Dysregulation of the actin scavenging system and inhibition of DNase activity following severe thermal injury

R J Dinsdale, J Hazeldine, K Al Tarrah, P Hampson, A Devi, C Ermogenous, A L Bamford, J Bishop, S Watts, E Kirkman, J J Dalle Lucca, M Midwinter, T Woolley, M Foster, J M Lord, N Moiemen, P Harrison, R J Dinsdale, J Hazeldine, K Al Tarrah, P Hampson, A Devi, C Ermogenous, A L Bamford, J Bishop, S Watts, E Kirkman, J J Dalle Lucca, M Midwinter, T Woolley, M Foster, J M Lord, N Moiemen, P Harrison

Abstract

Background: Circulating cell-free DNA (cfDNA) is not found in healthy subjects, but is readily detected after thermal injury and may contribute to the risk of multiple organ failure. The hypothesis was that a postburn reduction in DNase protein/enzyme activity could contribute to the increase in cfDNA following thermal injury.

Methods: Patients with severe burns covering at least 15 per cent of total body surface area were recruited to a prospective cohort study within 24 h of injury. Blood samples were collected from the day of injury for 12 months.

Results: Analysis of blood samples from 64 patients revealed a significant reduction in DNase activity on days 1-28 after injury, compared with healthy controls. DNase protein levels were not affected, suggesting the presence of an enzyme inhibitor. Further analysis revealed that actin (an inhibitor of DNase) was present in serum samples from patients but not those from controls, and concentrations of the actin scavenging proteins gelsolin and vitamin D-binding protein were significantly reduced after burn injury. In a pilot study of ten military patients with polytrauma, administration of blood products resulted in an increase in DNase activity and gelsolin levels.

Conclusion: The results of this study suggest a novel biological mechanism for the accumulation of cfDNA following thermal injury by which high levels of actin released by damaged tissue cause a reduction in DNase activity. Restoration of the actin scavenging system could therefore restore DNase activity, and reduce the risk of cfDNA-induced host tissue damage and thrombosis.

© 2019 The Authors. BJS published by John Wiley & Sons Ltd on behalf of BJS Society Ltd.

Figures

Figure 1
Figure 1
Thermal injury results in reduced DNase activity in the presence of stable or increased antigen levelsa DNase activity in serum taken from 64 patients on day (D) 1 to month (M) 12 after thermal injury, and from healthy controls (HC). Median (horizontal bars) and individual values are shown. *P < 0·001 versus HC (1‐way ANOVA followed by Kruskal–Wallis test). b DNase 1 antigen levels in serum taken from 24 patients on D1 to D28 after thermal injury, and from HC. Median (horizontal bars) and individual values are shown. *P < 0·010 versus HC (1‐way ANOVA followed by Bonferroni's multiple comparison test). c Fluorescence microscopic imaging of neutrophil extracellular trap (NET) degradation with phorbol myristate acetate (PMA)‐induced NET DNA stained with SYTOX® Green for untreated control (N = 3), and NETS treated with serum from patients with thermal injuries (N = 3), serum from healthy controls (N = 3) and DNAse 1 positive controls (N = 3) (original magnification ×20).
Figure 2
Figure 2
Thermal injury causes release of actin into the blooda Western blot of actin in plasma samples from nine patients (P1–P9) taken within 24 h of injury. Actin was not detected in plasma from healthy controls (HC). Data are from two independent western blots. b Western blot for a patient (P10) with a burn affecting 66 per cent of the total body surface area who developed multiple organ failure (MOF). Actin was detected within 24 h and up to day (D) 3 after thermal injury. Actin was detectable at later time points, day 20–23, corresponding to the later development of MOF. A second western blot for four independent patients (P11–14) shows actin at later time points.
Figure 3
Figure 3
Levels of vitamin D‐binding protein and gelsolin are reduced after thermal injury Levels of a vitamin D‐binding protein (VDBP) in 50 patients and b gelsolin in 64 patients from day (D) 1 to month (M) 12 after thermal injury, and in healthy controls (HC). Median (horizontal bars) and individual values are shown. *P < 0·010 versus HC (1‐way ANOVA followed by Bonferroni's multiple comparison test).
Figure 4
Figure 4
Blood products increase circulating gelsolin levels and protect against inhibition of DNase activity in patients with polytrauma Comparison of a DNase activity, b gelsolin, c vitamin D‐binding protein (VDBP) and d cell‐free (cf) DNA levels at hospital admission between five patients who had previously received blood products (fresh frozen plasma) and five who had not. Median (horizontal bars) and individual values are shown. *P = 0·028, †P = 0·032 (Mann–Whitney U test).
Figure 5
Figure 5
Hypothesis model suggesting how severe thermal injury results in disruption of the actin scavenging system which predisposes to DNase activity inhibition and accumulation of cell‐free DNA Following severe thermal injury, polymerized and monomeric actin is released, which immediately reduces vitamin D‐binding protein and gelsolin levels. The polymerized actin can then bind to DNase and inhibit its activity. Injury‐ and immune cell‐derived DNA can accumulate and potentially cause activation of the intrinsic clotting cascade, impaired fibrinolysis, bind to platelets, and cause cellular damage and/or inflammation. NET, neutrophil extracellular trap.

References

    1. Jackson PC, Hardwicke J, Bamford A, Nightingale P, Wilson Y, Papini R et al Revised estimates of mortality from the Birmingham Burn Centre, 2001–2010: a continuing analysis over 65 years. Ann Surg 2014; 259: 979–984.
    1. Mann EA, Baun MM, Meininger JC, Wade CE. Comparison of mortality associated with sepsis in the burn, trauma, and general intensive care unit patient: a systematic review of the literature. Shock 2012; 37: 4–16.
    1. Xiao W, Mindrinos MN, Seok J, Cuschieri J, Cuenca AG, Gao H et al; Inflammation and Host Response to Injury Large‐Scale Collaborative Research Program. A genomic storm in critically injured humans. J Exp Med 2011; 208: 2581–2590.
    1. Cohen J, Vincent JL, Adhikari NK, Machado FR, Angus DC, Calandra T et al Sepsis: a roadmap for future research. Lancet Infect Dis 2015; 15: 581–614.
    1. Dwivedi DJ, Toltl LJ, Swystun LL, Pogue J, Liaw KL, Weitz JI et al; Canadian Critical Care Translational Biology Group . Prognostic utility and characterization of cell‐free DNA in patients with severe sepsis. Crit Care 2012; 16: R151.
    1. Saukkonen K, Lakkisto P, Pettila V, Varpula M, Karlsson S, Ruokonen E et al; Finnsepsis Study Group . Cell‐free plasma DNA as a predictor of outcome in severe sepsis and septic shock. Clin Chem 2008; 54: 1000–1007.
    1. Rhodes A, Wort SJ, Thomas H, Collinson P, Bennett ED. Plasma DNA concentration as a predictor of mortality and sepsis in critically ill patients. Crit Care 2006; 10: R60.
    1. Shoham Y, Krieger Y, Perry ZH, Shaked G, Bogdanov‐Berezovsky A, Silberstein E et al Admission cell free DNA as a prognostic factor in burns: quantification by use of a direct rapid fluorometric technique. Biomed Res Int 2014; 2014: 306580.
    1. Lo YM, Rainer TH, Chan LY, Hjelm NM, Cocks RA. Plasma DNA as a prognostic marker in trauma patients. Clin Chem 2000; 46: 319–323.
    1. Naumann DN, Hazeldine J, Dinsdale RJ, Bishop JR, Midwinter MJ, Harrison P et al Endotheliopathy is associated with higher levels of cell‐free DNA following major trauma: a prospective observational study. PLoS One 2017; 12: e0189870.
    1. Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol 2013; 13: 34–45.
    1. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM et al Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med 2007; 13: 463–469.
    1. Boneschansker L, Inoue Y, Oklu R, Irimia D. Capillary plexuses are vulnerable to neutrophil extracellular traps. Integr Biol (Camb) 2016; 8: 149–155.
    1. Otawara M, Roushan M, Wang X, Ellett F, Yu YM, Irimia D. Microfluidic assay measures increased neutrophil extracellular traps circulating in blood after burn injuries. Sci Rep 2018; 8: 16983.
    1. Hampson P, Dinsdale RJ, Wearn CM, Bamford AL, Bishop JRB, Hazeldine J et al Neutrophil dysfunction, immature granulocytes, and cell‐free DNA are early biomarkers of sepsis in burn‐injured patients: a prospective observational cohort study. Ann Surg 2017; 265: 1241–1249.
    1. Napirei M, Wulf S, Eulitz D, Mannherz HG, Kloeckl T. Comparative characterization of rat deoxyribonuclease 1 (Dnase1) and murine deoxyribonuclease 1‐like 3 (Dnase1l3). Biochem J 2005; 389: 355–364.
    1. Napirei M, Ludwig S, Mezrhab J, Klockl T, Mannherz HG. Murine serum nucleases – contrasting effects of plasmin and heparin on the activities of Dnase1 and Dnase1‐like 3 (Dnase1l3). FEBS J 2009; 276: 1059–1073.
    1. Meng W, Paunel‐Görgülü A, Flohé S, Witte I, Schädel‐Höpfner M, Windolf J et al Deoxyribonuclease is a potential counter regulator of aberrant neutrophil extracellular traps formation after major trauma. Mediators Inflamm 2012; 2012: 149560.
    1. Hakkim A, Fürnrohr BG, Amann K, Laube B, Abed UA, Brinkmann V et al Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A 2010; 107: 9813–9818.
    1. Al‐Mayouf SM, Sunker A, Abdwani R, Abrawi SA, Almurshedi F, Alhashmi N et al Loss‐of‐function variant in DNASE1L3 causes a familial form of systemic lupus erythematosus. Nat Genet 2011; 43: 1186–1188.
    1. Jimenez‐Alcazar M, Napirei M, Panda R, Kohler EC, Kremer Hovinga JA, Mannherz HG et al Impaired DNase1‐mediated degradation of neutrophil extracellular traps is associated with acute thrombotic microangiopathies. J Thromb Haemost 2015; 13: 732–742.
    1. Lazarides E, Lindberg U. Actin is the naturally occurring inhibitor of deoxyribonuclease I. Proc Natl Acad Sci U S A 1974; 71: 4742–4746.
    1. Lee WM, Galbraith RM. The extracellular actin‐scavenger system and actin toxicity. N Engl J Med 1992; 326: 1335–1341.
    1. Kwiatkowski DJ, Janmey PA, Yin HL. Identification of critical functional and regulatory domains in gelsolin. J Cell Biol 1989; 108: 1717–1726.
    1. Yin HL, Iida K, Janmey PA. Identification of a polyphosphoinositide‐modulated domain in gelsolin which binds to the sides of actin filaments. J Cell Biol 1988; 106: 805–812.
    1. Tobiasen J, Hiebert JM, Edlich RF. The abbreviated burn severity index. Ann Emerg Med 1982; 11: 260–262.
    1. Osler T, Glance LG, Hosmer DW. Simplified estimates of the probability of death after burn injuries: extending and updating the Baux score. J Trauma 2010; 68: 690–697.
    1. Greenhalgh DG, Saffle JR, Holmes JH IV, Gamelli RL, Palmieri TL, Horton JW et al; American Burn Association Consensus Conference on Burn Sepsis and Infection Group . American Burn Association consensus conference to define sepsis and infection in burns. J Burn Care Res 2007; 28: 776–790.
    1. Thierry AR, El Messaoudi S, Gahan PB, Anker P, Stroun M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev 2016; 35: 347–376.
    1. Bhagirath VC, Dwivedi DJ, Liaw PC. Comparison of the proinflammatory and procoagulant properties of nuclear, mitochondrial, and bacterial DNA. Shock 2015; 44: 265–271.
    1. Kaplan MJ, Radic M. Neutrophil extracellular traps: double‐edged swords of innate immunity. J Immunol 2012; 189: 2689–2695.
    1. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD Jr et al Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A 2010; 107: 15 880–15 885.
    1. Fuchs TA, Brill A, Wagner DD. Neutrophil extracellular trap (net) impact on deep vein thrombosis. Arterioscler Thromb Vasc Biol 2012; 32: 1777–1783.
    1. Gupta AK, Joshi MB, Philippova M, Erne P, Hasler P, Hahn S et al Activated endothelial cells induce neutrophil extracellular traps and are susceptible to NETosis‐mediated cell death. FEBS Lett 2010; 584: 3193–3197.
    1. Mogilner A, Oster G. Cell motility driven by actin polymerization. Biophys J 1996; 71: 3030–3045.
    1. Ananthakrishnan R, Ehrlicher A. The forces behind cell movement. Int J Biol Sci 2007; 3: 303–317.
    1. Srinivasan N, Gordon O, Ahrens S, Franz A, Deddouche S, Chakravarty P et al Actin is an evolutionarily‐conserved damage‐associated molecular pattern that signals tissue injury in Drosophila melanogaster . Elife 2016; 5: pii: e19662.
    1. Lee PS, Patel SR, Christiani DC, Bajwa E, Stossel TP, Waxman AB. Plasma gelsolin depletion and circulating actin in sepsis: a pilot study. PLoS One 2008; 3: e3712.
    1. Lind SE, Smith DB, Janmey PA, Stossel TP. Depression of gelsolin levels and detection of gelsolin–actin complexes in plasma of patients with acute lung injury. Am Rev Respir Dis 1988; 138: 429–434.
    1. Ito H, Kambe H, Kimura Y, Nakamura H, Hayashi E, Kishimoto T et al Depression of plasma gelsolin level during acute liver injury. Gastroenterology 1992; 102: 1686–1692.
    1. Huang LF, Yao YM, Li JF, Dong N, Liu C, Yu Y et al Reduction of plasma gelsolin levels correlates with development of multiple organ dysfunction syndrome and fatal outcome in burn patients. PLoS One 2011; 6: e25748.
    1. Suhler E, Lin W, Yin HL, Lee WM. Decreased plasma gelsolin concentrations in acute liver failure, myocardial infarction, septic shock, and myonecrosis. Crit Care Med 1997; 25: 594–598.
    1. Osborn TM, Verdrengh M, Stossel TP, Tarkowski A, Bokarewa M. Decreased levels of the gelsolin plasma isoform in patients with rheumatoid arthritis. Arthritis Res Ther 2008; 10: R117.
    1. Hu Y, Li H, Li WH, Meng HX, Fan YZ, Li WJ et al The value of decreased plasma gelsolin levels in patients with systemic lupus erythematosus and rheumatoid arthritis in diagnosis and disease activity evaluation. Lupus 2013; 22: 1455–1461.
    1. Dahl B, Schiødt FV, Nielsen M, Kiaer T, Williams JG, Ott P. Admission level of Gc‐globulin predicts outcome after multiple trauma. Injury 1999; 30: 275–281.
    1. Dahl B, Schiødt FV, Ott P, Wians F, Lee WM, Balko J et al Plasma concentration of Gc‐globulin is associated with organ dysfunction and sepsis after injury. Crit Care Med 2003; 31: 152–156.
    1. Dahl B, Schiødt FV, Rudolph S, Ott P, Kiaer T, Heslet L. Trauma stimulates the synthesis of Gc‐globulin. Intensive Care Med 2001; 27: 394–399.
    1. Dahl B, Schiødt FV, 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.
    1. Mounzer KC, Moncure M, Smith YR, Dinubile MJ. Relationship of admission plasma gelsolin levels to clinical outcomes in patients after major trauma. Am J Respir Crit Care Med 1999; 160: 1673–1681.
    1. DiNubile MJ. Plasma gelsolin as a biomarker of inflammation. Arthritis Res Ther 2008; 10: 124.
    1. Mitra B, Mori A, Cameron PA, Fitzgerald M, Paul E, Street A. Fresh frozen plasma (FFP) use during massive blood transfusion in trauma resuscitation. Injury 2010; 41: 35–39.
    1. Chhabra D, Nosworthy NJ, dos Remedios CG. The N‐terminal fragment of gelsolin inhibits the interaction of DNase I with isolated actin, but not with the cofilin–actin complex. Proteomics 2005; 5: 3131–3136.
    1. Sillesen M, Johansson PI, Rasmussen LS, Jin G, Jepsen CH, Imam A et al Fresh frozen plasma resuscitation attenuates platelet dysfunction compared with normal saline in a large animal model of multisystem trauma. J Trauma Acute Care Surg 2014; 76: 998–1007.
    1. Watts S, Nordmann G, Brohi K, Midwinter M, Woolley T, Gwyther R et al Evaluation of prehospital blood products to attenuate acute coagulopathy of trauma in a model of severe injury and shock in anesthetized pigs. Shock 2015; 44(Suppl 1): 138–148.
    1. Halaweish I, Bambakidis T, Nikolian VC, Georgoff P, Bruhn P, Piascik P et al Early resuscitation with lyophilized plasma provides equal neuroprotection compared with fresh frozen plasma in a large animal survival model of traumatic brain injury and hemorrhagic shock. J Trauma Acute Care Surg 2016; 81: 1080–1087.
    1. Georgoff PE, Nikolian VC, Halaweish I, Chtraklin K, Bruhn PJ, Eidy H et al Resuscitation with lyophilized plasma is safe and improves neurological recovery in a long‐term survival model of swine subjected to traumatic brain injury, hemorrhagic shock, and polytrauma. J Neurotrauma 2017; 34: 2167–2175.
    1. Imam AM, Jin G, Sillesen M, Duggan M, Jepsen CH, Hwabejire JO et al Early treatment with lyophilized plasma protects the brain in a large animal model of combined traumatic brain injury and hemorrhagic shock. J Trauma Acute Care Surg 2013; 75: 976–983.
    1. Zhang LM, Li R, Zhao XC, Zhang Q, Luo XL. Increased transfusion of fresh frozen plasma is associated with mortality or worse functional outcomes after severe traumatic brain injury: a retrospective study. World Neurosurg 2017; 104: 381–389.
    1. Mesar T, Larentzakis A, Dzik W, Chang Y, Velmahos G, Yeh DD. Association between ratio of fresh frozen plasma to red blood cells during massive transfusion and survival among patients without traumatic injury. JAMA Surg 2017; 152: 574–580.
    1. Hagiwara A, Kushimoto S, Kato H, Sasaki J, Ogura H, Matsuoka T et al Can early aggressive administration of fresh frozen plasma improve outcomes in patients with severe blunt trauma? – A report by the Japanese Association for the Surgery of Trauma. Shock 2016; 45: 495–501.
    1. Rothenbach PA, Dahl B, Schwartz JJ, O'Keefe GE, Yamamoto M, Lee WM et al Recombinant plasma gelsolin infusion attenuates burn‐induced pulmonary microvascular dysfunction. J Appl Physiol (1985) 2004; 96: 25–31.

Source: PubMed

3
Abonnieren