Skip to main content
Erschienen in: Cardiovascular Drugs and Therapy 1/2020

Open Access 15.02.2020 | Review Article

The Role of Extracellular DNA and Histones in Ischaemia-Reperfusion Injury of the Myocardium

verfasst von: Mohammed Shah, Derek M. Yellon, Sean M. Davidson

Erschienen in: Cardiovascular Drugs and Therapy | Ausgabe 1/2020

Abstract

Despite an increase in the rates of survival in patients suffering myocardial infarction, as yet there is no therapy specifically targeting ischaemia and reperfusion injury of the myocardium. With a greater understanding of immune activation during infarction, more potential treatment targets are now being identified. The innate immune system is believed to play an important role in the myocardium after ischaemia-driven cardiomyocyte death. The release of intracellular contents including DNA into the extracellular space during necrosis and cell rupture is now believed to create a pro-inflammatory milieu which propagates the inflammatory process. DNA and DNA fragments have been shown to activate the innate immune system by acting as Danger-Associated Molecular Patterns (DAMPs), which act as ligands on toll-like receptors (TLRs). Stimulation of TLRs, in turn, can activate intracellular cell death pathways such as pyroptosis. Here, we review the role of DNA fragments during ischaemia and reperfusion, and assess their potential as a target in the quest to preserve cardiomyocyte viability following myocardial infarction.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Targeting Cell-Free DNA in Myocardial Infarction

Despite our increasing understanding of the pathogenesis of myocardial infarction, it remains a leading cause of premature death in the Western world [1, 2]. The widespread adoption of percutaneous coronary intervention has resulted in a significant reduction in the duration of coronary ischaemia once the clinical diagnosis of coronary artery occlusion and ST elevation myocardial infarction (STEMI) has been made. Restoring blood flow to the myocardium promptly can prevent excessive cardiomyocyte death. Conversely, delaying treatment is associated with worse outcome and death. As a consequence of the success in instigating rapid reperfusion therapy, there has been an increase in the survival of STEMI patients, but this has led to a greater incidence of subsequent heart failure [3]. Paradoxically, both ischaemia and the subsequent reperfusion lead to excessive cardiomyocyte death which results in profound “remodelling” of the heart associated with fibrotic replacement of the myocardial cytoskeleton, altering the geometry of the ventricle and resulting in impaired pump function and heart failure—also called ventricular remodelling [4].
In order to help preserve cardiomyocyte viability after ischaemia-reperfusion injury, attention has focussed for many years on trying to understanding the process of cardiomyocyte death and inflammation after myocardial reperfusion [58]. There is increasing evidence that inflammation induced by ischaemia-reperfusion may actually contribute to cardiomyocyte death, excessive scar formation, and poor ventricular remodelling [9, 10]. Unfortunately, the results of the majority of clinical trials into the use of anti-inflammatory therapies for treating MI have been disappointing, illustrating our lack of understanding of ischaemia-reperfusion-induced inflammation in the myocardium. One important target is the process by which cardiomyocytes, which are viable at the point of reperfusion, die during reperfusion. This type of cell death differs from phagocytosis or apoptosis in that it is more uncontrolled and results in the rupture of the sarcolemma and release of the intracellular contents into the extracellular space. Current well-established cardioprotective strategies such as ischaemic preconditioning preserve cardiomyocytes during ischaemia-reperfusion injury, thereby limiting the release of intracellular debris [10]. During this process, the dying cells propagate the inflammatory response throughout the reperfusion zone, as the intracellular debris act as Danger-Associated Molecular Patterns (DAMPs) which are ligands for activation of the innate immune system [11] (Fig. 1). This type of cell death is called “necrosis”. Examples of DAMPs include mobility group box-1 protein (HMGB1), heat shock proteins, adenosine, extracellular RNA, mitochondrial DNA, and interleukin (IL)-1α all of which may stimulate the innate immune response. Recently, it has been shown that cells can also undergo a type of programmed necrosis, referred to as “necroptosis” or “pyroptosis” [1214]. Identifying and targeting these DAMPs has provided varying results in attempts to save myocytes from the deleterious effects of reperfusion. This is often difficult because many identified DAMPs such as HMGB1 have complex, multifaceted roles, and inhibiting their function may instead be detrimental as inflammation is important in the process of cardiac repair after an insult. One potential DAMP that may be a promising target is DNA itself, which has the benefit of having no such multifaceted effect once outside the cell. There is now some evidence to suggest that cell death may be propagated by intracellular material such as DNA in an extracellular environment, contributing to excessive myocyte death in the myocardium after ischaemia-reperfusion injury [1518]. One potential DAMP that has been highlighted is DNA and its components. This review aims to highlight this potentially promising target for future cardioprotective therapies.

Inflammation and Ischaemia-Reperfusion Injury

Ischaemia causes the cardiomyocytes to switch to anaerobic glycolysis to generate ATP, but this increases lactate production which causes a rapid drop in intracellular pH, driving ion exchangers to extrude protons at the expensive of accumulating intracellular Na and subsequently Ca ions. While the acidic conditions inhibit the opening of the MPTP (mitochondrial permeability transition pore), the cytosolic calcium overload results in cardiomyocyte hypercontracture. With reperfusion, the arrival of oxygen permits the re-activation of the electron transport chain, but also results in a burst of ROS (reactive oxygen species) production. In combination with Ca overload, ROS induce the opening of the MPTP, depleting ATP and damaging intracellular structures. This can either directly cause cellular death via necrosis or can cause extensive damage to cell membranes by lipid peroxidation and enzyme denaturation as well as direct oxidative damage to DNA, such that the resultant non-viable cell activates intracellular cell death signalling pathways including pyroptosis. As a consequence, the necrotic or pyroptotic cell releases its intracellular contents into the extracellular milieu. At a later time-point, several hours after reperfusion, the pro-inflammatory milieu attracts neutrophils which invade the necrotic region [10, 14, 19, 20].
During the process of reperfusion, there is an intense inflammatory process activated within the ischaemic area, coordinated by the huge influx of immune cells that circulating blood brings with it. A crucial aspect of the activated innate immune response is leukocyte infiltration into the necrotic myocardium. Leukocytes, the vast majority of which reside in the circulation or lymphatic tissue, play an important role during the process of myocardial infarction and repair. Neutrophils are the most abundant leukocyte and they infiltrate the injured myocardium early during ischaemia [21, 22]. Within the milieu of the infarcted myocardium, they exert direct effects such as tissue infiltration [23, 24], proteolysis, generation of oxygen free radicals [25], the release of pro-inflammatory cytokines, and stimulation of the complement cascade [26]. The resulting immune cascade is beyond the scope of this review; however, its complexity and the presence of multifunctioning mediators are undoubtedly why an effective anti-inflammatory therapy is yet to be developed.

Targeting Inflammation to Reduce MI Injury

There have been attempts to target the immune response during and after an infarction; however, the results of the majority of clinical trials have been disappointing. Earlier attempts were focussed on using corticosteroids, an idea borne out of cell-based studies showing that they conferred protection upon cardiomyocytes during ischaemia [27]. However, despite this promising theory, clinical trials gave conflicting results and in some cases raised serious concerns into the use of corticosteroid therapy post-infarction [28, 29]. Indeed, there is growing evidence that corticosteroids may impede fibroblast function and thus prevent healthy repair [30].
As understanding of the immune process has improved, more specific immunomodulatory therapies have been trialled in the setting of ACS. Crucial to neutrophils entering the ischaemic zone is the interaction of circulating neutrophils and the vascular endothelium. One way to block this step is through inhibiting the binding of leukocyte surface adhesion molecules such as P-selectin [3133]. Inclacumab, a P-selectin inhibitor, was used in a randomised trial of 544 NSTEMI patients, but the inconclusive results only showed a trend towards a reduction in troponin levels in the treatment arm [34]. The HALT-MI study looked at antibody-mediated inhibition of the CD11/CD18 integrin receptor on leukocytes in 420 patients with an acute MI; however, treatment with the antibody to the integrin did not reduce infarct size [35]. Tocilizumab, the antibody to IL-6 receptor, reduced levels of troponin during in-hospital stay in 117 NSTEMI patients [36]. The APEX-AMI trial looked at using Pexelizumab, a humanised monoclonal antibody that binds to C5 component of the complement cascade, during PCI after myocardial infarction. A total of 5745 patients were recruited but there was no difference in all-cause mortality after 90 days between the treatment and non-treatment group [37].
Despite these disappointing findings, the search for an effective anti-inflammatory therapy for acute MI continues. The exciting results of the recent CANTOS trial in the setting of atherosclerosis have reignited the hope of developing an equivalent effective immunomodulatory therapy for ischaemia-reperfusion injury. In the CANTOS trial, Canakinumab, a human monoclonal antibody targeting interleukin 1 beta (IL-1β), was administered to patients who had previously suffered a myocardial infarct and raised circulating CRP levels. Treatment demonstrated a 15% reduction in relative risk for the composite primary endpoint of non-fatal MI, non-fatal stroke, and death from cardiovascular disease [38].
A huge body of animal studies offer further tantalising hope of identifying new treatment targets in preventing the inflammatory damage in ischaemia-reperfusion injuries. Modulating pro-inflammatory cytokines in animal models have yielded exciting results; IL-1β blockade in a mouse ischaemic cardiomyopathy model significantly improved LV function [39]. Similar studies targeting IL-2 or IL-18 in animal models have demonstrated significant reduction in cardiomyocyte death and improvement in LV function [40, 41]. Chemokines [42], inflammasomes, and TGF-β [43] are some of the other targets that have demonstrated potential benefit in animal models of ischaemia-reperfusion injury.
However, one avenue that has received limited coverage as a potential new target is the role of extracellular DNA and the innate immune system. Innate immunity is the first line of defence to tissue injury and represents an evolutionary older branch in comparison with the more specific and targeted adaptive immunity. The innate immune system first appeared 750 million years ago and has been remarkably conserved throughout the evolutionary tree of life [44, 45]. A key initiator of an innate immune response is immune stimulation via a DAMP [11]. Therefore, identification of DAMPs released during ischaemia-reperfusion injury may lead to the identification of a valuable drug target. The appearance of excessive amounts of intact, high-molecular-weight, extracellular DNA is one of many differences between controlled cell death pathways such as apoptosis and uncontrolled necrotic cell death [46, 47]. When cells undergo apoptosis, intracellular DNA is methodically degraded and shielded from the immune system by retention within plasma membrane vesicles (apoptotic bodies). However, during ischaemia, cells die primarily via a process of necrosis during which DNA is released into the extracellular space and blood. The nucleus of every eukaryotic cell (except red blood cells, which do not have a nucleus) contains approximately 6 pg of DNA. Therefore, since the human myocardium contains approximately 5 billion cells, a large left ventricular myocardial infarct could cause the death of ~ 1 billion cardiomyocytes which can potentially release ~ 1 mg of DNA and DNA fragments into the extracellular space [48, 49]. This huge quantity of DNA is then free to diffuse within the necrotic milieu of the infarct zone.
Another large source of extracellular DNA during ischaemia and reperfusion comes from infiltrating leukocytes. In response to a TLR-dependent process, neutrophils, the most abundant leukocytes in the myocardium during reperfusion, discharge their nuclear DNA forming an extracellular net of DNA rich in histones [50]. This process, termed NETosis, ultimately kills the neutrophil whilst it lays down a histone-rich mechanical mesh which traps debris. NETs (neutrophil extracellular traps) can break down and release histones causing further damage to tissue remote from the initial necrotic site [51]. The NETs also play a crucial role in thrombosis, platelet aggregation, and occluding blood vessel further exaggerating coronary ischaemia [52]. Thrombi aspirated from the coronary arteries of patients who suffered STEMI demonstrate that the burden of NETosis positively correlates with infarct size and negatively correlates with ST segment resolution [53].
The crucial role of DNA breakdown in normal physiology is highlighted by the fact that mice lacking the DNA cleaving enzyme DNAse II die shortly after birth [54]. Furthermore, deficiencies in the normal biological process of DNA digestion and processing are linked to diseases with inappropriately active innate immune systems or autoimmunity [5557]. Elevated levels of circulating DNA are also associated with a variety of conditions from trauma, tumour malignancy, and sepsis, all of which are themselves associated with a degree of immune activation or inflammation [58, 59]. Each DNA nucleosome core consists of superhelical DNA wound around an octamer of histones, composed of two copies of each of the core histones H2A, H2B, H3, and H4 [60]. The linker histone H1 binds to the complete nucleosome core particle and forms higher order structures [61]. After a necrotic event, cellular DNA may be released either as DNA fragments or as nucleosomes, both of which are well-recognised DAMPs [18]. The extent to which mammalian DNA components are cytotoxic was first investigated by Xu et al., who showed that intravenous injection of isolated histones into mice caused death through sepsis within minutes [62]. Curiously, administration of intact nucleosomes did not have this effect. It has now been shown that this effect is mediated through the toll-like receptors TLR2 and TLR4, two crucial receptors involved in activation of the innate immune system [63]. Unlike human DNA, foreign microbial DNA has also long been accepted as a potent stimulator of the innate immune system [64]. A heterogeneous group of pattern recognition receptors on immune cell surfaces detects foreign microbial nucleic acids, including TLR3, TLR7–TLR9 [65, 66].
A certain quantity of cell-free, circulating DNA is understood to be part of the normal physiological state in both humans and rodents; its concentration is tightly controlled by extracellular DNAases [67]. It is now believed that during normal cellular process such as cell division, the amount of extracellular DNA remains manageable through continuous degradation to maintain a level below the immuno-stimulatory threshold [68, 69]. If, however, the threshold is breached in conditions such as necrotic cell death, the DNA may act on the same pathogenic receptor pathways stimulating an innate immune response [7072]. A well-documented finding in auto-inflammatory conditions is the presence of circulating cell-free DNA incorporated into immune complexes [73], confirming the ability of DNA to act as an auto-antigen. Previously, it was believed that the presence of unmethylated CpG dinucleotides in microbial DNA conferred foreign DNAs ability to interact with TLR9 and stimulate the innate immune response. Mammalian purified DNA dinucleotides are mostly methylated so in theory they should not exhibit an immuno-stimulatory response, but mammalian complexed DNA, either within histones or DNA-binding proteins, has been demonstrated to induce TLR9-mediated signalling [74, 75]. Extracellular mammalian DNA has been shown to have this effect both by interacting with TLR9 [76] to activate the innate immune system and by TLR-independent mechanism which increase the transcription of type 1 interferons a potent pro-inflammatory cytokine [7779]. Unlike naked DNA, cell-free chromatin contains abundant proteins that may expose epitopes for helper T cells to identify them as foreign. Indeed, the appearance of antibodies to chromatin precedes the occurrence of anti-DNA antibodies suggesting chromatin plays a crucial role in developing an auto-inflammatory response to self-DNA [59].

The Role of Extracellular Histones in Innate Immunity

Apoptotic or necrotic cells induced by ischaemia release histones [80] either as part of nucleosome fragments or on their own. These extracellular histones have also been shown to trigger inflammation and cell death, either by stimulating pro-inflammatory cytokines resulting in the activation of cell death pathways or through the process of neutrophil extracellular traps. In human observational studies, raised histone serum levels have been demonstrated in multiple trauma patients and correlate with severity of coagulopathy, endothelial damage, and inflammation [81]. A large body of evidence demonstrates that histone-induced cell toxicity plays a crucial role in cell death during ischaemia-reperfusion injury of the myocardium. Histones are known to activate TLR2 and TLR4; furthermore, TLR knockout mice are protected from the lethal effects of histones [63]. In an ischaemic stroke model, histone infusion is correlated with large infarct size and conversely histone neutralisation via an antibody infusion results in a reduction in infarct size [82]. In a toxic liver injury model, free histones mediated cytotoxicity of liver cells via a TLR-dependent process—an effect that was abrogated by anti-histone antibodies [63]. Furthermore, in liver cells, histone-stimulated TLR activation results in activation of the intracellular NLRP3 inflammasome and subsequent pyroptosis [83]. Histones have been shown to mediate endothelial cell cytotoxicity, resulting in acute lung haemorrhage, thrombosis, and oedema [84]. Similar cytotoxic effects of histones have been demonstrated in kidney injury [85], sepsis [62], and even hair follicle death [86]. There is also increasing evidence that histones can cause cytotoxicity independent of immunostimulation, damaging endothelial cells and stimulating an influx of intracellular calcium and subsequent necrosis [84]. Thus, increasing evidence suggests free histones function as DAMPs, leading to both inflammatory and toxic responses culminating in cell death.

DNA and Cell Death (Inflammasome Activation and Pyroptosis)

It has been shown that extracellular DNA and histones could function as an alarmin or DAMP by causing activation of TLRs. As well as playing a crucial role in the innate immune system, activation of TLR can activate cell death pathways such as pyroptosis. It is now believed that during ischaemia and reperfusion, pyroptosis may contribute to infarct size and subsequent poor remodelling of the myocardium [12, 87, 88]. This has culminated in a great deal of interest in targeting intracellular cell death pathways to limit the degree of cell death during myocardial infarction [8, 89, 90].
During ischaemia, necrotic cell debris including DNA fragments circulate in the extracellular matrix creating a pro-inflammatory milieu. Activation of the TLRs on surviving cardiac cells in the border zone of the infarct area leads to activation of downstream intracellular signalling pathways, which convene to result in NF-κB mediated expression of the protein components that make up the NLRP3 inflammasome [91, 92]. Following a secondary trigger, the individual protein components which now exist in the cytoplasm of the cell aggregate to form a multiprotein oligomer also called the inflammasome complex [93, 94]. This complex is now able to interact with pro-caspase-1 and leads to its conversion into its active caspase-1 form [95]. Caspase-1 begins the subsequent autocatalytic activation of the pro-inflammatory cytokines IL-1β and IL-18 [96]. An additional substrate of caspase-1 is the cytosolic protein, gasdermin D (GSDMD) [9799]. Following cleavage by caspase-1, the N-terminal fragments of GSDMD (GSDMD-N) oligomerise within the cell membrane to form pores. These pores result in loss of cell membrane integrity, leading pyroptotic cell death [100]. The pores also increase membrane permeability to IL-1β and IL-18 leading to their extracellular release, and these cytokines amplify the inflammatory response and mediate further injury [42, 87, 88, 101108].

Evidence for a Role of Extracellular DNA in Ischaemia-Reperfusion Injury of the Myocardium

A number of animal studies have proven that self-DNA may be an effective target to inhibit inflammation and myocyte death during ischaemia-reperfusion injury. In a murine model, it was demonstrated that histones caused cardiomyocyte toxicity and an in vivo heart ischaemia-reperfusion model, DNAse 1 treatment, disrupted extracellular cytotoxic chromatin resulting in a reduction myocardial histone concentration [17]. This correlated with a significant improvement in left ventricular remodelling and cardiomyocyte survival. Ge et al. supported this finding in a murine model of ischaemia-reperfusion showing that DNAse with the addition of recombinant tissue-type plasminogen activator resulted in a reduction of infarct size as well as decreasing the density of neutrophil-associated NETs [109]. This also leads to an improvement in left ventricular remodelling. Curiously, this effect was not observed when DNAse or rt-PA was administered on its own [109]. Savchenko et al. administered DNAse to PAD4-/- mice which do not produce NETs as well as wild type mice [110].The study demonstrated that myocardial ischaemia-reperfusion injury caused an increase in nucleosomes, neutrophil infiltration, and histone H3 at the site of injury. Treatment with DNAse improved cardiac contractile function to a similar degree in both wild type and PAD4-/- deficient mice. This suggests that DNA fragments contribute to cardiomyocyte dysfunction during reperfusion irrespective of NETs, possibly by acting as a DAMP. Using an in vivo rat model, Downey’s group have made similar findings, demonstrating that DNAse administered after 30 min of coronary artery occlusion resulted in a significant reduction in infarct size [111]. Interestingly, the addition of mitochondrial DNA inhibitor with DNAse resulted in a greater reduction in infarct size then that which was seen with DNAse alone [111]. This would suggest that nuclear DNA could be acting through a different pathway than the well-recognised DAMP, mitochondrial DNA.
Endothelial dysfunction plays a crucial role in ischaemia-reperfusion injury, contributing to myocardial stunning, microvascular obstruction, and exposing the myocytes to toxic stimuli which contribute to lethal myocardial injury [112114]. Heparin consists of a high volume of negatively charged sulphated proteoglycans, binds to histones, and inactivates them through high-affinity electrostatic interactions [115]. It has long been shown that heparin protects the coronary endothelium and myocardium from ischaemia-reperfusion injury [116119]. Both heparin and chondroitin sulphate have both been shown to protect vascular endothelial cells from histone-induced cytotoxicity in vitro [120, 121]. Furthermore, heparin derivatives reduce infarct size in a rat model of ischaemia-reperfusion by inhibiting caspase-dependent, cell death pathways [122].

Summary

In the quest to protect cardiomyocytes from the deleterious effects of ischaemia-reperfusion injury, identification of pyroptosis as a contributing factor to infarct size has revealed a target for future cardioprotective therapies. Extracellular DNA fragments from dead cells and neutrophils are potent instigators of TLR-dependent inflammasome activation and subsequent pyroptosis. The use of DNAse and DNA-neutralising therapies has shown some promise in animal models of preventing the damaging effects of ischaemia-reperfusion injury. Identification and targeting the instigators of pyroptosis may provide benefit in limiting cell death post-infarction and preventing the morbidity and mortality associated with ischaemia and reperfusion injury.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

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

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

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat World Health Organization. Global status report on noncommunicable diseases 2014. World Health. 2014;176–9. World Health Organization. Global status report on noncommunicable diseases 2014. World Health. 2014;176–9.
2.
Zurück zum Zitat Joseph P, Leong D, McKee M, Anand SS, Schwalm JD, Teo K, et al. Reducing the global burden of cardiovascular disease, part 1: the epidemiology and risk factors. Circ Res. 2017;121:677–94.CrossRefPubMed Joseph P, Leong D, McKee M, Anand SS, Schwalm JD, Teo K, et al. Reducing the global burden of cardiovascular disease, part 1: the epidemiology and risk factors. Circ Res. 2017;121:677–94.CrossRefPubMed
3.
Zurück zum Zitat Bansilal S, Castellano JM, Fuster V. Global burden of CVD: focus on secondary prevention of cardiovascular disease. Int J Cardiol. 2015;201:S1–7.CrossRefPubMed Bansilal S, Castellano JM, Fuster V. Global burden of CVD: focus on secondary prevention of cardiovascular disease. Int J Cardiol. 2015;201:S1–7.CrossRefPubMed
4.
Zurück zum Zitat Briceno N, Schuster A, Lumley M, Perera D. Ischaemic cardiomyopathy: pathophysiology, assessment and the role of revascularisation. Heart. 2016;102:396–406. Briceno N, Schuster A, Lumley M, Perera D. Ischaemic cardiomyopathy: pathophysiology, assessment and the role of revascularisation. Heart. 2016;102:396–406.
5.
Zurück zum Zitat Andreadou I, Cabrera-Fuentes H, Devaux Y, Frangogiannis NG, Frantz S, Guzik T, et al. Immune cells as targets for cardioprotection: new players and novel therapeutic opportunities. Cardiovasc Res. 2019;115:1117–30.CrossRefPubMedPubMedCentral Andreadou I, Cabrera-Fuentes H, Devaux Y, Frangogiannis NG, Frantz S, Guzik T, et al. Immune cells as targets for cardioprotection: new players and novel therapeutic opportunities. Cardiovasc Res. 2019;115:1117–30.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Davidson SM, Arjun S, Basalay MV, Bell RM, Bromage DI, Bøtker HE, et al. The 10th biennial hatter cardiovascular institute workshop: cellular protection—evaluating new directions in the setting of myocardial infarction, ischaemic stroke, and cardio-oncology. Basic Res Cardiol. 2018;113:43.CrossRefPubMedPubMedCentral Davidson SM, Arjun S, Basalay MV, Bell RM, Bromage DI, Bøtker HE, et al. The 10th biennial hatter cardiovascular institute workshop: cellular protection—evaluating new directions in the setting of myocardial infarction, ischaemic stroke, and cardio-oncology. Basic Res Cardiol. 2018;113:43.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Hausenloy DJ, Garcia-Dorado D, Bøtker HE, Davidson SM, Downey J, Engel FB, et al. Novel targets and future strategies for acute cardioprotection: position paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart. Cardiovasc Res. 2017;113:564–85.CrossRefPubMed Hausenloy DJ, Garcia-Dorado D, Bøtker HE, Davidson SM, Downey J, Engel FB, et al. Novel targets and future strategies for acute cardioprotection: position paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart. Cardiovasc Res. 2017;113:564–85.CrossRefPubMed
8.
Zurück zum Zitat Hausenloy DJ, Barrabes JA, Bøtker HE, Davidson SM, Di Lisa F, Downey J, et al. Ischaemic conditioning and targeting reperfusion injury: a 30 year voyage of discovery. Basic Res Cardiol. 2016;111:70.CrossRefPubMedPubMedCentral Hausenloy DJ, Barrabes JA, Bøtker HE, Davidson SM, Di Lisa F, Downey J, et al. Ischaemic conditioning and targeting reperfusion injury: a 30 year voyage of discovery. Basic Res Cardiol. 2016;111:70.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Bell RM, Bøtker HE, Carr RD, Davidson SM, Downey JM, Dutka DP, et al. 9th Hatter Biannual Meeting: position document on ischaemia/reperfusion injury, conditioning and the ten commandments of cardioprotection. Basic Res Cardiol. 2016;111:41.CrossRefPubMedPubMedCentral Bell RM, Bøtker HE, Carr RD, Davidson SM, Downey JM, Dutka DP, et al. 9th Hatter Biannual Meeting: position document on ischaemia/reperfusion injury, conditioning and the ten commandments of cardioprotection. Basic Res Cardiol. 2016;111:41.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Hausenloy DJ, Yellon DM. Ischaemic conditioning and reperfusion injury. Nat Rev Cardiol. 2016;13:193–209.CrossRefPubMed Hausenloy DJ, Yellon DM. Ischaemic conditioning and reperfusion injury. Nat Rev Cardiol. 2016;13:193–209.CrossRefPubMed
11.
Zurück zum Zitat Matzinger P. The danger model: a renewed sense of self. Science (80-). 2002;296:301–5.CrossRef Matzinger P. The danger model: a renewed sense of self. Science (80-). 2002;296:301–5.CrossRef
12.
13.
Zurück zum Zitat Takahashi M. NLRP3 inflammasome as a novel player in myocardial infarction. Int Heart J. 2014;55:101–5.CrossRefPubMed Takahashi M. NLRP3 inflammasome as a novel player in myocardial infarction. Int Heart J. 2014;55:101–5.CrossRefPubMed
14.
Zurück zum Zitat Rauf A, Shah M, Yellon DM, Davidson SM. Role of caspase 1 in ischemia/reperfusion injury of the myocardium. J Cardiovasc Pharmacol. 2019;74:194–200.CrossRefPubMed Rauf A, Shah M, Yellon DM, Davidson SM. Role of caspase 1 in ischemia/reperfusion injury of the myocardium. J Cardiovasc Pharmacol. 2019;74:194–200.CrossRefPubMed
15.
Zurück zum Zitat Kalbitz M, Grailer JJ, Fattahi F, Jajou L, Herron TJ, Campbell KF, et al. Role of extracellular histones in the cardiomyopathy of sepsis. FASEB J. 2015;29:2185–93.CrossRefPubMedPubMedCentral Kalbitz M, Grailer JJ, Fattahi F, Jajou L, Herron TJ, Campbell KF, et al. Role of extracellular histones in the cardiomyopathy of sepsis. FASEB J. 2015;29:2185–93.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Fattahi F, Russell MW, Malan EA, Parlett M, Abe E, Zetoune FS, et al. Harmful roles of TLR3 and TLR9 in cardiac dysfunction developing during polymicrobial Sepsis. Biomed Res Int. 2018;2018:1–10.CrossRef Fattahi F, Russell MW, Malan EA, Parlett M, Abe E, Zetoune FS, et al. Harmful roles of TLR3 and TLR9 in cardiac dysfunction developing during polymicrobial Sepsis. Biomed Res Int. 2018;2018:1–10.CrossRef
17.
Zurück zum Zitat Vogel B, Shinagawa H, Hofmann U, Ertl G, Frantz S. Acute DNase1 treatment improves left ventricular remodeling after myocardial infarction by disruption of free chromatin. Basic Res Cardiol. 2015;110:15.CrossRefPubMed Vogel B, Shinagawa H, Hofmann U, Ertl G, Frantz S. Acute DNase1 treatment improves left ventricular remodeling after myocardial infarction by disruption of free chromatin. Basic Res Cardiol. 2015;110:15.CrossRefPubMed
19.
20.
Zurück zum Zitat Hausenloy DJ, Yellon DM. The mitochondrial permeability transition pore: its fundamental role in mediating cell death during ischaemia and reperfusion. J Mol Cell Cardiol. 2003;35:339–41.CrossRefPubMed Hausenloy DJ, Yellon DM. The mitochondrial permeability transition pore: its fundamental role in mediating cell death during ischaemia and reperfusion. J Mol Cell Cardiol. 2003;35:339–41.CrossRefPubMed
21.
Zurück zum Zitat Engler RL, Dahlgren MD, Peterson MA, Dobbs A, Schmid-Schönbein GW. Accumulation of polymorphonuclear leukocytes during 3-h experimental myocardial ischemia. Am J Phys. 1986;251:H93–100. Engler RL, Dahlgren MD, Peterson MA, Dobbs A, Schmid-Schönbein GW. Accumulation of polymorphonuclear leukocytes during 3-h experimental myocardial ischemia. Am J Phys. 1986;251:H93–100.
22.
Zurück zum Zitat Jolly SR, Kane WJ, Hook BG, Abrams GD, Kunkel SL, Lucchesi BR. Reduction of myocardial infarct size by neutrophil depletion: effect of duration of occlusion. Am Heart J. 1986;112:682–90.CrossRefPubMed Jolly SR, Kane WJ, Hook BG, Abrams GD, Kunkel SL, Lucchesi BR. Reduction of myocardial infarct size by neutrophil depletion: effect of duration of occlusion. Am Heart J. 1986;112:682–90.CrossRefPubMed
23.
Zurück zum Zitat Bienvenu K, Granger DN. Molecular determinants of shear rate-dependent leukocyte adhesion in postcapillary venules. Am J Phys. 1993;264:H1504–8. Bienvenu K, Granger DN. Molecular determinants of shear rate-dependent leukocyte adhesion in postcapillary venules. Am J Phys. 1993;264:H1504–8.
24.
Zurück zum Zitat Gasic AC, McGuire G, Krater S, Farhood AI, Goldstein MA, Smith CW, et al. Hydrogen peroxide pretreatment of perfused canine vessels induces ICAM-1 and CD18-dependent neutrophil adherence. Circulation. 1991;84:2154–66.CrossRefPubMed Gasic AC, McGuire G, Krater S, Farhood AI, Goldstein MA, Smith CW, et al. Hydrogen peroxide pretreatment of perfused canine vessels induces ICAM-1 and CD18-dependent neutrophil adherence. Circulation. 1991;84:2154–66.CrossRefPubMed
25.
Zurück zum Zitat Duilio C, Ambrosio G, Kuppusamy P, DiPaula A, Becker LC, Zweier JL. Neutrophils are primary source of O2 radicals during reperfusion after prolonged myocardial ischemia. Am J Physiol Heart Circ Physiol. 2001;280:H2649–57.CrossRefPubMed Duilio C, Ambrosio G, Kuppusamy P, DiPaula A, Becker LC, Zweier JL. Neutrophils are primary source of O2 radicals during reperfusion after prolonged myocardial ischemia. Am J Physiol Heart Circ Physiol. 2001;280:H2649–57.CrossRefPubMed
27.
Zurück zum Zitat Libby P, Maroko PR, Bloor CM, Sobel BE, Braunwald E. Reduction of experimental myocardial infarct size by corticosteroid administration. J Clin Invest. 1973;52:599–607.CrossRefPubMedPubMedCentral Libby P, Maroko PR, Bloor CM, Sobel BE, Braunwald E. Reduction of experimental myocardial infarct size by corticosteroid administration. J Clin Invest. 1973;52:599–607.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Giugliano GR, Giugliano RP, Gibson CM, Kuntz RE. Meta-analysis of corticosteroid treatment in acute myocardial infarction. Am J Cardiol. 2003;91:1055–9.CrossRefPubMed Giugliano GR, Giugliano RP, Gibson CM, Kuntz RE. Meta-analysis of corticosteroid treatment in acute myocardial infarction. Am J Cardiol. 2003;91:1055–9.CrossRefPubMed
29.
Zurück zum Zitat Roberts R, DeMello V, Sobel BE, Koerting A, Ren G, Abou-Khamis T, et al. Deleterious effects of methylprednisolone in patients with myocardial infarction. Circulation. 1976;53:I204–6.CrossRefPubMed Roberts R, DeMello V, Sobel BE, Koerting A, Ren G, Abou-Khamis T, et al. Deleterious effects of methylprednisolone in patients with myocardial infarction. Circulation. 1976;53:I204–6.CrossRefPubMed
30.
Zurück zum Zitat Kloner RA, Fishbein MC, Lew H, Maroko PR, Braunwald E. Mummification of the infarcted myocardium by high dose corticosteroids. Circulation. 1978;57:56–63.CrossRefPubMed Kloner RA, Fishbein MC, Lew H, Maroko PR, Braunwald E. Mummification of the infarcted myocardium by high dose corticosteroids. Circulation. 1978;57:56–63.CrossRefPubMed
31.
Zurück zum Zitat Diacovo TG, Roth SJ, Buccola JM, Bainton DF, Springer T A. Neutrophil rolling, arrest, and transmigration across activated, surface-adherent platelets via sequential action of P-selectin and the beta 2-integrin CD11b/CD18. Blood. 1996;88:146–57.CrossRefPubMed Diacovo TG, Roth SJ, Buccola JM, Bainton DF, Springer T A. Neutrophil rolling, arrest, and transmigration across activated, surface-adherent platelets via sequential action of P-selectin and the beta 2-integrin CD11b/CD18. Blood. 1996;88:146–57.CrossRefPubMed
32.
Zurück zum Zitat Burns AR, Bowden RA, Abe Y, Walker DC, Simon SI, Entman ML, et al. P-selectin mediates neutrophil adhesion to endothelial cell borders. J Leukoc Biol. 1999;65:299–306.CrossRefPubMed Burns AR, Bowden RA, Abe Y, Walker DC, Simon SI, Entman ML, et al. P-selectin mediates neutrophil adhesion to endothelial cell borders. J Leukoc Biol. 1999;65:299–306.CrossRefPubMed
33.
Zurück zum Zitat Jones DA, Abbassi O, McIntire LV, McEver RP, Smith CW. P-selectin mediates neutrophil rolling on histamine-stimulated endothelial cells. Biophys J. 1993;65:1560–9.CrossRefPubMedPubMedCentral Jones DA, Abbassi O, McIntire LV, McEver RP, Smith CW. P-selectin mediates neutrophil rolling on histamine-stimulated endothelial cells. Biophys J. 1993;65:1560–9.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Tardif JC, Tanguay JF, Wright SS, Duchatelle V, Petroni T, Grégoire JC, et al. Effects of the P-selectin antagonist inclacumab on myocardial damage after percutaneous coronary intervention for non-st-segment elevation myocardial infarction: results of the SELECT-ACS trial. J Am Coll Cardiol. 2013;61:2048–55.CrossRefPubMed Tardif JC, Tanguay JF, Wright SS, Duchatelle V, Petroni T, Grégoire JC, et al. Effects of the P-selectin antagonist inclacumab on myocardial damage after percutaneous coronary intervention for non-st-segment elevation myocardial infarction: results of the SELECT-ACS trial. J Am Coll Cardiol. 2013;61:2048–55.CrossRefPubMed
35.
Zurück zum Zitat Faxon DP, Gibbons RJ, Chronos NAF, Gurbel PA, Sheehan F. The effect of blockade of the CD11/CD18 integrin receptor on infarct size in patients with acute myocardial infarction treated with direct angioplasty: the results of the HALT-MI study. J Am Coll Cardiol. 2002;40:1199–204.CrossRefPubMed Faxon DP, Gibbons RJ, Chronos NAF, Gurbel PA, Sheehan F. The effect of blockade of the CD11/CD18 integrin receptor on infarct size in patients with acute myocardial infarction treated with direct angioplasty: the results of the HALT-MI study. J Am Coll Cardiol. 2002;40:1199–204.CrossRefPubMed
36.
Zurück zum Zitat Kleveland G, Bratlie M, Ueland T, Amundsen B, Aakhus S, Damaas JK, et al. The interleukin-6 receptor antagonist tocilizumab reduces inflammation and myocardial damage in non-ST elevation myocardial infarction-a randomized, double-blind, placebo controlled study. Eur Heart J. 2015;36:27. Kleveland G, Bratlie M, Ueland T, Amundsen B, Aakhus S, Damaas JK, et al. The interleukin-6 receptor antagonist tocilizumab reduces inflammation and myocardial damage in non-ST elevation myocardial infarction-a randomized, double-blind, placebo controlled study. Eur Heart J. 2015;36:27.
37.
Zurück zum Zitat Armstrong PW, Granger CB, Adams PX, Hamm C, Holmes D, O’Neill WW, et al. Pexelizumab for acute ST-elevation myocardial infarction in patients undergoing primary percutaneous coronary intervention: a randomized controlled trial. JAMA. 2007;297:43–51.CrossRefPubMed Armstrong PW, Granger CB, Adams PX, Hamm C, Holmes D, O’Neill WW, et al. Pexelizumab for acute ST-elevation myocardial infarction in patients undergoing primary percutaneous coronary intervention: a randomized controlled trial. JAMA. 2007;297:43–51.CrossRefPubMed
38.
Zurück zum Zitat Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, et al. Antiinflammatory therapy with Canakinumab for atherosclerotic disease. N Engl J Med. 2017;377:1119–31.CrossRefPubMed Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, et al. Antiinflammatory therapy with Canakinumab for atherosclerotic disease. N Engl J Med. 2017;377:1119–31.CrossRefPubMed
39.
Zurück zum Zitat Toldo S, Mezzaroma E, Bressi E, Marchetti C, Carbone S, Sonnino C, et al. Interleukin-1beta blockade improves left ventricular systolic/diastolic function and restores contractility reserve in severe ischemic cardiomyopathy in the mouse. J Cardiovasc Pharmacol. 2014;64:1–6.CrossRefPubMed Toldo S, Mezzaroma E, Bressi E, Marchetti C, Carbone S, Sonnino C, et al. Interleukin-1beta blockade improves left ventricular systolic/diastolic function and restores contractility reserve in severe ischemic cardiomyopathy in the mouse. J Cardiovasc Pharmacol. 2014;64:1–6.CrossRefPubMed
40.
Zurück zum Zitat Abbate A, Salloum FN, van Tassell BW, Vecile E, Toldo S, Seropian I, et al. Alterations in the interleukin-1/interleukin-1 receptor antagonist balance modulate cardiac remodeling following myocardial infarction in the mouse. PLoS One. 2011;6:11. Abbate A, Salloum FN, van Tassell BW, Vecile E, Toldo S, Seropian I, et al. Alterations in the interleukin-1/interleukin-1 receptor antagonist balance modulate cardiac remodeling following myocardial infarction in the mouse. PLoS One. 2011;6:11.
41.
Zurück zum Zitat Venkatachalam K, Prabhu SD, Reddy VS, Boylston WH, Valente AJ, Chandrasekar B. Neutralization of interleukin-18 ameliorates ischemia/reperfusion-induced myocardial injury. J Biol Chem. 2009;284:7853–65.CrossRefPubMedPubMedCentral Venkatachalam K, Prabhu SD, Reddy VS, Boylston WH, Valente AJ, Chandrasekar B. Neutralization of interleukin-18 ameliorates ischemia/reperfusion-induced myocardial injury. J Biol Chem. 2009;284:7853–65.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Dobaczewski M, Xia Y, Bujak M, Gonzalez-Quesada C, Frangogiannis NG. CCR5 signaling suppresses inflammation and reduces adverse remodeling of the infarcted heart, mediating recruitment of regulatory T cells. Am J Pathol. 2010;176:2177–87.CrossRefPubMedPubMedCentral Dobaczewski M, Xia Y, Bujak M, Gonzalez-Quesada C, Frangogiannis NG. CCR5 signaling suppresses inflammation and reduces adverse remodeling of the infarcted heart, mediating recruitment of regulatory T cells. Am J Pathol. 2010;176:2177–87.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Bujak M, Ren G, Kweon HJ, Dobaczewski M, Reddy A, Taffet G, et al. Essential role of Smad3 in infarct healing and in the pathogenesis of cardiac remodeling. Circulation. 2007;116:2127–38.CrossRefPubMed Bujak M, Ren G, Kweon HJ, Dobaczewski M, Reddy A, Taffet G, et al. Essential role of Smad3 in infarct healing and in the pathogenesis of cardiac remodeling. Circulation. 2007;116:2127–38.CrossRefPubMed
44.
Zurück zum Zitat Kimbrell DA, Beutler B. The evolution and genetics of innate immunity. Nat Rev Genet. 2001;2:256–67.CrossRefPubMed Kimbrell DA, Beutler B. The evolution and genetics of innate immunity. Nat Rev Genet. 2001;2:256–67.CrossRefPubMed
45.
Zurück zum Zitat Cooper MD, Herrin BR. How did our complex immune system evolve? Nat Rev Immunol. 2010;10:2–3.CrossRefPubMed Cooper MD, Herrin BR. How did our complex immune system evolve? Nat Rev Immunol. 2010;10:2–3.CrossRefPubMed
46.
Zurück zum Zitat Fadeel B, Orrenius S. Apoptosis: a basic biological phenomenon with wide-ranging implications in human disease. J Intern Med. 2005;258:479–517.CrossRefPubMed Fadeel B, Orrenius S. Apoptosis: a basic biological phenomenon with wide-ranging implications in human disease. J Intern Med. 2005;258:479–517.CrossRefPubMed
47.
Zurück zum Zitat Kerr JFR, Wyllie A, Currie A. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. J Intern Med. 1972;258:479–517. Kerr JFR, Wyllie A, Currie A. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. J Intern Med. 1972;258:479–517.
48.
Zurück zum Zitat Nagata S, Hanayama R, Kawane K. Autoimmunity and the clearance of dead cells. Cell. 2010;140:619–30.CrossRefPubMed Nagata S, Hanayama R, Kawane K. Autoimmunity and the clearance of dead cells. Cell. 2010;140:619–30.CrossRefPubMed
49.
Zurück zum Zitat Michlewska S, McColl A, Rossi A, Megson I, Dransfield I. Clearance of dying cells and autoimmunity. Autoimmunity. 2007;40(4):267–73.CrossRefPubMed Michlewska S, McColl A, Rossi A, Megson I, Dransfield I. Clearance of dying cells and autoimmunity. Autoimmunity. 2007;40(4):267–73.CrossRefPubMed
50.
Zurück zum Zitat Silk E, Zhao H, Weng H, Ma D. The role of extracellular histone in organ injury. Cell Death Dis. 2017;8:2812.CrossRef Silk E, Zhao H, Weng H, Ma D. The role of extracellular histone in organ injury. Cell Death Dis. 2017;8:2812.CrossRef
51.
Zurück zum Zitat Pfeiler S, Stark K, Massberg S, Engelmann B. Propagation of thrombosis by neutrophils and extracellular nucleosome networks. Haematologica. 2017;102:206–13.CrossRefPubMedPubMedCentral Pfeiler S, Stark K, Massberg S, Engelmann B. Propagation of thrombosis by neutrophils and extracellular nucleosome networks. Haematologica. 2017;102:206–13.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Hoeksema M, Van Eijk M, Haagsman HP, Hartshorn KL. Histones as mediators of host defense, inflammation and thrombosis. Future Microbiol. 2016;11:441–53.CrossRefPubMedPubMedCentral Hoeksema M, Van Eijk M, Haagsman HP, Hartshorn KL. Histones as mediators of host defense, inflammation and thrombosis. Future Microbiol. 2016;11:441–53.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Mangold A, Alias S, Scherz T, Hofbauer T, Jakowitsch J, Panzenböck A, et al. Coronary neutrophil extracellular trap burden and deoxyribonuclease activity in ST-elevation acute coronary syndrome are predictors of ST-segment resolution and infarct size. Circ Res. 2015;116:1182–92.CrossRefPubMed Mangold A, Alias S, Scherz T, Hofbauer T, Jakowitsch J, Panzenböck A, et al. Coronary neutrophil extracellular trap burden and deoxyribonuclease activity in ST-elevation acute coronary syndrome are predictors of ST-segment resolution and infarct size. Circ Res. 2015;116:1182–92.CrossRefPubMed
54.
Zurück zum Zitat Kawane K, Fukuyama H, Yoshida H, Nagase H, Ohsawa Y, Uchiyama Y, et al. Impaired thymic development in mouse embryos deficient in apoptotic DNA degradation. Nat Immunol. 2003;4:138–44.CrossRefPubMed Kawane K, Fukuyama H, Yoshida H, Nagase H, Ohsawa Y, Uchiyama Y, et al. Impaired thymic development in mouse embryos deficient in apoptotic DNA degradation. Nat Immunol. 2003;4:138–44.CrossRefPubMed
55.
Zurück zum Zitat Nishimoto S, Kawane K, Watanabe-Fukunaga R, Fukuyama H, Ohsawa Y, Uchiyama Y, et al. Nuclear cataract caused by a lack of DNA degradation in the mouse eye lens. Nature. 2003;424:1071–4.CrossRefPubMed Nishimoto S, Kawane K, Watanabe-Fukunaga R, Fukuyama H, Ohsawa Y, Uchiyama Y, et al. Nuclear cataract caused by a lack of DNA degradation in the mouse eye lens. Nature. 2003;424:1071–4.CrossRefPubMed
56.
Zurück zum Zitat Crow YJ, Rehwinkel J. Aicardi-Goutie’res syndrome and related phenotypes: linking nucleic acid metabolism with autoimmunity. Hum Mol Genet. 2009;18:130–6.CrossRef Crow YJ, Rehwinkel J. Aicardi-Goutie’res syndrome and related phenotypes: linking nucleic acid metabolism with autoimmunity. Hum Mol Genet. 2009;18:130–6.CrossRef
57.
Zurück zum Zitat Krieser RJ, MacLea KS, Park JP, Eastman A. The cloning, genomic structure, localization, and expression of human deoxyribonuclease IIbeta. Gene. 2001;269:205–16.CrossRefPubMed Krieser RJ, MacLea KS, Park JP, Eastman A. The cloning, genomic structure, localization, and expression of human deoxyribonuclease IIbeta. Gene. 2001;269:205–16.CrossRefPubMed
58.
Zurück zum Zitat Kaplan MJ, Radic M. Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol. 2012;189:2689–95.CrossRefPubMed Kaplan MJ, Radic M. Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol. 2012;189:2689–95.CrossRefPubMed
60.
61.
Zurück zum Zitat Kirmes I, Szczurek A, Prakash K, Charapitsa I, Heiser C, Musheev M, et al. A transient ischemic environment induces reversible compaction of chromatin. Genome Biol. 2015;16:1–19.CrossRef Kirmes I, Szczurek A, Prakash K, Charapitsa I, Heiser C, Musheev M, et al. A transient ischemic environment induces reversible compaction of chromatin. Genome Biol. 2015;16:1–19.CrossRef
62.
Zurück zum Zitat Xu J, Zhang X, Pelayo R, Monestier M, Ammollo CT, Semeraro F, et al. Extracellular histones are major mediators of death in sepsis. Nat Med. 2009;15:1318–21.CrossRefPubMedPubMedCentral Xu J, Zhang X, Pelayo R, Monestier M, Ammollo CT, Semeraro F, et al. Extracellular histones are major mediators of death in sepsis. Nat Med. 2009;15:1318–21.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Xu J, Zhang X, Monestier M, Esmon NL, Esmon CT. Extracellular histones are mediators of death through TLR2 and TLR4 in mouse fatal liver injury. J Immunol. 2011;187:2626–31.CrossRefPubMed Xu J, Zhang X, Monestier M, Esmon NL, Esmon CT. Extracellular histones are mediators of death through TLR2 and TLR4 in mouse fatal liver injury. J Immunol. 2011;187:2626–31.CrossRefPubMed
64.
Zurück zum Zitat Tokunaga T, Yamamoto H, Shimada S, Abe H, Fukuda T, Fujisawa Y, et al. Antitumor activity of deoxyribonucleic acid fraction from Mycobacterium bovis BCG. I. Isolation, physicochemical characterization, and antitumor activity. J Natl Cancer Inst. 1984;72:955–62.PubMed Tokunaga T, Yamamoto H, Shimada S, Abe H, Fukuda T, Fujisawa Y, et al. Antitumor activity of deoxyribonucleic acid fraction from Mycobacterium bovis BCG. I. Isolation, physicochemical characterization, and antitumor activity. J Natl Cancer Inst. 1984;72:955–62.PubMed
65.
Zurück zum Zitat Engel A, Barton GM. Compartment-specific control of signaling from a DNA-sensing immune receptor. Sci Signal. 2010;3:150.CrossRef Engel A, Barton GM. Compartment-specific control of signaling from a DNA-sensing immune receptor. Sci Signal. 2010;3:150.CrossRef
66.
Zurück zum Zitat Lim KH, Staudt LM. Toll-like receptor signaling. Cold Spring Harb Perspect Biol. 2013;5:1. Lim KH, Staudt LM. Toll-like receptor signaling. Cold Spring Harb Perspect Biol. 2013;5:1.
67.
Zurück zum Zitat Yasutomo K, Horiuchi T, Kagami S, Tsukamoto H, Hashimura C, Urushihara M, et al. Mutation of DNASE1 in people with systemic lupus erythematosus. Nat Genet. 2001;28:313–4.CrossRefPubMed Yasutomo K, Horiuchi T, Kagami S, Tsukamoto H, Hashimura C, Urushihara M, et al. Mutation of DNASE1 in people with systemic lupus erythematosus. Nat Genet. 2001;28:313–4.CrossRefPubMed
68.
Zurück zum Zitat Nagata S, Nagase H, Kawane K, Mukae N, Fukuyama H. Degradation of chromosomal DNA during apoptosis. Cell Death Differ. 2003;10:108–16.CrossRefPubMed Nagata S, Nagase H, Kawane K, Mukae N, Fukuyama H. Degradation of chromosomal DNA during apoptosis. Cell Death Differ. 2003;10:108–16.CrossRefPubMed
69.
Zurück zum Zitat Viorritto ICB, Nikolov NP, Siegel RM. Autoimmunity versus tolerance: can dying cells tip the balance? Clin Immunol. 2007;122:125–34.CrossRefPubMed Viorritto ICB, Nikolov NP, Siegel RM. Autoimmunity versus tolerance: can dying cells tip the balance? Clin Immunol. 2007;122:125–34.CrossRefPubMed
70.
Zurück zum Zitat Bamboat ZM, Balachandran VP, Ocuin LM, Obaid H, Plitas G, DeMatteo RP. Toll-like receptor 9 inhibition confers protection from liver ischemia-reperfusion injury. Hepatology. 2010;51:621–32.CrossRefPubMed Bamboat ZM, Balachandran VP, Ocuin LM, Obaid H, Plitas G, DeMatteo RP. Toll-like receptor 9 inhibition confers protection from liver ischemia-reperfusion injury. Hepatology. 2010;51:621–32.CrossRefPubMed
71.
Zurück zum Zitat Chen C, Feng Y, Zou L, Wang L, Chen HH, Cai JY, et al. Role of extracellular RNA and TLR3-Trif signaling in myocardial ischemia-reperfusion injury. J Am Heart Assoc 2014;3. Chen C, Feng Y, Zou L, Wang L, Chen HH, Cai JY, et al. Role of extracellular RNA and TLR3-Trif signaling in myocardial ischemia-reperfusion injury. J Am Heart Assoc 2014;3.
72.
Zurück zum Zitat Gregorio J, Meller S, Conrad C, Di Nardo A, Homey B, Lauerma A, et al. Plasmacytoid dendritic cells sense skin injury and promote wound healing through type I interferons. J Exp Med. 2010;207:2921–30.CrossRefPubMedPubMedCentral Gregorio J, Meller S, Conrad C, Di Nardo A, Homey B, Lauerma A, et al. Plasmacytoid dendritic cells sense skin injury and promote wound healing through type I interferons. J Exp Med. 2010;207:2921–30.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Chan RWY, Jiang P, Peng X, Tam L-S, Liao GJW, Li EKM, et al. Plasma DNA aberrations in systemic lupus erythematosus revealed by genomic and methylomic sequencing. Proc Natl Acad Sci. 2014;111:E5302–11.CrossRefPubMedPubMedCentral Chan RWY, Jiang P, Peng X, Tam L-S, Liao GJW, Li EKM, et al. Plasma DNA aberrations in systemic lupus erythematosus revealed by genomic and methylomic sequencing. Proc Natl Acad Sci. 2014;111:E5302–11.CrossRefPubMedPubMedCentral
74.
Zurück zum Zitat Suzuki K, Mori A, Ishii KJ, Saito J, Singer DS, Klinman DM, et al. Activation of target-tissue immune-recognition molecules by double-stranded polynucleotides. Proc Natl Acad Sci. 1999;96:2285–90.CrossRefPubMedPubMedCentral Suzuki K, Mori A, Ishii KJ, Saito J, Singer DS, Klinman DM, et al. Activation of target-tissue immune-recognition molecules by double-stranded polynucleotides. Proc Natl Acad Sci. 1999;96:2285–90.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Park JH, Chang SH, Kim MC, Shin SH, Youn HJ, Kim JK, et al. Up-regulation of the expression of major histocompatibility complex class I antigens by plasmid DNA transfection in non-hematopoietic cells. FEBS Lett. 1998;436:55–60.CrossRefPubMed Park JH, Chang SH, Kim MC, Shin SH, Youn HJ, Kim JK, et al. Up-regulation of the expression of major histocompatibility complex class I antigens by plasmid DNA transfection in non-hematopoietic cells. FEBS Lett. 1998;436:55–60.CrossRefPubMed
76.
Zurück zum Zitat Yasuda K, Yu P, Kirschning CJ, Schlatter B, Schmitz F, Heit A, et al. Endosomal translocation of vertebrate DNA activates dendritic cells via TLR9-dependent and -independent pathways. J Immunol. 2005;174:6129–36.CrossRefPubMed Yasuda K, Yu P, Kirschning CJ, Schlatter B, Schmitz F, Heit A, et al. Endosomal translocation of vertebrate DNA activates dendritic cells via TLR9-dependent and -independent pathways. J Immunol. 2005;174:6129–36.CrossRefPubMed
77.
Zurück zum Zitat Okabe Y, Kawane K, Akira S, Taniguchi T, Nagata S. Toll-like receptor-independent gene induction program activated by mammalian DNA escaped from apoptotic DNA degradation. J Exp Med. 2005;202:1333–9.CrossRefPubMedPubMedCentral Okabe Y, Kawane K, Akira S, Taniguchi T, Nagata S. Toll-like receptor-independent gene induction program activated by mammalian DNA escaped from apoptotic DNA degradation. J Exp Med. 2005;202:1333–9.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Yasuda K, Rutz M, Schlatter B, Metzger J, Luppa PB, Schmitz F, et al. CpG motif-independent activation of TLR9 upon endosomal translocation of “natural” phosphodiester DNA. Eur J Immunol. 2006;36(2):431–6.CrossRefPubMed Yasuda K, Rutz M, Schlatter B, Metzger J, Luppa PB, Schmitz F, et al. CpG motif-independent activation of TLR9 upon endosomal translocation of “natural” phosphodiester DNA. Eur J Immunol. 2006;36(2):431–6.CrossRefPubMed
79.
Zurück zum Zitat Roers A, Hiller B, Hornung V. Recognition of endogenous nucleic acids by the innate immune system. Immunity. 2016;44:739–54.CrossRefPubMed Roers A, Hiller B, Hornung V. Recognition of endogenous nucleic acids by the innate immune system. Immunity. 2016;44:739–54.CrossRefPubMed
80.
Zurück zum Zitat Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61:1659–65.PubMed Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61:1659–65.PubMed
81.
Zurück zum Zitat Kaufman T, Magosevich D, Moreno MC, Guzman MA, D’Atri LP, Carestia A, et al. Nucleosomes and neutrophil extracellular traps in septic and burn patients. Clin Immunol. 2017;183:254–62.CrossRefPubMed Kaufman T, Magosevich D, Moreno MC, Guzman MA, D’Atri LP, Carestia A, et al. Nucleosomes and neutrophil extracellular traps in septic and burn patients. Clin Immunol. 2017;183:254–62.CrossRefPubMed
82.
Zurück zum Zitat De Meyer SF, Suidan GL, Fuchs TA, Monestier M, Wagner DD. Extracellular chromatin is an important mediator of ischemic stroke in mice. Arterioscler Thromb Vasc Biol. 2012;32:1884–91.CrossRefPubMedPubMedCentral De Meyer SF, Suidan GL, Fuchs TA, Monestier M, Wagner DD. Extracellular chromatin is an important mediator of ischemic stroke in mice. Arterioscler Thromb Vasc Biol. 2012;32:1884–91.CrossRefPubMedPubMedCentral
83.
Zurück zum Zitat Huang H, Chen H-W, Evankovich J, Yan W, Rosborough BR, Nace GW, et al. Histones activate the NLRP3 inflammasome in Kupffer cells during sterile inflammatory liver injury. J Immunol. 2013;191:2665–79.CrossRefPubMed Huang H, Chen H-W, Evankovich J, Yan W, Rosborough BR, Nace GW, et al. Histones activate the NLRP3 inflammasome in Kupffer cells during sterile inflammatory liver injury. J Immunol. 2013;191:2665–79.CrossRefPubMed
84.
Zurück zum Zitat Abrams ST, Zhang N, Manson J, Liu T, Dart C, Baluwa F, et al. Circulating histones are mediators of trauma-associated lung injury. Am J Respir Crit Care Med. 2013;187:160–9.CrossRefPubMedPubMedCentral Abrams ST, Zhang N, Manson J, Liu T, Dart C, Baluwa F, et al. Circulating histones are mediators of trauma-associated lung injury. Am J Respir Crit Care Med. 2013;187:160–9.CrossRefPubMedPubMedCentral
85.
Zurück zum Zitat Allam R, Scherbaum CR, Darisipudi MN, Mulay SR, Hagele H, Lichtnekert J, et al. Histones from dying renal cells aggravate kidney injury via TLR2 and TLR4. J Am Soc Nephrol. 2012;23:1375–88.CrossRefPubMedPubMedCentral Allam R, Scherbaum CR, Darisipudi MN, Mulay SR, Hagele H, Lichtnekert J, et al. Histones from dying renal cells aggravate kidney injury via TLR2 and TLR4. J Am Soc Nephrol. 2012;23:1375–88.CrossRefPubMedPubMedCentral
86.
Zurück zum Zitat Shin SH, Joo HW, Kim MK, Kim JC, Sung YK. Extracellular histones inhibit hair shaft elongation in cultured human hair follicles and promote regression of hair follicles in mice. Exp Dermatol. 2012;21:956–8.CrossRefPubMed Shin SH, Joo HW, Kim MK, Kim JC, Sung YK. Extracellular histones inhibit hair shaft elongation in cultured human hair follicles and promote regression of hair follicles in mice. Exp Dermatol. 2012;21:956–8.CrossRefPubMed
89.
Zurück zum Zitat Zhu H, Sun A. Programmed necrosis in heart disease: molecular mechanisms and clinical implications. J Mol Cell Cardiol. 2018;116:125–34.CrossRefPubMed Zhu H, Sun A. Programmed necrosis in heart disease: molecular mechanisms and clinical implications. J Mol Cell Cardiol. 2018;116:125–34.CrossRefPubMed
90.
Zurück zum Zitat Ibáñez B, Heusch G, Ovize M, Van De Werf F. Evolving therapies for myocardial ischemia/reperfusion injury. J Am Coll Cardiol. 2015;65:1454–71.CrossRefPubMed Ibáñez B, Heusch G, Ovize M, Van De Werf F. Evolving therapies for myocardial ischemia/reperfusion injury. J Am Coll Cardiol. 2015;65:1454–71.CrossRefPubMed
91.
Zurück zum Zitat Hall G, Hasday JD, Rogers TB. Regulating the regulator: NF-κB signaling in heart. J Mol Cell Cardiol. 2006;41:580–91.CrossRefPubMed Hall G, Hasday JD, Rogers TB. Regulating the regulator: NF-κB signaling in heart. J Mol Cell Cardiol. 2006;41:580–91.CrossRefPubMed
92.
Zurück zum Zitat Boyd JH, Mathur S, Wang Y, Bateman RM, Walley KR. Toll-like receptor stimulation in cardiomyoctes decreases contractility and initiates an NF-κB dependent inflammatory response. Cardiovasc Res. 2006;72:384–93.CrossRefPubMed Boyd JH, Mathur S, Wang Y, Bateman RM, Walley KR. Toll-like receptor stimulation in cardiomyoctes decreases contractility and initiates an NF-κB dependent inflammatory response. Cardiovasc Res. 2006;72:384–93.CrossRefPubMed
93.
Zurück zum Zitat Dick MS, Sborgi L, Rühl S, Hiller S, Broz P. ASC filament formation serves as a signal amplification mechanism for inflammasomes. Nat Commun. 2016;7:11929.CrossRefPubMedPubMedCentral Dick MS, Sborgi L, Rühl S, Hiller S, Broz P. ASC filament formation serves as a signal amplification mechanism for inflammasomes. Nat Commun. 2016;7:11929.CrossRefPubMedPubMedCentral
94.
Zurück zum Zitat Stutz A, Horvath GL, Monks BG, Latz E. ASC speck formation as a readout for inflammasome activation. Methods Mol Biol. 2013;1040:91–101.CrossRefPubMed Stutz A, Horvath GL, Monks BG, Latz E. ASC speck formation as a readout for inflammasome activation. Methods Mol Biol. 2013;1040:91–101.CrossRefPubMed
95.
Zurück zum Zitat Lu A, Magupalli VG, Ruan J, Yin Q, Atianand MK, Vos MR, et al. Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell. 2014;156:1193–206.CrossRefPubMedPubMedCentral Lu A, Magupalli VG, Ruan J, Yin Q, Atianand MK, Vos MR, et al. Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell. 2014;156:1193–206.CrossRefPubMedPubMedCentral
96.
Zurück zum Zitat Brydges SD, Mueller JL, McGeough MD, Pena CA, Misaghi A, Gandhi C, et al. Inflammasome-mediated disease animal models reveal roles for innate but not adaptive immunity. Immunity. 2009;30:875–87.CrossRefPubMedPubMedCentral Brydges SD, Mueller JL, McGeough MD, Pena CA, Misaghi A, Gandhi C, et al. Inflammasome-mediated disease animal models reveal roles for innate but not adaptive immunity. Immunity. 2009;30:875–87.CrossRefPubMedPubMedCentral
98.
Zurück zum Zitat Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature. 2016;535:111–6.CrossRefPubMed Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature. 2016;535:111–6.CrossRefPubMed
99.
Zurück zum Zitat Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526:660–5.CrossRefPubMed Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526:660–5.CrossRefPubMed
100.
Zurück zum Zitat Russo HM, Rathkey J, Boyd-Tressler A, Katsnelson MA, Abbott DW, Dubyak GR. Active caspase-1 induces plasma membrane pores that precede pyroptotic lysis and are blocked by lanthanides. J Immunol. 2016;197:1353–67.CrossRefPubMed Russo HM, Rathkey J, Boyd-Tressler A, Katsnelson MA, Abbott DW, Dubyak GR. Active caspase-1 induces plasma membrane pores that precede pyroptotic lysis and are blocked by lanthanides. J Immunol. 2016;197:1353–67.CrossRefPubMed
101.
Zurück zum Zitat Linkermann A, Stockwell BR, Krautwald S, Anders HJ. Regulated cell death and inflammation: an auto-amplification loop causes organ failure. Nat Rev Immunol. 2014;14:759–67.CrossRefPubMed Linkermann A, Stockwell BR, Krautwald S, Anders HJ. Regulated cell death and inflammation: an auto-amplification loop causes organ failure. Nat Rev Immunol. 2014;14:759–67.CrossRefPubMed
102.
Zurück zum Zitat Dinarello CA. A clinical perspective of IL-1β as the gatekeeper of inflammation. Eur J Immunol. 2011;41:1203–17.CrossRefPubMed Dinarello CA. A clinical perspective of IL-1β as the gatekeeper of inflammation. Eur J Immunol. 2011;41:1203–17.CrossRefPubMed
104.
Zurück zum Zitat Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, et al. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation. 2011;123:594–604.CrossRefPubMed Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, et al. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation. 2011;123:594–604.CrossRefPubMed
105.
Zurück zum Zitat Shao BZ, Xu ZQ, Han BZ, Su DF, Liu C. NLRP3 inflammasome and its inhibitors: a review. Front Pharmacol. 2015;6:1–9.CrossRef Shao BZ, Xu ZQ, Han BZ, Su DF, Liu C. NLRP3 inflammasome and its inhibitors: a review. Front Pharmacol. 2015;6:1–9.CrossRef
107.
Zurück zum Zitat Toldo S, Abbate A. The NLRP3 inflammasome in acute myocardial infarction. Nat Rev Cardiol. 2018;15(4):203–14.CrossRefPubMed Toldo S, Abbate A. The NLRP3 inflammasome in acute myocardial infarction. Nat Rev Cardiol. 2018;15(4):203–14.CrossRefPubMed
108.
Zurück zum Zitat Sandanger Ø, Ranheim T, Vinge LE, Bliksøen M, Alfsnes K, Finsen AV, et al. The NLRP3 inflammasome is up-regulated in cardiac fibroblasts and mediates myocardial ischaemia-reperfusion injury. Cardiovasc Res. 2013;99:164–74.CrossRefPubMed Sandanger Ø, Ranheim T, Vinge LE, Bliksøen M, Alfsnes K, Finsen AV, et al. The NLRP3 inflammasome is up-regulated in cardiac fibroblasts and mediates myocardial ischaemia-reperfusion injury. Cardiovasc Res. 2013;99:164–74.CrossRefPubMed
109.
Zurück zum Zitat Ge L, Zhou X, Ji W-J, Lu R-Y, Zhang Y, Zhang Y-D, et al. Neutrophil extracellular traps in ischemia-reperfusion injury-induced myocardial no-reflow: therapeutic potential of DNase-based reperfusion strategy. Am J Physiol Heart Circ Physiol. 2015;308:500–9.CrossRef Ge L, Zhou X, Ji W-J, Lu R-Y, Zhang Y, Zhang Y-D, et al. Neutrophil extracellular traps in ischemia-reperfusion injury-induced myocardial no-reflow: therapeutic potential of DNase-based reperfusion strategy. Am J Physiol Heart Circ Physiol. 2015;308:500–9.CrossRef
110.
Zurück zum Zitat Savchenko AS, Borissoff JI, Martinod K, De Meyer SF, Gallant M, Erpenbeck L, et al. VWF-mediated leukocyte recruitment with chromatin decondensation by PAD4 increases myocardial ischemia/reperfusion injury in mice. Blood. 2014;123(1):141–8.CrossRefPubMedPubMedCentral Savchenko AS, Borissoff JI, Martinod K, De Meyer SF, Gallant M, Erpenbeck L, et al. VWF-mediated leukocyte recruitment with chromatin decondensation by PAD4 increases myocardial ischemia/reperfusion injury in mice. Blood. 2014;123(1):141–8.CrossRefPubMedPubMedCentral
111.
Zurück zum Zitat Yang XM, Cui L, White J, Kuck J, Ruchko MV, Wilson GL, et al. Mitochondrially targeted endonuclease III has a powerful anti-infarct effect in an in vivo rat model of myocardial ischemia/reperfusion. Basic Res Cardiol. 2015;110:3.CrossRefPubMedPubMedCentral Yang XM, Cui L, White J, Kuck J, Ruchko MV, Wilson GL, et al. Mitochondrially targeted endonuclease III has a powerful anti-infarct effect in an in vivo rat model of myocardial ischemia/reperfusion. Basic Res Cardiol. 2015;110:3.CrossRefPubMedPubMedCentral
112.
Zurück zum Zitat Vinten-Johansen J, Zatta AJ, Jiang R, Shi W. Lethal myocardial reperfusion injury. Manag Myocard Reperfus Inj 2013;51–85. Vinten-Johansen J, Zatta AJ, Jiang R, Shi W. Lethal myocardial reperfusion injury. Manag Myocard Reperfus Inj 2013;51–85.
113.
Zurück zum Zitat Eltzschig HK, Collard CD. Vascular ischaemia and reperfusion injury. Br Med Bull. 2004;70:71–86.CrossRefPubMed Eltzschig HK, Collard CD. Vascular ischaemia and reperfusion injury. Br Med Bull. 2004;70:71–86.CrossRefPubMed
114.
Zurück zum Zitat Piper HM, García-Dorado D, Ovize M. A fresh look at reperfusion injury. Cardiovasc Res. 1998;38:291–300.CrossRefPubMed Piper HM, García-Dorado D, Ovize M. A fresh look at reperfusion injury. Cardiovasc Res. 1998;38:291–300.CrossRefPubMed
115.
Zurück zum Zitat Alcantara FF, Iglehart DJ, Ochs RL. Heparin in plasma samples causes nonspecific binding to histones on Western blots. J Immunol Methods. 1999;226:11–8.CrossRefPubMed Alcantara FF, Iglehart DJ, Ochs RL. Heparin in plasma samples causes nonspecific binding to histones on Western blots. J Immunol Methods. 1999;226:11–8.CrossRefPubMed
116.
Zurück zum Zitat Thourani VH, Brar SS, Kennedy TP, Thornton LR, Watts JA, Ronson RS, et al. Nonanticoagulant heparin inhibits NF-kappaB activation and attenuates myocardial reperfusion injury. Am J Physiol Heart Circ Physiol. 2000;278:2084–93.CrossRef Thourani VH, Brar SS, Kennedy TP, Thornton LR, Watts JA, Ronson RS, et al. Nonanticoagulant heparin inhibits NF-kappaB activation and attenuates myocardial reperfusion injury. Am J Physiol Heart Circ Physiol. 2000;278:2084–93.CrossRef
117.
Zurück zum Zitat Kouretas PC, Myers AK, Kim YD, Cahill PA, Myers JL, Wang N, et al. Heparin and nonanticoagulant heparin preserve regional myocardial contractility after ischemia-reperfusion injury: role of nitric oxide. J Thorac Cardiovasc Surg. 1998;115:440–9.CrossRefPubMed Kouretas PC, Myers AK, Kim YD, Cahill PA, Myers JL, Wang N, et al. Heparin and nonanticoagulant heparin preserve regional myocardial contractility after ischemia-reperfusion injury: role of nitric oxide. J Thorac Cardiovasc Surg. 1998;115:440–9.CrossRefPubMed
118.
Zurück zum Zitat Kouretas PC, Kim YD, Cahill PA, Myers AK, To LN, Wang YN, et al. Nonanticoagulant heparin prevents coronary endothelial dysfunction after brief ischemia-reperfusion injury in the dog. Circulation. 1999;99:1062–8.CrossRefPubMed Kouretas PC, Kim YD, Cahill PA, Myers AK, To LN, Wang YN, et al. Nonanticoagulant heparin prevents coronary endothelial dysfunction after brief ischemia-reperfusion injury in the dog. Circulation. 1999;99:1062–8.CrossRefPubMed
119.
Zurück zum Zitat Pevni D, Frolkis I, Shapira I, Schwartz D, Yuhas Y, Schwartz IF, et al. Heparin added to cardioplegic solution inhibits tumor necrosis factor-α production and attenuates myocardial ischemic-reperfusion injury. Chest. 2005;128:1805–11.CrossRefPubMed Pevni D, Frolkis I, Shapira I, Schwartz D, Yuhas Y, Schwartz IF, et al. Heparin added to cardioplegic solution inhibits tumor necrosis factor-α production and attenuates myocardial ischemic-reperfusion injury. Chest. 2005;128:1805–11.CrossRefPubMed
120.
Zurück zum Zitat Iba T, Hashiguchi N, Nagaoka I, Tabe Y, Kadota K, Sato K. Heparins attenuated histone-mediated cytotoxicity in vitro and improved the survival in a rat model of histone-induced organ dysfunction. Intensive Care Med Exp. 2015;3:36.CrossRefPubMedPubMedCentral Iba T, Hashiguchi N, Nagaoka I, Tabe Y, Kadota K, Sato K. Heparins attenuated histone-mediated cytotoxicity in vitro and improved the survival in a rat model of histone-induced organ dysfunction. Intensive Care Med Exp. 2015;3:36.CrossRefPubMedPubMedCentral
121.
Zurück zum Zitat Nagano F, Mizuno T, Mizumoto S, Yoshioka K, Takahashi K, Tsuboi N, et al. Chondroitin sulfate protects vascular endothelial cells from toxicities of extracellular histones. Eur J Pharmacol. 2018;826:48–55.CrossRefPubMed Nagano F, Mizuno T, Mizumoto S, Yoshioka K, Takahashi K, Tsuboi N, et al. Chondroitin sulfate protects vascular endothelial cells from toxicities of extracellular histones. Eur J Pharmacol. 2018;826:48–55.CrossRefPubMed
122.
Zurück zum Zitat Collino M, Pini A, Mastroianni R, Benetti E, Lanzi C, Bani D, et al. The non-anticoagulant heparin-like K5 polysaccharide derivative K5-N,OSepi attenuates myocardial ischaemia/reperfusion injury. J Cell Mol Med. 2012;16:2196–207.CrossRefPubMedPubMedCentral Collino M, Pini A, Mastroianni R, Benetti E, Lanzi C, Bani D, et al. The non-anticoagulant heparin-like K5 polysaccharide derivative K5-N,OSepi attenuates myocardial ischaemia/reperfusion injury. J Cell Mol Med. 2012;16:2196–207.CrossRefPubMedPubMedCentral
Metadaten
Titel
The Role of Extracellular DNA and Histones in Ischaemia-Reperfusion Injury of the Myocardium
verfasst von
Mohammed Shah
Derek M. Yellon
Sean M. Davidson
Publikationsdatum
15.02.2020
Verlag
Springer US
Erschienen in
Cardiovascular Drugs and Therapy / Ausgabe 1/2020
Print ISSN: 0920-3206
Elektronische ISSN: 1573-7241
DOI
https://doi.org/10.1007/s10557-020-06946-6

Weitere Artikel der Ausgabe 1/2020

Cardiovascular Drugs and Therapy 1/2020 Zur Ausgabe

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Strenge Blutdruckeinstellung lohnt auch im Alter noch

30.04.2024 Arterielle Hypertonie Nachrichten

Ältere Frauen, die von chronischen Erkrankungen weitgehend verschont sind, haben offenbar die besten Chancen, ihren 90. Geburtstag zu erleben, wenn ihr systolischer Blutdruck < 130 mmHg liegt. Das scheint selbst für 80-Jährige noch zu gelten.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Dihydropyridin-Kalziumantagonisten können auf die Nieren gehen

30.04.2024 Hypertonie Nachrichten

Im Vergleich zu anderen Blutdrucksenkern sind Kalziumantagonisten vom Diyhdropyridin-Typ mit einem erhöhten Risiko für eine Mikroalbuminurie und in Abwesenheit eines RAS-Blockers auch für ein terminales Nierenversagen verbunden.

Update Kardiologie

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