Immunology of Ocular Allergy Manfred Zierhut, Tilo Biedermann, Santa Jeremy Ono
INDEX
A
Allergic asthma 107
mechanisms of immune privilege and ACAID 108
tolerogenic APC therapy 109
Allergic conjunctivitis 123, 156
Allergic contact eczema 116
cosmetics 119
drug vehicles 119
metals 119
pharmaceutical products 117
plant materials 120
preservative agents 119
rubber additives 119
Allergy hypersensitivity 153
type I (immediate) hypersensitivity 129, 153
type IV (delayed-type) hypersensitivity 137, 156
Angioedema 115
Angiogenesis 82
Anterior chamber associated immune deviation (ACAID) 108
Anti-IgE 31, 39, 60, 83, 84, 203, 205, 206, 215
Antigen-presenting cells 43
Antihistamines 192, 195
effects on conjunctival tissues 193
new antihistamines 197
receptors 194
Atopic aczema 120
Atopic dermatitis 138
Atopic keratoconjunctivitis 125, 160, 179
corneal ulcer 183
B
B-cells 38
maturation and differentiation 38
Immunoglobulin class switch 36
Basophils 77, 82
involvement in allergic eye disorders 81
mediators 78
membrane receptors 80
morphologic and ultrastructural characteristics 77
pharmacologic modulation 83
adenylate cyclase activators 84
blockade of activating receptors 83
glucocorticoids 84
immunophilin ligands 84
inhibition of signal transduction 83
phosphodiesterase inhibitors 84
receptor antagonists 85
role for allegic angiogenesis 82
surface markers 79
C
Chemosis 180
Cicatricial conjunctivitis 182
Conjunctival allergen challenge (CAC) 168, 171, 174, 196
Contact dermatitis or eczema 120, 138
Contact urticaria 116
Corneal allograft rejection 184
adoptive transfer of graft rejection 189
atopy as risk factors 184
TH2-mediated airway hyperreactivity 186
Cortisone 202
D
Dendritic cells (DC) 30, 43, 45, 130, 163, 164, 222
delayed type hypersensitivity (DTH) 184
differentiation 43, 45
T-cells 43
E
Environment and allergy 95
adjuvant effects of air pollutants 98
air pollutants 99
causative factors in development 95
genetic and environmental determinants 96
influence of air pollutants in allergic bioavailability 96
tobacco smoke 99100
Eosinophils 48
activation 57
degranulation 58
mediators 49
chemokines 53
cytokines 53
granule proteins 49
lipid mediators 53
protease and other enzymes 52
recruitment in allergic diseases 55
response to innate immunological stimuli 61
receptor 55
Eosinophil cationic protein (ECP) 49, 54, 55, 203
G
Giant papillary conjunctivitis 162
Granule protease proteoglycan complex 2
Granule proteins 49
H
Histamine 1, 6, 810, 2729, 49, 50, 64, 7780, 82, 84, 85, 96, 99, 100, 103, 130, 131, 146, 154, 163, 164, 168, 174, 192199, 202206, 215
Hypersensitivity vasculitis 136
I
ICAM 45, 55, 58, 168, 172, 174, 193, 203206
Immunoglobulin E (IgE) 20
diseases related to 130
inert nature of IgE binding 23
monomeric IgE as an effector molecule 25, 48
effect of monomeric IgE 27
mast cell survival 25, 27, 28
pathophysiology 20
relevance of monomeric IgE 30
receptor 1, 20
requirement of aggregation 23
specific IgE 173
structure 21, 22
Immunomodutation 220
aspirin 225, 231
inhibiting cyclo-oxygenase 168, 223
NSAID targets 227
integrin 29, 55, 58, 61, 64, 136
intercellular adhesion molecule (ICAM) 168
Interferon (IFN) 37, 40, 44, 5658, 79, 99, 107, 137, 139, 156, 172, 184186, 188, 203, 206, 213, 216, 231, 233, 234
alpha 21, 57, 205, 206
gamma 40, 172
Interleukin (IL) 48
IL-1 57, 79, 174, 193, 206
IL-2 54, 58, 137, 161, 164, 170, 171, 184, 225, 231234
IL-3 4, 27, 28, 48, 54, 57, 58, 77, 79, 91, 161
IL-4 29, 36, 4446, 5457, 79, 91, 84, 85, 96, 99, 100, 107, 110, 161, 168174, 184, 212, 213, 222, 223, 233, 234
IL-5 8, 49, 51, 5458, 60, 64, 79, 81, 99, 107, 161, 170, 171, 174, 184, 231
IL-6 44, 45, 54, 55, 170, 171, 174, 193, 196, 225
IL-8 10, 37, 49, 50, 54, 64, 79, 82, 99, 168, 170172, 174, 193, 196, 206, 225
IL-9 44, 57
IL-10 8, 37, 44, 54, 55, 171, 174, 213
IL-12 37, 54, 137, 169171, 220, 223, 232, 234
IL-13 8, 29, 44, 5557, 79, 81, 85, 96, 99, 107, 154, 170174, 184, 223, 233
IL-16 54
IL-17 44
IL-22 44
L
Lid swelling 180
Limbal mass 180
Long-lived plasma cells 37, 38
Long-term modulation of disease 45
M
Major basic protein (MBP) 49, 54, 110
Mast cell stabilizers 202
Mast cells (MC) 1, 143
activated 6
degranulation 78
development 4
functions 144
immune responses to non-bacterial pathogens 145
induction of innate immune responses to bacterial infections 144
granule protease proteoglycan complex 2
histochemistry 2
immune responses to pathogens 147
innate immune system 130
morphology 2
proteases (MCP) 2
react to environmental threats 145
receptors 6
skin mast cells 143
tryptase mediators 9
N
neutrophil 2, 5, 7, 9, 10, 48, 50, 51, 53, 5961, 67, 68, 97, 101, 130, 135, 136, 143, 145, 147, 155, 156, 162, 168, 172, 174, 204, 206
O
Ocular allergy 178
differential diagnosis 179
chemosis 180
cicatricial conjunctivitis 182
conjunctival congestion 181
corneal ulcer 183
lid swelling 180
limbal mass 180
pannus 181
proteases 173
pseudomembranous conjunctivitis 181
Reaction in ocular allergy 167
early-and late-phase 167
TH1-and TH2-type cytokines 169
epidemiology management 163
Omalizumab (see also “anti-IgE”) 39
P
Pannus 181
Piecemeal degranulation 78
Plasma cells
Pseudomembranous conjunctivitis 181
Q
Quincke's edema 115
S
Systemic immune therapy for allergic disorders 212
long-term effects 216
mechanism of action 206
new indications 216
safety concern 214
T
Tacrolimus 84
Th1 40, 4446, 56, 57, 79, 103, 107111, 129, 130, 137, 139, 147, 156, 169, 170, 175, 184, 185, 188, 189, 195, 212, 213, 216, 217, 231234
Th2 40, 4446, 51, 5557, 61, 63, 77, 7981, 85, 86, 96, 99101, 107111, 123, 129, 130, 138, 139, 154, 156, 160, 161, 168170, 174, 175, 184189, 195, 204, 212, 213, 216, 223, 224, 231234
Th17 4446, 86
T regulatory cells 4446, 107, 109, 110, 213, 214, 221, 222, 224
thrombocytopenic purpura 133
TNF-α 8, 10, 54, 55, 57, 58, 79, 99, 168, 170174, 205, 206, 225, 231
Tryptase 24, 6, 911, 79, 82, 85, 96, 133, 144, 154, 168, 203205, 215
Toll-like receptor 8, 39, 44, 51
U
Urticaria 132
V
vascular cell adhesion molecule (VCAM) 55, 168, 174
Vernal keratoconjunctivitis 124, 158, 178
Very late antigen (VLA) 29, 55, 61, 168
Vitamin D 40
×
Chapter Notes

Save Clear


Mast Cells and Innate Immunity1

Matthew J Hamilton,
Richard L Stevens
 
INTRODUCTION
Mast cells (MCs) are immune cells that have prominent adverse roles in allergic inflammation and systemic anaphylaxis. Cross-linking of the high-affinity IgE receptors (Fc∊RI) on the MC's plasma membrane results in the rapid release of granule mediators (e.g., histamine and protease•proteoglycan complexes) and lipid mediators (e.g., leukotriene B4, leukotriene C4, prostaglandin D2, platelet-activating factor, and thromboxanes). Starting 1–4 h later, Fc∊RI-activated MCs generate substantial amounts of a diverse array of cytokines and chemokines. The accumulated effects of these exocytosed mediators result in the classical acute- and late-phase reactions seen in allergic individuals. Because of their abundance in the eye, MCs play a significant role in IgE/allergen-dependent ocular inflammation.
The release of too many MC-derived mediators at an inappropriate time can result in life-threatening anaphylaxis. While this is a serious adverse consequence of MC activation, recent studies uncovered beneficial roles for MCs in innate immunity. In this regard, it was recently shown that the “test” cells in the Styela plicata1 and Ciona intestinalis (Stevens, unpublished data) sea squirts store substantial amounts of histamine and protease•heparin proteoglycan complexes in their cytoplasmic granules analogous to mouse, rat, and human cutaneous MCs. These primitive MC-like cells in urochordates also generate prostaglandin D2 when exposed ex vivo to the MC-activating factor compound 48/80. Thus, cutaneous-like MCs evolved more than 500 million years ago, predating the appearance of B cells, T cells, and the development of acquired immunity. Presumably, sea squirts use their MC-like test cells to help combat the infectious organisms encountered in their ocean environments.
In support of the perceived importance of MCs for our survival, no human has been found to lack MCs. The participation of MCs in innate immunity explains why these connective tissue-residing immune cells developed prior to adaptive immunity and why they have been conserved throughout evolution. MCs are strategically positioned to confront and combat potential environmental 2pathogens. MCs recognize bacteria and their products, and these immune cells are equipped with a complex armament to carry out their vital roles in immunity. MCs display numerous receptors on their surfaces that allow these cells to respond to pathogen-induced changes in their environments. Once activated, MCs can respond in a calculated manner using a variety of amines, proteases, eicosanoids, cytokines, and chemokines. MCs therefore possess the ability to combat bacteria and other infectious organisms in a direct manner or in an indirect manner by recruiting other cell types into the infected tissue. Here, we review data that document the roles of MCs in innate immunity, with particular emphasis on the participation of the cell's tryptases in bacterial infections.
 
MORPHOLOGY, HISTOCHEMISTRY, AND GRANULE PROTEASE•PROTEOGLYCAN COMPLEXES OF MOUSE AND HUMAN MCs
Mature mammalian MCs are unique granulocytes that reside in connective tissues in close proximity to nerves and blood vessels, in contrast to eosinophils, neutrophils, and basophils that are preferentially found in peripheral blood. Providing the initial clue as to their immunoprotective roles, MCs are most prevalent at sites of interface with the outside environment. The lung, skin, and gastrointestinal tract have 500–4,000, 7,000–12,000, and ∼20,000 MCs/mm³, respectively. In regard to the eye, MCs are present in the choroid (the vascular layer between the retina and the sclera), but they are most abundant in the conjunctiva and eyelid.
MCs can be distinguished from other cell types based on their morphology26 and histochemistry when stained with cationic dyes.7,8 Mature MCs generally possess a monolobed nucleus and a surface architecture composed of prominent ridge-like folds of plasmalemma known as microplicae. MCs also can be distinguished from other cell types due to their numerous electron-dense, cytoplasmic secretory granules. These granules in human MCs usually contain crystalline structures that exhibit novel scrolls, gratings, and lattice patterns.6 Using different histochemistry approaches, Enerbäck first demonstrated the heterogeneous nature of MCs.8 It is now known that the histochemical differences seen in different populations of rodent MCs primarily are a consequence of serglycin proteoglycans9 that possess different types of glycosaminoglycan side chains [e.g., heparin10, chondroitin sulfate diB11, and chondroitin sulfate E12].
Mature mouse and human MCs have distinct phenotypes based on the panel of proteases they store in their secretory granules bound to serglycin proteoglycans. For example, it is now known that mouse MCs store varied combinations of at least sixteen proteases in their secretory granules [designated as mouse MC protease (mMCP) 1-10, mMCP-11/Prss34, transmembrane tryptase/tryptase γ/Prss31, carboxypeptidase A3 (CPA3), cathepsin G, 3granzyme B, and neuropsin/Prss19]. The genes that encode cathepsin G, granzyme B, and mMCPs 1-5 and mMCPs 8-10 reside on chromosome 14C3. This is the second largest group of protease genes in the mouse's genome. Because these serine proteases are more similar to chymotrypsin than trypsin, the chromosome 14C3 locus historically has been named the “chymase” locus. However, it must be pointed out that some chromosome 14C3 family members (e.g., granzyme B) lack chymotryptic activity. mMCP-6,13,14 mMCP-7,15 Prss31,16 and Prss3417 are MC-restricted tryptases, and their genes reside on chromosome 17A3.3, which contains the third largest cluster of protease genes in the mouse's genome. Fourteen genes are present on chromosome 17A3.3 that encode thirteen functional tryptic-like serine proteases, each of which exhibit distinct tissue and cell expression patterns.17
Human MCs also store numerous proteases in their secretory granules. These include cathepsin G, granzyme B, CPA3, chymase-1, Prss31, and multiple tetramer-forming tryptases. In regard to the latter family of serine proteases, five cDNAs have been isolated that encode the homologous human tryptases known as α, β1, β2, β3, and δ.1821 It tentatively has been concluded that these cDNAs originate from three genes on chromosome 16p13.3.22 The uncertainty is due to the fact that this region of the human genome is undergoing a high rate of mutation and recombination.23 While it is likely that there is evolutionary pressure to expand the number of beneficial tryptases and to eliminate potentially harmful ones, the high rate of recombination at chromosome 16p13.3 probably is due to a chromosomal break and inversion event that took place at the locus after mice and humans separated in evolution.17
hTryptase-β120,24,25 is the functional ortholog of mMCP-6.13,14,26 Both MC-restricted proteases are initially expressed as glycoprotein zymogens that in each instance have a signal peptide that is removed in the endoplasmic reticulum and a 10-mer propeptide that is removed in the secretory granule. The resulting mature domain of each tryptase consists of 245 amino acids. When activated, mMCP-6 and hTryptase-β1 spontaneously form tetramers with the active site of each monomer facing inside the doughnut-shaped tetramer unit. Formation of the novel tetramer unit is dependent on conserved Tyr-, Pro­-, and Trp-rich domains within each monomer,2729 whereas interaction of the tetramer unit with the glycosaminoglycan side chain of a serglycin proteoglycan is dependent on spatially conserved His, Arg, and Lys residues.27,28,30 The functional significance of the binding of the MC's granule proteases to serglycin proteoglycans was conclusively demonstrated when it was discovered that the MCs in the skin and peritoneal cavity of serglycin-null31 and heparin-null32,33 mice are protease deficient. Because their proteoglycan-binding domains are Lys/Arg-rich rather than His-rich as in mMCP-7, exocytosed mMCP-6 and hTryptase-β1 remain tightly bound to their serglycin proteoglycans.34 This feature prevents diffusion of exocytosed mMCP-46/hTryptase-β1•proteoglycan macromolecular complexes, thereby allowing these tryptases to carry out their functions locally for hours. Serglycin proteoglycans also play significant roles in regulating the substrate specificity of their bound proteases.26,35
mMCP-6 and mMCP-7 are packaged in MCs as homotypic and heterotypic tetramers ionically bound to serglycin proteoglycans.29 The fact that these tryptases can be released as heterotypic complexes suggests they have complementary activity in vivo. The role of mMCP-7 is explained by its potent anticoagulant activity. In this regard, the α chain of fibrinogen is a physiologic substrate of mMCP-7, and no protease inhibitor in blood exists to rapidly inactivate this tryptase.36 mMCP-7 therefore works in concert with mMCP-6 to assist in the recruitment of other immune cells into the inflammatory area by preventing the deposition of fibrin-platelet clots, which have the ability to block cellular extravasation physically.
 
MC DEVELOPMENT
All mouse MCs originate from hematopoietic progenitors that reside in the bone marrow and fetal liver.37,38 The systemic and local factors that regulate the tissue recruitment of these progenitors, and subsequently their proliferation and development into mature MCs, are major areas of investigation. Much of this work has been carried out on cultured cells, and many of the factors that regulate the development of mouse and human MCs have been identified. In the mouse, interleukin (IL) 339,40 and Kit ligand (KitL)/stem cell factor/steel factor4143 are the two most important cytokines that regulate the retention of MC-committed progenitors in tissues and/or their proliferation, differentiation, and granule maturation. However, accessory cytokines such as IL-4,4446 IL-6,47 IL-9,48,49 IL-10,50,51 IL-16,52 and transforming growth factor-β1,53,54 also play significant roles in the final stages of development of human and mouse MCs. Although human MCs and their progenitors are less responsive to IL-3 than mouse MCs, IL-3 is routinely used to generate mouse MCs by culturing their unfractionated bone marrow cells in the presence of the cytokine for 3-6 weeks.39,40,55,56 Arguably, the most important discovery in the MC field in the last 30 years was the identification of IL-3 and the use of this T cell-derived cytokine to generate large numbers of mouse bone marrow-derived MCs (mBMMCs) for varied in vitro and in vivo studies. Although IL-3 is not needed for the development of constitutive heparin+ MCs, this cytokine is required for the rapid expansion of chondroitin sulfate+ mouse MCs in the small intestine during a helminth infection.57
KitL4143 binds to the tyrosine kinase-type surface receptor CD117/Kit58, and KitL/Kit signaling is essential for the development of the heparin+ population of MCs in mice and humans.59 In this regard, WBB6F1KitW/KitW-v (W/Wv) mice are MC-deficient due to an inactivating point mutation in the intracellular domain of 5Kit.60 WCB6F1-KitlSl/KitlSl-d mice are MC-deficient because their mesenchymal cells can not produce the membrane isoform of the KitL, which is needed to retain MC-committed progenitors in tissues.61,62 On the other hand, tg/tg mice are MC-deficient due to a defect in expression of microphthalmia-associated transcription factor (MITF)63,64, which acts downstream of Kit. MITF is essential for transcription of the mMCP-6 gene65 and numerous other MC-restricted genes. The discovery of an activating mutation at residue 816 in Kit that leads to systemic mastocytosis in humans66,67 supports the mouse data, which led to the conclusion that KitL/Kit/MITF-signaling pathways control the development of the constitutive MCs found in most tissues.
MCs exhibit substantial plasticity in their development, and it is now known that the phenotype of a MC at any stage of its life-cycle is highly dependent on the combination of factors the mature cell and its progenitor encounter in different tissue microenvironments. Mature MCs differ from other immune cells in that they can reversibly alter what mediators they express. This property was elegantly shown in the small intestine of Trichinella spiralis-infected mice.5,68,69 A transient mastocytosis occurs in the jejunum of helminth-infected mice5,70 and rats.71,72 The expanded MCs display specific combinations of proteases depending on their location within the jejunum and the time course of the infection. Most mature MCs in the jejunum reside in the muscle and submucosa at baseline, and these cells express mMCP-5, mMCP-6, and mMCP-7, but not mMCP-1 or mMCP-2. During the inductive phase of the infection, jejunal MCs cease expressing mMCP-5, mMCP-6, and mMCP-7, and begin expressing mMCP-1 and mMCP-2 as they transverse from the submucosa to the tips of the villus. This differentiation process is reversed during the expulsion phase of the infection as the senescent MCs slowly make their way from the mucosa to draining lymph nodes and ultimately to the spleen. Based on these and other data, it is now apparent that MCs can reversibly alter their protease phenotype in a time- and strata-dependent manner. The observed changes in protease expression have functional significance, as the expulsion of T. spiralis is delayed in mMCP-1-null mice.73 Similar to their protease mediators, MCs also can reversibly alter their expression of prostaglandins and leukotrienes in a cytokine-dependent manner.74,75
The experimental model of MC adaptation in the response to a helminth infection has not yet been studied in a bacterial infection and in different inflammatory disorders. Nevertheless, the ability of a MC to alter its expression of the proinflammatory tryptases mMCP-6 and mMCP-7 in a reversible manner is likely to impact the animal's ability to recruit peripheral blood neutrophils and eosinophils into a tissue to combat an infectious organism. It also remains to be determined if the MCs in the conjunctiva can reversibly alter their phenotype in response to different pathogens as occurs in the jejunum. However, at the mRNA level, the two most abundant MC-restricted proteases in the conjunctiva 6are the tryptases mMCP-6 and mMCP-7 (Miyazaki, Stevens, and Ono, unpublished data), thereby suggesting a prominent role for these tryptases in ocular inflammation.
 
RECEPTORS THAT MCs USE TO RECOGNIZE BACTERIA AND OTHER INFECTIOUS ORGANISMS
Mature MCs use numerous receptors to recognize and respond to factors produced by bacteria and other infectious organisms (Fig. 1-1). The MCs response to these foreign factors can be opsonin-dependent or opsonin-independent.
zoom view
Fig. 1-1: MC participation in innate and acquired immunity. The MCs in the eye and other tissue sites express a diverse array of receptors (e.g., TLR2, TLR4, TLR6, CD48, PAR-1, and PAR-2) that recognize factors that originate from bacteria, yeast, and other infectious organisms. MCs also possess receptors (e.g., Fc∊RI, FcγRII, and FcγRIII) that recognize immunoglobulins generated in the host against pathogens. Finally, the coating of bacteria and other infectious organisms with complement proteins results in the generation of anaphylatoxins which bind to the C3a and C5a/CD88 receptors on the MC's surface. MCs release varied combinations of biologically active factors when they become activated via these signaling pathways. Cross-linking of a surface receptor such as Fc∊RI results in the rapid release of the preformed mediators stored in the cell's granules [e.g., histamine, serotonin, and protease•serglycin proteoglycan (PG) complexes]. Mouse MCs store 16 proteases in their granules, and mMCP-6 and mMCP-7 are the cell's tetramer-forming tryptases. Homotypic mMCP-6 tetramers remain bound to serglycin proteoglycans when exocytosed from activated MCs, whereas homotypic mMCP-7 tetramers dissociate from their macromolecular complexes. A few minutes after the cell's high-affinity IgE receptors are cross-linked, activated MCs release varied combinations of prostaglandins, leukotrienes, and other lipid mediators. Hours later, Fc∊RI-activated MCs markedly increase their expression of varied combinations of at least 31 different cytokines and chemokines. Thus, activated MCs produce more biologically active factors than any other cell type in the body
7
As an example of their participation in acquired immunity, MCs can bind to helminths in an opsonin-dependent manner. In this scenario, helminths evoke a humoral response that results in the generation of helminth-specific IgE. The binding of this immunoglobulin to surface Fc∊RI receptors allows MCs the ability to recognize and bind to the helminth.76 After a bacterial infection, mice and humans normally produce pathogen-specific IgG. Nevertheless, in some instances, MCs also can respond to bacteria in an IgE/Fc∊RI-dependent manner. For example, the generation of IgE that recognizes Staphylococcus aureus-derived exotoxin can induce the release of mediators from Fc∊RI-bearing MCs and basophils, which, in turn, can lead to atopic dermatitis.77 Mouse78 and human79 MCs also express the low-affinity IgG receptors FcγRII and FcγRIII on their surfaces. Some populations of MCs become activated via these Fcγ receptors when they encounter immune complexes,80 and the inflammatory Arthus reaction to antibody-antigen complexes is a consequence of the activation of cutaneous MCs via their FcγRIII receptors.81 Bullous pemphigoid is an autoimmune disorder in which autoreactive IgG recognizes the hemidesmosomal proteins BP230 and BP180. The binding of anti-BP230 and/or anti-BP180 IgG to the MC's FcγR receptors results in MC degranulation, neutrophil infiltration, and skin blistering.82
In regard to their role in innate immunity, MCs can respond to complement-derived anaphylatoxins due to their C3a and C5a/CD88 receptors. Because sea squirts express complement83, it is likely that their MC-like “test” cells also are regulated by complement-derived factors like C3a and C5a. Sepsis occurs in the mouse when its cecum is ligated and punctured (CLP). The resultant accumulation of neutrophils into the bacteria-infected peritoneal cavity is highly dependent on factors released from the peritoneal MCs.84 Prodeus and coworkers85 noted that C3-deficient mice are more sensitive to CLP-induced sepsis than wild-type mice. In this bacterial infection model, fewer neutrophils and fewer degranulated MCs were found in CLP-treated C3-deficient mice. The ability to rescue the mouse's impaired neutrophil response to the bacterial infection by treating the C3-deficient mice with purified C3 protein confirmed the key role for complement pathways in MC-dependent sepsis.
MCs respond to fimbriated Escherichia coli in an opsonin-independent manner. FimH is the mannose-binding subunit of type 1 fimbriae, which are rod-like adhesive structures that project from the outer surface of E. coli and other enterobacteria. Malaviya and coworkers reported that MCs express CD48 and use this SLAM (signaling lymphocyte-activation molecule) family member to recognize and phagocytose FimH-expressing E. coli.86,87 The biologic relevance of the CD48-FimH interaction remains to be determined. Although it was initially proposed that MCs use CD48 primarily to kill E. coli, neutrophils are the primary cells in the body that eliminate bacteria. Thus, a more likely scenario is that FimH-CD48-dependent signaling events induce MCs to release factors that induce the recruitment of large numbers of neutrophils into the bacteria-infected site.8
MCs also can recognize factors produced by infectious organisms via their Toll-like receptors (TLRs). In vitro-differentiated mBMMCs express TLR2, TLR4, and TLR6.88,89 TLR2 forms a dimer with TLR6, and this receptor complex recognizes bacterial peptidoglycan and yeast zymosan, whereas TLR4 recognizes bacterial lipopolysaccharide (LPS). mBMMCs increase their expression of granulocyte-macrophage-colony stimulating factor, IL-1β, and cysteinyl leukotrienes when exposed to peptidoglycan and zymosan, but not LPS. In contrast, in vitro-differentiated human MCs that had been exposed to LPS increased their expression of tumor necrosis factor-α (TNF-α), IL-5, IL-10, and IL-13, as did peptidoglycan-treated human MCs.90 Peptidoglycan additionally induced histamine release from this population of cells. It therefore is apparent that the mediators released from MCs by bacterial products are dependent on what TLR is used in the signal-transduction event. Because MCs are heterogeneous, it remains to be determined if the MCs in every human and every mouse respond to TLR and CD48 agonists in the same way. In this regard, mouse fetal skin-derived MCs express much more TLR3 and TLR7 than mBMMCs. Nevertheless, the cytokine responses that have been observed so far in TLR-activated mouse and human MCs are distinct from the histamine and eicosanoid responses evoked by cross-linking of the cell's high-affinity IgE receptors. The accumulated CD48 and TLR data highlight the many ways in which mouse and human MCs are able to recognize various bacteria and illicit diverse responses.
An unexplored area of investigation is the ability of MCs to recognize bacteria via NOD1/CARD4 and NOD2/CARD15. NOD2 recognizes bacterial muramyldipeptide, whereas NOD1 recognizes the peptidoglycan-specific dipeptide γ-D-glutamyl-meso-diaminopimelic acid. MCs have been implicated in inflammatory bowel disease91, and many patients with Crohn's disease have a mutated NOD2 gene.92,93 While it remains to be seen if MCs use NOD1 and/or NOD2 to recognize microbial products, we recently discovered that in vitro-differentiated mouse and human MCs express NOD1 and NOD2 (Hamilton, Krilis, and Stevens, unpublished findings).
Lastly, MCs express proteinase-activated receptor (PAR) 1 and PAR-2, and the cellular activation via these receptors results in degranulation and increased expression of cytokines.9498 While the emphasis in this area of investigation has been on the ability of thrombin, trypsin, and other endogenous proteases to activate MCs via their PARs, it is now apparent that numerous proteases produced by infectious organisms and biting insects activate mammalian cells via this family of receptors. For example, the house dust mite proteases Der p3 and Der p9 are potent activators of PAR-2.99 House dust mites live in beds, sofas, and carpets, and feed on human skin. Their protease-rich fecal droppings elicit extreme allergic reactions in many humans who live in moist climates where the insect infestation is greatest. The accumulated data suggest that MCs also use the receptor PAR-2 in immune surveillance.9
 
ROLE OF MCs AND THEIR TRYPTASE MEDIATORS IN BACTERIAL INFECTIONS
Earlier in vitro studies revealed that MCs release their mediators when exposed to bacteria. However, the importance of MCs in bacterial infections did not become apparent until in vivo studies were carried out on MC-deficient W/Wv mice before and after their reconstitution with wild-type +/+ mBMMCs. Using the CLP model of sepsis, Echtenacher and coworkers84 noted that W/Wv mice are more susceptible to E. coli than their MC-sufficient +/+ littermates. Malaviya and coworkers100 obtained similar data when Klebsiella pneumoniae was injected into the peritoneal cavity or lungs of a W/Wv mouse. In the latter infection model, W/Wv mice were ∼20-fold less efficient than +/+ mice in clearing the enterobacteria. The MC-deficiency of W/Wv mice can be rescued using an adoptive transfer approach in which the mice are given unfractionated bone marrow cells or in vitro-differentiated mBMMCs from their histocompatible +/+ littermates.101,102 The ability of reconstituted W/Wv mice to combat K. pneumoniae confirmed a key role for MCs in the anti-bacteria response.100 When evaluating for a potential mechanism to explain the increased lethality seen in the W/Wv mice, Malaviya and coworkers100 discovered a significant difference in the number of neutrophils in the infected peritoneal cavities of W/Wv mice compared with that in +/+ mice. Because neutrophils are the professional killers of bacteria in the body, it became evident from these and other experiments that MCs primarily were indirectly involved in combating bacterial infections by inducing the recruitment of neutrophils.
It is likely that MCs produce a diverse array of factors that work in concert to orchestrate an effective anti-bacteria response. mBMMCs can produce modest amounts of leukotriene B4,103 which is a potent chemotactic factor for neutrophils.104 However, the fact that the MCs in the mouse and rat peritoneal cavity preferentially metabolize arachidonic acid to prostaglandin D2105 suggested that leukotriene B4 was not the primary antimicrobial factor produced by peritoneal MCs. While MCs also release histamine when activated, this vasopermeability factor turned out not to be the MC-restricted factor needed for eliminating bacteria because histamine-null mice actually combated bacteria better than histamine-sufficient mice.106 The MCs in the peritoneal cavity contain substantial amounts of mMCP-4 and mMCP-5.13,107,108 These two chromosome 14C3 family members (as well as mMCP-1, mMCP-2, mMCP-3/4L, mMCP-8, mMCP-9, mMCP-10, cathepsin G, and granzyme B) are expressed as zymogens that contain 2-mer propeptides. Because dipeptidyl peptidase I (DPPI)/cathepsin C is used to activate the chromosome 14C3 family of serine proteases109, the MCs in DPPI-null mice have very little, if any, enzymatically active mMCP-4 and mMCP-5. Like histamine-null mice, DDPI-null mice fared better than their wild-type counterparts with respect to a bacterial challenge.110 The reasons for these unexpected findings remain to be determined. However, it is presumed 10that histamine, mMCP-4, and/or mMCP-5 play adverse roles in bacterial infections by inducing vasopermeability, which, in turn, allows more harmful bacteria to enter the circulation. Whatever the mechanism, none of the MC's chromosome 14C3 family members appear to have a beneficial role in bacterial clearance.
TNF-α is a neutrophil chemoattractant transiently produced by activated MCs.111113 Using anti-TNF-α antibody blocking approaches, Malaviya100 and Echtenacher84 and their coworkers tentatively concluded that TNF-α was the critical MC-derived factor required for efficient bacterial clearance. However, macrophages greatly outnumber MCs in the peritoneal cavity and lung. When activated with LPS, macrophages produce more TNF-α than activated MCs. Furthermore, TNF-α-null mice successfully combated a bacterial infection when their MCs were activated.114 It therefore became apparent that another factor more restricted to MCs had to be involved in the neutrophil recruitment seen in bacterial infections.
Administration of recombinant mMCP-626,115 or hTryptase-β124 into varied mouse tissues resulted in the preferential accumulation of neutrophils. Moreover, hTryptase-β1-treated W/Wv mice could combat a K. pneumoniae infection better than pro-Tryptase β1-treated W/Wv mice.24 In support of a key role for the MC's tryptases in granulocyte accumulation, Oh and coworkers116 were able to block airway inflammation in mice given the tryptase inhibitor MOL6131. Transcription of the mMCP-6 gene is greatly diminished in tg/tg mice.65 The fact that tg/tg mice also cannot combat bacterial infections efficiently supported the conclusion that mMCP-6 was likely the critical MC-restricted anti-microbial factor.117 Definitive proof that this tryptase has a critical immunoprotective role in bacterial infections occurred when it was shown that a newly created mMCP-6-null mouse could not survive a K. pneumoniae infection of its peritoneal cavity.118
The mechanisms by which mMCP-6 and its human ortholog hTryptase-β1 regulate granulocyte accumulation in tissues at the molecular level remain to be determined, but in vitro studies revealed that mMCP-6 can induce human endothelial cells to increase their expression of IL-8, which is a potent chemoattractant for neutrophils.26 The accumulated data suggest that mMCP-6 in mice and hTryptase-β1 in humans control bacterial infections in an indirect manner by inducing bystander cells to secrete factors that recruit bactericidal neutrophils.
 
CONCLUSION
While recent studies on the MC's tryptase tetramers have provided exciting new insight into the beneficial roles of MCs in immunity, many questions remain unanswered. This review has highlighted some of the pertinent studies that have brought to the forefront the importance of MC-restricted tryptases in 11bacterial infections. Nevertheless, little is known about how these proteases are activated inside MCs from their zymogen precursors, how the enzymatically active mature proteases are metabolized after their exocytosis from the MC to dampen their pro-inflammatory effects, and how they interact with other mediators and cells in the body. MCs and their progenitors are highly susceptible to M-tropic strains of HIV-1119121, and the number of hTryptase-β1+ MCs are greatly reduced in the gastrointestinal tract of AIDS patients.122 Because AIDS patients are highly susceptible to bacteria and other infectious organisms, it is possible that recombinant hTryptase-β1 could provide beneficial therapy to these immunocompromised patients. In contrast, pharmacologic inhibition of the MC's tryptases might have benefit in various inflammatory disorders characterized by prominent neutrophil accumulation after MC activation as occurs in rheumatoid arthritis and ocular inflammation.
The ability of MC-restricted tryptases to help mammals fight off bacteria explains why their genes have been conserved. Nevertheless, mouse MCs store varied combinations of 14 other serine proteases in their granules. Some of these proteases (e.g., CPA3, Prss31, and mMCP-5) have human orthologs. The mMCP-6/hTryptase-β1 studies noted in this review raise the possibility that CPA3, Prss31, and mMCP-5/chymase-1 have been conserved for millions of years because they also possess yet unexplored beneficial roles in innate and/or acquired immunity. Mature MCs display a wide range of phenotypes in tissues and can adapt to changes in the local environment. The functional significance of MC heterogeneity has not been adequately explored. It therefore is anticipated that future studies will illuminate the importance of MCs and their diverse array of mediators in various infections, and perhaps even in situations where MCs respond inappropriately. After all, MCs today encounter a far different world than that which existed when these cells first appeared in evolution.
REFERENCES
  1. Cavalcante MC, Allodi S, Valente AP, Straus AH, Takahashi HK, Mourao PA, Pavao MS. Occurrence of heparin in the invertebrate Styela plicata (Tunicata) is restricted to cell layers facing the outside environment. An ancient role in defense? J Biol Chem. 2000; 275: 36189–96.
  1. Enerbäck L, Lundin PM. Ultrastructure of mucosal mast cells in normal and compound 48-80-treated rats. Cell Tissue Res. 1974; 150: 95–105.
  1. Craig SS, Schechter NM, Schwartz LB. Ultrastructural analysis of human T and TC mast cells identified by immunoelectron microscopy. Lab Invest. 1988; 58: 682–91.
  1. Weidner N, Austen, KF. Evidence for morphologic diversity of human mast cells: an ultrastructural study of mast cells from multiple body sites. Lab Invest. 1990; 63: 63–72.
  1. Friend DS, Ghildyal N, Austen KF, Gurish MF, Matsumoto R, Stevens RL. Mast cells that reside at different locations in the jejunum of mice infected with Trichinella spiralis exhibit sequential changes in their granule ultrastructure and chymase phenotype. J Cell Biol. 1996; 135: 279–90.

  1. 12 Caulfield JP, Lewis RA, Hein A, Austen KF. Secretion in dissociated human pulmonary mast cells: evidence for solubilization of granule contents before discharge. J Cell Biol. 1980; 85: 299–312.
  1. Ehrlich P. Beitrage zur Theorie und Praxis der Histologischen Farbung. Doctoral thesis, University of Leipzig, Germany. 1878.
  1. Enerbäck L. Mast cells in rat gastrointestinal mucosa. 2. Dye-binding and metachromatic properties. Acta Pathol Microbiol Scand. 1966; 66: 303–12.
  1. Stevens RL, Avraham S, Gartner MC, Bruns GA, Austen KF, Weis JH. Isolation and characterization of a cDNA that encodes the peptide core of the secretory granule proteoglycan of human promyelocytic leukemia HL-60 cells. J Biol Chem. 1988; 263: 7287–91.
  1. Lindahl U, Kusche M, Lidholt K, Oscarsson LG. Biosynthesis of heparin and heparan sulfate. Ann N Y Acad Sci. 1989; 556: 36–50.
  1. Seldin DC, Austen KF, Stevens RL. Purification and characterization of protease-resistant secretory granule proteoglycans containing chondroitin sulfate di-B and heparin-like glycosaminoglycans from rat basophilic leukemia cells. J Biol Chem. 1985; 260: 11131–39.
  1. Razin E, Stevens RL, Akiyama F, Schmid K, Austen KF. Culture from mouse bone marrow of a subclass of mast cells possessing a distinct chondroitin sulfate proteoglycan with glycosaminoglycans rich in N-acetylgalactosamine-4,6-disulfate. J Biol Chem. 1982; 257: 7229–36.
  1. Reynolds DS, Stevens RL, Lane WS, Carr MH, Austen KF, Serafin WE. Different mouse mast cell populations express various combinations of at least six distinct mast cell serine proteases. Proc Natl Acad Sci USA. 1990; 87: 3230–34.
  1. Reynolds DS, Gurley DS, Austen KF, Serafin WE. Cloning of the cDNA and gene of mouse mast cell protease 6: transcription by progenitor mast cells and mast cells of the connective tissue subclass. J Biol Chem. 1991; 266: 3847–53.
  1. McNeil HP, Reynolds DS, Schiller V, Ghildyal N, Gurley DS, Austen KF, Stevens RL. Isolation, characterization, and transcription of the gene encoding mouse mast cell protease 7. Proc Natl Acad Sci USA. 1992; 89: 11174–78.
  1. Wong GW, Foster PS, Yasuda S, Qi JC, Mahalingam S, Mellor EA, Katsoulotos G, Li L, Boyce JA, Krilis SA, Stevens RL. Biochemical and functional characterization of human transmembrane tryptase (TMT)/tryptase γ: TMT is an exocytosed mast cell protease that induces airway hyperresponsiveness in vivo via an IL-13/IL-4Rα/STAT6-dependent pathway. J Biol Chem. 2002; 277: 41906–15.
  1. Wong GW, Yasuda S, Morokawa N, Li L, Stevens RL. Mouse chromosome 17A3.3 contains thirteen genes that encode functional tryptic-like serine proteases with distinct tissue and cell expression patterns. J Biol Chem. 2004; 279: 2438–52.
  1. Miller JS, Westin EH, Schwartz LB. Cloning and characterization of complementary DNA for human tryptase. J Clin Invest. 1989; 84: 1188–95.
  1. Miller JS, Moxley G, Schwartz LB. Cloning and characterization of a second complementary DNA for human tryptase. J Clin Invest. 1990; 86: 864–70.
  1. Vanderslice P, Ballinger SM, Tam EK Goldstein SM, Craik CS, Caughey GH. Human mast cell tryptase: multiple cDNAs and genes reveal a multi-gene serine protease family. Proc Natl Acad Sci USA. 1990; 87: 3811–15.
  1. Wang HW, McNeil HP, Husain A, Liu K, Tedla N, Thomas PS, Raftery M, King GC, Cai ZY, Hunt JE. δ tryptase is expressed in multiple human tissues, and a recombinant form has proteolytic activity. J Immunol. 2002; 169: 5145–52.
  1. Pallaoro M, Fejzo MS, Shayesteh L, Blount JL, Caughey GH. Characterization of genes encoding known and novel human mast cell tryptases on chromosome 16p13.3. J Biol Chem. 1999; 274: 3355–62.

  1. 13 Badge RM, Yardley J, Jeffreys AJ, Armour JA. Crossover breakpoint mapping identifies a subtelomeric hotspot for male meiotic recombination. Hum Mol Genet. 2000; 9: 1239–44.
  1. Huang C, De Sanctis GT, O'Brien PJ Mizgerd JP, Friend DS, Drazen JM, Brass LF, Stevens RL. Evaluation of the substrate specificity of human mast cell tryptase β1 and demonstration of its importance in bacterial infections of the lung. J Biol Chem. 2001; 276: 26276–84.
  1. Harris JL, Niles A, Burdick K, Maffitt M, Backes BJ, Ellman JA, Kuntz I, Haak-Frendscho M, Craik CS. Definition of the extended substrate specificity determinants for β tryptases I and II. J Biol Chem. 2001; 276: 34941–47.
  1. Huang C, Friend DS, Qiu WT, Wong GW, Morales G, Hunt J, Stevens RL. Induction of a selective and persistent extravasation of neutrophils into the peritoneal cavity by tryptase mouse mast cell protease 6. J Immunol. 1998; 160: 1910–19.
  1. Johnson DA, Barton GJ. Mast cell tryptases: examination of unusual characteristics by multiple sequence alignment and molecular modeling. Protein Sci. 1992; 1: 370–77.
  1. Pereira PJ, Bergner A, Macedo-Ribeiro S, Huber R, Matschiner G, Fritz H, Sommerhoff CP, Bode W. Human β-tryptase is a ring-like tetramer with active sites facing a central pore. Nature. 1998; 392: 306–11.
  1. Huang C, Morales G, Vagi A, Chanasyk K, Ferrazzi M, Burklow C, Qiu WT, Feyfant E, Šali A, Stevens RL. Formation of enzymatically active, homotypic, and heterotypic tetramers of mouse mast cell tryptases: dependence on a conserved Trp-rich domain on the surface. J Biol Chem. 2000; 275: 351–58.
  1. Matsumoto R, Sali A, Ghildyal N, Karplus M, Stevens RL. Packaging of proteases and proteoglycans in the granules of mast cells and other hematopoietic cells: a cluster of histidines on mouse mast cell protease 7 regulates its binding to heparin serglycin proteoglycans. J Biol Chem. 1995; 270: 19524–31.
  1. Abrink M, Grujic M, Pejler G. Serglycin is essential for maturation of mast cell secretory granule. J Biol Chem. 2004; 279: 40897–905.
  1. Humphries DE, Wong GW, Friend DS, Gurish MF, Qiu WT, Huang C, Sharpe AH, Stevens RL. Heparin is essential for the storage of specific granule proteases in mast cells. Nature. 1999; 400: 769–72.
  1. Forsberg E, Pejler G, Ringvall M, Lunderius C, Tomasini-Johansson B, Kusche-Gullberg M, Eriksson I, Ledin J, Hellman L, Kjellén L. Abnormal mast cells in mice deficient in a heparin-synthesizing enzyme. Nature. 1999; 400: 773–76.
  1. Ghildyal N, Friend DS, Stevens RL, Austen KF, Huang C, Penrose JF, Sali A, Gurish MF. Fate of two mast cell tryptases in V3 mastocytosis and normal BALB/c mice undergoing passive systemic anaphylaxis: prolonged retention of exocytosed mMCP-6 in connective tissues, and rapid accumulation of enzymatically active mMCP-7 in the blood. J Exp Med. 1996; 184: 1061–73.
  1. Le Trong H, Neurath H, Woodbury RG. Substrate specificity of the chymotrypsin-like protease in secretory granules isolated from rat mast cells. Proc Natl Acad Sci USA. 1987; 84: 364–67.
  1. Huang C, Wong GW, Ghildyal N, Gurish MF, Šali A, Matsumoto R, Qiu WT, Stevens RL. 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; 272: 31885–93.
  1. Kitamura Y, Shimada M, Hatanaka K, Miyano Y. Development of mast cells from grafted bone marrow cells in irradiated mice. Nature. 1977; 268: 442–43.

  1. 14 Kitamura Y, Shimada M, Go S, Matsuda H, Hatanaka K, Seki M. Distribution of mast-cell precursors in hematopoeitic and lymphopoietic tissues of mice. J Exp Med. 1979; 150: 482–90.
  1. Ihle JN, Keller J, Oroszlan S, Henderson LE, Copeland TD, Fitch F, Prystowsky MB, Goldwasser E, Schrader JW, Palaszynski E, Dy M, Lebel B. Biologic properties of homogeneous interleukin 3. I. Demonstration of WEHI-3 growth factor activity, mast cell growth factor activity, P cell-stimulating factor activity, colony-stimulating factor activity, and histamine-producing cell-stimulating factor activity. J Immunol. 1983; 131: 282–87.
  1. Razin E, Ihle JN, Seldin D, Mencia-Huerta JM, Katz HR, Le Blanc PA, Hein A, Caulfield JP, Austen KF, Stevens RL. Interleukin 3: a differentiation and growth factor for the mouse mast cell that contains chondroitin sulfate E proteoglycan. J Immunol. 1984; 132: 1479–86.
  1. Huang E, Nocka K, Beier DR, Chu TY, Buck J, Lahm HW, Wellner D, Leder P, Besmer P. The hematopoietic growth factor KL is encoded by the Sl locus and is the ligand of the c-kit receptor, the gene product of the W locus. Cell 1990; 63: 225–33.
  1. Martin FH, Suggs SV, Langley KE, Lu HS, Ting J, Okino KH, Morris CF, McNiece IK, Jacobsen FW, Mendiaz EA et al. Primary structure and functional expression of rat and human stem cell factor DNAs. Cell. 1990; 63: 203–11.
  1. Zsebo KM, Wypych J, McNiece IK, Lu HS, Smith KA, Karkare SB, Sachdev RK, Yuschenkoff VN, Birkett NC, Williams LR et al. Identification, purification, and biological characterization of hematopoietic stem cell factor from buffalo rat liver-conditioned medium. Cell. 1990; 63: 195–201.
  1. Mosmann TR, Bond MW, Coffman RL, Ohara J, Paul WE. T-cell and mast cell lines respond to B-cell stimulatory factor 1. Proc Natl Acad Sci USA. 1986; 83: 5654–58.
  1. Hamaguchi Y, Kanakura Y, Fujita J, Takedo S, Nakano T, Tarui S, Honjo T, Kitamura Y. Interleukin 4 as an essential factor for in vitro clonal growth of murine connective tissue-type mast cells. J Exp Med. 1987; 165: 268–73.
  1. Lorentz A, Bischoff, SC. Regulation of human intestinal mast cells by stem cell factor and IL-4. Immunol Rev. 2001; 179: 57–60.
  1. Kinoshita T, Sawai N, Hidaka E, Yamashita T, Koike K. Interleukin-6 directly modulates stem cell factor-dependent development of human mast cells derived from CD34+ cord blood cells. Blood. 1999; 94: 496–508.
  1. Hültner L, Druez C, Moeller J, Uyttenhove C, Schmitt E, Rude E, Dormer P, van Snick J. Mast cell growth-enhancing activity (MEA) is structurally related and functionally identical to the novel mouse T cell growth factor P40/TCGFIII (interleukin 9). Eur J Immunol. 1990; 20: 1413–16.
  1. Eklund KK, Ghildyal N, Austen KF, Stevens RL. Induction by IL-9 and suppression by IL-3 and IL-4 of the levels of chromosome 14-derived transcripts that encode late-expressed mouse mast cell proteases. J Immunol. 1993; 151: 4266–73.
  1. Ghildyal N, McNeil HP, Gurish MF, Austen KF, Stevens RL. Transcriptional regulation of the mucosal mast cell-specific protease gene, mMCP-2, by interleukin 10 and interleukin 3. J Biol Chem. 1992; 267: 8473–77.
  1. Ghildyal N, Friend DS, Nicodemus CF, Austen KF, Stevens RL. Reversible expression of mouse mast cell protease 2 mRNA and protein in cultured mast cells exposed to IL-10. J Immunol. 1993; 151: 3206–14.
  1. Qi JC, Stevens RL, Wadley R, Collins A, Cooley M, Naif HM, Nasr N, Cunningham A, Katsoulotos G, Wanigasek Y, Roufogalis B, Krilis SA. IL-16 regulation of human mast cells/basophils and their susceptibility to HIV-1. J Immunol. 2002; 168: 4127–34.

  1. 15 Wright SH, Brown J, Knight PA, Thornton EM, Kilshaw PJ, Miller HR. Transforming growth factor-β1 mediates coexpression of the integrin subunit αE and the chymase mouse mast cell protease-1 during the early differentiation of bone marrow-derived mucosal mast cell homologues. Clin Exp Allergy. 2002; 32: 315–24.
  1. Funaba M, Ikeda T, Murakami M, Ogawa K, Abe M. Up-regulation of mouse mast cell protease-6 gene by transforming growth factor-β and activin in mast cell progenitors. Cell Signal. 2005; 17: 121–28.
  1. Razin E, Cordon-Cardo C, Good RA. Growth of a pure population of mouse mast cells in vitro with conditioned medium derived from concanavalin A-stimulated splenocytes. Proc Natl Acad Sci USA. 1981; 78: 2559–61.
  1. Schrader JW, Lewis SJ, Clark-Lewis I, Culvenor JG. The persisting (P) cell: histamine content, regulation by a T cell-derived factor, origin from a bone marrow precursor, and relationship to mast cells. Proc Natl Acad Sci USA. 1981; 78: 323–27.
  1. Madden KB, Urban Jr, JF Ziltener HJ, Schrader JW, Finkelman FD, Katona IM. Antibodies to IL-3 and IL-4 suppress helminth-induced intestinal mastocytosis. J Immunol. 1991; 147: 1387–91.
  1. Yarden Y, Kuang WJ, Yang-Feng T, Coussens L, Munemitsu S, Dull TJ, Chen E, Schlessinger J, Francke U, Ullrich A. Human proto-oncogene c-kit: a new cell surface receptor tyrosine kinase for an unidentified ligand. EMBO J. 1987; 6: 3341–51.
  1. Galli SJ, Zsebo KM, Geissler EN. The kit ligand, stem cell factor. Adv Immunol. 1994; 55: 1–96.
  1. Geissler EN, Ryan MA, Housman DE. The dominant-white spotting (W) locus of the mouse encodes the c-kit proto-oncogene. Cell. 1988; 55: 185–92.
  1. Flanagan JG, Leder, P. The kit ligand: a cell surface molecule altered in steel mutant fibroblasts. Cell. 1990; 63: 185–94.
  1. Flanagan JG, Chan DC, Leder P. Transmembrane form of the c-kit ligand growth factor is determined by alternative splicing and is missing in the Slα mutant. Cell. 1991; 64: 1025–35.
  1. Hodgkinson CA, Moore KJ, Nakayama A, Steingrimsson E, Copeland NG, Jenkins NA, Arnheiter H. Mutations at the mouse microphthalmia locus are associated with defects in a gene encoding a novel basic-helix-loop-helix-zipper protein. Cell. 1993; 74: 395–404.
  1. Ito A, Morii E, Kim DK, Kataoka TR, Jippo T, Maeyama K, Nojima H, Kitamura Y. Inhibitory effect of the transcription factor encoded by the mi mutant allele in cultured mast cells of mice. Blood. 1999; 93: 1189–96.
  1. Morii E, Tsujimura T, Jippo T, Hashimoto K, Takebayashi K, Tsujino K, Nomura S, Yamamoto M, Kitamura Y. Regulation of mouse mast cell protease 6 gene expression by transcription factor encoded by the mi locus. Blood. 1996; 88: 2488–94.
  1. Kanakura Y, Furitsu T, Tsujimura T, Butterfield JH, Ashman LK, Ikeda H, Kitayama H, Kanayama Y, Matsuzawa Y, Kitamura Y. Activating mutations of the c-kit proto-oncogene in a human mast cell leukemia cell line. Leukemia. 1994; 8 Suppl 1: S18–S22.
  1. Nagata H, Worobec AS, Oh CK, Chowdhury BA, Tannenbaum S, Suzuki Y, Metcalfe DD. Identification of a point mutation in the catalytic domain of the protooncogene c-kit in peripheral blood mononuclear cells of patients who have mastocytosis with an associated hematologic disorder. Proc Natl Acad Sci USA. 1995; 92: 10560–64.

  1. 16 Friend DS, Ghildyal N, Gurish MF, Hunt J, Hu X, Austen KF, Stevens RL. Reversible expression of tryptases and chymases in the jejunal mast cells of mice infected with Trichinella spiralis. J Immunol. 1998; 160: 5537–45.
  1. Friend DS, Gurish MF, Austen KF, Hunt J, Stevens RL. Senescent jejunal mast cells and eosinophils in the mouse preferentially translocate to the spleen and draining lymph node, respectively, during the recovery phase of helminth infection. J Immunol. 2000; 165: 344–352.
  1. Miller HR, Huntley JF, Newlands GF, Mackellar A, Lammas DA, Wakelin D. Granule proteinases define mast cell heterogeneity in the serosa and the gastrointestinal mucosa of the mouse. Immunology. 1988; 65: 559–66.
  1. Wells PD. Mast cell, eosinophil, and histamine levels in Nippostrongylus brasiliensis-infected rats. Exp Parasitol. 1962; 12: 82–101.
  1. Miller HR, Jarrett WF. Immune reactions in mucous membranes: intestinal mast cell response during helminth expulsion in the rat. Immunology. 1971; 20: 277–88.
  1. 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; 192: 1849–56.
  1. Murakami M, Matsumoto R, Urade Y, Austen KF, Arm JP. c-kit ligand mediates increased expression of cytosolic phospholipase A2, prostaglandin endoperoxide synthase-1, and hematopoietic prostaglandin D2 synthase and increased IgE-dependent prostaglandin D2 generation in immature mouse mast cells. J Biol Chem. 1995; 270: 3239–46.
  1. Murakami M, Penrose JF, Urade Y, Austen KF, Arm JP. Interleukin 4 suppresses c-kit ligand-induced expression of cytosolic phospholipase A2 and prostaglandin endoperoxide synthase 2 and their roles in separate pathways of eicosanoid synthesis in mouse bone marrow-derived mast cells. Proc Natl Acad Sci USA. 1995; 92: 6107–11.
  1. Metzger H. The high affinity receptor for IgE on mast cells. Clin Exp Allergy. 1991; 21: 269–79.
  1. Leung DY, Harbeck R, Bina P, Reiser RF, Yang E, Norris DA, Hanifin JM, Sampson HA. Presence of IgE antibodies to staphylococcal exotoxins on the skin of patients with atopic dermatitis: evidence for a new group of allergens. J Clin Invest. 1993; 92: 1374–80.
  1. Katz HR, Arm JP, Benson AC, Austen KF. Maturation-related changes in the expression of FcγRII and FcγRIII on mouse mast cells derived in vitro and in vivo. J Immunol. 1990; 145: 3412–17.
  1. Okayama Y, Hagaman DD, Woolhiser M, Metcalfe DD. Further characterization of FcγRII and FcγRIII expression by cultured human mast cells. Int Arch Allergy Immunol. 2001; 124: 155–57.
  1. Malbec O, Daeron, M. The mast cell IgG receptors and their roles in tissue inflammation. Immunol Rev. 2007; 217: 206–21.
  1. Sylvestre DL, Ravetch, JV. Dominant role for mast cell Fc receptors in the Arthus reaction. Immunity. 1996; 5: 387–90.
  1. Leighty L, Li N, Diaz LA, Liu Z. Experimental models for the autoimmune and inflammatory blistering disease, Bullous pemphigoid. Arch Dermatol Res. 2007; 299: 417–22.
  1. Nonaka M, Kimura, A. Genomic view of the evolution of the complement system. Immunogenetics. 2006; 58: 701–13.
  1. Echtenacher B, Männel DN, Hültner L. Critical protective role of mast cells in a model of acute septic peritonitis. Nature. 1996; 381: 75–77.

  1. 17 Prodeus AP, Zhou X, Maurer M, Galli SJ, Carroll MC. Impaired mast cell-dependent natural immunity in complement C3-deficient mice. Nature. 1997; 390: 172–75.
  1. Malaviya R, Ross E, Jakschik BA, Abraham SN. Mast cell degranulation induced by type 1 fimbriated Escherichia coli in mice. J Clin Invest. 1994; 93: 1645–53.
  1. Malaviya R, Mast cell phagocytosis of FimH-expressing enterobacteria. J Immunol. 1994; 152: 1907–14.
  1. McCurdy JD, Lin TJ, Marshall JS. Toll-like receptor 4-mediated activation of murine mast cells. J Leukoc Biol. 2001; 70: 977–84.
  1. McCurdy JD, Olynych TJ, Maher LH, Marshall JS. Cutting edge: distinct Toll-like receptor 2 activators selectively induce different classes of mediator production from human mast cells. J Immunol. 2003; 170: 1625–29.
  1. Varadaradjalou S, Feger F, Thieblemont N, Hamouda NB, Pleau JM, Dy M, Arock M. Toll-like receptor 2 (TLR2) and TLR4 differentially activate human mast cells. Eur J Immunol. 2003; 33: 899–906.
  1. He SH. Key role of mast cells and their major secretory products in inflammatory bowel disease. World J Gastroenterol. 2004; 10: 309–18.
  1. Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, Britton H, Moran T, Karaliuskas R, Duerr RH, Achkar JP, Brant SR, Bayless TM, Kirschner BS, Hanauer SB, Nunez G, Cho JH. A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature. 2001; 411: 603–06.
  1. Hugot JP, Chamaillard M, Zouali H, Lesage S, Cezard JP, Belaiche J, Almer S, Tysk C, O'Morain CA, Gassull M, Binder V, Finkel Y, Cortot A, Modigliani R, Laurent-Puig P, Gower-Rousseau C, Macry J, Colombel JF, Sahbatou M, Thomas G. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature. 2001; 411: 599–603.
  1. Razin E, Marx, G. Thrombin-induced degranulation of cultured bone marrow-derived mast cells. J Immunol. 1984; 133: 3282–85.
  1. D'Andrea MR, Rogahn CJ, Andrade-Gordon P. Localization of protease-activated receptors-1 and −2 in human mast cells: indications for an amplified mast cell degranulation cascade. Biotech Histochem. 2000; 75: 85–90.
  1. He SH, Xie H, Fu YL. Activation of human tonsil and skin mast cells by agonists of proteinase activated receptor-2. Acta Pharmacol Sin. 2005; 26: 568–74.
  1. Moormann C, Artuc M, Pohl E, Varga G, Buddenkotte J, Vergnolle N, Brehler R, Henz BM, Schneider SW, Luger TA, Steinhoff M. Functional characterization and expression analysis of the proteinase-activated receptor-2 in human cutaneous mast cells. J Invest Dermatol. 2006; 126: 746–55.
  1. Gordon JR, Zhang X, Stevenson K, Cosford K. Thrombin induces IL-6 but not TNF-α secretion by mouse mast cells: threshold-level thrombin receptor and very low level Fc∊RI signaling synergistically enhance IL-6 secretion. Cell Immunol. 2000; 205: 128–35.
  1. Sun G, Stacey MA, Schmidt M, Mori L, Mattoli S. Interaction of mite allergens Der p3 and Der p9 with protease-activated receptor-2 expressed by lung epithelial cells. J Immunol. 2001; 167: 1014–21.
  1. Malaviya R, Ikeda T, Ross E, Abraham SN. Mast cell modulation of neutrophil influx and bacterial clearance at sites of infection through TNF-α. Nature. 1996; 381: 77–80.
  1. Nakano T, Sonoda T, Hayashi C, Yamatodani A, Kanayama Y, Yamamura T, Asai H, Yonezawa T, Kitamura Y, Galli SJ. Fate of bone marrow-derived cultured mast cells after intracutaneous, intraperitoneal, and intravenous transfer into genetically mast cell-deficient W/Wv mice: evidence that cultured mast cells can give rise to both connective tissue and mucosal mast cells. 18 J Exp Med. 1985; 162: 1025–43.
  1. Otsu K, Nakano T, Kanakura Y, Asai H, Katz HR, Austen KF, Stevens RL, Galli SJ, Kitamura Y. Phenotypic changes of bone marrow-derived mast cells after intraperitoneal transfer into W/Wv mice that are genetically deficient in mast cells. J Exp Med. 1987; 165: 615–27.
  1. Mencia-Huerta JM, Razin E, Ringel EW, Corey EJ, Hoover D, Austen KF, Lewis RA. Immunologic and ionophore-induced generation of leukotriene B4 from mouse bone marrow-derived mast cells. J Immunol. 1983; 130: 1885–90.
  1. Ford-Hutchinson AW. The role of leukotriene B4 as a mediator of leucocyte function. Agents Actions Suppl. 1983; 12: 154–66.
  1. Lewis RA, Soter NA, Diamond PT, Austen KF, Oates JA; Roberts LJ 2nd. Prostaglandin D2 generation after activation of rat and human mast cells with anti-IgE. J Immunol. 1982; 129: 1627–31.
  1. Hori Y, Nihei Y, Kurokawa Y, Kuramasu A, Makabe-Kobayashi Y, Terui T, Doi H, Satomi S, Sakurai E, Nagy A, Watanabe T, Ohtsu H. Accelerated clearance of Escherichia coli in experimental peritonitis of histamine-deficient mice. J Immunol. 2002; 169: 1978–83.
  1. Serafin WE, Sullivan TP, Conder GA, Ebrahimi A, Marcham P, Jozhnson SS, Austen KF, Reynolds DS. Cloning of the cDNA and gene for mouse mast cell protease 4: demonstration of its late transcription in mast cell subclasses and analysis of its homology to subclass-specific neutral proteases of the mouse and rat. J Biol Chem. 1991; 266: 1934–41.
  1. McNeil HP, Austen KF, Somerville LL, Gurish MF, Stevens RL. Molecular cloning of the mouse mast cell protease 5 gene: a novel secretory granule protease expressed early in the differentiation of serosal mast cells. J Biol Chem. 1991; 266: 20316–22.
  1. 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; 276: 18551–56.
  1. Mallen-St Clair J, Pham CT, Villalta SA, Caughey GH, Wolters PJ. Mast cell dipeptidyl peptidase I mediates survival from sepsis. J Clin Invest. 2004; 113: 628–34.
  1. Young JD, Liu CC, Butler G, Cohn ZA, Galli SJ. Identification, purification, and characterization of a mast cell-associated cytolytic factor related to tumor necrosis factor-α. Proc Natl Acad Sci USA. 1987; 84: 9175–79.
  1. Richards AL, Okuno T, Takagaki Y, Djeu JY. Natural cytotoxic cell-specific cytotoxic factor produced by IL-3- dependent basophilic/mast cells: relationship to TNF-α. J Immunol. 1988; 141: 3061–66.
  1. Gurish MF, Ghildyal N, Arm J, Austen KF, Avraham S, Reynolds D, Stevens RL. Cytokine mRNA are preferentially increased relative to secretory granule protein mRNA in mouse bone marrow-derived mast cells that have undergone IgE-mediated activation and degranulation. J Immunol. 1991; 146: 1527–33.
  1. Maurer M, Echtenacher B, Hültner L, Kollias G, Männel DN, Langley KE, Galli SJ. The c-kit ligand, stem cell factor, can enhance innate immunity through effects on mast cells. J Exp Med. 1998; 188: 2343–48.
  1. 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; 39: 13068–77.

  1. 19 Oh SW, Pae CI, Lee DK, Jones F, Chiang GK, Kim HO, Moon SH, Cao B, Ogbu C, Jeong KW, Kozu G, Nakanishi H, Kahn M, Chi EY, Henderson JR. WR Tryptase inhibition blocks airway inflammation in a mouse asthma model. J Immunol. 2002; 168: 1992–2000.
  1. Jippo T, Morii E, Ito A, Kitamura Y. Effect of anatomical distribution of mast cells on their defense function against bacterial infections: demonstration using partially mast cell-deficient tg/tg mice. J Exp Med. 2003; 197: 1417–25.
  1. Thakurdas SM, Melicoff E, Sansores-Garcia L, Moreira DC, Petrova Y, Stevens RL, Adachi R. The mast cell-restricted tryptase mMCP-6 has a critical immunoprotective role in bacterial infections. J Biol Chem. 2007; 207: 20809–15.
  1. Li Y, Li L, Wadley R, Reddel SW, Qi JC, Archis C, Collins A, Clark E, Cooley M, Kouts S, Naif HM, Alali M, Cunningham A, Wong GW, Stevens RL, Krilis SA. Mast cells/basophils in the peripheral blood of allergic individuals who are HIV-1 susceptible due to their surface expression of CD4 and the chemokine receptors CCR3, CCR5, and CXCR4. Blood. 2001; 97: 3484–90.
  1. Bannert N, Farzan M, Friend DS, Ochi H, Price KS, Sodroski J, Boyce JA. Human mast cell progenitors can be infected by macrophagotropic human immunodeficiency virus type 1 and retain virus with maturation in vitro. J Virol. 2001; 75: 10808–14.
  1. Taub DD, Mikovits JA, Nilsson G, Schaffer EM, Key ML, Petrow-Sadowski C, Ruscetti FW. Alterations in mast cell function and survival following in vitro infection with human immunodeficiency viruses-1 through CXCR4. Cell Immunol. 2004; 230: 65–80.
  1. Irani AM, Craig SS, De Blois G, Elson CO, Schechter NM, Schwartz LB. Deficiency of the tryptase+, chymase mast cell type in gastrointestinal mucosa of patients with defective T lymphocyte function. J Immunol. 1987; 138: 4381–86.