Skip to main content

Mechanisms of endothelial activation, hypercoagulation and thrombosis in COVID-19: a link with diabetes mellitus

Abstract

Early since the onset of the COVID-19 pandemic, the medical and scientific community were aware of extra respiratory actions of SARS-CoV-2 infection. Endothelitis, hypercoagulation, and hypofibrinolysis were identified in COVID-19 patients as subsequent responses of endothelial dysfunction. Activation of the endothelial barrier may increase the severity of the disease and contribute to long-COVID syndrome and post-COVID sequelae. Besides, it may cause alterations in primary, secondary, and tertiary hemostasis. Importantly, these responses have been highly decisive in the evolution of infected patients also diagnosed with diabetes mellitus (DM), who showed previous endothelial dysfunction. In this review, we provide an overview of the potential triggers of endothelial activation related to COVID-19 and COVID-19 under diabetic milieu. Several mechanisms are induced by both the viral particle itself and by the subsequent immune-defensive response (i.e., NF-κB/NLRP3 inflammasome pathway, vasoactive peptides, cytokine storm, NETosis, activation of the complement system). Alterations in coagulation mediators such as factor VIII, fibrin, tissue factor, the von Willebrand factor: ADAMST-13 ratio, and the kallikrein-kinin or plasminogen-plasmin systems have been reported. Moreover, an imbalance of thrombotic and thrombolytic (tPA, PAI-I, fibrinogen) factors favors hypercoagulation and hypofibrinolysis. In the context of DM, these mechanisms can be exacerbated leading to higher loss of hemostasis. However, a series of therapeutic strategies targeting the activated endothelium such as specific antibodies or inhibitors against thrombin, key cytokines, factor X, complement system, the kallikrein-kinin system or NETosis, might represent new opportunities to address this hypercoagulable state present in COVID-19 and DM. Antidiabetics may also ameliorate endothelial dysfunction, inflammation, and platelet aggregation. By improving the microvascular pathology in COVID-19 and post-COVID subjects, the associated comorbidities and the risk of mortality could be reduced.

COVID-19 and the disruption of hemostasis

Hemostasis is a finely tuned physiological process that leads to the cessation of bleeding from a blood vessel. It begins with trauma to the lining of the vessel wall and involves multiple interlinked steps that allow formation of a fibrin clot, which finally dissolves after injury is repaired. However, alterations in these processes may lead to hemostasis diseases [1, 2]. In this sense, COVID-19 infected patients have exhibited higher risk of arterial or venous thrombosis associated with disease severity [3] (Fig. 1). Higher plasma levels of hemostatic markers were linked to a worse prognosis and higher mortality [4]. Also, 25–85% of COVID-19 patients admitted to the intensive care unit (ICU) experienced a thrombotic complication, and postmortem analysis revealed the presence of endothelitis [5,6,7]. Importantly, both hypercoagulation and thrombotic events have been observed not only in acute COVID-19. They have been experienced in some few patients suffering from long-COVID syndrome [8], what is currently acknowledged as a challenging component of post-COVID-19 sequelae [9, 10]. Moreover, thrombocytopenia or thrombocytopenic purpura have been rare but serious adverse effects of SARS-CoV-2 spike (S) protein mRNA-based vaccines [11, 12] (see later).

Fig. 1
figure 1

COVID-19 and hemostasis regulation. SARS-CoV-2 may infect endothelial cells (EC) causing endothelitis and directly disrupting endothelial homeostasis, leading to cytokine release, and favoring a pro-coagulant micro-environment. Then, primary hemostasis can be induced by fast vasoconstriction and release of pro-inflammatory and pro-contractile endothelial factors. Activation of coagulation cascades weaves thrombin and fibrin networks that immobilize erythrocytes and activated platelets to from a blood clot in the secondary hemostasis. The resolution of coagulation (tertiary hemostasis) may be also damaged in COVID-19 by alterations in the plasminogen-plasmin and thrombin/thrombomodulin-EPCRP-aPC pathways. Hyperinflammation, hypercoagulation, and hypofibrinolysis could be responsible for thrombotic events in COVID-19 subjects. TXA2 (thromboxane A2), aPC (activated protein C), PC (protein C), EPCR (endothelial protein C receptor)

Interestingly, among different regulators, the endothelium and its highly specialized EC placed at the inner layer of blood vessels tightly regulate vascular reactivity, cell growth, and inflammation by releasing and modulating specific factors [13]. Endothelial activation and dysfunction can be crucial for COVID-19 patients and its major related comorbidities like Diabetes Mellitus (DM) [14,15,16]. This narrative review aims to discuss the main triggers of endothelial dysfunction in COVID-19 and how subsequent disruption in endothelium-dependent hemostasis may promote hypercoagulation and thrombosis. This endothelial dysfunction can be a common event linking the hemostatic alterations observed in COVID-19 with those present in DM. Novel opportunities for therapeutical interventions will be also explored.

Endothelial cell dysfunction and hypercoagulation in COVID-19

Besides pulmonary complications, endothelial injury was established as a primary finding in patients infected by SARS-CoV-2. This virus leads to a complex and multifactorial EC activation, progressive loss of anti-thrombotic factors, and promotion of local pro-angiogenesis (Fig. 1). Postmortem histology revealed viral inclusions in apoptotic EC, infiltration of inflammatory immune cells around the vessels and endothelial layer, and microvascular lymphocytic endothelitis [17, 18]. Clinical observations have identified the vasculature as one of the main trans-organ systems affected by SARS-CoV-2 infection as well as a major trigger of sequelae following COVID-19 [19]. In fact, viral tropism for vascular lesions has been identified in the most severe cases of COVID-19 [18]. While the endothelium basally favors an anti-thrombotic environment by preventing platelet activation and the onset of the coagulation cascade, both physiological and pathological stimuli may shift this balance towards pro-thrombotic and hypercoagulative states [20]. Indeed, presence of SARS-CoV-2 particles undergoes EC activation. A large body of clinical and experimental evidence currently supports the crucial role for EC activation in the pathological changes induced by SARS-CoV-2 in different territories, particularly, in terms of inflammation and thrombotic alterations [21,22,23]. As the intensity and/or the duration of the activation increases, the endothelial dysfunction, as an early sign of vascular disease, takes place.

Endothelial dysfunction runs with vasoconstriction, hyperpermeability, loss of integrity of the endothelial layer, and over-production of chemokines and cytokines together with upregulation of adhesion molecules for leukocytes [20, 24]. Activation of the endothelial monolayer also implies a phenotypic change from an anti-thrombotic to a pro-thrombotic surface more prone to platelet adhesion, together with deregulated synthesis and release of hemostatic factors and the onset of fibrin clots [20, 24]. Enhanced levels of adhesion molecules as well as platelet hyperactivation were observed in COVID-19 in correlation with severity of disease [25]. Thus, COVID-19-related vascular complications, including lung injury, stroke, myocardial dysfunction, or deep vein thrombosis, among others, share the common basis of endothelial dysfunction [20].

Endothelial cell activators in COVID-19

The pathophysiological activation of EC in the context of COVID-19 is triggered by a variety of stimuli, including pro-inflammatory cytokines, vasoactive compounds, components of the immune system, or even by direct actions of SARS-CoV-2 and its isolated viral components (Fig. 2).

Fig. 2
figure 2

The EC activation and dysfunction, as a central pathophysiological mechanism of COVID-19 coagulopathy. SARS-CoV-2 infection and its concomitant local and systemic immunogenic stimuli (cytokine storm, vasoactive compounds, NETosis, and activated complement system) disrupt endothelial homeostasis leading to EC activation. This activation comprises over-inflammation, loss of endothelial barrier integrity and altered hemostasis, favoring coagulation and thrombosis. In red, specific drugs against mediators of endothelial activation. ARDS (acute respiratory distress syndrome), IL (interleukin), TNF-α (tumor necrosis factor alpha), Ang-(1-7) (angiotensin-(1-7)), NO (nitric oxide), Ang II (angiotensin II), TXA2 (thromboxane A2), MAC (membrane attack complex), TLR-4 (toll-like receptor 4)

Pro-inflammatory cytokines and vasoactive compounds

Endothelial activation becomes particularly relevant in acute COVID-19, where the acute respiratory distress syndrome (ARDS) and other complications are triggered by a cytokine storm, i.e., a burst of pro-inflammatory cytokines such as interleukin (IL)-6, IL-1α, IL-1β, IL-17, and tumor necrosis factor-alpha (TNF-α), among others [9, 26]. A large amount of clinical evidence shows a strong association between the inflammatory cell infiltration with increased thrombo-inflammatory biomarkers and COVID-19 severity [27, 28]. Enhanced levels of these cytokines, C-reactive protein, and ferritin were also associated with hyper-coagulation [29]. Moreover, the failure to resolve this inflammatory response could generate a cycle of unregulated events that contribute to endothelial activation and coagulopathy in acute and perhaps long-COVID [30]. In this regard, pro-inflammatory molecules such as platelet factor 4, α-2 antiplasmin, and the von Willebrand factor (WVF) were found increased in long-COVID, which may contribute to the failed fibrinolysis response and explain why these individuals suffer from constant fatigue, cognitive impairment, depression/anxiety, or dyspnea [31].

In addition, enhanced levels of vasoactive compounds, such as angiotensin II (Ang II) or thromboxane A2 (TXA2), which are potent vasoconstrictors and effectors of endothelial dysfunction with pro-inflammatory, pro-adhesive and pro-coagulant properties can play a role in the endotheliopathy associated to COVID-19 [32, 33] (Fig. 2). On the contrary, reduced levels or bioavailability of their physiological counterparts, such as angiotensin-(1–7), which is formed by a main receptor for SARS-CoV-2 (i.e., angiotensin converting enzyme 2; ACE2), prostacyclin, and nitric oxide (NO), can also contribute to the endothelial dysfunction and the hypercoagulation state [32, 34, 35].

NETosis

An abnormal interrelation with immune components can also promote endothelial dysfunction. Neutrophils, as the largest population of myeloid leukocytes, are abundantly recruited in COVID-19 [36]. These phagocytes can act as endothelial activators through the release of extracellular neutrophil traps (NETs). NETs are generated by oxidative stress after stimulation of NADPH oxidase by NLRP3 inflammasome and pro-inflammatory cytokines (i.e., IL-1β and IL-18). Once released, NET components that include chromatin associated with bactericidal proteins from granules and cytoplasm, further intensify the pro-inflammatory response [37,38,39,40,41]. Although NETs mainly exhibit an antibacterial function, in excess, they can cause cell inflammation and tissue damage, increasing thrombogenicity of the endothelial layer [42, 43]. Indeed, NETs play an active role in the pathogenesis of coagulation and thrombosis of various origins by eliciting both extrinsic and intrinsic coagulation pathways [44]. In COVID-19, stimulated NET formation was associated with ARDS and hypercoagulability, as predictors of disease severity [42, 45] (Fig. 2).

The complement system

The complement system, as an integral part of the innate immune response, also participates in the activation of the endothelium and contributes to the formation of a positive feedback loop between inflammation and thrombosis. The main activity of this system is to build up a multiprotein membrane attack complex (MAC) that ends with the death of pathogens by osmotic lysis or macrophage-mediated phagocytosis [46] (Fig. 2). In particular, high amounts of circulating C5a and soluble MAC (C5b-9), as well as processed fragments of C3, were seen in patients with severe COVID-19 [47,48,49]. The SARS-CoV-2 envelope proteins stimulate the lectin pathway of complement activation leading to C3 activation [50]. This activated C3a modulates the expression of endothelial adhesion molecules contributing to immune cell infiltration [51], and C5a directly stimulates secretion of IL-6, IL-8 and vascular endothelial growth factor (VEGF) [52]. Interestingly, a correlation between the activation of the complement and that of the endothelium has been also observed in chronic heart failure, suggesting the interrelated implication of both systems in vascular disease. In fact, complement and EC activation draw a bidirectional loop, since activated EC might also secrete complement components, mostly C7, and contribute to its plasmatic pool [53].

SARS-CoV-2 and isolated viral components

Not only the pathophysiological responses triggered by SARS-CoV-2 have been identified as potential direct endothelial activators capable of triggering hemostatic abnormalities [54, 55]. Viral components like the S protein, formed by two domains named S1 and S2, are able to exert direct effects on EC by binding to cell surface receptors [56] (Fig. 2). Previous reports have described particles of SARS-CoV-2 inside EC [55, 57], and S1 presence was linked to endothelial dysfunction [56, 58]. We have recently observed a direct activation of endothelial pro-inflammatory pathways, including NF-κB and the NLRP3 inflammasome, and a disbalanced production of endothelial hemostatic regulators by the S protein [59]. Moreover, a persistent endothelial injury and inflammation were proposed as potential mechanisms of long-COVID and post-COVID sequelae related to cardiovascular events and dysregulated coagulation [60]. Indeed, in the arterial wall of severe COVID-19 patients, mRNA from SARS-CoV-2 was detected within endothelium, vascular smooth muscle, and infiltrated macrophages, representing a potential viral reservoir [18].

Disbalanced endothelial-derived hemostatic regulators in COVID-19

After COVID-19, dysfunctional EC can synthesize and release several factors that are crucial regulators of coagulation and thrombosis [61]. The role of such regulators and how their delicate balance is disrupted in the context of COVID-19 is briefly reviewed over the next sections.

The tissue factor

The contact and intrinsic pathways may contribute to the pro-thrombotic state of COVID-19 [62]. The tissue factor (TF) is a transmembrane receptor that initiates the extrinsic coagulation cascade [63]. EC together with platelets, T lymphocytes, polymorphonuclear cells, monocytes, macrophages, dendritic cells, and fibroblasts are the main sources of TF [64,65,66,67,68]. Under physiological conditions TF is not typically expressed in active form by EC [69]. However, vessel injury and tissue trauma are major physiological activators of TF by promoting a change from an encrypted inactive to a decrypted active conformation. Then, TF acts as a high affinity receptor and cofactor for factor VII (FVII) and factor VIIa (FVIIa) at the site of tissue damage [70], and the TF:FVIIa complex activates factor X (FX) and factor IX (FIX), producing thrombin and fibrin, activation of platelet, and thrombosis [63]. To prevent excessive coagulation, EC also express tissue factor pathway inhibitor (TFPI) [71]. However, increased TF expression and fibrin enriched thrombi were reported in lung tissues from COVID-19 autopsies [72, 73] (Fig. 3). In human lung epithelial cells and EC, SARS-CoV-2 infection also enhanced TF expression and triggered pro-coagulant and pro-inflammatory responses [74, 75]. These actions were linked to the activation of the complement system, which could amplify and perpetuate endothelial dysfunction [41]. Also, the S protein inhibited TFPI and induced thrombogenic factors in human EC and neutrophils [55]. As consequence, higher circulating TF levels were associated with COVID-19 severity and associated mortality [76].

Fig. 3
figure 3

Hypercoagulability and hypofibrinolysis in COVID-19 and DM. Disrupted hemostasis during COVID-19 may be further intensified under diabetic milieu, resulting in a hypercoagulant phenotype of the activated EC. Elevated levels of pro-coagulant factors (VWF, FVIII, TF/TFPI, thrombin and fibrin) and diminished or insufficient anti-coagulant mediators (ADAMST-13, tPA-plasminogen, KKS, aPC-EPCR) may alter the thrombosis and thrombolysis equilibrium towards formation of blood clots. Arrows indicate over- or down-regulation of factors in COVID-19 (blue) or in DM (green) pathology. Drugs against specific mediators are shown in red beside its target of action. VWF (von Willebrand factor), FVIII (factor VIII), TF (tissue factor), HMWK (high molecular weight kininogen), PK (plasma kallikrein), LMWH (low molecular weight heparin), FXa (activated factor X), aPC (activated protein C), EPCR (endothelial cell protein C receptor), tPA (tissue plasminogen activator), PAI-1 (tissue plasminogen activator), TAFI (thrombin activatable fibrinolysis inhibitor)

Factor VIII

By non-covalent interaction in the Weibel-Palade (WP) bodies, factor VIII (FVIII) is protected from degradation by VWF [77, 78]. Once FVIII is activated to FVIIIa, it is detached from VWF and proteolized [79]. FVIIIa acts as a cofactor in the tenase complex (FIXa/FVIIIa), favoring conversion of FX to FXa [80]. Remarkedly, FVIII has been found disrupted in COVID-19 patients [81] (Fig. 3). Its plasma levels and activity were greatly elevated in parallel to disease severity [3, 81, 82]. In fact, FVIII was reported as an independent predictor of COVID-19 associated mortality [83].

Interstingly, the elevation of FVIII has been also reported in long-COVID in association with thrombo-inflammatory manifestations and vascular dysfunction [84, 85]. Also, very few cases of acquired hemophilia A, a bleeding condition caused by the development of auto-antibodies against FVIII, has been observed after SARS-CoV-2 S mRNA-based vaccination, perhaps as the result of a cross-reaction between antibodies against S protein and endogenous FVIII [86]. More commonly, antiphospholipid antibodies (aPL) derived from B1-cells induced up-regulation of C-reactive protein and D-dimer, and were related to fatal outcomes in COVID-19 patients. This lipid-binding aPL isolated from COVID-19 patients could target monocytes and EC to induce pro-thrombotic and inflammatory responses [87].

The kallikrein-kinin system

In COVID-19, the intrinsic pathway of coagulation not only induces fibrin generation but also links to inflammation by release of kallikrein and bradykinin [88]. The kallikrein-kinin system (KKS) is a family of proteins that can effectively counteract blood pressure and eliminate pathogens by recruiting neutrophils to the site of injury. Kallikrein is mainly synthesized by the liver and endothelium (requiring factor XIIa; FXIIa) and released as plasma kallikrein (PK), which can be activated on the EC surface [89, 90]. Once active, kallikreins cleave kininogens (i.e., high molecular weight kininogen; HMWK) to ultimately synthesize bradykinin (BK) to mediates the release of NO and pro-inflammatory cytokines [91]. However, in COVID-19, the higher concentrations of some components of the KKS correlated with the severity of the disease [92] (Fig. 3). Thus, the hyperactivity of the KKS has become a prognostic marker of poorer outcomes in critically ill patients [93].

The VWF: ADAMST-13 ratio

The VWF is a large multimeric glycoprotein synthesized and stored in EC and megakaryocytes/platelets [94]. After activation of EC or platelet, plasma VWF facilitate platelet aggregation and adhesion to the sites of vascular injury reinforcing the pro-coagulation effect of primary and secondary hemostasis [95]. The VWF form pro-coagulant multimers of different length interacting with angiopoietin-2 (Angpt-2) and osteoprotegerin at the WP bodies [96]. Remarkably, the VWF has been observed elevated in COVID-19 patients, acting as a marker of acute and sustained EC activation and predictor of poor outcomes [97, 98] (Fig. 3). Moreover, the VWF could be involved in local angiogenesis in severe COVID-19. Incubation of plasma from acute COVID-19 patients with EC triggered VWF secretion and Angpt-2 expression, as well as EC tube formation and angiogenesis [99].

In addition, one of the main regulators of the VWF activity is the disintegrin and metalloproteinase ADAMST-13, which is generated by hepatic stellate cells, but also from EC and megakaryocytes/platelets. ADAMST-13 cleaves VWF multimers reducing their pro-adhesive and pro-coagulant activity [100]. However, a higher VWF:ADAMST-13 ratio together with endothelial injury, coagulopathy, and poor prognosis has been found in acute and long-COVID syndrome [84, 101,102,103] (Fig. 3). In this line, COVID-19 vaccination could have originated pro-thrombotic complications in individuals with extremely low ADAMST-13 [104]. A deficiency in ADAMST-13 might be unable to counteract the VWF over-activity found after SARS-CoV-2 infection, long-COVID syndrome and, very rarely, COVID-19 vaccination [105].

Fibrinolytic regulators

The fibrinolytic activity is essential to dissolve the fibrin clot in tertiary hemostasis and it is mainly determined by the balance between the endothelial tissue plasminogen activator (tPA) and its inhibitor, the plasminogen activator inhibitor type 1 (PAI-1) [106]. By producing tPA, the liver-derived plasminogen (PLG) converts into plasmin that eventually breaks up fibrin (releasing D-dimer) to dissolve blood clots. However, under SARS-CoV-2 infection endothelial dysfunction may also imbalance fibrinolysis [107] (Fig. 3). A time-dependent variation of the plasmin-dependent fibrinolytic system has been observed in COVID-19 progression, with initial activation followed by suppression in patients with more severe cases [108]. The plasma concentration of PAI-1 and the tPA-PAI-1 complex were elevated in COVID-19 subjects in comparison to healthy controls [109, 110], and the tPA levels at hospital admission were associated with lower survival rates [111]. Also, increased levels of the endothelial tPA receptor, annexin A2, correlated with inflammatory markers (i.e., IL-1β, IL-6, and TNF-α) and COVID-19 magnitude [112].

Furthermore, evidence suggests the existence of a circulating anti-coagulant factor, activated protein C (aPC) , which is triggered by the thrombin-thrombomodulin complex when bound to the EC protein C receptor (EPCR). aPC can suppress thrombin formation by proteolytical degradation or inactivation of coagulation factor Va (FVa) and FVIIIa and increases fibrinolytic activity by neutralizing PAI-1. However, the thrombin-thrombomodulin-EPCR complex is dysfunctional under COVID-19, affecting the aPC synthesis [113, 114]. Also, an enhanced thrombin generation, decreased fibrinolytic activity, and elevated levels of PAI-1 were identified in patients with long-COVID [115]. Likely, a complex disruption of the balance between thrombogenesis and thrombolysis is not properly solved in some cases or is excessively perpetuated in other patients. In this sense, micro-clots composed by fibrin amyloid and hyperactivated platelets might block capillaries and inhibit O2 transport to tissues, leading to some of the symptoms of the long-COVID and related post-acute sequelae [116].

In addition, elevated fibrinogen and D-dimer levels were correlated with hypercoagulable states, inflammation, and unfavorable outcomes after COVID-19 [117]. The increased D-dimer paradoxically coincided with decreased fibrinolytic capacity [117, 118]. Possibly, the presence of elevated D-dimer, especially during the early stages of pulmonary disease, could indicate the efforts of the local fibrinolytic system to eliminate fibrin and the necrotic tissue from the affected pulmonary parenchyma [118]. Thus, hyperfibrinolysis may be adequate at tissue level but systemically insufficient, delaying the resolution of fibrin deposition. In this context, the regulatory role of PAI-1 may further contribute to explain this effect since PAI-1, tPA, and the thrombin activatable fibrinolysis inhibitor (TAFI) augmented in parallel with COVID-19 severity [110, 119]. Further investigation on the interaction of pro-coagulant and anti-fibrinolytic factors may reveal novel issues in COVID-19 pathogenesis [120].

Diabetes mellitus, a comorbidity in COVID-19

DM is a non-infectious epidemic disease whose prevalence in adults has risen from 108 million in 1980 to 537 million in 2021, and may reach 783 million by 2045 [121]. In fact, DM was responsible for 6.7 million deaths in 2021, a similar number to the fatalities associated to COVID-19 so far [122]. In this sense, the presence of DM has been recognized as a significant risk factor for the rapid progression and poor prognosis of COVID-19, ranging from 5.7% in non-severe cases up to 58% in critical individuals [123, 124]. More unfavorable outcomes have been observed in COVID-19-infected patients with DM than in those without DM. COVID-19 with DM leads to a twofold risk of ICU hospitalization and a two–threefold risk of mortality compared to COVID-19 alone [125, 126]. In a recent meta-analysis, which included 2,987,938 subjects with COVID-19, 10.4% of non-hospitalized individuals were diagnosed with DM, while DM was present in 28.9% of subjects requiring hospitalization and experiencing severe infection. In the same study, 34.6% of deceased patients exhibited both pathologies [127], and in another meta-analysis, the risk of mortality after infection was 54% higher in DM patients compared to non-DM patients [128]. Finally, a relative increased risk of 7–342% for the development of post-acute sequelae of COVID-19 has been reported in DM subjects. Thus, a potential correlation between DM and the long-COVID syndrome may affect vulnerable patients [129].

Hyperglycemia and COVID-19

In the diabetic milieu, hyperglycemia can elicit changes in the immune system, favoring production and release of pro-inflammatory cytokines. After SARS-CoV-2 infection, serum levels of IL-6, C-reactive protein, ferritin, and D-dimer were significantly higher in DM patients compared to non-DM, enforcing the cytokine storm and contributing to the rapid deterioration of patients [130, 131]. In fact, hyperglycemic individuals treated with insulin showed lower risk to develop severe COVID-19. A poor glycemic control promoted higher levels of inflammatory markers during COVID-19 [130], and an adequate glycemic range (3.9–10 mmol/L) was associated with a reduction of adverse COVID-19 outcomes, including death [132]. In this regard, anti-hyperglycemic drugs decreased by 41% the rate of incidence for long-COVID syndrome [133]. Also, the level of SARS-CoV-2 replication was higher in the presence of serum from patients with DM than that from non-diabetics [134]. Thus, individuals with DM and COVID-19, even without other comorbidities, exhibit an increased risk of severe complications, such as pneumonia, uncontrolled inflammatory responses, hypercoagulability, and elevated mortality associated to dysregulated glucose metabolism [131].

On the other hand, SARS-CoV-2 infection may induce a diabetogenic actions. 17% of severe COVID-19 cases exhibited pancreatic lesions that could affect glycemic metabolism and inflammation [135, 136]. Administration of the S protein to type 2 DM-ACE2 knockout mice intensified cerebrovascular complications and cognitive dysfunction through activation of the renin–angiotensin–aldosterone system (RAAS) and toll-like receptor (TLR) signaling [137]. Individuals recovering from COVID-19 faced an elevated risk and burden of DM, leading to an increased usage of anti-hyperglycemic agents [135, 136]. Among fourteen studies that documented new-onset DM after COVID-19, twelve of them found a significant association between both pathologies, suggesting an increased risk of DM of 11–276% [129]. However, DM does not increase the risk of SARS-CoV-2 infection [138].

Endothelial cell activation, a link between COVID-19 and diabetes

The coexistence of DM and COVID-19 could favor deleterious additive or synergistic effects related to endothelial dysfunction and coagulopathy (Fig. 3). In DM, endothelial activation and dysfunction underlies the associated cardiovascular complications. 80% of the cardiovascular deaths observed in DM are attributed to thrombotic events [139]. These responses are favored by factors also present in COVID-19. Pro-inflammatory cytokines may influence the disbalanced release of vasoactive factors and provoke over-activation of the complement system that amplifies endothelial dysfunction [140,141,142,143]. Moreover, COVID-19 associated NETosis can be also part of the pathogenesis of DM and its complications [144, 145]. In the other way, several stimuli related to the metabolic dysregulation can exert deleterious actions on the COVID-19 affected endothelium. The excess of plasma glucose and free fatty acids reduce NO and activate the NLRP3 inflammasome and NF-κB pathways favoring endothelial permeability [146, 147]. Stimulation of coagulation factors in DM may accentuate the risk of thrombotic events. In this sense, the thrombin-thrombomodulin-EPCR complex has been often observed dysfunctional under both DM and COVID-19, affecting aPC synthesis [113, 114]. Therefore, COVID-19 and DM, by sharing or accumulating mechanisms of endothelial activation and dysfunction, may stimulate more severe vascular-driven complications than these entities alone.

Hemostasis disruption in DM and COVID-19

Hypercoagulability and hypofibrinolysis can be common features of both DM and COVID-19 due to the overactivation of the endothelium and variations of hemostatic factors [110, 148]. However, there is still limited evidence regarding how all hemostatic mediators vary in the combined context of these pathologies (Table 1). Through intricate interplays between the pathophysiological mechanisms inherent to both conditions, concomitancy of DM and COVID-19 could exacerbate inflammatory responses. As a result, subsequent alterations in hemostasis and coagulation worsen the disease progression and outcomes, leading to DM as a major risk factor for hemostasis disease in both acute and long-COVID patients [141, 149]. In fact, patients with both pathologies exhibited higher hypercoagulability and thrombotic complications than those infected patients without DM [150].

Table 1 Pro-coagulation and fibrinolytic factors in DM and COVID-19

Specifically, in primary hemostasis, alterations in platelet activation and aggregation, as well as in platelet interaction with endothelium were exacerbated in DM compared to control patients [151]. Hyperglycemia induced overexpression of vascular cell adhesion molecule-1 (VCAM-1) and P-, E- and L-selectins [152, 153]. Also, increased FVIII and other factors of the coagulation cascade, as well as the KKS activity were reported in patients with DM [154,155,156,157] (Table 1). Other authors demonstrated an elevation of TF in both type 1 and type 2 DM [158], or even after glucose variability [159]. Plasma VWF levels were also augmented in diabetics as compared to healthy controls, while ADAMST-13 was diminished [160, 161]. On tertiary hemostasis, up-regulated D-dimer and reduction of plasmin activity were described in DM [162,163,164]. Also, these patients displayed higher FXIII-induced crosslinking of plasmin inhibitor into the fibrin networks, and up-regulation of PAI-1 and TAFI [165,166,167,168].

Importantly, when both DM and COVID-19 overlapped, pro-coagulant factors and deficient fibrinolytic mechanisms were exacerbated. The combination of pre-existing hypofibrinolysis in DM with alterations in severe COVID-19 resulted in significant reduction in the body’s capability to dissolve clots. In these patients, higher levels of C-reactive protein and D-dimer were associated with lethality, and the cutoff value for D-dimer as a predictor of mortality was 2.8 ug/mL [169, 170]. Nevertheless, potential synergisms or additional mechanism of action for pro-coagulant and hypofibrinolytic profiles may be activated in diabetic COVID-19 subjects.

Novel opportunities for therapeutic interventions

For COVID-19, the non-replicating adenoviral vectors were promising carriers for viral antigenic material (i.e., S protein) to induce safe and effective immunity against the virus. However, in 2021 several countries suspended vaccinations due to occurrence of vaccine-induced immune thrombotic thrombocytopenia events [199]. Some vaccines favoured hypercoagulopathy in specific patients with anatomical variants of cerebral venous outflow by inducing a transient inflammatory response and endothelial activation [200]. Although this thromboembolic complication was very rare, more preventive and therapeutic approaches could be suggested, particularly in patients with pro-coagulant comorbidities such as DM [201].

Therapeutic approaches for COVID-19 may depend on disease evolution and symptoms, as well as presence of complications and comorbidities. Asymptomatic infection includes patients with positive virologic test for SARS-CoV-2 but who have no symptoms consistent with COVID-19 [202]. Mild illness comprises individuals with several symptoms such as fever, cough, loss of taste and smell, muscle pain, and diarrhea, but not dyspnea. Herein, only patients aged ≥ 50 years old or with underlying comorbidities are at higher risk of disease progression [203]. In this sense, moderate illness, which includes subjects with lower respiratory disease, requires anti-SARS-CoV-2 treatment (antiviral, immunomodulator, anti-coagulant) [202]. In severe illness, patients show oxygen saturation < 94%, a respiratory rate > 30 breaths/min, a ratio of arterial partial pressure of oxygen to fraction of inspired oxygen < 300 mmHg, or lung infiltrates > 50% [203] . These patients can rapidly exhibit clinical deterioration and require additional oxygen therapy [202]. In critical illness, individuals exhibit ARDS, virus-induced distributive (septic) shock, cardiac shock, an exaggerated inflammatory response, thrombotic disease, and exacerbation of underlying comorbidities such as DM. They are admitted at the ICU to receive treatment for COVID-19 and comorbidities. In addition, reinfection with SARS-CoV-2 may occur as initial immune responses to the primary infection wane over time [204]. Data regarding the prevalence, risk factors, timing, and severity of reinfection likely vary depending on the SARS-CoV-2 variants. Nevertheless, no evidence suggests that the treatment should be different [205].

Novel approaches for diabetic COVID-19

By unveiling the mechanisms that underlie the hypercoagulation/hypofibrinolytic responses in acute or long-COVID, novel therapeutic interventions might be suggested particularly favorable in high-risk patients like diabetics.

Antidiabetics

Administration of insulin improved outcomes in patients with COVID-19 by achieving glycemic goals [130], but this hormone might increase mortality and complications in patients with both COVID-19 and DM [206]. However, metformin has exhibited therapeutic attributes beyond glycemic control. It ameliorated endothelial dysfunction by reduction of ROS production, the activation of NLRP3 inflammasome pathway, and downregulation of pro-inflammatory and adhesion molecules [207,208,209,210]. Also, metformin showed anti-thrombotic actions by attenuation of TF and platelet activation [211, 212]. Thus, it emerges as a promising candidate for enhancing survival in diabetic COVID-19 subjects [213]. Indeed, a recent meta-analysis established a correlation between metformin usage and reduction of mortality among diabetic COVID-19 subjects [214]. Metformin can also limit the replication of SARS-CoV-2 and the inflammatory response mediated by its S1 protein [215, 216]. Interestingly, although it did not reduce mortality nor ICU admission rates in non-diabetic patients with COVID-19 [217], outpatient treatment demonstrated an absolute reduction of 4.1% in the incidence of long COVID-19 [133]. Thus, positive effects of metformin may be also linked to modulatory effects on immune and hemostatic responses [218]. Other antidiabetics such as dipeptidyl peptisase-4 (DPP-4) and sodium-glucose cotransporter 2 (SGLT2) inhibitors, glucagon-like peptide-1 (GLP-1) agonists, thiazolidinediones, and sulfonylureas also reduced the risk of mortality in diabetic patients with COVID-19 [219, 220]. However, some contradictory results have been also reported since administration of DPP-4 inhibitors and sulfonylureas was associated with adverse outcomes and mortality in COVID-19 patients [220, 221].

Immune-thrombotics

Multiple-target strategies may effectively dampen the immune-thrombotic response. The currently established anti-coagulant therapies include low-molecular-weight heparin (LMWH), warfarin, thrombin inhibitors (i.e. hirudin, bivalirudin, argatroban, dabigatran), or FXa inhibitors (i.e. rivaroxaban, apixaban, edoxaban) [222, 223]. Apixaban, a direct oral anti-coagulant, not only blocked both the free and clot-bound FXa and the activity of prothrombinase, it also inhibited the activity of SARS-CoV-2 protease M implicated in viral replication [224].

Novel approaches may target the complement system. Monoclonal antibodies like eculizumab, ravulizumab or zilucoplan inhibited the C5 cleavage or block the resulting fragments reducing generation of the MAC [225], and PK  and FIXa [88]. Inhibitors of C1 esterase are currently being assayed [225], and inhibitors of KKS, such as lanadelumab attenuated both the cytokine storm and hypercoagulation by blocking the PK activity. Also, antagonists for the bradykinin B2 receptor (i.e., icatibant) might be use for acute COVID-19 and perhaps for long-term manifestations of the disease [226]. In these cases, a major therapeutical challenge may consist in clearing viral particles from tissues or at least interfering with their potential receptors and signaling pathways. The viral S protein has been found in post-COVID tissues as a direct pro-inflammatory and pro-coagulant trigger of EC by interacting with a number of cell surface receptors including TLR-4 [227], [228]. In this sense, several TLR-4 inhibitors may be tested in this population [229,230,231]. Moreover, administration of aPC attenuated organ dysfunction and host death caused by ischemia–reperfusion in brain, heart, kidney, and lung in COVID-19 and other pathologies [232]. Inhibitors of the neutrophil elastase, which is released to the extracellular medium upon NETs formation, or agonists for the adenosine, which produce cyclic AMP, may mitigate NETosis [233,234,235]. The use of anti-cytokine drugs (i.e., against ILs, type-1 interferon (IFN)-γ) reduced the cytokine storm in COVID-19 [236, 237], and could indirectly attenuate NETosis and the complement system. In this sense, tocilizumab was approved by FDA as a candidate treatment of severe hospitalized COVID-19 patients [238], and canakinumab lowered the use of antidiabetic drugs in patients with COVID-19 and DM inducing prolonged reduction of systemic inflammation [239].

Limitations

Despite numerous efforts to understand the variability in the severity of the disease, the specific relationship between different variants of SARS-CoV-2 and their impact on diabetic patients is still not fully elucidated. More comprehensive and coordinated research is needed, including genomic analysis, clinical studies, and epidemiological investigations, to better understand the connection between SARS-CoV-2 variants and the severity of infection in individuals with DM [240]. In addition, other limitation of this review is the selective use of literature, unavoidable because of the huge number of papers that emerged during and after this pandemic.

Conclusions

Thrombotic complications are leading causes of hospitalization and death among COVID-19 and post-COVID-affected patients. Those DM patients infected with SARS-CoV-2 may exhibit exacerbated alterations in primary, secondary, and tertiary hemostasis by induction of endothelitis and endothelial dysfunction. As consequence of virus infection and the immune-defense response, increased activation of NF-κB/NLRP3 inflammasome pathways, vasoactive peptides, cytokines, NETosis, and the complement system, finally damage endothelial vasculature and stimulate coagulation mediators. This hypercoagulable state is favored by the lack of fibrinolytic factors, affecting blood irrigation in all tissues. However, several anti-coagulant therapies might be beneficial for these patients, however, therapeutic approaches reducing the initial triggers of pathological endothelial activation (i.e., by antidiabetics, immune-thrombotics) may improve vascular function and ameliorate risk of COVID-19 associated comorbidities.

Availability of data and materials

Not applicable.

Abbreviations

ACE2:

Angiotensin converting enzyme 2

Angpt-2:

Angiopoietin-2

Ang II:

Angiotensin II

aPC:

Activated protein C

ARDS:

Acute respiratory distress syndrome

aPL:

Antiphospholipid antibodies

BK:

Bradykinin

DM:

Diabetes mellitus

DPP-4:

Dipeptidyl peptidase-4

EC:

Endothelial cell

EPCR:

Endothelial cell protein C receptor

FVIII:

Factor FVIII

GLP-1:

Glucagon like peptide-1

HMWK:

High molecular weight kininogen

ICU:

Intensive care unit

IL:

Interleukin

IFN:

Type-1 interferon

KKS:

Kallikrein-kinin system

LMWH:

Low molecular weight heparin

MAC:

Membrane attack complex

MCP-1:

Monocyte chemoattractant protein-1

NETs:

Extracellular neutrophil traps

NO:

Nitric oxide

PAI-1:

Plasminogen activator inhibitor type 1

PK:

Plasma kallikrein

PLG:

Liver-derived plasminogen

RAAS:

Renin–angiotensin–aldosterone system

ROS:

Reactive oxygen species

TAFI:

Thrombin-activatable fribrinolysis inhibitor

SGLT2:

Sodium-glucose cotransporter-2

TF:

Tissue factor

TFPI:

Tissue factor pathway inhibitor

TNF-α:

Tumor necrosis factor alpha

tPA:

Tissue plasminogen

TXA2 :

Thromboxane A2

TLR-4:

Toll-like receptor 4 - S (spike protein)

VCAM-1:

Vascular cell adhesion molecule-1

VEGF:

Vascular endothelial growth factor

VWF:

Von Willebrand factor

WP:

Weibel-Palade

References

  1. Borissoff JI, Spronk HMH, ten Cate H. The hemostatic system as a modulator of atherosclerosis. N Engl J Med. 2011;364(18):1746–60. https://doi.org/10.1056/NEJMra1011670.

    Article  CAS  PubMed  Google Scholar 

  2. van der Meijden PEJ, Heemskerk JWM. Platelet biology and functions: new concepts and clinical perspectives. Nat Rev Cardiol. 2019;16(3):166–79. https://doi.org/10.1038/s41569-018-0110-0.

    Article  CAS  PubMed  Google Scholar 

  3. Panigada M, Bottino N, Tagliabue P, Grasselli G, Novembrino C, Chantarangkul V, et al. Hypercoagulability of COVID-19 patients in intensive care unit: a report of thromboelastography findings and other parameters of hemostasis. J Thromb Haemost. 2020;18(7):1738–42. https://doi.org/10.1111/jth.14850.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. von Meijenfeldt FA, Havervall S, Adelmeijer J, Lundström A, Rudberg A, Magnusson M, et al. Prothrombotic changes in patients with COVID-19 are associated with disease severity and mortality. Res Pract Thromb Haemost. 2021;5(1):132–41. https://doi.org/10.1002/rth2.12462.

    Article  CAS  Google Scholar 

  5. Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F, et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in Covid-19. N Engl J Med. 2020;383(2):120–8. https://doi.org/10.1056/NEJMoa2015432.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Cui S, Chen S, Li X, Liu S, Wang F. Prevalence of venous thromboembolism in patients with severe novel coronavirus pneumonia. J Thromb Haemost. 2020;18(6):1421–4. https://doi.org/10.1111/jth.14830.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ren B, Yan F, Deng Z, Zhang S, Xiao L, Wu M, et al. Extremely high incidence of lower extremity deep venous thrombosis in 48 Patients with severe COVID-19 in Wuhan. Circulation. 2020;142(2):181–3. https://doi.org/10.1161/CIRCULATIONAHA.120.047407.

    Article  CAS  PubMed  Google Scholar 

  8. Vassiliou AG, Vrettou CS, Keskinidou C, Dimopoulou I, Kotanidou A, Orfanos SE. Endotheliopathy in Acute COVID-19 and Long COVID. Int J Mol Sci. 2023;24(9):8237. https://doi.org/10.3390/ijms24098237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Jose RJ, Manuel A. COVID-19 cytokine storm: the interplay between inflammation and coagulation. Lancet Respir Med. 2020;8(6):e46–7. https://doi.org/10.1016/S2213-2600(20)30216-2.

    Article  CAS  PubMed  Google Scholar 

  10. Kole C, Stefanou Ε, Karvelas N, Schizas D, Toutouzas KP. Acute and post-acute COVID-19 cardiovascular complications: a comprehensive review. Cardiovasc Drugs Ther. 2023. https://doi.org/10.1007/s10557-023-07465-w.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Saluja P, Gautam N, Yadala S, Venkata AN. Thrombotic thrombocytopenic purpura (TTP) after COVID-19 vaccination: a systematic review of reported cases. Thromb Res. 2022;214:115–21. https://doi.org/10.1016/j.thromres.2022.04.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yasmin F, Najeeb H, Naeem U, Moeed A, Atif AR, Asghar MS, et al. Adverse events following COVID-19 mRNA vaccines: a systematic review of cardiovascular complication, thrombosis, and thrombocytopenia. Immun Inflamm Dis. 2023;11(3):e807. https://doi.org/10.1002/iid3.807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation. Nat Rev Immunol. 2007;7(10):803–15. https://doi.org/10.1038/nri2171.

    Article  CAS  PubMed  Google Scholar 

  14. Nassar M, Daoud A, Nso N, Medina L, Ghernautan V, Bhangoo H, et al. Diabetes mellitus and COVID-19: review article. Diabetes Metab Syndr. 2021;15(6):102268. https://doi.org/10.1016/j.dsx.2021.102268.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Goshua G, Pine AB, Meizlish ML, Chang C, Zhang H, Bahel P, et al. Endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study. Lancet Haematol. 2020;7(8):e575–82. https://doi.org/10.1016/S2352-3026(20)30216-7.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zhang N, Wang C, Zhu F, Mao H, Bai P, Chen L, et al. Risk factors for poor outcomes of diabetes patients with COVID-19: a single-center, retrospective study in early outbreak in China. Front Endocrinol. 2020;11:571037. https://doi.org/10.3389/fendo.2020.571037.

    Article  Google Scholar 

  17. Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R, Zinkernagel AS, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020;395(10234):1417–8. https://doi.org/10.1016/S0140-6736(20)30937-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Eberhardt N, Noval MG, Kaur R, Amadori L, Gildea M, Sajja S, et al. SARS-CoV-2 infection triggers pro-atherogenic inflammatory responses in human coronary vessels. Nat Cardiovasc Res. 2023;2(10):899–916. https://doi.org/10.1038/s44161-023-00336-5.

  19. Martínez-Salazar B, Holwerda M, Stüdle C, Piragyte I, Mercader N, Engelhardt B, et al. COVID-19 and the vasculature: current aspects and long-term consequences. Front Cell Dev Biol. 2022;10:824851. https://doi.org/10.3389/fcell.2022.824851.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Xu S, Ilyas I, Little PJ, Li H, Kamato D, Zheng X, et al. Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: from mechanism to pharmacotherapies. Pharmacol Rev. 2021;73(3):924–67. https://doi.org/10.1124/pharmrev.120.000096.

    Article  CAS  PubMed  Google Scholar 

  21. Birnhuber A, Fließer E, Gorkiewicz G, Zacharias M, Seeliger B, David S, et al. Between inflammation and thrombosis: endothelial cells in COVID-19. Eur Respir J. 2021;58(3):2100377. https://doi.org/10.1183/13993003.00377-2021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. D’Ardes D, Boccatonda A, Cocco G, Fabiani S, Rossi I, Bucci M, et al. Impaired coagulation, liver dysfunction and COVID-19: discovering an intriguing relationship. World J Gastroenterol. 2022;28(11):1102–12. https://doi.org/10.3748/wjg.v28.i11.1102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. D’Agnillo F, Walters K, Xiao Y, Sheng Z, Scherler K, Park J, et al. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci Transl Med. 2021;13(620):7790. https://doi.org/10.1126/scitranslmed.abj7790.

    Article  CAS  Google Scholar 

  24. Hunt BJ, Jurd KM. Endothelial cell activation. BMJ. 1998;316(7141):1328–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Watany MM, Abdou S, Elkolaly R, Elgharbawy N, Hodeib H. Evaluation of admission levels of P, E and L selectins as predictors for thrombosis in hospitalized COVID-19 patients. Clin Exp Med. 2022;22(4):567–75. https://doi.org/10.1007/s10238-021-00787-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Montazersaheb S, Hosseiniyan Khatibi SM, Hejazi MS, Tarhriz V, Farjami A, Ghasemian Sorbeni F, et al. COVID-19 infection: an overview on cytokine storm and related interventions. Virol J. 2022;19(1):92. https://doi.org/10.1186/s12985-022-01814-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hu B, Huang S, Yin L. The cytokine storm and COVID-19. J Med Virol. 2021;93(1):250–6. https://doi.org/10.1002/jmv.26232.

    Article  CAS  PubMed  Google Scholar 

  28. Fajgenbaum DC, June CH. Cytokine Storm. N Engl J Med. 2020;383(23):2255–73. https://doi.org/10.1056/NEJMra2026131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Moore JB, June CH. Cytokine release syndrome in severe COVID-19. Science. 2020;368(6490):473–4. https://doi.org/10.1126/science.abb8925.

    Article  ADS  CAS  PubMed  Google Scholar 

  30. Low RN, Low RJ, Akrami A. A review of cytokine-based pathophysiology of Long COVID symptoms. Front Med. 2023;10:1011936. https://doi.org/10.3389/fmed.2023.1011936.

    Article  Google Scholar 

  31. Kruger A, Vlok M, Turner S, Venter C, Laubscher GJ, Kell DB, et al. Proteomics of fibrin amyloid microclots in long COVID/post-acute sequelae of COVID-19 (PASC) shows many entrapped pro-inflammatory molecules that may also contribute to a failed fibrinolytic system. Cardiovasc Diabetol. 2022;21(1):190. https://doi.org/10.1186/s12933-022-01623-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Henry BM, Vikse J, Benoit S, Favaloro EJ, Lippi G. Hyperinflammation and derangement of renin-angiotensin-aldosterone system in COVID-19: a novel hypothesis for clinically suspected hypercoagulopathy and microvascular immunothrombosis. Clin Chim Acta. 2020;507:167–73. https://doi.org/10.1016/j.cca.2020.04.027.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Conti P, Caraffa A, Gallenga CE, Ross R, Kritas SK, Frydas I, et al. IL-1 induces throboxane-A2 (TxA2) in COVID-19 causing inflammation and micro-thrombi: inhibitory effect of the IL-1 receptor antagonist (IL-1Ra). J Biol Regul Homeost Agents. 2020;34(5):1623–7. https://doi.org/10.23812/20-34-4EDIT-65.

    Article  CAS  PubMed  Google Scholar 

  34. Carpenter RM, Young MK, Petri WAO, Lyons GR, Gilchrist C, Carey RM, et al. Repressed Ang 1–7 in COVID-19 Is inversely associated with inflammation and coagulation. mSphere. 2022;7(4):e0022022. https://doi.org/10.1128/msphere.00220-22.

    Article  CAS  PubMed  Google Scholar 

  35. Peiró C, Moncada S. Substituting angiotensin-(1–7) to prevent lung damage in SARS-CoV-2 infection? Circulation. 2020;141(21):1665–6. https://doi.org/10.1161/CIRCULATIONAHA.120.047297.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Qin G, Liu S, Yang L, Yu W, Zhang Y. Myeloid cells in COVID-19 microenvironment. Signal Transduct Target Ther. 2021;6(1):372. https://doi.org/10.1038/s41392-021-00792-0.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Vorobjeva NV, Chernyak BV. NETosis: molecular mechanisms, role in physiology and pathology. Biochemistry. 2020;85(10):1178–90. https://doi.org/10.1134/S0006297920100065.

    Article  CAS  PubMed  Google Scholar 

  38. Thiam HR, Wong SL, Wagner DD, Waterman CM. Cellular mechanisms of NETosis. Annu Rev Cell Dev Biol. 2020;36:191–218. https://doi.org/10.1146/annurev-cellbio-020520-111016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532–5. https://doi.org/10.1126/science.1092385.

    Article  ADS  CAS  PubMed  Google Scholar 

  40. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD, et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A. 2010;107(36):15880–5. https://doi.org/10.1073/pnas.1005743107.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  41. Folco EJ, Mawson TL, Vromman A, Bernardes-Souza B, Franck G, Persson O, et al. Neutrophil extracellular traps induce endothelial cell activation and tissue factor production through interleukin-1α and cathepsin G. Arterioscler Thromb Vasc Biol. 2018;38(8):1901–12. https://doi.org/10.1161/ATVBAHA.118.311150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Barnes BJ, Adrover JM, Baxter-Stoltzfus A, Borczuk A, Cools-Lartigue J, Crawford JM, et al. Targeting potential drivers of COVID-19: neutrophil extracellular traps. J Exp Med. 2020;217(6):e20200652. https://doi.org/10.1084/jem.20200652.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Yu S, Liu J, Yan N. Endothelial dysfunction induced by extracellular neutrophil traps plays important role in the occurrence and treatment of extracellular neutrophil traps-related disease. Int J Mol Sci. 2022;23(10):5626. https://doi.org/10.3390/ijms23105626.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Moschonas IC, Tselepis AD. The pathway of neutrophil extracellular traps towards atherosclerosis and thrombosis. Atherosclerosis. 2019;288:9–16. https://doi.org/10.1016/j.atherosclerosis.2019.06.919.

    Article  CAS  PubMed  Google Scholar 

  45. Zuo Y, Yalavarthi S, Shi H, Gockman K, Zuo M, Madison JA, et al. Neutrophil extracellular traps in COVID-19. JCI Insight. 2020;5(11):e138999. https://doi.org/10.1172/jci.insight.138999.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Walport MJ. Complement first of two parts. N Engl J Med. 2001;344(14):1058–66. https://doi.org/10.1056/NEJM200104053441406.

    Article  CAS  PubMed  Google Scholar 

  47. Cugno M, Meroni PL, Gualtierotti R, Griffini S, Grovetti E, Torri A, et al. Complement activation in patients with COVID-19: a novel therapeutic target. J Allergy Clin Immunol. 2020;146(1):215–7. https://doi.org/10.1016/j.jaci.2020.05.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Skendros P, Mitsios A, Chrysanthopoulou A, Mastellos DC, Metallidis S, Rafailidis P, et al. Complement and tissue factor-enriched neutrophil extracellular traps are key drivers in COVID-19 immunothrombosis. J Clin Invest. 2020;130(11):6151–7. https://doi.org/10.1172/JCI141374.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ma L, Sahu SK, Cano M, Kuppuswamy V, Bajwa J, McPhatter J, et al. Increased complement activation is a distinctive feature of severe SARS-CoV-2 infection. Sci Immunol. 2021;6(59):abh2259. https://doi.org/10.1126/sciimmunol.abh2259.

    Article  Google Scholar 

  50. Sinkovits G, Mező B, Réti M, Müller V, Iványi Z, Gál J, et al. Complement overactivation and consumption predicts in-hospital mortality in SARS-CoV-2 infection. Front Immunol. 2021;12:663187. https://doi.org/10.3389/fimmu.2021.663187.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Propson NE, Roy ER, Litvinchuk A, Köhl J, Zheng H. Endothelial C3a receptor mediates vascular inflammation and blood-brain barrier permeability during aging. J Clin Invest. 2021;131(1):e140966. https://doi.org/10.1172/JCI140966.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Albrecht EA, Chinnaiyan AM, Varambally S, Kumar-Sinha C, Barrette TR, Sarma JV, et al. C5a-induced gene expression in human umbilical vein endothelial cells. Am J Pathol. 2004;164(3):849–59. https://doi.org/10.1016/S0002-9440(10)63173-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Fischetti F, Tedesco F. Cross-talk between the complement system and endothelial cells in physiologic conditions and in vascular diseases. Autoimmunity. 2006;39(5):417–28. https://doi.org/10.1080/08916930600739712.

    Article  CAS  PubMed  Google Scholar 

  54. Otifi HM, Adiga BK. Endothelial dysfunction in Covid-19 infection. Am J Med Sci. 2022;363(4):281–7. https://doi.org/10.1016/j.amjms.2021.12.010.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Bhargavan B, Kanmogne GD. SARS-CoV-2 spike proteins and cell-cell communication inhibits TFPI and induces thrombogenic factors in human lung microvascular endothelial cells and neutrophils: implications for COVID-19 coagulopathy pathogenesis. Int J Mol Sci. 2022;23(18):10436. https://doi.org/10.3390/ijms231810436.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Perico L, Benigni A, Remuzzi G. SARS-CoV-2 and the spike protein in endotheliopathy. Trends Microbiol. 2023. https://doi.org/10.1016/j.tim.2023.06.004.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Monteil V, Kwon H, Prado P, Hagelkrüys A, Wimmer RA, Stahl M, et al. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell. 2020;181(4):905-913.e7. https://doi.org/10.1016/j.cell.2020.04.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Rotoli BM, Barilli A, Visigalli R, Ferrari F, Dall’Asta V. Endothelial cell activation by SARS-CoV-2 spike S1 protein: a crosstalk between endothelium and innate immune cells. Biomedicines. 2021;9(9):1220. https://doi.org/10.3390/biomedicines9091220.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Villacampa A, Alfaro E, Morales C, Díaz-García E, López-Fernández C, Bartha JL, et al. SARS-CoV-2 protein activates NLRP3 inflammasome and deregulates coagulation factor in endothelial and immune cells. Cell Commun Signal. 2023. https://doi.org/10.1186/s12964-023-01397-6.

    Article  Google Scholar 

  60. Ahamed J, Laurence J. Long COVID endotheliopathy: hypothesized mechanisms and potential therapeutic approaches. J Clin Invest. 2022;132(15):e161167. https://doi.org/10.1172/JCI161167.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Jin Y, Ji W, Yang H, Chen S, Zhang W, Duan G. Endothelial activation and dysfunction in COVID-19: from basic mechanisms to potential therapeutic approaches. Signal Transduct Target Ther. 2020;5(1):293. https://doi.org/10.1038/s41392-020-00454-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Henderson MW, Lima F, Moraes CRP, Ilich A, Huber SC, Barbosa MS, et al. Contact and intrinsic coagulation pathways are activated and associated with adverse clinical outcomes in COVID-19. Blood Adv. 2022;6(11):3367–77. https://doi.org/10.1182/bloodadvances.2021006620.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Grover SP, Mackman N. Tissue factor: an essential mediator of hemostasis and trigger of thrombosis. Arterioscler Thromb Vasc Biol. 2018;38(4):709–25. https://doi.org/10.1161/ATVBAHA.117.309846.

    Article  CAS  PubMed  Google Scholar 

  64. Parry GC, Mackman N. Transcriptional regulation of tissue factor expression in human endothelial cells. Arterioscler Thromb Vasc Biol. 1995;15(5):612–21. https://doi.org/10.1161/01.atv.15.5.612.

    Article  CAS  PubMed  Google Scholar 

  65. De Palma R, Cirillo P, Ciccarelli G, Barra G, Conte S, Pellegrino G, et al. Expression of functional tissue factor in activated T-lymphocytes in vitro and in vivo: a possible contribution of immunity to thrombosis? Int J Cardiol. 2016;218:188–95. https://doi.org/10.1016/j.ijcard.2016.04.177.

    Article  PubMed  Google Scholar 

  66. Mandal SK, Iakhiaev A, Pendurthi UR, Mohan Rao LV. Acute cholesterol depletion impairs functional expression of tissue factor in fibroblasts: modulation of tissue factor activity by membrane cholesterol. Blood. 2005;105(1):153–60. https://doi.org/10.1182/blood-2004-03-0990.

    Article  CAS  PubMed  Google Scholar 

  67. Musgrave KM, Scott J, Sendama W, Gardner AI, Dewar F, Lake CJ, et al. Tissue factor expression in monocyte subsets during human immunothrombosis, endotoxemia and sepsis. Thromb Res. 2023;228:10–20. https://doi.org/10.1016/j.thromres.2023.05.018.

    Article  CAS  PubMed  Google Scholar 

  68. Kothari H, Pendurthi UR, Rao LVM. Analysis of tissue factor expression in various cell model systems: cryptic vs. active. J Thromb Haemost. 2013;11(7):1353–63. https://doi.org/10.1111/jth.12272.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Owens AP, Mackman N. Role of tissue factor in atherothrombosis. Curr Atheroscler Rep. 2012;14(5):394–401. https://doi.org/10.1007/s11883-012-0269-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Mackman N. The role of tissue factor and factor VIIa in hemostasis. Anesth Analg. 2009;108(5):1447–52. https://doi.org/10.1213/ane.0b013e31819bceb1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Girard TJ, Broze GJ. Tissue factor pathway inhibitor. Methods Enzymol. 1993;222:195–209. https://doi.org/10.1016/0076-6879(93)22014-7.

    Article  CAS  PubMed  Google Scholar 

  72. Subrahmanian S, Borczuk A, Salvatore SP, Laurence J, Ahamed J. Higher tissue factor (TF) expression in the lungs of COVID-19 pneumonia patients than patients with acute respiratory distress syndrome: association with thrombi formation. Blood. 2020;136:4. https://doi.org/10.1182/blood-2020-142827.

    Article  Google Scholar 

  73. Subrahmanian S, Borczuk A, Salvatore S, Fung K, Merrill JT, Laurence J, et al. Tissue factor upregulation is associated with SARS-CoV-2 in the lungs of COVID-19 patients. J Thromb Haemost. 2021;19(9):2268–74. https://doi.org/10.1111/jth.15451.

    Article  CAS  PubMed  Google Scholar 

  74. Nascimento Conde J, Schutt WR, Gorbunova EE, Mackow ER. Recombinant ACE2 expression is required for SARS-CoV-2 To infect primary human endothelial cells and induce inflammatory and procoagulative responses. MBio. 2020;11(6):3185. https://doi.org/10.1128/mBio.03185-20.

    Article  Google Scholar 

  75. FitzGerald ES, Chen Y, Fitzgerald KA, Jamieson AM. Lung epithelial cell transcriptional regulation as a factor in COVID-19-associated coagulopathies. Am J Respir Cell Mol Biol. 2021;64(6):687–97. https://doi.org/10.1165/rcmb.2020-0453OC.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Sachetto ATA, Mackman N. Tissue factor and COVID-19: an update. Curr Drug Targets. 2022;23(17):1573–7. https://doi.org/10.2174/1389450123666220926144432.

    Article  CAS  PubMed  Google Scholar 

  77. Rosenberg JB, Foster PA, Kaufman RJ, Vokac EA, Moussalli M, Kroner PA, et al. Intracellular trafficking of factor VIII to von Willebrand factor storage granules. J Clin Invest. 1998;101(3):613–24. https://doi.org/10.1172/JCI1250.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Grover SP, Mackman N. Intrinsic pathway of coagulation and thrombosis. Arterioscler Thromb Vasc Biol. 2019;39(3):331–8. https://doi.org/10.1161/ATVBAHA.118.312130.

    Article  CAS  PubMed  Google Scholar 

  79. Mazurkiewicz-Pisarek A, Płucienniczak G, Ciach T, Płucienniczak A. The factor VIII protein and its function. Acta Biochim Pol. 2016;63(1):11–6. https://doi.org/10.18388/abp.2015_1056.

    Article  CAS  PubMed  Google Scholar 

  80. Nogami K, Shima M, Hosokawa K, Suzuki T, Koide T, Saenko EL, et al. Role of factor VIII C2 domain in factor VIII binding to factor Xa. J Biol Chem. 1999;274(43):31000–7. https://doi.org/10.1074/jbc.274.43.31000.

    Article  CAS  PubMed  Google Scholar 

  81. Martín-Rojas RM, Pérez-Rus G, Delgado-Pinos VE, Domingo-González A, Regalado-Artamendi I, Alba-Urdiales N, et al. COVID-19 coagulopathy: an in-depth analysis of the coagulation system. Eur J Haematol. 2020;105(6):741–50. https://doi.org/10.1111/ejh.13501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Voicu S, Delrue M, Chousterman BG, Stépanian A, Bonnin P, Malissin I, et al. Imbalance between procoagulant factors and natural coagulation inhibitors contributes to hypercoagulability in the critically ill COVID-19 patient: clinical implications. Eur Rev Med Pharmacol Sci. 2020;24(17):9161–8. https://doi.org/10.26355/eurrev_202009_22866.

    Article  CAS  PubMed  Google Scholar 

  83. Abd El-Lateef AE, Alghamdi S, Ebid G, Khalil K, Kabrah S, Abdel Ghafar MT. Coagulation profile in COVID-19 patients and its relation to disease severity and overall survival: a single-center study. Br J Biomed Sci. 2022;79:10098. https://doi.org/10.3389/bjbs.2022.10098.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Prasannan N, Heightman M, Hillman T, Wall E, Bell R, Kessler A, et al. Impaired exercise capacity in post-COVID-19 syndrome: the role of VWF-ADAMTS13 axis. Blood Adv. 2022;6(13):4041–8. https://doi.org/10.1182/bloodadvances.2021006944.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Nicolai L, Kaiser R, Stark K. Thromboinflammation in long COVID—the elusive key to postinfection sequelae? J Thromb Haemost. 2023. https://doi.org/10.1016/j.jtha.2023.04.039.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Duminuco A, Calagna M, Markovic U, Esposito B, Grasso S, Riccobene C, et al. Acquired hemophilia A following COVID-19 vaccination—the importance of prompt diagnosis: a case report. Transfus Apher Sci. 2022. https://doi.org/10.1016/j.transci.2022.103577.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Hollerbach A, Müller-Calleja N, Pedrosa D, Canisius A, Sprinzl MF, Falter T, et al. Pathogenic lipid-binding antiphospholipid antibodies are associated with severity of COVID-19. J Thromb Haemost. 2021;19(9):2335–47. https://doi.org/10.1111/jth.15455.

    Article  CAS  PubMed  Google Scholar 

  88. Busch MH, Timmermans SAMEG, Nagy M, Visser M, Huckriede J, Aendekerk JP, et al. Neutrophils and contact activation of coagulation as potential drivers of COVID-19. Circulation. 2020;142(18):1787–90. https://doi.org/10.1161/CIRCULATIONAHA.120.050656.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Bryant JW, Shariat-Madar Z. Human plasma kallikrein-kinin system: physiological and biochemical parameters. Cardiovasc Hematol Agents Med Chem. 2009;7(3):234–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Didiasova M, Wujak L, Schaefer L, Wygrecka M. Factor XII in coagulation, inflammation and beyond. Cell Signal. 2018;51:257–65. https://doi.org/10.1016/j.cellsig.2018.08.006.

    Article  CAS  PubMed  Google Scholar 

  91. Marcondes S, Antunes E. The plasma and tissue kininogen-kallikrein-kinin system: role in the cardiovascular system. Curr Med Chem Cardiovasc Hematol Agents. 2005;3(1):33–44. https://doi.org/10.2174/1568016052773351.

    Article  CAS  PubMed  Google Scholar 

  92. Alfaro E, Díaz-García E, García-Tovar S, Zamarrón E, Mangas A, Galera R, et al. Impaired kallikrein-kinin system in COVID-19 patients’ severity. Front Immunol. 2022;13:909342. https://doi.org/10.3389/fimmu.2022.909342.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Lipcsey M, Persson B, Eriksson O, Blom AM, Fromell K, Hultström M, et al. The outcome of critically Ill COVID-19 patients is linked to thromboinflammation dominated by the Kallikrein/Kinin system. Front Immunol. 2021;12:627579. https://doi.org/10.3389/fimmu.2021.627579.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Kanaji S, Fahs SA, Shi Q, Haberichter SL, Montgomery RR. Contribution of platelet versus endothelial VWF to platelet adhesion and hemostasis. J Thromb Haemost. 2012;10(8):1646–52. https://doi.org/10.1111/j.1538-7836.2012.04797.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Verhenne S, Denorme F, Libbrecht S, Vandenbulcke A, Pareyn I, Deckmyn H, et al. Platelet-derived VWF is not essential for normal thrombosis and hemostasis but fosters ischemic stroke injury in mice. Blood. 2015;126(14):1715–22. https://doi.org/10.1182/blood-2015-03-632901.

    Article  CAS  PubMed  Google Scholar 

  96. Valentijn KM, Sadler JE, Valentijn JA, Voorberg J, Eikenboom J. Functional architecture of weibel-palade bodies. Blood. 2011;117(19):5033–43. https://doi.org/10.1182/blood-2010-09-267492.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Ward SE, Curley GF, Lavin M, Fogarty H, Karampini E, McEvoy NL, et al. Von Willebrand factor propeptide in severe coronavirus disease 2019 (COVID-19): evidence of acute and sustained endothelial cell activation. Br J Haematol. 2021;192(4):714–9. https://doi.org/10.1111/bjh.17273.

    Article  CAS  PubMed  Google Scholar 

  98. Wibowo A, Pranata R, Lim MA, Akbara MR, Martha JW. Endotheliopathy marked by high von Willebrand factor (vWF) antigen in COVID-19 is associated with poor outcome: a systematic review and meta-analysis. Int J Infect Dis. 2022;117:267–73. https://doi.org/10.1016/j.ijid.2021.06.051.

    Article  CAS  PubMed  Google Scholar 

  99. Karampini E, Fogarty H, Elliott S, Morrin H, Bergin C, O’Sullivan JM, et al. Endothelial cell activation, Weibel-Palade body secretion, and enhanced angiogenesis in severe COVID-19. Res Pract Thromb Haemost. 2023;7(2):100085. https://doi.org/10.1016/j.rpth.2023.100085.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Zheng XL. ADAMTS13 and von willebrand factor in thrombotic thrombocytopenic purpura. Annu Rev Med. 2015;66:211–25. https://doi.org/10.1146/annurev-med-061813-013241.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Mancini I, Baronciani L, Artoni A, Colpani P, Biganzoli M, Cozzi G, et al. The ADAMTS13-von willebrand factor axis in COVID-19 patients. J Thromb Haemost. 2021;19(2):513–21. https://doi.org/10.1111/jth.15191.

    Article  CAS  PubMed  Google Scholar 

  102. Xu X, Feng Y, Jia Y, Zhang X, Li L, Bai X, et al. Prognostic value of von willebrand factor and ADAMTS13 in patients with COVID-19: a systematic review and meta-analysis. Thromb Res. 2022;218:83–98. https://doi.org/10.1016/j.thromres.2022.08.017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Mei ZW, van Wijk Xander MR, Pham HP, Marin MJ. Role of von willebrand factor in COVID-19 associated coagulopathy. J Appl Lab Med. 2021;6(5):1305–15. https://doi.org/10.1093/jalm/jfab042.

    Article  PubMed  Google Scholar 

  104. Shah H, Kim A, Sukumar S, Mazepa M, Kohli R, Braunstein EM, et al. SARS-CoV-2 vaccination and immune thrombotic thrombocytopenic purpura. Blood. 2022;139(16):2570–3. https://doi.org/10.1182/blood.2022015545.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Fogarty H, Ward SE, Townsend L, Karampini E, Elliott S, Conlon N, et al. Sustained VWF-ADAMTS-13 axis imbalance and endotheliopathy in long COVID syndrome is related to immune dysfunction. J Thromb Haemost. 2022;20(10):2429–38. https://doi.org/10.1111/jth.15830.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Medcalf RL, Keragala CB. The fibrinolytic system: mysteries and opportunities. Hemasphere. 2021;5(6):e570. https://doi.org/10.1097/HS9.0000000000000570.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Neubauer K, Zieger B. Endothelial cells and coagulation. Cell Tissue Res. 2022;387(3):391–8. https://doi.org/10.1007/s00441-021-03471-2.

    Article  CAS  PubMed  Google Scholar 

  108. Keragala CB, Medcalf RL, Myles PS. Fibrinolysis and COVID-19: a tale of two sites? J Thromb Haemost. 2020;18(9):2430–2. https://doi.org/10.1111/jth.15017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Zuo Y, Warnock M, Harbaugh A, Yalavarthi S, Gockman K, Zuo M, et al. Plasma tissue plasminogen activator and plasminogen activator inhibitor-1 in hospitalized COVID-19 patients. Sci Rep. 2021;11(1):1580. https://doi.org/10.1038/s41598-020-80010-z.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  110. Nougier C, Benoit R, Simon M, Desmurs-Clavel H, Marcotte G, Argaud L, et al. Hypofibrinolytic state and high thrombin generation may play a major role in SARS-COV2 associated thrombosis. J Thromb Haemost. 2020;18(9):2215–9. https://doi.org/10.1111/jth.15016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Cabrera-Garcia D, Miltiades A, Yim P, Parsons S, Elisman K, Mansouri MT, et al. Plasma biomarkers associated with survival and thrombosis in hospitalized COVID-19 patients. Int J Hematol. 2022;116(6):937–46. https://doi.org/10.1007/s12185-022-03437-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Shenoy RD, Nithin Kuriakose N, Vijaykrishnaraj MN, Patil P, Jayaswamy PK, Alagundagi DB, et al. Tissue plasminogen activator receptor ANXA2 and its complementary regulator anti-inflammatory ANXA1 as prognostic indicators of inflammatory response in COVID-19 pathogenesis. Immunobiology. 2023;228(5):152728. https://doi.org/10.1016/j.imbio.2023.152728.

    Article  CAS  PubMed  Google Scholar 

  113. Santos Silva BR, Poblete Jara C, Sidarta-Oliveira D, Velloso LA, Velander WH, Araújo EP. Downregulation of the protein C signaling system is associated with COVID-19 hypercoagulability-A single-cell transcriptomics analysis. Viruses. 2022;14(12):2753. https://doi.org/10.3390/v14122753.

    Article  CAS  Google Scholar 

  114. Isermann B, Vinnikov IA, Madhusudhan T, Herzog S, Kashif M, Blautzik J, et al. Activated protein C protects against diabetic nephropathy by inhibiting endothelial and podocyte apoptosis. Nat Med. 2007;13(11):1349–58. https://doi.org/10.1038/nm1667.

    Article  CAS  PubMed  Google Scholar 

  115. Prasannan N, Heightman M, Hillman T, Wall E, Bell R, Kessler A, et al. Impaired exercise capacity in post–COVID-19 syndrome: the role of VWF-ADAMTS13 axis. Blood Adv. 2022;6(13):4041–8. https://doi.org/10.1182/bloodadvances.2021006944.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Pretorius E, Venter C, Laubscher GJ, Kotze MJ, Oladejo SO, Watson LR, et al. Prevalence of symptoms, comorbidities, fibrin amyloid microclots and platelet pathology in individuals with Long COVID/Post-Acute Sequelae of COVID-19 (PASC). Cardiovasc Diabetol. 2022;21(1):148. https://doi.org/10.1186/s12933-022-01579-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Calvet L, Thouy F, Mascle O, Sapin A, Grapin K, Liteaudon JM, et al. Hypercoagulability in critically ill patients with COVID 19, an observational prospective study. PLoS ONE. 2022;17(11):e0277544. https://doi.org/10.1371/journal.pone.0277544.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Ibañez C, Perdomo J, Calvo A, Ferrando C, Reverter JC, Tassies D, et al. High D dimers and low global fibrinolysis coexist in COVID19 patients: what is going on in there? J Thromb Thrombolysis. 2021;51(2):308–12. https://doi.org/10.1007/s11239-020-02226-0.

    Article  CAS  PubMed  Google Scholar 

  119. Baycan OF, Barman HA, Bolen F, Atici A, Erman H, Korkmaz R, et al. Plasminogen activator inhibitor-1 levels as an indicator of severity and mortality for COVID-19. North Clin Istanb. 2023;10(1):1–9. https://doi.org/10.14744/nci.2022.09076.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Nielsen ND, Rollins-Raval MA, Raval JS, Thachil J. Is it hyperfibrinolysis or fibrinolytic shutdown in severe COVID-19? Thromb Res. 2022;210:1–3. https://doi.org/10.1016/j.thromres.2021.12.012.

    Article  CAS  PubMed  Google Scholar 

  121. diabetes Lw, Policy P. IDF Diabetes Atlas | Tenth Edition. https://diabetesatlas.org/. Accessed 8 Oct 2023.

  122. WHO Coronavirus (COVID-19) Dashboard. https://covid19.who.int. Accessed 8 Oct 2023.

  123. Guan W, Ni Z, Hu Y, Liang W, Ou C, He J, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382(18):1708–20. https://doi.org/10.1056/NEJMoa2002032.

    Article  CAS  PubMed  Google Scholar 

  124. Bhatraju PK, Ghassemieh BJ, Nichols M, Kim R, Jerome KR, Nalla AK, et al. Covid-19 in critically Ill patients in the seattle region—case series. N Engl J Med. 2020;382(21):2012–22. https://doi.org/10.1056/NEJMoa2004500.

    Article  CAS  PubMed  Google Scholar 

  125. Roncon L, Zuin M, Rigatelli G, Zuliani G. Diabetic patients with COVID-19 infection are at higher risk of ICU admission and poor short-term outcome. J Clin Virol. 2020;127:104354. https://doi.org/10.1016/j.jcv.2020.104354.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Kumar A, Arora A, Sharma P, Anikhindi SA, Bansal N, Singla V, et al. Is diabetes mellitus associated with mortality and severity of COVID-19? A meta-analysis. Diabetes Metab Syndr. 2020;14(4):535–45. https://doi.org/10.1016/j.dsx.2020.04.044.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Li R, Shen M, Yang Q, Fairley CK, Chai Z, McIntyre R, et al. Global diabetes prevalence in COVID-19 patients and contribution to COVID-19- related severity and mortality: a systematic review and meta-analysis. Diabetes Care. 2023;46(4):890–7. https://doi.org/10.2337/dc22-1943.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Mahamat-Saleh Y, Fiolet T, Rebeaud ME, Mulot M, Guihur A, El Fatouhi D, et al. Diabetes, hypertension, body mass index, smoking and COVID-19-related mortality: a systematic review and meta-analysis of observational studies. BMJ Open. 2021;11(10):e052777. https://doi.org/10.1136/bmjopen-2021-052777.

    Article  PubMed  Google Scholar 

  129. Harding JL, Oviedo SA, Ali MK, Ofotokun I, Gander JC, Patel SA, et al. The bidirectional association between diabetes and long-COVID-19—a systematic review. Diabetes Res Clin Pract. 2023;195:110202. https://doi.org/10.1016/j.diabres.2022.110202.

    Article  CAS  PubMed  Google Scholar 

  130. Sardu C, D’Onofrio N, Balestrieri ML, Barbieri M, Rizzo MR, Messina V, et al. Outcomes in patients with hyperglycemia affected by COVID-19: can we do more on glycemic control? Diabetes Care. 2020;43(7):1408–15. https://doi.org/10.2337/dc20-0723.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Guo W, Li M, Dong Y, Zhou H, Zhang Z, Tian C, et al. Diabetes is a risk factor for the progression and prognosis of COVID-19. Diabetes Metab Res Rev. 2020;36(7):e3319. https://doi.org/10.1002/dmrr.3319.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Zhu L, She Z, Cheng X, Qin J, Zhang X, Cai J, et al. Association of blood glucose control and outcomes in patients with COVID-19 and pre-existing type 2 diabetes. Cell Metab. 2020;31(6):1068-1077.e3. https://doi.org/10.1016/j.cmet.2020.04.021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Bramante CT, Buse JB, Liebovitz DM, Nicklas JM, Puskarich MA, Cohen K, et al. Outpatient treatment of COVID-19 and incidence of post-COVID-19 condition over 10 months (COVID-OUT): a multicentre, randomised, quadruple-blind, parallel-group, phase 3 trial. Lancet Infect Dis. 2023;23(10):1119–29. https://doi.org/10.1016/S1473-3099(23)00299-2.

    Article  PubMed  Google Scholar 

  134. Tong L, Xiao X, Li M, Fang S, Ma E, Yu X, et al. A glucose-like metabolite deficient in diabetes inhibits cellular entry of SARS-CoV-2. Nat Metab. 2022;4(5):547–58. https://doi.org/10.1038/s42255-022-00567-z.

    Article  CAS  PubMed  Google Scholar 

  135. Xie Y, Al-Aly Z. Risks and burdens of incident diabetes in long COVID: a cohort study. Lancet Diabetes Endocrinol. 2022;10(5):311–21. https://doi.org/10.1016/S2213-8587(22)00044-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Liu F, Long X, Zhang B, Zhang W, Chen X, Zhang Z. ACE2 expression in pancreas may cause pancreatic damage after SARS-CoV-2 infection. Clin Gastroenterol Hepatol. 2020;18(9):2128-2130.e2. https://doi.org/10.1016/j.cgh.2020.04.040.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Burnett FN, Coucha M, Bolduc DR, Hermanns VC, Heath SP, Abdelghani M, et al. SARS-CoV-2 spike protein intensifies cerebrovascular complications in diabetic hACE2 mice through RAAS and TLR signaling activation. Int J Mol Sci. 2023;24(22):16394. https://doi.org/10.3390/ijms242216394.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Fadini GP, Morieri ML, Longato E, Avogaro A. Prevalence and impact of diabetes among people infected with SARS-CoV-2. J Endocrinol Invest. 2020;43(6):867–9. https://doi.org/10.1007/s40618-020-01236-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Polat G, Güçsav MO, Özdemir Ö, Türk MA, Unat DS, Tatar D. The association between glycemia and clinical outcomes in patients with diabetes mellitus and pulmonary thromboembolism. Arch Endocrinol Metab. 2023;67(3):341–7. https://doi.org/10.20945/2359-3997000000544.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Gęca T, Wojtowicz K, Guzik P, Góra T. Increased risk of COVID-19 in patients with diabetes mellitus-current challenges in pathophysiology, treatment and prevention. Int J Environ Res Public Health. 2022;19(11):6555. https://doi.org/10.3390/ijerph19116555.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Varikasuvu SR, Varshney S, Dutt N. Markers of coagulation dysfunction and inflammation in diabetic and non-diabetic COVID-19. J Thromb Thrombolysis. 2021;51(4):941–6. https://doi.org/10.1007/s11239-020-02270-w.

    Article  CAS  PubMed  Google Scholar 

  142. Alosaimi B, Mubarak A, Hamed ME, Almutairi AZ, Alrashed AA, AlJuryyan A, et al. Complement anaphylatoxins and inflammatory cytokines as prognostic markers for COVID-19 severity and in-hospital mortality. Front Immunol. 2021;12:668725. https://doi.org/10.3389/fimmu.2021.668725.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. George TP, Joy SS, Rafiullah M, Siddiqui K. Cytokines involved in COVID-19 patients with diabetes: a systematic review. Curr Diabetes Rev. 2023;19(3):e180122200321. https://doi.org/10.2174/1573399818666220118100743.

    Article  CAS  PubMed  Google Scholar 

  144. Menegazzo L, Scattolini V, Cappellari R, Bonora BM, Albiero M, Bortolozzi M, et al. The antidiabetic drug metformin blunts NETosis in vitro and reduces circulating NETosis biomarkers in vivo. Acta Diabetol. 2018;55(6):593–601. https://doi.org/10.1007/s00592-018-1129-8.

    Article  CAS  PubMed  Google Scholar 

  145. Njeim R, Azar WS, Fares AH, Azar ST, Kfoury Kassouf H, Eid AA. NETosis contributes to the pathogenesis of diabetes and its complications. J Mol Endocrinol. 2020;65(4):R65–76. https://doi.org/10.1530/JME-20-0128.

    Article  CAS  PubMed  Google Scholar 

  146. Ghosh A, Gao L, Thakur A, Siu PM, Lai CWK. Role of free fatty acids in endothelial dysfunction. J Biomed Sci. 2017;24:50. https://doi.org/10.1186/s12929-017-0357-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Matuschik L, Riabov V, Schmuttermaier C, Sevastyanova T, Weiss C, Klüter H, et al. Hyperglycemia induces inflammatory response of human macrophages to CD163-mediated scavenging of hemoglobin-haptoglobin complexes. Int J Mol Sci. 2022;23(3):1385. https://doi.org/10.3390/ijms23031385.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Li X, Weber NC, Cohn DM, Hollmann MW, DeVries JH, Hermanides J, et al. Effects of hyperglycemia and diabetes mellitus on coagulation and hemostasis. J Clin Med. 2021;10(11):2419. https://doi.org/10.3390/jcm10112419.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Chourasia P, Goyal L, Kansal D, Roy S, Singh R, Mahata I, et al. Risk of new-onset diabetes mellitus as a post-COVID-19 condition and possible mechanisms: a scoping review. J Clin Med. 2023;12(3):1159. https://doi.org/10.3390/jcm12031159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Calvisi SL, Ramirez GA, Scavini M, Da Prat V, Di Lucca G, Laurenzi A, et al. Thromboembolism risk among patients with diabetes/stress hyperglycemia and COVID-19. Metabolism. 2021;123:154845. https://doi.org/10.1016/j.metabol.2021.154845.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Schiattarella GG, Carrizzo A, Ilardi F, Damato A, Ambrosio M, Madonna M, et al. Rac1 modulates endothelial function and platelet aggregation in diabetes mellitus. J Am Heart Assoc. 2018;7(8):e007322. https://doi.org/10.1161/JAHA.117.007322.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Siddiqui K, George TP, Mujammami M, Isnani A, Alfadda AA. The association of cell adhesion molecules and selectins (VCAM-1, ICAM-1, E-selectin, L-selectin, and P-selectin) with microvascular complications in patients with type 2 diabetes: a follow-up study. Front Endocrinol (Lausanne). 2023;14:1072288. https://doi.org/10.3389/fendo.2023.1072288.

    Article  PubMed  Google Scholar 

  153. Palella E, Cimino R, Pullano SA, Fiorillo AS, Gulletta E, Brunetti A, et al. Laboratory parameters of hemostasis, adhesion molecules, and inflammation in type 2 diabetes mellitus: correlation with glycemic control. Int J Environ Res Public Health. 2020;17(1):300. https://doi.org/10.3390/ijerph17010300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Kim HK, Kim JE, Park SH, Kim YI, Nam-Goong IS, Kim ES. High coagulation factor levels and low protein C levels contribute to enhanced thrombin generation in patients with diabetes who do not have macrovascular complications. J Diabetes Complications. 2014;28(3):365–9. https://doi.org/10.1016/j.jdiacomp.2014.01.006.

    Article  PubMed  Google Scholar 

  155. van der Toorn FA, de Mutsert R, Lijfering WM, Rosendaal FR, van Hylckama VA. Glucose metabolism affects coagulation factors: the NEO study. J Thromb Haemost. 2019;17(11):1886–97. https://doi.org/10.1111/jth.14573.

    Article  CAS  PubMed  Google Scholar 

  156. Barillari G, Fabbro E, Pasca S, Bigotto E. Coagulation and oxidative stress plasmatic levels in a type 2 diabetes population. Blood Coagul Fibrinolysis. 2009;20(4):290–6. https://doi.org/10.1097/MBC.0b013e328329e49b.

    Article  CAS  PubMed  Google Scholar 

  157. Rothschild AM, Reis ML, Melo VL, Foss MC, Gallo L. Increased kininogen levels observed in plasma of diabetic patients are corrected by the administration of insulin. Horm Metab Res. 1999;31(5):326–8. https://doi.org/10.1055/s-2007-978746.

    Article  CAS  PubMed  Google Scholar 

  158. Sambola A, Osende J, Hathcock J, Degen M, Nemerson Y, Fuster V, et al. Role of risk factors in the modulation of tissue factor activity and blood thrombogenicity. Circulation. 2003;107(7):973–7. https://doi.org/10.1161/01.cir.0000050621.67499.7d.

    Article  CAS  PubMed  Google Scholar 

  159. Kietsiriroje N, Pearson SM, O’Mahoney LL, West DJ, Ariëns RA, Ajjan RA, et al. Glucose variability is associated with an adverse vascular profile but only in the presence of insulin resistance in individuals with type 1 diabetes: an observational study. Diab Vasc Dis Res. 2022;19(3):14791641221103216. https://doi.org/10.1177/14791641221103217.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. De Mattia G, Bravi MC, Laurenti O, Moretti A, Cipriani R, Gatti A, et al. Endothelial dysfunction and oxidative stress in type 1 and type 2 diabetic patients without clinical macrovascular complications. Diabetes Res Clin Pract. 2008;79(2):337–42. https://doi.org/10.1016/j.diabres.2007.09.005.

    Article  CAS  PubMed  Google Scholar 

  161. Dhanesha N, Doddapattar P, Chorawala MR, Nayak MK, Kokame K, Staber JM, et al. ADAMTS13 retards progression of diabetic nephropathy by inhibiting intrarenal thrombosis in mice. Arterioscler Thromb Vasc Biol. 2017;37(7):1332–8. https://doi.org/10.1161/ATVBAHA.117.309539.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Dunn EJ, Philippou H, Ariëns RAS, Grant PJ. Molecular mechanisms involved in the resistance of fibrin to clot lysis by plasmin in subjects with type 2 diabetes mellitus. Diabetologia. 2006;49(5):1071–80. https://doi.org/10.1007/s00125-006-0197-4.

    Article  CAS  PubMed  Google Scholar 

  163. Cheng L, Fu Q, Zhou L, Fan Y, Liu F, Fan Y, et al. D-dimer as a predictor of cardiovascular outcomes in patients with diabetes mellitus. BMC Cardiovasc Disord. 2022;22(1):82. https://doi.org/10.1186/s12872-022-02531-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Ajjan RA, Gamlen T, Standeven KF, Mughal S, Hess K, Smith KA, et al. Diabetes is associated with posttranslational modifications in plasminogen resulting in reduced plasmin generation and enzyme-specific activity. Blood. 2013;122(1):134–42. https://doi.org/10.1182/blood-2013-04-494641.

    Article  CAS  PubMed  Google Scholar 

  165. Uitte de Willige S, Malfliet Joyce JCM, Abdul S, Leebeek FWG, Rijken DC. The level of circulating fibroblast activation protein correlates with incorporation of alpha-2-antiplasmin into the fibrin clot. Thromb Res. 2018;166:19–21. https://doi.org/10.1016/j.thromres.2018.03.018.

    Article  CAS  PubMed  Google Scholar 

  166. Agren A, Jörneskog G, Elgue G, Henriksson P, Wallen H, Wiman B. Increased incorporation of antiplasmin into the fibrin network in patients with type 1 diabetes. Diabetes Care. 2014;37(7):2007–14. https://doi.org/10.2337/dc13-1776.

    Article  CAS  PubMed  Google Scholar 

  167. Akwiwu E, Edem M, Akpotuzor J, Asemota E, Isong I. Glycated haemogloin, fasting plasma glucose, plasminogen activator inhibitor type-1, and soluble thrombomodulin levels in patients with type 2 diabetes mellitus. Niger J Physiol Sci. 2021;36(2):159–64. https://doi.org/10.54548/njps.v36i2.3.

    Article  PubMed  Google Scholar 

  168. Kitagawa N, Yano Y, Gabazza EC, Bruno NE, Araki R, Matsumoto K, et al. Different metabolic correlations of thrombin-activatable fibrinolysis inhibitor and plasminogen activator inhibitor-1 in non-obese type 2 diabetic patients. Diabetes Res Clin Pract. 2006;73(2):150–7. https://doi.org/10.1016/j.diabres.2005.12.008.

    Article  CAS  PubMed  Google Scholar 

  169. Miri C, Charii H, Bouazzaoui M, Laouan Brem F, Boulouiz S, Abda N, et al. D-dimer level and diabetes in the COVID-19 infection. Clin Appl Thromb Hemost. 2021;27:10760296211045902. https://doi.org/10.1177/10760296211045902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Iskandar A, Mayashinta DK, Robert R, Samsu N, Endharti AT, Widjajanto E. Correlation between IL-8, C-reactive proteins (CRP) and neutrophil to lymphocyte ratio (NLR) as predictor of mortality in COVID-19 patients with diabetes mellitus comorbidity. Int J Gen Med. 2023;16:2349–54. https://doi.org/10.2147/IJGM.S412070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Francischetti IMB, Toomer K, Zhang Y, Jani J, Siddiqui Z, Brotman DJ, et al. Upregulation of pulmonary tissue factor, loss of thrombomodulin and immunothrombosis in SARS-CoV-2 infection. EClinicalMedicine. 2021;39:101069. https://doi.org/10.1016/j.eclinm.2021.101069.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Soma P, Swanepoel AC, Bester J, Pretorius E. Tissue factor levels in type 2 diabetes mellitus. Inflamm Res. 2017;66(5):365–8. https://doi.org/10.1007/s00011-017-1030-x.

    Article  CAS  PubMed  Google Scholar 

  173. Masi P, Hékimian G, Lejeune M, Chommeloux J, Desnos C, Pineton De Chambrun M, et al. Systemic inflammatory response syndrome is a major contributor to COVID-19-associated coagulopathy: insights from a prospective, single-center cohort study. Circulation. 2020;142(6):611–4. https://doi.org/10.1161/CIRCULATIONAHA.120.048925.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Zhang Y, Cao W, Jiang W, Xiao M, Li Y, Tang N, et al. Profile of natural anticoagulant, coagulant factor and anti-phospholipid antibody in critically ill COVID-19 patients. J Thromb Thrombolysis. 2020;50(3):580–6. https://doi.org/10.1007/s11239-020-02182-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Kedzierska K, Ciechanowski K, Gołembiewska E, Safranow K, Ciechanowicz A, Domański L, et al. Plasma prekallikrein as a risk factor for diabetic retinopathy. Arch Med Res. 2005;36(5):539–43. https://doi.org/10.1016/j.arcmed.2005.03.050.

    Article  CAS  PubMed  Google Scholar 

  176. Colhoun HM, Zito F, Norman Chan N, Rubens MB, Fuller JH, Humphries SE. Activated factor XII levels and factor XII 46C>T genotype in relation to coronary artery calcification in patients with type 1 diabetes and healthy subjects. Atherosclerosis. 2002;163(2):363–9. https://doi.org/10.1016/s0021-9150(02)00022-9.

    Article  CAS  PubMed  Google Scholar 

  177. Ceballos FC, Ryan P, Blancas R, Martin-Vicente M, Vidal-Alcántara EJ, Peréz-García F, et al. Are reduced levels of coagulation proteins upon admission linked to COVID-19 severity and mortality? Front Med. 2021;8:718053. https://doi.org/10.3389/fmed.2021.718053.

    Article  Google Scholar 

  178. Paszek E, Polak M, Bryk-Wiązania AH, Konieczyńska M, Undas A. Elevated plasma factor XI predicts cardiovascular events in patients with type 2 diabetes: a long-term observational study. Cardiovasc Diabetol. 2023;22(1):182. https://doi.org/10.1186/s12933-023-01905-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M, Delabranche X, et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med. 2020;46(6):1089–98. https://doi.org/10.1007/s00134-020-06062-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Ephraim RK, Awuku YA, Adu P, Ampomah LT, Adoba P, Panford S, et al. High risk of coagulopathy among Type-2 diabetes mellitus clients at a municipal hospital in Ghana. Ghana Med J. 2017;51(3):101–7. https://doi.org/10.4314/gmj.v51i3.2.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Althaus K, Marini I, Zlamal J, Pelzl L, Singh A, Häberle H, et al. Antibody-induced procoagulant platelets in severe COVID-19 infection. Blood. 2021;137(8):1061–71. https://doi.org/10.1182/blood.2020008762.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Marchetti M, Gomez-Rosas P, Russo L, Gamba S, Sanga E, Verzeroli C, et al. Fibrinolytic proteins and factor XIII as predictors of thrombotic and hemorrhagic complications in hospitalized COVID-19 patients. Front Cardiovasc Med. 2022;9:896362. https://doi.org/10.3389/fcvm.2022.896362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Sauls DL, Banini AE, Boyd LC, Hoffman M. Elevated prothrombin level and shortened clotting times in subjects with type 2 diabetes. J Thromb Haemost. 2007;5(3):638–9. https://doi.org/10.1111/j.1538-7836.2007.02366.x.

    Article  CAS  PubMed  Google Scholar 

  184. Zhao Y, Zhang J, Zhang J, Wu J. Diabetes mellitus is associated with shortened activated partial thromboplastin time and increased fibrinogen values. PLoS ONE. 2011;6(1):e16470. https://doi.org/10.1371/journal.pone.0016470.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  185. Sui J, Noubouossie DF, Gandotra S, Cao L. Elevated plasma fibrinogen is associated with excessive inflammation and disease severity in COVID-19 patients. Front Cell Infect Microbiol. 2021;11:734005. https://doi.org/10.3389/fcimb.2021.734005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Mansfield MW, Kohler HP, Ariëns RA, McCormack LJ, Grant PJ. Circulating levels of coagulation factor XIII in subjects with type 2 diabetes and in their first-degree relatives. Diabetes Care. 2000;23(5):703–5. https://doi.org/10.2337/diacare.23.5.703.

    Article  CAS  PubMed  Google Scholar 

  187. Lichter Y, Badelbayov T, Shalev I, Schvartz R, Szekely Y, Benisty D, et al. Low FXIII activity levels in intensive care unit hospitalized COVID-19 patients. Thromb J. 2021;19(1):79. https://doi.org/10.1186/s12959-021-00333-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Medina MG, Ledesma MD, Domínguez JE, Medina M, Zafra D, Alameda F, et al. Tissue plasminogen activator mediates amyloid-induced neurotoxicity via Erk1/2 activation. EMBO J. 2005;24(9):1706–16. https://doi.org/10.1038/sj.emboj.7600650.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Andrianto N, Al-Farabi MJ, Nugraha RA, Marsudi BA, Azmi Y. Biomarkers of endothelial dysfunction and outcomes in coronavirus disease 2019 (COVID-19) patients: a systematic review and meta-analysis. Microvasc Res. 2021;138:104224. https://doi.org/10.1016/j.mvr.2021.104224.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Carmassi F, Morale M, Puccetti R, De Negri F, Monzani F, Navalesi R, et al. Coagulation and fibrinolytic system impairment in insulin dependent diabetes mellitus. Thromb Res. 1992;67(6):643–54. https://doi.org/10.1016/0049-3848(92)90068-l.

    Article  CAS  PubMed  Google Scholar 

  191. Della-Morte D, Pacifici F, Ricordi C, Massoud R, Rovella V, Proietti S, et al. Low level of plasminogen increases risk for mortality in COVID-19 patients. Cell Death Dis. 2021;12(8):773. https://doi.org/10.1038/s41419-021-04070-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Bouck EG, Denorme F, Holle LA, Middelton EA, Blair AM, de Laat B, et al. COVID-19 and sepsis are associated with different abnormalities in plasma procoagulant and fibrinolytic activity. Arterioscler Thromb Vasc Biol. 2021;41(1):401–14. https://doi.org/10.1161/ATVBAHA.120.315338.

    Article  CAS  PubMed  Google Scholar 

  193. Boknäs N, Laine C, Hillarp A, Macwan AS, Gustafsson KM, Lindahl TL, et al. Associations between hemostatic markers and mortality in COVID-19—compounding effects of D-dimer, antithrombin and PAP complex. Thromb Res. 2022;213:97–104. https://doi.org/10.1016/j.thromres.2022.03.013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. De Jongh R, Ninivaggi M, Mesotten D, Bai C, Marcus B, Huskens D, et al. Vascular activation is a strong predictor of mortality in coronavirus disease 2019 patients on the ICU. Blood Coagul Fibrinolysis. 2021;32(4):290–3. https://doi.org/10.1097/MBC.0000000000001007.

    Article  CAS  PubMed  Google Scholar 

  195. Zhuang L, Yu C, Xu F, Zhao L, Wang X, Wang C, et al. Increased plasma D-dimer levels may be a promising indicator for diabetic peripheral neuropathy in type 2 diabetes. Front Endocrinol. 2022;13:930271. https://doi.org/10.3389/fendo.2022.930271.

    Article  Google Scholar 

  196. Pérez-García N, García-González J, Requena-Mullor M, Rodríguez-Maresca MÁ, Alarcón-Rodríguez R. Comparison of analytical values D-Dimer, glucose, ferritin and C-reactive protein of symptomatic and asymptomatic COVID-19 patients. Int J Environ Res Public Health. 2022;19(9):5354. https://doi.org/10.3390/ijerph19095354.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Fukahori S, Han JY, Vera I, Lockey R, Kim K, Cho S. Elevated PAI-1 levels are associated with severe COVID-19. J Allergy Clin Immunol. 2023;151(2):190. https://doi.org/10.1016/j.jaci.2022.12.595.

    Article  Google Scholar 

  198. Pandolfi A, Cetrullo D, Polishuck R, Alberta MM, Calafiore A, Pellegrini G, et al. Plasminogen activator inhibitor type 1 is increased in the arterial wall of type II diabetic subjects. Arterioscler Thromb Vasc Biol. 2001;21(8):1378–82. https://doi.org/10.1161/hq0801.093667.

    Article  CAS  PubMed  Google Scholar 

  199. Agosti F, Toffolutti V, Cavalli N, Nivakoski S, Mascherini M, Aassve A. Information and vaccine hesitancy: Evidence from the early stage of the vaccine roll-out in 28 European countries. PLoS One. 2022;17(9):e0273555. https://doi.org/10.1371/journal.pone.0273555.

  200. de Gregorio C, Calcaterra G, Kounis NG, Bassareo PP, Mehta JL. Cerebral venous thrombosis after COVID-19 vaccines: Do we know the mechanism? Lancet Reg Health Eur. 2022;16:100387. https://doi.org/10.1016/j.lanepe.2022.100387.

  201. Brambilla M, Canzano P, Valle PD, Becchetti A, Conti M, Alberti M, et al. Head-to-head comparison of four COVID-19 vaccines on platelet activation, coagulation and inflammation. The TREASURE study.  Thromb Res. 2023;223:24–33. https://doi.org/10.1016/j.thromres.2023.01.015.

  202. Kompaniyets L, Pennington AF, Goodman AB, Rosenblum HG, Belay B, Ko JY, et al. Underlying Medical Conditions and Severe Illness Among 540,667 Adults Hospitalized With COVID-19, March 2020-March 2021. Prev Chronic Dis. 2021;18:E66. https://doi.org/10.5888/pcd18.210123.

  203. Zhang R, Ouyang H, Fu L, Wang S, Han J, Huang K, et al. CT features of SARS-CoV-2 pneumonia according to clinical presentation: a retrospective analysis of 120 consecutive patients from Wuhan city. Eur Radiol. 2020;30(8):4417–26. https://doi.org/10.1007/s00330-020-06854-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Cohen JI, Burbelo PD. Reinfection with SARS-CoV-2: implications for vaccines. Clin Infect Dis. 2021;73(11):e4223–8. https://doi.org/10.1093/cid/ciaa1866.

    Article  CAS  PubMed  Google Scholar 

  205. Sun J, Zheng Q, Madhira V, Olex AL, Anzalone AJ, Vinson A, et al. Association between immune dysfunction and COVID-19 breakthrough infection after SARS-CoV-2 vaccination in the US. JAMA Intern Med. 2022;182(2):153–62. https://doi.org/10.1001/jamainternmed.2021.7024.

    Article  CAS  PubMed  Google Scholar 

  206. Yang Y, Cai Z, Zhang J. Insulin treatment may increase adverse outcomes in patients with COVID-19 and diabetes: a systematic review and meta-analysis. Front Endocrinol. 2021;12:696087. https://doi.org/10.3389/fendo.2021.696087.

    Article  Google Scholar 

  207. Liu J, Aylor KW, Chai W, Barrett EJ, Liu Z. Metformin prevents endothelial oxidative stress and microvascular insulin resistance during obesity development in male rats. Am J Physiol Endocrinol Metab. 2022;322(3):E293–306. https://doi.org/10.1152/ajpendo.00240.2021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Abad-Jiménez Z, López-Domènech S, Díaz-Rúa R, Iannantuoni F, Gómez-Abril SÁ, Periañez-Gómez D, et al. Systemic oxidative stress and visceral adipose tissue mediators of NLRP3 inflammasome and autophagy are reduced in obese type 2 diabetic patients treated with metformin. Antioxidants. 2020;9(9):892. https://doi.org/10.3390/antiox9090892.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Victor VM, Rovira-Llopis S, Bañuls C, Diaz-Morales N, Lopez-Domenech S, Escribano-López I, et al. Metformin modulates human leukocyte/endothelial cell interactions and proinflammatory cytokines in polycystic ovary syndrome patients. Atherosclerosis. 2015;242(1):167–73. https://doi.org/10.1016/j.atherosclerosis.2015.07.017.

    Article  CAS  PubMed  Google Scholar 

  210. Gongol B, Marin T, Peng I, Woo B, Martin M, King S, et al. AMPKα2 exerts its anti-inflammatory effects through PARP-1 and Bcl-6. Proc Natl Acad Sci U S A. 2013;110(8):3161–6. https://doi.org/10.1073/pnas.1222051110.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  211. Witkowski M, Friebel J, Tabaraie T, Grabitz S, Dörner A, Taghipour L, et al. Metformin is associated with reduced tissue factor procoagulant activity in patients with poorly controlled diabetes. Cardiovasc Drugs Ther. 2021;35(4):809–13. https://doi.org/10.1007/s10557-020-07040-7.

    Article  CAS  PubMed  Google Scholar 

  212. Xin G, Wei Z, Ji C, Zheng H, Gu J, Ma L, et al. Metformin uniquely prevents thrombosis by inhibiting platelet activation and mtDNA release. Sci Rep. 2016;6:36222. https://doi.org/10.1038/srep36222.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  213. Apicella M, Campopiano MC, Mantuano M, Mazoni L, Coppelli A, Del Prato S. COVID-19 in people with diabetes: understanding the reasons for worse outcomes. Lancet Diabetes Endocrinol. 2020;8(9):782–92. https://doi.org/10.1016/S2213-8587(20)30238-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Ganesh A, Randall MD. Does metformin affect outcomes in COVID-19 patients with new or pre-existing diabetes mellitus? A systematic review and meta-analysis. Br J Clin Pharmacol. 2022;88(6):2642–56. https://doi.org/10.1111/bcp.15258.

    Article  CAS  PubMed  Google Scholar 

  215. Parthasarathy H, Tandel D, Siddiqui AH, Harshan KH. Metformin suppresses SARS-CoV-2 in cell culture. Virus Res. 2023;323:199010. https://doi.org/10.1016/j.virusres.2022.199010.

    Article  CAS  PubMed  Google Scholar 

  216. Cory TJ, Emmons RS, Yarbro JR, Davis KL, Pence BD. Metformin suppresses monocyte immunometabolic activation by SARS-CoV-2 spike protein subunit 1. Front Immunol. 2021;12:733921. https://doi.org/10.3389/fimmu.2021.733921.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Shaseb E, Ghaffary S, Garjani A, Zoghi E, Maleki Dizaji N, Soltani S, et al. Long and short-term metformin consumption as a potential therapy to prevent complications of COVID-19. Adv Pharm Bull. 2023;13(3):621–6. https://doi.org/10.34172/apb.2023.066.

    Article  CAS  PubMed  Google Scholar 

  218. Wiernsperger N, Al-Salameh A, Cariou B, Lalau J. Protection by metformin against severe Covid-19: an in-depth mechanistic analysis. Diabetes Metab. 2022;48(4):101359. https://doi.org/10.1016/j.diabet.2022.101359.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Shestakova MV, Vikulova OK, Elfimova AR, Deviatkin AA, Dedov II, Mokrysheva NG. Risk factors for COVID-19 case fatality rate in people with type 1 and type 2 diabetes mellitus: a nationwide retrospective cohort study of 235,248 patients in the Russian Federation. Front Endocrinol. 2022;13:909874. https://doi.org/10.3389/fendo.2022.909874.

    Article  CAS  Google Scholar 

  220. Khunti K, Knighton P, Zaccardi F, Bakhai C, Barron E, Holman N, et al. Prescription of glucose-lowering therapies and risk of COVID-19 mortality in people with type 2 diabetes: a nationwide observational study in England. Lancet Diabetes Endocrinol. 2021;9(5):293–303. https://doi.org/10.1016/S2213-8587(21)00050-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Luk AOY, Yip TCF, Zhang X, Kong APS, Wong VW, Ma RCW, et al. Glucose-lowering drugs and outcome from COVID-19 among patients with type 2 diabetes mellitus: a population-wide analysis in Hong Kong. BMJ Open. 2021;11(10):e052310. https://doi.org/10.1136/bmjopen-2021-052310.

    Article  PubMed  Google Scholar 

  222. ASH Guidelines on Use of Anticoagulation in Patients with COVID-19—Hematology.org. https://www.hematology.org/education/clinicians/guidelines-and-quality-care/clinical-practice-guidelines/venous-thromboembolism-guidelines/ash-guidelines-on-use-of-anticoagulation-in-patients-with-covid-19. Accessed 28 Sep 2023.

  223. Cuker A, Tseng EK, Nieuwlaat R, Angchaisuksiri P, Blair C, Dane K, et al. American society of hematology 2021 guidelines on the use of anticoagulation for thromboprophylaxis in patients with COVID-19. Blood Adv. 2021;5(3):872–88. https://doi.org/10.1182/bloodadvances.2020003763.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Chaves OA, Sacramento CQ, Fintelman-Rodrigues N, Temerozo JR, Pereira-Dutra F, Mizurini DM, et al. Apixaban, an orally available anticoagulant, inhibits SARS-CoV-2 replication and its major protease in a non-competitive way. J Mol Cell Biol. 2022;14(6):mjac039. https://doi.org/10.1093/jmcb/mjac039.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Gianni P, Goldin M, Ngu S, Zafeiropoulos S, Geropoulos G, Giannis D. Complement-mediated microvascular injury and thrombosis in the pathogenesis of severe COVID-19: a review. World J Exp Med. 2022;12(4):53–67. https://doi.org/10.5493/wjem.v12.i4.53.

    Article  PubMed  PubMed Central  Google Scholar 

  226. van de Veerdonk FL, Kouijzer IJE, de Nooijer AH, van der Hoeven HG, Maas C, Netea MG, et al. Outcomes associated with use of a kinin B2 receptor antagonist among patients with COVID-19. JAMA Netw Open. 2020;3(8):e2017708. https://doi.org/10.1001/jamanetworkopen.2020.17708.

    Article  PubMed  PubMed Central  Google Scholar 

  227. Aboudounya MM, Heads RJ. COVID-19 and toll-like receptor 4 (TLR4): SARS-CoV-2 may bind and activate TLR4 to increase ACE2 expression, facilitating entry and causing hyperinflammation. Med Inflamm. 2021;2021:8874339. https://doi.org/10.1155/2021/8874339.

    Article  CAS  Google Scholar 

  228. Fontes-Dantas FL, Fernandes GG, Gutman EG, De Lima EV, Antonio LS, Hammerle MB, et al. SARS-CoV-2 Spike protein induces TLR4-mediated long-term cognitive dysfunction recapitulating post-COVID-19 syndrome in mice. Cell Rep. 2023;42(3):112189. https://doi.org/10.1016/j.celrep.2023.112189.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Zhao D, Qin Y, Liu J, Tang K, Lu S, Liu Z, et al. Orally administered BZL-sRNA-20 oligonucleotide targeting TLR4 effectively ameliorates acute lung injury in mice. Sci China Life Sci. 2023;66(7):1589–99. https://doi.org/10.1007/s11427-022-2219-0.

    Article  CAS  PubMed  Google Scholar 

  230. Wei W, Liaw C, Tsai K, Chiou C, Tseng Y, Chiou W, et al. Targeting spike protein-induced TLR/NET axis by COVID-19 therapeutic NRICM102 ameliorates pulmonary embolism and fibrosis. Pharmacol Res. 2022;184:106424. https://doi.org/10.1016/j.phrs.2022.106424.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Gadanec L, Qaradakhi T, Renee McSweeney K, Ashiana Ali B, Zulli A, Apostolopoulos V. Dual targeting of Toll-like receptor 4 and angiotensin-converting enzyme 2: a proposed approach to SARS-CoV-2 treatment. Future Microbiol. 2021;16:205–9. https://doi.org/10.2217/fmb-2021-0018.

    Article  CAS  Google Scholar 

  232. Griffin JH, Lyden P. COVID-19 hypothesis: activated protein C for therapy of virus-induced pathologic thromboinflammation. Res Pract Thromb Haemost. 2020;4(4):506–9. https://doi.org/10.1002/rth2.12362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Ali RA, Gandhi AA, Meng H, Yalavarthi S, Vreede AP, Estes SK, et al. Adenosine receptor agonism protects against NETosis and thrombosis in antiphospholipid syndrome. Nat Commun. 2019;10:1916. https://doi.org/10.1038/s41467-019-09801-x.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  234. Okeke EB, Louttit C, Fry C, Najafabadi AH, Han K, Nemzek J, et al. Inhibition of neutrophil elastase prevents neutrophil extracellular trap formation and rescues mice from endotoxic shock. Biomaterials. 2020;238:119836. https://doi.org/10.1016/j.biomaterials.2020.119836.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Wienkamp A, Erpenbeck L, Rossaint J. Platelets in the NETworks interweaving inflammation and thrombosis. Front Immunol. 2022;13:953129. https://doi.org/10.3389/fimmu.2022.953129.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Yang L, Xie X, Tu Z, Fu J, Xu D, Zhou Y. The signal pathways and treatment of cytokine storm in COVID-19. Sig Transduct Target Ther. 2021;6(1):1–20. https://doi.org/10.1038/s41392-021-00679-0.

    Article  CAS  Google Scholar 

  237. Kim JS, Lee JY, Yang JW, Lee KH, Effenberger M, Szpirt W, et al. Immunopathogenesis and treatment of cytokine storm in COVID-19. Theranostics. 2021;11(1):316–29. https://doi.org/10.7150/thno.49713.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Gordon AC, Mouncey PR, Al-Beidh F, Rowan KM, Nichol AD, Arabi YM, et al. Interleukin-6 receptor antagonists in critically Ill patients with Covid-19. N Engl J Med. 2021;384(16):1491–502. https://doi.org/10.1056/NEJMoa2100433.

    Article  PubMed  Google Scholar 

  239. Hepprich M, Mudry JM, Gregoriano C, Jornayvaz FR, Carballo S, Wojtusciszyn A, et al. Canakinumab in patients with COVID-19 and type 2 diabetes—A multicentre, randomised, double-blind, placebo-controlled trial. EClinicalMedicine. 2022;53:101649. https://doi.org/10.1016/j.eclinm.2022.101649.

    Article  PubMed  PubMed Central  Google Scholar 

  240. Yonekawa A, Shimono N. Clinical significance of COVID-19 and diabetes: in the pandemic situation of SARS-CoV-2 variants including omicron B.1.1.529. Biology. 2022;11(3):400. https://doi.org/10.3390/biology11030400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Supported by funds from REACT-EU-Comunidad de Madrid and the European Regional Development Fund (SPACE2-CV-COVID-CM) to C. Peiró and O. Lorenzo; from the Spanish Ministry of Economy (SAF2017-84776-R and PID2020-115590RB-100/AEI/10.13039/501100011033) to C.F. Sánchez-Ferrer and C. Peiró; from Instituto de Salud Carlos III and Fondo de Investigaciones Sanitarias (P120/00923) to O. Lorenzo; from Instituto de Salud Carlos III via Sara Borrell postdoctoral grant (CD22/00101) to I. Valencia; from Universidad Autónoma de Madrid via FPI-UAM PhD contract to J. Lumpuy-Castillo; from European Union-Next Generation EU via María Zambrano postdoctoral grant in the UAM to G. Magalhaes.

Author information

Authors and Affiliations

Authors

Contributions

CP, OL and IV conceived and edited the manuscript. IV, JL-C, GM, CFS-F, CP and OL wrote the sections of the manuscript. All authors approved the final version of the manuscript.

Corresponding authors

Correspondence to Inés Valencia, Óscar Lorenzo or Concepción Peiró.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Valencia, I., Lumpuy-Castillo, J., Magalhaes, G. et al. Mechanisms of endothelial activation, hypercoagulation and thrombosis in COVID-19: a link with diabetes mellitus. Cardiovasc Diabetol 23, 75 (2024). https://doi.org/10.1186/s12933-023-02097-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12933-023-02097-8

Keywords