Back to Journals » Journal of Inflammation Research » Volume 18
The Mechanisms of Sepsis Induced Coagulation Dysfunction and Its Treatment
Authors Zhu L, Dong H, Li L, Liu X
Received 4 November 2024
Accepted for publication 22 January 2025
Published 3 February 2025 Volume 2025:18 Pages 1479—1495
DOI https://doi.org/10.2147/JIR.S504184
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Professor Ning Quan
Lei Zhu,1,2 He Dong,1,2 Lin Li,1,2 Xiaojie Liu1,2
1Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China; 2Department of Anesthesiology, Shandong Provincial Key Medical and Heath Laboratory of Anesthesia and Brain Function, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
Correspondence: Xiaojie Liu, Email [email protected]
Abstract: Sepsis is a critical condition characterized by organ dysfunction due to a dysregulated response to infection that poses significant global health challenges. Coagulation dysfunction is nearly ubiquitous among sepsis patients. Its mechanisms involve platelet activation, coagulation cascade activation, inflammatory reaction imbalances, immune dysregulation, mitochondrial damage, neuroendocrine network disruptions, and endoplasmic reticulum (ER) stress. These factors not only interact but also exacerbate one another, leading to severe organ dysfunction. This review illustrates the mechanisms of sepsis-induced coagulopathy, with a focus on tissue factor activation, endothelial glycocalyx damage, and the release of neutrophil extracellular traps (NETs), all of which are potential targets for therapeutic interventions.
Keywords: sepsis, coagulation dysfunction, thrombosis
Introduction
The prevalence and mortality rates of sepsis remain extremely high, particularly in economically undeveloped regions such as Africa, South Asia, and Latin America.1,2 In China, One-fifth of ICU patients suffer from sepsis, with a 90-day mortality rate of 35.5%. A shortage of ICU beds exacerbates the situation, hindering optimal management and accurate data collection, thereby emphasizing the need for comprehensive research into its prevalence and mortality.3 In 2016, the third international consensus on sepsis redefined it as a life-threatening organ dysfunction caused by a dysregulated host response to infection.4 The coagulation dysfunction leading to microthrombosis caused by sepsis is a very serious disease, eventually leading to disseminated intravascular coagulation (DIC) and organ dysfunction. Thus, it has a high mortality rate.5
Currently, the mechanisms underlying coagulopathy in sepsis are focused on the imbalance of immune system and inflammatory response, activation of platelets and coagulation factors, endothelial cell injury, and disruption of the coagulation-fibrinolysis balance.6 Sepsis is typically caused by bacterial or other pathogenic microbial infections, leading to systemic inflammatory response.4 The infection initially triggers immune response, in which immune cells recognize the pathogen and release a large number of inflammatory cytokines. These cytokines not only provoke local inflammatory responses but also activate the coagulation system throughout the body.7,8 They further damage endothelial cells (ECs), exposing more tissue factor (TF) and simultaneously decreasing the synthesis of anticoagulant and antifibrinolytic factors,9 thus promoting the activation of the coagulation cascade and accelerating blood clotting.10,11 The activation of platelets and coagulation factors results in sepsis-induced coagulopathy (SIC), leading to microthrombi formation. As these elements are depleted, the balance between coagulation and fibrinolysis is disrupted, resulting in DIC.12,13 In addition, mitochondrial damage, neuroendocrine dysregulation, and endoplasmic reticulum (ER) stress may also contribute to coagulation dysfunction. However, current research has predominantly focused on the common mechanisms discussed above, without offering a systematic and comprehensive analysis.
Historically, the lack of a single diagnostic criterion and the high costs of coagulation tests have limited their use.14 The International Society on Thrombosis and Haemostasis (ISTH) established a clear definition and scoring system for SIC, an early phase of DIC, that includes the platelet count and international normalized ratio (INR) in 2017. The SOFA score can be used to diagnose DIC to meet the SIC criteria at the initial assessment.15,16 I Iba et al reported that the SIC scoring system was highly sensitive, as an effective screening tool for DIC, providing insights into mortality risk and guiding the initiation of heparin anticoagualtion therapy in 2019.17,18 Recently, advances in technology have facilitated the development of an increasing number of targeted therapies for sepsis-induced coagulopathy, providing new prospects for patient outcomes.
Mechanisms of Coagulation Dysfunction Induced by Sepsis
Coagulopathy, ranging from mild thrombocytopenia to severe conditions such as DIC, is a key determinant of mortality in sepsis patients. Most patients with sepsis undergo significant hemostatic alterations.19 The pathogenesis of sepsis involves various mechanisms, including inflammatory imbalances, immune dysregulation, disturbance of coagulation, mitochondrial damage, neuroendocrine disruption, and ER stress, all of which ultimately lead to organ dysfunction.20
Activation of Platelets and Coagulation Cascades
The physiological coagulation cascade is a highly regulated process that includes vascular constriction, platelet plug formation, and coagulation (Figure 1). These phases are sequential yet overlapping, each reinforcing the others.21 When damage occurs, vascular ECs are susceptible to various factors and pathogenic stimuli. Platelets attach to exposed collagen and von Willebrand factor in the subendothelial layer via their surface glycoprotein Ib-IX-V Complex (GPIb-IX-V), initiating activation.22 The subsequent release of adenosine diphosphate (ADP), thromboxane A2 (TXA2), and platelet factor 4 (PF4) further activates and aggregates platelets, leading to thrombus formation.23,24 Endothelial damage and trauma also trigger both intrinsic and extrinsic coagulation pathways. Foreign substances activate factor XII (FXII), sequentially activating activate factor XI (FXI), factor X (FX), factor IX (FIX), and factor VIII (FVIII), forming the intrinsic Xase complex in the presence of calcium, thereby converting FX to factor Xa (FXa).25,26 When combined with factor VIIa (FVIIa) and calcium, TF forms the TF-FVIIa complex, activating FX to FXa. The resulting FXa, along with factor Va (FVa) and calcium from both pathways, forms the prothrombin complex on phospholipid membranes, which then converts prothrombin to thrombin, transforming fibrinogen into fibrin and finalizing the coagulation process.27–30
Activation of TF
TF is a glycoprotein receptor on cell membranes that is expressed primarily on ECs (Figure 2). Upon tissue injury, TF is exposed to the bloodstream, initiating the extrinsic coagulation pathway.31 Pattern recognition receptors (PRRs) detect damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), activating the innate immune system.28 During sepsis, lipopolysaccharide (LPS) enters the bloodstream and binds to CD14 receptors on the surfaces of monocytes/macrophages and ECs, activating Toll-like receptor 4 (TLR4). This activation initiates signaling through Myeloid Differentiation Primary Response Gene 88 (MyD88) and Toll/Interleukin-1 Receptor (TIR) Domain-Containing Adapter Inducing IFN-β (TRIF) dependent pathways, leading to the activation of the Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) and Interferon Regulatory Factor 3 (IRF3) signaling cascades. This process promotes the production of proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), and interleukin-6 (IL-6) and upregulates TF gene expression, thereby triggering the coagulation cascade.32,33 Hepatocytes detect PAMPs in the circulation and release high mobility group box 1 protein (HMGB1), a key protein in the processes of proinflammatory responses and pyroptosis. HMGB1 binds to TLR4 receptors on the cell surface, promoting TF gene expression through the MyD88/NF-κB pathway. Additionally, HMGB1 can interact with the Receptor for Advanced Glycation End Products (RAGE), activating NF-κB via the Mitogen-Activated Protein Kinase (MAPK) pathways, including Extracellular Signal-Regulated Kinase 1/2 (ERK1/2), c-Jun N-terminal Kinase (JNK), and p38 Mitogen-Activated Protein Kinase (p38), as well as the Phosphoinositide 3-Kinase (PI3K)/Protein Kinase B (Akt) pathways. This activation, in turn, upregulates TF gene expression. Research by Deng indicated that HMGB1, upon binding to LPS, can enter lysosomes via the RAGE pathway. HMGB1 disrupts the lysosomal membrane structure, allowing LPS to enter the cytoplasm and activate caspase-11.32,34 Activated caspase-11 triggers the formation of gasdermin D (GSDMD) pores and phosphatidylserine exposure, thereby activating pathways such as the NF-κB and MAPK pathways to promote the expression of TF. Furthermore, GSDMD promotes the maturation of IL-1 by inducing the activation of inflammasomes.35 Inflammasomes, exemplified by NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3), not only mediate the maturation and secretion of IL-1 but also induce monocytes/macrophages to release TF.36 As proinflammatory factors, the expression of TF is continuously upregulated, and inflammation and coagulation are further activated.37
Antithrombin (AT) and Activated Protein C (APC)
AT is a crucial natural anticoagulant that regulates the coagulation process by interacting with key coagulation factors. Synthesized in the liver, AT circulates in the bloodstream, playing a vital role in maintaining hemostasis. Its primary targets are FIIa and FXa.38 The reactive peptide region of AT binds to the catalytic triad of thrombin, which includes serine (Ser195), histidine (His57), and aspartic acid (Asp102), thus inducing a conformational change in thrombin that accelerates its inactivation, followed by clearance in the liver.39 Heparin enhances the anticoagulant activity of AT by binding to it, inducing a conformational change that increases its affinity for thrombin, thus further accelerating thrombin inactivation and improving AT’s anticoagulant effect.40,41 Sepsis triggers a systemic inflammatory response that activates the coagulation cascade, leading to the activation of multiple coagulation factors. These activated factors require inhibition by AT, contributing to its rapid consumption. In addition, inflammatory mediators such as TNF-α and IL-6 suppress AT synthesis and promote its accelerated degradation. Simultaneously, the functionality of AT is compromised, primarily due to a decreased affinity for coagulation factors, which impairs its ability to effectively inhibit the coagulation process.42
APC is another important natural anticoagulant in the body, primarily regulating coagulation and inflammation through multiple mechanisms.43 Initially, the zymogen protein C is activated by thrombin in the presence of thrombomodulin (TM), a receptor on ECs, which converts it into active APC. Once activated, APC, in conjunction with its cofactor Protein S, cleaves coagulation FVa and FVIIIa, thereby inhibiting the amplification of the coagulation cascade.44 Specifically, APC cleaves distinct peptide bonds in FVa and FVIIIa,45 which reduces their ability to activate FXa and FIXa, ultimately inhibiting thrombin generation.43,46 Furthermore, APC promotes the release of nitric oxide (NO) from ECs, leading to vasodilation, improving blood flow, and a reduction in thrombus formation.47 In addition to its anticoagulant function, APC exerts anti-inflammatory effects by binding to Proteinase-Activated Receptor-1 (PAR-1), thereby inhibiting the activation of endothelial and immune cells.48 This interaction reduces the release of pro-inflammatory cytokines, such as TNF-α and IL-6, mitigating systemic inflammation.43 During sepsis, although thrombin activates protein C via the endothelial receptor TM, the generation of APC are significantly reduced due to suppression of protein C synthesis49 by elevated inflammatory cytokines (eg, TNF-α, IL-6).50 Sepsis-induced endothelial damage further impairs TM function, diminishing APC activation. The excessive activation of coagulation factors in sepsis leads to consumption of APC, depleting its levels. Similar to AT, inflammatory cytokines in sepsis can alter the conformation of APC, reducing its affinity for coagulation factors and impairing its ability to degrade FVa and FVIIIa,45 thereby exacerbating coagulation.51 Additionally, high concentrations of pro-inflammatory cytokines in sepsis inhibit APC binding to PAR-1,48 further diminishing its anti-inflammatory effects.
Inflammatory Imbalance Induces Coagulation Dysfunction
Systemic inflammatory response syndrome (SIRS) refers to the complex pathophysiologic response to an insult, such as infection, trauma, burns, pancreatitis, or a variety of other injuries, and comes from the Sepsis Definitions Consensus Conference held in Chicago, Interleukin-8 (IL-8), in August 1991.52 The clinical definition of sepsis that emerged from the consensus conference sponsored by the American College of Chest Physicians and the Society of Critical Care Medicine was met with mixed reactions.53,54 The focus of sepsis has shifted toward organ dysfunction following the 2017 international consensus.4 In sepsis, the immune system’s balance is disrupted, resulting in both excessive inflammation and immune suppression, with inflammatory responses potentially activating the coagulation cascade.36,55 PRRs recognize DAMPs and PAMPs, triggering inflammatory pathways that release cytokines, chemokines, and growth factors, such as TNF-α and IL-6, which activate coagulation factors VII and XII. Platelet-activating factor (PAF) and PF4 promote platelet aggregation.56 DAMPs and PAMPs also activate the complement cascade through the classical, alternative, and lectin pathways,57 generating Complement Component 3a (C3a) and Complement Component 5a (C5a), and forming the membrane attack complex (MAC), which promotes inflammation and damages cells.58,59 In 2013, it was reported that platelets could promote P-selectin expression to form complexes with P-selectin glycoprotein ligand 1 (PSGL-1) on leukocytes at inflammatory sites, enhancing the inflammatory response.60,61
Endothelial Damage
ECs play a critical role in regulating coagulation by both pro-coagulant and anti-coagulant mechanisms.27 During vascular injury or inflammation, ECs release pro-coagulant factors such as von Willebrand factor (vWF) and TF. vWF facilitates platelet adhesion to the damaged vascular wall, while TF activates the extrinsic coagulation pathway, leading to the activation of FX and the subsequent formation of thrombus. These mechanisms enable ECs to promote thrombosis, which is essential for preventing excessive bleeding. However, uncontrolled activation of this process can result in thrombotic disorders.62,63 Conversely, ECs regulate coagulation through negative feedback mechanisms to maintain blood flow stability and prevent unnecessary clotting.64 Under normal physiological conditions, ECs secrete anti-coagulant substances, such as NO and prostacyclin (PGI2), which dilate blood vessels and inhibit platelet activation, ensuring smooth blood flow.65 NO activates soluble guanylate cyclase (sGC), increasing intracellular cyclic Guanosine Monophosphate (cGMP) levels, which in turn inhibits platelet aggregation.66 ECs also express TM on their surface, which binds to thrombin and promotes the activation of protein C, resulting in the formation of APC.67 APC then degrades coagulation FVa and FVIIIa, thereby inhibiting the coagulation cascade.68 In addition, ECs secrete tissue-type plasminogen activator (tPA),69 further inhibiting coagulation factor activation and promoting fibrinolysis, thus preventing coagulation abnormalities.70 ECs prevent excessive coagulation under normal physiological conditions by these mechanisms, ensuring the stability of blood flow.
In sepsis, LPS stimulates ECs, exposing TF expression and triggering coagulation.71 The reduction in nitric oxide synthesis and promotion of endothelin-1 expression further contribute to the formation of thrombosis.72 The endothelial glycocalyx plays a crucial role in these processes,73 acting as a barrier against vascular permeability and allowing adhesion molecules to protect against oxidative stress (Figure 1).74,75 Damage to the glycocalyx during sepsis leads to increased vascular permeability and edema and facilitates the adhesion of leukocytes and platelets, promoting immune and coagulation responses.76,77 Glycocalyx injury is progressive, illustrating the worsening of microcirculatory dysfunction in DIC,9 even after fluid resuscitation and hemodynamic stabilization. There is variability in endothelial glycocalyx thickness across different vascular locations and conditions. The limitations of current technology in measuring its thickness make directly assessing glycocalyx damage challenging. Hypothetically, assessing glycocalyx degradation through circulating component levels could offer diagnostic and prognostic insights, although the reliability of this approach requires further validation.76,78
Coagulation Dysfunction Caused by Immune Disorders
The immune system activates the NF-κB and MAPK pathways via PRRs and DAMPs to detect pathogens and initiate the release of inflammatory cytokines such as IL-1, IL-6, and TNF-α, recruiting immune cells such as neutrophils and macrophages and triggering innate immunity.79 Neutrophils, as the primary defense cells involved in innate immunity, are recruited by activated ECs during infection and inflammation. However, in the dynamic context of blood flow, neutrophils release neutrophil extracellular traps (NETs) to increase pathogen capture.80 The systemic inflammatory response in sepsis can lead to hyperactivation of neutrophils, prolonging their lifespan and inhibiting their migration. Confined within the vasculature, neutrophils can release NETs, which not only activate ECs but also directly damage the endothelial glycocalyx, exacerbating vascular inflammation and thrombosis.81,82
Formation of NETs
NETs formation involves multiple processes (Figure 3). Upon stimulation by inflammation or pathogens, the Nncotinamide ndenine dinucleotide phosphate hydrogen (NADPH) oxidase within neutrophils is activated, leading to the generation of reactive oxygen species (ROS). These ROS can activate signaling pathways such as protein kinase C (PKC), which amplifies the signal and further activates MAPK pathways, including p38 and ERK, to mediate chromatin decondensation. Myeloperoxidase (MPO), neutrophil elastase, and cathepsins can facilitate nuclear membrane degradation. Once chromatin decondensation and nuclear membrane rupture occur, neutrophils release their DNA and associated proteins. NETs are formed in the cytoplasm through vesicular transport, without the occurrence of membrane rupture.83,84 Furthermore, depending on the type of pathogen component present, various Toll-like receptors on neutrophil surfaces can be activated, leading to the activation of the NF-κB and MAPK signaling pathways. This process initiates neutrophil activation and culminates in NETs formation. For example, Toll-Like Receptor 2 (TLR2) recognizes bacterial lipoproteins and peptidoglycans. TLR4 is the primary receptor for detecting LPS, and Toll-Like Receptor 9 (TLR9) can detect cytosine-phosphate-guanine motifs (CpG motifs) within bacterial and viral DNA.84–86 NETs not only participate in the body’s immune response but also interact with the coagulation system. MPO and histones within NETs can activate G protein-coupled receptors (GPCRs) on the surfaces of platelets, leading to the release of platelet factor 4 (PF4) and platelet-derived growth factor (PDGF), thereby promoting platelet aggregation and thrombosis. Moreover, platelets are also significant inducers of NETs formation. PF4 and PDGF released by activated platelets can bind to receptors on the surface of neutrophils, activating the MAPK and NF-κB signaling pathways via the PKC, Rat Sarcoma (Ras), and PI3K/Akt pathways, respectively, ultimately contributing to the formation of NETs.77,83,87,88 During the formation of NETs and through the catalytic action of MPO, a significant amount of ROS are produced. These ROS can affect the stability of coagulation factors (such as FV, FVII, and FVIII) through oxidative modifications, thereby influencing the coagulation process.89,90 Research by Silva and Wanderley suggested that the caspase-11/GSDMD pathway can induce NETs formation in sepsis patients. The inhibitor disulfiram may reduce NETs release, potentially mitigating organ damage. However, owing to contradictions involving immunopathology, further research is necessary.91 Mesenchymal stem cells with apoptotic vesicles can promote NETs formation, increasing survival rates in septic mice.92 Inhibitors of TLR4/Triggering Receptor Expressed on Myeloid Cells 1 (TREM1) signaling can alleviate NETs formation and mitigate its deleterious effects.93 Monocytes and macrophages secrete cytokines such as Interleukin-10 (IL-10) and transforming growth factor-β (TGF-β). The anti-inflammatory molecule indoleamine 2.3-dioxygenase (IDO) can regulate immunity and prevent excessive inflammation. The expression of TF and various proinflammatory factors is involved in the activation of the extrinsic coagulation pathway and thrombin generation.94
In adaptive immunity, lymphocytes enhance inflammation and coagulation by producing cytokines. B lymphocytes form immune complexes with antigens produced by pathogens, activating the complement system to promote coagulation by expressing FVII and FVIII, which directly participate in the coagulation cascade. PDGF released by T lymphocytes enhances platelet function, whereas interferon-γ (IFN-γ) produced by T cells inhibits TF expression on ECs, promoting anticoagulation.94
During sepsis, destruction of immune homeostasis leads to immune cell dysfunction characterized by excessive inflammation and immune suppression.8,55 Immune cell exhaustion, dysregulation of anti-inflammatory mechanisms, and a reduction in human leukocyte antigen-DR (HLA-DR) expression and that of other factors can induce immune suppression.95 Within this context, immune cells can protect or eliminate pathogens via “suicidal” methods such as apoptosis, pyroptosis, autophagy, and release.96 The release of substances such as histones, nucleosomes, proteases, and tissue factors from many dead immune cells significantly promotes inflammation and endothelial damage and facilitates coagulation.97
Mitochondrial Damage Induces Coagulation Abnormalities
Mitochondria are essential for maintaining cellular homeostasis and function. As the primary energy producers of the cell, they generate adenosine triphosphate (ATP) through oxidative phosphorylation, supporting cellular metabolism. Additionally, mitochondria regulate intracellular calcium levels, which are critical for various cellular processes, particularly blood coagulation. Calcium ions play a central role in converting prothrombin to thrombin, a key step in the coagulation cascade. Furthermore, mitochondria produce ROS under normal physiological conditions, which act as key signaling molecules involved in these processes such as cell growth, differentiation, and stress responses.36,98
In sepsis, however, the inflammatory response leads to the release of pro-inflammatory cytokines, which exacerbate oxidative stress and increase ROS production. These ROS cause damage to mitochondrial membrane and protein, impairing the mitochondrial electron transport chain (ETC), and reduce ATP production, compromising cellular energy.99 The accumulation of ROS also results in elevated intracellular calcium levels, further activating coagulation. Excessive calcium ions enhance the activity of prothrombinase complexes and platelets, and increase the activity of coagulation FV and FVIII, which intensify coagulation.100 Moreover, mitochondrial damage alters cellular metabolism, particularly by upregulating glycolysis, which leads to lactate accumulation and decrease blood pH. The resulting acidic environment further activates coagulation factors and enhances platelet aggregation, exacerbating coagulation abnormalities.101 Research by Zou in 2022 further revealed that DNA-dependent protein kinase catalytic subunit (DNA-PKcs) signaling exacerbates liver dysfunction by inducing mitochondrial impairment, consequently affecting the production of coagulation factors.102
Neuroendocrine Network Disorders Induce Coagulation Abnormalities
The neuroendocrine system regulates various physiological functions, through the interaction of hormones, neurotransmitters, and their receptors. Neuroendocrine factors, particularly norepinephrine (NE), cortisol, and adrenaline, play a pivotal role in stress response.103 Norepinephrine activates platelets and promotes platelet aggregation by binding to β-adrenergic receptors.104 Cortisol further enhances coagulation by upregulating the synthesis of coagulation FVII and fibrinogen, thereby promoting thrombus formation. Adrenaline indirectly exacerbates the coagulation response through vasoconstriction and increased blood flow velocity.105,106 Additionally, neuroendocrine hormones can activate NF-κB and MAPK signaling pathways in ECs, triggering the release of pro-coagulant factors and inflammatory cytokines. These cytokines not only drive the coagulation cascade but also amplify coagulation by altering vascular permeability and platelet activity.107
In sepsis, neuroendocrine dysregulation exacerbates coagulation by activating the sympathetic nervous system, resulting in vasoconstriction and altered blood flow velocity, which further aggravates coagulation.108 On the other hand, infection-or injury-associated molecules transmit signals through the vagus nerve, stimulating T cells to release acetylcholine. Acetylcholine binds to α7-nicotinic acetylcholine receptors (α7nAChR) on immune cells, thereby suppressing cytokine release from macrophages.109 Simultaneously, inflammatory mediators and damage-associated molecules interact with the central nervous system (CNS), altering the function of the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB), which triggers sterile neuroinflammation.110,111 Activation of glial cells leads to neuronal injury and suppression of the vagus nerve, further intensifying the inflammatory response in sepsis. Stimulation of the vagus nerve and activation of the cholinergic anti-inflammatory pathway can mitigate the release of pro-inflammatory factors, thereby reducing thrombus formation.112,113
ER Stress Induces Coagulation Abnormalities
ER is integral to the modification and transport of proteins and regulates numerous cellular pathways and physiological activities, including protein synthesis, protein quality control, and lipid synthesis. When the function of one or more ERs is dysregulated and saturated, the ER enters a state of stress, which in turn activates the highly conserved unfolded protein response (UPR). The UPR triggers pathways to restore ER homeostasis by sensing the accumulation of unfolded proteins or lipid bilayer stress on the ER. If stress remains unresolved, it ultimately induces apoptosis.114 During apoptosis, substances such as phosphatidylserine (PS) are released and serve as substrates for the coagulation reaction, promoting the generation of thrombin and the formation of thrombi.115
The UPR involves three main ER membrane protein sensors: inositol-requiring enzyme 1 (IRE1), PKR-like ER kinase (PERK), and activating transcription factor 6 (ATF6). These sensors are activated when unfolded or misfolded proteins accumulate, thereby initiating a series of intracellular signaling events. During sepsis, ER stress activates the UPR. If stress exceeds the compensatory capacity of the UPR, it accelerates calcium ion release, activates the caspase cascade, and eventually leads to apoptosis or necrosis. Additionally, the activation of the UPR is involved in inflammation and angiogenesis.114,116 Professor Zhang reported that transmembrane protein 173 (TMEM173) is an ER-associated membrane protein that mediates ER stress and can participate in the release of TF, a key initiator of the coagulation cascade, thereby activating coagulation.117
Microthrombus Formation in Sepsis
In sepsis, the activation of coagulation mechanisms results in the formation of microthrombi (Figure 4), which impair blood flow within the microcirculation, obstructing the supply to vital organs such as the brain, kidneys, and lungs. This disruption contributes to significant hemodynamic changes, typically marked by reducing cardiac output and blood pressure, which often progress to shock.118,119
During the initial stage of septic shock, inflammatory mediators in the circulation can lead to damage to vascular ECs, activation of coagulation, and increased capillary permeability, resulting in the leakage of fluid and proteins. The stimulation of inflammatory mediators also causes the dilation of both precapillary and postcapillary sphincters, which reduces the resistance of the capillary bed. The sympathetic‒adrenal axis is activated, and catecholamines cause systemic vasoconstriction, especially in the skin, kidneys, and gastrointestinal tract, to ensure blood supply to vital organs. At this point, tissues can still receive sufficient perfusion; therefore, hemodynamic changes are not significant.120–122
As the condition worsens, the release of numerous inflammatory mediators results in the impairment and potential death of vascular ECs, significantly increasing capillary permeability and leading to the leakage of fluid and proteins, which in turn causes severe tissue edema. The reduction in blood volume within the microcirculation, coupled with hyperactivation of the sympathetic‒adrenal axis, leads to the release of substantial amounts of catecholamine hormones. These hormones cause capillaries, particularly precapillary sphincters, to contract strongly, resulting in blood retention within the microcirculation. This exacerbates capillary hydrostatic pressure and permeability, facilitating the formation of numerous microthrombi and ultimately causing a continuous decline in effective blood flow.123–125
In the irreversible microcirculatory failure phase, persistent hypoxia and inflammation contribute to the death of a significant number of ECs, causing the microcirculation to lose its ability to self-regulate. Changes in intracellular calcium concentrations may exacerbate the constriction of both the precapillary and postcapillary sphincters. At this critical juncture, the microcirculatory blood volume is severely depleted, leading to critically inadequate tissue perfusion. Following the sustained activation of the sympathetic‒adrenal axis, the vascular responsiveness to catecholamines diminishes, resulting in a further drop in blood pressure. Blood flow becomes extensively trapped within the microcirculation, the coagulation system becomes hyperactive, fibrinolysis is inhibited, and DIC occurs, ultimately resulting in severe tissue hypoxia and organ failure.10,126–129
Treatment of Coagulation Dysfunction Caused by Sepsis
Early and accurate diagnosis of SIC is crucial. Initially, patients exhibit elevated thrombin levels and paradoxical reductions in platelet counts due to activation and aggregation, presenting a procoagulant state that leads to microvascular thrombosis and subsequent tissue ischemia. During this period, the balance between the production and consumption of coagulation factors and platelets is maintained. Clinical symptoms may not be evident, making laboratory results challenging and potentially leading to overdiagnoses. Combining SIC with thrombin-related marker testing may improve diagnostic accuracy12 but requires distinguishing it from noninfectious conditions, a common challenge with other DIC diagnostic methods.18
As the disease progresses, the consumption of coagulation factors and platelets surpasses their production. Low coagulation activity, such as bleeding symptoms, can occur. When coagulation factors and platelets are depleted and fibrinolysis becomes excessive, significant bleeding or even shock may occur. Therefore, patients with severe infection, thrombotic and/or bleeding symptoms, and laboratory test abnormalities indicate coagulopathy, and any organ dysfunction should be suspected of suffering DIC.130
TF Pathway Inhibitor (TFPI)
Currently, no drugs have been identified that can directly inhibit TF activation. However, TFPI is the primary inhibitor of the tissue factor-mediated extrinsic coagulation pathway and is the most critical and specific endogenous anticoagulant protein.131,132 It belongs to the Kunitz-type serine protease inhibitor family and possesses three consecutive functional domains: K1, K2, and K3. Primarily secreted by vascular ECs, TFPI can directly bind to FXa to inhibit its activity. The FXa-TFPI complex, through its K1 domain, binds to the active site of FVIIa within the FVIIa-TF complex, thereby inhibiting the FVIIa-TF complex133 and fundamentally preventing the activation of the extrinsic coagulation pathway.134 Furthermore, TFPI-related medications have concluded clinical trials in recent years and obtained regulatory approval for market release in 2024.
Endothelial Biomarkers and Endothelial Pathways
Therapeutic strategies targeting endothelial biomarkers and pathways focus on microRNAs (miRs), microparticles (MPs), and pharmacological interventions.9 miRs such as miR-146,135,136 miR-181,137 and miR-155138 could reduce endothelial activation and inflammation by inhibiting the NF-κB and MAPK pathways and by regulating adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1) and endothelial-leukocyte adhesion molecule 1 (E-selectin).139 miRs like miR-150135 and miR-147b could help maintain endothelial barrier function by modulating Angiopoietin-2 (Ang2) and A Disintegrin And Metalloproteinase Domain 15 (ADAM15). Additionally, miRs such as miR-30b and miR-181b137 regulate PAI-1 levels, thereby reducing endothelial injury during sepsis. MPs derived from ECs serve as biomarkers of damage and are involved in coagulation and inflammation during sepsis and DIC.9 Complement inhibitors such as eculizumab show promise in diseases characterized by complement activation. Pharmacological treatments, including statins, vitamin C, and anticoagulants, protect the endothelium by reducing oxidative stress, while agents like APC, recombinant TM, and Ang-1 have shown potential in restoring endothelial function in animal models.10,140 These strategies offer hope for treating sepsis and related diseases.
Inhibitors of NETs
In the treatment of coagulopathy caused by sepsis, intervention strategies targeting NETs have become an important area of research. Protein arginine deiminase 4 (PAD4) is a calcium-dependent enzyme that mediates the citrullination of histones. This chemical modification weakens the binding between histones and DNA, leading to chromatin loosening and decondensation, which could lead to NETs release. PAD4 inhibitors effectively suppress NETs formation through multiple mechanisms.141,142 First, they block histone citrullination, preventing chromatin decondensation and thus inhibiting NETs release. Secondly, PAD4 inhibitors indirectly reduce ROS production, attenuating oxidative stress-mediated activation of neutrophils, further suppressing NETs formation. Moreover, by restricting chromatin decondensation, PAD4 inhibitors reduce the space required for degranulation, leading to decreased release of enzymes such as elastase and myeloperoxidase, which in turn inhibits NETs formation.143,144
Another therapeutic strategy involves Toll-like receptor (TLR) inhibitors. These compounds block the signaling pathways that induce NETs formation, thereby reducing NETs release and suppressing inflammatory responses and pro-coagulant activities.10
Furthermore, vitamin D (Vit D) exerts inhibitory effects on NETs formation through several mechanisms. By binding to the vit D receptor (VDR) on neutrophils, Vit D inhibits their activation. It also downregulates the expression of ROS-producing enzymes such as NADPH oxidase, thereby inhibiting ROS-dependent formation of NETs.145 Additionally, Vit D suppresses the activity of the NF-κB signaling pathway, reducing the expression of pro-inflammatory cytokines146 while upregulating anti-inflammatory cytokines (such as IL-10). It also modulates the antioxidant enzyme system, enhancing cellular defense against oxidative stress, and further suppressing NET formation.147,148
Conventional Treatment
Aggressive treatment of the underlying condition and prompt administration of antibiotics and anticoagulants are pivotal in managing DIC. Although heparin has been traditionally used in DIC treatment, clinical trials have shown inconsistent results regarding its efficacy. In addition, its mechanism remains somewhat unclear.149,150 Antiplatelet drugs such as aspirin, clopidogrel, and ticagrelor may also reduce mortality events, but similar to heparin, their clinical efficacy needs further study.151 Additionally, research on fibrinolytic activator preparations has shown some potential.152
When DIC patients present with bleeding, supplementation of platelets and coagulation factors, along with adequate anticoagulation, is necessary. Fibrinolytic inhibitors, which can impede the resolution of microthrombi and may lead to irreversible kidney damage, should be used with caution. Antagonists targeting the aforementioned mechanisms could theoretically mitigate the induction and progression of DIC to a certain extent, but their effectiveness remains to be confirmed.20 Maintaining a perioperative oxygen partial pressure (PaO2) between 97–339 mmHg, a PaO2/ Fraction of inspiration O2 (FiO2) between 189–619, and a Pulse Oximeter Oxygen Saturation (SpO2) ≥ 93% can effectively prevent the occurrence of sepsis-related brain injury.153
The optimal strategy for treating DIC involves early diagnosis, timely intervention, and dynamic continuous management. Clinicians should assess patients’ disease progression accurately and initiate antibiotic and anticoagulant treatment as early as possible. It is prudent to select medications that have a relatively minor impact on coagulation function to avoid exacerbating the patient’s coagulopathy. Employing crystalloids, component transfusions, and vasoactive drugs to maintain stable vital signs in patients can minimize the risk of thrombus detachment due to blood pressure fluctuations.
Conclusion
Coagulopathy is a common complication of sepsis and strongly correlates with mortality. The pathogenesis of sepsis involves interconnected and interdependent events that collectively exacerbate coagulopathy. Inflammatory processes activate ECs and platelets, enhancing coagulation, while the activation of the coagulation system further promotes inflammation. The immune system also contributes to coagulation through various mechanisms, including increased inflammation, the release of NETs, and immune cell death. Moreover, mitochondrial damage, neuroendocrine network disruption, and ER stress initiate different pathways that induce coagulation. Additionally, disruption of the endothelial glycocalyx, release of NETs, and activation of TF are critical in the development of SIC. The complexities of infection, surgical stress, and shock pose significant challenges in managing sepsis-induced coagulopathy. We advocate the use of the SIC assessment system as a basis for the early diagnosis of DIC and recommend dynamic treatment strategies tailored to the various stages of DIC progression. Clinicians must be vigilant and prepared to ensure patient safety under these challenging conditions.
Abbreviations
ADAM15, A Disintegrin and Metalloproteinase Domain 15; ADP, Adenosine Diphosphate; Akt, Protein Kinase B; Ang2, Angiopoietin-2; APC, Activated Protein C; AT, Antithrombin; ATF6, Activating Transcription Factor 6; ATP, Adenosine Triphosphate; BBB, Blood-Brain Barrier; BCSFB, Blood-Cerebrospinal Fluid Barrier; C3a, Complement Component 3a; C5a, Complement Component 5a; cGMP, cyclic Guanosine Monophosphate; CNS, Central Nervous System; CpG motifs, Cytosine-Phosphate-Guanine motifs; DAMPs, Damage-Associated Molecular Patterns; DIC, Disseminated Intravascular Coagulation; DNA-PKcs, DNA-dependent protein kinase catalytic subunit; ECs, Endothelial Cells; ER, Endoplasmic Reticulum; ERK1/2, Extracellular Signal-Regulated Kinase 1/2; E-selectin, Endothelial-leukocyte adhesion molecule 1; ETC, Electron Transport Chain; FiO2, Fraction of inspiration O2; FIX, Factor IX; FVa, Factor Va; FVII, Factor VII; FVIIa, Factor VIIa; FVIII, Factor VIII; FVIIIa, Factor VIII; FX, Factor X; FXa, Factor Xa; FXI, Factor XI; FXII, Factor XII; GPCRs, G protein-coupled receptors; GPIb-IX-V, Glycoprotein Ib-IX-V Complex; GSDMD, Gasdermin D; HLA-DR, Human Leukocyte Antigen-DR; HMGB1, High Mobility Group Box 1 Protein; IDO, Indoleamine 2.3-Dioxygenase; IFN-γ, Interferon-γ; IL-1, Interleukin-1; IL-10, Interleukin-10; IL-6, Interleukin-6; IL-8, Interleukin-8; INR, International Normalized Ratio; IRF3, Interferon Regulatory Factor 3; ISTH, International Society on Thrombosis and Haemostasis; JNK, C-Jun N-terminal Kinase; LPS, Lipopolysaccharide; MAC, Membrane Attack Complex; MAPK, Mitogen-Activated Protein Kinase; miRs, MmicroRNAs; MPO, Myeloperoxidase; MPs, Microparticles; MyD88, Myeloid Differentiation Primary Response Gene 88; NADPH, Nicotinamide Adenine Dinucleotide Phosphate Hydrogen; NE, Norepinephrine; NETs, Neutrophil Extracellular Traps; NF-κB, Nuclear Factor kappa-light-chain-enhancer of activated B cells; NLRP3, NOD-, LRR-, and pyrin domain-containing protein 3; NO, Nitric Oxide; p38, p38 Mitogen-Activated Protein Kinase; PAD4, Protein arginine deiminase 4; PAF, Platelet-Activating Factor; PAMPs, Pathogen-Associated Molecular Patterns; PaO2, Perioperative oxygen partial pressure; PAR-1, Proteinase-Activated Receptor-1; PDGF, Platelet-Derived Growth Factor; PERK, PKR-like ER kinase; PF4, Platelet Factor 4; PF4, Platelet Factor 4; PGI2, Prostacyclin; PI3K, Phosphoinositide 3-Kinase; PKC, Protein Kinase C; PRRs, Pattern Recognition Receptors; PS, Phosphatidylserine; PSGL-1, P-selectin glycoprotein ligand 1; RAGE, Receptor for Advanced Glycation End Products; Ras, Rat Sarcoma; ROS, Reactive Oxygen Species; sGC, soluble Guanylate Cyclase; SIC, Sepsis-Induced Coagulopathy; SIRS, Systemic inflammatory response syndrome; SpO2, Pulse Oximeter Oxygen Saturation; TF, Tissue Factor; TFPI, TF Pathway Inhibitor; TGF-β, Transforming Growth Factor-β; TIR, Toll/Interleukin-1 Receptor; TLR, Toll-like receptor; TLR2, Toll-Like Receptor 2; TLR4, Toll-Like Receptor 4; TLR9, Toll-Like Receptor 9; TM, Thrombomodulin; TMEM173, Transmembrane protein 173; TNF-α, Tumor Necrosis Factor-alpha; tPA, Tissue-type Plasminogen Activator; TREM1, Triggering Receptor Expressed on Myeloid Cells 1; TRIF, TIR Domain-Containing Adapter Inducing IFN-β; TXA2, Thromboxane A2; UPR, Unfolded Protein Response; VCAM-1, Vascular Cell Adhesion Molecule-1; VDR, Vit D receptor; Vit D, Vitamin D; vWF, von Willebrand factor; α7nAChR, α7-nicotinic Acetylcholine receptors.
Acknowledgments
All figures were edited using the FIGDRAW 2.0 software. We would like to express our sincere gratitude to the online scientific illustration platform Figdraw. Natural Science Foundation of Shandong Province (ZR2022QH217).
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Fleischmann-Struzek C, Mellhammar L, Rose N, et al. Incidence and mortality of hospital- and ICU-treated sepsis: results from an updated and expanded systematic review and meta-analysis. Intensive Care Med. 2020;46(8):1552–1562. doi:10.1007/s00134-020-06151-x
2. Rudd KE, Johnson SC, Agesa KM, et al. Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the global burden of disease study. Lancet. 2020;395(10219):200–211. doi:10.1016/S0140-6736(19)32989-7
3. Xie J, Wang H, Kang Y, et al. The epidemiology of sepsis in Chinese ICUs: a national cross-sectional survey. Crit Care Med. 2020;48(3):e209–e218. doi:10.1097/CCM.0000000000004155
4. Singer M, Deutschman CS, Seymour CW, et al. The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. 2016;315(8):801–810. doi:10.1001/jama.2016.0287
5. Schmoch T, Möhnle P, Weigand MA, et al. The prevalence of sepsis-induced coagulopathy in patients with sepsis - a secondary analysis of two German multicenter randomized controlled trials. Ann Intens Care. 2023;13(1):3. doi:10.1186/s13613-022-01093-7
6. Maneta E, Aivalioti E, Tual-Chalot S, et al. Endothelial dysfunction and immunothrombosis in sepsis. Front Immunol. 2023;14:1144229. doi:10.3389/fimmu.2023.1144229
7. Bode C, Weis S, Sauer A, Wendel-Garcia P, David S. Targeting the host response in sepsis: current approaches and future evidence. Crit Carelondon. 2023;27(1):478. doi:10.1186/s13054-023-04762-6
8. van der Poll T, Shankar-Hari M, Wiersinga WJ. The immunology of sepsis. Immunity. 2021;54(11):2450–2464. doi:10.1016/j.immuni.2021.10.012
9. Dolmatova EV, Wang K, Mandavilli R, Griendling KK. The effects of sepsis on endothelium and clinical implications. Cardiovasc Res. 2021;117(1):60–73. doi:10.1093/cvr/cvaa070
10. Joffre J, Hellman J, Ince C, Ait-Oufella H. Endothelial responses in sepsis. Am J Respir Crit Care Med. 2020;202(3):361–370. doi:10.1164/rccm.201910-1911TR
11. Raia L, Zafrani L. Endothelial activation and microcirculatory disorders in sepsis. Front Med. 2022;9:907992. doi:10.3389/fmed.2022.907992
12. Tsantes AG, Parastatidou S, Tsantes EA, et al. Sepsis-induced coagulopathy: an update on pathophysiology, biomarkers, and current guidelines. Life. 2023;13(2):350. doi:10.3390/life13020350
13. Squizzato A, Gallo A, Levi M, et al. Underlying disorders of disseminated intravascular coagulation: communication from the ISTH SSC subcommittees on disseminated intravascular coagulation and perioperative and critical care thrombosis and hemostasis. J Thromb Haemost. 2020;18(9):2400–2407. doi:10.1111/jth.14946
14. Di Nisio M, Thachil J, Squizzato A. Management of disseminated intravascular coagulation: a survey of the international society on thrombosis and haemostasis. Thromb Res. 2015;136(2):239–242. doi:10.1016/j.thromres.2015.05.022
15. Iba T, Nisio MD, Levy JH, Kitamura N, Thachil J. New criteria for sepsis-induced coagulopathy (SIC) following the revised sepsis definition: a retrospective analysis of a nationwide survey. BMJ open. 2017;7(9):e017046. doi:10.1136/bmjopen-2017-017046
16. Iba T, Levy JH, Yamakawa K, Thachil J, Warkentin TE, Levi M. Proposal of a two-step process for the diagnosis of sepsis-induced disseminated intravascular coagulation. J Thromb Haemost. 2019;17(8):1265–1268. doi:10.1111/jth.14482
17. Iba T, Levy JH, Warkentin TE, Thachil J, van der Poll T, Levi M. Diagnosis and management of sepsis-induced coagulopathy and disseminated intravascular coagulation. J Thromb Haemost. 2019;17(11):1989–1994. doi:10.1111/jth.14578
18. Iba T, Levi M, Thachil J, Helms J, Scarlatescu E, Levy JH. Communication from the scientific and standardization committee of the international society on thrombosis and haemostasis on sepsis-induced coagulopathy in the management of sepsis. J Thromb Haemost. 2023;21(1):145–153. doi:10.1016/j.jtha.2022.10.022
19. Giustozzi M, Ehrlinder H, Bongiovanni D, et al. Coagulopathy and sepsis: pathophysiology, clinical manifestations and treatment. Blood Rev. 2021;50:100864. doi:10.1016/j.blre.2021.100864
20. Huang M, Cai S, Su J. The pathogenesis of sepsis and potential therapeutic targets. Int J mol Sci. 2019;20(21):5376. doi:10.3390/ijms20215376
21. Reininger AJ. Function of von Willebrand factor in haemostasis and thrombosis. Haemophilia. 2008;14(Suppl 5):11–26. doi:10.1111/j.1365-2516.2008.01848.x
22. Harrison P, Cramer EM. Platelet alpha-granules. Blood Rev. 1993;7(1):52–62. doi:10.1016/0268-960X(93)90024-X
23. Rodrigues M, Kosaric N, Bonham CA, Gurtner GC. Wound Healing: a Cellular Perspective. Physiol Rev. 2019;99(1):665–706. doi:10.1152/physrev.00067.2017
24. Bloom SI, Islam MT, Lesniewski LA, Donato AJ. Mechanisms and consequences of endothelial cell senescence. Nat Rev Cardiology. 2023;20(1):38–51. doi:10.1038/s41569-022-00739-0
25. Geng Y, Verhamme IM, Smith SB, et al. The dimeric structure of factor XI and zymogen activation. Blood. 2013;121(19):3962–3969. doi:10.1182/blood-2012-12-473629
26. Maas C, Meijers JC, Marquart JA, et al. Activated factor V is a cofactor for the activation of factor XI by thrombin in plasma. Proc Natl Acad Sci USA. 2010;107(20):9083–9087. doi:10.1073/pnas.1004741107
27. Sang Y, Roest M, de Laat B, de Groot PG, Huskens D. Interplay between platelets and coagulation. Blood Rev. 2021;46:100733. doi:10.1016/j.blre.2020.100733
28. Grover SP, Mackman N. Intrinsic Pathway of Coagulation and Thrombosis. Arteriosclerosis Thrombosis Vasc Biol. 2019;39(3):331–338. doi:10.1161/ATVBAHA.118.312130
29. Smith SA, Mutch NJ, Baskar D, Rohloff P, Docampo R, Morrissey JH. Polyphosphate modulates blood coagulation and fibrinolysis. Proc Natl Acad Sci USA. 2006;103(4):903–908. doi:10.1073/pnas.0507195103
30. van der Meijden PE, Munnix IC, Auger JM, et al. Dual role of collagen in factor XII-dependent thrombus formation. Blood. 2009;114(4):881–890. doi:10.1182/blood-2008-07-171066
31. Li X, Sim MMS, Wood JP. Recent insights into the regulation of coagulation and thrombosis. Arteriosclerosis Thrombosis Vasc Biol. 2020;40(5):e119–e125. doi:10.1161/ATVBAHA.120.312674
32. Deng M, Tang Y, Li W, et al. The endotoxin delivery protein HMGB1 mediates caspase-11-dependent lethality in sepsis. Immunity. 2018;49(4):740–753.e7. doi:10.1016/j.immuni.2018.08.016
33. Yang X, Cheng X, Tang Y, et al. Bacterial endotoxin activates the coagulation cascade through gasdermin D-dependent phosphatidylserine exposure. Immunity. 2019;51(6):983–996.e6. doi:10.1016/j.immuni.2019.11.005
34. Rathinam VAK, Zhao Y, Shao F. Innate immunity to intracellular LPS. Nat Immunol. 2019;20(5):527–533. doi:10.1038/s41590-019-0368-3
35. Cecconi M, Evans L, Levy M, Rhodes A. Sepsis and septic shock. Lancet. 2018;392(10141):75–87. doi:10.1016/S0140-6736(18)30696-2
36. Wu R, Wang N, Comish PB, Tang D, Kang R. Inflammasome-dependent coagulation activation in sepsis. Front Immunol. 2021;12:641750. doi:10.3389/fimmu.2021.641750
37. Sachetto ATA, Mackman N. Monocyte tissue factor expression: lipopolysaccharide induction and roles in pathological activation of coagulation. Thromb Hemost. 2023;123(11):1017–1033. doi:10.1055/a-2091-7006
38. Schlömmer C, Brandtner A, Bachler M. Antithrombin and its role in host defense and inflammation. Int J mol Sci. 2021;22(8):4283.
39. Rezaie AR, Giri H. Anticoagulant and signaling functions of antithrombin. J Thromb Haemost. 2020;18(12):3142–3153. doi:10.1111/jth.15052
40. Iba T, Maier CL, Tanigawa T, Levy JH. Risk stratification utilizing sequential organ failure assessment (SOFA) score, antithrombin activity, and demographic data in sepsis-associated disseminated intravascular coagulation (DIC). Sci Rep. 2023;13(1):22502. doi:10.1038/s41598-023-49855-y
41. Levy JH, Sniecinski RM, Welsby IJ, Levi M. Antithrombin: anti-inflammatory properties and clinical applications. Thromb Hemost. 2016;115(4):712–728. doi:10.1160/TH15-08-0687
42. Wiedermann CJ. Clinical review: molecular mechanisms underlying the role of antithrombin in sepsis. Critical Care. 2006;10(1):209. doi:10.1186/cc4822
43. Oto J, Fernández-Pardo Á, Miralles M, et al. Activated protein C assays: a review. Clinica Chimica Acta. 2020;502:227–232. doi:10.1016/j.cca.2019.11.005
44. Stojanovski BM, Pelc LA, Zuo X, Di Cera E. Zymogen and activated protein C have similar structural architecture. J Biol Chem. 2020;295(45):15236–15244. doi:10.1074/jbc.RA120.014789
45. Wilhelm AR, Parsons NA, Samelson-Jones BJ, et al. Activated protein C has a regulatory role in factor VIII function. Blood. 2021;137(18):2532–2543. doi:10.1182/blood.2020007562
46. Li X, Song X, Mahmood DFD, Sim MMS, Bidarian SJ, Wood JP. Activated protein C, protein S, and tissue factor pathway inhibitor cooperate to inhibit thrombin activation. Thromb Res. 2023;230:84–93. doi:10.1016/j.thromres.2023.08.012
47. Kohli S, Shahzad K, Jouppila A, Holthöfer H, Isermann B, Lassila R. Thrombosis and inflammation-A dynamic interplay and the role of glycosaminoglycans and activated protein C. Front Cardiovas Med. 2022;9:866751. doi:10.3389/fcvm.2022.866751
48. Guo H, Liu D, Gelbard H, et al. Activated protein C prevents neuronal apoptosis via protease activated receptors 1 and 3. Neuron. 2023;111(24):4116. doi:10.1016/j.neuron.2023.12.001
49. Petros S, Siegemund R, Siegemund A. Increased activated protein C to protein C ratio in sepsis and cirrhosis. Thromb Res. 2021;202:74–76. doi:10.1016/j.thromres.2021.03.017
50. Shorr AF, Bernard GR, Dhainaut JF, et al. Protein C concentrations in severe sepsis: an early directional change in plasma levels predicts outcome. Critical Care. 2006;10(3):R92. doi:10.1186/cc4946
51. Eliwan H, Omer M, McKenna E, et al. Protein C pathway in paediatric and neonatal sepsis. Front Pediatr. 2021;9:562495. doi:10.3389/fped.2021.562495
52. Bone RC, Balk RA, Cerra FB, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. the ACCP/SCCM consensus conference committee. American college of chest physicians/society of critical care medicine. Chest. 1992;101(6):1644–1655. doi:10.1378/chest.101.6.1644
53. American College of chest Physicians/Society of Critical Care Medicine Consensus Conference. definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Crit Care Med. 1992;20(6):864–874. doi:10.1097/00003246-199206000-00025
54. Vincent JL. Dear SIRS, I’m sorry to say that I don’t like you. Crit Care Med. 1997;25(2):372–374. doi:10.1097/00003246-199702000-00029
55. van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17(7):407–420. doi:10.1038/nri.2017.36
56. Zindel J, Kubes P. DAMPs, PAMPs, and LAMPs in immunity and sterile inflammation. Annu Rev Pathol. 2020;15:493–518. doi:10.1146/annurev-pathmechdis-012419-032847
57. de Nooijer AH, Kotsaki A, Kranidioti E, et al. Complement activation in severely ill patients with sepsis: no relationship with inflammation and disease severity. Critical Care. 2023;27(1):63. doi:10.1186/s13054-023-04344-6
58. Rawish E, Sauter M, Sauter R, Nording H, Langer HF. Complement, inflammation and thrombosis. Br J Pharmacol. 2021;178(14):2892–2904. doi:10.1111/bph.15476
59. Noris M, Galbusera M. The complement alternative pathway and hemostasis. Immunol Rev. 2023;313(1):139–161. doi:10.1111/imr.13150
60. Mandel J, Casari M, Stepanyan M, Martyanov A, Deppermann C. Beyond hemostasis: platelet innate immune interactions and thromboinflammation. Int J mol Sci. 2022;23(7). doi:10.3390/ijms23073868
61. Futosi K, Fodor S, Mócsai A. Neutrophil cell surface receptors and their intracellular signal transduction pathways. Int Immunopharmacol. 2013;17(3):638–650. doi:10.1016/j.intimp.2013.06.034
62. Navarrete S, Solar C, Tapia R, Pereira J, Fuentes E, Palomo I. Pathophysiology of deep vein thrombosis. Clin Exp Med. 2023;23(3):645–654. doi:10.1007/s10238-022-00829-w
63. Cohen CT, Turner NA, Moake JL. Human endothelial cells and fibroblasts express and produce the coagulation proteins necessary for thrombin generation. Sci Rep. 2021;11(1):21852. doi:10.1038/s41598-021-01360-w
64. Neubauer K, Zieger B. Endothelial cells and coagulation. Cell Tissue Res. 2022;387(3):391–398. doi:10.1007/s00441-021-03471-2
65. Leo F, Suvorava T, Heuser SK, et al. Red blood cell and endothelial eNOS independently regulate circulating nitric oxide metabolites and blood pressure. Circulation. 2021;144(11):870–889. doi:10.1161/CIRCULATIONAHA.120.049606
66. Cyr AR, Huckaby LV, Shiva SS, Zuckerbraun BS. Nitric Oxide and Endothelial Dysfunction. Crit Care Clin. 2020;36(2):307–321. doi:10.1016/j.ccc.2019.12.009
67. Kant R, Halder SK, Fernández JA, Griffin JH, Milner R. Activated protein C attenuates experimental autoimmune encephalomyelitis progression by enhancing vascular integrity and suppressing microglial activation. Front Neurosci. 2020;14:333. doi:10.3389/fnins.2020.00333
68. Giri H, Panicker SR, Cai X, Biswas I, Weiler H, Rezaie AR. Thrombomodulin is essential for maintaining quiescence in vascular endothelial cells. Proc Natl Acad Sci USA. 2021;118(11). doi:10.1073/pnas.2022248118
69. Morrow GB, Past MNJ. Present, and Future Perspectives of Plasminogen Activator Inhibitor 1 (PAI-1). Semin Thromb Hemost. 2023;49(3):305–313. doi:10.1055/s-0042-1758791
70. Wada T, Gando S. Phenotypes of Disseminated Intravascular Coagulation. Thromb Hemost. 2024;124(3):181–191. doi:10.1055/a-2165-1142
71. Vincent JL, Ince C, Pickkers P. Endothelial dysfunction: a therapeutic target in bacterial sepsis? Expert Opin Ther Targets. 2021;25(9):733–748. doi:10.1080/14728222.2021.1988928
72. Martín-Fernández M, Tamayo-Velasco Á, Aller R, Gonzalo-Benito H, Martínez-Paz P, Tamayo E. Endothelial dysfunction and neutrophil degranulation as central events in sepsis physiopathology. Int J mol Sci. 2021;22(12):6272. doi:10.3390/ijms22126272
73. Ying J, Zhang C, Wang Y, et al. Sulodexide improves vascular permeability via glycocalyx remodelling in endothelial cells during sepsis. Front Immunol. 2023;14:1172892. doi:10.3389/fimmu.2023.1172892
74. Fernández-Sarmiento J, Salazar-Peláez LM, Carcillo JA. The endothelial glycocalyx: a fundamental determinant of vascular permeability in sepsis. Pediatr Crit Care Med. 2020;21(5):e291–e300. doi:10.1097/PCC.0000000000002266
75. Tarbell JM, Cancel LM. The glycocalyx and its significance in human medicine. J Inter Med. 2016;280(1):97–113. doi:10.1111/joim.12465
76. Uchimido R, Schmidt EP, Shapiro NI. The glycocalyx: a novel diagnostic and therapeutic target in sepsis. Critical Care. 2019;23(1):16. doi:10.1186/s13054-018-2292-6
77. Zhang H, Wang Y, Qu M, et al. Neutrophil, neutrophil extracellular traps and endothelial cell dysfunction in sepsis. Clin trans med. 2023;13(1):e1170. doi:10.1002/ctm2.1170
78. Hippensteel JA, Uchimido R, Tyler PD, et al. Intravenous fluid resuscitation is associated with septic endothelial glycocalyx degradation. Critical Care. 2019;23(1):259. doi:10.1186/s13054-019-2534-2
79. Denning NL, Aziz M, Gurien SD, Wang P. DAMPs and NETs in Sepsis. Front Immunol. 2019;10:2536. doi:10.3389/fimmu.2019.02536
80. Liew PX, Kubes P. The neutrophil’s role during health and disease. Physiol Rev. 2019;99(2):1223–1248. doi:10.1152/physrev.00012.2018
81. Vago JP, Zaidan I, Perucci LO, et al. Plasmin and plasminogen prevent sepsis severity by reducing neutrophil extracellular traps and systemic inflammation. JCI Insight. 2023;8(8). doi:10.1172/jci.insight.166044
82. Chen Z, Zhang H, Qu M, et al. Review: the emerging role of neutrophil extracellular traps in sepsis and sepsis-associated thrombosis. Front Cell Infect Microbiol. 2021;11:653228. doi:10.3389/fcimb.2021.653228
83. Ravindran M, Khan MA, Palaniyar N. Neutrophil extracellular trap formation: physiology, pathology, and pharmacology. Biomolecules. 2019;9(8):365. doi:10.3390/biom9080365
84. Hidalgo A, Libby P, Soehnlein O, Aramburu IV, Papayannopoulos V, Silvestre-Roig C. Neutrophil extracellular traps: from physiology to pathology. Cardiovasc Res. 2022;118(13):2737–2753. doi:10.1093/cvr/cvab329
85. Breda LCD, Breda CNS, de Almeida JRF, et al. Fonsecaea pedrosoi conidia and hyphae activate neutrophils distinctly: requirement of TLR-2 and TLR-4 in neutrophil effector functions. Front Immunol. 2020;11:540064. doi:10.3389/fimmu.2020.540064
86. Shao Y, Guo Z, Yang Y, et al. Neutrophil extracellular traps contribute to myofibroblast differentiation and scar hyperplasia through the Toll-like receptor 9/nuclear factor Kappa-B/interleukin-6 pathway. Burns Trauma. 2022;10:044. doi:10.1093/burnst/tkac044
87. Zhang H, Zhou Y, Qu M, et al. Tissue factor-enriched neutrophil extracellular traps promote immunothrombosis and disease progression in sepsis-induced lung injury. Front Cell Infect Microbiol. 2021;11:677902. doi:10.3389/fcimb.2021.677902
88. McDonald B, Davis RP, Kim S-J, et al. Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice. Blood. 2017;129(10):1357–1367. doi:10.1182/blood-2016-09-741298
89. Herre M, Cedervall J, Mackman N, Olsson AK. Neutrophil extracellular traps in the pathology of cancer and other inflammatory diseases. Physiol Rev. 2023;103(1):277–312. doi:10.1152/physrev.00062.2021
90. Thiam HR, Wong SL, Wagner DD, Waterman CM. Cellular mechanisms of NETosis. Annu Rev Cell Dev Biol. 2020;36:191–218. doi:10.1146/annurev-cellbio-020520-111016
91. Silva CMS, Wanderley CWS, Veras FP, et al. Gasdermin D inhibition prevents multiple organ dysfunction during sepsis by blocking NET formation. Blood. 2021;138(25):2702–2713. doi:10.1182/blood.2021011525
92. Ou Q, Tan L, Shao Y, et al. Electrostatic charge-mediated apoptotic vesicle biodistribution attenuates sepsis by switching neutrophil NETosis to apoptosis. Small. 2022;18(20):e2200306. doi:10.1002/smll.202200306
93. Boufenzer A, Carrasco K, Jolly L, et al. Potentiation of NETs release is novel characteristic of TREM-1 activation and the pharmacological inhibition of TREM-1 could prevent from the deleterious consequences of NETs release in sepsis. Cell Molecul Immunol. 2021;18(2):452–460. doi:10.1038/s41423-020-00591-7
94. Wilhelm G, Mertowska P, Mertowski S, et al. The crossroads of the coagulation system and the immune system: interactions and connections. Int J mol Sci. 2023;24(16):12563. doi:10.3390/ijms241612563
95. Liu D, Huang SY, Sun JH, et al. Sepsis-induced immunosuppression: mechanisms, diagnosis and current treatment options. Military Med Res. 2022;9(1):56. doi:10.1186/s40779-022-00422-y
96. Iba T, Hashiguchi N, Nagaoka I, Tabe Y, Murai M. Neutrophil cell death in response to infection and its relation to coagulation. J Intensive Care. 2013;1(1):13. doi:10.1186/2052-0492-1-13
97. Wen X, Xie B, Yuan S, Zhang J. The ”self-sacrifice” of immunecells in sepsis. Front Immunol. 2022;13:833479. doi:10.3389/fimmu.2022.833479
98. Andrieux P, Chevillard C, Cunha-Neto E, Nunes JPS. Mitochondria as a cellular Hub in infection and inflammation. Int J mol Sci. 2021;22(21):11338. doi:10.3390/ijms222111338
99. Supinski GS, Schroder EA, Callahan LA. Mitochondria and critical illness. Chest. 2020;157(2):310–322. doi:10.1016/j.chest.2019.08.2182
100. Pinto BB, Dyson A, Umbrello M, et al. Improved survival in a long-term rat model of sepsis is associated with reduced mitochondrial calcium uptake despite increased energetic demand. Crit Care Med. 2017;45(8):e840–e848. doi:10.1097/CCM.0000000000002448
101. Flora GD, Nayak MK, Ghatge M, Kumskova M, Patel RB, Chauhan AK. Mitochondrial pyruvate dehydrogenase kinases contribute to platelet function and thrombosis in mice by regulating aerobic glycolysis. Blood Adv. 2023;7(11):2347–2359. doi:10.1182/bloodadvances.2023010100
102. Zou R, Tao J, Qiu J, et al. DNA-PKcs promotes sepsis-induced multiple organ failure by triggering mitochondrial dysfunction. J Adv Res. 2022;41:39–48. doi:10.1016/j.jare.2022.01.014
103. Tang KJ, Zhao Y, Tao X, et al. Catecholamine derivatives: natural occurrence, structural diversity, and biological activity. J Natural Prod. 2023;86(11):2592–2619. doi:10.1021/acs.jnatprod.3c00465
104. Saboory E, Ghasemi M, Mehranfard N. Norepinephrine, neurodevelopment and behavior. Neurochem Int. 2020;135:104706. doi:10.1016/j.neuint.2020.104706
105. Li L, Rana AN, Li EM, Feng J, Li Y, Bruchas MR. Activity-dependent constraints on catecholamine signaling. Cell Rep. 2023;42(12):113566. doi:10.1016/j.celrep.2023.113566
106. Motiejunaite J, Amar L, Vidal-Petiot E. Adrenergic receptors and cardiovascular effects of catecholamines. Ann Endocrinol. 2021;82(3–4):193–197. doi:10.1016/j.ando.2020.03.012
107. Shi X, Li P, Liu H, Prokosch V. Oxidative stress, vascular endothelium, and the pathology of neurodegeneration in retina. Antioxidants. 2022;11(3):543. doi:10.3390/antiox11030543
108. Liu T, Yang L, Han X, Ding X, Li J, Yang J. Local sympathetic innervations modulate the lung innate immune responses. Sci Adv. 2020;6(20):1497. doi:10.1126/sciadv.aay1497
109. Andersson U, Tracey KJ. Reflex principles of immunological homeostasis. Ann Rev Immunol. 2012;30:313–335. doi:10.1146/annurev-immunol-020711-075015
110. Millán Solano MV, Salinas Lara C, Sánchez-Garibay C, et al. Effect of systemic inflammation in the cns: a silent history of neuronal damage. Int J mol Sci. 2023;24(15):11902. doi:10.3390/ijms241511902
111. Da Mesquita S, Fu Z, Kipnis J. The meningeal lymphatic system: a new player in neurophysiology. Neuron. 2018;100(2):375–388. doi:10.1016/j.neuron.2018.09.022
112. Kohoutova M, Horak J, Jarkovska D, et al. Vagus nerve stimulation attenuates multiple organ dysfunction in resuscitated porcine progressive sepsis. Crit Care Med. 2019;47(6):e461–e469. doi:10.1097/CCM.0000000000003714
113. Rana M, Fei-Bloom Y, Son M, et al. Constitutive vagus nerve activation modulates immune suppression in sepsis survivors. Front Immunol. 2018;9:2032. doi:10.3389/fimmu.2018.02032
114. Celik C, Lee SYT, Yap WS, Thibault G. Endoplasmic reticulum stress and lipids in health and diseases. Prog lipid res. 2023;89:101198. doi:10.1016/j.plipres.2022.101198
115. Wang J, Yu C, Zhuang J, et al. The role of phosphatidylserine on the membrane in immunity and blood coagulation. Biomarker Res. 2022;10(1):4. doi:10.1186/s40364-021-00346-0
116. Khan MM, Yang WL, Wang P. Endoplasmic Reticulum Stress In Sepsis. Shock. 2015;44(4):294–304. doi:10.1097/SHK.0000000000000425
117. Zhang H, Zeng L, Xie M, et al. TMEM173 drives lethal coagulation in sepsis. Cell Host Microbe. 2020;27(4):556–570.e6. doi:10.1016/j.chom.2020.02.004
118. Iba T, Umemura Y, Wada H, Levy JH. Roles of coagulation abnormalities and microthrombosis in sepsis: pathophysiology, diagnosis, and treatment. Archiv Med Res. 2021;52(8):788–797. doi:10.1016/j.arcmed.2021.07.003
119. Shankar-Hari M, Phillips GS, Levy ML, et al. Developing a new definition and assessing new clinical criteria for septic shock: for the third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. 2016;315(8):775–787. doi:10.1001/jama.2016.0289
120. Bateman RM, Sharpe MD, Goldman D, Lidington D, Ellis CG. Inhibiting nitric oxide overproduction during hypotensive sepsis increases local oxygen consumption in rat skeletal muscle. Crit Care Med. 2008;36(1):225–231. doi:10.1097/01.CCM.0000295307.92027.2F
121. Yajnik V, Maarouf R. Sepsis and the microcirculation: the impact on outcomes. Curr Opin Anaesthesiol. 2022;35(2):230–235. doi:10.1097/ACO.0000000000001098
122. Rovas A, Seidel LM, Vink H, et al. Association of sublingual microcirculation parameters and endothelial glycocalyx dimensions in resuscitated sepsis. Critical Care. 2019;23(1):260. doi:10.1186/s13054-019-2542-2
123. Bateman RM, Sharpe MD, Jagger JE, Ellis CG. Sepsis impairs microvascular autoregulation and delays capillary response within hypoxic capillaries. Critical Care. 2015;19:389. doi:10.1186/s13054-015-1102-7
124. De Backer D, Ricottilli F, Ospina-Tascón GA. Septic shock: a microcirculation disease. Curr Opin Anaesthesiol. 2021;34(2):85–91. doi:10.1097/ACO.0000000000000957
125. Colbert JF, Schmidt EP. Endothelial and microcirculatory function and dysfunction in sepsis. Clinics Chest Med. 2016;37(2):263–275. doi:10.1016/j.ccm.2016.01.009
126. Mallat J, Rahman N, Hamed F, Hernandez G, Fischer MO. Pathophysiology, mechanisms, and managements of tissue hypoxia. Anaesth Crit Care Pain Med. 2022;41(4):101087. doi:10.1016/j.accpm.2022.101087
127. Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol. 2013;13(1):34–45. doi:10.1038/nri3345
128. Park I, Kim M, Choe K, et al. Neutrophils disturb pulmonary microcirculation in sepsis-induced acute lung injury. Eur Respir J. 2019;53(3).1.
129. Burban M, Hamel JF, Tabka M, et al. Renal macro- and microcirculation autoregulatory capacity during early sepsis and norepinephrine infusion in rats. Critical Care. 2013;17(4):R139. doi:10.1186/cc12818
130. Iba T, Helms J, Connors JM, Levy JH. The pathophysiology, diagnosis, and management of sepsis-associated disseminated intravascular coagulation. J Intensive Care. 2023;11(1):24. doi:10.1186/s40560-023-00672-5
131. Lehner GF, Tobiasch AK, Perschinka F, et al. Associations of tissue factor and tissue factor pathway inhibitor with organ dysfunctions in septic shock. Sci Rep. 2024;14(1):14468. doi:10.1038/s41598-024-65262-3
132. Stahl K, Lehner GF, Wendel-Garcia PD, et al. Effect of therapeutic plasma exchange on tissue factor and tissue factor pathway inhibitor in septic shock. Critical Care. 2024;28(1):351. doi:10.1186/s13054-024-05142-4
133. Raman R, Fallatah W, Qaryoute AA, Ryon M, Jagadeeswaran P. Knockdown and knockout of tissue factor pathway inhibitor in zebrafish. Thromb Hemost. 2022;122(7):1104–1114. doi:10.1055/a-1723-4075
134. Reitsma SE, Holle LA, Bouck EG, et al. Tissue factor pathway inhibitor is a potential modifier of bleeding risk in factor XI deficiency. J Thromb Haemost. 2023;21(3):467–479. doi:10.1016/j.jtha.2022.10.005
135. Formosa A, Turgeon P, Dos Santos CC. Role of miRNA dysregulation in sepsis. Mol Med. 2022;28(1):99. doi:10.1186/s10020-022-00527-z
136. Arroyo AB, Águila S, Fernández-Pérez MP, et al. miR-146a in cardiovascular diseases and sepsis: an additional burden in the inflammatory balance? Thromb Hemost. 2021;121(9):1138–1150. doi:10.1055/a-1342-3648
137. Yang C, Passos Gibson V, Hardy P. The role of MiR-181 family members in endothelial cell dysfunction and tumor angiogenesis. Cells. 2022;11(10):1.
138. Barker KR, Lu Z, Kim H, et al. miR-155 modifies inflammation, endothelial activation and blood-brain barrier dysfunction in cerebral malaria. Mol Med. 2017;23:24–33. doi:10.2119/molmed.2016.00139
139. Etzrodt V, Idowu TO, Schenk H, et al. Role of endothelial microRNA 155 on capillary leakage in systemic inflammation. Critical Care. 2021;25(1):76. doi:10.1186/s13054-021-03500-0
140. Palomo M, Moreno-Castaño AB, Salas MQ, et al. Endothelial activation and damage as a common pathological substrate in different pathologies and cell therapy complications. Front Med. 2023;10:1285898. doi:10.3389/fmed.2023.1285898
141. Liu X, Arfman T, Wichapong K, Reutelingsperger CPM, Voorberg J, Nicolaes GAF. PAD4 takes charge during neutrophil activation: impact of PAD4 mediated NET formation on immune-mediated disease. J Thromb Haemost. 2021;19(7):1607–1617. doi:10.1111/jth.15313
142. Li P, Li M, Lindberg MR, Kennett MJ, Xiong N, Wang Y. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Experiment Med. 2010;207(9):1853–1862. doi:10.1084/jem.20100239
143. Suzuki M, Ikari J, Anazawa R, et al. PAD4 deficiency improves bleomycin-induced neutrophil extracellular traps and fibrosis in mouse lung. Am J Respir Cell Mol Biol. 2020;63(6):806–818. doi:10.1165/rcmb.2019-0433OC
144. Liu X, Li T, Chen H, Yuan L, Ao H. Role and intervention of PAD4 in NETs in acute respiratory distress syndrome. Respir Res. 2024;25(1):63. doi:10.1186/s12931-024-02676-7
145. Moreira Leal LK A, Lima LA, de Aquino PE A, et al. Vitamin D (VD3) antioxidative and anti-inflammatory activities: peripheral and central effects. Eur J Pharmacol. 2020;879:173099. doi:10.1016/j.ejphar.2020.173099
146. Ahmad S, Zaki A, Manda K, Mohan A, Syed MA. Vitamin-D ameliorates sepsis-induced acute lung injury via augmenting miR-149-5p and downregulating ER stress. J Nutr Biochem. 2022;110:109130. doi:10.1016/j.jnutbio.2022.109130
147. Vanherwegen AS, Gysemans C, Mathieu C. Regulation of immune function by vitamin d and its use in diseases of immunity. Endocrinol Metabol Clin North Am. 2017;46(4):1061–1094. doi:10.1016/j.ecl.2017.07.010
148. Jones AW, Mironas A, Mur LAJ, Beckmann M, Thatcher R, Davison G. Vitamin D status modulates innate immune responses and metabolomic profiles following acute prolonged cycling. Eur J Nutr. 2023;62(7):2977–2990. doi:10.1007/s00394-023-03181-1
149. Inata Y. Should we treat sepsis-induced DIC with anticoagulants? J Intensive Care. 2020;8:18. doi:10.1186/s40560-020-0435-8
150. Levi M, Scully M. How I treat disseminated intravascular coagulation. Blood. 2018;131(8):845–854. doi:10.1182/blood-2017-10-804096
151. Omarjee L, Meilhac O, Perrot F, Janin A, Mahe G. Can Ticagrelor be used to prevent sepsis-induced coagulopathy in COVID-19? Clinical Immunology. 2020;216:108468. doi:10.1016/j.clim.2020.108468
152. Mihalko EP, Sandry M, Mininni N, et al. Fibrin-modulating nanogels for treatment of disseminated intravascular coagulation. Blood Adv. 2021;5(3):613–627. doi:10.1182/bloodadvances.2020003046
153. Li Y, Zhao L, Yu Y, et al. Conservative oxygen therapy in critically ill and perioperative period of patients with sepsis-associated encephalopathy. Front Immunol. 2022;13:1035298. doi:10.3389/fimmu.2022.1035298
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.