Next Article in Journal
Integrated Microarray-Based Data Analysis of miRNA Expression Profiles: Identification of Novel Biomarkers of Cisplatin-Resistance in Testicular Germ Cell Tumours
Next Article in Special Issue
Rho GTPase Signaling in Platelet Regulation and Implication for Antiplatelet Therapies
Previous Article in Journal
DDC-Promoter-Driven Chemogenetic Activation of SNpc Dopaminergic Neurons Alleviates Parkinsonian Motor Symptoms
Previous Article in Special Issue
Dissecting Platelet’s Role in Viral Infection: A Double-Edged Effector of the Immune System
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

Microvascular Thrombosis as a Critical Factor in Severe COVID-19

by
Patricia P. Wadowski
1,2,*,
Benjamin Panzer
3,
Alicja Józkowicz
2,
Christoph W. Kopp
1,
Thomas Gremmel
4,5,
Simon Panzer
6 and
Renate Koppensteiner
1,*
1
Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
2
Department of Medical Biotechnology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
3
Department of Cardiology, Wilhelminenspital, 1160 Vienna, Austria
4
Institute of Antithrombotic Therapy in Cardiovascular Disease, Karl Landsteiner Society, 3100 St. Pölten, Austria
5
Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
6
Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, 1090 Vienna, Austria
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2492; https://doi.org/10.3390/ijms24032492
Submission received: 17 December 2022 / Revised: 21 January 2023 / Accepted: 25 January 2023 / Published: 27 January 2023
(This article belongs to the Special Issue Advances in Platelet Biology and Functions)

Abstract

:
Platelet–endothelial interactions have a critical role in microcirculatory function, which maintains tissue homeostasis. The subtle equilibrium between platelets and the vessel wall is disturbed by the coronavirus disease 2019 (COVID-19), which affects all three components of Virchow’s triad (endothelial injury, stasis and a hypercoagulable state). Endotheliitis, vasculitis, glycocalyx degradation, alterations in blood flow and viscosity, neutrophil extracellular trap formation and microparticle shedding are only few pathomechanisms contributing to endothelial damage and microthrombosis resulting in capillary plugging and tissue ischemia. In the following opinion paper, we discuss major pathological processes leading to microvascular endothelial activation and thrombosis formation as a possible major adverse factor driving the deterioration of patient disease course in severe COVID-19.

Coronavirus disease 2019 (COVID-19) emerged recently as a burden of global scope deteriorating healthcare systems [1,2,3]. COVID-19 is a prothrombotic and proinflammatory disease, with many immune mechanisms resembling other diseases, in particular (viral) infections, but with distinct changes in pathophysiological reactions [4,5]. Herein, it promotes systemic hyper-inflammation with an increased risk of thromboembolism, and the occurrence of microthrombosis is discussed as a major driver of disease severity [6]. The thrombotic response in COVID-19 patients involves all components of Virchow’s triad, including endothelial injury, stasis and a hypercoagulable state [7,8,9]. As a central mechanism of viral infection, glycocalyx shedding is discussed as being a critical and accelerating factor for viral entry (Figure 1) [10,11]. The latter can cause direct tissue injury such as endotheliitis and myocarditis [12,13,14]. Indirect cellular lesions are mediated by the immune response and hypercoagulability, where thromboinflammation is a key driver [14,15]. Microthrombi are regarded to be an underlying cause of myocyte necrosis in COVID-19 [16]. These thrombi are different in comparison to the composition of thromboemboli in COVID-19 negative subjects, having higher deposition of fibrin and the complement factor C5b-9 [16,17]. Deposits of the complement system such as C5b-9 and C4d were detected in the microvasculature of the lungs and skin [18]. Moreover, skin samples showing characteristic changes of severe COVID-19 could help to identify individuals at risk [17]. Microthrombus formation could therefore also explain the severe disease courses of COVID-19 in patients with pre-existing cardiovascular illness [19], which are especially prone to microcirculatory complications, as systemic microvascular perfusion is much lower than in healthy volunteers [20,21,22].
Endothelial cell damage due to the viral infection results in subendothelial collagen exposure initiating von Willebrand factor (vWF) binding via its A3 domain [24,25]. This promotes platelet adhesion via the glycoprotein (GP) Ib-V-IX complex, leading to platelet activation and aggregation and the induction of the coagulation cascade (further details are discussed later in the text) [26,27,28]. Platelet activation can be propagated by several agonists, the strongest of which is thrombin [29]. Thrombin binds to the protease-activated receptors (PAR) 1 and 4, and also to GP Ib, while collagen binds to the GP VI receptor on platelets [29,30]. This causes platelets to become activated, resulting in a prothrombotic cascade. In the first step of processes, arachidonic acid is converted into thromboxane A2, a vasoconstrictive agent, which has further potent proaggregatory properties [31,32]. These aforementioned influences cause the platelets to degranulate. The released dense granules are rich in ADP, which binds to the P2Y12 and P2Y1 receptors increasing platelet activation [27,33]. Alpha granules fuse with the cell membrane and release further procoagulatory and proinflammatory factors [33,34,35]. Among these factors is P-selectin, which binds to neutrophils, monocytes and other proinflammatory cells [36]. In neutrophils, the interaction with P-selectin also promotes the formation of neutrophil extracellular traps (NETs) [37]. P-selectin is expressed on activated platelets or shed into the circulation by the latter and endothelial cells [15,38]. The shedding of platelet adhesion receptors is a modulator of thromboinflammatory processes, also during COVID-19 [39].
Platelets undergo shape changes as a result of calcium mobilization and the dephosphorylation of vasodilator-stimulated phosphoprotein, causing physical platelet aggregation [40]. GP IIb/IIIa receptors link to GP IIb/IIIa receptors on other platelets making the bonds between platelets stronger [41]. This also causes nearby platelets to become activated by a mechanism called outside-in signaling [41]. Among others, vascular endothelial cells, platelets and blood cells are capable of secreting extracellular vesicles [42,43]. These vesicles contain RNA (e.g., microRNA and mRNA), cytokines, growth factors, lipids and small amounts of DNA [43,44]. Some of these extracellular vesicles expose phosphatidylserine, which has an up to 100 times greater procoagulatory property than activated platelets [45].
The other main component in thrombosis is the activation of the coagulation cascade either by tissue factor (TF; extrinsic pathway) or the intrinsic pathway [27]. Both pathways are active in COVID-19 [46,47,48] and result in factor X activation, causing thrombin generation [27]. Thrombin cleaves soluble fibrinogen and turns it into insoluble fibrin [27]. Fibrin forms interlocked strands, and this complex is stabilized by factor XIII [27]. As previously mentioned, thrombin activates platelets, but platelets play a role in activating the production of thrombin by membrane scramblase action [49,50].
Thrombin activates platelets even in subnanomolar concentrations [51]. Herein, PAR-1 is first activated, and as the thrombin concentration rises, PAR-4 activation is needed, being the predominant pathway for acceleration of platelet activation [51,52]. The signaling pathways of the PARs are still activated in patients with platelet inhibition by dual antiplatelet therapy consisting of aspirin and a P2Y12 receptor antagonist despite adequate P2Y12 receptor inhibition [53,54,55]. Other platelet activation pathways, which also play a role during SARS-CoV-2 infection, are toll-like receptors (TLRs), which are not targeted by current antiplatelet therapy [56,57].
TLR2 has been shown to play a role in sensing the E-protein of ß-coronaviruses, and blocking of TLR2 signaling resulted in a survival-benefit of SARS-CoV-2 infected mice [56]. The TLR2 pathway promotes platelet hyperreactivity and accelerates thrombosis [58]. Interestingly, TLR2 signaling upregulates the release of vWF from Weibel–Palade bodies of endothelial cells [59], and from alpha granules in megakaryocytes/platelets [60,61] vWF has a central role in microthrombosis formation during COVID-19 [62]. After sensing of the SARS-CoV-2 spike protein, TLR2 dimerizes with TLR1 or TLR6 and activates the proinflammatory NF-κB pathway [63].
TLR4 has a major role in the inflammatory response by getting activated by lipopolysaccharides (LPS). During SARS-COV-2 infection; however, the spike protein directly binds to the extracellular domains of TLR1, TLR4 and TLR6, having the strongest affinity to TLR4 [64]. Therefore, a strong inflammatory effect and platelet activation through pattern recognition of the SARS-CoV-2 spike protein by TLR4 is discussed [65] as being potentially one of the main pathophysiological pathways of microthrombosis in COVID-19. Furthermore, SARS-CoV-2 is suggested to infect enterocytes and hereby induce a disruption of the gut barrier [66]. This promotes bacterial translocation, which can be measured by increased LPS levels [66]. Consequently, platelet TLR4 activation by LPS during inflammation promotes platelet–neutrophil interactions, which induce endothelial damage and lead to NET formation [67]. Hereby platelets remain the main limiting factor in NETosis and the process occurs primarily in liver and pulmonary capillaries [67]. However, further studies on COVID-19 are necessary to explore detailed pathomechanisms.
An important fact in understanding potential therapeutic approaches is that pathways, which are not targeted by dual antiplatelet therapy (DAPT), among those PAR-1, collagen and epinephrine signaling still account for stable platelet aggregate formation [68]. The above-discussed observations might be the underlying pathomechanisms, which explain that there was no clear benefit of antiplatelet therapy in the prevention of thrombotic events in COVID-19 patients [69,70,71]. Therapeutic anticoagulation using heparin seems to be most effective in non-critically ill COVID-19 patients [72], but not in those with critical illness [73], and further strategies to overcome the processes of immunothrombosis are needed. However, patient outcome with regard to different anticoagulation treatment regimens warrants further clinical trials [72,73,74]. In addition, impaired fibrinolysis has emerged as another challenging therapeutic aspect, requiring future research [75,76,77,78]. Interference with thrombin-induced pathways could also be of potential benefit; however, the occurrence of increased bleeding events might limit therapeutic success [79,80]. Moreover, anticoagulation regimens after recovery from COVID-19 to overcome the higher risk of deep vein thrombosis and pulmonary embolism are yet not well studied and should also consider a higher bleeding risk in the first months after SARS-CoV-2 infection [81].
The inflammatory properties associated with COVID-19 infection add to the activation of platelets and the induction of the coagulation cascade. Leukocytes are recruited to the sites of inflammation, causing the release of cytokines (such as interleukin (IL) 6 and 8) among other proinflammatory substances [82,83]. Acute inflammation is associated with increased levels of fibrin strand density [84]. Increased thrombin generation promotes leukocyte recruitment and augments inflammation [85].
The upregulation of tissue factor in different cells during COVID-19, including endothelial and epithelial cells, neutrophils, monocytes and macrophages as well as platelets, links inflammation and thrombosis [46,48,86,87]. Tissue-factor-bearing extracellular vesicles are increasingly released into the circulation and activate the extrinsic coagulation pathway [48]. Extracellular vesicle tissue factor activity has been shown to be associated with thrombotic events and disease severity, circulating leukocytes and inflammatory markers, prothrombin time and d-dimer levels, vWF, ADAMTS13 (a disintegrin-like metallopeptidase with thrombospondin motif type 1 member 13), fibrinogen and plasmin–antiplasmin complexes [48,88,89]. In addition, contact activation of the coagulation system is triggered by neutrophil and complement activation [47,48].
During systemic inflammation endothelial function is deteriorated as a result of elevated vWF, which is released from Weibel–Palade bodies upon endothelial activation and from platelet alpha granules [33]. vWF enhances platelet-to-endothelium and platelet-to-platelet binding [33]. The activity of vWF is influenced by the molecular size and ultra-large vWF (UL-vWF) highly promotes thrombosis [90]. The maintenance of the equilibrium of vWF and its specific cleaving protease ADAMTS13 is a limiting factor in the process of microthrombosis, and the equilibrium is disturbed in systemic inflammation such as also severe COVID-19 [62,90,91]. A higher vWF activity is linked to in-hospital mortality of infected patients [26,91]. Moreover, a decrease in ADAMTS13 activity was associated with lower survival [91]. Therefore, as therapeutic measures caplacizumab (a bivalent single-domain antibody targeting the A1 domain of the vWF), recombinant ADAMTS13 and N-acetyl cysteine, which is known to reduce the size of vWF multimers, were suggested [26]. Autopsy results of patients with severe COVID-19 have shown the occurrence of severe capillary congestion, in part by microthrombi, some of which exhibit vWF positivity [62,92]. After release, vWF assembles into long strands and forms scaffolds to attach NETs [15,93]. Long vWF multimers and herein especially UL-vWF exhibit prothrombotic effects, contributing to glycocalyx disintegration by disturbing glycocalyx integrity, as vWF associates with heparan sulfate of syndecan-1 [62,94]. Multimeric vWF binds also to NO synthase, thereby decreasing NO availability during hypoxic conditions [95]. Moreover, NETs interact electrostatically with vWF, and this causes the retention of NETs on the endothelial surface [96,97]. In turn, a massive tissue injury is induced, which could be reduced by nearly 80% after blocking of vWF in mice [97]. One of the primary components of NETs, histones, directly induce endothelial damage during inflammation [97,98]. Histones exhibit a high content of positively charged amino acids and therefore can easily interact with the polyanions of the glycocalyx [99]. Histone accumulation is a major process driving microvessel dysfunction in acute lung injury. Hence, the design of polyanions to neutralize circulating histones was suggested to prevent acute lung injury [99]. Furthermore, histones and the DNA of NETs activate the coagulation system through promotion of thrombin generation [93]. The latter can occur through multiple pathways such as, among others, the induction of platelet activation by TLR2 and TLR4 through NETs [93]. Furthermore, histones are also capable of inducing platelet microaggregate formation in a GPIIbIIIa- independent, but fibrinogen-dependent manner [93]. The clinical benefits of heparin administration in, e.g., non-critically ill COVID-19 patients [72] might also be explained by the prevention of histone-platelet interactions, which is achieved by high heparin concentrations and was shown to protect mice from histone-induced tissue damage, thrombocytopenia and death [100].
One of the linking elements between vWF and NET formation is possibly IL-6, having a central role in the cytokine release syndrome of COVID-19 [91,101]. IL-6 is suggested to orchestrate a feedback mechanism between NETs and the vWF/ADAMTS13 axis [91]. The application of the IL-6 receptor antagonists tocilizumab and sarilumab showed to improve survival of critically ill COVID-19 patients [102]. In addition, NETosis is triggered by other cytokines, e.g., IL-1β and IL-8 [103,104]. Blockage of IL-1β by canakinumab was recently tested in patients with SARS-CoV-2 infection [105]. The ‘Canakinumab in Patients With COVID-19 and Type 2 Diabetes’ (CanCovDia) trial included 116 diabetic patients with COVID-19, which were randomized into two groups with canakinumab vs. placebo in addition to standard treatment [105]. Although canakinumab did not result in an improvement in survival, length of ICU stay or hospitalization as well as ventilation time and glycemic control was more rapidly achieved, requiring less treatment with antidiabetic drugs [105]. As shown in this trial, blocking of IL-1β might be of most advantage for patients at high risk for severe COVID-19 disease course, including those with diabetes-related complications, kidney failure and adiposity [105].
The inflammation induced by excessive NET formation might explain the progress of cardiovascular diseases during COVID-19 and other acute infections [15]. Herein, NETs promote intimal smooth muscle cell apoptosis and drive the destabilization of atherosclerotic plaques [106]. The latter promotes further changes in local hemodynamics and hypercoagulability [107].
Unfolding of vWF leads to exposure of the A2 domain, which exhibits high sequence homology with the complement factor B [62]. This enables the binding of activated complement C3b initiating the alternative complement pathway [62]. In turn, the formation of tissue-factor-enriched NETs is promoted [62].
Interference with the activity of ADAMTS13 is also conferred by thrombospondin 1 and platelet factor 4, which are secreted from activated platelets [62]. Both molecules bind to the vWF A2 domain and cause steric hindrance for ADAMTS13 binding, which prevents the cleavage of vWF [108,109].
Another pathophysiological aspect leading to higher NET release in COVID-19 is the occurrence of autoantibodies [110]. Herein, eight different types of autoantibodies targeting phospholipids and phospholipid-binding proteins (aPL antibodies) were detected during SARS-CoV-2 infection [110]. These aPL antibodies occurred in about half of the measured patients, and although assumed to be transient, they exhibited a prothrombotic potential by triggering neutrophil activation and NET release [110]. The autoimmune response leading to aPL antibody formation is complexly orchestrated by genetic predisposition, previous antigen exposures and the hyperactivation of the host immune system in response to environmental triggers, in this case SARS-CoV-2 infection [110,111].
The persistence of neutrophil activation and NET formation seems to be involved in the development of long COVID [112]. These changes can be observed even months after SARS-CoV-2 infection [112], and viral persistence is suggested to trigger repeated inflammatory responses [113]. In post-COVID-associated changes, neutrophils are discussed to promote fibrosis by protease release and to drive pulmonary symptoms [112]. In addition, NETosis is discussed to contribute to tumor growth, neurological disorders and worsening of concomitant diseases (e.g., hypertension or diabetes) after SARS-CoV-2 infection [104]. Given the involvement of NETs in (micro)thrombosis and vasculitis [114,115], it can be speculated that NETosis promotes post-COVID endotheliopathy and procoagulability. Recently, there is increasing evidence for microthrombus formation as a contributing factor to cardiopulmonary long COVID syndromes [116]. Moreover, as during acute SARS-CoV-2 infection, key factors of post-COVID sequelae are based on endothelial injury, inflammation and procoagulability [116,117]. Virus-eliminating strategies might be helpful in the prevention of persistent COVID-associated complications [118].
Prevention of NET formation may be achieved by application of dipyridamole, an antiplatelet agent, which interferes with aPL antibody-mediated NET release by adenosine A2A receptor agonism [119]. Likewise, ticagrelor might exert its beneficial inhibitory effects on NET formation independently of P2Y12 inhibition by increasing adenosine levels [120]. NET release can also be abrogated by the JAK 1/2 inhibitor ruxolitinib, which is applied in myeloproliferative neoplasms [121]. Inhibitors of the complement system are further therapeutic options interfering with NET production and with the first reports of symptom improvement during COVID-19 [6,122,123].
NET degradation can be induced by dornase alpha, a recombinant DNase I, which can be inhaled and the application may reduce the occurrence of acute respiratory distress syndromes (ARDS) [6]. However, similarly to DNase, the administration of heparin was shown to dismantle NETs and to remove platelet aggregates from NETs [124].
However, future research should also emphasize preservation models of endothelial integrity, which provides a shield against thromboinflammatory reactions [10]. The endothelial surface layer and in particular the glycocalyx is a very fragile structure, which is very prone to adverse environmental changes [125]. Conformational changes of the glycocalyx can be conferred by proinflammatory stimuli, such as increased plasma Na+ levels [126,127]. These changes may also initiate pathological processes facilitating infections and immunothrombosis. Many clinical concepts for glycocalyx preservation are hardly feasible; however, some of those such as administration of albumin, antithrombin III, sulodexide, atrasentan (a selective endothelin A receptor antagonist), steroids or statins should be studied further [128,129,130,131,132,133,134,135,136].
In conclusion, endothelial dysfunction with glycocalyx disintegration can be regarded as an initial trigger of SARS-CoV-2-mediated microthrombosis. Signaling pathways contributing to immunothrombosis are complex, involving innate and adaptive mechanisms. COVID-19-associated prothrombotic potential relies on well-published findings, such as endothelial cell infection [13] with glycocalyx destruction, promoting subsequent collagen exposure with platelet and coagulation activation [137,138]. Microthromboses upon platelet–platelet and platelet–leukocyte interactions further augment this response [31,32,139]. These are specific therapeutic targets to interfere with prolonged procoagulant and prothrombotic responses to viral infection. Evidence for long COVID treatment is still scarce, although many aspects of the endothelial-based pathologies of the acute SARS-CoV-2 infection may apply to the long-term sequelae [116,117,118]. Research on the elimination of viral persistence as well as interference with processes of immunothrombosis is still ongoing and merits further study.

Author Contributions

Conceptualization: P.P.W.; Writing—Original Draft Preparation: P.P.W. and B.P.; Writing—Review and Editing: A.J., C.W.K., T.G., S.P. and R.K.; Supervision: S.P., C.W.K. and R.K. All authors have contributed substantially to the work. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nadarajah, R.; Wu, J.; Hurdus, B.; Asma, S.; Bhatt, D.L.; Biondi-Zoccai, G.; Mehta, L.S.; Ram, C.V.S.; Ribeiro, A.L.P.; Van Spall, H.G.C.; et al. The collateral damage of COVID-19 to cardiovascular services: A meta-analysis. Eur. Heart J. 2022, 43, 3164–3178. [Google Scholar] [CrossRef]
  2. Arsenault, C.; Gage, A.; Kim, M.K.; Kapoor, N.R.; Akweongo, P.; Amponsah, F.; Aryal, A.; Asai, D.; Awoonor-Williams, J.K.; Ayele, W.; et al. COVID-19 and resilience of healthcare systems in ten countries. Nat. Med. 2022, 28, 1314–1324. [Google Scholar] [CrossRef] [PubMed]
  3. Wadowski, P.P.; Piechota-Polańczyk, A.; Andreas, M.; Kopp, C.W. Cardiovascular Disease Management in the Context of Global Crisis. Int. J. Environ. Res. Public Health 2022, 20, 689. [Google Scholar] [CrossRef] [PubMed]
  4. Mathew, D.; Giles, J.R.; Baxter, A.E.; Oldridge, D.A.; Greenplate, A.R.; Wu, J.E.; Alanio, C.; Kuri-Cervantes, L.; Pampena, M.B.; D’Andrea, K.; et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 2020, 369, eabc8511. [Google Scholar] [CrossRef]
  5. Jonigk, D.; Werlein, C.; Acker, T.; Aepfelbacher, M.; Amann, K.U.; Baretton, G.; Barth, P.; Bohle, R.M.; Büttner, A.; Büttner, R.; et al. Organ manifestations of COVID-19: What have we learned so far (not only) from autopsies? Virchows Arch. 2022, 481, 139–159. [Google Scholar] [CrossRef]
  6. Bonaventura, A.; Vecchié, A.; Dagna, L.; Martinod, K.; Dixon, D.L.; Van Tassell, B.W.; Dentali, F.; Montecucco, F.; Massberg, S.; Levi, M.; et al. Endothelial dysfunction and immunothrombosis as key pathogenic mechanisms in COVID-19. Nat. Rev. Immunol. 2021, 21, 319–329. [Google Scholar] [CrossRef]
  7. Gonzalez-Gonzalez, F.J.; Ziccardi, M.R.; McCauley, M.D. Virchow’s Triad and the Role of Thrombosis in COVID-Related Stroke. Front. Physiol. 2021, 12, 769254. [Google Scholar] [CrossRef]
  8. Conway, E.M.; Mackman, N.; Warren, R.Q.; Wolberg, A.S.; Mosnier, L.O.; Campbell, R.A.; Gralinski, L.E.; Rondina, M.T.; van de Veerdonk, F.L.; Hoffmeister, K.M.; et al. Understanding COVID-19-associated coagulopathy. Nat. Rev. Immunol. 2022, 22, 639–649. [Google Scholar] [CrossRef] [PubMed]
  9. Flaumenhaft, R.; Enjyoji, K.; Schmaier, A.A. Vasculopathy in COVID-19. Blood 2022, 140, 222–235. [Google Scholar] [CrossRef]
  10. Wadowski, P.P.; Jilma, B.; Kopp, C.W.; Ertl, S.; Gremmel, T.; Koppensteiner, R. Glycocalyx as Possible Limiting Factor in COVID-19. Front. Immunol. 2021, 12, 607306. [Google Scholar] [CrossRef] [PubMed]
  11. Targosz-Korecka, M.; Kubisiak, A.; Kloska, D.; Kopacz, A.; Grochot-Przeczek, A.; Szymonski, M. Endothelial glycocalyx shields the interaction of SARS-CoV-2 spike protein with ACE2 receptors. Sci. Rep. 2021, 11, 12157. [Google Scholar] [CrossRef] [PubMed]
  12. Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in COVID-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef] [PubMed]
  13. Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef]
  14. Siripanthong, B.; Asatryan, B.; Hanff, T.C.; Chatha, S.R.; Khanji, M.Y.; Ricci, F.; Muser, D.; Ferrari, V.A.; Nazarian, S.; Santangeli, P.; et al. The Pathogenesis and Long-Term Consequences of COVID-19 Cardiac Injury. JACC Basic Transl. Sci. 2022, 7, 294–308. [Google Scholar] [CrossRef]
  15. Wagner, D.D.; Heger, L.A. Thromboinflammation: From Atherosclerosis to COVID-19. Arter. Thromb. Vasc. Biol. 2022, 42, 1103–1112. [Google Scholar] [CrossRef] [PubMed]
  16. Pellegrini, D.; Kawakami, R.; Guagliumi, G.; Sakamoto, A.; Kawai, K.; Gianatti, A.; Nasr, A.; Kutys, R.; Guo, L.; Cornelissen, A.; et al. Microthrombi as a Major Cause of Cardiac Injury in COVID-19. Circulation 2021, 143, 1031–1042. [Google Scholar] [CrossRef] [PubMed]
  17. Laurence, J.; Nuovo, G.; Racine-Brzostek, S.E.; Seshadri, M.; Elhadad, S.; Crowson, A.N.; Mulvey, J.J.; Harp, J.; Ahamed, J.; Magro, C. Premortem Skin Biopsy Assessing Microthrombi, Interferon Type I Antiviral and Regulatory Proteins, and Complement Deposition Correlates with Coronavirus Disease 2019 Clinical Stage. Am. J. Pathol. 2022, 192, 1282–1294. [Google Scholar] [CrossRef]
  18. Magro, C.; Mulvey, J.J.; Berlin, D.; Nuovo, G.; Salvatore, S.; Harp, J.; Baxter-Stoltzfus, A.; Laurence, J. 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. [Google Scholar] [CrossRef]
  19. Xu, J.; Xiao, W.; Liang, X.; Shi, L.; Zhang, P.; Wang, Y.; Wang, Y.; Yang, H. A meta-analysis on the risk factors adjusted association between cardiovascular disease and COVID-19 severity. BMC Public Health 2021, 21, 147. [Google Scholar] [CrossRef]
  20. Wadowski, P.P.; Hülsmann, M.; Schörgenhofer, C.; Lang, I.M.; Wurm, R.; Gremmel, T.; Koppensteiner, R.; Steinlechner, B.; Schwameis, M.; Jilma, B. Sublingual functional capillary rarefaction in chronic heart failure. Eur. J. Clin. Investig. 2017, 48, e12869. [Google Scholar] [CrossRef]
  21. Wadowski, P.P.; Steinlechner, B.; Zimpfer, D.; Schlöglhofer, T.; Schima, H.; Hülsmann, M.; Lang, I.M.; Gremmel, T.; Koppensteiner, R.; Zehetmayer, S.; et al. Functional capillary impairment in patients with ventricular assist devices. Sci. Rep. 2019, 9, 5909. [Google Scholar] [CrossRef] [Green Version]
  22. Wadowski, P.P.; Schörgenhofer, C.; Rieder, T.; Ertl, S.; Pultar, J.; Serles, W.; Sycha, T.; Mayer, F.; Koppensteiner, R.; Gremmel, T.; et al. Microvascular rarefaction in patients with cerebrovascular events. Microvasc. Res. 2022, 140, 104300. [Google Scholar] [CrossRef]
  23. Ataga, K.I. Hypercoagulability and thrombotic complications in hemolytic anemias. Haematologica 2009, 94, 1481–1484. [Google Scholar] [CrossRef] [Green Version]
  24. Constantinescu-Bercu, A.; Wang, Y.A.; Woollard, K.J.; Mangin, P.; Vanhoorelbeke, K.; Crawley, J.T.; Salles-Crawley, I.I. The GPIbα intracellular tail-role in transducing VWF- and collagen/GPVI-mediated signaling. Haematologica 2021, 107, 933–946. [Google Scholar] [CrossRef] [PubMed]
  25. Sadler, J. von Willebrand factor. J. Biol. Chem. 1991, 266, 22777–22780. [Google Scholar] [CrossRef] [PubMed]
  26. Philippe, A.; Gendron, N.; Bory, O.; Beauvais, A.; Mirault, T.; Planquette, B.; Sanchez, O.; Diehl, J.-L.; Chocron, R.; Smadja, D.M. Von Willebrand factor collagen-binding capacity predicts in-hospital mortality in COVID-19 patients: Insight from VWF/ADAMTS13 ratio imbalance. Angiogenesis 2021, 24, 407–411. [Google Scholar] [CrossRef] [PubMed]
  27. Andrade, S.A.D.; de Souza, D.A.; Torres, A.L.; de Lima, C.F.G.; Ebram, M.C.; Celano, R.M.G.; Schattner, M.; Chudzinski-Tavassi, A.M. Patho-physiology of COVID-19: Critical Role of Hemostasis. Front. Cell. Infect. Microbiol. 2022, 12, 696. [Google Scholar] [CrossRef] [PubMed]
  28. Li, R.; Emsley, J. The organizing principle of the platelet glycoprotein Ib–IX–V complex. J. Thromb. Haemost. 2013, 11, 605–614. [Google Scholar] [CrossRef] [Green Version]
  29. Yun, S.-H.; Sim, E.-H.; Goh, R.-Y.; Park, J.-I.; Han, J.-Y. Platelet Activation: The Mechanisms and Potential Biomarkers. BioMed Res. Int. 2016, 2016, 9060143. [Google Scholar] [CrossRef] [Green Version]
  30. Dörmann, D.; Clemetson, K.J.; Kehrel, B.E. The GPIb thrombin-binding site is essential for thrombin-induced platelet procoagulant activity. Blood 2000, 96, 2469–2478. [Google Scholar] [CrossRef]
  31. Parker, W.A.; Orme, R.C.; Hanson, J.; Stokes, H.M.; Bridge, C.M.; Shaw, P.A.; Sumaya, W.; Thorneycroft, K.; Petrucci, G.; Porro, B.; et al. Very-low-dose twice-daily aspirin maintains platelet inhibition and improves haemostasis during dual-antiplatelet therapy for acute coronary syndrome. Platelets 2019, 30, 148–157. [Google Scholar] [CrossRef]
  32. Paul, B.Z.S.; Jin, J.; Kunapuli, S.P. Molecular Mechanism of Thromboxane A2-induced Platelet Aggregation: Essential role for p2t(ac) and alpha(2a) receptors. J. Biol. Chem. 1999, 274, 29108–29114. [Google Scholar] [CrossRef] [Green Version]
  33. Gremmel, T.; Frelinger, A.L., III; Michelson, A.D. Platelet Physiology. Semin. Thromb. Hemost. 2016, 42, 191–204. [Google Scholar] [CrossRef] [Green Version]
  34. Parker, W.A.E.; Storey, R.F. Long-term antiplatelet therapy following myocardial infarction: Implications of PEGASUS-TIMI 54. Heart 2016, 102, 783–789. [Google Scholar] [CrossRef]
  35. Storey, R.F.; Sanderson, H.M.; White, A.E.; May, J.A.; Cameron, K.E.; Heptinstall, S. The central role of the P2T receptor in amplification of human platelet activation, aggregation, secretion and procoagulant activity. Br. J. Haematol. 2000, 110, 925–934. [Google Scholar] [CrossRef]
  36. Storey, R.F.; Judge, H.M.; Wilcox, R.G.; Heptinstall, S. Inhibition of ADP-induced P-selectin Expression and Platelet-Leukocyte Conjugate Formation by Clopidogrel and the P2Y12 Receptor Antagonist AR-C69931MX but not As-pirin. Thromb. Haemost. 2002, 88, 488–494. [Google Scholar]
  37. Etulain, J.; Martinod, K.; Wong, S.L.; Cifuni, S.M.; Schattner, M.; Wagner, D.D. P-selectin promotes neutrophil extracellular trap formation in mice. Blood 2015, 126, 242–246. [Google Scholar] [CrossRef] [Green Version]
  38. Michelson, A.D.; Barnard, M.R.; Hechtman, H.B.; MacGregor, H.; Connolly, R.J.; Loscalzo, J.; Valeri, C.R. In vivo tracking of platelets: Circulating degranulated platelets rapidly lose surface P-selectin but continue to circulate and function. Proc. Natl. Acad. Sci. USA 1996, 93, 11877–11882. [Google Scholar] [CrossRef] [Green Version]
  39. Garcia, C.; Duong, J.A.; Poëtte, M.; Ribes, A.; Payre, B.; Mémier, V.; Sié, P.; Minville, V.; Voisin, S.; Payrastre, B.; et al. Platelet activation and partial desensitization are associated with viral xenophagy in patients with severe COVID-19. Blood Adv. 2022, 6, 3884–3898. [Google Scholar] [CrossRef]
  40. Sanderson, H.M.; Heptinstall, S.; Vickers, J.; Lösche, W. Studies on the effects of agonists and antagonists on platelet shape change and platelet aggregation in whole blood. Blood Coagul. Fibrinolysis 1996, 7, 245–248. [Google Scholar] [CrossRef]
  41. Cho, M.J.; Liu, J.; Pestina, T.I.; Steward, S.A.; Jackson, C.W.; Gartner, T.K. αIIbβ3-mediated outside-in signal-ing induced by the agonist peptide LSARLAF utilizes ADP and thromboxane A2 receptors to cause α-granule secretion by platelets. J. Thromb. Haemost. 2003, 1, 363–373. [Google Scholar] [CrossRef]
  42. Brahmer, A.; Neuberger, E.; Esch-Heisser, L.; Haller, N.; Jørgensen, M.M.; Baek, R.; Möbius, W.; Simon, P.; Krämer-Albers, E.-M. Platelets, endothelial cells and leukocytes contribute to the exercise-triggered release of extracellular vesicles into the circulation. J. Extracell. Vesicles 2019, 8, 1615820. [Google Scholar] [CrossRef]
  43. Nederveen, J.P.; Warnier, G.; Di Carlo, A.; Nilsson, M.I.; Tarnopolsky, M.A. Extracellular Vesicles and Exo-somes: Insights from Exercise Science. Front. Physiol. 2021, 11, 604274. [Google Scholar] [CrossRef]
  44. Elzanowska, J.; Semira, C.; Costa-Silva, B. DNA in extracellular vesicles: Biological and clinical aspects. Mol. Oncol. 2020, 15, 1701–1714. [Google Scholar] [CrossRef]
  45. Sinauridze, E.I.; Kireev, D.A.; Popenko, N.Y.; Pichugin, A.; Panteleev, M.; Krymskaya, O.V.; Ataullakhanov, F.I. Platelet microparticle membranes have 50- to 100-fold higher specific procoagulant activity than activated platelets. Thromb. Haemost. 2007, 97, 425–434. [Google Scholar] [CrossRef]
  46. Subrahmanian, S.; Borczuk, A.; Salvatore, S.; Fung, K.M.; Merrill, J.T.; Laurence, J.; Ahamed, J. Tissue factor upregula-tion is associated with SARS-CoV-2 in the lungs of COVID-19 patients. J. Thromb. Haemost. 2021, 19, 2268–2274. [Google Scholar] [CrossRef]
  47. Busch, M.; Timmermans, S.A.; Nagy, M.; Visser, M.; Huckriede, J.; Aendekerk, J.P.; De Vries, F.; Potjewijd, J.; Jallah, B.; Ysermans, R.; et al. Neutrophils and Contact Activation of Coagulation as Potential Drivers of COVID-19. Circulation 2020, 142, 1787–1790. [Google Scholar] [CrossRef]
  48. Gorog, D.A.; Storey, R.F.; Gurbel, P.A.; Tantry, U.S.; Berger, J.S.; Chan, M.Y.; Duerschmied, D.; Smyth, S.S.; Parker, W.A.E.; Ajjan, R.A.; et al. Current and novel biomarkers of thrombotic risk in COVID-19: A Consensus Statement from the International COVID-19 Thrombosis Biomarkers Colloquium. Nat. Rev. Cardiol. 2022, 19, 475–495. [Google Scholar] [CrossRef]
  49. Bevers, E.M.; Comfurius, P.; Van Rijn, J.L.M.L.; Hemker, H.C. Generation of Prothrombin-Converting Activity and the Exposure of Phosphatidylserine at the Outer Surface of Platelets. Eur. J. Biochem. 1982, 122, 429–436. [Google Scholar] [CrossRef] [PubMed]
  50. Lhermusier, T.; Chap, H.; Payrastre, B. Platelet membrane phospholipid asymmetry: From the characterization of a scramblase activity to the identification of an essential protein mutated in Scott syndrome. J. Thromb. Haemost. 2011, 9, 1883–1891. [Google Scholar] [CrossRef]
  51. Ofosu, F.A.; Dewar, L.; Craven, S.J.; Song, Y.; Cedrone, A.; Freedman, J.; Fenton, J.W. Coordinate Activation of Human Platelet Protease-activated Receptor-1 and -4 in Response to Subnanomolar α-Thrombin. J. Biol. Chem. 2008, 283, 26886–26893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Duvernay, M.T.; Temple, K.J.; Maeng, J.G.; Blobaum, A.L.; Stauffer, S.R.; Lindsley, C.W.; Hamm, H.E. Contributions of Protease-Activated Receptors PAR1 and PAR4 to Thrombin-Induced GPIIbIIIa Activation in Human Platelets. Mol. Pharmacol. 2016, 91, 39–47. [Google Scholar] [CrossRef] [Green Version]
  53. Wadowski, P.P.; Pultar, J.; Weikert, C.; Eichelberger, B.; Panzer, B.; Huber, K.; Lang, I.M.; Koppensteiner, R.; Panzer, S.; Gremmel, T. Protease-activated receptor-mediated platelet aggregation in acute coronary syndrome patients on potent P2Y12 inhibitors. Res. Pract. Thromb. Haemost. 2019, 3, 383–390. [Google Scholar] [CrossRef] [Green Version]
  54. Gremmel, T.; Michelson, A.D.; Wadowski, P.P.; Pultar, J.; Weikert, C.; Tscharre, M.; Lee, S.; Panzer, S.; Frelinger, A.L. Sex-specific platelet activation through protease-activated receptor-1 in patients undergoing cardiac catheterization. Atherosclerosis 2021, 339, 12–19. [Google Scholar] [CrossRef]
  55. Wadowski, P.P.; Pultar, J.; Weikert, C.; Eichelberger, B.; Tscharre, M.; Koppensteiner, R.; Panzer, S.; Gremmel, T. Platelet-to-Lymphocyte Ratio as Marker of Platelet Activation in Patients on Potent P2Y12 Inhibitors. J. Cardiovasc. Pharmacol. Ther. 2022, 27, 10742484221096524. [Google Scholar] [CrossRef]
  56. Zheng, M.; Karki, R.; Williams, E.P.; Yang, D.; Fitzpatrick, E.; Vogel, P.; Jonsson, C.B.; Kanneganti, T.-D. TLR2 senses the SARS-CoV-2 envelope protein to produce inflammatory cytokines. Nat. Immunol. 2021, 22, 829–838. [Google Scholar] [CrossRef]
  57. Wadowski, P.P.; Weikert, C.; Pultar, J.; Lee, S.; Eichelberger, B.; Koppensteiner, R.; Lang, I.M.; Panzer, S.; Gremmel, T. Ticagrelor Inhibits Toll-Like and Protease-Activated Receptor Mediated Platelet Activation in Acute Coronary Syndromes. Cardiovasc. Drugs Ther. 2020, 34, 53–63. [Google Scholar] [CrossRef] [Green Version]
  58. Biswas, S.; Zimman, A.; Gao, D.; Byzova, T.V.; Podrez, E.A. TLR2 Plays a Key Role in Platelet Hyperreactivity and Accelerated Thrombosis Associated with Hyperlipidemia. Circ. Res. 2017, 121, 951–962. [Google Scholar] [CrossRef]
  59. Into, T.; Kanno, Y.; Dohkan, J.-I.; Nakashima, M.; Inomata, M.; Shibata, K.-I.; Lowenstein, C.J.; Matsushita, K. Pathogen Recognition by Toll-like Receptor 2 Activates Weibel-Palade Body Exocytosis in Human Aortic Endothelial Cells. J. Biol. Chem. 2007, 282, 8134–8141. [Google Scholar] [CrossRef] [Green Version]
  60. Singh, B.; Biswas, I.; Bhagat, S.; Surya Kumari, S.; Khan, G.A. HMGB1 facilitates hypoxia-induced vWF upregu-lation through TLR2-MYD88-SP1 pathway. Eur. J. Immunol. 2016, 46, 2388–2400. [Google Scholar] [CrossRef] [Green Version]
  61. Carestia, A.; Kaufman, T.; Rivadeneyra, L.; Landoni, V.I.; Pozner, R.G.; Negrotto, S.; D’Atri, L.P.; Gómez, R.M.; Schattner, M. Mediators and molec-ular pathways involved in the regulation of neutrophil extracellular trap formation mediated by activated platelets. J. Leukoc. Biol. 2016, 99, 153–162. [Google Scholar] [CrossRef] [Green Version]
  62. Fujimura, Y.; Holland, L.Z. COVID-19 microthrombosis: Unusually large VWF multimers are a platform for activation of the alternative complement pathway under cytokine storm. Int. J. Hematol. 2022, 115, 457–469. [Google Scholar] [CrossRef]
  63. Khan, S.; Shafiei, M.S.; Longoria, C.; Schoggins, J.W.; Savani, R.C.; Zaki, H. SARS-CoV-2 spike protein induces inflammation via TLR2-dependent activation of the NF-κB pathway. Elife 2021, 10, e68563. [Google Scholar] [CrossRef]
  64. Choudhury, A.; Mukherjee, S. In silico studies on the comparative characterization of the interactions of SARS-CoV-2 spike glycoprotein with ACE-2 receptor homologs and human TLRs. J. Med. Virol. 2020, 92, 2105–2113. [Google Scholar] [CrossRef]
  65. Fard, M.B.; Fard, S.B.; Ramazi, S.; Atashi, A.; Eslamifar, Z. Thrombosis in COVID-19 infection: Role of platelet activation-mediated immunity. Thromb. J. 2021, 19, 59. [Google Scholar] [CrossRef]
  66. Teixeira, P.C.; Dorneles, G.P.; Filho, P.C.S.; da Silva, I.M.; Schipper, L.L.; Postiga, I.A.; Neves, C.A.M.; Junior, L.C.R.; Peres, A.; de Souto, J.T.; et al. Increased LPS levels coexist with systemic inflammation and result in monocyte activation in severe COVID-19 patients. Int. Immunopharmacol. 2021, 100, 108125. [Google Scholar] [CrossRef]
  67. Clark, S.R.; Ma, A.C.; Tavener, S.A.; McDonald, B.; Goodarzi, Z.; Kelly, M.M.; Patel, K.D.; Chakrabarti, S.; McAvoy, E.; Sinclair, G.D.; et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat. Med. 2007, 13, 463–469. [Google Scholar] [CrossRef]
  68. Wadowski, P.P.; Eichelberger, B.; Kopp, C.W.; Pultar, J.; Seidinger, D.; Koppensteiner, R.; Lang, I.M.; Panzer, S.; Gremmel, T. Disaggregation Following Agonist-Induced Platelet Activation in Patients on Dual Antiplatelet Therapy. J. Cardiovasc. Transl. Res. 2017, 10, 359–367. [Google Scholar] [CrossRef] [Green Version]
  69. Bohula, E.A.; Berg, D.D.; Lopes, M.S.; Connors, J.M.; Babar, I.; Barnett, C.F.; Chaudhry, S.-P.; Chopra, A.; Ginete, W.; Ieong, M.H.; et al. Anticoagulation and Antiplatelet Therapy for Prevention of Venous and Arterial Thrombotic Events in Critically Ill Patients with COVID-19: COVID-PACT. Circulation 2022, 146, 1344–1356. [Google Scholar] [CrossRef]
  70. Abani, O.; Abbas, A.; Abbas, F.; Abbas, M.; Abbasi, S.; Abbass, H.; Abbott, A.; Abdallah, N.; Abdelaziz, A.; Abdelfattah, M.; et al. Aspirin in patients admitted to hospital with COVID-19 (RECOVERY): A randomised, controlled, open-label, platform trial. Lancet 2022, 399, 143–151. [Google Scholar] [CrossRef]
  71. Berger, J.S.; Kornblith, L.Z.; Gong, M.N.; Reynolds, H.R.; Cushman, M.; Cheng, Y.; McVerry, B.J.; Kim, K.S.; Lopes, R.D.; Atassi, B.; et al. Effect of P2Y12 Inhibitors on Survival Free of Organ Support among Non-Critically Ill Hospitalized Patients with COVID-19. JAMA 2022, 327, 227. [Google Scholar] [CrossRef]
  72. Lawler, P.R.; Goligher, E.C.; Berger, J.S.; Neal, M.D.; McVerry, B.J.; Nicolau, J.C. Therapeutic Anticoagulation with Heparin in Noncritically Ill Patients with COVID-19. N. Engl. J. Med. 2021, 385, 790–802. [Google Scholar] [CrossRef]
  73. Goligher, E.C.; Bradbury, C.A.; McVerry, B.J.; Lawler, P.R.; Berger, J.S.; Gong, M.N. Therapeutic Anticoagulation with Heparin in Critically Ill Patients with COVID-19. N. Engl. J. Med. 2021, 385, 777–789. [Google Scholar] [CrossRef]
  74. Ionescu, F.; Jaiyesimi, I.; Petrescu, I.; Lawler, P.R.; Castillo, E.; Munoz-Maldonado, Y.; Imam, Z.; Narasimhan, M.; Abbas, A.E.; Konde, A.; et al. Association of anti-coagulation dose and survival in hospitalized COVID-19 patients: A retrospective propensity score-weighted analysis. Eur. J. Haematol. 2021, 106, 165–174. [Google Scholar] [CrossRef]
  75. Bakchoul, T.; Hammer, S.; Lang, P.; Rosenberger, P. Fibrinolysis shut down in COVID-19 patients: Report on two severe cases with potential diagnostic and clinical relevance. Thromb. Updat. 2020, 1, 100008. [Google Scholar] [CrossRef]
  76. Wygrecka, M.; Birnhuber, A.; Seeliger, B.; Michalick, L.; Pak, O.; Schultz, A.-S.; Schramm, F.; Zacharias, M.; Gorkiewicz, G.; David, S.; et al. Altered fibrin clot structure and dysregulated fibrinolysis contribute to thrombosis risk in severe COVID-19. Blood Adv. 2022, 6, 1074–1087. [Google Scholar] [CrossRef]
  77. Ji, H.-L.; Dai, Y.; Zhao, R. Fibrinolytic therapy for COVID-19: A review of case series. Acta Pharmacol. Sin. 2021, 43, 2168–2170. [Google Scholar] [CrossRef]
  78. Zuo, Y.; Warnock, M.; Harbaugh, A.; Yalavarthi, S.; Gockman, K.; Zuo, M.; Madison, J.A.; Knight, J.S.; Kanthi, Y.; Lawrence, D.A. Plasma tissue plasminogen activator and plasminogen activator inhibitor-1 in hospitalized COVID-19 patients. Sci. Rep. 2021, 11, 1580. [Google Scholar] [CrossRef]
  79. Rovai, E.S.; Alves, T.; Holzhausen, M. Protease-activated receptor 1 as a potential therapeutic target for COVID-19. Exp. Biol. Med. 2020, 246, 688–694. [Google Scholar] [CrossRef]
  80. Pultar, J.; Wadowski, P.P.; Panzer, S.; Gremmel, T. Oral antiplatelet agents in cardiovascular disease. Vasa 2019, 48, 291–302. [Google Scholar] [CrossRef]
  81. Katsoularis, I.; Fonseca-Rodríguez, O.; Farrington, P.; Jerndal, H.; Lundevaller, E.H.; Sund, M.; Lindmark, K.; Connolly, A.-M.F. Risks of deep vein thrombosis, pulmonary embolism, and bleeding after COVID-19: Nationwide self-controlled cases series and matched cohort study. BMJ 2022, 377, e069590. [Google Scholar] [CrossRef] [PubMed]
  82. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef] [PubMed]
  83. Chaplin, D.D. Overview of the immune response. J. Allergy Clin. Immunol. 2010, 125, S3–S23. [Google Scholar] [CrossRef] [PubMed]
  84. Weisel, J.W.; Litvinov, R.I. Fibrin Formation, Structure and Properties. In Fibrous Proteins: Structures and Mechanisms; Springer International Publishing: Cham, Switzerland, 2017; pp. 405–456. [Google Scholar]
  85. Kaplan, Z.S.; Zarpellon, A.; Alwis, I.; Yuan, Y.; McFadyen, J.; Ghasemzadeh, M.; Schoenwaelder, S.M.; Ruggeri, Z.M.; Jackson, S.P. Thrombin-dependent intravascular leukocyte trafficking regulated by fibrin and the platelet receptors GPIb and PAR4. Nat. Commun. 2015, 6, 7835. [Google Scholar] [CrossRef] [Green Version]
  86. Nguyen, D.; Jeon, H.-M.; Lee, J. Tissue factor links inflammation, thrombosis, and senescence in COVID-19. Sci. Rep. 2022, 12, 19842. [Google Scholar] [CrossRef] [PubMed]
  87. Mackman, N.; Sachettor, A.T.A. Tissue factor and COVID-19: An update. Curr. Drug Targets 2022, 23, 1573–1577. [Google Scholar] [CrossRef]
  88. Rosell, A.; Havervall, S.; Von Meijenfeldt, F.; Hisada, Y.; Aguilera, K.; Grover, S.P.; Lisman, T.; Mackman, N.; Thålin, C. Patients With COVID-19 Have Elevated Levels of Circulating Extracellular Vesicle Tissue Factor Activity That Is Associated with Severity and Mortality—Brief Report. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 878–882. [Google Scholar] [CrossRef] [PubMed]
  89. Guervilly, C.; Bonifay, A.; Burtey, S.; Sabatier, F.; Cauchois, R.; Abdili, E.; Arnaud, L.; Lano, G.; Pietri, L.; Robert, T.; et al. Dissemination of extreme levels of extracellular vesicles: Tissue factor activity in patients with severe COVID-19. Blood Adv. 2021, 5, 628–634. [Google Scholar] [CrossRef]
  90. Claus, R.A.; Bockmeyer, C.L.; Sossdorf, M.; Lösche, W. The balance between von-Willebrand factor and its cleaving protease ADAMTS13: Biomarker in systemic inflammation and development of organ failure? Curr. Mol. Med. 2010, 10, 236–248. [Google Scholar] [CrossRef]
  91. Fernández-Pérez, M.P.; Águila, S.; Reguilón-Gallego, L.; de los Reyes-García, A.M.; Miñano, A.; Bravo-Pérez, C.; María, E.; Corral, J.; García-Barberá, N.; Gómez-Verdú, J.M.; et al. Neutrophil extracellular traps and von Willebrand factor are allies that negatively influence COVID-19 outcomes. Clin. Transl. Med. 2021, 11, e268. [Google Scholar] [CrossRef] [PubMed]
  92. Menter, T.; Haslbauer, J.D.; Nienhold, R.; Savic, S.; Hopfer, H.; Deigendesch, N.; Frank, S.; Turek, D.; Willi, N.; Pargger, H.; et al. Post-mortem examination of COVID19 patients reveals diffuse alveolar damage with severe capillary congestion and variegated findings of lungs and other organs suggesting vascular dysfunction. Histopathology 2020, 77, 198–209. [Google Scholar] [CrossRef]
  93. Yang, J.; Wu, Z.; Long, Q.; Huang, J.; Hong, T.; Liu, W.; Lin, J. Insights into Immunothrombosis: The Interplay among Neutrophil Extracellular Trap, von Willebrand Factor, and ADAMTS13. Front. Immunol. 2020, 11, 610696. [Google Scholar] [CrossRef]
  94. Furlan, M. Von Willebrand factor: Molecular size and functional activity. Ann. Hematol. 1996, 72, 341–348. [Google Scholar] [CrossRef]
  95. Singh, B.; Biswas, I.; Garg, I.; Sugadev, R.; Singh, A.K.; Dey, S.; Khan, G.A. Von Willebrand Factor Antagonizes Nitric Oxide Synthase to Promote Insulin Resistance during Hypoxia. Biochemistry 2013, 53, 115–126. [Google Scholar] [CrossRef]
  96. Grässle, S.; Huck, V.; Pappelbaum, K.I.; Gorzelanny, C.; Aponte-Santamaría, C.; Baldauf, C.; Gräter, F.; Schneppenheim, R.; Obser, T.; Schneider, S.W. Von Willebrand Factor Directly Interacts with DNA From Neutrophil Extracellular Traps. Arter. Thromb. Vasc. Biol. 2014, 34, 1382–1389. [Google Scholar] [CrossRef] [Green Version]
  97. Kolaczkowska, E.; Jenne, C.N.; Surewaard, B.G.J.; Thanabalasuriar, A.; Lee, W.-Y.; Sanz, M.-J.; Mowen, K.; Opdenakker, G.; Kubes, P. Molecular mechanisms of NET formation and degradation revealed by intravital imaging in the liver vasculature. Nat. Commun. 2015, 6, 6673. [Google Scholar] [CrossRef] [Green Version]
  98. Xu, J.; Zhang, X.; Pelayo, R.; Monestier, M.; Ammollo, C.T.; Semeraro, F.; Taylor, F.B.; Esmon, N.L.; Lupu, F.; Esmon, C.T. Extracellular histones are major mediators of death in sepsis. Nat. Med. 2009, 15, 1318–1321. [Google Scholar] [CrossRef] [Green Version]
  99. Freeman, C.G.; Parish, C.; Knox, K.J.; Blackmore, J.L.; Lobov, S.A.; King, D.W.; Senden, T.; Stephens, R.W. The accumulation of circulating histones on heparan sulphate in the capillary glycocalyx of the lungs. Biomaterials 2013, 34, 5670–5676. [Google Scholar] [CrossRef]
  100. Fuchs, T.A.; Bhandari, A.A.; Wagner, D.D. Histones induce rapid and profound thrombocytopenia in mice. Blood 2011, 118, 3708–3714. [Google Scholar] [CrossRef] [Green Version]
  101. Moore, B.J.B.; June, C.H. Cytokine release syndrome in severe COVID-19. Science 2020, 368, 473–474. [Google Scholar] [CrossRef] [Green Version]
  102. Gordon, A.C.; Mouncey, P.R.; Al-Beidh, F.; Rowan, K.M.; Nichol, A.D.; Arabi, Y.M.; Annane, D.; Beane, A.; Van Bentum-Puijk, W.; Berry, L.R.; et al. Interleukin-6 Receptor Antagonists in Critically Ill Patients with COVID-19. N. Engl. J. Med. 2021, 384, 1491–1502. [Google Scholar] [CrossRef] [PubMed]
  103. Thiam, H.R.; Wong, S.L.; Wagner, D.D.; Waterman, C.M. Cellular Mechanisms of NETosis. Annu. Rev. Cell Dev. Biol. 2020, 36, 191–218. [Google Scholar] [CrossRef] [PubMed]
  104. Zhu, Y.; Chen, X.; Liu, X. NETosis and Neutrophil Extracellular Traps in COVID-19: Immunothrombosis and Beyond. Front. Immunol. 2022, 13, 838011. [Google Scholar] [CrossRef]
  105. Hepprich, M.; Mudry, J.M.; Gregoriano, C.; Jornayvaz, F.R.; Carballo, S.; Wojtusciszyn, A.; Bart, P.A.; Chiche, J.D.; Fischli, S.; Baumgartner, T.; et al. Canakinumab in patients with COVID-19 and type 2 diabetes—A multicentre, randomised, double-blind, placebo-controlled trial. EClinicalMedicine 2022, 53, 101649. [Google Scholar] [CrossRef]
  106. Silvestre-Roig, C.; Braster, Q.; Wichapong, K.; Lee, E.Y.; Teulon, J.M.; Berrebeh, N.; Winter, J.; Adrover, J.M.; Santos, G.S.; Froese, A.; et al. Externalized histone H4 orchestrates chronic inflammation by inducing lytic cell death. Nature 2019, 569, 236–240. [Google Scholar] [CrossRef]
  107. Falk, E.; Shah, P.K.; Fuster, V. Coronary Plaque Disruption. Circulation 1995, 92, 657–671. [Google Scholar] [CrossRef]
  108. Nazy, I.; Elliott, T.D.; Arnold, D.M. Platelet factor 4 inhibits ADAMTS13 activity and regulates the multimeric distribution of von Willebrand factor. Br. J. Haematol. 2020, 190, 594–598. [Google Scholar] [CrossRef]
  109. Wang, A.; Liu, F.; Dong, N.; Ma, Z.; Zhang, J.; Su, J.; Zhao, Y.; Ruan, C. Thrombospondin-1 and ADAMTS13 competitively bind to VWF A2 and A3 domains in vitro. Thromb. Res. 2010, 126, e260–e265. [Google Scholar] [CrossRef]
  110. Zuo, Y.; Estes, S.K.; Ali, R.A.; Gandhi, A.A.; Yalavarthi, S.; Shi, H.; Sule, G.; Gockman, K.; Madison, J.A.; Zuo, M.; et al. Prothrombotic autoantibodies in serum from patients hospitalized with COVID-19. Sci. Transl. Med. 2020, 12, eabd3876. [Google Scholar] [CrossRef]
  111. Ehrenfeld, M.; Tincani, A.; Andreoli, L.; Cattalini, M.; Greenbaum, A.; Kanduc, D.; Alijotas-Reig, J.; Zinserling, V.; Semenova, N.; Amital, H.; et al. Covid-19 and autoimmunity. Autoimmun. Rev. 2020, 19, 102597. [Google Scholar] [CrossRef]
  112. George, P.M.; Reed, A.; Desai, S.R.; Devaraj, A.; Faiez, T.S.; Laverty, S.; Kanwal, A.; Esneau, C.; Liu, M.K.; Kamal, F.; et al. A persistent neutrophil-associated immune signature characterizes post–COVID-19 pulmonary sequelae. Sci. Transl. Med. 2022, 14, eabo5795. [Google Scholar] [CrossRef]
  113. Mantovani, A.; Morrone, M.C.; Patrono, C.; Santoro, M.G.; Schiaffino, S.; Remuzzi, G.; Bussolati, G.; Cappuccinelli, P.; Fitzgerald, G.; Bacci, M.L.; et al. Long Covid: Where we stand and challenges ahead. Cell Death Differ. 2022, 29, 1891–1900. [Google Scholar] [CrossRef] [PubMed]
  114. Thierry, A.R.; Roch, B. Neutrophil Extracellular Traps and By-Products Play a Key Role in COVID-19: Pathogenesis, Risk Factors, and Therapy. J. Clin. Med. 2020, 9, 2942. [Google Scholar] [CrossRef]
  115. Middleton, E.A.; He, X.-Y.; Denorme, F.; Campbell, R.A.; Ng, D.; Salvatore, S.P.; Mostyka, M.; Baxter-Stoltzfus, A.; Borczuk, A.C.; Loda, M.; et al. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 2020, 136, 1169–1179. [Google Scholar] [CrossRef]
  116. Ahamed, J.; Laurence, J. Long COVID endotheliopathy: Hypothesized mechanisms and potential therapeutic approaches. J. Clin. Investig. 2022, 132, e161167. [Google Scholar] [CrossRef]
  117. Wang, C.; Yu, C.; Jing, H.; Wu, X.; Novakovic, V.A.; Xie, R.; Shi, J. Long COVID: The Nature of Thrombotic Se-quelae Determines the Necessity of Early Anticoagulation. Front. Cell. Infect. Microbiol. 2022, 12, 861703. [Google Scholar] [CrossRef]
  118. Xie, Y.; Choi, T.; Al-Aly, Z. Nirmatrelvir and the Risk of Post-Acute Sequelae of COVID-19. medRxiv 2022. [Google Scholar] [CrossRef]
  119. Ali, R.A.; Gandhi, A.A.; Meng, H.; Yalavarthi, S.; Vreede, A.P.; Estes, S.K.; Palmer, O.R.; Bockenstedt, P.L.; Pinsky, D.J.; Greve, J.M.; et al. Adenosine receptor agonism protects against NETosis and thrombosis in antiphospholipid syndrome. Nat. Commun. 2019, 10, 1916. [Google Scholar] [CrossRef] [Green Version]
  120. Mitsios, A.; Chrysanthopoulou, A.; Arampatzioglou, A.; Angelidou, I.; Vidali, V.; Ritis, K.; Skendros, P.; Stakos, D. Ticagrelor Exerts Immune-Modulatory Effect by Attenuating Neutrophil Extracellular Traps. Int. J. Mol. Sci. 2020, 21, 3625. [Google Scholar] [CrossRef]
  121. Wolach, O.; Sellar, R.S.; Martinod, K.; Cherpokova, D.; McConkey, M.; Chappell, R.J.; Silver, A.J.; Adams, D.; Castellano, C.A.; Schneider, R.K.; et al. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci. Transl. Med. 2018, 10, eaan8292. [Google Scholar] [CrossRef] [Green Version]
  122. Annane, D.; Heming, N.; Grimaldi-Bensouda, L.; Frémeaux-Bacchi, V.; Vigan, M.; Roux, A.-L.; Marchal, A.; Michelon, H.; Rottman, M.; Moine, P. 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. [Google Scholar] [CrossRef] [PubMed]
  123. Diurno, F.; Numis, F.G.; Porta, G.; Cirillo, F.; Maddaluno, S.; Ragozzino, A.; De Negri, P.; Di Gennaro, C.; Pagano, A.; Allegorico, E.; 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, 4040–4047. [Google Scholar]
  124. Fuchs, T.A.; Brill, A.; Duerschmied, D.; Schatzberg, D.; Monestier, M.; Myers, D.D., Jr.; Wrobleski, S.K.; Wakefield, T.W.; Hartwig, J.H.; Wagner, D.D. Extracellular DNA traps promote thrombosis. Proc. Natl. Acad. Sci. USA 2010, 107, 15880–15885. [Google Scholar] [CrossRef] [Green Version]
  125. Reitsma, S.; Slaaf, D.W.; Vink, H.; van Zandvoort, M.A.M.J.; Oude Egbrink, M.G. The endothelial glycocalyx: Composition, functions, and visualization. Pflug. Arch. 2007, 454, 345–359. [Google Scholar] [CrossRef] [Green Version]
  126. Schierke, F.; Wyrwoll, M.J.; Wisdorf, M.; Niedzielski, L.; Maase, M.; Ruck, T.; Meuth, S.G.; Kusche-Vihrog, K. Nanomechanics of the endothelial glycocalyx contribute to Na+-induced vascular inflammation. Sci. Rep. 2017, 7, srep46476. [Google Scholar] [CrossRef] [Green Version]
  127. Min, B.; Fairchild, R.L. Over-salting ruins the balance of the immune menu. J. Clin. Investig. 2015, 125, 4002–4004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Zeng, Y.; Adamson, R.H.; Curry, F.-R.E.; Tarbell, J.M. Sphingosine-1-phosphate protects endothelial glycocalyx by inhibiting syndecan-1 shedding. Am. J. Physiol. Circ. Physiol. 2014, 306, H363–H372. [Google Scholar] [CrossRef]
  129. Becker, B.F.; Chappell, D.; Bruegger, D.; Annecke, T.; Jacob, M. Therapeutic strategies targeting the endothelial glycocalyx: Acute deficits, but great potential. Cardiovasc. Res. 2010, 87, 300–310. [Google Scholar] [CrossRef] [Green Version]
  130. Broekhuizen, L.N.; Lemkes, B.A.; Mooij, H.L.; Meuwese, M.C.; Verberne, H.; Holleman, F.; Schlingemann, R.O.; Nieuwdorp, M.; Stroes, E.S.G.; Vink, H. Effect of sulodexide on endothelial glycocalyx and vascular permeability in patients with type 2 diabetes mellitus. Diabetologia 2010, 53, 2646–2655. [Google Scholar] [CrossRef] [Green Version]
  131. Uchimido, R.; Schmidt, E.P.; Shapiro, N.I. The glycocalyx: A novel diagnostic and therapeutic target in sepsis. Crit. Care 2019, 23, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Boels, M.G.; Avramut, M.C.; Koudijs, A.; Dane, M.J.; Lee, D.H.; van der Vlag, J.; Koster, A.J.; van Zonneveld, A.J.; van Faassen, E.; Gröne, H.-J.; et al. Atrasentan Reduces Albuminuria by Restoring the Glomerular Endothelial Glycocalyx Barrier in Diabetic Nephropathy. Diabetes 2016, 65, 2429–2439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Chappell, D.; Jacob, M.; Hofmann-Kiefer, K.; Bruegger, D.; Rehm, M.; Conzen, P.; Welsch, U.; Becker, B.F. Hydrocortisone Preserves the Vascular Barrier by Protecting the Endothelial Glycocalyx. Anesthesiology 2007, 107, 776–784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Brettner, F.; Chappell, D.; Nebelsiek, T.; Hauer, D.; Schelling, G.; Becker, B.F.; Rehm, M.; Weis, F. Preinterventional hydrocortisone sustains the endothelial glycocalyx in cardiac surgery. Clin. Hemorheol. Microcirc. 2019, 71, 59–70. [Google Scholar] [CrossRef] [PubMed]
  135. Chappell, D.; Dörfler, N.; Jacob, M.; Rehm, M.; Welsch, U.; Conzen, P.; Becker, B.F. Glycocalyx protection reduces leukocyte adhesion after ischemia/reperfusion. Shock 2010, 34, 133–139. [Google Scholar] [CrossRef] [PubMed]
  136. Meuwese, M.C.; Mooij, H.L.; Nieuwdorp, M.; van Lith, B.; Marck, R.; Vink, H.; Kastelein, J.J.P.; Stroes, E.S.G. Partial recovery of the endothelial glycocalyx upon rosuvastatin therapy in patients with heterozygous familial hypercholesterolemia. J. Lipid Res. 2009, 50, 148–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Root-Bernstein, R.; Huber, J.; Ziehl, A. Complementary Sets of Autoantibodies Induced by SARS-CoV-2, Adenovirus and Bacterial Antigens Cross-React with Human Blood Protein Antigens in COVID-19 Coagulopathies. Int. J. Mol. Sci. 2022, 23, 11500. [Google Scholar] [CrossRef]
  138. Xu, S.-W.; Ilyas, I.; Weng, J.-P. Endothelial dysfunction in COVID-19: An overview of evidence, biomarkers, mechanisms and potential therapies. Acta Pharmacol. Sin. 2022, 1–15, (Epub ahead of print). [Google Scholar] [CrossRef]
  139. Caillon, A.; Trimaille, A.; Favre, J.; Jesel, L.; Morel, O.; Kauffenstein, G. Role of neutrophils, platelets, and ex-tracellular vesicles and their interactions in COVID-19-associated thrombopathy. J. Thromb. Haemost. 2022, 20, 17–31. [Google Scholar] [CrossRef]
Figure 1. Endothelial dysfunction with glycocalyx degradation is postulated as the central mechanism induced through and facilitating viral entry. The resulting inflammatory and procoagulative processes initiate a complex cascade destabilizing the equilibrium of Virchow’s triad resulting in (micro-) thrombosis and tissue necrosis. The concept of the figure is modified after Ataga et al.—‘Hypercoagulability and thrombotic complications in hemolytic anemias.’ [23].
Figure 1. Endothelial dysfunction with glycocalyx degradation is postulated as the central mechanism induced through and facilitating viral entry. The resulting inflammatory and procoagulative processes initiate a complex cascade destabilizing the equilibrium of Virchow’s triad resulting in (micro-) thrombosis and tissue necrosis. The concept of the figure is modified after Ataga et al.—‘Hypercoagulability and thrombotic complications in hemolytic anemias.’ [23].
Ijms 24 02492 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wadowski, P.P.; Panzer, B.; Józkowicz, A.; Kopp, C.W.; Gremmel, T.; Panzer, S.; Koppensteiner, R. Microvascular Thrombosis as a Critical Factor in Severe COVID-19. Int. J. Mol. Sci. 2023, 24, 2492. https://doi.org/10.3390/ijms24032492

AMA Style

Wadowski PP, Panzer B, Józkowicz A, Kopp CW, Gremmel T, Panzer S, Koppensteiner R. Microvascular Thrombosis as a Critical Factor in Severe COVID-19. International Journal of Molecular Sciences. 2023; 24(3):2492. https://doi.org/10.3390/ijms24032492

Chicago/Turabian Style

Wadowski, Patricia P., Benjamin Panzer, Alicja Józkowicz, Christoph W. Kopp, Thomas Gremmel, Simon Panzer, and Renate Koppensteiner. 2023. "Microvascular Thrombosis as a Critical Factor in Severe COVID-19" International Journal of Molecular Sciences 24, no. 3: 2492. https://doi.org/10.3390/ijms24032492

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop