Next Article in Journal
Blood–Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in Aging
Next Article in Special Issue
Tissue Engineering Approaches to Uncover Therapeutic Targets for Endothelial Dysfunction in Pathological Microenvironments
Previous Article in Journal
Novel Epoxides of Soloxolone Methyl: An Effect of the Formation of Oxirane Ring and Stereoisomerism on Cytotoxic Profile, Anti-Metastatic and Anti-Inflammatory Activities In Vitro and In Vivo
Previous Article in Special Issue
Hemin-Induced Endothelial Dysfunction and Endothelial to Mesenchymal Transition in the Pathogenesis of Pulmonary Hypertension Due to Chronic Hemolysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Exploratory Investigation of the Plasma Proteome Associated with the Endotheliopathy of Trauma

1
Center for Translational Injury Research, Department of Surgery, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030, USA
2
Center for Clinical Research and Evidence-Based Medicine, Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
3
Center for Endotheliomics CAG, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, 2200 Copenhagen, Denmark
4
Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Lyngby, Denmark
5
Center for Systems Biology, University of Iceland, 101 Reykjavik, Iceland
6
Center for Endotheliomics CAG, Department of Clinical Immunology, Rigshospitalet, & Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(11), 6213; https://doi.org/10.3390/ijms23116213
Submission received: 29 April 2022 / Revised: 27 May 2022 / Accepted: 30 May 2022 / Published: 1 June 2022
(This article belongs to the Special Issue Molecular Mechanisms of Endothelial Dysfunction)

Abstract

:
Background: The endotheliopathy of trauma (EoT) is associated with increased mortality following injury. Herein, we describe the plasma proteome related to EoT in order to provide insight into the role of the endothelium within the systemic response to trauma. Methods: 99 subjects requiring the highest level of trauma activation were included in the study. Enzyme-linked immunosorbent assays of endothelial and catecholamine biomarkers were performed on admission plasma samples, as well as untargeted proteome quantification utilizing high-performance liquid chromatography and tandem mass spectrometry. Results: Plasma endothelial and catecholamine biomarker abundance was elevated in EoT. Patients with EoT (n = 62) had an increased incidence of death within 24 h at 21% compared to 3% for non-EoT (n = 37). Proteomic analysis revealed that 52 out of 290 proteins were differentially expressed between the EoT and non-EoT groups. These proteins are involved in endothelial activation, coagulation, inflammation, and oxidative stress, and include known damage-associated molecular patterns (DAMPs) and intracellular proteins specific to several organs. Conclusions: We report a proteomic profile of EoT suggestive of a surge of DAMPs and inflammation driving nonspecific activation of the endothelial, coagulation, and complement systems with subsequent end-organ damage and poor clinical outcome. These findings support the utility of EoT as an index of cellular injury and delineate protein candidates for therapeutic intervention.

1. Introduction

Unintentional injury remains the third leading cause of death in the United States [1] and persists as the leading cause of mortality for individuals aged one to 44 years old [2]. The economic burden generated by trauma exceeds 189 billion dollars annually [2]. While there have been some advancements in the mitigation of early mortality in traumatic injury, progress has proven to be more difficult in improving long-term survival [3].
The endotheliopathy of trauma (EoT), a form of shock-induced endotheliopathy (SHINE), has been postulated to represent a major component driving outcomes in trauma [4,5,6]. Endotheliopathy signifies dysfunction of the endothelium as well as disruption of its overlying glycocalyx layer. The vascular endothelium is responsible for maintaining the permeability barrier, in part through structural components including adherens junctions [7]. The glycocalyx is composed of proteoglycans and glycoproteins fixed to the endothelial cell surface which serve as a scaffold for soluble proteins and other soluble components [8]. The unique composition of these glycosaminoglycans dictates interactions with extracellular ligands and modulates systemic inflammatory and coagulation profiles [8,9,10]. Some consider this intricate relationship between blood and the endothelium an organ system unto itself [7,11]. Endotheliopathy represents a contributor to ongoing physiologic dysfunction common to all forms of shock and a target for therapeutic intervention [6].
Circulating biomarkers of endotheliopathy have been utilized to reveal an inextricable link between the condition of the endothelium, the glycocalyx, and the physiology of the trauma patient. Trauma-induced endotheliopathy has been implicated in permeability barrier attenuation [12,13,14], sympathoadrenal hyperactivation [15,16,17,18,19,20], coagulopathy [17,21,22,23,24,25], hypoxia and metabolic derangement [17,19,26]. Endothelial dysfunction following injury has been associated with increased injury severity [17,19], multiple organ failure [26], increased intensive care unit stay [17,18], and increased mortality [17,18,19,27]. Despite these associations, the underlying etiology and clinical utility of endotheliopathy in trauma have not yet been fully elucidated. Proteomic profiling provides a more comprehensive representation of the systemic response to injury [28,29]. Herein, we investigate the plasma proteome of EoT in order to gain new insight into the molecular mechanisms underlying endothelial and glycocalyx dysfunction and to identify new targets for therapeutic intervention.

2. Methods

2.1. Patient Population

Patients presenting to the emergency department requiring the highest level of trauma activation between March 2012 and February 2018 at Hermann Memorial Hospital, a level-1 trauma center in Houston, Texas, were included in an ongoing prospective universal protocol and considered eligible for the study. The highest level of trauma activation requires at least one of the following: systolic blood pressure < 90 mm Hg, heart rate > 120 beats per minute, respiratory rate > 29 or <10 breaths per minute, Glasgow coma score < 10, intubation, prehospital blood product administration, uncontrolled external hemorrhage, penetrating injury to the head, neck, torso or groin, amputation proximal to the wrist or ankle, paraplegia, quadriplegia, pelvic fracture or two or more long bone fractures. Study subjects were selected retrospectively from this universal cohort by stratified randomization based on injury severity score and mechanism of injury to achieve a balanced patient population for this exploratory analysis. Exclusions included prisoners, pregnant patients, those less than 16 years of age, and those who declined to consent to the study. Traumatic brain injury (TBI) influences patient outcomes through mechanisms that are not necessarily related to endotheliopathy within the systemic vasculature. For this reason, subjects with a head abbreviated injury score > 2 representing moderate or severe traumatic brain injury were excluded, including TBI subjects with multi-trauma. Patient demographics, clinical information, and outcome data were obtained from the institutional trauma registry or medical record extraction by research personnel. Consent from patients or legally authorized representatives was obtained within 72 h of admission. A waiver of consent was procured for patients who expired, were discharged within 24 h of admission, or were lacking decision-making capacity without a surrogate decision-maker following at least three attempts to acquire consent over 72 h. This prospective, observational study was approved by the McGovern Medical School at UTHealth institutional review board (HSC-GEN-12-0059). Study subjects were randomly selected retrospectively based on injury severity score and mechanism of injury.

2.2. Sample Collection and Processing

Admission blood samples were collected for each patient upon emergency department arrival in 3.2% sodium citrate and ethylenediaminetetraacetic acid (EDTA) tubes and inverted several times. Samples were centrifuged at 1800× g at 5 °C for 10 min twice. The plasma layer was isolated, aliquoted, and stored at −80 °C.

2.3. Enzyme-Linked Immunosorbent Assay (ELISA)

The soluble biomarkers of sympathoadrenal activation (epinephrine and norepinephrine), endothelial glycocalyx shedding (Syn-1), and endothelial damage (soluble thrombomodulin; sTM) were measured by immunoassays according to the manufacturer’s recommendation (2-CAT ELISA, Labor Diagnostica Nord GmbH & Co. KG, Nordhorn, Germany and Diaclone Nordic Biosite, Copenhagen, Denmark) using citrate plasma.

2.4. High Performance Liquid Chromatography and Tandem Mass Spectrometry (MS)

2.4.1. Sample Preparation and Digestion

Sample preparation and digestion were performed without a high abundance protein depletion step using EDTA plasma according to methods previously published by Kulak et al. [30]. Briefly, plasma proteins were denatured using 20 µL of buffer consisting of 6 M guanidinium hydrochloride, 10 mM TCEP (Tris(2-carboxyethyl)phosphine hydrochloride; Sigma), 40 mM CAA (40 mM 2-Chloroacetamide; Sigma, Burlington, MA, USA), and 50 mM HEPES (N-(2-Hydroxyethyl)piperazine-N′-(2-ethanesulfonic acid), 4-(2-Hydroxyethyl)piperazine-1-ethanesulfonic acid; Sigma) at pH 8.5. Samples were then boiled at 95 °C for 5 min, and subsequently sonicated on high for 30 s five times in a Bioruptor sonication water bath (Diagenode, Denville, NJ, USA) at 4 °C. Protein concentration was determined by a Bradford assay (Sigma). Protein digestion was then performed with 5 µg of protein. Samples were diluted 1:3 in a solution consisting of digestion buffer (10% acetonitrile and 50 mM HEPES at pH 8.5) and Endoproteinase Lys-C (MS grade; Wako, Neuss, Germany) was added in a 1:50 enzyme-to-protein ratio. Samples were incubated at 37 °C for 4 h. A subsequent 1:10 dilution was completed with digestion buffer (10% acetonitrile and 50 mM HEPES at pH 8.5) and a 1:100 enzyme-to-protein ratio of trypsin (MS grade; Promega, Madison, WI, USA) was added. Samples were then incubated overnight at 37 °C. Enzyme activity was quenched with the addition of 2% trifluoroacetic acid (TFA) to create a final concentration of 1%. Subsequently, 500 ng of sample was loaded onto EvoSep stagetips according to the manufacturer’s protocol.

2.4.2. Data Acquisition

Samples underwent protein analysis utilizing an EvoSep One instrument on the default “30 samples per day” setting. A Q-Exactive HF-X instrument (ThermoFisher Scientific, Waltham, MA, USA) with a DD-MS2 top28 method was used for analysis following protein elution over a 44 min gradient. A resolution of 60,000 with an automatic gain control (AGC) target of 3 × 106 or a maximum injection time of 50 ms and a scan range of 350–1850 m/z was used for the recording of the full mass spectrometry (MS) spectra. A resolution of 15,000 with an AGC target value of 1 × 105 or a maximum injection time of 22 ms with a normalized collision energy of 28 and an intensity threshold of 2 × 104 was used for MS2 spectra acquisition. Exclusions included ions with a charge state of <2, >6, and unknown with a dynamic exclusion at 30 s. MS consistency was verified using complex cell lysate quality control standards and chromatography was evaluated for reproducibility.

2.4.3. Protein Quantitation

Raw MS data were analyzed using Proteome Discoverer 2.2. Processing and consensus analysis was completed with label-free quantitation. Spectra protein matching was completed using UniProt and the 9606 Human database. Dynamic modifications included oxidation (methionine), deamidation (asparagine, glutamine), and acetyl groups located on N-termini. Cysteine carbamidomethyl was considered a static modification. Protein quantification was completed using the built-in Minora Feature Detector with a false discovery rate (FDR) filter of 1% and a built-in T-test to evaluate statistical significance.

2.5. Statistical Analysis

2.5.1. Clinical Variables and ELISA

EoT was defined as a plasma Syn-1 concentration of ≥40 ng/mL [17,18,31,32]. Comparison of EoT vs non-EoT groups for clinical variables and ELISA quantification consisted of a Wilcoxon rank-sum test for continuous variables, a Fisher exact test for categorical variables, and an Armitage trend test for ordinal variables.

2.5.2. MS Protein Analysis

Following MS protein quantification at an FDR threshold of 0.01, proteins with excessive missing values (>30%) were excluded, as were proteins with ≥10 extreme outliers (i.e., values above or below Q 3 ± 3 × Q 3 Q 1 , where Q 1 and Q 3 are the first and third quartile, respectively). Patients without adequate plasma samples for proteomic analysis were excluded. Values below a detection limit (initially recorded as 0) were replaced by 10% of the smallest non-zero value of each protein. After applying a l o g 2 -transformation, the values of each protein were then standardized (centered at 0 with variance 1).

2.5.3. Identification of Differentially Expressed Proteins

Identification of differentially expressed proteins between EoT and non-EoT groups was initiated with a two-sided t-test to obtain a test statistic and a raw p-value of each protein. To account for high dimensional data, the raw p-values were then adjusted using the method of Benjamini and Hochberg at 5% to control the false discovery rate [33]. Fold change was calculated as log2 of the ratio of a mean protein level in the EoT group to that of the non-EoT group. Analysis of differentially expressed proteins was completed using the QIAGEN Ingenuity Pathway Analysis (IPA) database (https://www.qiagenbioinformatics.com/products/ingenuity-pathway-analysis; accessed on 1 November 2021).

2.5.4. Identification of Protein Predictors of EoT

To identify a set of proteins that best predict EoT, we performed elastic-net regularized logistic regression using the glmnet package in R (version 3.6.2). In the regression model, there are two user-defined hyper-parameters: elastic-net penalty, α , and tuning parameter, λ [34]. We chose an α that produced the highest prediction accuracy and a λ that provided the most regularized model such that the error is within one standard error of the minimum (usually denoted by lambda.1se). The optimal value of λ was obtained from leave-one-out cross-validation (LOOCV). To account for the relatively small sample size a bootstrap procedure was executed to obtain optimism-corrected performance metrics [35]. For details, see Section 5.3.4 of Clinical Prediction Models by Steyerberg and Ewout, 2019 [36]. Performance metrics included area under the curve (AUC), prediction accuracy, Brier scores, sensitivity, and specificity. No covariate for adjustment was considered. Proteins with non-zero coefficients were reported as strong predictors.

2.5.5. Identification of Intracellular Proteins Associated with End-Organ Damage

Intracellular proteins differentially expressed between EoT and non-EoT groups and expressed within organs of interest were evaluated. Correlation coefficients were generated between MS-derived plasma protein abundance for proteins preferentially expressed in the liver and serum liver aminotransferases. An analysis of variance test was used to evaluate associations between MS-derived proteins primarily expressed in the kidney and acute kidney injury development. Acute kidney injury was defined by stage 2 or 3 Kidney Disease Improving Global Outcomes (KDIGO) criteria using a Modification of Diet in Renal Disease-derived reference for creatinine described previously by Hatton et al. [37].

3. Results

3.1. Clinical Characteristics

One subject was excluded for inadequate plasma sample, leaving 99 subjects for study inclusion. The EoT group consisted of 62 subjects with plasma Syn-1 ≥ 40 ng/mL, while the 37 remaining subjects were considered non-EoT. Patient demographic information, clinical data, and laboratory values are summarized in Table 1. EoT subjects were more severely injured, had more severe chest and abdominal injuries, were less likely to have a mechanism of injury related to burns, and were more likely to have blunt and penetrating trauma compared to non-EoT subjects. The EoT group was associated with a lower Glasgow coma scale score and systolic blood pressure on emergency department arrival. EoT patients had increased serum creatinine and glucose concentrations, increased base deficit, and decreased pH. Thromboelastography (TEG) demonstrated associations between EoT and increased activated clotting time, increased K-time, decreased α-angle, decreased maximum amplitude, and decreased G-value. EoT subjects required increased blood product transfusion. The EoT group had increased mortality within 72 h of admission; however, there was no statistically significant difference in overall in-hospital mortality between those with and without EoT. EoT patients were more likely to develop pneumonia and pulmonary embolism.

3.2. ELISA

ELISA data are summarized in Table 2. Plasma epinephrine, norepinephrine, and sTM concentrations were increased in the EoT group.

3.3. MS Proteomics

Six hundred and five proteins were identified by MS for 99 trauma subjects with sufficient plasma samples. Four hundred and seventy-six proteins qualified as high confidence reads with an FDR < 0.01. The remaining 129 proteins were excluded. One hundred and seventy-eight proteins with missing values for >30% of subjects were omitted leaving 298 acceptable proteins. Eight proteins with >10 extreme outliers were then excluded resulting in 290 proteins for subsequent analysis. Fifty-two differentially expressed proteins between EoT and non-EoT groups were identified using a Benjamini and Hochberg FDR-adjusted p-value at 5%, including 39 upregulated proteins and 13 downregulated proteins associated with EoT. This information is summarized in Figure 1 and Table 3. The top ten proteins out of 42 identified by elastic-net regularized logistic regression with bootstrapping that best predict EoT in descending order included ACTC1, ADH1B, GSTA1, C4A, S100A9, HIST1H4A, ACTBL2, SERPINF2, CAT, and CFH. The regression model produced a prediction accuracy of 0.71 (95% CI: 0.62–0.78), a sensitivity of 0.81, a specificity of 0.70, and an area under the curve of 0.84 (95% CI: 0.80–0.89). Within the 52 proteins differentially expressed in the EoT group, ADH1A, MAT1A, BHMT, HPD, and FABP1 were selected for further analysis based on intracellular hepatocyte expression [38,39]. Serum liver transaminase data within three days of admission were available for 51 of the 99 patients. Each of the five hepatocyte proteins displayed statistically significant positive correlation coefficients between plasma protein abundance and serum aspartate transaminase (AST) and alanine aminotransferase (ALT) concentration, see Table 4. BHMT, HPD, and FABP1 were selected based on known intracellular kidney abundance [38,39]. None of these proteins demonstrated a statistically significant association with acute kidney injury defined by KDIGO criteria, see Table 5.

4. Discussion

Associations between EoT and clinical characteristics are consistent with previous studies for many parameters, including increased injury severity [17,18], hypotension [14,18], increased base deficit [17], hypocoaguable TEG profile [17], increased blood transfusion requirement [17,18], and increased early mortality [17,18]. A statistically significant increase in early death was observed in the EoT group, but not in overall in-hospital mortality. The majority of the burn patients were within the non-EoT group, many of whom succumbed to their injuries later in their hospital course which may contribute to this finding. The ELISA data is in agreement with previously reported associations between endothelial activation and sympathoadrenal hyperactivation, as increased serum concentrations of sTM [14,17,18], epinephrine [17], and norepinephrine [17] were observed in the EoT group. The 39 upregulated and 13 downregulated proteins that differentiate EoT from non-EoT have several significant clinical correlations.

4.1. Damage Associated Molecular Patterns

Four prominent damage-associated molecular pattern (DAMP) proteins were found to have elevated concentrations in the EoT group, including S100A8, S100A9, HIST1H4A, and PPIA. Protein S100A8 and protein S100A9, also known as myeloid-related protein-8 (MRP8) and myeloid-related protein-14 (MRP14), respectively, as well as the S100A8/A9 heterodimer known as calprotectin, represent mediators of acute inflammation [40]. When secreted these proteins induce leukocyte activation and chemotaxis through toll-like receptor 4 (TLR4) and receptor for advanced glycation end products (RAGE) pattern recognition receptors [40]. These pleiotropic proteins possess anti-inflammatory properties as well which may be suppressed by oxidative stress [41,42,43,44,45,46,47,48].
Histone H4 (HIST1H4A) is a regulator of chromatin packing and gene expression within cell nuclei [49]. However, this protein functions as a potent alarmin when introduced into circulation following cellular damage [50]. Histones induce cytokine release through interactions with toll-like receptors [51,52,53]. Extracellular histones represent the predominant contributor of cytotoxicity within neutrophil extracellular traps (NETs) [54], a network of molecules with thrombotic and antimicrobial functions involved in the innate immune response [55]. Glycocalyx constituents, including glycosaminoglycans and heparin, inhibit the cytotoxic effects of extracellular histones [56,57]. Circulating inter-α-trypsin inhibitor (ITIH1) is involved in histone sequestration in states of acute inflammation [58]. We find in the current EoT cohort a diminished abundance of circulating ITIH1 paired with increased plasma histone H4. Elevated circulating histone H4 specifically has been observed in trauma subjects previously [59,60], and among all histone components, H4 is considered the most cytotoxic and the greatest contributor to fibrinolysis [61,62].
Cyclophilin A, also known as peptidyl-prolyl cis-trans isomerase A, CYPA, or PPIA, is a ubiquitous intracellular protein that is released into circulation by vascular smooth muscle cells in response to oxidative stress [63]. This protein can also be released by endothelial cells [64], platelets [65], and macrophages [66]. PPIA promotes cytokine release through interactions with cell surface receptor CD147 [67].
A summary of previously established associations between these DAMPs and pathophysiology relevant to the systemic response to trauma is summarized in Table 6. DAMPs represent a potential driver of ongoing endothelial injury and activation, as well as attenuation of the permeability barrier. In addition to the physiologic challenges associated with hyper-permeable vessels, these proteins contribute to organ injury through direct cytotoxic effects and microvascular thrombosis. This EoT phenotype likely feeds self-amplifying loops involving sympathoadrenal hyperactivation and cytokine release [68]. DAMPs have been implicated in the etiology of immune failure and immune-mediated host injury in trauma subjects [69,70]. A Danger Model has been proposed based on the assertion that tissue damage governs activation of the immune system, rather than an innate sense of self versus non-self [71,72,73]. DAMPs likely represent the source of immune-mediated host injury and attenuated host immune defense in a manner analogous to the nonspecific activation and subsequent exhaustion of coagulation factors in the acute coagulopathy of trauma. DAMP-induced damage can, in turn, generate DAMPs and precipitate self-perpetuating physiologic disarray in injured patients. A variety of potential pharmacological therapeutics exist with the potential to arrest these DAMP-related pathophysiological processes in trauma [54,74,75,76,77,78,79,80,81,82,83,84,85,86].

4.2. End-Organ Damage

Several proteins demonstrating an increased abundance in the EoT group are suggestive of end-organ damage. Actin α cardiac muscle-1 (ACTC1) represents the single best predictor of EoT of the 290 MS-derived proteins determined by elastic net regularized regression. ACTC1 is typically an intracellular protein and represents the major form of actin within cardiac sarcomeres [38,161,162,163], although it may also be found in smooth muscle and skeletal muscle [39,164,165,166]. Elevated serum ACTC1 has been associated with endotoxemia, potentially due to sarcomere disruption [167]. There is little data available regarding the molecular and clinical significance of circulating ACTC1. However, its characteristics can be inferred based on similarities to comparable proteins including troponin and other subtypes of actin. Circulating α-actin has been utilized as an early biomarker reflecting cardiac damage secondary to myocardial infarction in a manner similar to troponin [168]. Elevated circulating troponin has been used to diagnose secondary cardiac injury without related thoracic injury in trauma cohorts, even in younger patients unlikely to have had pre-injury cardiac disease [169]. This trauma-induced secondary cardiac injury has been associated with mortality [169,170,171]. The etiology underlying cardiac impairment is believed to involve DAMP and pro-inflammatory mediators as well as catecholamine surge resulting in cardiomyocyte cytotoxicity and dysfunction [169,172,173,174]. Histologic changes in cardiomyocytes identified in trauma patients upon autopsy include congestion, interstitial edema, and myofibril degeneration [175,176]. Similar sarcomere disorganization has been observed in murine models of hemorrhagic shock [177].
In the present study an association between increased plasma concentration of DAMPs, epinephrine, norepinephrine, and EoT is observed. Taken together, the elevated abundance of ACTC1 may reflect DAMP and catecholamine-induced cardiomyocyte damage with subsequent leakage of myofilament components into plasma. The hypotension and metabolic acidosis associated with EoT may signify increased cardiac demand and heart strain contributing to the release of ACTC1 into circulation. In addition to representing a biomarker for cardiac damage, ACTC1 itself may contribute to poor outcomes in EoT. Circulating actin is cytotoxic to endothelial cells [178], impairs clearance of cytotoxic cell-free circulating DNA [179,180], promotes coagulopathy [181], and disrupts host defenses [182]. Exhaustion of the actin-scavenger system is a risk factor for mortality in trauma [183,184].
A statistically significant increase in pulmonary embolism (PE) and pneumonia was observed in the EoT group. Contemporary guidelines recommend pharmaceutical venous thromboembolism (VTE) prophylaxis in the majority of moderate to high-risk trauma patients [185]. Even with perfect adherence to prophylaxis guidelines breakthrough VTE following injury persists [186,187], particularly in those with severe injuries [188]. Breakthrough cases of PE in trauma appear to have unique characteristics and an etiology distinct from other forms of VTE. These blood clots are more likely to arise in the segmental and sub-segmental vasculature [189] and appear to arise de novo within the lung rather than from proximal pelvic vein embolization [189,190,191]. Several EoT-associated DAMPs are cytotoxic to the pulmonary endothelium and capable of inciting microthrombosis within the pulmonary vasculature. These proteins may be contributing to the etiology of breakthrough PE, as well as pneumonia, within the EoT group. There was no statistically significant difference in respiratory failure or ARDS development between the EoT and non-EoT groups. This may be related to insufficient sample size and censoring secondary to increased early mortality in the EoT group. Of note, both elevated ACTC1 and increased incidence of PE could be related to direct injury to the heart and lungs, respectively, as the EoT group demonstrated increased chest abbreviated injury score compared to non-EoT subjects.
An increased abundance of plasma fatty-acid binding protein 1 (liver type; FABP1) was observed in the EoT group. This protein is expressed in the intestines, liver, and kidney [38,39,192], and has many intracellular functions including antioxidant defense [193]. Elevated circulating FABP1 is associated with severe abdominal injury and acute renal failure in trauma subjects [194,195]. Increased plasma FABP1 may result from direct organ damage or ischemia [196,197,198,199,200]. The EoT group was associated with increased abdominal abbreviated injury score and increased blood transfusion requirement, suggesting both direct abdominal organ injury and hypoperfusion. Plasma FABP1 is positively correlated with serum transaminases indicating FABP1 leakage from hepatocytes. However, this does not preclude additional concurrent plasma FABP1 contributions from the kidney and the intestines. FABP1 possesses heme-binding and antioxidant properties [193], characteristics that may provide some benefit to the acutely injured trauma patient. This may represent an evolved adaptive response to oxidative damage in severe injury as the superoxide dismutase SOD1, a primarily intracellular antioxidant [201], is also elevated in the plasma of EoT subjects.
The EoT group was associated with increased plasma concentrations of four additional plasma proteins that are primarily expressed in hepatocytes and renal cells. ADH1A (alcohol dehydrogenase 1A) and MAT1A (methionine adenosyltransferase 1A) are preferentially expressed in the liver [38,39]. BHMT (betaine-homocysteine S-methyltransferase) and HPD (4-hydroxyphenylpyruvate dioxygenase) are principally expressed in both the liver and kidneys [38,39]. ADH1A, MAT1A, BHMT, and HPD each demonstrated a statistically significant positive correlation coefficient with serum AST and ALT concentration. There was no statistically significant association observed between BHMT or HPD and acute kidney injury. These findings suggest an association between liver injury, leakage of hepatocyte proteins into circulation, and EoT.
Elevated plasma BHMT may indicate underlying metabolic processes related to EoT. Increased abundance of intracellular liver proteins in circulating plasma is likely a function of both hepatocyte leakage and hepatocyte intracellular protein concentration. BHMT is a regulator of homocysteine metabolism and methionine biosynthesis through the facilitation of betaine methyl group donation [202]. BHMT expression is upregulated by the amino acid taurine [203], an osmolyte with important physiologic functions involving resistance to osmotic stress and maintenance of microvascular flow [204,205,206,207]. Increased plasma BHMT in the EoT group may reflect increased taurine abundance within hepatocytes with subsequent BHMT upregulation in addition to liver damage. This finding may denote a compensatory metabolic response in EoT subjects.

4.3. Coagulation

Coagulation factor V (FV) was the only coagulation factor with a statistically significant decrease in plasma abundance in EoT compared to non-EoT. EoT was associated with a hypocoaguable TEG profile, consistent with previous findings demonstrating an association between lower levels of circulating FV and acute coagulopathy following trauma [208,209,210,211,212,213,214]. A proportion of plasma FV undergoes endocytosis and is sequestered within platelet alpha granules [215,216,217,218], and this intra-platelet FV is integral for achieving hemostasis following traumatic hemorrhage [219,220]. The factor V Leiden (FVL) mutation results in an FV phenotype that is resistant to proteolysis by activated protein C (APC) [221] and represents the most common inherited form of thrombophilia in Caucasians [222]. The high frequency of this polymorphism is believed to be due to a conferred evolutionary advantage secondary to improved hemostasis in states of hemorrhage, including trauma [223,224,225]. This theory is supported by observations of decreased peri-partum bleeding in FVL carriers [226,227,228]. The effect of FVL on bleeding and nonbleeding traumatic injury is unknown. The influence of FVL on the function of APC, which is involved in many processes relevant to trauma including the degradation of extracellular histones [52,53,54,92], is not fully elucidated. However, FV may be the only coagulation factor deficit that can be identified at the scene of injury [209], is the most consistent coagulation factor deficiency observed on trauma admission blood samples [211], and regulators of FV abundance appear to be distinct from those involved in other coagulation proteins [213]. It may not be a coincidence that the strongest genotype candidate for improved outcomes in traumatic bleeding involves conservation of FV, the same coagulation factor deficiency that is most directly tied to the acute coagulopathy of trauma and endotheliopathy. Associations between EoT and increased blood transfusion requirements, decreased plasma FV, coagulopathy, and elevated plasma DAMPs reinforce the suspicion that FV may serve an important role in the systemic response to trauma.
We report decreased plasma concentration of α-2-plasmin inhibitor (SERPINF2; AAP) and plasminogen activator inhibitor (SERPINA5; PAI-1) in the EoT group. These proteins represent inhibitors of fibrinolysis and depletion of circulating AAP and PAI-1 is associated with the acute coagulopathy of trauma [229], and hyperfibrinolysis [230,231,232,233]. The primary driver of trauma-related hyperfibrinolysis is postulated to be insufficient PAI-1 concentration relative to increased tissue plasminogen activator (tPA) release by the microvascular endothelium [234]. Decreased AAP and PAI-1 indicates fibrinolysis within the endotheliopathy group despite the lack of an observed difference in TEG clot lysis between EoT and non-EoT in the present study. Varying degrees of fibrinolysis exist within trauma subjects [233], and a spectrum of coagulation phenotypes may be present within EoT as well. Despite this potential heterogeneity within the EoT group, decreased inhibitors of fibrinolysis and a hypocoaguable coagulation profile are suggestive of consumptive coagulopathy. These findings may represent widespread microthrombosis provoked by endothelial injury and DAMPs. Of note, a previous study has failed to identify a significant association between PAI-1 level and syndecan-1 shedding [27]. However, these findings were based on a higher plasma syndecan-1 concentration cutoff and a smaller sample size.
Decreased plasma high-molecular-weight kininogen (HMWK; KNG1) was associated with the EoT group. HMWK maintains an antithrombotic endothelial cell surface and patent blood flow within the microvasculature [235,236,237,238]. HMWK cleavage by factor XII (Hageman Factor) initiates the coagulation cascade and conversion of prekallikrein to kallikrein, subsequent release of bradykinin, and increased permeability and vasodilation [238,239,240]. This process can be incited by complement, neutrophils, and tPA [235,241,242]. Glycosaminoglycans on the endothelial surface preserve HMWK, and degradation of the glycocalyx results in HMWK proteolysis and bradykinin-mediated attenuation of the permeability barrier [243]. Consumption of HMWK indicates thrombosis formation and increased permeability secondary to glycocalyx deterioration.
Hemopexin (HPX) is a circulating free heme carrier protein that neutralizes oxidative damage and transports heme to the liver [244,245]. Hemopexin reserve can be diminished following traumatic hemorrhage and blood product resuscitation, and depletion is associated with increased vascular permeability and susceptibility to infection [246]. Decreased hemopexin abundance implies red blood cell lysis within the EoT group and is likely related to consumptive coagulopathy and red blood cell transfusion.

4.4. Complement

In the EoT group, we find decreased abundance of complement inhibitor CFH (complement factor H), complement activator FCN3 (ficolin-3), and complement components C8A, C8B, C8G, C4A, and C4B. CFH binds heparin and glycosaminoglycan linkages on endothelial cell surfaces and supports endothelial integrity by suppression of nonspecific alternative complement activation [247,248]. Mutations in the endothelial binding domain of CFH can manifest as an atypical hemolytic uremic syndrome (aHUS) resulting in alternative complement pathway-induced endothelial damage [248]. Endotheliopathy is believed to serve as the initiator of aHUS by provoking arteriole and capillary wall thickening, intraluminal platelet activation, microthrombosis, vessel occlusion, and end-organ damage [249]. Factor H provides specificity to alternative complement activation [250], and a deficiency of this protein within the EoT group may allow for unregulated complement-induced cytotoxicity of host endothelium and a clinical phenotype similar to aHUS.
Ficolin-3 is a known activator of the lectin complement pathway [251]. Cleavage of C4 into C4a and C4b is involved in the initiation of the classic and lectin complement pathways [252], while C8 is a constituent of the terminal complement complex. C4a is a potent anaphylatoxin capable of inducing endothelial activation [253], as is the terminal complement complex [254,255,256,257,258,259,260]. Complement activation following trauma has been associated with increased injury severity and red blood cell and platelet dysfunction [261,262,263].
Reduced plasma abundance of these complement components within EoT subjects, particularly the deficiency of CFH, is suggestive of nonspecific complement activation, widespread deposition of complement components into host endothelium, and subsequent exhaustion of the complement system. This process likely contributes to increased endothelial permeability, coagulopathy, and end-organ damage.

4.5. Limitations

The limitations of this study include those inherent to MS methodology, as no single proteomic assay can identify all proteins within a plasma sample. For this reason, proteins significant to EoT may not be included in this analysis, particularly low-abundance plasma proteins which can be better evaluated through a high abundance protein depletion step prior to MS. This data set is restricted by sample size which was not sufficient to accurately identify subgroups within EoT subjects. Evaluation of sub-phenotypes within EoT may allow for a more personalized approach in trauma management, as this strategy has shown promise in similar heterogeneous pathologies including ARDS [264,265]. Further, some relevant proteins may not yield significant associations with EoT due to type II error. Finally, we did not normalize total protein concentration to control for hemodilution. This method has been utilized in similar proteomic studies and may provide distinct information [159].

5. Conclusions

The clinical significance of endothelial permeability has been debated since early dog models of hemorrhagic shock in the 1940s [266,267]. Shedding of the glycocalyx does not imply endothelial cell death, but rather reflects endothelial activation and increased permeability [13,31]. This is an important distinction, as these dysfunctional living cells represent an opportunity for intervention that would not exist in nonviable endothelial cells. Within the proteomic profile of EoT, we present several DAMPs that contribute to endothelial activation, coagulopathy, and host tissue destruction. We report signs of non-specific coagulation and complement activation, as well as the exhaustion of vital regulatory components within these systems. We describe circulating intracellular proteins suggestive of end-organ damage. The proteomic profile of EoT reflects mechanisms evolutionarily intended for local tissue injury amplified to a pathologic level. This secondary injury represents a perpetual source of damage in itself and current management practices in trauma largely allow this process to continue unabated. The key to improving long-term outcomes in trauma depends on the mitigation of this secondary traumatic insult which may require a multimodal approach. Herein, we have identified proteins and pathways that are likely to be driving the pathologic processes underlying EoT which serve as targets for pharmacological therapeutics.

Author Contributions

Conceptualization, J.D.K., P.I.J. and C.E.W.; methodology, J.D.K. and E.M.S.; software, n/a; validation, E.M.S.; formal analysis, J.D.K., K.H.L. and C.P.; investigation, E.M.S.; resources, J.D.K., K.H.L., H.H.H., Y.-W.W.W., E.M.S., S.T.K., Ó.R., P.I.J., C.P. and C.E.W.; data curation, J.D.K., H.H.H., E.M.S., P.I.J. and C.E.W.; writing—original draft preparation, J.D.K., P.I.J. and C.E.W.; writing—review and editing, J.D.K., K.H.L., H.H.H., Y.-W.W.W., E.M.S., S.T.K., Ó.R., P.I.J., C.P. and C.E.W.; visualization, K.H.L., C.P. and J.D.K.; supervision, C.E.W. and P.I.J.; project administration, C.E.W. and P.I.J.; funding acquisition, C.E.W. and P.I.J. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institute of General Medical Sciences of the NIH (5T32GM008792). This project received funding from the William Stamps Farish Fund, the Howell Family Foundation, and the James H. “Red” Duke Professorship.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board of McGovern Medical School at UTHealth (HSC-GEN-12-0059), originally approved on 22 March 2012 and subsequently regularly reviewed.

Informed Consent Statement

Informed consent or a waiver of informed consent was obtained from all subjects involved in the study. Consent from patients or legally authorized representatives was obtained within 72 h of admission. A waiver of consent was procured for patients who expired, were discharged within 24 h of admission, or were lacking decision-making capacity without a surrogate decision-maker following at least three attempts to acquire consent over 72 h.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author, J.D.K., upon reasonable request.

Acknowledgments

The authors thank Veda Pa and Erin Fox for their assistance in clinical data acquisition, as well as Kimberly Mankiewicz for reviewing the manuscript.

Conflicts of Interest

The authors declare no conflict of interest. Dr. Wade holds a financial interest in Decisio Health, Inc. (3900 Essex Ln Suite 600, Houston, TX 77027, USA), is a consultant to CellPhire, and has patents licensed through the University of Texas to Decisio Health and Grifols. None of these are related to the present work. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Kochanek, K.D.; Murphy, S.L.; Xu, J.; Arias, E. Deaths: Final Data for 2017. Natl. Vital Stat. Rep. 2019, 68, 9. [Google Scholar]
  2. Centers for Disease Control and Prevention, National Center for Injury Prevention and Control. Injuries and Violence Are Leading Causes of Death. 2020. Available online: https://www.cdc.gov/injury/wisqars/animated-leading-causes.html (accessed on 20 October 2020).
  3. Frydrych, L.M.; Keeney-Bonthrone, T.P.; Gwinn, E.; Wakam, G.K.; Anderson, M.S.; Delano, M.J. Short-term versus long-term trauma mortality: A systematic review. J. Trauma Acute Care Surg. 2019, 87, 990–997. [Google Scholar] [CrossRef] [PubMed]
  4. Johansson, P.I.; Ostrowski, S.R. Acute coagulopathy of trauma: Balancing progressive catecholamine induced endothelial activation and damage by fluid phase anticoagulation. Med. Hypotheses 2010, 75, 564–567. [Google Scholar] [CrossRef] [PubMed]
  5. Holcomb, J.B. A novel and potentially unifying mechanism for shock induced early coagulopathy. Ann. Surg. 2011, 254, 201–202. [Google Scholar] [CrossRef]
  6. Johansson, P.I.; Stensballe, J.; Ostrowski, S.R. Shock induced endotheliopathy (SHINE) in acute critical illness—A unifying pathophysiologic mechanism. Crit. Care. 2017, 21, 25. [Google Scholar] [CrossRef] [Green Version]
  7. White, N.J.; Ward, K.R.; Pati, S.; Strandenes, G.; Cap, A.P. Hemorrhagic blood failure: Oxygen debt, coagulopathy, and endothelial damage. J. Trauma Acute Care Surg. 2017, 82 (Suppl. 1), S41–S49. [Google Scholar] [CrossRef]
  8. Reitsma, S.; Slaaf, D.W.; Vink, H.; van Zandvoort, M.A.; oude Egbrink, M.G. The endothelial glycocalyx: Composition, functions, and visualization. Pflug. Arch. 2007, 454, 345–359. [Google Scholar] [CrossRef] [Green Version]
  9. Reine, T.M.; Lanzalaco, F.; Kristiansen, O.; Enget, A.R.; Satchell, S.; Jenssen, T.G.; Kolset, S.O. Matrix metalloproteinase-9 mediated shedding of syndecan-4 in glomerular endothelial cells. Microcirculation 2019, 26, e12534. [Google Scholar] [CrossRef]
  10. Stepp, M.A.; Pal-Ghosh, S.; Tadvalkar, G.; Pajoohesh-Ganji, A. Syndecan-1 and Its Expanding List of Contacts. Adv. Wound Care 2015, 4, 235–249. [Google Scholar] [CrossRef] [Green Version]
  11. Bjerkvig, C.K.; Strandenes, G.; Eliassen, H.S.; Spinella, P.C.; Fosse, T.K.; Cap, A.P.; Ward, K.R. ‘Blood failure‘ time to view blood as an organ: How oxygen debt contributes to blood failure and its implications for remote damage control resuscitation. Transfusion 2016, 56 (Suppl. 2), S182–S189. [Google Scholar] [CrossRef] [Green Version]
  12. Rahbar, E.; Baer, L.A.; Cotton, B.A.; Holcomb, J.B.; Wade, C.E. Plasma colloid osmotic pressure is an early indicator of injury and hemorrhagic shock. Shock 2014, 41, 181–187. [Google Scholar] [CrossRef]
  13. Rahbar, E.; Cardenas, J.C.; Baimukanova, G.; Usadi, B.; Bruhn, R.; Pati, S.; Ostrowski, S.R.; Johansson, P.I.; Holcomb, J.B.; Wade, C.E. Endothelial glycocalyx shedding and vascular permeability in severely injured trauma patients. J. Transl. Med. 2015, 13, 117. [Google Scholar] [CrossRef] [Green Version]
  14. Gonzalez Rodriguez, E.; Cardenas, J.C.; Lopez, E.; Cotton, B.A.; Tomasek, J.S.; Ostrowski, S.R.; Baer, L.A.; Stensballe, J.; Holcomb, J.B.; Johansson, P.I.; et al. Early Identification of the Patient with Endotheliopathy of Trauma by Arrival Serum Albumin. Shock 2018, 50, 31–37. [Google Scholar] [CrossRef]
  15. Hofmann, N.; Zipperle, J.; Jafarmadar, M.; Ashmwe, M.; Keibl, C.; Penzenstadler, C.; Ponschab, M.; Jafarmadar, B.; Redl, H.; Bahrami, S.; et al. Experimental Models of Endotheliopathy: Impact of Shock Severity. Shock 2018, 49, 564–571. [Google Scholar] [CrossRef]
  16. Di Battista, A.P.; Rizoli, S.B.; Lejnieks, B.; Min, A.; Shiu, M.Y.; Peng, H.T.; Baker, A.J.; Hutchison, M.G.; Churchill, N.; Inaba, K.; et al. Sympathoadrenal Activation is Associated with Acute Traumatic Coagulopathy and Endotheliopathy in Isolated Brain Injury. Shock 2016, 46 (Suppl. 1), 96–103. [Google Scholar] [CrossRef]
  17. Gonzalez Rodriguez, E.; Cardenas, J.C.; Cox, C.S.; Kitagawa, R.S.; Stensballe, J.; Holcomb, J.B.; Johansson, P.I.; Wade, C.E. Traumatic brain injury is associated with increased syndecan-1 shedding in severely injured patients. Scand. J. Trauma Resusc. Emerg. Med. 2018, 26, 102. [Google Scholar] [CrossRef]
  18. Gonzalez Rodriguez, E.; Ostrowski, S.R.; Cardenas, J.C.; Baer, L.A.; Tomasek, J.S.; Henriksen, H.H.; Stensballe, J.; Cotton, B.A.; Holcomb, J.B.; Johansson, P.I.; et al. Syndecan-1: A Quantitative Marker for the Endotheliopathy of Trauma. J. Am. Coll. Surg. 2017, 225, 419–427. [Google Scholar] [CrossRef] [Green Version]
  19. Johansson, P.I.; Henriksen, H.H.; Stensballe, J.; Gybel-Brask, M.; Cardenas, J.C.; Baer, L.A.; Cotton, B.A.; Holcomb, J.B.; Wade, C.E.; Ostrowski, S.R. Traumatic Endotheliopathy: A Prospective Observational Study of 424 Severely Injured Patients. Ann. Surg. 2017, 265, 597–603. [Google Scholar] [CrossRef] [Green Version]
  20. Johansson, P.I.; Stensballe, J.; Rasmussen, L.S.; Ostrowski, S.R. High circulating adrenaline levels at admission predict increased mortality after trauma. J. Trauma Acute Care Surg. 2012, 72, 428–436. [Google Scholar] [CrossRef]
  21. Albert, V.; Subramanian, A.; Agrawal, D.; Pati, H.P.; Gupta, S.D.; Mukhopadhyay, A.K. Acute Traumatic Endotheliopathy in Isolated Severe Brain Injury and Its Impact on Clinical Outcome. Med. Sci. 2018, 6, 5. [Google Scholar] [CrossRef] [Green Version]
  22. Ostrowski, S.R.; Henriksen, H.H.; Stensballe, J.; Gybel-Brask, M.; Cardenas, J.C.; Baer, L.A.; Cotton, B.A.; Holcomb, J.B.; Wade, C.E.; Johansson, P.I. Sympathoadrenal activation and endotheliopathy are drivers of hypocoagulability and hyperfibrinolysis in trauma: A prospective observational study of 404 severely injured patients. J. Trauma Acute Care Surg. 2017, 82, 293–301. [Google Scholar] [CrossRef]
  23. Ostrowski, S.R.; Johansson, P.I. Endothelial glycocalyx degradation induces endogenous heparinization in patients with severe injury and early traumatic coagulopathy. J. Trauma Acute Care Surg. 2012, 73, 60–66. [Google Scholar] [CrossRef]
  24. Johansson, P.I.; Sørensen, A.M.; Perner, A.; Welling, K.L.; Wanscher, M.; Larsen, C.F.; Ostrowski, S.R. Disseminated intravascular coagulation or acute coagulopathy of trauma shock early after trauma? An observational study. Crit. Care 2011, 15, R272. [Google Scholar] [CrossRef] [Green Version]
  25. Johansson, P.I.; Sørensen, A.M.; Perner, A.; Welling, K.L.; Wanscher, M.; Larsen, C.F.; Ostrowski, S.R. High sCD40L levels early after trauma are associated with enhanced shock, sympathoadrenal activation, tissue and endothelial damage, coagulopathy and mortality. J. Thromb Haemost. 2012, 10, 207–216. [Google Scholar] [CrossRef]
  26. Naumann, D.N.; Hazeldine, J.; Davies, D.J.; Bishop, J.; Midwinter, M.J.; Belli, A.; Harrison, P.; Lord, J.M. Endotheliopathy of Trauma is an on-Scene Phenomenon, and is Associated with Multiple Organ Dysfunction Syndrome: A Prospective Observational Study. Shock 2018, 49, 420–428. [Google Scholar] [CrossRef] [Green Version]
  27. Johansson, P.I.; Stensballe, J.; Rasmussen, L.S.; Ostrowski, S.R. A high admission syndecan-1 level, a marker of endothelial glycocalyx degradation, is associated with inflammation, protein C depletion, fibrinolysis, and increased mortality in trauma patients. Ann. Surg. 2011, 254, 194–200. [Google Scholar] [CrossRef]
  28. Cobb, J.P.; O’Keefe, G.E. Injury research in the genomic era. Lancet 2004, 363, 2076–2083. [Google Scholar] [CrossRef]
  29. Alpantaki, K.; Tsiridis, E.; Pape, H.C.; Giannoudis, P.V. Application of clinical proteomics in diagnosis and management of trauma patients. Injury 2007, 38, 263–271. [Google Scholar] [CrossRef]
  30. Kulak, N.A.; Pichler, G.; Paron, I.; Nagaraj, N.; Mann, M. Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells. Nat. Methods 2014, 11, 319–324. [Google Scholar] [CrossRef]
  31. Wade, C.E.; Matijevic, N.; Wang, Y.W.; Rodriguez, E.G.; Lopez, E.; Ostrowski, S.R.; Cardenas, J.C.; Baer, L.A.; Chen, T.A.; Tomasek, J.S.; et al. Absences of Endothelial Microvesicle Changes in the Presence of the Endotheliopathy of Trauma. Shock 2019, 51, 180–184. [Google Scholar] [CrossRef]
  32. Wei, S.; Gonzalez Rodriguez, E.; Chang, R.; Holcomb, J.B.; Kao, L.S.; Wade, C.E. Elevated Syndecan-1 after Trauma and Risk of Sepsis: A Secondary Analysis of Patients from the Pragmatic, Randomized Optimal Platelet and Plasma Ratios (PROPPR) Trial. J. Am. Coll. Surg. 2018, 227, 587–595. [Google Scholar] [CrossRef]
  33. Benjamini, Y.; Hochberg, Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J. R. Stat. Soc. Ser. B 1995, 57, 289–300. [Google Scholar] [CrossRef]
  34. Hastie, T.; Qian, J. Glmnet vignette. Stanford 2014, 9, 1–30. Available online: https://hastie.su.domains/Papers/Glmnet_Vignette.pdf (accessed on 11 January 2021).
  35. Steyerberg, E.W.; Harrell, F.E., Jr.; Borsboom, G.J.; Eijkemans, M.J.; Vergouwe, Y.; Habbema, J.D. Internal validation of predictive models: Efficiency of some procedures for logistic regression analysis. J. Clin. Epidemiol. 2001, 54, 774–781. [Google Scholar] [CrossRef]
  36. Steyerberg, E.W. Clinical Prediction Models: A Practical Approach to Development, Validation, and Updating; Springer International Publishing AG: Cham, Switzerland, 2019. [Google Scholar]
  37. Hatton, G.E.; Isbell, K.D.; Henriksen, H.H.; Stensballe, J.; Brummerstedt, M.; Johansson, P.I.; Kao, L.S.; Wade, C.E. Endothelial Dysfunction is Associated with Increased Incidence, Worsened Severity, and Prolonged Duration of Acute Kidney Injury after Severe Trauma. Shock 2021, 55, 311–315. [Google Scholar] [CrossRef]
  38. Thul, P.J.; Åkesson, L.; Wiking, M.; Mahdessian, D.; Geladaki, A.; Ait Blal, H.; Alm, T.; Asplund, A.; Björk, L.; Breckels, L.M.; et al. A subcellular map of the human proteome. Science 2017, 356, eaal3321. [Google Scholar] [CrossRef]
  39. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Proteomics. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
  40. Wang, S.; Song, R.; Wang, Z.; Jing, Z.; Wang, S.; Ma, J. S100A8/A9 in Inflammation. Front. Immunol. 2018, 9, 1298. [Google Scholar] [CrossRef]
  41. Sroussi, H.Y.; Berline, J.; Dazin, P.; Green, P.; Palefsky, J.M. S100A8 triggers oxidation-sensitive repulsion of neutrophils. J. Dent. Res. 2006, 85, 829–833. [Google Scholar] [CrossRef]
  42. Hiroshima, Y.; Hsu, K.; Tedla, N.; Chung, Y.M.; Chow, S.; Herbert, C.; Geczy, C.L. S100A8 induces IL-10 and protects against acute lung injury. J. Immunol. 2014, 192, 2800–2811. [Google Scholar] [CrossRef]
  43. Raftery, M.J.; Yang, Z.; Valenzuela, S.M.; Geczy, C.L. Novel intra- and inter-molecular sulfinamide bonds in S100A8 produced by hypochlorite oxidation. J. Biol. Chem. 2001, 276, 33393–33401. [Google Scholar] [CrossRef] [Green Version]
  44. Harrison, C.A.; Raftery, M.J.; Walsh, J.; Alewood, P.; Iismaa, S.E.; Thliveris, S.; Geczy, C.L. Oxidation regulates the inflammatory properties of the murine S100 protein S100A8. J. Biol. Chem. 1999, 274, 8561–8569. [Google Scholar] [CrossRef] [Green Version]
  45. Lim, S.Y.; Raftery, M.J.; Geczy, C.L. Oxidative modifications of DAMPs suppress inflammation: The case for S100A8 and S100A9. Antioxid Redox Signal. 2011, 15, 2235–2248. [Google Scholar] [CrossRef]
  46. Goyette, J.; Geczy, C.L. Inflammation-associated S100 proteins: New mechanisms that regulate function. Amino Acids. 2011, 41, 821–842. [Google Scholar] [CrossRef]
  47. Lim, S.Y.; Raftery, M.J.; Goyette, J.; Hsu, K.; Geczy, C.L. Oxidative modifications of S100 proteins: Functional regulation by redox. J. Leukoc. Biol. 2009, 86, 577–587. [Google Scholar] [CrossRef]
  48. Sroussi, H.Y.; Berline, J.; Palefsky, J.M. Oxidation of methionine 63 and 83 regulates the effect of S100A9 on the migration of neutrophils in vitro. J. Leukoc. Biol. 2007, 81, 818–824. [Google Scholar] [CrossRef]
  49. Smith, M.M. Histone structure and function. Curr. Opin. Cell Biol. 1991, 3, 429–437. [Google Scholar] [CrossRef]
  50. Huang, H.; Evankovich, J.; Yan, W.; Nace, G.; Zhang, L.; Ross, M.; Liao, X.; Billiar, T.; Xu, J.; Esmon, C.T.; et al. Endogenous histones function as alarmins in sterile inflammatory liver injury through Toll-like receptor 9 in mice. Hepatology 2011, 54, 999–1008. [Google Scholar] [CrossRef]
  51. Xu, J.; Zhang, X.; Monestier, M.; Esmon, N.L.; Esmon, C.T. Extracellular histones are mediators of death through TLR2 and TLR4 in mouse fatal liver injury. J. Immunol. 2011, 187, 2626–2631. [Google Scholar] [CrossRef] [Green Version]
  52. Xu, J.; Zhang, X.; Pelayo, R.; Monestier, M.; Ammollo, C.T.; Semeraro, F.; Taylor, F.B.; Esmon, N.L.; Lupu, F.; Esmon, C.T. Extracellular histones are major mediators of death in sepsis. Nat. Med. 2009, 15, 1318–1321. [Google Scholar] [CrossRef] [Green Version]
  53. Dinarvand, P.; Hassanian, S.M.; Qureshi, S.H.; Manithody, C.; Eissenberg, J.C.; Yang, L.; Rezaie, A.R. Polyphosphate amplifies proinflammatory responses of nuclear proteins through interaction with receptor for advanced glycation end products and P2Y1 purinergic receptor. Blood 2014, 123, 935–945. [Google Scholar] [CrossRef] [PubMed]
  54. Saffarzadeh, M.; Juenemann, C.; Queisser, M.A.; Lochnit, G.; Barreto, G.; Galuska, S.P.; Lohmeyer, J.; Preissner, K.T. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: A predominant role of histones. PLoS ONE 2012, 7, e32366. [Google Scholar] [CrossRef] [PubMed]
  55. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef] [PubMed]
  56. Chaaban, H.; Keshari, R.S.; Silasi-Mansat, R.; Popescu, N.I.; Mehta-D’Souza, P.; Lim, Y.P.; Lupu, F. Inter-α inhibitor protein and its associated glycosaminoglycans protect against histone-induced injury. Blood 2015, 125, 2286–2296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Wildhagen, K.C.; García de Frutos, P.; Reutelingsperger, C.P.; Schrijver, R.; Aresté, C.; Ortega-Gómez, A.; Deckers, N.M.; Hemker, H.C.; Soehnlein, O.; Nicolaes, G.A. Nonanticoagulant heparin prevents histone-mediated cytotoxicity in vitro and improves survival in sepsis. Blood 2014, 123, 1098–1101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Pemberton, A.D.; Brown, J.K.; Inglis, N.F. Proteomic identification of interactions between histones and plasma proteins: Implications for cytoprotection. Proteomics 2010, 10, 1484–1493. [Google Scholar] [CrossRef]
  59. Johansson, P.I.; Windeløv, N.A.; Rasmussen, L.S.; Sørensen, A.M.; Ostrowski, S.R. Blood levels of histone-complexed DNA fragments are associated with coagulopathy, inflammation and endothelial damage early after trauma. J. Emerg. Trauma Shock 2013, 6, 171–175. [Google Scholar] [CrossRef]
  60. Abrams, S.T.; Zhang, N.; Manson, J.; Liu, T.; Dart, C.; Baluwa, F.; Wang, S.S.; Brohi, K.; Kipar, A.; Yu, W.; et al. Circulating histones are mediators of trauma-associated lung injury. Am. J. Respir. Crit. Care Med. 2013, 187, 160–169. [Google Scholar] [CrossRef] [Green Version]
  61. Esmon, C.T. Molecular circuits in thrombosis and inflammation. Thromb. Haemost. 2013, 109, 416–420. [Google Scholar] [CrossRef]
  62. Semeraro, F.; Ammollo, C.T.; Semeraro, N.; Colucci, M. Extracellular histones promote fibrinolysis by single-chain urokinase-type plasminogen activator in a factor seven activating protease-dependent way. Thromb. Res. 2020, 196, 193–199. [Google Scholar] [CrossRef]
  63. Jin, Z.G.; Melaragno, M.G.; Liao, D.F.; Yan, C.; Haendeler, J.; Suh, Y.A.; Lambeth, J.D.; Berk, B.C. Cyclophilin A is a secreted growth factor induced by oxidative stress. Circ. Res. 2000, 87, 789–796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Koh, M.W.; Baldi, R.F.; Soni, S.; Handslip, R.; Tan, Y.Y.; O’Dea, K.P.; Malesevic, M.; McAuley, D.F.; O’Kane, C.M.; Patel, B.V.; et al. Secreted Extracellular Cyclophilin A Is a Novel Mediator of Ventilator-induced Lung Injury. Am. J. Respir. Crit. Care Med. 2021, 204, 421–430. [Google Scholar] [CrossRef] [PubMed]
  65. Coppinger, J.A.; Cagney, G.; Toomey, S.; Kislinger, T.; Belton, O.; McRedmond, J.P.; Cahill, D.J.; Emili, A.; Fitzgerald, D.J.; Maguire, P.B. Characterization of the proteins released from activated platelets leads to localization of novel platelet proteins in human atherosclerotic lesions. Blood 2004, 103, 2096–2104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sherry, B.; Yarlett, N.; Strupp, A.; Cerami, A. Identification of cyclophilin as a proinflammatory secretory product of lipopolysaccharide-activated macrophages. Proc. Natl. Acad. Sci. USA 1992, 89, 3511–3515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Dawar, F.U.; Xiong, Y.; Khattak, M.N.K.; Li, J.; Lin, L.; Mei, J. Potential role of cyclophilin A in regulating cytokine secretion. J. Leukoc. Biol. 2017, 102, 989–992. [Google Scholar] [CrossRef]
  68. Staedtke, V.; Bai, R.Y.; Kim, K.; Darvas, M.; Davila, M.L.; Riggins, G.J.; Rothman, P.B.; Papadopoulos, N.; Kinzler, K.W.; Vogelstein, B.; et al. Disruption of a self-amplifying catecholamine loop reduces cytokine release syndrome. Nature 2018, 564, 273–277. [Google Scholar] [CrossRef]
  69. Fontaine, M.; Lepape, A.; Piriou, V.; Venet, F.; Friggeri, A. Innate danger signals in acute injury: From bench to bedside. Anaesth. Crit. Care Pain Med. 2016, 35, 283–292. [Google Scholar] [CrossRef]
  70. Relja, B.; Land, W.G. Damage-associated molecular patterns in trauma. Eur. J. Trauma Emerg. Surg. 2020, 46, 751–775. [Google Scholar] [CrossRef] [Green Version]
  71. Matzinger, P. Tolerance, danger, and the extended family. Annu. Rev. Immunol. 1994, 12, 991–1045. [Google Scholar] [CrossRef]
  72. Matzinger, P. Essay 1: The Danger model in its historical context. Scand. J Immunol. 2001, 54, 4–9. [Google Scholar] [CrossRef]
  73. Matzinger, P. The danger model: A renewed sense of self. Science 2002, 296, 301–305. [Google Scholar] [CrossRef] [Green Version]
  74. Marinković, G.; Koenis, D.S.; de Camp, L.; Jablonowski, R.; Graber, N.; de Waard, V.; de Vries, C.J.; Goncalves, I.; Nilsson, J.; Jovinge, S.; et al. S100A9 Links Inflammation and Repair in Myocardial Infarction. Circ. Res. 2020, 127, 664–676. [Google Scholar] [CrossRef]
  75. Araki, K.; Kinoshita, R.; Tomonobu, N.; Gohara, Y.; Tomida, S.; Takahashi, Y.; Senoo, S.; Taniguchi, A.; Itano, J.; Yamamoto, K.I.; et al. The heterodimer S100A8/A9 is a potent therapeutic target for idiopathic pulmonary fibrosis. J. Mol. Med. 2021, 99, 131–145. [Google Scholar] [CrossRef]
  76. Rodrigues, R.M.; He, Y.; Hwang, S.; Bertola, A.; Mackowiak, B.; Ahmed, Y.A.; Seo, W.; Ma, J.; Wang, X.; Park, S.H.; et al. E-Selectin-Dependent Inflammation and Lipolysis in Adipose Tissue Exacerbate Steatosis-to-NASH Progression via S100A8/9. Cell Mol. Gastroenterol. Hepatol. 2022, 13, 151–171. [Google Scholar] [CrossRef]
  77. Lu, G.; Jia, Z.; Zu, Q.; Zhang, J.; Zhao, L.; Shi, H. Inhibition of the cyclophilin A-CD147 interaction attenuates right ventricular injury and dysfunction after acute pulmonary embolism in rats. J. Biol. Chem. 2018, 293, 12199–12208. [Google Scholar] [CrossRef]
  78. Seizer, P.; Schönberger, T.; Schött, M.; Lang, M.R.; Langer, H.F.; Bigalke, B.; Krämer, B.F.; Borst, O.; Daub, K.; Heidenreich, O.; et al. EMMPRIN and its ligand cyclophilin A regulate MT1-MMP, MMP-9 and M-CSF during foam cell formation. Atherosclerosis 2010, 209, 51–57. [Google Scholar] [CrossRef]
  79. Balsley, M.A.; Malesevic, M.; Stemmy, E.J.; Gigley, J.; Jurjus, R.A.; Herzog, D.; Bukrinsky, M.I.; Fischer, G.; Constant, S.L. A cell-impermeable cyclosporine A derivative reduces pathology in a mouse model of allergic lung inflammation. J. Immunol. 2010, 185, 7663–7670. [Google Scholar] [CrossRef]
  80. Von Ungern-Sternberg, S.N.I.; Vogel, S.; Walker-Allgaier, B.; Geue, S.; Maurer, A.; Wild, A.M.; Münzer, P.; Chatterjee, M.; Heinzmann, D.; Kremmer, E.; et al. Extracellular Cyclophilin A Augments Platelet-Dependent Thrombosis and Thromboinflammation. Thromb. Haemost. 2017, 117, 2063–2078. [Google Scholar] [CrossRef]
  81. Heinzmann, D.; Bangert, A.; Müller, A.M.; von Ungern-Sternberg, S.N.; Emschermann, F.; Schönberger, T.; Chatterjee, M.; Mack, A.F.; Klingel, K.; Kandolf, R.; et al. The Novel Extracellular Cyclophilin A (CyPA)—Inhibitor MM284 Reduces Myocardial Inflammation and Remodeling in a Mouse Model of Troponin I -Induced Myocarditis. PLoS ONE 2015, 10, e0124606. [Google Scholar] [CrossRef] [Green Version]
  82. Liu, S.Y.; Zhang, Q.Z.; Hu, M.Q.; Li, F.X.; Fu, J.M.; Zhu, Z.D.; Li, Q.K.; Yang, Z.; Quan, J.M. Targeting Extracellular Cyclophilin A via an Albumin-Binding Cyclosporine A Analogue. ChemMedChem 2021, 16, 3649–3652. [Google Scholar] [CrossRef]
  83. Hernandez, I.; Tesoro, L.; Ramirez-Carracedo, R.; Diez-Mata, J.; Sanchez, S.; Saura, M.; Zamorano, J.L.; Zaragoza, C.; Botana, L. Ivabradine Induces Cardiac Protection against Myocardial Infarction by Preventing Cyclophilin-A Secretion in Pigs under Coronary Ischemia/Reperfusion. Int. J. Mol. Sci. 2021, 22, 2902. [Google Scholar] [CrossRef]
  84. Zhou, J.; Liu, K.; Feng, C.; Peng, Z.; Luo, W. Therapeutic effect of SP-8356 on pulmonary embolism-associated cardiac injury is mediated by its ability to suppress apoptosis and inflammation. J. Cell Mol. Med. 2021, 25, 5260–5268. [Google Scholar] [CrossRef]
  85. Nakahara, M.; Ito, T.; Kawahara, K.; Yamamoto, M.; Nagasato, T.; Shrestha, B.; Yamada, S.; Miyauchi, T.; Higuchi, K.; Takenaka, T.; et al. Recombinant thrombomodulin protects mice against histone-induced lethal thromboembolism. PLoS ONE 2013, 8, e75961. [Google Scholar] [CrossRef] [Green Version]
  86. Shi, H.; Gandhi, A.A.; Smith, S.A.; Wang, Q.; Chiang, D.; Yalavarthi, S.; Ali, R.A.; Liu, C.; Sule, G.; Tsou, P.S.; et al. Endothelium-protective, histone-neutralizing properties of the polyanionic agent defibrotide. JCI Insight 2021, 6, e149149. [Google Scholar] [CrossRef]
  87. Viemann, D.; Strey, A.; Janning, A.; Jurk, K.; Klimmek, K.; Vogl, T.; Hirono, K.; Ichida, F.; Foell, D.; Kehrel, B.; et al. Myeloid-related proteins 8 and 14 induce a specific inflammatory response in human microvascular endothelial cells. Blood 2005, 105, 2955–2962. [Google Scholar] [CrossRef] [Green Version]
  88. Ryckman, C.; Vandal, K.; Rouleau, P.; Talbot, M.; Tessier, P.A. Proinflammatory activities of S100: Proteins S100A8, S100A9, and S100A8/A9 induce neutrophil chemotaxis and adhesion. J. Immunol. 2003, 170, 3233–3242. [Google Scholar] [CrossRef] [Green Version]
  89. Vogl, T.; Ludwig, S.; Goebeler, M.; Strey, A.; Thorey, I.S.; Reichelt, R.; Foell, D.; Gerke, V.; Manitz, M.P.; Nacken, W.; et al. MRP8 and MRP14 control microtubule reorganization during transendothelial migration of phagocytes. Blood 2004, 104, 4260–4268. [Google Scholar] [CrossRef] [Green Version]
  90. Simard, J.C.; Girard, D.; Tessier, P.A. Induction of neutrophil degranulation by S100A9 via a MAPK-dependent mechanism. J. Leukoc. Biol. 2010, 87, 905–914. [Google Scholar] [CrossRef] [Green Version]
  91. Riva, M.; Källberg, E.; Björk, P.; Hancz, D.; Vogl, T.; Roth, J.; Ivars, F.; Leanderson, T. Induction of nuclear factor-κB responses by the S100A9 protein is Toll-like receptor-4-dependent. Immunology 2012, 137, 172–182. [Google Scholar] [CrossRef]
  92. Ekaney, M.L.; Otto, G.P.; Sossdorf, M.; Sponholz, C.; Boehringer, M.; Loesche, W.; Rittirsch, D.; Wilharm, A.; Kurzai, O.; Bauer, M.; et al. Impact of plasma histones in human sepsis and their contribution to cellular injury and inflammation. Crit. Care 2014, 18, 543. [Google Scholar] [CrossRef] [Green Version]
  93. Xu, Q.; Leiva, M.C.; Fischkoff, S.A.; Handschumacher, R.E.; Lyttle, C.R. Leukocyte chemotactic activity of cyclophilin. J. Biol. Chem. 1992, 267, 11968–11971. [Google Scholar] [CrossRef]
  94. Jin, Z.G.; Lungu, A.O.; Xie, L.; Wang, M.; Wong, C.; Berk, B.C. Cyclophilin A is a proinflammatory cytokine that activates endothelial cells. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 1186–1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Lim, S.Y.; Raftery, M.; Cai, H.; Hsu, K.; Yan, W.X.; Hseih, H.L.; Watts, R.N.; Richardson, D.; Thomas, S.; Perry, M.; et al. S-nitrosylated S100A8: Novel anti-inflammatory properties. J. Immunol. 2008, 181, 5627–5636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Koike, A.; Arai, S.; Yamada, S.; Nagae, A.; Saita, N.; Itoh, H.; Uemoto, S.; Totani, M.; Ikemoto, M. Dynamic mobility of immunological cells expressing S100A8 and S100A9 in vivo: A variety of functional roles of the two proteins as regulators in acute inflammatory reaction. Inflammation 2012, 35, 409–419. [Google Scholar] [CrossRef]
  97. Sun, Y.; Lu, Y.; Engeland, C.G.; Gordon, S.C.; Sroussi, H.Y. The anti-oxidative, anti-inflammatory, and protective effect of S100A8 in endotoxemic mice. Mol. Immunol. 2013, 53, 443–449. [Google Scholar] [CrossRef] [Green Version]
  98. Sui, J.; Lu, R.; Halkidis, K.; Kocher, N.K.; Cao, W.; Marques, M.B.; Zheng, X.L. Plasma levels of S100A8/A9, histone/DNA complexes, and cell-free DNA predict adverse outcomes of immune thrombotic thrombocytopenic purpura. J. Thromb. Haemost. 2021, 19, 370–379. [Google Scholar] [CrossRef]
  99. Fuchs, T.A.; Kremer Hovinga, J.A.; Schatzberg, D.; Wagner, D.D.; Lämmle, B. Circulating DNA and myeloperoxidase indicate disease activity in patients with thrombotic microangiopathies. Blood 2012, 120, 1157–1164. [Google Scholar] [CrossRef] [Green Version]
  100. Zuo, Y.; Zuo, M.; Yalavarthi, S.; Gockman, K.; Madison, J.A.; Shi, H.; Woodard, W.; Lezak, S.P.; Lugogo, N.L.; Knight, J.S.; et al. Neutrophil extracellular traps and thrombosis in COVID-19. J. Thromb. Thrombolysis 2021, 51, 446–453. [Google Scholar] [CrossRef]
  101. Wang, X.; Guan, M.; Zhang, X.; Ma, T.; Wu, M.; Li, Y.; Chen, X.; Zheng, Y. The Association Between S100A8/A9 and the Development of Very Late Stent Thrombosis in Patients with Acute Myocardial Infarction. Clin. Appl. Thromb. Hemost. 2020, 26, 1076029620943295. [Google Scholar] [CrossRef]
  102. Semeraro, F.; Ammollo, C.T.; Morrissey, J.H.; Dale, G.L.; Friese, P.; Esmon, N.L.; Esmon, C.T. Extracellular histones promote thrombin generation through platelet-dependent mechanisms: Involvement of platelet TLR2 and TLR4. Blood 2011, 118, 1952–1961. [Google Scholar] [CrossRef] [Green Version]
  103. Fuchs, T.A.; Bhandari, A.A.; Wagner, D.D. Histones induce rapid and profound thrombocytopenia in mice. Blood 2011, 118, 3708–3714. [Google Scholar] [CrossRef] [Green Version]
  104. Lam, F.W.; Cruz, M.A.; Leung, H.C.; Parikh, K.S.; Smith, C.W.; Rumbaut, R.E. Histone induced platelet aggregation is inhibited by normal albumin. Thromb. Res. 2013, 132, 69–76. [Google Scholar] [CrossRef]
  105. Ammollo, C.T.; Semeraro, F.; Xu, J.; Esmon, N.L.; Esmon, C.T. Extracellular histones increase plasma thrombin generation by impairing thrombomodulin-dependent protein C activation. J. Thromb. Haemost. 2011, 9, 1795–1803. [Google Scholar] [CrossRef]
  106. Vulliamy, P.; Gillespie, S.; Armstrong, P.C.; Allan, H.E.; Warner, T.D.; Brohi, K. Histone H4 induces platelet ballooning and microparticle release during trauma hemorrhage. Proc. Natl. Acad. Sci. USA 2019, 116, 17444–17449. [Google Scholar] [CrossRef] [Green Version]
  107. Pozzi, N.; Di Cera, E. Dual effect of histone H4 on prothrombin activation. J. Thromb. Haemost. 2016, 14, 1814–1818. [Google Scholar] [CrossRef] [Green Version]
  108. Seizer, P.; Ungern-Sternberg, S.N.; Schönberger, T.; Borst, O.; Münzer, P.; Schmidt, E.M.; Mack, A.F.; Heinzmann, D.; Chatterjee, M.; Langer, H.; et al. Extracellular cyclophilin A activates platelets via EMMPRIN (CD147) and PI3K/Akt signaling, which promotes platelet adhesion and thrombus formation in vitro and in vivo. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 655–663. [Google Scholar] [CrossRef] [Green Version]
  109. Kawasaki, Y.; Ohara, S.; Abe, Y.; Watanabe, M.; Suyama, K.; Sato, M.; Hashimoto, K.; Hosoya, M. The role of serum myeloid-related protein 8/14 complex in Henoch-Schönlein purpura nephritis. Pediatr. Nephrol. 2012, 27, 65–71. [Google Scholar] [CrossRef]
  110. Chen, X.; Tao, T.; Wang, H.; Zhao, H.; Lu, L.; Wu, F. Arterial Thrombosis Is Accompanied by Elevated Mitogen-Activated Protein Kinase (MAPK) and Cyclooxygenase-2 (COX-2) Expression via Toll-Like Receptor 4 (TLR-4) Activation by S100A8/A9. Med. Sci. Monit. 2018, 24, 7673–7681. [Google Scholar] [CrossRef]
  111. Stocca, A.; O’Toole, D.; Hynes, N.; Hynes, S.O.; Mashayekhi, K.; McGinley, L.; O’Connell, E.; Coleman, C.; Sultan, S.; Duffy, A.; et al. A role for MRP8 in in stent restenosis in diabetes. Atherosclerosis 2012, 221, 325–332. [Google Scholar] [CrossRef]
  112. Collier, D.M.; Villalba, N.; Sackheim, A.; Bonev, A.D.; Miller, Z.D.; Moore, J.S.; Shui, B.; Lee, J.C.; Lee, F.K.; Reining, S.; et al. Extracellular histones induce calcium signals in the endothelium of resistance-sized mesenteric arteries and cause loss of endothelium-dependent dilation. Am. J. Physiol. Heart Circ. Physiol. 2019, 316, H1309–H1322. [Google Scholar] [CrossRef]
  113. Xie, Y.; Li, X.; Ge, J. Cyclophilin A-FoxO1 signaling pathway in endothelial cell apoptosis. Cell Signal. 2019, 61, 57–65. [Google Scholar] [CrossRef] [PubMed]
  114. Kim, S.H.; Lessner, S.M.; Sakurai, Y.; Galis, Z.S. Cyclophilin A as a novel biphasic mediator of endothelial activation and dysfunction. Am. J. Pathol. 2004, 164, 1567–1574. [Google Scholar] [CrossRef] [Green Version]
  115. Soe, N.N.; Sowden, M.; Baskaran, P.; Kim, Y.; Nigro, P.; Smolock, E.M.; Berk, B.C. Acetylation of cyclophilin A is required for its secretion and vascular cell activation. Cardiovasc. Res. 2014, 101, 444–453. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, L.; Luo, H.; Chen, X.; Jiang, Y.; Huang, Q. Functional characterization of S100A8 and S100A9 in altering monolayer permeability of human umbilical endothelial cells. PLoS ONE 2014, 9, e90472. [Google Scholar] [CrossRef] [Green Version]
  117. Wang, A.; Guo, B.; Jia, Q.; Chen, Y.U.; Gao, X.; Xu, S. S100A9-containing serum exosomes of burn injury patients promote permeability of pulmonary microvascular endothelial cells. J. Biosci. 2021, 46, 33. [Google Scholar] [CrossRef]
  118. Rasmuson, J.; Kenne, E.; Wahlgren, M.; Soehnlein, O.; Lindbom, L. Heparinoid sevuparin inhibits Streptococcus-induced vascular leak through neutralizing neutrophil-derived proteins. Faseb. J. 2019, 33, 10443–10452. [Google Scholar] [CrossRef] [Green Version]
  119. Abrams, S.T.; Zhang, N.; Dart, C.; Wang, S.S.; Thachil, J.; Guan, Y.; Wang, G.; Toh, C. Human CRP defends against the toxicity of circulating histones. J. Immunol. 2013, 191, 2495–2502. [Google Scholar] [CrossRef] [Green Version]
  120. Li, Y.; Chen, B.; Yang, X.; Zhang, C.; Jiao, Y.; Li, P.; Liu, Y.; Li, Z.; Qiao, B.; Bond Lau, W.; et al. S100a8/a9 Signaling Causes Mitochondrial Dysfunction and Cardiomyocyte Death in Response to Ischemic/Reperfusion Injury. Circulation 2019, 140, 751–764. [Google Scholar] [CrossRef]
  121. Sreejit, G.; Abdel-Latif, A.; Athmanathan, B.; Annabathula, R.; Dhyani, A.; Noothi, S.K.; Quaife-Ryan, G.A.; Al-Sharea, A.; Pernes, G.; Dragoljevic, D.; et al. Neutrophil-Derived S100A8/A9 Amplify Granulopoiesis After Myocardial Infarction. Circulation 2020, 141, 1080–1094. [Google Scholar] [CrossRef]
  122. Bruhn, L.V.; Lauridsen, K.G.; Schmidt, A.S.; Rickers, H.; Bach, L.F.; Løfgren, B.; Hornung, N. Elevated calprotectin in patients with atrial fibrillation with and without heart failure. Scand. J. Clin. Lab. Investig. 2017, 77, 210–215. [Google Scholar] [CrossRef]
  123. Imbalzano, E.; Mandraffino, G.; Casciaro, M.; Quartuccio, S.; Saitta, A.; Gangemi, S. Pathophysiological mechanism and therapeutic role of S100 proteins in cardiac failure: A systematic review. Heart Fail. Rev. 2016, 21, 463–473. [Google Scholar] [CrossRef]
  124. Lu, N.F.; Jiang, L.; Zhu, B.; Yang, D.G.; Zheng, R.Q.; Shao, J.; Yuan, J.; Xi, X.M. Elevated Plasma Histone H4 Levels Are an Important Risk Factor in the Development of Septic Cardiomyopathy. Balk. Med. J. 2020, 37, 72–78. [Google Scholar] [CrossRef]
  125. Shah, M.; He, Z.; Rauf, A.; Kalkhoran, S.B.; Heiestad, C.M.; Stensløkken, K.O.; Parish, C.R.; Soehnlein, O.; Arjun, S.; Davidson, S.M.; et al. Extracellular histones are a target in myocardial ischaemia reperfusion injury. Cardiovasc. Res. 2021, 118, 1115–1125. [Google Scholar] [CrossRef]
  126. Lu, N.F.; Jiang, L.; Zhu, B.; Yang, D.G.; Zheng, R.Q.; Shao, J.; Xi, X.M. Elevated plasma histone H4 level predicts increased risk of mortality in patients with sepsis. Ann. Palliat. Med. 2020, 9, 1084–1091. [Google Scholar] [CrossRef]
  127. Yu, W.; Jianhong, L.; Weili, W.; Peng, S.; Zhongyang, S. Effects of CyPA signal pathway in myocardial tissue after cardiopulmonary resuscitation in rats. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 2015, 27, 965–969. [Google Scholar]
  128. Seizer, P.; Ochmann, C.; Schönberger, T.; Zach, S.; Rose, M.; Borst, O.; Klingel, K.; Kandolf, R.; MacDonald, H.R.; Nowak, R.A.; et al. Disrupting the EMMPRIN (CD147)-cyclophilin A interaction reduces infarct size and preserves systolic function after myocardial ischemia and reperfusion. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 1377–1386. [Google Scholar] [CrossRef] [Green Version]
  129. Seko, Y.; Fujimura, T.; Taka, H.; Mineki, R.; Murayama, K.; Nagai, R. Hypoxia followed by reoxygenation induces secretion of cyclophilin A from cultured rat cardiac myocytes. Biochem. Biophys. Res. Commun. 2004, 317, 162–168. [Google Scholar] [CrossRef]
  130. Yan, J.; Zang, X.; Chen, R.; Yuan, W.; Gong, J.; Wang, C.; Li, Y. The clinical implications of increased cyclophilin A levels in patients with acute coronary syndromes. Clin. Chim. Acta 2012, 413, 691–695. [Google Scholar] [CrossRef]
  131. Shi, H.; Zuo, Y.; Yalavarthi, S.; Gockman, K.; Zuo, M.; Madison, J.A.; Blair, C.; Woodward, W.; Lezak, S.P.; Lugogo, N.L.; et al. Neutrophil calprotectin identifies severe pulmonary disease in COVID-19. J. Leukoc. Biol. 2021, 109, 67–72. [Google Scholar] [CrossRef]
  132. Hasenauer, A.; Bédat, B.; Parapanov, R.; Lugrin, J.; Debonneville, A.; Abdelnour-Berchtold, E.; Gonzalez, M.; Perentes, J.Y.; Piquilloud, L.; Szabo, C.; et al. Effects of cold or warm ischemia and ex-vivo lung perfusion on the release of damage associated molecular patterns and inflammatory cytokines in experimental lung transplantation. J. Heart Lung Transplant. 2021, 40, 905–916. [Google Scholar] [CrossRef]
  133. Saito, T.; Liu, M.; Binnie, M.; Sato, M.; Hwang, D.; Azad, S.; Machuca, T.N.; Zamel, R.; Waddell, T.K.; Cypel, M.; et al. Distinct expression patterns of alveolar “alarmins” in subtypes of chronic lung allograft dysfunction. Am. J. Transplant. 2014, 14, 1425–1432. [Google Scholar] [CrossRef] [PubMed]
  134. Lou, Y.; Zheng, Y.; Fan, B.; Zhang, L.; Zhu, F.; Wang, X.; Chen, Z.; Tan, X.; Wei, Q. Serum levels of interleukins and S100A8/A9 correlate with clinical severity in patients with dermatomyositis-associated interstitial lung disease. BMC Pulm. Med. 2020, 20, 196. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, Y.; Zhao, J.; Guan, L.; Mao, L.; Li, S.; Zhao, J. Histone H4 aggravates inflammatory injury through TLR4 in chlorine gas-induced acute respiratory distress syndrome. J. Occup. Med. Toxicol. 2020, 15, 31. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, Y.; Guan, L.; Yu, J.; Zhao, Z.; Mao, L.; Li, S.; Zhao, J. Pulmonary endothelial activation caused by extracellular histones contributes to neutrophil activation in acute respiratory distress syndrome. Respir. Res. 2016, 17, 155. [Google Scholar] [CrossRef] [Green Version]
  137. Gwinn, W.M.; Damsker, J.M.; Falahati, R.; Okwumabua, I.; Kelly-Welch, A.; Keegan, A.D.; Vanpouille, C.; Lee, J.J.; Dent, L.A.; Leitenberg, D.; et al. Novel approach to inhibit asthma-mediated lung inflammation using anti-CD147 intervention. J. Immunol. 2006, 177, 4870–4879. [Google Scholar] [CrossRef] [Green Version]
  138. Nikolakopoulou, Z.; Hector, L.R.; Creagh-Brown, B.C.; Evans, T.W.; Quinlan, G.J.; Burke-Gaffney, A. Plasma S100A8/A9 heterodimer is an early prognostic marker of acute kidney injury associated with cardiac surgery. Biomark. Med. 2019, 13, 205–218. [Google Scholar] [CrossRef]
  139. Fujiu, K.; Manabe, I.; Nagai, R. Renal collecting duct epithelial cells regulate inflammation in tubulointerstitial damage in mice. J. Clin. Investig. 2011, 121, 3425–3441. [Google Scholar] [CrossRef] [Green Version]
  140. Dessing, M.C.; Tammaro, A.; Pulskens, W.P.; Teske, G.J.; Butter, L.M.; Claessen, N.; van Eijk, M.; van der Poll, T.; Vogl, T.; Roth, J.; et al. The calcium-binding protein complex S100A8/A9 has a crucial role in controlling macrophage-mediated renal repair following ischemia/reperfusion. Kidney Int. 2015, 87, 85–94. [Google Scholar] [CrossRef]
  141. Tammaro, A.; Florquin, S.; Brok, M.; Claessen, N.; Butter, L.M.; Teske, G.J.D.; de Boer, O.J.; Vogl, T.; Leemans, J.C.; Dessing, M.C. S100A8/A9 promotes parenchymal damage and renal fibrosis in obstructive nephropathy. Clin. Exp. Immunol. 2018, 193, 361–375. [Google Scholar] [CrossRef] [Green Version]
  142. Pepper, R.J.; Wang, H.H.; Rajakaruna, G.K.; Papakrivopoulou, E.; Vogl, T.; Pusey, C.D.; Cook, H.T.; Salama, A.D. S100A8/A9 (calprotectin) is critical for development of glomerulonephritis and promotes inflammatory leukocyte-renal cell interactions. Am. J. Pathol. 2015, 185, 1264–1274. [Google Scholar] [CrossRef] [Green Version]
  143. Allam, R.; Scherbaum, C.R.; Darisipudi, M.N.; Mulay, S.R.; Hägele, H.; Lichtnekert, J.; Hagemann, J.H.; Rupanagudi, K.V.; Ryu, M.; Schwarzenberger, C.; et al. Histones from dying renal cells aggravate kidney injury via TLR2 and TLR4. J. Am. Soc. Nephrol. 2012, 23, 1375–1388. [Google Scholar] [CrossRef] [Green Version]
  144. Nakazawa, D.; Kumar, S.V.; Marschner, J.; Desai, J.; Holderied, A.; Rath, L.; Kraft, F.; Lei, Y.; Fukasawa, Y.; Moeckel, G.W.; et al. Histones and Neutrophil Extracellular Traps Enhance Tubular Necrosis and Remote Organ Injury in Ischemic AKI. J. Am. Soc. Nephrol. 2017, 28, 1753–1768. [Google Scholar] [CrossRef] [Green Version]
  145. Leong, K.G.; Ozols, E.; Kanellis, J.; Badal, S.S.; Liles, J.T.; Nikolic-Paterson, D.J.; Ma, F.Y. Cyclophilin Inhibition Protects against Experimental Acute Kidney Injury and Renal Interstitial Fibrosis. Int. J. Mol. Sci. 2020, 22, 271. [Google Scholar] [CrossRef]
  146. Leong, K.G.; Ozols, E.; Kanellis, J.; Nikolic-Paterson, D.J.; Ma, F.Y. Cyclophilin A Promotes Inflammation in Acute Kidney Injury but Not in Renal Fibrosis. Int. J. Mol. Sci. 2020, 21, 3667. [Google Scholar] [CrossRef]
  147. Lee, C.C.; Chang, C.H.; Cheng, Y.L.; Kuo, G.; Chen, S.W.; Li, Y.J.; Chen, Y.T.; Tian, Y.C. Diagnostic Performance of Cyclophilin A in Cardiac Surgery-Associated Acute Kidney Injury. J. Clin. Med. 2019, 9, 108. [Google Scholar] [CrossRef] [Green Version]
  148. Mizobuchi, H.; Fujii, W.; Isokawa, S.; Ishizuka, K.; Wang, Y.; Watanabe, S.; Sanjoba, C.; Matsumoto, Y.; Goto, Y. Exacerbation of hepatic injury during rodent malaria by myeloid-related protein 14. PLoS ONE 2018, 13, e0199111. [Google Scholar] [CrossRef] [Green Version]
  149. Moles, A.; Murphy, L.; Wilson, C.L.; Chakraborty, J.B.; Fox, C.; Park, E.J.; Mann, J.; Oakley, F.; Howarth, R.; Brain, J.; et al. A TLR2/S100A9/CXCL-2 signaling network is necessary for neutrophil recruitment in acute and chronic liver injury in the mouse. J. Hepatol. 2014, 60, 782–791. [Google Scholar] [CrossRef] [Green Version]
  150. Takai, S.; Higuchi, S.; Yano, A.; Tsuneyama, K.; Fukami, T.; Nakajima, M.; Yokoi, T. Involvement of immune- and inflammatory-related factors in flucloxacillin-induced liver injury in mice. J. Appl. Toxicol. 2015, 35, 142–151. [Google Scholar] [CrossRef]
  151. Wen, Z.; Liu, Y.; Li, F.; Ren, F.; Chen, D.; Li, X.; Wen, T. Circulating histones exacerbate inflammation in mice with acute liver failure. J. Cell Biochem. 2013, 114, 2384–2391. [Google Scholar] [CrossRef]
  152. Huang, H.; Chen, H.W.; Evankovich, J.; Yan, W.; Rosborough, B.R.; Nace, G.W.; Ding, Q.; Loughran, P.; Beer-Stolz, D.; Billiar, T.R.; et al. Histones activate the NLRP3 inflammasome in Kupffer cells during sterile inflammatory liver injury. J. Immunol. 2013, 191, 2665–2679. [Google Scholar] [CrossRef]
  153. Dear, J.W.; Simpson, K.J.; Nicolai, M.P.; Catterson, J.H.; Street, J.; Huizinga, T.; Craig, D.G.; Dhaliwal, K.; Webb, S.; Bateman, D.N.; et al. Cyclophilin A is a damage-associated molecular pattern molecule that mediates acetaminophen-induced liver injury. J. Immunol. 2011, 187, 3347–3352. [Google Scholar] [CrossRef] [Green Version]
  154. Mutlu, H.H.; Caklili, O.T.; Coskunpinar, E. Serum concentrations of Cyclophilin A in patients with Nonalcoholic Fatty Liver Disease. Acta Gastroenterol. Belg. 2017, 80, 3–7. [Google Scholar]
  155. Tian, X.; Zhao, C.; Zhu, H.; She, W.; Zhang, J.; Liu, J.; Li, L.; Zheng, S.; Wen, Y.M.; Xie, Y. Hepatitis B virus (HBV) surface antigen interacts with and promotes cyclophilin a secretion: Possible link to pathogenesis of HBV infection. J. Virol. 2010, 84, 3373–3381. [Google Scholar] [CrossRef] [Green Version]
  156. Naoumov, N.V. Cyclophilin inhibition as potential therapy for liver diseases. J. Hepatol. 2014, 61, 1166–1174. [Google Scholar] [CrossRef] [Green Version]
  157. Joly, P.; Marshall, J.C.; Tessier, P.A.; Massé, C.; Page, N.; Frenette, A.J.; Khazoom, F.; Le Guillan, S.; Berthiaume, Y.; Charbonney, E. S100A8/A9 and sRAGE kinetic after polytrauma; an explorative observational study. Scand. J. Trauma Resusc. Emerg. Med. 2017, 25, 114. [Google Scholar] [CrossRef] [Green Version]
  158. Kutcher, M.E.; Xu, J.; Vilardi, R.F.; Ho, C.; Esmon, C.T.; Cohen, M.J. Extracellular histone release in response to traumatic injury: Implications for a compensatory role of activated protein C. J. Trauma Acute Care Surg. 2012, 73, 1389–1394. [Google Scholar] [CrossRef] [Green Version]
  159. St John, A.; Wang, Y.; Chen, J.; Osborn, W.; Wang, X.; Lim, E.; Chung, D.; Stern, S.; White, N.; Fu, X.; et al. Plasma proteomic profile associated with platelet dysfunction after trauma. J. Thromb. Haemost. 2021, 19, 1666–1675. [Google Scholar] [CrossRef]
  160. Gao, Y.; Duan, J.; Ji, H.; Lu, W. Levels of S100 calcium binding protein B (S100B), neuron-specific enolase (NSE), and cyclophilin A (CypA) in the serum of patients with severe craniocerebral injury and multiple injuries combined with delirium transferred from the ICU and their prognostic value. Ann. Palliat. Med. 2021, 10, 3371–3378. [Google Scholar]
  161. Smith, J.G.W.; Owen, T.; Bhagwan, J.R.; Mosqueira, D.; Scott, E.; Mannhardt, I.; Patel, A.; Barriales-Villa, R.; Monserrat, L.; Hansen, A.; et al. Isogenic Pairs of hiPSC-CMs with Hypertrophic Cardiomyopathy/LVNC-Associated ACTC1 E99K Mutation Unveil Differential Functional Deficits. Stem Cell Rep. 2018, 11, 1226–1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Olson, T.M.; Michels, V.V.; Thibodeau, S.N.; Tai, Y.S.; Keating, M.T. Actin mutations in dilated cardiomyopathy, a heritable form of heart failure. Science 1998, 280, 750–752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Lu, M.H.; DiLullo, C.; Schultheiss, T.; Holtzer, S.; Murray, J.M.; Choi, J.; Fischman, D.A.; Holtzer, H. The vinculin/sarcomeric-alpha-actinin/alpha-actin nexus in cultured cardiac myocytes. J. Cell Biol. 1992, 117, 1007–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Willis-Owen, S.A.G.; Thompson, A.; Kemp, P.R.; Polkey, M.I.; Cookson, W.; Moffatt, M.F.; Natanek, S.A. COPD is accompanied by co-ordinated transcriptional perturbation in the quadriceps affecting the mitochondria and extracellular matrix. Sci. Rep. 2018, 8, 12165. [Google Scholar] [CrossRef] [PubMed]
  165. Feng, H.; Li, H.; Zhang, D.; Zhao, Y.; Jiang, N.; Zhao, X.; Zhang, Y.U.; Tan, J.; Fang, W.; Zhang, Y.; et al. Aortic wall proteomic analysis in spontaneously hypertensive rats with a blood pressure decrease induced by 6-week load-free swimming. Biomed. Rep. 2015, 3, 681–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Liu, Y.; Wang, Y.; Teng, Z.; Zhang, X.; Ding, M.; Zhang, Z.; Chen, J.; Xu, Y. DNA Microarray Analysis in Screening Features of Genes Involved in Spinal Cord Injury. Med. Sci. Monit. 2016, 22, 1571–1581. [Google Scholar] [CrossRef] [Green Version]
  167. Yilmaz, Z.; Eralp Inan, O.; Kocaturk, M.; Baykal, A.T.; Hacariz, O.; Hatipoglu, I.; Tvarijonaviciute, A.; Cansev, M.; Ceron, J.; Ulus, I.H. Changes in serum proteins after endotoxin administration in healthy and choline-treated calves. BMC Vet. Res. 2016, 12, 210. [Google Scholar] [CrossRef] [Green Version]
  168. Aránega, A.E.; Reina, A.; Muros, M.A.; Alvarez, L.; Prados, J.; Aránega, A. Circulating alpha-actin protein in acute myocardial infarction. Int. J. Cardiol. 1993, 38, 49–55. [Google Scholar] [CrossRef]
  169. De’Ath, H.D.; Rourke, C.; Davenport, R.; Manson, J.; Renfrew, I.; Uppal, R.; Davies, L.C.; Brohi, K. Clinical and biomarker profile of trauma-induced secondary cardiac injury. Br. J. Surg. 2012, 99, 789–797. [Google Scholar] [CrossRef]
  170. Martin, M.; Mullenix, P.; Rhee, P.; Belzberg, H.; Demetriades, D.; Salim, A. Troponin increases in the critically injured patient: Mechanical trauma or physiologic stress? J. Trauma Acute Care Surg. 2005, 59, 1086–1091. [Google Scholar] [CrossRef]
  171. Baur, M.; Weber, B.; Lackner, I.; Gebhard, F.; Pfeifer, R.; Cinelli, P.; Halvachizadeh, S.; Teuben, M.; Lipiski, M.; Cesarovic, N.; et al. Structural alterations and inflammation in the heart after multiple trauma followed by reamed versus non-reamed femoral nailing. PLoS ONE 2020, 15, e0235220. [Google Scholar] [CrossRef]
  172. De’Ath, H.D.; Manson, J.; Davenport, R.; Glasgow, S.; Renfrew, I.; Davies, L.C.; Uppal, R.; Brohi, K. Trauma-induced secondary cardiac injury is associated with hyperacute elevations in inflammatory cytokines. Trauma-induced secondary cardiac injury is associated with hyperacute elevations in inflammatory cytokines. Shock 2013, 39, 415–420. [Google Scholar] [CrossRef]
  173. Wilson, N.M.; Wall, J.; Naganathar, V.; Brohi, K.; De’Ath, H.D. Mechanisms Involved in Secondary Cardiac Dysfunction in Animal Models of Trauma and Hemorrhagic Shock. Shock 2017, 48, 401–410. [Google Scholar] [CrossRef]
  174. Naganathar, S.; De’Ath, H.D.; Wall, J.; Brohi, K. Admission biomarkers of trauma-induced secondary cardiac injury predict adverse cardiac events and are associated with plasma catecholamine levels. J. Trauma Acute Care Surg. 2015, 79, 71–77. [Google Scholar] [CrossRef]
  175. Gawande, N.B.; Tumram, N.K.; Dongre, A.P. Cardiac changes in hospitalized patients of trauma. Shock 2014, 42, 211–217. [Google Scholar] [CrossRef]
  176. Cebelin, M.S.; Hirsch, C.S. Human stress cardiomyopathy. Myocardial lesions in victims of homicidal assaults without internal injuries. Hum. Pathol. 1980, 11, 123–132. [Google Scholar] [CrossRef]
  177. Wall, J.; Naganathar, S.; Praditsuktavorn, B.; Bugg, O.F.; McArthur, S.; Thiemermann, C.; Tremoleda, J.L.; Brohi, K. Modeling Cardiac Dysfunction Following Traumatic Hemorrhage Injury: Impact on Myocardial Integrity. Front. Immunol. 2019, 10, 2774. [Google Scholar] [CrossRef] [Green Version]
  178. Erukhimov, J.A.; Tang, Z.L.; Johnson, B.A.; Donahoe, M.P.; Razzack, J.A.; Gibson, K.F.; Lee, W.M.; Wasserloos, K.J.; Watkins, S.A.; Pitt, B.R. Actin-containing sera from patients with adult respiratory distress syndrome are toxic to sheep pulmonary endothelial cells. Am. J. Respir. Crit. Care Med. 2000, 162, 288–294. [Google Scholar] [CrossRef]
  179. Hazeldine, J.; Dinsdale, R.J.; Naumann, D.N.; Acharjee, A.; Bishop, J.R.B.; Lord, J.M.; Harrison, P. Traumatic injury is associated with reduced deoxyribonuclease activity and dysregulation of the actin scavenging system. Burns Trauma 2021, 9, tkab001. [Google Scholar] [CrossRef]
  180. Dinsdale, R.J.; Hazeldine, J.; Al Tarrah, K.; Hampson, P.; Devi, A.; Ermogenous, C.; Bamford, A.L.; Bishop, J.; Watts, S.; Kirkman, E.; et al. Dysregulation of the actin scavenging system and inhibition of DNase activity following severe thermal injury. Br. J. Surg. 2020, 107, 391–401. [Google Scholar] [CrossRef] [Green Version]
  181. Coleman, J.R.; Moore, E.E.; Freeman, K.; Grubinger, N.D.; Hennig, G.W.; Cohen, M.J.; Samuels, J.M.; Hansen, K. Actin is associated with tissue injury in trauma patients and produces a hypercoagulable profile in vitro. J. Trauma Acute Care Surg. 2020, 89, 87–95. [Google Scholar] [CrossRef]
  182. Ordija, C.M.; Chiou, T.T.; Yang, Z.; Deloid, G.M.; de Oliveira Valdo, M.; Wang, Z.; Bedugnis, A.; Noah, T.L.; Jones, S.; Koziel, H.; et al. Free actin impairs macrophage bacterial defenses via scavenger receptor MARCO interaction with reversal by plasma gelsolin. Am. J. Physiol. Lung Cell Mol. Physiol. 2017, 312, L1018–L1028. [Google Scholar] [CrossRef]
  183. Dahl, B.; Schiødt, F.V.; Rudolph, S.; Ott, P.; Kiaer, T.; Heslet, L. Trauma stimulates the synthesis of Gc-globulin. Intensive Care Med. 2001, 27, 394–399. [Google Scholar] [CrossRef]
  184. Dahl, B.; Schiødt, F.V.; Kiaer, T.; Ott, P.; Bondesen, S.; Tygstrup, N. Serum Gc-globulin in the early course of multiple trauma. Crit. Care Med. 1998, 26, 285–289. [Google Scholar] [CrossRef]
  185. Ley, E.J.; Brown, C.V.R.; Moore, E.E.; Sava, J.A.; Peck, K.; Ciesla, D.J.; Sperry, J.L.; Rizzo, A.G.; Rosen, N.G.; Brasel, K.J.; et al. Updated guidelines to reduce venous thromboembolism in trauma patients: A Western Trauma Association critical decisions algorithm. J. Trauma Acute Care Surg. 2020, 89, 971–981. [Google Scholar] [CrossRef]
  186. Shackford, S.R.; Cipolle, M.D.; Badiee, J.; Mosby, D.L.; Knudson, M.M.; Lewis, P.R.; McDonald, V.S.; Olson, E.J.; Thompson, K.A.; Van Gent, J.M.; et al. Determining the magnitude of surveillance bias in the assessment of lower extremity deep venous thrombosis: A prospective observational study of two centers. J. Trauma Acute Care Surg. 2016, 80, 734–739. [Google Scholar] [CrossRef]
  187. Thorson, C.M.; Ryan, M.L.; Van Haren, R.M.; Curia, E.; Barrera, J.M.; Guarch, G.A.; Busko, A.M.; Namias, N.; Livingstone, A.S.; Proctor, K.G. Venous thromboembolism after trauma: A never event?*. Crit. Care Med. 2012, 40, 2967–2973. [Google Scholar] [CrossRef] [PubMed]
  188. Hamada, S.R.; Espina, C.; Guedj, T.; Buaron, R.; Harrois, A.; Figueiredo, S.; Duranteau, J. High level of venous thromboembolism in critically ill trauma patients despite early and well-driven thromboprophylaxis protocol. Ann. Intensive Care 2017, 7, 97. [Google Scholar] [CrossRef] [PubMed]
  189. Velmahos, G.C.; Spaniolas, K.; Tabbara, M.; Abujudeh, H.H.; de Moya, M.; Gervasini, A.; Alam, H.B. Pulmonary embolism and deep venous thrombosis in trauma: Are they related? Arch. Surg. 2009, 144, 928–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Van Gent, J.M.; Zander, A.L.; Olson, E.J.; Shackford, S.R.; Dunne, C.E.; Sise, C.B.; Badiee, J.; Schechter, M.S.; Sise, M.J. Pulmonary embolism without deep venous thrombosis: De novo or missed deep venous thrombosis? J. Trauma Acute Care Surg. 2014, 76, 1270–1274. [Google Scholar] [CrossRef] [PubMed]
  191. Van Langevelde, K.; Srámek, A.; Vincken, P.W.; van Rooden, J.K.; Rosendaal, F.R.; Cannegieter, S.C. Finding the origin of pulmonary emboli with a total-body magnetic resonance direct thrombus imaging technique. Haematologica 2013, 98, 309–315. [Google Scholar] [CrossRef] [Green Version]
  192. Pelsers, M.M.; Namiot, Z.; Kisielewski, W.; Namiot, A.; Januszkiewicz, M.; Hermens, W.T.; Glatz, J.F. Intestinal-type and liver-type fatty acid-binding protein in the intestine. Tissue distribution and clinical utility. Clin. Biochem. 2003, 36, 529–535. [Google Scholar] [CrossRef]
  193. Wang, G.; Bonkovsky, H.L.; de Lemos, A.; Burczynski, F.J. Recent insights into the biological functions of liver fatty acid binding protein 1. J. Lipid Res. 2015, 56, 2238–2247. [Google Scholar] [CrossRef] [Green Version]
  194. Relja, B.; Szermutzky, M.; Henrich, D.; Maier, M.; de Haan, J.J.; Lubbers, T.; Buurman, W.A.; Marzi, I. Intestinal-FABP and liver-FABP: Novel markers for severe abdominal injury. Acad. Emerg. Med. 2010, 17, 729–735. [Google Scholar] [CrossRef]
  195. Voth, M.; Holzberger, S.; Auner, B.; Henrich, D.; Marzi, I.; Relja, B. I-FABP and L-FABP are early markers for abdominal injury with limited prognostic value for secondary organ failures in the post-traumatic course. Clin. Chem. Lab. Med. 2015, 53, 771–780. [Google Scholar] [CrossRef]
  196. Van de Poll, M.C.; Derikx, J.P.; Buurman, W.A.; Peters, W.H.; Roelofs, H.M.; Wigmore, S.J.; Dejong, C.H. Liver manipulation causes hepatocyte injury and precedes systemic inflammation in patients undergoing liver resection. World J. Surg. 2007, 31, 2033–2038. [Google Scholar] [CrossRef] [Green Version]
  197. Van den Broek, M.A.; Bloemen, J.G.; Dello, S.A.; van de Poll, M.C.; Olde Damink, S.W.; Dejong, C.H. Randomized controlled trial analyzing the effect of 15 or 30 min intermittent Pringle maneuver on hepatocellular damage during liver surgery. J. Hepatol. 2011, 55, 337–345. [Google Scholar] [CrossRef]
  198. Van den Broek, M.A.; Shiri-Sverdlov, R.; Schreurs, J.J.; Bloemen, J.G.; Bieghs, V.; Rensen, S.S.; Dejong, C.H.; Olde Damink, S.W. Liver manipulation during liver surgery in humans is associated with hepatocellular damage and hepatic inflammation. Liver Int. 2013, 33, 633–641. [Google Scholar] [CrossRef]
  199. Hanssen, S.J.; Derikx, J.P.; Vermeulen Windsant, I.C.; Heijmans, J.H.; Koeppel, T.A.; Schurink, G.W.; Buurman, W.A.; Jacobs, M.J. Visceral injury and systemic inflammation in patients undergoing extracorporeal circulation during aortic surgery. Ann. Surg. 2008, 248, 117–125. [Google Scholar] [CrossRef]
  200. Kunihara, T.; Kubota, S.; Shiiya, N.; Iizuka, K.; Sasaki, S.; Wakasa, S.; Matsuzaki, K.; Matsui, Y. Cytokine balance in hepatosplanchnic system during thoracoabdominal aortic aneurysm repair. J. Artif. Organs 2011, 14, 192–200. [Google Scholar] [CrossRef]
  201. Crapo, J.D.; Oury, T.; Rabouille, C.; Slot, J.W.; Chang, L.Y. Copper, zinc superoxide dismutase is primarily a cytosolic protein in human cells. Proc. Natl. Acad. Sci. USA 1992, 89, 10405–10409. [Google Scholar] [CrossRef] [Green Version]
  202. Pérez-Miguelsanz, J.; Vallecillo, N.; Garrido, F.; Reytor, E.; Pérez-Sala, D.; Pajares, M.A. Betaine homocysteine S-methyltransferase emerges as a new player of the nuclear methionine cycle. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1165–1182. [Google Scholar] [CrossRef] [Green Version]
  203. Jurkowska, H.; Niewiadomski, J.; Hirschberger, L.L.; Roman, H.B.; Mazor, K.M.; Liu, X.; Locasale, J.W.; Park, E.; Stipanuk, M.H. Downregulation of hepatic betaine:homocysteine methyltransferase (BHMT) expression in taurine-deficient mice is reversed by taurine supplementation in vivo. Amino Acids 2016, 48, 665–676. [Google Scholar] [CrossRef] [Green Version]
  204. Lambert, I.H. Reactive oxygen species regulate swelling-induced taurine efflux in NIH3T3 mouse fibroblasts. J. Membr. Biol. 2003, 192, 19–32. [Google Scholar] [CrossRef]
  205. Abebe, W.; Mozaffari, M.S. Taurine depletion alters vascular reactivity in rats. Can. J. Physiol. Pharmacol. 2003, 81, 903–909. [Google Scholar] [CrossRef]
  206. Sun, Q.; Wang, B.; Li, Y.; Sun, F.; Li, P.; Xia, W.; Zhou, X.; Li, Q.; Wang, X.; Chen, J.; et al. Taurine Supplementation Lowers Blood Pressure and Improves Vascular Function in Prehypertension: Randomized, Double-Blind, Placebo-Controlled Study. Hypertension 2016, 67, 541–549. [Google Scholar] [CrossRef] [Green Version]
  207. Ristori, M.T.; Verdetti, J. Effects of taurine on rat aorta in vitro. Fundam. Clin. Pharmacol. 1991, 5, 245–258. [Google Scholar] [CrossRef]
  208. Innes, D.; Sevitt, S. Coagulation and fibrinolysis in injured patients. J. Clin. Pathol. 1964, 17, 1–13. [Google Scholar] [CrossRef] [Green Version]
  209. Floccard, B.; Rugeri, L.; Faure, A.; Saint Denis, M.; Boyle, E.M.; Peguet, O.; Levrat, A.; Guillaume, C.; Marcotte, G.; Vulliez, A.; et al. Early coagulopathy in trauma patients: An on-scene and hospital admission study. Injury 2012, 43, 26–32. [Google Scholar] [CrossRef] [PubMed]
  210. Carroll, R.C.; Craft, R.M.; Langdon, R.J.; Clanton, C.R.; Snider, C.C.; Wellons, D.D.; Dakin, P.A.; Lawson, C.M.; Enderson, B.L.; Kurek, S.J. Early evaluation of acute traumatic coagulopathy by thrombelastography. Transl. Res. 2009, 154, 34–39. [Google Scholar] [CrossRef] [PubMed]
  211. Rizoli, S.B.; Scarpelini, S.; Callum, J.; Nascimento, B.; Mann, K.G.; Pinto, R.; Jansen, J.; Tien, H.C. Clotting factor deficiency in early trauma-associated coagulopathy. J. Trauma 2011, 71 (Suppl. 1), S427–S434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Woolley, T.; Gwyther, R.; Parmar, K.; Kirkman, E.; Watts, S.; Midwinter, M.; Lucca, J.D.; Hunt, B.J. A prospective observational study of acute traumatic coagulopathy in traumatic bleeding from the battlefield. Transfusion 2020, 60 (Suppl. 3), S52–S61. [Google Scholar] [CrossRef] [PubMed]
  213. Jansen, J.O.; Scarpelini, S.; Pinto, R.; Tien, H.C.; Callum, J.; Rizoli, S.B. Hypoperfusion in severely injured trauma patients is associated with reduced coagulation factor activity. J. Trauma 2011, 71 (Suppl. 1), S435–S440. [Google Scholar] [CrossRef]
  214. Theusinger, O.M.; Baulig, W.; Seifert, B.; Müller, S.M.; Mariotti, S.; Spahn, D.R. Changes in coagulation in standard laboratory tests and ROTEM in trauma patients between on-scene and arrival in the emergency department. Anesth. Analg. 2015, 120, 627–635. [Google Scholar] [CrossRef] [Green Version]
  215. Camire, R.M.; Pollak, E.S.; Kaushansky, K.; Tracy, P.B. Secretable human platelet-derived factor V originates from the plasma pool. Blood 1998, 92, 3035–3041. [Google Scholar] [CrossRef]
  216. Tracy, P.B.; Eide, L.L.; Bowie, E.J.; Mann, K.G. Radioimmunoassay of factor V in human plasma and platelets. Blood 1982, 60, 59–63. [Google Scholar] [CrossRef] [Green Version]
  217. Chesney, C.M.; Pifer, D.; Colman, R.W. Subcellular localization and secretion of factor V from human platelets. Proc. Natl. Acad. Sci. USA 1981, 78, 5180–5184. [Google Scholar] [CrossRef] [Green Version]
  218. Wencel-Drake, J.D.; Dahlback, B.; White, J.G.; Ginsberg, M.H. Ultrastructural localization of coagulation factor V in human platelets. Blood 1986, 68, 244–249. [Google Scholar] [CrossRef]
  219. Tracy, P.B.; Giles, A.R.; Mann, K.G.; Eide, L.L.; Hoogendoorn, H.; Rivard, G.E. Factor V (Quebec): A bleeding diathesis associated with a qualitative platelet Factor V deficiency. J. Clin. Investig. 1984, 74, 1221–1228. [Google Scholar] [CrossRef]
  220. Kahr, W.H.; Zheng, S.; Sheth, P.M.; Pai, M.; Cowie, A.; Bouchard, M.; Podor, T.J.; Rivard, G.E.; Hayward, C.P. Platelets from patients with the Quebec platelet disorder contain and secrete abnormal amounts of urokinase-type plasminogen activator. Blood 2001, 98, 257–265. [Google Scholar] [CrossRef]
  221. Bertina, R.M.; Koeleman, B.P.; Koster, T.; Rosendaal, F.R.; Dirven, R.J.; de Ronde, H.; van der Velden, P.A.; Reitsma, P.H. Mutation in blood coagulation factor V associated with resistance to activated protein C. Nature 1994, 369, 64–67. [Google Scholar] [CrossRef]
  222. Ridker, P.M.; Miletich, J.P.; Hennekens, C.H.; Buring, J.E. Ethnic distribution of factor V Leiden in 4047 men and women. Implications for venous thromboembolism screening. JAMA 1997, 277, 1305–1307. [Google Scholar] [CrossRef]
  223. Zivelin, A.; Griffin, J.H.; Xu, X.; Pabinger, I.; Samama, M.; Conard, J.; Brenner, B.; Eldor, A.; Seligsohn, U. A single genetic origin for a common Caucasian risk factor for venous thrombosis. Blood 1997, 89, 397–402. [Google Scholar] [CrossRef]
  224. Van Mens, T.E.; Levi, M.; Middeldorp, S. Evolution of Factor V Leiden. Thromb. Haemost. 2013, 110, 23–30. [Google Scholar]
  225. Lindqvist, P.G.; Dahlbäck, B. Carriership of Factor V Leiden and evolutionary selection advantage. Curr. Med. Chem. 2008, 15, 1541–1544. [Google Scholar] [CrossRef]
  226. Lindqvist, P.G.; Svensson, P.J.; Dahlbäck, B.; Marsál, K. Factor V Q506 mutation (activated protein C resistance) associated with reduced intrapartum blood loss—A possible evolutionary selection mechanism. Thromb. Haemost. 1998, 79, 69–73. [Google Scholar]
  227. Lindqvist, P.G.; Svensson, P.J.; Marsaál, K.; Grennert, L.; Luterkort, M.; Dahlbäck, B. Activated protein C resistance (FV:Q506) and pregnancy. Thromb. Haemost. 1999, 81, 532–537. [Google Scholar] [CrossRef]
  228. Lindqvist, P.G.; Zöller, B.; Dahlbäck, B. Improved hemoglobin status and reduced menstrual blood loss among female carriers of factor V Leiden--an evolutionary advantage? Thromb. Haemost. 2001, 86, 1122–1123. [Google Scholar] [PubMed]
  229. Gando, S.; Shiraishi, A.; Wada, T.; Yamakawa, K.; Fujishima, S.; Saitoh, D.; Kushimoto, S.; Ogura, H.; Abe, T.; Otomo, Y. A multicenter prospective validation study on disseminated intravascular coagulation in trauma-induced coagulopathy. J. Thromb. Haemost. 2020, 18, 2232–2244. [Google Scholar] [CrossRef] [PubMed]
  230. Raza, I.; Davenport, R.; Rourke, C.; Platton, S.; Manson, J.; Spoors, C.; Khan, S.; De’Ath, H.D.; Allard, S.; Hart, D.P.; et al. The incidence and magnitude of fibrinolytic activation in trauma patients. J. Thromb. Haemost. 2013, 11, 307–314. [Google Scholar] [CrossRef] [PubMed]
  231. Blombäck, M.; Eklund, J.; Hellgren, M.; Lagerkranser, M.; Swedenborg, J. Blood coagulation and fibrinolytic factors as well as their inhibitors in trauma. Scand. J. Clin. Lab. Investig. Suppl. 1985, 178, 15–23. [Google Scholar]
  232. Cardenas, J.C.; Matijevic, N.; Baer, L.A.; Holcomb, J.B.; Cotton, B.A.; Wade, C.E. Elevated tissue plasminogen activator and reduced plasminogen activator inhibitor promote hyperfibrinolysis in trauma patients. Shock 2014, 41, 514–521. [Google Scholar] [CrossRef]
  233. Cardenas, J.C.; Wade, C.E.; Cotton, B.A.; George, M.J.; Holcomb, J.B.; Schreiber, M.A.; White, N.J. TEG Lysis Shutdown Represents Coagulopathy in Bleeding Trauma Patients: Analysis of the PROPPR Cohort. Shock 2019, 51, 273–283. [Google Scholar] [CrossRef]
  234. Chapman, M.P.; Moore, E.E.; Moore, H.B.; Gonzalez, E.; Gamboni, F.; Chandler, J.G.; Mitra, S.; Ghasabyan, A.; Chin, T.L.; Sauaia, A.; et al. Overwhelming tPA release, not PAI-1 degradation, is responsible for hyperfibrinolysis in severely injured trauma patients. J. Trauma Acute Care Surg. 2016, 80, 16–23. [Google Scholar] [CrossRef] [Green Version]
  235. Hasan, A.A.; Cines, D.B.; Ngaiza, J.R.; Jaffe, E.A.; Schmaier, A.H. High-molecular-weight kininogen is exclusively membrane bound on endothelial cells to influence activation of vascular endothelium. Blood 1995, 85, 3134–3143. [Google Scholar] [CrossRef]
  236. Kolte, D.; Osman, N.; Yang, J.; Shariat-Madar, Z. High molecular weight kininogen activates B2 receptor signaling pathway in human vascular endothelial cells. J. Biol. Chem. 2011, 286, 24561–24571. [Google Scholar] [CrossRef] [Green Version]
  237. Kaplan, A.P.; Ghebrehiwet, B. The plasma bradykinin-forming pathways and its interrelationships with complement. Mol. Immunol. 2010, 47, 2161–2169. [Google Scholar] [CrossRef]
  238. Reddigari, S.; Silverberg, M.; Kaplan, A.P. Assembly of the human plasma kinin-forming cascade along the surface of vascular endothelial cells. Int. Arch. Allergy Immunol. 1995, 107, 93–94. [Google Scholar] [CrossRef]
  239. Murray, M.A.; Heistad, D.D.; Mayhan, W.G. Role of protein kinase C in bradykinin-induced increases in microvascular permeability. Circ. Res. 1991, 68, 1340–1348. [Google Scholar] [CrossRef] [Green Version]
  240. Oh-ishi, S.; Hayashi, I.; Yamaki, K.; Utsunomiya, I.; Hayashi, M.; Yamasu, A.; Nakano, T. Role of high molecular weight (HMW)-kininogen in inflammatory exudation: Evidence with the studies of the HMW-kininogen deficient rat. Adv. Exp. Med. Biol. 1989, 247a, 145–152. [Google Scholar]
  241. Kaplan, A.P.; Ghebrehiwet, B.; Silverberg, M.; Sealey, J.E. The intrinsic coagulation-kinin pathway, complement cascades, plasma renin-angiotensin system, and their interrelationships. Crit. Rev. Immunol. 1981, 3, 75–93. [Google Scholar]
  242. Cap, A.P. Plasmin: A driver of hemovascular dysfunction. Blood 2016, 128, 2375–2376. [Google Scholar] [CrossRef] [Green Version]
  243. Renné, T.; Schuh, K.; Müller-Esterl, W. Local bradykinin formation is controlled by glycosaminoglycans. J. Immunol. 2005, 175, 3377–3385. [Google Scholar] [CrossRef] [Green Version]
  244. Smith, A.; Ledford, B.E. Expression of the haemopexin-transport system in cultured mouse hepatoma cells. Links between haemopexin and iron metabolism. Biochem. J. 1988, 256, 941–950. [Google Scholar] [CrossRef] [Green Version]
  245. Montecinos, L.; Eskew, J.D.; Smith, A. What Is Next in This “Age” of Heme-Driven Pathology and Protection by Hemopexin? An Update and Links with Iron. Pharmaceuticals 2019, 12, 144. [Google Scholar] [CrossRef] [Green Version]
  246. Wagener, B.M.; Hu, P.J.; Oh, J.Y.; Evans, C.A.; Richter, J.R.; Honavar, J.; Brandon, A.P.; Creighton, J.; Stephens, S.W.; Morgan, C.; et al. Role of heme in lung bacterial infection after trauma hemorrhage and stored red blood cell transfusion: A preclinical experimental study. PLoS Med. 2018, 15, e1002522. [Google Scholar] [CrossRef] [Green Version]
  247. Morgan, H.P.; Schmidt, C.Q.; Guariento, M.; Blaum, B.S.; Gillespie, D.; Herbert, A.P.; Kavanagh, D.; Mertens, H.D.; Svergun, D.I.; Johansson, C.M.; et al. Structural basis for engagement by complement factor H of C3b on a self surface. Nat. Struct. Mol. Biol. 2011, 18, 463–470. [Google Scholar]
  248. Jokiranta, T.S.; Cheng, Z.Z.; Seeberger, H.; Jòzsi, M.; Heinen, S.; Noris, M.; Remuzzi, G.; Ormsby, R.; Gordon, D.L.; Meri, S.; et al. Binding of complement factor H to endothelial cells is mediated by the carboxy-terminal glycosaminoglycan binding site. Am. J. Pathol. 2005, 167, 1173–1181. [Google Scholar] [CrossRef]
  249. Ruggenenti, P.; Noris, M.; Remuzzi, G. Thrombotic microangiopathy, hemolytic uremic syndrome, and thrombotic thrombocytopenic purpura. Kidney Int. 2001, 60, 831–846. [Google Scholar] [CrossRef] [Green Version]
  250. Dopler, A.; Guntau, L.; Harder, M.J.; Palmer, A.; Höchsmann, B.; Schrezenmeier, H.; Simmet, T.; Huber-Lang, M.; Schmidt, C.Q. Self versus Nonself Discrimination by the Soluble Complement Regulators Factor H and FHL-1. J. Immunol. 2019, 202, 2082–2094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Matsushita, M.; Kuraya, M.; Hamasaki, N.; Tsujimura, M.; Shiraki, H.; Fujita, T. Activation of the lectin complement pathway by H-ficolin (Hakata antigen). J. Immunol. 2002, 168, 3502–3506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Merle, N.S.; Church, S.E.; Fremeaux-Bacchi, V.; Roumenina, L.T. Complement System Part I—Molecular Mechanisms of Activation and Regulation. Front. Immunol. 2015, 6, 262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Gorski, J.P.; Hugli, T.E.; Müller-Eberhard, H.J. C4a: The third anaphylatoxin of the human complement system. Proc. Natl. Acad. Sci. USA 1979, 76, 5299–5302. [Google Scholar] [CrossRef] [Green Version]
  254. Saadi, S.; Holzknecht, R.A.; Patte, C.P.; Stern, D.M.; Platt, J.L. Complement-mediated regulation of tissue factor activity in endothelium. J. Exp. Med. 1995, 182, 1807–1814. [Google Scholar] [CrossRef]
  255. Hattori, R.; Hamilton, K.K.; McEver, R.P.; Sims, P.J. Complement proteins C5b-9 induce secretion of high molecular weight multimers of endothelial von Willebrand factor and translocation of granule membrane protein GMP-140 to the cell surface. J. Biol. Chem. 1989, 264, 9053–9060. [Google Scholar] [CrossRef]
  256. Kilgore, K.S.; Shen, J.P.; Miller, B.F.; Ward, P.A.; Warren, J.S. Enhancement by the complement membrane attack complex of tumor necrosis factor-alpha-induced endothelial cell expression of E-selectin and ICAM-1. J. Immunol. 1995, 155, 1434–1441. [Google Scholar]
  257. Kilgore, K.S.; Flory, C.M.; Miller, B.F.; Evans, V.M.; Warren, J.S. The membrane attack complex of complement induces interleukin-8 and monocyte chemoattractant protein-1 secretion from human umbilical vein endothelial cells. Am. J. Pathol. 1996, 149, 953–961. [Google Scholar]
  258. Kilgore, K.S.; Schmid, E.; Shanley, T.P.; Flory, C.M.; Maheswari, V.; Tramontini, N.L.; Cohen, H.; Ward, P.A.; Friedl, H.P.; Warren, J.S. Sublytic concentrations of the membrane attack complex of complement induce endothelial interleukin-8 and monocyte chemoattractant protein-1 through nuclear factor-kappa B activation. Am. J. Pathol. 1997, 150, 2019–2031. [Google Scholar]
  259. Dobrina, A.; Pausa, M.; Fischetti, F.; Bulla, R.; Vecile, E.; Ferrero, E.; Mantovani, A.; Tedesco, F. Cytolytically inactive terminal complement complex causes transendothelial migration of polymorphonuclear leukocytes in vitro and in vivo. Blood 2002, 99, 185–192. [Google Scholar] [CrossRef]
  260. Tedesco, F.; Pausa, M.; Nardon, E.; Introna, M.; Mantovani, A.; Dobrina, A. The cytolytically inactive terminal complement complex activates endothelial cells to express adhesion molecules and tissue factor procoagulant activity. J. Exp. Med. 1997, 185, 1619–1627. [Google Scholar] [CrossRef] [Green Version]
  261. Atefi, G.; Aisiku, O.; Shapiro, N.; Hauser, C.; Dalle Lucca, J.; Flaumenhaft, R.; Tsokos, G.C. Complement Activation in Trauma Patients Alters Platelet Function. Shock 2016, 46 (Suppl. 1), 83–88. [Google Scholar] [CrossRef]
  262. Del Conde, I.; Crúz, M.A.; Zhang, H.; López, J.A.; Afshar-Kharghan, V. Platelet activation leads to activation and propagation of the complement system. J. Exp. Med. 2005, 201, 871–879. [Google Scholar] [CrossRef] [Green Version]
  263. Muroya, T.; Kannan, L.; Ghiran, I.C.; Shevkoplyas, S.S.; Paz, Z.; Tsokos, M.; Dalle Lucca, J.J.; Shapiro, N.I.; Tsokos, G.C. C4d deposits on the surface of RBCs in trauma patients and interferes with their function. Crit. Care Med. 2014, 42, e364–e372. [Google Scholar] [CrossRef] [PubMed]
  264. Calfee, C.S.; Delucchi, K.; Parsons, P.E.; Thompson, B.T.; Ware, L.B.; Matthay, M.A. Subphenotypes in acute respiratory distress syndrome: Latent class analysis of data from two randomised controlled trials. Lancet Respir. Med. 2014, 2, 611–620. [Google Scholar] [CrossRef] [Green Version]
  265. Calfee, C.S.; Delucchi, K.L.; Sinha, P.; Matthay, M.A.; Hackett, J.; Shankar-Hari, M.; McDowell, C.; Laffey, J.G.; O’Kane, C.M.; McAuley, D.F. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: Secondary analysis of a randomised controlled trial. Lancet Respir. Med. 2018, 6, 691–698. [Google Scholar] [CrossRef] [Green Version]
  266. Wiggers, H.C.; Ingraham, R.C.; Dille, J. Hemorrhagic-hypotension shock in locally anesthetized dogs. Am. J. Physiol. Leg. Content 1945, 143, 126–133. [Google Scholar] [CrossRef] [Green Version]
  267. Fine, J.; Seligman, A.M. Traumatic shock: Iv. a study of the problem of the “lost plasma” in hemorrhagic shock by the use of radioactive plasma protein. J. Clin. Investig. 1943, 22, 285–303. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Volcano plot featuring differentially expressed proteins between the EoT and non-EoT group. EoT: endotheliopathy of trauma defined by plasma syndecan-1 ≥ 40 ng/mL; p-value: Benjamini and Hochberg false discovery rate-adjusted p-value; labeled proteins represent an adjusted p-value < 0.05.
Figure 1. Volcano plot featuring differentially expressed proteins between the EoT and non-EoT group. EoT: endotheliopathy of trauma defined by plasma syndecan-1 ≥ 40 ng/mL; p-value: Benjamini and Hochberg false discovery rate-adjusted p-value; labeled proteins represent an adjusted p-value < 0.05.
Ijms 23 06213 g001
Table 1. Demographics, clinical data, laboratory values and outcomes of trauma patients by EoT status. Data are presented as the median (IQR) for continuous variables and n (%) for categorical and ordinal variables.
Table 1. Demographics, clinical data, laboratory values and outcomes of trauma patients by EoT status. Data are presented as the median (IQR) for continuous variables and n (%) for categorical and ordinal variables.
nAll Trauma Subjects
(n = 99)
nNon-EoT
(n = 37)
nEoT
(n = 62)
p-Value
Demographics
Age (years)9944 (34, 54)3746 (36, 58)6244 (32, 51)0.265
Body mass index9627.95 (25, 31)3728.3 (25, 31)5927.2 (25, 31)0.698
Male9973 (74%)3725 (68%)6248 (77%)0.347
Race99 37 62 0.616
White 70 (71%) 42 (67.74%) 28 (75.68%)
African American 27 (27%) 9 (24%) 18 (29%)
Asian/Pacific Islander 2 (2%) 1 (2.7%) 1 (1.6%)
Ethnicity99 37 62 1
Hispanic 23 (24%) 8 (22%) 15 (25%)
Injury Characteristics
Injury severity score9925 (16, 29)3722 (9, 25)6226 (22, 34)<0.001 *
Head AIS 0 (0, 0) 0 (0,0) 0 (0,0)0.907
Face AIS 0 (0, 0) 0 (0,0) 0 (0,0)0.154
Chest AIS 3 (0, 4) 0 (0, 3) 3 (0,4)0.026 *
Abdomen AIS 0 (0, 4) 0 (0, 2) 2 (0, 4)0.001 *
Extremity AIS 0 (0, 3) 0 (0, 3) 0 (0, 3)0.177
External AIS 1 (0, 2) 1 (0, 3) 1 (1, 2)0.188
Mechanism of injury99 37 62 0.005 *
Blunt 55 (56%) 16 (43%) 39 (63%)
Penetrating 25 (25%) 8 (22%) 17 (27%)
Burn 19 (19%) 13 (35%) 6 (10%)
Admission Vital Signs and Labs
Glasgow Coma Scale99 37 62 0.018 *
3 37 (37.4%) 10 (27.0%) 27 (43.5%)
7 2 (2.0%) 0 (0.0%) 2 (3.2%)
8 1 (1.0%) 0 (0.0%) 1 (1.6%)
9 2 (2.0%) 0 (0.0%) 2 (3.2%)
11 1 (1.0%) 0 (0.0%) 1 (1.6%)
13 2 (2.0%) 1 (2.7%) 1 (1.6%)
14 5 (5.1%) 1 (2.7%) 4 (6.5%)
15 49 (49.5%) 25 (67.6%) 24 (38.7%)
Systolic BP (mm Hg)97118 (99, 137)35125 (113, 139)62110 (97, 130)0.018
Heart rate (bpm)97100 (83, 116)3596 (79, 110)62101 (84, 121)0.252
Creatinine (mg/dL)971.21 (1.00, 1.47)371.08 (0.95, 1.30)601.32 (1.01, 1.57)0.003 *
Glucose (mg/dL)97163 (121, 213)37153 (117, 180)60180 (133, 230)0.020 *
Albumin (g/dL)343.10 (2.73, 3.40)133.00 (2.90, 3.50)213.10 (2.70, 3.40)0.845
Base excess (mmol/L)98−6 (−9, −2)37−2 (−6, −2)61−8 (−12, −4)<0.001 *
pH967.26 (7.18, 7.35)367.34 (7.28, 7.35)607.20 (7.10, 7.28)<0.001 *
Platelet count (K/uL)95230 (187, 281)37242 (199, 314)58215 (164, 277)0.127
ACT (s)91113 (105, 121)34105 (105, 113)57113 (105, 121)0.047 *
TEG R-time (min)910.7 (0.6, 0.8)340.6 (0.6, 0.7)570.70 (0.6, 0.8)0.091
TEG K-time (min)901.30 (1.10, 1.80)341.20 (1.02, 1.50)561.45 (1.20, 1.80)0.023 *
TEG α-angle (degrees)9174 (70, 77)3476 (72,78)5773 (69, 76)0.016 *
TEG maximum amplitude (mm)9164 (59, 68)3466 (62, 69)5762 (58, 66)0.002 *
TEG G-value (Kdynes/cm2)918.70 (7.15, 10.50)349.75 (8.03, 11.30)578.20 (6.80, 9.50)0.002 *
TEG estimated lysis (%)911 (0.1, 2.4)341.2 (0.2, 2.5)570.9 (0.1, 2.5)0.961
Transfusions
Any blood product prehospital to 24 h9971 (72%)3721 (57%)6250 (81%)0.020 *
Outcomes
ICU-free days (30 days)9919 (0, 28)3726 (0, 30)629 (0, 27)0.058
ICU days993, (0, 11)372 (0, 10)624 (0, 13)0.450
12-h mortality9913 (13%)371 (3%)6212 (19%)0.028 *
24-h mortality9914 (14%)371 (3%)6213 (21%)0.015 *
72-h mortality9915 (15%)371 (3%)6214 (23%)0.008 *
In-hospital mortality9937 (37%)3710 (27%)6227 (44%)0.133
Complications
Acute renal failure9927 (27%)3710 (27%)6217 (27%)1
Deep vein thrombosis991 (1%)371 (3%)620 (0%)0.374
Pulmonary embolism997 (7%)370 (0%)627 (11%)0.043 *
Pneumonia9916 (16%)372 (5%)6214 (23%)0.026 *
Sepsis9919 (19%)377 (19%)6212 (20%)1
Urinary tract infection996 (6%)374 (11%)622 (3%)0.193
ARDS996 (6%)373 (8%)623 (5%)0.668
Respiratory failure9940 (40%)3714 (38%)6226 (42%)0.833
SIRS997 (7%)374 (11%)623 (5%)0.419
Decubitus ulcer993 (3%)370 (0%)623 (5%)0.291
Multiple organ failure9917 (17%)377 (19%)6210 (16%)0.786
EoT: endotheliopathy of trauma defined by plasma syndecan-1 ≥ 40 ng/mL; AIS: abbreviated injury scale; TEG: thromboelastography; ICU: intensive care unit; ARDS: acute respiratory distress syndrome; SIRS: systemic inflammatory response syndrome; * p-value < 0.05.
Table 2. Plasma enzyme linked immunosorbent assay results presented as median (IQR).
Table 2. Plasma enzyme linked immunosorbent assay results presented as median (IQR).
nAll Trauma Subjects
(n = 99)
Non-EoT
(n = 62)
EoT
(n = 37)
p-Value
Epinephrine99315 (140, 906)241 (89, 392)458 (201, 2136)0.004 *
Norepinephrine991295 (595, 2432)780 (388, 1308)1799 (803, 3642)<0.001 *
Soluble Thrombomodulin996.6 (5.2, 9.5)5.2 (4.7, 6.5)7.5 (6.1, 11.4)<0.001 *
Syndecan-19967 (28, 184)25 (19, 31)165 (79, 195)<0.001 *
EoT: endotheliopathy of trauma defined by plasma syndecan-1 ≥ 40 ng/mL; biomarker concentrations are reported in ng/mL; * p-value < 0.05.
Table 3. Differentially expressed plasma proteins between EoT and non-EoT groups based on adjusted p-value < 0.05.
Table 3. Differentially expressed plasma proteins between EoT and non-EoT groups based on adjusted p-value < 0.05.
Gene SymbolProtein DescriptionRaw p-Value ¥Adjusted
p-Value £
Fold Change ±
HIST1H4AHistone H45.97 × 10−33.69 × 10−23.34
ACTC1Actin, alpha cardiac muscle 13.90 × 10−76.61 × 10−52.84
TPI1Isoform 2 of triosephosphate isomerase1.86 × 10−31.54 × 10−22.78
LDHAL-lactate dehydrogenase A chain3.33 × 10−43.86 × 10−32.09
ADH1AAlcohol dehydrogenase 1A3.48 × 10−58.94 × 10−41.99
S100A9Protein S100-A91.80 × 10−42.49 × 10−31.93
SOD1Superoxide dismutase1.95 × 10−42.58 × 10−31.90
GSTA1Glutathione S-transferase2.00 × 10−56.71 × 10−41.87
FABP1Fatty acid-binding protein, liver3.20 × 10−43.86 × 10−31.67
ADH1BAlcohol dehydrogenase 1B2.87 × 10−62.78 × 10−41.62
UGP2UTP--glucose-1-phosphate uridylyltransferase8.35 × 10−51.50 × 10−31.61
FAHFumarylacetoacetase7.17 × 10−51.39 × 10−31.58
HIST2H2BFHistone H2B type 2-F3.29 × 10−32.33 × 10−21.55
DDTD-dopachrome decarboxylase8.78 × 10−51.50 × 10−31.55
PRDX1Peroxiredoxin-12.51 × 10−43.17 × 10−31.49
ALDOBFructose-bisphosphate aldolase B7.04 × 10−63.58 × 10−41.44
ALDH1A1Retinal dehydrogenase 12.46 × 10−57.12 × 10−41.27
ASS1Argininosuccinate synthase6.42 × 10−51.33 × 10−31.25
HPD4-hydroxyphenylpyruvate dioxygenase4.40 × 10−44.40 × 10−31.24
MAT1AS-adenosylmethionine synthase isoform type-13.99 × 10−32.75 × 10−21.24
ACTBL2Beta-actin-like protein 2 O4.56 × 10−76.61 × 10−51.16
BHMTBetaine--homocysteine S-methyltransferase 18.64 × 10−63.58 × 10−41.12
YWHAE14-3-3 protein epsilon3.77 × 10−44.20 × 10−31.11
GSTM1Glutathione S-transferase Mu 15.97 × 10−33.69 × 10−21.10
ENO1Alpha-enolase2.08 × 10−56.71 × 10−41.08
HSPA8Heat shock cognate 71 kDa protein4.22 × 10−32.77 × 10−21.02
GOT1Aspartate aminotransferase, cytoplasmic1.02 × 10−41.64 × 10−30.80
ALDH1L1Cytosolic 10-formyltetrahydrofolate dehydrogenase5.02 × 10−51.12 × 10−30.78
PPIAPeptidyl-prolyl cis-trans isomerase A1.11 × 10−31.01 × 10−20.72
FTCDFormimidoyltransferase-cyclodeaminase3.22 × 10−32.33 × 10−20.63
S100A8Protein S100-A82.85 × 10−32.17 × 10−20.62
GAPDHGlyceraldehyde-3-phosphate dehydrogenase4.33 × 10−44.40 × 10−30.59
IDH1Isocitrate dehydrogenase [NADP] cytoplasmic4.15 × 10−44.40 × 10−30.56
DCXRL-xylulose reductase8.75 × 10−34.88 × 10−20.54
ACTBActin, cytoplasmic 14.90 × 10−63.55 × 10−40.53
B2MBeta-2-microglobulin9.11 × 10−48.52 × 10−30.41
FBP1Fructose-1,6-bisphosphatase 11.48 × 10−31.30 × 10−20.36
FCN3Ficolin-3 OS = Homo sapiens1.54 × 10−42.24 × 10−30.32
PGM1Phosphoglucomutase-12.38 × 10−31.87 × 10−20.23
HPXHemopexin2.99 × 10−32.22 × 10−2−0.12
ITIH2Inter-alpha-trypsin inhibitor heavy chain H27.85 × 10−34.55 × 10−2−0.15
KNG1Kininogen-18.33 × 10−34.73 × 10−2−0.19
C8BComplement component C8 beta chain6.92 × 10−34.18 × 10−2−0.19
CFHComplement factor H3.70 × 10−58.94 × 10−4−0.20
ITIH1Inter-alpha-trypsin inhibitor heavy chain H12.21 × 10−31.78 × 10−2−0.20
C8GComplement component C8 gamma chain7.23 × 10−34.28 × 10−2−0.23
C8AComplement component C8 alpha chain4.99 × 10−44.82 × 10−3−0.25
C4BComplement C4-B4.27 × 10−32.77 × 10−2−0.28
F5Coagulation factor V1.76 × 10−31.50 × 10−2−0.37
SERPINF2Alpha-2-antiplasmin7.66 × 10−63.58 × 10−4−0.38
C4A; C4BComplement C4-A1.40 × 10−42.14 × 10−3−0.48
SERPINA5Plasma serine protease inhibitor4.30 × 10−32.77 × 10−2−0.58
EoT: endotheliopathy of trauma defined by plasma syndecan-1 ≥ 40 ng/mL; ¥ raw p-value based on a two-sided t-test; £ Benjamini and Hochberg false discovery rate-adjusted p-value; ± fold-change based on EoT/non-EoT ratio.
Table 4. Correlation with 95% confidence interval between plasma abundance of upregulated intracellular hepatic proteins associated with the EoT group and serum transaminases.
Table 4. Correlation with 95% confidence interval between plasma abundance of upregulated intracellular hepatic proteins associated with the EoT group and serum transaminases.
ASTALT
rp-Valuerp-Value
BHMT0.837 (0. 0.729, 0.904)<0.001 *0.894 (0.820, 0.938)<0.001 *
ADH1A0.907 (0. 842, 0.946)<0.001 *0.795 (0.666, 0.878)<0.001 *
MAT1A0.441 (0.188, 0.639)0.001 *0.310 (0.038, 0.539)0.027 *
HPD0.631 (0.430, 0.772)<0.001 *0.654 (0.461, 0.787)<0.001 *
FABP10.828 (0.716, 0.899)<0.001 *0.732 (0.572, 0.838)<0.001 *
EoT: endotheliopathy of trauma defined by plasma syndecan-1 ≥ 40 ng/mL; r: correlation coefficient; AST: aspartate transaminase; ALT: alanine aminotransferase; * p-value < 0.05; transaminase data were available for 51 subjects.
Table 5. Comparison between plasma abundance of upregulated intracellular renal proteins associated with the EoT group and acute kidney injury development presented as median (IQR).
Table 5. Comparison between plasma abundance of upregulated intracellular renal proteins associated with the EoT group and acute kidney injury development presented as median (IQR).
non-AKI
n = 72
AKI
n = 27
p-Value
BHMT2,039,625 (1,498,128, 5,608,227)1,276,029 (1,285,297, 2,118,322)0.226
HPD3,018,214 (2,053,260, 8,459,705)3,118,767 (1,450,326, 5,226,505)0.188
FABP13,362,220 (1,877,758, 16,830,020)3,032,475 (2,430,624, 5,302,253)0.997
EoT: endotheliopathy of trauma defined by plasma syndecan-1 ≥ 40 ng/mL; AKI: acute kidney injury defined by stage 2 or 3 Kidney Disease Improving Global Outcomes group criteria using a Modification of Diet in Renal Disease derived reference creatinine.
Table 6. Damage-associated molecular patterns with increased abundance in the EoT group and associations with pathophysiology relevant to the systemic response to trauma.
Table 6. Damage-associated molecular patterns with increased abundance in the EoT group and associations with pathophysiology relevant to the systemic response to trauma.
S100-A8, S-100A9,
S100-A8/A9
HistonesPPIA
Pro-inflammatory[87,88,89,90,91][50,51,53,60,92][66,93,94]
Anti-inflammatory[41,42,95,96,97]
Thrombosis[87,98,99,100,101][52,85,102,103,104,105,106,107][65,80,108]
Endothelial impairment and activation[109,110,111][52,53,54,57,60,92,112][94,113,114,115]
Endothelial permeability[87,116,117,118][56,118,119]
Cardiac injury[74,120,121,122,123][124,125,126][127,128,129,130]
Pulmonary injury[52,131,132,133,134][56,60,135,136][64,137]
Renal injury[138,139,140,141,142][52,143,144][145,146,147]
Hepatic injury[76,148,149,150][50,51,151,152][153,154,155,156]
Trauma populations
-
Post injury elevations in circulating S100-A8/9 [157]
-
Decreased rate of post-injury circulating S100-A8/9 elevation is associated with increased risk of infection [157]
-
Increased injury severity [59]
-
Decreased admission Glasgow coma scale [158]
-
Increased sympatho-adrenal activation [59]
-
Increased inflammatory cytokine release [59]
-
Coagulopathy [25,59,60,106,159]
-
Increased ventilator days [158]
-
Multiple organ failure [158]
-
Acute lung injury [60,158]
-
Mortality [158]
-
Mortality [160]
EoT: endotheliopathy of trauma defined by plasma syndecan-1 ≥ 40 ng/mL.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Krocker, J.D.; Lee, K.H.; Henriksen, H.H.; Wang, Y.-W.W.; Schoof, E.M.; Karvelsson, S.T.; Rolfsson, Ó.; Johansson, P.I.; Pedroza, C.; Wade, C.E. Exploratory Investigation of the Plasma Proteome Associated with the Endotheliopathy of Trauma. Int. J. Mol. Sci. 2022, 23, 6213. https://doi.org/10.3390/ijms23116213

AMA Style

Krocker JD, Lee KH, Henriksen HH, Wang Y-WW, Schoof EM, Karvelsson ST, Rolfsson Ó, Johansson PI, Pedroza C, Wade CE. Exploratory Investigation of the Plasma Proteome Associated with the Endotheliopathy of Trauma. International Journal of Molecular Sciences. 2022; 23(11):6213. https://doi.org/10.3390/ijms23116213

Chicago/Turabian Style

Krocker, Joseph D., Kyung Hyun Lee, Hanne H. Henriksen, Yao-Wei Willa Wang, Erwin M. Schoof, Sigurdur T. Karvelsson, Óttar Rolfsson, Pär I. Johansson, Claudia Pedroza, and Charles E. Wade. 2022. "Exploratory Investigation of the Plasma Proteome Associated with the Endotheliopathy of Trauma" International Journal of Molecular Sciences 23, no. 11: 6213. https://doi.org/10.3390/ijms23116213

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop