585.
    1. Tedesco F, Pausa M, Nardon E, Introna M, Mantovani A, Dobrina A. The cytolytically inactive terminal complement complex activates endothelial cells to express adhesion molecules and tissue factor procoagulant activity. J Exp Med. 1997;185(9):1619-1627.
    1. Saadi S, Holzknecht RA, Patte CP, Stern DM, Platt JL. Complement-mediated regulation of tissue factor activity in endothelium. J Exp Med. 1995;182(6):1807-1814.
    1. Frimat M, Tabarin F, Dimitrov JD, et al. . Complement activation by heme as a secondary hit for atypical hemolytic uremic syndrome. Blood. 2013;122(2):282-292.
    1. Van Avondt K, Nur E, Zeerleder S. Mechanisms of haemolysis-induced kidney injury. Nat Rev Nephrol. 2019;15(11):671-692.
    1. Yan C, Liu D, Li L, et al. . Discovery and characterization of small molecules that target the GTPase Ral. Nature. 2014;515(7527):443-447.
    1. Moghadam AR, Patrad E, Tafsiri E, et al. . Ral signaling pathway in health and cancer. Cancer Med. 2017;6(12):2998-3013.

Source: PubMed

3
Sottoscrivi