Next Article in Journal
Modelling the In Vivo and Ex Vivo DNA Damage Response after Internal Irradiation of Blood from Patients with Thyroid Cancer
Previous Article in Journal
Kinase Inhibitors and Kinase-Targeted Cancer Therapies: Recent Advances and Future Perspectives
Previous Article in Special Issue
Neutrophil Elastase Degrades Histone Deacetylases and Sirtuin 1 in Primary Human Monocyte Derived Macrophages
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects

by
Coby J. Cheetham
1,
Michael C. McKelvey
1,
Daniel F. McAuley
2 and
Clifford C. Taggart
1,*
1
Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK
2
Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(10), 5492; https://doi.org/10.3390/ijms25105492
Submission received: 23 April 2024 / Revised: 10 May 2024 / Accepted: 15 May 2024 / Published: 17 May 2024
(This article belongs to the Special Issue New Pathogenic Mechanism of Proteases in Inflammatory Lung Diseases)

Abstract

:
Neutrophil-derived proteases are critical to the pathology of many inflammatory lung diseases, both chronic and acute. These abundant enzymes play roles in key neutrophil functions, such as neutrophil extracellular trap formation and reactive oxygen species release. They may also be released, inducing tissue damage and loss of tissue function. Historically, the neutrophil serine proteases (NSPs) have been the main subject of neutrophil protease research. Despite highly promising cell-based and animal model work, clinical trials involving the inhibition of NSPs have shown mixed results in lung disease patients. As such, the cutting edge of neutrophil-derived protease research has shifted to proteases that have had little-to-no research in neutrophils to date. These include the cysteine and serine cathepsins, the metzincins and the calpains, among others. This review aims to outline the previous work carried out on NSPs, including the shortcomings of some of the inhibitor-orientated clinical trials. Our growing understanding of other proteases involved in neutrophil function and neutrophilic lung inflammation will then be discussed. Additionally, the potential of targeting these more obscure neutrophil proteases will be highlighted, as they may represent new targets for inhibitor-based treatments of neutrophil-mediated lung inflammation.

1. Introduction

Inflammatory lung diseases, both chronic and acute, are major causes of morbidity and mortality worldwide, and consequently put huge pressure on healthcare systems and hospitals [1,2,3,4,5,6]. As the most abundant circulating leukocyte [7,8], neutrophils have a critical role in inflammatory responses, and neutrophilia is a key component of lung diseases such as bronchiectasis, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF) and acute respiratory distress syndrome (ARDS) [9,10,11,12,13,14,15,16]. Neutrophils arise from progenitor cells in the bone marrow, progressing from pluripotent stem cells to promyelocytes, band cells and finally mature neutrophils [17,18]. Neutrophil lineage cells can be identified by several surface markers [18], but the expression of surface markers, such as Ly6G an CD11b, changes drastically over the course of maturation [19]. Neutrophils have also been shown to exhibit different phenotypes in the context of various lung diseases. For example, in bronchiectasis, neutrophils display increased survival, decreased surface expression of CD11b, and impaired phagocytosis [9]. Additionally, neutrophils from patients with COPD seem to have altered chemotactic migration with decreased migratory accuracy [10]. As such, understanding how neutrophils function in the context of lung disease, and the mechanisms behind alterations in neutrophil phenotypes, is critical to developing better treatments.
Neutrophils have multiple unique mechanisms by which they influence infection and inflammation. For example, neutrophils can undergo a unique form of cell death known as NETosis where they release a ‘web’ of DNA called a neutrophil extracellular trap (NET) [20,21]. This process requires the decondensation of chromatin in the nucleus, as well as oxidative burst within the cell [22]. The aggregation of these structures can lead to the entrapment of pathogenic microorganisms, as well as degradation of some inflammatory mediators, facilitating the resolution of inflammation [23]. Neutrophils are also among the first cells to respond to inflammatory signals [8], particularly in the lung. Chemo-attractants such as interleukin 8 (IL-8), leukotriene B4 (LTB4), and more recently IL-17, are known to contribute to the chemotaxis and transmigration of neutrophils into the airways [24,25,26,27,28]. The levels of neutrophil-attracting chemokines have been shown to increase in acute lung inflammation [13,14], as well as in chronic lung diseases, such as CF [29,30]. Interestingly, some of these migrating neutrophils are known to remain in the lung microvasculature, forming a marginated neutrophil pool, which exists in dynamic equilibrium with the circulating neutrophil pool [31,32]. Additionally, when transmigrating into the airways of patients with CF, neutrophils have been shown to gain a unique inflammatory phenotype known as GRIM (granule releasing, immunomodulatory and metabolically active) [33,34].
Within the armament of neutrophils is an array of proteases, which act as both regulators and effectors of neutrophil function [35,36,37]. Proteases are known to contribute to the pathology of many muco-obstructive [38,39,40], and acute inflammatory lung diseases [41,42], so targeting neutrophil-derived proteases has garnered much attention as a means by which to alleviate tissue damage and loss of lung function. This review will outline the roles of neutrophil proteases in lung inflammation, both those commonly associated with neutrophils, and proteases that have less well understood roles in neutrophils. The current therapeutic options targeting neutrophilic proteases will also be highlighted, as well as their shortcomings and potential alternatives.

2. Old Players: Neutrophil Serine Proteases

The neutrophil serine proteases (NSPs) are the proteases most often associated with neutrophils and neutrophilic inflammatory diseases [8]. They include neutrophil elastase (NE), cathepsin G (CTSG) and proteinase 3 (PR3), as well as the more recently discovered NSP4 [43]. They are produced during neutrophil development and are stored in the azurophilic (primary) granules (Figure 1) [44]. Interestingly, their expression levels vary throughout the maturation and life of a neutrophil [45,46], with expression first being detectable in blast cells, peaking in promyelocytic cells and waning once cells fully differentiate. As such, mature neutrophils do not synthesise NSPs, but simply store them in large amounts in the primary granules [47]. NSP proteins are initially synthesised as zymogens that are activated by cathepsin C (CTSC) via the modification of the N-terminal region of the proteins [48]. NSPs can act both intracellularly, where they degrade phagocytosed material and microorganisms, and extracellularly, where they are secreted during degranulation, and can contribute to extracellular matrix (ECM) remodelling by cleaving components, such as elastin, collagen, fibronectin and laminin [49,50]. Within the neutrophil, NSPs potentially contribute to NETosis [35,36], though their importance in this process is not fully clear due to conflicting data [21]. Dysregulation of overall NSP levels or activity contributes to the pathology of lung diseases. However, each NSP acts in a unique manner, and as such it is important to look at the function and dysregulation of each individually.

2.1. Neutrophil Elastase

NE was first described in 1965 by Janoff and Scherer, who found that an extract of frozen granules contained proteases, which increased vascular permeability in the skin and muscle [51]. Since then, there has been a vast amount of research into NE due to it being the most abundant NSP in the azurophilic granules [52]. NE is encoded by the gene ELANE, and mutations in this gene can lead to neutropaenia, likely due to a maturation arrest in the promyelocytes [53]. NE acts alongside reactive oxygen species (ROS) to mediate bacterial killing and sepsis defence, where it can cleave outer membrane proteins of bacteria, such as Escherichia coli [54,55,56]. As such, mutations in ELANE may also result in increased incidence and severity of infection.
Although initially observed in granules [51], further research has shown that NE can bind to proteoglycans on the cell surface, in a reversible and dynamic manner [57]. This mode of cell surface binding leaves the NE active site available, meaning that surface-bound NE is catalytically active. Furthermore, neutrophil exosomes are able to uptake this NE from the cell surface [58]. Critically, this surface-bound NE seems to be resistant to inhibition, and instilled NE-containing exosomes can confer destructive lung disease to mice. This highlights that NE localisation is not only more complex than originally believed, but also potentially important in the pathology and propagation of lung inflammation. The overall activity of NE in the lung not only depends on the level of NE in the neutrophils and the levels of secreted NE, but also on the levels of endogenous inhibitors in the tissues [52]. These naturally-occurring inhibitors of NSPs include the serpins [59,60,61], α1-antitrypsin (A1AT) [62], and α1-antichymotrypsin [63], as well as secretory leukocyte protease inhibitor (SLPI) and elafin [64,65,66]. Interestingly, both SLPI and elafin appear to be cleaved and inactivated by high levels of NE activity [66,67]. Therefore, in lung disease with high levels of NE, there is not only an increased level of NE but a decreased level of these endogenous inhibitors [68].
NE appears to directly contribute to mucus obstruction, which is a key component of many respiratory diseases. For example, NE upregulates the expression of secretory mucins, such as MUC5AC in the airways [69,70], altering the levels of MUC5AC mRNA and modulating the levels and activity of micro RNAs (miRNAs). NE is also suggested to induce MUC5AC secretion from the airway epithelium, via the cleavage of PAR2 and the metaplasia of goblet cells [71,72,73]. This leads to thickening of the mucus and impaired mucociliary clearance. This thickening is exacerbated by NE-mediated cleavage of cystic fibrosis transmembrane conductance regulator (CFTR) and epithelial sodium (ENaC) channels in the airways [39,74]. NE is also closely associated with pro-inflammatory signalling within the context of lung disease, where it can activate cytokines, such as IL-1α, IL-8, and IL-36δ [75,76], and produce fibrin cleavage products that promote the migration of neutrophils [52]. Despite enhancing many aspects of the immune response, NE also interferes with other aspects of immunity in the lung. Interestingly, NE can cleave antimicrobial proteins, like lactoferrin [77], and may also contribute to the defective phagocytosis seen in diseases such as CF [78]. Toll-like receptor (TLR) signalling also seems to be affected by NE, which can cleave TLRs in some pneumonias, leading to altered cytokine transcription [79]. Environmental stimuli, such as cigarette smoke and E-cigarette vapour, also induce a neutrophil-mediated inflammation, which involves NE [80,81]. Together, the years of mechanistic work regarding NE in the lung have shown a complex relationship between this protease and the immune function of the lung, which explains its involvement in a number of lung diseases [42,82,83,84,85].

2.2. Cathepsin G

CTSG is a serine cathepsin produced by immune cells, such as monocytes and neutrophils [86,87]. CTSG can not only be secreted from neutrophils, but can also be expressed on the cell surface, which preserves its activity in the presence of inhibitors [88]. Like NE, CTSG has several important roles in cellular and immune functions related to lung inflammation, such as cytokine processing [89,90], pathogen clearance [91,92,93], and cell-surface receptor modification [94]. However, dysregulation of CTSG activity is known to be detrimental, especially in the lung. For example, CTSG cleaves ECM proteins, such as elastin, in COPD, contributing to the loss of lung function [95,96]. CTSG is also thought to affect lung microbiome composition in patients with CF by affecting the amino acid levels in the airway environment and by cleaving important antimicrobial proteins [97,98]. There also appears to be a role for CTSG in the formation of NETs via a PAD4-mediated mechanism involving neutrophil-platelet interactions [22,99]. The association of CTSG with lung inflammation has even implicated CTSG as a potential biomarker for airway tissue damage [100]. Therefore, despite NE being the major interest regarding NSP research, CTSG also has crucial and independent roles that are worth considering in lung inflammation.

2.3. Proteinase 3 and NSP4

PR3 is the third classical NSP, which is known to be stored both in neutrophil azurophilic granules and in secretory vesicles [101]. The roles of PR3 within cells are varied, ranging from cytokine and antimicrobial peptide activation [102,103] to the activation of pro-apoptotic signalling pathways [104]. PR3 dysregulation is also associated with lung disease. PR3 is thought to contribute to the ECM degradation seen in emphysema [95,105] and is also detectable at high concentrations in CF sputum, where it contributes to the secretion of characteristically thick mucus [106].
The fourth NSP is the more recently discovered NSP4, which appears to be found in neutrophil granules, like the classical NSPs [43,107]. The abundance of this NSP in the granules is low, though the processing and secretion seems to be comparable to the other NSPs [107]. Apart from this, very little is known about NSP4, and its role in lung disease has been unexplored to date.

2.4. NSP Inhibition

Due to the involvement of the NSPs, particularly NE, in lung disease pathology, synthetic inhibitors that regulate their activity have previously been investigated in both animal models and human clinical trials. In the context of COPD, the NE inhibitor AZD9668 showed promising results in animal models utilising smoke-inhalation [108]; however, two clinical trials investigating the compound found no significant therapeutic effect for patients [109,110]. This inhibitor was also trialled in patients with bronchiectasis, and similarly variable results were observed [111], though there were slight improvements in forced expiratory volume. Similarly mixed efficacy of other NE inhibitors has also been reported in CF and non-CF bronchiectasis clinical trials [112,113]. The only NE inhibitor licenced for use is Sivelestat, which is licenced for clinical use in Japan [114]. This inhibitor has been widely investigated in the context of acute lung inflammation [115,116], and a meta-analysis of these trials showed no overall effect on mortality or length of ICU stay [117]. There are some other NE inhibitors that are still in the early trial stages, such as POL6014, which has shown effective pharmacokinetics in patients with CF [118], but has yet to be investigated regarding efficacy. A summary of these clinical studies can be found in Table 1.
Overall, patients treated with NE inhibitors seem to have a significant variability in their response to the drugs, resulting in the poor results seen in these studies. The mode of inhibition may be a key reason behind these failures, and more novel inhibition methods, such as antibody-based and miRNA-based therapeutics, are therefore being researched [122,123]. These failures of NE inhibition may also be in part due to the high day-to-day variability seen in NSP and NE activity in lung disease patients [124]. This may suggest that a more personalised approach to NSP inhibition, considering the individual NSP activity of each patient, would be much more effective. The limited success of NSP inhibition has prompted the consideration of alternative or complementary proteases that could be targeted therapeutically. Indeed, the cutting edge of protease research in lung inflammation is now moving towards proteases of which we have limited understanding in the context of neutrophil function.

3. New Prospects: Other Neutrophil Proteases

3.1. Cysteine Cathepsins

The cathepsins are a group of related proteases that were first discovered in 1968 when Drenth and colleagues described the protease papain from papayas [125]. They found that this protease had an active site consisting of an α-helical heavy chain and a short β-chain, and this form of active ‘papain-like’ domain is the link between the various cathepsin proteases [126]. The cathepsins can be organised into several families, such as the serine cathepsins, the aspartyl cathepsins, and the cysteine cathepsins [127]. Cathepsins are endopeptidases for the most part, though there are some exceptions, such as cathepsin C [48]. Cathepsins may be expressed in a ubiquitous or tissue-specific manner [127], for example, cathepsin K is expressed mainly in osteoclasts due to its involvement in bone resorption [128]. The regulation of cathepsin activity is controlled by both endogenous antiproteases as well as other factors. Some endogenous inhibitors of cathepsin activity include the cystatins, thyropins and serpins [129], and these may act either in a regulatory or emergency manner. Due to the need for reduced residues in the cathepsin active site for activity, modification of the redox environment can also lead to changes in cathepsin activity [130]. This can be done by both reactive nitrogen and oxygen species [131,132]. Additionally, some data suggest that the cleaved pro-peptides from cathepsin zymogens can act as selective inhibitors for their cognate proteases [133]. The removal of the pro-peptide seems to be promoted by some glycosaminoglycans [134], implicating molecules such as chondroitin-4-sulfate in the regulation of cathepsin activity [135,136]. Zymogens are normally processed to their mature forms in the lysosomes and are localised to these compartments via the pro-peptide [137]. The regulation of cathepsin activity is therefore complex, and highly important due to the involvement of cathepsins in many cellular functions.
Cathepsins are able to degrade and cleave ECM components, such as collagen, laminin, and fibronectin [138], and are also involved in the remodelling of tissues in the blood vessels, such as arteries [139]. Cathepsins are also important in apoptosis, where they cleave Bid and anti-apoptotic proteins [140,141], facilitating the release of cytochrome C and subsequent cell death. Cathepsins also play key roles in immunological signalling pathways, including toll-like receptor (TLR) signalling and antigen presentation [142,143], and other key cellular functions, such as autophagy, cell cycle progression and cell adhesion [144,145,146]. Their close involvement with a variety of critical cellular functions therefore explains why their dysregulation is associated with pathology; most dysregulation relating to lung diseases is aberrantly high cathepsin activity. In CF for example, the epithelial lining of the airways is acidified [147], resulting in increased cathepsin activity [148], and subsequent degradation of anti-microbial peptides [149]. This therefore implicates cathepsin activity in the tissue damage and increased incidence of infection seen in CF [150]. The upregulated cathepsin activity in COPD can also result in increased cleavage of antiproteases such as SLPI [151]. Additionally, murine models of asthma have shown a pro-inflammatory role for cathepsins in allergic airway inflammation [152,153]. These are conditions that also have a significant neutrophil component to their pathology [154,155] and understanding the roles of specific cathepsins expressed in neutrophils is therefore key to understanding neutrophil contribution to lung disease.

3.2. Cathepsin C

Cathepsin C (CTSC, also known as Dipeptidyl peptidase I) is a cysteine cathepsin that is expressed primarily in neutrophils and mast cells [48]. It was first described in 1948 by Gutman and Fruton [156] and was later linked to the papain-like family of cathepsin proteases after their discovery [157]. The addition of CTSC to the cathepsin family of proteases came despite its highly unique, tetrameric structure, which has not been observed for other cathepsins [158]. CTSC monomers are synthesised as zymogens in haemopoietic precursor cells [159]. These monomers contain a papain-like domain, which confers activity, a pro-peptide and an exclusion domain. The papain-like domain further consists of a heavy and a light chain. Two of these 60 kDa monomers associate to form a homodimer [160], which is then subsequently cleaved into several mature chains of CTSC [48]. This processing step results in the excision of the pro-peptide, and the remaining domains are held together by tight non-covalent interactions. Two of these processed dimers can then associate to form the active 200 kDa homo-tetramer, a process that is mediated by the exclusion domains [161]. This processing pathway is suggested to be facilitated by other cathepsins, such as cathepsins L, S, K, V and F [160], though there is little research in cell lines or animal models to confirm this. Mature CTSC plays a role in the catalytic processing of other proteases, such as the NSPs [162,163]. This happens early in the maturation process of the neutrophil [164]; as these cells mature, more CTSC is secreted into the extracellular space, as there are no new NSP mRNA transcripts being produced [45,46,159].
CTSC therefore plays a critical role in the protective inflammatory responses during infection, and some pathogens, such as Helicobacter pylori, are known to impair CTSC expression to promote infection [165]. Loss-of-function genetic mutations in the CTSC gene are also detrimental to normal immune function and cause a condition known as Papillon–Lefèvre syndrome (PLS) [166]. Many PLS-associated mutations result in impaired CTSC processing, which is detectable by the lack of heavy or light chain CTSC in urine samples [167]. The associated phenotype for this condition primarily presents as severe periodontitis and palmoplantar keratosis [166], but the levels of NSPs are also significantly reduced in PLS patients [168]. This results in reduced downstream proteolysis, such as reduced processing of LL-37 [169], which can have significant effects on inflammatory and anti-microbial responses. Neutrophil functions that rely on NSP activity are also impaired in PLS [169]. NET release and chemotaxis appear impaired in PLS neutrophils, whereas the levels of pro-inflammatory cytokines released are higher [170]. As such, PLS results in an increased susceptibility to infection in around 25% of cases [171]. Other CTSC mutations result in a more severe condition known as Haim–Munk syndrome (HMS) [172], however some evidence suggests that they are the same condition, presenting with different phenotypes [173]. Together, PLS and HMS demonstrate that CTSC is a critical component not only of neutrophil function, but cellular and immune function as a whole.
Like most proteases, CTSC activity within neutrophils must be balanced, and aberrantly high CTSC activity may also be detrimental in inflammatory diseases of the lungs. CTSC is mainly linked to NSP-mediated lung diseases, which include ARDS, CF, and COPD [80,82,85,95,106,174,175], due to its role in NSP activation. However, CTSC may also contribute to lung inflammation through alternative means. Lysosomal destabilisation is one of the key processes by which cathepsins are released into the cytosol, and lead to various forms of cell death [176]. Some interesting data suggest that CTSC is important in leucyl-L-leucine methyl ester (LLOME) mediated lysosomal destabilisation [177], leading to a rapid release of lysosomal contents. This may implicate CTSC in the action and secretion of other cathepsins, which are known to cause lung tissue damage [149,151,152]. CTSC also appears to have roles in IL-1β secretion, through a caspase-1-independent mechanism [178]. CTSC may consequently play a role in important inflammatory signalling pathways, such as the inflammasome pathway [179]. Whether this effect is directly mediated by CTSC, or if it is mediated by a downstream protease, is unknown in neutrophils, though it has been shown that mast cell chymase, another protease activated by CTSC, can cleave IL-1β in an atypical way [180]. It may therefore be the case that NSPs are involved in this alternative pathway of IL-1β activation in neutrophils.
Because of these pro-inflammatory links, targeting CTSC activity using inhibitors has been investigated in both animal models and clinical trials regarding lung disease. Most of the currently developed CTSC inhibitors are nitrile inhibitors [181,182,183,184], for example Brensocatib [185]. In rodent models, this inhibitor achieved a dose-dependent plasma exposure and a dose-dependent reduction in NSP activity in the bone marrow [186]. The success of Brensocatib in animal models led to several clinical trials in lung diseases, as well as animal models of inflammatory diseases of other organ systems [187]. While work in patients with bronchiectasis showed promising results, with a reduction in the frequency of exacerbations [188,189], work in patients with COVID-19 showed negative effects and increased mortality [190] (Table 1). Despite these issues in COVID-19, Brensocatib has been considered a success and, following promising preclinical results [191], Chalmers and colleagues have started investigating a new CTSC inhibitor BI1291583 in patients with bronchiectasis [192]. Other CTSC inhibitors have not shown strong clinical translation, for example GSK-2793660 [193], which failed due to minimal effects on NSP activity. More recent work has shown that non-peptidyl, non-covalent inhibitors of CTSC can also effectively reduce NSP activity and may be more metabolically stable than their covalent counterparts [194,195]. Overall, CTSC inhibition represents a promising option for the treatment of inflammatory lung diseases, potentially having more consistent and far-reaching effects than NSP-specific inhibitors [110,111,112,116].

3.3. Cathepsin S

Cathepsin S (CTSS) is a cysteine cathepsin that has somewhat restricted expression, being produced mainly in immune cells such as macrophages and neutrophils [146,196]; however, it can also be expressed in non-immune cells such as airway epithelial cells [197]. In the context of the neutrophil, though not thought to be present in primary, secondary or tertiary granules, Rørvig and colleagues have previously noted that CTSS is located in a subset of Ficolin-1-rich granules, which are likely to be among the first granules exocytosed upon stimulation [198]. Like most cathepsins, CTSS is synthesised as a zymogen, which is subsequently cleaved to its active and mature form [199]. The processing of CTSS zymogens can be facilitated by other proteases, as well as CTSS itself, in an autocatalytic process [200]. Regarding function, CTSS has the unique ability to retain its activity at neutral and even slightly alkaline pH [201], allowing CTSS to exhibit activity in the extracellular space [202]. The secretion of CTSS is usually as a result of overexpression [203] in pro-inflammatory contexts [204]. The regulation of CTSS activity both in cells and in the extracellular space is highly complex. The regulation of CTSS mRNA at a transcriptional and post-transcriptional level is reported to be mediated by molecules such as gamma interferon-inducible lysosomal thiol reductase (GLiT), human antigen R (HuR) and tristetrapolin (TTP) [205,206,207]. The activity of the CTSS protease itself is also subject to strict regulation, both through the action of endogenous inhibitors [208,209], and other mechanisms such as oxidation [210,211]. The regulation of CTSS can even be altered by certain microorganisms to facilitate their survival and spread. For example, several Mycobacteria species regulate CTSS through upregulating certain micro RNAs (miRNAs) and inhibiting CTSS processing through IL-10 signalling [212,213].
CTSS plays a critical role in multiple cellular functions, including the fusion of autophagosomes and lysosomes [214], and the processing of major histocompatibility complex II [143]. Interestingly, CTSS also appears to play a role in the pain signalling associated with inflammation [215]. Like most cathepsins, however, CTSS dysregulation plays a role in the pathogenesis of many diseases, including several neutrophilic lung diseases [41,154,216,217]. CTSS is also upregulated in lung inflammation induced by environmental stimuli [218,219,220]. Although the role of CTSS in tissue damage and increased airway permeability in these diseases is well-documented [119,219], the role of CTSS in neutrophils themselves is relatively unexplored to date. Some previous work looking at murine models of acute lung inflammation demonstrated that CTSS inhibition reduced neutrophil influx into the airways, but the mechanisms behind this are not understood [41]. Adding to this are recent data regarding stress responses in mice, which also found a role for CTSS in the recruitment of immune cells, such as neutrophils [120]. CTSS has previously been shown to affect migration of other cells [121], and it can activate several chemokines, such as CXCL2, via cleavage [221]. CTSS may also influence neutrophil migration and function by affecting the processing and activity of NSPs, though there is limited, conflicting evidence of its involvement [159,160,222,223]. CTSS has also recently been linked to NETosis in the context of cancer immunotherapy [224], where CTSS was found within NETs cleaving arginase to facilitate anti-tumour immunity [225,226]. Together, these data show that the role of CTSS in the inflammatory response is relatively unknown at this stage, but its critical role in many neutrophilic lung diseases highlights the need for further research in this area.

3.4. Other Cysteine Cathepsins

Neutrophils express other cathepsins, some at high levels and some at very low levels [227]. The involvement of many of these in neutrophil function has not been investigated to date. Cathepsin B (CTSB) has been identified as playing a role in neutrophil migration [228], by cleaving Mac-1 integrins on the endothelial surfaces of blood vessels. This gives CTSB an anti-adhesive and pro-migratory role, which has not been explored outside of this work. CTSB is known to be upregulated in various pro-inflammatory lung diseases, such as CF, COPD, and pneumonia [229,230,231], though further research is needed to investigate its roles in neutrophil function and inflammatory processes.

3.5. Calpains

The calpains are a family of non-lysosomal cysteine proteases, with a very broad expression profile. They exist as cytosolic inactive precursors until intracellular Ca2+ reaches a threshold level, triggering their activation. They are mostly associated with promoting cell death in response to Ca2+ through their cleavage of a wide range of proteins including cytoskeletal proteins, Bid, caspase-3, and proteins of the electron transport chain [232]. Overexuberant calpain activity has been associated with chronic pulmonary disease [233,234], and calpains of neutrophilic provenance may contribute specially to pathology. For example, calpains have previously been linked to cigarette smoke-mediated airway epithelium disruption, potentially increasing susceptibility to infection [235]. Within the neutrophil, along with their role in mediating apoptosis and necrosis [236], calpains appear to be important regulators of neutrophil movement. Calpain I (μ-calpain) inhibition promotes Rho GTPase activity, rapid random chemo-kinesis and pseudopod formation; interestingly, this inhibition reduces the directional fidelity of the neutrophil in response to IL-8 [237]. A similar phenomenon is observed when calpain I is inactivated in neutrophils treated with A1AT [238]. Thus, calpains facilitate the direct and accurate movement of neutrophils to the site of inflammation.

3.6. Metzincin Proteases

Proteases from the metzincin superfamily are characterised by the presence of a Zn2+ ion at the catalytic centre, bound by three histidine residues, which facilitates the hydrolysation of proteins. This superfamily comprises the matrix metalloprotease (MMP), a distintegrin and metalloprotease (ADAM), and ADAM proteases with a thrombospondin motif (ADAMTS) families, which have well-documented roles in lung homeostasis and pathogenesis, especially in the remodelling of the pulmonary ECM [239,240,241]. Despite the traditional association of metzincins with parenchymal and mesenchymal lung cells, expression of all three families has been reported in immune cells [242]. However, only a handful of specific metzincins are known to be expressed by neutrophils.
During homeostasis, metzincin expression tends to be low, and is upregulated during pathologies, such as COPD, ARDS, and CF [243,244,245,246]. MMP-8 (neutrophil collagenase) is perhaps the best-known neutrophil-derived metzincin, though neutrophils are also a major source of MMP-9 (gelatinase B) [247]. Stored in secondary and tertiary neutrophil granules [198], these MMPs are readily exocytosed during stimulation, and their potent proteolytic abilities facilitate the migration of neutrophils through tissues by the degradation of ECM and basement membranes. Destruction and remodelling of the ECM by proteases, derived not exclusively but significantly from neutrophils, is a key aspect in the development of many major lung diseases [240,248]. Some of the released MMPs are soluble and diffuse, while others remain membrane-bound [249], meaning that high MMP activity is retained on migrating neutrophils. Moreover, neutrophils do not appear to release the tissue inhibitor of metalloproteinase (TIMP)-1 [250,251], which may also contribute to high MMP activity in the environs of the neutrophil during stimulation and migration. MMP-8 is involved in ECM remodelling during migration, where it degrades collagen [252]. MMP-8 also cleaves non-structural substrates, such as A1AT [253], which compromises the antiprotease defence against serine proteases. Furthermore, the inflammatory ability of an array of cytokines and chemokines can be proteolytically enhanced by MMP cleavage. For instance, CXCL8 can be processed by MMP-8 and -9 [254,255], while N-acetyl proline-glycine-proline (Ac-PGP) fragments are produced by MMP-9-mediated cleavage of collagen [256]. Both cleavage processes produce highly chemotactic molecules that enhance neutrophil recruitment during lung disease. Interestingly, both CXCL8 and Ac-PGP also upregulate MMP-9 release from neutrophils, stimulating a feed-forward mechanism that propagates inflammation. Beyond MMP-8 and -9, the roles of other neutrophil-derived MMPs have not been investigated. Gene expression analysis indicates that at least two membrane-type (MT-)MMPs are expressed by primary human neutrophils (MMP-24 and -25, also known as MT5-MMP and MT6-MMP, respectively) [227]. Few clear functions have been attributed to these proteases, though MT6-MMP can cleave numerous CXC and CC chemokines, possibly indicating a role in the inflammatory process [257].
ADAM proteases are best-known for their functions in cell adhesion and the processing of ectodomains of cell surface proteins (so-called ‘sheddase’ activity), though several members of this family lack catalytic activity. Like the MT-MMPs, ADAMs contain transmembrane and cytoplasmic domains, as well as the extracellular MMP domain, though they are distinguished by the presence of extracellular EGF-like, cysteine-rich and disintegrin-like motifs [258]. Because of these various domains, ADAMs can exert complex pleiotropic effects, such as proteolysis, cytoskeletal manipulation, and cell signalling. Uniquely among the ADAMs, ADAM-8 is constitutively expressed in neutrophils and is, thus far, the most intriguing ADAM in neutrophil biology [259,260]. Recent work in the context of ARDS has highlighted that ADAM-8 plays an important role in neutrophil adhesion and transmigration via its intracellular interactions with myosin and the cytoskeleton, independent of its catalytic domain [246]. Although not a direct consequence of its protease activity, this function of ADAM-8 has important implications for neutrophil proteolysis in general, since transmigration into the lung facilitates the delivery of granule proteases to the site of inflammation [261]. ADAM-8 shares most of its extracellular substrates with other ADAMs, including pro-TNFα and CD23, indicating a degree of redundancy in protease function [258,262]. However, its resistance to endogenous control by TIMP-1 and its high expression in the context of neutrophilic disease suggest that ADAM-8 may have potential to contribute specially to disease progression [263]. The more broadly expressed ADAM-10 also seems to have a role in promoting neutrophil adhesion to fibronectin and migration in response to CXCL8, as well as contributing to the development of pulmonary oedema [264]. On the other hand, ADAM-17, with which ADAM-10 shares many functions, may play an antagonistic role by shedding pro-adhesive L-selectin from neutrophils [264,265], highlighting the importance of a finely tuned protease balance in neutrophil function. At the time of writing, the authors are unaware of any reports linking neutrophil-derived ADAMTS proteases with lung inflammation.

3.7. Aspartyl Proteases

Human aspartyl proteases use an aspartate-bound activated water molecule for catalysis and are relatively few in number when compared to other protease families. Most that are not found in the digestive system are stored in the lysosomes of cells, though notably cathepsin D is localised in the primary granules of neutrophils [266]. In these granules, cathepsin D appears to play a central role in promoting neutrophil apoptosis by activating caspase-8 when primary granules are released into the cytoplasm following, for instance, bacterial phagocytosis. This proapoptotic mechanism is important in the resolution of inflammation and can be blocked by protease inhibition or treatment of neutrophils with pro-survival factors, like GM-CSF [266,267]. Cathepsin D is processed by cathepsins L and B and auto-activation [268], once again illustrating the intricate interactions that occur between these enzymes to regulate activity. Optimal cathepsin D activity is facilitated by the acidic pH of lysosomes or neutrophil granules, most probably limiting its influence in the extracellular environment [269]; it is unclear whether cathepsin D secreted from neutrophils has a role in pulmonary ECM processing. Other aspartyl proteases, such as cathepsin E and legumain, do not appear to exhibit activity in neutrophils.

3.8. Granzymes

Granzymes are serine proteases typically released from the granules of NK and cytotoxic T lymphocytes. Their primary function is the induction of apoptosis in targeted cells, via the activation of caspases and apoptotic proteins, such as Bid. Granzymes also mediate the killing of intracellular pathogens [270]. However, the status and function of neutrophil granzymes remains controversial. Naïve spleen or blood neutrophils are not thought to express granzymes [271], though some groups report that colon tumour-associated neutrophils and neutrophils exposed to inflammatory cytokines, such as IFN-γ, or bacteria demonstrate upregulated expression of granzymes A and B [272,273,274]. The targets of neutrophil-derived extracellular granzymes have not been elucidated in the context of the lung, though it has been suggested that they may affect ECM structure and initiate anoikis in nearby cells [275].

4. Conclusions and Future Directions

Altogether, the future of neutrophil protease research in lung disease is at a crossroads. Despite being highly researched, the NSPs have been poor therapeutic targets, with inhibitors of NE showing mixed efficacy in human trials. These issues may be resolved by implementing a targeted, personalised approach depending on NSP levels within the airways of each patient. Future clinical trials of NSP inhibitors could therefore consider stratifying patients based on the baseline activity of individual NSPs in the lung to identify the patients most likely to benefit from inhibitor therapy. However, new tools would be required to assess NSP status at the bedside, especially in the context of critical illness, where treatment decisions must be made rapidly. Furthermore, pulmonary sampling techniques (bronchoalveolar lavage, sputum collection, etc.) would have to be carefully selected to ensure that accurate and disease-relevant readouts are obtained to inform treatment selection.
An alternative approach is to target some of the less-researched proteases in the neutrophil, which may prove to have equally or more profound roles in disease. This approach is already showing promising results, with CTSC inhibitors, such as Brensocatib, having beneficial effects not seen with NE inhibitors. The success of this drug and others should encourage future research to target the cathepsins and other less well-researched neutrophil proteases in both acute and chronic lung inflammation.
To date, there is relatively little research elucidating the role of many of these neutrophil-expressed proteases in neutrophil function, and for some proteases there is a complete lack of data [227]. It is important that the roles of these proteases in neutrophil function are investigated to determine their potential as therapeutic targets in lung inflammation. It is worth noting that, since proteases have such important homeostatic roles, there is also a case for the focused development of organ-directed (direct application to the lung), neutrophil-specific or extracellular-only inhibitors, to mitigate lingering concerns over systemic protease inhibition.
As our understanding of how neutrophil proteases process one another and participate in parallel and interacting signalling pathways continues to develop, we will increasingly appreciate which proteases represent the best targets; these may be proteases with well-known effector functions, those that act upstream as master regulators, or both. With this broader understanding, there will be the possibility of developing novel specific inhibitors, combination inhibitors or inhibitors of master regulators that control an array of downstream proteases, as is appropriate in each disease context to achieve the maximal benefit with minimal off-target effects.

Author Contributions

Planning, design, drafting and revision of the article C.J.C., M.C.M., D.F.M. and C.C.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Medical Research Council (MR/X001504/1, MR/T016760/1 and MR/P022847/1—to C.C.T.) and the Department for the Economy Northern Ireland PhD studentship funding to C.J.C.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Adeloye, D.; Song, P.; Zhu, Y.; Campbell, H.; Sheikh, A.; Rudan, I. Global, Regional, and National Prevalence of, and Risk Factors for, Chronic Obstructive Pulmonary Disease (COPD) in 2019: A Systematic Review and Modelling Analysis. Lancet Respir. Med. 2022, 10, 447–458. [Google Scholar] [CrossRef]
  2. Safiri, S.; Carson-Chahhoud, K.; Noori, M.; Nejadghaderi, S.A.; Sullman, M.J.M.; Ahmadian Heris, J.; Ansarin, K.; Mansournia, M.A.; Collins, G.S.; Kolahi, A.-A.; et al. Burden of Chronic Obstructive Pulmonary Disease and Its Attributable Risk Factors in 204 Countries and Territories, 1990–2019: Results from the Global Burden of Disease Study 2019. BMJ 2022, 15, e069679. [Google Scholar] [CrossRef]
  3. Burki, T.K. The Economic Cost of Respiratory Disease in the UK. Lancet Respir. Med. 2017, 5, 381. [Google Scholar] [CrossRef]
  4. Diamond, M.; Peniston, H.L.; Sanghavi, D.; Mahapatra, S. Acute Respiratory Distress Syndrome. Available online: https://www.ncbi.nlm.nih.gov/books/NBK436002/ (accessed on 5 June 2022).
  5. Ramirez, J.A.; Wiemken, T.L.; Peyrani, P.; Arnold, F.W.; Kelley, R.; Mattingly, W.A.; Nakamatsu, R.; Pena, S.; Guinn, B.E.; Furmanek, S.P.; et al. Adults Hospitalized with Pneumonia in the United States: Incidence, Epidemiology, and Mortality. Clin. Infect. Dis. 2017, 65, 1806–1812. [Google Scholar] [CrossRef]
  6. Koulenti, D.; Tsigou, E.; Rello, J. Nosocomial Pneumonia in 27 ICUs in Europe: Perspectives from the EU-VAP/CAP Study. Eur. J. Clin. Microbiol. Infect. Dis. 2017, 36, 1999–2006. [Google Scholar] [CrossRef]
  7. Jasper, A.E.; McIver, W.J.; Sapey, E.; Walton, G.M. Understanding the Role of Neutrophils in Chronic Inflammatory Airway Disease. F1000Research 2019, 8, 557. [Google Scholar] [CrossRef]
  8. Korkmaz, B.; Horwitz, M.S.; Jenne, D.E.; Gauthier, F. Neutrophil Elastase, Proteinase 3, and Cathepsin G as Therapeutic Targets in Human Diseases. Pharmacol. Rev. 2010, 62, 726–759. [Google Scholar] [CrossRef]
  9. Bedi, P.; Davidson, D.J.; McHugh, B.J.; Rossi, A.G.; Hill, A.T. Blood Neutrophils Are Reprogrammed in Bronchiectasis. Am. J. Respir. Crit. Care Med. 2018, 198, 880–890. [Google Scholar] [CrossRef]
  10. Sapey, E.; Stockley, J.A.; Greenwood, H.; Ahmad, A.; Bayley, D.; Lord, J.M.; Insall, R.H.; Stockley, R.A. Behavioral and Structural Differences in Migrating Peripheral Neutrophils from Patients with Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2011, 183, 1176–1186. [Google Scholar] [CrossRef]
  11. Milara, J.; Juan, G.; Peiró, T.; Serrano, A.; Cortijo, J. Neutrophil Activation in Severe, Early-Onset COPD Patients versus Healthy Non-Smoker Subjects in Vitro: Effects of Antioxidant Therapy. Respiration 2012, 83, 147–158. [Google Scholar] [CrossRef]
  12. Kindt, G.C.; Gadek, J.E.; Weiland, J.E. Initial Recruitment of Neutrophils to Alveolar Structures in Acute Lung Injury. J. Appl. Physiol. 1991, 70, 1575–1585. [Google Scholar] [CrossRef]
  13. Mercer, P.F.; Williams, A.E.; Scotton, C.J.; José, R.J.; Sulikowski, M.; Moffatt, J.D.; Murray, L.A.; Chambers, R.C. Proteinase-Activated Receptor-1, CCL2, and CCL7 Regulate Acute Neutrophilic Lung Inflammation. Am. J. Respir. Cell Mol. Biol. 2014, 50, 144–157. [Google Scholar] [CrossRef]
  14. Williams, A.E.; José, R.J.; Mercer, P.F.; Brealey, D.; Parekh, D.; Thickett, D.R.; O’Kane, C.; McAuley, D.F.; Chambers, R.C. Evidence for Chemokine Synergy during Neutrophil Migration in ARDS. Thorax 2017, 72, 66–73. [Google Scholar] [CrossRef]
  15. Houston, N.; Stewart, N.; Smith, D.S.; Bell, S.C.; Champion, A.C.; Reid, D.W. Sputum Neutrophils in Cystic Fibrosis Patients Display a Reduced Respiratory Burst. J. Cyst. Fibros. 2013, 12, 352–362. [Google Scholar] [CrossRef]
  16. Gray, R.D.; Hardisty, G.; Regan, K.H.; Smith, M.; Robb, C.T.; Duffin, R.; MacKellar, A.; Felton, J.M.; Paemka, L.; McCullagh, B.N.; et al. Delayed Neutrophil Apoptosis Enhances NET Formation in Cystic Fibrosis. Thorax 2018, 73, 134–144. [Google Scholar] [CrossRef]
  17. Akashi, K.; Traver, D.; Miyamoto, T.; Weissman, I.L. A Clonogenic Common Myeloid Progenitor That Gives Rise to All Myeloid Lineages. Nature 2000, 404, 193–197. [Google Scholar] [CrossRef]
  18. Giladi, A.; Paul, F.; Herzog, Y.; Lubling, Y.; Weiner, A.; Yofe, I.; Jaitin, D.; Cabezas-Wallscheid, N.; Dress, R.; Ginhoux, F.; et al. Single-Cell Characterization of Haematopoietic Progenitors and Their Trajectories in Homeostasis and Perturbed Haematopoiesis. Nat. Cell Biol. 2018, 20, 836–846. [Google Scholar] [CrossRef]
  19. Mondemé, M.; Zeroual, Y.; Soulard, D.; Hennart, B.; Beury, D.; Saliou, J.-M.; Carnoy, C.; Sirard, J.-C.; Faveeuw, C. Amoxicillin Treatment of Pneumococcal Pneumonia Impacts Bone Marrow Neutrophil Maturation and Function. J. Leukoc. Biol. 2023, 115, 463–475. [Google Scholar] [CrossRef]
  20. Papayannopoulos, V.; Zychlinsky, A. NETs: A New Strategy for Using Old Weapons. Trends Immunol. 2009, 30, 513–521. [Google Scholar] [CrossRef]
  21. Kasperkiewicz, P.; Hempel, A.; Janiszewski, T.; Kołt, S.; Snipas, S.J.; Drag, M.; Salvesen, G.S. NETosis Occurs Independently of Neutrophil Serine Proteases. J. Biol. Chem. 2020, 295, 17624–17631. [Google Scholar] [CrossRef]
  22. Carnevale, R.; Leopizzi, M.; Dominici, M.; D’Amati, G.; Bartimoccia, S.; Nocella, C.; Cammisotto, V.; D’Amico, A.; Castellani, V.; Baratta, F.; et al. PAD4-Induced NETosis Via Cathepsin G-Mediated Platelet-Neutrophil Interaction in ChAdOx1 Vaccine-Induced Thrombosis—Brief Report. Arterioscler. Thromb. Vasc. Biol. 2023, 43, E396–E403. [Google Scholar] [CrossRef]
  23. Schauer, C.; Janko, C.; Munoz, L.E.; Zhao, Y.; Kienhöfer, 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] [PubMed]
  24. Baggiolini, M.; Walz, A.; Kunkel, S.L. Neutrophil-Activating Peptide-1/Interleukin 8, a Novel Cytokine That Activates Neutrophils. J. Clin. Investig. 1989, 84, 1045–1049. [Google Scholar] [CrossRef]
  25. Liu, R.; Bai, J.; Xu, G.; Xuan, L.; Zhang, T.; Meng, A.; Hou, Q. Multi-Allergen Challenge Stimulates Steriod-Resistant Airway Inflammation via NF-ΚB-Mediated IL-8 Expression. Inflammation 2013, 36, 845–854. [Google Scholar] [CrossRef]
  26. Guan, X.; Hou, Y.; Sun, F.; Yang, Z.; Li, C. Dysregulated Chemokine Signaling in Cystic Fibrosis Lung Disease: A Potential Therapeutic Target. Curr. Drug Targets 2016, 17, 1535–1544. [Google Scholar] [CrossRef]
  27. Gabrijelcic, J.; Acuña, A.; Profita, M.; Paternò, A.; Chung, K.F.; Vignola, A.M.; Rodríguez-Roisin, R. Neutrophil Airway Influx by Platelet-Activating Factor in Asthma: Role of Adhesion Molecules and LTB4 Expression. Eur. Respir. J. 2003, 22, 290–297. [Google Scholar] [CrossRef]
  28. Theriot, H.M.; Malaviarachchi, P.A.; Scott, M.G.; Appell, K.T.; Banerjee, S.K.; Pechous, R.D. Pulmonary Expression of Interleukin-17 Contributes to Neutrophil Infiltration into the Lungs during Pneumonic Plague. Infect. Immun. 2023, 91, e0013123. [Google Scholar] [CrossRef]
  29. Yonker, L.M.; Marand, A.; Muldur, S.; Hopke, A.; Leung, H.M.; De La Flor, D.; Park, G.; Pinsky, H.; Guthrie, L.B.; Tearney, G.J.; et al. Neutrophil Dysfunction in Cystic Fibrosis. J. Cyst. Fibros. 2021, 20, 1062–1071. [Google Scholar] [CrossRef]
  30. Ringholz, F.C.; Buchanan, P.J.; Clarke, D.T.; Millar, R.G.; McDermott, M.; Linnane, B.; Harvey, B.J.; McNally, P.; Urbach, V. Reduced 15-Lipoxygenase 2 and Lipoxin A4/Leukotriene B4 Ratio in Children with Cystic Fibrosis. Eur. Respir. J. 2014, 44, 394–404. [Google Scholar] [CrossRef]
  31. Martin, B.A.; Wright, J.L.; Thommasen, H.; Hogg, J.C. Effect of Pulmonary Blood Flow on the Exchange between the Circulating and Marginating Pool of Polymorphonuclear Leukocytes in Dog Lungs. J. Clin. Investig. 1982, 69, 1277–1285. [Google Scholar] [CrossRef]
  32. Doerschuk, C.M.; Allard, M.F.; Martin, B.A.; MacKenzie, A.; Autor, A.P.; Hogg, J.C. Marginated Pool of Neutrophils in Rabbit Lungs. J. Appl. Physiol. 1987, 63, 1806–1815. [Google Scholar] [CrossRef]
  33. Tirouvanziam, R.; Gernez, Y.; Conrad, C.K.; Moss, R.B.; Schrijver, I.; Dunn, C.E.; Davies, Z.A.; Herzenberg, L.A.; Herzenberg, L.A. Profound Functional and Signaling Changes in Viable Inflammatory Neutrophils Homing to Cystic Fibrosis Airways. Proc. Natl. Acad. Sci. USA 2008, 105, 4335–4339. [Google Scholar] [CrossRef]
  34. Margaroli, C.; Moncada-Giraldo, D.; Gulick, D.A.; Dobosh, B.; Giacalone, V.D.; Forrest, O.A.; Sun, F.; Gu, C.; Gaggar, A.; Kissick, H.; et al. Transcriptional Firing Represses Bactericidal Activity in Cystic Fibrosis Airway Neutrophils. Cell Rep. Med. 2021, 2, 100239. [Google Scholar] [CrossRef]
  35. Metzler, K.D.; Goosmann, C.; Lubojemska, A.; Zychlinsky, A.; Papayannopoulos, V. Myeloperoxidase-Containing Complex Regulates Neutrophil Elastase Release and Actin Dynamics during NETosis. Cell Rep. 2014, 8, 883–896. [Google Scholar] [CrossRef]
  36. Farley, K.; Stolley, J.M.; Zhao, P.; Cooley, J.; Remold-O’Donnell, E. A SerpinB1 Regulatory Mechanism Is Essential for Restricting Neutrophil Extracellular Trap Generation. J. Immunol. 2012, 189, 4574–4581. [Google Scholar] [CrossRef]
  37. Margaroli, C.; Garratt, L.W.; Horati, H.; Dittrich, A.S.; Rosenow, T.; Montgomery, S.T.; Frey, D.L.; Brown, M.R.; Schultz, C.; Guglani, L.; et al. Elastase Exocytosis by Airway Neutrophils Is Associated with Early Lung Damage in Children with Cystic Fibrosis. Am. J. Respir. Crit. Care Med. 2019, 199, 873–881. [Google Scholar] [CrossRef]
  38. Hughey, R.P.; Bruns, J.B.; Kinlough, C.L.; Harkleroad, K.L.; Tong, Q.; Carattino, M.D.; Johnson, J.P.; Stockand, J.D.; Kleyman, T.R. Epithelial Sodium Channels Are Activated by Furin-Dependent Proteolysis. J. Biol. Chem. 2004, 279, 18111–18114. [Google Scholar] [CrossRef]
  39. Caldwell, R.A.; Boucher, R.C.; Stutts, M.J. Neutrophil Elastase Activates Near-Silent Epithelial Na+ Channels and Increases Airway Epithelial Na+ Transport. Am. J. Physiol. Lung Cell. Mol. Physiol. 2005, 288, 813–819. [Google Scholar] [CrossRef]
  40. Haerteis, S.; Krappitz, M.; Bertog, M.; Krappitz, A.; Baraznenok, V.; Henderson, I.; Lindström, E.; Murphy, J.E.; Bunnett, N.W.; Korbmacher, C. Proteolytic Activation of the Epithelial Sodium Channel (ENaC) by the Cysteine Protease Cathepsin-S. Pflug. Arch. 2012, 464, 353–365. [Google Scholar] [CrossRef] [PubMed]
  41. McKelvey, M.C.; Abladey, A.A.; Small, D.M.; Doherty, D.F.; Williams, R.; Scott, A.; Spek, C.A.; Borensztajn, K.S.; Holsinger, L.; Booth, R.; et al. Cathepsin S Contributes to Lung Inflammation in Acute Respiratory Distress Syndrome. Am. J. Respir. Crit. Care Med. 2022, 205, 769–782. [Google Scholar] [CrossRef]
  42. Wang, T.; Zhu, Z.; Liu, Z.; Yi, L.; Yang, Z.; Bian, W.; Chen, W.; Wang, S.; Li, G.; Li, A.; et al. Plasma Neutrophil Elastase and Elafin as Prognostic Biomarker for Acute Respiratory Distress Syndrome: A Multicenter Survival and Longitudinal Prospective Observation Study. Shock 2017, 48, 168–174. [Google Scholar] [CrossRef]
  43. Perera, N.C.; Schilling, O.; Kittel, H.; Back, W.; Kremmer, E.; Jenne, D.E. NSP4, an Elastase-Related Protease in Human Neutrophils with Arginine Specificity. Proc. Natl. Acad. Sci. USA 2012, 109, 6229–6234. [Google Scholar] [CrossRef]
  44. Pham, C.T.N. Neutrophil Serine Proteases: Specific Regulators of Inflammation. Nat. Rev. Immunol. 2006, 6, 541–550. [Google Scholar] [CrossRef] [PubMed]
  45. Fouret, P.; du Bois, R.M.; Bernaudin, J.F.; Takahashi, H.; Ferrans, V.J.; Crystal, R.G. Expression of the Neutrophil Elastase Gene during Human Bone Marrow Cell Differentiation. J. Exp. Med. 1989, 169, 833–845. [Google Scholar] [CrossRef] [PubMed]
  46. Yoshimura, K.; Crystal, R.G. Transcriptional and Posttranscriptional Modulation of Human Neutrophil Elastase Gene Expression. Blood 1992, 79, 2733–2740. [Google Scholar] [CrossRef] [PubMed]
  47. Takahashi, H.; Nukiwa, T.; Basset, P.; Crystal, R.G. Myelomonocytic Cell Lineage Expression of the Neutrophil Elastase Gene. J. Biol. Chem. 1988, 263, 2543–2547. [Google Scholar] [CrossRef] [PubMed]
  48. Turk, D.; Janjić, V.; Stern, I.; Podobnik, M.; Lamba, D.; Dahl, S.W.; Lauritzen, C.; Pedersen, J.; Turk, V.; Turk, B. Structure of Human Dipeptidyl Peptidase I (Cathepsin C): Exclusion Domain Added to an Endopeptidase Framework Creates the Machine for Activation of Granular Serine Proteases. EMBO J. 2001, 20, 6570–6582. [Google Scholar] [CrossRef] [PubMed]
  49. Heinz, A.; Jung, M.C.; Jahreis, G.; Rusciani, A.; Duca, L.; Debelle, L.; Weiss, A.S.; Neubert, R.H.H.; Schmelzer, C.E.H. The Action of Neutrophil Serine Proteases on Elastin and Its Precursor. Biochimie 2012, 94, 192–202. [Google Scholar] [CrossRef] [PubMed]
  50. Skoreński, M.; Torzyk, K.; Sieńczyk, M. Neutrophil Serine Proteases. Rare Dis. Orphan Drugs J. 2023, 2, 6. [Google Scholar] [CrossRef]
  51. Janoff, A.; Scherer, J. Mediators of Inflammation in Leukocyte Lysosomes. J. Exp. Med. 1968, 128, 1137–1155. [Google Scholar] [CrossRef]
  52. Voynow, J.A.; Shinbashi, M. Neutrophil Elastase and Chronic Lung Disease. Biomolecules 2021, 11, 1065. [Google Scholar] [CrossRef] [PubMed]
  53. Makaryan, V.; Zeidler, C.; Bolyard, A.A.; Skokowa, J.; Rodger, E.; Kelley, M.L.; Boxer, L.A.; Bonilla, M.A.; Newburger, P.E.; Shimamura, A.; et al. The Diversity of Mutations and Clinical Outcomes for ELANE-Associated Neutropenia. Curr. Opin. Hematol. 2015, 22, 3–11. [Google Scholar] [CrossRef] [PubMed]
  54. Belaaouaj, A.; McCarthy, R.; Baumann, M.; Gao, Z.; Ley, T.J.; Abraham, S.N.; Shapiro, S.D. Mice Lacking Neutrophil Elastase Reveal Impaired Host Defense against Gram Negative Bacterial Sepsis. Nat. Med. 1998, 4, 615–618. [Google Scholar] [CrossRef]
  55. Tadié, J.-M.; Bae, H.-B.; Banerjee, S.; Zmijewski, J.W.; Abraham, E. Differential Activation of RAGE by HMGB1 Modulates Neutrophil-Associated NADPH Oxidase Activity and Bacterial Killing. Am. J. Physiol. Cell Physiol. 2012, 302, 249–256. [Google Scholar] [CrossRef] [PubMed]
  56. Belaaouaj, A.A.; Kim, K.S.; Shapiro, S.D. Degradation of Outer Membrane Protein A in Escherichia coli Killing by Neutrophil Elastase. Science 2000, 289, 1185–1187. [Google Scholar] [CrossRef] [PubMed]
  57. Campbell, E.J.; Owen, C.A. The Sulfate Groups of Chondroitin Sulfate- and Heparan Sulfate-Containing Proteoglycans in Neutrophil Plasma Membranes Are Novel Binding Sites for Human Leukocyte Elastase and Cathepsin G. J. Biol. Chem. 2007, 282, 14645–14654. [Google Scholar] [CrossRef]
  58. Genschmer, K.R.; Russell, D.W.; Lal, C.; Szul, T.; Bratcher, P.E.; Noerager, B.D.; Abdul Roda, M.; Xu, X.; Rezonzew, G.; Viera, L.; et al. Activated PMN Exosomes: Pathogenic Entities Causing Matrix Destruction and Disease in the Lung. Cell 2019, 176, 113–126.e15. [Google Scholar] [CrossRef] [PubMed]
  59. Huntington, J.A.; Read, R.J.; Carrell, R.W. Structure of a Serpin–Protease Complex Shows Inhibition by Deformation. Nature 2000, 407, 923–926. [Google Scholar] [CrossRef] [PubMed]
  60. Cooley, J.; Takayama, T.K.; Shapiro, S.D.; Schechter, N.M.; Remold-O’Donnell, E. The Serpin MNEI Inhibits Elastase-like and Chymotrypsin-like Serine Proteases through Efficient Reactions at Two Active Sites. Biochemistry 2001, 40, 15762–15770. [Google Scholar] [CrossRef]
  61. Cooley, J.; Sontag, M.K.; Accurso, F.J.; Remold-O’Donnell, E. SerpinB1 in Cystic Fibrosis Airway Fluids: Quantity, Molecular Form and Mechanism of Elastase Inhibition. Eur. Respir. J. 2011, 37, 1083–1090. [Google Scholar] [CrossRef]
  62. Korkmaz, B.; Poutrain, P.; Hazouard, E.; De Monte, M.; Attucci, S.; Gauthier, F.L. Competition between Elastase and Related Proteases from Human Neutrophil for Binding to A1-Protease Inhibitor. Am. J. Respir. Cell Mol. Biol. 2005, 32, 553–559. [Google Scholar] [CrossRef] [PubMed]
  63. Bai, X.; Hippensteel, J.; Leavitt, A.; Maloney, J.P.; Beckham, D.; Garcia, C.; Li, Q.; Freed, B.M.; Ordway, D.; Sandhaus, R.A.; et al. Hypothesis: Alpha-1-Antitrypsin Is a Promising Treatment Option for COVID-19. Med. Hypotheses 2021, 146, 110394. [Google Scholar] [CrossRef]
  64. Jacobsen, L.C.; Sørensen, O.E.; Cowland, J.B.; Borregaard, N.; Theilgaard-Mönch, K. The Secretory Leukocyte Protease Inhibitor (SLPI) and the Secondary Granule Protein Lactoferrin Are Synthesized in Myelocytes, Colocalize in Subcellular Fractions of Neutrophils, and Are Coreleased by Activated Neutrophils. J. Leukoc. Biol. 2008, 83, 1155–1164. [Google Scholar] [CrossRef] [PubMed]
  65. Cane, J.; Tregidgo, L.; Thulborn, S.; Finch, D.; Bafadhel, M. Antimicrobial Peptides Slpi and Beta Defensin-1 in Sputum Are Negatively Correlated with Fev1. Int. J. COPD 2021, 16, 1437–1447. [Google Scholar] [CrossRef] [PubMed]
  66. Guyot, N.; Butler, M.W.; McNally, P.; Weldon, S.; Greene, C.M.; Levine, R.L.; O’Neill, S.J.; Taggart, C.C.; McElvaney, N.G. Elafin, an Elastase-Specific Inhibitor, Is Cleaved by Its Cognate Enzyme Neutrophil Elastase in Sputum from Individuals with Cystic Fibrosis. J. Biol. Chem. 2008, 283, 32377–32385. [Google Scholar] [CrossRef] [PubMed]
  67. Weldon, S.; McNally, P.; McElvaney, N.G.; Elborn, J.S.; McAuley, D.F.; Wartelle, J.; Belaaouaj, A.; Levine, R.L.; Taggart, C.C. Decreased Levels of Secretory Leucoprotease Inhibitor in the Pseudomonas -Infected Cystic Fibrosis Lung Are Due to Neutrophil Elastase Degradation. J. Immunol. 2009, 183, 8148–8156. [Google Scholar] [CrossRef] [PubMed]
  68. Sagel, S.D.; Wagner, B.D.; Anthony, M.M.; Emmett, P.; Zemanick, E.T. Sputum Biomarkers of Inflammation and Lung Function Decline in Children with Cystic Fibrosis. Am. J. Respir. Crit. Care Med. 2012, 186, 857–865. [Google Scholar] [CrossRef]
  69. Yan, D.; Ye, Y.; Zhang, J.; Zhao, J.; Yu, J.; Luo, Q. Human Neutrophil Elastase Induces MUC5AC Overexpression in Chronic Rhinosinusitis Through MiR-146a. Am. J. Rhinol. Allergy 2020, 34, 59–69. [Google Scholar] [CrossRef]
  70. Voynow, J.A.; Young, L.R.; Wang, Y.; Horger, T.; Rose, M.C.; Fischer, B.M. Neutrophil Elastase Increases MUC5AC MRNA and Protein Expression in Respiratory Epithelial Cells. Am. J. Physiol.-Lung Cell. Mol. Physiol. 1999, 276, L835–L843. [Google Scholar] [CrossRef]
  71. Zhou, J.; Perelman, J.M.; Kolosov, V.P.; Zhou, X. Neutrophil Elastase Induces MUC5AC Secretion via Protease-Activated Receptor 2. Mol. Cell. Biochem. 2013, 377, 75–85. [Google Scholar] [CrossRef]
  72. Meyer, M.L.; Potts-Kant, E.N.; Ghio, A.J.; Fischer, B.M.; Foster, W.M.; Voynow, J.A. NAD(P)H Quinone Oxidoreductase 1 Regulates Neutrophil Elastase-Induced Mucous Cell Metaplasia. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012, 303, 181–188. [Google Scholar] [CrossRef]
  73. Park, J.-A.; Sharif, A.S.; Shiomi, T.; Kobzik, L.; Kasahara, D.I.; Tschumperlin, D.J.; Voynow, J.; Drazen, J.M. Human Neutrophil Elastase-Mediated Goblet Cell Metaplasia Is Attenuated in TACE-Deficient Mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2013, 304, 701–707. [Google Scholar] [CrossRef]
  74. Le Gars, M.; Descamps, D.; Roussel, D.; Saussereau, E.; Guillot, L.; Ruffin, M.; Tabary, O.; Hong, S.S.; Boulanger, P.; Paulais, M.; et al. Neutrophil Elastase Degrades Cystic Fibrosis Transmembrane Conductance Regulator via Calpains and Disables Channel Function in Vitro and in Vivo. Am. J. Respir. Crit. Care Med. 2013, 187, 170–179. [Google Scholar] [CrossRef] [PubMed]
  75. Walsh, D.E.; Greene, C.M.; Carroll, T.P.; Taggart, C.C.; Gallagher, P.M.; O’Neill, S.J.; McElvaney, N.G. Interleukin-8 Up-Regulation by Neutrophil Elastase Is Mediated by MyD88/IRAK/TRAF-6 in Human Bronchial Epithelium. J. Biol. Chem. 2001, 276, 35494–35499. [Google Scholar] [CrossRef] [PubMed]
  76. Clancy, D.M.; Sullivan, G.P.; Moran, H.B.T.; Henry, C.M.; Reeves, E.P.; McElvaney, N.G.; Lavelle, E.C.; Martin, S.J. Extracellular Neutrophil Proteases Are Efficient Regulators of IL-1, IL-33, and IL-36 Cytokine Activity but Poor Effectors of Microbial Killing. Cell Rep. 2018, 22, 2937–2950. [Google Scholar] [CrossRef] [PubMed]
  77. Rogan, M.P.; Taggart, C.C.; Greene, C.M.; Murphy, P.G.; O’Neill, S.J.; McElvaney, N.G. Loss of Microbicidal Activity and Increased Formation of Biofilm Due to Decreased Lactoferrin Activity in Patients with Cystic Fibrosis. J. Infect. Dis. 2004, 190, 1245–1253. [Google Scholar] [CrossRef] [PubMed]
  78. Ma, J.; Kummarapurugu, A.B.; Hawkridge, A.; Ghosh, S.; Zheng, S.; Voynow, J.A. Neutrophil Elastase-Regulated Macrophage Sheddome/Secretome and Phagocytic Failure. Am. J. Physiol. Lung Cell. Mol. Physiol. 2021, 321, L555–L565. [Google Scholar] [CrossRef] [PubMed]
  79. Domon, H.; Nagai, K.; Maekawa, T.; Oda, M.; Yonezawa, D.; Takeda, W.; Hiyoshi, T.; Tamura, H.; Yamaguchi, M.; Kawabata, S.; et al. Neutrophil Elastase Subverts the Immune Response by Cleaving Toll-like Receptors and Cytokines in Pneumococcal Pneumonia. Front. Immunol. 2018, 9, 732. [Google Scholar] [CrossRef]
  80. Shapiro, S.D.; Goldstein, N.M.; Houghton, A.M.; Kobayashi, D.K.; Kelley, D.; Belaaouaj, A. Neutrophil Elastase Contributes to Cigarette Smoke-Induced Emphysema in Mice. Am. J. Pathol. 2003, 163, 2329–2335. [Google Scholar] [CrossRef]
  81. Ghosh, A.; Coakley, R.D.; Ghio, A.J.; Muhlebach, M.S.; Esther, C.R.; Alexis, N.E.; Tarran, R. Chronic E-Cigarette Use Increases Neutrophil Elastase and Matrix Metalloprotease Levels in the Lung. Am. J. Respir. Crit. Care Med. 2019, 200, 1392–1401. [Google Scholar] [CrossRef]
  82. Thulborn, S.J.; Mistry, V.; Brightling, C.E.; Moffitt, K.L.; Ribeiro, D.; Bafadhel, M. Neutrophil Elastase as a Biomarker for Bacterial Infection in COPD. Respir. Res. 2019, 20, 170. [Google Scholar] [CrossRef]
  83. Lodge, K.M.; Vassallo, A.; Liu, B.; Long, M.; Tong, Z.; Newby, P.R.; Agha-Jaffa, D.; Paschalaki, K.; Green, C.E.; Belchamber, K.B.R.; et al. Hypoxia Increases the Potential for Neutrophil-Mediated Endothelial Damage in Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2022, 205, 903–916. [Google Scholar] [CrossRef]
  84. Koga, H.; Miyahara, N.; Fuchimoto, Y.; Ikeda, G.; Waseda, K.; Ono, K.; Tanimoto, Y.; Kataoka, M.; Gelfand, E.W.; Tanimoto, M.; et al. Inhibition of Neutrophil Elastase Attenuates Airway Hyperresponsiveness and Inflammation in a Mouse Model of Secondary Allergen Challenge: Neutrophil Elastase Inhibition Attenuates Allergic Airway Responses. Respir. Res. 2013, 14, 8. [Google Scholar] [CrossRef]
  85. Majka, G.; Mazurek, H.; Strus, M.; Ciszek-Lenda, M.; Szatanek, R.; Pac, A.; Golińska, E.; Marcinkiewicz, J. Chronic Bacterial Pulmonary Infections in Advanced Cystic Fibrosis Differently Affect the Level of Sputum Neutrophil Elastase, IL-8 and IL-6. Clin. Exp. Immunol. 2021, 205, 391–405. [Google Scholar] [CrossRef]
  86. Delgado, M.B.; Clark-Lewis, I.; Loetscher, P.; Langen, H.; Thelen, M.; Baggiolini, M.; Wolf, M. Rapid Inactivation of Stromal Cell-Derived Factor-1 by Cathepsin G Associated with Lymphocytes. Eur. J. Immunol. 2001, 31, 699–707. [Google Scholar] [CrossRef]
  87. Rivera-Marrero, C.A.; Stewart, J.; Shafer, W.M.; Roman, J. The Down-Regulation of Cathepsin G in THP-1 Monocytes after Infection with Mycobacterium Tuberculosis Is Associated with Increased Intracellular Survival of Bacilli. Infect. Immun. 2004, 72, 5712–5721. [Google Scholar] [CrossRef] [PubMed]
  88. Owen, C.A.; Campbell, M.A.; Sannes, P.L.; Boukedes, S.S.; Campbell, E.J. Cell Surface-Bound Elastase and Cathepsin G on Human Neutrophils: A Novel, Non-Oxidative Mechanism by Which Neutrophils Focus and Preserve Catalytic Activity of Serine Proteinases. J. Cell Biol. 1995, 131, 775–789. [Google Scholar] [CrossRef] [PubMed]
  89. Guo, J.; Tu, J.; Hu, Y.; Song, G.; Yin, Z. Cathepsin G Cleaves and Activates IL-36γ and Promotes the Inflammation of Psoriasis. Drug Des. Dev. Ther. 2019, 13, 581–588. [Google Scholar] [CrossRef] [PubMed]
  90. Richter, R.; Bistrian, R.; Escher, S.; Forssmann, W.-G.; Vakili, J.; Henschler, R.; Spodsberg, N.; Frimpong-Boateng, A.; Forssmann, U. Quantum Proteolytic Activation of Chemokine CCL15 by Neutrophil Granulocytes Modulates Mononuclear Cell Adhesiveness. J. Immunol. 2005, 175, 1599–1608. [Google Scholar] [CrossRef]
  91. Steinwede, K.; Maus, R.; Bohling, J.; Voedisch, S.; Braun, A.; Ochs, M.; Schmiedl, A.; Länger, F.; Gauthier, F.; Roes, J.; et al. Cathepsin G and Neutrophil Elastase Contribute to Lung-Protective Immunity against Mycobacterial Infections in Mice. J. Immunol. 2012, 188, 4476–4487. [Google Scholar] [CrossRef]
  92. Zhao, Y.; Olonisakin, T.F.; Xiong, Z.; Hulver, M.; Sayeed, S.; Yu, M.T.; Gregory, A.D.; Kochman, E.J.; Chen, B.B.; Mallampalli, R.K.; et al. Thrombospondin-1 Restrains Neutrophil Granule Serine Protease Function and Regulates the Innate Immune Response during Klebsiella Pneumoniae Infection. Mucosal Immunol. 2015, 8, 896–905. [Google Scholar] [CrossRef] [PubMed]
  93. Kavanaugh, J.S.; Leidal, K.G.; Nauseef, W.M.; Horswill, A.R. Cathepsin G Degrades Staphylococcus Aureus Biofilms. J. Infect. Dis. 2021, 223, 1865–1869. [Google Scholar] [CrossRef] [PubMed]
  94. Giese, M.; Turiello, N.; Molenda, N.; Palesch, D.; Meid, A.; Schroeder, R.; Basilico, P.; Benarafa, C.; Halatsch, M.-E.; Zimecki, M.; et al. Exogenous Cathepsin G Upregulates Cell Surface MHC Class I Molecules on Immune and Glioblastoma Cells. Oncotarget 2016, 7, 74602–74611. [Google Scholar] [CrossRef] [PubMed]
  95. Gudmann, N.S.; Manon-Jensen, T.; Sand, J.M.B.; Diefenbach, C.; Sun, S.; Danielsen, A.; Karsdal, M.A.; Leeming, D.J. Lung Tissue Destruction by Proteinase 3 and Cathepsin G Mediated Elastin Degradation Is Elevated in Chronic Obstructive Pulmonary Disease. Biochem. Biophys. Res. Commun. 2018, 503, 1284–1290. [Google Scholar] [CrossRef] [PubMed]
  96. Guyot, N.; Wartelle, J.; Malleret, L.; Todorov, A.A.; Devouassoux, G.; Pacheco, Y.; Jenne, D.E.; Belaaouaj, A. Unopposed Cathepsin G, Neutrophil Elastase, and Proteinase 3 Cause Severe Lung Damage and Emphysema. Am. J. Pathol. 2014, 184, 2197–2210. [Google Scholar] [CrossRef] [PubMed]
  97. Quinn, R.A.; Adem, S.; Mills, R.H.; Comstock, W.; DeRight Goldasich, L.; Humphrey, G.; Aksenov, A.A.; Melnik, A.V.; da Silva, R.; Ackermann, G.; et al. Neutrophilic Proteolysis in the Cystic Fibrosis Lung Correlates with a Pathogenic Microbiome. Microbiome 2019, 7, 23. [Google Scholar] [CrossRef] [PubMed]
  98. Farberman, M.M.; Akers, K.T.; Malone, J.P.; Erdman-Gilmore, P.; Townsend, R.R.; Ferkol, T. Airway Proteins Involved in Bacterial Clearance Susceptible to Cathepsin G Proteolysis. Eur. Respir. J. 2010, 35, 410–417. [Google Scholar] [CrossRef] [PubMed]
  99. Elaskalani, O.; Abdol Razak, N.B.; Metharom, P. Neutrophil Extracellular Traps Induce Aggregation of Washed Human Platelets Independently of Extracellular DNA and Histones. Cell Commun. Signal. 2018, 16, 24. [Google Scholar] [CrossRef] [PubMed]
  100. Guerra, M.; Frey, D.; Hagner, M.; Dittrich, S.; Paulsen, M.; Mall, M.A.; Schultz, C. Cathepsin G Activity as a New Marker for Detecting Airway Inflammation by Microscopy and Flow Cytometry. ACS Cent. Sci. 2019, 5, 539–548. [Google Scholar] [CrossRef]
  101. Witko-Sarsat, V.; Cramer, E.M.; Hieblot, C.; Guichard, J.; Nusbaum, P.; Lopez, S.; Lesavre, P.; Halbwachs-Mecarelli, L. Presence of Proteinase 3 in Secretory Vesicles: Evidence of a Novel, Highly Mobilizable Intracellular Pool Distinct from Azurophil Granules. Blood 1999, 94, 2487–2496. [Google Scholar] [CrossRef]
  102. Armstrong, L.; Godinho, S.I.H.; Uppington, K.M.; Whittington, H.A.; Millar, A.B. Tumour Necrosis Factor-α Processing in Interstitial Lung Disease: A Potential Role for Exogenous Proteinase-3. Clin. Exp. Immunol. 2009, 156, 336–343. [Google Scholar] [CrossRef] [PubMed]
  103. Sørensen, O.E.; Follin, P.; Johnsen, A.H.; Calafat, J.; Tjabringa, G.S.; Hiemstra, P.S.; Borregaard, N. Human Cathelicidin, HCAP-18, Is Processed to the Antimicrobial Peptide LL-37 by Extracellular Cleavage with Proteinase 3. Blood 2001, 97, 3951–3959. [Google Scholar] [CrossRef] [PubMed]
  104. Loison, F.; Zhu, H.; Karatepe, K.; Kasorn, A.; Liu, P.; Ye, K.; Zhou, J.; Cao, S.; Gong, H.; Jenne, D.E.; et al. Proteinase 3-Dependent Caspase-3 Cleavage Modulates Neutrophil Death and Inflammation. J. Clin. Investig. 2014, 124, 4445–4458. [Google Scholar] [CrossRef] [PubMed]
  105. Kao, R.C.; Wehner, N.G.; Skubitz, K.M.; Gray, B.H.; Hoidal, J.R. Proteinase 3. A Distinct Human Polymorphonuclear Leukocyte Proteinase That Produces Emphysema in Hamsters. J. Clin. Investig. 1988, 82, 1963–1973. [Google Scholar] [CrossRef] [PubMed]
  106. Witko-Sarsat, V.; Halbwachs-Mecarelli, L.; Schuster, A.; Nusbaum, P.; Ueki, I.; Canteloup, S.; Lenoir, G.; Descamps-Latscha, B.; Nadel, J.A. Proteinase 3, a Potent Secretagogue in Airways, Is Present in Cystic Fibrosis Sputum. Am. J. Respir. Cell Mol. Biol. 1999, 20, 729–736. [Google Scholar] [CrossRef] [PubMed]
  107. Perera, N.C.; Wiesmüller, K.-H.; Larsen, M.T.; Schacher, B.; Eickholz, P.; Borregaard, N.; Jenne, D.E. NSP4 Is Stored in Azurophil Granules and Released by Activated Neutrophils as Active Endoprotease with Restricted Specificity. J. Immunol. 2013, 191, 2700–2707. [Google Scholar] [CrossRef] [PubMed]
  108. Stevens, T.; Ekholm, K.; Gränse, M.; Lindahl, M.; Kozma, V.; Jungar, C.; Ottosson, T.; Falk-Håkansson, H.; Churg, A.; Wright, J.L.; et al. AZD9668: Pharmacological Characterization of a Novel Oral Inhibitor of Neutrophil Elastase. J. Pharmacol. Exp. Ther. 2011, 339, 313–320. [Google Scholar] [CrossRef] [PubMed]
  109. Kuna, P.; Jenkins, M.; O’Brien, C.D.; Fahy, W.A. AZD9668, a Neutrophil Elastase Inhibitor, plus Ongoing Budesonide/Formoterol in Patients with COPD. Respir. Med. 2012, 106, 531–539. [Google Scholar] [CrossRef]
  110. Vogelmeier, C.; Aquino, T.O.; O’Brien, C.D.; Perrett, J.; Gunawardena, K.A. A Randomised, Placebo-Controlled, Dose-Finding Study of AZD9668, an Oral Inhibitor of Neutrophil Elastase, in Patients with Chronic Obstructive Pulmonary Disease Treated with Tiotropium. COPD J. Chronic Obstr. Pulm. Dis. 2012, 9, 111–120. [Google Scholar] [CrossRef]
  111. Stockley, R.; De Soyza, A.; Gunawardena, K.; Perrett, J.; Forsman-Semb, K.; Entwistle, N.; Snell, N. Phase II Study of a Neutrophil Elastase Inhibitor (AZD9668) in Patients with Bronchiectasis. Respir. Med. 2013, 107, 524–533. [Google Scholar] [CrossRef]
  112. Elborn, J.S.; Perrett, J.; Forsman-Semb, K.; Marks-Konczalik, J.; Gunawardena, K.; Entwistle, N. Efficacy, Safety and Effect on Biomarkers of AZD9668 in Cystic Fibrosis. Eur. Respir. J. 2012, 40, 969–976. [Google Scholar] [CrossRef] [PubMed]
  113. Watz, H.; Nagelschmitz, J.; Kirsten, A.; Pedersen, F.; van der Mey, D.; Schwers, S.; Bandel, T.J.; Rabe, K.F. Safety and Efficacy of the Human Neutrophil Elastase Inhibitor BAY 85-8501 for the Treatment of Non-Cystic Fibrosis Bronchiectasis: A Randomized Controlled Trial. Pulm. Pharmacol. Ther. 2019, 56, 86–93. [Google Scholar] [CrossRef] [PubMed]
  114. Aikawa, N.; Kawasaki, Y. Clinical Utility of the Neutrophil Elastase Inhibitor Sivelestat for the Treatment of Acute Respiratory Distress Syndrome. Ther. Clin. Risk Manag. 2014, 10, 621–629. [Google Scholar] [CrossRef] [PubMed]
  115. Tagami, T.; Tosa, R.; Omura, M.; Fukushima, H.; Kaneko, T.; Endo, T.; Rinka, H.; Murai, A.; Yamaguchi, J.; Yoshikawa, K.; et al. Effect of a Selective Neutrophil Elastase Inhibitor on Mortality and Ventilator-Free Days in Patients with Increased Extravascular Lung Water: A Post Hoc Analysis of the PiCCO Pulmonary Edema Study. J. Intensive Care 2014, 2, 67. [Google Scholar] [CrossRef] [PubMed]
  116. Hayashida, K.; Fujishima, S.; Sasao, K.; Orita, T.; Toyoda, Y.; Kitano, M.; Hori, S. Early Administration of Sivelestat, the Neutrophil Elastase Inhibitor, in Adults for Acute Lung Injury following Gastric Aspiration. Shock 2011, 36, 223–227. [Google Scholar] [CrossRef]
  117. Pu, S.; Wang, D.; Liu, D.; Zhao, Y.; Qi, D.; He, J.; Zhou, G. Effect of Sivelestat Sodium in Patients with Acute Lung Injury or Acute Respiratory Distress Syndrome: A Meta-Analysis of Randomized Controlled Trials. BMC Pulm. Med. 2017, 17, 148. [Google Scholar] [CrossRef] [PubMed]
  118. Barth, P.; Bruijnzeel, P.; Wach, A.; Sellier Kessler, O.; Hooftman, L.; Zimmermann, J.; Naue, N.; Huber, B.; Heimbeck, I.; Kappeler, D.; et al. Single Dose Escalation Studies with Inhaled POL6014, a Potent Novel Selective Reversible Inhibitor of Human Neutrophil Elastase, in Healthy Volunteers and Subjects with Cystic Fibrosis. J. Cyst. Fibros. 2020, 19, 299–304. [Google Scholar] [CrossRef]
  119. Olivera, D.S.; Boggs, S.E.; Beenhouwer, C.; Aden, J.; Knall, C. Cellular Mechanisms of Mainstream Cigarette Smoke-Induced Lung Epithelial Tight Junction Permeability Changes in Vitro. Inhal. Toxicol. 2007, 19, 13–22. [Google Scholar] [CrossRef]
  120. Wan, Y.; Piao, L.; Xu, S.; Inoue, A.; Meng, X.; Lei, Y.; Huang, Z.; Wang, H.; Yue, X.; Shi, G.P.; et al. Cathepsin S Deficiency Improves Muscle Mass Loss and Dysfunction via the Modulation of Protein Metabolism in Mice under Pathological Stress Conditions. FASEB J. 2023, 37, e23086. [Google Scholar] [CrossRef]
  121. Staun-Ram, E.; Miller, A. Cathepsins (S and B) and Their Inhibitor Cystatin C in Immune Cells: Modulation by Interferon-β and Role Played in Cell Migration. J. Neuroimmunol. 2011, 232, 200–206. [Google Scholar] [CrossRef]
  122. Chu, X.; Sun, Z.; Baek, D.S.; Li, W.; Mellors, J.W.; Shapiro, S.D.; Dimitrov, D.S. Human Antibody Domains and Fragments Targeting Neutrophil Elastase as Candidate Therapeutics for Cancer and Inflammation-Related Diseases. Int. J. Mol. Sci. 2021, 22, 11136. [Google Scholar] [CrossRef]
  123. Gaggar, A.; Chen, J.; Chmiel, J.F.; Dorkin, H.L.; Flume, P.A.; Griffin, R.; Nichols, D.; Donaldson, S.H. Inhaled Alpha 1 -Proteinase Inhibitor Therapy in Patients with Cystic Fibrosis. J. Cyst. Fibros. 2016, 15, 227–233. [Google Scholar] [CrossRef] [PubMed]
  124. Keir, H.R.; Fong, C.J.; Crichton, M.L.; Barth, P.; Chevalier, E.; Brady, G.; Kennedy, G.; Zimmermann, J.; Bruijnzeel, P.L.B.; Dicker, A.J.; et al. Personalised Anti-Inflammatory Therapy for Bronchiectasis and Cystic Fibrosis: Selecting Patients for Controlled Trials of Neutrophil Elastase Inhibition. ERJ Open Res. 2019, 5, 00252–02018. [Google Scholar] [CrossRef] [PubMed]
  125. Drenth, J.; Jansonius, J.N.; Koekoek, R.; Swen, H.M.; Wolthers, B.G. Structure of Papain. Nature 1968, 218, 929–932. [Google Scholar] [CrossRef] [PubMed]
  126. Musil, D.; Zucic, D.; Turk, D.; Engh, R.A.; Mayr, I.; Huber, R.; Popovic, T.; Turk Towatari, V.T.; Katunuma, N.; Bode, W. The Refined 2.15 Å X-Ray Crystal Structure of Human Liver Cathepsin B: The Structural Basis for Its Specificity. EMBO J. 1991, 10, 2321–2330. [Google Scholar] [CrossRef] [PubMed]
  127. Patel, S.; Homaei, A.; El-Seedi, H.R.; Akhtar, N. Cathepsins: Proteases That Are Vital for Survival but Can Also Be Fatal. Biomed. Pharmacother. 2018, 105, 526–532. [Google Scholar] [CrossRef]
  128. Gowen, M.; Lazner, F.; Dodds, R.; Kapadia, R.; Feild, J.; Tavaria, M.; Bertoncello, I.; Drake, F.; Zavarselk, S.; Tellis, I.; et al. Cathepsin K Knockout Mice Develop Osteopetrosis Due to a Deficit in Matrix Degradation but Not Demineralization. J. Bone Miner. Res. 1999, 14, 1654–1663. [Google Scholar] [CrossRef] [PubMed]
  129. Turk, V.; Stoka, V.; Vasiljeva, O.; Renko, M.; Sun, T.; Turk, B.; Turk, D. Cysteine Cathepsins: From Structure, Function and Regulation to New Frontiers. Biochim. Biophys. Acta (BBA) Proteins Proteom. 2012, 1824, 68–88. [Google Scholar] [CrossRef]
  130. Lalmanach, G.; Saidi, A.; Bigot, P.; Chazeirat, T.; Lecaille, F.; Wartenberg, M. Regulation of the Proteolytic Activity of Cysteine Cathepsins by Oxidants. Int. J. Mol. Sci. 2020, 21, 1944. [Google Scholar] [CrossRef]
  131. Xian, M.; Chen, X.; Liu, Z.; Wang, K.; Wang, P.G. Inhibition of Papain by S-Nitrosothiols: Formation of Mixed Disulfides. J. Biol. Chem. 2000, 275, 20467–20473. [Google Scholar] [CrossRef]
  132. Poole, L.B.; Klomsiri, C.; Knaggs, S.A.; Furdui, C.M.; Nelson, K.J.; Thomas, M.J.; Fetrow, J.S.; Daniel, L.W.; King, S.B. Fluorescent and Affinity-Based Tools To Detect Cysteine Sulfenic Acid Formation in Proteins. Bioconjug. Chem. 2007, 18, 2004–2017. [Google Scholar] [CrossRef] [PubMed]
  133. Guay, J.; Falgueyret, J.P.; Ducret, A.; Percival, M.D.; Mancini, J.A. Potency and Selectivity of Inhibition of Cathepsin K, L and S by Their Respective Propeptides. Eur. J. Biochem. 2000, 267, 6311–6318. [Google Scholar] [CrossRef] [PubMed]
  134. Caglič, D.; Pungerčar, J.R.; Pejler, G.; Turk, V.; Turk, B. Glycosaminoglycans Facilitate Procathepsin B Activation through Disruption of Propeptide-Mature Enzyme Interactions. J. Biol. Chem. 2007, 282, 33076–33085. [Google Scholar] [CrossRef] [PubMed]
  135. Sage, J.; Mallèvre, F.; Barbarin-Costes, F.; Samsonov, S.A.; Gehrcke, J.P.; Pisabarro, M.T.; Perrier, E.; Schnebert, S.; Roget, A.; Livache, T.; et al. Binding of Chondroitin 4-Sulfate to Cathepsin S Regulates Its Enzymatic Activity. Biochemistry 2013, 52, 6487–6498. [Google Scholar] [CrossRef] [PubMed]
  136. Li, Z.; Yasuda, Y.; Li, W.; Bogyo, M.; Katz, N.; Gordon, R.E.; Fields, G.B.; Brömme, D. Regulation of Collagenase Activities of Human Cathepsins by Glycosaminoglycans. J. Biol. Chem. 2004, 279, 5470–5479. [Google Scholar] [CrossRef] [PubMed]
  137. Yasuda, Y.; Tsukuba, T.; Okamoto, K.; Kadowaki, T.; Yamamoto, K. The Role of the Cathepsin E Propeptide in Correct Folding, Maturation and Sorting to the Endosome. J. Biochem. 2005, 138, 621–630. [Google Scholar] [CrossRef]
  138. Buck, M.R.; Karustis, D.G.; Day, N.A.; Honn, K.V.; Sloane, B.F. Degradation of Extracellular-Matrix Proteins by Human Cathepsin B from Normal and Tumour Tissues. Biochem. J. 1992, 282, 273–278. [Google Scholar] [CrossRef] [PubMed]
  139. Yasuda, Y.; Li, Z.; Greenbaum, D.; Bogyo, M.; Weber, E.; Brömme, D. Cathepsin V, a Novel and Potent Elastolytic Activity Expressed in Activated Macrophages. J. Biol. Chem. 2004, 279, 36761–36770. [Google Scholar] [CrossRef] [PubMed]
  140. Stoka, V.; Turk, B.; Schendel, S.L.; Kim, T.H.; Cirman, T.; Snipas, S.J.; Ellerby, L.M.; Bredesen, D.; Freeze, H.; Abrahamson, M.; et al. Lysosomal Protease Pathways to Apoptosis: Cleavage of Bid, Not pro-Caspases, Is the Most Likely Route. J. Biol. Chem. 2001, 276, 3149–3157. [Google Scholar] [CrossRef]
  141. Droga-Mazovec, G.; Bojič, L.; Petelin, A.; Ivanova, S.; Romih, R.; Repnik, U.; Salvesen, G.S.; Stoka, V.; Turk, V.; Turk, B. Cysteine Cathepsins Trigger Caspase-Dependent Cell Death through Cleavage of Bid and Antiapoptotic Bcl-2 Homologues. J. Biol. Chem. 2008, 283, 19140–19150. [Google Scholar] [CrossRef]
  142. Creasy, B.M.; McCoy, K.L. Cytokines Regulate Cysteine Cathepsins during TLR Responses. Cell. Immunol. 2011, 267, 56–66. [Google Scholar] [CrossRef]
  143. Gunčar, G.; Pungerčič, G.; Klemenčič, I.; Turk, V.; Turk, D. Crystal Structure of MHC Class II-Associated P41 Ii Fragment Bound to Cathepsin L Reveals the Structural Basis for Differentiation between Cathepsins L and S. EMBO J. 1999, 18, 793–803. [Google Scholar] [CrossRef] [PubMed]
  144. Xu, T.; Nicolson, S.; Sandow, J.J.; Dayan, S.; Jiang, X.; Manning, J.A.; Webb, A.I.; Kumar, S.; Denton, D. Cp1/Cathepsin L Is Required for Autolysosomal Clearance in Drosophila. Autophagy 2021, 17, 2734–2749. [Google Scholar] [CrossRef] [PubMed]
  145. Kos, J.; Jevnikar, Z.; Obermajer, N. The Role of Cathepsin X in Cell Signaling. Cell Adhes. Migr. 2009, 3, 164–166. [Google Scholar] [CrossRef] [PubMed]
  146. Maciewicz, R.A.; Etherington, D.J. A Comparison of Four Cathepsins (B, L, N and S) with Collagenolytic Activity from Rabbit Spleen. Biochem. J. 1988, 256, 433–440. [Google Scholar] [CrossRef] [PubMed]
  147. Tate, S.; MacGregor, G.; Davis, M.; Innes, J.A.; Greening, A.P. Airways in Cystic Fibrosis Are Acidified: Detection by Exhaled Breath Condensate. Thorax 2002, 57, 926–929. [Google Scholar] [CrossRef] [PubMed]
  148. Turk, B.; Dolenc, I.; Turk, V.; Bieth, J.G. Kinetics of the PH-Induced Inactivation of Human Cathepsin L. Biochemistry 1993, 32, 375–380. [Google Scholar] [CrossRef] [PubMed]
  149. Taggart, C.C.; Greene, C.M.; Smith, S.G.; Levine, R.L.; McCray, P.B.; O’Neill, S.; McElvaney, N.G. Inactivation of Human β-Defensins 2 and 3 by Elastolytic Cathepsins. J. Immunol. 2003, 171, 931–937. [Google Scholar] [CrossRef] [PubMed]
  150. Davis, P.B. Cystic Fibrosis since 1938. Am. J. Respir. Crit. Care Med. 2006, 173, 475–482. [Google Scholar] [CrossRef]
  151. Taggart, C.C.; Lowe, G.J.; Greene, C.M.; Mulgrew, A.T.; O’Neill, S.J.; Levine, R.L.; McElvaney, N.G. Cathepsin B, L, and S Cleave and Inactivate Secretory Leucoprotease Inhibitor. J. Biol. Chem. 2001, 276, 33345–33352. [Google Scholar] [CrossRef]
  152. Layton, G.T.; Harris, S.J.; Bland, F.A.; Lee, S.R.; Fearn, S.; Kaleta, J.; Wood, M.L.; Bond, A.; Ward, G. Therapeutic Effects of Cysteine Protease Inhibition in Allergic Lung Inflammation: Inhibition of Allergen-Specific T Lymphocyte Migration. Inflamm. Res. 2001, 50, 400–408. [Google Scholar] [CrossRef] [PubMed]
  153. Deschamps, K.; Cromlish, W.; Weicker, S.; Lamontagne, S.; Huszar, S.L.; Gauthier, J.Y.; Mudgett, J.S.; Guimond, A.; Romand, R.; Frossard, N.; et al. Genetic and Pharmacological Evaluation of Cathepsin S in a Mouse Model of Asthma. Am. J. Respir. Cell Mol. Biol. 2011, 45, 81–87. [Google Scholar] [CrossRef] [PubMed]
  154. Small, D.M.; Brown, R.R.; Doherty, D.F.; Abladey, A.; Zhou-Suckow, Z.; Delaney, R.J.; Kerrigan, L.; Dougan, C.M.; Borensztajn, K.S.; Holsinger, L.; et al. Targeting of Cathepsin S Reduces Cystic Fibrosis-like Lung Disease. Eur. Respir. J. 2019, 53, 1801523. [Google Scholar] [CrossRef]
  155. 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]
  156. Gutmann, H.R.; Fruton, J.S. On the Proteolytic Enzymes of Animal Tissues. J. Biol. Chem. 1948, 174, 851–858. [Google Scholar] [CrossRef] [PubMed]
  157. Rao, N.V.; Rao, G.V.; Hoidal, J.R. Human Dipeptidyl-Peptidase I. J. Biol. Chem. 1997, 272, 10260–10265. [Google Scholar] [CrossRef] [PubMed]
  158. Dolenc, I.; Turk, B.; Pungercic, G.; Ritonja, A.; Turk, V. Oligomeric Structure and Substrate Induced Inhibition of Human Cathepsin C. J. Biol. Chem. 1995, 270, 21626–21631. [Google Scholar] [CrossRef] [PubMed]
  159. Hamon, Y.; Legowska, M.; Hervé, V.; Dallet-Choisy, S.; Marchand-Adam, S.; Vanderlynden, L.; Demonte, M.; Williams, R.; Scott, C.J.; Si-Tahar, M.; et al. Neutrophilic Cathepsin C Is Maturated by a Multistep Proteolytic Process and Secreted by Activated Cells during Inflammatory Lung Diseases. J. Biol. Chem. 2016, 291, 8486–8499. [Google Scholar] [CrossRef]
  160. Lamort, A.-S.; Hamon, Y.; Czaplewski, C.; Gieldon, A.; Seren, S.; Coquet, L.; Lecaille, F.; Lesner, A.; Lalmanach, G.; Gauthier, F.; et al. Processing and Maturation of Cathepsin C Zymogen: A Biochemical and Molecular Modeling Analysis. Int. J. Mol. Sci. 2019, 20, 4747. [Google Scholar] [CrossRef]
  161. Rebernik, M.; Lenarčič, B.; Novinec, M. The Catalytic Domain of Cathepsin C (Dipeptidyl-Peptidase I) Alone Is a Fully Functional Endoprotease. Protein Expr. Purif. 2019, 157, 21–27. [Google Scholar] [CrossRef]
  162. McGuire, M.J.; Lipsky, P.E.; Thiele, D.L. Generation of Active Myeloid and Lymphoid Granule Serine Proteases Requires Processing by the Granule Thiol Protease Dipeptidyl Peptidase I. J. Biol. Chem. 1993, 268, 2458–2467. [Google Scholar] [CrossRef] [PubMed]
  163. Adkison, A.M.; Raptis, S.Z.; Kelley, D.G.; Pham, C.T.N. Dipeptidyl Peptidase I Activates Neutrophil-Derived Serine Proteases and Regulates the Development of Acute Experimental Arthritis. J. Clin. Investig. 2002, 109, 363–371. [Google Scholar] [CrossRef] [PubMed]
  164. Gardiner, P.; Wikell, C.; Clifton, S.; Shearer, J.; Benjamin, A.; Peters, S.A. Neutrophil Maturation Rate Determines the Effects of Dipeptidyl Peptidase 1 Inhibition on Neutrophil Serine Protease Activity. Br. J. Pharmacol. 2016, 173, 2390–2401. [Google Scholar] [CrossRef] [PubMed]
  165. Liu, Y.G.; Teng, Y.S.; Cheng, P.; Kong, H.; Lv, Y.P.; Mao, F.Y.; Wu, X.L.; Hao, C.J.; Chen, W.; Yang, S.M.; et al. Abrogation of Cathepsin C by Helicobacter Pylori Impairs Neutrophil Activation to Promote Gastric Infection. FASEB J. 2019, 33, 5018–5033. [Google Scholar] [CrossRef]
  166. Toomes, C.; James, J.; Wood, A.J.; Wu, C.L.; McCormick, D.; Lench, N.; Hewitt, C.; Moynihan, L.; Roberts, E.; Woods, C.G.; et al. Loss-of-Function Mutations in the Cathepsin C Gene Result in Periodontal Disease and Palmoplantar Keratosis. Nat. Genet. 1999, 23, 421–424. [Google Scholar] [CrossRef] [PubMed]
  167. Sabry, S.; Abouzaid, M.R.; Mostafa, M.I.; Abdel-Hamid, M.S.; Saad, A.K.; Soliman, H.N.; Ahmed, N.E.-M.B. Abnormal Profiles of Cathepsin C Secreted in Urine of Papillon Lefevre Syndrome Patients. Eur. J. Med. Genet. 2022, 65, 104605. [Google Scholar] [CrossRef]
  168. Seren, S.; Abouzaid, M.R.; Eulenberg-Gustavus, C.; Hirschfeld, J.; Soliman, H.N.; Jerke, U.; N’Guessan, K.; Dallet-Choisy, S.; Lesner, A.; Lauritzen, C.; et al. Consequences of Cathepsin C Inactivation for Membrane Exposure of Proteinase 3, the Target Antigen in Autoimmune Vasculitis. J. Biol. Chem. 2018, 293, 12415–12428. [Google Scholar] [CrossRef] [PubMed]
  169. Roberts, H.; White, P.; Dias, I.; McKaig, S.; Veeramachaneni, R.; Thakker, N.; Grant, M.; Chapple, I. Characterization of Neutrophil Function in Papillon-Lefèvre Syndrome. J. Leukoc. Biol. 2016, 100, 433–444. [Google Scholar] [CrossRef] [PubMed]
  170. Sadik, C.D.; Noack, B.; Schacher, B.; Pfeilschifter, J.; Mühl, H.; Eickholz, P. Cytokine Production by Leukocytes of Papillon-Lefèvre Syndrome Patients in Whole Blood Cultures. Clin. Oral Investig. 2012, 16, 591–597. [Google Scholar] [CrossRef] [PubMed]
  171. Ikeshima, A. Papillon-Lefévre Syndrome: A Highly-Suspected Case. J. Oral Sci. 2006, 48, 257–260. [Google Scholar] [CrossRef]
  172. Hart, T.C.; Soskolne, W.A.; Hart, P.S.; Michalec, M.D.; Zhang, Y.; Firatli, E.; Van Dyke, T.E.; Stabholz, A.; Zlorogorski, A.; Shapira, L. Haim-Munk Syndrome and Papillon-Lefevre Syndrome Are Allelic Mutations in Cathepsin C. J. Med. Genet. 2000, 37, 88–94. [Google Scholar] [CrossRef]
  173. Pap, M.; Farkas, K.; Tóth, L.; Fábos, B.; Széll, M.; Németh, G.; Nagy, N. Identification of Putative Genetic Modifying Factors That Influence the Development of Papillon–Lefévre or Haim–Munk Syndrome Phenotypes. Clin. Exp. Dermatol. 2020, 45, 555–559. [Google Scholar] [CrossRef] [PubMed]
  174. Seren, S.; Derian, L.; Keles, I.; Guillon, A.; Lesner, A.; Gonzalez, L.; Baranek, T.; Si-Tahar, M.; Marchand-Adam, S.; Jenne, D.E.; et al. Proteinase Release from Activated Neutrophils in Mechanically Ventilated Patients with Non-COVID-19 and COVID-19 Pneumonia. Eur. Respir. J. 2021, 57, 2003755. [Google Scholar] [CrossRef] [PubMed]
  175. Benjamin, J.T.; Plosa, E.J.; Sucre, J.M.S.; van der Meer, R.; Dave, S.; Gutor, S.; Nichols, D.S.; Gulleman, P.M.; Jetter, C.S.; Han, W.; et al. Neutrophilic Inflammation during Lung Development Disrupts Elastin Assembly and Predisposes Adult Mice to COPD. J. Clin. Investig. 2021, 131, e139481. [Google Scholar] [CrossRef]
  176. Repnik, U.; Stoka, V.; Turk, V.; Turk, B. Lysosomes and Lysosomal Cathepsins in Cell Death. Biochim. Biophys. Acta Proteins Proteom. 2012, 1824, 22–33. [Google Scholar] [CrossRef]
  177. Thiele, D.L.; Lipsky, P.E. Mechanism of L-Leucyl-L-Leucine Methyl Ester-Mediated Killing of Cytotoxic Lymphocytes: Dependence on a Lysosomal Thiol Protease, Dipeptidyl Peptidase I, That Is Enriched in These Cells. Proc. Natl. Acad. Sci. USA 1990, 87, 83–87. [Google Scholar] [CrossRef]
  178. Kono, H.; Orlowski, G.M.; Patel, Z.; Rock, K.L. The IL-1–Dependent Sterile Inflammatory Response Has a Substantial Caspase-1–Independent Component That Requires Cathepsin C. J. Immunol. 2012, 189, 3734–3740. [Google Scholar] [CrossRef]
  179. Martinon, F.; Burns, K.; Tschopp, J. The Inflammasome: A Molecular Platform Triggering Activation of Inflammatory Caspases and Processing of ProIL-β. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef] [PubMed]
  180. Morales-Maldonado, A.; Humphry, M.; Figg, N.; Clarke, M.C. Human Vascular Smooth Muscle Cells Utilise Chymase for the Atypical Cleavage and Activation of Interleukin-1β. Atherosclerosis 2023, 390, 117308. [Google Scholar] [CrossRef]
  181. Guay, D.; Beaulieu, C.; Jagadeeswar Reddy, T.; Zamboni, R.; Methot, N.; Rubin, J.; Ethier, D.; David Percival, M. Design and Synthesis of Dipeptidyl Nitriles as Potent, Selective, and Reversible Inhibitors of Cathepsin C. Bioorg. Med. Chem. Lett. 2009, 19, 5392–5396. [Google Scholar] [CrossRef]
  182. Bondebjerg, J.; Fuglsang, H.; Valeur, K.R.; Pedersen, J.; Nærum, L. Dipeptidyl Nitriles as Human Dipeptidyl Peptidase I Inhibitors. Bioorg. Med. Chem. Lett. 2006, 16, 3614–3617. [Google Scholar] [CrossRef] [PubMed]
  183. Korkmaz, B.; Lesner, A.; Wysocka, M.; Gieldon, A.; Håkansson, M.; Gauthier, F.; Logan, D.T.; Jenne, D.E.; Lauritzen, C.; Pedersen, J. Structure-Based Design and in Vivo Anti-Arthritic Activity Evaluation of a Potent Dipeptidyl Cyclopropyl Nitrile Inhibitor of Cathepsin C. Biochem. Pharmacol. 2019, 164, 349–367. [Google Scholar] [CrossRef] [PubMed]
  184. Méthot, N.; Rubin, J.; Guay, D.; Beaulieu, C.; Ethier, D.; Reddy, T.J.; Riendeau, D.; Percival, M.D. Inhibition of the Activation of Multiple Serine Proteases with a Cathepsin C Inhibitor Requires Sustained Exposure to Prevent Pro-Enzyme Processing. J. Biol. Chem. 2007, 282, 20836–20846. [Google Scholar] [CrossRef] [PubMed]
  185. Doyle, K.; Lönn, H.; Käck, H.; Van De Poël, A.; Swallow, S.; Gardiner, P.; Connolly, S.; Root, J.; Wikell, C.; Dahl, G.; et al. Discovery of Second Generation Reversible Covalent DPP1 Inhibitors Leading to an Oxazepane Amidoacetonitrile Based Clinical Candidate (AZD7986). J. Med. Chem. 2016, 59, 9457–9472. [Google Scholar] [CrossRef]
  186. Basso, J.; Chen, K.J.; Zhou, Y.; Mark, L.; LaSala, D.; Dorfman, A.; Atalla, M.; Chun, D.; Viramontes, V.; Chang, C.; et al. The Pharmacokinetic Profile of Brensocatib and Its Effect on Pharmacodynamic Biomarkers Including NE, PR3, and CatG in Various Rodent Species. Front. Pharmacol. 2023, 14, 1208780. [Google Scholar] [CrossRef] [PubMed]
  187. McDonald, P.P.; Leifer, F.G.; Basso, J.; Lasala, D.; Li, D.; Chen, K.J.; Zhang, J.; Perkins, W.R.; Cipolla, D.C. Brensocatib (an Oral, Reversible Inhibitor of Dipeptidyl Peptidase-1) Attenuates Disease Progression in Two Animal Models of Rheumatoid Arthritis. Front. Immunol. 2023, 14, 1231047. [Google Scholar] [CrossRef] [PubMed]
  188. Chalmers, J.D.; Haworth, C.S.; Metersky, M.L.; Loebinger, M.R.; Blasi, F.; Sibila, O.; O’Donnell, A.E.; Sullivan, E.J.; Mange, K.C.; Fernandez, C.; et al. Phase 2 Trial of the DPP-1 Inhibitor Brensocatib in Bronchiectasis. N. Engl. J. Med. 2020, 383, 2127–2137. [Google Scholar] [CrossRef] [PubMed]
  189. Palmér, R.; Mäenpää, J.; Jauhiainen, A.; Larsson, B.; Mo, J.; Russell, M.; Root, J.; Prothon, S.; Chialda, L.; Forte, P.; et al. Dipeptidyl Peptidase 1 Inhibitor AZD7986 Induces a Sustained, Exposure-Dependent Reduction in Neutrophil Elastase Activity in Healthy Subjects. Clin. Pharmacol. Ther. 2018, 104, 1155–1164. [Google Scholar] [CrossRef]
  190. Keir, H.R.; Long, M.B.; Abo-Leyah, H.; Giam, Y.H.; Vadiveloo, T.; Pembridge, T.; Hull, R.C.; Delgado, L.; Band, M.; McLaren-Neil, F.; et al. Dipeptidyl Peptidase-1 Inhibition in Patients Hospitalised with COVID-19: A Multicentre, Double-Blind, Randomised, Parallel-Group, Placebo-Controlled Trial. Lancet Respir. Med. 2022, 10, 1119–1128. [Google Scholar] [CrossRef] [PubMed]
  191. Kreideweiss, S.; Schänzle, G.; Schnapp, G.; Vintonyak, V.; Grundl, M.A. BI 1291583: A Novel Selective Inhibitor of Cathepsin C with Superior in Vivo Profile for the Treatment of Bronchiectasis. Inflamm. Res. 2023, 72, 1709–1717. [Google Scholar] [CrossRef]
  192. Chalmers, J.D.; Gupta, A.; Chotirmall, S.H.; Armstrong, A.; Eickholz, P.; Hasegawa, N.; McShane, P.J.; O’donnell, A.E.; Shteinberg, M.; Watz, H.; et al. A Phase 2 Randomised Study to Establish Efficacy, Safety and Dosing of a Novel Oral Cathepsin C Inhibitor, BI 1291583, in Adults with Bronchiectasis: Airleaf. ERJ Open Res. 2023, 9, 00633–2022. [Google Scholar] [CrossRef] [PubMed]
  193. Miller, B.E.; Mayer, R.J.; Goyal, N.; Bal, J.; Dallow, N.; Boyce, M.; Carpenter, D.; Churchill, A.; Heslop, T.; Lazaar, A.L. Epithelial Desquamation Observed in a Phase I Study of an Oral Cathepsin C Inhibitor (GSK2793660). Br. J. Clin. Pharmacol. 2017, 83, 2813–2820. [Google Scholar] [CrossRef]
  194. Chen, X.; Yan, Y.; Zhang, Z.; Zhang, F.; Liu, M.; Du, L.; Zhang, H.; Shen, X.; Zhao, D.; Shi, J.B.; et al. Discovery and in Vivo Anti-Inflammatory Activity Evaluation of a Novel Non-Peptidyl Non-Covalent Cathepsin C Inhibitor. J. Med. Chem. 2021, 64, 11857–11885. [Google Scholar] [CrossRef]
  195. Chen, X.; Yan, Y.; Du, J.; Shen, X.; He, C.; Pan, H.; Zhu, J.; Liu, X. Non-Peptidyl Non-Covalent Cathepsin C InhibitoEEr Bearing a Unique Thiophene-Substituted Pyridine: Design, Structure-Activity Relationship and Anti-Inflammatory Activity in Vivo. Eur. J. Med. Chem. 2022, 236, 114368. [Google Scholar] [CrossRef] [PubMed]
  196. Veilleux, A.; Black, W.C.; Gauthier, J.Y.; Mellon, C.; Percival, M.D.; Tawa, P.; Falgueyret, J.P. Probing Cathepsin S Activity in Whole Blood by the Activity-Based Probe BIL-DMK: Cellular Distribution in Human Leukocyte Populations and Evidence of Diurnal Modulation. Anal. Biochem. 2011, 411, 43–49. [Google Scholar] [CrossRef] [PubMed]
  197. Weldon, S.; McNally, P.; McAuley, D.F.; Oglesby, I.K.; Wohlford-Lenane, C.L.; Bartlett, J.A.; Scott, C.J.; McElvaney, N.G.; Greene, C.M.; McCray, P.B.; et al. MiR-31 Dysregulation in Cystic Fibrosis Airways Contributes to Increased Pulmonary Cathepsin S Production. Am. J. Respir. Crit. Care Med. 2014, 190, 165–174. [Google Scholar] [CrossRef] [PubMed]
  198. Rørvig, S.; Østergaard, O.; Heegaard, N.H.H.; Borregaard, N. Proteome Profiling of Human Neutrophil Granule Subsets, Secretory Vesicles, and Cell Membrane: Correlation with Transcriptome Profiling of Neutrophil Precursors. J. Leukoc. Biol. 2013, 94, 711–721. [Google Scholar] [CrossRef] [PubMed]
  199. Shi, G.P.; Munger, J.S.; Meara, J.P.; Rich, D.H.; Chapman, H.A. Molecular Cloning and Expression of Human Alveolar Macrophage Cathepsin S, an Elastinolytic Cysteine Protease. J. Biol. Chem. 1992, 267, 7258–7262. [Google Scholar] [CrossRef] [PubMed]
  200. Vasiljeva, O.; Dolinar, M.; Pungerčar, J.R.; Turk, V.; Turk, B. Recombinant Human Procathepsin S Is Capable of Autocatalytic Processing at Neutral PH in the Presence of Glycosaminoglycans. FEBS Lett. 2005, 579, 1285–1290. [Google Scholar] [CrossRef]
  201. Kirschke, H.; Wiederanders, B.; Bromme, D.; Rinne, A. Cathepsin S from Bovine Spleen. Purification, Distribution, Intracellular Localization and Action on Proteins. Biochem. J. 1989, 264, 467–473. [Google Scholar] [CrossRef]
  202. Sukhova, G.K.; Shi, G.P.; Simon, D.I.; Chapman, H.A.; Libby, P. Expression of the Elastolytic Cathepsins S and K in Human Atheroma and Regulation of Their Production in Smooth Muscle Cells. J. Clin. Investig. 1998, 102, 576–583. [Google Scholar] [CrossRef]
  203. Caglic, D.; Turk, B.; Repnik, U.; Kosec, G.; Vasiljeva, O.; Turk, V.; Jedeszko, C.; Sloane, B.F.; Miniejew, C.; Kindermann, M.; et al. The Proinflammatory Cytokines Interleukin-1 α and Tumor Necrosis Factor α Promote the Expression and Secretion of Proteolytically Active Cathepsin S from Human Chondrocytes. Biol. Chem. 2013, 394, 307–316. [Google Scholar] [CrossRef] [PubMed]
  204. Sobotič, B.; Vizovišek, M.; Vidmar, R.; Van Damme, P.; Gocheva, V.; Joyce, J.A.; Gevaert, K.; Turk, V.; Turk, B.; Fonović, M. Proteomic Identification of Cysteine Cathepsin Substrates Shed from the Surface of Cancer Cells. Mol. Cell. Proteom. 2015, 14, 2213–2228. [Google Scholar] [CrossRef] [PubMed]
  205. Phipps-Yonas, H.; Semik, V.; Hastings, K.T. GILT Expression in B Cells Diminishes Cathepsin S Steady-State Protein Expression and Activity. Eur. J. Immunol. 2013, 43, 65–74. [Google Scholar] [CrossRef] [PubMed]
  206. Ross, E.A.; Smallie, T.; Ding, Q.; O’Neil, J.D.; Cunliffe, H.E.; Tang, T.; Rosner, D.R.; Klevernic, I.; Morrice, N.A.; Monaco, C.; et al. Dominant Suppression of Inflammation via Targeted Mutation of the MRNA Destabilizing Protein Tristetraprolin. J. Immunol. 2015, 195, 265–276. [Google Scholar] [CrossRef] [PubMed]
  207. Stellos, K.; Gatsiou, A.; Stamatelopoulos, K.; Perisic Matic, L.; John, D.; Lunella, F.F.; Jaé, N.; Rossbach, O.; Amrhein, C.; Sigala, F.; et al. Adenosine-to-Inosine RNA Editing Controls Cathepsin S Expression in Atherosclerosis by Enabling HuR-Mediated Post-Transcriptional Regulation. Nat. Med. 2016, 22, 1140–1150. [Google Scholar] [CrossRef]
  208. Xie, Y.; Fontenot, L.; Chupina Estrada, A.; Nelson, B.; Wang, J.; Shih, D.Q.; Ho, W.; Mattai, S.A.; Rieder, F.; Jensen, D.D.; et al. Elafin Reverses Intestinal Fibrosis by Inhibiting Cathepsin S-Mediated Protease-Activated Receptor 2. CMGH 2022, 14, 841–876. [Google Scholar] [CrossRef] [PubMed]
  209. Nit, J.; Abrahamson, M.; Zhang, M.; Fernandez, M.A.; Grubb, A.; Su, J.; Yu, G.L.; Li, Y.; Parmelee, D.; Xing, L.; et al. Cystatin E Is a Novel Human Cysteine Proteinase Inhibitor with Structural Resemblance to Family 2 Cystatins. J. Biol. Chem. 1997, 272, 10853–10858. [Google Scholar] [CrossRef] [PubMed]
  210. Wartenberg, M.; Andrault, P.M.; Saidi, A.; Bigot, P.; Nadal-Desbarats, L.; Lecaille, F.; Lalmanach, G. Oxidation of Cathepsin S by Major Chemicals of Cigarette Smoke. Free Radic. Biol. Med. 2020, 150, 53–65. [Google Scholar] [CrossRef]
  211. Headlam, H.A.; Gracanin, M.; Rodgers, K.J.; Davies, M.J. Inhibition of Cathepsins and Related Proteases by Amino Acid, Peptide, and Protein Hydroperoxides. Free Radic. Biol. Med. 2006, 40, 1539–1548. [Google Scholar] [CrossRef]
  212. Pires, D.; Bernard, E.M.; Pombo, J.P.; Carmo, N.; Fialho, C.; Gutierrez, M.G.; Bettencourt, P.; Anes, E. Mycobacterium Tuberculosis Modulates MiR-106b-5p to Control Cathepsin S Expression Resulting in Higher Pathogen Survival and Poor T-Cell Activation. Front. Immunol. 2017, 8, 1819. [Google Scholar] [CrossRef] [PubMed]
  213. Sendide, K.; Deghmane, A.-E.; Pechkovsky, D.; Av-Gay, Y.; Talal, A.; Hmama, Z. Mycobacterium Bovis BCG Attenuates Surface Expression of Mature Class II Molecules through IL-10-Dependent Inhibition of Cathepsin S. J. Immunol. 2005, 175, 5324–5332. [Google Scholar] [CrossRef] [PubMed]
  214. Pan, L.; Li, Y.; Jia, L.; Qin, Y.; Qi, G.; Cheng, J.; Qi, Y.; Li, H.; Du, J. Cathepsin S Deficiency Results in Abnormal Accumulation of Autophagosomes in Macrophages and Enhances Ang II-Induced Cardiac Inflammation. PLoS ONE 2012, 7, e35315. [Google Scholar] [CrossRef] [PubMed]
  215. Zhao, P.; Lieu, T.; Barlow, N.; Metcalf, M.; Veldhuis, N.A.; Jensen, D.D.; Kocan, M.; Sostegni, S.; Haerteis, S.; Baraznenok, V.; et al. Cathepsin S Causes Inflammatory Pain via Biased Agonism of PAR2 and TRPV4. J. Biol. Chem. 2014, 289, 27215–27234. [Google Scholar] [CrossRef]
  216. Zhou, P.P.; Zhang, W.Y.; Li, Z.F.; Chen, Y.R.; Kang, X.C.; Jiang, Y.X. Association between SNPs in the Promoter Region in Cathepsin S and Risk of Asthma in Chinese Han Population. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 2070–2076. [Google Scholar] [PubMed]
  217. Zheng, T.; Zhu, Z.; Wang, Z.; Homer, R.J.; Ma, B.; Riese, R.J.; Chapman, H.A.; Shapiro, S.D.; Elias, J.A. Inducible Targeting of IL-13 to the Adult Lung Causes Matrix Metalloproteinase- and Cathepsin-Dependent Emphysema. J. Clin. Investig. 2000, 106, 1081–1093. [Google Scholar] [CrossRef] [PubMed]
  218. Doherty, D.F.; Nath, S.; Poon, J.; Foronjy, R.F.; Ohlmeyer, M.; Dabo, A.J.; Salathe, M.; Birrell, M.; Belvisi, M.; Baumlin, N.; et al. Protein Phosphatase 2A Reduces Cigarette Smoke-Induced Cathepsin S and Loss of Lung Function. Am. J. Respir. Crit. Care Med. 2019, 200, 51–62. [Google Scholar] [CrossRef] [PubMed]
  219. Bigot, P.; Chesseron, S.; Saidi, A.; Sizaret, D.; Parent, C.; Petit-Courty, A.; Courty, Y.; Lecaille, F.; Lalmanach, G. Cleavage of Occludin by Cigarette Smoke-Elicited Cathepsin S Increases Permeability of Lung Epithelial Cells. Antioxidants 2023, 12, 5. [Google Scholar] [CrossRef] [PubMed]
  220. Andrault, P.M.; Schamberger, A.C.; Chazeirat, T.; Sizaret, D.; Renault, J.; Staab-Weijnitz, C.A.; Hennen, E.; Petit-Courty, A.; Wartenberg, M.; Saidi, A.; et al. Cigarette Smoke Induces Overexpression of Active Human Cathepsin S in Lungs from Current Smokers with or without COPD. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 317, L625–L638. [Google Scholar] [CrossRef]
  221. Repnik, U.; Starr, A.E.; Overall, C.M.; Turk, B. Cysteine Cathepsins Activate ELR Chemokines and Inactivate Non-ELR Chemokines. J. Biol. Chem. 2015, 290, 13800–13811. [Google Scholar] [CrossRef]
  222. Clair, J.M.-S.; Shi, G.-P.; Sutherland, R.E.; Chapman, H.A.; Caughey, G.H.; Wolters, P.J. Cathepsins L and S Are Not Required for Activation of Dipeptidyl Peptidase I (Cathepsin C) in Mice. Biol. Chem. 2006, 387, 1143–1146. [Google Scholar] [CrossRef] [PubMed]
  223. Dahl, S.W.; Halkier, T.; Lauritzen, C.; Dolenc, I.; Pedersen, J.; Turk, V.; Turk, B. Human Recombinant Pro-Dipeptidyl Peptidase I (Cathepsin C) Can Be Activated by Cathepsins L and S but Not by Autocatalytic Processing. Biochemistry 2001, 40, 1671–1678. [Google Scholar] [CrossRef] [PubMed]
  224. Canè, S.; Barouni, R.M.; Fabbi, M.; Cuozzo, J.; Fracasso, G.; Adamo, A.; Ugel, S.; Trovato, R.; De Sanctis, F.; Giacca, M.; et al. Neutralization of NET-Associated Human ARG1 Enhances Cancer Immunotherapy. Sci. Transl. Med. 2023, 15, eabq6221. [Google Scholar] [CrossRef] [PubMed]
  225. Geiger, R.; Rieckmann, J.C.; Wolf, T.; Basso, C.; Feng, Y.; Fuhrer, T.; Kogadeeva, M.; Picotti, P.; Meissner, F.; Mann, M.; et al. L-Arginine Modulates T Cell Metabolism and Enhances Survival and Anti-Tumor Activity. Cell 2016, 167, 829–842.e13. [Google Scholar] [CrossRef] [PubMed]
  226. Rotondo, R.; Bertolotto, M.; Barisione, G.; Astigiano, S.; Mandruzzato, S.; Ottonello, L.; Dallegri, F.; Bronte, V.; Ferrini, S.; Barbieri, O. Exocytosis of Azurophil and Arginase 1-Containing Granules by Activated Polymorphonuclear Neutrophils Is Required to Inhibit T Lymphocyte Proliferation. J. Leukoc. Biol. 2011, 89, 721–727. [Google Scholar] [CrossRef] [PubMed]
  227. Rincón, E.; Rocha-Gregg, B.L.; Collins, S.R. A Map of Gene Expression in Neutrophil-like Cell Lines. BMC Genom. 2018, 19, 573. [Google Scholar] [CrossRef]
  228. Akenhead, M.L.; Fukuda, S.; Schmid-Schoünbein, G.W.; Shin, H.Y. Fluid Shear-Induced Cathepsin B Release in the Control of Mac1-Dependent Neutrophil Adhesion. J. Leukoc. Biol. 2017, 102, 117–126. [Google Scholar] [CrossRef]
  229. Watson, A.; Öberg, L.; Angermann, B.; Spalluto, C.M.; Hühn, M.; Burke, H.; Cellura, D.; Freeman, A.; Muthas, D.; Etal, D.; et al. Dysregulation of COVID-19 Related Gene Expression in the COPD Lung. Respir. Res. 2021, 22, 164. [Google Scholar] [CrossRef]
  230. Lee, Y.T.; Chen, S.C.; Shyu, L.Y.; Lee, M.C.; Wu, T.C.; Tsao, S.M.; Yang, S.F. Significant Elevation of Plasma Cathepsin B and Cystatin C in Patients with Community-Acquired Pneumonia. Clin. Chim. Acta 2012, 413, 630–635. [Google Scholar] [CrossRef]
  231. Martin, S.L.; Moffitt, K.L.; McDowell, A.; Greenan, C.; Bright-Thomas, R.J.; Jones, A.M.; Webb, A.K.; Elborn, J.S. Association of Airway Cathepsin B and S with Inflammation in Cystic Fibrosis. Pediatr. Pulmonol. 2010, 45, 860–868. [Google Scholar] [CrossRef]
  232. Smith, M.A.; Schnellmann, R.G. Calpains, Mitochondria, and Apoptosis. Cardiovasc. Res. 2012, 96, 32–37. [Google Scholar] [CrossRef] [PubMed]
  233. Kovacs, L.; Su, Y. Redox-Dependent Calpain Signalling in Airway and Pulmonary Vascular and Remodelling in COPD. Adv. Exp. Med. Biol. 2017, 967, 139–160. [Google Scholar] [CrossRef] [PubMed]
  234. White, M.M.; Geraghty, P.; Hayes, E.; Cox, S.; Leitch, W.; Alfawaz, B.; Lavelle, G.M.; McElvaney, O.J.; Flannery, R.; Keenan, J.; et al. Neutrophil Membrane Cholesterol Content Is a Key Factor in Cystic Fibrosis Lung Disease. eBioMedicine 2017, 23, 173–184. [Google Scholar] [CrossRef] [PubMed]
  235. Heijink, I.H.; Brandenburg, S.M.; Postma, D.S.; Van Oosterhout, A.J.M. Cigarette Smoke Impairs Airway Epithelial Barrier Function and Cell-Cell Contact Recovery. Eur. Respir. J. 2012, 39, 419–428. [Google Scholar] [CrossRef] [PubMed]
  236. Francis, R.J.; Kotecha, S.; Hallett, M.B. Ca2+ Activation of Cytosolic Calpain Induces the Transition from Apoptosis to Necrosis in Neutrophils with Externalized Phosphatidylserine. J. Leukoc. Biol. 2013, 93, 95–100. [Google Scholar] [CrossRef]
  237. Lokuta, M.A.; Nuzzit, P.A.; Huttenlocher, A. Calpain Regulates Neutrophil Chemotaxis. Proc. Natl. Acad. Sci. USA 2003, 100, 4006–4011. [Google Scholar] [CrossRef] [PubMed]
  238. Al-Omari, M.; Korenbaum, E.; Ballmaier, M.; Lehmann, U.; Jonigk, D.; Manstein, D.J.; Welte, T.; Mahadeva, R.; Janciauskiene, S. Acute-Phase Protein A1-Antitrypsin Inhibits Neutrophil Calpain I and Induces Random Migration. Mol. Med. 2011, 17, 865–874. [Google Scholar] [CrossRef]
  239. Raeeszadeh-Sarmazdeh, M.; Do, L.D.; Hritz, B.G. Metalloproteinases and Their Inhibitors: Potential for the Development of New Therapeutics. Cells 2020, 9, 1313. [Google Scholar] [CrossRef]
  240. McGarry Houghton, A. Matrix Metalloproteinases in Destructive Lung Disease. Matrix Biol. 2015, 44–46, 167–174. [Google Scholar] [CrossRef]
  241. Dreymueller, D.; Uhlig, S.; Ludwig, A. ADAM-Family Metalloproteinases in Lung Inflammation: Potential Therapeutic Targets. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015, 308, L325–L343. [Google Scholar] [CrossRef]
  242. Paulissen, G.; Rocks, N.; Gueders, M.M.; Crahay, C.; Quesada-Calvo, F.; Bekaert, S.; Hacha, J.; Hour, M.E.; Foidart, J.-M.; Noel, A.; et al. Role of ADAM and ADAMTS Metalloproteinases in Airway Diseases. Respir. Res. 2009, 10, 127. [Google Scholar] [CrossRef]
  243. Christopoulou, M.E.; Papakonstantinou, E.; Stolz, D. Matrix Metalloproteinases in Chronic Obstructive Pulmonary Disease. Int. J. Mol. Sci. 2023, 24, 3786. [Google Scholar] [CrossRef] [PubMed]
  244. Davey, A.; McAuley, D.F.; O’Kane, C.M. Matrix Metalloproteinases in Acute Lung Injury: Mediators of Injury and Drivers of Repair. Eur. Respir. J. 2011, 38, 959–970. [Google Scholar] [CrossRef]
  245. Esposito, R.; Mirra, D.; Spaziano, G.; Panico, F.; Gallelli, L.; D’Agostino, B. The Role of MMPs in the Era of CFTR Modulators: An Additional Target for Cystic Fibrosis Patients? Biomolecules 2023, 13, 350. [Google Scholar] [CrossRef] [PubMed]
  246. Conrad, C.; Yildiz, D.; Cleary, S.J.; Margraf, A.; Cook, L.; Schlomann, U.; Panaretou, B.; Bowser, J.L.; Karmouty-Quintana, H.; Li, J.; et al. ADAM8 Signaling Drives Neutrophil Migration and ARDS Severity. JCI Insight 2022, 7, e149870. [Google Scholar] [CrossRef] [PubMed]
  247. Bradley, L.M.; Douglass, M.F.; Chatterjee, D.; Akira, S.; Baaten, B.J.G. Matrix Metalloprotease 9 Mediates Neutrophil Migration into the Airways in Response to Influenza Virus-Induced Toll-Like Receptor Signaling. PLoS Pathog. 2012, 8, e1002641. [Google Scholar] [CrossRef]
  248. Burgess, J.K.; Harmsen, M.C. Chronic Lung Diseases: Entangled in Extracellular Matrix. Eur. Respir. Rev. 2022, 31, 210202. [Google Scholar] [CrossRef]
  249. Owen, C.A.; Hu, Z.; Lopez-Otin, C.; Shapiro, S.D. Membrane-Bound Matrix Metalloproteinase-8 on Activated Polymorphonuclear Cells Is a Potent, Tissue Inhibitor of Metalloproteinase-Resistant Collagenase and Serpinase. J. Immunol. 2004, 172, 7791–7803. [Google Scholar] [CrossRef]
  250. Ardi, V.C.; Kupriyanova, T.A.; Deryugina, E.I.; Quigley, J.P. Human Neutrophils Uniquely Release TIMP-Free MMP-9 to Provide a Potent Catalytic Stimulator of Angiogenesis. Proc. Natl. Acad. Sci. USA 2007, 104, 20262–20267. [Google Scholar] [CrossRef]
  251. Deryugina, E.I.; Zajac, E.; Juncker-Jensen, A.; Kupriyanova, T.A.; Welter, L.; Quigley, J.P. Tissue-Infiltrating Neutrophils Constitute the Major In Vivo Source of Angiogenesis-Inducing MMP-9 in the Tumor Microenvironment. Neoplasia 2014, 16, 771–788. [Google Scholar] [CrossRef]
  252. García-Prieto, E.; González-López, A.; Cabrera, S.; Astudillo, A.; Gutiérrez-Fernández, A.; Fanjul-Fernandez, M.; Batalla-Solís, E.; Puente, X.S.; Fueyo, A.; López-Otín, C.; et al. Resistance to Bleomycin-Induced Lung Fibrosis in MMP-8 Deficient Mice Is Mediated by Interleukin-10. PLoS ONE 2010, 5, e13242. [Google Scholar] [CrossRef] [PubMed]
  253. Michaelis, J.; Vissers, M.C.; Winterbourn, C.C. Human Neutrophil Collagenase Cleaves Alpha 1-Antitrypsin. Biochem. J. 1990, 270, 809–814. [Google Scholar] [CrossRef]
  254. Tester, A.M.; Cox, J.H.; Connor, A.R.; Starr, A.E.; Dean, R.A.; Puente, X.S.; López-Otín, C.; Overall, C.M. LPS Responsiveness and Neutrophil Chemotaxis In Vivo Require PMN MMP-8 Activity. PLoS ONE 2007, 2, e312. [Google Scholar] [CrossRef] [PubMed]
  255. Van Den Steen, P.E.; Proost, P.; Wuyts, A.; Van Damme, J.; Opdenakker, G. Neutrophil Gelatinase B Potentiates Interleukin-8 Tenfold by Aminoterminal Processing, Whereas It Degrades CTAP-III, PF-4, and GRO-and Leaves RANTES and MCP-2 Intact. Blood 2000, 96, 2673–2681. [Google Scholar] [CrossRef] [PubMed]
  256. Xu, X.; Jackson, P.L.; Tanner, S.; Hardison, M.T.; Roda, M.A.; Blalock, J.E.; Gaggar, A. A Self-Propagating Matrix Metalloprotease-9 (MMP-9) Dependent Cycle of Chronic Neutrophilic Inflammation. PLoS ONE 2011, 6, e15781. [Google Scholar] [CrossRef] [PubMed]
  257. Starr, A.E.; Bellac, C.L.; Dufour, A.; Goebeler, V.; Overall, C.M. Biochemical Characterization and N-Terminomics Analysis of Leukolysin, the Membrane-Type 6 Matrix Metalloprotease (MMP25): Chemokine and Vimentin Cleavages Enhance Cell Migration and Macrophage Phagocytic Activities. J. Biol. Chem. 2012, 287, 13382–13395. [Google Scholar] [CrossRef]
  258. Edwards, D.R.; Handsley, M.M.; Pennington, C.J. The ADAM Metalloproteinases. Mol. Asp. Med. 2009, 29, 258–289. [Google Scholar] [CrossRef] [PubMed]
  259. Gómez-Gaviro, M.; Domínguez-Luis, M.; Canchado, J.; Calafat, J.; Janssen, H.; Lara-Pezzi, E.; Fourie, A.; Tugores, A.; Valenzuela-Fernández, A.; Mollinedo, F.; et al. Expression and Regulation of the Metalloproteinase ADAM-8 during Human Neutrophil Pathophysiological Activation and Its Catalytic Activity on L-Selectin Shedding. J. Immunol. 2007, 178, 8053–8063. [Google Scholar] [CrossRef] [PubMed]
  260. Dreymueller, D.; Pruessmeyer, J.; Schumacher, J.; Fellendorf, S.; Hess, F.M.; Seifert, A.; Babendreyer, A.; Bartsch, J.W.; Ludwig, A. The Metalloproteinase ADAM8 Promotes Leukocyte Recruitment in Vitro and in Acute Lung Inflammation. Am. J. Physiol. Lung Cell. Mol. Physiol. 2017, 313, L602–L614. [Google Scholar] [CrossRef]
  261. Yang, S.C.; Tsai, Y.F.; Pan, Y.L.; Hwang, T.L. Understanding the Role of Neutrophils in Acute Respiratory Distress Syndrome. Biomed. J. 2021, 44, 439–446. [Google Scholar] [CrossRef]
  262. Cook, L.; Gharzia, F.G.; Bartsch, J.W.; Yildiz, D. A Jack of All Trades—ADAM8 as a Signaling Hub in Inflammation and Cancer. FEBS J. 2023. [Google Scholar] [CrossRef] [PubMed]
  263. Amour, A.; English, W.R.; Knäuper, V.; Murphy, G.; Webster, A.; Slocombe, P.M.; Docherty, A.J.P.; Becherer, J.D. The Enzymatic Activity of ADAM8 and ADAM9 Is Not Regulated by TIMPs. FEBS Lett. 2002, 524, 154–158. [Google Scholar] [CrossRef] [PubMed]
  264. Pruessmeyer, J.; Hess, F.M.; Alert, H.; Groth, E.; Pasqualon, T.; Schwarz, N.; Nyamoya, S.; Kollert, J.; Van Der Vorst, E.; Donners, M.; et al. Leukocytes Require ADAM10 but Not ADAM17 for Their Migration and Inflammatory Recruitment into the Alveolar Space. Blood 2014, 123, 4077–4088. [Google Scholar] [CrossRef] [PubMed]
  265. Tang, J.; Zarbock, A.; Gomez, I.; Wilson, C.L.; Lefort, C.T.; Stadtmann, A.; Bell, B.; Huang, L.C.; Ley, K.; Raines, E.W. Adam17-Dependent Shedding Limits Early Neutrophil Influx but Does Not Alter Early Monocyte Recruitment to Inflammatory Sites. Blood 2011, 118, 786–794. [Google Scholar] [CrossRef] [PubMed]
  266. Conus, S.; Perozzo, R.; Reinheckel, T.; Peters, C.; Scapozza, L.; Yousefi, S.; Simon, H.U. Caspase-8 Is Activated by Cathepsin D Initiating Neutrophil Apoptosis during the Resolution of Inflammation. J. Exp. Med. 2008, 205, 685–698. [Google Scholar] [CrossRef] [PubMed]
  267. Conus, S.; Pop, C.; Snipas, S.J.; Salvesen, G.S.; Simon, H.U. Cathepsin D Primes Caspase-8 Activation by Multiple Intra-Chain Proteolysis. J. Biol. Chem. 2012, 287, 21142–21151. [Google Scholar] [CrossRef]
  268. Laurent-Matha, V.; Derocq, D.; Prébois, C.; Katunuma, N.; Liaudet-Coopman, E. Processing of Human Cathepsin D Is Independent of Its Catalytic Function and Auto-Activation: Involvement of Cathepsins L and B. J. Biochem. 2006, 139, 371. [Google Scholar] [CrossRef]
  269. Yadati, T.; Houben, T.; Bitorina, A.; Shiri-Sverdlov, R. The Ins and Outs of Cathepsins: Physiological Function and Role in Disease Management. Cells 2020, 9, 1679. [Google Scholar] [CrossRef] [PubMed]
  270. Chowdhury, D.; Lieberman, J. Death by a Thousand Cuts: Granzyme Pathways of Programmed Cell Death. Annu. Rev. Immunol. 2008, 26, 389–420. [Google Scholar] [CrossRef]
  271. Metkar, S.S.; Froelich, C.J. Human Neutrophils Lack Granzyme A, Granzyme B, and Perforin. Blood 2004, 104, 905–906. [Google Scholar] [CrossRef]
  272. Wagner, C.; Iking-Konert, C.; Denefleh, B.; Stegmaier, S.; Hug, F.; Hä, G.M. Granzyme B and Perforin: Constitutive Expression in Human Polymorphonuclear Neutrophils. Blood 2004, 103, 1099–1104. [Google Scholar] [CrossRef] [PubMed]
  273. Hochegger, K.; Eller, P.; Huber, J.M.; Bernhard, D.; Mayer, G.; Zlabinger, G.J.; Rosenkranz, A.R. Expression of Granzyme A in Human Polymorphonuclear Neutrophils. Immunology 2007, 121, 166–173. [Google Scholar] [CrossRef] [PubMed]
  274. Martin, A.; Seignez, C.; Racoeur, C.; Isambert, N.; Mabrouk, N.; Scagliarini, A.; Reveneau, S.; Arnould, L.; Bettaieb, A.; Jeannin, J.F.; et al. Tumor-Derived Granzyme B-Expressing Neutrophils Acquire Antitumor Potential after Lipid A Treatment. Oncotarget 2018, 9, 28378. [Google Scholar] [CrossRef] [PubMed]
  275. Mattila, J.T.; Maiello, P.; Sun, T.; Via, L.E.; Flynn, J.L. Granzyme B-Expressing Neutrophils Correlate with Bacterial Load in Granulomas from Mycobacterium Tuberculosis-Infected Cynomolgus Macaques. Cell. Microbiol. 2015, 17, 1085–1097. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A figure illustrating the subcellular locations of various neutrophil-derived proteases. Many proteases are stored in neutrophil granules and subsequently released into the extracellular environment during degranulation. Some are neutrophil membrane- or exosome-bound, and others, while known to be produced by neutrophils, are not known to reside in specific subcellular locations.
Figure 1. A figure illustrating the subcellular locations of various neutrophil-derived proteases. Many proteases are stored in neutrophil granules and subsequently released into the extracellular environment during degranulation. Some are neutrophil membrane- or exosome-bound, and others, while known to be produced by neutrophils, are not known to reside in specific subcellular locations.
Ijms 25 05492 g001
Table 1. Table summarising current clinical data regarding the efficacy of neutrophil elastase and cathepsin C inhibitors in inflammatory lung diseases. Abbreviations: NE—Neutrophil elastase, COPD—Chronic obstructive pulmonary disease, ARDS—acute respiratory distress syndrome, FEV1—Forced expiratory volume in 1 s, QoL—Quality of life, N/A—Not Available.
Table 1. Table summarising current clinical data regarding the efficacy of neutrophil elastase and cathepsin C inhibitors in inflammatory lung diseases. Abbreviations: NE—Neutrophil elastase, COPD—Chronic obstructive pulmonary disease, ARDS—acute respiratory distress syndrome, FEV1—Forced expiratory volume in 1 s, QoL—Quality of life, N/A—Not Available.
InhibitorCOPDCystic FibrosisBronchiectasisARDS
AZD9668No significant effects on pre-bronchodilator FEV1 or QoL. No significant effect on urine desmosine [107,108].No significant effects on sputum neutrophils, NE activity, lung function or QoL. Some statistically significant changes in IL-6 and urine desmosine [110].No significant effects on sputum neutrophils. FEV1 improved and levels of IL-6 and IL-8 decreased significantly [109].N/A.
SivelestatN/A.N/A.N/A.Approved for use in Japan [112]. Meta-analysis of six randomised control trials suggests no significant effects on 28–30-day mortality or ventilation days. Some evidence suggests Sivelestat may improve P/F level and may be more effective in mild-to-moderate ARDS [114,115].
POL6014N/A.Good results in pre-clinical investigations and good pharmacokinetics and dynamics. Clinical investigation of efficacy not yet performed [116].N/A.N/A.
BAY 85-8501N/A.N/A.No significant effects on FEV1. Significant reduction in NE activity; however, this had no clinical effects [111].N/A.
BrensocatibN/A.N/A.Increased time until first exacerbation and reduced overall incidence of exacerbations by around 40%. Number of severe exacerbations in treatment group were 50% lower [119,120].Severe COVID-19 patients treated with Brensocatib had increased mortality and increased need for supplemental oxygen. Treatment did reduce NE activity in blood however [121].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cheetham, C.J.; McKelvey, M.C.; McAuley, D.F.; Taggart, C.C. Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects. Int. J. Mol. Sci. 2024, 25, 5492. https://doi.org/10.3390/ijms25105492

AMA Style

Cheetham CJ, McKelvey MC, McAuley DF, Taggart CC. Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects. International Journal of Molecular Sciences. 2024; 25(10):5492. https://doi.org/10.3390/ijms25105492

Chicago/Turabian Style

Cheetham, Coby J., Michael C. McKelvey, Daniel F. McAuley, and Clifford C. Taggart. 2024. "Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects" International Journal of Molecular Sciences 25, no. 10: 5492. https://doi.org/10.3390/ijms25105492

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