Thromb Haemost 2022; 122(11): 1858-1868
DOI: 10.1055/a-1910-4538
Review Article

Current View on the Molecular Mechanisms Underlying Fibrin(ogen)-Dependent Inflammation

Sergiy Yakovlev
1   Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States
2   Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
,
Dudley K. Strickland
1   Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States
3   Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
4   Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States
,
Leonid Medved
1   Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States
2   Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
› Author Affiliations
Funding This work was supported by National Institutes of Health grants R01 HL056051 to L.M. and R35 HL135743 to D.K.S.

Abstract

Numerous studies have revealed the involvement of fibrinogen in the inflammatory response. To explain the molecular mechanisms underlying fibrinogen-dependent inflammation, two bridging mechanisms have been proposed in which fibrin(ogen) bridges leukocytes to endothelial cells. The first mechanism suggests that bridging occurs via the interaction of fibrinogen with the leukocyte receptor Mac-1 and the endothelial receptor ICAM-1 (intercellular adhesion molecule-1), which promotes leukocyte transmigration and enhances inflammation. The second mechanism includes bridging of leukocytes to the endothelium by fibrin degradation product E1 fragment through its interaction with leukocyte receptor CD11c and endothelial VE-cadherin to promote leukocyte transmigration. The role of E1 in promoting inflammation is inhibited by the fibrin-derived β15–42 fragment, and this has been suggested to result from its ability to compete for the E1–VE-cadherin interaction and to trigger signaling pathways through the src kinase Fyn. Our recent study revealed that the β15–42 fragment is ineffective in inhibiting the E1– or fibrin–VE-cadherin interaction, leaving the proposed signaling mechanism as the only viable explanation for the inhibitory function of β15–42. We have discovered that fibrin interacts with the very-low-density lipoprotein (VLDL) receptor, and this interaction triggers a signaling pathway that promotes leukocyte transmigration through inhibition of the src kinase Fyn. This pathway is inhibited by another pathway induced by the interaction of β15–42 with a putative endothelial receptor. In this review, we briefly describe the previously proposed molecular mechanisms underlying fibrin-dependent inflammation and their advantages/disadvantages and summarize our recent studies of the novel VLDL receptor-dependent pathway of leukocyte transmigration which plays an important role in fibrin-dependent inflammation.



Publication History

Received: 23 May 2022

Accepted: 22 July 2022

Accepted Manuscript online:
27 July 2022

Article published online:
17 October 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Riddle JM, Barnhart MI. Ultrastructural study of fibrin dissolution via emigrated polymorphonuclear neutrophils. Am J Pathol 1964; 45 (05) 805-823
  • 2 Riddle JM, Barnhart MI. The eosinophil as a source for profibrinolysin in acute inflammation. Blood 1965; 25 (05) 776-794
  • 3 Barnhart MI, Riddle JM, Bluhm GB, Quintana C. Fibrin promotion and lysis in arthritic joints. Ann Rheum Dis 1967; 26 (03) 206-218
  • 4 Colvin RB, Johnson RA, Mihm Jr MC, Dvorak HF. Role of the clotting system in cell-mediated hypersensitivity. I. Fibrin deposition in delayed skin reactions in man. J Exp Med 1973; 138 (03) 686-698
  • 5 McRitchie DI, Girotti MJ, Glynn MF, Goldberg JM, Rotstein OD. Effect of systemic fibrinogen depletion on intraabdominal abscess formation. J Lab Clin Med 1991; 118 (01) 48-55
  • 6 Tang L, Eaton JW. Fibrin(ogen) mediates acute inflammatory responses to biomaterials. J Exp Med 1993; 178 (06) 2147-2156
  • 7 Hurley JV. Substances promoting leukocyte emigration. Ann N Y Acad Sci 1964; 116 (03) 918-935
  • 8 Jennewein C, Tran N, Paulus P, Ellinghaus P, Eble JA, Zacharowski K. Novel aspects of fibrin(ogen) fragments during inflammation. Mol Med 2011; 17 (5-6): 568-573
  • 9 Farrell DH. γ′ Fibrinogen as a novel marker of thrombotic disease. Clin Chem Lab Med 2012; 50 (11) 1903-1909
  • 10 Rein-Smith CM, Anderson NW, Farrell DH. Differential regulation of fibrinogen γ chain splice isoforms by interleukin-6. Thromb Res 2013; 131 (01) 89-93
  • 11 Ugarova TP, Solovjov DA, Zhang L. et al. Identification of a novel recognition sequence for integrin αMβ2 within the γ-chain of fibrinogen. J Biol Chem 1998; 273 (35) 22519-22527
  • 12 Luyendyk JP, Schoenecker JG, Flick MJ. The multifaceted role of fibrinogen in tissue injury and inflammation. Blood 2019; 133 (06) 511-520
  • 13 Petersen MA, Ryu JK, Akassoglou K. Fibrinogen in neurological diseases: mechanisms, imaging and therapeutics. Nat Rev Neurosci 2018; 19 (05) 283-301
  • 14 Akassoglou K. The immunology of blood: connecting the dots at the neurovascular interface. Nat Immunol 2020; 21 (07) 710-712
  • 15 Henschen A, McDonagh J. Fibrinogen, fibrin and factor XIII. In: Zwaal RFA, Hemker HC. eds. Blood Coagulation. Amsterdam: Elsevier Science Publishers; 1986: 171-241
  • 16 Medved L, Weisel JW. Fibrinogen and Factor XIII Subcommittee of Scientific Standardization Committee of International Society on Thrombosis and Haemostasis. Recommendations for nomenclature on fibrinogen and fibrin. J Thromb Haemost 2009; 7 (02) 355-359
  • 17 Kollman JM, Pandi L, Sawaya MR, Riley M, Doolittle RF. Crystal structure of human fibrinogen. Biochemistry 2009; 48 (18) 3877-3886
  • 18 Olexa SA, Budzynski AZ. Primary soluble plasmic degradation product of human cross-linked fibrin. Isolation and stoichiometry of the (DD)E complex. Biochemistry 1979; 18 (06) 991-995
  • 19 Moskowitz KA, Budzynski AZ. The (DD)E complex is maintained by a composite fibrin polymerization site. Biochemistry 1994; 33 (44) 12937-12944
  • 20 Yakovlev S, Makogonenko E, Kurochkina N, Nieuwenhuizen W, Ingham K, Medved L. Conversion of fibrinogen to fibrin: mechanism of exposure of tPA- and plasminogen-binding sites. Biochemistry 2000; 39 (51) 15730-15741
  • 21 Olexa SA, Budzynski AZ. Binding phenomena of isolated unique plasmic degradation products of human cross-linked fibrin. J Biol Chem 1979; 254 (11) 4925-4932
  • 22 Carlos TM, Harlan JM. Leukocyte-endothelial adhesion molecules. Blood 1994; 84 (07) 2068-2101
  • 23 Springer TA. Traffic signals on endothelium for lymphocyte recirculation and leukocyte emigration. Annu Rev Physiol 1995; 57: 827-872
  • 24 Languino LR, Plescia J, Duperray A. et al. Fibrinogen mediates leukocyte adhesion to vascular endothelium through an ICAM-1-dependent pathway. Cell 1993; 73 (07) 1423-1434
  • 25 Altieri DC, Bader R, Mannucci PM, Edgington TS. Oligospecificity of the cellular adhesion receptor Mac-1 encompasses an inducible recognition specificity for fibrinogen. J Cell Biol 1988; 107 (05) 1893-1900
  • 26 Trezzini C, Jungi TW, Kuhnert P, Peterhans E. Fibrinogen association with human monocytes: evidence for constitutive expression of fibrinogen receptors and for involvement of Mac-1 (CD18, CR3) in the binding. Biochem Biophys Res Commun 1988; 156 (01) 477-484
  • 27 Wright SD, Weitz JI, Huang AJ, Levin SM, Silverstein SC, Loike JD. Complement receptor type three (CD11b/CD18) of human polymorphonuclear leukocytes recognizes fibrinogen. Proc Natl Acad Sci U S A 1988; 85 (20) 7734-7738
  • 28 Altieri DC. Regulation of leukocyte-endothelium interaction by fibrinogen. Thromb Haemost 1999; 82 (02) 781-786
  • 29 Sans E, Delachanal E, Duperray A. Analysis of the roles of ICAM-1 in neutrophil transmigration using a reconstituted mammalian cell expression model: implication of ICAM-1 cytoplasmic domain and Rho-dependent signaling pathway. J Immunol 2001; 166 (01) 544-551
  • 30 Roche Y, Pasquier D, Rambeaud JJ, Seigneurin D, Duperray A. Fibrinogen mediates bladder cancer cell migration in an ICAM-1-dependent pathway. Thromb Haemost 2003; 89 (06) 1089-1097
  • 31 Adamson P, Etienne S, Couraud PO, Calder V, Greenwood J. Lymphocyte migration through brain endothelial cell monolayers involves signaling through endothelial ICAM-1 via a Rho-dependent pathway. J Immunol 1999; 162 (05) 2964-2973
  • 32 Lishko VK, Kudryk B, Yakubenko VP, Yee VC, Ugarova TP. Regulated unmasking of the cryptic binding site for integrin αMβ2 in the γ C-domain of fibrinogen. Biochemistry 2002; 41 (43) 12942-12951
  • 33 Yakovlev S, Zhang L, Ugarova T, Medved L. Interaction of fibrin(ogen) with leukocyte receptor αMβ2 (Mac-1): further characterization and identification of a novel binding region within the central domain of the fibrinogen γ-module. Biochemistry 2005; 44 (02) 617-626
  • 34 Yakovlev S, Medved L. Effect of fibrinogen, fibrin, and fibrin degradation products on transendothelial migration of leukocytes. Thromb Res 2018; 162: 93-100
  • 35 Petzelbauer P, Zacharowski PA, Miyazaki Y. et al. The fibrin-derived peptide Bβ15–42 protects the myocardium against ischemia-reperfusion injury. Nat Med 2005; 11 (03) 298-304
  • 36 Zacharowski K, Zacharowski P, Reingruber S, Petzelbauer P. Fibrin(ogen) and its fragments in the pathophysiology and treatment of myocardial infarction. J Mol Med (Berl) 2006; 84 (06) 469-477
  • 37 Blombäck B, Blombäck M, Henschen A, Hessel B, Iwanaga S, Woods KR. N-terminal disulphide knot of human fibrinogen. Nature 1968; 218 (5137): 130-134
  • 38 Medved L, Yakovlev S. Structure and function of fibrinogen BβN-domains. Ukr Biochem J 2020; 92 (03) 22-32
  • 39 Loike JD, Sodeik B, Cao L. et al. CD11c/CD18 on neutrophils recognizes a domain at the N terminus of the Aα chain of fibrinogen. Proc Natl Acad Sci U S A 1991; 88 (03) 1044-1048
  • 40 Chalupowicz DG, Chowdhury ZA, Bach TL, Barsigian C, Martinez J. Fibrin II induces endothelial cell capillary tube formation. J Cell Biol 1995; 130 (01) 207-215
  • 41 Martinez J, Ferber A, Bach TL, Yaen CH. Interaction of fibrin with VE-cadherin. Ann N Y Acad Sci 2001; 936 (01) 386-405
  • 42 Zacharowski K, Zacharowski PA, Friedl P. et al. The effects of the fibrin-derived peptide Bβ15–42 in acute and chronic rodent models of myocardial ischemia-reperfusion. Shock 2007; 27 (06) 631-637
  • 43 Roesner JP, Petzelbauer P, Koch A. et al. The fibrin-derived peptide Bβ15–42 is cardioprotective in a pig model of myocardial ischemia-reperfusion injury. Crit Care Med 2007; 35 (07) 1730-1735
  • 44 Ahrens I, Peter K. FX-06, a fibrin-derived Bβ15–42 peptide for the potential treatment of reperfusion injury following myocardial infarction. Curr Opin Investig Drugs 2009; 10 (09) 997-1003
  • 45 Atar D, Petzelbauer P, Schwitter J. et al; F.I.R.E. Investigators. Effect of intravenous FX06 as an adjunct to primary percutaneous coronary intervention for acute ST-segment elevation myocardial infarction results of the F.I.R.E. (Efficacy of FX06 in the Prevention of Myocardial Reperfusion Injury) trial. J Am Coll Cardiol 2009; 53 (08) 720-729
  • 46 Hallén J, Petzelbauer P, Schwitter J, Geudelin B, Buser P, Atar D. Impact of time to therapy and presence of collaterals on the efficacy of FX06 in acute ST elevation myocardial infarction: a substudy of the F.I.R.E., the Efficacy of FX06 in the prevention of myocardial reperfusion injury trial. EuroIntervention 2010; 5 (08) 946-952
  • 47 Gorlatov S, Medved L. Interaction of fibrin(ogen) with the endothelial cell receptor VE-cadherin: mapping of the receptor-binding site in the NH2-terminal portions of the fibrin β chains. Biochemistry 2002; 41 (12) 4107-4116
  • 48 Yakovlev S, Gao Y, Cao C. et al. Interaction of fibrin with VE-cadherin and anti-inflammatory effect of fibrin-derived fragments. J Thromb Haemost 2011; 9 (09) 1847-1855
  • 49 Roesner JP, Petzelbauer P, Koch A. et al. Bβ15–42 (FX06) reduces pulmonary, myocardial, liver, and small intestine damage in a pig model of hemorrhagic shock and reperfusion. Crit Care Med 2009; 37 (02) 598-605
  • 50 Roesner JP, Petzelbauer P, Koch A. et al. A double blind, single centre, sub-chronic reperfusion trial evaluating FX06 following haemorrhagic shock in pigs. Resuscitation 2009; 80 (02) 264-271
  • 51 Wiedemann D, Schneeberger S, Friedl P. et al. The fibrin-derived peptide Bβ15–42 significantly attenuates ischemia-reperfusion injury in a cardiac transplant model. Transplantation 2010; 89 (07) 824-829
  • 52 Sörensen I, Rong S, Susnik N. et al. Bβ15–42 attenuates the effect of ischemia-reperfusion injury in renal transplantation. J Am Soc Nephrol 2011; 22 (10) 1887-1896
  • 53 Krishnamoorthy A, Ajay AK, Hoffmann D. et al. Fibrinogen β-derived Bβ15–42 peptide protects against kidney ischemia/reperfusion injury. Blood 2011; 118 (07) 1934-1942
  • 54 Liu A, Fang H, Yang Y. et al. The fibrin-derived peptide Bβ15-42 attenuates liver damage in a rat model of liver ischemia/reperfusion injury. Shock 2013; 39 (04) 397-403
  • 55 Urbschat A, Rupprecht K, Zacharowski K. et al. Combined peri-ischemic administration of Bβ15-42 in treating ischemia reperfusion injury of the mouse kidney. Microvasc Res 2015; 101: 48-54
  • 56 Matt U, Warszawska JM, Bauer M. et al. Bβ15–42 protects against acid-induced acute lung injury and secondary pseudomonas pneumonia in vivo. Am J Respir Crit Care Med 2009; 180 (12) 1208-1217
  • 57 Gröger M, Pasteiner W, Ignatyev G. et al. Peptide Bβ15–42 preserves endothelial barrier function in shock. PLoS One 2009; 4 (04) e5391
  • 58 Bergt S, Gruenewald M, Beltschany C. et al. The fibrin-derived peptide Bβ15–42 (FX06) ameliorates vascular leakage and improves survival and neurocognitive recovery: Implications from two animal models of cardiopulmonary resuscitation. Crit Care Med 2016; 44 (10) e988-e995
  • 59 Wolf T, Kann G, Becker S. et al. Severe Ebola virus disease with vascular leakage and multiorgan failure: treatment of a patient in intensive care. Lancet 2015; 385 (9976): 1428-1435
  • 60 Holinstat M, Knezevic N, Broman M, Samarel AM, Malik AB, Mehta D. Suppression of RhoA activity by focal adhesion kinase-induced activation of p190RhoGAP: role in regulation of endothelial permeability. J Biol Chem 2006; 281 (04) 2296-2305
  • 61 Yakovlev S, Medved L. Interaction of fibrin(ogen) with the endothelial cell receptor VE-cadherin: localization of the fibrin-binding site within the third extracellular VE-cadherin domain. Biochemistry 2009; 48 (23) 5171-5179
  • 62 Yakovlev S, Cao C, Galisteo R, Zhang L, Strickland DK, Medved L. Fibrin-VLDL receptor-dependent pathway promotes leukocyte transmigration by inhibiting Src kinase Fyn and is a target for fibrin β15–42 peptide. Thromb Haemost 2019; 119 (11) 1816-1826
  • 63 Yakovlev S, Mikhailenko I, Cao C, Zhang L, Strickland DK, Medved L. Identification of VLDLR as a novel endothelial cell receptor for fibrin that modulates fibrin-dependent transendothelial migration of leukocytes. Blood 2012; 119 (02) 637-644
  • 64 Herz J, Goldstein JL, Strickland DK, Ho YK, Brown MS. 39-kDa protein modulates binding of ligands to low density lipoprotein receptor-related protein/α2-macroglobulin receptor. J Biol Chem 1991; 266 (31) 21232-21238
  • 65 Williams SE, Ashcom JD, Argraves WS, Strickland DK. A novel mechanism for controlling the activity of α2-macroglobulin receptor/low density lipoprotein receptor-related protein. Multiple regulatory sites for 39-kDa receptor-associated protein. J Biol Chem 1992; 267 (13) 9035-9040
  • 66 Yakovlev S, Medved L. Interaction of fibrin with the very low density lipoprotein receptor: further characterization and localization of the fibrin-binding site. Biochemistry 2015; 54 (30) 4751-4761
  • 67 Yakovlev S, Medved L. Interaction of fibrin with the very low-density lipoprotein (VLDL) receptor: further characterization and localization of the VLDL receptor-binding site in fibrin βN-domains. Biochemistry 2017; 56 (19) 2518-2528
  • 68 Banerjee K, Yakovlev S, Gruschus JM, Medved L, Tjandra N. Nuclear magnetic resonance solution structure of the recombinant fragment containing three fibrin-binding cysteine-rich domains of the very low density lipoprotein receptor. Biochemistry 2018; 57 (30) 4395-4403
  • 69 Gruschus JM, Yakovlev S, Banerjee K, Medved L, Tjandra N. Structural basis for the interaction of fibrin with the very low-density lipoprotein receptor revealed by NMR and site-directed mutagenesis. Biochemistry 2021; 60 (33) 2537-2548
  • 70 Fisher C, Beglova N, Blacklow SC. Structure of an LDLR-RAP complex reveals a general mode for ligand recognition by lipoprotein receptors. Mol Cell 2006; 22 (02) 277-283
  • 71 Ruiz J, Kouiavskaia D, Migliorini M. et al. The apoE isoform binding properties of the VLDL receptor reveal marked differences from LRP and the LDL receptor. J Lipid Res 2005; 46 (08) 1721-1731
  • 72 Yakovlev S, Belkin AM, Chen L. et al. Anti-VLDL receptor monoclonal antibodies inhibit fibrin-VLDL receptor interaction and reduce fibrin-dependent leukocyte transmigration. Thromb Haemost 2016; 116 (06) 1122-1130
  • 73 Yakovlev S, Medved L. Dual functions of the fibrin βN-domains in the VLDL receptor-dependent pathway of transendothelial migration of leukocytes. Thromb Res 2022; 214: 1-7
  • 74 Sörensen-Zender I, Chen R, Rong S. et al. Binding to carboxypeptidase M mediates protective effects of fibrinopeptide Bβ15-42 . Transl Res 2019; 213: 124-135
  • 75 Kirchhofer D, Tschopp TB, Hadváry P, Baumgartner HR. Endothelial cells stimulated with tumor necrosis factor-α express varying amounts of tissue factor resulting in inhomogenous fibrin deposition in a native blood flow system. Effects of thrombin inhibitors. J Clin Invest 1994; 93 (05) 2073-2083
  • 76 Kirchhofer D, Sakariassen KS, Clozel M. et al. Relationship between tissue factor expression and deposition of fibrin, platelets, and leukocytes on cultured endothelial cells under venous blood flow conditions. Blood 1993; 81 (08) 2050-2058
  • 77 Campbell RA, Overmyer KA, Bagnell CR, Wolberg AS. Cellular procoagulant activity dictates clot structure and stability as a function of distance from the cell surface. Arterioscler Thromb Vasc Biol 2008; 28 (12) 2247-2254
  • 78 Campbell RA, Overmyer KA, Selzman CH, Sheridan BC, Wolberg AS. Contributions of extravascular and intravascular cells to fibrin network formation, structure, and stability. Blood 2009; 114 (23) 4886-4896