Skip to main content
Erschienen in: European Journal of Trauma and Emergency Surgery 4/2020

Open Access 14.10.2019 | Review Article

Damage-associated molecular patterns in trauma

verfasst von: Borna Relja, Walter Gottlieb Land

Erschienen in: European Journal of Trauma and Emergency Surgery | Ausgabe 4/2020

Abstract

In 1994, the “danger model” argued that adaptive immune responses are driven rather by molecules released upon tissue damage than by the recognition of “strange” molecules. Thus, an alternative to the “self versus non-self recognition model” has been provided. The model, which suggests that the immune system discriminates dangerous from safe molecules, has established the basis for the future designation of damage-associated molecular patterns (DAMPs), a term that was coined by Walter G. Land, Seong, and Matzinger. The pathological importance of DAMPs is barely somewhere else evident as in the posttraumatic or post-surgical inflammation and regeneration. Since DAMPs have been identified to trigger specific immune responses and inflammation, which is not necessarily detrimental but also regenerative, it still remains difficult to describe their “friend or foe” role in the posttraumatic immunogenicity and healing process. DAMPs can be used as biomarkers to indicate and/or to monitor a disease or injury severity, but they also may serve as clinically applicable parameters for optimized indication of the timing for, i.e., secondary surgeries. While experimental studies allow the detection of these biomarkers on different levels including cellular, tissue, and circulatory milieu, this is not always easily transferable to the human situation. Thus, in this review, we focus on the recent literature dealing with the pathophysiological importance of DAMPs after traumatic injury. Since dysregulated inflammation in traumatized patients always implies disturbed resolution of inflammation, so-called model of suppressing/inhibiting inducible DAMPs (SAMPs) will be very briefly introduced. Thus, an update on this topic in the field of trauma will be provided.
Abkürzungen
AM
Alveolar macrophage
APC
Activated protein C
ARDS
Acute respiratory distress syndrome
ATP
Adenosine triphosphate
BAL
Bronchoalveolar lavage fluid
CARS
Compensatory anti-inflammatory response syndrome
CD
Cluster of differentiation
cfDNA
Cell-free DNA
COPD
Chronic obstructive pulmonary disease
CLP
Cecal ligation and puncture
CLR
C-type lectin receptor
CNS
Central nervous system
CSF
Cerebrospinal fluid
CXCL
Chemokine (C-X-C motif) ligand
CypA
Cyclophilin A
CytC
Cytochome C
DAMP
Damage-associated molecular pattern
DIC
Disseminated intravascular coagulation
DNA
Deoxyribonucleic acid
EV
Extracellular vesicles
HMGB
High-mobility group box
HSP
Heat shock protein
ICU
Intensive care unit
IL
Interleukin
IL-1R
Interleukin-1 receptor
IS
Immunosuppression
LPS
Lipopolysaccharide
MAPK
Mitogen-activated protein kinase
MBL
Mannose binding lectine
MHC
Major histocompatibility complex
Mincle
Macrophage-inducible C-type lectin
MIP
Macrophage-inflammatory protein
miRNA
MicroRNA
MODS
Multiple organ dysfunction syndrome
MOF
Multiple organ failure
MPO
Myeloperoxidase
mRNA
Messenger RNA
mtDNA
Mitochondrial DNA
MyD88
Myeloid differentiation primary response 88
NET
Neutrophil extracellular traps
NF-κB
Nuclear factor “kappa-light-chain-enhancer” of activated B cells
NLR
NOD-like receptor
NLRP
Pyrin domain-containing nucleotide oligomerization receptor (NLR)
NOD
Nucleotide oligomerization domain
nRNA
Nuclear RNA
PAMP
Pathogen-associated molecular pattern
PMNL
Polymorphonuclear leukocytes
PRR
Pattern recognition receptor
RAGE
Receptor for advanced glycation end products of proteins
ROS
Reactive oxygen species
RNA
Ribonucleic acid
SAMP
Suppressing/inhibiting inducible DAMP
SAP130
Spliceosome-associated protein 130
SCI
Spinal cord injury
SIRS
Systematic inflammatory response syndrome
ST2
Orphan receptor serum stimulation 2
TBI
Traumatic brain injury
Th
T-helper cell
TLR
Toll-like receptor
TNF
Tumor necrosis factor
WT
Wild type

Introduction

Traumatic injury is one of the major contributors to worldwide mortality as well as one of the world’s most relevant but neglected health concerns [1, 2]. Multiply injured or polytraumatized patients most frequently die either immediately or early within only of a few hours after the traumatic insult due to their injury severity, severe traumatic brain injuries (TBI), and/or exsanguination, or in the later post-injury phase due to inflammation-related complications, which affect the immune system homeostasis, resulting in, e.g., sepsis, septic shock, or multiple organ failure (MOF) [26]. Adjacent to physical factors, which are intensely treated in the intensive care units (ICU), in case of survival, the multiply injured patient may suffer from serious impairments in cognitive, psychological, or psychosocial issues [79].
Over the last decades, several experimental (poly)trauma models which combine different injury patterns have been developed and applied to study and understand the basic pathophysiology of the posttraumatic sequelae in polytraumatized patients. While the primary impact can only be addressed by injury prevention, the later detrimental sequalae may be prevented by an abrogation of the posttraumatic course [10, 11]. The typical pathophysiological course of traumatized patients under ICU treatment is characterized by a systemic inflammatory response to severe injury that begins immediately after trauma, and which involves complex interactions across the hemostatic, inflammatory, endocrine, and neurological systems [3, 12, 13]. Modern notions hold that this clinically observed so-called systemic inflammatory response syndrome (SIRS) reflects an uncontrolled overshooting systemic innate immune response caused by polytrauma-induced emission of large amounts of so-called damage-associated molecular patterns (DAMPs) which circulate systemically to affect the whole body of the patient [3, 1215]. The term DAMP was coined in 2003 by Land [15]. And it is apparently the DAMPs which lead to the catastrophe of the sterile hyperinflammatory, sepsis-like, life-threatening SIRS, which is most often associated with organ failure, MOF, or multiple organ dysfunction syndrome (MODS) [12, 13, 16]. Typically, this DAMP-induced systemic cytokine/chemokine-mediated hyperinflammatory response to severe trauma is associated with an intense and long-lasting counterbalancing compensatory anti-inflammatory response syndrome (CARS) resulting in posttraumatic immunosuppression (IS) [3, 5, 1719]. CARS can be regarded as a disorder caused by a hyper-resolution of hyperinflammation in terms of a mirror-imaged counter-regulation of SIRS. Thus, for several decades research on posttraumatic complications was based on a biphasic posttraumatic inflammation model. The central pathophysiological principle there was to find the balance between a qualitative defense against invasive putative pathogens and additionally reducing collateral damage by immune cells [2022]. Of note, however, instead of a previously assumed biphasic inflammatory response consisting of an initial proinflammatory and a subsequent counterbalancing anti-inflammatory response, hyper-resolving processes are now considered to occur simultaneously with hyperinflammation. There is presumably only a slight delay between initial hyperinflammation and the following hyper-resolution, which is possibly caused by the production of suppressing/inhibiting inducible DAMPs (SAMPs) by already DAMP-activated/initialized responses of innate immune cells [23]. Recently, this novel term accomplishing the DAMPs/SAMPs post-injury response was coined by Land [23]. The posttraumatic course of this inflammatory response is briefly represented in Fig. 1.

SIRS

The recognition of DAMPs by immune cells initiates an acute hyperinflammatory condition of SIRS that induces physiological changes such as hypothermia or hyperthermia, elevated heart rate, and leukocytosis/leukocytopenia [24, 25]. The uncontrolled or protracted clinical signs of SIRS constitute a risk factor for organ failure, delayed hospital-acquired infections as well as occasionally MOF after trauma [2628]. The concept of SIRS was introduced in early 1990s and charged with the task of developing an easy-to-apply set of clinical parameters, as well as biomarkers, to improve the early identification of potential candidates for clinical trials and new treatment strategies for sepsis, as well as for the identification of patients at risk of infectious complications, respectively [25]. Thus, since three decades, the knowledge on the complex pathophysiological and posttraumatic inflammatory response contributing to the development of post-injury complications is growing [2932]. Numerous cellular and humoral proinflammatory mediators including complement factors are markedly elevated in patients suffering from SIRS and associated complications on the other hand. In general, SIRS is classically dominated by overwhelming hyperinflammatory processes which are characterized by elevated circulating levels of multiple powerful cellular and humoral proinflammatory mediators. Of those, notably in the experimental and clinical sepsis-associated SIRS, cytokines (e.g., tumor necrosis factor (TNF), interleukin (IL)-1α, IL-1β, and IL-6), chemokines, growth factors, adhesion molecules, gaseous substances, vasoactive peptides, and cell stress markers have been identified and analyzed [33]. After the concept of SIRS was created, a number of publications emerged heralding the efficacy of SIRS in predicting outcomes after trauma [34, 35]. Accumulating evidence indicates that SIRS associated with MOF is the result of traumatic injury-induced either accidental or regulated cell death such as necroptosis, serving as a robust source for emission of DAMPs, while the impact of apoptosis may be presumably less significant here [3639]. Today, it is widely accepted that DAMPs are the key predominant molecules which trigger the biological host response to trauma including SIRS, by activating and recruiting effector cells including antigen-presenting cells of the immune system [12, 4046]. Thus, basically, every DAMP proven to promote efferent proinflammatory pathways can be considered to be involved in the development of SIRS. In 2012, in accordance with the newly recognized “sterile SIRS” as a severe complication form in polytraumatized patients, first clues to a critical role of DAMPs in the promotion of those hyperinflammatory responses have been reported [47]. Among those molecules, endogenous DAMPs including high-mobility group box (HMGB) protein 1, heat shock proteins (HSPs), certain members of the S100A family, histones, free nucleic acids, members of the IL-1 cytokine as well as complement family as bona fide intracellular effectors, which upon abundant rapid release alert the environment about cell stress and danger, have already been reported to activate the posttraumatic innate immune responses and drive organ dysfunction(s) after trauma [12, 13, 46, 48].
Although a large number of endogenous nuclear or cytosolic DAMPs have been described in the context of the local/systemic posttraumatic and/or noninfectious “sterile” inflammatory response, which represents the key driver of the late occurring post-injury complications and fatal outcome rates, the knowledge on their variety and underlying mechanisms still remains unknown [4951]. In particular, it is unknown whether there is a distinct pattern of DAMPs emission that may be helpful in differentiating between, i.e., sterile and infectious (bacterial or viral) sepsis form. The knowledge on posttraumatic DAMPs pattern may allow to further define the exact roles of DAMPs in hyperinflammation and develop therapeutic inhibitors targeting specific DAMPs to improve outcomes. Thus, some of most critically involved DAMPs in both sterile and infectious posttraumatic disorders will be briefly described in this review.

CARS and SAMPs

The recognition of an overshooting SIRS which may be associated with septic complications has provided several attractive targets for new anti-inflammatory drugs which were designed to prevent further propagation of inflammation in past. Yet, nowadays, treatment of sepsis in critically ill patients is mainly based on the prompt intravenous administration of adequate antimicrobial agents and support of organ functions. For the last four decades, in theory, many different targets for treatment were designed to inhibit, bind, or neutralize the proinflammatory mediators that were assumed to be responsible for the network of events that culminated in detrimental clinical outcomes. Disappointingly, however, targeted clinical trials have shown that most of these agents failed to cure the septic disease [5256]. Even the promising US FDA-approved drug, recombinant human-activated protein C or drotrecogin-α failed in clinical trials and showed similar effects as placebo. Intriguingly, these observations paved the way to another typical symptomatic feature of the disorder: the CARS [24]. The high susceptibility to secondary infections after the initial phase of sepsis or after trauma has been attributable to CARS causing the posttraumatic IS [17, 19, 57]. Interestingly, for a long time, intensive care physicians have observed that patients with SIRS were prone to severe, sometimes lethal infectious diseases. However, in search of the causes during the 1970s and 1980s, researchers found that many of those patients had acquired an impaired immune state characterized by various markers of IS. The concept was actually underlined from the beginning as the treatment strategies with anti-inflammatory agents failed in clinical trials on septic patients.
The phenomenon of impaired immunity that was frequently observed in posttraumatic as well as in septic conditions essentially reverses many of the typical events encountered in SIRS. However, research over the last several decades uncovered the anti-inflammatory and immunosuppressive mechanisms that govern the clinical post-injury disorders. Initially, IS corresponds to a homeostatic phenomenon to avoid the remote organ injuries caused by the early SIRS; however, it becomes deleterious when it persists, rendering patients prone to secondary infections [17, 58]. Nowadays, CARS is not regarded as part of a biphasic inflammatory response consisting of an initial SIRS with a subsequent CARS. It became evident that the hyper-resolving processes of inflammation (CARS, IS) occur nearly in parallel with the hyperinflammatory response, but also that they can exist separately from the SIRS [28, 59]. Moreover, CARS is not only a simplified anti-inflammatory process, but rather a disorder caused by a hyper-resolution of inflammation.
Remarkably, next to alterations of the innate immune system, posttraumatic IS has been closely linked to modulations of the adaptive immune system. Here, among others, i.e., a shift from a T-helper cell type (Th)1- to a Th2-mediated immune response, T cell anergy in traumatized patients, suppression of T cell effector functions, modulated frequency of T regulatory cells have been observed [19, 21, 22, 6064]. This posttraumatic response suggests that in reverse with the proinflammatory action of DAMPs in SIRS, SAMP triggers the proresolving pathways in CARS and IS. Besides activating DAMPs, potentially suppressing DAMPs, that is SAMPs, are mainly produced by activated leukocytes and macrophages upon stress and injury, which—in parallel to proinflammatory responses, convey anti-inflammatory processes and shape immunosuppressive responses. In 2015, it was shown that the gene expression of proresolving lipid mediator pathways was closely associated with clinical outcomes in traumatized patients [65]. Patients with uncomplicated recoveries had differential and higher resolving scores [65]. The SAMPs prostaglandin E2 or AnxA1 plays significant roles in proresolving CARS [6668]. Accordingly, DAMPs and SAMPs constitute valuable tools in diagnosis and therapy of severely injured and/or infected patients that are expected to find their way into clinical routine at the ICUs. Hyper-resolution with the following IS may be the most critical factor for the immune status of patients with either sterile or infectious SIRS. Thus, here as well, measurement of DAMPs with their pattern, but also continuous determination of SAMPs with their pattern, respectively, may be used as hospital admission criteria for decisions supporting further diagnostic, prognostic, and therapeutic modalities. Although a closer look into both DAMPs and SAMPs is necessary to understand the posttraumatic response, in the underlying review only DAMPs will be addressed.

DAMPS

DAMPs are released from a damaged or diseased cell, and upon their release, they can stimulate a sterile immune or inflammatory response [13, 49, 69]. Across the tree of life, DAMP-induced immune responses serve as defense strategies aimed at maintaining and restoring homeostasis. However, despite their initially beneficial character, when dysregulated, uncontrolled, and exaggerated, the inflammatory and tissue repairing processes may become pathogenic and can lead to serious pathologies, i.e., sepsis, cardiovascular diseases, neurodegenerative diseases, diabetes, obesity, and asthma, as well as classic inflammatory diseases. Notably in the last two decades, the biological host response to trauma, which initially has been characterized by massive cytokine release as well as the activation and recruitment of effector cells including antigen-presenting cells, has further employed a large number of both microbial and host alarmins [12, 4044]. In general, on the biochemical level, the posttraumatic immune response is not only activated by foreign non-self material, but includes also endogenous factors, DAMPs, which are released from necrotic or physiologically “stressed” cells, with the aim of initiating and recruiting effector cells of the immune system [7072]. A large number of those endogenous nuclear or cytosolic triggers have been described to initiate and perpetuate the systemic posttraumatic and/or noninfectious inflammatory response; however, the knowledge on their precise role still remains unknown [13, 4951]. They signal “danger” to the host and trigger an inflammatory response, which in physiological healing leads over to tissue regeneration, but when dysregulated can cause pathologic inflammation and related disorders.
In the pathology of severe trauma, trauma-induced generation/emission of DAMPs can refer to cell-intrinsic modified DAMPs such as cell-free deoxyribonucleic acid cfDNA or DAMPs passively released from dying cells. Thus, any kind of traumatic cell stress/tissue injury provoked by mechanical trauma (e.g., blow, crush, penetrating wound, or surgical procedures usually related to fracture, hemorrhage, and/or infections), thermal trauma, or metabolic trauma such as promoted by ischemia/reperfusion injury, acidosis, hypoxia/hypoxemia, provokes a DAMP release [23]. Clinically, SIRS is characterized by local and systemic release of proinflammatory cytokines produced by DAMP-activated innate immune cells including IL-1β, TNF, and IL-6, arachidonic acid metabolites, proteins of the contact phase and coagulation systems, and complement factors as well as hormonal mediators [3, 34, 73, 74]. Among other factors, proinflammatory cytokines also activate coagulation pathways, whereby enhanced thrombin formation increases fibrinogen cleavage and reduces fibrin monomers from polymerizing to form stable fibrin clots (hypercoagulability), which is why fibrinogen depletion is observed [75]. Disseminated intravascular coagulation (DIC) is a heterogeneous group of disorders, which manifests as a spectrum of hemorrhage and thrombosis complicating many primary conditions including, i.e., sepsis and trauma [76]. Although its pathophysiology is complex, there is growing evidence that DAMPs emitted in excess play a crucial role in the pathogenesis of DIC [76]. Here, i.e., cfDNA, also known as extracellular DNA, which can be released by various host cells including neutrophils, macrophages, eosinophils, and tumor cells, as well as by certain strains of bacteria plays a significant role [7782]. Elevated levels of cfDNA have been found in various pathologic conditions including trauma [83, 84]. Interestingly, cfDNA that appears to exert both pro- and anti-fibrinolytic effects is a good predictor of trauma patient’s outcome in the ICU [84]. Activation of neutrophils with microbial or inflammatory stimuli results in the release of neutrophil extracellular traps (NETs) [77]. Upon cell death or specific cell activation of hematopoietic and parenchymal cells, extracellular cfDNA as well as DNA-binding proteins (e.g., histones and HMGB1) are released into circulation [13]. Those DNA-binding proteins are also strongly procoagulant and are involved in the pathogenesis of DIC [76, 85]. Another factor that can be directly influenced by DAMPs and contribute to DIC is the cellular migratory behavior with barrier loss. DAMP-induced endothelial expression of adhesion molecules facilitates leukocyte adhesion, promoting extravasation of leukocytes from the circulation into damaged tissue [86]. Mitochondrial (mt) DAMPs like mitochondrial DNA (mtDNA) and peptides appear in the blood after injury or shock, activate human leukocytes and might contribute to increased endothelial permeability during systemic inflammation [87]. The various DAMP motifs from mitochondria can act on endothelia and/or leukocytes via multiple pathways by enhancing leukocyte adherence to endothelial cells, activating cell–cell interactions, and subsequently increasing systemic endothelial permeability [87]. Mitochondrial DAMPs may be important therapeutic targets in conditions where inflammation pathologically increases endothelial permeability [87]. The mechanisms of a concerted action between endothelial cells and leukocytes, endothelial cell damage, leukocytes extravasation, microcirculatory disturbances, or DIC frequently lead to cell loss of parenchymal cells with the following organ failure. Increased systemic levels of DAMPs have been positively correlated with mortality and morbidity of patients or animals with trauma or surgical insults, while blocking or neutralizing DAMPs with specific small molecules or antibodies ameliorated sepsis and SOF in vivo [88]. As briefly introduced above, the clinical manifestation of severe multiple tissue injury including burn damage is characterized by a systemic cytokine/chemokine-induced hyperinflammatory response resulting in SIRS which is accomplished by an intense and long-lasting CARS associated with posttraumatic immunosuppression that predisposes patients to infectious sepsis and MOF [3, 17, 19, 57]. For example, in a murine burn injury model, IL-10 levels were significantly increased at 24 h, 7 days, and 21 days post-injury as compared with IL-10 levels in sham mice [89]. Thus, accumulating evidence suggests that clinically observed infectious complications are associated with high susceptibility of the patient to secondary bacterial, fungal, and viral infections and that this state is mediated by suppressing DAMPs by SAMPs which control homeostasis following injury. Yet, in the following, a closer look at the role of exemplary DAMPs in traumatic setting is taken. Since DAMPs are a culmination of highly heterogenous mediators, it is nearly impossible to choose the most important ones. Thus, in this review, we focus on the most prominent DAMPs involved in the posttraumatic pathophysiology, but other DAMPs of equal importance, which have been neglected here, may be addressed in further studies. This review sought to primarily describe the exemplary variety of DAMPs in their preclinical and clinical context.

High-mobility group box protein

HMGB1 as one of best studied DAMPs is a non-histone chromatin nuclear peptide that acts as a DNA chaperon, which is involved in binding of proteins and in DNA transcription, replication, but also repair [90, 91]. HMGB1 is composed of two positively charged DNA-binding motifs and a C-terminal acidic tail [92]. Nearby, all cells constitutively express HMGB1. It is located in the nucleus, but it will be released either passively after necrotic cell death, or secreted actively by living cells undergoing stress in response to angiogenic and inflammatory triggers [9396]. HMGB1 acts as an endogenous ligand of toll-like receptors (TLRs) 2, 4, 9 and receptor for advanced glycation end products (RAGE), thereby inducing nuclear factor “kappa-light-chain-enhancer” of activated B cells (NF-κB) activation [97, 98]. Although the mechanisms of HMGB1 secretion are elusive, it has been shown that its processing via inflammasomes is involved [99].
The innate immune stimulatory activity of HMGB1 has been regulated by the redox state of cysteine residues C23, C45, and C106 [100]. Interestingly, while a reduction in cysteine residues makes HMGB1 rather a chemoattractant than a cytokine inducer, oxidization of the cysteine residues via reactive oxygen species abrogates both activities of HMGB1 [101]. Thus, it remains elusive, if HMGB1 levels indeed reflect its innate immunity-inducing potential, since the HMGB1 levels do not necessarily reflect the complexity of inflammatory complications with underlying downstream signaling.
Under physiological conditions, HMGB1 is predominantly anchored inside the nucleus via nuclear localization sites and accordingly its binding with nuclear cargo carrier proteins [102, 103]. Systemic levels of HMGB1 increased within 30 min after severe trauma in patients [104] although its regulated activity HMGB1 levels correlated with injury severity, tissue hypoperfusion, early posttraumatic coagulopathy, hyperfibrinolysis, with complement activation and with SIRS [104]. Early increase in systemic HMGB1 levels indicated patients who developed acute lung injury (ALI) or acute renal failure in the later posttraumatic course [104]. Increased levels of HMGB1 were found to stratify non-survivors from trauma as compared to survivors [104]. Deng et al. [103, 105] have recently demonstrated that hepatocytes are the main source of circulating HMGB1 in LPS-induced endotoxemia and cecal ligation and puncture (CLP)-induced polymicrobial sepsis, as well as hemorrhagic shock. They have demonstrated that cell-specific depletion of HMGB1 in hepatocytes dramatically reduced circulating HMGB1 level and conferred protection from sepsis lethality in mice [105]. With regard to trauma, Levy et al. [106] have demonstrated transient elevations in systemic HMGB1 levels within 1 h post bilateral femur fracture. Interestingly, they have shown that after trauma, treatment with neutralizing antibodies to HMGB1 lowered systemic IL-6 and IL-10 levels as compared to controls treated with nonimmune control antibody [106]. Similarly to the above-mentioned study, with regard to hepatic changes, HMGB1 neutralization decreased serum alanine aminotransferase levels, and hepatic as well as gut mucosal NF-κB DNA binding [106]. The authors conclude that HMGB1, more precisely the TLR4-HMGB1 pathway, constitutes as an early mediator of systemic inflammation and end-organ injury after peripheral tissue injury after trauma. In line with these observations, it has been demonstrated in vivo that HMGB1 levels significantly increased in muscle 12 h after crush injury [107]. As already indicated, HMGB1 neutralization by an antibody reduced the cellular apoptosis in the renal cortex, thus indicating a positive feedback cycle [107]. In a systematic review on the role of HMGB1 danger signaling in TBI, HMGB1 was found to be released from damaged neurons, and furthermore, it was elevated in patient’s serum and cerebrospinal fluid (CSF) [108]. Moreover, HMGB1 increased in CSF associated with neuronal death in subarachnoid hemorrhage [109, 110]. The elaborated studies show that HMGB1 may serve as a prognostic biomarker and therapeutic target in patients with TBI [108]. Adjacent to traumatic injury, it was demonstrated before that HMGB1 plays an important role in the initiation and propagation of inflammation and organ injury under conditions of sterile inflammation which involve ischemic processes [111113]. Also in patients with ventilator-associated pneumonia during mechanical ventilation increased HMGB1 levels in bronchoalveolar lavage fluid (BAL) were detected [114]. In vivo models have demonstrated that intratracheal or intranasal administration of HMGB1 caused ALI, which has been reflected by enhanced acute inflammatory injury to the lungs, neutrophil accumulation, development of lung edema, and increased pulmonary production of IL-1β, TNFα, and macrophage-inflammatory protein (MIP)-2 [115, 116]. Also in different ischemic heart diseases a positive correlation between increased HMGB1 levels and worsen ventricular function upon ischemia/reperfusion injury has been demonstrated [117]. As briefly described in Introduction, DAMPs are playing a role in coagulation processes. HMGB1 itself is also a potent procoagulant that can directly stimulate and recruit platelets through TLR4 and RAGE [88, 118, 119]. Mice with HMGB1-deficient platelets exhibited increased bleeding times and reduced thrombus formation, platelet aggregation, inflammation, and organ damage during experimental trauma/hemorrhagic shock [118]. Extracellular HMGB1 coordinates numerous cellular functions, including migration, chemotaxis, activation, maturation, and proliferation but also the redox status of its target cells [120123]. However, the binding of HMGB1 to its receptors can be potentiated by binding to a whole variety of other factors, including PAMP and cytokines [123]. Thus, in summary, HMGB1 is an important pathogenic factor of inflammatory and thrombotic complications.

Interleukin-1

Some of the numerous DAMPs are cytokines, small messenger molecules, which are produced, activated, and released upon trauma [124]. In 1974, Dinarello [125] discovered the first cytokines IL-1α and IL-1β as members of the IL-1 family. IL-1α and IL-1β are encoded by different genes; however, they can be bound by the same IL-1 receptor (IL-1R) [125]. IL-1α has an higher affinity for IL-1R1 and IL-1β for the soluble IL-1R2 [126].
In contrast to IL-1β, IL-1α is constitutively expressed mainly in resting nonhematopoietic cells, which line the gastrointestinal tract, liver, kidney, and skin, but it can also be expressed in most cells and, furthermore, can be biologically active in its full-length form without its previous processing through inflammasomes, as it is mandatory for IL-1β activity [127129]. IL-1α and IL-33 are expressed in their precursor forms in the nucleus of various hematopoietic cells, and these nuclear proteins play important roles in the regulation of gene expression [130]. Since IL-1α does not contain a secretory signal peptide, it is released into extracellular space through either non-canonical vesicular secretion pathway or passive necrotic release [130]. The release of IL-1α into the extracellular space in stimulated cells occurs after processing of the membrane-bound IL-1α by the membrane-bound calcium-dependent cysteine protease calpain [130133]. Upon extracellular IL-1α interaction with ubiquitously expressed IL-1R1 and IL-1 receptor accessory protein downstream signaling proteins, such as myeloid differentiation primary response gene 88 (MyD88) and IL-1 receptor-activated protein kinase 4, an inflammatory response is induced [134]. In general, members of the IL-1 family induce similar signaling cascades in their target cells via mitogen-activated protein kinase (MAPK) or NF-κB pathways [135]. IL-1α as a dual function protein is a proinflammatory activator of transcription as chromatin-associated protein, and above that, it is acting as a cytokine [136138]. The latter is exerting its function after being released from apoptotic or necrotic cells, thereby alerting the immune system to tissue damage [139].
Thus, IL-1α has been considered a potential pathogenic factor involved in the development and progression of several disease including diabetes, inflammatory bowel disease, myocardial inflammation, and cancer [140143]. Interleukin-1 can initiate many important immunological responses such as fever, prostaglandin synthesis, mobilization of neutrophils into tissues, activation of B and T cell lymphocytes, fibroblast proliferation as well as the production of antibodies, collagen, and cytokines [126, 127]. With regard to trauma, only few studies on IL-1α are available. Jackman et al. [144] observed a mixed early systemic inflammatory response in trauma patients with elevated levels of IL-6, IL-10, IL-1Ra, macrophage migration inhibitory factor, myeloperoxidase (MPO), monocyte chemotactic protein-1, matrix metalloproteinase-9, and soluble Fas ligand, but also with simultaneously decreased levels of fractalkine, epidermal growth factor, IL-7, IL-9, IL-17, TNFbeta (TNFβ), MIP-1α, macrophage-derived chemokine and notably IL-1α. While in vivo DNA microarray data confirmed this highly complex inflammatory response in lung tissue following blunt chest trauma, interestingly, IL-1α has shown elevated expression levels, which were concomitant with increased levels of other inflammatory and coagulatory proteins, including TNFα receptor, IL-1β, C3, NF-κB, and plasminogen activator [145]. In contrast to the above-mentioned study, increased levels of IL-1 have been associated with the pathogenesis of acute respiratory distress syndrome (ARDS), and subsequent idiopathic pulmonary fibrosis, sarcoidosis, as well as certain other inflammatory diseases [130, 146]. Notably alveolar macrophages (AM) from patients with ARDS were shown to release significantly more IL-1 and IL-1β as compared with controls, indicating that increased IL-1 release by AM may be related to the progression of ALI [146]. Also inflammation caused by necrosis or tissue damage upon ischemia or hypoxia caused by poor oxygen supply has been related to IL-1 [130]. Hypoxia upregulated IL-1α transcription in epithelial cells [147]. Ischemia induced IL-1α release in activated platelets of the brain vasculature, and thereby stimulated endothelial cells to secrete the chemokine CXCL1 and express cell adhesion proteins vascular cell adhesion molecule-1 and intercellular adhesion molecule-1, thus promoting the transendothelial migration of neutrophils which are major contributors to inflammation-mediated brain injury [148]. IL-1α-stimulated proinflammatory cytokine expression in necrotic myocytes from ischemic heart following myocardial infarction has been reported as well [149]. It is known that IL-1α is a significant driver of the proinflammatory mechanisms, which are often linked to chronic inflammatory diseases or cancer [130]. However, only little is known about the role of IL-1α during the post-injury inflammatory response. Thus, uncovering its role in trauma and/or trauma-related pathologies may be promising in regard to specific targeting of IL-1α with clinically approved drugs, i.e., anakinra under pathological conditions.

Interleukin-33

Interleukin-33, a nuclear alarmin, is the youngest member of the IL-1 family, and it is mainly expressed in the cells which are in contact with the environment including stromal cells, such as endothelial cells, fibroblasts, and epithelial cells [130, 150, 151]. Thus, IL-33 is highly expressed in the lungs of both mice and humans [152]. Similar to IL-1α, IL-33 can be released upon the loss of cell integrity [153, 154]. Upon binding to its orphan receptor serum stimulation (ST)2 of the TLR/IL1R superfamily of receptors (also known as IL-1RL1), IL-33 initiates the potential signaling pathway via NF-κB and MyD88 [155, 156]. Interestingly, regulatory T cells that express the IL-33 receptor, ST2, upregulate amphiregulin after IL-33 stimulation, and this mechanism has been implicated recently in epithelial tissue repair after viral infection of the lung [157]. Again similar to IL-1α, IL-33 does not require processing to maintain its activity [158]. However, IL-33 can be cleaved to a mature and more proinflammatory form [159]. Interestingly, unlike other DAMPs, i.e., HMGB1 and IL-1α, nuclear IL-33 represses gene expression, and its nuclear localization alters the subcellular localization of IL-33, thereby regulating its extracellular functions and affecting immune homeostasis [160, 161]. On the other hand, comparable to other alarmins, posttranslational modification of IL-33 via its oxidation can regulate its activity, and thereby restrict its inflammatory function to the local environment [130, 162]. IL-33 plays an important role in inflammatory and autoimmune diseases such as asthma, inflammatory bowel disease, or autoimmune hepatitis by influencing neutrophilic infiltration into tissues, modulating the type 2 immune response, including Th2 cells, mast cells, eosinophils, basophils, and group 2 innate lymphoid cells [158, 163166]. The relevance of IL-33 processing in a pathological environment is still not clear, but it is evident that an inflammatory microenvironment can exacerbate disease-associated functions of IL-33 via generating more active mature form [159].
Recently, Liu et al. [167] have shown that the sensitivity of IL-33-deficient mice to bleomycin-induced ALI and that this phenotype was reversible by complementation with rIL-33. The authors conclude that IL-33 is a crucial local factor necessary to control and limit the early inflammatory response after chemically induced ALI [167]. On the other hand, depending on the context of certain infections, IL-33 was either protective or even deleterious, because of its either beneficial role in the resolution of inflammatory processes, or detrimental function in aggravating inflammation [130, 154, 168]. Interestingly, IL-33 reduced mortality in septic mice due to increased neutrophil influx into the peritoneal cavity and thereby enhanced bacterial clearance as compared to controls [169]. The data are significant, since they show that IL-33 reduced systemic but not local proinflammatory responses, and thereby limited the sepsis-induced systemic inflammation [169]. Under certain conditions of chronic inflammation, IL-33 was deleterious during the development of, e.g., asthma, or in the context of chronic obstructive pulmonary disease (COPD) and arthritic joint inflammation [170172]. The role of IL-33 in traumatic setting is completely elusive. However, there are some studies dealing with IL-33 during injury to the central nervous system (CNS) or spinal cord. It has been shown that IL-33 was highly expressed in white matter in the CNS, where it colocalized with oligodendrocyte markers as well as with astrocyte markers in gray matter [173]. Immediately after CNS injury IL-33 was released from damaged oligodendrocytes, acting on local astrocytes and microglia to induce chemokines critical for monocyte recruitment [173]. Mice lacking IL-33 had impaired recovery after CNS injury, which is associated with reduced myeloid cell infiltrates and decreased induction of M2 genes at the injury site [173]. Wicher et al. (2017) used samples from human TBI microdialysate, tissue sections from human TBI, and mouse models of central nervous system injury and found that expression of IL-33 in the brain was elevated from nondetectable levels, reaching a maximum after 72 h in both human samples and mouse models [174]. The authors have shown that astrocytes and oligodendrocytes were the main producers of IL-33 [174]. Post-TBI brains of mice deficient in the IL-33 receptor, ST2, contained fewer microglia/macrophages in the injured region as compared to the wild-type controls and had an altered cytokine/chemokine profile in response to injury [174]. Taken together, these observations indicate that IL-33 plays a role in neuroinflammation with microglia/macrophages being cellular targets for this interleukin post-TBI. Other in vivo experiments have shown before that after traumatic spinal cord injury (SCI) administration of recombinant IL-33 turned beneficial by significantly decreasing tissue loss, demyelination and astrogliosis in the contused mouse spinal cord, finally resulting in dramatically improved functional recovery [175]. Taken together, the recent findings indicate that IL-33 plays a role in response to TBI and drives chemokines that recruit monocytes and polarize macrophages toward an M2 phenotype, thereby potentially protecting neurons from further damage and promoting recovery after CNS injury [176]. Maybe a role of IL-33 as a potential SAMP should be carefully considered.

S100A proteins

S100 proteins or calgranulins are a family of low molecular weight calcium-binding homodimeric proteins, consisting of at least 25 distinct members with a large variety of intracellular and/or extracellular functions [130]. They are localized intracellularly in the cytoplasm, and here, they interact with various effector molecules to regulate cell proliferation, differentiation, migration, energy metabolism, scavenging of reactive oxygen species (ROS) and nitric oxide, calcium homeostasis, inflammation, apoptosis, transcriptional regulation as well as DNA repair and others [16, 130]. S100 proteins are mainly expressed in cells of myeloid origin, predominantly in neutrophils and are induced in several cell types which mediate inflammatory responses and recruit inflammatory cells to sites of tissue damage [177]. These small proteins are known to be either passively released from damaged cells or actively secreted from activated cells, and they have been detected in various body fluids, such as serum, urine, sputum, cerebrospinal fluid, and feces of patients with cancer, inflammatory and autoimmune disease, or cardiovascular complications, atherosclerosis or stroke [130, 178183]. Upon their release, they act as alarmins via their interaction with different receptors to orchestrate innate and adaptive immune responses [130, 184]. As example, S100A8, S100A9, and S100A12 family have been found extracellularly at high concentrations in inflamed tissue, where they exert their proinflammatory effects via binding to RAGE, TLR4 or upon interaction with other receptors [177, 185]. The signaling pathways trigger the activation of kinases including p38 MAPK, ERK1/2, and again transcription factor NF-κB [186188]. S100A1 released from damaged cardiomyocytes during myocardial infarction triggers TLR4-dependent proinflammatory responses, leading to induction of myocardial damage [189]. Several studies have reported an increase in S100A8/A9 levels in sepsis [190192]. However, certain anti-inflammatory properties in case of, e.g., S100A8 and S100A9 have been reported as well [193, 194].
With specific regard to traumatic injury, it has been demonstrated that S100B in CSF and plasma increases after injury and, even more, that the increasing levels were negatively correlated to outcome from human TBI [195197]. It was shown that S100B can counteract and reduce some negative cellular consequences of injury [184, 196, 198]. However, more recent studies imply the negative role of S100B in TBI and represent that S100B in CSF, as an astrocytic protein specific to the central nervous system and a useful marker in outcome prediction for TBI [199, 200]. Thus, S100 s protein have been established as useful marker for inflammatory diseases and blood brain barrier damage, which plays an important role in the development and recovery of normal CNS after injury [201203]. Recently, results of a meta-analysis showed a significant difference in S100B levels between survived and died subjects with head injuries on overall follow-up timeline, during treatment, or 6 months with an average threshold value that varies according to the study method used [203]. In terms of other S100 proteins and nervous injury, following peripheral nerve injury an immediate acute immune response distally and proximally to the lesion site with rapid transcriptional activation of S100a8 and S100a9 genes resulting in S100A8/A9 hetero- and homodimers has been observed [204]. The subsequently provoked release of chemokines and cytokines by activated Schwann cells induced an initial chemotactic gradient, which was responsible for the transmigration of hematogenous immune cells toward the traumatized site of the injured nerve [204]. Also after severe burn injury, S100A8 and S100A9 levels in bloodstream were correlated with poor outcome [205]. These proteins were found to be increased in a biphasic manner with the early peak at initial presentation representing the innate response to injury [205]. Similar to HMGB1, S100A8 and S100A9 levels in the bloodstream increase early in the acute phase of trauma or brain injury. Thus, although large number of studies deals with neurological trauma, there is some evidence that other S100 protein, i.e., S100B is also elevated in patients with major extracranial trauma [203, 206208]. S100B concentrations upon admission correlated positively with injury severity and decreased survival in major trauma patients, observations which were independent of head injury [206, 208]. In general, circulating levels of S100B in severely injured trauma patients were increased as compared to healthy volunteers [206]. Even more, S100B levels correlated with systemic levels of sE-selectin and von willebrand factor, which are indicating endothelial cell injury [206, 209]. Further mechanistical analyses of human endothelial cells transfected with S100B uncovered an increase in apoptosis and levels of proinflammatory cytokines IL-6 and IL-8 [206]. Thus, S100B apparently correlates with endothelial damage indicating important pathomechanistical influence of S100B in the recovery from trauma. However, it cannot be ruled out that increased S100B after soft tissue trauma might occur due to peripheral nerve injury. On the other hand, currently a protective role of certain members of the S100 family as briefly addressed above cannot be ruled out. Notably in a study on human trauma victims, human blunt trauma survivors were seen to produce higher levels of S100A8 and S100A9 than non-survivors, and a preliminary screen suggested that interferon-γ inducible protein 10 (CXCL10) was significantly increased in monocytes/macrophages stimulated with S100A8/S100A9 [210]. Thus, heat shock protein (HSP)70, S100A8/S100A9 DAMPs appear to exert a protective role after trauma. This remains to be further elucidated in future.

Histones

Other critical members of DAMPs are the circulating extracellular histones which act as clinically relevant mediators of multiple organ injuries [211, 212]. These nucleoproteins enable DNA compaction into nucleosomes, thereby contributing to the structural organization and stability of chromatin. As they localize in the nucleus, their release into the extracellular space, as free histones, DNA-bound histones (nucleosomes), or part of NETs, has been recently recognized as candidate of the DAMP family, and as such in all three forms, they can be detected in serum after significant cellular death which is occurring in infective or sterile SIRS [213, 214]. Their extracellular activity has a long history, while more than 50 years ago the first observations uncovered the antibacterial potential of these molecules [215, 216]. Since Xu et al. [217] have demonstrated that extracellular histones released in response to inflammatory challenge contributed to endothelial dysfunction, organ failure and death during sepsis, the mechanisms of histone-mediated injury in certain organs have been extensively studied. Clinically, elevated levels of circulating histones and histone–DNA complexes were associated with the incidence of MOF, disseminated intravascular coagulation, cardiac injury, arrhythmia, and ventricular dysfunction in patients with sepsis [218, 219]. Therefore, the extracellular histone also acts as an immunothrombotic agent. Furthermore, pharmacological targeting by antibodies to histones or by activated protein C (APC) reduced the mortality of septic mice [217]. Thus, the authors proposed extracellular histones as potential molecular targets for therapeutical treatment of sepsis and other inflammatory diseases.
Pathomechanistically, histone administration resulted in neutrophil margination, vacuolated endothelium, intra-alveolar hemorrhage and macro- and microvascular thrombosis in vivo [217]. Histones mainly bind and activate TLR, e.g., TLR2, TLR4 or TLR9 on various cells, and similar to other DAMP, they subsequently trigger the inflammatory response [213, 220, 221]. This has been confirmed in vivo, by showing that histone administration led to death after a dose-dependent aggravation of multiple organ injury in mice [211]. The causative effects for organ injuries were histone-driven endothelial damage, and the associated release of HMGB1 [211]. The authors concluded that extracellular histones induce multiple organ injury in two progressive stages via direct endothelia disruption, and the subsequent release of other DAMPs [211]. Circulating levels of histones were significantly increased after severe non-thoracic blunt trauma in patients [222]. Furthermore, enhanced histone levels positively correlated with severe complications such as the incidence of ALI, and dismal prognosis, as well as with markers of endothelial damage and coagulation activation [222]. The harmful impact of histones on endothelia has been confirmed in vitro and in vivo demonstrating that histones directly damaged endothelial cells, stimulated cytokine release (e.g., TNFα, IL-6, and IL-10), induced NET formation and MPO release [222]. An anti-histone antibody reduced the harmful changes and protected mice from histone-induced lethality [222]. Also in a large in vivo model of polytrauma, increased extracellular histone levels could have been linked to cardiac dysfunction after porcine multiple trauma [223]. Thus, histone release plays an important pathological role in trauma-associated injuries [222, 223].

Heat shock proteins

So far, only a few studies have investigated the relation of HSP to trauma-induced tissue damage [224]. Heat shock or stress response is a highly preserved cell response to injury which is actually a cell defense mechanism characterized by an increased expression of heat shock or stress proteins. HSP constitutes a group of proteins that primarily act as molecular chaperones in the cytosol which are constitutively expressed by all cells. HSPs are essential for significant cell processes such as protein folding, protein protection from denaturation or aggregation, and facilitation of protein transport through membrane channels [225, 226]. Thus, their specific elevated expression during the heat shock response stands for their essential role in protecting cells from stress, and preparing them to survive under environmental challenges [227]. HSPs are induced by a variety of cellular stress factors, including hypo- and hyperthermia, UV radiation, pathogens, and other forms of stress [16, 227, 228]. HSP family members are named according to their molecular mass [229]. Although HSPs were considered for some time as intracellular molecules that could only be released from necrotic cells by a passive mechanism [230], meanwhile it is known that they may be released by non-necrotic cells via an active mechanism including the non-classic protein release pathway, through which HSPs are released both as free proteins and within highly immunogenic exosomes [231]. It has been under discussion, if HSP in general constitute DAMPs, or if the most widely studied HSP70 is rather an exception [224]. This is not easy to evaluate since very often contamination with immunogenic molecules, i.e., LPS or DNA, can generate false-positive responses and results.
Upon release, HSP70 can stimulate monocytes/macrophages, microglia, and dendritic cells via the TLR2 and 4 and cluster of differentiation (CD)14 pathways, subsequently leading to the activation of intracellular signaling pathways [229]. Next to TLR, HSP70 binds to CD36, CD40, CD91, siglec-5, and siglec-14, lectin-like oxidized low-density lipoprotein receptor 1, and scavenger receptor class A to induce pro- or anti-inflammatory responses on a range of cells, mainly those of the innate immune system with exception of T lymphocytes [232234]. Notably in studies of cerebral ischemia, neurodegenerative diseases, and epilepsy, HSP70 reduces protein aggregates, intracellular inclusions, and apoptosis improving neurological outcomes [229, 235237]. However, conflictive results are found regarding HSP functions. On the one hand increased levels of HSP27, HSP60, HSP70, and HSP90 in patients with infectious septic complications [238240], and significant improvement in mortality, lung function, local and systemic inflammation in a mouse model of severe sepsis-induced ALI after therapeutic HSP90 inhibition have been observed [241]. On the other hand, in a CLP-induced sepsis model in HSP70.1/3 knockout mice, NF-κB binding/activation, TNFα and IL-6 in lungs and mortality were increased [242]. Thus, HSP70 may confer protection from ARDS via acting at least partly through the NF-κB pathway and thereby reducing the proinflammatory cytokine response [242]. In line with these observations, HSP70 played a protective role in an age-dependent response to sepsis by preventing excessive gut apoptosis and both pulmonary and systemic inflammation [243]. Thus, multiple data are available suggesting that HSP are potent regulators of inflammatory events.
Severe trauma causes enhanced expression of HSP in polymorphonuclear leukocytes (PMNL) during the acute post-injury phase [244]. In comparison with healthy volunteers increased expressions of HSP27, HSP60, HSP70, and HSP90 in PMNL from trauma patients or patients with burn trauma were found, indicating that this enhanced expression of HSP may regulate PMNL functions [244, 245]. Concomitant with these changes was an increase in oxidative activity in PMNL, and markedly inhibited cell apoptosis after thermal injury [245]. Extracellular HSP60 release was observed within 30 min after trauma and correlated with the development of ALI [246]. Extracellular HSP have powerful immune properties by activating the classical complement pathway, participating in exogenous antigen processing and presentation, and showing immune reactivity to endogenous HSP [247]. Thus, HSP can also have an immunostimulatory effect and activate the host inflammatory response [224]. In mechanistical in vitro studies, it was shown that HSP60 caused the release of nitric oxide by macrophages [246]. Taken together with previously described in vivo observations, the authors suggest that the extracellular release of the immature HSP60 may be associated with traumatic cell necrosis, and could be involved in the release of NO by immune competent cells, subsequently inducing an activation of the local inflammatory response [246]. Similarly, systemic HSP72 levels were markedly elevated immediately after admission of severely traumatized patients to the emergency department as compared to healthy volunteers [248]. Interestingly, elevated initial HSP72 levels were associated with improved survival in severely traumatized patients, without showing any association to the overall injury severity [248]. Notably concerning HSP72 conflictive reposts are shown. Pittet et al. [248] reported that HSP72 was neither related to the incidence or severity of the inflammatory response nor to post-injury organ dysfunction.
HSP70 serum concentrations have been up to ten times higher immediately after injury in polytraumatized patients versus control subjects, levels that remained elevated until 48 h after the accident following a time kinetics concordant with that previously described [75, 248]. The magnitude of this increase was related to injury severity and prediction of secondary infection [249]. If HSP70 levels decreased in the period from 60 to 90 h after trauma, the patient had a better outcome as compared to those patients without a decrease in HSP70 levels [249]. The high extracellular concentration of HSP70 was even greater in patients with MODS [224]. Although the authors did not include follow-up of survival after hospital discharge and all patients except one survived during admission, the greater elevation of extracellular HSP70 in patients with MODS corroborates the proinflammatory character of this protein in severe trauma as reported by others as well, who also correlated HSP70 with survival and morbidity [250, 251]. In summary, these data support the hypothesis that HSP70 is produced as a danger signal to stimulate the immune systems of trauma patients [224, 252].

Nucleic acids

All human cells contain nucleic acids as deoxynucleic acid (DNA), messenger ribonucleic acid (RNA), or mtDNA while mature erythrocytes may also retain some residual non-functional mtDNA [253, 254]. The endosymbiotic theory suggesting that mitochondria originate from bacteria, provide the cellular content of mtDNA. Usually, nucleic acids are released into the circulation after cell necrosis and nuclear destruction, but their active release has been reported as well [16, 255, 256]. This so-called extracellular cfDNA can be built up by either DNA or different species of RNA, and based on its origin, whether it is host or pathogen derived, it accounts for a DAMP or a PAMP, respectively [255]. TLRs are evolutionary conserved pattern recognition receptors (PRR) for detection of multiple microbial products including bacterial DNA and RNA, sense invading microbes and initiate a rapid immune response [257, 258]. However, adjacent to membrane-bound TLR3, TLR7, TLR8, TLR9, and RAGE, soluble mainly cytosolic receptors retinoic acid-inducible gene I, melanoma differentiation-associated protein 5, and cyclic guanosine monophosphate-adenosine monophosphate can recognize the cfDNA signals [255, 259, 260]. Intracellular mechanisms of mtDNA inflammation include inflammasome activation and stimulator of interferon gene pathway activation [261]. However, although these mechanisms have not been confirmed directly in the trauma setting, based on available scientific research plausible mechanisms exist [262]. The cfDNA released from mammalian cells and bacteria is a potent stimulator of the innate immunity. The ability of the cell to distinguish dangerous nucleic acids from safe counterparts may be determined by the unique cellular location of nucleic acid-sensing PRR [263265]. In addition to proinflammatory activity, elevated plasma DNA and mtDNA as well as nDNA in patients with pulmonary embolism as compared to those patients with submassive pulmonary embolism or other diagnoses (pneumonia, myocardial infarction, thrombophlebitis, or normal lung scans) suggest that cfDNA may induce potent prothrombotic effects as well [266, 267]. NETs, which constitute a meshwork of DNA fibers comprising histones and antimicrobial proteins, stimulated thrombus formation both in vitro and in vivo, while the treatment with deoxyribonuclease (DNase) or anticoagulant heparin prevented NET-mediated thrombus formation [268]. Thus, cfDNA acts as potent immunothrombotic agents [265]. Increased levels of cfDNA in the form of nuclear DNA (nDNA), mtDNA, or NETs were found and correlated with detrimental outcomes in patients with sepsis [269, 270], cancer [271], autoimmune disease [272], cardiopulmonary bypass surgery [273], and solid organ transplantation [274].
As recently reviewed by Thurairajah et al. [262], clinicians have regarded DNA and mitochondria as intracellular structures, unrelated to the pathophysiology of trauma. Meanwhile, we are beginning to understand the large impact of cfDNA on posttraumatic inflammation. Release of cfDNA plays an important role in trauma [84, 256]. The group around Hauser [256] has shown that traumatic injury causes a release of mtDAMPs into the circulation, which have functionally important immune consequences. These DAMPs include formyl peptides and mtDNA as well, which can activate human polymorphonuclear neutrophils via TLR [256]. Briefly, tissue injury caused by trauma results in uncontrolled rupture of cell membranes and cell contents are spilled into the extracellular space [275, 276]. Adjacent to the release of mtDNA, numerous other DAMPs and ROS from damaged neighboring cells promote further necrosis and mtDNA release [50, 277]. As observed in traumatized patients, cf-mtDNA concentration positively correlated with injury severity, incidence of SIRS, and mortality [278281]. Plasma levels of DNA were increased concomitant with the injury severity caused by trauma, and the concentration of mtDNA correlated not only with SIRS but also with the occurrence of ALI and MODS [256, 278, 280282]. In vivo studies confirmed that hemorrhagic shock and trauma raised plasma mtDNA concentration [283]. The raised mtDNA concentration persisted throughout the 7 days of sampling. Not only traumatic insults but also surgical interventions result in tissue injury and can also increase cf-mtDNA release [284]. Also upon femoral reaming or cardiopulmonary bypass surgery, high quantities of mtDAMPs were identified [285]. Even more, the higher concentrations of mtDNA correlated with postoperative complications [273]. Interestingly, endotracheal tube placement in surgery has been associated with higher concentrations of mtDNA in throat lavage fluid, and a correlation between non-infected sore throat post-intubation and elevated throat lavage mtDNA concentration was found [286]. Although it appears that the inflammatory effects of mtDNA can be beneficial and harmful, in the trauma setting, the increase in cf-mtDNA associated with MOF proposes rather a harmful scenario of mtDNA-induced inflammation [80, 278, 287]. Interestingly, Prikhodko et al. [288] have demonstrated that the levels of circulating mtDNA significantly increased in trauma patients compared to those in healthy volunteers, but that purified mtDNA could not stimulate innate immune cells. These data suggest that the level of extracellular DNA in the blood may be a suboptimal marker for human disease. Development of new approaches to detect circulating extracellular DNA that actually activates innate immune cells would be beneficial. Although enormous research and efforts to develop therapeutic agents that neutralize cf-mtDNA are ongoing, there is still insufficient knowledge to including this into clinical management. Current goals are limited to the validation of cf-mtDNA as a biomarker of injury severity, and/or predictor of SIRS/MOF to improve the clinical management of patients.

Adenosine triphosphate

Adenosine 5′ triphosphate (ATP), another DAMP originating from mitochondria, can be released passively due to tissue damage or actively via channels or vesicular exocytosis, contributing to the induction of inflammation by activation and recruitment of various inflammatory cells, e.g., macrophages, neutrophils, and dendritic cells [46, 50, 289293]. Stimulation of virtually any mammalian cell type leads to the release of cellular ATP and autocrine feedback through a diverse set of different purinergic receptors [294]. Extracellular ATP signaling is transduced, e.g., via the purinergic receptor P2X7 with subsequent efflux of potassium ions, with following aggregation and activation of inflammasomes [290, 291, 294]. Thus, ATP gating of P2X7R with the subsequent assembly of the NLRP3 inflammasome/caspase-1 complex is critical for IL-1β maturation as well as its release [295]. Human neutrophils, which play an important role in tissue damage and repair, release ATP from the leading edge of the cell surface to amplify chemotactic signals and direct cell orientation and migration by feedback through P2Y2 nucleotide receptors [293, 296]. Thus, ATP release and autocrine feedback through P2Y2 and A3 receptors provide signal amplification, controlling gradient sensing and migration of neutrophils [293]. It was shown that systemic ATP impairs polymorphonuclear neutrophil functions by disrupting the endogenous purinergic signaling mechanisms that regulate cell activation and chemotaxis [297]. Based on their data, the authors suggest that removal of systemic ATP improves polymorphonuclear neutrophil function and host defenses, making this a promising new treatment strategy for sepsis [297].
In case of, e.g., overaggressive mechanical ventilation strategies, direct injury or cyclic deformation can release massive amounts of extracellular ATP from type I alveolar epithelial cells [298]. High levels of ATP may saturate ATP degradation resulting in increased alveolar ATP levels despite the ongoing enzymatic conversion to the immunosuppressive purine, adenosine [298]. The authors suggest that this sequence of events may be an important contributor to both pneumonia and clinical ALI.

Complement factors

Exposure to traumatic or infectious insults results in an immediate activation of the complement cascade as major fluid defense system of innate immunity [48, 299]. As nicely described by Huber-Lang, the complement system acts as a master alarm system during the molecular danger response after trauma and significantly contributes to the clearance of DAMPs and PAMPs [48, 299]. Activation of complement in trauma patients has been verified by detection of elevated plasma C3a, C5a, and C5b-9 levels which correlate with disease severity [73, 300305]. In septic patients, there is abundant evidence for complement activation and production of the anaphylatoxin C5a. In an experimental model of CLP-induced sepsis, interception of C5a or its receptors was shown to greatly improve survival in rodents, an effect that was associated with mitigation of the consumptive septic coagulopathy as well as reduction in the hyperinflammatory response, thereby reducing intensity of MOF, and septic shock [306, 307]. However, depending on the origin or extent of the damaged macro- and micromilieu, not only activation but also inhibition of the complement system leading to a maladaptive immune response and subsequent cellular and organ dysfunctions was observed [48, 299].

Extracellular vesicles

Extracellular vesicles (EVs) comprise various small, membranous vesicles that are released from activated or dying cells. Presently, there are two distinct populations of EVs based on their size: exosomes (30–100 nm) and microparticles (100–1000 nm). Different types of EVs are generated by different mechanisms of biogenesis [308]. Exosomes form by inward budding of multivesicular bodies membranes, while microparticles and apoptotic bodies are generated by outward budding of plasma membrane [308]. This process requires cytoskeletal reformations and sequential assembly of the endosomal sorting complex on the multivesicular bodies membranes [309311].
Upon release, EVs transfer their cargo by multiple mechanisms, such as endocytosis, phagocytosis, micropinocytosis, and membrane fusion [312, 313]. EVs carry the molecular signature of their origination cells including proteins, mRNAs, miRNAs (miRs), and lipids which make EVs effective cell-to-cell communicators [314319]. Therefore, specific EVs from activated or dying cells can be used as biomarkers. For example, circulating EVs with liver-specific proteome markers and miRs are released from lipotoxic hepatocytes in nonalcoholic fatty liver disease [320, 321]. Recently, it was shown that damaged hepatocytes from alcoholic liver disease released a key source of EVs containing a specific microRNA “barcode” [322]. This EVs specific, i.e., microRNA “barcode” is detectable in the blood [320, 322, 323]. Next to their biomarker character, EVs are potent immune modulators. Dendritic cell-derived exosomes express major histocompatibility complex (MHC) I, MHC II, and costimulatory molecules, and they can induce antigen-specific T cell responses [324326]. In case of cancer, it has been shown that exosomes and microparticles released from cancer cells contributed to the suppression of host immune surveillance, cancer progression and metastasis, and angiogenesis [327329]. In various other diseases, graft-versus-host disease, chronic kidney disease, and acute radiation injury EVs have been proposed as therapeutic to modulate the inflammatory responses and tissue regeneration [330332]. However, this remains to be considered carefully, since EVs themselves can express various procoagulants promoting the vascular thrombosis [333, 334]. Exosomes isolated from septic patients induced vascular dysfunction by inducing reactive oxygen species generation and endothelial cell apoptosis [335]. Inhibition of the exosome release prior to endotoxin challenge or CLP in mice significantly reduced the levels of circulating exosomes and diminished sepsis-induced cardiac inflammation, myocardial dysfunction, and mortality [336, 337]. Interestingly, increasing evidence is demonstrating that the positive effects of cell-based therapies may be mediated by exosomes released from the administered cells and that the corresponding microRNA cargo in these exosomes is largely responsible for the therapeutic effects [338].
With regard to traumatic injury, recently Eguchi et al. [339] have demonstrated that liver injury in alcohol-intoxicated trauma patients with severe injury was reflected by increased systemic EV numbers, their specific miR barcode, and the correlated increase in systemic inflammatory markers IL-6 and IL-33, with IL-33 being a marker of alcoholic liver disease. Interestingly, severely injured trauma patients without liver injury were found to have a reduced number of liver-derived EVs, no observed inflammatory infiltration, and reduced specific miR “barcode” [339]. Trauma/hemorrhagic shock causes a release of proinflammatory mediators into the mesenteric lymph that may trigger a systemic inflammatory response and subsequent organ failure. Recently, it has been shown that exosomes in post-shock mesenteric lymph were biologically active mediators of this inflammation and that they carry a distinct, proinflammatory protein cargo [340, 341]. Interestingly, a stimulation of the vagus nerve prevented the T/HS-induced changes in mesenteric lymph exosome protein [341]. Subsequently, Williams et al. [341] propose a novel mechanism by which the neuroenteric axis may limit the systemic inflammatory response after injury protein. Neuroinflammation is a response against harmful effects of diverse stimuli and participates in the pathogenesis of brain and spinal cord injury. The innate immune response plays an important role in neuroinflammation following CNS injury via activation of inflammasomes [342, 343]. Interestingly, exosomes derived from neurons can deliver short-interfering RNA into the CNS to significantly decrease inflammasome activation after injury [343]. Thus, exosomes offer a new therapeutic approach to deliver RNA-based drugs to block inflammation after CNS injury. Several studies have characterized EVs in blood collected after traumatic injury, demonstrating their levels, cellular origins, adhesion molecule expression, and procoagulant activity [344348]. Blood from patients who developed sepsis after trauma increased levels of platelet microparticles and platelet-leukocyte aggregates as compared to healthy controls were detected [344]. Kuravi et al. [349] have shown that a significant increase in plasma EVs after severe traumatic injury had procoagulant and proinflammatory effects that may influence outcomes. Thus, EVs as DAMPs may play dual roles in tissue repair and damage.

Other DAMPs

Several other DAMPs playing important roles in the initiation and regulation of immune responses as well as in the post-injury regeneration have not been addressed here. Of those, notably other mitochondria-derived DAMPs including mitochondrial formyl peptides, heme, and cytochrome (Cyt)C have not been described [46].
Heme, a tetrapyrrole containing a central iron ion, is synthesized in the mitochondria and constitutes a versatile signaling molecule controlling the activities of diverse regulators ranging from transcription factors to MAP kinases [350]. Upon severe trauma, this DAMP can bind to TLR4 and trigger a number of downstream stress response pathways, and it was suggested to play a central role in the pathogenesis of severe sepsis [351].
CytC which is also released from dying cells during cellular trauma can act as a danger signal in the extracellular space. In experiments on rabbits, oxidative injury to mitochondria has been demonstrated to be associated with high mitochondrial expression of CytC [352]. In studies on a burn model in mice, circulating CytC levels were found to be elevated eightfold within 3 h post-injury and remained elevated at 24 h [353].
Upon destruction of the red cells in the blood vessels, a significant quantity of hemoglobin and other contents of these cells are released into the circulation [354]. In case that this cell-free hemoglobin is not neutralized to its inert, non-toxic form by its scavenger proteins, significant damage in the vascular, perivascular and endothelial spaces occurs [354, 355].
Extracellular cyclophilin A (CypA) is a DAMP that has been associated with rheumatoid arthritis, liver injury and severe sepsis as well [356358]. It can act as a chemotactic agent for inflammatory cells via the CD147 receptor, and it can directly stimulate the proinflammatory response. However, in an in vitro shock tube model of blast TBI with human neuroblastoma cells, a potentially neuroprotective mechanism involving released CypA has been proposed, since the accumulation of CypA in the culture medium after repeated blast exposures supported the hypothesis that extracellular CypA-mediated neuroprotection [359]. Post-exposure treatment of the cells with purified recombinant CypA confirmed this protection against blast-induced neuronal injury [359].
Uric acid can be released from injured cells as other DAMPs as well [360]. Inside the cells, it is soluble, but it precipitates to monosodium urate microcrystals in its extracellular form, exerting inflammatory properties, as evident by its accumulation in tissues and gout [49, 361]. Uric acid crystals act via inflammasomes, resulting in the production of active proinflammatory cytokines IL-1β and IL-18 and neutrophilic influx [362]. Elevated levels of serum uric acid correlated with early acute kidney injury after severe burns [363]. The authors propose uric acid-related aberrant inflammation to be one of the pathogenic factors [363].

Recognition, signaling and cellular response

The “danger model” was proposed as an alternative to the “self versus non-self recognition model” [364, 365]. Briefly, the immune system discriminates not only between self and non-self, but also between safe and dangerous as well, as clinically proved by Walter Land and presented by Polly Matzinger in the “Danger Model,” expanding the work of Janeway and others [70, 71, 364367]. This complex response to stress employs numerous equivalent or comparable components of PAMPs, DAMPs, or newly introduced SAMPs, which can be found in most vertebrates, invertebrates, and even plants [23, 366369]. These molecules are sensitized and recognized via PRR mediating very potent immune responses [370, 371]. Several classes of PRR have been identified so far, including the most prominent group of toll-like receptors (TLR), nucleotide oligomerization domain (NOD)-like receptors (NLR), members of the c-type lectin receptors (CLR) like mannose binding lectine (MBL) or receptor for advanced glycation end products (RAGE) among others [187, 370, 372, 373].

TLRs

Among TLRs, membrane-bound TLRs recognize microbial components and environmental danger signals, such as lipopolysaccharide (LPS) of gram negative, lipoteichoic acid, or peptidoglycan of Gram-positive bacteria, or even liporabinomannan of mycoplasma, and, e.g., endogenous HMGB proteins or HEME [370, 374378]. Intracellular TLRs recognize predominantly nucleic acids derived from bacteria and viruses, such as single- or double-stranded RNA from viruses, unmethylated CpG motifs, or purine analogs and other components of cellular stress [370]. PRR signaling is intracellularly transduced, e.g., via MAPK pathways to nuclei, with subsequent activation of diverse transcription factors, including NF-κB, and thereby induced a cellular responses [379, 380]. The cellular responses include, i.e., expression of cytokines or adhesion molecules to accelerate inflammation and diapedesis of the immune effector cells [380]. Interestingly, in a feedback loop, those inflammatory mediators themselves can induce, e.g., DAMPs to potentiate inflammation [381].
The cell surface receptor RAGE belongs to the immunoglobulin superfamily and was first described in 1992. Interestingly, RAGE is a multiligand receptor that can bind structurally diverse array of several molecules, including DAMPs like HMGB1, but also S100 family members and amyloid-β proteins [382384]. RAGE is predominantly involved in the recognition of endogenous molecules released in the context of infection, chronic inflammation, or physiological stress, and its activation has been confirmed in various diseases, including sepsis and cardiovascular disease among others [187, 385387]. Since RAGE shares numerous TLR ligands, its activation can induce numerous cellular signaling pathways, which among other transcription factors modulate NF-κB, activator protein-1, or signal transducers and activators of transcription-3 [187, 388390]. In general, RAGE triggers a proinflammatory response, generation of nitric oxide, and several adhesion molecules but also RAGE itself causing and enhancing a continuous inflammatory response [389, 391394].

NLR

Adjacent to TLRs, NLR sensitizes signals of cellular stress, such as ATP-induced activation of P2X7 channels and the efflux of potassium ions, host cell-free nuclear DNA, ROS as well as bacterial peptidoglycans, crystalline material, peptide aggregates, bacterial toxins and numerous others [395397]. As part of the multiprotein inflammasome complexes, they mediate the processing of biologically inactive precursors of, e.g., IL-1β or IL-18 into their respective bioactive forms, which furthermore promote pyroptosis, a specific form of cell death [396, 398402]. A direct link for the NLR sensor molecule-mediated inflammasome activation via binding of either a PAMP or a DAMP is still under discussion [397, 402]. The proposed classical mechanism implies two signals, the first one mediated by a TLR [403, 404], and in parallel, the potential DAMP signal for inflammasome induction by, e.g., potassium influx or binding of ATP to P2X7 [126, 402407]. Apart from activating certain cytokines, the inflammasome complex induces cell lysis in form of pyroptosis, which has been introduced in 2001 [408]. Pyroptosis describes the well-orchestrated lysis of the cell, which is initiated by an inflammasome activation and consecutive formation of caspase-1-dependent pores of 1–2 nm width [408]. As a consequence, cells are prone to swelling and lysis, but also to the release of intracellular molecules, which act as DAMPs, e.g., HMGB1 or ATP, which are described above. Briefly, pyroptosis is an important mode of cell death in DAMP-mediated enhancing and spreading of the inflammatory response, which mainly affects the cells of the myeloid lineage but also occurs in epithelial, endothelial cells and neurons.

CLR

CLR has been mainly described in the context of binding to PAMPs [409]. There are two groups of transmembrane CLR, like dectin-1 or dectin-2 subgroups, and a group of soluble CLR including MBL, which comprise this large family of receptors that are expressed on most cell types including macrophages and dendritic cells [409, 410]. The signaling pathways are either mediated directly via, e.g., NF-κB, or modulated signaling by TLR, triggering cellular phagocytosis, maturation of dendritic cells, chemotaxis, respiratory burst, and cytokine production [410]. More than 10 years ago, Yamasaki et al. [411] found that macrophage-inducible c-type lectin (Mincle) senses nonhomeostatic necrotic dead of cells due to a component of small nuclear ribonucleoprotein, spliceosome-associated protein 130 (SAP130), and induces thereby the production of inflammatory cytokines and chemokines to potentiate the neutrophilic infiltration of damaged tissue. The endogenous Mincle ligand SAP130 was confirmed as a danger signal, which can be released by damaged cells, thereby activating inflammatory responses including inflammasome activation [412].

Conclusions

This review discusses only a few of the currently discussed DAMPs that play important roles in the inflammatory or regenerative responses upon trauma. Although the provided list is certainly both incomplete and provides only a limited overview to the concept of trauma-induced DAMPs, it remains evident that due to the bivalent character and often pleotropic effects of a DAMP, its functions in the posttraumatic inflammation and regeneration still remain to be studied in detail. However, the new insights into the contribution of DAMPs to the sepsis-like inflammation and immune paralysis-like states that can be observed in severely injured patients will be helpful in further specifying the clinical picture presented by, e.g., polytrauma. Considering the “SIRS and CARS paradigm,” and the terms mixed antagonist response syndrome and persistent inflammation, immunosuppression and catabolism syndrome, the role of these injury-induced activating and suppressing molecules clearly marks their predominant role in posttraumatic pathologies. It is indisputable that both systemic as well local presence of DAMPs is obligatory for the immune response upon traumatic insult. Yet, adjacent to their ability to be used as biomarkers to indicate or monitor disease or injury severity, and for eventual improving the timing of secondary surgery, they remain either negative or positive contributing factors for the disease development. In polytrauma, there is tentative evidence of a model suggesting that, simultaneously or somewhat retardedly with uncontrolled overshooting emission of proinflammatory DAMPs in excess (resulting in hyperinflammatory SIRS and MOF), uncontrolled overshooting generation/emission of SAMPs in excess takes place. Those “suppressing molecules” may lead to an exaggerated and long-lasting CARS associated with a stage of immunosuppression—still aimed at maintaining tissue homeostasis. Considering this scenario, it certainly remains to be further discussed if the biologically natural attempt of the innate immune defense system to reach homeostasis via emission of SAMPs in case of severe injury-induced SIRS potentially and tragically ends up with the creation of an overshooting anti-inflammatory/proresolving and immunosuppressive milieu rendering the patient susceptible to secondary life-threatening, often lethal infections. Although the data from SAMPs involved in polytrauma are sparse and, though, the various suppressing molecules are not systemically investigated, there is preliminary evidence suggesting a role of this class of suppressing DAMPs in posttraumatic inflammation. Therefore, increased knowledge exploring the dual role of DAMPs and the interplay with SAMPs after trauma, and with their nuclear functions, transcriptional targets, release mechanisms, cellular sources, and other functions and interactions in traumatized patients is warranted. Adjacent to in vivo and in vitro studies, and furthermore, based on some contradictory findings, which often simply originate from differences between the immune system of animals and human, further clinical research is necessary to resolve this puzzle.

Compliance with ethical standards

Conflict of interest

The authors declare no conflicts of interest.
No informed consent is necessary.

Human and animal rights

This study does not include any human participants and/or animals.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Orthopädie & Unfallchirurgie

Kombi-Abonnement

Mit e.Med Orthopädie & Unfallchirurgie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Neuer Inhalt

Print-Titel

Literatur
2.
3.
Zurück zum Zitat Wutzler S, et al. Pathophysiology of multiple trauma: intensive care medicine and timing of treatment. Chirurg. 2013;84(9):753–8.PubMed Wutzler S, et al. Pathophysiology of multiple trauma: intensive care medicine and timing of treatment. Chirurg. 2013;84(9):753–8.PubMed
4.
Zurück zum Zitat Wafaisade A, et al. Epidemiology and risk factors of sepsis after multiple trauma: an analysis of 29,829 patients from the Trauma Registry of the German Society for Trauma Surgery. Crit Care Med. 2011;39(4):621–8.PubMed Wafaisade A, et al. Epidemiology and risk factors of sepsis after multiple trauma: an analysis of 29,829 patients from the Trauma Registry of the German Society for Trauma Surgery. Crit Care Med. 2011;39(4):621–8.PubMed
5.
Zurück zum Zitat Osuka A, Ogura H, Ueyama M, Shimazu T, Lederer JA. Immune response to traumatic injury: harmony and discordance of immune system homeostasis. Acute Med Surg. 2014;1(2):6. Osuka A, Ogura H, Ueyama M, Shimazu T, Lederer JA. Immune response to traumatic injury: harmony and discordance of immune system homeostasis. Acute Med Surg. 2014;1(2):6.
6.
Zurück zum Zitat Rose S, Marzi I. Mediators in polytrauma–pathophysiological significance and clinical relevance. Langenbecks Arch Surg. 1998;383(3–4):199–208.PubMed Rose S, Marzi I. Mediators in polytrauma–pathophysiological significance and clinical relevance. Langenbecks Arch Surg. 1998;383(3–4):199–208.PubMed
7.
Zurück zum Zitat Charters E, Gillett L, Simpson GK. Efficacy of electronic portable assistive devices for people with acquired brain injury: a systematic review. Neuropsychol Rehabil. 2015;25(1):82–121.PubMed Charters E, Gillett L, Simpson GK. Efficacy of electronic portable assistive devices for people with acquired brain injury: a systematic review. Neuropsychol Rehabil. 2015;25(1):82–121.PubMed
8.
Zurück zum Zitat Bridgeland HJ, et al. Polytrauma in veterans: what does it mean for the medical-surgical nurse? Medsurg Nurs. 2014;23(4):213-7. Bridgeland HJ, et al. Polytrauma in veterans: what does it mean for the medical-surgical nurse? Medsurg Nurs. 2014;23(4):213-7.
9.
Zurück zum Zitat Gironda RJ, et al. Traumatic brain injury, polytrauma, and pain: challenges and treatment strategies for the polytrauma rehabilitation. Rehabil Psychol. 2009;54(3):247–58.PubMed Gironda RJ, et al. Traumatic brain injury, polytrauma, and pain: challenges and treatment strategies for the polytrauma rehabilitation. Rehabil Psychol. 2009;54(3):247–58.PubMed
10.
Zurück zum Zitat Bouillon B, Marzi I. The updated German “Polytrauma - Guideline”: an extensive literature evaluation and treatment recommendation for the care of the critically injured patient. Eur J Trauma Emerg Surg. 2018;44(Suppl 1):1.PubMed Bouillon B, Marzi I. The updated German “Polytrauma - Guideline”: an extensive literature evaluation and treatment recommendation for the care of the critically injured patient. Eur J Trauma Emerg Surg. 2018;44(Suppl 1):1.PubMed
11.
Zurück zum Zitat Maier B, et al. Early versus late onset of multiple organ failure is associated with differing patterns of plasma cytokine biomarker expression and outcome after severe trauma. Shock. 2007;28(6):668–74.PubMed Maier B, et al. Early versus late onset of multiple organ failure is associated with differing patterns of plasma cytokine biomarker expression and outcome after severe trauma. Shock. 2007;28(6):668–74.PubMed
12.
14.
Zurück zum Zitat Lord JM, et al. The systemic immune response to trauma: an overview of pathophysiology and treatment. Lancet. 2014;384(9952):1455–65.PubMedPubMedCentral Lord JM, et al. The systemic immune response to trauma: an overview of pathophysiology and treatment. Lancet. 2014;384(9952):1455–65.PubMedPubMedCentral
15.
Zurück zum Zitat Land WG. Allograft injury mediated by reactive oxygen species: from conserved proteins of Drosophila to acute and chronic rejection of human transplants. Part III: interaction of (oxidative) stress-induced heat shock proteins with toll-like receptor-bearing cells of innate immunity and its consequences for the development of acute and chronic allograft rejection. Transplant Rev. 2003;17(2):20. Land WG. Allograft injury mediated by reactive oxygen species: from conserved proteins of Drosophila to acute and chronic rejection of human transplants. Part III: interaction of (oxidative) stress-induced heat shock proteins with toll-like receptor-bearing cells of innate immunity and its consequences for the development of acute and chronic allograft rejection. Transplant Rev. 2003;17(2):20.
16.
Zurück zum Zitat Timmermans K, et al. Danger in the intensive care unit: damps in critically ill patients. Shock. 2016;45(2):108–16.PubMed Timmermans K, et al. Danger in the intensive care unit: damps in critically ill patients. Shock. 2016;45(2):108–16.PubMed
17.
Zurück zum Zitat Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis. 2013;13(3):260–8.PubMedPubMedCentral Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis. 2013;13(3):260–8.PubMedPubMedCentral
18.
Zurück zum Zitat Osuchowski MF, et al. Circulating cytokine/inhibitor profiles reshape the understanding of the SIRS/CARS continuum in sepsis and predict mortality. J Immunol. 2006;177(3):1967–74.PubMed Osuchowski MF, et al. Circulating cytokine/inhibitor profiles reshape the understanding of the SIRS/CARS continuum in sepsis and predict mortality. J Immunol. 2006;177(3):1967–74.PubMed
20.
Zurück zum Zitat Adib-Conquy M, Cavaillon JM. Compensatory anti-inflammatory response syndrome. Thromb Haemost. 2009;101(1):36–47.PubMed Adib-Conquy M, Cavaillon JM. Compensatory anti-inflammatory response syndrome. Thromb Haemost. 2009;101(1):36–47.PubMed
21.
Zurück zum Zitat Bhan C, et al. Role of cellular events in the pathophysiology of sepsis. Inflamm Res. 2016;65(11):853–68.PubMed Bhan C, et al. Role of cellular events in the pathophysiology of sepsis. Inflamm Res. 2016;65(11):853–68.PubMed
22.
Zurück zum Zitat Reikeras O. Immune depression in musculoskeletal trauma. Inflamm Res. 2010;59(6):409–14.PubMed Reikeras O. Immune depression in musculoskeletal trauma. Inflamm Res. 2010;59(6):409–14.PubMed
23.
Zurück zum Zitat Land WG. Damage-associated molecular patterns in human diseases, vol. 1. Berlin: Springer; 2018. Land WG. Damage-associated molecular patterns in human diseases, vol. 1. Berlin: Springer; 2018.
24.
Zurück zum Zitat Bone RC. Sir Isaac Newton, sepsis, SIRS, and CARS. Crit Care Med. 1996;24(7):1125–8.PubMed Bone RC. Sir Isaac Newton, sepsis, SIRS, and CARS. Crit Care Med. 1996;24(7):1125–8.PubMed
25.
Zurück zum Zitat Bone RC, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101(6):1644–55.PubMed Bone RC, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101(6):1644–55.PubMed
26.
Zurück zum Zitat Hoover L, et al. Systemic inflammatory response syndrome and nosocomial infection in trauma. J Trauma. 2006;61(2):310–6 (discussion 316-7).PubMed Hoover L, et al. Systemic inflammatory response syndrome and nosocomial infection in trauma. J Trauma. 2006;61(2):310–6 (discussion 316-7).PubMed
27.
Zurück zum Zitat Bochicchio GV, et al. Persistent systemic inflammatory response syndrome is predictive of nosocomial infection in trauma. J Trauma. 2002;53(2):245–50 (discussion 250-1).PubMed Bochicchio GV, et al. Persistent systemic inflammatory response syndrome is predictive of nosocomial infection in trauma. J Trauma. 2002;53(2):245–50 (discussion 250-1).PubMed
28.
Zurück zum Zitat Sauaia A, Moore FA, Moore EE. Postinjury inflammation and organ dysfunction. Crit Care Clin. 2017;33(1):167–91.PubMedPubMedCentral Sauaia A, Moore FA, Moore EE. Postinjury inflammation and organ dysfunction. Crit Care Clin. 2017;33(1):167–91.PubMedPubMedCentral
29.
Zurück zum Zitat Azevedo LC. The many facets of sepsis pathophysiology and treatment. Shock. 2013;39(Suppl 1):1–2.PubMed Azevedo LC. The many facets of sepsis pathophysiology and treatment. Shock. 2013;39(Suppl 1):1–2.PubMed
30.
Zurück zum Zitat Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. 2003;348(2):138–50.PubMed Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. 2003;348(2):138–50.PubMed
31.
Zurück zum Zitat Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13(12):862–74.PubMedPubMedCentral Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13(12):862–74.PubMedPubMedCentral
32.
Zurück zum Zitat Balk RA. Systemic inflammatory response syndrome (SIRS): where did it come from and is it still relevant today? Virulence. 2014;5(1):20–6.PubMed Balk RA. Systemic inflammatory response syndrome (SIRS): where did it come from and is it still relevant today? Virulence. 2014;5(1):20–6.PubMed
33.
Zurück zum Zitat Mosevoll KA, et al. Inflammatory mediator profiles differ in sepsis patients with and without bacteremia. Front Immunol. 2018;9:691.PubMedPubMedCentral Mosevoll KA, et al. Inflammatory mediator profiles differ in sepsis patients with and without bacteremia. Front Immunol. 2018;9:691.PubMedPubMedCentral
34.
Zurück zum Zitat Keel M, Trentz O. Pathophysiology of polytrauma. Injury. 2005;36(6):691–709.PubMed Keel M, Trentz O. Pathophysiology of polytrauma. Injury. 2005;36(6):691–709.PubMed
35.
Zurück zum Zitat Lustenberger T, et al. The severity of injury and the extent of hemorrhagic shock predict the incidence of infectious complications in trauma patients. Eur J Trauma Emerg Surg. 2009;35(6):538–46.PubMed Lustenberger T, et al. The severity of injury and the extent of hemorrhagic shock predict the incidence of infectious complications in trauma patients. Eur J Trauma Emerg Surg. 2009;35(6):538–46.PubMed
37.
Zurück zum Zitat Sarabhai T, et al. Serum alpha-1 Antitrypsin (AAT) antagonizes intrinsic apoptosis induction in neutrophils from patients with systemic inflammatory response syndrome. PLoS ONE. 2017;12(5):e0177450.PubMedPubMedCentral Sarabhai T, et al. Serum alpha-1 Antitrypsin (AAT) antagonizes intrinsic apoptosis induction in neutrophils from patients with systemic inflammatory response syndrome. PLoS ONE. 2017;12(5):e0177450.PubMedPubMedCentral
38.
Zurück zum Zitat Yang R, et al. HMGB1 and extracellular histones significantly contribute to systemic inflammation and multiple organ failure in acute liver failure. Mediat Inflamm. 2017;2017:5928078. Yang R, et al. HMGB1 and extracellular histones significantly contribute to systemic inflammation and multiple organ failure in acute liver failure. Mediat Inflamm. 2017;2017:5928078.
39.
Zurück zum Zitat Yatim N, Cullen S, Albert ML. Dying cells actively regulate adaptive immune responses. Nat Rev Immunol. 2017;17(4):262–75.PubMed Yatim N, Cullen S, Albert ML. Dying cells actively regulate adaptive immune responses. Nat Rev Immunol. 2017;17(4):262–75.PubMed
40.
Zurück zum Zitat Raymond SL, et al. Microbial recognition and danger signals in sepsis and trauma. Biochim Biophys Acta. 2017;1863:2564–73.PubMedCentral Raymond SL, et al. Microbial recognition and danger signals in sepsis and trauma. Biochim Biophys Acta. 2017;1863:2564–73.PubMedCentral
41.
Zurück zum Zitat Tsukamoto T, Chanthaphavong RS, Pape HC. Current theories on the pathophysiology of multiple organ failure after trauma. Injury. 2010;41(1):21–6.PubMed Tsukamoto T, Chanthaphavong RS, Pape HC. Current theories on the pathophysiology of multiple organ failure after trauma. Injury. 2010;41(1):21–6.PubMed
42.
Zurück zum Zitat Horst K, et al. Impact of haemorrhagic shock intensity on the dynamic of alarmins release in porcine poly-trauma animal model. Eur J Trauma Emerg Surg. 2016;42(1):67–75.PubMed Horst K, et al. Impact of haemorrhagic shock intensity on the dynamic of alarmins release in porcine poly-trauma animal model. Eur J Trauma Emerg Surg. 2016;42(1):67–75.PubMed
43.
Zurück zum Zitat Qiao Z, et al. Using IL-6 concentrations in the first 24 h following trauma to predict immunological complications and mortality in trauma patients: a meta-analysis. Eur J Trauma Emerg Surg. 2017;44:679–87.PubMed Qiao Z, et al. Using IL-6 concentrations in the first 24 h following trauma to predict immunological complications and mortality in trauma patients: a meta-analysis. Eur J Trauma Emerg Surg. 2017;44:679–87.PubMed
44.
Zurück zum Zitat Ahmed Ali M, et al. Interleukin-17 as a predictor of sepsis in polytrauma patients: a prospective cohort study. Eur J Trauma Emerg Surg. 2017;44:621–6.PubMed Ahmed Ali M, et al. Interleukin-17 as a predictor of sepsis in polytrauma patients: a prospective cohort study. Eur J Trauma Emerg Surg. 2017;44:621–6.PubMed
45.
Zurück zum Zitat Gentile LF, et al. Persistent inflammation and immunosuppression: a common syndrome and new horizon for surgical intensive care. J Trauma Acute Care Surg. 2012;72(6):1491–501.PubMedPubMedCentral Gentile LF, et al. Persistent inflammation and immunosuppression: a common syndrome and new horizon for surgical intensive care. J Trauma Acute Care Surg. 2012;72(6):1491–501.PubMedPubMedCentral
46.
Zurück zum Zitat Hauser CJ, Otterbein LE. Danger signals from mitochondrial DAMPS in trauma and post-injury sepsis. Eur J Trauma Emerg Surg. 2018;44(3):317–24.PubMed Hauser CJ, Otterbein LE. Danger signals from mitochondrial DAMPS in trauma and post-injury sepsis. Eur J Trauma Emerg Surg. 2018;44(3):317–24.PubMed
48.
Zurück zum Zitat Karasu E, et al. Targeting complement pathways in polytrauma- and sepsis-induced multiple-organ dysfunction. Front Immunol. 2019;10:543.PubMedPubMedCentral Karasu E, et al. Targeting complement pathways in polytrauma- and sepsis-induced multiple-organ dysfunction. Front Immunol. 2019;10:543.PubMedPubMedCentral
49.
Zurück zum Zitat Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol. 2007;81(1):1–5.PubMed Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol. 2007;81(1):1–5.PubMed
50.
Zurück zum Zitat Manson J, Thiemermann C, Brohi K. Trauma alarmins as activators of damage-induced inflammation. Br J Surg. 2012;99(Suppl 1):12–20.PubMed Manson J, Thiemermann C, Brohi K. Trauma alarmins as activators of damage-induced inflammation. Br J Surg. 2012;99(Suppl 1):12–20.PubMed
51.
Zurück zum Zitat Zedler S, Faist E. The impact of endogenous triggers on trauma-associated inflammation. Curr Opin Crit Care. 2006;12(6):595–601.PubMed Zedler S, Faist E. The impact of endogenous triggers on trauma-associated inflammation. Curr Opin Crit Care. 2006;12(6):595–601.PubMed
52.
Zurück zum Zitat Hosac AM. Drotrecogin alfa (activated): the first FDA-approved treatment for severe sepsis. Proc (Bayl Univ Med Cent). 2002;15(2):224–7. Hosac AM. Drotrecogin alfa (activated): the first FDA-approved treatment for severe sepsis. Proc (Bayl Univ Med Cent). 2002;15(2):224–7.
53.
Zurück zum Zitat Kumar V. Immunometabolism: another road to sepsis and its therapeutic targeting. Inflammation. 2018;42(3):765–88. Kumar V. Immunometabolism: another road to sepsis and its therapeutic targeting. Inflammation. 2018;42(3):765–88.
55.
Zurück zum Zitat Russell JA. Management of sepsis. N Engl J Med. 2006;355(16):1699–713.PubMed Russell JA. Management of sepsis. N Engl J Med. 2006;355(16):1699–713.PubMed
56.
Zurück zum Zitat Giuliani A, Pirri G, Rinaldi AC. Antimicrobial peptides: the LPS connection. Methods Mol Biol. 2010;618:137–54.PubMed Giuliani A, Pirri G, Rinaldi AC. Antimicrobial peptides: the LPS connection. Methods Mol Biol. 2010;618:137–54.PubMed
57.
Zurück zum Zitat Asehnoune K, Roquilly A, Abraham E. Innate immune dysfunction in trauma patients: from pathophysiology to treatment. Anesthesiology. 2012;117(2):411–6.PubMed Asehnoune K, Roquilly A, Abraham E. Innate immune dysfunction in trauma patients: from pathophysiology to treatment. Anesthesiology. 2012;117(2):411–6.PubMed
58.
Zurück zum Zitat Roquilly A, et al. Hydrocortisone therapy for patients with multiple trauma: the randomized controlled HYPOLYTE study. JAMA. 2011;305(12):1201–9.PubMed Roquilly A, et al. Hydrocortisone therapy for patients with multiple trauma: the randomized controlled HYPOLYTE study. JAMA. 2011;305(12):1201–9.PubMed
59.
Zurück zum Zitat Ward NS, Casserly B, Ayala A. The compensatory anti-inflammatory response syndrome (CARS) in critically ill patients. Clin Chest Med. 2008;29(4):617–25.PubMedPubMedCentral Ward NS, Casserly B, Ayala A. The compensatory anti-inflammatory response syndrome (CARS) in critically ill patients. Clin Chest Med. 2008;29(4):617–25.PubMedPubMedCentral
60.
Zurück zum Zitat Islam MN, Bradley BA, Ceredig R. Sterile post-traumatic immunosuppression. Clin Transl Immunol. 2016;5(4):e77. Islam MN, Bradley BA, Ceredig R. Sterile post-traumatic immunosuppression. Clin Transl Immunol. 2016;5(4):e77.
61.
Zurück zum Zitat Bandyopadhyay G, et al. Negative signaling contributes to T-cell anergy in trauma patients. Crit Care Med. 2007;35(3):794–801.PubMed Bandyopadhyay G, et al. Negative signaling contributes to T-cell anergy in trauma patients. Crit Care Med. 2007;35(3):794–801.PubMed
62.
Zurück zum Zitat Hensler T, et al. Distinct mechanisms of immunosuppression as a consequence of major surgery. Infect Immun. 1997;65(6):2283–91.PubMedPubMedCentral Hensler T, et al. Distinct mechanisms of immunosuppression as a consequence of major surgery. Infect Immun. 1997;65(6):2283–91.PubMedPubMedCentral
63.
Zurück zum Zitat Hamers L, Kox M, Pickkers P. Sepsis-induced immunoparalysis: mechanisms, markers, and treatment options. Minerva Anestesiol. 2015;81(4):426–39.PubMed Hamers L, Kox M, Pickkers P. Sepsis-induced immunoparalysis: mechanisms, markers, and treatment options. Minerva Anestesiol. 2015;81(4):426–39.PubMed
64.
Zurück zum Zitat Serve R, et al. Comparative analysis of the regulatory T cells dynamics in peripheral blood in Human and porcine polytrauma. Front Immunol. 2018;9:435.PubMedPubMedCentral Serve R, et al. Comparative analysis of the regulatory T cells dynamics in peripheral blood in Human and porcine polytrauma. Front Immunol. 2018;9:435.PubMedPubMedCentral
65.
Zurück zum Zitat Orr SK, et al. Gene expression of proresolving lipid mediator pathways is associated with clinical outcomes in trauma patients. Crit Care Med. 2015;43(12):2642–50.PubMedPubMedCentral Orr SK, et al. Gene expression of proresolving lipid mediator pathways is associated with clinical outcomes in trauma patients. Crit Care Med. 2015;43(12):2642–50.PubMedPubMedCentral
66.
Zurück zum Zitat Nicolete R, et al. Prostaglandin E(2)-loaded microspheres as strategy to inhibit phagocytosis and modulate inflammatory mediators release. Eur J Pharm Biopharm. 2008;70(3):784–90.PubMed Nicolete R, et al. Prostaglandin E(2)-loaded microspheres as strategy to inhibit phagocytosis and modulate inflammatory mediators release. Eur J Pharm Biopharm. 2008;70(3):784–90.PubMed
67.
Zurück zum Zitat Nemeth K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15(1):42–9.PubMed Nemeth K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15(1):42–9.PubMed
68.
Zurück zum Zitat Tsai WH, et al. Serial changes in plasma annexin A1 and cortisol levels in sepsis patients. Chin J Physiol. 2014;57(1):1–7.PubMed Tsai WH, et al. Serial changes in plasma annexin A1 and cortisol levels in sepsis patients. Chin J Physiol. 2014;57(1):1–7.PubMed
69.
Zurück zum Zitat Oppenheim JJ, Yang D. Alarmins: chemotactic activators of immune responses. Curr Opin Immunol. 2005;17(4):359–65.PubMed Oppenheim JJ, Yang D. Alarmins: chemotactic activators of immune responses. Curr Opin Immunol. 2005;17(4):359–65.PubMed
70.
Zurück zum Zitat Janeway CA Jr, Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216.PubMed Janeway CA Jr, Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216.PubMed
71.
Zurück zum Zitat Matzinger P. The danger model: a renewed sense of self. Science. 2002;296(5566):301–5.PubMed Matzinger P. The danger model: a renewed sense of self. Science. 2002;296(5566):301–5.PubMed
72.
Zurück zum Zitat Lotze MT, et al. The grateful dead: damage-associated molecular pattern molecules and reduction/oxidation regulate immunity. Immunol Rev. 2007;220:60–81.PubMed Lotze MT, et al. The grateful dead: damage-associated molecular pattern molecules and reduction/oxidation regulate immunity. Immunol Rev. 2007;220:60–81.PubMed
73.
74.
Zurück zum Zitat Bortolotti P, Faure E, Kipnis E. Inflammasomes in tissue damages and immune disorders after trauma. Front Immunol. 2018;9:1900.PubMedPubMedCentral Bortolotti P, Faure E, Kipnis E. Inflammasomes in tissue damages and immune disorders after trauma. Front Immunol. 2018;9:1900.PubMedPubMedCentral
75.
Zurück zum Zitat Guisasola MC, et al. Early inflammatory response in polytraumatized patients: cytokines and heat shock proteins. A pilot study. Orthop Traumatol Surg Res. 2015;101(5):607–11.PubMed Guisasola MC, et al. Early inflammatory response in polytraumatized patients: cytokines and heat shock proteins. A pilot study. Orthop Traumatol Surg Res. 2015;101(5):607–11.PubMed
76.
Zurück zum Zitat Liaw PC, et al. DAMP and DIC: the role of extracellular DNA and DNA-binding proteins in the pathogenesis of DIC. Blood Rev. 2016;30(4):257–61.PubMed Liaw PC, et al. DAMP and DIC: the role of extracellular DNA and DNA-binding proteins in the pathogenesis of DIC. Blood Rev. 2016;30(4):257–61.PubMed
77.
Zurück zum Zitat Brinkmann V, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532–5.PubMed Brinkmann V, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532–5.PubMed
78.
Zurück zum Zitat Marin-Esteban V, et al. Afa/Dr diffusely adhering Escherichia coli strain C1845 induces neutrophil extracellular traps that kill bacteria and damage human enterocyte-like cells. Infect Immun. 2012;80(5):1891–9.PubMedPubMedCentral Marin-Esteban V, et al. Afa/Dr diffusely adhering Escherichia coli strain C1845 induces neutrophil extracellular traps that kill bacteria and damage human enterocyte-like cells. Infect Immun. 2012;80(5):1891–9.PubMedPubMedCentral
79.
Zurück zum Zitat Choi JJ, Reich CF 3rd, Pisetsky DS. The role of macrophages in the in vitro generation of extracellular DNA from apoptotic and necrotic cells. Immunology. 2005;115(1):55–62.PubMedPubMedCentral Choi JJ, Reich CF 3rd, Pisetsky DS. The role of macrophages in the in vitro generation of extracellular DNA from apoptotic and necrotic cells. Immunology. 2005;115(1):55–62.PubMedPubMedCentral
80.
Zurück zum Zitat Yousefi S, et al. Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense. Nat Med. 2008;14(9):949–53.PubMed Yousefi S, et al. Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense. Nat Med. 2008;14(9):949–53.PubMed
81.
Zurück zum Zitat Diehl F, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14(9):985–90.PubMed Diehl F, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14(9):985–90.PubMed
82.
Zurück zum Zitat Moscoso M, Garcia E, Lopez R. Biofilm formation by Streptococcus pneumoniae: role of choline, extracellular DNA, and capsular polysaccharide in microbial accretion. J Bacteriol. 2006;188(22):7785–95.PubMedPubMedCentral Moscoso M, Garcia E, Lopez R. Biofilm formation by Streptococcus pneumoniae: role of choline, extracellular DNA, and capsular polysaccharide in microbial accretion. J Bacteriol. 2006;188(22):7785–95.PubMedPubMedCentral
83.
Zurück zum Zitat Gogenur M, Burcharth J, Gogenur I. The role of total cell-free DNA in predicting outcomes among trauma patients in the intensive care unit: a systematic review. Crit Care. 2017;21(1):14.PubMedPubMedCentral Gogenur M, Burcharth J, Gogenur I. The role of total cell-free DNA in predicting outcomes among trauma patients in the intensive care unit: a systematic review. Crit Care. 2017;21(1):14.PubMedPubMedCentral
84.
Zurück zum Zitat Ahmed AI, Soliman RA, Samir S. Cell free DNA and procalcitonin as early markers of complications in ICU patients with multiple trauma and major surgery. Clin Lab. 2016;62(12):2395–404.PubMed Ahmed AI, Soliman RA, Samir S. Cell free DNA and procalcitonin as early markers of complications in ICU patients with multiple trauma and major surgery. Clin Lab. 2016;62(12):2395–404.PubMed
86.
Zurück zum Zitat Vestweber D. How leukocytes cross the vascular endothelium. Nat Rev Immunol. 2015;15(11):692–704.PubMed Vestweber D. How leukocytes cross the vascular endothelium. Nat Rev Immunol. 2015;15(11):692–704.PubMed
87.
Zurück zum Zitat Sun S, et al. Mitochondrial DAMPs increase endothelial permeability through neutrophil dependent and independent pathways. PLoS ONE. 2013;8(3):e59989.PubMedPubMedCentral Sun S, et al. Mitochondrial DAMPs increase endothelial permeability through neutrophil dependent and independent pathways. PLoS ONE. 2013;8(3):e59989.PubMedPubMedCentral
88.
Zurück zum Zitat Eppensteiner J, et al. Immunothrombotic activity of damage-associated molecular patterns and extracellular vesicles in secondary organ failure induced by trauma and sterile insults. Front Immunol. 2018;9:190.PubMedPubMedCentral Eppensteiner J, et al. Immunothrombotic activity of damage-associated molecular patterns and extracellular vesicles in secondary organ failure induced by trauma and sterile insults. Front Immunol. 2018;9:190.PubMedPubMedCentral
89.
Zurück zum Zitat Ahmad A, Druzhyna N, Szabo C. Effect of 3-mercaptopyruvate sulfurtransferase deficiency on the development of multiorgan failure, inflammation, and wound healing in mice subjected to burn injury. J Burn Care Res. 2019;40(2):148–56.PubMed Ahmad A, Druzhyna N, Szabo C. Effect of 3-mercaptopyruvate sulfurtransferase deficiency on the development of multiorgan failure, inflammation, and wound healing in mice subjected to burn injury. J Burn Care Res. 2019;40(2):148–56.PubMed
90.
Zurück zum Zitat Bianchi ME, Agresti A. HMG proteins: dynamic players in gene regulation and differentiation. Curr Opin Genet Dev. 2005;15(5):496–506.PubMed Bianchi ME, Agresti A. HMG proteins: dynamic players in gene regulation and differentiation. Curr Opin Genet Dev. 2005;15(5):496–506.PubMed
91.
Zurück zum Zitat Pandolfi F, et al. Key role of DAMP in inflammation, cancer, and tissue repair. Clin Ther. 2016;38(5):1017–28.PubMed Pandolfi F, et al. Key role of DAMP in inflammation, cancer, and tissue repair. Clin Ther. 2016;38(5):1017–28.PubMed
92.
Zurück zum Zitat Belgrano FS, et al. Role of the acidic tail of high mobility group protein B1 (HMGB1) in protein stability and DNA bending. PLoS ONE. 2013;8(11):e79572.PubMedPubMedCentral Belgrano FS, et al. Role of the acidic tail of high mobility group protein B1 (HMGB1) in protein stability and DNA bending. PLoS ONE. 2013;8(11):e79572.PubMedPubMedCentral
94.
Zurück zum Zitat Bellussi LM, et al. Are HMGB1 protein expression and secretion markers of upper airways inflammatory diseases? J Biol Regul Homeost Agents. 2013;27(3):791–804.PubMed Bellussi LM, et al. Are HMGB1 protein expression and secretion markers of upper airways inflammatory diseases? J Biol Regul Homeost Agents. 2013;27(3):791–804.PubMed
95.
Zurück zum Zitat Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418(6894):191–5.PubMed Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418(6894):191–5.PubMed
96.
Zurück zum Zitat Andersson U, Yang H, Harris H. High-mobility group box 1 protein (HMGB1) operates as an alarmin outside as well as inside cells. Semin Immunol. 2018;38:40–8.PubMed Andersson U, Yang H, Harris H. High-mobility group box 1 protein (HMGB1) operates as an alarmin outside as well as inside cells. Semin Immunol. 2018;38:40–8.PubMed
97.
Zurück zum Zitat Yang H, et al. A critical cysteine is required for HMGB1 binding to Toll-like receptor 4 and activation of macrophage cytokine release. Proc Natl Acad Sci U S A. 2010;107(26):11942–7.PubMedPubMedCentral Yang H, et al. A critical cysteine is required for HMGB1 binding to Toll-like receptor 4 and activation of macrophage cytokine release. Proc Natl Acad Sci U S A. 2010;107(26):11942–7.PubMedPubMedCentral
98.
Zurück zum Zitat Yu L, Wang L, Chen S. Endogenous toll-like receptor ligands and their biological significance. J Cell Mol Med. 2010;14(11):2592–603.PubMedPubMedCentral Yu L, Wang L, Chen S. Endogenous toll-like receptor ligands and their biological significance. J Cell Mol Med. 2010;14(11):2592–603.PubMedPubMedCentral
99.
100.
Zurück zum Zitat Yang H, et al. Redox modification of cysteine residues regulates the cytokine activity of high mobility group box-1 (HMGB1). Mol Med. 2012;18:250–9.PubMed Yang H, et al. Redox modification of cysteine residues regulates the cytokine activity of high mobility group box-1 (HMGB1). Mol Med. 2012;18:250–9.PubMed
101.
Zurück zum Zitat Venereau E, et al. Mutually exclusive redox forms of HMGB1 promote cell recruitment or proinflammatory cytokine release. J Exp Med. 2012;209(9):1519–28.PubMedPubMedCentral Venereau E, et al. Mutually exclusive redox forms of HMGB1 promote cell recruitment or proinflammatory cytokine release. J Exp Med. 2012;209(9):1519–28.PubMedPubMedCentral
102.
Zurück zum Zitat Schulman IG, et al. Macronuclei and micronuclei in Tetrahymena thermophila contain high-mobility-group-like chromosomal proteins containing a highly conserved eleven-amino-acid putative DNA-binding sequence. Mol Cell Biol. 1991;11(1):166–74.PubMedPubMedCentral Schulman IG, et al. Macronuclei and micronuclei in Tetrahymena thermophila contain high-mobility-group-like chromosomal proteins containing a highly conserved eleven-amino-acid putative DNA-binding sequence. Mol Cell Biol. 1991;11(1):166–74.PubMedPubMedCentral
103.
Zurück zum Zitat Deng M, et al. Location is the key to function: HMGB1 in sepsis and trauma-induced inflammation. J Leukoc Biol. 2019;106(1):161–9.PubMed Deng M, et al. Location is the key to function: HMGB1 in sepsis and trauma-induced inflammation. J Leukoc Biol. 2019;106(1):161–9.PubMed
104.
Zurück zum Zitat Cohen MJ, et al. Early release of high mobility group box nuclear protein 1 after severe trauma in humans: role of injury severity and tissue hypoperfusion. Crit Care. 2009;13(6):R174.PubMedPubMedCentral Cohen MJ, et al. Early release of high mobility group box nuclear protein 1 after severe trauma in humans: role of injury severity and tissue hypoperfusion. Crit Care. 2009;13(6):R174.PubMedPubMedCentral
105.
Zurück zum Zitat Deng M, et al. The endotoxin delivery protein HMGB1 mediates caspase-11-dependent lethality in sepsis. Immunity. 2018;49(4):740–53.PubMedPubMedCentral Deng M, et al. The endotoxin delivery protein HMGB1 mediates caspase-11-dependent lethality in sepsis. Immunity. 2018;49(4):740–53.PubMedPubMedCentral
106.
Zurück zum Zitat Levy RM, et al. Systemic inflammation and remote organ injury following trauma require HMGB1. Am J Physiol Regul Integr Comp Physiol. 2007;293(4):R1538–44.PubMed Levy RM, et al. Systemic inflammation and remote organ injury following trauma require HMGB1. Am J Physiol Regul Integr Comp Physiol. 2007;293(4):R1538–44.PubMed
107.
Zurück zum Zitat Zhang BF, et al. Anti-high mobility group box-1 (HMGB1) antibody attenuates kidney damage following experimental crush injury and the possible role of the tumor necrosis factor-alpha and c-Jun N-terminal kinase pathway. J Orthop Surg Res. 2017;12(1):110.PubMedPubMedCentral Zhang BF, et al. Anti-high mobility group box-1 (HMGB1) antibody attenuates kidney damage following experimental crush injury and the possible role of the tumor necrosis factor-alpha and c-Jun N-terminal kinase pathway. J Orthop Surg Res. 2017;12(1):110.PubMedPubMedCentral
108.
Zurück zum Zitat Parker TM, et al. The danger zone: systematic review of the role of HMGB1 danger signalling in traumatic brain injury. Brain Inj. 2017;31(1):2–8.PubMed Parker TM, et al. The danger zone: systematic review of the role of HMGB1 danger signalling in traumatic brain injury. Brain Inj. 2017;31(1):2–8.PubMed
109.
Zurück zum Zitat Tian X, et al. HMGB1 promotes neurovascular remodeling via Rage in the late phase of subarachnoid hemorrhage. Brain Res. 2017;1670:135–45.PubMed Tian X, et al. HMGB1 promotes neurovascular remodeling via Rage in the late phase of subarachnoid hemorrhage. Brain Res. 2017;1670:135–45.PubMed
110.
Zurück zum Zitat Wang KC, et al. Cerebrospinal fluid high mobility group box 1 is associated with neuronal death in subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2017;37(2):435–43.PubMed Wang KC, et al. Cerebrospinal fluid high mobility group box 1 is associated with neuronal death in subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2017;37(2):435–43.PubMed
111.
Zurück zum Zitat Kim JY, et al. HMGB1 contributes to the development of acute lung injury after hemorrhage. Am J Physiol Lung Cell Mol Physiol. 2005;288(5):L958–65.PubMed Kim JY, et al. HMGB1 contributes to the development of acute lung injury after hemorrhage. Am J Physiol Lung Cell Mol Physiol. 2005;288(5):L958–65.PubMed
112.
Zurück zum Zitat Ogawa EN, et al. Contribution of high-mobility group box-1 to the development of ventilator-induced lung injury. Am J Respir Crit Care Med. 2006;174(4):400–7.PubMed Ogawa EN, et al. Contribution of high-mobility group box-1 to the development of ventilator-induced lung injury. Am J Respir Crit Care Med. 2006;174(4):400–7.PubMed
113.
Zurück zum Zitat Tsung A, et al. The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J Exp Med. 2005;201(7):1135–43.PubMedPubMedCentral Tsung A, et al. The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J Exp Med. 2005;201(7):1135–43.PubMedPubMedCentral
114.
Zurück zum Zitat van Zoelen MA, et al. Pulmonary levels of high-mobility group box 1 during mechanical ventilation and ventilator-associated pneumonia. Shock. 2008;29(4):441–5.PubMed van Zoelen MA, et al. Pulmonary levels of high-mobility group box 1 during mechanical ventilation and ventilator-associated pneumonia. Shock. 2008;29(4):441–5.PubMed
115.
Zurück zum Zitat Ueno H, et al. Contributions of high mobility group box protein in experimental and clinical acute lung injury. Am J Respir Crit Care Med. 2004;170(12):1310–6.PubMed Ueno H, et al. Contributions of high mobility group box protein in experimental and clinical acute lung injury. Am J Respir Crit Care Med. 2004;170(12):1310–6.PubMed
116.
Zurück zum Zitat Abraham E, et al. HMG-1 as a mediator of acute lung inflammation. J Immunol. 2000;165(6):2950–4.PubMed Abraham E, et al. HMG-1 as a mediator of acute lung inflammation. J Immunol. 2000;165(6):2950–4.PubMed
117.
Zurück zum Zitat Pellegrini L, et al. HMGB1 and repair: focus on the heart. Pharmacol Ther. 2019;196:160–82.PubMed Pellegrini L, et al. HMGB1 and repair: focus on the heart. Pharmacol Ther. 2019;196:160–82.PubMed
118.
Zurück zum Zitat Vogel S, et al. Platelet-derived HMGB1 is a critical mediator of thrombosis. J Clin Invest. 2015;125(12):4638–54.PubMedPubMedCentral Vogel S, et al. Platelet-derived HMGB1 is a critical mediator of thrombosis. J Clin Invest. 2015;125(12):4638–54.PubMedPubMedCentral
119.
Zurück zum Zitat Ahrens I, et al. HMGB1 binds to activated platelets via the receptor for advanced glycation end products and is present in platelet rich human coronary artery thrombi. Thromb Haemost. 2015;114(5):994–1003.PubMed Ahrens I, et al. HMGB1 binds to activated platelets via the receptor for advanced glycation end products and is present in platelet rich human coronary artery thrombi. Thromb Haemost. 2015;114(5):994–1003.PubMed
120.
Zurück zum Zitat Arshad MI, Piquet-Pellorce C, Samson M. IL-33 and HMGB1 alarmins: sensors of cellular death and their involvement in liver pathology. Liver Int. 2012;32(8):1200–10.PubMed Arshad MI, Piquet-Pellorce C, Samson M. IL-33 and HMGB1 alarmins: sensors of cellular death and their involvement in liver pathology. Liver Int. 2012;32(8):1200–10.PubMed
121.
Zurück zum Zitat Yang H, Tracey KJ. Targeting HMGB1 in inflammation. Biochim Biophys Acta. 2010;1799(1–2):149–56.PubMed Yang H, Tracey KJ. Targeting HMGB1 in inflammation. Biochim Biophys Acta. 2010;1799(1–2):149–56.PubMed
122.
Zurück zum Zitat Tang D, et al. High-mobility group box 1, oxidative stress, and disease. Antioxid Redox Signal. 2011;14(7):1315–35.PubMedPubMedCentral Tang D, et al. High-mobility group box 1, oxidative stress, and disease. Antioxid Redox Signal. 2011;14(7):1315–35.PubMedPubMedCentral
123.
Zurück zum Zitat Keyel PA. How is inflammation initiated? Individual influences of IL-1, IL-18 and HMGB1. Cytokine. 2014;69(1):136–45.PubMed Keyel PA. How is inflammation initiated? Individual influences of IL-1, IL-18 and HMGB1. Cytokine. 2014;69(1):136–45.PubMed
124.
Zurück zum Zitat van Griensven M. Cytokines as biomarkers in polytraumatized patients. Unfallchirurg. 2014;117(8):699–702.PubMed van Griensven M. Cytokines as biomarkers in polytraumatized patients. Unfallchirurg. 2014;117(8):699–702.PubMed
125.
Zurück zum Zitat Dinarello CA, Goldin NP, Wolff SM. Demonstration and characterization of two distinct human leukocytic pyrogens. J Exp Med. 1974;139(6):1369–81.PubMedPubMedCentral Dinarello CA, Goldin NP, Wolff SM. Demonstration and characterization of two distinct human leukocytic pyrogens. J Exp Med. 1974;139(6):1369–81.PubMedPubMedCentral
126.
Zurück zum Zitat Dinarello CA. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood. 2011;117(14):3720–32.PubMedPubMedCentral Dinarello CA. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood. 2011;117(14):3720–32.PubMedPubMedCentral
127.
Zurück zum Zitat Dinarello CA. A clinical perspective of IL-1beta as the gatekeeper of inflammation. Eur J Immunol. 2011;41(5):1203–17.PubMed Dinarello CA. A clinical perspective of IL-1beta as the gatekeeper of inflammation. Eur J Immunol. 2011;41(5):1203–17.PubMed
128.
Zurück zum Zitat Aden N, et al. Epithelial cells promote fibroblast activation via IL-1alpha in systemic sclerosis. J Invest Dermatol. 2010;130(9):2191–200.PubMed Aden N, et al. Epithelial cells promote fibroblast activation via IL-1alpha in systemic sclerosis. J Invest Dermatol. 2010;130(9):2191–200.PubMed
129.
Zurück zum Zitat Rosenwasser LJ, Dinarello CA, Rosenthal AS. Adherent cell function in murine T-lymphocyte antigen recognition. IV. Enhancement of murine T-cell antigen recognition by human leukocytic pyrogen. J Exp Med. 1979;150(3):709–14.PubMed Rosenwasser LJ, Dinarello CA, Rosenthal AS. Adherent cell function in murine T-lymphocyte antigen recognition. IV. Enhancement of murine T-cell antigen recognition by human leukocytic pyrogen. J Exp Med. 1979;150(3):709–14.PubMed
130.
Zurück zum Zitat Bertheloot D, Latz E. HMGB1, IL-1alpha, IL-33 and S100 proteins: dual-function alarmins. Cell Mol Immunol. 2017;14(1):43–64.PubMed Bertheloot D, Latz E. HMGB1, IL-1alpha, IL-33 and S100 proteins: dual-function alarmins. Cell Mol Immunol. 2017;14(1):43–64.PubMed
131.
Zurück zum Zitat Kobayashi Y, et al. Identification of calcium-activated neutral protease as a processing enzyme of human interleukin 1 alpha. Proc Natl Acad Sci U S A. 1990;87(14):5548–52.PubMedPubMedCentral Kobayashi Y, et al. Identification of calcium-activated neutral protease as a processing enzyme of human interleukin 1 alpha. Proc Natl Acad Sci U S A. 1990;87(14):5548–52.PubMedPubMedCentral
132.
Zurück zum Zitat Kavita U, Mizel SB. Differential sensitivity of interleukin-1 alpha and -beta precursor proteins to cleavage by calpain, a calcium-dependent protease. J Biol Chem. 1995;270(46):27758–65.PubMed Kavita U, Mizel SB. Differential sensitivity of interleukin-1 alpha and -beta precursor proteins to cleavage by calpain, a calcium-dependent protease. J Biol Chem. 1995;270(46):27758–65.PubMed
133.
Zurück zum Zitat Carruth LM, Demczuk S, Mizel SB. Involvement of a calpain-like protease in the processing of the murine interleukin 1 alpha precursor. J Biol Chem. 1991;266(19):12162–7.PubMed Carruth LM, Demczuk S, Mizel SB. Involvement of a calpain-like protease in the processing of the murine interleukin 1 alpha precursor. J Biol Chem. 1991;266(19):12162–7.PubMed
134.
Zurück zum Zitat Weber A, Wasiliew P, Kracht M. Interleukin-1 (IL-1) pathway. Sci Signal. 2010;3(105):cm1.PubMed Weber A, Wasiliew P, Kracht M. Interleukin-1 (IL-1) pathway. Sci Signal. 2010;3(105):cm1.PubMed
135.
Zurück zum Zitat Lefrancais E, Cayrol C. Mechanisms of IL-33 processing and secretion: differences and similarities between IL-1 family members. Eur Cytokine Netw. 2012;23(4):120–7.PubMed Lefrancais E, Cayrol C. Mechanisms of IL-33 processing and secretion: differences and similarities between IL-1 family members. Eur Cytokine Netw. 2012;23(4):120–7.PubMed
136.
Zurück zum Zitat Maier JA, Statuto M, Ragnotti G. Endogenous interleukin 1 alpha must be transported to the nucleus to exert its activity in human endothelial cells. Mol Cell Biol. 1994;14(3):1845–51.PubMedPubMedCentral Maier JA, Statuto M, Ragnotti G. Endogenous interleukin 1 alpha must be transported to the nucleus to exert its activity in human endothelial cells. Mol Cell Biol. 1994;14(3):1845–51.PubMedPubMedCentral
137.
Zurück zum Zitat Werman A, et al. The precursor form of IL-1alpha is an intracrine proinflammatory activator of transcription. Proc Natl Acad Sci U S A. 2004;101(8):2434–9.PubMedPubMedCentral Werman A, et al. The precursor form of IL-1alpha is an intracrine proinflammatory activator of transcription. Proc Natl Acad Sci U S A. 2004;101(8):2434–9.PubMedPubMedCentral
138.
Zurück zum Zitat Sakurai T, et al. Hepatocyte necrosis induced by oxidative stress and IL-1 alpha release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis. Cancer Cell. 2008;14(2):156–65.PubMedPubMedCentral Sakurai T, et al. Hepatocyte necrosis induced by oxidative stress and IL-1 alpha release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis. Cancer Cell. 2008;14(2):156–65.PubMedPubMedCentral
139.
Zurück zum Zitat Carta S, Lavieri R, Rubartelli A. Different members of the IL-1 family come out in different ways: DAMPs vs. cytokines? Front Immunol. 2013;4:123.PubMedPubMedCentral Carta S, Lavieri R, Rubartelli A. Different members of the IL-1 family come out in different ways: DAMPs vs. cytokines? Front Immunol. 2013;4:123.PubMedPubMedCentral
140.
Zurück zum Zitat Ballak DB, et al. IL-1 family members in the pathogenesis and treatment of metabolic disease: focus on adipose tissue inflammation and insulin resistance. Cytokine. 2015;75(2):280–90.PubMedPubMedCentral Ballak DB, et al. IL-1 family members in the pathogenesis and treatment of metabolic disease: focus on adipose tissue inflammation and insulin resistance. Cytokine. 2015;75(2):280–90.PubMedPubMedCentral
141.
142.
Zurück zum Zitat Lugrin J, et al. Cutting edge: IL-1alpha is a crucial danger signal triggering acute myocardial inflammation during myocardial infarction. J Immunol. 2015;194(2):499–503.PubMed Lugrin J, et al. Cutting edge: IL-1alpha is a crucial danger signal triggering acute myocardial inflammation during myocardial infarction. J Immunol. 2015;194(2):499–503.PubMed
143.
Zurück zum Zitat Scarpa M, et al. The epithelial danger signal IL-1alpha is a potent activator of fibroblasts and reactivator of intestinal inflammation. Am J Pathol. 2015;185(6):1624–37.PubMedPubMedCentral Scarpa M, et al. The epithelial danger signal IL-1alpha is a potent activator of fibroblasts and reactivator of intestinal inflammation. Am J Pathol. 2015;185(6):1624–37.PubMedPubMedCentral
144.
Zurück zum Zitat Jackman RP, et al. Distinct roles of trauma and transfusion in induction of immune modulation after injury. Transfusion. 2012;52(12):2533–50.PubMedPubMedCentral Jackman RP, et al. Distinct roles of trauma and transfusion in induction of immune modulation after injury. Transfusion. 2012;52(12):2533–50.PubMedPubMedCentral
145.
Zurück zum Zitat Ehrnthaller C, et al. The molecular fingerprint of lung inflammation after blunt chest trauma. Eur J Med Res. 2015;20:70.PubMedPubMedCentral Ehrnthaller C, et al. The molecular fingerprint of lung inflammation after blunt chest trauma. Eur J Med Res. 2015;20:70.PubMedPubMedCentral
146.
Zurück zum Zitat Jacobs RF, et al. Elevated interleukin-1 release by human alveolar macrophages during the adult respiratory distress syndrome. Am Rev Respir Dis. 1989;140(6):1686–92.PubMed Jacobs RF, et al. Elevated interleukin-1 release by human alveolar macrophages during the adult respiratory distress syndrome. Am Rev Respir Dis. 1989;140(6):1686–92.PubMed
147.
Zurück zum Zitat Rider P, et al. The transcription of the alarmin cytokine interleukin-1 alpha is controlled by hypoxia inducible factors 1 and 2 alpha in hypoxic cells. Front Immunol. 2012;3:290.PubMedPubMedCentral Rider P, et al. The transcription of the alarmin cytokine interleukin-1 alpha is controlled by hypoxia inducible factors 1 and 2 alpha in hypoxic cells. Front Immunol. 2012;3:290.PubMedPubMedCentral
148.
Zurück zum Zitat Thornton P, et al. Platelet interleukin-1alpha drives cerebrovascular inflammation. Blood. 2010;115(17):3632–9.PubMed Thornton P, et al. Platelet interleukin-1alpha drives cerebrovascular inflammation. Blood. 2010;115(17):3632–9.PubMed
149.
Zurück zum Zitat Turner NA, et al. Interleukin-1alpha stimulates proinflammatory cytokine expression in human cardiac myofibroblasts. Am J Physiol Heart Circ Physiol. 2009;297(3):H1117–27.PubMed Turner NA, et al. Interleukin-1alpha stimulates proinflammatory cytokine expression in human cardiac myofibroblasts. Am J Physiol Heart Circ Physiol. 2009;297(3):H1117–27.PubMed
150.
Zurück zum Zitat Pichery M, et al. Endogenous IL-33 is highly expressed in mouse epithelial barrier tissues, lymphoid organs, brain, embryos, and inflamed tissues: in situ analysis using a novel Il-33-LacZ gene trap reporter strain. J Immunol (Baltimore, Md.: 1950). 2012;188(7):3488–95. Pichery M, et al. Endogenous IL-33 is highly expressed in mouse epithelial barrier tissues, lymphoid organs, brain, embryos, and inflamed tissues: in situ analysis using a novel Il-33-LacZ gene trap reporter strain. J Immunol (Baltimore, Md.: 1950). 2012;188(7):3488–95.
151.
Zurück zum Zitat Baekkevold ES, et al. Molecular characterization of NF-HEV, a nuclear factor preferentially expressed in human high endothelial venules. Am J Pathol. 2003;163(1):69–79.PubMedPubMedCentral Baekkevold ES, et al. Molecular characterization of NF-HEV, a nuclear factor preferentially expressed in human high endothelial venules. Am J Pathol. 2003;163(1):69–79.PubMedPubMedCentral
152.
Zurück zum Zitat Liew FY, Girard JP, Turnquist HR. Interleukin-33 in health and disease. Nat Rev Immunol. 2016;16(11):676–89.PubMed Liew FY, Girard JP, Turnquist HR. Interleukin-33 in health and disease. Nat Rev Immunol. 2016;16(11):676–89.PubMed
153.
Zurück zum Zitat Carriere V, et al. IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc Natl Acad Sci USA. 2007;104(1):282–7.PubMed Carriere V, et al. IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc Natl Acad Sci USA. 2007;104(1):282–7.PubMed
154.
Zurück zum Zitat Hirsiger S, et al. Danger signals activating the immune response after trauma. Mediat Inflamm. 2012;2012:315941. Hirsiger S, et al. Danger signals activating the immune response after trauma. Mediat Inflamm. 2012;2012:315941.
155.
Zurück zum Zitat Schmitz J, et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity. 2005;23(5):479–90.PubMed Schmitz J, et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity. 2005;23(5):479–90.PubMed
156.
Zurück zum Zitat Palmer G, et al. The IL-1 receptor accessory protein (AcP) is required for IL-33 signaling and soluble AcP enhances the ability of soluble ST2 to inhibit IL-33. Cytokine. 2008;42(3):358–64.PubMed Palmer G, et al. The IL-1 receptor accessory protein (AcP) is required for IL-33 signaling and soluble AcP enhances the ability of soluble ST2 to inhibit IL-33. Cytokine. 2008;42(3):358–64.PubMed
157.
158.
Zurück zum Zitat Molofsky AB, Savage AK, Locksley RM. Interleukin-33 in tissue homeostasis, injury, and inflammation. Immunity. 2015;42(6):1005–19.PubMedPubMedCentral Molofsky AB, Savage AK, Locksley RM. Interleukin-33 in tissue homeostasis, injury, and inflammation. Immunity. 2015;42(6):1005–19.PubMedPubMedCentral
159.
Zurück zum Zitat Lefrancais E, et al. IL-33 is processed into mature bioactive forms by neutrophil elastase and cathepsin G. Proc Natl Acad Sci U S A. 2012;109(5):1673–8.PubMedPubMedCentral Lefrancais E, et al. IL-33 is processed into mature bioactive forms by neutrophil elastase and cathepsin G. Proc Natl Acad Sci U S A. 2012;109(5):1673–8.PubMedPubMedCentral
160.
Zurück zum Zitat Roussel L, et al. Molecular mimicry between IL-33 and KSHV for attachment to chromatin through the H2A-H2B acidic pocket. EMBO Rep. 2008;9(10):1006–12.PubMedPubMedCentral Roussel L, et al. Molecular mimicry between IL-33 and KSHV for attachment to chromatin through the H2A-H2B acidic pocket. EMBO Rep. 2008;9(10):1006–12.PubMedPubMedCentral
161.
Zurück zum Zitat Bessa J, et al. Altered subcellular localization of IL-33 leads to non-resolving lethal inflammation. J Autoimmun. 2014;55:33–41.PubMed Bessa J, et al. Altered subcellular localization of IL-33 leads to non-resolving lethal inflammation. J Autoimmun. 2014;55:33–41.PubMed
162.
Zurück zum Zitat Cohen ES, et al. Oxidation of the alarmin IL-33 regulates ST2-dependent inflammation. Nat Commun. 2015;6:8327.PubMed Cohen ES, et al. Oxidation of the alarmin IL-33 regulates ST2-dependent inflammation. Nat Commun. 2015;6:8327.PubMed
163.
Zurück zum Zitat Cayrol C, Girard J-P. IL-33: an alarmin cytokine with crucial roles in innate immunity, inflammation and allergy. Curr Opin Immunol. 2014;31:31–7.PubMed Cayrol C, Girard J-P. IL-33: an alarmin cytokine with crucial roles in innate immunity, inflammation and allergy. Curr Opin Immunol. 2014;31:31–7.PubMed
164.
Zurück zum Zitat Ali S, et al. IL-1 receptor accessory protein is essential for IL-33-induced activation of T lymphocytes and mast cells. Proc Natl Acad Sci U S A. 2007;104(47):18660–5.PubMedPubMedCentral Ali S, et al. IL-1 receptor accessory protein is essential for IL-33-induced activation of T lymphocytes and mast cells. Proc Natl Acad Sci U S A. 2007;104(47):18660–5.PubMedPubMedCentral
165.
Zurück zum Zitat Kurowska-Stolarska M, et al. IL-33 induces antigen-specific IL-5+ T cells and promotes allergic-induced airway inflammation independent of IL-4. J Immunol. 2008;181(7):4780–90.PubMed Kurowska-Stolarska M, et al. IL-33 induces antigen-specific IL-5+ T cells and promotes allergic-induced airway inflammation independent of IL-4. J Immunol. 2008;181(7):4780–90.PubMed
166.
Zurück zum Zitat Jovanovic IP, et al. IL-33/ST2 axis in innate and acquired immunity to tumors. Oncoimmunology. 2012;1(2):229–31.PubMedPubMedCentral Jovanovic IP, et al. IL-33/ST2 axis in innate and acquired immunity to tumors. Oncoimmunology. 2012;1(2):229–31.PubMedPubMedCentral
168.
Zurück zum Zitat Humphreys NE, et al. IL-33, a potent inducer of adaptive immunity to intestinal nematodes. J Immunol. 2008;180(4):2443–9.PubMed Humphreys NE, et al. IL-33, a potent inducer of adaptive immunity to intestinal nematodes. J Immunol. 2008;180(4):2443–9.PubMed
169.
Zurück zum Zitat Alves-Filho JC, et al. Interleukin-33 attenuates sepsis by enhancing neutrophil influx to the site of infection. Nat Med. 2010;16(6):708–12.PubMed Alves-Filho JC, et al. Interleukin-33 attenuates sepsis by enhancing neutrophil influx to the site of infection. Nat Med. 2010;16(6):708–12.PubMed
170.
Zurück zum Zitat Kurowska-Stolarska M, et al. IL-33 amplifies the polarization of alternatively activated macrophages that contribute to airway inflammation. J Immunol. 2009;183(10):6469–77.PubMed Kurowska-Stolarska M, et al. IL-33 amplifies the polarization of alternatively activated macrophages that contribute to airway inflammation. J Immunol. 2009;183(10):6469–77.PubMed
171.
Zurück zum Zitat Sethi S, Murphy TF. Infection in the pathogenesis and course of chronic obstructive pulmonary disease. N Engl J Med. 2008;359(22):2355–65.PubMed Sethi S, Murphy TF. Infection in the pathogenesis and course of chronic obstructive pulmonary disease. N Engl J Med. 2008;359(22):2355–65.PubMed
172.
Zurück zum Zitat Palmer G, et al. Inhibition of interleukin-33 signaling attenuates the severity of experimental arthritis. Arthritis Rheum. 2009;60(3):738–49.PubMed Palmer G, et al. Inhibition of interleukin-33 signaling attenuates the severity of experimental arthritis. Arthritis Rheum. 2009;60(3):738–49.PubMed
173.
Zurück zum Zitat Gadani SP, et al. The glia-derived alarmin IL-33 orchestrates the immune response and promotes recovery following CNS injury. Neuron. 2015;85(4):703–9.PubMed Gadani SP, et al. The glia-derived alarmin IL-33 orchestrates the immune response and promotes recovery following CNS injury. Neuron. 2015;85(4):703–9.PubMed
174.
Zurück zum Zitat Wicher G, et al. Interleukin-33 promotes recruitment of microglia/macrophages in response to traumatic brain injury. J Neurotrauma. 2017;34(22):3173–82.PubMed Wicher G, et al. Interleukin-33 promotes recruitment of microglia/macrophages in response to traumatic brain injury. J Neurotrauma. 2017;34(22):3173–82.PubMed
175.
Zurück zum Zitat Pomeshchik Y, et al. Interleukin-33 treatment reduces secondary injury and improves functional recovery after contusion spinal cord injury. Brain Behav Immun. 2015;44:68–81.PubMed Pomeshchik Y, et al. Interleukin-33 treatment reduces secondary injury and improves functional recovery after contusion spinal cord injury. Brain Behav Immun. 2015;44:68–81.PubMed
176.
Zurück zum Zitat Foster SL, Talbot S, Woolf CJ. CNS injury: IL-33 sounds the alarm. Immunity. 2015;42(3):403–5.PubMed Foster SL, Talbot S, Woolf CJ. CNS injury: IL-33 sounds the alarm. Immunity. 2015;42(3):403–5.PubMed
177.
Zurück zum Zitat Foell D, et al. S100 proteins expressed in phagocytes: a novel group of damage-associated molecular pattern molecules. J Leukoc Biol. 2007;81(1):28–37.PubMed Foell D, et al. S100 proteins expressed in phagocytes: a novel group of damage-associated molecular pattern molecules. J Leukoc Biol. 2007;81(1):28–37.PubMed
179.
Zurück zum Zitat Oesterle A, Bowman MA. S100A12 and the S100/calgranulins: emerging biomarkers for atherosclerosis and possibly therapeutic targets. Arterioscler Thromb Vasc Biol. 2015;35(12):2496–507.PubMedPubMedCentral Oesterle A, Bowman MA. S100A12 and the S100/calgranulins: emerging biomarkers for atherosclerosis and possibly therapeutic targets. Arterioscler Thromb Vasc Biol. 2015;35(12):2496–507.PubMedPubMedCentral
180.
Zurück zum Zitat Dassan P, Keir G, Brown MM. Criteria for a clinically informative serum biomarker in acute ischaemic stroke: a review of S100B. Cerebrovasc Dis. 2009;27(3):295–302.PubMed Dassan P, Keir G, Brown MM. Criteria for a clinically informative serum biomarker in acute ischaemic stroke: a review of S100B. Cerebrovasc Dis. 2009;27(3):295–302.PubMed
182.
Zurück zum Zitat Gazzolo D, Michetti F. Perinatal S100B protein assessment in human unconventional biological fluids: a minireview and new perspectives. Cardiovasc Psychiatry Neurol. 2010;2010:703563.PubMedPubMedCentral Gazzolo D, Michetti F. Perinatal S100B protein assessment in human unconventional biological fluids: a minireview and new perspectives. Cardiovasc Psychiatry Neurol. 2010;2010:703563.PubMedPubMedCentral
183.
Zurück zum Zitat Mocellin S, Zavagno G, Nitti D. The prognostic value of serum S100B in patients with cutaneous melanoma: a meta-analysis. Int J Cancer. 2008;123(10):2370–6.PubMed Mocellin S, Zavagno G, Nitti D. The prognostic value of serum S100B in patients with cutaneous melanoma: a meta-analysis. Int J Cancer. 2008;123(10):2370–6.PubMed
184.
Zurück zum Zitat Ellis EF, et al. S100B protein is released from rat neonatal neurons, astrocytes, and microglia by in vitro trauma and anti-S100 increases trauma-induced delayed neuronal injury and negates the protective effect of exogenous S100B on neurons. J Neurochem. 2007;101(6):1463–70.PubMed Ellis EF, et al. S100B protein is released from rat neonatal neurons, astrocytes, and microglia by in vitro trauma and anti-S100 increases trauma-induced delayed neuronal injury and negates the protective effect of exogenous S100B on neurons. J Neurochem. 2007;101(6):1463–70.PubMed
185.
Zurück zum Zitat Pruenster M, et al. S100A8/A9: from basic science to clinical application. Pharmacol Ther. 2016;167:120–31.PubMed Pruenster M, et al. S100A8/A9: from basic science to clinical application. Pharmacol Ther. 2016;167:120–31.PubMed
186.
Zurück zum Zitat Ghavami S, et al. S100A8/A9 at low concentration promotes tumor cell growth via RAGE ligation and MAP kinase-dependent pathway. J Leukoc Biol. 2008;83(6):1484–92.PubMed Ghavami S, et al. S100A8/A9 at low concentration promotes tumor cell growth via RAGE ligation and MAP kinase-dependent pathway. J Leukoc Biol. 2008;83(6):1484–92.PubMed
187.
Zurück zum Zitat Ibrahim ZA, et al. RAGE and TLRs: relatives, friends or neighbours? Mol Immunol. 2013;56(4):739–44.PubMed Ibrahim ZA, et al. RAGE and TLRs: relatives, friends or neighbours? Mol Immunol. 2013;56(4):739–44.PubMed
188.
Zurück zum Zitat Loeser RF, et al. Articular chondrocytes express the receptor for advanced glycation end products: potential role in osteoarthritis. Arthritis Rheum. 2005;52(8):2376–85.PubMedPubMedCentral Loeser RF, et al. Articular chondrocytes express the receptor for advanced glycation end products: potential role in osteoarthritis. Arthritis Rheum. 2005;52(8):2376–85.PubMedPubMedCentral
189.
Zurück zum Zitat Rohde D, et al. S100A1 is released from ischemic cardiomyocytes and signals myocardial damage via Toll-like receptor 4. EMBO Mol Med. 2014;6(6):778–94.PubMedPubMedCentral Rohde D, et al. S100A1 is released from ischemic cardiomyocytes and signals myocardial damage via Toll-like receptor 4. EMBO Mol Med. 2014;6(6):778–94.PubMedPubMedCentral
190.
Zurück zum Zitat Payen D, et al. Gene profiling in human blood leucocytes during recovery from septic shock. Intensive Care Med. 2008;34(8):1371–6.PubMed Payen D, et al. Gene profiling in human blood leucocytes during recovery from septic shock. Intensive Care Med. 2008;34(8):1371–6.PubMed
191.
Zurück zum Zitat Zhang LN, et al. Diagnostic and predictive levels of calcium-binding protein A8 and tumor necrosis factor receptor-associated factor 6 in sepsis-associated encephalopathy: a prospective observational study. Chin Med J (Engl). 2016;129(14):1674–81. Zhang LN, et al. Diagnostic and predictive levels of calcium-binding protein A8 and tumor necrosis factor receptor-associated factor 6 in sepsis-associated encephalopathy: a prospective observational study. Chin Med J (Engl). 2016;129(14):1674–81.
192.
Zurück zum Zitat Uhel F, et al. Early expansion of circulating granulocytic myeloid-derived suppressor cells predicts development of nosocomial infections in patients with sepsis. Am J Respir Crit Care Med. 2017;196(3):315–27.PubMed Uhel F, et al. Early expansion of circulating granulocytic myeloid-derived suppressor cells predicts development of nosocomial infections in patients with sepsis. Am J Respir Crit Care Med. 2017;196(3):315–27.PubMed
193.
Zurück zum Zitat Fontaine M, et al. Innate danger signals in acute injury: from bench to bedside. Anaesth Crit Care Pain Med. 2016;35(4):283–92.PubMed Fontaine M, et al. Innate danger signals in acute injury: from bench to bedside. Anaesth Crit Care Pain Med. 2016;35(4):283–92.PubMed
194.
Zurück zum Zitat De Lorenzo BH, et al. Macrophage suppression following phagocytosis of apoptotic neutrophils is mediated by the S100A9 calcium-binding protein. Immunobiology. 2010;215(5):341–7.PubMed De Lorenzo BH, et al. Macrophage suppression following phagocytosis of apoptotic neutrophils is mediated by the S100A9 calcium-binding protein. Immunobiology. 2010;215(5):341–7.PubMed
195.
Zurück zum Zitat Azmitia EC. Cajal’s hypotheses on neurobiones and neurotropic factor match properties of microtubules and S-100 beta. Prog Brain Res. 2002;136:87–100.PubMed Azmitia EC. Cajal’s hypotheses on neurobiones and neurotropic factor match properties of microtubules and S-100 beta. Prog Brain Res. 2002;136:87–100.PubMed
196.
Zurück zum Zitat Van Eldik LJ, Wainwright MS. The Janus face of glial-derived S100B: beneficial and detrimental functions in the brain. Restor Neurol Neurosci. 2003;21(3–4):97–108.PubMed Van Eldik LJ, Wainwright MS. The Janus face of glial-derived S100B: beneficial and detrimental functions in the brain. Restor Neurol Neurosci. 2003;21(3–4):97–108.PubMed
197.
Zurück zum Zitat Thelin EP, et al. Assessing bicycle-related trauma using the biomarker S100B reveals a correlation with total injury severity. Eur J Trauma Emerg Surg. 2016;42(5):617–25.PubMed Thelin EP, et al. Assessing bicycle-related trauma using the biomarker S100B reveals a correlation with total injury severity. Eur J Trauma Emerg Surg. 2016;42(5):617–25.PubMed
198.
Zurück zum Zitat Adami C, et al. S100B expression in and effects on microglia. Glia. 2001;33(2):131–42.PubMed Adami C, et al. S100B expression in and effects on microglia. Glia. 2001;33(2):131–42.PubMed
199.
Zurück zum Zitat Goyal A, et al. S100b as a prognostic biomarker in outcome prediction for patients with severe traumatic brain injury. J Neurotrauma. 2013;30(11):946–57.PubMedPubMedCentral Goyal A, et al. S100b as a prognostic biomarker in outcome prediction for patients with severe traumatic brain injury. J Neurotrauma. 2013;30(11):946–57.PubMedPubMedCentral
200.
Zurück zum Zitat Kellermann I, et al. Early CSF and serum S100B concentrations for outcome prediction in traumatic brain injury and subarachnoid hemorrhage. Clin Neurol Neurosurg. 2016;145:79–83.PubMed Kellermann I, et al. Early CSF and serum S100B concentrations for outcome prediction in traumatic brain injury and subarachnoid hemorrhage. Clin Neurol Neurosurg. 2016;145:79–83.PubMed
202.
Zurück zum Zitat Holzinger D, Tenbrock K, Roth J. Alarmins of the S100-family in juvenile autoimmune and auto-inflammatory diseases. Front Immunol. 2019;10:182.PubMedPubMedCentral Holzinger D, Tenbrock K, Roth J. Alarmins of the S100-family in juvenile autoimmune and auto-inflammatory diseases. Front Immunol. 2019;10:182.PubMedPubMedCentral
203.
Zurück zum Zitat Golden N, et al. S100B serum level as a mortality predictor for traumatic brain injury: a meta-analysis. Open Access Maced J Med Sci. 2018;6(11):2239–44.PubMedPubMedCentral Golden N, et al. S100B serum level as a mortality predictor for traumatic brain injury: a meta-analysis. Open Access Maced J Med Sci. 2018;6(11):2239–44.PubMedPubMedCentral
204.
Zurück zum Zitat Chernov AV, et al. The calcium-binding proteins S100A8 and S100A9 initiate the early inflammatory program in injured peripheral nerves. J Biol Chem. 2015;290(18):11771–84.PubMedPubMedCentral Chernov AV, et al. The calcium-binding proteins S100A8 and S100A9 initiate the early inflammatory program in injured peripheral nerves. J Biol Chem. 2015;290(18):11771–84.PubMedPubMedCentral
205.
Zurück zum Zitat Austermann J, et al. Alarmins MRP8 and MRP14 induce stress tolerance in phagocytes under sterile inflammatory conditions. Cell Rep. 2014;9(6):2112–23.PubMed Austermann J, et al. Alarmins MRP8 and MRP14 induce stress tolerance in phagocytes under sterile inflammatory conditions. Cell Rep. 2014;9(6):2112–23.PubMed
206.
Zurück zum Zitat Dang X, et al. S100B ranks as a new marker of multiple traumas in patients and may accelerate its development by regulating endothelial cell dysfunction. Int J Clin Exp Pathol. 2014;7(7):3818–26.PubMedPubMedCentral Dang X, et al. S100B ranks as a new marker of multiple traumas in patients and may accelerate its development by regulating endothelial cell dysfunction. Int J Clin Exp Pathol. 2014;7(7):3818–26.PubMedPubMedCentral
207.
Zurück zum Zitat Anderson RE, et al. High serum S100B levels for trauma patients without head injuries. Neurosurgery. 2001;48(6):1255–8 (discussion 1258-60).PubMed Anderson RE, et al. High serum S100B levels for trauma patients without head injuries. Neurosurgery. 2001;48(6):1255–8 (discussion 1258-60).PubMed
208.
Zurück zum Zitat Pfortmueller CA, et al. S-100 B concentrations are a predictor of decreased survival in patients with major trauma, independently of head injury. PLoS ONE. 2016;11(3):e0152822.PubMedPubMedCentral Pfortmueller CA, et al. S-100 B concentrations are a predictor of decreased survival in patients with major trauma, independently of head injury. PLoS ONE. 2016;11(3):e0152822.PubMedPubMedCentral
209.
Zurück zum Zitat Greven J, et al. Update on the role of endothelial cells in trauma. Eur J Trauma Emerg Surg. 2017;44:667–77.PubMed Greven J, et al. Update on the role of endothelial cells in trauma. Eur J Trauma Emerg Surg. 2017;44:667–77.PubMed
210.
Zurück zum Zitat Wang J, et al. Injury-induced MRP8/MRP14 stimulates IP-10/CXCL10 in monocytes/macrophages. FASEB J. 2015;29(1):250–62.PubMed Wang J, et al. Injury-induced MRP8/MRP14 stimulates IP-10/CXCL10 in monocytes/macrophages. FASEB J. 2015;29(1):250–62.PubMed
211.
Zurück zum Zitat Kawai C, et al. Circulating extracellular histones are clinically relevant mediators of multiple organ injury. Am J Pathol. 2016;186(4):829–43.PubMed Kawai C, et al. Circulating extracellular histones are clinically relevant mediators of multiple organ injury. Am J Pathol. 2016;186(4):829–43.PubMed
212.
213.
Zurück zum Zitat Huang H, et al. Endogenous histones function as alarmins in sterile inflammatory liver injury through Toll-like receptor 9 in mice. Hepatology. 2011;54(3):999–1008.PubMed Huang H, et al. Endogenous histones function as alarmins in sterile inflammatory liver injury through Toll-like receptor 9 in mice. Hepatology. 2011;54(3):999–1008.PubMed
214.
Zurück zum Zitat Tang D, et al. PAMPs and DAMPs: signal 0 s that spur autophagy and immunity. Immunol Rev. 2012;249(1):158–75.PubMedPubMedCentral Tang D, et al. PAMPs and DAMPs: signal 0 s that spur autophagy and immunity. Immunol Rev. 2012;249(1):158–75.PubMedPubMedCentral
216.
Zurück zum Zitat Pisetsky DS. Immune activation by histones: plusses and minuses in inflammation. Eur J Immunol. 2013;43(12):3163–6.PubMed Pisetsky DS. Immune activation by histones: plusses and minuses in inflammation. Eur J Immunol. 2013;43(12):3163–6.PubMed
217.
218.
Zurück zum Zitat Toh CH, Alhamdi Y, Abrams ST. Current pathological and laboratory considerations in the diagnosis of disseminated intravascular coagulation. Ann Lab Med. 2016;36(6):505–12.PubMedPubMedCentral Toh CH, Alhamdi Y, Abrams ST. Current pathological and laboratory considerations in the diagnosis of disseminated intravascular coagulation. Ann Lab Med. 2016;36(6):505–12.PubMedPubMedCentral
219.
Zurück zum Zitat Alhamdi Y, et al. Circulating histones are major mediators of cardiac injury in patients with sepsis. Crit Care Med. 2015;43(10):2094–103.PubMed Alhamdi Y, et al. Circulating histones are major mediators of cardiac injury in patients with sepsis. Crit Care Med. 2015;43(10):2094–103.PubMed
220.
Zurück zum Zitat Allam R, et al. Histones from dying renal cells aggravate kidney injury via TLR2 and TLR4. J Am Soc Nephrol. 2012;23(8):1375–88.PubMedPubMedCentral Allam R, et al. Histones from dying renal cells aggravate kidney injury via TLR2 and TLR4. J Am Soc Nephrol. 2012;23(8):1375–88.PubMedPubMedCentral
221.
Zurück zum Zitat Semeraro F, et al. Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4. Blood. 2011;118(7):1952–61.PubMedPubMedCentral Semeraro F, et al. Extracellular histones promote thrombin generation through platelet-dependent mechanisms: involvement of platelet TLR2 and TLR4. Blood. 2011;118(7):1952–61.PubMedPubMedCentral
222.
Zurück zum Zitat Abrams ST, et al. Circulating histones are mediators of trauma-associated lung injury. Am J Respir Crit Care Med. 2013;187(2):160–9.PubMedPubMedCentral Abrams ST, et al. Circulating histones are mediators of trauma-associated lung injury. Am J Respir Crit Care Med. 2013;187(2):160–9.PubMedPubMedCentral
223.
Zurück zum Zitat Kalbitz M, et al. Cardiac depression in pigs after multiple trauma—characterization of posttraumatic structural and functional alterations. Sci Rep. 2017;7(1):17861.PubMedPubMedCentral Kalbitz M, et al. Cardiac depression in pigs after multiple trauma—characterization of posttraumatic structural and functional alterations. Sci Rep. 2017;7(1):17861.PubMedPubMedCentral
224.
Zurück zum Zitat Guisasola MC, et al. An overview of cytokines and heat shock response in polytraumatized patients. Cell Stress Chaperones. 2018;23(4):483–9.PubMed Guisasola MC, et al. An overview of cytokines and heat shock response in polytraumatized patients. Cell Stress Chaperones. 2018;23(4):483–9.PubMed
225.
Zurück zum Zitat De Maio A. Heat shock proteins: facts, thoughts, and dreams. Shock. 1999;11(1):1–12.PubMed De Maio A. Heat shock proteins: facts, thoughts, and dreams. Shock. 1999;11(1):1–12.PubMed
226.
Zurück zum Zitat DeMeester SL, Buchman TG, Cobb JP. The heat shock paradox: does NF-kappaB determine cell fate? FASEB J. 2001;15(1):270–4.PubMed DeMeester SL, Buchman TG, Cobb JP. The heat shock paradox: does NF-kappaB determine cell fate? FASEB J. 2001;15(1):270–4.PubMed
228.
Zurück zum Zitat Schopf FH, Biebl MM, Buchner J. The HSP90 chaperone machinery. Nat Rev Mol Cell Biol. 2017;18(6):345–60.PubMed Schopf FH, Biebl MM, Buchner J. The HSP90 chaperone machinery. Nat Rev Mol Cell Biol. 2017;18(6):345–60.PubMed
229.
Zurück zum Zitat Kim JY, Yenari MA. The immune modulating properties of the heat shock proteins after brain injury. Anat Cell Biol. 2013;46(1):1–7.PubMedPubMedCentral Kim JY, Yenari MA. The immune modulating properties of the heat shock proteins after brain injury. Anat Cell Biol. 2013;46(1):1–7.PubMedPubMedCentral
230.
Zurück zum Zitat Calderwood SK, et al. Extracellular heat shock proteins in cell signaling. FEBS Lett. 2007;581(19):3689–94.PubMed Calderwood SK, et al. Extracellular heat shock proteins in cell signaling. FEBS Lett. 2007;581(19):3689–94.PubMed
231.
Zurück zum Zitat Csermely P. The biology of extracellular molecular chaperones. Chair’s introduction. Novartis Found Symp. 2008;291:1–2.PubMed Csermely P. The biology of extracellular molecular chaperones. Chair’s introduction. Novartis Found Symp. 2008;291:1–2.PubMed
232.
Zurück zum Zitat Arnold-Schild D, et al. Cutting edge: receptor-mediated endocytosis of heat shock proteins by professional antigen-presenting cells. J Immunol. 1999;162(7):3757–60.PubMed Arnold-Schild D, et al. Cutting edge: receptor-mediated endocytosis of heat shock proteins by professional antigen-presenting cells. J Immunol. 1999;162(7):3757–60.PubMed
233.
Zurück zum Zitat Fong JJ, et al. Immunomodulatory activity of extracellular Hsp70 mediated via paired receptors Siglec-5 and Siglec-14. EMBO J. 2015;34(22):2775–88.PubMedPubMedCentral Fong JJ, et al. Immunomodulatory activity of extracellular Hsp70 mediated via paired receptors Siglec-5 and Siglec-14. EMBO J. 2015;34(22):2775–88.PubMedPubMedCentral
234.
Zurück zum Zitat Tang D, et al. The anti-inflammatory effects of heat shock protein 72 involve inhibition of high-mobility-group box 1 release and proinflammatory function in macrophages. J Immunol. 2007;179(2):1236–44.PubMed Tang D, et al. The anti-inflammatory effects of heat shock protein 72 involve inhibition of high-mobility-group box 1 release and proinflammatory function in macrophages. J Immunol. 2007;179(2):1236–44.PubMed
235.
Zurück zum Zitat Turturici G, Sconzo G, Geraci F. Hsp70 and its molecular role in nervous system diseases. Biochem Res Int. 2011;2011:618127.PubMedPubMedCentral Turturici G, Sconzo G, Geraci F. Hsp70 and its molecular role in nervous system diseases. Biochem Res Int. 2011;2011:618127.PubMedPubMedCentral
236.
Zurück zum Zitat Giffard RG, Yenari MA. Many mechanisms for hsp70 protection from cerebral ischemia. J Neurosurg Anesthesiol. 2004;16(1):53–61.PubMed Giffard RG, Yenari MA. Many mechanisms for hsp70 protection from cerebral ischemia. J Neurosurg Anesthesiol. 2004;16(1):53–61.PubMed
237.
Zurück zum Zitat Adachi H, et al. Heat shock proteins in neurodegenerative diseases: pathogenic roles and therapeutic implications. Int J Hyperth. 2009;25(8):647–54. Adachi H, et al. Heat shock proteins in neurodegenerative diseases: pathogenic roles and therapeutic implications. Int J Hyperth. 2009;25(8):647–54.
238.
Zurück zum Zitat Wheeler DS, et al. Extracellular heat shock protein 60 (Hsp60) levels in children with septic shock. Inflamm Res. 2007;56(5):216–9.PubMed Wheeler DS, et al. Extracellular heat shock protein 60 (Hsp60) levels in children with septic shock. Inflamm Res. 2007;56(5):216–9.PubMed
239.
Zurück zum Zitat Wheeler DS, et al. Extracellular hsp70 levels in children with septic shock. Pediatr Crit Care Med. 2005;6(3):308–11.PubMed Wheeler DS, et al. Extracellular hsp70 levels in children with septic shock. Pediatr Crit Care Med. 2005;6(3):308–11.PubMed
240.
Zurück zum Zitat Hashiguchi N, et al. Enhanced expression of heat shock proteins in activated polymorphonuclear leukocytes in patients with sepsis. J Trauma. 2001;51(6):1104–9.PubMed Hashiguchi N, et al. Enhanced expression of heat shock proteins in activated polymorphonuclear leukocytes in patients with sepsis. J Trauma. 2001;51(6):1104–9.PubMed
241.
Zurück zum Zitat Chatterjee A, et al. Heat shock protein 90 inhibitors prolong survival, attenuate inflammation, and reduce lung injury in murine sepsis. Am J Respir Crit Care Med. 2007;176(7):667–75.PubMedPubMedCentral Chatterjee A, et al. Heat shock protein 90 inhibitors prolong survival, attenuate inflammation, and reduce lung injury in murine sepsis. Am J Respir Crit Care Med. 2007;176(7):667–75.PubMedPubMedCentral
242.
Zurück zum Zitat Singleton KD, Wischmeyer PE. Effects of HSP70.1/3 gene knockout on acute respiratory distress syndrome and the inflammatory response following sepsis. Am J Physiol Lung Cell Mol Physiol. 2006;290(5):L956–61.PubMed Singleton KD, Wischmeyer PE. Effects of HSP70.1/3 gene knockout on acute respiratory distress syndrome and the inflammatory response following sepsis. Am J Physiol Lung Cell Mol Physiol. 2006;290(5):L956–61.PubMed
243.
Zurück zum Zitat McConnell KW, et al. The role of heat shock protein 70 in mediating age-dependent mortality in sepsis. J Immunol. 2011;186(6):3718–25.PubMed McConnell KW, et al. The role of heat shock protein 70 in mediating age-dependent mortality in sepsis. J Immunol. 2011;186(6):3718–25.PubMed
244.
Zurück zum Zitat Hashiguchi N, et al. Enhanced expression of heat shock proteins in leukocytes from trauma patients. J Trauma. 2001;50(1):102–7.PubMed Hashiguchi N, et al. Enhanced expression of heat shock proteins in leukocytes from trauma patients. J Trauma. 2001;50(1):102–7.PubMed
245.
Zurück zum Zitat Ogura H, et al. Long-term enhanced expression of heat shock proteins and decelerated apoptosis in polymorphonuclear leukocytes from major burn patients. J Burn Care Rehabil. 2002;23(2):103–9.PubMed Ogura H, et al. Long-term enhanced expression of heat shock proteins and decelerated apoptosis in polymorphonuclear leukocytes from major burn patients. J Burn Care Rehabil. 2002;23(2):103–9.PubMed
246.
Zurück zum Zitat Pespeni M, et al. Serum levels of Hsp60 correlate with the development of acute lung injury after trauma. J Surg Res. 2005;126(1):41–7.PubMed Pespeni M, et al. Serum levels of Hsp60 correlate with the development of acute lung injury after trauma. J Surg Res. 2005;126(1):41–7.PubMed
247.
Zurück zum Zitat Pockley AG, Muthana M, Calderwood SK. The dual immunoregulatory roles of stress proteins. Trends Biochem Sci. 2008;33(2):71–9.PubMed Pockley AG, Muthana M, Calderwood SK. The dual immunoregulatory roles of stress proteins. Trends Biochem Sci. 2008;33(2):71–9.PubMed
248.
Zurück zum Zitat Pittet JF, et al. Serum levels of Hsp 72 measured early after trauma correlate with survival. J Trauma. 2002;52(4):611–7 (discussion 617).PubMed Pittet JF, et al. Serum levels of Hsp 72 measured early after trauma correlate with survival. J Trauma. 2002;52(4):611–7 (discussion 617).PubMed
249.
Zurück zum Zitat Ren B, et al. Serum levels of HSP70 and other DAMP proteins can aid in patient diagnosis after traumatic injury. Cell Stress Chaperones. 2016;21(4):677–86.PubMedPubMedCentral Ren B, et al. Serum levels of HSP70 and other DAMP proteins can aid in patient diagnosis after traumatic injury. Cell Stress Chaperones. 2016;21(4):677–86.PubMedPubMedCentral
250.
Zurück zum Zitat Dehbi M, et al. Hsp-72, a candidate prognostic indicator of heatstroke. Cell Stress Chaperones. 2010;15(5):593–603.PubMedPubMedCentral Dehbi M, et al. Hsp-72, a candidate prognostic indicator of heatstroke. Cell Stress Chaperones. 2010;15(5):593–603.PubMedPubMedCentral
251.
Zurück zum Zitat Vardas K, et al. Early response roles for prolactin cortisol and circulating and cellular levels of heat shock proteins 72 and 90alpha in severe sepsis and SIRS. Biomed Res Int. 2014;2014:803561.PubMedPubMedCentral Vardas K, et al. Early response roles for prolactin cortisol and circulating and cellular levels of heat shock proteins 72 and 90alpha in severe sepsis and SIRS. Biomed Res Int. 2014;2014:803561.PubMedPubMedCentral
253.
Zurück zum Zitat Ma KC, et al. The role of danger signals in the pathogenesis and perpetuation of critical illness. Am J Respir Crit Care Med. 2018;197(3):300–9.PubMedPubMedCentral Ma KC, et al. The role of danger signals in the pathogenesis and perpetuation of critical illness. Am J Respir Crit Care Med. 2018;197(3):300–9.PubMedPubMedCentral
254.
Zurück zum Zitat Lee YL, et al. Blood transfusion products contain mitochondrial DNA damage-associated molecular patterns: a potential effector of transfusion-related acute lung injury. J Surg Res. 2014;191(2):286–9.PubMedPubMedCentral Lee YL, et al. Blood transfusion products contain mitochondrial DNA damage-associated molecular patterns: a potential effector of transfusion-related acute lung injury. J Surg Res. 2014;191(2):286–9.PubMedPubMedCentral
255.
Zurück zum Zitat Preissner KT, Herwald H. Extracellular nucleic acids in immunity and cardiovascular responses: between alert and disease. Thromb Haemost. 2017;117(7):1272–82.PubMed Preissner KT, Herwald H. Extracellular nucleic acids in immunity and cardiovascular responses: between alert and disease. Thromb Haemost. 2017;117(7):1272–82.PubMed
256.
Zurück zum Zitat Zhang Q, et al. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010;464(7285):104–7.PubMedPubMedCentral Zhang Q, et al. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010;464(7285):104–7.PubMedPubMedCentral
257.
Zurück zum Zitat Liew FY, et al. Negative regulation of toll-like receptor-mediated immune responses. Nat Rev Immunol. 2005;5(6):446–58.PubMed Liew FY, et al. Negative regulation of toll-like receptor-mediated immune responses. Nat Rev Immunol. 2005;5(6):446–58.PubMed
258.
Zurück zum Zitat Miyake K, et al. Mechanisms controlling nucleic acid-sensing Toll-like receptors. Int Immunol. 2018;30(2):43–51.PubMed Miyake K, et al. Mechanisms controlling nucleic acid-sensing Toll-like receptors. Int Immunol. 2018;30(2):43–51.PubMed
259.
Zurück zum Zitat Roers A, Hiller B, Hornung V. Recognition of endogenous nucleic acids by the innate immune system. Immunity. 2016;44(4):739–54.PubMed Roers A, Hiller B, Hornung V. Recognition of endogenous nucleic acids by the innate immune system. Immunity. 2016;44(4):739–54.PubMed
260.
Zurück zum Zitat West AP, Shadel GS. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat Rev Immunol. 2017;17(6):363–75.PubMedPubMedCentral West AP, Shadel GS. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat Rev Immunol. 2017;17(6):363–75.PubMedPubMedCentral
261.
Zurück zum Zitat Barber GN. STING-dependent cytosolic DNA sensing pathways. Trends Immunol. 2014;35(2):88–93.PubMed Barber GN. STING-dependent cytosolic DNA sensing pathways. Trends Immunol. 2014;35(2):88–93.PubMed
262.
Zurück zum Zitat Thurairajah K, Briggs GD, Balogh ZJ. The source of cell-free mitochondrial DNA in trauma and potential therapeutic strategies. Eur J Trauma Emerg Surg. 2018;44(3):325–34.PubMedPubMedCentral Thurairajah K, Briggs GD, Balogh ZJ. The source of cell-free mitochondrial DNA in trauma and potential therapeutic strategies. Eur J Trauma Emerg Surg. 2018;44(3):325–34.PubMedPubMedCentral
263.
Zurück zum Zitat Deane JA, Bolland S. Nucleic acid-sensing TLRs as modifiers of autoimmunity. J Immunol. 2006;177(10):6573–8.PubMed Deane JA, Bolland S. Nucleic acid-sensing TLRs as modifiers of autoimmunity. J Immunol. 2006;177(10):6573–8.PubMed
264.
Zurück zum Zitat Saitoh S, Miyake K. Regulatory molecules required for nucleotide-sensing Toll-like receptors. Immunol Rev. 2009;227(1):32–43.PubMed Saitoh S, Miyake K. Regulatory molecules required for nucleotide-sensing Toll-like receptors. Immunol Rev. 2009;227(1):32–43.PubMed
265.
Zurück zum Zitat Barton GM, Kagan JC, Medzhitov R. Intracellular localization of Toll-like receptor 9 prevents recognition of self DNA but facilitates access to viral DNA. Nat Immunol. 2006;7(1):49–56.PubMed Barton GM, Kagan JC, Medzhitov R. Intracellular localization of Toll-like receptor 9 prevents recognition of self DNA but facilitates access to viral DNA. Nat Immunol. 2006;7(1):49–56.PubMed
266.
Zurück zum Zitat Sipes JN, et al. A prospective study of plasma DNA in the diagnosis of pulmonary embolism. Am Rev Respir Dis. 1978;118(3):475–8.PubMed Sipes JN, et al. A prospective study of plasma DNA in the diagnosis of pulmonary embolism. Am Rev Respir Dis. 1978;118(3):475–8.PubMed
267.
Zurück zum Zitat Arnalich F, et al. Plasma levels of mitochondrial and nuclear DNA in patients with massive pulmonary embolism in the emergency department: a prospective cohort study. Crit Care. 2013;17(3):R90.PubMedPubMedCentral Arnalich F, et al. Plasma levels of mitochondrial and nuclear DNA in patients with massive pulmonary embolism in the emergency department: a prospective cohort study. Crit Care. 2013;17(3):R90.PubMedPubMedCentral
268.
269.
Zurück zum Zitat Kung CT, et al. Plasma nuclear and mitochondrial DNA levels as predictors of outcome in severe sepsis patients in the emergency room. J Transl Med. 2012;10:130.PubMedPubMedCentral Kung CT, et al. Plasma nuclear and mitochondrial DNA levels as predictors of outcome in severe sepsis patients in the emergency room. J Transl Med. 2012;10:130.PubMedPubMedCentral
270.
Zurück zum Zitat Itagaki K, et al. Mitochondrial DNA released by trauma induces neutrophil extracellular traps. PLoS ONE. 2015;10(3):e0120549.PubMedPubMedCentral Itagaki K, et al. Mitochondrial DNA released by trauma induces neutrophil extracellular traps. PLoS ONE. 2015;10(3):e0120549.PubMedPubMedCentral
271.
Zurück zum Zitat Cedervall J, Zhang Y, Olsson AK. Tumor-induced NETosis as a risk factor for metastasis and organ failure. Cancer Res. 2016;76(15):4311–5.PubMed Cedervall J, Zhang Y, Olsson AK. Tumor-induced NETosis as a risk factor for metastasis and organ failure. Cancer Res. 2016;76(15):4311–5.PubMed
272.
Zurück zum Zitat Lee KH, et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: a comprehensive review. Autoimmun Rev. 2017;16(11):1160–73.PubMed Lee KH, et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: a comprehensive review. Autoimmun Rev. 2017;16(11):1160–73.PubMed
273.
Zurück zum Zitat Sandler N, et al. Mitochondrial DAMPs are released during cardiopulmonary bypass surgery and are associated with postoperative atrial fibrillation. Heart Lung Circ. 2018;27(1):122–9.PubMed Sandler N, et al. Mitochondrial DAMPs are released during cardiopulmonary bypass surgery and are associated with postoperative atrial fibrillation. Heart Lung Circ. 2018;27(1):122–9.PubMed
274.
Zurück zum Zitat Zou J, et al. Rapid detection of donor cell free DNA in lung transplant recipients with rejections using donor-recipient HLA mismatch. Hum Immunol. 2017;78(4):342–9.PubMedPubMedCentral Zou J, et al. Rapid detection of donor cell free DNA in lung transplant recipients with rejections using donor-recipient HLA mismatch. Hum Immunol. 2017;78(4):342–9.PubMedPubMedCentral
277.
Zurück zum Zitat Wilkins HM, et al. Mitochondria-derived damage-associated molecular patterns in neurodegeneration. Front Immunol. 2017;8:508.PubMedPubMedCentral Wilkins HM, et al. Mitochondria-derived damage-associated molecular patterns in neurodegeneration. Front Immunol. 2017;8:508.PubMedPubMedCentral
278.
Zurück zum Zitat Simmons JD, et al. Elevated levels of plasma mitochondrial DNA DAMPs are linked to clinical outcome in severely injured human subjects. Ann Surg. 2013;258(4):591–6 (discussion 596-8).PubMed Simmons JD, et al. Elevated levels of plasma mitochondrial DNA DAMPs are linked to clinical outcome in severely injured human subjects. Ann Surg. 2013;258(4):591–6 (discussion 596-8).PubMed
279.
Zurück zum Zitat Lam NY, et al. Plasma mitochondrial DNA concentrations after trauma. Clin Chem. 2004;50(1):213–6.PubMed Lam NY, et al. Plasma mitochondrial DNA concentrations after trauma. Clin Chem. 2004;50(1):213–6.PubMed
280.
Zurück zum Zitat Gu X, et al. The plasma mitochondrial DNA is an independent predictor for post-traumatic systemic inflammatory response syndrome. PLoS ONE. 2013;8(8):e72834.PubMedPubMedCentral Gu X, et al. The plasma mitochondrial DNA is an independent predictor for post-traumatic systemic inflammatory response syndrome. PLoS ONE. 2013;8(8):e72834.PubMedPubMedCentral
281.
Zurück zum Zitat Lo YM, et al. Plasma DNA as a prognostic marker in trauma patients. Clin Chem. 2000;46(3):319–23.PubMed Lo YM, et al. Plasma DNA as a prognostic marker in trauma patients. Clin Chem. 2000;46(3):319–23.PubMed
282.
Zurück zum Zitat Gan L, et al. Significance of serum mtDNA concentration in lung injury induced by hip fracture. Shock. 2015;44(1):52–7.PubMed Gan L, et al. Significance of serum mtDNA concentration in lung injury induced by hip fracture. Shock. 2015;44(1):52–7.PubMed
283.
Zurück zum Zitat Zhang Q, Itagaki K, Hauser CJ. Mitochondrial DNA is released by shock and activates neutrophils via p38 map kinase. Shock. 2010;34(1):55–9.PubMed Zhang Q, Itagaki K, Hauser CJ. Mitochondrial DNA is released by shock and activates neutrophils via p38 map kinase. Shock. 2010;34(1):55–9.PubMed
284.
Zurück zum Zitat McIlroy DJ, et al. Cell necrosis-independent sustained mitochondrial and nuclear DNA release following trauma surgery. J Trauma Acute Care Surg. 2015;78(2):282–8.PubMedPubMedCentral McIlroy DJ, et al. Cell necrosis-independent sustained mitochondrial and nuclear DNA release following trauma surgery. J Trauma Acute Care Surg. 2015;78(2):282–8.PubMedPubMedCentral
285.
Zurück zum Zitat Hauser CJ, et al. Mitochondrial damage associated molecular patterns from femoral reamings activate neutrophils through formyl peptide receptors and P44/42 MAP kinase. J Orthop Trauma. 2010;24(9):534–8.PubMedPubMedCentral Hauser CJ, et al. Mitochondrial damage associated molecular patterns from femoral reamings activate neutrophils through formyl peptide receptors and P44/42 MAP kinase. J Orthop Trauma. 2010;24(9):534–8.PubMedPubMedCentral
286.
Zurück zum Zitat Puyo CA, et al. Endotracheal tube-induced sore throat pain and inflammation is coupled to the release of mitochondrial DNA. Mol Pain. 2017;13:1744806917731696.PubMedPubMedCentral Puyo CA, et al. Endotracheal tube-induced sore throat pain and inflammation is coupled to the release of mitochondrial DNA. Mol Pain. 2017;13:1744806917731696.PubMedPubMedCentral
287.
Zurück zum Zitat Yousefi S, et al. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 2009;16(11):1438–44.PubMed Yousefi S, et al. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 2009;16(11):1438–44.PubMed
288.
Zurück zum Zitat Prikhodko AS, et al. Pure mitochondrial DNA does not activate human neutrophils in vitro. Biochemistry (Mosc). 2015;80(5):629–35. Prikhodko AS, et al. Pure mitochondrial DNA does not activate human neutrophils in vitro. Biochemistry (Mosc). 2015;80(5):629–35.
289.
Zurück zum Zitat Willart MA, Lambrecht BN. The danger within: endogenous danger signals, atopy and asthma. Clin Exp Allergy. 2009;39(1):12–9.PubMed Willart MA, Lambrecht BN. The danger within: endogenous danger signals, atopy and asthma. Clin Exp Allergy. 2009;39(1):12–9.PubMed
290.
Zurück zum Zitat Novak I. ATP as a signaling molecule: the exocrine focus. News Physiol Sci. 2003;18:12–7.PubMed Novak I. ATP as a signaling molecule: the exocrine focus. News Physiol Sci. 2003;18:12–7.PubMed
291.
Zurück zum Zitat Martinon F. Detection of immune danger signals by NALP3. J Leukoc Biol. 2008;83(3):507–11.PubMed Martinon F. Detection of immune danger signals by NALP3. J Leukoc Biol. 2008;83(3):507–11.PubMed
292.
Zurück zum Zitat Elliott MR, et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature. 2009;461(7261):282–6.PubMedPubMedCentral Elliott MR, et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature. 2009;461(7261):282–6.PubMedPubMedCentral
293.
Zurück zum Zitat Chen Y, et al. ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors. Science. 2006;314(5806):1792–5.PubMed Chen Y, et al. ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors. Science. 2006;314(5806):1792–5.PubMed
294.
296.
Zurück zum Zitat Peiseler M, Kubes P. More friend than foe: the emerging role of neutrophils in tissue repair. J Clin Invest. 2019;129(7):2629–39.PubMedPubMedCentral Peiseler M, Kubes P. More friend than foe: the emerging role of neutrophils in tissue repair. J Clin Invest. 2019;129(7):2629–39.PubMedPubMedCentral
297.
Zurück zum Zitat Li X, et al. Systemic adenosine triphosphate impairs neutrophil chemotaxis and host defense in sepsis. Crit Care Med. 2017;45(1):e97–104.PubMedPubMedCentral Li X, et al. Systemic adenosine triphosphate impairs neutrophil chemotaxis and host defense in sepsis. Crit Care Med. 2017;45(1):e97–104.PubMedPubMedCentral
298.
Zurück zum Zitat Hasan D, Blankman P, Nieman GF. Purinergic signalling links mechanical breath profile and alveolar mechanics with the pro-inflammatory innate immune response causing ventilation-induced lung injury. Purinergic Signal. 2017;13(3):363–86.PubMedPubMedCentral Hasan D, Blankman P, Nieman GF. Purinergic signalling links mechanical breath profile and alveolar mechanics with the pro-inflammatory innate immune response causing ventilation-induced lung injury. Purinergic Signal. 2017;13(3):363–86.PubMedPubMedCentral
299.
Zurück zum Zitat Chakraborty S, Karasu E, Huber-Lang M. Complement after trauma: suturing innate and adaptive immunity. Front Immunol. 2018;9:2050.PubMedPubMedCentral Chakraborty S, Karasu E, Huber-Lang M. Complement after trauma: suturing innate and adaptive immunity. Front Immunol. 2018;9:2050.PubMedPubMedCentral
300.
Zurück zum Zitat Frohlich M, et al. Epidemiology and risk factors of multiple-organ failure after multiple trauma: an analysis of 31,154 patients from the TraumaRegister DGU. J Trauma Acute Care Surg. 2014;76(4):921–7 (discussion 927-8).PubMed Frohlich M, et al. Epidemiology and risk factors of multiple-organ failure after multiple trauma: an analysis of 31,154 patients from the TraumaRegister DGU. J Trauma Acute Care Surg. 2014;76(4):921–7 (discussion 927-8).PubMed
301.
Zurück zum Zitat Hecke F, et al. Circulating complement proteins in multiple trauma patients–correlation with injury severity, development of sepsis, and outcome. Crit Care Med. 1997;25(12):2015–24.PubMed Hecke F, et al. Circulating complement proteins in multiple trauma patients–correlation with injury severity, development of sepsis, and outcome. Crit Care Med. 1997;25(12):2015–24.PubMed
302.
Zurück zum Zitat Ganter MT, et al. Role of the alternative pathway in the early complement activation following major trauma. Shock. 2007;28(1):29–34.PubMed Ganter MT, et al. Role of the alternative pathway in the early complement activation following major trauma. Shock. 2007;28(1):29–34.PubMed
303.
304.
Zurück zum Zitat Paredes RM, et al. Generation of complement molecular complex C5b-9 (C5b-9) in response to poly-traumatic hemorrhagic shock and evaluation of C5 cleavage inhibitors in non-human primates. Int Immunopharmacol. 2018;54:221–5.PubMed Paredes RM, et al. Generation of complement molecular complex C5b-9 (C5b-9) in response to poly-traumatic hemorrhagic shock and evaluation of C5 cleavage inhibitors in non-human primates. Int Immunopharmacol. 2018;54:221–5.PubMed
305.
Zurück zum Zitat Li Y, et al. Complement inhibition ameliorates blast-induced acute lung injury in rats: potential role of complement in intracellular HMGB1-mediated inflammation. PLoS ONE. 2018;13(8):e0202594.PubMedPubMedCentral Li Y, et al. Complement inhibition ameliorates blast-induced acute lung injury in rats: potential role of complement in intracellular HMGB1-mediated inflammation. PLoS ONE. 2018;13(8):e0202594.PubMedPubMedCentral
306.
308.
Zurück zum Zitat Colombo M, Raposo G, Thery C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.PubMed Colombo M, Raposo G, Thery C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.PubMed
309.
Zurück zum Zitat Muralidharan-Chari V, et al. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr Biol. 2009;19(22):1875–85.PubMedPubMedCentral Muralidharan-Chari V, et al. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr Biol. 2009;19(22):1875–85.PubMedPubMedCentral
310.
Zurück zum Zitat Lima LG, et al. Tumor-derived microvesicles modulate the establishment of metastatic melanoma in a phosphatidylserine-dependent manner. Cancer Lett. 2009;283(2):168–75.PubMed Lima LG, et al. Tumor-derived microvesicles modulate the establishment of metastatic melanoma in a phosphatidylserine-dependent manner. Cancer Lett. 2009;283(2):168–75.PubMed
311.
Zurück zum Zitat Iraci N, et al. Focus on extracellular vesicles: physiological role and signalling properties of extracellular membrane vesicles. Int J Mol Sci. 2016;17(2):171.PubMedPubMedCentral Iraci N, et al. Focus on extracellular vesicles: physiological role and signalling properties of extracellular membrane vesicles. Int J Mol Sci. 2016;17(2):171.PubMedPubMedCentral
312.
Zurück zum Zitat Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3:24641. Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3:24641.
313.
Zurück zum Zitat Montecalvo A, et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood. 2012;119(3):756–66.PubMedPubMedCentral Montecalvo A, et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood. 2012;119(3):756–66.PubMedPubMedCentral
314.
Zurück zum Zitat Morel O, et al. Microparticles: a critical component in the nexus between inflammation, immunity, and thrombosis. Semin Immunopathol. 2011;33(5):469–86.PubMed Morel O, et al. Microparticles: a critical component in the nexus between inflammation, immunity, and thrombosis. Semin Immunopathol. 2011;33(5):469–86.PubMed
315.
Zurück zum Zitat Rautou PE, et al. Microparticles, vascular function, and atherothrombosis. Circ Res. 2011;109(5):593–606.PubMed Rautou PE, et al. Microparticles, vascular function, and atherothrombosis. Circ Res. 2011;109(5):593–606.PubMed
316.
Zurück zum Zitat Diehl P, et al. Microparticles: major transport vehicles for distinct microRNAs in circulation. Cardiovasc Res. 2012;93(4):633–44.PubMedPubMedCentral Diehl P, et al. Microparticles: major transport vehicles for distinct microRNAs in circulation. Cardiovasc Res. 2012;93(4):633–44.PubMedPubMedCentral
317.
Zurück zum Zitat Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373–83.PubMedPubMedCentral Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373–83.PubMedPubMedCentral
318.
Zurück zum Zitat Eguchi A, et al. Microparticles release by adipocytes act as “find-me” signals to promote macrophage migration. PLoS ONE. 2015;10(4):e0123110.PubMedPubMedCentral Eguchi A, et al. Microparticles release by adipocytes act as “find-me” signals to promote macrophage migration. PLoS ONE. 2015;10(4):e0123110.PubMedPubMedCentral
319.
Zurück zum Zitat Yanez-Mo M, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066.PubMed Yanez-Mo M, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066.PubMed
320.
Zurück zum Zitat Povero D, et al. Lipid-induced toxicity stimulates hepatocytes to release angiogenic microparticles that require Vanin-1 for uptake by endothelial cells. Sci Signal. 2013;6(296):ra88.PubMedPubMedCentral Povero D, et al. Lipid-induced toxicity stimulates hepatocytes to release angiogenic microparticles that require Vanin-1 for uptake by endothelial cells. Sci Signal. 2013;6(296):ra88.PubMedPubMedCentral
321.
Zurück zum Zitat Povero D, et al. Circulating extracellular vesicles with specific proteome and liver microRNAs are potential biomarkers for liver injury in experimental fatty liver disease. PLoS ONE. 2014;9(12):e113651.PubMedPubMedCentral Povero D, et al. Circulating extracellular vesicles with specific proteome and liver microRNAs are potential biomarkers for liver injury in experimental fatty liver disease. PLoS ONE. 2014;9(12):e113651.PubMedPubMedCentral
322.
Zurück zum Zitat Eguchi A, et al. Extracellular vesicles released by hepatocytes from gastric infusion model of alcoholic liver disease contain a MicroRNA barcode that can be detected in blood. Hepatology. 2017;65(2):475–90.PubMed Eguchi A, et al. Extracellular vesicles released by hepatocytes from gastric infusion model of alcoholic liver disease contain a MicroRNA barcode that can be detected in blood. Hepatology. 2017;65(2):475–90.PubMed
323.
Zurück zum Zitat Momen-Heravi F, et al. Exosomes derived from alcohol-treated hepatocytes horizontally transfer liver specific miRNA-122 and sensitize monocytes to LPS. Sci Rep. 2015;5:9991.PubMedPubMedCentral Momen-Heravi F, et al. Exosomes derived from alcohol-treated hepatocytes horizontally transfer liver specific miRNA-122 and sensitize monocytes to LPS. Sci Rep. 2015;5:9991.PubMedPubMedCentral
324.
Zurück zum Zitat Zitvogel L, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4(5):594–600.PubMed Zitvogel L, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4(5):594–600.PubMed
325.
Zurück zum Zitat Liu Q, et al. Donor dendritic cell-derived exosomes promote allograft-targeting immune response. J Clin Invest. 2016;126(8):2805–20.PubMedPubMedCentral Liu Q, et al. Donor dendritic cell-derived exosomes promote allograft-targeting immune response. J Clin Invest. 2016;126(8):2805–20.PubMedPubMedCentral
326.
Zurück zum Zitat Marino J, et al. Donor exosomes rather than passenger leukocytes initiate alloreactive T cell responses after transplantation. Sci Immunol. 2016;1(1):aaf8759.PubMedPubMedCentral Marino J, et al. Donor exosomes rather than passenger leukocytes initiate alloreactive T cell responses after transplantation. Sci Immunol. 2016;1(1):aaf8759.PubMedPubMedCentral
327.
Zurück zum Zitat Zhang HG, Grizzle WE. Exosomes and cancer: a newly described pathway of immune suppression. Clin Cancer Res. 2011;17(5):959–64.PubMedPubMedCentral Zhang HG, Grizzle WE. Exosomes and cancer: a newly described pathway of immune suppression. Clin Cancer Res. 2011;17(5):959–64.PubMedPubMedCentral
328.
Zurück zum Zitat Rak J. Microparticles in cancer. Semin Thromb Hemost. 2010;36(8):888–906.PubMed Rak J. Microparticles in cancer. Semin Thromb Hemost. 2010;36(8):888–906.PubMed
329.
Zurück zum Zitat Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011;71(11):3792–801.PubMed Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011;71(11):3792–801.PubMed
330.
Zurück zum Zitat Chang PY, et al. The potential of mesenchymal stem cells in the management of radiation enteropathy. Cell Death Dis. 2015;6:e1840.PubMedPubMedCentral Chang PY, et al. The potential of mesenchymal stem cells in the management of radiation enteropathy. Cell Death Dis. 2015;6:e1840.PubMedPubMedCentral
331.
Zurück zum Zitat Kordelas L, et al. MSC-derived exosomes: a novel tool to treat therapy-refractory graft-versus-host disease. Leukemia. 2014;28(4):970–3.PubMed Kordelas L, et al. MSC-derived exosomes: a novel tool to treat therapy-refractory graft-versus-host disease. Leukemia. 2014;28(4):970–3.PubMed
332.
Zurück zum Zitat Nassar W, et al. Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases. Biomater Res. 2016;20:21.PubMedPubMedCentral Nassar W, et al. Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases. Biomater Res. 2016;20:21.PubMedPubMedCentral
333.
Zurück zum Zitat Gardiner C, et al. Extracellular vesicles, tissue factor, cancer and thrombosis—discussion themes of the ISEV 2014 Educational Day. J Extracell Vesicles. 2015;4:26901.PubMed Gardiner C, et al. Extracellular vesicles, tissue factor, cancer and thrombosis—discussion themes of the ISEV 2014 Educational Day. J Extracell Vesicles. 2015;4:26901.PubMed
334.
Zurück zum Zitat Muhsin-Sharafaldine MR, et al. Procoagulant and immunogenic properties of melanoma exosomes, microvesicles and apoptotic vesicles. Oncotarget. 2016;7(35):56279–94.PubMedPubMedCentral Muhsin-Sharafaldine MR, et al. Procoagulant and immunogenic properties of melanoma exosomes, microvesicles and apoptotic vesicles. Oncotarget. 2016;7(35):56279–94.PubMedPubMedCentral
335.
Zurück zum Zitat Janiszewski M, et al. Platelet-derived exosomes of septic individuals possess proapoptotic NAD(P)H oxidase activity: a novel vascular redox pathway. Crit Care Med. 2004;32(3):818–25.PubMed Janiszewski M, et al. Platelet-derived exosomes of septic individuals possess proapoptotic NAD(P)H oxidase activity: a novel vascular redox pathway. Crit Care Med. 2004;32(3):818–25.PubMed
336.
Zurück zum Zitat Essandoh K, et al. Blockade of exosome generation with GW4869 dampens the sepsis-induced inflammation and cardiac dysfunction. Biochim Biophys Acta. 2015;1852(11):2362–71.PubMedPubMedCentral Essandoh K, et al. Blockade of exosome generation with GW4869 dampens the sepsis-induced inflammation and cardiac dysfunction. Biochim Biophys Acta. 2015;1852(11):2362–71.PubMedPubMedCentral
337.
Zurück zum Zitat Trajkovic K, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244–7.PubMed Trajkovic K, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244–7.PubMed
338.
Zurück zum Zitat Zhang ZG, Buller B, Chopp M. Exosomes—beyond stem cells for restorative therapy in stroke and neurological injury. Nat Rev Neurol. 2019;15(4):193–203.PubMed Zhang ZG, Buller B, Chopp M. Exosomes—beyond stem cells for restorative therapy in stroke and neurological injury. Nat Rev Neurol. 2019;15(4):193–203.PubMed
339.
Zurück zum Zitat Eguchi A, et al. Circulating extracellular vesicles and their miR “Barcode” differentiate alcohol drinkers with liver injury and those without liver injury in severe trauma patients. Front Med (Lausanne). 2019;6:30. Eguchi A, et al. Circulating extracellular vesicles and their miR “Barcode” differentiate alcohol drinkers with liver injury and those without liver injury in severe trauma patients. Front Med (Lausanne). 2019;6:30.
340.
Zurück zum Zitat Kojima M, et al. Exosomes, not protein or lipids, in mesenteric lymph activate inflammation: unlocking the mystery of post-shock multiple organ failure. J Trauma Acute Care Surg. 2017;82(1):42–50.PubMed Kojima M, et al. Exosomes, not protein or lipids, in mesenteric lymph activate inflammation: unlocking the mystery of post-shock multiple organ failure. J Trauma Acute Care Surg. 2017;82(1):42–50.PubMed
341.
Zurück zum Zitat Williams EC, et al. Precious cargo: modulation of the mesenteric lymph exosome payload after hemorrhagic shock. J Trauma Acute Care Surg. 2019;86(1):52–61.PubMedPubMedCentral Williams EC, et al. Precious cargo: modulation of the mesenteric lymph exosome payload after hemorrhagic shock. J Trauma Acute Care Surg. 2019;86(1):52–61.PubMedPubMedCentral
342.
Zurück zum Zitat Haneklaus M, O’Neill LA, Coll RC. Modulatory mechanisms controlling the NLRP3 inflammasome in inflammation: recent developments. Curr Opin Immunol. 2013;25(1):40–5.PubMed Haneklaus M, O’Neill LA, Coll RC. Modulatory mechanisms controlling the NLRP3 inflammasome in inflammation: recent developments. Curr Opin Immunol. 2013;25(1):40–5.PubMed
343.
Zurück zum Zitat de Rivero Vaccari JP, et al. Exosome-mediated inflammasome signaling after central nervous system injury. J Neurochem. 2016;136(Suppl 1):39–48.PubMed de Rivero Vaccari JP, et al. Exosome-mediated inflammasome signaling after central nervous system injury. J Neurochem. 2016;136(Suppl 1):39–48.PubMed
344.
Zurück zum Zitat Ogura H, et al. Activated platelets enhance microparticle formation and platelet-leukocyte interaction in severe trauma and sepsis. J Trauma. 2001;50(5):801–9.PubMed Ogura H, et al. Activated platelets enhance microparticle formation and platelet-leukocyte interaction in severe trauma and sepsis. J Trauma. 2001;50(5):801–9.PubMed
345.
Zurück zum Zitat Curry N, et al. Levels of procoagulant microvesicles are elevated after traumatic injury and platelet microvesicles are negatively correlated with mortality. J Extracell Vesicles. 2014;3:25625.PubMed Curry N, et al. Levels of procoagulant microvesicles are elevated after traumatic injury and platelet microvesicles are negatively correlated with mortality. J Extracell Vesicles. 2014;3:25625.PubMed
346.
Zurück zum Zitat Matijevic N, et al. Cellular microparticle and thrombogram phenotypes in the Prospective Observational Multicenter Major Trauma Transfusion (PROMMTT) study: correlation with coagulopathy. Thromb Res. 2014;134(3):652–8.PubMedPubMedCentral Matijevic N, et al. Cellular microparticle and thrombogram phenotypes in the Prospective Observational Multicenter Major Trauma Transfusion (PROMMTT) study: correlation with coagulopathy. Thromb Res. 2014;134(3):652–8.PubMedPubMedCentral
347.
Zurück zum Zitat Morel N, et al. Generation of procoagulant microparticles in cerebrospinal fluid and peripheral blood after traumatic brain injury. J Trauma. 2008;64(3):698–704.PubMed Morel N, et al. Generation of procoagulant microparticles in cerebrospinal fluid and peripheral blood after traumatic brain injury. J Trauma. 2008;64(3):698–704.PubMed
348.
Zurück zum Zitat Fujimi S, et al. Increased production of leukocyte microparticles with enhanced expression of adhesion molecules from activated polymorphonuclear leukocytes in severely injured patients. J Trauma. 2003;54(1):114–9 (discussion 119-20).PubMed Fujimi S, et al. Increased production of leukocyte microparticles with enhanced expression of adhesion molecules from activated polymorphonuclear leukocytes in severely injured patients. J Trauma. 2003;54(1):114–9 (discussion 119-20).PubMed
349.
Zurück zum Zitat Kuravi SJ, et al. Changes in the pattern of plasma extracellular vesicles after severe trauma. PLoS ONE. 2017;12(8):e0183640.PubMedPubMedCentral Kuravi SJ, et al. Changes in the pattern of plasma extracellular vesicles after severe trauma. PLoS ONE. 2017;12(8):e0183640.PubMedPubMedCentral
350.
Zurück zum Zitat Mense SM, Zhang L. Heme: a versatile signaling molecule controlling the activities of diverse regulators ranging from transcription factors to MAP kinases. Cell Res. 2006;16(8):681–92.PubMed Mense SM, Zhang L. Heme: a versatile signaling molecule controlling the activities of diverse regulators ranging from transcription factors to MAP kinases. Cell Res. 2006;16(8):681–92.PubMed
351.
Zurück zum Zitat Larsen R, et al. A central role for free heme in the pathogenesis of severe sepsis. Sci Transl Med. 2010;2(51):51ra71.PubMed Larsen R, et al. A central role for free heme in the pathogenesis of severe sepsis. Sci Transl Med. 2010;2(51):51ra71.PubMed
352.
Zurück zum Zitat Li W, et al. Oxidative damage and mitochondrial injuries differ following pneumoperitoneum pressure in rabbit models of varying degrees of hydronephrosis. Mol Med Rep. 2018;17(5):6819–27.PubMed Li W, et al. Oxidative damage and mitochondrial injuries differ following pneumoperitoneum pressure in rabbit models of varying degrees of hydronephrosis. Mol Med Rep. 2018;17(5):6819–27.PubMed
353.
Zurück zum Zitat Rani M, et al. Damage-associated molecular patterns (DAMPs) released after burn are associated with inflammation and monocyte activation. Burns. 2017;43(2):297–303.PubMed Rani M, et al. Damage-associated molecular patterns (DAMPs) released after burn are associated with inflammation and monocyte activation. Burns. 2017;43(2):297–303.PubMed
354.
Zurück zum Zitat Mendonca R, Silveira AA, Conran N. Red cell DAMPs and inflammation. Inflamm Res. 2016;65(9):665–78.PubMed Mendonca R, Silveira AA, Conran N. Red cell DAMPs and inflammation. Inflamm Res. 2016;65(9):665–78.PubMed
355.
Zurück zum Zitat Schaer DJ, et al. Haptoglobin, hemopexin, and related defense pathways-basic science, clinical perspectives, and drug development. Front Physiol. 2014;5:415.PubMedPubMedCentral Schaer DJ, et al. Haptoglobin, hemopexin, and related defense pathways-basic science, clinical perspectives, and drug development. Front Physiol. 2014;5:415.PubMedPubMedCentral
356.
Zurück zum Zitat Billich A, et al. Presence of cyclophilin A in synovial fluids of patients with rheumatoid arthritis. J Exp Med. 1997;185(5):975–80.PubMedPubMedCentral Billich A, et al. Presence of cyclophilin A in synovial fluids of patients with rheumatoid arthritis. J Exp Med. 1997;185(5):975–80.PubMedPubMedCentral
357.
Zurück zum Zitat Dear JW, et al. Cyclophilin A is a damage-associated molecular pattern molecule that mediates acetaminophen-induced liver injury. J Immunol. 2011;187(6):3347–52.PubMed Dear JW, et al. Cyclophilin A is a damage-associated molecular pattern molecule that mediates acetaminophen-induced liver injury. J Immunol. 2011;187(6):3347–52.PubMed
358.
Zurück zum Zitat Tegeder I, et al. Elevated serum cyclophilin levels in patients with severe sepsis. J Clin Immunol. 1997;17(5):380–6.PubMed Tegeder I, et al. Elevated serum cyclophilin levels in patients with severe sepsis. J Clin Immunol. 1997;17(5):380–6.PubMed
359.
Zurück zum Zitat Arun P, et al. Extracellular cyclophilin A protects against blast-induced neuronal injury. Neurosci Res. 2013;76(1–2):98–100.PubMed Arun P, et al. Extracellular cyclophilin A protects against blast-induced neuronal injury. Neurosci Res. 2013;76(1–2):98–100.PubMed
360.
Zurück zum Zitat Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003;425(6957):516–21.PubMed Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003;425(6957):516–21.PubMed
361.
Zurück zum Zitat Ragab G, Elshahaly M, Bardin T. Gout: an old disease in new perspective—a review. J Adv Res. 2017;8(5):495–511.PubMedPubMedCentral Ragab G, Elshahaly M, Bardin T. Gout: an old disease in new perspective—a review. J Adv Res. 2017;8(5):495–511.PubMedPubMedCentral
362.
Zurück zum Zitat Martinon F, et al. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440(7081):237–41.PubMed Martinon F, et al. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440(7081):237–41.PubMed
363.
Zurück zum Zitat Liang J, et al. Elevated serum uric acid after injury correlates with the early acute kidney in severe burns. Burns. 2015;41(8):1724–31.PubMed Liang J, et al. Elevated serum uric acid after injury correlates with the early acute kidney in severe burns. Burns. 2015;41(8):1724–31.PubMed
364.
Zurück zum Zitat Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12(1):991–1045.PubMed Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12(1):991–1045.PubMed
365.
Zurück zum Zitat Land W, et al. The beneficial effect of human recombinant superoxide dismutase on acute and chronic rejection events in recipients of cadaveric renal transplants. Transplantation. 1994;57(2):211–7.PubMed Land W, et al. The beneficial effect of human recombinant superoxide dismutase on acute and chronic rejection events in recipients of cadaveric renal transplants. Transplantation. 1994;57(2):211–7.PubMed
366.
Zurück zum Zitat Medzhitov R, Preston-Hurlburt P, Janeway CA. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997;388(6640):394–7.PubMed Medzhitov R, Preston-Hurlburt P, Janeway CA. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997;388(6640):394–7.PubMed
367.
Zurück zum Zitat Janeway CA. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb Symp Quant Biol. 1989;54(Pt 1):1–13.PubMed Janeway CA. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb Symp Quant Biol. 1989;54(Pt 1):1–13.PubMed
368.
369.
370.
Zurück zum Zitat Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140(6):805–20.PubMed Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140(6):805–20.PubMed
371.
Zurück zum Zitat Bryant CE, et al. Advances in Toll-like receptor biology: modes of activation by diverse stimuli. Crit Rev Biochem Mol Biol. 2015;50(5):359–79.PubMed Bryant CE, et al. Advances in Toll-like receptor biology: modes of activation by diverse stimuli. Crit Rev Biochem Mol Biol. 2015;50(5):359–79.PubMed
372.
Zurück zum Zitat Creagh EM, O’Neill LAJ. TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol. 2006;27(8):352–7.PubMed Creagh EM, O’Neill LAJ. TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol. 2006;27(8):352–7.PubMed
373.
Zurück zum Zitat Gay NJ, et al. A leucine-rich repeat peptide derived from the Drosophila Toll receptor forms extended filaments with a beta-sheet structure. FEBS Lett. 1991;291(1):87–91.PubMed Gay NJ, et al. A leucine-rich repeat peptide derived from the Drosophila Toll receptor forms extended filaments with a beta-sheet structure. FEBS Lett. 1991;291(1):87–91.PubMed
374.
Zurück zum Zitat Wegiel B, Hauser CJ, Otterbein LE. Heme as a danger molecule in pathogen recognition. Free Radic Biol Med. 2015;89:651–61.PubMed Wegiel B, Hauser CJ, Otterbein LE. Heme as a danger molecule in pathogen recognition. Free Radic Biol Med. 2015;89:651–61.PubMed
375.
Zurück zum Zitat Schwandner R, et al. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2. J Biol Chem. 1999;274(25):17406–9.PubMed Schwandner R, et al. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2. J Biol Chem. 1999;274(25):17406–9.PubMed
376.
Zurück zum Zitat Poltorak A, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998;282(5396):2085–8.PubMed Poltorak A, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998;282(5396):2085–8.PubMed
377.
Zurück zum Zitat Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity. 2011;34(5):637–50.PubMed Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity. 2011;34(5):637–50.PubMed
378.
Zurück zum Zitat Yu M, et al. HMGB1 signals through toll-like receptor (TLR) 4 and TLR2. Shock. 2006;26(2):174–9.PubMed Yu M, et al. HMGB1 signals through toll-like receptor (TLR) 4 and TLR2. Shock. 2006;26(2):174–9.PubMed
379.
Zurück zum Zitat Franchi L, et al. Function of Nod-like receptors in microbial recognition and host defense. Immunol Rev. 2009;227(1):106–28.PubMedPubMedCentral Franchi L, et al. Function of Nod-like receptors in microbial recognition and host defense. Immunol Rev. 2009;227(1):106–28.PubMedPubMedCentral
380.
Zurück zum Zitat Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol. 2009;1(6):a001651.PubMedPubMedCentral Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol. 2009;1(6):a001651.PubMedPubMedCentral
381.
Zurück zum Zitat Tang D, et al. Hydrogen peroxide stimulates macrophages and monocytes to actively release HMGB1. J Leukoc Biol. 2007;81(3):741–7.PubMed Tang D, et al. Hydrogen peroxide stimulates macrophages and monocytes to actively release HMGB1. J Leukoc Biol. 2007;81(3):741–7.PubMed
382.
Zurück zum Zitat Hori O, et al. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. J Biol Chem. 1995;270(43):25752–61.PubMed Hori O, et al. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. J Biol Chem. 1995;270(43):25752–61.PubMed
383.
Zurück zum Zitat Hofmann MA, et al. RAGE mediates a novel proinflammatory axis: a central cell surface receptor for S100/calgranulin polypeptides. Cell. 1999;97(7):889–901.PubMed Hofmann MA, et al. RAGE mediates a novel proinflammatory axis: a central cell surface receptor for S100/calgranulin polypeptides. Cell. 1999;97(7):889–901.PubMed
384.
Zurück zum Zitat Du Yan S, et al. Amyloid-beta peptide-receptor for advanced glycation endproduct interaction elicits neuronal expression of macrophage-colony stimulating factor: a proinflammatory pathway in Alzheimer disease. Proc Natl Acad Sci U S A. 1997;94(10):5296–301.PubMedPubMedCentral Du Yan S, et al. Amyloid-beta peptide-receptor for advanced glycation endproduct interaction elicits neuronal expression of macrophage-colony stimulating factor: a proinflammatory pathway in Alzheimer disease. Proc Natl Acad Sci U S A. 1997;94(10):5296–301.PubMedPubMedCentral
385.
Zurück zum Zitat Yan SF, Ramasamy R, Schmidt AM. The receptor for advanced glycation endproducts (RAGE) and cardiovascular disease. Expert Rev Mol Med. 2009;11:e9.PubMedPubMedCentral Yan SF, Ramasamy R, Schmidt AM. The receptor for advanced glycation endproducts (RAGE) and cardiovascular disease. Expert Rev Mol Med. 2009;11:e9.PubMedPubMedCentral
386.
Zurück zum Zitat Chen X, et al. RAGE: a potential target for Abeta-mediated cellular perturbation in Alzheimer’s disease. Curr Mol Med. 2007;7(8):735–42.PubMed Chen X, et al. RAGE: a potential target for Abeta-mediated cellular perturbation in Alzheimer’s disease. Curr Mol Med. 2007;7(8):735–42.PubMed
387.
Zurück zum Zitat Sims GP, et al. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28:367–88.PubMed Sims GP, et al. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28:367–88.PubMed
388.
Zurück zum Zitat Sorci G, et al. RAGE in tissue homeostasis, repair and regeneration. Biochim Biophys Acta. 2013;1833(1):101–9.PubMed Sorci G, et al. RAGE in tissue homeostasis, repair and regeneration. Biochim Biophys Acta. 2013;1833(1):101–9.PubMed
389.
Zurück zum Zitat Kierdorf K, Fritz G. RAGE regulation and signaling in inflammation and beyond. J Leukoc Biol. 2013;94(1):55–68.PubMed Kierdorf K, Fritz G. RAGE regulation and signaling in inflammation and beyond. J Leukoc Biol. 2013;94(1):55–68.PubMed
390.
391.
Zurück zum Zitat Schmidt AM, et al. The multiligand receptor RAGE as a progression factor amplifying immune and inflammatory responses. J Clin Invest. 2001;108(7):949–55.PubMedPubMedCentral Schmidt AM, et al. The multiligand receptor RAGE as a progression factor amplifying immune and inflammatory responses. J Clin Invest. 2001;108(7):949–55.PubMedPubMedCentral
392.
Zurück zum Zitat Bierhaus A, et al. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med (Berl). 2005;83(11):876–86. Bierhaus A, et al. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med (Berl). 2005;83(11):876–86.
393.
Zurück zum Zitat Wu X, et al. The activation of HMGB1 as a progression factor on inflammation response in normal human bronchial epithelial cells through RAGE/JNK/NF-kappaB pathway. Mol Cell Biochem. 2013;380(1–2):249–57.PubMed Wu X, et al. The activation of HMGB1 as a progression factor on inflammation response in normal human bronchial epithelial cells through RAGE/JNK/NF-kappaB pathway. Mol Cell Biochem. 2013;380(1–2):249–57.PubMed
394.
Zurück zum Zitat Chen L, et al. Overexpression of RAGE contributes to cigarette smoke-induced nitric oxide generation in COPD. Lung. 2014;192(2):267–75.PubMed Chen L, et al. Overexpression of RAGE contributes to cigarette smoke-induced nitric oxide generation in COPD. Lung. 2014;192(2):267–75.PubMed
395.
Zurück zum Zitat Bauernfeind F, et al. Inflammasomes: current understanding and open questions. Cell Mol Life Sci. 2011;68(5):765–83.PubMed Bauernfeind F, et al. Inflammasomes: current understanding and open questions. Cell Mol Life Sci. 2011;68(5):765–83.PubMed
396.
Zurück zum Zitat Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10(2):417–26.PubMed Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10(2):417–26.PubMed
397.
Zurück zum Zitat Muruve DA, et al. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature. 2008;452(7183):103–7.PubMed Muruve DA, et al. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature. 2008;452(7183):103–7.PubMed
398.
Zurück zum Zitat Agostini L, et al. NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle–Wells autoinflammatory disorder. Immunity. 2004;20(3):319–25.PubMed Agostini L, et al. NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle–Wells autoinflammatory disorder. Immunity. 2004;20(3):319–25.PubMed
399.
Zurück zum Zitat Arend WP, Palmer G, Gabay C. IL-1, IL-18, and IL-33 families of cytokines. Immunol Rev. 2008;223:20–38.PubMed Arend WP, Palmer G, Gabay C. IL-1, IL-18, and IL-33 families of cytokines. Immunol Rev. 2008;223:20–38.PubMed
400.
Zurück zum Zitat Cerretti DP, et al. Molecular cloning of the interleukin-1 beta converting enzyme. Science. 1992;256(5053):97–100.PubMed Cerretti DP, et al. Molecular cloning of the interleukin-1 beta converting enzyme. Science. 1992;256(5053):97–100.PubMed
401.
Zurück zum Zitat Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13(6):397–411.PubMed Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13(6):397–411.PubMed
402.
Zurück zum Zitat Martinon F, Mayor A, Tschopp J. The inflammasomes: guardians of the body. Annu Rev Immunol. 2009;27:229–65.PubMed Martinon F, Mayor A, Tschopp J. The inflammasomes: guardians of the body. Annu Rev Immunol. 2009;27:229–65.PubMed
403.
Zurück zum Zitat Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009;27(1):519–50.PubMed Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009;27(1):519–50.PubMed
404.
Zurück zum Zitat van de Veerdonk FL, et al. Inflammasome activation and IL-1beta and IL-18 processing during infection. Trends Immunol. 2011;32(3):110–6.PubMed van de Veerdonk FL, et al. Inflammasome activation and IL-1beta and IL-18 processing during infection. Trends Immunol. 2011;32(3):110–6.PubMed
405.
Zurück zum Zitat Rathinam VA, Fitzgerald KA. Inflammasome complexes: emerging mechanisms and effector functions. Cell. 2016;165(4):792–800.PubMedPubMedCentral Rathinam VA, Fitzgerald KA. Inflammasome complexes: emerging mechanisms and effector functions. Cell. 2016;165(4):792–800.PubMedPubMedCentral
406.
Zurück zum Zitat Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821–32.PubMed Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821–32.PubMed
407.
Zurück zum Zitat Vladimer GI, et al. Inflammasomes and host defenses against bacterial infections. Curr Opin Microbiol. 2013;16(1):23–31.PubMedPubMedCentral Vladimer GI, et al. Inflammasomes and host defenses against bacterial infections. Curr Opin Microbiol. 2013;16(1):23–31.PubMedPubMedCentral
408.
Zurück zum Zitat Walle LV, Lamkanfi M. Pyroptosis. Curr Biol. 2016;26(13):R568–72. Walle LV, Lamkanfi M. Pyroptosis. Curr Biol. 2016;26(13):R568–72.
409.
Zurück zum Zitat Hoving JC, Wilson GJ, Brown GD. Signalling C-type lectin receptors, microbial recognition and immunity. Cell Microbiol. 2014;16(2):185–94.PubMedPubMedCentral Hoving JC, Wilson GJ, Brown GD. Signalling C-type lectin receptors, microbial recognition and immunity. Cell Microbiol. 2014;16(2):185–94.PubMedPubMedCentral
410.
411.
Zurück zum Zitat Yamasaki S, et al. Mincle is an ITAM-coupled activating receptor that senses damaged cells. Nat Immunol. 2008;9(10):1179–88.PubMed Yamasaki S, et al. Mincle is an ITAM-coupled activating receptor that senses damaged cells. Nat Immunol. 2008;9(10):1179–88.PubMed
412.
Zurück zum Zitat Zhou H, et al. IRAKM-Mincle axis links cell death to inflammation: pathophysiological implications for chronic alcoholic liver disease. Hepatology. 2016;64(6):1978–93.PubMed Zhou H, et al. IRAKM-Mincle axis links cell death to inflammation: pathophysiological implications for chronic alcoholic liver disease. Hepatology. 2016;64(6):1978–93.PubMed
Metadaten
Titel
Damage-associated molecular patterns in trauma
verfasst von
Borna Relja
Walter Gottlieb Land
Publikationsdatum
14.10.2019
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Trauma and Emergency Surgery / Ausgabe 4/2020
Print ISSN: 1863-9933
Elektronische ISSN: 1863-9941
DOI
https://doi.org/10.1007/s00068-019-01235-w

Weitere Artikel der Ausgabe 4/2020

European Journal of Trauma and Emergency Surgery 4/2020 Zur Ausgabe

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Update Orthopädie und Unfallchirurgie

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