Previous Article in Journal
Assessing the Onset of Regional Anaesthesia: The Role of Thermographic Imaging
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Damage-Associated Molecular Patterns in Perioperative Anesthesia Care: A Clinical Perspective

1
Department of Anesthesiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
2
Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
3
Department of Anaesthesia and Immunology, Harvard Medical School, Boston, MA 02115, USA
*
Authors to whom correspondence should be addressed.
Anesth. Res. 2026, 3(1), 1; https://doi.org/10.3390/anesthres3010001 (registering DOI)
Submission received: 8 October 2025 / Revised: 26 November 2025 / Accepted: 11 December 2025 / Published: 20 December 2025

Abstract

Damage-associated molecular patterns (DAMPs) are endogenous molecules released during cellular stress or injury that trigger sterile inflammation. In perioperative settings, common triggers include surgical trauma, ischemia–reperfusion injury, cardiopulmonary bypass, blood transfusion, and mechanical ventilation. When released extracellularly, DAMPs activate innate immune receptors such as Toll-like receptors (TLRs) and the receptor for advanced glycation end products (RAGE), initiating signaling cascades that amplify inflammation, disrupt endothelial integrity, and promote coagulation and metabolic imbalance. This sterile inflammatory response may extend local tissue injury into systemic organ dysfunction, manifesting clinically as acute lung injury, acute kidney injury, myocardial dysfunction, disseminated intravascular coagulation, and perioperative neurocognitive disorders. Recognizing the central role of DAMPs reframes these complications as predictable consequences of endogenous danger signaling rather than solely as results of infection or hemodynamic instability. This understanding supports the use of established strategies such as protective ventilation and restrictive transfusion to minimize DAMP release. Emerging evidence also suggests that anesthetic agents may influence DAMP-mediated inflammation: propofol and dexmedetomidine appear to exert anti-inflammatory effects, whereas volatile anesthetics show variable results. Although clinical data remain limited, anesthetic choice and perioperative management may significantly affect systemic inflammatory burden and recovery. Future research validating DAMPs as biomarkers and therapeutic targets may inform precision anesthetic strategies aimed at modulating sterile inflammation, ultimately enhancing perioperative outcome.

Graphical Abstract

1. Introduction

Anesthesiology has evolved far beyond the roles of inducing unconsciousness and analgesia. Modern anesthesiologists function as perioperative physicians, managing complex physiological challenges and preventing complications that may arise long after anesthesia ends. Despite improvement in surgical techniques, anesthesia management, monitoring, and perioperative care, postoperative complications such as acute lung injury (ALI), acute kidney injury (AKI), myocardial dysfunction, and delirium remain common and continue to affect patient recovery. Traditionally, these issues have been attributed to perioperative ischemia, hemodynamic instability, infection, or drug effects. However, emerging research suggests that damage-associated molecular patterns (DAMPs), molecules released endogenously as danger signals, may fundamentally contribute to the development of systemic inflammation and organ [1].
The concept of DAMPs originated from the recognition that inflammation can occur independently of infection [2]. These molecules are normally contained within cells but are released following cellular stress, injury, or necrosis. Once outside the cell, DAMPs are recognized mainly by the innate immune system as signals of tissue damage, provoking inflammatory responses that resembles sepsis, even in the absence of microbial infection.
Although the terminology around DAMPs may seem abstract or overly focused on molecular immunology, their clinical importance is direct and tangible. DAMPs are released during common perioperative events, such as surgical incisions, transfusions, ischemia followed by reperfusion, and the use of cardiopulmonary bypass (CPB). After their release, these molecules can initiate or amplify inflammation, contributing to postoperative complications. These mechanisms may also help explain why some patients develop systemic complications despite apparently uneventful intraoperative courses. Differences in both the extent of tissue injury and the individual’s immune response may influence postoperative trajectories.
This review provides a clinician-oriented perspective on DAMPs, with a focus on their perioperative sources, pathophysiological consequences, and relevance to anesthetic management. By translating emerging scientific knowledge into practical clinical understanding, we aim to offer anesthesiologists a practical framework to better anticipate, recognize, and potentially mitigate DAMP-related complications.

2. Methods

This narrative review was conducted to examine the role of DAMPs in perioperative inflammation and organ dysfunction, with an emphasis on their relevance to anesthesia care. A targeted literature search was performed across MEDLINE, PubMed, and Google Scholar for articles published up to September 2025. Primary search terms included: “damage-associated molecular patterns”, “DAMPs”, “sterile inflammation”, “postoperative complications”, and “anesthesia”. Additional keywords were combined to refine the search, such as “ischemia–reperfusion injury”, “cardiopulmonary bypass”, “mechanical ventilation”, “transfusion”, “acute lung injury”, “acute kidney injury”, “myocardial dysfunction”, and “perioperative neurocognitive disorders”.
To capture relevant mechanistic and translational insights, supplementary searches were performed for specific DAMP molecules, pattern recognition receptors, and inflammatory mediators. Eligible studies included basic science experiments, animal models, translational research, clinical trials, systematic reviews, and narrative reviews. Articles were selected based on their relevance to perioperative DAMP release, signaling, and pathophysiological consequences, as well as the influence of anesthetic agents and perioperative practices. Case reports, commentaries, and correspondence pieces were excluded. Reference lists of included articles were also screened to identify additional relevant studies. The initial search identified approximately 7930 articles, of which 130 were selected for inclusion in this review.
Given the narrative nature of this review, a formal systematic review methodology was not employed. As such, the possibility of selection bias and incomplete coverage of the literature remains a limitation of this work.

3. Understanding Damage-Associated Molecular Patterns (DAMPs)

DAMPs are endogenous molecules that are released from the body’s own cells when they are stressed, injured, or undergo unregulated forms of cell death [3]. Under normal conditions, these molecules remain confined within cells, where they play essential roles in metabolism, structural organization, and gene regulation. However, when they are released into the extracellular environment, they are detected by pattern recognition receptors (PRRs) on immune and non-immune cells, which perceive their presence as a sign of tissue injury. This interaction initiates a cascade of immune responses that can lead to sterile inflammation, even when no pathogens are present [4].

3.1. Historical Background and the Danger Model of Immunity

Consider a patient who develops systemic inflammatory response syndrome (SIRS) following major surgery, severe trauma, acute pancreatitis, or ischemic-reperfusion injury after organ transplantation. Despite the absence of identifiable infection, the patient presents with fever, hypotension, leukocytosis, and signs of multi-organ dysfunction. Blood cultures remain negative, and no microbial source is identified. The clinical presentation, however, closely resembles sepsis. Such scenarios have long challenged the traditional views that inflammation arises primarily in response to infection.
Earlier concepts in immunology proposed that the immune system functions primarily by distinguishing between self and non-self. In this model, immune responses were directed against foreign entities such as bacteria, viruses, or transplanted tissues, while endogenous components were presumed to be tolerated. This framework provided a useful basis for understanding host defense and transplant rejection. The subsequent discovery of pathogen-associated molecular patterns (PAMPs) added molecular details to the theory. PAMPs are conserved microbial structures absent from host cells, recognized by PRRs on immune cells, which then initiate protective responses to infection [5]. While this model clarified how the innate immune system detects pathogens, it did not adequately explain inflammation triggered by sterile tissue injury [6].
To address this gap, the “danger model” of immunity was proposed [7]. This model suggests that immune responses can be triggered not only by microbial components but also by endogenous signals released during cellular stress or injury [8,9]. These signals, now recognized as DAMPs, are normally contained within healthy cells. When released into the extracellular environment, they are perceived as misplaced and interpreted by PRRs as indicators of danger. Thus, immune activation reflects not only the detection of non-self, but also the recognition of tissue injury within self.

3.2. Perioperative DAMPs and Immune Recognition

The term DAMP refers to endogenous molecules that alert the immune system when they appear outside their normal intracellular locations. They are sometimes also called alarmins, a term that emphasizes their role as rapid alarm signals, often released actively by stressed or immune cells [10]. Although the terminology differs slightly, both concepts describe the same phenomenon, and in this review, we will use the term DAMPs for clarity.
DAMPs include molecules with diverse structures and functions, such as nuclear proteins, mitochondrial components, cytosolic enzymes, extracellular matrix fragments, and transfusion-derived products [3,11] (Table 1, Figure 1). Within healthy cells they contribute to essential biological processes but once released into the extracellular space they are interpreted as signals of tissue injury and can provoke an immune response.
Recognition of DAMPs occurs through PRRs, a broad family of innate immune sensors. These receptors are expressed on immune cells as well as on endothelial and parenchymal cells, enabling both systemic and tissue-specific responses [24]. Their physiological role is to detect danger signals, whether derived from invading microbes or from injured host tissues. In the perioperative setting, the most relevant PRR families are Toll-like receptors (TLRs), the receptor for advanced glycation end products (RAGE), and NOD-like receptors (NLRs). Engagement of these receptors activates intracellular signaling cascades. For TLRs, this involves recruitment of the adaptor protein myeloid differentiation primary response 88 (MyD88), which activates downstream pathways including nuclear factor kappa B (NF-κB) and the mitogen-activated protein kinases (MAPKs). RAGE also activates MAPKs and NF-κB through distinct adaptors. RAGE also helps to mobilize some DAMPs to the intracellular space and present to TLRs. Once activated, NF-κB translocates to the nucleus, while MAPKs stimulate transcription factors such as activator protein-1 (AP-1). Together, these promote the transcription of pro-inflammatory cytokines, chemokines, and adhesion molecules that sustain inflammation (Figure 2). In addition, certain DAMPs contribute to inflammasome activation. For example, high mobility group box 1 (HMGB1) can enhance NOD-like receptor protein 3 (NLRP3) priming through NF-κB signaling, while extracellular adenosine triphosphate (ATP) engages P2X7 receptors and mitochondrial stress releases reactive oxygen species (ROS) and mitochondrial DNA (mtDNA), all of which promote inflammasome assembly. The resulting caspase-1 activation drives interleukin (IL)-1β and IL-18 maturation and pyroptotic cell death, further amplifying inflammation and DAMP release.

3.3. Dual Roles of DAMPs: Healing and Harm

DAMPs play a dual role in the host response to tissue injury. At physiological levels, they locally serve as an important alarm signal that helps initiate healing. By activating the innate immune system, DAMPs promote leukocyte recruitment, clearance of cellular debris, and tissue regeneration. These local responses are essential for recovery after surgery or trauma, and in this context, DAMPs contribute to tissue repair rather than harm the host (Figure 2).
However, when DAMP release is excessive, sustained, or dysregulated, the same signals that support healing can become pathogenic. Instead of resolving local injury, they may drive persistent systemic inflammation, damage the vascular endothelium, disrupt coagulation, and impair organ function. These systemic effects can occur even when the initial insult is localized. For example, sterile tissue injury from surgical trauma or limb ischemia–reperfusion can trigger distant organ dysfunction through DAMP-mediated pathways.

4. Cell Death as a Source of DAMPs

The type of cell death that occurs during surgical injury strongly influences the nature and quantity of DAMPs released. Some modes of death are relatively silent, while others provoke robust inflammatory responses that can contribute to perioperative complications. Figure 3 illustrates types of cell death and their pathological feature.
  • Necrosis
  • Necrosis is an uncontrolled form of cell death that results from direct trauma, ischemia, or toxic injury. It is characterized by cellular swelling and membrane rupture, which cause the release of nuclear, mitochondrial, and cytosolic components into the extracellular space [25]. Necrosis is therefore a major contributor to DAMP release during surgical dissection and manipulation, liberating molecules such as HMGB1, ATP, histones, heat shock proteins (HSPs), mtDNA, and cell-free DNA (cfDNA) [26].
  • Apoptosis
  • Apoptosis is a tightly regulated process that normally avoids inflammation. Cellular content are packaged into apoptotic bodies that are cleared by phagocytes without DAMP release [27]. However, if clearance is delayed or overwhelmed, apoptotic cells progress to secondary necrosis, resulting in the release of DAMPs, particularly, HMGB1 [28]. Clinically, it has been shown that the rate of apoptosis is significantly increased in patients undergoing surgical stress [29].
  • Autophagy-related death
  • Autophagy is usually a protective mechanism that helps cells survive stress by removing damaged cytoplasmic components and preserving cellular homeostasis. In the perioperative setting, this function can reduce the release of DAMPs during tissue stress. However, when stress is excessive or prolonged, autophagy may fail and progress to cell death, allowing intracellular material to escape and contribute to extracellular pool of DAMPs [30].
  • Regulated inflammatory cell death
  • This group of programmed cell death pathways differs from apoptosis in that it provokes inflammation rather than suppressing it. Several regulated inflammatory cell death mechanisms are increasingly recognized as important in perioperative injury:
    Pyroptosis—a pro-inflammatory form of cell death driven by NLRP3 inflammasome activation. A common trigger is extracellular ATP acting on P2X7 receptors. The process results in membrane rupture and the release of cytokines (IL-1β, IL-18) together with DAMPs, amplifying inflammation [31].
    Necroptosis—a regulated form of cell death that resembles necrosis in its outcome but proceeds though a regulated signaling cascade. It culminates in membrane disruption and extracellular release of HMGB1, ATP, and S100 proteins [32].
    Ferroptosis—associated with iron overload and lipid peroxidation, this process releases DAMPs such as HMGB1 and has been implicated in ischemia–reperfusion injury [33].
    NETosis—a neutrophil-specific pathway in which neutrophils release neutrophil extracellular traps (NETs) composed of DNA, histones, and granule proteins. Although NETs immobilize pathogens, they are also rich in DAMPs that promote endothelial injury, thrombosis, and organ dysfunction [34]. NETosis has also been implicated in complications such as transfusion-related lung injury and thromboinflammation [35,36].

5. Perioperative Sources of DAMPs

Cell death processes become particularly relevant in the perioperative setting, where surgical and anesthetic interventions often create conditions that favor their activation. Necrosis, apoptosis, necroptosis, and other inflammatory cell death can be triggered by direct tissue injury, ischemia–reperfusion (I/R) injury, extracorporeal circulation, transfusion, and mechanical stress. Each of these perioperative events promotes the release of distinct DAMPs, which in turn contribute to systemic inflammation and postoperative complications.

5.1. Surgical Trauma

Surgical incision, dissection, and cauterization inevitably disrupt cells and extracellular structures. This causes the release of nuclear protein, cytosolic enzymes, mitochondrial component, and extracellular matrix fragments. While a controlled inflammatory response is necessary for wound healing, extensive surgical trauma can generate excessive release and propagate systemic inflammation. This mechanistic link provides a biological explanation for the higher inflammatory burden and complication rates seen with longer or more invasive procedures.

5.2. Ischemia–Reperfusion Injury

Temporary interruption of blood flow is common in vascular, transplant, and cardiac surgery, as well as during the use of tourniquets. In the ischemic phase, oxygen and nutrient deprivation deplete ATP stores, impair ion gradients, and promote mitochondrial dysfunction. These changes initiate necrosis and apoptosis in some cells, while others undergo necroptosis if the insult is severe or prolonged [37]. Reperfusion, although restoring circulation, paradoxically worsens injury by generating ROS, which destabilize cell membranes and activate regulated inflammatory cell death pathways such as pyroptosis and ferroptosis [38]. These processes release DAMPs including HMGB1, mtDNA, ATP, and HSPs [37,39], which amplify sterile inflammation beyond the ischemic tissue and potentially contribute to perioperative complications such as ALI, myocardial dysfunction, and AKI, even after otherwise successful operations.

5.3. Cardiopulmonary Bypass and Extracorporeal Circulation

CPB is among the most intense stimuli for perioperative DAMP release because it exposes blood to artificial surfaces and non-physiological flow conditions. Blood contact with artificial surfaces and non-physiological flow conditions activates leukocytes, platelets, and complement, while ischemia–reperfusion of the myocardium and other organs adds further injury [40]. High circulating concentrations of HMGB1, histones, S100A8/A9, and HSPs have been consistently documented during and after CPB [34,41]. The magnitude of this response correlates with bypass duration and ischemic severity, providing a mechanistic rationale for the poorer outcomes seen after prolonged CPB and the relative advantages reported with off-pump coronary artery bypass (OPCAB) surgery. The frequent need for perioperative transfusion further compounds this DAMP burden.

5.4. Blood Product Transfusion

Blood products accumulate structural and biochemical changes during storage, known as “storage lesions” [42], which generate bioactive molecules such as heme, cfDNA, extracellular ATP, HMGB1, and microparticles [11,43,44]. These changes occur not only in red cells but also in plasma-rich components such as platelets and fresh frozen plasma [11]. When transfused, these molecules enter the circulation and activate PRRs on immune and endothelial cells. In the pulmonary vasculature, this process primes neutrophils, leading to adhesion, activation, and the release of ROS and proteolytic enzymes. The resulting endothelial injury and increased vascular permeability form the basis of transfusion-related acute lung injury (TRALI) [11]. Transfusion may also induce broader immune dysregulation, referred to as transfusion-related immunomodulation (TRIM) [45]. These insights provide a mechanistic explanation for why restrictive transfusion thresholds and patient blood management (PBM) strategies improve outcomes.

5.5. Mechanical Ventilation

Mechanical stress is another iatrogenic source of DAMPs. High tidal volumes and excessive airway pressures stretch alveoli, disrupting epithelial and endothelial barriers, and release of extracellular ATP and HSP70 [46,47]. These DAMPs activate immune pathways that promote cytokine production, leukocyte recruitment, and pulmonary inflammation, potentially contributing to ventilator-induced lung injury (VILI) [48]. Importantly, this inflammatory reaction is not confined to the lungs. Systemic spillover of inflammatory mediators may initiate and propagate systemic inflammation and multi-organ dysfunction [47]. Protective ventilation strategies mitigate these effects and have become standard practice in perioperative and critical care.

6. Pathophysiological Consequences of DAMP Release

The perioperative environment provides numerous triggers for DAMP release. These molecules do not remain confined to the site of injury but spill into the circulation, spread systemically, and interact with immune systems in ways that amplify inflammation and disrupt homeostasis. The downstream consequences include endothelial injury, coagulation disturbances, propagation of systemic inflammation, and ultimately organ dysfunction. Taken together, these processes form the pathophysiological bridge between intraoperative events and postoperative complications.

6.1. Endothelial Activation and Microvascular Dysfunction

The vascular endothelium is one of the earliest and most vulnerable targets of circulating DAMPs. Through PRR-mediated signaling [49], DAMPs disrupt endothelial barrier integrity, increase permeability, and induce the expression of adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) [50]. These changes promote leukocyte adhesion and transmigration into the site of injury [51].
Loss of barrier function results in capillary leak and tissue edema, while reduced nitric oxide (NO) bioavailability compromises vasodilation and microvascular perfusion [52]. Endothelial activation also promotes a shift toward a procoagulant phenotype, with increased tissue factor (TF) exposure and von Willebrand factor (vWF) release [53]. Collectively, these responses impair vascular homeostasis, reduce oxygen delivery, and predispose to postoperative inflammatory complications.

6.2. Thrombosis and Coagulopathy

Perioperative release of DAMPs exerts complex effects on hemostasis, with most molecules driving immunothrombosis while others exert anticoagulant influences.
On the prothrombotic side, DAMPs may act at multiple stages of coagulation. HMGB1 initiates clotting by upregulating TF expression on endothelial cells and inhibiting the protein C pathway [54]. mtDNA propagates the cascade through activation of the intrinsic pathway [55], while S100A9 enhance thrombus formation [56]. Clot stability is reinforced by histone-induced secretion of plasminogen activator inhibitor-1 (PAI-1) [57], which suppresses fibrinolysis, and by cfDNA, which impairs plasmin-mediated fibrin degradation [58]. In addition, DAMPs promote platelet activation and drive neutrophils to form extracellular traps (NETs), providing a structural scaffold for thrombin generation, fibrin deposition, and further platelet aggregation [36]. Together, these processes create a potent prothrombotic environment that can escalate into widespread microvascular thrombosis.
Excessive activation, however, may culminate in consumptive coagulopathy, where sustained thrombin generation and fibrin deposition deplete platelets and coagulation factors. Clinically, this manifests as disseminated intravascular coagulation (DIC), characterized by the paradoxical coexistence of microvascular thrombosis and bleeding.
In contrast, certain DAMPs can exert anticoagulant effects. Heparan sulfate has been associated with impaired thrombin generation in severely injured trauma patients [59], while elevated serum histone H3 levels were observed in septic patients with coagulopathy and predictive of their higher mortality risk [60]. Although these findings may reflect protective or compensatory mechanisms in isolation, their coexistence with strong prothrombotic signals contributes to the unstable hemostatic balance that characterizes perioperative critical illness.

6.3. Propagation of Systemic Inflammation

DAMP release initiates and sustains systemic inflammation through complex signaling pathways and cellular interactions. Activation of PRRs leads to production of pro-inflammatory cytokines and chemokines, such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, IL-8, and interferons. These mediators recruit and activate leukocytes and establish a positive feedback loop that promotes further DAMP release.
Certain DAMPs also stimulate neutrophils to undergo NETosis, generating extracellular traps rich in DNA, histones, and HMGB1 that act as additional DAMPs and sustain inflammation. Activation of the NLRP3 inflammasome further enhances secretion of IL-1β and IL-18 through pyroptosis, intensifying the systemic response. Collectively, these processes perpetuate sterile inflammation that can progress to SIRS and multi-organ dysfunction in susceptible patients.

6.4. Organ Dysfunction

The convergence of endothelial injury, coagulation disturbances, and sustained inflammation drives organ dysfunction. Capillary leak and microvascular thrombosis reduce oxygen and nutrient delivery, while mitochondrial dysfunction and oxidative stress impair cellular energy metabolism. NETs exacerbate this by obstructing capillary flow and propagating inflammation.
These mechanisms manifest clinically as ALI, acute respiratory distress syndrome (ARDS), AKI, myocardial dysfunction, DIC, or perioperative neurocognitive disorder (PND). Although the affected organ may differ, the underlying mechanism is unified, driven by sterile inflammation initiated by DAMP release. This model provides a coherent explanation for how apparently routine intraoperative events can progress to systemic complications and highlights the potential role of DAMPs in perioperative outcomes. Table 2 summarizes the major DAMPs implicated in perioperative organ dysfunction, their pathways, and associated clinical consequences.

7. Anesthetic Implications

Many perioperative events act as potent triggers for DAMP release, and anesthesia is closely intertwined with these processes. This positions anesthesiologists to influence not only immediate physiological stability but also the molecular signals that shape postoperative recovery. Unlike cytokines, which represent downstream mediators common to many inflammatory cascades, DAMPs function as upstream initiators of sterile inflammation. Strategies that reduce their release or limit their recognition therefore represent opportunities to interrupt the inflammatory cascade at an earlier and potentially more effective stage.

7.1. Anesthetic Agents

Experimental and early clinical evidence suggests that anesthetic agents may modulate DAMP release and signaling These effects are not yet strong enough to dictate practice guideline, but they may provide biological rationale for tailoring drug selection in high-risk settings. Table 3 provides an overview of current evidence on anesthetic agents and their potential interactions with DAMP biology.
Volatile anesthetics such as sevoflurane and isoflurane have shown mixed results. In experimental models, they can reduce HMGB1 release and attenuate I/R injury [98], but prolonged exposure has also been associated with impaired cognitive outcomes [125], particularly in vulnerable brains [97]. These discrepancies may be due to differences in study conditions, including the duration and depth of anesthesia, timing of biomarker collection, baseline inflammatory status, or tissue-specific susceptibility.
In contrast, propofol has demonstrated more consistent protective effects in preclinical studies, including reduced mtDNA and HMGB1 release, suppression of HMGB1-driven pathways, and reduction in oxidative stress. Clinical evidence is less consistent. For example, in posterior spinal fusion surgery, isoflurane anesthesia was associated with higher perioperative mtDNA levels compared with propofol, suggesting greater DAMP release [126]. In contrast, in a pediatric liver transplantation study reported similar rises in IL-6, TNF-α, and HMGB1 with both propofol and sevoflurane, with no clear difference between groups [127].
Among available agents, dexmedetomidine (DEX) has been studied most extensively for its anti-inflammatory potential. Experimental data show that DEX can suppress HMGB1 release, inhibit TLR4 activation, and reduce pyroptosis, necroptosis, and I/R injury. Clinical studies suggest that DEX reduces the incidence of PND, although this benefit may involve multiple pathways beyond HMGB1 alone. It is worth noting that some anesthetics do not directly alter DAMP molecules themselves but instead act by inhibiting TLR signaling, thereby limiting the immune system’s ability to amplify DAMP-driven inflammation.

7.2. Transfusion Practice

Blood products represent an important iatrogenic source of DAMPs. Recognition that stored blood products contain DAMPs reinforces the importance of PBM principles in anesthetic practice. Restrictive transfusion thresholds should be employed whenever possible, and unnecessary transfusions should be avoided. When transfusion is unavoidable, strategies such as using fresher units, preferring leuko-reduced or washed products, and minimizing exposure to plasma-rich components can reduce the DAMP burden. Careful intraoperative hemostasis, antifibrinolytic therapy, and blood conservation techniques further decrease reliance on transfusion. These measures, already advocated by PBM guidelines, gain additional mechanistic support from DAMP biology and highlight the anesthesiologist’s role in actively mitigating transfusion-related complications.

7.3. Cardiopulmonary Bypass and Extracorporeal Circuits

In the setting of CPB, strategies that minimize the inflammatory burden should be prioritized. Several strategies may attenuate this response. Minimizing bypass duration and using shorter or simplified circuits reduces the blood–surface interface and the extent of hemolysis. Optimizing perfusion parameters and oxygen delivery helps limit I/R injury, while biocompatible circuit coatings reduce leukocyte and platelet activation. Ultrafiltration during CPB can remove circulating inflammatory mediators, and newer approaches such as cytokine adsorption devices have shown promise in selectively reducing both cytokines and DAMP-related mediators from the circulation [128].

7.4. Ischemia–Reperfusion Injury

I/R injury is encountered not only in cardiac and vascular surgery but also in transplantation, abdominal procedures, and operations using tourniquets. The magnitude of injury can be influenced by perioperative management. Key strategies include minimizing the duration of ischemia, maintaining adequate perfusion and oxygen delivery, and avoiding sustained hypotension at critical periods. Experimental approaches such as ischemic preconditioning, postconditioning, and volatile anesthetic preconditioning have shown potential but remain controversial, with inconsistent results in clinical trials. At present, the most reliable preventive measures remain careful intraoperative monitoring, timely restoration of blood flow, and optimization of hemodynamics to reduce the burden of reperfusion-related inflammation.

7.5. Monitoring and Risk Stratification

Current perioperative risk assessment relies primarily on clinical evaluation and physiologic monitoring. However, DAMPs are increasingly recognized as potential biomarkers of perioperative risk. Elevated levels of HMGB1, S100 proteins, mtDNA, and histones have been associated with postoperative organ dysfunction and often precede clinical deterioration. Although measurement of these biomarkers is not yet part of routine practice, ongoing research suggests that they could contribute to earlier detection of complications and more refined risk stratification [34].
For example, in patients undergoing surgical aortic valve replacement with cardiopulmonary bypass, plasma mtDNA levels increased more than tenfold and were associated with postoperative bleeding, hepatic dysfunction, and prolonged hospitalization [129]. Similarly, postoperative increases in circulating mtDNA were correlated with the development of systemic inflammatory response syndrome and multiple organ dysfunction following pancreaticoduodenectomy [130]. Even though specific cutoffs remain undefined for clinical application, these studies demonstrate the potential of perioperative DAMP levels as biomarkers for monitoring inflammatory stress and improving risk stratification.

8. Future Directions

Much of the current knowledge on DAMPs comes from in vitro experiments and preclinical animal models. These studies have provided valuable mechanistic insights, but their direct translation to the clinical perioperative setting remains challenging. The complexity of surgical care, patient heterogeneity, and the interplay of multiple overlapping inflammatory pathways limit the extrapolation of experimental findings. Moreover, clinical studies measuring perioperative DAMPs are often small, methodologically heterogeneous, and rely on non-standardized assays, making cross-study comparisons difficult.
Future research should aim to bridge these gaps by clarifying how DAMP biology can move from mechanistic insight to clinical application. Large, well-designed studies are needed to validate perioperative DAMPs as reliable biomarkers for risk stratification and early detection of complications. Interventions that target DAMP release, signaling, or clearance, such as pharmacologic inhibitors, extracorporeal adsorption techniques, or tailored anesthetic regimens, hold promise but require rigorous clinical evaluation. Ultimately, integrating DAMP biology into perioperative care may enable more personalized anesthetic strategies, reduce postoperative morbidity, and strengthen the role of anesthesiologists in advancing perioperative inflammation and recovery science.

9. Conclusions

DAMPs represent upstream mediators that bridge perioperative cellular injury to systemic inflammation and organ dysfunction. Recognizing their role reframes postoperative complications not only as sequelae of hemodynamic or infectious events but also as consequences of sterile inflammation triggered by surgical and anesthetic stress.
For anesthesiologists, this perspective provides a mechanistic rationale for existing practices such as protective ventilation, restrictive transfusion strategies, and careful ischemia–reperfusion management, while also highlighting emerging opportunities for innovation in monitoring and intervention. Translating DAMP biology into perioperative care requires rigorous clinical validation, standardized measurement approaches, and integration with established risk stratification tools. Ultimately, embedding DAMPs into perioperative practice may facilitate more personalized anesthetic management and improved postoperative outcomes.

Author Contributions

Conceptualization, K.Y. and W.M.; writing—original draft preparation, W.M.; writing—review and editing, K.Y. and W.M. All authors have read and agreed to the published version of the manuscript.

Funding

This is in part supported by R21HD109119 (K.Y.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We did not use any artificial intelligence tools in the preparation of this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Land, W.G. The Role of Damage-Associated Molecular Patterns (DAMPs) in Human Diseases: Part II: DAMPs as diagnostics, prognostics and therapeutics in clinical medicine. Sultan Qaboos Univ. Med. J. 2015, 15, e157–e170. [Google Scholar] [CrossRef]
  2. Matzinger, P. Tolerance, danger, and the extended family. Annu. Rev. Immunol. 1994, 12, 991–1045. [Google Scholar] [CrossRef]
  3. Maisat, W.; Yuki, K. Narrative review of systemic inflammatory response mechanisms in cardiac surgery and immunomodulatory role of anesthetic agents. Ann. Card. Anaesth. 2023, 26, 133–142. [Google Scholar] [CrossRef]
  4. Chen, G.Y.; Nuñez, G. Sterile inflammation: Sensing and reacting to damage. Nat. Rev. Immunol. 2010, 10, 826–837. [Google Scholar] [CrossRef]
  5. Mogensen, T.H. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin. Microbiol. Rev. 2009, 22, 240–273. [Google Scholar] [CrossRef]
  6. Janeway, C.A., Jr. The immune system evolved to discriminate infectious nonself from noninfectious self. Immunol. Today 1992, 13, 11–16. [Google Scholar] [CrossRef]
  7. Matzinger, P. The danger model: A renewed sense of self. Science 2002, 296, 301–305. [Google Scholar] [CrossRef]
  8. Shi, Y.; Evans, J.E.; Rock, K.L. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature 2003, 425, 516–521. [Google Scholar] [CrossRef] [PubMed]
  9. Scaffidi, P.; Misteli, T.; Bianchi, M.E. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 2002, 418, 191–195. [Google Scholar] [CrossRef] [PubMed]
  10. Bianchi, M.E. DAMPs, PAMPs and alarmins: All we need to know about danger. J. Leukoc. Biol. 2007, 81, 1–5. [Google Scholar] [CrossRef] [PubMed]
  11. Land, W.G. Transfusion-Related Acute Lung Injury: The Work of DAMPs. Transfus. Med. Hemother. 2013, 40, 3–13. [Google Scholar] [CrossRef]
  12. Yu, M.; Wang, H.; Ding, A.; Golenbock, D.T.; Latz, E.; Czura, C.J.; Fenton, M.J.; Tracey, K.J.; Yang, H. HMGB1 signals through toll-like receptor (TLR) 4 and TLR2. Shock 2006, 26, 174–179. [Google Scholar] [CrossRef]
  13. Chen, R.; Kang, R.; Tang, D. The mechanism of HMGB1 secretion and release. Exp. Mol. Med. 2022, 54, 91–102. [Google Scholar] [CrossRef]
  14. Silk, E.; Zhao, H.; Weng, H.; Ma, D. The role of extracellular histone in organ injury. Cell Death Dis. 2017, 8, e2812. [Google Scholar] [CrossRef]
  15. Apel, F.; Andreeva, L.; Knackstedt, L.S.; Streeck, R.; Frese, C.K.; Goosmann, C.; Hopfner, K.P.; Zychlinsky, A. The cytosolic DNA sensor cGAS recognizes neutrophil extracellular traps. Sci. Signal 2021, 14, eaax7942. [Google Scholar] [CrossRef]
  16. Pisetsky, D.S. The origin and properties of extracellular DNA: From PAMP to DAMP. Clin. Immunol. 2012, 144, 32–40. [Google Scholar] [CrossRef]
  17. Sandler, N.; Kaczmarek, E.; Itagaki, K.; Zheng, Y.; Otterbein, L.; Khabbaz, K.; Liu, D.; Senthilnathan, V.; Gruen, R.L.; Hauser, C.J. Mitochondrial DAMPs Are Released During Cardiopulmonary Bypass Surgery and Are Associated with Postoperative Atrial Fibrillation. Heart Lung Circ. 2018, 27, 122–129. [Google Scholar] [CrossRef] [PubMed]
  18. Amores-Iniesta, J.; Barberà-Cremades, M.; Martínez, C.M.; Pons, J.A.; Revilla-Nuin, B.; Martínez-Alarcón, L.; Di Virgilio, F.; Parrilla, P.; Baroja-Mazo, A.; Pelegrín, P. Extracellular ATP Activates the NLRP3 Inflammasome and Is an Early Danger Signal of Skin Allograft Rejection. Cell Rep. 2017, 21, 3414–3426. [Google Scholar] [CrossRef] [PubMed]
  19. Dorward, D.A.; Lucas, C.D.; Chapman, G.B.; Haslett, C.; Dhaliwal, K.; Rossi, A.G. The role of formylated peptides and formyl peptide receptor 1 in governing neutrophil function during acute inflammation. Am. J. Pathol. 2015, 185, 1172–1184. [Google Scholar] [CrossRef]
  20. Ma, L.; Sun, P.; Zhang, J.C.; Zhang, Q.; Yao, S.L. Proinflammatory effects of S100A8/A9 via TLR4 and RAGE signaling pathways in BV-2 microglial cells. Int. J. Mol. Med. 2017, 40, 31–38. [Google Scholar] [CrossRef] [PubMed]
  21. Braga, T.T.; Forni, M.F.; Correa-Costa, M.; Ramos, R.N.; Barbuto, J.A.; Branco, P.; Castoldi, A.; Hiyane, M.I.; Davanso, M.R.; Latz, E.; et al. Soluble Uric Acid Activates the NLRP3 Inflammasome. Sci. Rep. 2017, 7, 39884. [Google Scholar] [CrossRef] [PubMed]
  22. Roedig, H.; Nastase, M.V.; Frey, H.; Moreth, K.; Zeng-Brouwers, J.; Poluzzi, C.; Hsieh, L.T.; Brandts, C.; Fulda, S.; Wygrecka, M.; et al. Biglycan is a new high-affinity ligand for CD14 in macrophages. Matrix Biol. 2019, 77, 4–22. [Google Scholar] [CrossRef]
  23. Oshima, K.; Haeger, S.M.; Hippensteel, J.A.; Herson, P.S.; Schmidt, E.P. More than a biomarker: The systemic consequences of heparan sulfate fragments released during endothelial surface layer degradation (2017 Grover Conference Series). Pulm. Circ. 2018, 8, 2045893217745786. [Google Scholar] [CrossRef] [PubMed]
  24. Takeuchi, O.; Akira, S. Pattern recognition receptors and inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef]
  25. Tang, D.; Kang, R.; Berghe, T.V.; Vandenabeele, P.; Kroemer, G. The molecular machinery of regulated cell death. Cell Res. 2019, 29, 347–364. [Google Scholar] [CrossRef]
  26. Murao, A.; Aziz, M.; Wang, H.; Brenner, M.; Wang, P. Release mechanisms of major DAMPs. Apoptosis 2021, 26, 152–162. [Google Scholar] [CrossRef]
  27. Eefting, F.; Rensing, B.; Wigman, J.; Pannekoek, W.J.; Liu, W.M.; Cramer, M.J.; Lips, D.J.; Doevendans, P.A. Role of apoptosis in reperfusion injury. Cardiovasc. Res. 2004, 61, 414–426. [Google Scholar] [CrossRef] [PubMed]
  28. Bell, C.W.; Jiang, W.; Reich, C.F., III; Pisetsky, D.S. The extracellular release of HMGB1 during apoptotic cell death. Am. J. Physiol. Cell Physiol. 2006, 291, C1318–C1325. [Google Scholar] [CrossRef]
  29. Delogu, G.; Moretti, S.; Antonucci, A.; Marcellini, S.; Masciangelo, R.; Famularo, G.; Signore, L.; De Simone, C. Apoptosis and surgical trauma: Dysregulated expression of death and survival factors on peripheral lymphocytes. Arch. Surg. 2000, 135, 1141–1147. [Google Scholar] [CrossRef]
  30. Denton, D.; Kumar, S. Autophagy-dependent cell death. Cell Death Differ. 2019, 26, 605–616. [Google Scholar] [CrossRef]
  31. Yu, P.; Zhang, X.; Liu, N.; Tang, L.; Peng, C.; Chen, X. Pyroptosis: Mechanisms and diseases. Signal Transduct. Target. Ther. 2021, 6, 128. [Google Scholar] [CrossRef] [PubMed]
  32. Kaczmarek, A.; Vandenabeele, P.; Krysko, D.V. Necroptosis: The release of damage-associated molecular patterns and its physiological relevance. Immunity 2013, 38, 209–223. [Google Scholar] [CrossRef] [PubMed]
  33. Davaanyam, D.; Lee, H.; Seol, S.I.; Oh, S.A.; Kim, S.W.; Lee, J.K. HMGB1 induces hepcidin upregulation in astrocytes and causes an acute iron surge and subsequent ferroptosis in the postischemic brain. Exp. Mol. Med. 2023, 55, 2402–2416. [Google Scholar] [CrossRef]
  34. Maisat, W.; Hou, L.; Sandhu, S.; Sin, Y.C.; Kim, S.; Van Pelt, H.; Chen, Y.; Emani, S.; Kong, S.W.; Emani, S.; et al. Neutrophil extracellular traps formation is associated with postoperative complications in congenital cardiac surgery. Pediatr. Res. 2024, 98, 1545–1558. [Google Scholar] [CrossRef]
  35. Caudrillier, A.; Kessenbrock, K.; Gilliss, B.M.; Nguyen, J.X.; Marques, M.B.; Monestier, M.; Toy, P.; Werb, Z.; Looney, M.R. Platelets induce neutrophil extracellular traps in transfusion-related acute lung injury. J. Clin. Investig. 2012, 122, 2661–2671. [Google Scholar] [CrossRef]
  36. Colicchia, M.; Perrella, G.; Gant, P.; Rayes, J. Novel mechanisms of thrombo-inflammation during infection: Spotlight on neutrophil extracellular trap-mediated platelet activation. Res. Pract. Thromb. Haemost. 2023, 7, 100116. [Google Scholar] [CrossRef] [PubMed]
  37. Salvadori, M.; Rosso, G.; Bertoni, E. Update on ischemia-reperfusion injury in kidney transplantation: Pathogenesis and treatment. World J. Transpl. 2015, 5, 52–67. [Google Scholar] [CrossRef]
  38. Lasorsa, F.; Rutigliano, M.; Milella, M.; d’Amati, A.; Crocetto, F.; Pandolfo, S.D.; Barone, B.; Ferro, M.; Spilotros, M.; Battaglia, M.; et al. Ischemia-Reperfusion Injury in Kidney Transplantation: Mechanisms and Potential Therapeutic Targets. Int. J. Mol. Sci. 2024, 25, 4332. [Google Scholar] [CrossRef]
  39. Silvis, M.J.M.; Kaffka Genaamd Dengler, S.E.; Odille, C.A.; Mishra, M.; van der Kaaij, N.P.; Doevendans, P.A.; Sluijter, J.P.G.; de Kleijn, D.P.V.; de Jager, S.C.A.; Bosch, L.; et al. Damage-Associated Molecular Patterns in Myocardial Infarction and Heart Transplantation: The Road to Translational Success. Front. Immunol. 2020, 11, 599511. [Google Scholar] [CrossRef]
  40. Squiccimarro, E.; Stasi, A.; Lorusso, R.; Paparella, D. Narrative review of the systemic inflammatory reaction to cardiac surgery and cardiopulmonary bypass. Artif. Organs 2022, 46, 568–577. [Google Scholar] [CrossRef]
  41. Dybdahl, B.; Wahba, A.; Lien, E.; Flo, T.H.; Waage, A.; Qureshi, N.; Sellevold, O.F.; Espevik, T.; Sundan, A. Inflammatory response after open heart surgery: Release of heat-shock protein 70 and signaling through toll-like receptor-4. Circulation 2002, 105, 685–690. [Google Scholar] [CrossRef]
  42. Kim-Shapiro, D.B.; Lee, J.; Gladwin, M.T. Storage lesion: Role of red blood cell breakdown. Transfusion 2011, 51, 844–851. [Google Scholar] [CrossRef]
  43. Yoshida, T.; Prudent, M.; D’Alessandro, A. Red blood cell storage lesion: Causes and potential clinical consequences. Blood Transfus. 2019, 17, 27–52. [Google Scholar] [CrossRef]
  44. Peltz, E.D.; Moore, E.E.; Eckels, P.C.; Damle, S.S.; Tsuruta, Y.; Johnson, J.L.; Sauaia, A.; Silliman, C.C.; Banerjee, A.; Abraham, E. HMGB1 is markedly elevated within 6 hours of mechanical trauma in humans. Shock 2009, 32, 17–22. [Google Scholar] [CrossRef]
  45. Neal, M.D.; Raval, J.S.; Triulzi, D.J.; Simmons, R.L. Innate immune activation after transfusion of stored red blood cells. Transfus. Med. Rev. 2013, 27, 113–118. [Google Scholar] [CrossRef]
  46. Wei, W.; Sun, Z.; He, S.; Zhang, W.; Chen, S.; Cao, Y.N.; Wang, N. Mechanical ventilation induces lung and brain injury through ATP production, P2Y1 receptor activation and dopamine release. Bioengineered 2022, 13, 2346–2359. [Google Scholar] [CrossRef] [PubMed]
  47. Plötz, F.B.; Slutsky, A.S.; van Vught, A.J.; Heijnen, C.J. Ventilator-induced lung injury and multiple system organ failure: A critical review of facts and hypotheses. Intensive Care Med. 2004, 30, 1865–1872. [Google Scholar] [CrossRef] [PubMed]
  48. Kuipers, M.T.; van der Poll, T.; Schultz, M.J.; Wieland, C.W. Bench-to-bedside review: Damage-associated molecular patterns in the onset of ventilator-induced lung injury. Crit. Care 2011, 15, 235. [Google Scholar] [CrossRef] [PubMed]
  49. Shao, Y.; Saredy, J.; Yang, W.Y.; Sun, Y.; Lu, Y.; Saaoud, F.; Drummer, C.T.; Johnson, C.; Xu, K.; Jiang, X.; et al. Vascular Endothelial Cells and Innate Immunity. Arter. Thromb. Vasc. Biol. 2020, 40, e138–e152. [Google Scholar] [CrossRef]
  50. Sun, S.; Sursal, T.; Adibnia, Y.; Zhao, C.; Zheng, Y.; Li, H.; Otterbein, L.E.; Hauser, C.J.; Itagaki, K. Mitochondrial DAMPs increase endothelial permeability through neutrophil dependent and independent pathways. PLoS ONE 2013, 8, e59989. [Google Scholar] [CrossRef]
  51. Singh, V.; Kaur, R.; Kumari, P.; Pasricha, C.; Singh, R. ICAM-1 and VCAM-1: Gatekeepers in various inflammatory and cardiovascular disorders. Clin. Chim. Acta 2023, 548, 117487. [Google Scholar] [CrossRef] [PubMed]
  52. Riedel, B.; Schier, R. Endothelial dysfunction in the perioperative setting. Semin. Cardiothorac. Vasc. Anesth. 2010, 14, 41–43. [Google Scholar] [CrossRef]
  53. Biswas, I.; Panicker, S.R.; Cai, X.; Mehta-D’souza, P.; Rezaie, A.R. Inorganic Polyphosphate Amplifies High Mobility Group Box 1-Mediated Von Willebrand Factor Release and Platelet String Formation on Endothelial Cells. Arter. Thromb. Vasc. Biol. 2018, 38, 1868–1877. [Google Scholar] [CrossRef]
  54. Ito, T.; Kawahara, K.; Nakamura, T.; Yamada, S.; Nakamura, T.; Abeyama, K.; Hashiguchi, T.; Maruyama, I. High-mobility group box 1 protein promotes development of microvascular thrombosis in rats. J. Thromb. Haemost. 2007, 5, 109–116. [Google Scholar] [CrossRef]
  55. Bhagirath, V.C.; Dwivedi, D.J.; Liaw, P.C. Comparison of the Proinflammatory and Procoagulant Properties of Nuclear, Mitochondrial, and Bacterial DNA. Shock 2015, 44, 265–271. [Google Scholar] [CrossRef]
  56. Wang, Y.; Fang, C.; Gao, H.; Bilodeau, M.L.; Zhang, Z.; Croce, K.; Liu, S.; Morooka, T.; Sakuma, M.; Nakajima, K.; et al. Platelet-derived S100 family member myeloid-related protein-14 regulates thrombosis. J. Clin. Investig. 2014, 124, 2160–2171. [Google Scholar] [CrossRef]
  57. Shimono, K.; Ito, T.; Kamikokuryo, C.; Niiyama, S.; Yamada, S.; Onishi, H.; Yoshihara, H.; Maruyama, I.; Kakihana, Y. Damage-associated molecular patterns and fibrinolysis perturbation are associated with lethal outcomes in traumatic injury. Thromb. J. 2023, 21, 91. [Google Scholar] [CrossRef]
  58. Gould, T.J.; Vu, T.T.; Stafford, A.R.; Dwivedi, D.J.; Kim, P.Y.; Fox-Robichaud, A.E.; Weitz, J.I.; Liaw, P.C. Cell-Free DNA Modulates Clot Structure and Impairs Fibrinolysis in Sepsis. Arter. Thromb. Vasc. Biol. 2015, 35, 2544–2553. [Google Scholar] [CrossRef]
  59. Rahbar, E.; Cardenas, J.C.; Baimukanova, G.; Usadi, B.; Bruhn, R.; Pati, S.; Ostrowski, S.R.; Johansson, P.I.; Holcomb, J.B.; Wade, C.E. Endothelial glycocalyx shedding and vascular permeability in severely injured trauma patients. J. Transl. Med. 2015, 13, 117. [Google Scholar] [CrossRef] [PubMed]
  60. Ito, T.; Totoki, T.; Yokoyama, Y.; Yasuda, T.; Furubeppu, H.; Yamada, S.; Maruyama, I.; Kakihana, Y. Serum histone H3 levels and platelet counts are potential markers for coagulopathy with high risk of death in septic patients: A single-center observational study. J. Intensive Care 2019, 7, 63. [Google Scholar] [CrossRef] [PubMed]
  61. Mao, J.Y.; Li, D.K.; Zhang, H.M.; Wang, X.T.; Liu, D.W. Plasma mitochondrial DNA levels are associated with acute lung injury and mortality in septic patients. BMC Pulm. Med. 2021, 21, 66. [Google Scholar] [CrossRef]
  62. Zhao, C.; Liang, F.; Ye, M.; Wu, S.; Qin, Y.; Zhao, L.; Zhang, L.; He, J.; Cen, L.; Lin, F. GSDMD promotes neutrophil extracellular traps via mtDNA-cGAS-STING pathway during lung ischemia/reperfusion. Cell Death Discov. 2023, 9, 368. [Google Scholar] [CrossRef]
  63. Peng, W.; Peng, F.; Lou, Y.; Li, Y.; Zhao, N.; Shao, Q.; Chen, J.; Qian, K.; Zeng, Z.; Zhan, Y.; et al. Autophagy alleviates mitochondrial DAMP-induced acute lung injury by inhibiting NLRP3 inflammasome. Life Sci. 2021, 265, 118833. [Google Scholar] [CrossRef]
  64. Zhang, Q.; Raoof, M.; Chen, Y.; Sumi, Y.; Sursal, T.; Junger, W.; Brohi, K.; Itagaki, K.; Hauser, C.J. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 2010, 464, 104–107. [Google Scholar] [CrossRef] [PubMed]
  65. van Zoelen, M.A.; Ishizaka, A.; Wolthuls, E.K.; Choi, G.; van der Poll, T.; Schultz, M.J. Pulmonary levels of high-mobility group box 1 during mechanical ventilation and ventilator-associated pneumonia. Shock 2008, 29, 441–445. [Google Scholar] [CrossRef] [PubMed]
  66. Hou, L.; Yang, Z.; Wang, Z.; Zhang, X.; Zhao, Y.; Yang, H.; Zheng, B.; Tian, W.; Wang, S.; He, Z.; et al. NLRP3/ASC-mediated alveolar macrophage pyroptosis enhances HMGB1 secretion in acute lung injury induced by cardiopulmonary bypass. Lab. Investig. 2018, 98, 1052–1064. [Google Scholar] [CrossRef] [PubMed]
  67. Deng, Y.; Hou, L.; Xu, Q.; Liu, Q.; Pan, S.; Gao, Y.; Dixon, R.A.F.; He, Z.; Wang, X. Cardiopulmonary Bypass Induces Acute Lung Injury via the High-Mobility Group Box 1/Toll-Like Receptor 4 Pathway. Dis. Markers 2020, 2020, 8854700. [Google Scholar] [CrossRef]
  68. Zhan, Y.; Ling, Y.; Deng, Q.; Qiu, Y.; Shen, J.; Lai, H.; Chen, Z.; Huang, C.; Liang, L.; Li, X.; et al. HMGB1-Mediated Neutrophil Extracellular Trap Formation Exacerbates Intestinal Ischemia/Reperfusion-Induced Acute Lung Injury. J. Immunol. 2022, 208, 968–978. [Google Scholar] [CrossRef]
  69. Zhang, Y.; Guan, L.; Yu, J.; Zhao, Z.; Mao, L.; Li, S.; Zhao, J. Pulmonary endothelial activation caused by extracellular histones contributes to neutrophil activation in acute respiratory distress syndrome. Respir. Res. 2016, 17, 155. [Google Scholar] [CrossRef]
  70. Yu, J.; Fu, Y.; Gao, J.; Zhang, Q.; Zhang, N.; Zhang, Z.; Jiang, X.; Chen, C.; Wen, Z. Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury. Redox Biol. 2024, 74, 103231. [Google Scholar] [CrossRef]
  71. Wittkowski, H.; Sturrock, A.; van Zoelen, M.A.; Viemann, D.; van der Poll, T.; Hoidal, J.R.; Roth, J.; Foell, D. Neutrophil-derived S100A12 in acute lung injury and respiratory distress syndrome. Crit. Care Med. 2007, 35, 1369–1375. [Google Scholar] [CrossRef]
  72. Zhao, B.; Lu, R.; Chen, J.; Xie, M.; Zhao, X.; Kong, L. S100A9 blockade prevents lipopolysaccharide-induced lung injury via suppressing the NLRP3 pathway. Respir. Res. 2021, 22, 45. [Google Scholar] [CrossRef]
  73. Alhamdi, Y.; Abrams, S.T.; Cheng, Z.; Jing, S.; Su, D.; Liu, Z.; Lane, S.; Welters, I.; Wang, G.; Toh, C.H. Circulating Histones Are Major Mediators of Cardiac Injury in Patients with Sepsis. Crit. Care Med. 2015, 43, 2094–2103. [Google Scholar] [CrossRef]
  74. Shah, M.; He, Z.; Rauf, A.; Beikoghli Kalkhoran, S.; Heiestad, C.M.; Stensløkken, K.O.; Parish, C.R.; Soehnlein, O.; Arjun, S.; Davidson, S.M.; et al. Extracellular histones are a target in myocardial ischaemia-reperfusion injury. Cardiovasc. Res. 2022, 118, 1115–1125. [Google Scholar] [CrossRef]
  75. Chen, R.; Duan, J.; Zhou, Y.; Cao, M.; Su, Y.; Jiang, D.; Tao, A.; Yuan, W.; Dai, Z. Endothelial HMGB1-AIM2 axis worsens myocardial ischemia-reperfusion injury by regulating endothelial pyroptosis. Apoptosis 2025, 30, 2466–2479. [Google Scholar] [CrossRef] [PubMed]
  76. Andrassy, M.; Volz, H.C.; Igwe, J.C.; Funke, B.; Eichberger, S.N.; Kaya, Z.; Buss, S.; Autschbach, F.; Pleger, S.T.; Lukic, I.K.; et al. High-mobility group box-1 in ischemia-reperfusion injury of the heart. Circulation 2008, 117, 3216–3226. [Google Scholar] [CrossRef]
  77. Ding, H.S.; Yang, J.; Chen, P.; Yang, J.; Bo, S.Q.; Ding, J.W.; Yu, Q.Q. The HMGB1-TLR4 axis contributes to myocardial ischemia/reperfusion injury via regulation of cardiomyocyte apoptosis. Gene 2013, 527, 389–393. [Google Scholar] [CrossRef] [PubMed]
  78. Li, Y.; Chen, B.; Yang, X.; Zhang, C.; Jiao, Y.; Li, P.; Liu, Y.; Li, Z.; Qiao, B.; Bond Lau, W.; et al. S100a8/a9 Signaling Causes Mitochondrial Dysfunction and Cardiomyocyte Death in Response to Ischemic/Reperfusion Injury. Circulation 2019, 140, 751–764. [Google Scholar] [CrossRef]
  79. Manghelli, J.L.; Kelly, M.O.; Carter, D.I.; Gauthier, J.M.; Scozzi, D.; Lancaster, T.S.; MacGregor, R.M.; Khiabani, A.J.; Schuessler, R.B.; Gelman, A.E.; et al. Pericardial Mitochondrial DNA Levels Are Associated with Atrial Fibrillation After Cardiac Surgery. Ann. Thorac. Surg. 2021, 111, 1593–1600. [Google Scholar] [CrossRef] [PubMed]
  80. Qu, C.; Wang, X.W.; Huang, C.; Qiu, F.; Xiang, X.Y.; Lu, Z.Q. High mobility group box 1 gene polymorphism is associated with the risk of postoperative atrial fibrillation after coronary artery bypass surgery. J. Cardiothorac. Surg. 2015, 10, 88. [Google Scholar] [CrossRef]
  81. Wu, H.; Ma, J.; Wang, P.; Corpuz, T.M.; Panchapakesan, U.; Wyburn, K.R.; Chadban, S.J. HMGB1 contributes to kidney ischemia reperfusion injury. J. Am. Soc. Nephrol. 2010, 21, 1878–1890. [Google Scholar] [CrossRef]
  82. Lau, A.; Wang, S.; Liu, W.; Haig, A.; Zhang, Z.X.; Jevnikar, A.M. Glycyrrhizic acid ameliorates HMGB1-mediated cell death and inflammation after renal ischemia reperfusion injury. Am. J. Nephrol. 2014, 40, 84–95. [Google Scholar] [CrossRef]
  83. Hu, Q.; Ren, J.; Wu, J.; Li, G.; Wu, X.; Liu, S.; Wang, G.; Gu, G.; Ren, H.; Hong, Z.; et al. Urinary Mitochondrial DNA Levels Identify Acute Kidney Injury in Surgical Critical Illness Patients. Shock 2017, 48, 11–17. [Google Scholar] [CrossRef] [PubMed]
  84. Whitaker, R.M.; Stallons, L.J.; Kneff, J.E.; Alge, J.L.; Harmon, J.L.; Rahn, J.J.; Arthur, J.M.; Beeson, C.C.; Chan, S.L.; Schnellmann, R.G. Urinary mitochondrial DNA is a biomarker of mitochondrial disruption and renal dysfunction in acute kidney injury. Kidney Int. 2015, 88, 1336–1344. [Google Scholar] [CrossRef]
  85. Nakazawa, D.; Kumar, S.V.; Marschner, J.; Desai, J.; Holderied, A.; Rath, L.; Kraft, F.; Lei, Y.; Fukasawa, Y.; Moeckel, G.W.; et al. Histones and Neutrophil Extracellular Traps Enhance Tubular Necrosis and Remote Organ Injury in Ischemic AKI. J. Am. Soc. Nephrol. 2017, 28, 1753–1768. [Google Scholar] [CrossRef] [PubMed]
  86. Tsung, A.; Sahai, R.; Tanaka, H.; Nakao, A.; Fink, M.P.; Lotze, M.T.; Yang, H.; Li, J.; Tracey, K.J.; Geller, D.A.; et al. The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J. Exp. Med. 2005, 201, 1135–1143. [Google Scholar] [CrossRef] [PubMed]
  87. Fuchs, T.A.; Bhandari, A.A.; Wagner, D.D. Histones induce rapid and profound thrombocytopenia in mice. Blood 2011, 118, 3708–3714. [Google Scholar] [CrossRef]
  88. Nakahara, M.; Ito, T.; Kawahara, K.; Yamamoto, M.; Nagasato, T.; Shrestha, B.; Yamada, S.; Miyauchi, T.; Higuchi, K.; Takenaka, T.; et al. Recombinant thrombomodulin protects mice against histone-induced lethal thromboembolism. PLoS ONE 2013, 8, e75961. [Google Scholar] [CrossRef]
  89. Hatada, T.; Wada, H.; Nobori, T.; Okabayashi, K.; Maruyama, K.; Abe, Y.; Uemoto, S.; Yamada, S.; Maruyama, I. Plasma concentrations and importance of High Mobility Group Box protein in the prognosis of organ failure in patients with disseminated intravascular coagulation. Thromb. Haemost. 2005, 94, 975–979. [Google Scholar] [CrossRef]
  90. Lin, G.X.; Wang, T.; Chen, M.H.; Hu, Z.H.; Ouyang, W. Serum high-mobility group box 1 protein correlates with cognitive decline after gastrointestinal surgery. Acta Anaesthesiol. Scand. 2014, 58, 668–674. [Google Scholar] [CrossRef]
  91. He, H.J.; Wang, Y.; Le, Y.; Duan, K.M.; Yan, X.B.; Liao, Q.; Liao, Y.; Tong, J.B.; Terrando, N.; Ouyang, W. Surgery upregulates high mobility group box-1 and disrupts the blood-brain barrier causing cognitive dysfunction in aged rats. CNS Neurosci. Ther. 2012, 18, 994–1002. [Google Scholar] [CrossRef] [PubMed]
  92. Li, R.L.; Zhang, Z.Z.; Peng, M.; Wu, Y.; Zhang, J.J.; Wang, C.Y.; Wang, Y.L. Postoperative impairment of cognitive function in old mice: A possible role for neuroinflammation mediated by HMGB1, S100B, and RAGE. J. Surg. Res. 2013, 185, 815–824. [Google Scholar] [CrossRef]
  93. Yang, N.S.; Zhong, W.J.; Sha, H.X.; Zhang, C.Y.; Jin, L.; Duan, J.X.; Xiong, J.B.; You, Z.J.; Zhou, Y.; Guan, C.X. mtDNA-cGAS-STING axis-dependent NLRP3 inflammasome activation contributes to postoperative cognitive dysfunction induced by sevoflurane in mice. Int. J. Biol. Sci. 2024, 20, 1927–1946. [Google Scholar] [CrossRef]
  94. Lu, S.M.; Yu, C.J.; Liu, Y.H.; Dong, H.Q.; Zhang, X.; Zhang, S.S.; Hu, L.Q.; Zhang, F.; Qian, Y.N.; Gui, B. S100A8 contributes to postoperative cognitive dysfunction in mice undergoing tibial fracture surgery by activating the TLR4/MyD88 pathway. Brain Behav. Immun. 2015, 44, 221–234. [Google Scholar] [CrossRef]
  95. Su, T.; Si, Y. PCSK9 exacerbates sevoflurane-induced neuroinflammatory response and apoptosis by up-regulating cGAS-STING signal. Tissue Cell 2025, 93, 102739. [Google Scholar] [CrossRef] [PubMed]
  96. Joe, Y.E.; Jun, J.H.; Oh, J.E.; Lee, J.R. Damage-associated molecular patterns as a mechanism of sevoflurane-induced neuroinflammation in neonatal rodents. Korean J. Anesth. 2024, 77, 468–479. [Google Scholar] [CrossRef]
  97. Shan, Y.; Liu, P.; Zhou, Y.; Ding, X.; Liu, H.; Yang, J. Prenatal Sevoflurane Exposure Impairs the Learning and Memory of Rat Offspring via HMGB1-Induced NLRP3/ASC Inflammasome Activation. ACS Chem. Neurosci. 2023, 14, 699–708. [Google Scholar] [CrossRef] [PubMed]
  98. Xu, L.; Ge, F.; Hu, Y.; Yu, Y.; Guo, K.; Miao, C. Sevoflurane Postconditioning Attenuates Hepatic Ischemia-Reperfusion Injury by Limiting HMGB1/TLR4/NF-κB Pathway via Modulating microRNA-142 in vivo and in vitro. Front. Pharmacol. 2021, 12, 646307. [Google Scholar] [CrossRef]
  99. Zhao, Z.; Li, Y.; Chi, F.; Ma, L.; Li, Y.; Hou, Z.; Wang, Q. Sevoflurane postconditioning ameliorates cerebral ischemia-reperfusion injury in rats via TLR4/MyD88/TRAF6 signaling pathway. Aging 2022, 14, 10153–10170. [Google Scholar] [CrossRef]
  100. Shan, Y.; Xie, K.; Zhou, Q.; He, R.; Chen, Z.; Feng, W. Sevoflurane alleviates myocardial ischemia/reperfusion injury via actitation of heat shock protein-70 in patients undergoing double valve replacement surgery. Am. J. Transl. Res. 2022, 14, 5529–5540. [Google Scholar]
  101. Gao, T.; Huang, Z. Effects of Isoflurane on the Cell Pyroptosis in the Lung Cancer Through the HMGB1/RAGE Pathway. Appl. Biochem. Biotechnol. 2024, 196, 3786–3799. [Google Scholar] [CrossRef]
  102. Han, D.; Zhou, Z.; Liu, J.; Wang, T.; Yin, J. Neuroprotective effects of isoflurane against lipopolysaccharide-induced neuroinflammation in BV2 microglial cells by regulating HMGB1/TLRs pathway. Folia Neuropathol. 2020, 58, 57–69. [Google Scholar] [CrossRef]
  103. Sun, M.; Deng, B.; Zhao, X.; Gao, C.; Yang, L.; Zhao, H.; Yu, D.; Zhang, F.; Xu, L.; Chen, L.; et al. Isoflurane preconditioning provides neuroprotection against stroke by regulating the expression of the TLR4 signalling pathway to alleviate microglial activation. Sci. Rep. 2015, 5, 11445. [Google Scholar] [CrossRef] [PubMed]
  104. Bian, W.Y.; Chen, Y.P.; Xu, B.; Tang, J. Pretreatment with Propofol Reduces Pulmonary Injury in a Pig Model of Intestinal Ischemia-Reperfusion via Suppressing the High-Mobility Group Box 1 Protein (HMGB1)/Toll-Like Receptor 4 (TLR4)/Protein Kinase R (PKR) Signaling Pathway. Med. Sci. Monit. 2021, 27, e930478. [Google Scholar] [CrossRef]
  105. Feng, Z.; Wang, J.W.; Wang, Y.; Dong, W.W.; Xu, Z.F. Propofol Protects Lung Endothelial Barrier Function by Suppression of High-Mobility Group Box 1 (HMGB1) Release and Mitochondrial Oxidative Damage Catalyzed by HMGB1. Med. Sci. Monit. 2019, 25, 3199–3211. [Google Scholar] [CrossRef] [PubMed]
  106. Zhao, H.; Gu, Y.; Chen, H. Propofol ameliorates endotoxin-induced myocardial cell injury by inhibiting inflammation and apoptosis via the PPARγ/HMGB1/NLRP3 axis. Mol. Med. Rep. 2021, 23, 176. [Google Scholar] [CrossRef]
  107. Li, Y.M.; Sun, J.G.; Hu, L.H.; Ma, X.C.; Zhou, G.; Huang, X.Z. Propofol-mediated cardioprotection dependent of microRNA-451/HMGB1 against myocardial ischemia-reperfusion injury. J. Cell Physiol. 2019, 234, 23289–23301. [Google Scholar] [CrossRef] [PubMed]
  108. Wu, M.; Zhao, L.; Wang, Y.; Guo, Q.; An, Q.; Geng, J.; Zhang, C.; Guo, Z. Ketamine Regulates the Autophagy Flux and Polarization of Microglia through the HMGB1-RAGE Axis and Exerts Antidepressant Effects in Mice. J. Neuropathol. Exp. Neurol. 2022, 81, 931–942. [Google Scholar] [CrossRef]
  109. Xu, D.; Sun, X.; Zhang, Y.; Cao, L. Ketamine alleviates HMGB1-induced acute lung injury through TLR4 signaling pathway. Adv. Clin. Exp. Med. 2020, 29, 813–817. [Google Scholar] [CrossRef]
  110. Zhang, Y.; Zhang, M.; Wang, C.Y.; Shen, A. Ketamine alleviates LPS induced lung injury by inhibiting HMGB1-RAGE level. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 1830–1836. [Google Scholar] [CrossRef]
  111. Li, K.; Yang, J.; Han, X. Ketamine attenuates sepsis-induced acute lung injury via regulation of HMGB1-RAGE pathways. Int. Immunopharmacol. 2016, 34, 114–128. [Google Scholar] [CrossRef] [PubMed]
  112. Liu, Z.; Wang, Z.; Han, G.; Huang, L.; Jiang, J.; Li, S. Ketamine attenuates high mobility group box-1-induced inflammatory responses in endothelial cells. J. Surg. Res. 2016, 200, 593–603. [Google Scholar] [CrossRef]
  113. Chen, J.; Liang, S.; Li, C.; Li, B.; He, M.; Li, K.; Fu, W.; Li, S.; Mi, H. Mitochondrial damage causes inflammation via cGAS-STING signaling in ketamine-induced cystitis. Inflamm. Res. 2025, 74, 6. [Google Scholar] [CrossRef]
  114. Wang, J.; Xin, Y.; Chu, T.; Liu, C.; Xu, A. Dexmedetomidine attenuates perioperative neurocognitive disorders by suppressing hippocampal neuroinflammation and HMGB1/RAGE/NF-κB signaling pathway. Biomed. Pharmacother. 2022, 150, 113006. [Google Scholar] [CrossRef] [PubMed]
  115. Xiong, W.; Zhou, R.; Qu, Y.; Yang, Y.; Wang, Z.; Song, N.; Liang, R.; Qian, J. Dexmedetomidine preconditioning mitigates myocardial ischemia/reperfusion injury via inhibition of mast cell degranulation. Biomed. Pharmacother. 2021, 141, 111853. [Google Scholar] [CrossRef]
  116. Zhang, B.; Zhang, J.; Ainiwaer, Y.; He, B.; Geng, Q.; Lin, L.; Li, X. Dexmedetomidine Attenuates Myocardial Injury Induced by Renal Ischemia/Reperfusion by Inhibiting the HMGB1-TLR4-MyD88-NF-κB Signaling Pathway. Ann. Clin. Lab. Sci. 2021, 51, 376–384. [Google Scholar]
  117. Xiao, S.; Wang, Q.; Gao, H.; Zhao, X.; Zhi, J.; Yang, D. Dexmedetomidine alleviates airway hyperresponsiveness and allergic airway inflammation through the TLR4/NF-κB signaling pathway in mice. Mol. Med. Rep. 2022, 25, 74. [Google Scholar] [CrossRef]
  118. Sun, H.; Hu, H.; Xu, X.; Fang, M.; Tao, T.; Liang, Z. Protective effect of dexmedetomidine in cecal ligation perforation-induced acute lung injury through HMGB1/RAGE pathway regulation and pyroptosis activation. Bioengineered 2021, 12, 10608–10623. [Google Scholar] [CrossRef]
  119. Chen, Y.; Huang, Y.; Xiong, B.; Luo, H.; Song, X. Dexmedetomidine ameliorates renal ischemia reperfusion-mediated activation of the NLRP3 inflammasome in alveolar macrophages. Gene 2020, 758, 144973. [Google Scholar] [CrossRef]
  120. Chen, J.; Jiang, Z.; Zhou, X.; Sun, X.; Cao, J.; Liu, Y.; Wang, X. Dexmedetomidine Preconditioning Protects Cardiomyocytes Against Hypoxia/Reoxygenation-Induced Necroptosis by Inhibiting HMGB1-Mediated Inflammation. Cardiovasc. Drugs Ther. 2019, 33, 45–54. [Google Scholar] [CrossRef] [PubMed]
  121. Wu, W.; He, Y.; Lin, D.; Zhang, G.; Zhang, X.; Zhang, N.; Xie, T.; Wei, H. Dexmedetomidine mitigates lipopolysaccharide-induced acute lung injury by modulating heat shock protein A12B to inhibit the toll-like receptor 4/nuclear factor-kappa B signaling pathway. Chem. Biol. Interact. 2024, 398, 111112. [Google Scholar] [CrossRef]
  122. Yoo, S.H.; Jue, M.J.; Kim, Y.H.; Cho, S.; Kim, W.J.; Kim, K.M.; Han, J.I.; Lee, H. The Effect of Dexmedetomidine on the Mini-Cog Score and High-Mobility Group Box 1 Levels in Elderly Patients with Postoperative Neurocognitive Disorders Undergoing Orthopedic Surgery. J. Clin. Med. 2023, 12, 6610. [Google Scholar] [CrossRef]
  123. Mao, Q.; Liang, B.; Leng, Z.; Ma, W.; Chen, Y.; Xie, Y. Remimazolam ameliorates postoperative cognitive dysfunction after deep hypothermic circulatory arrest through HMGB1-TLR4-NF-κB pathway. Brain Res. Bull. 2024, 217, 111086. [Google Scholar] [CrossRef]
  124. Seo, K.H.; Choi, J.W.; Jung, H.S.; Yoo, H.; Joo, J.D. The Effects of Remifentanil on Expression of High Mobility Group Box 1 in Septic Rats. J. Korean Med. Sci. 2017, 32, 542–551. [Google Scholar] [CrossRef]
  125. Zhang, M.; Yin, Y. Dual roles of anesthetics in postoperative cognitive dysfunction: Regulation of microglial activation through inflammatory signaling pathways. Front. Immunol. 2023, 14, 1102312. [Google Scholar] [CrossRef]
  126. Behnaz, F.; Erfanian, M.; Chegini, A. Comparison of the Effects of Isoflurane and Propofol as Anesthesia Maintenance on Plasma Mitochondrial DNA Levels in Posterior Spinal Fusion Surgeries. Anesth. Pain Med. 2025, 15, e161767. [Google Scholar] [CrossRef] [PubMed]
  127. Weng, Y.; Yuan, S.; Li, H.; Yu, W. Comparison of Cardioprotective Effects of Propofol versus Sevoflurane in Pediatric Living Donor Liver Transplantation. Ann. Transpl. 2020, 25, e923398. [Google Scholar] [CrossRef] [PubMed]
  128. Graf, H.; Gräfe, C.; Bruegel, M.; Happich, F.L.; Wustrow, V.; Wegener, A.; Wilfert, W.; Zoller, M.; Liebchen, U.; Paal, M.; et al. Extracorporeal Elimination of Pro- and Anti-inflammatory Modulators by the Cytokine Adsorber CytoSorb® in Patients with Hyperinflammation: A Prospective Study. Infect. Dis. Ther. 2024, 13, 2089–2101. [Google Scholar] [CrossRef] [PubMed]
  129. Hyslop, K.; Ki, K.K.; Naidoo, R.; O’Brien, D.; Prabhu, A.; Gill, D.; Anstey, C.; Rapchuk, I.L.; McDonald, C.I.; Marshall, L.; et al. Cell-free mitochondrial DNA may predict the risk of post-operative complications and outcomes in surgical aortic valve replacement patients. Sci. Rep. 2025, 15, 15857. [Google Scholar] [CrossRef]
  130. Pencovich, N.; Nevo, N.; Weiser, R.; Bonder, E.; Bogoch, Y.; Nachmany, I. Postoperative Rise of Circulating Mitochondrial DNA Is Associated with Inflammatory Response in Patients following Pancreaticoduodenectomy. Eur. Surg. Res. 2021, 62, 18–24. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the sources and types damage-associated molecular patterns (DAMPs). (A) Endogenous DAMPs released from an injured cell. The figure illustrates the major subcellular origins of DAMPs, including mitochondrial DAMPs (mitochondrial DNA, extracellular ATP, N-formyl peptides), cytosolic DAMPs (heat shock proteins, S100 proteins, uric acid), and nuclear DAMPs (extracellular DNA, histones, HMGB1). Elements of the extracellular matrix such as biglycan, low-molecular-weight hyaluronan, and heparan sulfate are also shown as extracellular-derived DAMPs. (B) Transfusion-derived DAMPs associated with different blood products. Red blood cell units may contain cell-free hemoglobin and heme, mitochondrial and nuclear DNA fragments, microparticles, and ATP. Platelet units may release platelet-derived microparticles and HMGB1. Plasma products, including fresh frozen plasma, may contain plasma-derived microparticles. Explanation in the text. Created in BioRender. Maisat, W. (2025) https://BioRender.com/7ldsmk0.
Figure 1. Schematic representation of the sources and types damage-associated molecular patterns (DAMPs). (A) Endogenous DAMPs released from an injured cell. The figure illustrates the major subcellular origins of DAMPs, including mitochondrial DAMPs (mitochondrial DNA, extracellular ATP, N-formyl peptides), cytosolic DAMPs (heat shock proteins, S100 proteins, uric acid), and nuclear DAMPs (extracellular DNA, histones, HMGB1). Elements of the extracellular matrix such as biglycan, low-molecular-weight hyaluronan, and heparan sulfate are also shown as extracellular-derived DAMPs. (B) Transfusion-derived DAMPs associated with different blood products. Red blood cell units may contain cell-free hemoglobin and heme, mitochondrial and nuclear DNA fragments, microparticles, and ATP. Platelet units may release platelet-derived microparticles and HMGB1. Plasma products, including fresh frozen plasma, may contain plasma-derived microparticles. Explanation in the text. Created in BioRender. Maisat, W. (2025) https://BioRender.com/7ldsmk0.
Anesthres 03 00001 g001
Figure 2. Recognition of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) by the innate immune system. The innate immune system integrates signals from both exogenous (PAMPs) and endogenous (DAMPs) ligands through PRRs. PAMPs, such as lipopolysaccharide (LPS), lipoteichoic acid, flagellin, and viral RNA, arise from invading microorganisms (stranger model). DAMPs, including HMGB1, histones, mitochondrial DNA (mtDNA), extracellular ATP (eATP), and S100A8/A9, are released during sterile injury such as surgery, ischemia–reperfusion, cardiopulmonary bypass, and blood transfusion (danger signals). PRRs (e.g., Toll-like receptors, NOD-like receptors, RAGE) activate intracellular signaling cascades (e.g., MyD88, MAPK), leading to cytokine and chemokine release, immune cell recruitment, and amplification of inflammation. Controlled responses facilitate wound healing, debris clearance, and tissue repair, whereas excessive or dysregulated responses cause systemic inflammation, endothelial injury, and organ dysfunction. Explanation in the text. Created in BioRender. Maisat, W. (2025) https://BioRender.com/skorejb.
Figure 2. Recognition of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) by the innate immune system. The innate immune system integrates signals from both exogenous (PAMPs) and endogenous (DAMPs) ligands through PRRs. PAMPs, such as lipopolysaccharide (LPS), lipoteichoic acid, flagellin, and viral RNA, arise from invading microorganisms (stranger model). DAMPs, including HMGB1, histones, mitochondrial DNA (mtDNA), extracellular ATP (eATP), and S100A8/A9, are released during sterile injury such as surgery, ischemia–reperfusion, cardiopulmonary bypass, and blood transfusion (danger signals). PRRs (e.g., Toll-like receptors, NOD-like receptors, RAGE) activate intracellular signaling cascades (e.g., MyD88, MAPK), leading to cytokine and chemokine release, immune cell recruitment, and amplification of inflammation. Controlled responses facilitate wound healing, debris clearance, and tissue repair, whereas excessive or dysregulated responses cause systemic inflammation, endothelial injury, and organ dysfunction. Explanation in the text. Created in BioRender. Maisat, W. (2025) https://BioRender.com/skorejb.
Anesthres 03 00001 g002
Figure 3. Overview of major cell death pathways and their characteristic morphological features. Necrosis involves early swelling of cells and organelles followed by rupture of the plasma membrane. Apoptosis progresses through cell shrinkage and membrane blebbing, formation of apoptotic bodies, and phagocytosis by neighboring cells. When clearance is delayed or insufficient, apoptotic cells may undergo secondary necrosis. Autophagy is characterized by formation of an autophagosome that fuses with a lysosome to generate an autolysosome. Excessive or inadequate autophagy may lead to autophagy-dependent cell death. The lower panel illustrates forms of regulated inflammatory cell death. Pyroptosis features pore formation, cell swelling, and membrane rupture. Necroptosis is marked by programmed necrotic swelling and membrane rupture. NETosis involves chromatin decondensation and extracellular release of DNA and histones. Ferroptosis is defined by iron-dependent lipid peroxidation and mitochondrial shrinkage. These regulated pathways differ in triggers and mechanisms but share the potential to generate DAMPs that propagate inflammatory responses. Explanation in the text. Created in BioRender. Maisat, W. (2025) https://BioRender.com/sij8drd.
Figure 3. Overview of major cell death pathways and their characteristic morphological features. Necrosis involves early swelling of cells and organelles followed by rupture of the plasma membrane. Apoptosis progresses through cell shrinkage and membrane blebbing, formation of apoptotic bodies, and phagocytosis by neighboring cells. When clearance is delayed or insufficient, apoptotic cells may undergo secondary necrosis. Autophagy is characterized by formation of an autophagosome that fuses with a lysosome to generate an autolysosome. Excessive or inadequate autophagy may lead to autophagy-dependent cell death. The lower panel illustrates forms of regulated inflammatory cell death. Pyroptosis features pore formation, cell swelling, and membrane rupture. Necroptosis is marked by programmed necrotic swelling and membrane rupture. NETosis involves chromatin decondensation and extracellular release of DNA and histones. Ferroptosis is defined by iron-dependent lipid peroxidation and mitochondrial shrinkage. These regulated pathways differ in triggers and mechanisms but share the potential to generate DAMPs that propagate inflammatory responses. Explanation in the text. Created in BioRender. Maisat, W. (2025) https://BioRender.com/sij8drd.
Anesthres 03 00001 g003
Table 1. Major perioperative damage-associated molecular patterns (DAMPs).
Table 1. Major perioperative damage-associated molecular patterns (DAMPs).
Location
/Source
AcronymFull NameFunction (Normal)PRRs/Recognition PathwaysReferences
NuclearHMGB1High-Mobility Group Box 1Chromatin binding; transcriptional regulationTLR2, TLR4, RAGE[12,13]
HistonesHistone proteinsStructural organization and stability of chromatinTLR2, TLR4, TLR9; direct endothelial binding[14]
Extracellular DNANuclear DNA (cfDNA)Genetic materialTLR9, cGAS–STING[15,16]
MitochondrialmtDNAMitochondrial DNAEncodes mitochondrial proteinsTLR9, cGAS–STING, NLRP3[17]
ATPAdenosine triphosphateEnergy currency of the cellP2X7; NLRP3 inflammasome[18]
N-formyl peptidesMitochondrial peptidesChemotactic signalsFPR-1[19]
CytosolicS100A8/A9S100 calcium-binding proteins A8/A9Stress response; intracellular signalingRAGE, TLR4[20]
S100A12S100 calcium-binding protein A12Proinflammatory mediatorRAGE, TLR4[10]
HSPsHeat-shock proteinsMolecular chaperones; protein foldingTLR2, TLR4[10]
UAUric acidEnd product of purine metabolismNLRP3 inflammasome[21]
Extracellular matrix (ECM)BiglycanSmall leucine-rich proteoglycanStructural support; released upon ECM damageTLR2, TLR4[22]
LMW-HALow molecular weight hyaluronanECM degradation fragmentTLR2, TLR4[22]
Heparan sulfateGlycosaminoglycan fragmentsECM component; endothelial injury signalTLR4[23]
Blood/Transfusion-relatedFree hemeCell-free hemeOxygen transport; pro-oxidant when releasedTLR4[11]
Lipid peroxidation productsn/an/aTLR4[11]
MicroparticlesPlatelet- or FFP- or RBC-derived vesiclesn/aNLRP3[11]
Abbreviations: cfDNA, cell-free DNA; ECM, extracellular matrix; FPR-1, formyl peptide receptor-1; LMW-HA, low molecular weight-hyaluronan; NLRP3, NOD-like receptor family pyrin domain-containing 3; PRR, pattern recognition receptor; RAGE, receptor for advanced glycation end products; TLR, Toll-like receptor.
Table 2. DAMPs implicated in perioperative organ dysfunction, their triggers, signaling pathways, and supporting evidence from clinical and preclinical studies.
Table 2. DAMPs implicated in perioperative organ dysfunction, their triggers, signaling pathways, and supporting evidence from clinical and preclinical studies.
Clinical ManifestationDAMPs InvolvedTriggersMechanisms/PathwaysClinical/Experimental RelevanceEvidence Type
ALI/ARDSmtDNA
  • Sepsis
  • n/a
  • Elevated plasma mtDNA in septic patients with ALI was associated with higher mortality.
Clinical observation [61]
  • Lung I/R injury
  • GSDMD-mediated pores promote mtDNA leakage into neutrophils, activating cGAS–STING and inducing NET formation
  • NET release driven by mtDNA contributed to lung injury during I/R.
Preclinical (mice) [62]
MTD
  • MTD injection
  • Activates NLRP3, FPR-1, and TLR9
  • Increases Ca2+ flux and MAPK activation in neutrophils
  • MTD promoted alveolar macrophage activation and acute lung injury.
Preclinical (mice) [63,64]
HGMB1
  • Postoperative mechanical ventilation
  • n/a
  • Elevated HMGB1 detected in BALF of ventilated patients without preexisting lung injury.
Clinical RCT [65]
  • CPB
  • NLRP3/ASC-mediated alveolar macrophage pyroptosis
  • TLR4 activation
  • Elevated HGMB1 secretion in serum and BALF
  • Anti-HMGB1 treatment reduced ALI after CPB. and lowered IL-1β, TNF-α, HGMB1, and TLR4 expression in lung tissue.
Preclinical (rat) [66,67]
  • Intestinal I/R
  • MyD88-mediated NET formation
  • HMGB1-induced NETosis exacerbated intestinal I/R-associated ALI.
  • NET degradation and inhibition mitigated injury.
Preclinical (mice) [68]
Histones
  • LPS-induced SIRS/ARDS
  • Induce pulmonary endothelial activation via TLRs, promoting neutrophil adhesion and activation
  • Histones act as potent proinflammatory mediators in ARDS.
Preclinical (mice) [69]
  • Lung I/R (lung transplantation)
  • Stimulate CTSC secretion from alveolar macrophages, triggering NETosis via NADPH oxidase-derived ROS
  • Increased NETosis observed in lung transplant patients with primary graft dysfunction.
  • CTSC-inhibition attenuated NETosis and I/R injury in mice.
Clinical case–control; preclinical (mice) [70]
S100A12
  • Pneumonia with ARDS
  • RAGE activation
  • S100A12 and RAGE are highly expressed in lung tissue during ALI.
  • BALF levels of S100A12 and sRAGE are elevated in ARDS patients.
  • S100A12 induces a proinflammatory response in lung endothelial cells in vitro.
Clinical case–control; experimental [71]
S100A9
  • LPS-induced SIRS/ALI
  • NLRP3 activation
  • S100A9 is upregulated in lung tissue; blockade attenuated inflammation and apoptosis.
Preclinical (mice) [72]
Extracellular ATP
  • Mechanical ventilation
  • P2Y1 receptor activation
  • Extracellular ATP in BALF and P2Y1 receptor expression in lung tissues are increased after mechanical ventilation (high tidal volume > low tidal volume > sham ventilation).
Preclinical (mice) [46]
Myocardial dysfunctionHistones
  • Sepsis
  • n/a
  • Circulating histone levels correlated with cTn levels in septic patients and mediate cardiomyocyte injury ex vivo.
  • High histone levels were associated with new-onset LV dysfunction and arrhythmias.
  • Anti-histone intervention attenuated cardiac injury and dysfunction in septic models.
Clinical case–control; preclinical (mice) [73]
  • Global I/R injury
  • Direct cytotoxicity to cardiomyocytes through a mechanism independent of TLR4 and NF-kB signaling
  • Circulating histones increased following I/R injury; histone H4 levels positively correlated with infarct size.
Preclinical (rat) [74]
HMGB1
  • Myocardial I/R
  • Endothelial HMGB1-AIM2 axis regulating endothelial pyroptosis
  • Elevated HMGB1 detected in cardiac tissue and circulation after I/R.
  • Endothelial HMGB1 knockout preserved function by reducing infarct size, maintaining barrier integrity, and attenuating inflammation and oxidative stress.
Preclinical (murine) [75]
  • Myocardial I/R
  • JNK/ERK/NF-κB and RAGE signaling activation
  • HMGB1 levels increased within 30 min after hypoxia in vitro, and during ischemic myocardial injury in vivo.
  • Inhibition of HMGB1 reduced infarct size and tissue damage.
Preclinical (mice) [76]
  • Myocardial I/R
  • HMGB1/TLR4 signaling
  • TLR4 mutation reduced cardiomyocyte apoptosis and infarct size after myocardial I/R.
Preclinical (mice) [77]
S100A8/A9
  • Myocardial I/R, PCI
  • Suppresses mitochondrial energy production by inhibiting complex I via TLR4/ERK signaling, leading to downregulation of PGC-1α/NRF1 pathways
  • Serum S100A8/A9 levels were significantly increased one day after PCI.
  • Higher levels were associated with the incidence of MACEs.
Clinical observation; preclinical (mice) [78]
POAFmtDNA
  • Cardiac surgery with CPB
  • n/a
  • Patients with POAF had higher pericardial mtDNA levels compared with those without.
Clinical observation [17,79]
HMGB1
  • Cardiac surgery with CPB
  • n/a
  • Elevated HMGB1 was associated with increased susceptibility to POAF after CABG with CPB.
Clinical observation [80]
AKIHGMB1
  • Renal I/R
  • HMGB1/TLR4 signaling
  • HMGB1 promoted renal damage after I/R injury.
  • Administration of neutralizing Ab to HMGB1 before or soon after I/R protected against AKI
Preclinical (mice) [81]
  • Renal I/R
  • n/a
  • HMGB1 expression progressively increased in ischemic kidneys.
  • HMGB1 neutralization preserved renal function and reduced tubular necrosis
Experimental; preclinical (mice) [82]
mtDNA
  • Surgical critical illness
  • n/a
  • Urinary mtDNA correlated with markers of renal injury/dysfunction.
  • Elevated levels identified new-onset AKI and predicted RRT need and hospital mortality.
Clinical observation [83]
  • Cardiac surgery with CPB
  • n/a
  • Urinary mtDNA was elevated in progressive AKI and was associated with worsening severity
Clinical observation [84]
Histone
  • Renal I/R
  • NET formation
  • Histone secreted from ischemic tubular cells primed neutrophils to form NETs in vitro.
  • NETs were observed in kidney biopsies from patients with post-transplant ATN after prolonged cold ischemic time.
  • Circulating histones promoted lung injury and remote organ dysfunction after renal I/R.
Clinical observation; preclinical (mice); experimental [85]
LiverHMGB1
  • Hepatic I/R
  • HMGB1/TRL4 signaling
  • HMGB1 expression was upregulated in hepatocytes after hypoxia in vitro and in the liver after I/R in vivo.
  • Neutralizing HMGB1 reduced inflammatory mediators and protected against hepatic I/R injury.
Experimental; preclinical [86]
CoagulopathyHistone
  • Sepsis
  • n/a
  • Serum histone H3 levels were elevated in septic patients with coagulopathy and predicted a higher risk of mortality.
Clinical observation [60]
Heparan sulfate
  • Trauma
  • n/a
  • Heparan sulfate was associated with impaired thrombin generation.
Clinical observation [59]
DICHistone
  • Histone injection
  • Histones bind platelets, induce calcium influx, and recruit adhesion molecules to promote platelet aggregation.
  • Histone–platelet interaction contributed to thrombosis in preclinical models.
Preclinical (mice) [87]
Histone
  • Sepsis
  • Histone injection
  • Extracellular histones trigger platelet aggregation and microvascular occlusion.
  • Histone H3 was markedly elevated in septic patients with DIC but absent in healthy controls.
  • Extracellular histones caused massive thromboembolism consumptive coagulopathy in mice.
Clinical case–control; preclinical [34,88]
HMBG1
  • Sepsis
  • n/a
  • Patients with DIC had significantly elevated plasma HMGB1.
  • HMGB1 levels correlated with DIC score and SOFA score.
Clinical observation [89]
PND (POD/POCD)HMGB1
  • Major GI surgery
  • n/a
  • Serum HMGB1 levels were elevated in elderly patients and associated with postoperative cognitive decline
Clinical [90]
  • Splenectomy
  • HMGB1/RAGE signaling disrupts the blood–brain barrier and promotes neuroinflammation.
  • Elevated HMGB1 and RAGE expression were observed with evidence of blood–brain barrier disruption and cognitive impairment
Preclinical (rat, mice) [91,92]
mtDNA
  • Prolonged anesthesia with sevoflurane
  • Microglial NLRP3 inflammasome activation
  • cGAS-STING signaling
  • Prolonged sevoflurane anesthesia triggered cognitive dysfunction and neurological impairment in mice
Preclinical (mice) [93]
S100A8/A9
  • Tibial fracture surgery
  • TLR4/MyD88 signaling promotes neuroinflammation and microgliosis
  • S100A8/A9 expression was elevated in PBMCs and hippocampus after surgery.
  • Antibody blockade reduced microgliosis and improved cognitive function 24 h after surgery.
Preclinical (mice) [94]
Abbreviations: ALI, acute lung injury; ARDS, acute respiratory distress syndrome; AIM2, absent in melanoma 2; AKI, acute kidney injury; ATN, acute tubular necrosis; BALF, bronchoalveolar lavage fluid; CABG, coronary artery bypass grafting; cGAS, cyclic GMP–AMP synthase; CPB, cardiopulmonary bypass; CTSC, cathepsin C; DAMP, damage-associated molecular pattern; DIC, disseminated intravascular coagulation; ERK, extracellular signal-regulated kinase; GSDMD, gasdermin D; HMGB1, high-mobility group box 1; IL, interleukin; I/R, ischemia–reperfusion; JNK, c-Jun N-terminal kinase; LV, left ventricle; MACE, major adverse cardiac event; MAPK, mitogen-activated protein kinase; mtDNA, mitochondrial DNA; NET, neutrophil extracellular trap; NF-κB, nuclear factor kappa B; NLRP3, NOD-like receptor family pyrin domain-containing 3; PBMC, peripheral blood mononuclear cell; PCI, percutaneous coronary intervention; PND, perioperative neurocognitive disorder; POD, postoperative delirium; POCD, postoperative cognitive dysfunction; POAF, postoperative atrial fibrillation; RAGE, receptor for advanced glycation end products; RRT, renal replacement therapy; ROS, reactive oxygen species; SOFA, sequential organ failure assessment; TLR, Toll-like receptor.
Table 3. Preclinical evidence for anesthetic and adjunctive modulation of DAMP pathways.
Table 3. Preclinical evidence for anesthetic and adjunctive modulation of DAMP pathways.
AgentDAMPs AffectedPathways/MechanismsReported Effects (Preclinical Study)
Volatile agents
Sevoflurane
  • cGAS-STING-NLRP3 inflammasome activation [93,95]
  • HMGB1-induced NLRP3/ASC activation [96,97]
  • TLR4/MyD88/NF-κB and TRAF signaling [97,98,99]
  • Protective effects: Postcondition reduced hepatic, cerebral, and myocardial I/R injury, suppressed neuroinflammation [98,99,100].
  • Adverse effects: Prolonged or prenatal exposure associated with neuroinflammation, impaired cognition, and memory deficits [93,95,96,97].
Isoflurane
  • ↑ HMGB1 [101]
  • ↓ HMGB1 [102]
  • ↑ HSP60 [103]
  • RAGE signaling [101]
  • TLR2 and TLR4 signaling [102,103]
  • Protective effects: Reduced microglial activation and neuroinflammation [102,103]
  • Adverse effects: May induce pyroptosis in lung cancer model [101].
Intravenous agents
Propofol
  • Inhibits TLR4/PKR [104] and HMGB1-NLRP3 inflammasome signaling [106]
  • Reduces mitochondrial oxidative stress [105]
  • Modulates miRNA-451/HMGB1 signaling [107]
  • Protective effects: Reduced pulmonary and myocardial injury in I/R models [104,106,107]; attenuated VILI [105].
Ketamine
  • HMGB1–RAGE signaling, with effects on autophagy and microglial polarization [108]
  • TLR4/MyD88/TRAF and NF-κB inhibition [109,110,111,112]
  • cGAS-STING pathway [113]
  • Protective effects: Reduced LPS- and sepsis-induced lung injury [109,110,111]; inhibited HMGB1-induced endothelial inflammation [112]; exerted antidepressant effect [108].
  • Adverse effects: In specific setting (e.g., ketamine-induced cystitis in rodents), mitochondrial damage and mtDNA release may worsen inflammation [113].
Adjuncts
Dexmedetomidine
(DEX)
  • Inhibits TLR4/MyD88/NF-κB and RAGE signaling [114,115,116,117,118]
  • Suppresses NLRP3 inflammasome activation [119]
  • Inhibit pyroptosis and necroptosis [120]
  • Enhances protective HSPA12B expression through TLR4/NF-κB signaling [121]
  • Protective effects: Attenuated hippocampal neuroinflammation and PNDs [114]; reduced myocardial and renal injury from ischemia–reperfusion [115,116,119,120]; mitigated renal I/R injury and LPS-induced ALI [118,121]; alleviated neuropathic pain and inflammatory response induced by a chronic constriction injury [117].
  • Clinical data: DEX showed anti-inflammatory effects and reduced the incidence of PNDs, but HMGB1 levels were not significantly correlated with the incidence of PND in patients [122].
Remimazolam
  • ↓ HMGB1 [123]
  • Inhibits TLR4/NF-κB signaling [123]
  • Protective effects: Attenuated POCD after DHCA [123].
Opioids
Remifentanil
  • ↓ HMGB1 [124]
  • Reduces NF-κB activation [124]
  • Protective effects: Anti-inflammatory effects in septic models [124].
Abbreviations: ASC, apoptosis-associated speck-like protein containing a CARD; cGAS, cyclic GMP–AMP synthase; DHCA, deep hypothermic circulatory arrest; DEX, dexmedetomidine; HMGB1, high-mobility group box 1; HSPA12B, heat shock protein family A member 12B; I/R, ischemia–reperfusion; LPS, lipopolysaccharide; miRNA, microRNA; mtDNA, mitochondrial DNA; NF-κB, nuclear factor kappa B; NLRP3, NOD-like receptor family pyrin domain containing 3; PND, perioperative neurocognitive disorder; POCD, postoperative cognitive dysfunction; RAGE, receptor for advanced glycation end products; TLR, Toll-like receptor; TRAF, TNF receptor-associated factor; VILI, ventilator-induced lung injury. Upper arrow increase, lower arrow decreases.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Maisat, W.; Yuki, K. Damage-Associated Molecular Patterns in Perioperative Anesthesia Care: A Clinical Perspective. Anesth. Res. 2026, 3, 1. https://doi.org/10.3390/anesthres3010001

AMA Style

Maisat W, Yuki K. Damage-Associated Molecular Patterns in Perioperative Anesthesia Care: A Clinical Perspective. Anesthesia Research. 2026; 3(1):1. https://doi.org/10.3390/anesthres3010001

Chicago/Turabian Style

Maisat, Wiriya, and Koichi Yuki. 2026. "Damage-Associated Molecular Patterns in Perioperative Anesthesia Care: A Clinical Perspective" Anesthesia Research 3, no. 1: 1. https://doi.org/10.3390/anesthres3010001

APA Style

Maisat, W., & Yuki, K. (2026). Damage-Associated Molecular Patterns in Perioperative Anesthesia Care: A Clinical Perspective. Anesthesia Research, 3(1), 1. https://doi.org/10.3390/anesthres3010001

Article Metrics

Back to TopTop