key: cord-0758605-ztqj120w authors: Bouchard, Beth A.; Colovos, Christos; Lawson, Michael A.; Osborn, Zachary T.; Sackheim, Adrian M.; Mould, Kara J.; Janssen, William J.; Cohen, Mitchell J.; Majumdar, Devdoot; Freeman, Kalev title: Increased histone-DNA complexes and endothelial-dependent thrombin generation in severe COVID-19 date: 2021-12-03 journal: Vascul Pharmacol DOI: 10.1016/j.vph.2021.106950 sha: c72260ca88746334c0a3fa81fc65cc9deca461dd doc_id: 758605 cord_uid: ztqj120w Coagulopathy in severe COVID-19 is common but poorly understood. The purpose of this study was to determine how SARS-CoV-2 infection impacts histone levels, fibrin structure, and endogenous thrombin potential in the presence and absence of endothelial cells. We studied individuals with SARS-CoV-2 infection and acute respiratory distress syndrome at the time of initiation of mechanical ventilation compared to healthy controls. Circulating histone-DNA complexes were elevated in the plasma of COVID-19 patients relative to healthy controls (n=6, each group). Using calibrated automated thrombinography, thrombin generation was altered in COVID-19 patient plasma samples. Despite having increased endogenous thrombin potential, patient plasma samples exhibited prolonged lag times and times to peak thrombin in the presence of added tissue factor and PCPS. Strikingly different results were observed when endothelial cells were used in place of tissue factor and PCPS. While healthy control plasma samples did not generate measurable thrombin after 60 min, plasma samples from COVID-19+ patients formed thrombin (mean lag time ~20 min). Consistent with the observed alterations in thrombin generation, clots from COVID-19 subjects exhibited a denser fibrin network, thinner fibers and lower fibrin resolvability. Elevated histones, aberrant fibrin formation, and increased endothelial-dependent thrombin generation may contribute to coagulopathy in COVID-19. The emergence of the severe acute respiratory syndrome novel corona virus 2 (SARS-CoV-2) resulted in a global pandemic that afflicted over 200 million people globally and over 40 million in the United States alone as of October 2021. The associated disease, coronavirus disease 2019 (COVID- 19) , accounts for over 4.8 million deaths worldwide and over 700,000 in the United States [1] . A consistent finding amongst patients with SARS-CoV-2 is derangements in coagulation markers and increased incidence of thrombotic complications [2] [3] [4] [5] [6] . Derangement in coagulation parameters was associated with a coagulation phenotype similar to disseminated intravascular coagulopathy and associated with death [5, 7] . Findings of micro and macro vascular thromboses resulting in organ failure are observed in autopsy studies. Emerging evidence of the benefits provided by therapeutic anticoagulation further supports the idea that coagulopathy contributes to SARS-CoV-2 disease progression. Plasma from patients and healthy donors was prepared from citrated (3.2%) whole blood obtained at the onset of mechanical ventilation from a central line or the antecubital vein using standard methods. All plasma samples were stored at -80°C in small aliquots. Cell-free histone levels were assessed in plasma from healthy controls and COVID-19 patients using a photometric enzyme immunoassay (Cell Death Detection ELISAPLUS kit, Roche, Indianapolis, IN), which measures histone-associated DNA fragments, according to the manufacturer's instructions. Fibrin clot formation was assessed as previously described [17] . Plasma was diluted 1:1 with 20 mM Hepes, 0.15 M NaCl, pH 7.4 (HBS) and incubated on glass chamber slides with AlexaFluor488-labeled fibrinogen (220 nM) (ThermoFisher Scientific, Waltham, MA), relipidated tissue factor 1-242 (8.7 pM) (a gift from Dr. R. Lunblad, Baxter Healthcare Corp.), and CaCl 2 , with HistoChoice (MilliporeSigma, Burlington, MA) and an anti-photobleaching agent (Agilent Technologies, Santa Clara, California). Clots were visualized by confocal microscopy using a Nikon A1R Confocal Microscope (Nikon Instruments Inc, Melville NY) with a 60X/1.5 oil immersion objective. For each clot, a three-dimensional Z-stack image series consisting of 40 images at 0.25 µm steps through each sample was obtained. All plasma clots were formed and imaged in duplicate. Fiber resolvability was quantified by standard deviation mapping of the images using custom in-house software as described previously [17] . Analysis of fiber resolvability, as determined by standard deviation mapping, provides a reliable assessment of the clot architecture that is unaffected by the overall brightness of the fluorescence [18] . Eagle Medium (DMEM) supplemented with 10% fetal bovine serum and 5 µg/mL gentamycin (complete medium) at 37°C, 5% CO 2 . Prior to use, the complete medium was removed and the cells incubated with DMEM for 1 hr at 37°C, 5% CO 2 . The cells were released from the tissue culture wells with trypsin and subjected to centrifugation (170 x g, 7 min). Cell pellets were washed one time by resuspension in HBS followed by centrifugation. The final cell pellets were resuspended in HBS and adjusted to a final concentration of 1x10 7 /mL. Thrombin generation was assessed by modified calibrated automated thrombography [19] . Plasma was thawed at 37°C in the presence of corn trypsin inhibitor (0.1 mg/mL final concentration). Plasma was incubated with the thrombin substrate Z-Gly-Gly-Arg 7-amido-4methylcoumarin hydrochloride (0.42 mM) (Bachem AG, Switzerland) and CaCl 2 (15 mM) (3 min, 37°C), and the reactions initiated by the addition of relipidated tissue factor 1-242 (6.5 pM) and synthetic vesicles consisting of 80% phosphatidylcholine and 20% phosphatidylserine (PCPS) (20 µM), or EA.hy926 cells (2x10 5 ). Fluorescence was measured (ex = 370 nm/em = 460 nm) for 1 hour with a Cytation 3 imaging reader (BioTek, Winooski, VT). Changes in fluorescence were converted to thrombin concentrations using a calibration curve created from sequential dilutions of human thrombin. If no change in fluorescence was noted after 60 min, the lag time for the sample was defined as >60 min. Data are expressed as mean ± standard error of the mean. Unpaired, two-tailed t-tests were performed to compare the mean histone levels and measures of thrombin generation between controls and patients. For comparison of clot resolvability, the Kolmogorov-Smirnov test was used to compare cumulative distributions with 95% confidence. A p-value less than 0.05 was considered significant. J o u r n a l P r e -p r o o f We enrolled patients diagnosed with COVID-19 (n=6 patients) and healthy controls (n=6). The patient characteristics are described in Table 1 . Using an assay that detects cell-free, histone-associated DNA fragments, it was observed that histones were significantly elevated in COVID-19 patient plasma relative to plasma from healthy controls ( Figure 1 ). Fibrin clot structure was also assessed using AlexaFluor488-labeled fibrinogen. Comparison of the clot structures by confocal microscopy demonstrated that fibrin clots formed using plasma from COVID-19 patients were markedly different from those formed using healthy control plasma. Control clots consistently demonstrated a robust fibrin network with high fiber resolvability ( Figure 2 ). In contrast, COVID-19 patient samples were densely packed with extremely thin, almost hair-like fibers that were significantly less resolvable (p<0.05) suggesting that thrombin generation in the COVID-19 plasma was altered [20] . Thrombin generation in healthy control plasma and plasma from COVID-19 patients were compared by calibrated automated thrombography in the presence of added tissue factor and PCPS (Figures 3a & 3b) or cultured EA.hy926 cells (Figures 4a & 4b) . In the presence of tissue factor and PCPS, the endogenous thrombin potential (ETP) (Figure 3c These combined observations suggest that thrombin generation by plasma from patients with severe COVID-19 infections is dysregulated, and that high levels of histones or another molecule(s) effect(s) blood coagulation in the presence of endothelial cells. Histones act as damage-associated molecular pattern molecules following their release from cells by NETs, cell apoptosis, or cell necrosis [21] . Circulating levels of nucleosomes and histones are significantly elevated and correlate with the severity or poor outcome of several pathophysiological processes such as acute bacterial infection, sepsis, autoimmune diseases, cerebral stroke, trauma, cancer [21] , and acute pulmonary embolism [22] . Here, we show increased histoneassociated DNA fragments in SARS-CoV-2 infection resulting in severe COVID-19. This is J o u r n a l P r e -p r o o f consistent with prior reports of elevated NETs in COVID-19, as evidenced by increased cell-free DNA, myeloperoxidase-DNA complexes, and citrullinated-histone H3 [23] [24] [25] . Similar observations were recently made in two independent cohorts of COVID-19 positive patients with a quantitative nucleosome immunoassay that measured cell-free H3.1 nucleosomes [26] . These investigators demonstrated that nucleosomes were highly elevated in plasma of COVID-19 patients with a severe course of the disease relative to healthy controls and that both the histone 3.1 variant and citrullinated nucleosomes increase with disease severity. In another study by Guéant and colleagues, circulating histone-DNA levels and other markers of neutrophil activation were increased in patients with COVID-19 [27] . Histone-DNA was associated with markers of disease severity including intensive care admission, body temperature, lung damage, markers of cardiovascular outcomes, renal failure, and increased interleukin-6, interleukin-8 and C-X-C motif chemokine receptor 2. Extracellular histones enhance plasma thrombin generation by reducing thrombomodulindependent protein C activation [14] . Additionally, isolated histone proteins bind to prothrombin via fragment 1 and fragment 2 (non-catalytic portions), and reduce the need for factor Xa in clotting [13] . Indeed, the overall higher amounts of thrombin generated by COVID-19 patient plasma is consistent with a more procoagulant state. However, unexpectedly, the mean lag time and time to peak thrombin were significantly reduced as compared to control patients suggesting that thrombin generation in COVID-19 patients is dysregulated. Consistent with our observations are very recent studies by Bouck and colleagues [28] who also demonstrated that compared with healthy donors, patients with SARS-CoV-2 infection had increased thrombin generation potential but a prolonged lag time. Additional experiments demonstrated increased endogenous plasmin potential and delayed plasmin formation. These perturbations led to increased fibrin formation. The fibrin clots formed in situ by plasma from patients with severe COVID-19 are denser with thinner fibers and lower fiber resolvability as compared to clots formed from the plasma of healthy controls. Patients with thromboembolic diseases also form structurally abnormal clots that are resistant to fibrinolysis [29] . Similar observations have been made in patients with severe trauma [17] , which is also characterized by a disseminated intravascular coagulation-like phenotype [30] . As patients with acute thromboembolic disease [22] and trauma [13] also have elevated circulating histone levels, these structurally-and functionally-altered clots may be the result of covalent (via factor XIIIa crosslinking) and noncovalent interactions of histones with fibrin [31] in addition to dysregulated thrombin formation. Additional studies are needed to examine the interactions of histones with fibrin. Histones can also promote thrombotic events by inducing endothelial cells to release proinflammatory cytokines [15] , increase cell surface adhesion molecules [16] , and express tissue factor [15] . When cultured endothelial cells were used in place of added tissue factor and PCPS, While the impact of this study is limited by its small sample size, its results confirm that circulating histones are elevated in severe COVID-19 and that thrombin generation using a plasma-based assay is dysregulated. This study extends these observations by demonstrating a role for endothelial cells in COVID-19 coagulopathy and that fibrin clot structure is altered. Based on these collective observations, we hypothesize that histones in COVID-19 patient plasma activate endothelial cells leading to dysregulated thrombin generation by release of von Willebrand factor and P-selectin from Weibel-Palade bodies [34] , activation and translocation of sphingomyelinase resulting in tissue factor decryption [15, 33] and procoagulant microparticle release [33] , and/or decrease of thrombomodulin expression [14] ( Figure 5 ). Histones may also mediate the formation of structurally abnormal clots directly or as a result of altered thrombin formation. Neutralization of histones with sulfated polyanions such as heparin may mitigate these coagulopathic responses and reduce morbidity and mortality due to thrombosis. Additional studies with a larger patient cohort and access to larger plasma volumes are warranted to test these hypotheses. This research was supported by grants (R01GM123010 to K.F., UM1HL120877 to K.F., R35HL140039 to W.J.) from the National Institutes of Health (NIH). It was also funded, in part, by the NIH Agreement 1OT2HL156812. The views and conclusions contained in this document are those of the authors and should not be interpreted as representing the official policies, either expressed or implied, of the NIH. An interactive web-based dashboard to track COVID-19 in real time Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study Clinical Characteristics of Coronavirus Disease 2019 in China Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia Potential Therapeutic Agents and Associated Bioassay Data for COVID-19 and Related Human Coronavirus Infections Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study Neutrophil extracellular traps in COVID-19 Targeting potential drivers of COVID-19: Neutrophil extracellular traps The pathogenesis of microthrombi in COVID-19 cannot be controlled by DOAC: NETosis should be the target The intriguing commonality of NETosis between COVID-19 & Periodontal disease How NETosis could drive "Post-COVID-19 syndrome" among survivors Assembly of alternative prothrombinase by extracellular histones initiates and disseminates intravascular coagulation Extracellular histones increase plasma thrombin generation by impairing thrombomodulin-dependent protein C activation Extracellular histones are major mediators of death in sepsis Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice Dense and dangerous: The tissue plasminogen activator-resistant fibrinolysis shutdown phenotype is due to abnormal fibrin polymerization Rapid quantification of pixel-wise fiber orientation data in micrographs Factor XIa and tissue factor activity in patients with coronary artery disease Thrombin generation and fibrin clot structure Release and activity of histone in diseases Prothrombotic fibrin clot properties associated with NETs formation characterize acute pulmonary embolism patients with higher mortality risk Neutrophils and Contact Activation of Coagulation as Potential Drivers of COVID-19 Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome Circulating Nucleosomes as Potential Markers to Monitor COVID-19 Disease Progression Elastase and exacerbation of neutrophil innate immunity are involved in multi-visceral manifestations of COVID-19 COVID-19 and Sepsis Are Associated With Different Abnormalities in Plasma Procoagulant and Fibrinolytic Activity Fibrin clot structure and function: a role in the pathophysiology of arterial and venous thromboembolic diseases Trauma-induced coagulopathy: The past, present, and future Extracellular Histones Inhibit Fibrinolysis through Noncovalent and Covalent Interactions with Fibrin Endothelial dysfunction and thrombosis in patients with COVID-19 -Brief Report SARS-CoV-2 infection induces the activation of tissue factor-mediate coagulation via activation of acid sphingomyelinase Histones link inflammation and thrombosis through the induction of Weibel-Palade body exocytosis This article is the authors' original work, hasn't received prior publication, and isn't under consideration for publication elsewhere. The authors declare that they have no conflicts of interest.