CC BY 4.0 · TH Open 2023; 07(04): e289-e293
DOI: 10.1055/a-2156-8048
Letter to the Editor

The Platelet Anaphylatoxin Receptor C5aR1 (CD88) Is a Promising Target for Modulating Vessel Growth in Response to Ischemiaa

Henry Nording
1   Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
2   DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
,
Lasse Baron
1   Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
,
Antje Lübken
1   Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
,
Hossein Emami
1   Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
,
Jacob von Esebeck
1   Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
,
Moritz Meusel
3   Medical Clinic II, University Hospital, University Heart Center Lübeck, Lübeck, Germany
,
Christian Sadik
4   Clinic for Dermatology, University of Lübeck, University Hospital, Lübeck, Germany
,
Nancy Schanze
5   Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
,
Daniel Duerschmied
5   Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
6   German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Germany
7   European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
,
Jörg Köhl
8   ISEF, University of Lübeck, Lübeck, Germany
,
Götz Münch
9   AdvanceCOR, Martinsried, Germany
,
Harald F. Langer
1   Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
2   DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
4   Clinic for Dermatology, University of Lübeck, University Hospital, Lübeck, Germany
5   Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
6   German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Germany
› Author Affiliations
 

Diseases featuring tissue ischemia are wide-spread and among the main cause of death in the Western world.[1] To date, the exact mechanisms of revascularization of tissues after arterial obstruction, usually as a result of atherosclerosis,[2] remain incompletely understood. This is underscored by the fact that pharmacological approaches to enhance vessel growth in patients with chronic vascular occlusions are not successfully established for clinical use, yet.[3] Several reports implicate complement activation as a contributor to ischemic tissue injury via interaction with reactive oxygen species,[4] for example, the lectin[5] or the classical pathway.[6] [7] Thus, the complement system may be a possible therapeutic target in this disease setting (reviewed in Markiewski et al[8]).

The complement system is a very well-preserved and phylogenetically old part of the immune system serving several functions from immune protection to tissue homeostasis and dysfunction.[9] Previously, the complement receptor C5aR1 was demonstrated to be pro-angiogenic,[10] but has also been attributed an inhibitory role for neovascularization.[11] Interestingly, platelet activation can colocalize with areas of increased complement activity and several functional links have been described connecting complement and platelets.[12] [13] [14]

Platelets play a decisive role in cardiovascular diseases featuring thrombosis, where they are the decisive cellular part of any thrombus and contribute to several mechanisms of thrombus formation.[15] Beyond that, platelets were recently implicated in tissue remodeling processes such as apoptosis,[16] [17] immune patrolling,[18] or adaptive immunity.[19] Furthermore, platelets contribute to the immediate response after vascular injury by promoting vascular inflammation,[15] immunomodulation,[20] [21] [22] and atherosclerosis.[23] [24] Recently, we could show that complement receptors are expressed on platelets, and that the anaphylatoxin C3a receptor modulates primary hemostasis.[25] [26] Moreover, platelets express C5a receptor 1 (C5aR1).[27] Absence of this anaphylatoxin receptor, however, had no effect on in vivo thrombus formation.[25] Thus, it might be of alternative fictional relevance for platelet effects in tissue remodeling.

The group of J. Italiano could visualize that platelets store pro- and antiangiogenic factors in distinct granules and can release them upon stimulation.[28] Recently, we demonstrated that C5aR1-induced CXCL4 release modulates revascularization.[14] Here, we present further data on the importance of C5aR1 on platelets for the modulation of tissue revascularization.

First, we analyzed C5aR1 expression on murine platelets ([Fig. 1A]). Then, we assessed receptor expression levels on platelets in a cohort of peripheral artery disease patients, which were not symptomatic. Previously, this condition has been linked to improved formation of collateral vessels, and presumably thereby lack of pain symptoms.[29] Interestingly, we observed an increased expression of C5aR1 on platelets of patients with an asymptomatic disease ([Fig. 1B]). This led us to investigate the platelet C5aR1 in the hindlimb ischemia model. In this mouse model of ischemic disease, the femoral artery is ligated to induce tissue ischemia (see [Supplementary Materials] for further details). One week after induction of tissue ischemia, the distal gastrocnemius muscle was explanted and processed for immunofluorescence microscopy. Co-staining of CD42b (red, “a platelet marker”) and C5aR1 (green) revealed expression of C5aR1 on DAPI (blue)-negative platelets in the ischemic tissue ([Fig. 1C]). To decipher the role of platelets in a controlled proangiogenic environment, we used a further mouse model, the Matrigel plug assay, whereby a gel-like substance is injected into the skin of mice forming an extracellular-space-like matrix. If the matrix is supplemented with vascular growth factors such as bFGF, vessels grow into the plugs, which can then be quantified. We injected Matrigel supplemented with bFGF and freshly isolated platelets with (wild type [WT]) or without C5aR1 (C5aR1−/−) into C57/Bl6 mice. Addition of platelets to the Matrigel resulted in reduced bFGF-mediated angiogenesis ([Fig. 1D]). Interestingly, presence of C5aR1 on WT platelets was associated with a reduced extent of vessel growth into the plugs and supplementation with C5aR1−/− platelets resulted in significantly more revascularization ([Fig. 1E]).

Zoom Image
Fig. 1 (A) Washed murine platelets were stained with a C5aR1-specific antibody or isotype control. Displayed is a histogram representative of four independent platelet samples. (B) In patients with peripheral artery disease (PAD), C5aR1 expression was increased in asymptomatic disease versus symptomatic PAD patients at Fontaine stage IIb. Shown are representative images of 20 patients. (C) Immunofluorescence co-staining of ischemic murine hindlimb gastrocnemius muscle sections at 630× magnification showing colocalization of the platelet markers CD42b (red) and C5aR1 (green) 1 week after induction of ischemia. Representative images confirm that the cells are anuclear (DAPI-negative, blue). Arrows point to platelets. Scale bars represent 5 µm. (D) In vivo Matrigel plug assay experiments were performed using WT mice. Matrigel was supplemented with bFGF and freshly isolated platelets from WT mice or vehicle control. After 7 days, Matrigels were explanted, and vessel density way assessed by H&E staining. We observed decreased vessel growth after addition of isolated platelets. Data are displayed as % of control. n = 6–7 plugs were analyzed. The group with vehicle control represents 100%. *p < 0.05. (E) Similarly, Matrigel was supplemented with bFGF and freshly isolated platelets from WT or C5aR1−/− mice were additionally injected. After 7 days, Matrigels were explanted, and vessel density was assessed by H&E staining. We observed increased vessel growth in the absence of the C5aR1 on platelets. Data are displayed as % of control. n = 7 plugs were analyzed. Data are displayed as % of control. The group with WT mice represents 100%. *p < 0.05. (F) Mice bearing a platelet-specific knockout of C5aR1 on platelets were generated (PF4-cre+C5aR1fl/fl). PF4-cre+C5aR1fl/fl and PF4-creC5aR1fl/fl mice were subjected to hindlimb ischemia and analyzed over 2 weeks. Revascularization of the hindlimbs after femoral artery ligation was visualized by laser Doppler fluximetry (LDI). We found increased revascularization in PF4-cre+C5aR1fl/fl compared with PF4-creC5aR1fl/fl animals, although this reached statistical significance only at some time points. Data are shown as the mean ± SEM (n = 3 animals per group) and are displayed as % of the perfusion in the contralateral control limb. *p < 0.05. (G) As prior experiments suggested a platelet-mediated mechanism of C5aR1 control of revascularization, we stimulated washed human platelets with C5a. The supernatant was analyzed by ELISA for the level of CXCL4. Preincubation with the C5aR1 inhibitor PMX205 inhibited C5a-induced CXCL4 secretion. Data are shown as the mean ± SEM. n = 4–8. The group with WT mice represents 100%. *p < 0.05. (H) Platelets from WT mice were stimulated with C5a or control and preincubated with PMX205 or control. The supernatant was co-incubated with endothelial cells (MHEC-5T). In some groups, an anti-CXCL4 antibody was added to the endothelial cells. C5a-stimulated WT platelet supernatant inhibited endothelial tube formation, which was not detectable when C5aR1 or CXCL4 was inhibited. Data represent mean ± SEM of the total tube length. Data are displayed as % of control and the group with control stimulated supernatant treatment represents 100%. n = 10–11. *p < 0.05. (I) Representative images of endothelial tube formation show the inhibitory effect of C5a-conditioned platelet supernatant compared with control. SEM, standard error of the mean; WT, wild type.

We then created a platelet-specific C5aR1-deficient mouse strain using the cre-lox system (for details please refer to the Material and Methods section in the [Supplementary Information] [14]). We assessed platelet reactivity by stimulating diluted whole blood with collagen-related protein and measured the platelet marker β3 integrin (GPIIIa, CD61), which mediates binding to fibrinogen. There was no significant difference in between both strains ([Supplementary Fig. S1A]), suggesting that there are no effects on the fibrinogen receptor. However, addition of C5a induced some activation in platelets on the level of fibrinogen binding and activated GPIIbIIIa (measured by activation-specific PAC1 antibody), but not on P-selectin upregulation ([Supplementary Fig. S2]). These effects could be blocked by the C5aR1 antagonist PMX205 ([Supplementary Fig. S3]). In the absence of C5a ligation, WT and C5aR1−/− platelets did not display differences in binding to fibrinogen ([Supplementary Fig. S4]). We monitored revascularization over 14 days post induction of hindlimb ischemia and observed that the C5aR1flox/floxPF4cre mice show a lower degree of revascularization than the C5aR1flox/floxPF4cre+ littermates, where C5aR1 is absent in platelets ([Fig. 1F]). Thus, the platelet C5aR1 is an important modulator of revascularization.

As all previous data suggest a platelet-mediated effect of C5aR1 on platelets, we stimulated platelets from WT mice with C5a in vitro. Using an ELISA-based analysis, we could measure a release of antiangiogenic CXCL4 (platelet factor 4, PF4) from platelets upon C5a stimulation, which could not be observed when C5aR1 was blocked by the C5aR1 antagonist PMX205 ([Fig. 1G]). To verify that the observed effect is indeed mediated through CXCL4 secreted from platelets, we assessed endothelial tube formation in vitro and co-incubated cells with C5a-conditioned platelet supernatant ([Fig. 1H, I]). Confirming our hypothesis, we could show that C5a-conditioned platelet supernatant inhibited endothelial tube formation, which could be blocked by administering the C5aR1 antagonist PMX205 ([Fig. 1H]). Interestingly, a neutralizing antibody against CXCL4 did not have an additive effect on top of PMX205 in reversing the effect of C5a-conditioned platelet supernatant ([Fig. 1H]). As the collagen pathway of platelet activation has gained recent attention,[30] we excluded involvement of glycoprotein VI in C5a-mediated release of CXCL4 from platelets ([Supplementary Fig. S5]). Together, we demonstrate that C5a-mediated release of CXCL4 is one mechanism how platelets modulate vessel growth.

Here, we identified a novel mechanism for inhibition of neovascularization via CXCL4 secretion induced by platelet C5aR1. Targeting the complement system may offer novel approaches to treat patients with diseases featuring tissue ischemia and inflammation. These diseases are largely caused by atherosclerosis.[2] Interestingly, the anaphylatoxin receptors C5aR1 and C3aR are expressed in atherosclerotic plaques.[31] Importantly, in patients with atherosclerosis, expression of C3aR and C5aR shows a significant correlation with platelet activation markers.[25] [27] [32] In a mouse model, anti-C5aR1 compounds have already proven effective in enhancing revascularization.[14] The potential for the treatment of human patients, however, will first have to be evaluated in future clinical studies.

Recently, the C5a–C5aR1 axis was suggested to be an important marker of inflammation associated with COVID-19.[33] Interestingly, the platelet C5aR1 has been recognized as an important player in the complex pathophysiology of SARS-CoV-2 infection.[34] Thus, evaluating the C5aR1–CXCL4 axis in this context may improve our understanding of disease mechanisms and provide new treatment options for COVID-19 and other diseases featuring thromboinflammation and complement involvement.

Complement-active drugs are available and already in use.[35] For instance, eculizumab, which inhibits the cleavage of C5 by the C5 convertase into C5a, has been clinically established for the treatment of aHUS (atypical hemolytic uremic syndrome) and for PNH (paroxysmal nocturnal hemoglobinuria).[36] Indeed, several novel pharmacological approaches have recently been introduced, i.e., Pegcetacoplan[37] or Avacopan,[38] a C5aR1 inhibitor, to the clinic. In light of our findings, implications of treatment regimen for vascular remodeling and regeneration should be assessed in future studies and should be taken into account when designing trials on complement therapeutics.

In conclusion, understanding the crosstalk of platelets with the complement system is important to apprehend the exact role of this interplay for platelet and complement activation and resulting diseases featuring thromboinflammation.


#

No conflict of interest has been declared by the author(s).

Acknowledgment

This work was supported by the Volkswagen Foundation (Lichtenberg program), the KFO 274-Platelets – basic mechanisms and clinical implications, and the DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel (STO Projekt F280404). H.N. was supported by the Clinician Scientist Programme of the DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel as well as the Clinician Scientist Programme of the University of Lübeck. H.N. and H.F.L. were supported by an ERA PerMed 2020 JTC grant “PROGRESS. ” H.F.L. and D.D. were furthermore supported by the SFB1366/2 (project B08) of the DFG and the DZHK partner site Mannheim/Heidelberg.

  • References

  • 1 Roth GA, Johnson C, Abajobir A. et al. Global, regional, and national burden of cardiovascular diseases for 10 causes, 1990 to 2015. J Am Coll Cardiol 2017; 70 (01) 1-25
  • 2 Galkina E, Ley K. Immune and inflammatory mechanisms of atherosclerosis (*). Annu Rev Immunol 2009; 27: 165-197
  • 3 Iyer SR, Annex BH. Therapeutic angiogenesis for peripheral artery disease: lessons learned in translational science. JACC Basic Transl Sci 2017; 2 (05) 503-512
  • 4 Noris M, Remuzzi G. Overview of complement activation and regulation. Semin Nephrol 2013; 33 (06) 479-492
  • 5 Panagiotou A, Trendelenburg M, Osthoff M. The lectin pathway of complement in myocardial ischemia/reperfusion injury-review of its significance and the potential impact of therapeutic interference by C1 esterase inhibitor. Front Immunol 2018; 9: 1151
  • 6 Yin W, Ghebrehiwet B, Weksler B, Peerschke EI. Classical pathway complement activation on human endothelial cells. Mol Immunol 2007; 44 (09) 2228-2234
  • 7 Ghebrehiwet B, CebadaMora C, Tantral L, Jesty J, Peerschke EI. gC1qR/p33 serves as a molecular bridge between the complement and contact activation systems and is an important catalyst in inflammation. Adv Exp Med Biol 2006; 586: 95-105
  • 8 Markiewski MM, Daugherity E, Reese B, Karbowniczek M. The role of complement in angiogenesis. Antibodies (Basel) 2020; 9 (04) 67
  • 9 Reis ES, Mastellos DC, Hajishengallis G, Lambris JD. New insights into the immune functions of complement. Nat Rev Immunol 2019; 19 (08) 503-516
  • 10 Nozaki M, Raisler BJ, Sakurai E. et al. Drusen complement components C3a and C5a promote choroidal neovascularization. Proc Natl Acad Sci U S A 2006; 103 (07) 2328-2333
  • 11 Langer HF, Chung KJ, Orlova VV. et al. Complement-mediated inhibition of neovascularization reveals a point of convergence between innate immunity and angiogenesis. Blood 2010; 116 (22) 4395-4403
  • 12 Del Conde I, Crúz MA, Zhang H, López JA, Afshar-Kharghan V. Platelet activation leads to activation and propagation of the complement system. J Exp Med 2005; 201 (06) 871-879
  • 13 Subramaniam S, Jurk K, Hobohm L. et al. Distinct contributions of complement factors to platelet activation and fibrin formation in venous thrombus development. Blood 2017; 129 (16) 2291-2302
  • 14 Nording H, Baron L, Haberthür D. et al. The C5a/C5a receptor 1 axis controls tissue neovascularization through CXCL4 release from platelets. Nat Commun 2021; 12 (01) 3352
  • 15 Gawaz M, Langer H, May AE. Platelets in inflammation and atherogenesis. J Clin Invest 2005; 115 (12) 3378-3384
  • 16 Schleicher RI, Reichenbach F, Kraft P. et al. Platelets induce apoptosis via membrane-bound FasL. Blood 2015; 126 (12) 1483-1493
  • 17 Kraft P, Schleicher R, Olbrich M. et al. Platelet derived FasL contributes to apoptosis in stroke. Thromb Haemost 2016; 116 (05) 998-1000
  • 18 Gaertner F, Ahmad Z, Rosenberger G. et al. Migrating platelets are mechano-scavengers that collect and bundle bacteria. Cell 2017; 171 (06) 1368-1382.e23
  • 19 Verschoor A, Neuenhahn M, Navarini AA. et al. A platelet-mediated system for shuttling blood-borne bacteria to CD8α+ dendritic cells depends on glycoprotein GPIb and complement C3. Nat Immunol 2011; 12 (12) 1194-1201
  • 20 Langer HF, Chavakis T. Leukocyte-endothelial interactions in inflammation. J Cell Mol Med 2009; 13 (07) 1211-1220
  • 21 Kapur R, Semple JW. Platelet functions beyond hemostasis. In: Molecular and Cellular Biology of Platelet Formation. Cham: Springer International Publishing; 2017: 221-237 DOI: 10.1007/978-3-319-39562-3_10
  • 22 Langer HF, Choi EY, Zhou H. et al. Platelets contribute to the pathogenesis of experimental autoimmune encephalomyelitis. Circ Res 2012; 110 (09) 1202-1210
  • 23 Huo Y, Schober A, Forlow SB. et al. Circulating activated platelets exacerbate atherosclerosis in mice deficient in apolipoprotein E. Nat Med 2003; 9 (01) 61-67
  • 24 Massberg S, Gawaz M, Grüner S. et al. A crucial role of glycoprotein VI for platelet recruitment to the injured arterial wall in vivo. J Exp Med 2003; 197 (01) 41-49
  • 25 Sauter RJ, Sauter M, Reis ES. et al. Functional relevance of the anaphylatoxin receptor C3aR for platelet function and arterial thrombus formation marks an intersection point between innate immunity and thrombosis. Circulation 2018; 138 (16) 1720-1735
  • 26 Sauter RJ, Sauter M, Obrich M. et al. Anaphylatoxin receptor C3aR contributes to platelet function, thrombus formation and in vivo haemostasis. Thromb Haemost 2019; 119 (01) 179-182
  • 27 Patzelt J, Mueller KAL, Breuning S. et al. Expression of anaphylatoxin receptors on platelets in patients with coronary heart disease. Atherosclerosis 2015; 238 (02) 289-295
  • 28 Klement GL, Yip TT, Cassiola F. et al. Platelets actively sequester angiogenesis regulators. Blood 2009; 113 (12) 2835-2842
  • 29 Keeling AN, Carroll TJ, McDermott MM. et al. Clinical correlates of size and number of collateral vessels in peripheral artery disease. Vasc Med 2012; 17 (04) 223-230
  • 30 Martins Lima A, Martins Cavaco AC, Fraga-Silva RA, Eble JA, Stergiopulos N. From patients to platelets and back again: pharmacological approaches to glycoprotein VI, a thrilling antithrombotic target with minor bleeding risks. Thromb Haemost 2019; 119 (11) 1720-1739
  • 31 Oksjoki R, Laine P, Helske S. et al. Receptors for the anaphylatoxins C3a and C5a are expressed in human atherosclerotic coronary plaques. Atherosclerosis 2007; 195 (01) 90-99
  • 32 Nording H, Giesser A, Patzelt J. et al. Platelet bound oxLDL shows an inverse correlation with plasma anaphylatoxin C5a in patients with coronary artery disease. Platelets 2016; 27 (06) 593-597
  • 33 Carvelli J, Demaria O, Vély F. et al; Explore COVID-19 IPH group, Explore COVID-19 Marseille Immunopole group. Association of COVID-19 inflammation with activation of the C5a-C5aR1 axis. Nature 2020; 588 (7836) 146-150
  • 34 Aiello S, Gastoldi S, Galbusera M. et al. C5a and C5aR1 are key drivers of microvascular platelet aggregation in clinical entities spanning from aHUS to COVID-19. Blood Adv 2022; 6 (03) 866-881
  • 35 Mastellos DC, Ricklin D, Lambris JD. Clinical promise of next-generation complement therapeutics. Nat Rev Drug Discov 2019; 18 (09) 707-729
  • 36 Rother RP, Rollins SA, Mojcik CF, Brodsky RA, Bell L. Discovery and development of the complement inhibitor eculizumab for the treatment of paroxysmal nocturnal hemoglobinuria. Nat Biotechnol 2007; 25 (11) 1256-1264
  • 37 Hillmen P, Szer J, Weitz I. et al. Pegcetacoplan versus eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med 2021; 384 (11) 1028-1037
  • 38 Jayne DRW, Merkel PA, Schall TJ, Bekker P. ADVOCATE Study Group. Avacopan for the treatment of ANCA-associated vasculitis. N Engl J Med 2021; 384 (07) 599-609

Address for correspondence

Henry Nording, MD
University Hospital, Medical Clinic II, University Heart Center Lübeck
Ratzeburger Allee 160, 23538 Lübeck
Germany   
Harald F. Langer, MD
Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University
Mannheim
Germany   

Publication History

Accepted Manuscript online:
18 August 2023

Article published online:
19 October 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Roth GA, Johnson C, Abajobir A. et al. Global, regional, and national burden of cardiovascular diseases for 10 causes, 1990 to 2015. J Am Coll Cardiol 2017; 70 (01) 1-25
  • 2 Galkina E, Ley K. Immune and inflammatory mechanisms of atherosclerosis (*). Annu Rev Immunol 2009; 27: 165-197
  • 3 Iyer SR, Annex BH. Therapeutic angiogenesis for peripheral artery disease: lessons learned in translational science. JACC Basic Transl Sci 2017; 2 (05) 503-512
  • 4 Noris M, Remuzzi G. Overview of complement activation and regulation. Semin Nephrol 2013; 33 (06) 479-492
  • 5 Panagiotou A, Trendelenburg M, Osthoff M. The lectin pathway of complement in myocardial ischemia/reperfusion injury-review of its significance and the potential impact of therapeutic interference by C1 esterase inhibitor. Front Immunol 2018; 9: 1151
  • 6 Yin W, Ghebrehiwet B, Weksler B, Peerschke EI. Classical pathway complement activation on human endothelial cells. Mol Immunol 2007; 44 (09) 2228-2234
  • 7 Ghebrehiwet B, CebadaMora C, Tantral L, Jesty J, Peerschke EI. gC1qR/p33 serves as a molecular bridge between the complement and contact activation systems and is an important catalyst in inflammation. Adv Exp Med Biol 2006; 586: 95-105
  • 8 Markiewski MM, Daugherity E, Reese B, Karbowniczek M. The role of complement in angiogenesis. Antibodies (Basel) 2020; 9 (04) 67
  • 9 Reis ES, Mastellos DC, Hajishengallis G, Lambris JD. New insights into the immune functions of complement. Nat Rev Immunol 2019; 19 (08) 503-516
  • 10 Nozaki M, Raisler BJ, Sakurai E. et al. Drusen complement components C3a and C5a promote choroidal neovascularization. Proc Natl Acad Sci U S A 2006; 103 (07) 2328-2333
  • 11 Langer HF, Chung KJ, Orlova VV. et al. Complement-mediated inhibition of neovascularization reveals a point of convergence between innate immunity and angiogenesis. Blood 2010; 116 (22) 4395-4403
  • 12 Del Conde I, Crúz MA, Zhang H, López JA, Afshar-Kharghan V. Platelet activation leads to activation and propagation of the complement system. J Exp Med 2005; 201 (06) 871-879
  • 13 Subramaniam S, Jurk K, Hobohm L. et al. Distinct contributions of complement factors to platelet activation and fibrin formation in venous thrombus development. Blood 2017; 129 (16) 2291-2302
  • 14 Nording H, Baron L, Haberthür D. et al. The C5a/C5a receptor 1 axis controls tissue neovascularization through CXCL4 release from platelets. Nat Commun 2021; 12 (01) 3352
  • 15 Gawaz M, Langer H, May AE. Platelets in inflammation and atherogenesis. J Clin Invest 2005; 115 (12) 3378-3384
  • 16 Schleicher RI, Reichenbach F, Kraft P. et al. Platelets induce apoptosis via membrane-bound FasL. Blood 2015; 126 (12) 1483-1493
  • 17 Kraft P, Schleicher R, Olbrich M. et al. Platelet derived FasL contributes to apoptosis in stroke. Thromb Haemost 2016; 116 (05) 998-1000
  • 18 Gaertner F, Ahmad Z, Rosenberger G. et al. Migrating platelets are mechano-scavengers that collect and bundle bacteria. Cell 2017; 171 (06) 1368-1382.e23
  • 19 Verschoor A, Neuenhahn M, Navarini AA. et al. A platelet-mediated system for shuttling blood-borne bacteria to CD8α+ dendritic cells depends on glycoprotein GPIb and complement C3. Nat Immunol 2011; 12 (12) 1194-1201
  • 20 Langer HF, Chavakis T. Leukocyte-endothelial interactions in inflammation. J Cell Mol Med 2009; 13 (07) 1211-1220
  • 21 Kapur R, Semple JW. Platelet functions beyond hemostasis. In: Molecular and Cellular Biology of Platelet Formation. Cham: Springer International Publishing; 2017: 221-237 DOI: 10.1007/978-3-319-39562-3_10
  • 22 Langer HF, Choi EY, Zhou H. et al. Platelets contribute to the pathogenesis of experimental autoimmune encephalomyelitis. Circ Res 2012; 110 (09) 1202-1210
  • 23 Huo Y, Schober A, Forlow SB. et al. Circulating activated platelets exacerbate atherosclerosis in mice deficient in apolipoprotein E. Nat Med 2003; 9 (01) 61-67
  • 24 Massberg S, Gawaz M, Grüner S. et al. A crucial role of glycoprotein VI for platelet recruitment to the injured arterial wall in vivo. J Exp Med 2003; 197 (01) 41-49
  • 25 Sauter RJ, Sauter M, Reis ES. et al. Functional relevance of the anaphylatoxin receptor C3aR for platelet function and arterial thrombus formation marks an intersection point between innate immunity and thrombosis. Circulation 2018; 138 (16) 1720-1735
  • 26 Sauter RJ, Sauter M, Obrich M. et al. Anaphylatoxin receptor C3aR contributes to platelet function, thrombus formation and in vivo haemostasis. Thromb Haemost 2019; 119 (01) 179-182
  • 27 Patzelt J, Mueller KAL, Breuning S. et al. Expression of anaphylatoxin receptors on platelets in patients with coronary heart disease. Atherosclerosis 2015; 238 (02) 289-295
  • 28 Klement GL, Yip TT, Cassiola F. et al. Platelets actively sequester angiogenesis regulators. Blood 2009; 113 (12) 2835-2842
  • 29 Keeling AN, Carroll TJ, McDermott MM. et al. Clinical correlates of size and number of collateral vessels in peripheral artery disease. Vasc Med 2012; 17 (04) 223-230
  • 30 Martins Lima A, Martins Cavaco AC, Fraga-Silva RA, Eble JA, Stergiopulos N. From patients to platelets and back again: pharmacological approaches to glycoprotein VI, a thrilling antithrombotic target with minor bleeding risks. Thromb Haemost 2019; 119 (11) 1720-1739
  • 31 Oksjoki R, Laine P, Helske S. et al. Receptors for the anaphylatoxins C3a and C5a are expressed in human atherosclerotic coronary plaques. Atherosclerosis 2007; 195 (01) 90-99
  • 32 Nording H, Giesser A, Patzelt J. et al. Platelet bound oxLDL shows an inverse correlation with plasma anaphylatoxin C5a in patients with coronary artery disease. Platelets 2016; 27 (06) 593-597
  • 33 Carvelli J, Demaria O, Vély F. et al; Explore COVID-19 IPH group, Explore COVID-19 Marseille Immunopole group. Association of COVID-19 inflammation with activation of the C5a-C5aR1 axis. Nature 2020; 588 (7836) 146-150
  • 34 Aiello S, Gastoldi S, Galbusera M. et al. C5a and C5aR1 are key drivers of microvascular platelet aggregation in clinical entities spanning from aHUS to COVID-19. Blood Adv 2022; 6 (03) 866-881
  • 35 Mastellos DC, Ricklin D, Lambris JD. Clinical promise of next-generation complement therapeutics. Nat Rev Drug Discov 2019; 18 (09) 707-729
  • 36 Rother RP, Rollins SA, Mojcik CF, Brodsky RA, Bell L. Discovery and development of the complement inhibitor eculizumab for the treatment of paroxysmal nocturnal hemoglobinuria. Nat Biotechnol 2007; 25 (11) 1256-1264
  • 37 Hillmen P, Szer J, Weitz I. et al. Pegcetacoplan versus eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med 2021; 384 (11) 1028-1037
  • 38 Jayne DRW, Merkel PA, Schall TJ, Bekker P. ADVOCATE Study Group. Avacopan for the treatment of ANCA-associated vasculitis. N Engl J Med 2021; 384 (07) 599-609

Zoom Image
Fig. 1 (A) Washed murine platelets were stained with a C5aR1-specific antibody or isotype control. Displayed is a histogram representative of four independent platelet samples. (B) In patients with peripheral artery disease (PAD), C5aR1 expression was increased in asymptomatic disease versus symptomatic PAD patients at Fontaine stage IIb. Shown are representative images of 20 patients. (C) Immunofluorescence co-staining of ischemic murine hindlimb gastrocnemius muscle sections at 630× magnification showing colocalization of the platelet markers CD42b (red) and C5aR1 (green) 1 week after induction of ischemia. Representative images confirm that the cells are anuclear (DAPI-negative, blue). Arrows point to platelets. Scale bars represent 5 µm. (D) In vivo Matrigel plug assay experiments were performed using WT mice. Matrigel was supplemented with bFGF and freshly isolated platelets from WT mice or vehicle control. After 7 days, Matrigels were explanted, and vessel density way assessed by H&E staining. We observed decreased vessel growth after addition of isolated platelets. Data are displayed as % of control. n = 6–7 plugs were analyzed. The group with vehicle control represents 100%. *p < 0.05. (E) Similarly, Matrigel was supplemented with bFGF and freshly isolated platelets from WT or C5aR1−/− mice were additionally injected. After 7 days, Matrigels were explanted, and vessel density was assessed by H&E staining. We observed increased vessel growth in the absence of the C5aR1 on platelets. Data are displayed as % of control. n = 7 plugs were analyzed. Data are displayed as % of control. The group with WT mice represents 100%. *p < 0.05. (F) Mice bearing a platelet-specific knockout of C5aR1 on platelets were generated (PF4-cre+C5aR1fl/fl). PF4-cre+C5aR1fl/fl and PF4-creC5aR1fl/fl mice were subjected to hindlimb ischemia and analyzed over 2 weeks. Revascularization of the hindlimbs after femoral artery ligation was visualized by laser Doppler fluximetry (LDI). We found increased revascularization in PF4-cre+C5aR1fl/fl compared with PF4-creC5aR1fl/fl animals, although this reached statistical significance only at some time points. Data are shown as the mean ± SEM (n = 3 animals per group) and are displayed as % of the perfusion in the contralateral control limb. *p < 0.05. (G) As prior experiments suggested a platelet-mediated mechanism of C5aR1 control of revascularization, we stimulated washed human platelets with C5a. The supernatant was analyzed by ELISA for the level of CXCL4. Preincubation with the C5aR1 inhibitor PMX205 inhibited C5a-induced CXCL4 secretion. Data are shown as the mean ± SEM. n = 4–8. The group with WT mice represents 100%. *p < 0.05. (H) Platelets from WT mice were stimulated with C5a or control and preincubated with PMX205 or control. The supernatant was co-incubated with endothelial cells (MHEC-5T). In some groups, an anti-CXCL4 antibody was added to the endothelial cells. C5a-stimulated WT platelet supernatant inhibited endothelial tube formation, which was not detectable when C5aR1 or CXCL4 was inhibited. Data represent mean ± SEM of the total tube length. Data are displayed as % of control and the group with control stimulated supernatant treatment represents 100%. n = 10–11. *p < 0.05. (I) Representative images of endothelial tube formation show the inhibitory effect of C5a-conditioned platelet supernatant compared with control. SEM, standard error of the mean; WT, wild type.