Next Article in Journal
PCR Detection of Toxoplasma gondii in European Wild Rabbit (Oryctolagus cuniculus) from Portugal
Next Article in Special Issue
Glucose-Binding of Periplasmic Protein GltB Activates GtrS-GltR Two-Component System in Pseudomonas aeruginosa
Previous Article in Journal
The Influence of pCO2-Driven Ocean Acidification on Open Ocean Bacterial Communities during A Short-Term Microcosm Experiment in the Eastern Tropical South Pacific (ETSP) off Northern Chile
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

“NETs and EETs, a Whole Web of Mess”

1
Department of Microbiology and Immunology, Louisiana State University (LSU), Health Science Center, Shreveport, LA 71103, USA
2
Department of Infectious Diseases, University of Georgia, Athens, GA 30302, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2020, 8(12), 1925; https://doi.org/10.3390/microorganisms8121925
Submission received: 15 October 2020 / Revised: 26 November 2020 / Accepted: 30 November 2020 / Published: 4 December 2020

Abstract

:
Neutrophils and eosinophils are granulocytes that have very distinct functions. Neutrophils are first responders to external threats, and they use different mechanisms to control pathogens. Phagocytosis, reactive oxygen species, and neutrophil extracellular traps (NETs) are some of the mechanisms that neutrophils utilize to fight pathogens. Although there is some controversy as to whether NETs are in fact beneficial or detrimental to the host, it mainly depends on the biological context. NETs can contribute to disease pathogenesis in certain types of diseases, while they are also undeniably critical components of the innate immune response. On the contrary, the role of eosinophils during host immune responses remains to be better elucidated. Eosinophils play an important role during helminthic infections and allergic responses. Eosinophils can function as effector cells in viral respiratory infections, gut bacterial infections, and as modulators of immune responses by driving the balance between Th1 and Th2 responses. In particular, eosinophils have biological activities that appear to be quite similar to those of neutrophils. Both possess bactericidal activity, can activate proinflammatory responses, can modulate adaptive immune responses, can form extracellular traps, and can be beneficial or detrimental to the host according to the underlying pathology. In this review we compare these two cell types with a focus on highlighting their numerous similarities related to extracellular traps.

1. Introduction

The immune system is a complex myriad of signals that orchestrates the timing, length, and quantity of the immune response [1]. A classic immune response consists of very well-coordinated steps and begins with innate immunity providing the first level of protection. The innate immune response includes phagocytic cells and physical barriers at mucosal sites [2,3,4,5,6]. After several steps, the innate response is followed by adaptive immunity that will be responsible for the ultimate, long-term clearance of the infection. The successful generation of protective immunity is dictated by a finely-tuned slew of immune signals beginning with those produced by innate cells [3,6,7,8,9]. Nevertheless, while host immunity fights to orchestrate the battle against the external aggression, microbes utilize means to disrupt this precise and well-adjusted communication. The manipulation and disruption of the immune homeostasis can lead to overreactive proinflammatory responses that can cause tissue damage, disease, and even death of the patient, as is currently happening during the COVID-19 pandemic.
Polymorphonuclear granulocytes are white blood cells characterized by harboring numerous cytoplasmic granules and segmented nuclei. Granulocytes are a very important part of the first line of the immune response against infections or external threats [10]. Beyond their classical role, their outstanding and recently-recognized versatility allows them to work as antigen presenting cells [10,11], modulators of adaptive responses [12,13,14,15], and overall shapers of the immune memory [10,16]. There are three major classes of granulocytes—neutrophils, eosinophils, and basophils, although mast cells can also be included. Neutrophils represent the most numerous and best-studied class [10].
In this review, we focus on a special function of these cells, which is the formation of extracellular traps (ETs). We first highlight neutrophils as we talk about their role as immune cells, the traps they form, and the functionality of these traps. This will be followed by a comparison between neutrophils and eosinophils as well as the traps they both form, and we outline similarities and differences between them. We will also dedicate a small section to exploring how pathogens can escape or evade neutrophils and their traps, as well as the recent discovery of bacterial modulation of eosinophils [17]. We give examples of some of the pathogens executing these defense mechanisms as well as some of the diseases that involve them (Figure 1). We conclude by discussing future directions focusing on the application of current knowledge for the development of therapies as well as guiding future research directions regarding these two cell types.

2. Neutrophils and Neutrophil Extracellular Traps

Neutrophils are a very abundant cell type in humans (55–70% of all peripheral white blood cells). They respond very quickly to infectious agents by trapping the microbes and releasing antimicrobial molecules with the purpose of killing them [18,19,20,21,22]. They are formed and differentiated in the bone marrow before being released into the circulatory system. Neutrophils contain many granules that are classified into three main groups, (1) azurophilic granules, that contain myeloperoxidase (MPO), bactericidal/permeability-increasing protein (BPI), defensins, neutrophil elastase (NE), and cathepsin G; (2) specific granules that encompass alkaline phosphatase, lysozyme, nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase), collagenase, lactoferrin, histamine, and cathelicidin; and, (3) tertiary granules that have cathepsin, gelatinase, and collagenase. There is a fourth type of granule called a secretory vesicle. These are not real granules, but vesicles derived from the plasma membrane. These vesicles are important in early events of polymorphonuclear activation and largely responsible for the many versatile functions of neutrophils [23].
Although neutrophils are short-lived, they are very motile and can travel through tight junctions between cells allowing them to reach a multitude of different body compartments. An interesting aspect of these cells is their unique ability to form neutrophil extracellular traps (NETs) [24,25,26,27] allowing them to catch pathogens in the extracellular space in order to control the spread of the infection. NETs can trap and kill microbes which significantly decreases the microbial burden [28,29,30] aiding the immune system in the clearance of the infection and in the individual’s overall survival.

2.1. Neutrophils in Infection

Neutrophil-mediated microbial clearance is required in infections caused not only by extracellular [31,32] but also intracellular pathogens such as Listeria monocytogenes [33]. Neutrophils’ classical role during the early innate immune response involves bacterial phagocytosis and killing facilitated by the generation of reactive oxygen species (ROS), NET formation, and production of proinflammatory cytokines [19,34,35]. However, current research has demonstrated that neutrophils can also acquire and perform other important immune functions including antigen presentation [11] and modulation of adaptive immunity [12].
Neutrophils are essential during the signaling cascade required to activate an efficient inflammatory response. They produce a plethora of chemokines and cytokines [36,37] highlighting the diversity of neutrophils in the modulation and overall conservancy of the immune homeostasis. While their job as first line of defense against pathogens is unquestionable, the formation of NETs remains a controversial function of neutrophils due to their critical role during bacterial clearance, while simultaneously associated with deleterious auto-inflammatory and autoimmune diseases.

2.2. NETs

NETs are made of extracellular fibers whose backbone consists of DNA [38,39]. NETs are complex structures formed not only by chromosomal and mitochondrial DNA, but also DNA-associated histones and granule proteins including neutrophil elastase (NE), cathepsin G, and MPO [25,40]. NETs can bind pathogens that adhere to the DNA through the bacterial lipopolysaccharides, for instance [40,41]. This mechanism of defense is inducible by Gram-positive and Gram-negative pathogens [28,29,30,42,43], such as Staphylococcus aureus [44,45,46], Escherichia coli [47,48,49], or Pseudomonas aeruginosa [41,50,51,52,53,54].
The NET formation process has been referred to as NETosis [55]. It was thought that neutrophils have to die to form NETs and the actual cell death is called NETosis. However, not all neutrophils have to die in order to release the mitochondrial DNA. Yousefi et al. [56] demonstrated that pure mitochondrial DNA can form NETs while neutrophils remain viable. This means that neutrophils have a cytolytic and non-cytolytic mechanism for NETosis [56,57]. However, the molecular mechanism by which NETosis is induced and mediated is still not fully elucidated, albeit many pathways have already been implicated. NET formation was first described to require the release of reactive oxygen species (ROS) produced by the NADPH oxidase enzyme complex [28,55]. The activation of the NADPH oxidase is one of the main effector responses of neutrophils to external pathogens and it is also critical for NET formation due to its ability to activate intracellular granular proteases [58,59,60]. The localization (intra- vs. extracellular) of ROS production in neutrophils has been proposed to drive effector responses of neutrophils including NET extrusion to microbes of different sizes [61]. While several stimuli trigger NETs in an NADPH oxidase- and ROS-dependent fashion, numerous reports have emerged proposing an NADPH oxidase-independent mechanism of NET formation in response to specific stimuli including microcrystals [27,62,63,64].
Protein arginine deiminase 4 (PAD4), an enzyme performing protein citrullination as a post-translational modification, has been shown to be critical for NET formation [65,66]. PAD4 is highly expressed in neutrophils and localizes to the cytosol in resting cells [67,68]. Upon neutrophil stimulation, PAD4 translocates to the nucleus to promote histone citrullination that mediates chromatin disassembly [65,69,70]. This is followed by the disruption of the cytoskeleton, endomembranes, and the nuclear envelope [71]. Different protein kinase C isoforms have been implicated in the mediation of PAD4- and NADPH oxidase-mediated NET formation [72]. But regardless of the path utilized to initiate NET formation, once the process is activated, NE, MPO, and other neutrophil granule proteins are released and decorate the DNA web [73].
NETosis was proposed to occur by two different pathways: suicidal NETosis and vital NETosis [26,40,55]. Suicidal NETosis refers to NET formation due to the release of DNA following the death of neutrophils [74]. Vital NETosis is activated by pathogens, bacterial lipopolysaccharide, TLR4-activated platelets, or complement proteins. It is a rapid process that does not result in immediate neutrophil death, however, the cell loses its DNA. More intriguingly, after vital NETosis neutrophils are still capable of phagocytosing bacteria [75] suggesting that albeit not containing nucleic acid they are still able to perform biological functions for some time. This is in line with classical observations of human neutrophil ‘ghosts’ or ‘cytoplasts’ that are isolated neutrophils whose nuclei were removed experimentally but still remained capable of migration, phagocytosis, and ROS production [76,77].

2.3. NETs and Infectious Diseases

NETs are highly conserved amongst different species in the animal kingdom, which features its cruciality. As with any other immune function, the ability to form NETs appears to be affected by age. Previous reports demonstrate that neonatal neutrophils are less capable of forming NETs than adult neutrophils indicating that the immune system needs to acquire a certain degree of maturity in order to establish proper NETs [78].
As previously mentioned, there is an extensive number of pathogens that trigger NET formation by diverse mechanisms including lipopolysaccharide and flagella [79,80]. NETs can have bacteriostatic and/or bactericidal effects [28]. Within NETs, bacteria cannot utilize the bacterial virulence factors to downregulate host immune response allowing for the immune system to better control the infection [28]. This undeniably powerful mechanism provides protection against bacterial infections [28,29,51,81,82], mycobacteria [40], fungi [83], viruses [84,85], and parasites [86,87,88] offering a basic universal method to combat microbes. Thus, this highlights the very significant role of NETs during clearance of multiple kinds of infections.
There is, nonetheless, debate in regard to the role of NETs during several chronic diseases such as cystic fibrosis (CF). CF is a lung disease where there is a build-up of chronic inflammation and thick mucus in the respiratory airways as well as robust bacterial colonization. Many different kinds of bacteria are found in CF patients including Haemophilus influenzae, Staphylococcus aureus, Pseudomonas aeruginosa, and Bordetella spp. [89]. As neutrophils release DNA, NETs are formed, and they promote the patients’ mucus to become thicker resulting in the build-up of a niche with a high amount of glycans that promotes a great bacterial diversity. NETs increase sputum viscosity causing the patients’ breathing capacity to decrease, negatively impacting their health status. Many CF patients are subjected to DNase treatment to disrupt the NETs so that sputum can be liquified and mucus can be cleared [90].
Neutrophils and their NETs play a very important role in the defense against fungal infections as well. Fungal infections are mostly controlled by neutrophils and they eradicate fungi by oxidative burst, phagocytosis, or releasing NETs. Candida albicans is a fungus that can cause infections in the gastrointestinal tract, mouth, throat, and the skin. While NETs bind to this fungal pathogen, neutrophils also release the protein calprotectin, which is a metal ion chelator. Calprotectin performs its antifungal activity by depleting Zn2+ and Mn2+. These two ions are very significant for Candida albicans to thrive, and NETs allow for calprotectin to better reach all fungi leading, then, to the control of the infection and its dissemination [83].
NET induction in viral infections has not been well-studied but scientists are now learning more and more about how NETs help protect the host during these infections. Neutrophils are able to recognize different viral pathogens and trigger NET formation via Toll-like receptors (TLR) like HIV-1, which promotes NETs via TLR7 and TLR 8 [84]. However, viruses can also promote NETosis indirectly without using pattern recognition receptors (PRRs). Chemokines and cytokines like interleukin-8 (IL-8) and type 1 interferons are present in virus-infected cells triggering NETosis in recruited neutrophils. These NETs help clear viral infections by binding to the viral particles and immobilizing them by electrostatic attraction, causing the virus to mechanically stop spreading [91]. But it is worth noting that bacterial and viral infections manipulate the host system in a very conservative way. This indicates that a better understanding of the precise mechanisms that pathogens utilize to modulate NETs can result in targets for therapeutics that can be applied to a broad spectrum of diseases.

NETs and COVID-19

In December 2019, a cluster of pneumonia of unknown etiology was identified in Wuhan, China. Rapidly, this novel virus known as SARS-CoV-2 emerged and it has spread worldwide causing a great pandemic during 2020. Since this pandemic started, many efforts have tried to characterize the pathology and immune responses to this novel coronavirus and a particular feature is the hyperreactive uncontrolled immune response that leads to the detriment of the patient’s health [92,93,94].
Increased neutrophil counts in the peripheral blood have been associated with a worsening of the patient’s prognosis. In fact, the neutrophil-to-lymphocyte ratio (NLR) has been used as a marker of severe disease [95,96,97]. In COVID-19 patients, reactive oxygen species (ROS) produced by neutrophils are augmented which can also contribute to organ failure [98].
Neutrophil degranulation and NET markers have been associated with severe illness during COVID-19 [99]. In fact, higher levels of NETs have been correlated with an increased death rate in COVID-19 patients, in which an overreactive neutrophilic response leads to a general multi-organ failure [97]. Neutrophils appear to be decorated with thrombocytes in severe cases of COVID-19, the number of NETs identified in the circulation is significantly increased in these patients [100,101,102] and this correlates with the evidence of increased platelet factor 4 detected in these patients [101]. This increased NET formation has also been associated with COVID-19-related acute respiratory distress syndrome and is a potential biomarker for disease severity [101]. SARS-CoV-2 has been shown to trigger NETs directly in neutrophils [103]. Because of all of this, neutrophils have emerged as potential, clinically relevant therapeutic targets in COVID-19 [104].

2.4. NETs and Inflammatory Diseases

While the benefit of NETs is to clear pathogens, the unchecked deposition of NETs has been associated with deleterious effects to health [105]. NETs have been related to several acute diseases such as acute respiratory distress syndrome [105], hepatic damage during bloodstream infection [106,107], as well as chronic diseases including cystic fibrosis lung disease [26,50,51,52] and chronic obstructive pulmonary disease (COPD) [108,109,110,111]. In CF patients, NETs associate, as previously mentioned, with an obstruction of the lung and decrease of lung function [80,112,113]. However, NETs are also involved in autoimmune diseases, cancer, and even fertility [114]. And excitingly, NETs and EETs, as discussed later, have also been associated with gout and its pathogenesis induced by the formation of monosodium urate microcrystals [27,62,115].
NETs play a very significant role in many diseases and health conditions including asthma and allergies. Unfortunately, NETs have a negative impact in asthmatic patients. In some cases when there is a disordered regulation in the formation of NETs, it can actually promote exacerbation of severe asthma and COPD112.
Furthermore, it has been proven that NETs have been found in the sputum of severe asthmatic patients. A previous study was completed to compare the sputum of severe asthmatic patients to healthy control subjects by measuring extracellular DNA (eDNA). This study resulted in the knowledge that a higher eDNA concentration was found in severe asthmatic patients and that NETs mediate inflammasome activation as well as cell injury. High eDNA in the sputum of severe asthmatic patients correlated with their levels of NETs that were also cytotoxic to airway epithelial cells [116].
NETs have also been shown to have a role in thrombosis. Thrombosis is the pathological deviation of hemostasis which involves the mechanisms that stops bleeding after blood vessels are injured [117]. In other words, thrombosis is the creation of a blood clot in a blood vessel which stops leakage of blood into surrounding tissues but can also result in the restriction of blood flow. It has been shown that thrombosis depends on platelet aggregation and adhesion, and NETs that are perfused within the blood can accelerate platelet aggregation [118,119]. Deep vein thrombosis is a serious issue because it develops pulmonary embolisms, referred to as venous thromboembolism [120], and there are occlusions of the pulmonary artery due to the thrombus breaking apart and traveling to the lungs [117]. Studies regarding deep vein thrombosis have shown that NETs stimulate thrombus formation through the entrapment of red blood cells which also promotes fibrin deposition [118]. The results of these studies also indicated that NETs in sepsis have the same function [118]. Other studies have illustrated the role of NETs in acute myocardial infarction, revealing that NETs at the site of plaque rupture were enveloped by tissue factor which is a significant in vivo initiator of coagulation [121]. Overall, NETs contribute to several pathologies via their prothrombotic effect.

2.5. Bacterial Mechanisms to Escape NETs

As NETs remained well-conserved across evolution, and due to the great selective pressure that this possesses on infectious agents, bacteria have evolved mechanisms to escape or manipulate NET mediated clearance [122,123,124,125,126,127,128]. Since this topic has been extensively covered in several reviews [50,84,128,129,130], only a few highlights are mentioned here. Bordetella pertussis harbors many immunomodulators that control several aspects of the host immune response [1,131,132] including NETs. B. pertussis suppresses the generation of ROS inhibiting NET formation. Also, adenylate cyclase toxin (ACT) of Bordetella spp. contributes to the suppression of ROS [126]. Similarly, Pseudomonas aeruginosa possesses means to trigger NET formation. Swimming motility mediated by the bacterial flagellum has been shown to be an essential factor in triggering NETs in neutrophils [80]. Interestingly, it has been demonstrated that NET formation is also linked to quorum sensing (QS). Quorum sensing is the mechanism by which bacteria communicate with each other via diffusible molecules [133,134,135]. LasR, a regulator of QS that controls expression of many bacterial virulence factors, provides an exciting target to explore the relationship between NETs and QS. Previous results demonstrated that LasR-deficient mutants presented a defect in NET formation. This implies that there is a two-way signaling mechanism involved in the formation and maturation of NETs [136]. On one side, the bacteria might be sensing the host inflammatory response and responding by increasing the expression of factors that allow the blockage of host immunity. In parallel, the host immunity is able to detect cues of the bacterial threat and counteract accordingly by orchestrating signals that will allow for the required response that leads to clearance. A better understanding of this universal language will enable us to design immunomodulators that can be used not only for treatments but also vaccine design.
These are only two examples that highlight the subtleties in the regulation of NETs in immune responses. The fact that SARS-CoV-2 is also manipulating NETs suggests that disturbing this mechanism of defense provides the pathogen with a very powerful tool to cause infection.

3. Eosinophils and Eosinophil Extracellular Traps

3.1. Eosinophil Biology

Eosinophils are terminally differentiated granulocytes that develop in the bone marrow from granulocytic/myeloid progenitors in mice and common myeloid progenitors in humans [137]. They become eosinophil progenitor cells (EoP) and differentiate into mature eosinophils prior to exiting the bone marrow [138]. Eosinophils rely on cytokines like IL-3, IL-5, and Granulocyte-macrophage colony-stimulating factor (GM-CSF) for hematopoiesis, survival, and enhanced activation by other stimuli. Homeostatic migration of eosinophils occurs through eotaxin-1 (CCL11), while eotaxins -2 (CCL24) and -3 (CCL26: human only) are the primary mediators of their migration into tissues in disease states. They are found in low numbers in the circulation (1–3% of all white blood cells) and can be resident cells in tissues such as the gastrointestinal tract, lung, thymus, adipose tissue, and endometrial tissue, as well as in secondary lymphoid tissues [139,140,141].
Eosinophils contain eosinophil-specific (secondary) granules [129] that are released by several mechanisms into the extracellular milieu. Within these secondary granules are proteins that are conserved across species like major basic protein (MBP) and eosinophil peroxidase (EPX). There are also granule matrix proteins like eosinophil cationic protein (ECP) and eosinophil-derived neurotoxin (EDN) that are divergent in mice forming a family of murine ribonucleases (mEARs) [142]. Importantly, eosinophil granules also contain immune signals such as chemokines, growth factors, and cytokines [143,144,145] within their granules that can be actively and specifically released without requiring previous RNA transcription, leading to a very rapid response. Granules and granule contents are released via several mechanisms in response to a variety of stimuli. This includes, for example, specific release of mediators through piecemeal degranulation or release of entire granules resulting in cell-free granules that deposit in tissue, or through lysis of the cell.
The classic role of the eosinophil is described as that of a cell that kills and damages other cells, including pathogens. For example, eosinophils contain antimicrobial peptides (AMPs) and generate reactive ROS that kill pathogens. Although eosinophils are known by these innate features that lead to tissue damage or cell death, eosinophils are increasingly being defined as cells with the potential to modulate immune responses as well as remodel and repair tissue as described by Lee and colleagues [146].
For example, eosinophils have been identified as cells that respond to innate cytokines such as IL-33, an IL-1 family cytokine [147], to induce type 2 cytokine production, macrophage polarization, and dendritic cell activation [148,149]. Moreover, eosinophils may modulate the polarization and activation of T cells through direct and indirect mechanisms. Tissue remodeling and repair by eosinophils has been shown to be somewhat dependent on the extracellular matrix components that, in turn, may activate or suppress eosinophils [150]. Finally, eosinophils have the capacity to resolve inflammation, whether directly, by the release of resolvins [151], or through activation of phagocytic macrophages [152]. The roles of eosinophils in infectious responses as well as non-allergic diseases have enhanced our understanding of these cells as complex participants in human health [137,139,153,154].

3.2. EETs and EETosis

The discovery of eosinophil extracellular traps (EETs) is a novel exciting area of investigation that promises to unravel important functions for these cells. EETs, similar to NETs, have many advantages when it comes to infectious diseases, although this comes with a price [73].
Eosinophil traps have been shown to occur in humans as well as other animal species [155,156]. The original hypothesis of how these eosinophil extracellular traps are formed was demonstrated by Yousefi and colleagues proposing that similarly to neutrophils, eosinophils can eject their mitochondrial DNA and granule content while still remaining viable cells [157]. A second mechanism was identified by Ukei and colleagues whereby lytic cells released nuclear DNA that contained histones and cell-free secondary granules. When the cells are presented with immobilized immunoglobulins (IgA and IgG), calcium ionophore, platelet activating factor, or phorbol myristate acetate, cellular death and degranulation are promoted [141,142] and EETosis takes place [143]. It is possible, as well, that NADPH oxidase-dependent mechanisms, similar to NETosis [158], are needed for EETosis. Recent reports suggest several mechanisms may give rise to either lytic or viable DNA release from eosinophils, all of which require further study. However, there is evidence that extracellular DNA trap formation for viable and lytic EETosis happens in vivo [148].
Monosodium urate crystals (MSU) also induce trap formation in neutrophils and eosinophils [159,160]. MSUs have been associated with the enhancement of the autoinflammatory disease gout [161], partially due to their ability to trigger NETs and EETs. Interestingly, this poses the question as to whether there is a feedback regulatory mechanism between both cell types and their traps.

3.3. Eosinophils and Their Traps in Infectious Diseases

Eosinophils have been shown to lead to the clearance of specific parasitic infections [162], bacterial infections, and viral infections [163,164,165,166,167,168]. Even though the role of eosinophils during infection has only started to be delineated, some evidence of their important involvement in immunity to bacterial infections has been noted mostly in two models of infection. During Staphylococcus aureus septicemia, the classical proinflammatory response involves production of type 1 and type 17 T helper cells [102]. Additionally, preliminary data have revealed that the cytokine secretion by group 2 innate lymphoid cells (ILC2) combined with eosinophil function can skew the response towards a type 2 response, and this appears to be a better defense mechanism against S. aureus infection [169]. Septicemia survivors present higher numbers of Th2 cells [169] indicating that eosinophils mediate the generation of efficient immunity during S. aureus systemic infections—by a still unknown mechanism.
An alternative model to study the role of eosinophils in inflammatory responses has also been revealed during Helicobacter pylori infection. H. pylori is a Gram-negative bacterium that is generally found in the digestive tract. During H. pylori infection, eosinophils promote immune responses by suppressing Th1 immune responses allowing for a robust Th2 response that aids clearance [170].
An important role of eosinophils and EETs is highlighted during fungal infections. Fungi are not only pathogens, but they can also be major allergens [171] triggering allergic responses classically associated with eosinophils. ABPA (allergic bronchopulmonary aspergillosis) is involved in an allergic response to Aspergillus fumigatus, a fungus that is commonly found in patients with cystic fibrosis and asthma [172]. ABPA can cause such a severe eosinophil-dependent asthmatic response that can lead to a significant decrease in lung function [171]. Following direct interaction with A. fumigatus, eosinophils go through EETosis contributing to the thickening of the mucus and eventually causing mucus plugging [173]. Mucus plugging is associated with recurrent relapse, and it is thought to be the cause of severe inflammation in the airways [173]. Furthermore, the induction of the EETs by this particular fungus culminates in an ROS-independent process [174]. Altogether, this indicates that eosinophils likely play an important role in the immune responses to A. fumigatus and their dysregulation may lead to increased morbidity associated with the primary infection [92].
Paracoccidioidomycosis (PCM) is caused by the fungus known as Paracoccidioides brasiliensis and eosinophils are also involved in the pathology of PCM disease [175,176,177]. Patients suffering from PCM have declining T cell immunity and an increasing number of blood eosinophils, which, combined, induces high levels of inflammatory response in the very first phase of the infection [178]. Moreover, animal experiments have revealed that increased levels of IL-5 result in the blocking of eosinophil maturation increasing the susceptibility to PCM. The association between eosinophils and disease severity implies that immunomodulators of eosinophils might be a good therapeutical candidate for these fungal diseases.
An example also related to S. aureus is eosinophilic chronic rhinosinusitis (CRS). Eosinophilic CRS is a refractory disease characterized by the presence of thick mucus, nasal polyps, and congestion. Eosinophil infiltration in these patients is often associated with secondary infections that triggers proinflammatory responses [179]. EETs are formed at the sites of airway damage and they protect CRS patients from external aggressions like infections by entrapping bacteria such as S. aureus [180].
Eosinophils also play a role in many viral infections [181,182,183]. A study was completed looking at how eosinophils respond to influenza A virus infection [184]. This study revealed that eosinophils could act directly against influenza A, but they also mediated CD8+ T cell responses against the virus [182]. Similarly, respiratory syncytial virus (RSV) also induces EET formation in vitro in a concentration-dependent manner [181]. Interestingly, RSV is frequently related to asthma [185] exacerbations, further implicating a role for eosinophils in these infections. RSV induces EETs in the bronchoalveolar lavage fluid in mice with asthma which is suggested to promote airway obstruction, mucus plugs, and inflammation in respiratory airways [181].
Altogether, this indicates that eosinophils likely play significant roles during infections, however, the recent attention focused on these cells will soon enlighten more of the interesting biology of eosinophils and EETs and their roles in the immune responses to infectious diseases. The function of EETs in pathogen clearance and the precise mechanisms that trigger EET formation and modulation still remain not fully understood.

Eosinophils and COVID-19 Infection

Persistent eosinopenia has been directly correlated with severe COVID-19 with a positive predictive value of over 72% [186,187,188,189,190,191,192,193,194,195,196], and eosinophil counts on peripheral blood have been proposed as biomarkers [187,197]. Eosinophil counts appear to be very low, with a minimum at day 4 [197] when patients arrive to the hospital and the numbers slowly increase in those patients that recover, while continuing at minimum levels for those with severe manifestations of the disease [196]. COVID-19 patients have low blood eosinophils, lung biopsies are characterized by the lack of eosinophils and massive macrophage infiltration. However, eosinopenia has also been thought of as a sign or consequence of host exhaustion from clearing the COVID-19 virus [198]. Dr. Iwasaki and colleagues characterized the immune responses to COVID-19 in a cohort of patients with different stages of severity where correlations between elevated type 2 responses in severe COVID-19 infections appear [199]. Patients with severe disease showed increases in eosinophils and levels of eotaxin-2, IL-5, and IL-13 compared to those with moderate disease. The apparent association with eosinophils and other signatures that are generally associated with Th2 responses, was unexpected [182].
Two eosinophilic proteins, eosinophilic cationic protein and eosinophil-derived neurotoxin (EDN), neutralize SARS-CoV-2 [200] and strategize to avoid clearance and increase persistence while keeping low levels of eosinophils [201]. In fact, elevated levels of EDN in serum have been observed in children with persistent wheeze-associated illness. And interestingly, IL-5 and IgE have also appeared to be augmented in the worse cases of COVID-19 [199], suggesting that eosinophil signatures could be considered as indicators of patient prognosis.
These, so far, are preliminary studies, and more investigations need to be done to evaluate the role of eosinophils during COVID-19 infections. Is the virus manipulating host response and promoting eosinopenia, are eosinophils forming traps, what are the consequences of this decrease in the numbers of eosinophils during early stages of infection? These and many other questions are still to be answer.

3.4. Bacterial Mechanisms to Escape EETs

The fact that EETs are a host mechanism to fight a variety of infectious diseases suggests that it is most likely that pathogens have evolved means to evade, manipulate, or escape EETs. The compelling evidence that EETs are involved in infectious pathologies such as septicemia or fungal and viral respiratory diseases leads to the question of the implication of these traps in other bacterial infections. A recent study reveals how well-adapted pathogens can efficiently block eosinophilic influx in the lungs, causing a defect in adaptive responses associated with an increased pathogen persistence [17]. The Bordetella bronchiseptica sigma factor, btrS, regulates many immunomodulators associated with suppressing different aspects of the immune response [202]. B. bronchiseptica btrS-null trigger a prompt and robust adaptive response [131]. Some insight into the possible mechanisms associated with this increase in adaptive responses points two different cells, macrophages and eosinophils. Transcriptomic data of macrophages challenged with this mutant indicate some Th2-related signatures [131]. Further studies in eosinophil-deficient mice, revealed that while the wildtype mice clear the infection in 14 days, mice lacking eosinophils present a long-term infections characteristic of the wildtype bacteria [17]. These results suggest that wildtype bacteria somehow block eosinophil influx in the lungs to prevent the generation of prompt and robust adaptive immunity and to promote long term bacterial persistence.
These findings open a novel perspective and understanding of bacterial ability to manipulate host immune responses using very sophisticated mechanisms. This suggests that we might need to reconsider the classical view that we have of some immune cells.

3.5. EETs and Inflammatory Diseases

While inflammatory diseases caused by NETs are very diverse, eosinophils and EETs have been mostly associated with allergic [203] and non-allergic respiratory diseases. EETs can negatively affect patients’ health due to the increased production of mucus secretions [155]. Unfortunately, dysregulations of eosinophilic functions can lead to pathological disorders, such as atopic dermatitis, rhinitis, asthma, and other inflammation-associated medical conditions [137].
Allergic asthma [178] is generally a very complex disease characterized by reversible bronchial hyperresponsiveness often associated with airway eosinophilia. In particular, severe eosinophilic asthma, which is often refractory to inhaled corticosteroids, has been found to present neutrophil and eosinophil traps that may play a role in disease pathologies as well as serve as biomarkers [204,205]. EETs contribute to asthmatic patients having respiratory problems likely due to the EETs promoting mucus plugging in the airways and epithelial damage [206]. Overall, these activities of eosinophils may contribute to asthmatic exacerbations [207]. One mechanism of EET activity in asthma may be through the functions of granule proteins and granules associated with the DNA NETs, resulting in tissue damage and cell-free granule signaling [205,208]. To further support these findings, when studying the role of EETs in the murine model, EETs injected into naïve wild-type mice induced epithelial activation that promoted group 2 innate lymphoid cell activities thus increasing type 2 cytokines IL-5 and IL-13 in the airways and lungs [206]. Overall, this study showed that EETs could potentially modulate type 2 immune responses in severe asthmatic patients with high airway and blood eosinophilia.
A further study indicated that indeed extracellular granules are found in tissues of asthmatic patients and this implies that eosinophils release cytotoxic cellular content that causes damage to the tissues. There are certain signaling pathways involved in triggering cytolysis, the bursting of a cell due to an osmotic imbalance, and the release of these free granules of eosinophils. Many of the granules are activated by interleukin-3 (IL-3). This study looked at IL-3-primed eosinophils seated on immunoglobulins like IgG. When eosinophils were exposed to IL-3, they degranulated early in the presence of IgG and the cytolysis of the IL-3-primed eosinophils was dependent on ROS production and involved eosinophil migration and adhesion [209].
Non-respiratory diseases including thrombosis and atherosclerotic plaque formation have also been associated with EETs [210,211]. Previous data have revealed that EETs could be used as targets for atherosclerosis treatment, for example using Siglec 8/F antibodies to block EET formation [210], providing a novel approach for a disease that is highly prevalent. In fact, in mice, SiglecF antibodies can reduce thrombus stability preventing arterial thrombosis [210]. In this study, the data suggested that eosinophil EETosis and MBP release promote platelet activation, thrombus formation, and production plaques in blood vessels. As EETs have been associated with thrombosis formation in atherosclerotic plaques it is not without reason to suspect EETs may be present in other diseases with thrombus formation, such as chronic urticaria [212] and other skin diseases [213]. Altogether these data suggest a novel mechanism of eosinophil-mediated damage to vasculature and tissue.

4. Future Directions

The role of eosinophils during immune responses against pathogens has been under-studied, nevertheless, when looking carefully, neutrophils and eosinophils share many functions. In this review we only explored one of the many aspects of this cell type, furthering the demonstration that EETs (of eosinophils) are similar to NETs (of neutrophils).
Interestingly, NETs and EETs possess many common mechanisms and functions. It could even be possible that NETs trigger the formation of EETs by inducing signals such as MSU crystals indicating that these two processes might complement each other and be interconnected. But the question remains as to whether, if the microbes are able to manipulate NET formation, are they also able to manipulate EET formation? How do pathogens disrupt immune signals orchestrating NETs and EETs and promoting inflammation in autoinflammatory disorders? If pathogens disrupt immune signals causing overreactive proinflammatory responses, can we manipulate them artificially as a treatment for some of the aforementioned diseases? Also, does viable and lytic EETosis and NETosis happen at the same time in vivo and in the same diseases?
Both EETs and NETs have advantageous and disadvantageous effects for the host. The great opportunity comes with the gain in knowledge of EETs, as it can provide not only a better understanding of the biology of eosinophils and their role during inflammatory responses, but it can generate targets for therapeutic agents of many infectious and eosinophilic diseases.

Author Contributions

Conceptualization, M.C.G.; brainstorming, M.C.G., T.L.W. and B.R., writing—original draft, T.L.W., E.T., and M.C.G.; writing—review and editing, T.L.W., B.R., and M.C.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the start-up funds provided by LSU Health, Shreveport.

Acknowledgments

The content is solely the responsibility of the authors. We would like to acknowledge the SMART program at Louisiana State University as one of the authors, Eshaan Tandon is part of this program. We also would like to acknowledge the help and guidance of Elizabeth A. Jacobsen, who has been providing great support during the preparation of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gestal, M.; Jonhson, H.; Harvill, E. Immunomodulation as a Novel Strategy for Prevention and Treatment of Bordetella spp. Infections. Front. Front. Immunol. 2019, 10, 2869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Fitzgerald, K.A.; Kagan, J.C. Toll-like Receptors and the Control of Immunity. Cell 2020, 180, 1044–1066. [Google Scholar] [CrossRef] [PubMed]
  3. Kogut, M.H.; Lee, A.; Santin, E. Microbiome and pathogen interaction with the immune system. Poult. Sci. 2020, 99, 1906–1913. [Google Scholar] [CrossRef] [PubMed]
  4. Milling, S. Sophisticated specificity in the innate immune response. Immunology 2019, 158, 61–62. [Google Scholar] [CrossRef] [Green Version]
  5. Rivera, A.; Siracusa, M.C.; Yap, G.S.; Gause, W.C. Innate cell communication kick-starts pathogen-specific immunity. Nat. Immunol. 2016, 17, 356–363. [Google Scholar] [CrossRef]
  6. Iwasaki, A.; Medzhitov, R. Toll-like receptor control of the adaptive immune responses. Nat. Immunol. 2004, 5, 987–995. [Google Scholar] [CrossRef]
  7. Rodriguez Cetina Biefer, H.; Heinbokel, T.; Uehara, H.; Camacho, V.; Minami, K.; Nian, Y.; Koduru, S.; El Fatimy, R.; Ghiran, I.; Trachtenberg, A.J.; et al. Mast cells regulate CD4. J. Allergy Clin. Immunol. 2018, 142, 1894–1908.e7. [Google Scholar] [CrossRef] [Green Version]
  8. Galli, S.J.; Tsai, M. Mast cells in allergy and infection: Versatile effector and regulatory cells in innate and adaptive immunity. Eur. J. Immunol. 2010, 40, 1843–1851. [Google Scholar] [CrossRef] [Green Version]
  9. Thornton, C.A.; Morgan, G. Innate and adaptive immune pathways to tolerance. Nestle Nutr. Workshop Ser. Pediatr. Program 2009, 64, 45–57; discussion 57–61, 251–257. [Google Scholar]
  10. Lin, A.; Loré, K. Granulocytes: New Members of the Antigen-Presenting Cell Family. Front. Immunol. 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  11. Vono, M.; Lin, A.; Norrby-Teglund, A.; Koup, R.A.; Liang, F.; Loré, K. Neutrophils acquire the capacity for antigen presentation to memory CD4. Blood 2017, 129, 1991–2001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Li, Y.; Wang, W.; Yang, F.; Xu, Y.; Feng, C.; Zhao, Y. The regulatory roles of neutrophils in adaptive immunity. Cell Commun. Signal. 2019, 17, 147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Leliefeld, P.H.; Koenderman, L.; Pillay, J. How Neutrophils Shape Adaptive Immune Responses. Front. Immunol. 2015, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ley, K.; Hoffman, H.M.; Kubes, P.; Cassatella, M.A.; Zychlinsky, A.; Hedrick, C.C.; Catz, S.D. Neutrophils: New insights and open questions. Sci. Immunol. 2018, 3. [Google Scholar] [CrossRef] [Green Version]
  15. Rosales, C.; Lowell, C.A.; Schnoor, M.; Uribe-Querol, E. Neutrophils: Their Role in Innate and Adaptive Immunity 2017. J. Immunol. Res. 2017, 2017. [Google Scholar] [CrossRef]
  16. Cerutti, A.; Puga, I.; Magri, G. The B cell helper side of neutrophils. J. Leukoc. Biol. 2013, 94, 677–682. [Google Scholar] [CrossRef] [Green Version]
  17. Gestal, M.C.; Blas-Machado, U.; Johnson, H.M.; Rubin, L.N.; Dewan, K.K.; Bryant, C.; Tiemeyer, M.; Harvill, E.T. Disrupting Bordetella Immunosuppression Reveals a Role for Eosinophils in Coordinating the Adaptive Immune Response in the Respiratory Tract. Microorganisms 2020, 8, 11. [Google Scholar] [CrossRef]
  18. Borregaard, N. Neutrophils, from marrow to microbes. Immunity 2010, 33, 657–670. [Google Scholar] [CrossRef] [Green Version]
  19. Nauseef, W.M. How human neutrophils kill and degrade microbes: An integrated view. Immunol. Rev. 2007, 219, 88–102. [Google Scholar] [CrossRef]
  20. Reynolds, H.Y. Phagocytic defense in the lung. Antibiot. Chemother. 1985, 36, 74–87. [Google Scholar]
  21. Root, R.K.; Isturiz, R.; Molavi, A.; Metcalf, J.A.; Malech, H.L. Interactions between antibiotics and human neutrophils in the killing of staphylococci. J. Clin. Investig. 1981, 67, 247–259. [Google Scholar] [CrossRef] [PubMed]
  22. Zakhireh, B.; Block, L.H.; Root, R.K. Neutrophil function and host resistance. Infection 1979, 7, 88–98. [Google Scholar] [CrossRef] [PubMed]
  23. Lacy, P. Mechanisms of degranulation in neutrophils. Allergy Asthma Clin. Immunol. 2006, 2, 98–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Daniel, C.; Leppkes, M.; Muñoz, L.E.; Schley, G.; Schett, G.; Herrmann, M. Extracellular DNA traps in inflammation, injury and healing. Nat. Rev. Nephrol. 2019, 15, 559–575. [Google Scholar] [CrossRef]
  25. Burgener, S.S.; Schroder, K. Neutrophil Extracellular Traps in Host Defense. Cold Spring Harb. Perspect. Biol. 2019, 12, a037028. [Google Scholar] [CrossRef]
  26. Rada, B. Neutrophil Extracellular Traps. Methods Mol. Biol. 2019, 1982, 517–528. [Google Scholar]
  27. Rada, B. Neutrophil Extracellular Traps and Microcrystals. J. Immunol. Res. 2017, 2017. [Google Scholar] [CrossRef] [Green Version]
  28. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef]
  29. Brinkmann, V.; Zychlinsky, A. Beneficial suicide: Why neutrophils die to make NETs. Nat. Rev. Microbiol. 2007, 5, 577–582. [Google Scholar] [CrossRef]
  30. Ermert, D.; Urban, C.F.; Laube, B.; Goosmann, C.; Zychlinsky, A.; Brinkmann, V. Mouse neutrophil extracellular traps in microbial infections. J. Innate Immun. 2009, 1, 181–193. [Google Scholar] [CrossRef]
  31. Tsai, W.C.; Strieter, R.M.; Mehrad, B.; Newstead, M.W.; Zeng, X.; Standiford, T.J. CXC chemokine receptor CXCR2 is essential for protective innate host response in murine Pseudomonas aeruginosa pneumonia. Infect. Immun. 2000, 68, 4289–4296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Guo, B.; Abdelraouf, K.; Ledesma, K.R.; Chang, K.T.; Nikolaou, M.; Tam, V.H. Quantitative impact of neutrophils on bacterial clearance in a murine pneumonia model. Antimicrob. Agents Chemother. 2011, 55, 4601–4605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Witter, A.R.; Okunnu, B.M.; Berg, R.E. The Essential Role of Neutrophils during Infection with the Intracellular Bacterial Pathogen Listeria monocytogenes. J. Immunol. 2016, 197, 1557–1565. [Google Scholar] [CrossRef] [Green Version]
  34. Malech, H.L.; DeLeo, F.R.; Quinn, M.T. The Role of Neutrophils in the Immune System: An Overview. Methods Mol. Biol. 2020, 2087, 3–10. [Google Scholar] [PubMed]
  35. Nauseef, W.M.; Borregaard, N. Neutrophils at work. Nat. Immunol. 2014, 15, 602–611. [Google Scholar] [CrossRef]
  36. Tecchio, C.; Micheletti, A.; Cassatella, M.A. Neutrophil-derived cytokines: Facts beyond expression. Front. Immunol. 2014, 5. [Google Scholar] [CrossRef] [Green Version]
  37. Bordon, J.; Aliberti, S.; Fernandez-Botran, R.; Uriarte, S.M.; Rane, M.J.; Duvvuri, P.; Peyrani, P.; Morlacchi, L.C.; Blasi, F.; Ramirez, J.A. Understanding the roles of cytokines and neutrophil activity and neutrophil apoptosis in the protective versus deleterious inflammatory response in pneumonia. Int. J. Infect. Dis. 2013, 17, e76–e83. [Google Scholar] [CrossRef] [Green Version]
  38. Lehman, H.K.; Segal, B.H. The role of neutrophils in host defense and disease. J. Allergy Clin. Immunol. 2020, 145, 1535–1544. [Google Scholar] [CrossRef]
  39. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef]
  40. Kaplan, M.J.; Radic, M. Neutrophil extracellular traps: Double-edged swords of innate immunity. J. Immunol. 2012, 189, 2689–2695. [Google Scholar] [CrossRef] [Green Version]
  41. Yoo, D.G.; Floyd, M.; Winn, M.; Moskowitz, S.M.; Rada, B. NET formation induced by Pseudomonas aeruginosa cystic fibrosis isolates measured as release of myeloperoxidase-DNA and neutrophil elastase-DNA complexes. Immunol. Lett. 2014, 160, 186–194. [Google Scholar] [CrossRef] [PubMed]
  42. Kenny, E.F.; Herzig, A.; Krüger, R.; Muth, A.; Mondal, S.; Thompson, P.R.; Brinkmann, V.; Bernuth, H.V.; Zychlinsky, A. Diverse stimuli engage different neutrophil extracellular trap pathways. Elife 2017, 6, e24437. [Google Scholar] [CrossRef] [PubMed]
  43. White, P.C.; Chicca, I.J.; Ling, M.R.; Wright, H.J.; Cooper, P.R.; Milward, M.R.; Chapple, I.L. Characterization, Quantification, and Visualization of Neutrophil Extracellular Traps. Methods Mol. Biol. 2017, 1537, 481–497. [Google Scholar] [PubMed]
  44. Wang, X.; Li, X.; Chen, L.; Yuan, B.; Liu, T.; Dong, Q.; Liu, Y.; Yin, H. Interleukin-33 facilitates cutaneous defense against Staphylococcus aureus by promoting the development of neutrophil extracellular trap. Int. Immunopharmacol. 2020, 81. [Google Scholar] [CrossRef]
  45. Bitschar, K.; Staudenmaier, L.; Klink, L.; Focken, J.; Sauer, B.; Fehrenbacher, B.; Herster, F.; Bittner, Z.; Bleul, L.; Schaller, M.; et al. Staphylococcus aureus Skin Colonization Is Enhanced by the Interaction of Neutrophil Extracellular Traps with Keratinocytes. J. Investig. Dermatol 2020, 140, 1054–1065.e4. [Google Scholar] [CrossRef]
  46. Hsu, C.C.; Hsu, R.B.; Ohniwa, R.L.; Chen, J.W.; Yuan, C.T.; Chia, J.S.; Jung, C.J. Neutrophil Extracellular Traps Enhance Staphylococcus Aureus Vegetation Formation through Interaction with Platelets in Infective Endocarditis. Thromb. Haemost. 2019, 119, 786–796. [Google Scholar] [CrossRef]
  47. Petretto, A.; Bruschi, M.; Pratesi, F.; Croia, C.; Candiano, G.; Ghiggeri, G.; Migliorini, P. Neutrophil extracellular traps (NET) induced by different stimuli: A comparative proteomic analysis. PLoS ONE 2019, 14, e0218946. [Google Scholar] [CrossRef]
  48. Carestia, A.; Kaufman, T.; Rivadeneyra, L.; Landoni, V.I.; Pozner, R.G.; Negrotto, S.; D’Atri, L.P.; Gómez, R.M.; Schattner, M. Mediators and molecular pathways involved in the regulation of neutrophil extracellular trap formation mediated by activated platelets. J. Leukoc. Biol. 2016, 99, 153–162. [Google Scholar] [CrossRef]
  49. Hashiba, M.; Huq, A.; Tomino, A.; Hirakawa, A.; Hattori, T.; Miyabe, H.; Tsuda, M.; Takeyama, N. Neutrophil extracellular traps in patients with sepsis. J. Surg. Res. 2015, 194, 248–254. [Google Scholar] [CrossRef]
  50. Rada, B. Interactions between Neutrophils and Pseudomonas aeruginosa in Cystic Fibrosis. Pathogens 2017, 6, 10. [Google Scholar] [CrossRef]
  51. Rada, B. Neutrophil extracellular trap release driven by bacterial motility: Relevance to cystic fibrosis lung disease. Commun. Integr. Biol. 2017, 10, e1296610. [Google Scholar] [CrossRef] [PubMed]
  52. Yoo, D.G.; Winn, M.; Pang, L.; Moskowitz, S.M.; Malech, H.L.; Leto, T.L.; Rada, B. Release of cystic fibrosis airway inflammatory markers from Pseudomonas aeruginosa-stimulated human neutrophils involves NADPH oxidase-dependent extracellular DNA trap formation. J. Immunol. 2014, 192, 4728–4738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Azzouz, L.; Cherry, A.; Riedl, M.; Khan, M.; Pluthero, F.G.; Kahr, W.H.A.; Palaniyar, N.; Licht, C. Relative antibacterial functions of complement and NETs: NETs trap and complement effectively kills bacteria. Mol. Immunol. 2018, 97, 71–81. [Google Scholar] [CrossRef] [PubMed]
  54. Wartha, F.; Beiter, K.; Normark, S.; Henriques-Normark, B. Neutrophil extracellular traps: Casting the NET over pathogenesis. Curr. Opin. Microbiol. 2007, 10, 52–56. [Google Scholar] [CrossRef] [PubMed]
  55. Fuchs, T.A.; Abed, U.; Goosmann, C.; Hurwitz, R.; Schulze, I.; Wahn, V.; Weinrauch, Y.; Brinkmann, V.; Zychlinsky, A. Novel cell death program leads to neutrophil extracellular traps. J. Cell Biol. 2007, 176, 231–241. [Google Scholar] [CrossRef]
  56. Yousefi, S.; Mihalache, C.; Kozlowski, E.; Schmid, I.; Simon, H.U. Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps. Cell Death Differ. 2009, 16, 1438–1444. [Google Scholar] [CrossRef]
  57. McIlroy, D.J.; Jarnicki, A.G.; Au, G.G.; Lott, N.; Smith, D.W.; Hansbro, P.M.; Balogh, Z.J. Mitochondrial DNA neutrophil extracellular traps are formed after trauma and subsequent surgery. J. Crit. Care 2014, 29, 1133.e1–1133.e5. [Google Scholar] [CrossRef] [Green Version]
  58. Almyroudis, N.G.; Grimm, M.J.; Davidson, B.A.; Röhm, M.; Urban, C.F.; Segal, B.H. NETosis and NADPH oxidase: At the intersection of host defense, inflammation, and injury. Front. Immunol. 2013, 4. [Google Scholar] [CrossRef] [Green Version]
  59. Metzler, K.D.; Goosmann, C.; Lubojemska, A.; Zychlinsky, A.; Papayannopoulos, V. A myeloperoxidase-containing complex regulates neutrophil elastase release and actin dynamics during NETosis. Cell Rep. 2014, 8, 883–896. [Google Scholar] [CrossRef] [Green Version]
  60. Röhm, M.; Grimm, M.J.; D’Auria, A.C.; Almyroudis, N.G.; Segal, B.H.; Urban, C.F. NADPH oxidase promotes neutrophil extracellular trap formation in pulmonary aspergillosis. Infect. Immun. 2014, 82, 1766–1777. [Google Scholar] [CrossRef] [Green Version]
  61. Warnatsch, A.; Tsourouktsoglou, T.D.; Branzk, N.; Wang, Q.; Reincke, S.; Herbst, S.; Gutierrez, M.; Papayannopoulos, V. Reactive Oxygen Species Localization Programs Inflammation to Clear Microbes of Different Size. Immunity 2017, 46, 421–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Sil, P.; Hayes, C.P.; Reaves, B.J.; Breen, P.; Quinn, S.; Sokolove, J.; Rada, B. P2Y6 Receptor Antagonist MRS2578 Inhibits Neutrophil Activation and Aggregated Neutrophil Extracellular Trap Formation Induced by Gout-Associated Monosodium Urate Crystals. J. Immunol. 2017, 198, 428–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Arai, Y.; Nishinaka, Y.; Arai, T.; Morita, M.; Mizugishi, K.; Adachi, S.; Takaori-Kondo, A.; Watanabe, T.; Yamashita, K. Uric acid induces NADPH oxidase-independent neutrophil extracellular trap formation. Biochem. Biophys. Res. Commun. 2014, 443, 556–561. [Google Scholar] [CrossRef] [PubMed]
  64. McHugh, J. Distinguishing NET subtypes. Nat. Rev. Rheumatol. 2018, 14, 560. [Google Scholar] [CrossRef] [PubMed]
  65. Leshner, M.; Wang, S.; Lewis, C.; Zheng, H.; Chen, X.A.; Santy, L.; Wang, Y. PAD4 mediated histone hypercitrullination induces heterochromatin decondensation and chromatin unfolding to form neutrophil extracellular trap-like structures. Front. Immunol. 2012, 3. [Google Scholar] [CrossRef] [Green Version]
  66. Li, P.; Li, M.; Lindberg, M.R.; Kennett, M.J.; Xiong, N.; Wang, Y. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J. Exp. Med. 2010, 207, 1853–1862. [Google Scholar] [CrossRef]
  67. Wong, S.L.; Wagner, D.D. Peptidylarginine deiminase 4: A nuclear button triggering neutrophil extracellular traps in inflammatory diseases and aging. FASEB J. 2018, 32, 6258–6370. [Google Scholar] [CrossRef]
  68. Anzilotti, C.; Pratesi, F.; Tommasi, C.; Migliorini, P. Peptidylarginine deiminase 4 and citrullination in health and disease. Autoimmun. Rev. 2010, 9, 158–160. [Google Scholar] [CrossRef]
  69. Neeli, I.; Khan, S.N.; Radic, M. Histone deimination as a response to inflammatory stimuli in neutrophils. J. Immunol. 2008, 180, 1895–1902. [Google Scholar] [CrossRef]
  70. Wang, Y.; Li, M.; Stadler, S.; Correll, S.; Li, P.; Wang, D.; Hayama, R.; Leonelli, L.; Han, H.; Grigoryev, S.A.; et al. Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J. Cell Biol. 2009, 184, 205–213. [Google Scholar] [CrossRef] [Green Version]
  71. Thiam, H.R.; Wong, S.L.; Qiu, R.; Kittisopikul, M.; Vahabikashi, A.; Goldman, A.E.; Goldman, R.D.; Wagner, D.D.; Waterman, C.M. NETosis proceeds by cytoskeleton and endomembrane disassembly and PAD4-mediated chromatin decondensation and nuclear envelope rupture. Proc. Natl. Acad. Sci. USA 2020, 117, 7326–7337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Neeli, I.; Radic, M. Opposition between PKC isoforms regulates histone deimination and neutrophil extracellular chromatin release. Front. Immunol. 2013, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Yang, F.; He, Y.; Zhai, Z.; Sun, E. Programmed Cell Death Pathways in the Pathogenesis of Systemic Lupus Erythematosus. J. Immunol. Res. 2019, 2019. [Google Scholar] [CrossRef] [PubMed]
  74. Thiam, H.R.; Wong, S.L.; Wagner, D.D.; Waterman, C.M. Cellular Mechanisms of NETosis. Annu. Rev. Cell Dev. Biol. 2020, 36, 191–218. [Google Scholar] [CrossRef] [PubMed]
  75. Jorch, S.K.; Kubes, P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat. Med. 2017, 23, 279–287. [Google Scholar] [CrossRef]
  76. Malawista, S.E.; Van Blaricom, G. Cytoplasts made from human blood polymorphonuclear leukocytes with or without heat: Preservation of both motile function and respiratory burst oxidase activity. Proc. Natl. Acad. Sci. USA 1987, 84, 454–458. [Google Scholar] [CrossRef] [Green Version]
  77. Omann, G.M.; Swann, W.N.; Oades, Z.G.; Parkos, C.A.; Jesaitis, A.J.; Sklar, L.A. N-formylpeptide-receptor dynamics, cytoskeletal activation, and intracellular calcium response in human neutrophil cytoplasts. J. Immunol. 1987, 139, 3447–3455. [Google Scholar]
  78. Yost, C.C.; Cody, M.J.; Harris, E.S.; Thornton, N.L.; McInturff, A.M.; Martinez, M.L.; Chandler, N.B.; Rodesch, C.K.; Albertine, K.H.; Petti, C.A.; et al. Impaired neutrophil extracellular trap (NET) formation: A novel innate immune deficiency of human neonates. Blood 2009, 113, 6419–6427. [Google Scholar] [CrossRef] [Green Version]
  79. Rada, B.; Jendrysik, M.A.; Pang, L.; Hayes, C.P.; Yoo, D.G.; Park, J.J.; Moskowitz, S.M.; Malech, H.L.; Leto, T.L. Pyocyanin-enhanced neutrophil extracellular trap formation requires the NADPH oxidase. PLoS ONE 2013, 8, e54205. [Google Scholar] [CrossRef] [Green Version]
  80. Floyd, M.; Winn, M.; Cullen, C.; Sil, P.; Chassaing, B.; Yoo, D.G.; Gewirtz, A.T.; Goldberg, J.B.; McCarter, L.L.; Rada, B. Swimming Motility Mediates the Formation of Neutrophil Extracellular Traps Induced by Flagellated Pseudomonas aeruginosa. PLoS Pathog. 2016, 12, e1005987. [Google Scholar] [CrossRef] [Green Version]
  81. Femling, J.K.; Cherny, V.V.; Morgan, D.; Rada, B.; Davis, A.P.; Czirják, G.; Enyedi, P.; England, S.K.; Moreland, J.G.; Ligeti, E.; et al. The antibacterial activity of human neutrophils and eosinophils requires proton channels but not BK channels. J. Gen. Physiol 2006, 127, 659–672. [Google Scholar] [CrossRef] [PubMed]
  82. Rada, B.; Leto, T.L. Oxidative innate immune defenses by Nox/Duox family NADPH oxidases. Contrib. Microbiol. 2008, 15, 164–187. [Google Scholar] [PubMed] [Green Version]
  83. Urban, C.F.; Ermert, D.; Schmid, M.; Abu-Abed, U.; Goosmann, C.; Nacken, W.; Brinkmann, V.; Jungblut, P.R.; Zychlinsky, A. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 2009, 5, e1000639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Schonrich, G.; Raftery, M.J. Neutrophil Extracellular Traps Go Viral. Front. Immunol. 2016, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Saitoh, T.; Komano, J.; Saitoh, Y.; Misawa, T.; Takahama, M.; Kozaki, T.; Uehata, T.; Iwasaki, H.; Omori, H.; Yamaoka, S.; et al. Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1. Cell Host Microb. 2012, 12, 109–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Bonne-Année, S.; Kerepesi, L.A.; Hess, J.A.; Wesolowski, J.; Paumet, F.; Lok, J.B.; Nolan, T.J.; Abraham, D. Extracellular traps are associated with human and mouse neutrophil and macrophage mediated killing of larval Strongyloides stercoralis. Microb. Infect. 2014, 16, 502–511. [Google Scholar] [CrossRef] [Green Version]
  87. Abi Abdallah, D.S.; Lin, C.; Ball, C.J.; King, M.R.; Duhamel, G.E.; Denkers, E.Y. Toxoplasma gondii triggers release of human and mouse neutrophil extracellular traps. Infect. Immun. 2012, 80, 768–777. [Google Scholar] [CrossRef] [Green Version]
  88. Kho, S.; Minigo, G.; Andries, B.; Leonardo, L.; Prayoga, P.; Poespoprodjo, J.R.; Kenangalem, E.; Price, R.N.; Woodberry, T.; Anstey, N.M.; et al. Circulating Neutrophil Extracellular Traps and Neutrophil Activation Are Increased in Proportion to Disease Severity in Human Malaria. J. Infect. Dis. 2019, 219, 1994–2004. [Google Scholar] [CrossRef]
  89. Bos, A.C.; Beemsterboer, P.; Wolfs, T.F.; Versteegh, F.G.; Arets, H.G. Bordetella species in children with cystic fibrosis: What do we know? The role in acute exacerbations and chronic course. J. Cyst. Fibros 2011, 10, 307–312. [Google Scholar] [CrossRef] [Green Version]
  90. Martinez-Aleman, S.R.; Campos-Garcia, L.; Palma-Nicolas, J.P.; Hernandez-Bello, R.; Gonzalez, G.M.; Sanchez-Gonzalez, A. Understanding the Entanglement: Neutrophil Extracellular Traps (NETs) in Cystic Fibrosis. Front. Cell Infect. Microbiol. 2017, 7. [Google Scholar] [CrossRef] [Green Version]
  91. Hiroki, C.H.; Toller-Kawahisa, J.E.; Fumagalli, M.J.; Colon, D.F.; Figueiredo, L.T.M.; Fonseca, B.A.L.D.; Franca, R.F.O.; Cunha, F.Q. Neutrophil Extracellular Traps Effectively Control Acute Chikungunya Virus Infection. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Hojyo, S.; Uchida, M.; Tanaka, K.; Hasebe, R.; Tanaka, Y.; Murakami, M.; Hirano, T. How COVID-19 induces cytokine storm with high mortality. Inflamm. Regen. 2020, 40, 37. [Google Scholar] [CrossRef] [PubMed]
  93. Tang, Y.; Liu, J.; Zhang, D.; Xu, Z.; Ji, J.; Wen, C. Cytokine Storm in COVID-19: The Current Evidence and Treatment Strategies. Front. Immunol. 2020, 11. [Google Scholar] [CrossRef] [PubMed]
  94. Sinha, P.; Matthay, M.A.; Calfee, C.S. Is a “Cytokine Storm” Relevant to COVID-19? JAMA Intern. Med. 2020, 180, 1152–1154. [Google Scholar] [CrossRef] [PubMed]
  95. Kong, M.; Zhang, H.; Cao, X.; Mao, X.; Lu, Z. Higher level of neutrophil-to-lymphocyte is associated with severe COVID-19. Epidemiol. Infect. 2020, 148, e139. [Google Scholar] [CrossRef]
  96. Liu, L.; Zheng, Y.; Cai, L.; Wu, W.; Tang, S.; Ding, Y.; Liu, W.; Kou, G.; Xiong, Z.; Wang, S.; et al. Neutrophil-to-lymphocyte ratio, a critical predictor for assessment of disease severity in patients with COVID-19. Int. J. Lab. Hematol. 2020. [Google Scholar] [CrossRef]
  97. Wang, J.; Li, Q.; Yin, Y.; Zhang, Y.; Cao, Y.; Lin, X.; Huang, L.; Hoffmann, D.; Lu, M.; Qiu, Y. Excessive Neutrophils and Neutrophil Extracellular Traps in COVID-19. Front. Immunol. 2020, 11. [Google Scholar] [CrossRef]
  98. Laforge, M.; Elbim, C.; Frère, C.; Hémadi, M.; Massaad, C.; Nuss, P.; Benoliel, J.J.; Becker, C. Tissue damage from neutrophil-induced oxidative stress in COVID-19. Nat. Rev. Immunol. 2020, 20, 579. [Google Scholar] [CrossRef]
  99. Didangelos, A. COVID-19 Hyperinflammation: What about Neutrophils? mSphere 2020, 5. [Google Scholar] [CrossRef]
  100. Leppkes, M.; Knopf, J.; Naschberger, E.; Lindemann, A.; Singh, J.; Herrmann, I.; Stürzl, M.; Staats, L.; Mahajan, A.; Schauer, C.; et al. Vascular occlusion by neutrophil extracellular traps in COVID-19. EBioMedicine 2020, 58. [Google Scholar] [CrossRef]
  101. Middleton, E.A.; He, X.Y.; Denorme, F.; Campbell, R.A.; Ng, D.; Salvatore, S.P.; Mostyka, M.; Baxter-Stoltzfus, A.; Borczuk, A.C.; Loda, M.; et al. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 2020, 136, 1169–1179. [Google Scholar] [CrossRef] [PubMed]
  102. Schulte-Schrepping, J.; Reusch, N.; Paclik, D.; Baßler, K.; Schlickeiser, S.; Zhang, B.; Krämer, B.; Krammer, T.; Brumhard, S.; Bonaguro, L.; et al. Severe COVID-19 Is Marked by a Dysregulated Myeloid Cell Compartment. Cell 2020, 182, 1419–1440.e23. [Google Scholar] [CrossRef] [PubMed]
  103. Arcanjo, A.; Logullo, J.; Menezes, C.C.B.; de Souza Carvalho Giangiarulo, T.C.; Dos Reis, M.C.; de Castro, G.M.M.; da Silva Fontes, Y.; Todeschini, A.R.; Freire-de-Lima, L.; Decoté-Ricardo, D.; et al. The emerging role of neutrophil extracellular traps in severe acute respiratory syndrome coronavirus 2 (COVID-19). Sci. Rep. 2020, 10, 19630. [Google Scholar] [CrossRef] [PubMed]
  104. Chiang, C.C.; Korinek, M.; Cheng, W.J.; Hwang, T.L. Targeting Neutrophils to Treat Acute Respiratory Distress Syndrome in Coronavirus Disease. Front. Pharmacol 2020, 11. [Google Scholar] [CrossRef]
  105. Castanheira, F.V.S.; Kubes, P. Neutrophils and NETs in modulating acute and chronic inflammation. Blood 2019, 133, 2178–2185. [Google Scholar] [CrossRef]
  106. Meng, W.; Paunel-Görgülü, A.; Flohé, S.; Hoffmann, A.; Witte, I.; MacKenzie, C.; Baldus, S.E.; Windolf, J.; Lögters, T.T. Depletion of neutrophil extracellular traps in vivo results in hypersusceptibility to polymicrobial sepsis in mice. Crit. Care 2012, 16, R137. [Google Scholar] [CrossRef] [Green Version]
  107. Czaikoski, P.G.; Mota, J.M.; Nascimento, D.C.; Sônego, F.; Castanheira, F.V.; Melo, P.H.; Scortegagna, G.T.; Silva, R.L.; Barroso-Sousa, R.; Souto, F.O.; et al. Neutrophil Extracellular Traps Induce Organ Damage during Experimental and Clinical Sepsis. PLoS ONE 2016, 11, e0148142. [Google Scholar] [CrossRef] [Green Version]
  108. Zha, Z.; Leng, R.; Xu, W.; Bao, H.; Chen, Y.; Fang, L.; Liu, Z.; Ye, D. Prevalence and risk factors of chronic obstructive pulmonary disease in Anhui Province, China: A population-based survey. BMC Pulm. Med. 2019, 19, 102. [Google Scholar] [CrossRef] [Green Version]
  109. Contoli, M.; Baraldo, S.; Conti, V.; Gnesini, G.; Marku, B.; Casolari, P.; Scrigner, P.; Morelli, P.; Saetta, M.; Spanevello, A.; et al. Airway inflammatory profile is correlated with symptoms in stable COPD: A longitudinal proof-of-concept cohort study. Respirology 2020, 25, 80–88. [Google Scholar] [CrossRef]
  110. Harries, T.H.; Rowland, V.; Corrigan, C.J.; Marshall, I.J.; McDonnell, L.; Prasad, V.; Schofield, P.; Armstrong, D.; White, P. Blood eosinophil count, a marker of inhaled corticosteroid effectiveness in preventing COPD exacerbations in post-hoc RCT and observational studies: Systematic review and meta-analysis. Respir. Res. 2020, 21, 3. [Google Scholar] [CrossRef] [PubMed]
  111. McDonald, C.F. Eosinophils in chronic obstructive pulmonary disease: Are they just another biomarker? Curr. Opin. Pulm. Med. 2020, 26, 169–174. [Google Scholar] [CrossRef] [PubMed]
  112. Sil, P.; Yoo, D.G.; Floyd, M.; Gingerich, A.; Rada, B. High Throughput Measurement of Extracellular DNA Release and Quantitative NET Formation in Human Neutrophils In Vitro. J. Vis. Exp. 2016. [Google Scholar] [CrossRef] [PubMed]
  113. Doskocil, I.; Havlik, J.; Verlotta, R.; Tauchen, J.; Vesela, L.; Macakova, K.; Opletal, L.; Kokoska, L.; Rada, V. In vitro immunomodulatory activity, cytotoxicity and chemistry of some central European polypores. Pharm. Biol. 2016, 54, 2369–2376. [Google Scholar] [CrossRef] [Green Version]
  114. Niedźwiedzka-Rystwej, P.; Repka, W.; Tokarz-Deptuła, B.; Deptuła, W. “In sickness and in health”—How neutrophil extracellular trap (NET) works in infections, selected diseases and pregnancy. J. Inflamm. 2019, 16. [Google Scholar] [CrossRef] [PubMed]
  115. Sil, P.; Wicklum, H.; Surell, C.; Rada, B. Macrophage-derived IL-1β enhances monosodium urate crystal-triggered NET formation. Inflamm. Res. 2017, 66, 227–237. [Google Scholar] [CrossRef] [Green Version]
  116. Lachowicz-Scroggins, M.E.; Dunican, E.M.; Charbit, A.R.; Raymond, W.; Looney, M.R.; Peters, M.C.; Gordon, E.D.; Woodruff, P.G.; Lefrançais, E.; Phillips, B.R.; et al. Extracellular DNA, Neutrophil Extracellular Traps, and Inflammasome Activation in Severe Asthma. Am. J. Respir. Crit. Care Med. 2019, 199, 1076–1085. [Google Scholar] [CrossRef]
  117. Engelmann, B.; Massberg, S. Thrombosis as an intravascular effector of innate immunity. Nat. Rev. Immunol. 2013, 13, 34–45. [Google Scholar] [CrossRef]
  118. Fuchs, T.A.; Brill, A.; Duerschmied, D.; Schatzberg, D.; Monestier, M.; Myers, D.D.; Wrobleski, S.K.; Wakefield, T.W.; Hartwig, J.H.; Wagner, D.D. Extracellular DNA traps promote thrombosis. Proc. Natl. Acad. Sci. USA 2010, 107, 15880–15885. [Google Scholar] [CrossRef] [Green Version]
  119. Brill, A.; Fuchs, T.A.; Savchenko, A.S.; Thomas, G.M.; Martinod, K.; De Meyer, S.F.; Bhandari, A.A.; Wagner, D.D. Neutrophil extracellular traps promote deep vein thrombosis in mice. J. Thromb. Haemost. 2012, 10, 136–144. [Google Scholar] [CrossRef] [Green Version]
  120. von Brühl, M.L.; Stark, K.; Steinhart, A.; Chandraratne, S.; Konrad, I.; Lorenz, M.; Khandoga, A.; Tirniceriu, A.; Coletti, R.; Köllnberger, M.; et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J. Exp. Med. 2012, 209, 819–835. [Google Scholar] [CrossRef]
  121. Stakos, D.A.; Kambas, K.; Konstantinidis, T.; Mitroulis, I.; Apostolidou, E.; Arelaki, S.; Tsironidou, V.; Giatromanolaki, A.; Skendros, P.; Konstantinides, S.; et al. Expression of functional tissue factor by neutrophil extracellular traps in culprit artery of acute myocardial infarction. Eur. Heart J. 2015, 36, 1405–1414. [Google Scholar] [CrossRef] [PubMed]
  122. Beiter, K.; Wartha, F.; Albiger, B.; Normark, S.; Zychlinsky, A.; Henriques-Normark, B. An endonuclease allows Streptococcus pneumoniae to escape from neutrophil extracellular traps. Curr. Biol. 2006, 16, 401–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Berends, E.T.; Horswill, A.R.; Haste, N.M.; Monestier, M.; Nizet, V.; von Köckritz-Blickwede, M. Nuclease expression by Staphylococcus aureus facilitates escape from neutrophil extracellular traps. J. Innate Immun. 2010, 2, 576–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kamoshida, G.; Kikuchi-Ueda, T.; Tansho-Nagakawa, S.; Nakano, R.; Nakano, A.; Kikuchi, H.; Ubagai, T.; Ono, Y. Acinetobacter baumannii escape from neutrophil extracellular traps (NETs). J. Infect. Chemother. 2015, 21, 43–49. [Google Scholar] [CrossRef]
  125. Urban, C.F.; Lourido, S.; Zychlinsky, A. How do microbes evade neutrophil killing? Cell Microbiol. 2006, 8, 1687–1696. [Google Scholar] [CrossRef]
  126. Eby, J.C.; Gray, M.C.; Hewlett, E.L. Cyclic AMP-mediated suppression of neutrophil extracellular trap formation and apoptosis by the Bordetella pertussis adenylate cyclase toxin. Infect. Immun. 2014, 82, 5256–5269. [Google Scholar] [CrossRef] [Green Version]
  127. Gorgojo, J.; Scharrig, E.; Gómez, R.M.; Harvill, E.T.; Rodríguez, M.E. Bordetella parapertussis Circumvents Neutrophil Extracellular Bactericidal Mechanisms. PLoS ONE 2017, 12, e0169936. [Google Scholar] [CrossRef] [Green Version]
  128. Storisteanu, D.M.; Pocock, J.M.; Cowburn, A.S.; Juss, J.K.; Nadesalingam, A.; Nizet, V.; Chilvers, E.R. Evasion of Neutrophil Extracellular Traps by Respiratory Pathogens. Am. J. Respir. Cell Mol. Biol. 2017, 56, 423–431. [Google Scholar] [CrossRef]
  129. Xia, X.; Qin, W.; Zhu, H.; Wang, X.; Jiang, J.; Hu, J. How Streptococcus suis serotype 2 attempts to avoid attack by host immune defenses. J. Microbiol. Immunol. Infect. 2019, 52, 516–525. [Google Scholar] [CrossRef]
  130. Palmer, A.; Criss, A.K. Gonococcal Defenses against Antimicrobial Activities of Neutrophils. Trends Microbiol. 2018, 26, 1022–1034. [Google Scholar] [CrossRef]
  131. Gestal, M.C.; Howard, L.K.; Dewan, K.; Johnson, H.M.; Barbier, M.; Bryant, C.; Soumana, I.H.; Rivera, I.; Linz, B.; Blas-Machado, U.; et al. Enhancement of immune response against Bordetella spp. by disrupting immunomodulation. Sci. Rep. 2019, 9, 20261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Gestal, M.C.; Whitesides, L.T.; Harvill, E.T. Integrated Signaling Pathways Mediate Bordetella Immunomodulation, Persistence, and Transmission. Trends Microbiol. 2019, 27, 118–130. [Google Scholar] [CrossRef] [PubMed]
  133. Cartelle Gestal, M. Vencer la Resistencia a Antibioticos; Investigacion y Ciencia: Barcelona, Spain, 2013. [Google Scholar]
  134. Holban, A.M.; Gestal, M.C.; Grumezescu, A.M. New molecular strategies for reducing implantable medical devices associated infections. Curr. Med. Chem. 2014, 21, 3375–3382. [Google Scholar] [CrossRef] [PubMed]
  135. Holban, A.M.; Gestal, M.C.; Grumezescu, A.M. Control of biofilm-associated infections by signaling molecules and nanoparticles. Int. J. Pharm. 2016, 510, 409–418. [Google Scholar] [CrossRef] [PubMed]
  136. Skopelja-Gardner, S.; Theprungsirikul, J.; Lewis, K.A.; Hammond, J.H.; Carlson, K.M.; Hazlett, H.F.; Nymon, A.; Nguyen, D.; Berwin, B.L.; Hogan, D.A.; et al. Regulation of Pseudomonas aeruginosa-Mediated Neutrophil Extracellular Traps. Front. Immunol. 2019, 10, 1670. [Google Scholar] [CrossRef] [Green Version]
  137. Klion, A.D.; Ackerman, S.J.; Bochner, B.S. Contributions of Eosinophils to Human Health and Disease. Annu. Rev. Pathol. 2020, 15, 179–209. [Google Scholar] [CrossRef] [Green Version]
  138. Lee, J.J.; Jacobsen, E.A.; Ochkur, S.I.; McGarry, M.P.; Condjella, R.M.; Doyle, A.D.; Luo, H.; Zellner, K.R.; Protheroe, C.A.; Willetts, L.; et al. Human versus mouse eosinophils: “that which we call an eosinophil, by any other name would stain as red”. J. Allergy Clin. Immunol. 2012, 130, 572–584. [Google Scholar] [CrossRef] [Green Version]
  139. Weller, P.F.; Spencer, L.A. Functions of tissue-resident eosinophils. Nat. Rev. Immunol. 2017, 17, 746–760. [Google Scholar] [CrossRef]
  140. Rosenberg, H.F.; Dyer, K.D.; Foster, P.S. Eosinophils: Changing perspectives in health and disease. Nat. Rev. Immunol. 2013, 13, 9–22. [Google Scholar] [CrossRef]
  141. Blanchard, C.; Rothenberg, M.E. Biology of the eosinophil. Adv. Immunol. 2009, 101, 81–121. [Google Scholar]
  142. Acharya, K.R.; Ackerman, S.J. Eosinophil granule proteins: Form and function. J. Biol. Chem. 2014, 289, 17406–17415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Spencer, L.A.; Bonjour, K.; Melo, R.C.; Weller, P.F. Eosinophil secretion of granule-derived cytokines. Front. Immunol. 2014, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Melo, R.C.N.; Weller, P.F. Contemporary understanding of the secretory granules in human eosinophils. J. Leukoc. Biol. 2018, 104, 85–93. [Google Scholar] [CrossRef] [PubMed]
  145. Shamri, R.; Xenakis, J.J.; Spencer, L.A. Eosinophils in innate immunity: An evolving story. Cell Tissue Res. 2011, 343, 57–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Lee, J.J.; Jacobsen, E.A.; McGarry, M.P.; Schleimer, R.P.; Lee, N.A. Eosinophils in health and disease: The LIAR hypothesis. Clin. Exp. Allergy 2010, 40, 563–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Liew, F.Y.; Girard, J.P.; Turnquist, H.R. Interleukin-33 in health and disease. Nat. Rev. Immunol. 2016, 16, 676–689. [Google Scholar] [CrossRef]
  148. Stolarski, B.; Kurowska-Stolarska, M.; Kewin, P.; Xu, D.; Liew, F.Y. IL-33 exacerbates eosinophil-mediated airway inflammation. J. Immunol. 2010, 185, 3472–3480. [Google Scholar] [CrossRef] [Green Version]
  149. Jacobsen, E.A.; Doyle, A.D.; Colbert, D.C.; Zellner, K.R.; Protheroe, C.A.; LeSuer, W.E.; Lee, N.A.; Lee, J.J. Differential activation of airway eosinophils induces IL-13-mediated allergic Th2 pulmonary responses in mice. Allergy 2015, 70, 1148–1159. [Google Scholar] [CrossRef] [Green Version]
  150. Doan, T.C.; Jeong, B.M.; Coden, M.E.; Loffredo, L.F.; Bhattacharyya, S.; Chiarella, S.E.; Varga, J.; Abdala-Valencia, H.; Berdnikovs, S. Matrix protein tenascin-C expands and reversibly blocks maturation of murine eosinophil progenitors. J. Allergy Clin. Immunol. 2018, 142, 695–698.e4. [Google Scholar] [CrossRef] [Green Version]
  151. Isobe, Y.; Kato, T.; Arita, M. Emerging roles of eosinophils and eosinophil-derived lipid mediators in the resolution of inflammation. Front. Immunol. 2012, 3. [Google Scholar] [CrossRef] [Green Version]
  152. Yamada, T.; Tani, Y.; Nakanishi, H.; Taguchi, R.; Arita, M.; Arai, H. Eosinophils promote resolution of acute peritonitis by producing proresolving mediators in mice. FASEB J. 2011, 25, 561–568. [Google Scholar] [CrossRef] [PubMed]
  153. Jacobsen, E.A.; Helmers, R.A.; Lee, J.J.; Lee, N.A. The expanding role(s) of eosinophils in health and disease. Blood 2012, 120, 3882–3890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Abdala-Valencia, H.; Coden, M.E.; Chiarella, S.E.; Jacobsen, E.A.; Bochner, B.S.; Lee, J.J.; Berdnikovs, S. Shaping eosinophil identity in the tissue contexts of development, homeostasis, and disease. J. Leukoc. Biol. 2018, 104, 95–108. [Google Scholar] [CrossRef] [PubMed]
  155. Mukherjee, M.; Lacy, P.; Ueki, S. Eosinophil Extracellular Traps and Inflammatory Pathologies-Untangling the Web! Front. Immunol. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
  156. Yousefi, S.; Simon, D.; Stojkov, D.; Karsonova, A.; Karaulov, A.; Simon, H.U. In vivo evidence for extracellular DNA trap formation. Cell Death Dis. 2020, 11, 300. [Google Scholar] [CrossRef]
  157. Yousefi, S.; Simon, D.; Simon, H.U. Eosinophil extracellular DNA traps: Molecular mechanisms and potential roles in disease. Curr. Opin. Immunol. 2012, 24, 736–739. [Google Scholar] [CrossRef]
  158. Rada, B.K.; Geiszt, M.; Kaldi, K.; Timar, C.; Ligeti, E. Dual role of phagocytic NADPH oxidase in bacterial killing. Blood 2004, 104, 2947–2953. [Google Scholar] [CrossRef]
  159. Schorn, C.; Janko, C.; Krenn, V.; Zhao, Y.; Munoz, L.E.; Schett, G.; Herrmann, M. Bonding the foe—NETting neutrophils immobilize the pro-inflammatory monosodium urate crystals. Front. Immunol. 2012, 3. [Google Scholar] [CrossRef] [Green Version]
  160. Schorn, C.; Janko, C.; Latzko, M.; Chaurio, R.; Schett, G.; Herrmann, M. Monosodium urate crystals induce extracellular DNA traps in neutrophils, eosinophils, and basophils but not in mononuclear cells. Front. Immunol. 2012, 3. [Google Scholar] [CrossRef] [Green Version]
  161. Kobayashi, T.; Kouzaki, H.; Kita, H. Human eosinophils recognize endogenous danger signal crystalline uric acid and produce proinflammatory cytokines mediated by autocrine ATP. J. Immunol. 2010, 184, 6350–6358. [Google Scholar] [CrossRef] [Green Version]
  162. Dixon, H.; Blanchard, C.; Deschoolmeester, M.L.; Yuill, N.C.; Christie, J.W.; Rothenberg, M.E.; Else, K.J. The role of Th2 cytokines, chemokines and parasite products in eosinophil recruitment to the gastrointestinal mucosa during helminth infection. Eur. J. Immunol. 2006, 36, 1753–1763. [Google Scholar] [CrossRef] [PubMed]
  163. Domachowske, J.B.; Rosenberg, H.F. Respiratory syncytial virus infection: Immune response, immunopathogenesis, and treatment. Clin. Microbiol. Rev. 1999, 12, 298–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Rosenberg, H.F.; Domachowske, J.B. Eosinophils, ribonucleases and host defense: Solving the puzzle. Immunol. Res. 1999, 20, 261–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. O’Bryan, L.; Pinkston, P.; Kumaraswami, V.; Vijayan, V.; Yenokida, G.; Rosenberg, H.F.; Crystal, R.; Ottesen, E.A.; Nutman, T.B. Localized eosinophil degranulation mediates disease in tropical pulmonary eosinophilia. Infect. Immun. 2003, 71, 1337–1342. [Google Scholar] [CrossRef] [Green Version]
  166. Moreau, J.M.; Dyer, K.D.; Bonville, C.A.; Nitto, T.; Vasquez, N.L.; Easton, A.J.; Domachowske, J.B.; Rosenberg, H.F. Diminished expression of an antiviral ribonuclease in response to pneumovirus infection in vivo. Antiviral. Res. 2003, 59, 181–191. [Google Scholar] [CrossRef]
  167. Dyer, K.D.; Percopo, C.M.; Fischer, E.R.; Gabryszewski, S.J.; Rosenberg, H.F. Pneumoviruses infect eosinophils and elicit MyD88-dependent release of chemoattractant cytokines and interleukin-6. Blood 2009, 114, 2649–2656. [Google Scholar] [CrossRef] [PubMed]
  168. Percopo, C.M.; Dyer, K.D.; Karpe, K.A.; Domachowske, J.B.; Rosenberg, H.F. Eosinophils and respiratory virus infection: A dual-standard curve qRT-PCR-based method for determining virus recovery from mouse lung tissue. Methods Mol. Biol. 2014, 1178, 257–266. [Google Scholar] [PubMed]
  169. Krishack, P.A.; Louviere, T.J.; Decker, T.S.; Kuzel, T.G.; Greenberg, J.A.; Camacho, D.F.; Hrusch, C.L.; Sperling, A.I.; Verhoef, P.A. Protection against Staphylococcus aureus bacteremia-induced mortality depends on ILC2s and eosinophils. JCI Insight 2019, 4, e124168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Berek, C. Eosinophils can more than kill. J. Exp. Med. 2018, 215, 1967–1969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Figueiredo, R.T.; Neves, J.S. Eosinophils in fungal diseases: An overview. J. Leukoc. Biol. 2018, 104, 49–60. [Google Scholar] [CrossRef]
  172. Ueki, S.; Hebisawa, A.; Kitani, M.; Asano, K.; Neves, J.S. Allergic Bronchopulmonary Aspergillosis-A Luminal Hypereosinophilic Disease With Extracellular Trap Cell Death. Front. Immunol. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
  173. Omokawa, A.; Ueki, S.; Kikuchi, Y.; Takeda, M.; Asano, M.; Sato, K.; Sano, M.; Ito, H.; Hirokawa, M. Mucus plugging in allergic bronchopulmonary aspergillosis: Implication of the eosinophil DNA traps. Allergol. Int. 2018, 67, 280–282. [Google Scholar] [CrossRef] [PubMed]
  174. Muniz, V.S.; Silva, J.C.; Braga, Y.A.V.; Melo, R.C.N.; Ueki, S.; Takeda, M.; Hebisawa, A.; Asano, K.; Figueiredo, R.T.; Neves, J.S. Eosinophils release extracellular DNA traps in response to Aspergillus fumigatus. J. Allergy Clin. Immunol. 2018, 141, 571–585.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Wagner, J.M.; Franco, M.; Kephart, G.M.; Gleich, G.J. Localization of eosinophil granule major basic protein in paracoccidioidomycosis lesions. Am. J. Trop. Med. Hyg. 1998, 59, 66–72. [Google Scholar] [CrossRef] [Green Version]
  176. Marques de Macedo, P.; de Oliveira, L.C.; Freitas, D.F.; da Rocha, J.A.; Freitas, A.D.; Nucci, M.; Zancopé-Oliveira, R.M.; Almeida-Paes, R.; do Valle, A.C. Acute Paracoccidioidomycosis Due to Paracoccidioides brasiliensis S1 Mimicking Hypereosinophilic Syndrome with Massive Splenomegaly: Diagnostic Challenge. PLoS Negl. Trop. Dis. 2016, 10, e0004487. [Google Scholar] [CrossRef] [PubMed]
  177. Diniz, S.N.; Cisalpino, P.S.; Freire, A.T.; Silva-Teixeira, D.N.; Contigli, C.; Rodrigues Júnior, V.; Goes, A.M. In vitro granuloma formation, NO production and cytokines profile from human mononuclear cells induced by fractionated antigens of Paracoccidioides brasiliensis. Hum. Immunol. 2001, 62, 799–808. [Google Scholar] [CrossRef]
  178. Braga, F.G.; Ruas, L.P.; Pereira, R.M.; Lima, X.T.; Antunes, E.; Mamoni, R.L.; Blotta, M.H.S.L. Functional and phenotypic evaluation of eosinophils from patients with the acute form of paracoccidioidomycosis. PLoS Negl. Trop. Dis. 2017, 11, e0005601. [Google Scholar] [CrossRef] [PubMed]
  179. Fujieda, S.; Imoto, Y.; Kato, Y.; Ninomiya, T.; Tokunaga, T.; Tsutsumiuchi, T.; Yoshida, K.; Kidoguchi, M.; Takabayashi, T. Eosinophilic chronic rhinosinusitis. Allergol. Int. 2019, 68, 403–412. [Google Scholar] [CrossRef]
  180. Gevaert, E.; Zhang, N.; Krysko, O.; Lan, F.; Holtappels, G.; De Ruyck, N.; Nauwynck, H.; Yousefi, S.; Simon, H.U.; Bachert, C. Extracellular eosinophilic traps in association with Staphylococcus aureus at the site of epithelial barrier defects in patients with severe airway inflammation. J. Allergy Clin. Immunol. 2017, 139, 1849–1860.e6. [Google Scholar] [CrossRef] [Green Version]
  181. Silveira, J.S.; Antunes, G.L.; Gassen, R.B.; Breda, R.V.; Stein, R.T.; Pitrez, P.M.; da Cunha, A.A. Respiratory syncytial virus increases eosinophil extracellular traps in a murine model of asthma. Asia Pac. Allergy 2019, 9, e32. [Google Scholar] [CrossRef]
  182. Sabogal Piñeros, Y.S.; Bal, S.M.; Dijkhuis, A.; Majoor, C.J.; Dierdorp, B.S.; Dekker, T.; Hoefsmit, E.P.; Bonta, P.I.; Picavet, D.; van der Wel, N.N.; et al. Eosinophils capture viruses, a capacity that is defective in asthma. Allergy 2019, 74, 1898–1909. [Google Scholar] [CrossRef]
  183. Flores-Torres, A.S.; Salinas-Carmona, M.C.; Salinas, E.; Rosas-Taraco, A.G. Eosinophils and Respiratory Viruses. Viral. Immunol. 2019, 32, 198–207. [Google Scholar] [CrossRef] [PubMed]
  184. Samarasinghe, A.E.; Melo, R.C.; Duan, S.; LeMessurier, K.S.; Liedmann, S.; Surman, S.L.; Lee, J.J.; Hurwitz, J.L.; Thomas, P.G.; McCullers, J.A. Eosinophils Promote Antiviral Immunity in Mice Infected with Influenza A Virus. J. Immunol. 2017, 198, 3214–3226. [Google Scholar] [CrossRef] [PubMed]
  185. Réfabert, L.; De Blic, J.; Scheinmann, P. Acute viral respiratory tract infections and childhood asthma. Rev. Fr. Allergol. Immunol. Clin. 1996, 36, 775–781. [Google Scholar] [PubMed]
  186. Abidi, K.; Belayachi, J.; Derras, Y.; Khayari, M.E.; Dendane, T.; Madani, N.; Khoudri, I.; Zeggwagh, A.A.; Abouqal, R. Eosinopenia, an early marker of increased mortality in critically ill medical patients. Intensive Care Med. 2011, 37, 1136–1142. [Google Scholar] [CrossRef]
  187. Lippi, G.; Henry, B.M. Eosinophil count in severe coronavirus disease 2019 (COVID-19). QJM 2020. [Google Scholar]
  188. Lindsley, A.W.; Schwartz, J.T.; Rothenberg, M.E. Eosinophil responses during COVID-19 infections and coronavirus vaccination. J. Allergy Clin. Immunol. 2020, 146, 1–7. [Google Scholar] [CrossRef]
  189. Sun, Y.; Dong, Y.; Wang, L.; Xie, H.; Li, B.; Chang, C.; Wang, F.S. Characteristics and prognostic factors of disease severity in patients with COVID-19: The Beijing experience. J. Autoimmun. 2020, 112, 102473. [Google Scholar] [CrossRef]
  190. Qian, G.Q.; Zhang, X.; Ma, A.H.Y.; Yang, N.B. Response letter to Eosinophil count in severe coronavirus disease 2019 (COVID-19). QJM 2020, 113, 7. [Google Scholar] [CrossRef] [Green Version]
  191. Perlini, S.; Ciprandi, G.; Castagnoli, R.; Licari, A.; Marseglia, G.L. Eosinopenia could be a relevant prognostic biomarker in patients with coronavirus disease 2019. Allergy Asthma Proc. 2020, 41, e80–e82. [Google Scholar] [CrossRef]
  192. Pereira, M.A.M.; Barros, I.C.A.; Jacob, A.L.V.; Assis, M.L.; Kanaan, S.; Kang, H.C. Laboratory findings in SARS-CoV-2 infections: State of the art. Rev. Assoc. Med. Bras. 2020, 66, 1152–1156. [Google Scholar] [CrossRef] [PubMed]
  193. Roca, E.; Ventura, L.; Zattra, C.M.; Lombardi, C. EOSINOPENIA: An early, effective, and relevant COVID-19 biomarker? QJM 2020. [Google Scholar] [CrossRef]
  194. Tanni, F.; Akker, E.; Zaman, M.M.; Figueroa, N.; Tharian, B.; Hupart, K.H. Eosinopenia and COVID-19. J. Am. Osteopath. Assoc. 2020, 120, 504–508. [Google Scholar] [CrossRef]
  195. Zhao, L.; Zhang, Y.P.; Yang, X.; Liu, X. Eosinopenia is associated with greater severity in patients with coronavirus disease 2019. Allergy 2020. [Google Scholar] [CrossRef] [PubMed]
  196. Xia, Z. Eosinopenia as an early diagnostic marker of COVID-19 at the time of the epidemic. EClinicalMedicine 2020, 23, 100398. [Google Scholar] [CrossRef] [PubMed]
  197. Xie, G.; Ding, F.; Han, L.; Yin, D.; Lu, H.; Zhang, M. The role of peripheral blood eosinophil counts in COVID-19 patients. Allergy 2020. [Google Scholar] [CrossRef] [PubMed]
  198. Rodrigo-Muñoz, J.M.; Sastre, B.; Cañas, J.A.; Gil-Martínez, M.; Redondo, N.; Del Pozo, V. Eosinophil Response Against Classical and Emerging Respiratory Viruses: COVID-19. J. Investig. Allergol. Clin. Immunol. 2020, 31. [Google Scholar] [CrossRef]
  199. Lucas, C.; Wong, P.; Klein, J.; Castro, T.B.R.; Silva, J.; Sundaram, M.; Ellingson, M.K.; Mao, T.; Oh, J.E.; Israelow, B.; et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature 2020, 584, 463–469. [Google Scholar] [CrossRef]
  200. Du, Y.; Tu, L.; Zhu, P.; Mu, M.; Wang, R.; Yang, P.; Wang, X.; Hu, C.; Ping, R.; Hu, P.; et al. Clinical Features of 85 Fatal Cases of COVID-19 from Wuhan. A Retrospective Observational Study. Am. J. Respir. Crit. Care Med. 2020, 201, 1372–1379. [Google Scholar] [CrossRef] [Green Version]
  201. Dosanjh, A. Eosinophil-Derived Neurotoxin and Respiratory Tract Infection and Inflammation: Implications for COVID-19 Management. J. Interferon. Cytokine Res. 2020, 40, 443–445. [Google Scholar] [CrossRef]
  202. Gestal, M.C.; Rivera, I.; Howard, L.K.; Dewan, K.K.; Soumana, I.H.; Dedloff, M.; Nicholson, T.L.; Linz, B.; Harvill, E.T. Blood or Serum Exposure Induce Global Transcriptional Changes, Altered Antigenic Profile, and Increased Cytotoxicity by Classical Bordetellae. Front. Microbiol. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
  203. Ueki, S.; Tokunaga, T.; Melo, R.C.N.; Saito, H.; Honda, K.; Fukuchi, M.; Konno, Y.; Takeda, M.; Yamamoto, Y.; Hirokawa, M.; et al. Charcot-Leyden crystal formation is closely associated with eosinophil extracellular trap cell death. Blood 2018, 132, 2183–2187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Granger, V.; Taillé, C.; Roach, D.; Letuvé, S.; Dupin, C.; Hamidi, F.; Noël, B.; Neukirch, C.; Aubier, M.; Pretolani, M.; et al. Circulating neutrophil and eosinophil extracellular traps are markers of severe asthma. Allergy 2020, 75, 699–702. [Google Scholar] [CrossRef] [PubMed]
  205. Dworski, R.; Simon, H.U.; Hoskins, A.; Yousefi, S. Eosinophil and neutrophil extracellular DNA traps in human allergic asthmatic airways. J. Allergy Clin. Immunol. 2011, 127, 1260–1266. [Google Scholar] [CrossRef] [Green Version]
  206. Choi, Y.; Kim, Y.M.; Lee, H.R.; Mun, J.; Sim, S.; Lee, D.H.; Pham, D.L.; Kim, S.H.; Shin, Y.S.; Lee, S.W.; et al. Eosinophil extracellular traps activate type 2 innate lymphoid cells through stimulating airway epithelium in severe asthma. Allergy 2020, 75, 95–103. [Google Scholar] [CrossRef]
  207. Nakagome, K.; Nagata, M. Involvement and Possible Role of Eosinophils in Asthma Exacerbation. Front. Immunol. 2018, 9. [Google Scholar] [CrossRef]
  208. Choi, Y.; Le Pham, D.; Lee, D.H.; Lee, S.H.; Kim, S.H.; Park, H.S. Biological function of eosinophil extracellular traps in patients with severe eosinophilic asthma. Exp. Mol. Med. 2018, 50, 104. [Google Scholar] [CrossRef] [Green Version]
  209. Esnault, S.; Leet, J.P.; Johansson, M.W.; Barretto, K.T.; Fichtinger, P.S.; Fogerty, F.J.; Bernau, K.; Mathur, S.K.; Mosher, D.F.; Sandbo, N.; et al. Eosinophil cytolysis on Immunoglobulin G is associated with microtubule formation and suppression of rho-associated protein kinase signalling. Clin. Exp. Allergy 2020, 50, 198–212. [Google Scholar] [CrossRef]
  210. Marx, C.; Novotny, J.; Salbeck, D.; Zellner, K.R.; Nicolai, L.; Pekayvaz, K.; Kilani, B.; Stockhausen, S.; Bürgener, N.; Kupka, D.; et al. Eosinophil-platelet interactions promote atherosclerosis and stabilize thrombosis with eosinophil extracellular traps. Blood 2019, 134, 1859–1872. [Google Scholar] [CrossRef] [Green Version]
  211. Pertiwi, K.R.; de Boer, O.J.; Mackaaij, C.; Pabittei, D.R.; de Winter, R.J.; Li, X.; van der Wal, A.C. Extracellular traps derived from macrophages, mast cells, eosinophils and neutrophils are generated in a time-dependent manner during atherothrombosis. J. Pathol. 2019, 247, 505–512. [Google Scholar] [CrossRef] [Green Version]
  212. Sakurai, Y.; Morioke, S.; Takeda, T.; Takahagi, S.; Hide, M.; Shima, M. Increased thrombin generation potential in patients with chronic spontaneous urticaria. Allergol. Int. 2015, 64, 96–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Simon, D.; Hoesli, S.; Roth, N.; Staedler, S.; Yousefi, S.; Simon, H.U. Eosinophil extracellular DNA traps in skin diseases. J. Allergy Clin. Immunol. 2011, 127, 194–199. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Involvement of NETs and EETs in diseases. The figure compares neutrophils and eosinophils as well as the extracellular traps they form. The right-hand side table summarizes autoimmune, autoinflammatory, and infectious diseases associated with the formation of NETs or EETs. The table also includes the ability of bacteria to modulate extracellular traps formation.
Figure 1. Involvement of NETs and EETs in diseases. The figure compares neutrophils and eosinophils as well as the extracellular traps they form. The right-hand side table summarizes autoimmune, autoinflammatory, and infectious diseases associated with the formation of NETs or EETs. The table also includes the ability of bacteria to modulate extracellular traps formation.
Microorganisms 08 01925 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Williams, T.L.; Rada, B.; Tandon, E.; Gestal, M.C. “NETs and EETs, a Whole Web of Mess”. Microorganisms 2020, 8, 1925. https://doi.org/10.3390/microorganisms8121925

AMA Style

Williams TL, Rada B, Tandon E, Gestal MC. “NETs and EETs, a Whole Web of Mess”. Microorganisms. 2020; 8(12):1925. https://doi.org/10.3390/microorganisms8121925

Chicago/Turabian Style

Williams, Tyler L., Balázs Rada, Eshaan Tandon, and Monica C. Gestal. 2020. "“NETs and EETs, a Whole Web of Mess”" Microorganisms 8, no. 12: 1925. https://doi.org/10.3390/microorganisms8121925

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

Article Metrics

Back to TopTop