Skip to main content
Erschienen in: Critical Care 1/2021

Open Access 01.12.2021 | COVID-19 | Review

Endotheliopathy in septic conditions: mechanistic insight into intravascular coagulation

verfasst von: Takashi Ito, Midori Kakuuchi, Ikuro Maruyama

Erschienen in: Critical Care | Ausgabe 1/2021

Abstract

Endothelial cells play a key role in maintaining intravascular patency through their anticoagulant properties. They provide a favorable environment for plasma anticoagulant proteins, including antithrombin, tissue factor pathway inhibitor, and protein C. Under septic conditions, however, the anticoagulant properties of endothelial cells are compromised. Rather, activated/injured endothelial cells can provide a scaffold for intravascular coagulation. For example, the expression of tissue factor, an important initiator of the coagulation pathway, is induced on the surface of activated endothelial cells. Phosphatidylserine, a high-affinity scaffold for gamma-carboxyglutamate domain containing coagulation factors, including FII, FVII, FIX, and FX, is externalized to the outer leaflet of the plasma membrane of injured endothelial cells. Hemodilution decreases not only coagulation factors but also plasma anticoagulant proteins, resulting in unleashed activation of coagulation on the surface of activated/injured endothelial cells. The aberrant activation of coagulation can be suppressed in part by the supplementation of recombinant antithrombin and recombinant thrombomodulin. This review aims to overview the physiological and pathological functions of endothelial cells along with proof-of-concept in vitro studies. The pathophysiology of COVID-19-associated thrombosis is also discussed.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s13054-021-03524-6.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
APC
Activated protein C
DIC
Disseminated intravascular coagulation
LPS
Lipopolysaccharide
NETs
Neutrophil extracellular traps
rAT
Recombinant antithrombin gamma
rTM
Recombinant thrombomodulin
TF
Tissue factor
TFPI
Tissue factor pathway inhibitor

Background

Blood must be in a fluid state while in blood vessels. However, it must change to a solid state when leaking out of blood vessels. Disseminated intravascular coagulation (DIC) is a pathogenic condition characterized by aberrant activation of coagulation within blood vessels and insufficient activation of coagulation outside blood vessels [1]. This paradoxical condition displays complex clinical manifestations of simultaneous thrombosis and bleeding.
Sepsis is defined as life-threatening organ dysfunction caused by dysregulated host responses to infection [2]. Among dysregulated host responses, including exaggerated inflammation, coagulation, vascular leakage, and tissue hypoperfusion [3], endotheliopathy might play a central role in the pathogenesis of sepsis [4]. For example, disruption of endothelial glycocalyx in septic conditions results in augmented leukocyte adhesion, intravascular coagulation, tissue edema, and dysregulated vasodilatation [5, 6]. In this review, we summarize physiological roles of endothelial cells in maintaining intravascular homeostasis, pathological roles of activated/injured endothelial cells in septic conditions, and proof-of-concept in vitro studies showing these differential roles of endothelial cells.

Inhibition of coagulation in the intravascular space

Thrombosis is a frequent complication of blood-contacting medical devices, such as vascular grafts, stents, heart valves, central venous catheters, and extracorporeal circuits [7]. Conversely, this indicates that blood-contacting endothelial cells actively resist thrombosis [8]. Endothelial cells synthesize and display heparan sulphate proteoglycans, a component of glycocalyx, which bind and potentiate plasma anticoagulant proteins, including tissue factor pathway inhibitor (TFPI) and antithrombin [9]. Endothelial cells also express thrombomodulin, which binds thrombin and converts its substrate specificity from procoagulant to anticoagulant. By binding to thrombomodulin, thrombin loses its affinity for fibrinogen, coagulation factor V (FV), FVIII, FXIII, and protease-activated receptors, and instead activates anticoagulant protein C [10]. Endothelial protein C receptor (EPCR) augments this reaction by positioning protein C so that thrombin-thrombomodulin complexes can effectively activate it [11]. Activated protein C (APC) then limits the amplification of coagulation by inactivating FVa and FVIIIa with support from cofactor protein S (Fig. 1). EPCR–APC complexes also elicit cytoprotective effects, including antiapoptotic and barrier stabilizing effects, in endothelial cells. Loss of function of antithrombin, protein C, protein S, and thrombomodulin leads to thrombophilia with varied clinical manifestations, which suggests that these anticoagulant proteins are essential for resistance to intravascular coagulation [1216]. Furthermore, endothelial cells synthesize and release tissue-type plasminogen activator (tPA) in constitutive and/or regulated manners, potentiating plasmin-mediated fibrinolysis within the vasculature [17].
To confirm the concept that an artificial surface is procoagulant while an endothelial surface is anticoagulant, we conducted proof-of-concept in vitro experiments. In these experiments, a synthetic fluorogenic substrate SN-20 was used for monitoring thrombin generation in normal human plasma in the presence of calcium. When plasma was placed on the artificial surface, thrombin was generated after a lag period of about 10 min (Fig. 2a, green). This reaction was completely inhibited by corn trypsin inhibitor, a potent and specific inhibitor of FXIIa (Fig. 2b), which suggests that thrombin generation on the artificial surface is mediated by the intrinsic coagulation pathway. Conversely, thrombin was not generated during the 30-min experimental period when plasma was placed on the surface of cultured endothelial cells (Fig. 2a, blue). These findings indicate that endothelial cells actually confer resistance to the activation of coagulation.

Activation of coagulation in the perivascular space

Coagulation properties are controlled not only by the coagulation factors but also by the cellular components. Tissue factor (TF)-bearing fibroblasts, which reside in the perivascular space but not in the intravascular space under physiologic conditions, play an important role in the initiation of hemostasis [18]. During the process of hemostasis, an injury to the vessel wall allows plasma coagulation factors, including FVII, to come into contact with TF-bearing fibroblasts (Fig. 1). The FVIIa/TF complex then activates FX and FIX. This results in the generation of a small amount of thrombin, insufficient to generate fibrin but enough to activate FV, FVIII, FXI, and platelets. This pathway serves as a propagation of coagulation, leading to the generation of large amounts of thrombin sufficient for fibrin formation [19]. Fibrin polymers are then stabilized by FXIIIa, which introduces fibrin–fibrin and fibrin–α2-antiplasmin (α2AP) cross-links [20]. In our proof-of-concept in vitro experiments, thrombin was not generated on the surface of endothelial cells (Fig. 2a, blue) but was rapidly generated on the surface of fibroblasts (Fig. 2a, red). These findings indicate that our experimental conditions may mimic rapid coagulation in the perivascular space, with no coagulation within the vasculature and gradual coagulation on the artificial surface.

Activation of coagulation in the intravascular space in septic conditions

While the intravascular space is normally free from thrombosis, intravascular coagulation may occur in septic conditions [21, 22]. In mouse models of infection with Staphylococcus aureus, Escherichia coli, or lipopolysaccharide (LPS), profound thrombin generation could be observed within the liver microcirculation [22]. Consistent with this, plasma levels of thrombin-antithrombin complex, a clinical biomarker of thrombin generation, were elevated in most patients with sepsis [23]. Activated leukocytes are prominently involved in the pathogenesis of intravascular coagulation as well as overwhelming inflammation in such conditions [24, 25]. In response to microbial stimuli, activated neutrophils release neutrophil extracellular traps (NETs), which can provide a scaffold and stimulus for intravascular coagulation (Fig. 3). NETs comprise DNA, histones, and neutrophil serine proteases, all of which are involved in the activation of coagulation. Negatively charged surfaces of DNA serve as a promoter of the intrinsic coagulation pathway by contact with FXII and FXI [26]. Neutrophil serine proteases inactivate anticoagulant TFPI [27]. Extracellular histones bind to prothrombin, to facilitate FXa-mediated cleavage of prothrombin to release active thrombin [28]. The TF-dependent extrinsic coagulation pathway is also involved in intravascular coagulation, which takes place on the surface of activated endothelial cells and microvesicles originating from activated monocytes [26, 29, 30]. Fibrinolysis shutdown in septic conditions further exacerbates microvascular thrombosis by preventing fibrin removal [31]. Increased levels of plasminogen activator inhibitor-1 (PAI-1) and thrombin-activatable fibrinolysis inhibitor (TAFI) are associated with organ failure and poor outcomes in patients with severe sepsis [32]. Disruption of the endothelial homeostasis by angiopoietin-2 also plays a fundamental role in the pathogenesis of sepsis-associated DIC [4].
To examine whether activated/injured endothelial cells can be procoagulant, we conducted proof-of-concept in vitro experiments. When plasma was placed on the surface of endothelial cells pretreated with LPS or histones, thrombin was generated over time (Fig. 2c, purple and navy). In contrast to the artificial surface, activated/injured endothelial surface exhibited shorter lag time but slower rate of thrombin generation (Fig. 2c, 2d), which suggests that activated/injured endothelial cells might provide both initiators and inhibitors of the coagulation pathway, although the latter might be insufficient compared with normal endothelial cells. Immunoblot analysis revealed that stimulation with LPS resulted in the induction of TF expression and a slight reduction of thrombomodulin expression in endothelial cells (Additional file 1). The induction of TF was responsible for thrombin generation on the surface of LPS-stimulated endothelial cells because treatment with anti-TF antibodies completely diminished thrombin generation (Fig. 2d). Stimulation with histones resulted in the profound reduction of thrombomodulin expression and the exposure of phosphatidylserine to the outer leaflet of the plasma membrane in endothelial cells (Additional file 1). Thus, the effects of LPS and histones on endothelial cells are mechanistically different although both stimulants compromise anticoagulant properties of endothelial cells.

Impact of hemodilution on procoagulant-anticoagulant balance

Fluid resuscitation decreases plasma levels of coagulation factors, which could eventually result in impaired hemostasis, called dilutional coagulopathy [33]. In our in vitro experiments using fibroblasts, which represent rapid coagulation in the perivascular space, threefold dilution of plasma delayed the time to onset of thrombin generation (Fig. 4a). By contrast, in our in vitro experiments using endothelial cells pretreated with LPS, which represent gradual coagulation in the intravascular space under pathological conditions, threefold dilution of plasma increased thrombin generation (Fig. 4b and Additional file 2). This might have occurred because activation of coagulation was unleashed by the decrease in plasma anticoagulant proteins. The anticoagulant pathways can be far more affected by dilution than the procoagulant pathways in the situation where gradual coagulation takes place [34, 35]. These findings indicate that patients with hemodilution are at risk of intravascular coagulation as well as perivascular bleeding.

Suppression of coagulation by recombinant thrombomodulin and antithrombin

In septic conditions, anticoagulant potential in the intravascular environment can be compromised because of disruption of endothelial glycocalyx, downregulation of endothelial thrombomodulin, and decline of plasma anticoagulant proteins such as TFPI and antithrombin. Recombinant thrombomodulin (rTM) and antithrombin gamma (rAT) are potential therapeutic agents that may restore anticoagulant potential within the septic microcirculation (Fig. 3). Similar to endogenous thrombomodulin, rTM binds to thrombin to generate APC, which can limit the amplification of coagulation without extending coagulation times [36]. rAT is an alternative to plasma-derived antithrombin, which traps activated coagulation factors, including thrombin and FXa [37]. In our proof-of-concept in vitro experiments using LPS-stimulated endothelial cells, thrombin generation was partially suppressed by the supplementation with rAT (Fig. 5a) and rTM (Fig. 5b). Concomitant use of rTM and rAT showed additive effects and efficiently suppressed thrombin generation on the surface of LPS-stimulated endothelial cells (Fig. 5c and Additional file 2). These findings offer valuable insight into the potential of combination therapy with these two drugs because it remains incompletely understood whether rAT may complement anticoagulant effects of the rTM-APC axis or may counteract the rTM-APC axis through inhibition of thrombin-mediated APC generation [38].

Perspective of anticoagulant therapy in sepsis-associated DIC

A large-scale, randomized, double-blind, placebo-controlled, phase 3 clinical trial, named the KyberSept trial, was undertaken to determine the clinical efficacy of antithrombin in patients with severe sepsis [39]. In this trial, a total of 2314 patients were randomized into two groups to receive either intravenous antithrombin (30,000 IU in total over 4 days) or a placebo (1% human albumin). Although high-dose administration of antithrombin offered no mortality advantage over standard care for sepsis (38.9% vs. 38.7%, P = 0.94), there was a trend toward reduced 28-day (37.8% vs. 43.6%, P = 0.08) and 90-day mortality (44.9% vs. 52.5%, P = 0.03) with antithrombin in the predefined subgroup of patients not receiving concomitant heparin. Furthermore, a post hoc analysis showed that a reduced 28-day mortality with antithrombin was observed in patients with DIC (25.4% vs. 40.0%, P = 0.024), whereas no effect was seen in patients without DIC (22.1% vs. 22.2%, P > 0.2) [40].
The efficacy and safety of recombinant human APC (rhAPC) in patients with severe sepsis was examined in a large-scale, randomized, double-blind, placebo-controlled, phase 3 clinical trial, named the PROWESS trial [41]. In this trial, a total of 1690 patients were randomized into two groups to receive either intravenous rhAPC (continuous infusion for 96 h) or a placebo. Administration of rhAPC significantly reduced 28-day all-cause mortality (24.7% vs. 30.8%, P = 0.005). However, subsequent trials failed to show the efficacy and safety of rhAPC [42, 43], due in part to patient selection and bleeding side effects, leading to its removal from clinical use.
The efficacy and safety of rTM in severe sepsis patients with low platelet counts and prolonged prothrombin times were examined in a randomized, double-blind, placebo-controlled, phase 3 clinical trial, named the SCARLET trial [44]. In this trial, a total of 816 patients were randomized into two groups to receive either intravenous rTM (once daily for 6 days) or a placebo. Administration of rTM did not significantly reduce 28-day all-cause mortality (26.8% vs. 29.4%, P = 0.32). A post hoc analysis indicated that the survival benefit with rTM was greater in subgroups with higher levels of thrombin generation at baseline [45]. This might be consistent with the previous finding of high-dose antithrombin, considering that the diagnosis of DIC is based on low platelet counts, prolonged prothrombin times, and increased coagulation biomarker levels [4648].
A meta-analysis of randomized controlled trials indicated that the survival benefit with anticoagulant therapy was not observed in the overall sepsis population but was observed in the population with sepsis-induced DIC [49]. Thus, the most important issue associated with anticoagulant therapy in septic patients is target selection. Thus far, it is suggested that an optimal target for anticoagulant therapy may be septic patients with DIC and high disease severity. However, this is not based on definitive evidence, and thus, further prospective studies are needed [50].

Perspective of COVID-19-associated endotheliopathy

Since the identification of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in China in late 2019, the coronavirus disease 2019 (COVID-19) has become pandemic. The clinical manifestation of COVID-19 varies substantially, ranging from almost asymptomatic to life-threatening. Thrombosis is a common and potentially lethal complication of COVID-19 [51, 52]. Endothelial cells are severely injured in alveolar capillaries in fatal COVID-19 cases [53], suggesting that endotheliopathy may play a pivotal role in the pathogenesis of COVID-19-associated thrombosis as in the case of sepsis-associated DIC. However, they differ from each other in several ways. First, thrombocytopenia, prolongation of prothrombin times, and elevation of PAI-1, which are typical in sepsis-associated DIC, are less common in COVID-19-associated thrombosis, at least in the early to mid-stage of this disease [54, 55]. Second, COVID-19-associated thrombosis manifests as not only microvascular thrombosis but also macrovascular thrombosis, such as stroke, myocardial infarction, and venous thromboembolism [56].
Key mechanisms that may cause endotheliopathy secondary to SARS-CoV-2 infection include direct viral toxicity and immune-mediated damage. Given that angiotensin-converting enzyme 2 and transmembrane serine protease 2, which are utilized by SARS-CoV-2 as tools for entry into host cells, are expressed on endothelial cells [57], direct viral toxicity against endothelial cells is a plausible mechanism [53]. Immune-mediated mechanisms may also play a key role particularly in cases where thrombotic organ damage develops in the absence of SARS-CoV-2 viremia or continues to worsen even after most of the virus has been cleared. Dysregulated cytokines, complement, platelets, and neutrophils cooperate to drive a systemic thrombo-inflammatory disorder in COVID-19, as well as traditional sepsis [52, 58].
In addition to these innate immune mediators, autoantibodies may have a significant role in the development of potentially lethal complications in COVID-19 [59, 60]. Compared to uninfected individuals, COVID-19 patients exhibit dramatic increases in autoantibodies against components of their blood vessels, heart, brain, and immune system [61]. Some of the autoantibodies target phospholipids and phospholipid-binding proteins on the surface of endothelial cells, platelets, and neutrophils, tipping the blood–endothelium interface toward thrombosis [59, 62]. Immunoglobulin G isolated from COVID-19 patients has the potential to induce NET release and accelerate thrombosis in mice. Given that the development of autoantibodies generally takes 1–2 weeks, the autoantibody theory might explain some of the delay in the onset of severe complications in COVID-19 [63]. It will be important to understand whether autoantibodies and neutralizing antibodies may persist for a long period after infection, whether these antibodies have direct pathogenic roles, and whether these immune responses have implications for the treatment of COVID-19.

Conclusions

Endothelial cells confer resistance to the activation of coagulation. Under septic conditions, however, the anticoagulant properties of endothelial cells are compromised, and activated/injured endothelial cells can provide a scaffold for intravascular coagulation. Hemodilution can more profoundly affect anticoagulant pathways than procoagulant pathways and further promote activation of coagulation on the surface of activated/injured endothelial cells. The aberrant activation of coagulation can be suppressed in part by the supplementation with rAT and rTM. These anticoagulants may provide survival benefit in a subpopulation of septic patients who suffer from DIC.

Acknowledgements

We thank Mark Abramovitz, PhD, from Edanz Group (https://​en-author-services.​edanz.​com/​ac) for editing a draft of this manuscript.

Declarations

Not applicable.
Not applicable.

Competing interests

TI and IM received research grants from Asahi Kasei Pharma, a pharmaceutical company manufacturing recombinant thrombomodulin. The fund is for academic promotion and is not directly related to this study. MK declares that she has no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Gando S, Levi M, Toh CH. Disseminated intravascular coagulation. Nat Rev Dis Primers. 2016;2:16037.PubMedCrossRef Gando S, Levi M, Toh CH. Disseminated intravascular coagulation. Nat Rev Dis Primers. 2016;2:16037.PubMedCrossRef
2.
Zurück zum Zitat Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, et al. The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016;315(8):801–10.PubMedPubMedCentralCrossRef Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, et al. The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016;315(8):801–10.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17(7):407–20.PubMedCrossRef van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17(7):407–20.PubMedCrossRef
4.
Zurück zum Zitat Higgins SJ, De Ceunynck K, Kellum JA, Chen X, Gu X, Chaudhry SA, Schulman S, Libermann TA, Lu S, Shapiro NI, et al. Tie2 protects the vasculature against thrombus formation in systemic inflammation. J Clin Investig. 2018;128(4):1471–84.PubMedCrossRefPubMedCentral Higgins SJ, De Ceunynck K, Kellum JA, Chen X, Gu X, Chaudhry SA, Schulman S, Libermann TA, Lu S, Shapiro NI, et al. Tie2 protects the vasculature against thrombus formation in systemic inflammation. J Clin Investig. 2018;128(4):1471–84.PubMedCrossRefPubMedCentral
5.
Zurück zum Zitat Okada H, Takemura G, Suzuki K, Oda K, Takada C, Hotta Y, Miyazaki N, Tsujimoto A, Muraki I, Ando Y, et al. Three-dimensional ultrastructure of capillary endothelial glycocalyx under normal and experimental endotoxemic conditions. Crit Care. 2017;21(1):261.PubMedPubMedCentralCrossRef Okada H, Takemura G, Suzuki K, Oda K, Takada C, Hotta Y, Miyazaki N, Tsujimoto A, Muraki I, Ando Y, et al. Three-dimensional ultrastructure of capillary endothelial glycocalyx under normal and experimental endotoxemic conditions. Crit Care. 2017;21(1):261.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Jaffer IH, Fredenburgh JC, Hirsh J, Weitz JI. Medical device-induced thrombosis: what causes it and how can we prevent it? J Thromb Haemost. 2015;13(Suppl 1):S72-81.PubMedCrossRef Jaffer IH, Fredenburgh JC, Hirsh J, Weitz JI. Medical device-induced thrombosis: what causes it and how can we prevent it? J Thromb Haemost. 2015;13(Suppl 1):S72-81.PubMedCrossRef
8.
Zurück zum Zitat Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation. Nat Rev Immunol. 2007;7(10):803–15.PubMedCrossRef Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation. Nat Rev Immunol. 2007;7(10):803–15.PubMedCrossRef
9.
Zurück zum Zitat Marcum JA, McKenney JB, Rosenberg RD. Acceleration of thrombin-antithrombin complex formation in rat hindquarters via heparinlike molecules bound to the endothelium. J Clin Investig. 1984;74(2):341–50.PubMedCrossRefPubMedCentral Marcum JA, McKenney JB, Rosenberg RD. Acceleration of thrombin-antithrombin complex formation in rat hindquarters via heparinlike molecules bound to the endothelium. J Clin Investig. 1984;74(2):341–50.PubMedCrossRefPubMedCentral
10.
Zurück zum Zitat Ito T, Maruyama I. Thrombomodulin: protectorate God of the vasculature in thrombosis and inflammation. J Thromb Haemost. 2011;9(Suppl 1):168–73.PubMedCrossRef Ito T, Maruyama I. Thrombomodulin: protectorate God of the vasculature in thrombosis and inflammation. J Thromb Haemost. 2011;9(Suppl 1):168–73.PubMedCrossRef
11.
Zurück zum Zitat Mohan Rao LV, Esmon CT, Pendurthi UR. Endothelial cell protein C receptor: a multiliganded and multifunctional receptor. Blood. 2014;124(10):1553–62.PubMedPubMedCentralCrossRef Mohan Rao LV, Esmon CT, Pendurthi UR. Endothelial cell protein C receptor: a multiliganded and multifunctional receptor. Blood. 2014;124(10):1553–62.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Patnaik MM, Moll S. Inherited antithrombin deficiency: a review. Haemophilia. 2008;14(6):1229–39.PubMedCrossRef Patnaik MM, Moll S. Inherited antithrombin deficiency: a review. Haemophilia. 2008;14(6):1229–39.PubMedCrossRef
13.
Zurück zum Zitat Stevens SM, Woller SC, Bauer KA, Kasthuri R, Cushman M, Streiff M, Lim W, Douketis JD. Guidance for the evaluation and treatment of hereditary and acquired thrombophilia. J Thromb Thrombolysis. 2016;41(1):154–64.PubMedPubMedCentralCrossRef Stevens SM, Woller SC, Bauer KA, Kasthuri R, Cushman M, Streiff M, Lim W, Douketis JD. Guidance for the evaluation and treatment of hereditary and acquired thrombophilia. J Thromb Thrombolysis. 2016;41(1):154–64.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Okada M, Tominaga N, Honda G, Nishioka J, Akita N, Hayashi T, Suzuki K, Moriuchi H. A case of thrombomodulin mutation causing defective thrombin binding with absence of protein C and TAFI activation. Blood Adv. 2020;4(12):2631–9.PubMedPubMedCentralCrossRef Okada M, Tominaga N, Honda G, Nishioka J, Akita N, Hayashi T, Suzuki K, Moriuchi H. A case of thrombomodulin mutation causing defective thrombin binding with absence of protein C and TAFI activation. Blood Adv. 2020;4(12):2631–9.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Isermann B, Hendrickson SB, Zogg M, Wing M, Cummiskey M, Kisanuki YY, Yanagisawa M, Weiler H. Endothelium-specific loss of murine thrombomodulin disrupts the protein C anticoagulant pathway and causes juvenile-onset thrombosis. J Clin Investig. 2001;108(4):537–46.PubMedCrossRefPubMedCentral Isermann B, Hendrickson SB, Zogg M, Wing M, Cummiskey M, Kisanuki YY, Yanagisawa M, Weiler H. Endothelium-specific loss of murine thrombomodulin disrupts the protein C anticoagulant pathway and causes juvenile-onset thrombosis. J Clin Investig. 2001;108(4):537–46.PubMedCrossRefPubMedCentral
16.
Zurück zum Zitat Manco-Johnson MJ, Bomgaars L, Palascak J, Shapiro A, Geil J, Fritsch S, Pavlova BG, Gelmont D. Efficacy and safety of protein C concentrate to treat purpura fulminans and thromboembolic events in severe congenital protein C deficiency. Thromb Haemost. 2016;116(1):58–68.PubMed Manco-Johnson MJ, Bomgaars L, Palascak J, Shapiro A, Geil J, Fritsch S, Pavlova BG, Gelmont D. Efficacy and safety of protein C concentrate to treat purpura fulminans and thromboembolic events in severe congenital protein C deficiency. Thromb Haemost. 2016;116(1):58–68.PubMed
17.
Zurück zum Zitat Urano T, Castellino FJ, Suzuki Y. Regulation of plasminogen activation on cell surfaces and fibrin. J Thromb Haemost. 2018;16:1487–97.PubMedCentralCrossRef Urano T, Castellino FJ, Suzuki Y. Regulation of plasminogen activation on cell surfaces and fibrin. J Thromb Haemost. 2018;16:1487–97.PubMedCentralCrossRef
18.
Zurück zum Zitat Hoffman M, Monroe DM 3rd. A cell-based model of hemostasis. Thromb Haemost. 2001;85(6):958–65.PubMedCrossRef Hoffman M, Monroe DM 3rd. A cell-based model of hemostasis. Thromb Haemost. 2001;85(6):958–65.PubMedCrossRef
19.
Zurück zum Zitat Swieringa F, Spronk HMH, Heemskerk JWM, van der Meijden PEJ. Integrating platelet and coagulation activation in fibrin clot formation. Res Pract Thrombosis Haemostasis. 2018;2(3):450–60.CrossRef Swieringa F, Spronk HMH, Heemskerk JWM, van der Meijden PEJ. Integrating platelet and coagulation activation in fibrin clot formation. Res Pract Thrombosis Haemostasis. 2018;2(3):450–60.CrossRef
20.
Zurück zum Zitat Pieters M, Wolberg AS. Fibrinogen and fibrin: An illustrated review. Res Pract Thrombosis Haemostasis. 2019;3(2):161–72.CrossRef Pieters M, Wolberg AS. Fibrinogen and fibrin: An illustrated review. Res Pract Thrombosis Haemostasis. 2019;3(2):161–72.CrossRef
21.
Zurück zum Zitat Levi M, Ten Cate H. Disseminated intravascular coagulation. N Engl J Med. 1999;341(8):586–92.PubMedCrossRef Levi M, Ten Cate H. Disseminated intravascular coagulation. N Engl J Med. 1999;341(8):586–92.PubMedCrossRef
22.
Zurück zum Zitat McDonald B, Davis RP, Kim SJ, Tse M, Esmon CT, Kolaczkowska E, Jenne CN. Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice. Blood. 2017;129(10):1357–67.PubMedPubMedCentralCrossRef McDonald B, Davis RP, Kim SJ, Tse M, Esmon CT, Kolaczkowska E, Jenne CN. Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice. Blood. 2017;129(10):1357–67.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Koyama K, Madoiwa S, Nunomiya S, Koinuma T, Wada M, Sakata A, Ohmori T, Mimuro J, Sakata Y. Combination of thrombin-antithrombin complex, plasminogen activator inhibitor-1, and protein C activity for early identification of severe coagulopathy in initial phase of sepsis: a prospective observational study. Crit Care. 2014;18(1):R13.PubMedPubMedCentralCrossRef Koyama K, Madoiwa S, Nunomiya S, Koinuma T, Wada M, Sakata A, Ohmori T, Mimuro J, Sakata Y. Combination of thrombin-antithrombin complex, plasminogen activator inhibitor-1, and protein C activity for early identification of severe coagulopathy in initial phase of sepsis: a prospective observational study. Crit Care. 2014;18(1):R13.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Bohn E, Muller-Berghaus G. The effect of leukocyte and platelet transfusion on the activation of intravascular coagulation by endotoxin in granulocytopenic and thrombocytopenic rabbits. Am J Pathol. 1976;84(2):239–58.PubMedPubMedCentral Bohn E, Muller-Berghaus G. The effect of leukocyte and platelet transfusion on the activation of intravascular coagulation by endotoxin in granulocytopenic and thrombocytopenic rabbits. Am J Pathol. 1976;84(2):239–58.PubMedPubMedCentral
26.
Zurück zum Zitat von Bruhl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, Khandoga A, Tirniceriu A, Coletti R, Kollnberger M, et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med. 2012;209(4):819–35.CrossRef von Bruhl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, Khandoga A, Tirniceriu A, Coletti R, Kollnberger M, et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med. 2012;209(4):819–35.CrossRef
27.
Zurück zum Zitat Massberg S, Grahl L, von Bruehl ML, Manukyan D, Pfeiler S, Goosmann C, Brinkmann V, Lorenz M, Bidzhekov K, Khandagale AB, et al. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med. 2010;16(8):887–96.PubMedCrossRef Massberg S, Grahl L, von Bruehl ML, Manukyan D, Pfeiler S, Goosmann C, Brinkmann V, Lorenz M, Bidzhekov K, Khandagale AB, et al. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med. 2010;16(8):887–96.PubMedCrossRef
28.
Zurück zum Zitat Abrams ST, Su D, Sahraoui Y, Lin Z, Cheng Z, Nesbitt K, Alhamdi Y, Harrasser M, Du M, Foley JH, et al. Assembly of alternative prothrombinase by extracellular histones initiates and disseminates intravascular coagulation. Blood. 2021;137(1):103–14.PubMedCrossRef Abrams ST, Su D, Sahraoui Y, Lin Z, Cheng Z, Nesbitt K, Alhamdi Y, Harrasser M, Du M, Foley JH, et al. Assembly of alternative prothrombinase by extracellular histones initiates and disseminates intravascular coagulation. Blood. 2021;137(1):103–14.PubMedCrossRef
29.
Zurück zum Zitat Lupu C, Westmuckett AD, Peer G, Ivanciu L, Zhu H, Taylor FB, Lupu F. Tissue factor-dependent coagulation is preferentially up-regulated within arterial branching areas in a baboon model of Escherichia coli Sepsis. Am J Pathol. 2005;167(4):1161–72.PubMedPubMedCentralCrossRef Lupu C, Westmuckett AD, Peer G, Ivanciu L, Zhu H, Taylor FB, Lupu F. Tissue factor-dependent coagulation is preferentially up-regulated within arterial branching areas in a baboon model of Escherichia coli Sepsis. Am J Pathol. 2005;167(4):1161–72.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Falati S, Liu Q, Gross P, Merrill-Skoloff G, Chou J, Vandendries E, Celi A, Croce K, Furie BC, Furie B. Accumulation of tissue factor into developing thrombi in vivo is dependent upon microparticle P-selectin glycoprotein ligand 1 and platelet P-selectin. J Exp Med. 2003;197(11):1585–98.PubMedPubMedCentralCrossRef Falati S, Liu Q, Gross P, Merrill-Skoloff G, Chou J, Vandendries E, Celi A, Croce K, Furie BC, Furie B. Accumulation of tissue factor into developing thrombi in vivo is dependent upon microparticle P-selectin glycoprotein ligand 1 and platelet P-selectin. J Exp Med. 2003;197(11):1585–98.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Madoiwa S. Recent advances in disseminated intravascular coagulation: endothelial cells and fibrinolysis in sepsis-induced DIC. J Intensive Care. 2015;3(1):8.PubMedPubMedCentralCrossRef Madoiwa S. Recent advances in disseminated intravascular coagulation: endothelial cells and fibrinolysis in sepsis-induced DIC. J Intensive Care. 2015;3(1):8.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Semeraro F, Colucci M, Caironi P, Masson S, Ammollo CT, Teli R, Semeraro N, Magnoli M, Salati G, Isetta M, et al. Platelet drop and fibrinolytic shutdown in patients with sepsis. Crit Care Med. 2018;46(3):e221–8.PubMedCrossRef Semeraro F, Colucci M, Caironi P, Masson S, Ammollo CT, Teli R, Semeraro N, Magnoli M, Salati G, Isetta M, et al. Platelet drop and fibrinolytic shutdown in patients with sepsis. Crit Care Med. 2018;46(3):e221–8.PubMedCrossRef
33.
Zurück zum Zitat Bolliger D, Görlinger K, Tanaka KA. Pathophysiology and treatment of coagulopathy in massive hemorrhage and hemodilution. Anesthesiology. 2010;113(5):1205–19.PubMedCrossRef Bolliger D, Görlinger K, Tanaka KA. Pathophysiology and treatment of coagulopathy in massive hemorrhage and hemodilution. Anesthesiology. 2010;113(5):1205–19.PubMedCrossRef
34.
Zurück zum Zitat De Smedt E, Wagenvoord R, Coen Hemker H. The technique of measuring thrombin generation with fluorogenic substrates: 3. The effects of sample dilution. Thrombosis Haemostasis. 2009;101(1):165–70.PubMedCrossRef De Smedt E, Wagenvoord R, Coen Hemker H. The technique of measuring thrombin generation with fluorogenic substrates: 3. The effects of sample dilution. Thrombosis Haemostasis. 2009;101(1):165–70.PubMedCrossRef
35.
Zurück zum Zitat Dunbar NM, Chandler WL. Thrombin generation in trauma patients. Transfusion. 2009;49(12):2652–60.PubMedCrossRef Dunbar NM, Chandler WL. Thrombin generation in trauma patients. Transfusion. 2009;49(12):2652–60.PubMedCrossRef
36.
Zurück zum Zitat Ito T, Maruyama I, Shimazaki S, Yamamoto Y, Aikawa N, Hirayama A, Honda G, Saito H. Effects of thrombomodulin alfa on hemostatic parameters in disseminated intravascular coagulation: Post hoc analysis of a phase 3 randomized controlled trial. Res Pract Thromb Haemost. 2020;4(7):1141–9.PubMedPubMedCentralCrossRef Ito T, Maruyama I, Shimazaki S, Yamamoto Y, Aikawa N, Hirayama A, Honda G, Saito H. Effects of thrombomodulin alfa on hemostatic parameters in disseminated intravascular coagulation: Post hoc analysis of a phase 3 randomized controlled trial. Res Pract Thromb Haemost. 2020;4(7):1141–9.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Endo S, Shimazaki R. An open-label, randomized, phase 3 study of the efficacy and safety of antithrombin gamma in patients with sepsis-induced disseminated intravascular coagulation syndrome. J Intensive Care. 2018;6:75.PubMedPubMedCentralCrossRef Endo S, Shimazaki R. An open-label, randomized, phase 3 study of the efficacy and safety of antithrombin gamma in patients with sepsis-induced disseminated intravascular coagulation syndrome. J Intensive Care. 2018;6:75.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Arishima T, Ito T, Yasuda T, Yashima N, Furubeppu H, Kamikokuryo C, Futatsuki T, Madokoro Y, Miyamoto S, Eguchi T, et al. Circulating activated protein C levels are not increased in septic patients treated with recombinant human soluble thrombomodulin. Thrombosis J. 2018;16:24.CrossRef Arishima T, Ito T, Yasuda T, Yashima N, Furubeppu H, Kamikokuryo C, Futatsuki T, Madokoro Y, Miyamoto S, Eguchi T, et al. Circulating activated protein C levels are not increased in septic patients treated with recombinant human soluble thrombomodulin. Thrombosis J. 2018;16:24.CrossRef
39.
Zurück zum Zitat Warren BL, Eid A, Singer P, Pillay SS, Carl P, Novak I, Chalupa P, Atherstone A, Penzes I, Kubler A, et al. Caring for the critically ill patient. High-dose antithrombin III in severe sepsis: a randomized controlled trial. JAMA. 2001;286(15):1869–78.PubMedCrossRef Warren BL, Eid A, Singer P, Pillay SS, Carl P, Novak I, Chalupa P, Atherstone A, Penzes I, Kubler A, et al. Caring for the critically ill patient. High-dose antithrombin III in severe sepsis: a randomized controlled trial. JAMA. 2001;286(15):1869–78.PubMedCrossRef
40.
Zurück zum Zitat Kienast J, Juers M, Wiedermann CJ, Hoffmann JN, Ostermann H, Strauss R, Keinecke HO, Warren BL, Opal SM. Treatment effects of high-dose antithrombin without concomitant heparin in patients with severe sepsis with or without disseminated intravascular coagulation. J Thromb Haemost. 2006;4(1):90–7.PubMedCrossRef Kienast J, Juers M, Wiedermann CJ, Hoffmann JN, Ostermann H, Strauss R, Keinecke HO, Warren BL, Opal SM. Treatment effects of high-dose antithrombin without concomitant heparin in patients with severe sepsis with or without disseminated intravascular coagulation. J Thromb Haemost. 2006;4(1):90–7.PubMedCrossRef
41.
Zurück zum Zitat Bernard GR, Vincent J-L, Laterre P-F, LaRosa SP, Dhainaut J-F, Lopez-Rodriguez A, Steingrub JS, Garber GE, Helterbrand JD, Ely EW, et al. Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med. 2001;344(10):699–709.PubMedCrossRef Bernard GR, Vincent J-L, Laterre P-F, LaRosa SP, Dhainaut J-F, Lopez-Rodriguez A, Steingrub JS, Garber GE, Helterbrand JD, Ely EW, et al. Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med. 2001;344(10):699–709.PubMedCrossRef
42.
Zurück zum Zitat Abraham E, Laterre P-F, Garg R, Levy H, Talwar D, Trzaskoma BL, François B, Guy JS, Brückmann M, Rea-Neto Á, et al. Drotrecogin alfa (activated) for adults with severe sepsis and a low risk of death. N Engl J Med. 2005;353(13):1332–41.PubMedCrossRef Abraham E, Laterre P-F, Garg R, Levy H, Talwar D, Trzaskoma BL, François B, Guy JS, Brückmann M, Rea-Neto Á, et al. Drotrecogin alfa (activated) for adults with severe sepsis and a low risk of death. N Engl J Med. 2005;353(13):1332–41.PubMedCrossRef
43.
Zurück zum Zitat Ranieri VM, Thompson BT, Barie PS, Dhainaut J-F, Douglas IS, Finfer S, Gårdlund B, Marshall JC, Rhodes A, Artigas A, et al. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med. 2012;366(22):2055–64.PubMedCrossRef Ranieri VM, Thompson BT, Barie PS, Dhainaut J-F, Douglas IS, Finfer S, Gårdlund B, Marshall JC, Rhodes A, Artigas A, et al. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med. 2012;366(22):2055–64.PubMedCrossRef
44.
Zurück zum Zitat Vincent JL, Francois B, Zabolotskikh I, Daga MK, Lascarrou JB, Kirov MY, Pettila V, Wittebole X, Meziani F, Mercier E, et al. Effect of a recombinant human soluble thrombomodulin on mortality in patients with sepsis-associated coagulopathy: the SCARLET randomized clinical trial. JAMA. 2019;321(20):1993–2002.PubMedPubMedCentralCrossRef Vincent JL, Francois B, Zabolotskikh I, Daga MK, Lascarrou JB, Kirov MY, Pettila V, Wittebole X, Meziani F, Mercier E, et al. Effect of a recombinant human soluble thrombomodulin on mortality in patients with sepsis-associated coagulopathy: the SCARLET randomized clinical trial. JAMA. 2019;321(20):1993–2002.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Levi M, Vincent J-L, Tanaka K, Radford AH, Kayanoki T, Fineberg DA, Hoppensteadt D, Fareed J. Effect of a recombinant human soluble thrombomodulin on baseline coagulation biomarker levels and mortality outcome in patients with sepsis-associated coagulopathy. Crit Care Med. 2020;48(8):1140–7.PubMedPubMedCentralCrossRef Levi M, Vincent J-L, Tanaka K, Radford AH, Kayanoki T, Fineberg DA, Hoppensteadt D, Fareed J. Effect of a recombinant human soluble thrombomodulin on baseline coagulation biomarker levels and mortality outcome in patients with sepsis-associated coagulopathy. Crit Care Med. 2020;48(8):1140–7.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Taylor FB Jr, Toh CH, Hoots WK, Wada H, Levi M. Scientific Subcommittee on Disseminated Intravascular Coagulation of the International Society on T, Haemostasis: Towards definition, clinical and laboratory criteria, and a scoring system for disseminated intravascular coagulation. Thromb Haemost. 2001;86(5):1327–30.PubMedCrossRef Taylor FB Jr, Toh CH, Hoots WK, Wada H, Levi M. Scientific Subcommittee on Disseminated Intravascular Coagulation of the International Society on T, Haemostasis: Towards definition, clinical and laboratory criteria, and a scoring system for disseminated intravascular coagulation. Thromb Haemost. 2001;86(5):1327–30.PubMedCrossRef
47.
Zurück zum Zitat Wada H, Gabazza EC, Asakura H, Koike K, Okamoto K, Maruyama I, Shiku H, Nobori T. Comparison of diagnostic criteria for disseminated intravascular coagulation (DIC): diagnostic criteria of the International Society of Thrombosis and Hemostasis and of the Japanese Ministry of Health and Welfare for overt DIC. Am J Hematol. 2003;74(1):17–22.PubMedCrossRef Wada H, Gabazza EC, Asakura H, Koike K, Okamoto K, Maruyama I, Shiku H, Nobori T. Comparison of diagnostic criteria for disseminated intravascular coagulation (DIC): diagnostic criteria of the International Society of Thrombosis and Hemostasis and of the Japanese Ministry of Health and Welfare for overt DIC. Am J Hematol. 2003;74(1):17–22.PubMedCrossRef
48.
Zurück zum Zitat Asakura H, Takahashi H, Uchiyama T, Eguchi Y, Okamoto K, Kawasugi K, Madoiwa S, Wada H. Proposal for new diagnostic criteria for DIC from the Japanese Society on Thrombosis and Hemostasis. Thrombosis Journal. 2016;14:42.PubMedPubMedCentralCrossRef Asakura H, Takahashi H, Uchiyama T, Eguchi Y, Okamoto K, Kawasugi K, Madoiwa S, Wada H. Proposal for new diagnostic criteria for DIC from the Japanese Society on Thrombosis and Hemostasis. Thrombosis Journal. 2016;14:42.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Umemura Y, Yamakawa K, Ogura H, Yuhara H, Fujimi S. Efficacy and safety of anticoagulant therapy in three specific populations with sepsis: a meta-analysis of randomized controlled trials. J Thromb Haemost. 2016;14(3):518–30.PubMedCrossRef Umemura Y, Yamakawa K, Ogura H, Yuhara H, Fujimi S. Efficacy and safety of anticoagulant therapy in three specific populations with sepsis: a meta-analysis of randomized controlled trials. J Thromb Haemost. 2016;14(3):518–30.PubMedCrossRef
50.
Zurück zum Zitat Umemura Y, Yamakawa K. Optimal patient selection for anticoagulant therapy in sepsis: an evidence-based proposal from Japan. J Thromb Haemost. 2018;16(3):462–4.PubMedCrossRef Umemura Y, Yamakawa K. Optimal patient selection for anticoagulant therapy in sepsis: an evidence-based proposal from Japan. J Thromb Haemost. 2018;16(3):462–4.PubMedCrossRef
51.
Zurück zum Zitat Wichmann D, Sperhake J-P, Lütgehetmann M, Steurer S, Edler C, Heinemann A, Heinrich F, Mushumba H, Kniep I, Schröder AS, et al. Autopsy findings and venous thromboembolism in patients with COVID-19. Ann Intern Med. 2020;173(4):268–77.PubMedCrossRef Wichmann D, Sperhake J-P, Lütgehetmann M, Steurer S, Edler C, Heinemann A, Heinrich F, Mushumba H, Kniep I, Schröder AS, et al. Autopsy findings and venous thromboembolism in patients with COVID-19. Ann Intern Med. 2020;173(4):268–77.PubMedCrossRef
52.
Zurück zum Zitat Aid M, Busman-Sahay K, Vidal SJ, Maliga Z, Bondoc S, Starke C, Terry M, Jacobson CA, Wrijil L, Ducat S, et al. Vascular disease and thrombosis in SARS-CoV-2-infected rhesus macaques. Cell. 2020;183(5):1354-1366.e1313.PubMedPubMedCentralCrossRef Aid M, Busman-Sahay K, Vidal SJ, Maliga Z, Bondoc S, Starke C, Terry M, Jacobson CA, Wrijil L, Ducat S, et al. Vascular disease and thrombosis in SARS-CoV-2-infected rhesus macaques. Cell. 2020;183(5):1354-1366.e1313.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Ackermann M, Verleden SE, 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(2):120–8.PubMedPubMedCentralCrossRef Ackermann M, Verleden SE, 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(2):120–8.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat McGonagle D, O’Donnell JS, Sharif K, Emery P, Bridgewood C. Immune mechanisms of pulmonary intravascular coagulopathy in COVID-19 pneumonia. Lancet Rheumatol. 2020;2(7):e437–45.PubMedPubMedCentralCrossRef McGonagle D, O’Donnell JS, Sharif K, Emery P, Bridgewood C. Immune mechanisms of pulmonary intravascular coagulopathy in COVID-19 pneumonia. Lancet Rheumatol. 2020;2(7):e437–45.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Umemura Y, Yamakawa K, Kiguchi T, Nishida T, Kawada M, Fujimi S. Hematological phenotype of COVID-19-induced coagulopathy: far from typical sepsis-induced coagulopathy. J Clin Med. 2020;9(9):2875.PubMedCentralCrossRef Umemura Y, Yamakawa K, Kiguchi T, Nishida T, Kawada M, Fujimi S. Hematological phenotype of COVID-19-induced coagulopathy: far from typical sepsis-induced coagulopathy. J Clin Med. 2020;9(9):2875.PubMedCentralCrossRef
56.
Zurück zum Zitat Gupta A, Madhavan MV, Sehgal K, Nair N, Mahajan S, Sehrawat TS, Bikdeli B, Ahluwalia N, Ausiello JC, Wan EY, et al. Extrapulmonary manifestations of COVID-19. Nat Med. 2020;26(7):1017–32.CrossRefPubMed Gupta A, Madhavan MV, Sehgal K, Nair N, Mahajan S, Sehrawat TS, Bikdeli B, Ahluwalia N, Ausiello JC, Wan EY, et al. Extrapulmonary manifestations of COVID-19. Nat Med. 2020;26(7):1017–32.CrossRefPubMed
57.
Zurück zum Zitat Pons S, Fodil S, Azoulay E, Zafrani L. The vascular endothelium: the cornerstone of organ dysfunction in severe SARS-CoV-2 infection. Crit Care. 2020;24(1):353.PubMedPubMedCentralCrossRef Pons S, Fodil S, Azoulay E, Zafrani L. The vascular endothelium: the cornerstone of organ dysfunction in severe SARS-CoV-2 infection. Crit Care. 2020;24(1):353.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Nicolai L, Leunig A, Brambs S, Kaiser R, Weinberger T, Weigand M, Muenchhoff M, Hellmuth JC, Ledderose S, Schulz H, et al. Immunothrombotic dysregulation in COVID-19 pneumonia is associated with respiratory failure and coagulopathy. Circulation. 2020;142(12):1176–89.PubMedPubMedCentralCrossRef Nicolai L, Leunig A, Brambs S, Kaiser R, Weinberger T, Weigand M, Muenchhoff M, Hellmuth JC, Ledderose S, Schulz H, et al. Immunothrombotic dysregulation in COVID-19 pneumonia is associated with respiratory failure and coagulopathy. Circulation. 2020;142(12):1176–89.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Zuo Y, Estes SK, Ali RA, Gandhi AA, Yalavarthi S, Shi H, Sule G, Gockman K, Madison JA, Zuo M, et al. Prothrombotic autoantibodies in serum from patients hospitalized with COVID-19. Sci Transl Med. 2020;12(570):eabd3876.PubMedPubMedCentralCrossRef Zuo Y, Estes SK, Ali RA, Gandhi AA, Yalavarthi S, Shi H, Sule G, Gockman K, Madison JA, Zuo M, et al. Prothrombotic autoantibodies in serum from patients hospitalized with COVID-19. Sci Transl Med. 2020;12(570):eabd3876.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Bastard P, Rosen LB, Zhang Q, Michailidis E, Hoffmann H-H, Zhang Y, Dorgham K, Philippot Q, Rosain J, Béziat V, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020;370(6515):eabd4585.PubMedPubMedCentralCrossRef Bastard P, Rosen LB, Zhang Q, Michailidis E, Hoffmann H-H, Zhang Y, Dorgham K, Philippot Q, Rosain J, Béziat V, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020;370(6515):eabd4585.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Wang EY, Mao T, Klein J, Dai Y, Huck JD, Liu F, Zheng NS, Zhou T, Israelow B, Wong P, et al. Diverse functional autoantibodies in patients with COVID-19. medRxiv 2020:2020.2012.2010.20247205. (not yet peer reviewed) Wang EY, Mao T, Klein J, Dai Y, Huck JD, Liu F, Zheng NS, Zhou T, Israelow B, Wong P, et al. Diverse functional autoantibodies in patients with COVID-19. medRxiv 2020:2020.2012.2010.20247205. (not yet peer reviewed)
62.
Zurück zum Zitat Zuniga M, Gomes C, Carsons SE, Bender MT, Cotzia P, Miao QR, Lee DC, Rodriguez A. Autoimmunity to the lung protective phospholipid-binding protein Annexin A2 predicts mortality among hospitalized COVID-19 patients. medRxiv 2021:2020.2012.2028.20248807. (not yet peer reviewed) Zuniga M, Gomes C, Carsons SE, Bender MT, Cotzia P, Miao QR, Lee DC, Rodriguez A. Autoimmunity to the lung protective phospholipid-binding protein Annexin A2 predicts mortality among hospitalized COVID-19 patients. medRxiv 2021:2020.2012.2028.20248807. (not yet peer reviewed)
63.
Metadaten
Titel
Endotheliopathy in septic conditions: mechanistic insight into intravascular coagulation
verfasst von
Takashi Ito
Midori Kakuuchi
Ikuro Maruyama
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Schlagwort
COVID-19
Erschienen in
Critical Care / Ausgabe 1/2021
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-021-03524-6

Weitere Artikel der Ausgabe 1/2021

Critical Care 1/2021 Zur Ausgabe

Häusliche Gewalt in der orthopädischen Notaufnahme oft nicht erkannt

28.05.2024 Traumatologische Notfälle Nachrichten

In der Notaufnahme wird die Chance, Opfer von häuslicher Gewalt zu identifizieren, von Orthopäden und Orthopädinnen offenbar zu wenig genutzt. Darauf deuten die Ergebnisse einer Fragebogenstudie an der Sahlgrenska-Universität in Schweden hin.

Fehlerkultur in der Medizin – Offenheit zählt!

Darüber reden und aus Fehlern lernen, sollte das Motto in der Medizin lauten. Und zwar nicht nur im Sinne der Patientensicherheit. Eine negative Fehlerkultur kann auch die Behandelnden ernsthaft krank machen, warnt Prof. Dr. Reinhard Strametz. Ein Plädoyer und ein Leitfaden für den offenen Umgang mit kritischen Ereignissen in Medizin und Pflege.

Mehr Frauen im OP – weniger postoperative Komplikationen

21.05.2024 Allgemeine Chirurgie Nachrichten

Ein Frauenanteil von mindestens einem Drittel im ärztlichen Op.-Team war in einer großen retrospektiven Studie aus Kanada mit einer signifikanten Reduktion der postoperativen Morbidität assoziiert.

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.