Next Article in Journal
Premature Macrophage Activation by Stored Red Blood Cell Transfusion Halts Liver Regeneration Post-Partial Hepatectomy in Rats
Next Article in Special Issue
Timing of Interleukin-4 Stimulation of Macrophages Determines Their Anti-Microbial Activity during Infection with Salmonella enterica Serovar Typhimurium
Previous Article in Journal
Arginine Reduces Glycation in γ2 Subunit of AMPK and Pathologies in Alzheimer’s Disease Model Mice
Previous Article in Special Issue
Increased Adipose Tissue Expression of IL-23 Associates with Inflammatory Markers in People with High LDL Cholesterol
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neutrophil Extracellular Traps in Asthma: Friends or Foes?

1
Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
2
Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
3
World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
4
Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair, Development, National Heart and Lung Institute, Imperial College, London SW7 2AZ, UK
5
Imperial College NIHR Biomedical Research Centre, Asthma UK Centre in Allergic Mechanisms of Asthma, London SW7 2AZ, UK
6
Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), 80131 Naples, Italy
7
Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College, London SW3 6LY, UK
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(21), 3521; https://doi.org/10.3390/cells11213521
Submission received: 18 October 2022 / Revised: 4 November 2022 / Accepted: 5 November 2022 / Published: 7 November 2022
(This article belongs to the Special Issue Metabolic Inflammation and Cellular Immunity)

Abstract

:
Asthma is a chronic inflammatory disease characterized by variable airflow limitation and airway hyperresponsiveness. A plethora of immune and structural cells are involved in asthma pathogenesis. The roles of neutrophils and their mediators in different asthma phenotypes are largely unknown. Neutrophil extracellular traps (NETs) are net-like structures composed of DNA scaffolds, histones and granular proteins released by activated neutrophils. NETs were originally described as a process to entrap and kill a variety of microorganisms. NET formation can be achieved through a cell-death process, termed NETosis, or in association with the release of DNA from viable neutrophils. NETs can also promote the resolution of inflammation by degrading cytokines and chemokines. NETs have been implicated in the pathogenesis of various non-infectious conditions, including autoimmunity, cancer and even allergic disorders. Putative surrogate NET biomarkers (e.g., double-strand DNA (dsDNA), myeloperoxidase-DNA (MPO-DNA), and citrullinated histone H3 (CitH3)) have been found in different sites/fluids of patients with asthma. Targeting NETs has been proposed as a therapeutic strategy in several diseases. However, different NETs and NET components may have alternate, even opposite, consequences on inflammation. Here we review recent findings emphasizing the pathogenic and therapeutic potential of NETs in asthma.

1. Introduction

Asthma is a chronic inflammatory airway disease characterized by variable airflow limitation and airway hyperresponsiveness [1,2]. Asthma can be classified into two subgroups based on the type of inflammation: T (type) 2-high and T2-low [3]. T2-high asthma is the most frequent endophenotype, characterized by the activation of T2 helper cells (Th2), T2 innate lymphoid cells (ILC2), mast cells, basophils, and eosinophils, as well as the production of T2 cytokines (e.g., IL-3, IL-4, IL-5, and IL-13) [4,5,6]. In T2-high and T2-low asthma, several environmental (e.g., respiratory viruses, bacteria, fungi and cigarette smoke) and endogenous stimuli (e.g., cytokines and chemokines) can induce the release of epithelium-derived alarmins (i.e., thymic stromal lymphopoietin (TSLP), IL-33, and IL-25), which activate both innate and adaptive immune cells [4,7]. T2-low asthma biomarkers are currently lacking and represent an unmet need. Thus, T2-low asthma has a diagnosis of exclusion identified/characterized by the absolute and/or relative absence of T2 biomarkers [8].
Neutrophils, also called polymorphonuclear leukocytes (PMNs), are the first line of defense in fighting infections and maintaining tissue homeostasis [9]. PMNs account for approximately 65% of all leukocytes in humans and 10–20% in mice [10,11]. It has been estimated that humans produce ≅1 billion neutrophils per kilogram of body weight every day, and this may increase to 10 billion during inflammation [12]. Among their primary functions are degranulation, phagocytosis, and neutrophil extracellular trap (NET) release [13,14].
NETs were originally described as a defense mechanism to entrap and kill invading microorganisms [15]. During the last decades, NETs have been implicated in various conditions such as cancer [16,17,18,19], cardiovascular diseases [20,21,22], and autoimmune [19,23,24,25] and respiratory disorders [26]. A growing body of literature has emerged exploring the relationship between NETs and chronic inflammatory airway diseases, including asthma [27,28,29] and chronic obstructive pulmonary disease (COPD) [30]. In this review, we provide an overview of recent progress in understanding the formation of different types of NETs, the relationship between neutrophils and NETs in asthma, and the biomarkers associated with NETs, as well as a discussion of the possible therapeutic implications of NET inhibition in asthma.

2. Neutrophils in Inflammation

Neutrophils are fundamental actors in the host’s immune response [9]. Circulating PMNs are recruited to the site of inflammation, where they release a wide range of preformed and de novo synthesized mediators including several granular enzymes (e.g., neutrophil elastase (NE), myeloperoxidase (MPO), proteinase 3 (PR3), pentraxin 3 (PTX3)), lipid mediators (leukotriene B4), and reactive oxygen species (ROS) [31,32]. Beyond killing by degranulation and phagocytosis [33], neutrophils can use their chromatin to trap and kill microbes [12,15].
Neutrophils play an important role in the modulation of both innate and adaptive immune responses through their ability to modulate the behavior of several immune cells. These cells can act as antigen-presenting cells (APCs) [34,35,36], drive the recruitment, maturation and activation of macrophages and dendritic cells (DCs) [37,38,39,40], and release activating signals that contribute to the recruitment, activation, and differentiation of T cells [41] and B cells [42]. Therefore, neutrophils play a role in orchestrating inflammatory response [43]. At the site of inflammation, they are involved in complex and bidirectional interactions with other immune and stromal cells through the release of soluble mediators or direct cell-to-cell contact [44]. Furthermore, neutrophils seem to have more transcriptional activity than initially thought.
There is compelling evidence that human [45,46,47,48] and mouse [46,49,50,51] neutrophils express high levels of heterogeneity. Several neutrophil phenotypes have been identified in different experimental models and appear to display distinct functional properties. For example, neutrophils could undergo polarization toward antitumoral (N1) and protumoral (N2) phenotypes which differ in morphology and function [49,51,52]. There is also evidence that aged neutrophils express a certain degree of heterogeneity driven by the microbiota via toll-like receptor (TLR) activation [53].
Neutrophils can facilitate the resolution of inflammation through the release of pro-resolving lipid mediators [54,55], anti-inflammatory cytokines, such as IL-10 [56], and decoy receptors that act as scavengers of chemokines and cytokines [57,58,59,60].
Therefore, neutrophils should not be viewed as solely destructive cells, but rather their pleiotropic properties, both beneficial and detrimental, should be considered in the context of infections and environmental triggers that could also modulate the neutrophil decision-making algorithm.

3. NET Formation

NETs are net-like structures composed of DNA scaffolds associated with histones and several cytoplasmic granular proteins. Several immunologic and non-immunologic stimuli, including various bacteria [15], fungi [61] and viruses [62,63] activate human neutrophils to release NETs. Epithelial-derived alarmins (e.g., IL-33) can also activate neutrophils to produce ROS and NETs [64,65]. Similarly, TSLP stimulates eosinophils to produce other NET-like structures, known as eosinophil extracellular traps (EETs) [66].
NET formation was initially proposed to be a process of programmed cell death initiated by the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase followed by chromatin decondensation, breakdown of the nuclear membrane, and mixing of the chromatin with granular proteins [67]. This process is mediated by MPO, NE, and peptidyl arginine deiminase (PAD)-4 [68,69], which leads to the extrusion of DNA scaffolds decorated with citrullinated histones and granular proteins [70]. Further research has questioned whether all steps in this pathway are essential for the development of NETosis. Indeed, human saliva can induce NETosis independently of NADPH oxidase and NE [71]. Therefore, NET extrusion can be NADPH enzyme-dependent or independent, and the formation process can be divided into NETosis involving cell death (suicidal NETosis) and NET formation in which neutrophils remain alive (vital NET formation) [18,72,73,74]. Figure 1 schematically illustrates the biochemical events and morphological changes associated with NETosis following the activation of human neutrophils.
The release of extracellular traps can also occur without cell death [18,72,73,74,80,81]. NETs released during vital NET formation are composed of nuclear and mitochondrial DNA (mtDNA) and can be formed independently of NADH oxidase or ROS. In contrast to suicidal NETosis, vital NET formation is a rapid process occurring within 60 min and without affecting neutrophil viability [66,72,73,82]. Figure 2 schematically illustrates the fundamental mechanisms of suicidal neutrophils cytolysis (also termed NETosis) and of vital NET formation.
NET clearance is an endocytic process promoted by macrophages and DCs [83,84]. These cells are able to degrade NETs by interplaying with extracellular and secreted DNases followed by intracellular uptake [85].

4. Neutrophils in Asthma

The role of neutrophils in asthma pathobiology is complex, multifactorial, and largely unknown [2,100]. Earlier studies showed that circulating neutrophils are activated in patients with asthma during exercise [101] and following allergen inhalation challenge [102] as shown by enhanced surface complement receptor expression. Recently, we demonstrated that highly purified neutrophils isolated from the peripheral blood of patients with asthma and healthy individuals produced ROS in response to lipopolysaccharide (LPS) and N-Formyl-Met-Leu-Phe (fMLP), which activate distinct surface receptors (TLR4 and FPR, respectively) [19,103,104]. ROS production was lower in PMNs from patients with asthma compared to healthy subjects. Moreover, the mobilization of two neutrophil surface markers, CD11b and CD62L, induced by LPS and fMLP was more prominent in cells of healthy individuals compared to cells obtained from patients with asthma. The latter results are compatible with the hypothesis that circulating neutrophils from patients with asthma are chronically stimulated by numerous stimuli in vivo, which render these cells hyporesponsive to stimulation in vitro [103].
Neutrophils contain several preformed granular components that can be rapidly released [105]. Plasma concentrations of the neutrophil-derived mediators MPO, matrix metallopeptidase 9 (MMP-9) and CXCL8 were augmented in patients with asthma compared to healthy subjects [103], which suggests that PMNs are activated in vivo. MPO is a cationic enzyme located in primary azurophilic granules that catalyzes ROS production [106] and is involved in NET formation [19,68]. MPO also stabilizes NET structure by promoting the incorporation of chlorinated polyamines and the formation of covalent crosslinks [107]. Circulating levels [103] and sputum concentrations of MPO [108] were increased in patients with asthma compared to healthy individuals and correlated with extracellular DNA, a putative NET biomarker [108].
MMP-9, present within tertiary granules of neutrophils, has been detected in the bronchoalveolar lavage (BAL) of patients with asthma [109] and is associated with neutrophil counts [110]. In a murine model of asthma, MMP-9 knockout mice exhibited a reduced level of immune cell infiltration as well as decreased bronchial hyperresponsiveness compared to wild-type mice [111]. MMP-9 levels were increased in the BAL and sputum of patients with asthma after allergen challenge [111,112]. Moreover, neutrophil-derived MMP-9 was elevated in patients with severe asthma [110]. Higher levels of CXCL8 in circulation [103,113,114,115,116], sputum [117], and BAL fluid [16,118] were found in patients with asthma compared to healthy subjects.
The S100 family of calcium binding proteins is localized in the cytoplasm and/or nucleus of a wide range of cells, including neutrophils [119]. The calcium-binding protein A9 (S100A9), also known as calgranulin B, is secreted by activated neutrophils and can induce NET formation [120]. Increased levels of S100A9 were observed in the serum and sputum of patients with neutrophilic asthma, especially in those with uncontrolled disease [121,122]. S100A12, known as calgranulin C and secreted upon neutrophil activation, has also been associated with mast cell activation and allergic responses [123]. Surprisingly, S100A12 has been reported to reduce airway smooth muscle cells and dampen airway inflammation and hyperresponsiveness in a murine model of allergic lung inflammation [124].
Angiogenesis, the formation of new blood vessels, and lymphangiogenesis, the formation of new lymphatic vessels, occur in different inflammatory disorders including asthma [125,126,127] and cancer [52]. Vascular endothelial growth factors (VEGFs) are the most specific growth factors for blood and lymphatic endothelial cells [128,129]. Angiopoietins (ANGPTs) are members of another family of promoters of embryonic and post-natal neovascularization [130]. Human neutrophils can release several angiogenic factors (e.g., VEGF-A, ANGPT1) [52,131].

5. NETs in Asthma

NETs can be induced by several immunologic and non-immunologic stimuli. IL-33 [64,65], CXCL8 [18,132,133,134], IL-17 [135], C3a/C5a [72,136,137], IL-1β [99,108] and LPS [18,72,80,138], associated with asthma pathophysiology [108,139], are important inducers of NET formation. Moreover, several bacterial products (e.g., fMLP) [136,140,141], rhinovirus and influenza virus can induce NET formation [139,142].
The airway epithelium is the first line of defense for the lungs, which detects inhaled environmental threats such as allergens, superallergens, bacterial, fungal and viral products, and smoke extracts [143]. In patients with asthma, environmental and endogenous insults damage the epithelial cells, representing the first immunologic event in bronchial asthma [144,145]. Epithelial-derived cytokines [i.e., TSLP, IL-33, IL-25] act as upstream cytokines in asthma pathobiology [4,144,145,146,147]. NETs disrupt bronchial epithelial tight junctions, leading to intracellular component spillover [148]. NETs directly act on bronchial epithelial cells (BECs) to secrete inflammatory mediators that increase airway inflammation and provoke respiratory symptoms [108,149]. NET components, such as high-mobility group box 1 protein (HMBG1), can activate BECs to release several mediators involved in asthma pathogenesis, including TSLP, TNF-α, MMP-9, and VEGF [150]. Therefore, NETs could contribute to asthma pathobiology and exacerbations through the loss of integrity of the bronchial epithelium and the release of “upstream cytokines’’ such as alarmins (e.g., TSLP, IL-33).
In a murine model of asthma, Toussaint et al. found increased neutrophil numbers and NET formation on day 1 following rhinovirus exposure [139]. Akk and collaborators found that NET formation peaked two days following infection with Sendai virus, and that NETs played a critical role in the early phases of the immune response by recruiting and activating CD4+ and CD8+ T cells, increasing TNF-α and IL-6, and causing airway hyperresponsiveness [151]. NETs stimulated the presentation of antigens by DCs promoting a Th2 inflammatory response in a murine asthma model induced by LPS and house dust mites (HDM), as suggested by increased expression of T2 cytokines, eosinophil infiltration, mucus hypersecretion, and airway hyperresponsiveness (AHR) [152].
Recently, NETs have been implicated in an experimental model of air pollutant-induced asthma [153]. Diesel exhaust particles (DEPs) induced the release of adenosine triphosphate (ATP), promoting the expression of SiglecF on neutrophils. SiglecF+ PMNs produced increased NETs, cysteinyl leukotrienes (CysLTs) and ROS. Interestingly, a dual inhibitor of CysLTs and NETs reduced DEP-mediated asthma exacerbations [153]. The authors also found Siglec8+ (which corresponds to murine SiglecF+) PMNs in the sputum of patients with asthma-COPD overlap (ACO). Collectively these results indicate that early infiltration of neutrophils and the formation of NETs appear to be important factors in the initiation and development of experimental models of asthma.
NETs were identified in bronchial biopsy specimens of patients with allergic asthma [154]. NETs have also been proposed as biomarkers of asthma severity. The levels of extracellular DNA (eDNA), a NET component, were increased in sputum samples from patients with severe asthma compared to those with mild or moderate disease [27]. Plasma concentrations of dsDNA were increased in children with mild persistent asthma compared to healthy controls [155].
IL-4 and IL-13, the prototypic type 2 cytokines, signal through the activation of type 1 and type 2 IL-4Rs on human neutrophils [156]. IL-4 and IL-13 inhibited both phorbol 12-myristate 13-acetate (PMA)-induced NETosis and CXCL8-mediated chemotaxis of neutrophils. Moreover, NET formation induced by PMA in neutrophils from allergic subjects was reduced compared to healthy donors [156]. Finally, sera from allergic subjects inhibited PMA-induced NET formation and chemotaxis of healthy human neutrophils. Thus, it appears that NETs may have both protective and harmful roles in asthma pathogenesis. Moreover, these data indicate that neutrophils stimulated by Th2-like cytokines enter a state of activation or differentiation that differs from that of neutrophils from healthy subjects. Notably, PMA is a non-physiological stimulus recognized to induce NETosis. It would be interesting to evaluate whether similar phenomena are observed using stimuli relevant to the pathobiology of asthma.
Allergen immunotherapy (AIT) is a disease-modifying treatment for allergic rhinitis, allergic asthma, and Hymenoptera venom allergy [157,158]. A role for neutrophils in allergen processing and presentation to T cells has recently been demonstrated [159]. Recently, it has been demonstrated that aluminum hydroxide (alum) and monophosphoryl-lipid A (MPLA), traditional AIT adjuvants, can trigger the release of NETs from human neutrophils [160]. Moreover, vaccine formulations containing alum or MPLA can induce strong NET formation. Finally, NETs and alum can synergistically enhance the expression of molecules (i.e., CD80, CD86 and CD83) implicated in antigen presentation by peripheral blood monocytes [160]. Considering the dual effects of NETs (i.e., proinflammatory and anti-inflammatory), it will be interesting to evaluate the presence and role of NETs during AIT in patients with allergic disorders.

6. The Anti-Inflammatory Role of NETs

NETs are fundamental components of the innate immune system and play a key role in capturing and/or killing bacteria [15], viruses [161], fungi [162] and parasites [163]. Recent evidence suggests that NETs, under certain circumstances, can also have an anti-inflammatory role [9] and can modulate acute and chronic inflammation [164]. NETs have been shown to form barriers to prevent the spread of infections [165,166,167]. NETs and aggregated NETs (aggNETs) can promote the resolution of neutrophilic inflammation by degrading cytokines and chemokines, thus disrupting neutrophil recruitment and activation [168]. NETs and aggNETs also orchestrate the resolution of sterile crystal-mediated (e.g., monosodium urate, calcium pyrophosphate) inflammation [169]. Reduced NETosis leads to more severe disease in murine models of systemic lupus erythematosus (SLE) [170] and gout [168]. Degradation of NETs by DNase leads to a reduction of their antimicrobial properties [15]. NE decays virulence factors produced by Gram-negative bacteria [15]; whereas, serine proteases penetrate and destroy the bacterial membrane [171]. PTX3, a NET component, exerts antifungal activity [172], protects against extracellular histone-mediated cytotoxicity and mitigates the detrimental effect of NETs [172]. Furthermore, NETs can inhibit GM-CSF/IL-4-induced differentiation of monocytes into DCs in vitro, resulting in an alternatively activated macrophage phenotype [173]. This subset of macrophages is important in resolving and preventing chronic inflammation. Additionally, aggNETs prevent inflammation on the human neutrophil-rich ocular surface [10]. Collectively, these studies suggest that, under certain circumstances, NETs may provide platforms for degrading proinflammatory mediators that would otherwise drive inflammation.
Figure 3 schematically summarizes the proinflammatory and anti-inflammatory effects of NETs in humans and experimental models of inflammation.

7. Physiologic States That Influence NET Biology

Neutrophil biology is influenced by several variables in the host, such as age, sex, and circadian rhythms [9,186]. For instance, neutrophils expressing an immature phenotype are increased in young males compared to females [187,188]. Interestingly, women have an increased capacity to form NETosis [187,188]. Moreover, neutrophil phenotype is altered during pregnancy as estrogen levels increase [189]. In both males and females, neutrophils are diurnally replenished and there is a distinct circadian regulation of neutrophil aging [190,191]. Neutrophil aging is modulated in part by the microbiome through the engagement of TLR4 by LPS [53], a potent stimulus for NET formation [18,72,80,138]. NET formation induced by inflammatory stimuli is impaired in older individuals compared to young adults [192]. Although age [193,194] and sex [194,195,196] can affect asthma prevalence and/or severity, the relationship between these factors and NET formation remains unexplored. From a practical point of view, these findings suggest that studies on the role of NETs in asthma should take into account age, sex, and the timing of blood and other tissue sampling.

8. NET Biomarkers

In recent decades, strong efforts have been made to identify reliable biomarkers of different phenotypes of asthma, such as T2-high and T2-low asthma [197,198,199]. In particular, biomarkers to assess non-type 2 asthma still represent an unmet need [200]. There is potential for NETs to become novel biomarkers of asthma progression or severity as well as therapeutic targets. A variety of different analytical approaches (e.g., double-strand DNA (dsDNA), myeloperoxidase-DNA (MPO-DNA), and citrullinated histone H3 (CitH3)) have been used as a surrogate measurement of NETs in asthma [27,103,108,139,201,202,203]. Putative NET biomarkers have been found in different sites/biological fluids of patients with asthma. The presence of DNA was initially detected in bronchial biopsies of patients with asthma [154]. Additionally, dsDNA has been found in the sputum [27,108], nasal lavage [139] and BAL of patients with asthma [179,202]. The measurement of dsDNA has been widely used as a NET biomarker [27,108,139]. However, the quantitative determination of circulating DNA does not necessarily reflect the concentration of NETs in vivo. For instance, DNA complexes may result from other forms of cell death related to neutrophilic inflammation [201,204]. MPO-DNA has been proposed as a circulating biomarker of NETs in severe asthma [202]. However, this assay is error-prone and does not offer complete information about NETs in vivo [203]. We demonstrated that plasma concentrations of two NET components, CitH3 and dsDNA, were increased in asthma patients compared to healthy donors and inversely correlated with the percent decrease in FEV1/FVC [103].
It should be emphasized that no validated assay available today has proven to be clinically useful. Indeed, there is a lack of standardization in the measurement of putative NET biomarkers as well as problems related to sensitivity and specificity. Thus, there are no currently accepted values for these parameters in relation to NETs in asthma or in other diseases. In conclusion, the evaluation of different NET biomarkers in different phenotypes of asthma (alone or in combination with other parameters) and during acute asthma exacerbations requires further study.

9. Future Perspective: NET Inhibitors

Inhibition of NET formation might represent a possible target for treating certain endophenotypes of asthma. A variety of inhibitors that prevent NET formation, as well as molecules that degrade NETs, are currently under investigation for the treatment of cancer and inflammatory diseases [19]. Recombinant human protease inhibitors (e.g., NE inhibitors) and DNase can neutralize and degrade NET-derived DNA and mediators, reducing their proinflammatory effects [205]. DNase, degrading chromatin within the NETs, is a promising strategy to interfere with NET formation and activity [52,206]. Anti-histone antibodies also have some proven efficacy in treating autoimmune disorders [207]. PAD4 appears to be an interesting target in several mouse models to reduce NET-mediated inflammation. PAD inhibitor Cl-amidine also inhibits histone citrullination, which is a pivotal step in NETosis [207]. However, NETosis may not always depend on PAD4 [208] and PAD4 inhibitor efficacy may have species-specific variations [209].
The blockade of NE has also been shown to inhibit NET-induced disruption of endothelial cell–cell integrity [210]. Other molecules are being examined for their potential to inhibit NETosis. The inhibition of NADPH oxidase is effective in preventing suicidal NETosis in vitro. However, studies on experimental murine models of SLE [170] and gout [168], both of which lack NADPH oxidase, have shown more severe disease. Similarly, patients with chronic granulomatous disease, who have defects in NADPH oxidase, have a greater incidence of autoimmune disorders [211].
It should be pointed out that although these pharmacologic strategies have advantages in the treatment of possible NET-mediated diseases, complete NET inhibition in animal models increases susceptibility to infections and decreases neutrophil functions involved in innate immunity [212,213]. There is also evidence that ROS signaling and NET presence are important to modulate inflammation; whereas, excessive NETs can actually be responsible for disease. Several compounds have been tested for their ability to limit NETosis without interfering with ROS production in vitro. An example of such a compound is tetrahydroisoquinoline [214], which inhibits both spontaneous and PMA-induced NETosis in neutrophils from SLE patients. Further studies and comprehension of the regulation and balance of NET formation, inhibition, and degradation using NET inhibitors will be mandatory to avoid compromising the patient’s immune system [205].

10. Extracellular Traps (ETs) from Other Immune Cells

Compelling evidence indicates that several immune cells (i.e., eosinophils, mast cells, macrophages, and basophils) and their mediators contribute to airway inflammation in different asthma phenotypes [215,216,217]. The first description of extracellular traps (ETs) from neutrophils occurred in 2004 [15]. Since then, ETs have been identified in several other cells of the innate immune system, including eosinophils [218,219], basophils [66,220,221], mast cells [222,223,224,225,226,227], and macrophages [228,229,230]. Therefore, the term ETosis has been coined as a conventional/generic term for immune cells releasing ETs. Increasing evidence suggests that a single cell type can display multiple ET formation mechanisms and that different immune cells can display similar ET formation mechanisms [97,231,232,233].
Eosinophil extracellular traps (EETs) are composed of a meshwork of DNA and eosinophil granule proteins, such as major basic protein (MBP) and eosinophil cationic protein (ECP) [201,219]. Notably, the release of mtDNA by eosinophils occurs rapidly in a catapult-like manner [182]. Eosinophils and their powerful mediators play a key role in the pathogenesis of allergic and eosinophilic asthma [234]. EETs can be released by activated eosinophils contributing to airway inflammation in severe asthma [218]. Vital and suicide EETs have also been associated with other allergic eosinophilic diseases, including chronic rhinosinusitis with nasal polyps and eosinophilic esophagitis [154,235,236,237,238].
Macrophage extracellular traps (METs) contain DNA (mtDNA and/or nuclear DNA) but also CitH3, MPO and lysozyme [228,239]. Macrophages are the predominant immune cells in the human lung and release a plethora of proinflammatory mediators that are involved in asthma. [240,241]. Several infectious and non-infectious stimuli can induce the release of METs [220,229,230].
Mast cell extracellular traps (MCETs) are composed of nuclear DNA, histones, tryptase, and antimicrobial peptides such as cathelicidins [224,225,227,242]. Mast cells and their mediators play a key role in the pathobiology of different asthma phenotypes [215,243,244]. MCETs are released from mast cells in response to a variety of immunologic (e.g., anti-IgE, substance P) [245] and chemical stimuli (e.g., H2O2, PMA) [242], and to various pathogens including bacteria, fungi, viruses and protozoa [222,223,224,225,226,227].
Even though basophils account for ≅1% of circulating leukocytes, these cells can play critical roles in the activation of type 2 immune responses in allergic asthma [6,52,246]. Basophil extracellular traps (BETs) are released within a few minutes of stimulation by IgE, chemokines, cytokines, TLRs, monosodium urate crystals and lipid mediators [220,221]. BETs are composed of mtDNA and are formed by a mtROS-dependent and NADPH oxidase-independent mechanism [66]. Further studies in experimental models and using human biological materials will be necessary to verify the involvement and the effects of different ETs in asthma pathobiology.

11. Conclusions and Future Perspectives

Asthma is a complex and heterogeneous disease with variable clinical manifestations [247,248,249]. The role of NETs or possibly other forms of ETosis (e.g., EETs, METs, MCETs, BETs) in different asthma endophenotypes is still largely unknown. The discovery of NETs has changed the paradigm of neutrophil-mediated innate immunity. Although NET formation is now recognized as a powerful antimicrobial defense system [250], its dysregulation may contribute to chronic airway inflammation. There is experimental evidence in preclinical models that NET formation is also implicated in asthma exacerbations and that a unique population of SiglecF+ PMNs is involved [153]. The role of Siglec8+ neutrophils and NET formation should also be investigated during asthma exacerbations.
NETopathic inflammation can be avoided or reversed by inhibiting NET formation, blocking the granular proteins decorating NETs, or clearing NETs that have already been released. Inhibition of NET formation has started to be investigated in experimental models of asthma. Vargas et al. found that glucocorticoids decreased NET formation in vitro and in vivo in the lungs of asthmatic horses [251]. Inhibition of NE, a NET component, can attenuate rhinovirus-induced airway hyperreactivity in a mouse model of asthma [139]. Patients with asthma on inhaled glucocorticoid (ICS) treatment showed lower circulating NET levels compared to patients who did not or just occasionally used ICS [252]. Future studies should investigate whether pharmacological or biological therapies for asthma could modify the expression of NETs and their circulating biomarkers in vivo.
Despite the growing evidence, important questions remain unanswered. Table 1 summarizes some limitations in studying NETs in asthma. It remains unclear whether NETs are a double-edged sword in asthma. It is not known whether or how NET formation contributes to the pathogenesis of asthma or whether stabilizing this process could be an effective therapeutic strategy. It is also crucial to consider that NETs, and presumably other forms of ET, play an important role in host defense. Therefore, therapeutic intervention should be cautiously targeted to modulate the potential of NETs toward maintaining homeostasis rather than attempting to neutralize them completely. In conclusion, we anticipate that a better knowledge of the biochemical and immunological features of NETopathic inflammation could lead to the development of innovative and personalized therapies for specific asthma endophenotypes.

Author Contributions

Conceptualization, R.P., M.S., G.M. and G.V; writing—original draft preparation, R.P., M.S., G.M., S.R.D. and G.V.; writing—review and editing, R.P., M.S., G.M., S.R.D., G.W.S. and G.V.; funding acquisition, G.M. and G.V. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by grants from the CISI-Lab Project (University of Naples Federico II) and TIMING Project and Campania Bioscience (Regione Campania).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Gjada Criscuolo for her excellent managerial assistance in preparing this manuscript and the administrative staff (Roberto Bifulco, Anna Ferraro and Maria Cristina Fucci), without whom it would not be possible to work as a team.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACO, asthma-COPD overlap; aggNET, aggregated NET; AIT, allergen immunotherapy; alum, aluminum hydroxide; AMP, antimicrobial peptide; ANGPT1, angiopoietin-1; APC, antigen-presenting cell; ATP, adenosine triphosphate; BAL, bronchoalveolar lavage; BEC, bronchial epithelial cell; BET, basophil extracellular trap; CitH3, citrullinated histone H3; COPD, chronic obstructive pulmonary disease; CysLT, cysteinyl leukotriene; DAMP, Damage-associated molecular pattern; DC, dendritic cell; DEP, diesel exhaust particle; dsDNA, double strand DNA; E. coli, Escherichia coli; ECM, extracellular matrix; ECP, eosinophil cationic protein; eDNA, extracellular DNA; EET, eosinophil extracellular trap; ET, extracellular trap; FEV1, forced expired volume; fMLP, N-formyl peptide; FVC, forced vital capacity; HDM, house dust mite; HMBG1, high mobility group box 1 protein; ICS, inhaled glucocorticoid; ILC2, innate lymphoid cells type 2; LPS, lipopolysaccharide; MBP, major basic protein; MCET, mast cell extracellular trap; MET, macrophage extracellular trap; MMP9, matrix metallopeptidase 9; mtDNA, mitochondrial DNA; MPLA, monophosphoryl-lipid A; MPO, myeloperoxidase; MPO-DNA, myeloperoxidase-DNA; mROS, mitochondrial ROS; NADPH, nicotinamide adenine dinucleotide phosphate; NE, neutrophils elastase; NET, neutrophil extracellular trap; PAD, peptidyl arginine deiminase; PKC, protein kinase C; PMA, phorbol 12-myristate 13-acetate; PMN, polymorphonuclear leukocyte; PR3, proteinase 3; PTX3, pentraxin 3; Th2, T2 helper cell; ROS, reactive oxygen species; S. aureus, Staphylococcus aureus; SLE, systemic lupus erythematosus, TSLP, thymic stromal lymphopoietin; VEGF, vascular endothelial growth factor.

References

  1. Khalfaoui, L.; Symon, F.A.; Couillard, S.; Hargadon, B.; Chaudhuri, R.; Bicknell, S.; Mansur, A.H.; Shrimanker, R.; Hinks, T.S.C.; Pavord, I.D.; et al. Airway remodelling rather than cellular infiltration characterizes both type2 cytokine biomarker-high and -low severe asthma. Allergy 2022, 77, 2974. [Google Scholar] [CrossRef] [PubMed]
  2. Al Heialy, S.; Ramakrishnan, R.K.; Hamid, Q. Recent advances in the immunopathogenesis of severe asthma. J. Allergy Clin. Immunol. 2022, 149, 455–465. [Google Scholar] [CrossRef] [PubMed]
  3. Tabatabaian, F.; Ledford, D.K.; Casale, T.B. Biologic and New Therapies in Asthma. Immunol. Allergy Clin. N. Am. 2017, 37, 329–343. [Google Scholar] [CrossRef] [PubMed]
  4. Marone, G.; Spadaro, G.; Braile, M.; Poto, R.; Criscuolo, G.; Pahima, H.; Loffredo, S.; Levi-Schaffer, F.; Varricchi, G. Tezepelumab: A novel biological therapy for the treatment of severe uncontrolled asthma. Expert Opin. Investig. Drugs 2019, 28, 931–940. [Google Scholar] [CrossRef]
  5. Pepper, A.N.; Renz, H.; Casale, T.B.; Garn, H. Biologic Therapy and Novel Molecular Targets of Severe Asthma. J. Allergy Clin. Immunol. Pract. 2017, 5, 909–916. [Google Scholar] [CrossRef]
  6. Marone, G.; Borriello, F.; Varricchi, G.; Genovese, A.; Granata, F. Basophils: Historical reflections and perspectives. Chem. Immunol. Allergy 2014, 100, 172–192. [Google Scholar] [CrossRef]
  7. Bel, E.H.; Brinke, A.T. New Anti-Eosinophil Drugs for Asthma and COPD: Targeting the Trait! Chest 2017, 152, 1276–1282. [Google Scholar] [CrossRef]
  8. Kaur, R.; Chupp, G. Phenotypes and endotypes of adult asthma: Moving toward precision medicine. J. Allergy Clin. Immunol. 2019, 144, 1–12. [Google Scholar] [CrossRef]
  9. Quail, D.F.; Amulic, B.; Aziz, M.; Barnes, B.J.; Eruslanov, E.; Fridlender, Z.G.; Goodridge, H.S.; Granot, Z.; Hidalgo, A.; Huttenlocher, A.; et al. Neutrophil phenotypes and functions in cancer: A consensus statement. J. Exp. Med. 2022, 219, e20220011. [Google Scholar] [CrossRef] [PubMed]
  10. Mestas, J.; Hughes, C.C. Of mice and not men: Differences between mouse and human immunology. J. Immunol. 2004, 172, 2731–2738. [Google Scholar] [CrossRef]
  11. McKenna, E.; Mhaonaigh, A.U.; Wubben, R.; Dwivedi, A.; Hurley, T.; Kelly, L.A.; Stevenson, N.J.; Little, M.A.; Molloy, E.J. Neutrophils: Need for Standardized Nomenclature. Front. Immunol. 2021, 12, 602963. [Google Scholar] [CrossRef] [PubMed]
  12. 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, eaat4579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Miralda, I.; Uriarte, S.M.; McLeish, K.R. Multiple Phenotypic Changes Define Neutrophil Priming. Front. Cell. Infect. Microbiol. 2017, 7, 217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Honda, M.; Kubes, P. Neutrophils and neutrophil extracellular traps in the liver and gastrointestinal system. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 206–221. [Google Scholar] [CrossRef]
  15. 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]
  16. Hakkim, A.; Furnrohr, B.G.; Amann, K.; Laube, B.; Abed, U.A.; Brinkmann, V.; Herrmann, M.; Voll, R.E.; Zychlinsky, A. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc. Natl. Acad. Sci. USA 2010, 107, 9813–9818. [Google Scholar] [CrossRef] [Green Version]
  17. van der Linden, M.; van den Hoogen, L.L.; Westerlaken, G.H.A.; Fritsch-Stork, R.D.E.; van Roon, J.A.G.; Radstake, T.; Meyaard, L. Neutrophil extracellular trap release is associated with antinuclear antibodies in systemic lupus erythematosus and anti-phospholipid syndrome. Rheumatology 2018, 57, 1228–1234. [Google Scholar] [CrossRef] [Green Version]
  18. Cristinziano, L.; Modestino, L.; Loffredo, S.; Varricchi, G.; Braile, M.; Ferrara, A.L.; de Paulis, A.; Antonelli, A.; Marone, G.; Galdiero, M.R. Anaplastic Thyroid Cancer Cells Induce the Release of Mitochondrial Extracellular DNA Traps by Viable Neutrophils. J. Immunol. 2020, 204, 1362–1372. [Google Scholar] [CrossRef]
  19. Cristinziano, L.; Modestino, L.; Antonelli, A.; Marone, G.; Simon, H.U.; Varricchi, G.; Galdiero, M.R. Neutrophil extracellular traps in cancer. Semin. Cancer Biol. 2022, 79, 91–104. [Google Scholar] [CrossRef]
  20. Perez-Olivares, L.; Soehnlein, O. Contemporary Lifestyle and Neutrophil Extracellular Traps: An Emerging Link in Atherosclerosis Disease. Cells 2021, 10, 1985. [Google Scholar] [CrossRef]
  21. Bonaventura, A.; Vecchie, A.; Abbate, A.; Montecucco, F. Neutrophil Extracellular Traps and Cardiovascular Diseases: An Update. Cells 2020, 9, 231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Qi, H.; Yang, S.; Zhang, L. Neutrophil Extracellular Traps and Endothelial Dysfunction in Atherosclerosis and Thrombosis. Front. Immunol. 2017, 8, 928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Berger-Achituv, S.; Brinkmann, V.; Abed, U.A.; Kuhn, L.I.; Ben-Ezra, J.; Elhasid, R.; Zychlinsky, A. A proposed role for neutrophil extracellular traps in cancer immunoediting. Front. Immunol. 2013, 4, 48. [Google Scholar] [CrossRef] [Green Version]
  24. Gregory, A.D.; Houghton, A.M. Tumor-associated neutrophils: New targets for cancer therapy. Cancer Res. 2011, 71, 2411–2416. [Google Scholar] [CrossRef] [Green Version]
  25. Cools-Lartigue, J.; Spicer, J.; Najmeh, S.; Ferri, L. Neutrophil extracellular traps in cancer progression. Cell. Mol. Life Sci. 2014, 71, 4179–4194. [Google Scholar] [CrossRef] [PubMed]
  26. Jorch, S.K.; Kubes, P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat. Med. 2017, 23, 279–287. [Google Scholar] [CrossRef]
  27. Wright, T.K.; Gibson, P.G.; Simpson, J.L.; McDonald, V.M.; Wood, L.G.; Baines, K.J. Neutrophil extracellular traps are associated with inflammation in chronic airway disease. Respirology 2016, 21, 467–475. [Google Scholar] [CrossRef]
  28. Chen, F.; Yu, M.; Zhong, Y.; Wang, L.; Huang, H. Characteristics and Role of Neutrophil Extracellular Traps in Asthma. Inflammation 2022, 45, 6–13. [Google Scholar] [CrossRef]
  29. Porto, B.N.; Stein, R.T. Neutrophil Extracellular Traps in Pulmonary Diseases: Too Much of a Good Thing? Front. Immunol. 2016, 7, 311. [Google Scholar] [CrossRef] [Green Version]
  30. Poto, R.; Loffredo, S.; Palestra, F.; Marone, G.; Patella, V.; Varricchi, G. Angiogenesis, Lymphangiogenesis, and Inflammation in Chronic Obstructive Pulmonary Disease (COPD): Few Certainties and Many Outstanding Questions. Cells 2022, 11, 1720. [Google Scholar] [CrossRef]
  31. Nathan, C. Immunology. Catalytic antibody bridges innate and adaptive immunity. Science 2002, 298, 2143–2144. [Google Scholar] [CrossRef] [PubMed]
  32. Borregaard, N. Neutrophils, from marrow to microbes. Immunity 2010, 33, 657–670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Nauseef, W.M. Neutrophils, from cradle to grave and beyond. Immunol. Rev. 2016, 273, 5–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Beauvillain, C.; Delneste, Y.; Scotet, M.; Peres, A.; Gascan, H.; Guermonprez, P.; Barnaba, V.; Jeannin, P. Neutrophils efficiently cross-prime naive T cells in vivo. Blood 2007, 110, 2965–2973. [Google Scholar] [CrossRef]
  35. Abi Abdallah, D.S.; Egan, C.E.; Butcher, B.A.; Denkers, E.Y. Mouse neutrophils are professional antigen-presenting cells programmed to instruct Th1 and Th17 T-cell differentiation. Int. Immunol. 2011, 23, 317–326. [Google Scholar] [CrossRef] [Green Version]
  36. Chtanova, T.; Schaeffer, M.; Han, S.J.; van Dooren, G.G.; Nollmann, M.; Herzmark, P.; Chan, S.W.; Satija, H.; Camfield, K.; Aaron, H.; et al. Dynamics of neutrophil migration in lymph nodes during infection. Immunity 2008, 29, 487–496. [Google Scholar] [CrossRef] [Green Version]
  37. Chertov, O.; Ueda, H.; Xu, L.L.; Tani, K.; Murphy, W.J.; Wang, J.M.; Howard, O.M.; Sayers, T.J.; Oppenheim, J.J. Identification of human neutrophil-derived cathepsin G and azurocidin/CAP37 as chemoattractants for mononuclear cells and neutrophils. J. Exp. Med. 1997, 186, 739–747. [Google Scholar] [CrossRef]
  38. Bennouna, S.; Bliss, S.K.; Curiel, T.J.; Denkers, E.Y. Cross-talk in the innate immune system: Neutrophils instruct recruitment and activation of dendritic cells during microbial infection. J. Immunol. 2003, 171, 6052–6058. [Google Scholar] [CrossRef] [Green Version]
  39. Wittamer, V.; Bondue, B.; Guillabert, A.; Vassart, G.; Parmentier, M.; Communi, D. Neutrophil-mediated maturation of chemerin: A link between innate and adaptive immunity. J. Immunol. 2005, 175, 487–493. [Google Scholar] [CrossRef] [Green Version]
  40. Bennouna, S.; Denkers, E.Y. Microbial antigen triggers rapid mobilization of TNF-alpha to the surface of mouse neutrophils transforming them into inducers of high-level dendritic cell TNF-alpha production. J. Immunol. 2005, 174, 4845–4851. [Google Scholar] [CrossRef]
  41. Pelletier, M.; Maggi, L.; Micheletti, A.; Lazzeri, E.; Tamassia, N.; Costantini, C.; Cosmi, L.; Lunardi, C.; Annunziato, F.; Romagnani, S.; et al. Evidence for a cross-talk between human neutrophils and Th17 cells. Blood 2010, 115, 335–343. [Google Scholar] [CrossRef] [PubMed]
  42. Scapini, P.; Bazzoni, F.; Cassatella, M.A. Regulation of B-cell-activating factor (BAFF)/B lymphocyte stimulator (BLyS) expression in human neutrophils. Immunol. Lett. 2008, 116, 1–6. [Google Scholar] [CrossRef] [PubMed]
  43. Burn, G.L.; Foti, A.; Marsman, G.; Patel, D.F.; Zychlinsky, A. The Neutrophil. Immunity 2021, 54, 1377–1391. [Google Scholar] [CrossRef]
  44. Nemeth, T.; Sperandio, M.; Mocsai, A. Neutrophils as emerging therapeutic targets. Nat. Rev. Drug Discov. 2020, 19, 253–275. [Google Scholar] [CrossRef] [PubMed]
  45. Tak, T.; Wijten, P.; Heeres, M.; Pickkers, P.; Scholten, A.; Heck, A.J.R.; Vrisekoop, N.; Leenen, L.P.; Borghans, J.A.M.; Tesselaar, K.; et al. Human CD62L(dim) neutrophils identified as a separate subset by proteome profiling and in vivo pulse-chase labeling. Blood 2017, 129, 3476–3485. [Google Scholar] [CrossRef] [Green Version]
  46. Veglia, F.; Hashimoto, A.; Dweep, H.; Sanseviero, E.; De Leo, A.; Tcyganov, E.; Kossenkov, A.; Mulligan, C.; Nam, B.; Masters, G.; et al. Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor-bearing mice. J. Exp. Med. 2021, 218, e20201803. [Google Scholar] [CrossRef]
  47. Xie, X.; Shi, Q.; Wu, P.; Zhang, X.; Kambara, H.; Su, J.; Yu, H.; Park, S.Y.; Guo, R.; Ren, Q.; et al. Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat. Immunol. 2020, 21, 1119–1133. [Google Scholar] [CrossRef]
  48. Zilionis, R.; Engblom, C.; Pfirschke, C.; Savova, V.; Zemmour, D.; Saatcioglu, H.D.; Krishnan, I.; Maroni, G.; Meyerovitz, C.V.; Kerwin, C.M.; et al. Single-Cell Transcriptomics of Human and Mouse Lung Cancers Reveals Conserved Myeloid Populations across Individuals and Species. Immunity 2019, 50, 1317–1334.e10. [Google Scholar] [CrossRef]
  49. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [Green Version]
  50. Hua, X.; Hu, G.; Hu, Q.; Chang, Y.; Hu, Y.; Gao, L.; Chen, X.; Yang, P.C.; Zhang, Y.; Li, M.; et al. Single-Cell RNA Sequencing to Dissect the Immunological Network of Autoimmune Myocarditis. Circulation 2020, 142, 384–400. [Google Scholar] [CrossRef]
  51. Sagiv, J.Y.; Michaeli, J.; Assi, S.; Mishalian, I.; Kisos, H.; Levy, L.; Damti, P.; Lumbroso, D.; Polyansky, L.; Sionov, R.V.; et al. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 2015, 10, 562–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Poto, R.; Cristinziano, L.; Modestino, L.; de Paulis, A.; Marone, G.; Loffredo, S.; Galdiero, M.R.; Varricchi, G. Neutrophil Extracellular Traps, Angiogenesis and Cancer. Biomedicines 2022, 10, 431. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, D.; Chen, G.; Manwani, D.; Mortha, A.; Xu, C.; Faith, J.J.; Burk, R.D.; Kunisaki, Y.; Jang, J.E.; Scheiermann, C.; et al. Neutrophil ageing is regulated by the microbiome. Nature 2015, 525, 528–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Serhan, C.N. Novel omega–3-derived local mediators in anti-inflammation and resolution. Pharmacol. Ther. 2005, 105, 7–21. [Google Scholar] [CrossRef]
  55. Serhan, C.N.; Savill, J. Resolution of inflammation: The beginning programs the end. Nat. Immunol. 2005, 6, 1191–1197. [Google Scholar] [CrossRef]
  56. De Santo, C.; Arscott, R.; Booth, S.; Karydis, I.; Jones, M.; Asher, R.; Salio, M.; Middleton, M.; Cerundolo, V. Invariant NKT cells modulate the suppressive activity of IL-10-secreting neutrophils differentiated with serum amyloid A. Nat. Immunol. 2010, 11, 1039–1046. [Google Scholar] [CrossRef] [Green Version]
  57. Ariel, A.; Fredman, G.; Sun, Y.P.; Kantarci, A.; Van Dyke, T.E.; Luster, A.D.; Serhan, C.N. Apoptotic neutrophils and T cells sequester chemokines during immune response resolution through modulation of CCR5 expression. Nat. Immunol. 2006, 7, 1209–1216. [Google Scholar] [CrossRef] [Green Version]
  58. McKimmie, C.S.; Fraser, A.R.; Hansell, C.; Gutierrez, L.; Philipsen, S.; Connell, L.; Rot, A.; Kurowska-Stolarska, M.; Carreno, P.; Pruenster, M.; et al. Hemopoietic cell expression of the chemokine decoy receptor D6 is dynamic and regulated by GATA1. J. Immunol. 2008, 181, 8171–8181. [Google Scholar] [CrossRef] [Green Version]
  59. Bourke, E.; Cassetti, A.; Villa, A.; Fadlon, E.; Colotta, F.; Mantovani, A. IL-1 beta scavenging by the type II IL-1 decoy receptor in human neutrophils. J. Immunol. 2003, 170, 5999–6005. [Google Scholar] [CrossRef] [Green Version]
  60. Bazzoni, F.; Tamassia, N.; Rossato, M.; Cassatella, M.A. Understanding the molecular mechanisms of the multifaceted IL-10-mediated anti-inflammatory response: Lessons from neutrophils. Eur. J. Immunol. 2010, 40, 2360–2368. [Google Scholar] [CrossRef]
  61. Urban, C.F.; Reichard, U.; Brinkmann, V.; Zychlinsky, A. Neutrophil extracellular traps capture and kill Candida albicans yeast and hyphal forms. Cell Microbiol. 2006, 8, 668–676. [Google Scholar] [CrossRef] [PubMed]
  62. 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 Microbe 2012, 12, 109–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zuo, Y.; Yalavarthi, S.; Shi, H.; Gockman, K.; Zuo, M.; Madison, J.A.; Blair, C.; Weber, A.; Barnes, B.J.; Egeblad, M.; et al. Neutrophil extracellular traps in COVID-19. JCI Insight 2020, 5, e138999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Jin, R.; Xu, J.; Gao, Q.; Mao, X.; Yin, J.; Lu, K.; Guo, Y.; Zhang, M.; Cheng, R. IL-33-induced neutrophil extracellular traps degrade fibronectin in a murine model of bronchopulmonary dysplasia. Cell Death Discov. 2020, 6, 33. [Google Scholar] [CrossRef]
  65. 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, 106256. [Google Scholar] [CrossRef]
  66. Morshed, M.; Hlushchuk, R.; Simon, D.; Walls, A.F.; Obata-Ninomiya, K.; Karasuyama, H.; Djonov, V.; Eggel, A.; Kaufmann, T.; Simon, H.U.; et al. NADPH oxidase-independent formation of extracellular DNA traps by basophils. J. Immunol. 2014, 192, 5314–5323. [Google Scholar] [CrossRef] [Green Version]
  67. 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]
  68. Papayannopoulos, V.; Metzler, K.D.; Hakkim, A.; Zychlinsky, A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J. Cell Biol. 2010, 191, 677–691. [Google Scholar] [CrossRef] [Green Version]
  69. 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]
  70. Raftery, M.J.; Lalwani, P.; Krautkrmer, E.; Peters, T.; Scharffetter-Kochanek, K.; Kruger, R.; Hofmann, J.; Seeger, K.; Kruger, D.H.; Schonrich, G. beta2 integrin mediates hantavirus-induced release of neutrophil extracellular traps. J. Exp. Med. 2014, 211, 1485–1497. [Google Scholar] [CrossRef]
  71. Mohanty, T.; Sjogren, J.; Kahn, F.; Abu-Humaidan, A.H.; Fisker, N.; Assing, K.; Morgelin, M.; Bengtsson, A.A.; Borregaard, N.; Sorensen, O.E. A novel mechanism for NETosis provides antimicrobial defense at the oral mucosa. Blood 2015, 126, 2128–2137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. 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] [PubMed]
  73. Pilsczek, F.H.; Salina, D.; Poon, K.K.; Fahey, C.; Yipp, B.G.; Sibley, C.D.; Robbins, S.M.; Green, F.H.; Surette, M.G.; Sugai, M.; et al. A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J. Immunol. 2010, 185, 7413–7425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Yipp, B.G.; Kubes, P. NETosis: How vital is it? Blood 2013, 122, 2784–2794. [Google Scholar] [CrossRef] [PubMed]
  75. Kenny, E.F.; Herzig, A.; Kruger, 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]
  76. Metzler, K.D.; Fuchs, T.A.; Nauseef, W.M.; Reumaux, D.; Roesler, J.; Schulze, I.; Wahn, V.; Papayannopoulos, V.; Zychlinsky, A. Myeloperoxidase is required for neutrophil extracellular trap formation: Implications for innate immunity. Blood 2011, 117, 953–959. [Google Scholar] [CrossRef] [Green Version]
  77. Christophorou, M.A.; Castelo-Branco, G.; Halley-Stott, R.P.; Oliveira, C.S.; Loos, R.; Radzisheuskaya, A.; Mowen, K.A.; Bertone, P.; Silva, J.C.; Zernicka-Goetz, M.; et al. Citrullination regulates pluripotency and histone H1 binding to chromatin. Nature 2014, 507, 104–108. [Google Scholar] [CrossRef] [Green Version]
  78. Sollberger, G.; Choidas, A.; Burn, G.L.; Habenberger, P.; Di Lucrezia, R.; Kordes, S.; Menninger, S.; Eickhoff, J.; Nussbaumer, P.; Klebl, B.; et al. Gasdermin D plays a vital role in the generation of neutrophil extracellular traps. Sci. Immunol. 2018, 3, eaar6689. [Google Scholar] [CrossRef] [Green Version]
  79. 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]
  80. Yipp, B.G.; Petri, B.; Salina, D.; Jenne, C.N.; Scott, B.N.; Zbytnuik, L.D.; Pittman, K.; Asaduzzaman, M.; Wu, K.; Meijndert, H.C.; et al. Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo. Nat. Med. 2012, 18, 1386–1393. [Google Scholar] [CrossRef]
  81. van Dam, L.S.; Kraaij, T.; Kamerling, S.W.A.; Bakker, J.A.; Scherer, U.H.; Rabelink, T.J.; van Kooten, C.; Teng, Y.K.O. Intrinsically Distinct Role of Neutrophil Extracellular Trap Formation in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Compared to Systemic Lupus Erythematosus. Arthritis Rheumatol. 2019, 71, 2047–2058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Rochael, N.C.; Guimaraes-Costa, A.B.; Nascimento, M.T.; DeSouza-Vieira, T.S.; Oliveira, M.P.; Garcia e Souza, L.F.; Oliveira, M.F.; Saraiva, E.M. Classical ROS-dependent and early/rapid ROS-independent release of Neutrophil Extracellular Traps triggered by Leishmania parasites. Sci. Rep. 2015, 5, 18302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Lazzaretto, B.; Fadeel, B. Intra- and Extracellular Degradation of Neutrophil Extracellular Traps by Macrophages and Dendritic Cells. J. Immunol. 2019, 203, 2276–2290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Farrera, C.; Fadeel, B. Macrophage clearance of neutrophil extracellular traps is a silent process. J. Immunol. 2013, 191, 2647–2656. [Google Scholar] [CrossRef] [Green Version]
  85. Haider, P.; Kral-Pointner, J.B.; Mayer, J.; Richter, M.; Kaun, C.; Brostjan, C.; Eilenberg, W.; Fischer, M.B.; Speidl, W.S.; Hengstenberg, C.; et al. Neutrophil Extracellular Trap Degradation by Differently Polarized Macrophage Subsets. Arter. Thromb. Vasc. Biol. 2020, 40, 2265–2278. [Google Scholar] [CrossRef]
  86. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef]
  87. Rada, B. Neutrophil Extracellular Traps and Microcrystals. J. Immunol. Res. 2017, 2017, 2896380. [Google Scholar] [CrossRef] [Green Version]
  88. Naffah de Souza, C.; Breda, L.C.D.; Khan, M.A.; de Almeida, S.R.; Camara, N.O.S.; Sweezey, N.; Palaniyar, N. Alkaline pH Promotes NADPH Oxidase-Independent Neutrophil Extracellular Trap Formation: A Matter of Mitochondrial Reactive Oxygen Species Generation and Citrullination and Cleavage of Histone. Front. Immunol. 2017, 8, 1849. [Google Scholar] [CrossRef] [Green Version]
  89. Parker, H.; Dragunow, M.; Hampton, M.B.; Kettle, A.J.; Winterbourn, C.C. Requirements for NADPH oxidase and myeloperoxidase in neutrophil extracellular trap formation differ depending on the stimulus. J. Leukoc. Biol. 2012, 92, 841–849. [Google Scholar] [CrossRef]
  90. 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]
  91. Luo, Y.; Arita, K.; Bhatia, M.; Knuckley, B.; Lee, Y.H.; Stallcup, M.R.; Sato, M.; Thompson, P.R. Inhibitors and inactivators of protein arginine deiminase 4: Functional and structural characterization. Biochemistry 2006, 45, 11727–11736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. 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, 307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Kayagaki, N.; Stowe, I.B.; Lee, B.L.; O’Rourke, K.; Anderson, K.; Warming, S.; Cuellar, T.; Haley, B.; Roose-Girma, M.; Phung, Q.T.; et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 2015, 526, 666–671. [Google Scholar] [CrossRef] [PubMed]
  94. Shi, J.; Zhao, Y.; Wang, K.; Shi, X.; Wang, Y.; Huang, H.; Zhuang, Y.; Cai, T.; Wang, F.; Shao, F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015, 526, 660–665. [Google Scholar] [CrossRef]
  95. Clark, S.R.; Ma, A.C.; Tavener, S.A.; McDonald, B.; Goodarzi, Z.; Kelly, M.M.; Patel, K.D.; Chakrabarti, S.; McAvoy, E.; Sinclair, G.D.; et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat. Med. 2007, 13, 463–469. [Google Scholar] [CrossRef] [PubMed]
  96. Amini, P.; Stojkov, D.; Felser, A.; Jackson, C.B.; Courage, C.; Schaller, A.; Gelman, L.; Soriano, M.E.; Nuoffer, J.M.; Scorrano, L.; et al. Neutrophil extracellular trap formation requires OPA1-dependent glycolytic ATP production. Nat. Commun. 2018, 9, 2958. [Google Scholar] [CrossRef] [Green Version]
  97. Lood, C.; Blanco, L.P.; Purmalek, M.M.; Carmona-Rivera, C.; De Ravin, S.S.; Smith, C.K.; Malech, H.L.; Ledbetter, J.A.; Elkon, K.B.; Kaplan, M.J. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat. Med. 2016, 22, 146–153. [Google Scholar] [CrossRef] [Green Version]
  98. Tong, M.; Potter, J.A.; Mor, G.; Abrahams, V.M. Lipopolysaccharide-Stimulated Human Fetal Membranes Induce Neutrophil Activation and Release of Vital Neutrophil Extracellular Traps. J. Immunol. 2019, 203, 500–510. [Google Scholar] [CrossRef]
  99. Meher, A.K.; Spinosa, M.; Davis, J.P.; Pope, N.; Laubach, V.E.; Su, G.; Serbulea, V.; Leitinger, N.; Ailawadi, G.; Upchurch, G.R., Jr. Novel Role of IL (Interleukin)-1beta in Neutrophil Extracellular Trap Formation and Abdominal Aortic Aneurysms. Arter. Thromb. Vasc. Biol. 2018, 38, 843–853. [Google Scholar] [CrossRef] [Green Version]
  100. Nair, P.; Surette, M.G.; Virchow, J.C. Neutrophilic asthma: Misconception or misnomer? Lancet Respir. Med. 2021, 9, 441–443. [Google Scholar] [CrossRef]
  101. Papageorgiou, N.; Carroll, M.; Durham, S.R.; Lee, T.H.; Walsh, G.M.; Kay, A.B. Complement receptor enhancement as evidence of neutrophil activation after exercise-induced asthma. Lancet 1983, 2, 1220–1223. [Google Scholar] [CrossRef]
  102. Durham, S.R.; Carroll, M.; Walsh, G.M.; Kay, A.B. Leukocyte activation in allergen-induced late-phase asthmatic reactions. N. Engl. J. Med. 1984, 311, 1398–1402. [Google Scholar] [CrossRef] [PubMed]
  103. Varricchi, G.; Modestino, L.; Poto, R.; Cristinziano, L.; Gentile, L.; Postiglione, L.; Spadaro, G.; Galdiero, M.R. Neutrophil extracellular traps and neutrophil-derived mediators as possible biomarkers in bronchial asthma. Clin. Exp. Med. 2021, 22, 285. [Google Scholar] [CrossRef] [PubMed]
  104. Braile, M.; Cristinziano, L.; Marcella, S.; Varricchi, G.; Marone, G.; Modestino, L.; Ferrara, A.L.; De Ciuceis, A.; Scala, S.; Galdiero, M.R.; et al. LPS-mediated neutrophil VEGF-A release is modulated by cannabinoid receptor activation. J. Leukoc. Biol. 2021, 109, 621–631. [Google Scholar] [CrossRef] [PubMed]
  105. 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]
  106. Aratani, Y. Myeloperoxidase: Its role for host defense, inflammation, and neutrophil function. Arch. Biochem. Biophys. 2018, 640, 47–52. [Google Scholar] [CrossRef]
  107. Csomos, K.; Kristof, E.; Jakob, B.; Csomos, I.; Kovacs, G.; Rotem, O.; Hodrea, J.; Bagoly, Z.; Muszbek, L.; Balajthy, Z.; et al. Protein cross-linking by chlorinated polyamines and transglutamylation stabilizes neutrophil extracellular traps. Cell Death Dis. 2016, 7, e2332. [Google Scholar] [CrossRef] [Green Version]
  108. Lachowicz-Scroggins, M.E.; Dunican, E.M.; Charbit, A.R.; Raymond, W.; Looney, M.R.; Peters, M.C.; Gordon, E.D.; Woodruff, P.G.; Lefrancais, 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]
  109. Wenzel, S.E.; Balzar, S.; Cundall, M.; Chu, H.W. Subepithelial basement membrane immunoreactivity for matrix metalloproteinase 9: Association with asthma severity, neutrophilic inflammation, and wound repair. J. Allergy Clin. Immunol. 2003, 111, 1345–1352. [Google Scholar] [CrossRef]
  110. Cundall, M.; Sun, Y.; Miranda, C.; Trudeau, J.B.; Barnes, S.; Wenzel, S.E. Neutrophil-derived matrix metalloproteinase-9 is increased in severe asthma and poorly inhibited by glucocorticoids. J. Allergy Clin. Immunol. 2003, 112, 1064–1071. [Google Scholar] [CrossRef]
  111. Cataldo, D.D.; Tournoy, K.G.; Vermaelen, K.; Munaut, C.; Foidart, J.M.; Louis, R.; Noel, A.; Pauwels, R.A. Matrix metalloproteinase-9 deficiency impairs cellular infiltration and bronchial hyperresponsiveness during allergen-induced airway inflammation. Am. J. Pathol. 2002, 161, 491–498. [Google Scholar] [CrossRef] [Green Version]
  112. Kelly, E.A.; Busse, W.W.; Jarjour, N.N. Increased matrix metalloproteinase-9 in the airway after allergen challenge. Am. J. Respir. Crit. Care Med. 2000, 162, 1157–1161. [Google Scholar] [CrossRef] [PubMed]
  113. Huang, A.X.; Lu, L.W.; Liu, W.J.; Huang, M. Plasma Inflammatory Cytokine IL-4, IL-8, IL-10, and TNF-alpha Levels Correlate with Pulmonary Function in Patients with Asthma-Chronic Obstructive Pulmonary Disease (COPD) Overlap Syndrome. Med. Sci. Monit. 2016, 22, 2800–2808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ding, Q.; Sun, S.; Zhang, Y.; Tang, P.; Lv, C.; Ma, H.; Yu, Y.; Xu, S.; Deng, Z. Serum IL-8 and VEGFA are Two Promising Diagnostic Biomarkers of Asthma-COPD Overlap Syndrome. Int. J. Chron. Obstruct. Pulmon. Dis. 2020, 15, 357–365. [Google Scholar] [CrossRef] [Green Version]
  115. Dimitrova, D.; Youroukova, V.; Ivanova-Todorova, E.; Tumangelova-Yuzeir, K.; Velikova, T. Serum levels of IL-5, IL-6, IL-8, IL-13 and IL-17A in pre-defined groups of adult patients with moderate and severe bronchial asthma. Respir. Med. 2019, 154, 144–154. [Google Scholar] [CrossRef]
  116. Jiang, X.G.; Yang, X.D.; Lv, Z.; Zhuang, P.H. Elevated serum levels of TNF-alpha, IL-8, and ECP can be involved in the development and progression of bronchial asthma. J. Asthma 2018, 55, 111–118. [Google Scholar] [CrossRef]
  117. Norzila, M.Z.; Fakes, K.; Henry, R.L.; Simpson, J.; Gibson, P.G. Interleukin-8 secretion and neutrophil recruitment accompanies induced sputum eosinophil activation in children with acute asthma. Am. J. Respir. Crit. Care Med. 2000, 161, 769–774. [Google Scholar] [CrossRef]
  118. Hosoki, K.; Ying, S.; Corrigan, C.; Qi, H.; Kurosky, A.; Jennings, K.; Sun, Q.; Boldogh, I.; Sur, S. Analysis of a Panel of 48 Cytokines in BAL Fluids Specifically Identifies IL-8 Levels as the Only Cytokine that Distinguishes Controlled Asthma from Uncontrolled Asthma, and Correlates Inversely with FEV1. PLoS ONE 2015, 10, e0126035. [Google Scholar] [CrossRef] [Green Version]
  119. Singh, P.; Ali, S.A. Multifunctional Role of S100 Protein Family in the Immune System: An Update. Cells 2022, 11, 2274. [Google Scholar] [CrossRef]
  120. Quoc, Q.L.; Choi, Y.; Thi Bich, T.C.; Yang, E.M.; Shin, Y.S.; Park, H.S. S100A9 in adult asthmatic patients: A biomarker for neutrophilic asthma. Exp. Mol. Med. 2021, 53, 1170–1179. [Google Scholar] [CrossRef]
  121. Hur, G.Y.; Ye, Y.M.; Yang, E.; Park, H.S. Serum potential biomarkers according to sputum inflammatory cell profiles in adult asthmatics. Korean J. Intern. Med. 2020, 35, 988–997. [Google Scholar] [CrossRef] [PubMed]
  122. Lee, T.H.; Jang, A.S.; Park, J.S.; Kim, T.H.; Choi, Y.S.; Shin, H.R.; Park, S.W.; Uh, S.T.; Choi, J.S.; Kim, Y.H.; et al. Elevation of S100 calcium binding protein A9 in sputum of neutrophilic inflammation in severe uncontrolled asthma. Ann. Allergy Asthma Immunol. 2013, 111, 268–275.e261. [Google Scholar] [CrossRef] [PubMed]
  123. Yang, Z.; Yan, W.X.; Cai, H.; Tedla, N.; Armishaw, C.; Di Girolamo, N.; Wang, H.W.; Hampartzoumian, T.; Simpson, J.L.; Gibson, P.G.; et al. S100A12 provokes mast cell activation: A potential amplification pathway in asthma and innate immunity. J. Allergy Clin. Immunol. 2007, 119, 106–114. [Google Scholar] [CrossRef] [PubMed]
  124. Camoretti-Mercado, B.; Karrar, E.; Nunez, L.; Bowman, M.A. S100A12 and the Airway Smooth Muscle: Beyond Inflammation and Constriction. J. Allergy Ther. 2012, 3 (Suppl. S1), S1-007. [Google Scholar] [CrossRef] [PubMed]
  125. Hashimoto, M.; Tanaka, H.; Abe, S. Quantitative analysis of bronchial wall vascularity in the medium and small airways of patients with asthma and COPD. Chest 2005, 127, 965–972. [Google Scholar] [CrossRef]
  126. Hoshino, M.; Takahashi, M.; Aoike, N. Expression of vascular endothelial growth factor, basic fibroblast growth factor, and angiogenin immunoreactivity in asthmatic airways and its relationship to angiogenesis. J. Allergy Clin. Immunol. 2001, 107, 295–301. [Google Scholar] [CrossRef]
  127. Chetta, A.; Zanini, A.; Foresi, A.; Del Donno, M.; Castagnaro, A.; D’Ippolito, R.; Baraldo, S.; Testi, R.; Saetta, M.; Olivieri, D. Vascular component of airway remodeling in asthma is reduced by high dose of fluticasone. Am. J. Respir. Crit. Care Med. 2003, 167, 751–757. [Google Scholar] [CrossRef] [Green Version]
  128. Tas, S.W.; Maracle, C.X.; Balogh, E.; Szekanecz, Z. Targeting of proangiogenic signalling pathways in chronic inflammation. Nat. Rev. Rheumatol. 2016, 12, 111–122. [Google Scholar] [CrossRef]
  129. Zheng, W.; Aspelund, A.; Alitalo, K. Lymphangiogenic factors, mechanisms, and applications. J. Clin. Invest. 2014, 124, 878–887. [Google Scholar] [CrossRef]
  130. Akwii, R.G.; Mikelis, C.M. Targeting the Angiopoietin/Tie Pathway: Prospects for Treatment of Retinal and Respiratory Disorders. Drugs 2021, 81, 1731–1749. [Google Scholar] [CrossRef]
  131. Loffredo, S.; Borriello, F.; Iannone, R.; Ferrara, A.L.; Galdiero, M.R.; Gigantino, V.; Esposito, P.; Varricchi, G.; Lambeau, G.; Cassatella, M.A.; et al. Group V Secreted Phospholipase A2 Induces the Release of Proangiogenic and Antiangiogenic Factors by Human Neutrophils. Front. Immunol. 2017, 8, 443. [Google Scholar] [CrossRef] [Green Version]
  132. Nie, M.; Yang, L.; Bi, X.; Wang, Y.; Sun, P.; Yang, H.; Liu, P.; Li, Z.; Xia, Y.; Jiang, W. Neutrophil Extracellular Traps Induced by IL8 Promote Diffuse Large B-cell Lymphoma Progression via the TLR9 Signaling. Clin. Cancer Res. 2019, 25, 1867–1879. [Google Scholar] [CrossRef] [PubMed]
  133. An, Z.; Li, J.; Yu, J.; Wang, X.; Gao, H.; Zhang, W.; Wei, Z.; Zhang, J.; Zhang, Y.; Zhao, J.; et al. Neutrophil extracellular traps induced by IL-8 aggravate atherosclerosis via activation NF-kappaB signaling in macrophages. Cell Cycle 2019, 18, 2928–2938. [Google Scholar] [CrossRef] [PubMed]
  134. Alfaro, C.; Teijeira, A.; Onate, C.; Perez, G.; Sanmamed, M.F.; Andueza, M.P.; Alignani, D.; Labiano, S.; Azpilikueta, A.; Rodriguez-Paulete, A.; et al. Tumor-Produced Interleukin-8 Attracts Human Myeloid-Derived Suppressor Cells and Elicits Extrusion of Neutrophil Extracellular Traps (NETs). Clin. Cancer Res. 2016, 22, 3924–3936. [Google Scholar] [CrossRef] [Green Version]
  135. Zhang, Y.; Chandra, V.; Sanchez, E.R.; Dutta, P.; Quesada, P.R.; Rakoski, A.; Zoltan, M.; Arora, N.; Baydogan, S.; Horne, W.; et al. Interleukin-17-induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer. J. Exp. Med. 2020, 217, e20190354. [Google Scholar] [CrossRef] [PubMed]
  136. Teijeira, A.; Garasa, S.; Gato, M.; Alfaro, C.; Migueliz, I.; Cirella, A.; de Andrea, C.; Ochoa, M.C.; Otano, I.; Etxeberria, I.; et al. CXCR1 and CXCR2 Chemokine Receptor Agonists Produced by Tumors Induce Neutrophil Extracellular Traps that Interfere with Immune Cytotoxicity. Immunity 2020, 52, 856–871.e858. [Google Scholar] [CrossRef]
  137. Guglietta, S.; Chiavelli, A.; Zagato, E.; Krieg, C.; Gandini, S.; Ravenda, P.S.; Bazolli, B.; Lu, B.; Penna, G.; Rescigno, M. Coagulation induced by C3aR-dependent NETosis drives protumorigenic neutrophils during small intestinal tumorigenesis. Nat. Commun. 2016, 7, 11037. [Google Scholar] [CrossRef] [Green Version]
  138. Robledo-Avila, F.H.; Ruiz-Rosado, J.D.; Brockman, K.L.; Kopp, B.T.; Amer, A.O.; McCoy, K.; Bakaletz, L.O.; Partida-Sanchez, S. Dysregulated Calcium Homeostasis in Cystic Fibrosis Neutrophils Leads to Deficient Antimicrobial Responses. J. Immunol. 2018, 201, 2016–2027. [Google Scholar] [CrossRef]
  139. Toussaint, M.; Jackson, D.J.; Swieboda, D.; Guedan, A.; Tsourouktsoglou, T.D.; Ching, Y.M.; Radermecker, C.; Makrinioti, H.; Aniscenko, J.; Bartlett, N.W.; et al. Host DNA released by NETosis promotes rhinovirus-induced type-2 allergic asthma exacerbation. Nat. Med. 2017, 23, 681–691. [Google Scholar] [CrossRef] [Green Version]
  140. Albrengues, J.; Shields, M.A.; Ng, D.; Park, C.G.; Ambrico, A.; Poindexter, M.E.; Upadhyay, P.; Uyeminami, D.L.; Pommier, A.; Kuttner, V.; et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018, 361, eaao4227. [Google Scholar] [CrossRef]
  141. Marino, F.; Scanzano, A.; Pulze, L.; Pinoli, M.; Rasini, E.; Luini, A.; Bombelli, R.; Legnaro, M.; de Eguileor, M.; Cosentino, M. beta2 -Adrenoceptors inhibit neutrophil extracellular traps in human polymorphonuclear leukocytes. J. Leukoc. Biol. 2018, 104, 603–614. [Google Scholar] [CrossRef]
  142. Tcherniuk, S.; Cenac, N.; Comte, M.; Frouard, J.; Errazuriz-Cerda, E.; Galabov, A.; Morange, P.E.; Vergnolle, N.; Si-Tahar, M.; Alessi, M.C.; et al. Formyl Peptide Receptor 2 Plays a Deleterious Role During Influenza A Virus Infections. J. Infect. Dis. 2016, 214, 237–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Varricchi, G.; Ferri, S.; Pepys, J.; Poto, R.; Spadaro, G.; Nappi, E.; Paoletti, G.; Virchow, J.C.; Heffler, E.; Canonica, W.G. Biologics and airway remodeling in severe asthma. Allergy 2022. [CrossRef] [PubMed]
  144. Hammad, H.; Lambrecht, B.N. The basic immunology of asthma. Cell 2021, 184, 1469–1485. [Google Scholar] [CrossRef] [PubMed]
  145. Roan, F.; Obata-Ninomiya, K.; Ziegler, S.F. Epithelial cell-derived cytokines: More than just signaling the alarm. J. Clin. Invest. 2019, 129, 1441–1451. [Google Scholar] [CrossRef] [Green Version]
  146. Varricchi, G.; Pecoraro, A.; Marone, G.; Criscuolo, G.; Spadaro, G.; Genovese, A. Thymic Stromal Lymphopoietin Isoforms, Inflammatory Disorders, and Cancer. Front. Immunol. 2018, 9, 1595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Whetstone, C.E.; Ranjbar, M.; Omer, H.; Cusack, R.P.; Gauvreau, G.M. The Role of Airway Epithelial Cell Alarmins in Asthma. Cells 2022, 11, 1105. [Google Scholar] [CrossRef]
  148. Li, Y.; Yang, Y.; Gan, T.; Zhou, J.; Hu, F.; Hao, N.; Yuan, B.; Chen, Y.; Zhang, M. Extracellular RNAs from lung cancer cells activate epithelial cells and induce neutrophil extracellular traps. Int. J. Oncol. 2019, 55, 69–80. [Google Scholar] [CrossRef] [Green Version]
  149. Pham, D.L.; Ban, G.Y.; Kim, S.H.; Shin, Y.S.; Ye, Y.M.; Chwae, Y.J.; Park, H.S. Neutrophil autophagy and extracellular DNA traps contribute to airway inflammation in severe asthma. Clin. Exp. Allergy 2017, 47, 57–70. [Google Scholar] [CrossRef]
  150. Liang, Y.; Hou, C.; Kong, J.; Wen, H.; Zheng, X.; Wu, L.; Huang, H.; Chen, Y. HMGB1 binding to receptor for advanced glycation end products enhances inflammatory responses of human bronchial epithelial cells by activating p38 MAPK and ERK1/2. Mol. Cell Biochem. 2015, 405, 63–71. [Google Scholar] [CrossRef]
  151. Akk, A.; Springer, L.E.; Pham, C.T. Neutrophil Extracellular Traps Enhance Early Inflammatory Response in Sendai Virus-Induced Asthma Phenotype. Front. Immunol. 2016, 7, 325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Radermecker, C.; Sabatel, C.; Vanwinge, C.; Ruscitti, C.; Marechal, P.; Perin, F.; Schyns, J.; Rocks, N.; Toussaint, M.; Cataldo, D.; et al. Locally instructed CXCR4(hi) neutrophils trigger environment-driven allergic asthma through the release of neutrophil extracellular traps. Nat. Immunol. 2019, 20, 1444–1455. [Google Scholar] [CrossRef] [PubMed]
  153. Shin, J.W.; Kim, J.; Ham, S.; Choi, S.M.; Lee, C.H.; Lee, J.C.; Kim, J.H.; Cho, S.H.; Kang, H.R.; Kim, Y.M.; et al. A unique population of neutrophils generated by air pollutant-induced lung damage exacerbates airway inflammation. J. Allergy Clin. Immunol. 2022, 149, 1253–1269.e1258. [Google Scholar] [CrossRef] [PubMed]
  154. 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] [PubMed] [Green Version]
  155. Li, W.X.; Wang, F.; Zhu, Y.Q.; Zhang, L.M.; Zhang, Z.H.; Wang, X.M. Inhibitors of nitric oxide synthase can reduce extracellular traps from neutrophils in asthmatic children in vitro. Pediatr. Pulmonol. 2020, 55, 68–75. [Google Scholar] [CrossRef] [PubMed]
  156. Impellizzieri, D.; Ridder, F.; Raeber, M.E.; Egholm, C.; Woytschak, J.; Kolios, A.G.A.; Legler, D.F.; Boyman, O. IL-4 receptor engagement in human neutrophils impairs their migration and extracellular trap formation. J. Allergy Clin. Immunol. 2019, 144, 267–279.e264. [Google Scholar] [CrossRef] [Green Version]
  157. Penagos, M.; Durham, S.R. Allergen immunotherapy for long-term tolerance and prevention. J. Allergy Clin. Immunol. 2022, 149, 802–811. [Google Scholar] [CrossRef]
  158. Pfaar, O.; Bousquet, J.; Durham, S.R.; Kleine-Tebbe, J.; Larche, M.; Roberts, G.; Shamji, M.H.; Gerth van Wijk, R. One hundred and ten years of Allergen Immunotherapy: A journey from empiric observation to evidence. Allergy 2022, 77, 454–468. [Google Scholar] [CrossRef]
  159. Polak, D.; Hafner, C.; Briza, P.; Kitzmuller, C.; Elbe-Burger, A.; Samadi, N.; Gschwandtner, M.; Pfutzner, W.; Zlabinger, G.J.; Jahn-Schmid, B.; et al. A novel role for neutrophils in IgE-mediated allergy: Evidence for antigen presentation in late-phase reactions. J. Allergy Clin. Immunol. 2019, 143, 1143–1152.e1144. [Google Scholar] [CrossRef] [Green Version]
  160. Karacs, J.; Reithofer, M.; Kitzmuller, C.; Kraller, M.; Schmalz, S.; Bleichert, S.; Huppa, J.B.; Stockinger, H.; Bohle, B.; Jahn-Schmid, B. Adjuvants and Vaccines Used in Allergen-Specific Immunotherapy Induce Neutrophil Extracellular Traps. Vaccines 2021, 9, 321. [Google Scholar] [CrossRef]
  161. Drescher, B.; Bai, F. Neutrophil in viral infections, friend or foe? Virus Res. 2013, 171, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. 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]
  163. Hermosilla, C.; Caro, T.M.; Silva, L.M.; Ruiz, A.; Taubert, A. The intriguing host innate immune response: Novel anti-parasitic defence by neutrophil extracellular traps. Parasitology 2014, 141, 1489–1498. [Google Scholar] [CrossRef] [PubMed]
  164. Castanheira, F.V.S.; Kubes, P. Neutrophils and NETs in modulating acute and chronic inflammation. Blood 2019, 133, 2178–2185. [Google Scholar] [CrossRef]
  165. Meegan, J.E.; Yang, X.; Coleman, D.C.; Jannaway, M.; Yuan, S.Y. Neutrophil-mediated vascular barrier injury: Role of neutrophil extracellular traps. Microcirculation 2017, 24, 12352. [Google Scholar] [CrossRef] [Green Version]
  166. Jenne, C.N.; Kubes, P. Virus-induced NETs--critical component of host defense or pathogenic mediator? PLoS Pathog. 2015, 11, e1004546. [Google Scholar] [CrossRef] [Green Version]
  167. Jenne, C.N.; Wong, C.H.; Zemp, F.J.; McDonald, B.; Rahman, M.M.; Forsyth, P.A.; McFadden, G.; Kubes, P. Neutrophils recruited to sites of infection protect from virus challenge by releasing neutrophil extracellular traps. Cell Host Microbe 2013, 13, 169–180. [Google Scholar] [CrossRef] [Green Version]
  168. Schauer, C.; Janko, C.; Munoz, L.E.; Zhao, Y.; Kienhofer, D.; Frey, B.; Lell, M.; Manger, B.; Rech, J.; Naschberger, E.; et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat. Med. 2014, 20, 511–517. [Google Scholar] [CrossRef]
  169. Li, Y.; Cao, X.; Liu, Y.; Zhao, Y.; Herrmann, M. Neutrophil Extracellular Traps Formation and Aggregation Orchestrate Induction and Resolution of Sterile Crystal-Mediated Inflammation. Front. Immunol. 2018, 9, 1559. [Google Scholar] [CrossRef]
  170. Kienhofer, D.; Hahn, J.; Stoof, J.; Csepregi, J.Z.; Reinwald, C.; Urbonaviciute, V.; Johnsson, C.; Maueroder, C.; Podolska, M.J.; Biermann, M.H.; et al. Experimental lupus is aggravated in mouse strains with impaired induction of neutrophil extracellular traps. JCI Insight 2017, 2, e92920. [Google Scholar] [CrossRef]
  171. Papayannopoulos, V.; Zychlinsky, A. NETs: A new strategy for using old weapons. Trends Immunol. 2009, 30, 513–521. [Google Scholar] [CrossRef] [PubMed]
  172. Daigo, K.; Takamatsu, Y.; Hamakubo, T. The Protective Effect against Extracellular Histones Afforded by Long-Pentraxin PTX3 as a Regulator of NETs. Front. Immunol. 2016, 7, 344. [Google Scholar] [CrossRef] [Green Version]
  173. Guimaraes-Costa, A.B.; Rochael, N.C.; Oliveira, F.; Echevarria-Lima, J.; Saraiva, E.M. Neutrophil Extracellular Traps Reprogram IL-4/GM-CSF-Induced Monocyte Differentiation to Anti-inflammatory Macrophages. Front. Immunol. 2017, 8, 523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Rawat, K.; Syeda, S.; Shrivastava, A. Hyperactive neutrophils infiltrate vital organs of tumor bearing host and contribute to gradual systemic deterioration via upregulated NE, MPO and MMP-9 activity. Immunol. Lett. 2022, 241, 35–48. [Google Scholar] [CrossRef] [PubMed]
  175. Pan, S.; Conaway, S., Jr.; Deshpande, D.A. Mitochondrial regulation of airway smooth muscle functions in health and pulmonary diseases. Arch. Biochem. Biophys. 2019, 663, 109–119. [Google Scholar] [CrossRef]
  176. Yu, H.; Yang, J.; Xiao, Q.; Lu, Y.; Zhou, X.; Xia, L.; Nie, D. Regulation of high glucose-mediated mucin expression by matrix metalloproteinase-9 in human airway epithelial cells. Exp. Cell Res. 2015, 333, 127–135. [Google Scholar] [CrossRef]
  177. Allam, R.; Kumar, S.V.; Darisipudi, M.N.; Anders, H.J. Extracellular histones in tissue injury and inflammation. J. Mol. Med. 2014, 92, 465–472. [Google Scholar] [CrossRef]
  178. Zhu, Y.; Huang, Y.; Ji, Q.; Fu, S.; Gu, J.; Tai, N.; Wang, X. Interplay between Extracellular Matrix and Neutrophils in Diseases. J. Immunol. Res. 2021, 2021, 8243378. [Google Scholar] [CrossRef]
  179. Krishnamoorthy, N.; Douda, D.N.; Bruggemann, T.R.; Ricklefs, I.; Duvall, M.G.; Abdulnour, R.E.; Martinod, K.; Tavares, L.; Wang, X.; Cernadas, M.; et al. Neutrophil cytoplasts induce TH17 differentiation and skew inflammation toward neutrophilia in severe asthma. Sci. Immunol. 2018, 3, eaao4747. [Google Scholar] [CrossRef] [Green Version]
  180. Wan, R.; Jiang, J.; Hu, C.; Chen, X.; Chen, C.; Zhao, B.; Hu, X.; Zheng, Z.; Li, Y. Neutrophil extracellular traps amplify neutrophil recruitment and inflammation in neutrophilic asthma by stimulating the airway epithelial cells to activate the TLR4/ NF-kappaB pathway and secrete chemokines. Aging 2020, 12, 16820–16836. [Google Scholar] [CrossRef]
  181. Chen, X.; Li, Y.; Qin, L.; He, R.; Hu, C. Neutrophil Extracellular Trapping Network Promotes the Pathogenesis of Neutrophil-associated Asthma through Macrophages. Immunol. Investig. 2021, 50, 544–561. [Google Scholar] [CrossRef] [PubMed]
  182. Yousefi, S.; Gold, J.A.; Andina, N.; Lee, J.J.; Kelly, A.M.; Kozlowski, E.; Schmid, I.; Straumann, A.; Reichenbach, J.; Gleich, G.J.; et al. Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense. Nat. Med. 2008, 14, 949–953. [Google Scholar] [CrossRef] [PubMed]
  183. Hoeksema, M.; Tripathi, S.; White, M.; Qi, L.; Taubenberger, J.; van Eijk, M.; Haagsman, H.; Hartshorn, K.L. Arginine-rich histones have strong antiviral activity for influenza A viruses. Innate Immun. 2015, 21, 736–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. McCormick, A.; Heesemann, L.; Wagener, J.; Marcos, V.; Hartl, D.; Loeffler, J.; Heesemann, J.; Ebel, F. NETs formed by human neutrophils inhibit growth of the pathogenic mold Aspergillus fumigatus. Microbes Infect. 2010, 12, 928–936. [Google Scholar] [CrossRef]
  185. Walter, L.; Canup, B.; Pujada, A.; Bui, T.A.; Arbasi, B.; Laroui, H.; Merlin, D.; Garg, P. Matrix metalloproteinase 9 (MMP9) limits reactive oxygen species (ROS) accumulation and DNA damage in colitis-associated cancer. Cell Death Dis. 2020, 11, 767. [Google Scholar] [CrossRef]
  186. Bencivenga, L.; Rengo, G.; Varricchi, G. Elderly at time of COronaVIrus disease 2019 (COVID-19): Possible role of immunosenescence and malnutrition. Geroscience 2020, 42, 1089–1092. [Google Scholar] [CrossRef]
  187. Blazkova, J.; Gupta, S.; Liu, Y.; Gaudilliere, B.; Ganio, E.A.; Bolen, C.R.; Saar-Dover, R.; Fragiadakis, G.K.; Angst, M.S.; Hasni, S.; et al. Multicenter Systems Analysis of Human Blood Reveals Immature Neutrophils in Males and During Pregnancy. J. Immunol. 2017, 198, 2479–2488. [Google Scholar] [CrossRef] [Green Version]
  188. Gupta, S.; Nakabo, S.; Blanco, L.P.; O’Neil, L.J.; Wigerblad, G.; Goel, R.R.; Mistry, P.; Jiang, K.; Carmona-Rivera, C.; Chan, D.W.; et al. Sex differences in neutrophil biology modulate response to type I interferons and immunometabolism. Proc. Natl. Acad. Sci. USA 2020, 117, 16481–16491. [Google Scholar] [CrossRef]
  189. Lu, R.J.; Taylor, S.; Contrepois, K.; Kim, M.; Bravo, J.I.; Ellenberger, M.; Sampathkumar, N.K.; Benayoun, B.A. Multi-omic profiling of primary mouse neutrophils predicts a pattern of sex and age-related functional regulation. Nat. Aging 2021, 1, 715–733. [Google Scholar] [CrossRef]
  190. Adrover, J.M.; Aroca-Crevillen, A.; Crainiciuc, G.; Ostos, F.; Rojas-Vega, Y.; Rubio-Ponce, A.; Cilloniz, C.; Bonzon-Kulichenko, E.; Calvo, E.; Rico, D.; et al. Programmed ‘disarming’ of the neutrophil proteome reduces the magnitude of inflammation. Nat. Immunol. 2020, 21, 135–144. [Google Scholar] [CrossRef]
  191. Casanova-Acebes, M.; Nicolas-Avila, J.A.; Li, J.L.; Garcia-Silva, S.; Balachander, A.; Rubio-Ponce, A.; Weiss, L.A.; Adrover, J.M.; Burrows, K.; N, A.G.; et al. Neutrophils instruct homeostatic and pathological states in naive tissues. J. Exp. Med. 2018, 215, 2778–2795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Hazeldine, J.; Harris, P.; Chapple, I.L.; Grant, M.; Greenwood, H.; Livesey, A.; Sapey, E.; Lord, J.M. Impaired neutrophil extracellular trap formation: A novel defect in the innate immune system of aged individuals. Aging Cell 2014, 13, 690–698. [Google Scholar] [CrossRef] [PubMed]
  193. Piasecka, B.; Duffy, D.; Urrutia, A.; Quach, H.; Patin, E.; Posseme, C.; Bergstedt, J.; Charbit, B.; Rouilly, V.; MacPherson, C.R.; et al. Distinctive roles of age, sex, and genetics in shaping transcriptional variation of human immune responses to microbial challenges. Proc. Natl. Acad. Sci. USA 2018, 115, E488–E497. [Google Scholar] [CrossRef] [Green Version]
  194. Varricchi, G.; Poto, R.; Covelli, B.; Di Spigna, G.; Galdiero, M.R.; Marone, G.; Postiglione, L.; Spadaro, G. Gender dimorphism in IgA subclasses in T2-high asthma. Clin. Exp. Med. 2022. [Google Scholar] [CrossRef] [PubMed]
  195. Mauvais-Jarvis, F.; Bairey Merz, N.; Barnes, P.J.; Brinton, R.D.; Carrero, J.J.; DeMeo, D.L.; De Vries, G.J.; Epperson, C.N.; Govindan, R.; Klein, S.L.; et al. Sex and gender: Modifiers of health, disease, and medicine. Lancet 2020, 396, 565–582. [Google Scholar] [CrossRef]
  196. Pignataro, F.S.; Bonini, M.; Forgione, A.; Melandri, S.; Usmani, O.S. Asthma and gender: The female lung. Pharmacol. Res. 2017, 119, 384–390. [Google Scholar] [CrossRef]
  197. Wenzel, S.E. Severe Adult Asthmas: Integrating Clinical Features, Biology, and Therapeutics to Improve Outcomes. Am. J. Respir. Crit. Care Med. 2021, 203, 809–821. [Google Scholar] [CrossRef]
  198. Fahy, J.V. Type 2 inflammation in asthma--present in most, absent in many. Nat. Rev. Immunol. 2015, 15, 57–65. [Google Scholar] [CrossRef] [Green Version]
  199. Frossing, L.; Silberbrandt, A.; Von Bulow, A.; Backer, V.; Porsbjerg, C. The Prevalence of Subtypes of Type 2 Inflammation in an Unselected Population of Patients with Severe Asthma. J. Allergy Clin. Immunol. Pract. 2021, 9, 1267–1275. [Google Scholar] [CrossRef]
  200. Diamant, Z.; Vijverberg, S.; Alving, K.; Bakirtas, A.; Bjermer, L.; Custovic, A.; Dahlen, S.E.; Gaga, M.; van Wijk, R.G.; Giacco, S.D.; et al. Toward clinically applicable biomarkers for asthma: An EAACI position paper. Allergy 2019, 74, 1835–1851. [Google Scholar] [CrossRef]
  201. 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] [PubMed]
  202. Granger, V.; Taille, C.; Roach, D.; Letuve, S.; Dupin, C.; Hamidi, F.; Noel, 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]
  203. Hayden, H.; Ibrahim, N.; Klopf, J.; Zagrapan, B.; Mauracher, L.M.; Hell, L.; Hofbauer, T.M.; Ondracek, A.S.; Schoergenhofer, C.; Jilma, B.; et al. ELISA detection of MPO-DNA complexes in human plasma is error-prone and yields limited information on neutrophil extracellular traps formed in vivo. PLoS ONE 2021, 16, e0250265. [Google Scholar] [CrossRef] [PubMed]
  204. Boeltz, S.; Amini, P.; Anders, H.J.; Andrade, F.; Bilyy, R.; Chatfield, S.; Cichon, I.; Clancy, D.M.; Desai, J.; Dumych, T.; et al. To NET or not to NET:current opinions and state of the science regarding the formation of neutrophil extracellular traps. Cell Death Differ. 2019, 26, 395–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Chamardani, T.M.; Amiritavassoli, S. Inhibition of NETosis for treatment purposes: Friend or foe? Mol. Cell Biochem. 2022, 477, 673–688. [Google Scholar] [CrossRef]
  206. Yang, C.; Montgomery, M. Dornase alfa for cystic fibrosis. Cochrane Database Syst. Rev. 2021, 3, CD001127. [Google Scholar] [CrossRef]
  207. Kumar, S.V.; Kulkarni, O.P.; Mulay, S.R.; Darisipudi, M.N.; Romoli, S.; Thomasova, D.; Scherbaum, C.R.; Hohenstein, B.; Hugo, C.; Muller, S.; et al. Neutrophil Extracellular Trap-Related Extracellular Histones Cause Vascular Necrosis in Severe GN. J. Am. Soc. Nephrol. 2015, 26, 2399–2413. [Google Scholar] [CrossRef] [Green Version]
  208. Claushuis, T.A.M.; van der Donk, L.E.H.; Luitse, A.L.; van Veen, H.A.; van der Wel, N.N.; van Vught, L.A.; Roelofs, J.; de Boer, O.J.; Lankelma, J.M.; Boon, L.; et al. Role of Peptidylarginine Deiminase 4 in Neutrophil Extracellular Trap Formation and Host Defense during Klebsiella pneumoniae-Induced Pneumonia-Derived Sepsis. J. Immunol. 2018, 201, 1241–1252. [Google Scholar] [CrossRef] [Green Version]
  209. Arpinati, L.; Shaul, M.E.; Kaisar-Iluz, N.; Mali, S.; Mahroum, S.; Fridlender, Z.G. NETosis in cancer: A critical analysis of the impact of cancer on neutrophil extracellular trap (NET) release in lung cancer patients vs. mice. Cancer Immunol. Immunother. 2020, 69, 199–213. [Google Scholar] [CrossRef]
  210. Lin, F.; Wang, N.; Zhang, T.C. The role of endothelial-mesenchymal transition in development and pathological process. IUBMB Life 2012, 64, 717–723. [Google Scholar] [CrossRef]
  211. Winkelstein, J.A.; Marino, M.C.; Johnston, R.B., Jr.; Boyle, J.; Curnutte, J.; Gallin, J.I.; Malech, H.L.; Holland, S.M.; Ochs, H.; Quie, P.; et al. Chronic granulomatous disease. Report on a national registry of 368 patients. Medicine 2000, 79, 155–169. [Google Scholar] [CrossRef] [PubMed]
  212. Curran, A.M.; Naik, P.; Giles, J.T.; Darrah, E. PAD enzymes in rheumatoid arthritis: Pathogenic effectors and autoimmune targets. Nat. Rev. Rheumatol. 2020, 16, 301–315. [Google Scholar] [CrossRef] [PubMed]
  213. Thanabalasuriar, A.; Scott, B.N.V.; Peiseler, M.; Willson, M.E.; Zeng, Z.; Warrener, P.; Keller, A.E.; Surewaard, B.G.J.; Dozier, E.A.; Korhonen, J.T.; et al. Neutrophil Extracellular Traps Confine Pseudomonas aeruginosa Ocular Biofilms and Restrict Brain Invasion. Cell Host Microbe 2019, 25, 526–536.e524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Martinez, N.E.; Zimmermann, T.J.; Goosmann, C.; Alexander, T.; Hedberg, C.; Ziegler, S.; Zychlinsky, A.; Waldmann, H. Tetrahydroisoquinolines: New Inhibitors of Neutrophil Extracellular Trap (NET) Formation. Chembiochem 2017, 18, 888–893. [Google Scholar] [CrossRef] [PubMed]
  215. Marone, G.; Galdiero, M.R.; Pecoraro, A.; Pucino, V.; Criscuolo, G.; Triassi, M.; Varricchi, G. Prostaglandin D2 receptor antagonists in allergic disorders: Safety, efficacy, and future perspectives. Expert Opin. Investig. Drugs 2019, 28, 73–84. [Google Scholar] [CrossRef]
  216. Cristinziano, L.; Poto, R.; Criscuolo, G.; Ferrara, A.L.; Galdiero, M.R.; Modestino, L.; Loffredo, S.; de Paulis, A.; Marone, G.; Spadaro, G.; et al. IL-33 and Superantigenic Activation of Human Lung Mast Cells Induce the Release of Angiogenic and Lymphangiogenic Factors. Cells 2021, 10, 145. [Google Scholar] [CrossRef]
  217. Puzzovio, P.G.; Eliashar, R.; Levi-Schaffer, F. Tezepelumab administration in moderate-to-severe uncontrolled asthma: Is it all about eosinophils? J. Allergy Clin. Immunol. 2022, 149, 1582–1584. [Google Scholar] [CrossRef]
  218. 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]
  219. 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]
  220. Conceicao-Silva, F.; Reis, C.S.M.; De Luca, P.M.; Leite-Silva, J.; Santiago, M.A.; Morrot, A.; Morgado, F.N. The Immune System Throws Its Traps: Cells and Their Extracellular Traps in Disease and Protection. Cells 2021, 10, 1891. [Google Scholar] [CrossRef]
  221. 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, 277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Campillo-Navarro, M.; Leyva-Paredes, K.; Donis-Maturano, L.; Rodriguez-Lopez, G.M.; Soria-Castro, R.; Garcia-Perez, B.E.; Puebla-Osorio, N.; Ullrich, S.E.; Luna-Herrera, J.; Flores-Romo, L.; et al. Mycobacterium tuberculosis Catalase Inhibits the Formation of Mast Cell Extracellular Traps. Front. Immunol. 2018, 9, 1161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Nija, R.J.; Sanju, S.; Sidharthan, N.; Mony, U. Extracellular Trap by Blood Cells: Clinical Implications. Tissue Eng. Regen Med. 2020, 17, 141–153. [Google Scholar] [CrossRef] [PubMed]
  224. Clark, M.; Kim, J.; Etesami, N.; Shimamoto, J.; Whalen, R.V.; Martin, G.; Okumura, C.Y.M. Group A Streptococcus Prevents Mast Cell Degranulation to Promote Extracellular Trap Formation. Front. Immunol. 2018, 9, 327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Lopes, J.P.; Stylianou, M.; Nilsson, G.; Urban, C.F. Opportunistic pathogen Candida albicans elicits a temporal response in primary human mast cells. Sci. Rep. 2015, 5, 12287. [Google Scholar] [CrossRef] [Green Version]
  226. Naqvi, N.; Ahuja, K.; Selvapandiyan, A.; Dey, R.; Nakhasi, H.; Puri, N. Role of Mast Cells in clearance of Leishmania through extracellular trap formation. Sci. Rep. 2017, 7, 13240. [Google Scholar] [CrossRef] [Green Version]
  227. Campillo-Navarro, M.; Leyva-Paredes, K.; Donis-Maturano, L.; Gonzalez-Jimenez, M.; Paredes-Vivas, Y.; Cerbulo-Vazquez, A.; Serafin-Lopez, J.; Garcia-Perez, B.; Ullrich, S.E.; Flores-Romo, L.; et al. Listeria monocytogenes induces mast cell extracellular traps. Immunobiology 2017, 222, 432–439. [Google Scholar] [CrossRef]
  228. Zhang, X.; Zhang, L.; Tan, Y.M.; Liu, Y.P.; Li, J.J.; Deng, Q.M.; Yan, S.B.; Zhang, W.; Han, L.; Zhong, M. Hepcidin gene silencing ameliorated inflammation and insulin resistance in adipose tissue of db/db mice via inhibiting METs formation. Mol. Immunol. 2021, 133, 110–121. [Google Scholar] [CrossRef]
  229. Kummarapurugu, A.B.; Zheng, S.; Ma, J.; Ghosh, S.; Hawkridge, A.; Voynow, J.A. Neutrophil Elastase Triggers the Release of Macrophage Extracellular Traps: Relevance to Cystic Fibrosis. Am. J. Respir. Cell Mol. Biol. 2022, 66, 76–85. [Google Scholar] [CrossRef]
  230. Okubo, K.; Kurosawa, M.; Kamiya, M.; Urano, Y.; Suzuki, A.; Yamamoto, K.; Hase, K.; Homma, K.; Sasaki, J.; Miyauchi, H.; et al. Macrophage extracellular trap formation promoted by platelet activation is a key mediator of rhabdomyolysis-induced acute kidney injury. Nat. Med. 2018, 24, 232–238. [Google Scholar] [CrossRef]
  231. Tan, C.; Aziz, M.; Wang, P. The vitals of NETs. J. Leukoc. Biol. 2021, 110, 797–808. [Google Scholar] [CrossRef] [PubMed]
  232. Ingelsson, B.; Soderberg, D.; Strid, T.; Soderberg, A.; Bergh, A.C.; Loitto, V.; Lotfi, K.; Segelmark, M.; Spyrou, G.; Rosen, A. Lymphocytes eject interferogenic mitochondrial DNA webs in response to CpG and non-CpG oligodeoxynucleotides of class C. Proc. Natl. Acad. Sci. USA 2018, 115, E478–E487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. West, A.P.; Shadel, G.S. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat. Rev. Immunol 2017, 17, 363–375. [Google Scholar] [CrossRef] [PubMed]
  234. Carr, T.F.; Zeki, A.A.; Kraft, M. Eosinophilic and Noneosinophilic Asthma. Am. J. Respir. Crit. Care Med. 2018, 197, 22–37. [Google Scholar] [CrossRef] [PubMed]
  235. Simon, D.; Radonjic-Hosli, S.; Straumann, A.; Yousefi, S.; Simon, H.U. Active eosinophilic esophagitis is characterized by epithelial barrier defects and eosinophil extracellular trap formation. Allergy 2015, 70, 443–452. [Google Scholar] [CrossRef]
  236. Ueki, S.; Konno, Y.; Takeda, M.; Moritoki, Y.; Hirokawa, M.; Matsuwaki, Y.; Honda, K.; Ohta, N.; Yamamoto, S.; Takagi, Y.; et al. Eosinophil extracellular trap cell death-derived DNA traps: Their presence in secretions and functional attributes. J. Allergy Clin. Immunol 2016, 137, 258–267. [Google Scholar] [CrossRef] [Green Version]
  237. 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, 1–8. [Google Scholar] [CrossRef] [Green Version]
  238. Hwang, C.S.; Park, S.C.; Cho, H.J.; Park, D.J.; Yoon, J.H.; Kim, C.H. Eosinophil extracellular trap formation is closely associated with disease severity in chronic rhinosinusitis regardless of nasal polyp status. Sci. Rep. 2019, 9, 8061. [Google Scholar] [CrossRef] [Green Version]
  239. Liu, P.; Wu, X.; Liao, C.; Liu, X.; Du, J.; Shi, H.; Wang, X.; Bai, X.; Peng, P.; Yu, L.; et al. Escherichia coli and Candida albicans induced macrophage extracellular trap-like structures with limited microbicidal activity. PLoS ONE 2014, 9, e90042. [Google Scholar] [CrossRef] [Green Version]
  240. Braile, M.; Fiorelli, A.; Sorriento, D.; Di Crescenzo, R.M.; Galdiero, M.R.; Marone, G.; Santini, M.; Varricchi, G.; Loffredo, S. Human Lung-Resident Macrophages Express and Are Targets of Thymic Stromal Lymphopoietin in the Tumor Microenvironment. Cells 2021, 10, 2012. [Google Scholar] [CrossRef]
  241. Balestrieri, B.; Granata, F.; Loffredo, S.; Petraroli, A.; Scalia, G.; Morabito, P.; Cardamone, C.; Varricchi, G.; Triggiani, M. Phenotypic and Functional Heterogeneity of Low-Density and High-Density Human Lung Macrophages. Biomedicines 2021, 9, 505. [Google Scholar] [CrossRef] [PubMed]
  242. von Kockritz-Blickwede, M.; Goldmann, O.; Thulin, P.; Heinemann, K.; Norrby-Teglund, A.; Rohde, M.; Medina, E. Phagocytosis-independent antimicrobial activity of mast cells by means of extracellular trap formation. Blood 2008, 111, 3070–3080. [Google Scholar] [CrossRef] [PubMed]
  243. Varricchi, G.; Rossi, F.W.; Galdiero, M.R.; Granata, F.; Criscuolo, G.; Spadaro, G.; de Paulis, A.; Marone, G. Physiological Roles of Mast Cells: Collegium Internationale Allergologicum Update 2019. Int. Arch. Allergy Immunol. 2019, 179, 247–261. [Google Scholar] [CrossRef] [PubMed]
  244. Varricchi, G.; de Paulis, A.; Marone, G.; Galli, S.J. Future Needs in Mast Cell Biology. Int. J. Mol. Sci. 2019, 20, 4397. [Google Scholar] [CrossRef] [Green Version]
  245. Zhang, B.; Asadi, S.; Weng, Z.; Sismanopoulos, N.; Theoharides, T.C. Stimulated human mast cells secrete mitochondrial components that have autocrine and paracrine inflammatory actions. PLoS ONE 2012, 7, e49767. [Google Scholar] [CrossRef]
  246. Varricchi, G.; Poto, R.; Marone, G.; Schroeder, J.T. IL-3 in the development and function of basophils. Semin. Immunol. 2021, 54, 101510. [Google Scholar] [CrossRef]
  247. Canonica, G.W.; Senna, G.; Mitchell, P.D.; O’Byrne, P.M.; Passalacqua, G.; Varricchi, G. Therapeutic interventions in severe asthma. World Allergy Organ. J. 2016, 9, 40. [Google Scholar] [CrossRef] [Green Version]
  248. Ferrando, M.; Bagnasco, D.; Varricchi, G.; Bernardi, S.; Bragantini, A.; Passalacqua, G.; Canonica, G.W. Personalized Medicine in Allergy. Allergy Asthma Immunol. Res. 2017, 9, 15–24. [Google Scholar] [CrossRef] [Green Version]
  249. Brusselle, G.G.; Koppelman, G.H. Biologic Therapies for Severe Asthma. N. Engl. J. Med. 2022, 386, 157–171. [Google Scholar] [CrossRef]
  250. Domer, D.; Walther, T.; Moller, S.; Behnen, M.; Laskay, T. Neutrophil Extracellular Traps Activate Proinflammatory Functions of Human Neutrophils. Front. Immunol. 2021, 12, 636954. [Google Scholar] [CrossRef]
  251. Vargas, A.; Boivin, R.; Cano, P.; Murcia, Y.; Bazin, I.; Lavoie, J.P. Neutrophil extracellular traps are downregulated by glucocorticosteroids in lungs in an equine model of asthma. Respir. Res. 2017, 18, 207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Gal, Z.; Gezsi, A.; Pallinger, E.; Visnovitz, T.; Nagy, A.; Kiss, A.; Sultesz, M.; Csoma, Z.; Tamasi, L.; Galffy, G.; et al. Plasma neutrophil extracellular trap level is modified by disease severity and inhaled corticosteroids in chronic inflammatory lung diseases. Sci. Rep. 2020, 10, 4320. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of biochemical and cellular events driving lytic NET formation. A multitude of extracellular pathogens, including viruses and bacteria, can induce lytic NET formation, also called NETosis [18,75]. Activation of neutrophils induces actin filament disassembly, an increase in intracellular Ca2+ and ROS production. Neutrophil elastase (NE) [68] and myeloperoxidase (MPO) [76] contribute to nuclear membrane permeabilization and unfolding of chromatin. Peptidyl arginine deiminase 4 (PAD4), mainly expressed in the nucleus of granulocytes, mediates citrullination of the nucleosome histones leading to chromatin decondensation [77]. These events favor nuclear rounding and nuclear envelope permeabilization. The following event is the chromatin swelling into the cytoplasm and the activation of gasdermin D [78]. A final step is the plasma membrane rupture and NET release [79].
Figure 1. Schematic representation of biochemical and cellular events driving lytic NET formation. A multitude of extracellular pathogens, including viruses and bacteria, can induce lytic NET formation, also called NETosis [18,75]. Activation of neutrophils induces actin filament disassembly, an increase in intracellular Ca2+ and ROS production. Neutrophil elastase (NE) [68] and myeloperoxidase (MPO) [76] contribute to nuclear membrane permeabilization and unfolding of chromatin. Peptidyl arginine deiminase 4 (PAD4), mainly expressed in the nucleus of granulocytes, mediates citrullination of the nucleosome histones leading to chromatin decondensation [77]. These events favor nuclear rounding and nuclear envelope permeabilization. The following event is the chromatin swelling into the cytoplasm and the activation of gasdermin D [78]. A final step is the plasma membrane rupture and NET release [79].
Cells 11 03521 g001
Figure 2. The most potent inducer of NETosis in vitro is phorbol 12-myristate 13-acetate (PMA), which activates protein kinase C (PKC) in human neutrophils. [15]. Several other stimuli such as agonists of FcγRIIIB [86], crystals [86,87], and the Ca2+ ionophore A23187 or ionomycin [88] can induce suicidal NETosis. NET formation induced by PMA requires NADPH (nicotinamide adenine dinucleotide phosphate) oxidase activity, but the requirement for this enzymatic activity differs depending on the stimulus [89]. Neutrophil elastase (NE) translocates from the cytoplasmic granules to the nucleus and activates its proteolytic activity in a myeloperoxidase (MPO)-dependent manner [90]. In the nucleus, NE degrades specific histones promoting chromatin decondensation [68]. Peptidyl arginine deiminase 4 (PAD4), which converts arginine residues into citrulline [91], catalyzes histone citrullination [69]. This results in a loss of positive charges on arginine residues in histones, which loosen the forces between DNA and histones and contributes to chromatin decondensation [92]. After nuclear envelope disruption, the chromatin comes into contact with several cytoplasmic granules (e.g., NE, MPO, PR3 (proteinase 3), PTX3 (pentraxin 3)) [12]. The rupture of the plasma membrane and the consequent NET release are mediated by gasdermin D [78,93,94]. In vital NET formation, the cell remains intact and normal cellular functions of neutrophils, such as chemotaxis and phagocytosis, can still be carried out [18,72,73,80,95]. This process is biochemically distinct from suicidal NETosis. A variety of stimuli such as bacterial products [73,95], GM-CSF and C5a [72,96], immune complexes [81,96,97], lipopolysaccharide (LPS) [80,98,99], and conditioned media of cancer cells [18] can rapidly (within 30 min) induce vital NET formation. Vital NET formation occurs without nuclear membrane disruption and has been reported with DNA leaving the cytoplasm in vesicles [73,80,95]. DNA becomes decorated with granule proteins (e.g., NE, MPO, PR3) [72,96]. Mitochondria can mediate mitochondrial ROS (mROS) formation [18,97]. (DAMP (damage-associated molecular pattern); E. coli, Escherichia coli; S. aureus, Staphylococcus aureus).
Figure 2. The most potent inducer of NETosis in vitro is phorbol 12-myristate 13-acetate (PMA), which activates protein kinase C (PKC) in human neutrophils. [15]. Several other stimuli such as agonists of FcγRIIIB [86], crystals [86,87], and the Ca2+ ionophore A23187 or ionomycin [88] can induce suicidal NETosis. NET formation induced by PMA requires NADPH (nicotinamide adenine dinucleotide phosphate) oxidase activity, but the requirement for this enzymatic activity differs depending on the stimulus [89]. Neutrophil elastase (NE) translocates from the cytoplasmic granules to the nucleus and activates its proteolytic activity in a myeloperoxidase (MPO)-dependent manner [90]. In the nucleus, NE degrades specific histones promoting chromatin decondensation [68]. Peptidyl arginine deiminase 4 (PAD4), which converts arginine residues into citrulline [91], catalyzes histone citrullination [69]. This results in a loss of positive charges on arginine residues in histones, which loosen the forces between DNA and histones and contributes to chromatin decondensation [92]. After nuclear envelope disruption, the chromatin comes into contact with several cytoplasmic granules (e.g., NE, MPO, PR3 (proteinase 3), PTX3 (pentraxin 3)) [12]. The rupture of the plasma membrane and the consequent NET release are mediated by gasdermin D [78,93,94]. In vital NET formation, the cell remains intact and normal cellular functions of neutrophils, such as chemotaxis and phagocytosis, can still be carried out [18,72,73,80,95]. This process is biochemically distinct from suicidal NETosis. A variety of stimuli such as bacterial products [73,95], GM-CSF and C5a [72,96], immune complexes [81,96,97], lipopolysaccharide (LPS) [80,98,99], and conditioned media of cancer cells [18] can rapidly (within 30 min) induce vital NET formation. Vital NET formation occurs without nuclear membrane disruption and has been reported with DNA leaving the cytoplasm in vesicles [73,80,95]. DNA becomes decorated with granule proteins (e.g., NE, MPO, PR3) [72,96]. Mitochondria can mediate mitochondrial ROS (mROS) formation [18,97]. (DAMP (damage-associated molecular pattern); E. coli, Escherichia coli; S. aureus, Staphylococcus aureus).
Cells 11 03521 g002
Figure 3. Schematic representation of the pleiotropic and potentially conflicting roles of NETs in human and experimental models of asthma. NETs and their components can exert a pathogenic role in asthma. In particular, they can contribute to epithelial and endothelial damage mediated by ROS, MMP-9 or NE [174]. ROS and NE can also favor airway smooth muscle cell proliferation [175]. MMP-9 contributes to airway edema and remodeling [112] and, together with ROS, stimulates mucus production [176]. High-mobility group box 1 protein (HMBG1) induces the release of TSLP, TNF-α, MMP-9 and VEGF-A from bronchial epithelial cells [150]. Histones exert a cytotoxic effect [177], and MMP-9 and NE participate in airway remodeling of the extracellular matrix (ECM) [178]. In an experimental model, NETs promoted rhinovirus-induced type 2 asthma exacerbations [139]. Neutrophil cytoplasts induced TH17 differentiation in a non-type 2 model of asthma [179]. NETs amplify neutrophil recruitment in a model of neutrophilic asthma [180]. NETs promoted neutrophil-associated asthma through the activation of macrophages [181]. In experimental models of asthma, NETs promoted the expression of Th2-like cytokines, airway eosinophil infiltration and airway hyperresponsiveness (AHR) [152]. NETs may not only have pathogenic effects but may also exert beneficial effects in human and experimental models of asthma [9]. NETs can capture and/or kill bacteria [15,182], viruses [161,167,183], and fungi [162,172,184]. NETs can also promote the resolution of inflammation by degrading cytokines and chemokines [168,169]. NETs inhibit GM-CSF/IL-4-induced dendritic cell (DC) differentiation [173]. In experimental models of asthma, MMP-9 reduced ROS accumulation and DNA damage [185].
Figure 3. Schematic representation of the pleiotropic and potentially conflicting roles of NETs in human and experimental models of asthma. NETs and their components can exert a pathogenic role in asthma. In particular, they can contribute to epithelial and endothelial damage mediated by ROS, MMP-9 or NE [174]. ROS and NE can also favor airway smooth muscle cell proliferation [175]. MMP-9 contributes to airway edema and remodeling [112] and, together with ROS, stimulates mucus production [176]. High-mobility group box 1 protein (HMBG1) induces the release of TSLP, TNF-α, MMP-9 and VEGF-A from bronchial epithelial cells [150]. Histones exert a cytotoxic effect [177], and MMP-9 and NE participate in airway remodeling of the extracellular matrix (ECM) [178]. In an experimental model, NETs promoted rhinovirus-induced type 2 asthma exacerbations [139]. Neutrophil cytoplasts induced TH17 differentiation in a non-type 2 model of asthma [179]. NETs amplify neutrophil recruitment in a model of neutrophilic asthma [180]. NETs promoted neutrophil-associated asthma through the activation of macrophages [181]. In experimental models of asthma, NETs promoted the expression of Th2-like cytokines, airway eosinophil infiltration and airway hyperresponsiveness (AHR) [152]. NETs may not only have pathogenic effects but may also exert beneficial effects in human and experimental models of asthma [9]. NETs can capture and/or kill bacteria [15,182], viruses [161,167,183], and fungi [162,172,184]. NETs can also promote the resolution of inflammation by degrading cytokines and chemokines [168,169]. NETs inhibit GM-CSF/IL-4-induced dendritic cell (DC) differentiation [173]. In experimental models of asthma, MMP-9 reduced ROS accumulation and DNA damage [185].
Cells 11 03521 g003
Table 1. Limitations in studying NETs in asthma.
Table 1. Limitations in studying NETs in asthma.
•Most if not all studies on the role of NETs in asthma have been performed on bulk analysis of
neutrophils. Human neutrophils are highly heterogeneous [45,46,47,48], and different subsets of
neutrophils could produce different forms of NETs.
•We do not know the role of neutrophil subsets and their specific mediators, including NETs,
in different phenotypes of asthma.
•We do not know whether subsets of neutrophils make different NETs.
•We do not know whether NETs in different phases (early vs. late) or phenotypes of asthma
(T2-high vs. T2-low) are proinflammatory or anti-inflammatory.
•In human studies, most experimental approaches are limited to ex vivo analyses of NET
formation by blood neutrophils and basic correlations with clinical outcomes. However, these
approaches do not provide insights into the underlying causes of disease.
•Approximately 25% of neutrophils release vital NETs [74]. We do not know the role of non-lytic
and lytic NETs in asthma.
•Primary human neutrophils cannot undergo transfection and it is difficult to specifically inhibit
pathways that lead to NET formation in vivo.
•Several immune cells, such as eosinophils, mast cells, macrophages, and basophils can release
extracellular DNA traps.
•In vivo identification of NETs requires the colocalization of at least three molecules: extracellular
DNA, histones, and neutrophil elastase.
•We do not know whether allergen immunotherapy modulates NET formation in vivo.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Poto, R.; Shamji, M.; Marone, G.; Durham, S.R.; Scadding, G.W.; Varricchi, G. Neutrophil Extracellular Traps in Asthma: Friends or Foes? Cells 2022, 11, 3521. https://doi.org/10.3390/cells11213521

AMA Style

Poto R, Shamji M, Marone G, Durham SR, Scadding GW, Varricchi G. Neutrophil Extracellular Traps in Asthma: Friends or Foes? Cells. 2022; 11(21):3521. https://doi.org/10.3390/cells11213521

Chicago/Turabian Style

Poto, Remo, Mohamed Shamji, Gianni Marone, Stephen R. Durham, Guy W. Scadding, and Gilda Varricchi. 2022. "Neutrophil Extracellular Traps in Asthma: Friends or Foes?" Cells 11, no. 21: 3521. https://doi.org/10.3390/cells11213521

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