Next Article in Journal
SIRT1: A Key Player in Male Reproduction
Previous Article in Journal
Association between Serum Adipocyte Fatty Acid Binding Protein Level and Endothelial Dysfunction in Chronic Hemodialysis Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neutrophil Extracellular Traps in ANCA-Associated Vasculitis and Interstitial Lung Disease: A Scoping Review

1
Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, University of Siena, 53100 Siena, Italy
2
Rheumatology Unit, Department of Medicine, Surgery & Neurosciences, University of Siena, 53100 Siena, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Life 2022, 12(2), 317; https://doi.org/10.3390/life12020317
Submission received: 12 January 2022 / Revised: 10 February 2022 / Accepted: 19 February 2022 / Published: 20 February 2022
(This article belongs to the Section Medical Research)

Abstract

:
Background: Deregulated neutrophil extracellular traps (NETs) formation is implicated in various diseases, including ANCA-associated vasculitis and pulmonary fibrosis (PF). Lung involvement is frequent in AAV, and interstitial lung diseases (ILDs) are strongly related to MPO-ANCA positivity and mainly reported in microscopic polyangiitis. The association between AAV and ILD is a strong indicator of poor prognosis and limited survival. Neutrophils, ANCA and NET interplay in PF development in AAV. This study aimed to review the literature concerning the implications of NET in lung fibrogenesis specifically focused on AAV associated with ILD, and the potential of NET as a theranostic marker. Methods: Through scoping review methodology, we used a descriptive thematic analysis to understand the pathogenic role of NETs in patients with AAV and pulmonary fibrosis and their further role as a theranostic marker of this disease. Results: The implications of NET in the pathogenesis of AAV and ILD, as well as an association between these two diseases, have been identified, but the underlying pathophysiological mechanisms are still unknown. The pharmacological or genetic inhibition of NET release reduces disease severity in multiple inflammatory disease models, indicating that NETs are potential therapeutic targets. In this regard, despite the lack of clinical data, we may hypothesise that an optimal management of AAV-ILD patients would require not only B-cells targeted therapy, but also NETs inhibition. Conclusion: Preliminary findings seem to display a lack of efficacy of traditional immunosuppressants, such as Rituximab, in this subset of patients, while to date no patients suffering from a definite ILD have been enrolled in clinical trials. Further insights would be provided by their employment, as a combination treatment, in common clinical practice. Although we can imagine that the inhibition of NETs in patients with AAV-ILD could reduce severity and mortality, we still lack the scientific basis that could improve our understanding of the disease from a molecular point of view.

Graphical Abstract

1. Introduction

Innate immune cell responses have been linked to fibroblast and myofibroblast biology and fibrogenesis [1]. In particular, the alteration of immune cells (macrophages, neutrophils) in pulmonary fibrosis (PF) significantly impairs antimicrobial host defence functions, promoting susceptibility to infection [2,3].
Pulmonary fibrosis is a chronic progressive respiratory condition determined by an exaggerated dysregulated tissue repair response triggered by infections, cigarette smoke, environmental and occupational pollutants, obesity, diabetes mellitus, gastroesophageal reflux, pulmonary hypertension, chemotherapy and autoimmune disorders (including rheumatoid arthritis, scleroderma, Sjögren’s syndrome and vasculitis) [4,5,6,7,8,9,10,11,12,13,14]. However, it can also manifest without any known aetiology, as in the case of idiopathic pulmonary fibrosis (IPF), the most common and lethal fibrotic interstitial lung disease (ILD) [15,16].
Neutrophils are not only antibacterial effectors, but also influence their tissue environment by releasing proteases, oxidants, cytokines and chemokines [17]. They produce elastase which activates tumour growth factor-beta (TGF-β) and recruits inflammatory cells to the lung, thereby promoting PF [18,19,20].
Neutrophil-mediated injury plays a key role in interstitial fibrosis and abnormal lung repair in IPF [21,22,23]. Several studies have investigated the contribution of neutrophils to the development of fibrosis in terms of extracellular matrix (ECM) homeostasis, which regulates the balance between metal proteinases and metal proteinase inhibitors [24]. Neutrophils are reported to be increased in bronchoalveolar lavage (BAL) fluid from IPF patients and BAL neutrophil percentage has been linked to early mortality [20,25].
Neutrophils react against pathogens through three mechanisms including NETosis [26], which is a form of cell death induced by neutrophils through the release of neutrophil extracellular traps (NETs), which are fibrous DNA structures that protrude from the membranes of activated neutrophils [27,28]. Under physiological conditions, neutrophils release NETs as an antimicrobial defence. However, sterile stimuli, such as phorbol 12-myristate 13-acetate (PMA), monosodium urate and calcium pyrophosphate dehydrate crystals, may also induce NET formation [29]. It was recently demonstrated that neutrophils may remain viable and functional even after NET extrusion, principally when the scaffold is composed of mitochondrial DNA [30].
Two main processes trigger NET formation through chromatin decompensation: (i) the disintegration of nuclear and granular membranes and the release of enzymes, such as neutrophil elastase and myeloperoxidase (MPO) which alter the neutrophil cytoskeleton and decompensate chromatin [31]; and (ii) the enzyme peptidyl arginine deiminase IV (PAD4), which converts arginine residues into citrulline residues via nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase (ΝOΧ), leading to histone deamination, loss of positive charge and chromatin decompensation [32].
In addition to the above, autophagy is also involved in NET formation through mTOR pathway inhibition, which increases autophagosome formation and accelerates NET formation [33,34].
Deregulated NET formation is implicated in various diseases, including autoimmune and autoinflammatory diseases, vein and arterial thrombosis, acute myocardial infarction, cancer and fibrosis [35,36]. Depending on the pathophysiological context of the disease, NETs are composed of different proteins that may account for their differential contribution to disease pathogenesis and phenotype [37,38]. In rheumatoid arthritis [39,40], anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) [41,42] and systemic lupus erythematosus [43,44], NETs are rich in immunogenic autoantigens and damage-associated molecular patterns, while NETs enriched in IL-17 promote fibrosis in ILD [45].
Here, we reviewed the literature concerning the implications of NETs in lung fibrogenesis. We also specifically focused on AAV associated with ILD and the potential of NET as a theranostic marker. There is growing interest in the involvement of NETs in inflammation and in the ensuing fibrosis.

2. Methods

A scoping review methodology was used according to the scoping review protocol [46]. We used descriptive thematic analysis to understand the pathogenic role of NETs in patients with AAV and pulmonary fibrosis and their further role as a theranostic marker of this disease. This article conforms with the Scale for Assessment of Narrative Review Articles (SANRA) guidelines [47].

2.1. Eligibility Criteria

The inclusion criteria were peer-reviewed, empirical or perspective papers with (a) relevance to the study topic: neutrophil extracellular traps, ANCA-associated vasculitis, pulmonary fibrosis and interstitial lung diseases; (b) language: English; (c) type of journal: preferences for journals related to pneumology and rheumatology with full text; (d) type of study: original article and multicentre study. Studies were excluded if (a) they were not relevant to the study topic; (b) they were written in languages other than English; (c) they did not adequately report objectives and conclusions; (d) they did not carry full text; (e) they were reviews, systematic reviews, meta-analyses, case reports, case series, correspondence or clinical trials.

2.2. Information Sources and Search

A systematic search of the literature was conducted in the PubMed online database. The Boolean search syntax was: neutrophil extracellular traps AND (“pulmonary fibrosis” OR “interstitial lung disease” OR “pulmonary fibrosis, vasculitis” OR “ANCA-associated vasculitis”). The search was limited to the English language and full texts. We did not include the grey literature (e.g., official reports from international organisations).

2.3. Selection Process

The abstract and title screenings and the full-text assessments were made against the eligibility criteria and were conducted by two independent reviewers (M.d. and E.C.) following pilot screenings with at least 80% agreement, and overseen by the leading review author (M.d.). Any discrepancies were resolved through consensus or the leading author’s input.

2.4. Data Charting and Items

Applying a coding structure designed by members of the research team, one author (M.d.) extracted the formal data elements (publication type, sources, geographies, objectives and main findings) with a random 5% verified by another author (L.B.). Regarding the included literature content, three independent reviewers (B.F., L.C. and E.B.) extracted the text quotations on (1) NETs in lung fibrosis or (2) NETs in vasculitis or (3) NETs in AAV lung fibrosis. These independent extractions were later paired for qualitative data synthesis, which was also independently informed by a brief synthesis of the papers by two reviewers. Then, the content of these extractions was merged (i.e., presented as the combined extractions of all reviewers) and the reviewers’ syntheses of the papers were combined. Figure 1 shows a flowchart of the selected articles.

3. Results

3.1. Synthesis of the Results: Descriptive Data

The search yielded entire texts. We selected 20 studies: 6 included analyses of NETs in pulmonary fibrosis (including ILD, COVID-19 and post-COVID-19 fibrosis); 1 of which proposed the involvement of NETs in the pathogenesis of IPF; 11 analysed the role of NETs in the pathogenesis of AAV (including GPA, MPA and EGPA); and 3 articles suggested the potential of NET inhibition as a therapeutic strategy for human fibrosis diseases. All these articles presented original data, albeit sometimes in experimental models.

3.2. Quality Assessment by SANRA Guidelines

The results of SANRA are reported in Table 1. All 60 ratings (3 raters × 20 manuscripts) were used for statistical analysis. The mean sum score across all 20 manuscripts was 8.0 out of 12 possible points (SD 0.0, range 7–8, median 8). The highest scores were rated for item 5 (scientific reasoning) (mean 1.8; SD 0.44); item 1 (justification of the article’s importance for the readership) (mean 1.7; SD 0.46); item 6 (appropriate presentation of data) (mean 1.62; SD 0.49); and item 2 (statement of concrete aims or formulation of questions) (mean 1.5; SD 0.51)—whereas item 4 had the lowest score (mean 1.3; SD 0.46). Item 3 had a score of 0 because one of the eligibility criteria excluded reviews of the literature.

3.3. NETs in Fibrotic Interstitial Lung Diseases

A pathogenic role of NETs has been described for many human diseases and the detrimental effects of excessive release of NETs are particularly significant for lung diseases, since this expands the alveoli, causing lung injury in asthma, chronic obstructive pulmonary disease, ILD and post-COVID-19-fibrosis, etc. [48,49,50,51,52,53]. Moreover, NETs and their associated molecules can directly induce epithelial and endothelial cell death [54]. NET formation must therefore be closely regulated to prevent NET-mediated tissue damage [31]. Recent therapies targeting NETs in lung diseases include the disintegration of DNA with human recombinant DNase, and the neutralisation of NET proteins with anti-histone antibodies and protease inhibitors [55]. ILD is often associated with specific environmental exposure or an underlying connective tissue disease [31]. An increase in circulating cell-free NETs and plasma LL-37 (a NET component) as well as lower DNase activity were reported in autoimmune rheumatic diseases associated with ILD, suggesting that prolonged exposure to NETs is involved in the development of ILD [56].
In vitro, NETs have been demonstrated to promote the activation of lung fibroblasts and their differentiation into a myofibroblast phenotype. Interestingly, these fibrotic effects were significantly less frequent after the degradation of NETs with DNase [45]. Confirming these findings, the detection of NETs close to alpha-smooth muscle actin-expressing fibroblasts was demonstrated in biopsies from patients with fibrotic ILD [31], probably mediated by the directed effect of neutrophil elastase [57]. Accordingly, a neutrophil elastase inhibitor attenuated pulmonary fibrosis in a murine model through the inhibition of TGF-β1 and inflammatory cell recruitment to the lungs [58]. Some authors demonstrated that the release of NETs by neutrophils treated with fibrosis-related agents (i.e., bleomycin) or with generic NET inducers (i.e., PMA) induced the activation of fibroblasts and their differentiation into the myofibroblast phenotype [59]. Altogether, these findings point to a key role of NETs in the development and progression of PF. Recently, Suzuki and colleagues identified NETs in the alveolar and interstitial lung space of mice undergoing bleomycin-induced lung fibrosis, which was suppressed by a pan-PAD inhibitor, demonstrating that Padi4 gene knockout in mice led to the alleviation of bleomycin-induced NETs and pulmonary fibrosis; this finding suggests that this pathway could be useful as a therapeutic target in the treatment of pulmonary fibrosis [60].

3.4. NETs in ANCA-Associated Vasculitis

ANCA-associated vasculitis (AAV) is life-threatening small vessel inflammation, affecting the kidneys, upper and lower airways, skin and central and peripheral nervous systems [61,62]. Three different clinical types of AAV can be distinguished: granulomatosis with polyangiitis (GPA); microscopic polyangiitis (MPA); and eosinophilic granulomatosis with polyangiitis (EGPA) [63]. Most evidence on the role of ANCA in the pathophysiology of these diseases comes from animal models [64]. ANCA participates in the migration and activation of neutrophils causing ROS and protease release through cell degranulation [65]. Various stimuli activate neutrophils, causing damage to the endothelium with positive feedback from activated endothelial cells that recruit monocytes and T cells, releasing pro-inflammatory mediators [19,21,24]. Monocytes express myeloperoxidase (MPO) and proteinase 3 (PR-3) on their surfaces, and ANCA binds to these molecules, activating monocytes and secreting cytokines (such as IL-8 and monocyte chemoattractant protein-1). In the case of MPO-AAV and PR3-AAV, there is a loss of immunological tolerance to neutrophil enzymes MPO and PR3, which in turn generates ANCA- and MPO-specific T cells. Neutrophils activated by ANCA induce cell cytotoxicity and cause direct tissue injury, thus inducing systemic vasculitis. In up to 90% of patients, ANCA is directed against MPO and PR-3 in neutrophil granules and macrophage lysosomes [66].
It has been reported that neutrophil priming and activation via a complement system (C5a and C5aR interactions) exteriorise MPO (or PR3) on their cell surfaces which MPO-ANCA (or PR3 ANCA) binds to. These neutrophils are home to the microvasculature of the glomeruli involving selectins which play a role in reducing the speed of the neutrophils in the circulation which allows it to roll. The second signal comes from the neutrophil surface integrins β1 and β2 which interact with the ICAM-1 and ICAM-2 ligands on the inflamed endothelium; then, the rolling neutrophils slow down and crawl along the endothelium mediated by integrins to the site of transmigration from the capillaries into the interstitium. Neutrophils either degranulate or release NETs, causing direct injury through the release of enzymes, ROS and proteases [67]. Excess neutrophils, MPO-specific T cells, and macrophages (via a delayed type of hypersensitivity mechanism) generate a vicious cycle of injury which causes glomerular injury.
NETs contain proinflammatory proteins and are thought to directly contribute to vessel inflammation by damaging endothelial cells and by activating the complement system and indirectly contribute to vessel inflammation by acting as a link between the innate and adaptive immune system through the generation of PR3- and MPO-ANCA.
Interestingly, increased levels of NETosis-derived products (or NET remnants) in circulation have been reported in a small number of patients with active vasculitis [68]. Soderberg and colleagues analysed for the first time increased levels of NET remnants in a larger cohort of AAV patients with active disease and an inverse correlation between such levels and ANCA at least during remission [69].
The production and release of NETs by activated neutrophils have also recently been implicated in the pathophysiology of AAV [70,71,72,73,74,75], although the precise mechanisms are not yet known. It is speculated that aberrant NETs could be autoantigens for ANCA underlying the development of MPA, and that the in vivo inhibition of NETs could be a therapeutic strategy for the disease [76]. Kimura and colleagues devised a mouse model of MPA, induced by the administration of a novel extract from Candida albicans and lacking phosphoinositide 3-kinase gamma (PI3K-gamma), and demonstrated the accumulation of NETs in vivo, the elevation of ANCA titres, small-vessel vasculitis and crescentic glomerulonephdritis [76]. The blockade of PI3K-gamma reduced these abnormalities, indicating its potential as a therapeutic molecule [76]. A group of Japanese researchers demonstrated the efficacy of intravenous immunoglobulin therapy in the inhibition of NET formation induced by PMA in vitro, improving the development of MPO-AAV [77]. A role of platelets stimulated via Toll-like receptor (TLR) pathways in NET formation was recently reported, although the underlying mechanism is unclear [78]. Matsumoto et al. cultured platelets from AAV patients, treating them with a TLR-agonist CXCL4 and/or with anti-CXCL4 antibody. They showed that neutralising the anti-CXCL4 antibody significantly inhibited NET formation [79].
Interestingly, some authors showed that neutrophils from AAV patients are less susceptible to apoptosis [80], suggesting that these neutrophils are more prone to other forms of cell death. Indeed, in vitro studies have shown that neutrophils from AAV patients spontaneously release more NETs than neutrophils from healthy blood donors [69,81,82]. These populations of neutrophils were reported by Greyson et al. as low-density granulocytes (LDGs) and have been proposed to be the major source of NETs in AAV [81]. However, the same authors showed that normal-density neutrophils from AAV patients spontaneously released more NETs than normal-density neutrophils from healthy blood donors.
Thus, future in vitro studies of NETs need to take account that during the neutrophil’s isolation procedures from peripheral whole blood, LDGs will not be included because they will be found in the fraction of peripheral blood mononuclear cells.
Few papers reported the implication of NETs in two clinically and pathologically different diseases, AAV and SLE. Van Dam and colleagues compared the mechanisms of NET formation and the composition of NETs in AAV and SLE. They revealed the differences between AAV and SLE in terms of NET formation and provided a better understanding of the pathophysiologic role of NETs in these different autoimmune diseases [83].
Alternative complement pathway activation plays a role in the pathogenesis of AAV [84]; its inhibition, mainly targeting C5a component, is of interest due to its role in neutrophil activation and migration [85]. This provides a rationale for using avacopan (CCX168), an oral C5a inhibitor recently approved by the FDA for the treatment of GPA and MPA, on the basis of an ADVOCATE trial which displayed a non-inferiority to prednisone with respect to remission at week 26 and superiority with respect to sustained remission at week 52 [86].

3.5. NETs in ANCA-Associated Vasculitis with Pulmonary Fibrosis

Pulmonary manifestations are frequent in AAV [87]. They range from nodules and transient infiltrates, which are more frequent in GPA and EGPA [88], respectively, alveolar haemorrhage, which can be the presenting sign of MPA, to ILD, which is mainly reported in MPA and strongly related to MPO-ANCA positivity [89]. The association between AAV and ILD is a strong indicator of poor prognosis and limited survival. Early detection of this association is crucial to the management of these patients, although no specific treatments have been proposed to date.
Pulmonary fibrosis in patients with MPA can lead to chronic respiratory failure and increased risk of potentially fatal acute exacerbations. The exact mechanisms involved in fibrosis development in AAV patients have been suggested to include the relation between the release of NETs [90] and the development of PF, although the pathobiological pathways are far from clear. It is speculated that neutrophils, ANCA and NETs interact to cause PF in AAV [91]: activated neutrophils release NETs that reach the endothelium or interstitium, participating in all stages of the development of PF. This assumption is in line with the general pathogenetic model of CTD-ILD proposed by Wells and Denton [92]: in CTD-ILD patients, the lung disorder develops in three stages: initiation, progression and failed resolution. After inflammation and endothelial activation, which result in epithelial damage, fibroblasts are recruited, and their proliferation and activation mark the transition to the progression phase. The specific molecular pathways of ILD development in AAV are still largely unknown, however, Negreros and Flores-Suárez postulated that NET-associated proteins (NAPs) may act as mediators in the development of fibrosis through the modification of ECM composition, increasing levels of collagen, osteopontin and fibronectin [91]. NET-associated proteins increase the number and activity of fibroblasts and profibrotic cytokines and inhibited the fibroblast apoptosis. The more NETs persist in the interstitium, the more NAPs affect fibroblasts. In the event of failure to remove NETs due to their resistance to degradation and low DNAse I activity in serum, AAV develops, and the effects of NAPs are maintained [91].
A few months ago, Chirivi and colleagues described the inhibition of NETs with therapeutic anti-citrullinated protein antibody (tACPA) for the first time in mice models. NET-mediated inflammatory diseases (including inflammatory arthritis and pulmonary fibrosis) enable eliminating the noxious triggers that lead to persistent inflammation and tissue damage [93].
In the initial stage of lung fibrosis in patients with AAV, any modifications of the interstitial microenvironment and resident cell behaviour exerted by neutrophils and NETs in the interstitial lung space have often been overlooked. Although some authors have demonstrated the effects of NETs on lung fibroblast proliferation and differentiation [91], the mechanisms by which NETs exert these effects remain unclear.

4. Conclusions

The implications of NETs in the pathogenesis of AAV and ILD, as well as an association between these two diseases, have been identified, however, the underlying pathophysiological mechanisms are still unknown. NETs contain proinflammatory proteins and are thought to directly contribute to vessel inflammation by damaging endothelial cells and by activating the complement system and indirectly contribute to vessel inflammation by acting as a link between the innate and adaptive immune system through the generation of PR3- and MPO-ANCA.
Although not strictly related to a complement pathway nor to the direct neutrophils’ activation, it is worth mentioning that several other cytokines seem to play a crucial role in the pathogenesis of AAV. In this regard, serum IL-1, IL-32 and IL-6 [94,95,96] seem to be elevated in patients suffering from AAV and directly correlate with disease activity [97], namely skin vasculitis in patients with GPA and high levels of ANCA-PR3 [95], also displaying a direct correlation with the risk of relapse [98] among the patients treated with anti-CD20 agents.
On the other hand, despite such interesting insights which may have paved the way for the use of Tocilizumab in AAV [96,97], anti-IL-6 agents have provided scanty and disappointing data [99] and their real-life use is restricted to case reports with doubtful diagnosis [100] or small case series.
The pharmacological or genetic inhibition of NET release reduces disease severity in multiple inflammatory disease models, indicating that NETs are potential therapeutic targets.
Despite the lack of clinical data, we may hypothesise that an optimal management of AAV-ILD patients would require not only B-cells’ targeted therapy but also NET inhibition.
Preliminary insights from avacopan registrative studies [101,102,103] suggest that the inhibition of a complement pathway, which is in turn related to NET activation, may be a promising option in the treatment of AAV. Nevertheless, the exact role of a complement pathway in the pathogenesis and disease activity of AAV is still matter of debate: while some papers have suggested that hypocomplementemia may be associated with a poorer renal outcome [104,105], other have not evidenced any difference in terms of C3 and C4 serum levels in AAV patients when compared with subjects affected by other kidney diseases [106].
Preliminary findings seem to display a lack of efficacy of traditional immunosuppressants, such as Rituximab, in this subset of patients, while to date, no patients suffering from a definite ILD has been enrolled in clinical trials. Further insights would be provided by their employment, as a combination treatment, in common clinical practice.
Although it can be speculated that NETs in patients with AAV-ILD could reduce severity and mortality, a scientific basis that could improve our understanding of the disease from a molecular point of view is lacking.

Author Contributions

Conceptualisation, E.C. and M.d.; methodology, M.d. and L.B.; software, M.d.; validation, M.d., P.C., L.C., E.B. and B.F.; writing—original draft preparation, E.C., M.d., L.B., P.C., L.C., B.F. and E.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available upon request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Huang, E.; Peng, N.; Xiao, F.; Hu, D.; Wang, X.; Lu, L. The Roles of Immune Cells in the Pathogenesis of Fibrosis. Int. J. Mol. Sci. 2020, 21, 5203. [Google Scholar] [CrossRef] [PubMed]
  2. Wynn, T.A. Cellular and Molecular Mechanisms of Fibrosis. J. Pathol. 2008, 214, 199–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Del Principe, D.; Vona, R.; Giordani, L.; Straface, E.; Giammarioli, A.M. Defective Autophagy in Fibroblasts May Contribute to Fibrogenesis in Autoimmune Processes. Curr. Pharm. Des. 2011, 17, 3878–3887. [Google Scholar] [CrossRef] [PubMed]
  4. Meyer, K.C. Pulmonary Fibrosis, Part I: Epidemiology, Pathogenesis, and Diagnosis. Expert Rev. Respir. Med. 2017, 11, 343–359. [Google Scholar] [CrossRef] [PubMed]
  5. Thannickal, V.J.; Toews, G.B.; White, E.S.; Lynch, J.P.; Martinez, F.J. Mechanisms of Pulmonary Fibrosis. Annu. Rev. Med. 2004, 55, 395–417. [Google Scholar] [CrossRef]
  6. Noble, P.W.; Barkauskas, C.E.; Jiang, D. Pulmonary Fibrosis: Patterns and Perpetrators. J. Clin. Investig. 2012, 122, 2756–2762. [Google Scholar] [CrossRef] [Green Version]
  7. d’Alessandro, M.; Bergantini, L.; Cameli, P.; Lanzarone, N.; Perillo, F.; Perrone, A.; Bargagli, E. BAL and Serum Multiplex Lipid Profiling in Idiopathic Pulmonary Fibrosis and Fibrotic Hypersensitivity Pneumonitis. Life Sci. 2020, 256, 117995. [Google Scholar] [CrossRef]
  8. d’Alessandro, M.; Bergantini, L.; Refini, R.M.; Cameli, P.; Perillo, F.; Landi, C.; Icorne, F.; Perrone, A.; Sestini, P.; Bonella, F.; et al. Adiponectin and Leptin Levels in Idiopathic Pulmonary Fibrosis: A New Method for BAL and Serum Assessment. Immunobiology 2020, 225, 151997. [Google Scholar] [CrossRef]
  9. d’Alessandro, M.; Carleo, A.; Cameli, P.; Bergantini, L.; Perrone, A.; Vietri, L.; Lanzarone, N.; Vagaggini, C.; Sestini, P.; Bargagli, E. BAL Biomarkers’ Panel for Differential Diagnosis of Interstitial Lung Diseases. Clin. Exp. Med. 2020, 20, 207–216. [Google Scholar] [CrossRef]
  10. d’Alessandro, M.; Bergantini, L.; Cameli, P.; Vietri, L.; Lanzarone, N.; Alonzi, V.; Pieroni, M.; Refini, R.M.; Sestini, P.; Bonella, F.; et al. Krebs von Den Lungen-6 as a Biomarker for Disease Severity Assessment in Interstitial Lung Disease: A Comprehensive Review. Biomark. Med. 2020, 14, 665–674. [Google Scholar] [CrossRef]
  11. d’Alessandro, M.; Bergantini, L.; Cameli, P.; Fanetti, M.; Alderighi, L.; Armati, M.; Refini, R.M.; Alonzi, V.; Sestini, P.; Bargagli, E. Immunologic Responses to Antifibrotic Treatment in IPF Patients. Int. Immunopharmacol. 2021, 95, 107525. [Google Scholar] [CrossRef] [PubMed]
  12. d’Alessandro, M.; Bergantini, L.; Cameli, P.; Pieroni, M.; Refini, R.M.; Sestini, P.; Bargagli, E. Serum Concentrations of KL-6 in Patients with IPF and Lung Cancer and Serial Measurements of KL-6 in IPF Patients Treated with Antifibrotic Therapy. Cancers 2021, 13, 689. [Google Scholar] [CrossRef] [PubMed]
  13. d’Alessandro, M.; Bergantini, L.; Torricelli, E.; Cameli, P.; Lavorini, F.; Pieroni, M.; Refini, R.M.; Sestini, P.; Bargagli, E. Systematic Review and Metanalysis of Oncomarkers in IPF Patients and Serial Changes of Oncomarkers in a Prospective Italian Real-Life Case Series. Cancers 2021, 13, 539. [Google Scholar] [CrossRef] [PubMed]
  14. Bargagli, E.; Conticini, E.; Mazzei, M.A.; Cameli, P.; Guerrini, S.; d’Alessandro, M.; Frediani, B.; Pleuroparenchymal Fibroelastosis (PPFE) Siena Unit, Italy; Rheumatology Unit Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Italy; Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy. Pleuroparenchymal Fibroelastosis in Interstitial Lung Disease with Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Clin. Exp. Rheumatol. 2021, 39 (Suppl. 129), 190. [Google Scholar]
  15. Richeldi, L.; Collard, H.R.; Jones, M.G. Idiopathic Pulmonary Fibrosis. Lancet 2017, 389, 1941–1952. [Google Scholar] [CrossRef]
  16. King, T.E.; Pardo, A.; Selman, M. Idiopathic Pulmonary Fibrosis. Lancet 2011, 378, 1949–1961. [Google Scholar] [CrossRef]
  17. Kumar, S.; Dikshit, M. Metabolic Insight of Neutrophils in Health and Disease. Front. Immunol. 2019, 10, 2099. [Google Scholar] [CrossRef]
  18. Qing, C. The Molecular Biology in Wound Healing & Non-Healing Wound. Chin. J. Traumatol. 2017, 20, 189–193. [Google Scholar] [CrossRef]
  19. Wang, J. Neutrophils in Tissue Injury and Repair. Cell Tissue Res. 2018, 371, 531–539. [Google Scholar] [CrossRef] [Green Version]
  20. D’alessandro, M.; Bergantini, L.; Carleo, A.; Cameli, P.; Perrone, A.; Fossi, A.; Sestini, P.; Bargagli, E. Neutrophil-to-Lymphocyte Ratio in Bronchoalveolar Lavage from IPF Patients: A Novel Prognostic Biomarker? Minerva Med. 2020. [Google Scholar] [CrossRef]
  21. Kruger, P.; Saffarzadeh, M.; Weber, A.N.R.; Rieber, N.; Radsak, M.; von Bernuth, H.; Benarafa, C.; Roos, D.; Skokowa, J.; Hartl, D. Neutrophils: Between Host Defence, Immune Modulation, and Tissue Injury. PLoS Pathog. 2015, 11, e1004651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Peiseler, M.; Kubes, P. More Friend than Foe: The Emerging Role of Neutrophils in Tissue Repair. J. Clin. Investig. 2019, 129, 2629–2639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Nathan, S.D.; Brown, A.W.; Mogulkoc, N.; Soares, F.; Collins, A.C.; Cheng, J.; Peterson, J.; Cannon, B.; King, C.S.; Barnett, S.D. The Association between White Blood Cell Count and Outcomes in Patients with Idiopathic Pulmonary Fibrosis. Respir. Med. 2020, 170, 106068. [Google Scholar] [CrossRef] [PubMed]
  24. Germann, M.; Zangger, N.; Sauvain, M.-O.; Sempoux, C.; Bowler, A.D.; Wirapati, P.; Kandalaft, L.E.; Delorenzi, M.; Tejpar, S.; Coukos, G.; et al. Neutrophils Suppress Tumor-Infiltrating T Cells in Colon Cancer via Matrix Metalloproteinase-Mediated Activation of TGFβ. EMBO Mol. Med. 2020, 12, e10681. [Google Scholar] [CrossRef]
  25. Kinder, B.W.; Brown, K.K.; Schwarz, M.I.; Ix, J.H.; Kervitsky, A.; King, T.E. Baseline BAL Neutrophilia Predicts Early Mortality in Idiopathic Pulmonary Fibrosis. Chest 2008, 133, 226–232. [Google Scholar] [CrossRef]
  26. Sørensen, O.E.; Borregaard, N. Neutrophil Extracellular Traps—The Dark Side of Neutrophils. J. Clin. Investig. 2016, 126, 1612–1620. [Google Scholar] [CrossRef] [Green Version]
  27. Vorobjeva, N.V.; Chernyak, B.V. NETosis: Molecular Mechanisms, Role in Physiology and Pathology. Biochemistry 2020, 85, 1178–1190. [Google Scholar] [CrossRef]
  28. 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]
  29. Masuda, S.; Shimizu, S.; Matsuo, J.; Nishibata, Y.; Kusunoki, Y.; Hattanda, F.; Shida, H.; Nakazawa, D.; Tomaru, U.; Atsumi, T.; et al. Measurement of NET Formation in Vitro and in Vivo by Flow Cytometry. Cytometry A 2017, 91, 822–829. [Google Scholar] [CrossRef] [Green Version]
  30. Takishita, Y.; Yasuda, H.; Shimizu, M.; Matsuo, A.; Morita, A.; Tsutsumi, T.; Tsuchiya, M.; Sato, E.F. Formation of Neutrophil Extracellular Traps in Mitochondrial DNA-Deficient Cells. J. Clin. Biochem. Nutr. 2020, 66, 15–23. [Google Scholar] [CrossRef]
  31. 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] [PubMed] [Green Version]
  32. Lewis, H.D.; Liddle, J.; Coote, J.E.; Atkinson, S.J.; Barker, M.D.; Bax, B.D.; Bicker, K.L.; Bingham, R.P.; Campbell, M.; Chen, Y.H.; et al. Inhibition of PAD4 Activity Is Sufficient to Disrupt Mouse and Human NET Formation. Nat. Chem. Biol. 2015, 11, 189–191. [Google Scholar] [CrossRef] [PubMed]
  33. Mao, J.-Y.; Zhang, J.-H.; Cheng, W.; Chen, J.-W.; Cui, N. Effects of Neutrophil Extracellular Traps in Patients With Septic Coagulopathy and Their Interaction With Autophagy. Front. Immunol. 2021, 12, 757041. [Google Scholar] [CrossRef]
  34. Tang, H.; Tan, C.; Cao, X.; Liu, Y.; Zhao, H.; Liu, Y.; Zhao, Y. NFIL3 Facilitates Neutrophil Autophagy, Neutrophil Extracellular Trap Formation and Inflammation During Gout via REDD1-Dependent MTOR Inactivation. Front. Med. 2021, 8, 692781. [Google Scholar] [CrossRef] [PubMed]
  35. Papayannopoulos, V. Neutrophil Extracellular Traps in Immunity and Disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef] [PubMed]
  36. Dąbrowska, D.; Jabłońska, E.; Garley, M.; Ratajczak-Wrona, W.; Iwaniuk, A. New Aspects of the Biology of Neutrophil Extracellular Traps. Scand. J. Immunol. 2016, 84, 317–322. [Google Scholar] [CrossRef]
  37. Hasler, P.; Giaglis, S.; Hahn, S. Neutrophil Extracellular Traps in Health and Disease. Swiss. Med. Wkly. 2016, 146, w14352. [Google Scholar] [CrossRef] [Green Version]
  38. Grayson, P.C.; Kaplan, M.J. At the Bench: Neutrophil Extracellular Traps (NETs) Highlight Novel Aspects of Innate Immune System Involvement in Autoimmune Diseases. J. Leukoc. Biol. 2016, 99, 253–264. [Google Scholar] [CrossRef] [Green Version]
  39. Karmakar, U.; Vermeren, S. Crosstalk between B Cells and Neutrophils in Rheumatoid Arthritis. Immunology 2021, 164, 689–700. [Google Scholar] [CrossRef]
  40. Carmona-Rivera, C.; Carlucci, P.M.; Goel, R.R.; James, E.; Brooks, S.R.; Rims, C.; Hoffmann, V.; Fox, D.A.; Buckner, J.H.; Kaplan, M.J. Neutrophil Extracellular Traps Mediate Articular Cartilage Damage and Enhance Cartilage Component Immunogenicity in Rheumatoid Arthritis. JCI Insight 2020, 5, 139388. [Google Scholar] [CrossRef]
  41. Söderberg, D.; Segelmark, M. Neutrophil Extracellular Traps in ANCA-Associated Vasculitis. Front. Immunol. 2016, 7, 256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Lee, K.H.; Kronbichler, A.; Park, D.D.-Y.; Park, Y.; Moon, H.; Kim, H.; Choi, J.H.; Choi, Y.; Shim, S.; Lyu, I.S.; et al. Neutrophil Extracellular Traps (NETs) in Autoimmune Diseases: A Comprehensive Review. Autoimmun. Rev. 2017, 16, 1160–1173. [Google Scholar] [CrossRef] [PubMed]
  43. Bruschi, M.; Bonanni, A.; Petretto, A.; Vaglio, A.; Pratesi, F.; Santucci, L.; Migliorini, P.; Bertelli, R.; Galetti, M.; Belletti, S.; et al. Neutrophil Extracellular Traps Profiles in Patients with Incident Systemic Lupus Erythematosus and Lupus Nephritis. J. Rheumatol. 2020, 47, 377–386. [Google Scholar] [CrossRef] [PubMed]
  44. Chapman, E.A.; Lyon, M.; Simpson, D.; Mason, D.; Beynon, R.J.; Moots, R.J.; Wright, H.L. Caught in a Trap? Proteomic Analysis of Neutrophil Extracellular Traps in Rheumatoid Arthritis and Systemic Lupus Erythematosus. Front. Immunol. 2019, 10, 423. [Google Scholar] [CrossRef]
  45. Chrysanthopoulou, A.; Mitroulis, I.; Apostolidou, E.; Arelaki, S.; Mikroulis, D.; Konstantinidis, T.; Sivridis, E.; Koffa, M.; Giatromanolaki, A.; Boumpas, D.T.; et al. Neutrophil Extracellular Traps Promote Differentiation and Function of Fibroblasts. J. Pathol. 2014, 233, 294–307. [Google Scholar] [CrossRef]
  46. Colquhoun, H.L.; Jesus, T.S.; O’Brien, K.K.; Tricco, A.C.; Chui, A.; Zarin, W.; Lillie, E.; Hitzig, S.L.; Straus, S. Study Protocol for a Scoping Review on Rehabilitation Scoping Reviews. Clin. Rehabil. 2017, 31, 1249–1256. [Google Scholar] [CrossRef]
  47. Baethge, C.; Goldbeck-Wood, S.; Mertens, S. SANRA-a Scale for the Quality Assessment of Narrative Review Articles. Res. Integr. Peer Rev. 2019, 4, 5. [Google Scholar] [CrossRef] [Green Version]
  48. Racanelli, A.C.; Kikkers, S.A.; Choi, A.M.K.; Cloonan, S.M. Autophagy and Inflammation in Chronic Respiratory Disease. Autophagy 2018, 14, 221–232. [Google Scholar] [CrossRef]
  49. Khawaja, A.A.; Chong, D.L.W.; Sahota, J.; Mikolasch, T.A.; Pericleous, C.; Ripoll, V.M.; Booth, H.L.; Khan, S.; Rodriguez-Justo, M.; Giles, I.P.; et al. Identification of a Novel HIF-1α-AMβ2 Integrin-NET Axis in Fibrotic Interstitial Lung Disease. Front. Immunol. 2020, 11, 2190. [Google Scholar] [CrossRef]
  50. Uddin, M.; Watz, H.; Malmgren, A.; Pedersen, F. NETopathic Inflammation in Chronic Obstructive Pulmonary Disease and Severe Asthma. Front. Immunol. 2019, 10, 47. [Google Scholar] [CrossRef]
  51. Pandolfi, L.; Bozzini, S.; Frangipane, V.; Percivalle, E.; De Luigi, A.; Violatto, M.B.; Lopez, G.; Gabanti, E.; Carsana, L.; D’Amato, M.; et al. Neutrophil Extracellular Traps Induce the Epithelial-Mesenchymal Transition: Implications in Post-COVID-19 Fibrosis. Front. Immunol. 2021, 12, 663303. [Google Scholar] [CrossRef] [PubMed]
  52. Radermecker, C.; Detrembleur, N.; Guiot, J.; Cavalier, E.; Henket, M.; d’Emal, C.; Vanwinge, C.; Cataldo, D.; Oury, C.; Delvenne, P.; et al. Neutrophil Extracellular Traps Infiltrate the Lung Airway, Interstitial, and Vascular Compartments in Severe COVID-19. J. Exp. Med. 2020, 217, e20201012. [Google Scholar] [CrossRef] [PubMed]
  53. Skopelja-Gardner, S.; Theprungsirikul, J.; Lewis, K.A.; Hammond, J.H.; Carlson, K.M.; Hazlett, H.F.; Nymon, A.; Nguyen, D.; Berwin, B.L.; Hogan, D.A.; et al. Regulation of Pseudomonas Aeruginosa-Mediated Neutrophil Extracellular Traps. Front. Immunol. 2019, 10, 1670. [Google Scholar] [CrossRef] [Green Version]
  54. Saffarzadeh, M.; Juenemann, C.; Queisser, M.A.; Lochnit, G.; Barreto, G.; Galuska, S.P.; Lohmeyer, J.; Preissner, K.T. Neutrophil Extracellular Traps Directly Induce Epithelial and Endothelial Cell Death: A Predominant Role of Histones. PLoS ONE 2012, 7, e32366. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, Y.; Guan, L.; Yu, J.; Zhao, Z.; Mao, L.; Li, S.; Zhao, J. Pulmonary Endothelial Activation Caused by Extracellular Histones Contributes to Neutrophil Activation in Acute Respiratory Distress Syndrome. Respir. Res. 2016, 17, 155. [Google Scholar] [CrossRef] [Green Version]
  56. Hosoda, H.; Nakamura, K.; Hu, Z.; Tamura, H.; Reich, J.; Kuwahara-Arai, K.; Iba, T.; Tabe, Y.; Nagaoaka, I. Antimicrobial Cathelicidin Peptide LL-37 Induces NET Formation and Suppresses the Inflammatory Response in a Mouse Septic Model. Mol. Med. Rep. 2017, 16, 5618–5626. [Google Scholar] [CrossRef]
  57. Gregory, A.D.; Kliment, C.R.; Metz, H.E.; Kim, K.-H.; Kargl, J.; Agostini, B.A.; Crum, L.T.; Oczypok, E.A.; Oury, T.A.; Houghton, A.M. Neutrophil Elastase Promotes Myofibroblast Differentiation in Lung Fibrosis. J. Leukoc. Biol. 2015, 98, 143–152. [Google Scholar] [CrossRef] [Green Version]
  58. Takemasa, A.; Ishii, Y.; Fukuda, T. A Neutrophil Elastase Inhibitor Prevents Bleomycin-Induced Pulmonary Fibrosis in Mice. Eur. Respir. J. 2012, 40, 1475–1482. [Google Scholar] [CrossRef] [Green Version]
  59. Zhang, S.; Jia, X.; Zhang, Q.; Zhang, L.; Yang, J.; Hu, C.; Shi, J.; Jiang, X.; Lu, J.; Shen, H. Neutrophil Extracellular Traps Activate Lung Fibroblast to Induce Polymyositis-Related Interstitial Lung Diseases via TLR9-MiR-7-Smad2 Pathway. J. Cell. Mol. Med. 2020, 24, 1658–1669. [Google Scholar] [CrossRef]
  60. Suzuki, M.; Ikari, J.; Anazawa, R.; Tanaka, N.; Katsumata, Y.; Shimada, A.; Suzuki, E.; Tatsumi, K. PAD4 Deficiency Improves Bleomycin-Induced Neutrophil Extracellular Traps and Fibrosis in Mouse Lung. Am. J. Respir. Cell Mol. Biol. 2020, 63, 806–818. [Google Scholar] [CrossRef]
  61. Geetha, D.; Jefferson, J.A. ANCA-Associated Vasculitis: Core Curriculum 2020. Am. J. Kidney Dis. 2020, 75, 124–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kitching, A.R.; Anders, H.-J.; Basu, N.; Brouwer, E.; Gordon, J.; Jayne, D.R.; Kullman, J.; Lyons, P.A.; Merkel, P.A.; Savage, C.O.S.; et al. ANCA-Associated Vasculitis. Nat. Rev. Dis. Primers 2020, 6, 71. [Google Scholar] [CrossRef] [PubMed]
  63. Cornec, D.; Cornec-Le Gall, E.; Fervenza, F.C.; Specks, U. ANCA-Associated Vasculitis—Clinical Utility of Using ANCA Specificity to Classify Patients. Nat. Rev. Rheumatol. 2016, 12, 570–579. [Google Scholar] [CrossRef]
  64. Little, M.A.; Al-Ani, B.; Ren, S.; Al-Nuaimi, H.; Leite, M.; Alpers, C.E.; Savage, C.O.; Duffield, J.S. Anti-Proteinase 3 Anti-Neutrophil Cytoplasm Autoantibodies Recapitulate Systemic Vasculitis in Mice with a Humanized Immune System. PLoS ONE 2012, 7, e28626. [Google Scholar] [CrossRef] [PubMed]
  65. Lamprecht, P.; Kerstein, A.; Klapa, S.; Schinke, S.; Karsten, C.M.; Yu, X.; Ehlers, M.; Epplen, J.T.; Holl-Ulrich, K.; Wiech, T.; et al. Pathogenetic and Clinical Aspects of Anti-Neutrophil Cytoplasmic Autoantibody-Associated Vasculitides. Front. Immunol. 2018, 9, 680. [Google Scholar] [CrossRef] [Green Version]
  66. Yoshida, M.; Yamada, M.; Sudo, Y.; Kojima, T.; Tomiyasu, T.; Yoshikawa, N.; Oda, T.; Yamada, M. Myeloperoxidase Anti-Neutrophil Cytoplasmic Antibody Affinity Is Associated with the Formation of Neutrophil Extracellular Traps in the Kidney and Vasculitis Activity in Myeloperoxidase Anti-Neutrophil Cytoplasmic Antibody-Associated Microscopic Polyangiitis. Nephrology 2016, 21, 624–629. [Google Scholar] [CrossRef] [Green Version]
  67. O’Sullivan, K.M.; Holdsworth, S.R. Neutrophil Extracellular Traps: A Potential Therapeutic Target in MPO-ANCA Associated Vasculitis? Front. Immunol. 2021, 12, 637. [Google Scholar] [CrossRef]
  68. Kessenbrock, K.; Krumbholz, M.; Schönermarck, U.; Back, W.; Gross, W.L.; Werb, Z.; Gröne, H.-J.; Brinkmann, V.; Jenne, D.E. Netting Neutrophils in Autoimmune Small-Vessel Vasculitis. Nat. Med. 2009, 15, 623–625. [Google Scholar] [CrossRef]
  69. Söderberg, D.; Kurz, T.; Motamedi, A.; Hellmark, T.; Eriksson, P.; Segelmark, M. Increased Levels of Neutrophil Extracellular Trap Remnants in the Circulation of Patients with Small Vessel Vasculitis, but an Inverse Correlation to Anti-Neutrophil Cytoplasmic Antibodies during Remission. Rheumatology 2015, 54, 2085–2094. [Google Scholar] [CrossRef] [Green Version]
  70. Roitsch, S.; Gößwein, S.; Neurath, M.F.; Leppkes, M. Detection by Flow Cytometry of Anti-Neutrophil Cytoplasmic Antibodies in a Novel Approach Based on Neutrophil Extracellular Traps. Autoimmunity 2018, 51, 288–296. [Google Scholar] [CrossRef]
  71. Takeuchi, H.; Kawasaki, T.; Shigematsu, K.; Kawamura, K.; Oka, N. Neutrophil Extracellular Traps in Neuropathy with Anti-Neutrophil Cytoplasmic Autoantibody-Associated Microscopic Polyangiitis. Clin. Rheumatol. 2017, 36, 913–917. [Google Scholar] [CrossRef] [PubMed]
  72. Nakazawa, D.; Shida, H.; Tomaru, U.; Yoshida, M.; Nishio, S.; Atsumi, T.; Ishizu, A. Enhanced Formation and Disordered Regulation of NETs in Myeloperoxidase-ANCA-Associated Microscopic Polyangiitis. J. Am. Soc. Nephrol. 2014, 25, 990–997. [Google Scholar] [CrossRef] [PubMed]
  73. Hattanda, F.; Nakazawa, D.; Watanabe-Kusunoki, K.; Kusunoki, Y.; Shida, H.; Masuda, S.; Nishio, S.; Tomaru, U.; Atsumi, T.; Ishizu, A. The Presence of Anti-Neutrophil Extracellular Trap Antibody in Patients with Microscopic Polyangiitis. Rheumatology 2019, 58, 1293–1298. [Google Scholar] [CrossRef] [PubMed]
  74. Su, B.; Mao, X.; Yin, B.; Chen, C.; Zhang, M.; Cui, T.; Hao, Y. TIM-3 Regulates the NETs-Mediated Dendritic Cell Activation in Myeloperoxidase-ANCA-Associated Vasculitis. Clin. Exp. Rheumatol. 2021, 39 (Suppl. 129), 13–20. [Google Scholar]
  75. Surmiak, M.; Hubalewska-Mazgaj, M.; Wawrzycka-Adamczyk, K.; Szczeklik, W.; Musiał, J.; Brzozowski, T.; Sanak, M. Neutrophil-Related and Serum Biomarkers in Granulomatosis with Polyangiitis Support Extracellular Traps Mechanism of the Disease. Clin. Exp. Rheumatol. 2016, 34, S98–S104. [Google Scholar]
  76. Kimura, H.; Matsuyama, Y.; Araki, S.; Koizumi, A.; Kariya, Y.; Takasuga, S.; Eguchi, S.; Nakanishi, H.; Sasaki, J.; Sasaki, T. The Effect and Possible Clinical Efficacy of in Vivo Inhibition of Neutrophil Extracellular Traps by Blockade of PI3K-Gamma on the Pathogenesis of Microscopic Polyangiitis. Mod. Rheumatol. 2018, 28, 530–541. [Google Scholar] [CrossRef]
  77. Uozumi, R.; Iguchi, R.; Masuda, S.; Nishibata, Y.; Nakazawa, D.; Tomaru, U.; Ishizu, A. Pharmaceutical Immunoglobulins Reduce Neutrophil Extracellular Trap Formation and Ameliorate the Development of MPO-ANCA-Associated Vasculitis. Mod. Rheumatol. 2020, 30, 544–550. [Google Scholar] [CrossRef]
  78. Cedervall, J.; Hamidi, A.; Olsson, A.-K. Platelets, NETs and Cancer. Thromb. Res. 2018, 164 (Suppl. 1), S148–S152. [Google Scholar] [CrossRef]
  79. Matsumoto, K.; Yasuoka, H.; Yoshimoto, K.; Suzuki, K.; Takeuchi, T. Platelet CXCL4 Mediates Neutrophil Extracellular Traps Formation in ANCA-Associated Vasculitis. Sci. Rep. 2021, 11, 222. [Google Scholar] [CrossRef]
  80. Abdgawad, M.; Pettersson, Å.; Gunnarsson, L.; Bengtsson, A.A.; Geborek, P.; Nilsson, L.; Segelmark, M.; Hellmark, T. Decreased Neutrophil Apoptosis in Quiescent ANCA-Associated Systemic Vasculitis. PLoS ONE 2012, 7, e32439. [Google Scholar] [CrossRef] [Green Version]
  81. Grayson, P.C.; Carmona-Rivera, C.; Xu, L.; Lim, N.; Gao, Z.; Asare, A.L.; Specks, U.; Stone, J.H.; Seo, P.; Spiera, R.F.; et al. Neutrophil-Related Gene Expression and Low-Density Granulocytes Associated With Disease Activity and Response to Treatment in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Arthritis Rheumatol. 2015, 67, 1922–1932. [Google Scholar] [CrossRef] [PubMed]
  82. Tang, S.; Zhang, Y.; Yin, S.-W.; Gao, X.-J.; Shi, W.-W.; Wang, Y.; Huang, X.; Wang, L.; Zou, L.-Y.; Zhao, J.-H.; et al. Neutrophil Extracellular Trap Formation Is Associated with Autophagy-Related Signalling in ANCA-Associated Vasculitis. Clin. Exp. Immunol. 2015, 180, 408–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. 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]
  84. Schreiber, A.; Rousselle, A.; Becker, J.U.; von Mässenhausen, A.; Linkermann, A.; Kettritz, R. Necroptosis Controls NET Generation and Mediates Complement Activation, Endothelial Damage, and Autoimmune Vasculitis. Proc. Natl. Acad. Sci. USA 2017, 114, E9618–E9625. [Google Scholar] [CrossRef] [Green Version]
  85. Jayne, D. Complement Inhibition in ANCA Vasculitis. Nephrol. Ther. 2019, 15, 409–412. [Google Scholar] [CrossRef]
  86. Tesar, V.; Hruskova, Z. Avacopan in the Treatment of ANCA-Associated Vasculitis. Expert Opin. Investig. Drugs 2018, 27, 491–496. [Google Scholar] [CrossRef]
  87. Thickett, D.R.; Richter, A.G.; Nathani, N.; Perkins, G.D.; Harper, L. Pulmonary Manifestations of Anti-Neutrophil Cytoplasmic Antibody (ANCA)-Positive Vasculitis. Rheumatology 2006, 45, 261–268. [Google Scholar] [CrossRef] [Green Version]
  88. Frankel, S.K.; Schwarz, M.I. The Pulmonary Vasculitides. Am. J. Respir. Crit. Care Med. 2012, 186, 216–224. [Google Scholar] [CrossRef] [Green Version]
  89. Alba, M.A.; Flores-Suárez, L.F.; Henderson, A.G.; Xiao, H.; Hu, P.; Nachman, P.H.; Falk, R.J.; Charles Jennette, J. Interstital Lung Disease in ANCA Vasculitis. Autoimmun. Rev. 2017, 16, 722–729. [Google Scholar] [CrossRef]
  90. Kraaij, T.; Kamerling, S.W.A.; van Dam, L.S.; Bakker, J.A.; Bajema, I.M.; Page, T.; Brunini, F.; Pusey, C.D.; Toes, R.E.M.; Scherer, H.U.; et al. Excessive Neutrophil Extracellular Trap Formation in ANCA-Associated Vasculitis Is Independent of ANCA. Kidney Int. 2018, 94, 139–149. [Google Scholar] [CrossRef]
  91. Negreros, M.; Flores-Suárez, L.F. A Proposed Role of Neutrophil Extracellular Traps and Their Interplay with Fibroblasts in ANCA-Associated Vasculitis Lung Fibrosis. Autoimmun. Rev. 2021, 20, 102781. [Google Scholar] [CrossRef]
  92. Wells, A.U.; Denton, C.P. Interstitial Lung Disease in Connective Tissue Disease--Mechanisms and Management. Nat. Rev. Rheumatol. 2014, 10, 728–739. [Google Scholar] [CrossRef]
  93. Chirivi, R.G.S.; van Rosmalen, J.W.G.; van der Linden, M.; Euler, M.; Schmets, G.; Bogatkevich, G.; Kambas, K.; Hahn, J.; Braster, Q.; Soehnlein, O.; et al. Therapeutic ACPA Inhibits NET Formation: A Potential Therapy for Neutrophil-Mediated Inflammatory Diseases. Cell. Mol. Immunol. 2021, 18, 1528–1544. [Google Scholar] [CrossRef] [Green Version]
  94. O’Brien, E.C.; Abdulahad, W.H.; Rutgers, A.; Huitema, M.G.; O’Reilly, V.P.; Coughlan, A.M.; Harrington, M.; Heeringa, P.; Little, M.A.; Hickey, F.B. Intermediate Monocytes in ANCA Vasculitis: Increased Surface Expression of ANCA Autoantigens and IL-1β Secretion in Response to Anti-MPO Antibodies. Sci. Rep. 2015, 5, 11888. [Google Scholar] [CrossRef] [Green Version]
  95. Krajewska Wojciechowska, J.; Kościelska-Kasprzak, K.; Krajewski, W.; Morawski, K. Serum Levels of Interleukin-32 and Interleukin-6 in Granulomatosis with Polyangiitis and Microscopic Polyangiitis: Association with Clinical and Biochemical Findings. Eur. Cytokine Netw. 2019, 30, 151–159. [Google Scholar] [CrossRef]
  96. Berti, A.; Warner, R.; Johnson, K.; Cornec, D.; Schroeder, D.; Kabat, B.; Langford, C.A.; Hoffman, G.S.; Fervenza, F.C.; Kallenberg, C.G.M.; et al. Brief Report: Circulating Cytokine Profiles and Antineutrophil Cytoplasmic Antibody Specificity in Patients With Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Arthritis Rheumatol. 2018, 70, 1114–1121. [Google Scholar] [CrossRef]
  97. Berti, A.; Cavalli, G.; Campochiaro, C.; Guglielmi, B.; Baldissera, E.; Cappio, S.; Sabbadini, M.G.; Doglioni, C.; Dagna, L. Interleukin-6 in ANCA-Associated Vasculitis: Rationale for Successful Treatment with Tocilizumab. Semin. Arthritis Rheum. 2015, 45, 48–54. [Google Scholar] [CrossRef]
  98. Berti, A.; Warner, R.; Johnson, K.; Cornec, D.; Schroeder, D.R.; Kabat, B.F.; Langford, C.A.; Kallenberg, C.G.M.; Seo, P.; Spiera, R.F.; et al. The Association of Serum Interleukin-6 Levels with Clinical Outcomes in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. J. Autoimmun. 2019, 105, 102302. [Google Scholar] [CrossRef]
  99. Sakai, R.; Kondo, T.; Kurasawa, T.; Nishi, E.; Okuyama, A.; Chino, K.; Shibata, A.; Okada, Y.; Takei, H.; Nagasawa, H.; et al. Current Clinical Evidence of Tocilizumab for the Treatment of ANCA-Associated Vasculitis: A Prospective Case Series for Microscopic Polyangiitis in a Combination with Corticosteroids and Literature Review. Clin. Rheumatol. 2017, 36, 2383–2392. [Google Scholar] [CrossRef]
  100. Iijima, T.; Suwabe, T.; Sumida, K.; Hayami, N.; Hiramatsu, R.; Hasegawa, E.; Yamanouchi, M.; Hoshino, J.; Sawa, N.; Takaichi, K.; et al. Tocilizumab Improves Systemic Rheumatoid Vasculitis with Necrotizing Crescentic Glomerulonephritis. Mod. Rheumatol. 2015, 25, 138–142. [Google Scholar] [CrossRef]
  101. Jayne, D.R.W.; Bruchfeld, A.N.; Harper, L.; Schaier, M.; Venning, M.C.; Hamilton, P.; Burst, V.; Grundmann, F.; Jadoul, M.; Szombati, I.; et al. Randomized Trial of C5a Receptor Inhibitor Avacopan in ANCA-Associated Vasculitis. J. Am. Soc. Nephrol. 2017, 28, 2756–2767. [Google Scholar] [CrossRef] [Green Version]
  102. Merkel, P.A.; Jayne, D.R.; Wang, C.; Hillson, J.; Bekker, P. Evaluation of the Safety and Efficacy of Avacopan, a C5a Receptor Inhibitor, in Patients With Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Treated Concomitantly With Rituximab or Cyclophosphamide/Azathioprine: Protocol for a Randomized, Double-Blind, Active-Controlled, Phase 3 Trial. JMIR Res. Protoc. 2020, 9, e16664. [Google Scholar] [CrossRef] [Green Version]
  103. Merkel, P.A.; Niles, J.; Jimenez, R.; Spiera, R.F.; Rovin, B.H.; Bomback, A.; Pagnoux, C.; Potarca, A.; Schall, T.J.; Bekker, P.; et al. Adjunctive Treatment With Avacopan, an Oral C5a Receptor Inhibitor, in Patients With Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. ACR Open Rheumatol. 2020, 2, 662–671. [Google Scholar] [CrossRef]
  104. Chalkia, A.; Thomas, K.; Giannou, P.; Panagiotopoulos, A.; Hadziyannis, E.; Kapota, A.; Gakiopoulou, H.; Vassilopoulos, D.; Petras, D. Hypocomplementemia Is Associated with More Severe Renal Disease and Worse Renal Outcomes in Patients with ANCA-Associated Vasculitis: A Retrospective Cohort Study. Ren. Fail. 2020, 42, 845–852. [Google Scholar] [CrossRef]
  105. Johansson, L.; Berglin, E.; Eriksson, O.; Mohammad, A.J.; Dahlqvist, J.; Rantapää-Dahlqvist, S. Complement Activation Prior to Symptom Onset in Myeloperoxidase ANCA-Associated Vasculitis but Not Proteinase 3 ANCA Associated Vasculitis—A Swedish Biobank Study. Scand. J. Rheumatol. 2022, 1–6. [Google Scholar] [CrossRef]
  106. Hakroush, S.; Tampe, D.; Korsten, P.; Ströbel, P.; Tampe, B. Complement Components C3 and C4 Indicate Vasculitis Manifestations to Distinct Renal Compartments in ANCA-Associated Glomerulonephritis. Int. J. Mol. Sci. 2021, 22, 6588. [Google Scholar] [CrossRef]
Figure 1. Flowchart of the selected articles used for the review.
Figure 1. Flowchart of the selected articles used for the review.
Life 12 00317 g001
Table 1. SANRA score for quality assessment.
Table 1. SANRA score for quality assessment.
N°.Title and AuthorsJustification of the Article’s Importance for the ReadershipStatement of Concrete Aims or Formulation of QuestionsDescription of the Literature SearchReferencingScientific ReasoningAppropriate Presentation of DataTotal Score
1Neutrophil Extracellular Traps Induce the Epithelial—n Mesenchymal Transition: Implications in Post-COVID-19 Fibrosis. Pandolfi L., Bozzini S., Frangipane V. et al., 20212201229
2Therapeutic ACPA inhibits NET formation: a potential therapy for neutrophil-mediated inflammatory diseases. Chirivi R.G.S., van Rosmalen J.W.G., van der Linden M. et al., 202122022210
3PAD4 Deficiency Improves Bleomycin-induced Neutrophil Extracellular Traps and Fibrosis in Mouse Lung. Suzuki M., Ikari J., Anazawa R. et al., 2020 2101127
4Neutrophil extracellular traps infiltrate the lung airway, interstitial, and vascular compartments in severe COVID-19. Radermecker C., Detrembleur N., Guiot J. et al., 20201202218
5Neutrophil extracellular traps activate lung fibroblast to induce polymyositis-related interstitial lung diseases via TLR9-miR-7-Smad2 pathway. Zhang S., Jia X., Zhang Q. et al., 20192102218
6Regulation of Pseudomonas aeruginosa-Mediated Neutrophil Extracellular Traps. Skopelja-Gardner S., Theprungsirikul J., Lewis K.A. et al., 20191101227
7Identification of a Novel HIF-1α-αMβ2 Integrin-NET Axis in Fibrotic Interstitial Lung Disease. Khawaja A.A., Chong D.L.W., Sahota J. et al., 2020 2201218
8Neutrophil extracellular traps promote differentiation and function of fibroblasts. Chrysanthopoulou A., Mitroulis I., Apostolidou E. et al., 20142102218
9Excessive neutrophil extracellular trap formation in ANCA-associated vasculitis is independent of ANCA. Kraaij T., Kamerling S.W.A., van Dam L.S. et al., 20182102128
10Neutrophil extracellular trap formation is associated with autophagy-related signalling in ANCA-associated vasculitis. Tang S., Zhang Y., Yin S.W. et al., 20152101217
11Myeloperoxidase anti-neutrophil cytoplasmic antibody affinity is associated with the formation of neutrophil extracellular traps in the kidney and vasculitis activity in myeloperoxidase anti-neutrophil cytoplasmic antibody-associated microscopic polyangiitis. Yoshida M., Yamada M., Sudo Y. et al., 20161201217
12Enhanced formation and disordered regulation of NETs in myeloperoxidase-ANCA-associated microscopic polyangiitis. Nakazawa D., Shida H., Tomaru U. et al., 20142102229
13The presence of anti-neutrophil extracellular trap antibody in patients with microscopic polyangiitis. Hattanda F., Nakazawa D., Watanabe-Kusunoki K. et al., 20192201218
14Necroptosis controls NET generation and mediates complement activation, endothelial damage, and autoimmune vasculitis. Schreiber A., Rousselle A., Becker J.U. et al., 20172201128
15Intrinsically Distinct Role of Neutrophil Extracellular Trap Formation in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Compared to Systemic Lupus Erythematosus. Van Dam L.S., Kraaij T., Kamerling S.W.A. et al., 20192201117
16Detection by flow cytometry of anti-neutrophil cytoplasmic antibodies in a novel approach based on neutrophil extracellular traps. Roitsch S., Gößwein S., Neurath M.F. et al., 20181201228
17Neutrophil extracellular traps in neuropathy with anti-neutrophil cytoplasmic autoantibody-associated microscopic polyangiitis. Takeuchi H., Kawasaki T., Shigematsu K. et al., 201722022210
18TIM-3 regulates the NETs-mediated dendritic cell activation in myeloperoxidase-ANCA-associated vasculitis. Su B., Mao X., Yin B. et al., 20212101228
19Increased levels of neutrophil extracellular trap remnants in the circulation of patients with small vessel vasculitis, but an inverse correlation to anti-neutrophil cytoplasmic antibodies during remission. Söderberg D., Kurz T., Motamedi A. et al., 20152101217
20Neutrophil-related and serum biomarkers in granulomatosis with polyangiitis support extracellular traps mechanism of the disease. Surmiak M., Hubalewska-Mazgaj M., Wawrzycka-Adamczyk K. et al., 20161201228
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

d’Alessandro, M.; Conticini, E.; Bergantini, L.; Cameli, P.; Cantarini, L.; Frediani, B.; Bargagli, E. Neutrophil Extracellular Traps in ANCA-Associated Vasculitis and Interstitial Lung Disease: A Scoping Review. Life 2022, 12, 317. https://doi.org/10.3390/life12020317

AMA Style

d’Alessandro M, Conticini E, Bergantini L, Cameli P, Cantarini L, Frediani B, Bargagli E. Neutrophil Extracellular Traps in ANCA-Associated Vasculitis and Interstitial Lung Disease: A Scoping Review. Life. 2022; 12(2):317. https://doi.org/10.3390/life12020317

Chicago/Turabian Style

d’Alessandro, Miriana, Edoardo Conticini, Laura Bergantini, Paolo Cameli, Luca Cantarini, Bruno Frediani, and Elena Bargagli. 2022. "Neutrophil Extracellular Traps in ANCA-Associated Vasculitis and Interstitial Lung Disease: A Scoping Review" Life 12, no. 2: 317. https://doi.org/10.3390/life12020317

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