Mast cells (MCs) are currently receiving increased attention among the scientific community, largely because of the recent identification of crucial functions for MCs in a variety of disorders. However, it is in many cases not clear exactly how MCs contribute in the respective settings. MCs express extraordinarily high levels of a number of proteases of chymase, tryptase, and carboxypeptidase A type, and these are stored in high amounts as active enzymes in the MC secretory granules. Hence, MC degranulation leads to the massive release of fully active MC proteases, which probably have a major impact on any condition in which MC degranulation occurs. Indeed, the recent generation and evaluation of mouse strains lacking individual MC proteases have indicated crucial contributions of these to a number of different disorders. MC proteases may thus account for many of the effects ascribed to MCs and are currently emerging as promising candidates for treatment of MC-driven disease. In this review, we discuss these findings.

Mast cells (MCs) are still mainly known for their harmful effects in connection with allergic reactions. However, this simplistic view is currently being extensively modified, and it is becoming more and more established that MCs have a complex array of functions and can contribute to a number of additional disorders.1  In addition to being detrimental, MCs also carry out a number of beneficial functions, most notably in connection with innate immune responses toward various pathogens.2  Moreover, recent studies indicate that MCs can play important roles in down-regulating adaptive immune responses.3,4  As a consequence of this progress, MCs are gaining a massively increased interest worldwide, and much effort is invested into investigations of the mechanisms by which MCs contribute to different disorders.

A striking morphologic feature of MCs is their abundance of electron-dense secretory granules, which contain large amounts of preformed compounds commonly referred to as “MC mediators.” These include biogenic amines (histamine and serotonin), certain preformed cytokines (most notably tumor necrosis factor), serglycin proteoglycans, various lysosomal enzymes, and a number of MC-specific proteases of chymase, tryptase, or carboxypeptidase A (MC-CPA) type.5-8  When MCs are induced to undergo degranulation, for example, through engagement of FcϵRI-bound IgE by polyvalent antigen, these mediators are thus released.9 

The MC proteases are expressed at exceptionally high levels, with mRNA levels approaching and even exceeding those of classic housekeeping genes,10  and they are stored in remarkably high amounts. Indeed, it has been calculated that MC proteases may account for more than 25% of the total MC protein.11,12  Importantly, in contrast to, for example, the pancreatic digestive proteases, the MC proteases are stored in fully active form; and when MCs undergo degranulation, large amounts of enzymatically active proteases are thus released into the extracellular space and probably have a profound impact on any condition in which MC degranulation occurs (Figure 1). Indeed, a plethora of potential functions of the MC proteases have previously been outlined, based on various approaches.6  However, it is not until relatively recently that the in vivo functions of these enzymes have started to become unraveled through experimental approaches involving MC protease-deficient mice. In this review, we summarize these findings and discuss their implications.

Figure 1

Storage and release of MC proteases. Mast cell (MC) proteases are synthesized as preproenzymes where both the signal (pre-) and activation (pro-) peptides are cleaved off intracellularly, a process in which dipeptidyl peptidase I/cathepsin C has a key role.13  Hence, enzymatically active proteases are stored in the MC granule. Major MC tryptases are tetrameric enzymes, whereas chymases and mast cell carboxypeptidase A (MC-CPA) are monomeric. Notably, for many MC proteases, the storage is strongly dependent on electrostatic interaction with serglycin proteoglycan, whereas others are stored independently of serglycin. After MC degranulation, MC proteases are released in complexes with serglycin. At exposure to extracellular pH, MC proteases may either be dissociated from serglycin or remain associated.

Figure 1

Storage and release of MC proteases. Mast cell (MC) proteases are synthesized as preproenzymes where both the signal (pre-) and activation (pro-) peptides are cleaved off intracellularly, a process in which dipeptidyl peptidase I/cathepsin C has a key role.13  Hence, enzymatically active proteases are stored in the MC granule. Major MC tryptases are tetrameric enzymes, whereas chymases and mast cell carboxypeptidase A (MC-CPA) are monomeric. Notably, for many MC proteases, the storage is strongly dependent on electrostatic interaction with serglycin proteoglycan, whereas others are stored independently of serglycin. After MC degranulation, MC proteases are released in complexes with serglycin. At exposure to extracellular pH, MC proteases may either be dissociated from serglycin or remain associated.

Close modal

The term “MC proteases” usually refers to the proteases that are expressed specifically by MCs and are stored in their secretory granules, that is, chymases, tryptases, and MC-CPA. However, it should be pointed out that MCs express a number of additional, non–MC-specific, proteases, such as lysosomal cathepsins, granzymes, neurolysin, and, possibly, cathepsin G.6  Recently, the strictly MC-specific expression of the chymases has been used to generate mouse strains in which the Cre recombinase is expressed under the control of chymase promoters, strains that can be used to specifically inactivate genes of interest in MCs.14,15 

Of the 3 classes of MC proteases, the tryptases and chymases are serine proteases, whereas MC-CPA is a Zn-dependent metalloprotease.6,16  Tryptases are denoted so because of their trypsin-like substrate specificity; that is, they cleave preferentially after Lys/Arg residues (Table 1). A unique feature of the secreted MC tryptases is that they are tetrameric, with all of the active sites facing a narrow central pore17  (Figure 1). Accordingly, their active sites are largely inaccessible for macromolecular protease inhibitors, and tryptase therefore resists inhibition by any of the proteinaceous protease inhibitors found in mammals. Further, this also limits the access to potential substrates.17  In contrast to the tryptases, chymases are monomeric and have chymotrypsin-like specificities; that is, they cleave preferentially after aromatic amino acid residues. MC-CPA is a monomeric exopeptidase, cleaving amino acid residues from the C-terminal end of peptides/proteins.16 

Table 1

Designation, chromosomal location, and primary substrate specificities of major human and murine MC proteases

Protein designationGene nameChromosomal locationPrimary substrate specificity*
Chymase (human) CMA1 14q11.2 Chymotrypsin-like (aromatic amino acid) 
mMCP-1 Mcpt1 14C1/2 Chymotrypsin-like (aromatic amino acid) 
mMCP-2 Mcpt2 14C1/2 Unknown (low enzymatic activity) 
mMCP-4 Mcpt4 14C1/2 Chymotrypsin-like (aromatic amino acid) 
mMCP-5 Cma1/Mcpt5 14C1/2 Elastase-like (Val/Ala/Ile) 
MC-CPA (human) CPA3 3q24 CPA-like (aromatic amino acid); exopeptidase 
MC-CPA (mouse) Cpa3 3A3 CPA-like (aromatic amino acid); exopeptidase 
Tryptase βII/βIII (human) TPSAB1 16p13.3 Trypsin-like (Arg/Lys) 
Tryptase αI/βI (human) TPSA1 16p13.3 Trypsin-like (Arg/Lys) 
mMCP-6 Mcpt6 17A3.3 Trypsin-like (Arg/Lys) 
mMCP-7 Tpsab1 17A3.3 Trypsin-like (Arg/Lys) 
Protein designationGene nameChromosomal locationPrimary substrate specificity*
Chymase (human) CMA1 14q11.2 Chymotrypsin-like (aromatic amino acid) 
mMCP-1 Mcpt1 14C1/2 Chymotrypsin-like (aromatic amino acid) 
mMCP-2 Mcpt2 14C1/2 Unknown (low enzymatic activity) 
mMCP-4 Mcpt4 14C1/2 Chymotrypsin-like (aromatic amino acid) 
mMCP-5 Cma1/Mcpt5 14C1/2 Elastase-like (Val/Ala/Ile) 
MC-CPA (human) CPA3 3q24 CPA-like (aromatic amino acid); exopeptidase 
MC-CPA (mouse) Cpa3 3A3 CPA-like (aromatic amino acid); exopeptidase 
Tryptase βII/βIII (human) TPSAB1 16p13.3 Trypsin-like (Arg/Lys) 
Tryptase αI/βI (human) TPSA1 16p13.3 Trypsin-like (Arg/Lys) 
mMCP-6 Mcpt6 17A3.3 Trypsin-like (Arg/Lys) 
mMCP-7 Tpsab1 17A3.3 Trypsin-like (Arg/Lys) 

MC indicates mast cell; mMCP, mouse mast cell protease; and CPA, carboxypeptidase A.

*

Refers to the preferred amino acid residue N-terminal of the cleavage site (P1 position).

The exact repertoire of expressed MC proteases varies among species and also among different MC subsets6-8  (Table 2). In humans, MCs are classified according to their protease content, with the MCT subclass expressing tryptase (α and β) only and the MCTC subclass expressing all types of MC proteases, that is, tryptase, chymase, and MC-CPA (Table 2). In mouse, MCs are divided into the connective tissue MC (CTMC) and mucosal MC (MMC) subtypes. CTMCs express predominantly 2 different chymases (the β-chymase mouse mast cell protease 4 [mMCP-4] and the α-chymase mMCP-5), whereas MMCs express predominantly 2 types of β-chymases (mMCP-1 and mMCP-2). CTMCs also express 2 tetrameric tryptases (mMCP-6 and mMCP-7) as well as MC-CPA (Table 2).

Table 2

Distribution of human and murine MC proteases among MC subclasses

SpeciesMC subclassTryptases*ChymasesMC-CPA
Human MCT α   
  β (I, II, III)   
 MCTC α CMA1 MC-CPA 
  β (I, II, III)   
Mouse MMC  mMCP-1  
   mMCP-2  
 CTMC mMCP-6 mMCP-4 MC-CPA 
  mMCP-7 mMCP-5  
   mMCP-9  
SpeciesMC subclassTryptases*ChymasesMC-CPA
Human MCT α   
  β (I, II, III)   
 MCTC α CMA1 MC-CPA 
  β (I, II, III)   
Mouse MMC  mMCP-1  
   mMCP-2  
 CTMC mMCP-6 mMCP-4 MC-CPA 
  mMCP-7 mMCP-5  
   mMCP-9  

MC indicates mast cell; MMC, mucosal mast cell; CTMC, connective tissue mast cell; mMCP, mouse mast cell protease; and CPA, carboxypeptidase A.

*

Refers to tetrameric, secreted tryptases. Various MC populations express a number of other trypsin-like proteases.6-8 

Preferentially expressed in uterine mast cells.18 

The highly efficient storage of the MC proteases is made possible through their tight packaging in complexes with serglycin proteoglycan, the latter consisting of a small protein core to which highly negatively charged glycosaminoglycans of chondroitin sulfate or heparin type are attached19  (Figure 1). Notably, the knockout of the serglycin gene resulted in an almost complete inability of MCs to store several MC proteases.20  Most probably, the storage-promoting function of serglycin is explained by the high extent of sulfation of its glycosaminoglycan components, with the sulfate groups engaging in electrostatic interactions with corresponding basic regions of the MC proteases. In support of this notion, targeting of N-deacetylase/N-sulfotransferase 2, an enzyme that is essential for the sulfation of heparin, caused similar storage defects as those imposed by serglycin deficiency.21,22  However, it should be pointed out that not all of the MC proteases depend on serglycin for storage: whereas mMCP-4, -5, -6, and MC-CPA20  are strictly serglycin-dependent, mMCP-2 storage is only partially dependent, and mMCP-1 and -7 storage is unaffected by the absence of serglycin.23  The most probable reason for the differential serglycin dependence of the various MC proteases lies within their exposure of surface basic charge. For example, mMCP-1 has a considerably lower net positive charge than mMCP-4 and -5, well explaining why mMCP-4 and -5 are strongly dependent on serglycin for storage whereas mMCP-1 is not.

Human MCs express 2 main, enzymatically active tryptases that are secreted as tetramers during MC degranulation: α- and β-tryptases (β I- III). The murine tryptases (mMCP-6 and mMCP-7) are also tetrameric and most probably constitute the counterparts to human α- and β-tryptase. In addition, both human and mouse MCs express a monomeric, membrane-bound tryptase as well as a number of other trypsin-like serine proteases.6-8  To study the biologic function of MC tryptase, several different approaches have been used, including instillation of purified or recombinant tryptases and assessment of tryptase inhibitors in animal models for disease.6-8  Recently, a more precise evaluation of the biologic function of MC tryptase has been made possible through the generation of mouse strains deficient in mMCP-6, the murine tryptase that may constitute the closest homolog to human β-tryptase.24,25  In the mMCP-6−/− strain generated by Thakurdas et al,25  the targeting of the mMCP-6 gene also resulted in inactivation of mMCP-7 expression and, hence, this strain is double deficient in mMCP-6 and -7. In contrast, the mMCP-6−/− strain reported by Shin et al retained mMCP-7 expression.24  As a tool for studying mMCP-7 function, several mouse strains, including the C57BL/6 strain, are known to lack the expression of mMCP-7 because of spontaneous mutations at various positions of the mMCP-7 gene.26 

Parasite infection

MCs are widely recognized as effector cells involved in clearance of diverse parasites, including Trichinella spiralis,27  although the precise effector mechanisms are poorly understood. Using the mMCP-6–deficient (but mMCP-7–sufficient) strain, Shin et al showed that MC tryptase contributes to the immune response toward T spiralis,24  and it was demonstrated that mMCP-6 had a role in eosinophil recruitment into infected skeletal muscle. Notably, the absence of mMCP-6 did not adversely affect the rate of parasite clearance, and there was no effect of mMCP-6 deficiency on the IgE response toward T spiralis. Thus, mMCP-6 contributes to the innate immune response toward T spiralis, although not being essential for combating infection.

Bacterial infection

A number of studies have shown that MCs are of primary importance in the defense toward bacterial infection,2  but the mechanisms by which MCs exert this function have not been completely clarified. Given the ability of mMCP-6 to recruit neutrophils,28,29  it may be a candidate for mediating MC-dependent effects during bacterial infection. Indeed, Thakurdas et al reported that mMCP-6 contributes to the defense toward intraperitoneal Klebsiella pneumoniae infection.25  mMCP-6 null animals displayed a significantly reduced survival rate, and it was also shown that the neutrophil recruitment was dependent on mMCP-6. Importantly, the defective neutrophil recruitment was only seen in the early phases of infection, in line with a primary role of MCs in the first-line innate defense toward bacterial insults.

Arthritis

In a hallmark study, Lee et al reported that MC-deficient mice are resistant to arthritis induced by administration of arthritogenic serum from K/BxN mice,30  although it should be emphasized that a role for MCs in experimentally induced arthritis has been questioned.31  A role for MCs in arthritis is also supported by clinical observations.32  In 2 recent studies, the possibility that tryptase could account for the MC-dependent contribution to arthritis has been investigated. In one of these, arthritis was passively induced using methyl-bovine serum albumin/interleukin-1β (IL-1β).33  The results indicated that, indeed, mMCP-6−/− mice developed less severe joint inflammation than did corresponding wild-type (WT) mice. Interestingly, mMCP-7 was shown to compensate for an absence of mMCP-6 and vice versa, indicating that mMCP-6 and -7 show a redundancy in this model.33 

In a more recent study, the same authors assessed the mMCP-6–deficient mouse strains in the K/BxN model for arthritis. It was shown that mMCP-6−/− animals developed lower clinical scores, inflammation, and bone/cartilage erosion than did WT mice.34  Notably, the clinical scores and inflammatory parameters were affected to a relatively moderate extent compared with the effects seen in globally MC-deficient mice, suggesting that other MC-mediated mechanisms contribute to disease.

Together, these studies have clearly implicated MC tryptase in the pathology of arthritis. However, it should be pointed out that both of the models used involve passive induction of disease, whereas the involvement of tryptase in actively induced disease, for example, by immunization with collagen II, remains to be investigated.

To date, knockout strains deficient in mMCP-1, mMCP-4, and mMCP-5 have been generated (Table 3). Importantly, though, because only one MC chymase gene is expressed in humans (CMA1), it is critical to ascertain which of the murine chymases that is the closest homolog of human chymase to extrapolate functions into the human situation. Based on sequence similarity, mMCP-5 is the closest homolog to human chymase. However, an examination of the cleavage specificity of mMCP-5 revealed that it has elastase- rather than chymotrypsin-like substrate specificity.48  Thus, mMCP-5 cannot be considered as a functional homolog to human chymase. mMCP-1 has a tissue distribution different from that of human chymase, has markedly lower affinity for serglycin than does human chymase, and its substrate cleavage specificity is slightly different from that of human chymase.49  In addition, mMCP-2 has a tissue location that is different from that of human chymase; moreover, mMCP-2 has been shown to be virtually without enzymatic activity.50  mMCP-4, on the other hand, has a substrate specificity highly similar to that of human chymase51,52 ; in addition, mMCP-4 and human chymase have similar tissue distribution and serglycin-binding properties. It has also been demonstrated that mMCP-4 accounts for essentially all stored chymotrypsin-like activity in skin and peritoneum.38  Taken together, these facts indicate that, of the murine chymases, mMCP-4 could be considered as the closest functional homolog to human chymase. Therefore, the mMCP-4–deficient strain may be a relevant tool for studies aimed at clarifying the function of human chymase. However, it cannot be excluded that some of the corresponding functions of chymase in humans are executed by murine chymases other than mMCP-4 and that all of the murine chymases thus contribute to the total impact of chymase function. Therefore, to provide a comprehensive insight into the biologic function of MC chymase based on murine studies, it is important to discuss the function of additional murine chymases, that is, also mMCP-1, -2, and -5.

Table 3

In vivo function of MC proteases, based on MC protease-deficient mouse strains

MC protease deficiencyPhenotypeImplicated mechanismReference
mMCP-4 Less severe collagen induced arthritis Effects on anticollagen II IgG generation, inflammation 35  
 Less development of AAAs Effects on inflammation, apoptosis, elastin degradation 36  
 Increased susceptibility to allergic airway inflammation Effects on smooth muscle cells, inflammation 37  
 Tissue accumulation of ECM Defective fibronectin and proMMP-9 processing 38,39  
 Reduced basal intestinal permeability Effects on epithelial cell migration 40  
mMCP-5 Decreased ischemia reperfusion injury Unknown 41  
mMCP-1 Increased susceptibility to T spiralis infection Promotion of capillary permeability; degradation of cell-cell junctions 42,44  
mMCP-6 Less severe passively induced arthritis (K/BxN model) Effect on expression of the neutrophil chemotactic factors CXCL1/KC, CXCL5/LIX, and CXCL8/IL-8; effect on aggrecan degradation 34  
 Nonadequate response to T spiralis infection Effect on eosinophil recruitment 24  
 Increased susceptibility to K pneumoniae infection Effect on neutrophil recruitment and production of inflammatory cytokines 25  
 Less severe passively induced arthritis (methyl-BSA/IL-1β model) Unknown 33  
mMCP-7* Less severe passively induced arthritis (methyl-BSA/IL-1β model) Unknown 33  
MC-CPA Increased susceptibility to snake poison envenomation Degradation of sarafotoxins 45,46  
 Defective granule staining Effects on serglycin proteoglycan solubility 47  
MC protease deficiencyPhenotypeImplicated mechanismReference
mMCP-4 Less severe collagen induced arthritis Effects on anticollagen II IgG generation, inflammation 35  
 Less development of AAAs Effects on inflammation, apoptosis, elastin degradation 36  
 Increased susceptibility to allergic airway inflammation Effects on smooth muscle cells, inflammation 37  
 Tissue accumulation of ECM Defective fibronectin and proMMP-9 processing 38,39  
 Reduced basal intestinal permeability Effects on epithelial cell migration 40  
mMCP-5 Decreased ischemia reperfusion injury Unknown 41  
mMCP-1 Increased susceptibility to T spiralis infection Promotion of capillary permeability; degradation of cell-cell junctions 42,44  
mMCP-6 Less severe passively induced arthritis (K/BxN model) Effect on expression of the neutrophil chemotactic factors CXCL1/KC, CXCL5/LIX, and CXCL8/IL-8; effect on aggrecan degradation 34  
 Nonadequate response to T spiralis infection Effect on eosinophil recruitment 24  
 Increased susceptibility to K pneumoniae infection Effect on neutrophil recruitment and production of inflammatory cytokines 25  
 Less severe passively induced arthritis (methyl-BSA/IL-1β model) Unknown 33  
mMCP-7* Less severe passively induced arthritis (methyl-BSA/IL-1β model) Unknown 33  
MC-CPA Increased susceptibility to snake poison envenomation Degradation of sarafotoxins 45,46  
 Defective granule staining Effects on serglycin proteoglycan solubility 47  

MC indicates mast cell; mMCP, mouse mast cell protease; CPA, carboxypeptidase A; AAA, abdominal aortic aneurysm; ECM, extracellular matrix; and proMMP, pro-matrix metalloprotease.

*

Refers to the spontaneous mMCP-7-deficiency of C57BL/6 mice.26 

Data obtained by shRNA-based technology.

Parasite infection

In the first report of the mMCP-1 knockout, it was shown that the absence of mMCP-1 affected the kinetics of MMC recruitment after Nippostrongylus brasiliensis infection,53  and follow-up studies demonstrated that mMCP-1−/− mice display defective clearance of T spiralis,42  with the latter effect being accompanied by reduced enteropathy and cytokine (tumor necrosis factor) responses.43  These results are thus in concordance with previous studies showing that mice with global MC deficiency are more susceptible to T spiralis infection than are WT mice,54  and suggest that chymase partly accounts for the MC-dependent protection toward T spiralis infection. Although the underlying mechanism has not been fully established, it has been suggested that mMCP-1 may promote inflammation by cleaving cell-cell contact proteins, such as occludin and ZO-1.44,55,56  In line with such a notion, infusion of rats with rMCP-2, that is, a rat homolog of mMCP-1, causes increased epithelial permeability.57 

Bacterial infection

Interesting insight into the function of chymase in bacterial disease came through a study where the role of interleukin-15 (IL-15) in regulating sepsis was studied.58  The authors reported that IL-15−/− mice showed a markedly higher survival rate in sepsis induced by cecal ligation and puncture compared with WT counterparts and that this effect was largely the result of IL-15 expressed in MCs. It was found that a main effect of IL-15 in MCs was to suppress the expression of mMCP-2, but not of other chymases58 ; collectively, these results suggest a role for chymase in regulating bacterial infection.

Arthritis

Because MCs have been implicated in rheumatoid arthritis,30,33  it was of interest to evaluate the contribution of chymase. Indeed, using a model where arthritis is induced actively by immunization with collagen II, mMCP-4−/− mice showed a reduced clinical score and fewer signs of tissue damage compared with WT controls. Moreover, the absence of mMCP-4 resulted in lower levels of anticollagen II IgG in plasma, suggesting that mMCP-4 indeed may contribute to the sensitization step.35  In line with these findings, previous in vitro–based experiments have suggested that purified chymase can enhance the production of IgE and IgG,59  and epidemiologic studies have suggested a connection between a chymase promoter polymorphism and IgE levels in atopic dermatitis.60 

AAA

Animals lacking MCs altogether (C57BL/6-KitW-sh/W-sh) are largely protected in a model for abdominal aortic aneurysm (AAA) formation61 ; and when assessing the mMCP-4−/− mice in this model, the degree of AAA formation was markedly lower in the mMCP-4 null compared with WT mice.36  Strikingly, the extent of protection resulting from mMCP-4 deficiency was similar to that seen in C57BL/6-KitW-sh/W-sh mice, indicating that mMCP-4 is the main effector molecule in MC-mediated promotion of AAA formation.36  A dissection of the underlying mechanisms showed that mMCP-4 contributed to the inflammatory reaction seen in AAA and also that mMCP-4 has a role in inducing apoptosis, angiogenesis, and elastase degradation.36 

Allergic airway inflammation

Because MCs are strongly implicated in asthma and in animal models of allergic airway inflammation, it may be expected that chymase, if anything, may have a pathogenic role. However, in a recent study, it was shown that mMCP-4 indeed protects toward extensive allergic airway inflammation in an ovalbumin-based model and that it protects toward bronchial hyper-reactivity.37  Evidence was also presented suggesting that mMCP-4 may exert this activity by limiting the thickening of the airway smooth muscle cell layer that accompanies allergic airway responses. These data thus indicate that, although the MC as such contributes to the pathology of allergic airway inflammation, one of the individual granule compounds may actually have an activity that counteracts the overall harmful impact of the MCs. Notably, a protective function for MC chymase in allergic airway responses is also supported by clinical observations in which chymase presence has been linked to preserved lung function in asthmatics.62 

Ischemia reperfusion injury

It has been reported that mice lacking mMCP-5 show reduced ischemia reperfusion injury in skeletal muscle.41  Noteworthy, because the mMCP-5−/− animals also lack MC-CPA at the protein level,63  it cannot be ruled out that MC-CPA accounted at least partially for the effects reported. To fully evaluate the individual impact of mMCP-5, it would therefore be valuable to generate a mouse strain in which the active site of mMCP-5 has been mutated, a strategy that presumably would not compromise the storage of MC-CPA.

Tissue homeostasis

In the initial report of the mMCP-4 knockout, evidence was presented suggesting that the lack of chymase causes impaired fibronectin turnover38 ; and in a subsequent study, it was shown that mMCP-4−/− mice show an excessive accumulation of fibronectin in various tissues.39  Intriguingly, these findings were obtained using naive animals, suggesting that MC chymase can contribute to normal tissue homeostasis under conditions not associated with extensive MC degranulation. In line with a homeostatic function of chymase, it was recently shown that the absence of mMCP-4 also leads to a reduced basal intestinal permeability,40  an effect that at least partly could be explained by effects of mMCP-4 on epithelial cell migration.

In all investigated species, only one MC-CPA gene has been identified (Table 1). The tissue location and other properties of human and murine MC-CPA are similar.16  Hence, the interspecies variation that applies for the tryptase and chymase families is less of an issue for MC-CPA, and it is therefore probable that functions identified for murine MC-CPA are mimicked by corresponding functions of the human counterpart.

Two different mouse strains with impaired MC-CPA expression have been reported.45,47  In the original report, a knockout for MC-CPA was generated.47  An important finding was that the absence of MC-CPA resulted in a secondary loss of mMCP-5 protein. This finding, together with the secondary lack of MC-CPA protein in mMCP-5−/− MCs (see “Chymase function”) suggests that mMCP-5 and MC-CPA storage is strongly interdependent. Another finding was that the absence of MC-CPA resulted in altered granule-staining properties, the latter most probably being explained by effects on the storage of serglycin proteoglycan.47  Thus, it appears that MC-CPA has an important role in maintaining normal secretory granule homeostasis, apparently by imposing secondary effects on other compounds stored within the granule. However, because of these secondary effects, it is difficult to discriminate between those mediated by MC-CPA as opposed to those mediated by compounds that are dependent on MC-CPA for their proper storage. To circumvent this issue, a strain with a mutated MC-CPA active site (MC-CPA knock-in) has also been generated.45  This strain does not exhibit any secondary effects on mMCP-5 storage and is therefore a more relevant tool for studies of MC-CPA function.

Using the MC-CPA–deficient strains and using an shRNA-based approach, it has been shown that MC-CPA plays an important role in degradation of certain toxic peptides in vivo. It had previously been shown that MCs are of central importance for clearance of endothelin 1, an endogenous peptide released during bacterial infection, and that MC-dependent endothelin 1 degradation prevents its toxic effects.64  Using the MC-CPA knock-in strain, it was shown that MC-CPA was responsible for this effect.45  This indicates that MC-CPA may have a role in regulating sepsis by preventing excessive accumulation of toxic peptides. It has also been shown, first using an shRNA-based approach46  and subsequently using the MC-CPA knock-in strain,45  that MC-CPA is of central importance for degrading certain snake venom toxins, in this way preventing their lethal effects. Together, these findings suggest an important function for MC-CPA in regulating innate immunologic reactions toward external insults.

Although recent research efforts have implicated the MC proteases in various novel settings, there is still limited knowledge of their true in vivo substrates. Important insight into this critical issue could potentially be obtained by determining their exact cleavage specificities, using unbiased approaches, such as peptide phage display-based techniques.49,51,52,65-67  Sequences that are preferentially cleaved by a given protease can then be used to identify matching sequences in known proteins, inferring that the protein is a preferred substrate for the respective MC protease. However, the MC proteases are relatively promiscuous in terms of cleavage specificity, except for the stringent requirements at the amino acid residue N-terminal of the cleavage site (the P1 position), and it is therefore apparent that each of the MC proteases has the capacity to cleave a multitude of substrates and even can cleave the same substrate at multiple positions. Hence, this type of strategy has only provided partial insight into the identities of their in vivo substrates. To resolve this critical issue, it will therefore be necessary to use alternative strategies, for example, the use of proteomics to identify differentially processed proteins in WT versus MC protease-deficient animals.

Given the broad cleavage specificity of the MC proteases, it is highly probable that a single MC protease may cleave a number of different substrates during one isolated condition, depending on the available repertoires of potential substrates. Moreover, there is also the likelihood that the preferred substrate(s) will vary during different phases of a given pathologic condition. For example, during the early stages of an inflammatory reaction, proteins expressed by resident tissue cells may be preferentially cleaved; whereas, in a later stage of the process, compounds secreted by extravasated inflammatory cells may be the preferred substrates (Figure 2). Clearly, a firm identification of the in vivo substrates for the MC proteases will therefore be a substantial challenge.

Figure 2

Dual roles of MC proteases in the regulation of inflammatory processes. MC proteases may affect inflammation at different levels, either by promoting inflammatory processes (A) or by offering protection (B). (A) Proinflammatory activities include (1) activating cleavages of proinflammatory cytokines/chemokines, (2) degradation of endothelial cell-cell contacts, (3) activation of extracellular matrix (ECM)–degrading enzymes, (4) recruitment of eosinophils/neutrophils, and (5) effects on gene expression (eg, via cleavage of protease-activated receptor 2). (B) Protective activities include (1) degradation of proinflammatory cytokines/chemokines, (2) degradation of toxic peptides, and (3) suppression of smooth muscle cell expansion.

Figure 2

Dual roles of MC proteases in the regulation of inflammatory processes. MC proteases may affect inflammation at different levels, either by promoting inflammatory processes (A) or by offering protection (B). (A) Proinflammatory activities include (1) activating cleavages of proinflammatory cytokines/chemokines, (2) degradation of endothelial cell-cell contacts, (3) activation of extracellular matrix (ECM)–degrading enzymes, (4) recruitment of eosinophils/neutrophils, and (5) effects on gene expression (eg, via cleavage of protease-activated receptor 2). (B) Protective activities include (1) degradation of proinflammatory cytokines/chemokines, (2) degradation of toxic peptides, and (3) suppression of smooth muscle cell expansion.

Close modal

So far, efforts aimed at clarifying the function of MC proteases have predominantly focused on the contribution of individual MC proteases, without evaluating the possibility that the different MC proteases may have cooperative activities. For example, it cannot be excluded that different MC proteases may act on the same substrate but cleave it at distinct sites, potentially leading to a more efficient cleavage/degradation and enhanced biologic impact. To address this issue, it will therefore be important to evaluate mouse strains deficient in multiple MC proteases.

Activation/inactivation of inflammatory compounds

Previous studies have identified a wide array of potential substrates for the various MC proteases, mostly derived from in vitro–based strategies, but also through in vivo experimentation. A survey of identified MC substrates reveals that they fall into distinct categories. Interestingly, a large number of them are proinflammatory compounds; and in many cases, MC protease–catalyzed cleavages are activating, as exemplified by the cleavage of proIL-1β,68  proIL-18,69  CTAP-III,70  CCL6, CCL9, CCL15, and CCL2371  by chymase (Figure 2A). In other cases, MC protease–catalyzed cleavage leads to degradation and destroyed activity, as exemplified by the degradation of CCL3, CCL5,71  IL-6, and IL-1372  by chymase, the degradation of IgE by tryptase,73  and the degradation of endothelin-1 by MC-CPA45  (Figure 2B). Hence, MC proteases may have the ability both to promote and dampen an inflammatory reaction, depending on the availability of cleavable substrates during the given condition. Clearly, because MC proteases are mainly implicated as proinflammatory agents, activating cleavages may be a dominating function. However, as discussed in “Tryptase function” and “Chymase function,” MC proteases have in some cases been shown to have protective/anti-inflammatory functions, and we may thus envisage that such activities can be attributed to degradation of proinflammatory substances.

Effects on ECM

Another plausible scenario is that MC proteases affect the inflammatory process by modifying the extracellular matrix (ECM), either by direct effects on ECM components or indirectly by regulating the activities of ECM-processing enzymes (Figure 2). Direct effects on the ECM include tryptase-catalyzed degradation of type VI collagen,74  denatured collagen (gelatin),75  as well as chymase-catalyzed degradation of vitronectin76  and procollagen.77  Moreover, both tryptase and chymase have been shown to degrade fibronectin38,78 ; and importantly, there is even in vivo evidence supporting that fibronectin is a chymase substrate.39  One notable example of indirect effects on the ECM is the ability of chymase to convert pro-matrix metalloprotease 9 (proMMP-9) into active form, a notion that is supported both by in vitro79  and in vivo39  evidence. It has also been shown that MC proteases can activate yet other MMPs.80-82  Potentially, MC protease–mediated ECM modification could lead to a more efficient influx of inflammatory cells into the tissue and to effects on cellular adhesion to the ECM. It is also noteworthy that degradation of the ECM can unleash proinflammatory cleavage products, such as fibronectin fragments or chemokines/growth factor bound to proteoglycans present in the ECM. Finally, MC proteases may indirectly promote ECM accumulation by acting as mitogens for fibroblasts83,84  (Figure 2).

Effects on cell surface proteins

Another mode by which MC proteases could affect inflammation is to enhance capillary permeability by degrading cell-cell contact components.44,55,56  Further, MC proteases may regulate the local gene expression pattern, as exemplified by the ability of tryptase to induce chemokine secretion.34  It is also well known that tryptase can cleave and thereby activate protease-activated receptor 2,85-87  a notion that is also supported by in vivo evidence.88 

An obvious extension of the findings reviewed here is the possible use of MC protease inhibitors as therapeutic agents. Generally, proteases are attractive targets for inhibition, considering their defined active sites and the relative ease to design low molecular weight, synthetic inhibitors with high selectivity.89  Because the MC proteases are cell-type specific, their inhibition will only affect activities mediated by MCs, with a low risk of side effects because of interference with actions of other cell types. This is in contrast to many other proteases being evaluated as potential drug targets, for example, the MMPs, which are expressed by many cell types. Moreover because the MC proteases under normal/unprovoked conditions are sequestered within the MC granule, MC protease inhibitors may be designed to selectively target MC proteases at the site of a pathologic condition, that is, where MC degranulation has occurred.

Tryptase inhibitors

During last decade, much effort has been invested into the development of MC protease inhibitors. After the implication of tryptase in allergic asthma,90-92  tryptase inhibitors were evaluated in models of asthma and were proved to be effective,93  although a subsequent clinical trial was only moderately successful.94  However, it should be noted that the first-generation tryptase inhibitor used in the clinical trial was rather unselective, of low potency, and extremely slow-acting.95  Hence, the failure of the clinical trial may have been related to these circumstances, and it cannot be excluded that a more favorable outcome may have been reached using the much more efficient and selective tryptase inhibitors that are available today.96-98  Based on the recent findings described in this article, we may also foresee that tryptase inhibitors may be effective for treatment of arthritis. Indeed, because chymase also appears to have a pathogenic role in arthritis, a combination of tryptase and chymase inhibitors may constitute an attractive, future therapeutic regimen.

Chymase inhibitors

A large number of chymase inhibitors, with various potency and selectivity, have recently been developed6,99  and assessed for ability to ameliorate experimentally induced disease. In particular, chymase inhibitors have been assessed in various models for cardiovascular disease, with the background being the strong implication for chymase in angiotensin I conversion in vivo.100  Indeed, chymase inhibitors have in many cases proved to be effective. For example, it has been shown that chymase inhibitors reduce AAA lesions.101  This finding, along with the smaller degree of AAA formation in mMCP-4−/− mice,36  clearly indicates that chymase inhibition may be effective in prevention of aneurysms. Chymase inhibitors are also effective in models for myocardial infarction,102,103  intimal hyperplasia after balloon injury,104  Big endothelin-1 conversion,105  and ventricular remodeling induced by intermittent hypoxia.106  In addition, chymase inhibitors have anti-inflammatory functions, for example, by reducing the eosinophilia during N brasiliensis infection107  and by preventing eosinophil accumulation in a model for atopic dermatitis.108  Together, this development strongly implicates chymase as a promising therapeutic target in variety of clinical settings, and we anticipate that this notion will be evaluated through clinical trials in the near future. However, it is important to use MC protease inhibitors with caution because they may interfere with beneficial functions of MC proteases, for example, in connection with bacterial and parasite infection.

Currently, MC tryptase is frequently used as biomarker both for systemic anaphylaxis and mastocytosis,109  and it has also been suggested that MC-CPA can serve as a biomarker for anaphylaxis.110  Based on the recent implication of MC proteases in additional types of diseases, we anticipate that MC proteases may be evaluated as biomarkers in number of novel settings. For example, it was recently shown that plasma levels of chymase correlated with AAA growth rate, implicating chymase as a potential biomarker for cardiovascular disease.36 

In conclusion, as described in this review article, it is now becoming clear that the MC proteases may account for many of the detrimental and beneficial activities that have been attributed to MCs. However, many important questions remain to be answered. For example, there is still limited knowledge concerning the downstream effects of MC protease action: that is, which substrates are cleaved by the MC proteases in vivo and how do MC protease-catalyzed cleavages of these substrates lead to (or protect from) disease? Certainly, a major future direction will therefore be to address these issues. Another major direction will be to further evaluate the clinical applications of these findings, that is, whether MC proteases can be used as targets for therapy. Clearly, we anticipate that the near future will reveal an even more detailed picture of how the MC proteases contribute to inflammatory disease. We think that this knowledge will enhance our understanding of the inflammatory process and lead to novel therapeutic regimens for inflammatory disease.

This work was supported by the Swedish Research Council, Formas, the Vårdal Foundation, King Gustaf V's 80-year Anniversary Fund, the Swedish Society of Medicine, Åke Wiberg Foundation, Torsten and Ragnar Söderberg Foundation, and the Swedish Cancer Foundation.

Contribution: G.P. conceived the article and wrote it; E.R. and I.W. contributed to the writing and designed figures; and S.W. contributed to the writing.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Gunnar Pejler, Swedish University of Agricultural Sciences, Department of Anatomy, Physiology and Biochemistry, BMC, Box 575, 75123 Uppsala, Sweden; e-mail: Gunnar.Pejler@afb.slu.se.

1
Galli
 
SJ
Grimbaldeston
 
M
Tsai
 
M
Immunomodulatory mast cells: negative, as well as positive, regulators of immunity.
Nat Rev Immunol
2008
, vol. 
8
 
6
(pg. 
478
-
486
)
2
Marshall
 
JS
Mast-cell responses to pathogens.
Nat Rev Immunol
2004
, vol. 
4
 
10
(pg. 
787
-
799
)
3
Grimbaldeston
 
MA
Nakae
 
S
Kalesnikoff
 
J
Tsai
 
M
Galli
 
SJ
Mast cell-derived interleukin 10 limits skin pathology in contact dermatitis and chronic irradiation with ultraviolet B.
Nat Immunol
2007
, vol. 
8
 
10
(pg. 
1095
-
1104
)
4
Lu
 
LF
Lind
 
EF
Gondek
 
DC
, et al. 
Mast cells are essential intermediaries in regulatory T-cell tolerance.
Nature
2006
, vol. 
442
 
7106
(pg. 
997
-
1002
)
5
Metcalfe
 
DD
Baram
 
D
Mekori
 
YA
Mast cells.
Physiol Rev
1997
, vol. 
77
 
4
(pg. 
1033
-
1079
)
6
Pejler
 
G
Åbrink
 
M
Ringvall
 
M
Wernersson
 
S
Mast cell proteases.
Adv Immunol
2007
, vol. 
95
 (pg. 
167
-
255
)
7
Caughey
 
GH
Mast cell tryptases and chymases in inflammation and host defense.
Immunol Rev
2007
, vol. 
217
 (pg. 
141
-
154
)
8
Stevens
 
RL
Adachi
 
R
Protease-proteoglycan complexes of mouse and human mast cells and importance of their beta-tryptase-heparin complexes in inflammation and innate immunity.
Immunol Rev
2007
, vol. 
217
 (pg. 
155
-
167
)
9
Blank
 
U
Rivera
 
J
The ins and outs of IgE-dependent mast-cell exocytosis.
Trends Immunol
2004
, vol. 
25
 
5
(pg. 
266
-
273
)
10
Lützelschwab
 
C
Pejler
 
G
Aveskogh
 
M
Hellman
 
L
Secretory granule proteases in rat mast cells: cloning of 10 different serine proteases and a carboxypeptidase A from various rat mast cell populations.
J Exp Med
1997
, vol. 
185
 
1
(pg. 
13
-
29
)
11
Schwartz
 
LB
Lewis
 
RA
Austen
 
KF
Tryptase from human pulmonary mast cells: purification and characterization.
J Biol Chem
1981
, vol. 
256
 
22
(pg. 
11939
-
11943
)
12
Schwartz
 
LB
Irani
 
AM
Roller
 
K
Castells
 
MC
Schechter
 
NM
Quantitation of histamine, tryptase, and chymase in dispersed human T and TC mast cells.
J Immunol
1987
, vol. 
138
 
8
(pg. 
2611
-
2615
)
13
Wolters
 
PJ
Pham
 
CT
Muilenburg
 
DJ
Ley
 
TJ
Caughey
 
GH
Dipeptidyl peptidase I is essential for activation of mast cell chymases, but not tryptases, in mice.
J Biol Chem
2001
, vol. 
276
 
21
(pg. 
18551
-
18556
)
14
Scholten
 
J
Hartmann
 
K
Gerbaulet
 
A
, et al. 
Mast cell-specific Cre/loxP-mediated recombination in vivo.
Transgenic Res
2008
, vol. 
17
 
2
(pg. 
307
-
315
)
15
Müsch
 
W
Wege
 
AK
Mannel
 
DN
Hehlgans
 
T
Generation and characterization of alpha-chymase-Cre transgenic mice.
Genesis
2008
, vol. 
46
 
3
(pg. 
163
-
166
)
16
Pejler
 
G
Knight
 
SD
Henningsson
 
F
Wernersson
 
S
Novel insights into the biological function of mast cell carboxypeptidase A.
Trends Immunol
2009
, vol. 
30
 
8
(pg. 
401
-
408
)
17
Pereira
 
PJ
Bergner
 
A
Macedo-Ribeiro
 
S
, et al. 
Human beta-tryptase is a ring-like tetramer with active sites facing a central pore.
Nature
1998
, vol. 
392
 
6673
(pg. 
306
-
311
)
18
Hunt
 
JE
Friend
 
DS
Gurish
 
MF
, et al. 
Mouse mast cell protease 9, a novel member of the chromosome 14 family of serine proteases that is selectively expressed in uterine mast cells.
J Biol Chem
1997
, vol. 
272
 
46
(pg. 
29158
-
29166
)
19
Pejler
 
G
Abrink
 
M
Wernersson
 
S
Serglycin proteoglycan: regulating the storage and activities of hematopoietic proteases.
Biofactors
2009
, vol. 
35
 
1
(pg. 
61
-
68
)
20
Åbrink
 
M
Grujic
 
M
Pejler
 
G
Serglycin is essential for maturation of mast cell secretory granule.
J Biol Chem
2004
, vol. 
279
 
39
(pg. 
40897
-
40905
)
21
Forsberg
 
E
Pejler
 
G
Ringvall
 
M
, et al. 
Abnormal mast cells in mice deficient in a heparin-synthesizing enzyme.
Nature
1999
, vol. 
400
 
6746
(pg. 
773
-
776
)
22
Humphries
 
DE
Wong
 
GW
Friend
 
DS
, et al. 
Heparin is essential for the storage of specific granule proteases in mast cells [see comments].
Nature
1999
, vol. 
400
 
6746
(pg. 
769
-
772
)
23
Braga
 
T
Grujic
 
M
Lukinius
 
A
, et al. 
Serglycin proteoglycan is required for secretory granule integrity in mucosal mast cells.
Biochem J
2007
, vol. 
403
 
1
(pg. 
49
-
57
)
24
Shin
 
K
Watts
 
GF
Oettgen
 
HC
, et al. 
Mouse mast cell tryptase mMCP-6 is a critical link between adaptive and innate immunity in the chronic phase of Trichinella spiralis infection.
J Immunol
2008
, vol. 
180
 
7
(pg. 
4885
-
4891
)
25
Thakurdas
 
SM
Melicoff
 
E
Sansores-Garcia
 
L
, et al. 
The mast cell-restricted tryptase mMCP-6 has a critical immunoprotective role in bacterial infections.
J Biol Chem
2007
, vol. 
282
 
29
(pg. 
20809
-
20815
)
26
Hunt
 
JE
Stevens
 
RL
Austen
 
KF
, et al. 
Natural disruption of the mouse mast cell protease 7 gene in the C57BL/6 mouse.
J Biol Chem
1996
, vol. 
271
 
5
(pg. 
2851
-
2855
)
27
Dawicki
 
W
Marshall
 
JS
New and emerging roles for mast cells in host defence.
Curr Opin Immunol
2007
, vol. 
19
 
1
(pg. 
31
-
38
)
28
Hallgren
 
J
Karlson
 
U
Poorafshar
 
M
Hellman
 
L
Pejler
 
G
Mechanism for activation of mouse mast cell tryptase: dependence on heparin and acidic pH for formation of active tetramers of mouse mast cell protease 6.
Biochemistry
2000
, vol. 
39
 
42
(pg. 
13068
-
13077
)
29
Huang
 
C
Friend
 
DS
Qiu
 
WT
, et al. 
Induction of a selective and persistent extravasation of neutrophils into the peritoneal cavity by tryptase mouse mast cell protease 6.
J Immunol
1998
, vol. 
160
 
4
(pg. 
1910
-
1919
)
30
Lee
 
DM
Friend
 
DS
Gurish
 
MF
, et al. 
Mast cells: a cellular link between autoantibodies and inflammatory arthritis.
Science
2002
, vol. 
297
 
5587
(pg. 
1689
-
1692
)
31
Zhou
 
JS
Xing
 
W
Friend
 
DS
Austen
 
KF
Katz
 
HR
Mast cell deficiency in Kit(W-sh) mice does not impair antibody-mediated arthritis.
J Exp Med
2007
, vol. 
204
 
12
(pg. 
2797
-
2802
)
32
Maruotti
 
N
Crivellato
 
E
Cantatore
 
FP
Vacca
 
A
Ribatti
 
D
Mast cells in rheumatoid arthritis.
Clin Rheumatol
2007
, vol. 
26
 
1
(pg. 
1
-
4
)
33
McNeil
 
HP
Shin
 
K
Campbell
 
IK
, et al. 
The mouse mast cell-restricted tetramer-forming tryptases mouse mast cell protease 6 and mouse mast cell protease 7 are critical mediators in inflammatory arthritis.
Arthritis Rheum
2008
, vol. 
58
 
8
(pg. 
2338
-
2346
)
34
Shin
 
K
Nigrovic
 
PA
Crish
 
J
, et al. 
Mast cells contribute to autoimmune inflammatory arthritis via their tryptase/heparin complexes.
J Immunol
2009
, vol. 
182
 
1
(pg. 
647
-
656
)
35
Magnusson
 
SE
Pejler
 
G
Kleinau
 
S
Abrink
 
M
Mast cell chymase contributes to the antibody response and the severity of autoimmune arthritis.
FASEB J
2009
, vol. 
23
 
3
(pg. 
875
-
882
)
36
Sun
 
J
Zhang
 
J
Lindholt
 
JS
, et al. 
Critical role of mast cell chymase in mouse abdominal aortic aneurysm formation.
Circulation
2009
, vol. 
120
 
11
(pg. 
973
-
982
)
37
Waern
 
I
Jonasson
 
S
Hjoberg
 
J
, et al. 
Mouse mast cell protease 4 is the major chymase in murine airways and has a protective role in allergic airway inflammation.
J Immunol
2009
, vol. 
183
 
10
(pg. 
6369
-
6376
)
38
Tchougounova
 
E
Pejler
 
G
Abrink
 
M
The chymase, mouse mast cell protease 4, constitutes the major chymotrypsin-like activity in peritoneum and ear tissue: a role for mouse mast cell protease 4 in thrombin regulation and fibronectin turnover.
J Exp Med
2003
, vol. 
198
 
3
(pg. 
423
-
431
)
39
Tchougounova
 
E
Lundequist
 
A
Fajardo
 
I
, et al. 
A key role for mast cell chymase in the activation of pro-matrix metalloprotease-9 and pro-matrix metalloprotease-2.
J Biol Chem
2005
, vol. 
280
 
10
(pg. 
9291
-
9296
)
40
Groschwitz
 
KR
Ahrens
 
R
Osterfeld
 
H
, et al. 
Mast cells regulate homeostatic intestinal epithelial migration and barrier function by a chymase/Mcpt4-dependent mechanism.
Proc Natl Acad Sci U S A
2009
, vol. 
106
 
52
(pg. 
22381
-
22386
)
41
Abonia
 
JP
Friend
 
DS
Austen
 
WG
, et al. 
Mast cell protease 5 mediates ischemia-reperfusion injury of mouse skeletal muscle.
J Immunol
2005
, vol. 
174
 
11
(pg. 
7285
-
7291
)
42
Knight
 
PA
Wright
 
SH
Lawrence
 
CE
Paterson
 
YY
Miller
 
HR
Delayed expulsion of the nematode Trichinella spiralis in mice lacking the mucosal mast cell-specific granule chymase, mouse mast cell protease-1.
J Exp Med
2000
, vol. 
192
 
12
(pg. 
1849
-
1856
)
43
Lawrence
 
CE
Paterson
 
YY
Wright
 
SH
Knight
 
PA
Miller
 
HR
Mouse mast cell protease-1 is required for the enteropathy induced by gastrointestinal helminth infection in the mouse.
Gastroenterology
2004
, vol. 
127
 
1
(pg. 
155
-
165
)
44
McDermott
 
JR
Bartram
 
RE
Knight
 
PA
, et al. 
Mast cells disrupt epithelial barrier function during enteric nematode infection.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 
13
(pg. 
7761
-
7766
)
45
Schneider
 
LA
Schlenner
 
SM
Feyerabend
 
TB
Wunderlin
 
M
Rodewald
 
HR
Molecular mechanism of mast cell mediated innate defense against endothelin and snake venom sarafotoxin.
J Exp Med
2007
, vol. 
204
 
11
(pg. 
2629
-
2639
)
46
Metz
 
M
Piliponsky
 
AM
Chen
 
CC
, et al. 
Mast cells can enhance resistance to snake and honeybee venoms.
Science
2006
, vol. 
313
 
5786
(pg. 
526
-
530
)
47
Feyerabend
 
TB
Hausser
 
H
Tietz
 
A
, et al. 
Loss of histochemical identity in mast cells lacking carboxypeptidase A.
Mol Cell Biol
2005
, vol. 
25
 
14
(pg. 
6199
-
6210
)
48
Kunori
 
Y
Koizumi
 
M
Masegi
 
T
, et al. 
Rodent alpha-chymases are elastase-like proteases.
Eur J Biochem
2002
, vol. 
269
 
23
(pg. 
5921
-
5930
)
49
Andersson
 
MK
Pemberton
 
AD
Miller
 
HR
Hellman
 
L
Extended cleavage specificity of mMCP-1, the major mucosal mast cell protease in mouse-high specificity indicates high substrate selectivity.
Mol Immunol
2008
, vol. 
45
 
9
(pg. 
2548
-
2558
)
50
Pemberton
 
AD
Brown
 
JK
Wright
 
SH
, et al. 
Purification and characterization of mouse mast cell proteinase-2 and the differential expression and release of mouse mast cell proteinase-1 and -2 in vivo.
Clin Exp Allergy
2003
, vol. 
33
 
7
(pg. 
1005
-
1012
)
51
Andersson
 
MK
Karlson
 
U
Hellman
 
L
The extended cleavage specificity of the rodent beta-chymases rMCP-1 and mMCP-4 reveal major functional similarities to the human mast cell chymase.
Mol Immunol
2008
, vol. 
45
 
3
(pg. 
766
-
775
)
52
Andersson
 
MK
Enoksson
 
M
Gallwitz
 
M
Hellman
 
L
The extended substrate specificity of the human mast cell chymase reveals a serine protease with well-defined substrate recognition profile.
Int Immunol
2009
, vol. 
21
 
1
(pg. 
95
-
104
)
53
Wastling
 
JM
Knight
 
P
Ure
 
J
, et al. 
Histochemical and ultrastructural modification of mucosal mast cell granules in parasitized mice lacking the beta-chymase, mouse mast cell protease-1.
Am J Pathol
1998
, vol. 
153
 
2
(pg. 
491
-
504
)
54
Ha
 
TY
Reed
 
ND
Crowle
 
PK
Delayed expulsion of adult Trichinella spiralis by mast cell-deficient W/Wv mice.
Infect Immun
1983
, vol. 
41
 
1
(pg. 
445
-
447
)
55
Scudamore
 
CL
Jepson
 
MA
Hirst
 
BH
Miller
 
HR
The rat mucosal mast cell chymase, RMCP-II, alters epithelial cell monolayer permeability in association with altered distribution of the tight junction proteins ZO-1 and occludin.
Eur J Cell Biol
1998
, vol. 
75
 
4
(pg. 
321
-
330
)
56
Ebihara
 
N
Funaki
 
T
Murakami
 
A
Takai
 
S
Miyazaki
 
M
Mast cell chymase decreases the barrier function and inhibits the migration of corneal epithelial cells.
Curr Eye Res
2005
, vol. 
30
 
12
(pg. 
1061
-
1069
)
57
Scudamore
 
CL
Thornton
 
EM
McMillan
 
L
Newlands
 
GF
Miller
 
HR
Release of the mucosal mast cell granule chymase, rat mast cell protease-II, during anaphylaxis is associated with the rapid development of paracellular permeability to macromolecules in rat jejunum.
J Exp Med
1995
, vol. 
182
 
6
(pg. 
1871
-
1881
)
58
Orinska
 
Z
Maurer
 
M
Mirghomizadeh
 
F
, et al. 
IL-15 constrains mast cell-dependent antibacterial defenses by suppressing chymase activities.
Nat Med
2007
, vol. 
13
 
8
(pg. 
927
-
934
)
59
Yoshikawa
 
T
Imada
 
T
Nakakubo
 
H
Nakamura
 
N
Naito
 
K
Rat mast cell protease-I enhances immunoglobulin E production by mouse B cells stimulated with interleukin-4.
Immunology
2001
, vol. 
104
 
3
(pg. 
333
-
340
)
60
Iwanaga
 
T
McEuen
 
A
Walls
 
AF
, et al. 
Polymorphism of the mast cell chymase gene (CMA1) promoter region: lack of association with asthma but association with serum total immunoglobulin E levels in adult atopic dermatitis.
Clin Exp Allergy
2004
, vol. 
34
 
7
(pg. 
1037
-
1042
)
61
Sun
 
J
Sukhova
 
GK
Yang
 
M
, et al. 
Mast cells modulate the pathogenesis of elastase-induced abdominal aortic aneurysms in mice.
J Clin Invest
2007
, vol. 
117
 
11
(pg. 
3359
-
3368
)
62
Balzar
 
S
Chu
 
HW
Strand
 
M
Wenzel
 
S
Relationship of small airway chymase-positive mast cells and lung function in severe asthma.
Am J Respir Crit Care Med
2005
, vol. 
171
 
5
(pg. 
431
-
439
)
63
Stevens
 
RL
Qui
 
D
McNeil
 
HP
, et al. 
Transgenic mice that possess a disrupted mast cell protease 5 (mMCP-5) gene can not store carboxypeptidase A (mMC-CPA) protein in their granules.
FASEB J
1996
pg. 
1017772
 
64
Maurer
 
M
Wedemeyer
 
J
Metz
 
M
, et al. 
Mast cells promote homeostasis by limiting endothelin-1-induced toxicity.
Nature
2004
, vol. 
432
 
7016
(pg. 
512
-
516
)
65
Harris
 
JL
Niles
 
A
Burdick
 
K
, et al. 
Definition of the extended substrate specificity determinants for beta- tryptases I and II.
J Biol Chem
2001
, vol. 
276
 
37
(pg. 
34941
-
34947
)
66
Huang
 
C
Wong
 
GW
Ghildyal
 
N
, et al. 
The tryptase, mouse mast cell protease 7, exhibits anticoagulant activity in vivo and in vitro due to its ability to degrade fibrinogen in the presence of the diverse array of protease inhibitors in plasma.
J Biol Chem
1997
, vol. 
272
 
50
(pg. 
31885
-
31893
)
67
Karlson
 
U
Pejler
 
G
Tomasini-Johansson
 
B
Hellman
 
L
Extended substrate specificity of rat mast cell protease 5, a rodent alpha-chymase with elastase-like primary specificity.
J Biol Chem
2003
, vol. 
278
 
41
(pg. 
39625
-
39631
)
68
Mizutani
 
H
Schechter
 
N
Lazarus
 
G
Black
 
RA
Kupper
 
TS
Rapid and specific conversion of precursor interleukin 1 beta (IL-1 beta) to an active IL-1 species by human mast cell chymase.
J Exp Med
1991
, vol. 
174
 
4
(pg. 
821
-
825
)
69
Omoto
 
Y
Tokime
 
K
Yamanaka
 
K
, et al. 
Human mast cell chymase cleaves pro-IL-18 and generates a novel and biologically active IL-18 fragment.
J Immunol
2006
, vol. 
177
 
12
(pg. 
8315
-
8319
)
70
Schiemann
 
F
Grimm
 
TA
Hoch
 
J
, et al. 
Mast cells and neutrophils proteolytically activate chemokine precursor CTAP-III and are subject to counterregulation by PF-4 through inhibition of chymase and cathepsin G.
Blood
2006
, vol. 
107
 
6
(pg. 
2234
-
2242
)
71
Berahovich
 
RD
Miao
 
Z
Wang
 
Y
, et al. 
Proteolytic activation of alternative CCR1 ligands in inflammation.
J Immunol
2005
, vol. 
174
 
11
(pg. 
7341
-
7351
)
72
Zhao
 
W
Oskeritzian
 
CA
Pozez
 
AL
Schwartz
 
LB
Cytokine production by skin-derived mast cells: endogenous proteases are responsible for degradation of cytokines.
J Immunol
2005
, vol. 
175
 
4
(pg. 
2635
-
2642
)
73
Rauter
 
I
Krauth
 
MT
Westritschnig
 
K
, et al. 
Mast cell-derived proteases control allergic inflammation through cleavage of IgE.
J Allergy Clin Immunol
2008
, vol. 
121
 
1
(pg. 
197
-
202
)
74
Kielty
 
CM
Lees
 
M
Shuttleworth
 
CA
Woolley
 
D
Catabolism of intact type VI collagen microfibrils: susceptibility to degradation by serine proteinases.
Biochem Biophys Res Commun
1993
, vol. 
191
 
3
(pg. 
1230
-
1236
)
75
Fajardo
 
I
Pejler
 
G
Human mast cell beta-tryptase is a gelatinase.
J Immunol
2003
, vol. 
171
 
3
(pg. 
1493
-
1499
)
76
Banovac
 
K
Banovac
 
F
Yang
 
J
Koren
 
E
Interaction of osteoblasts with extracellular matrix: effect of mast cell chymase.
Proc Soc Exp Biol Med
1993
, vol. 
203
 
2
(pg. 
221
-
235
)
77
Kofford
 
MW
Schwartz
 
LB
Schechter
 
NM
, et al. 
Cleavage of type I procollagen by human mast cell chymase initiates collagen fibril formation and generates a unique carboxyl-terminal propeptide.
J Biol Chem
1997
, vol. 
272
 
11
(pg. 
7127
-
7131
)
78
Vartio
 
T
Seppa
 
H
Vaheri
 
A
Susceptibility of soluble and matrix fibronectins to degradation by tissue proteinases, mast cell chymase and cathepsin G.
J Biol Chem
1981
, vol. 
256
 
1
(pg. 
471
-
477
)
79
Fang
 
KC
Raymond
 
WW
Lazarus
 
SC
Caughey
 
GH
Dog mastocytoma cells secrete a 92-kD gelatinase activated extracellularly by mast cell chymase.
J Clin Invest
1996
, vol. 
97
 
7
(pg. 
1589
-
1596
)
80
Saarinen
 
J
Kalkkinen
 
N
Welgus
 
HG
Kovanen
 
PT
Activation of human interstitial procollagenase through direct cleavage of the Leu83-Thr84 bond by mast cell chymase.
J Biol Chem
1994
, vol. 
269
 
27
(pg. 
18134
-
18140
)
81
Lees
 
M
Taylor
 
DJ
Woolley
 
DE
Mast cell proteinases activate precursor forms of collagenase and stromelysin, but not of gelatinases A and B.
Eur J Biochem
1994
, vol. 
223
 
1
(pg. 
171
-
177
)
82
Margulis
 
A
Nocka
 
KH
Brennan
 
AM
, et al. 
Mast cell-dependent contraction of human airway smooth muscle cell-containing collagen gels: influence of cytokines, matrix metalloproteases, and serine proteases.
J Immunol
2009
, vol. 
183
 
3
(pg. 
1739
-
1750
)
83
Algermissen
 
B
Hermes
 
B
Feldmann-Boeddeker
 
I
Bauer
 
F
Henz
 
BM
Mast cell chymase and tryptase during tissue turnover: analysis on in vitro mitogenesis of fibroblasts and keratinocytes and alterations in cutaneous scars.
Exp Dermatol
1999
, vol. 
8
 
3
(pg. 
193
-
198
)
84
Ruoss
 
SJ
Hartmann
 
T
Caughey
 
GH
Mast cell tryptase is a mitogen for cultured fibroblasts.
J Clin Invest
1991
, vol. 
88
 
2
(pg. 
493
-
499
)
85
Corvera
 
CU
Dery
 
O
McConalogue
 
K
, et al. 
Mast cell tryptase regulates rat colonic myocytes through proteinase-activated receptor 2.
J Clin Invest
1997
, vol. 
100
 
6
(pg. 
1383
-
1393
)
86
Mirza
 
H
Schmidt
 
VA
Derian
 
CK
Jesty
 
J
Bahou
 
WF
Mitogenic responses mediated through the proteinase-activated receptor-2 are induced by expressed forms of mast cell alpha- or beta-tryptases.
Blood
1997
, vol. 
90
 
10
(pg. 
3914
-
3922
)
87
Molino
 
M
Barnathan
 
ES
Numerof
 
R
, et al. 
Interactions of mast cell tryptase with thrombin receptors and PAR-2.
J Biol Chem
1997
, vol. 
272
 
7
(pg. 
4043
-
4049
)
88
Kelso
 
EB
Lockhart
 
JC
Hembrough
 
T
, et al. 
Therapeutic promise of Par2 antagonism in joint inflammation.
J Pharmacol Exp Ther
2006
, vol. 
316
 
3
(pg. 
1017
-
1024
)
89
Turk
 
B
Targeting proteases: successes, failures and future prospects.
Nat Rev Drug Discov
2006
, vol. 
5
 
9
(pg. 
785
-
799
)
90
Molinari
 
JF
Scuri
 
M
Moore
 
WR
, et al. 
Inhaled tryptase causes bronchoconstriction in sheep via histamine release.
Am J Respir Crit Care Med
1996
, vol. 
154
 
3
(pg. 
649
-
653
)
91
Barrios
 
VE
Middleton
 
SC
Kashem
 
MA
, et al. 
Tryptase mediates hyperresponsiveness in isolated guinea pig bronchi.
Life Sci
1998
, vol. 
63
 
26
(pg. 
2295
-
2303
)
92
Berger
 
P
Compton
 
SJ
Molimard
 
M
, et al. 
Mast cell tryptase as a mediator of hyperresponsiveness in human isolated bronchi.
Clin Exp Allergy
1999
, vol. 
29
 
6
(pg. 
804
-
812
)
93
Clark
 
JM
Abraham
 
WM
Fishman
 
CE
, et al. 
Tryptase inhibitors block allergen-induced airway and inflammatory responses in allergic sheep.
Am J Respir Crit Care Med
1995
, vol. 
152
 
6
(pg. 
2076
-
2083
)
94
Krishna
 
MT
Chauhan
 
A
Little
 
L
, et al. 
Inhibition of mast cell tryptase by inhaled APC 366 attenuates allergen-induced late-phase airway obstruction in asthma.
J Allergy Clin Immunol
2001
, vol. 
107
 
6
(pg. 
1039
-
1045
)
95
Rice
 
KD
Tanaka
 
RD
Katz
 
BA
Numerof
 
RP
Moore
 
WR
Inhibitors of tryptase for the treatment of mast cell-mediated diseases.
Curr Pharm Des
1998
, vol. 
4
 
5
(pg. 
381
-
396
)
96
Oh
 
SW
Pae
 
CI
Lee
 
DK
, et al. 
Tryptase inhibition blocks airway inflammation in a mouse asthma model.
J Immunol
2002
, vol. 
168
 
4
(pg. 
1992
-
2000
)
97
Rice
 
KD
Wang
 
VR
Gangloff
 
AR
, et al. 
Dibasic inhibitors of human mast cell tryptase: 2. Structure-activity relationships and requirements for potent activity.
Bioorg Med Chem Lett
2000
, vol. 
10
 
20
(pg. 
2361
-
2366
)
98
Wright
 
CD
Havill
 
AM
Middleton
 
SC
, et al. 
Inhibition of allergen-induced pulmonary responses by the selective tryptase inhibitor 1,5-bis-[4-[(3-carbamimidoyl-benzenesulfonylamino)-methyl]-phenoxy]-pen tane (AMG-126737).
Biochem Pharmacol
1999
, vol. 
58
 
12
(pg. 
1989
-
1996
)
99
Takai
 
S
Jin
 
D
Muramatsu
 
M
Okamoto
 
Y
Miyazaki
 
M
Therapeutic applications of chymase inhibitors in cardiovascular diseases and fibrosis.
Eur J Pharmacol
2004
, vol. 
501
 
1
(pg. 
1
-
8
)
100
Dell'Italia
 
LJ
Husain
 
A
Dissecting the role of chymase in angiotensin II formation and heart and blood vessel diseases.
Curr Opin Cardiol
2002
, vol. 
17
 
4
(pg. 
374
-
379
)
101
Tsunemi
 
K
Takai
 
S
Nishimoto
 
M
, et al. 
A specific chymase inhibitor, 2-(5-formylamino-6-oxo-2-phenyl-1,6-dihydropyrimidine-1-yl)-N-[[3,4-dioxo-1-phenyl-7-(2-pyridyloxy)]-2-heptyl]acetamide (NK3201), suppresses development of abdominal aortic aneurysm in hamsters.
J Pharmacol Exp Ther
2004
, vol. 
309
 
3
(pg. 
879
-
883
)
102
Hoshino
 
F
Urata
 
H
Inoue
 
Y
, et al. 
Chymase inhibitor improves survival in hamsters with myocardial infarction.
J Cardiovasc Pharmacol
2003
, vol. 
41
 
suppl 1
(pg. 
S11
-
S18
)
103
Jin
 
D
Takai
 
S
Yamada
 
M
, et al. 
Impact of chymase inhibitor on cardiac function and survival after myocardial infarction.
Cardiovasc Res
2003
, vol. 
60
 
2
(pg. 
413
-
420
)
104
Takai
 
S
Sakonjo
 
H
Fukuda
 
K
, et al. 
A novel chymase inhibitor, 2-(5-formylamino-6-oxo-2-phenyl-1,6-dihydropyrimidine-1-yl)-N-[[,4-dioxo-1-phenyl-7-(2-pyridyloxy)]2-heptyl]acetamide (NK3201), suppressed intimal hyperplasia after balloon injury.
J Pharmacol Exp Ther
2003
, vol. 
304
 
2
(pg. 
841
-
844
)
105
Simard
 
E
Jin
 
D
Takai
 
S
, et al. 
Chymase-dependent conversion of Big endothelin-1 in the mouse in vivo.
J Pharmacol Exp Ther
2009
, vol. 
328
 
2
(pg. 
540
-
548
)
106
Matsumoto
 
C
Hayashi
 
T
Kitada
 
K
, et al. 
Chymase plays an important role in left ventricular remodeling induced by intermittent hypoxia in mice.
Hypertension
2009
, vol. 
54
 
1
(pg. 
164
-
171
)
107
Watanabe
 
N
Miura
 
K
Fukuda
 
Y
Chymase inhibitor ameliorates eosinophilia in mice infected with Nippostrongylus brasiliensis.
Int Arch Allergy Immunol
2002
, vol. 
128
 
3
(pg. 
235
-
239
)
108
Watanabe
 
N
Tomimori
 
Y
Saito
 
K
, et al. 
Chymase inhibitor improves dermatitis in NC/Nga mice.
Int Arch Allergy Immunol
2002
, vol. 
128
 
3
(pg. 
229
-
234
)
109
Schwartz
 
LB
Diagnostic value of tryptase in anaphylaxis and mastocytosis.
Immunol Allergy Clin North Am
2006
, vol. 
26
 
3
(pg. 
451
-
463
)
110
Simons
 
FE
Frew
 
AJ
Ansotegui
 
IJ
, et al. 
Risk assessment in anaphylaxis: current and future approaches.
J Allergy Clin Immunol
2007
, vol. 
120
 
suppl
(pg. 
S2
-
S24
)

Author notes

*

E.R. and I.W. contributed equally to this study.

Sign in via your Institution