ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Mast cells: Development, identification, and physiologic roles

Mast cells: Development, identification, and physiologic roles
Literature review current through: Jan 2024.
This topic last updated: Feb 28, 2023.

INTRODUCTION — Mast cells are found in varying numbers in practically all tissues. They are positioned as sentinels at the body's portals of entry within mucosal membranes lining the respiratory, digestive, and urogenital systems, throughout the dermis, and surrounding blood vessels [1-3]. They are found in invertebrates as well as lower vertebrates, suggesting that they serve a fundamental role within the immune system [4,5].

Mast cells have long been recognized for their participation in allergic disease, including asthma, rhinitis, conjunctivitis, atopic dermatitis, urticaria, and anaphylaxis. However, they also have a variety of protective and regenerative roles throughout the body.

The anatomic location, development, cellular identification, and physiologic roles of mast cells are discussed in this topic review. Their activating and inhibitory surface receptors, signal transduction, and mediators are reviewed separately. (See "Mast cells: Surface receptors and signal transduction" and "Mast cell-derived mediators".)

The contributions of mast cells to specific atopic diseases and mastocytosis are discussed elsewhere. (See "Pathogenesis of asthma" and "Mastocytosis (cutaneous and systemic) in adults: Epidemiology, pathogenesis, clinical manifestations, and diagnosis".)

ANATOMIC LOCATIONS — Mast cells reside within the connective tissue of a variety of tissues and all vascularized organs. Their numbers and densities are highest at interfaces between the internal and external environments where they can respond to foreign organisms and antigens, providing a sentinel function [6]. Sites include the dermis, skeletal muscle, oral mucosa, gut mucosa and submucosa, conjunctiva, pulmonary alveoli and airways, and the atrial appendage of the heart [6-14]. Dermal mast cells are often located in close proximity to blood vessels, nerves, and lymphatics, with an estimated density of 7000 to 20,000 mast cells per cubic millimeter of skin [9,15]. From this strategic location, they can influence the function of vascular structures, monitor the blood for inflammatory and infectious changes, capture circulating immunoglobulin E (IgE) molecules onto their surface, and distribute their mediators throughout the body [16,17]. Mast cells are found in the choroid plexus of the brain and in the vascular bed of the meninges. Low numbers of mast cells are found in the kidneys and bone marrow.

Mast cell numbers increase at inflammatory sites in different diseases, including asthma, nasal polyposis, allergic rhinitis, eosinophilic esophagitis, psoriasis, rheumatoid synovitis, inflammatory bowel diseases, pulmonary fibrosis, cancer, atherosclerotic cardiovascular diseases, and cardiomyopathy, although it is not clear if their role in these diseases is pathogenic, regenerative, or both [18-29]. (See 'Physiologic roles' below.)

Mast cell numbers also increase dramatically at multiple anatomic sites in the rare condition known as mastocytosis and are directly responsible for the pathologic changes observed in this disorder. (See "Mastocytosis (cutaneous and systemic) in adults: Epidemiology, pathogenesis, clinical manifestations, and diagnosis".)

COLLECTION AND CULTURE — Mast cells are challenging to collect and study [30]. Mast cell progenitors (MCps) pass transiently through the circulation, at which point they are difficult to distinguish from other cell types. A rare circulating committed MCp that is expanded in those with severe asthma has been identified in humans; however, this cell has very limited proliferative capacity, and its relationship to mature tissue resident mast cells is not yet clear [31]. In systemic mastocytosis where mast cells carry the D816V KIT mutation, early MCps were identified in peripheral blood [32].

For studies on mast cell function, mature human mast cells can be isolated from solid tissues and cultured through a relatively complicated series of enzymatic and other techniques. Alternatively, transformed human mast cell lines can be used, although the cellular functions of these cells may be abnormal. Uncommitted progenitor cells can be collected from human cord blood or peripheral blood and developed in culture, although the resultant cells differ significantly from natural tissue mast cells.

Because of the difficulties in collecting human mast cells, mast cells from mice are often utilized instead. However, the distribution and phenotype of murine mast cells are substantively different from that of human cells [30]. Mice have much lower numbers of mature mast cells in their tissues but large numbers in the peritoneal cavity, whereas humans normally do not have significant numbers of peritoneal mast cells. Mouse mast cell precursors can be obtained from bone marrow and cultured to relative maturity with the same issues regarding the resultant cells that are present with human cell cultures.

DEVELOPMENT — Mast cells arise from pluripotential CD34+ stem cells that reside in the bone marrow and spleen [33-36]. They differentiate from the common myeloid progenitor, sharing intermediates with the megakaryocyte/erythrocyte progenitor, and are closely linked to basophils and eosinophils [37,38]. When mast cell progenitors (MCps) leave the hematopoietic tissues, some are committed to a mast cell lineage [31], and others appear to have multilineage capabilities [35,39,40]. They are then influenced by various signals (only some of which have been characterized), which direct their migration out of the circulation and into tissues where they subsequently mature [41,42]. They display considerable variability both within and across tissues compared with other cell types [43].

Kit and stem cell factor — An important growth factor for mast cells is stem cell factor (SCF), which is constitutively produced in both a soluble and membrane-bound form by stromal cells, particularly endothelial cells and fibroblasts [44-46]. SCF is also called kit ligand (KL) or steel factor [44,47-50]. The surface receptor for SCF, the receptor tyrosine kinase kit (CD117) generated from the gene KIT, is expressed by hematopoietic stem cells and is maintained during myeloid differentiation. It is thus an early marker for mast cell precursors. Mast cells continue to express kit throughout the lifetime of the cell, unlike most other cells (including basophils), which lose this marker during their development and become unresponsive to SCF [1,35,51].

SCF and kit were previously believed to be critical for the development of mast cells, and this seems true for the appearance of the mature, tissue-resident mast cell. However, the maturation of mast cell precursors to pre-mast cells may not be totally dependent on kit, as evidenced by the survival of circulating mast cell progenitors in patients undergoing therapy with imatinib, a drug that inhibits signaling through kit and other tyrosine kinases [52]. This interpretation was supported by in vitro developmental studies and is consistent with development of immature but not mature cells in mice in the absence of SCF-kit signaling. Note that imatinib does not inhibit mutated KIT, in particular at the D816V position, and early expression of KIT on pre-mast cells may not be accessible to imatinib.

With the above noted limited exception, SCF has been shown in vitro to participate in nearly every stage of growth and differentiation of mast cells, including chemotaxis, adhesion, survival, proliferation, and differentiation [53]. This global influence, combined with the constitutive and widespread production of SCF, results in the ubiquitous presence of mast cells throughout the tissues [33].

Other growth and differentiation factors — Numerous other factors (in addition to SCF) have been shown to influence proliferation and/or differentiation of mast cells. Both human and mouse MCps respond in vitro to several of the T helper type 2 (Th2) cell cytokines, including interleukin (IL) 3, IL-4, IL-5, IL-6, IL-9, and IL-10, although responses to these cytokines are better studied for mouse mast cells than for human mast cells [1]. Defects in the gene for phosphatase and tensin homolog (PTEN) can also result in hyperproliferation and heightened allergic responses in mice, although the prevalence of these defects in humans has not been extensively studied [54].

Migration into tissues — The exit of mast cell precursors from the circulation and the accumulation of differentiated mast cells in various tissues depends upon the interaction with endothelial cells initially and then with various tissue factors via adhesion molecules, including integrins and immunoglobulin supergene family receptors [55-58]. As an example, murine bone marrow-derived mast cells (BMMCs) exposed to the matrix protein, vitronectin, will spontaneously adhere and increase their proliferation [59]. A more general discussion of leukocyte movement and trafficking can be found elsewhere. (See "Leukocyte-adhesion deficiency" and "Leukocyte-endothelial adhesion in the pathogenesis of inflammation".)

Based principally on studies in the mouse with some limited support from human investigations, homing of MCps is highly organ specific [56,60,61]. In the mouse, MCp expression of the integrin alpha-4-beta-7 directs migration of MCps into normal intestine and inflamed lung. Alpha-4-beta-7 recognizes mucosal addressin cell adhesion molecule 1 (MAdCAM-1) and vascular cell adhesion molecule 1 (VCAM-1, CD106), both of which are present on normal vascular endothelium in Peyer's patches and lamina propria, while only VCAM-1 is found on vascular epithelium in the lung. The expression of the IL-8 receptor on both MCps and the vascular endothelium directs the specific migration of the precursors to the intestinal mucosa [56,57]. These processes appear to occur constitutively, with MCps demonstrating a tissue half-life in the intestine of approximately one week. These mechanisms result in the accumulation of a large pool of MCps, especially after inflammation, that is poised to mature into effector mast cells [58,62].

In contrast to the intestine, the submucosa of the mouse lung contains few mast cells and MCps in the absence of inflammation. With allergic inflammation, a rapid (within three days) and large (>25-fold) influx of MCps occurs, directed by alpha-4-beta-1 and alpha-4-beta-7 integrins on the MCps and by VCAM-1 and CXC chemokine receptor 2 (CXCR2) on the endothelium [58,63]. Studies indicate that this large pool of MCps gives rise to the mast cells that are found associated with the bronchial epithelium in the mouse and in association with both bronchial epithelium and smooth muscle in the human with chronic inflammation, such as asthma [63]. Studies in the mouse suggest a tissue-dependent variable lifespan for these inflammation-induced mature cells of anywhere from one week to several months [64].

Chemoattractants — Mast cell chemotaxis has been studied predominantly in vitro and occurs in response to a variety of growth and differentiation factors, such as SCF and IL-3, and to numerous chemokines. Transforming growth factor (TGF) beta is among the most potent mast cell chemotactic factors, active at femtomolar concentrations in both mouse and human [65,66]. The C-C chemokines RANTES (regulated on activation normal T expressed and secreted, CCL5) and CCL2 or monocyte chemotactic protein 1 (MCP-1) cause directed migration of cultured human mast cells and mouse BMMCs on vitronectin and laminin [67,68]. Also, CCL3 or macrophage inflammatory protein 1 (MIP-1) alpha and CXCL4 or platelet factor 4 (PF-4) elicit migration of mouse BMMCs after Fc-epsilon-RI activation on laminin, vitronectin, and fibronectin [67]. CXCR2 controls the chemotaxis of a human mast cell line in response to IL-8 (CXCL8) [69]. The neurotransmitter serotonin, acting through a serotonin receptor, is chemotactic for human mast cells [70]. The chemoattractant signals mediating MCp recruitment to peripheral tissue in vivo are less well understood. The murine IL-8 homologs (CXCL1 and CXCL2) acting on the single IL-8 receptor in the mouse, CXCR2, control homing of MCps to the small intestine of the mouse as noted above. CXCR2 expression by MCp is not required for recruitment to the murine lung; however, CXCR2 knockout mice exhibit impaired pulmonary MCp recruitment due to an unexpected role for this receptor in mediating VCAM-1 induction in pulmonary endothelial cells [63].

Mature phenotypes — Most investigational work on mast cells has been performed in mouse models to enable more sophisticated analyses. Using these models, subtypes of mature mast cells have been identified, which differ in location, mediator production, granule contents (and therefore staining properties), and behavior upon activation [1]. The properties of two prominent subtypes found in the mouse are presented here. It is not certain to what degree these findings apply to human mast cells. There are many similarities, but there are also differences, some of which may be due to the fact that the mice are housed in pathogen-limited environments, unlike the human situation.

In the mouse intestine, two primary forms of mature mast cells have been identified: murine mucosal mast cells (mMMCs), which appear mostly with inflammation, and murine connective tissue mast cells (mCTMCs), which are constitutive in most tissues [71]. Murine lineage tracing studies indicate that these two forms arise at different developmental time points and from different progenitor origins. Two sequential waves of progenitors arising from the yolk sac and aorta-gonado-mesonephros (AGM) develop into mCTMC, while mMMCs develop from adult bone marrow-derived progenitors [72,73]. In the small intestine, these differ most dramatically in their expression of the secretory granule proteases with proteases 1 and 2 associated with mast cells in the mucosal epithelium and proteases 4, 5, 6, 7, and the carboxypeptidase associated with mast cells in submucosal sites. However, this difference is tissue dependent as this does not hold true in the mouse lung [74]. Differences in eicosanoid synthesis have also been noted, with the mMMC producing both cysteinyl leukotrienes as well as prostanoids, while the mCTMC produces principally prostanoids upon activation [1].

Similarly, in the human, the principal division has been based upon secretory granule contents. The mast cells whose granules contain mostly tryptase (MC-T) are more prominent in the mucosa, while those containing tryptase, chymase, and carboxypeptidase A3 (MC-TC) are more prominent in the submucosal and connective tissue sites. In addition, the MC-T is the cell type depleted in human immunodeficiency virus (HIV) 1 infection, indicating that it is T cell dependent like the mMMC [75]. Thus, the MC-T and MC-TC are comparable with mMMC and mCTMC, although, due to the plasticity of this cell and differences in the proteases between the two species, this is an oversimplification, and caution is advised in drawing too strong an analogy between the murine and human systems. As in the mouse, subsequent studies have identified other tissue-dependent phenotypes, such as an intestinal mast cell containing only chymase and labeled MCc [76]. Th2-associated inflammatory diseases can also alter mast cell protease expression, with intraepithelial MC-T containing both tryptase and carboxypeptidase A3 in bronchial epithelium in asthma [77], apical epithelium in nasal polyposis [78], and esophageal epithelium during eosinophilic esophagitis [79].

Transcriptomics and proteomics studies have indicated that the MC-T/MC-TC paradigm is also somewhat of an oversimplification. Within human nasal polyps, MC-T and MC-TC are not transcriptionally discrete cell types but rather exist as two ends to a transcriptional gradient, suggesting that they may arise from a common progenitor and polarize in response to microenvironmental signals [80,81]. The nasal polyp MC-TC were also found to be highly distinct from those within the skin at the transcript level. In human lung tissue, flow cytometry-based proteomics analysis has indicated a high degree of heterogeneity across the mast cell compartment, with surface expression gradients observed for at least 10 cell surface markers, none of which exhibited a bimodal distribution [82]. Thus, while classification of mast cells based on the presence or absence of chymase can be useful under certain circumstances, it vastly underestimates the true degree of human mast cell heterogeneity.

Mucosal mast cells – MMCs in mice are found in the mucosal epithelial surfaces of the intestine and respiratory tract, typically in association with inflammation associated with Th2 responses. Intestinal infections and allergic diseases, such as allergic rhinitis and asthma, result in dramatic increases in the numbers of MMCs and, to a lesser extent, CTMCs at the involved sites [83-85].

As MMCs expand in number, they also differentiate in ways that are defined by the tissue microenvironment [86]. As an example, in murine Trichinella spiralis infection, the MMC population undergoes sequential changes in the secretory granule phenotype as it migrates through the lamina propria to an intraepithelial residence and then reverses its migration and phenotype with the resolution of infection [83]. Thus, the phenotype of these cells is dynamic and defined by the inflammatory perturbations at a tissue site, likely allowing this cell to play roles in both the induction/amplification phase and the resolution/healing phase of the response [83,86].

The development of MMCs is dependent on T cell-derived factors, such as IL-3 and IL-4, as evidenced by the selective absence of MMCs in athymic mice and studies in cytokine-deficient mice [87-90]. Similarly, patients with HIV and acquired immunodeficiency syndrome (AIDS) who have low CD4+ T cell counts are profoundly deficient in intestinal mast cells of the MC-T phenotype, illustrating the dependency of this human mast cells' phenotype on T cells [75].

Connective tissue mast cells – CTMCs in mice are found in the submucosal connective tissues of the intestine and throughout the body in association with various other connective tissues. Their maturation is T cell independent in mice as normal numbers are seen in lymphocyte-deficient strains [33]. Also, their numbers do not increase as significantly in acute reactions as the mMMC in the intestine or the lung of mice following Th2-mediated inflammations, such as in helminthic infection or atopic diseases [83].

In humans, increased numbers of mast cells are noted in association with bronchial smooth muscle in patients with asthma and with the synovium in rheumatoid arthritis, while mast cells with the MC-TC phenotype can be found in bronchial epithelium in severe asthma [84,91-93].

As mentioned previously, mast cell populations within tissues can expand severalfold during various disease states, including the following [94]:

Parasitic infection

Mastocytosis

Atopic diseases (including urticaria, eosinophilic esophagitis, acute allergic reactions, rhinitis, nasal polyposis, and asthma)

Rheumatologic disease (rheumatoid arthritis, scleroderma)

Nonparasitic infectious disease (tuberculosis and syphilis)

Neoplastic disease (melanoma and intestinal neoplasms)

Chronic diseases (chronic kidney disease, chronic liver disease, and osteoporosis)

CELLULAR IDENTIFICATION — Mast cells are normally round mononuclear cells, which exhibit phenotypic variation in cytoplasmic morphology. Cell diameter ranges up to 25 micrometers. A unilobed nucleus may be round or oval in shape and is typically eccentrically positioned.

Staining properties — The mast cell can be recognized by its numerous cytoplasmic granules, which may occupy the majority of the cell volume [6,95]. Poorly granulated mast cells (eg, immature progenitors) or mast cells that have recently degranulated if stained with Giemsa or toluidine blue will stain poorly, making them difficult to pick out in tissue sections and can cause them to be mistaken for monocytes. However, mast cells may be found using the more sensitive immunohistochemistry for tryptase.

The following staining techniques can be used to identify human mast cells:

Toluidine blue – Mast cell granules stain purple with the basic dye toluidine blue. This property of staining purple upon application of a blue dye is referred to as "metachromatic staining" and is due to the presence of highly sulfated, anionic proteoglycans (eg, heparin) complexed with the various secretory granule proteases. Metachromatic staining is unique to mast cells and basophils.

Eosinophils are usually easily differentiated from mast cells. Eosinophils stain red with Giemsa and blue with toluidine blue, and eosinophils have a bilobed nucleus. However, one of the authors (MC) has seen a case of cutaneous mastocytoma that was initially thought to be an eosinophilic granuloma [96]. Staining with monoclonal antibodies that reliably identify mast cells is critical in such situations.

Monoclonal antibodies – Monoclonal antibodies directed against the membrane receptors (kit and the high-affinity IgE receptor) and the granule proteases (tryptase, chymase, and mast cell carboxypeptidase A3) are useful in identifying mast cells [97-100]. However, the monoclonal antibody for tryptase will also stain basophils, although basophils are not normally located in the tissues [101]. Basophils contain much less tryptase, and the nuclear morphology of basophils and mast cells is different.

Fluorescently conjugated avidin – Avidin conjugated with a fluorophore, such as fluorescein isothiocyanate (FITC), can be used histologically to fluorescently label the granules of mast cells containing tryptase, chymase, and carboxypeptidase A3 (MC-TC) but not mast cells whose granules contain mostly tryptase (MC-T), likely due to interactions with heparin within the mast cell granule [76].

Electron microscopy — Mast cells examined with electron microscopy show abundant cytoplasmic granules, ranging in size from 0.3 to 0.8 micrometers (picture 1). The cytoplasmic granules of mast cells contain macromolecular complexes of proteoglycans and proteases that exhibit discrete patterns on electron microscopy. These patterns are related to the type of protease expressed (picture 2).

All granules of human mast cells contain trypsin-like proteases. In unstimulated MC-T, the secretory granules exhibit an ultrastructural pattern on electron microscopy that resembles scrolls viewed in cross-section. Histologically, the granules do not stain with safranin and will not stain after formalin fixation [53,102].

Granules containing chymase and carboxypeptidase A3 have a grating or lattice ultrastructure and are found in mast cells with the MC-TC phenotype [102,103]. Mast cells with this type of granule predominate in the skin and intestinal submucosa. The mast cell-specific carboxypeptidase A3 is also selectively found in MC-TC cells (containing tryptase, chymase, and carboxypeptidase A3) that are associated with connective tissue locations [99].

Mast cells also have numerous cytoplasmic projections called pseudopods that may interdigitate with other cells.

PHYSIOLOGIC ROLES — Primarily in mice, mast cells have been implicated in both physiologic and pathogenic processes. Mast cells are important in defense against some bacteria and viruses and contribute to defense against parasites [2-4,104,105]. They are key effector cells in both innate and acquired immunity and are capable of inducing and amplifying both types of responses [53,106]. Specifically, mast cells are capable of detecting microbial products through surface pattern recognition receptors, and they are involved in recruitment of other leukocytes, containment of bacterial infections, and tissue repair [2,53,105].

Innate immunity — Mast cells have emerged as important effector cells of the innate immune system [33,53,107]. (See "An overview of the innate immune system", section on 'Mast cells'.)

Mast cells assist in innate defense against certain bacterial (and likely also viral) infections by releasing cytokines in response to bacterial or viral products. These products include lipopolysaccharide (LPS), peptidoglycan, and single- or double-stranded nucleic acids [108-115]. Activation by microbial products is mediated through Toll-like receptors (TLRs) on the mast cell surface [109,110] and also involves complement activation that may amplify mast cell activation [116]. (See "Toll-like receptors: Roles in disease and therapy".)

A G-protein-coupled receptor has been described in mice and human mast cells, Mrgprb2/Mas-related G protein-coupled receptor X2 (MRGPRX2), which is responsible for secretagogue-induced histamine release and non-IgE-mediated reactions to quinolones, Hymenoptera mastoparan, and general anesthetics [117]. This receptor also mediates mast cell activation by the neuropeptide substance p, central to the development of experimental atopic dermatitis in mice, and pro-adrenomedullin peptide 9-20, regulating nonhistaminergic itch [118,119]. In humans, MRGPRX2 is highly expressed on skin MC-TC but is absent from MC-TC within nasal polyp tissue.

In mice, mast cells have important antibacterial responses. Mast cell-deficient mice succumb to experimentally induced peritonitis, whereas normal mice can survive [3]. Reconstitution of the peritoneal mast cells in the mast cell-deficient mice restores the ability of these mice to withstand this insult. Humans do not have peritoneal mast cells, and it is unclear if there are clinical correlates in human disease.

Mast cells may phagocytose bacteria, such as Salmonella typhi, under some circumstances [112-114].

Mast cells degranulate upon exposure to some viruses and release various cytokines and chemotactic mediators in response to others, again suggesting a role in both early defense and recruitment of other effector cells to sites of infection [115,120-122].

Mast cells produce antimicrobial peptides, such as cathelicidins, and can amplify complement activation, resulting in disruption of microbial membranes and lysis [123]. These may be important in defense against common bacteria, such as streptococci.

The proteases synthesized and released by mast cells are able to degrade and thus protect against some toxins. This was demonstrated in a study in which normal and mast cell-deficient mice were injected with snake and honey bee venoms [124,125]. Mast cell-deficient mice were 10-fold more sensitive to the toxins' effects, and this was ameliorated by restoration of mast cells. Similar studies have been performed with venoms from scorpions and Gila monsters [126].

Reperfusion injury results in an inflammatory response mediated by immunoglobulin M (IgM), complement, and mast cells. The mast cell secretory granule proteases have been implicated as critical mediators in this reaction [127].

Mast cell-derived cytokines are crucial to the early and effective recruitment of neutrophils and other leukocytes to sites of infection [111,128,129]. Specifically, mast cell-derived tumor necrosis factor (TNF) has been shown to be involved in neutrophil recruitment to sites of inflammation, as well as in dendritic cell migration to lymph nodes. The mast cell secretory granule proteases (tryptases specifically) can recruit both eosinophils and neutrophils [104,105,129]. The actions of these and other mast cell mediators are discussed separately. (See "Mast cell-derived mediators".)

Acquired immunity — Mast cells are thought to be important in defense against parasitic infections, although the mechanisms involved are not fully elucidated [87,88,130-133]. IgE antibody plays a role in the expulsion of some intestinal parasites in experimental animals. However, it does not account fully for the mast cell response [130,132,133].

Mast cell degranulation may also affect other cell types in ways that promote the development of a T helper type 2 (Th2) acquired immune response. In mice, stimuli that result in mast cell degranulation also enhance interleukin (IL) 4 generation by dendritic cells and suppress interferon (IFN) gamma production by T cells [134].

Other physiologic functions — Mast cells have been implicated in various physiologic and regenerative processes.

The role of mast cells in triggering increased intestinal motility, bronchoconstriction, and epithelial sloughing may have protective effects in ridding the body's epithelial surfaces of invaders and infectious agents, although these responses are more commonly viewed as manifestations of allergic reactions [2,135].

Mast cells stimulate fibroblast proliferation and collagen synthesis and are believed to play a role in "walling off" and limiting the spread of infections, as well as in wound healing and scar formation [136-139].

The number of mast cells normally increases dramatically within uterine tissues during pregnancy [140]. It is proposed that this influx of mast cells may contribute to a strengthening of local immunity through the secretion of cytokines and unique proteases based upon the mouse [141], as well as influence the contractility of myometrial cells.

Mast cells, together with T regulatory cells, may mediate allograft tolerance in mice and control inflammation in response to premalignant events in the intestine, suggesting a role for mast cells in immune surveillance [142,143].

Mast cells release carboxypeptidase A3, which may be important in the degradation of angiotensin and generation of angiotensin II [144,145].

Mast cell protease cleavage of the cytokine IL-33 significantly increases its bioactivity, which may be important for in vivo signal amplification [146].

Mast cell chymase has been implicated in blood pressure and intestinal homeostasis. Mice lacking the human mast cell chymase homolog mMCP4 exhibited decreased cleavage of big endothelin-1 to endothelin-1 (1-31) in the blood and in the intestine, decreased basal permeability, and altered epithelial cell migrations and villus morphology [147,148].

Potentially detrimental functions

Allergic diseases – Mast cells play a prominent and in some cases, a primary role in allergic responses, including anaphylaxis, urticaria, asthma, rhinitis, and atopic dermatitis [28,149-151]. (See 'Mast cells in allergy' below.)

Mastocytosis – Mastocytosis is due to a mutation in KIT, resulting in chronic activation and clonal expansion of mast cells. The disorder may affect multiple organ systems or be confined to the skin. (See "Mastocytosis (cutaneous and systemic) in adults: Epidemiology, pathogenesis, clinical manifestations, and diagnosis".)

Atherosclerosis – Increased mast cell numbers have been noted for decades within human atherosclerotic lesions, although their functions within plaques were unclear [152]. Mouse models suggest that mast cells promote atherosclerotic changes through the release of proinflammatory cytokines and perhaps proteases [153,154].

Autoimmune disease – Experiments employing a number of models of autoimmunity, including multiple sclerosis and rheumatoid arthritis, have implicated mast cells. In both instances, mast cell-deficient mice showed reduced pathology that was reversed upon reconstitution of the mast cells in the mice [155,156]. (See "Synovial pathology in rheumatoid arthritis".)

Burn lesions – Mast cells in the skin are rapidly activated following thermal trauma, and the release of the proteases is critical to the development of the inflammatory response that exacerbates the damage caused by first- and second-degree burns [157].

Mast cells in allergy — Mast cells play a central role in the response to allergen challenge. The activation of mast cells results in both an early and a delayed phase of inflammation. The various chemical mediators released from mast cells during an allergic reaction are discussed separately. (See "Mast cell-derived mediators".)

Early-phase response — Following activation of cutaneous mast cells, the early phase of the response occurs within 10 minutes and resolves within one hour. Cutaneous allergen challenge in sensitized subjects (ie, those with IgE specific to the allergen) elicits an early-phase response characterized by a "wheal-and-flare." The "wheal" is a central area of pruritic skin edema, caused predominantly by histamine-mediated increased vasopermeability [158]. The "flare" is erythema due to local vasodilation, which spreads out from the central area due to an axonal reflex, likely mediated by neuropeptides. Histology demonstrates dermal edema, characterized by separation of collagen bundles and degranulated mast cells, without an inflammatory infiltrate. The early phase of the response may be even more rapid (ie, seconds) in mucosal mast cells (MMCs).

Late-phase response — The early-phase response is followed in some patients by a delayed phase, which involves cutaneous erythema, warmth, and induration. This phase begins at six to eight hours and resolves within 24 hours. Histology at this point demonstrates dermal edema with the influx of mononuclear cells, eosinophils, basophils, and neutrophils [159]. A similar biphasic clinical and histologic process occurs in the airways of some patients with asthma when challenged with specific antigen inhalation [151].

SUMMARY

Anatomic locations – Mast cells are positioned as sentinels at sites that pathogens might invade the body (the dermis, gut mucosa and submucosa, conjunctiva, pulmonary alveoli, and airways). Dermal mast cells are often located in close proximity to blood vessels, nerves, and lymphatics. (See 'Anatomic locations' above.)

Collection and culture – Human mast cells are difficult to collect and study. Immature forms pass transiently through the circulation and are difficult to distinguish from other cell types. Progenitors develop into mature mast cells in the tissues, which do not recirculate through the blood. For these and other technical reasons, mouse mast cells are often used in research, but findings from studies of mouse mast cells do not always apply to humans. (See 'Collection and culture' above.)

Development – Mast cells arise from pluripotential CD34+ stem cells originating in the bone marrow and spleen and differentiate along the myeloid pathway. Stem cell factor (SCF or kit ligand [KL]) produced by stromal cells is critical for almost every stage of mast cell development and survival. The surface receptor for SCF is kit (CD117), which is expressed by many hematopoietic stem cells early in development but is later lost by all cell types except mast cells. Only mast cells remain responsive to SCF. (See 'Development' above.)

Identification and staining – The mast cell can be recognized by its numerous cytoplasmic granules, which may occupy the majority of the cell volume. Mast cell granules stain purple with the basic dye toluidine blue, a property referred to as metachromatic staining. Monoclonal antibodies directed against the membrane receptors kit, the high-affinity immunoglobulin E (IgE) receptor, and granule proteases (tryptase, chymase, and mast cell carboxypeptidase A3) are useful in identifying mast cells. (See 'Cellular identification' above.)

Roles in innate immunity – Mast cells participate in innate defense against certain bacterial and viral infections by releasing cytokines in response to lipopolysaccharide (LPS), peptidoglycan, and nucleic acids, which are recognized through Toll-like receptors (TLRs) on the mast cell surface. In response to these microbial products, mast cells secrete cytokines, activate complement, and recruit other effector cells to the site of invasion. (See 'Physiologic roles' above.)

Roles in disease and health – Mast cells are critical effector cells in allergic responses, including anaphylaxis, urticaria, asthma, rhinitis, and atopic dermatitis. Mast cell mediators trigger increased intestinal motility, bronchoconstriction, and epithelial sloughing. These responses may have protective effects by ridding the body's epithelial surfaces of invaders and infectious agents when properly controlled. However, in excess, these responses are manifestations of allergic reactions. (See 'Physiologic roles' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Michael Gurish, PhD, who contributed to earlier versions of this topic review.

  1. Boyce JA. The biology of the mast cell. Allergy Asthma Proc 2004; 25:27.
  2. Marshall JS, Jawdat DM. Mast cells in innate immunity. J Allergy Clin Immunol 2004; 114:21.
  3. Echtenacher B, Männel DN, Hültner L. Critical protective role of mast cells in a model of acute septic peritonitis. Nature 1996; 381:75.
  4. Silphaduang U, Noga EJ. Peptide antibiotics in mast cells of fish. Nature 2001; 414:268.
  5. de Barros CM, Andrade LR, Allodi S, et al. The Hemolymph of the ascidian Styela plicata (Chordata-Tunicata) contains heparin inside basophil-like cells and a unique sulfated galactoglucan in the plasma. J Biol Chem 2007; 282:1615.
  6. Dvorak AM. The fine structure of human basophils and mast cells. In: Mast cells, mediators, and disease, Holgate ST (Ed), Kluwer Academic Publishers, London 1988.
  7. Humphries DE, Wong GW, Friend DS, et al. Heparin is essential for the storage of specific granule proteases in mast cells. Nature 1999; 400:769.
  8. WISLOCKI GB, SOGNNAES RF. Histochemical reactions of normal teeth. Am J Anat 1950; 87:239.
  9. Eady RA, Cowen T, Marshall TF, et al. Mast cell population density, blood vessel density and histamine content in normal human skin. Br J Dermatol 1979; 100:623.
  10. Strobel S, Miller HR, Ferguson A. Human intestinal mucosal mast cells: evaluation of fixation and staining techniques. J Clin Pathol 1981; 34:851.
  11. Irani AM, Butrus SI, Tabbara KF, Schwartz LB. Human conjunctival mast cells: distribution of MCT and MCTC in vernal conjunctivitis and giant papillary conjunctivitis. J Allergy Clin Immunol 1990; 86:34.
  12. Baddeley SM, Bacon AS, McGill JI, et al. Mast cell distribution and neutral protease expression in acute and chronic allergic conjunctivitis. Clin Exp Allergy 1995; 25:41.
  13. Schulman ES, MacGlashan DW Jr, Peters SP, et al. Human lung mast cells: purification and characterization. J Immunol 1982; 129:2662.
  14. Sperr WR, Bankl HC, Mundigler G, et al. The human cardiac mast cell: localization, isolation, phenotype, and functional characterization. Blood 1994; 84:3876.
  15. Cowen T, Trigg P, Eady RA. Distribution of mast cells in human dermis: development of a mapping technique. Br J Dermatol 1979; 100:635.
  16. Kunder CA, St John AL, Abraham SN. Mast cell modulation of the vascular and lymphatic endothelium. Blood 2011; 118:5383.
  17. Cheng LE, Hartmann K, Roers A, et al. Perivascular mast cells dynamically probe cutaneous blood vessels to capture immunoglobulin E. Immunity 2013; 38:166.
  18. Kawabori S, Denburg JA, Schwartz LB, et al. Histochemical and immunohistochemical characteristics of mast cells in nasal polyps. Am J Respir Cell Mol Biol 1992; 6:37.
  19. Goodfield M, Hull SM, Holland D, et al. Investigations of the 'active' edge of plaque psoriasis: vascular proliferation precedes changes in epidermal keratin. Br J Dermatol 1994; 131:808.
  20. Naukkarinen A, Harvima IT, Aalto ML, Horsmanheimo M. Mast cell tryptase and chymase are potential regulators of neurogenic inflammation in psoriatic skin. Int J Dermatol 1994; 33:361.
  21. Harvima IT, Naukkarinen A, Paukkonen K, et al. Mast cell tryptase and chymase in developing and mature psoriatic lesions. Arch Dermatol Res 1993; 285:184.
  22. Gotis-Graham I, McNeil HP. Mast cell responses in rheumatoid synovium. Association of the MCTC subset with matrix turnover and clinical progression. Arthritis Rheum 1997; 40:479.
  23. de Paulis A, Marinò I, Ciccarelli A, et al. Human synovial mast cells. I. Ultrastructural in situ and in vitro immunologic characterization. Arthritis Rheum 1996; 39:1222.
  24. Fox CC, Lazenby AJ, Moore WC, et al. Enhancement of human intestinal mast cell mediator release in active ulcerative colitis. Gastroenterology 1990; 99:119.
  25. Nolte H, Spjeldnaes N, Kruse A, Windelborg B. Histamine release from gut mast cells from patients with inflammatory bowel diseases. Gut 1990; 31:791.
  26. Church MK, Levi-Schaffer F. The human mast cell. J Allergy Clin Immunol 1997; 99:155.
  27. Ryan JJ, Huff TF. Biology of mast cells. In: Middleton's allergy: Principles and practice, 6th ed, Adkinson NF, Yunginger JW, Busse WW, et al (Eds), Mosby, St. Louis 2003. p.333.
  28. Maurer M, Wedemeyer J, Metz M, et al. Mast cells promote homeostasis by limiting endothelin-1-induced toxicity. Nature 2004; 432:512.
  29. Levick SP, Loch DC, Taylor SM, Janicki JS. Arachidonic acid metabolism as a potential mediator of cardiac fibrosis associated with inflammation. J Immunol 2007; 178:641.
  30. Bischoff SC. Role of mast cells in allergic and non-allergic immune responses: comparison of human and murine data. Nat Rev Immunol 2007; 7:93.
  31. Dahlin JS, Malinovschi A, Öhrvik H, et al. Lin- CD34hi CD117int/hi FcεRI+ cells in human blood constitute a rare population of mast cell progenitors. Blood 2016; 127:383.
  32. Castells MC, Friend DS, Bunnell CA, et al. The presence of membrane-bound stem cell factor on highly immature nonmetachromatic mast cells in the peripheral blood of a patient with aggressive systemic mastocytosis. J Allergy Clin Immunol 1996; 98:831.
  33. Gurish MF, Boyce JA. Mast cells: ontogeny, homing, and recruitment of a unique innate effector cell. J Allergy Clin Immunol 2006; 117:1285.
  34. Kirshenbaum AS, Kessler SW, Goff JP, Metcalfe DD. Demonstration of the origin of human mast cells from CD34+ bone marrow progenitor cells. J Immunol 1991; 146:1410.
  35. Arinobu Y, Iwasaki H, Gurish MF, et al. Developmental checkpoints of the basophil/mast cell lineages in adult murine hematopoiesis. Proc Natl Acad Sci U S A 2005; 102:18105.
  36. Maaninka K, Lappalainen J, Kovanen PT. Human mast cells arise from a common circulating progenitor. J Allergy Clin Immunol 2013; 132:463.
  37. Velten L, Haas SF, Raffel S, et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat Cell Biol 2017; 19:271.
  38. Zheng S, Papalexi E, Butler A, et al. Molecular transitions in early progenitors during human cord blood hematopoiesis. Mol Syst Biol 2018; 14:e8041.
  39. Saenz SA, Siracusa MC, Perrigoue JG, et al. IL25 elicits a multipotent progenitor cell population that promotes T(H)2 cytokine responses. Nature 2010; 464:1362.
  40. Kirshenbaum AS, Goff JP, Semere T, et al. Demonstration that human mast cells arise from a progenitor cell population that is CD34(+), c-kit(+), and expresses aminopeptidase N (CD13). Blood 1999; 94:2333.
  41. Toru H, Ra C, Nonoyama S, et al. Induction of the high-affinity IgE receptor (Fc epsilon RI) on human mast cells by IL-4. Int Immunol 1996; 8:1367.
  42. Kitamura Y, Oboki K, Ito A. Molecular mechanisms of mast cell development. Immunol Allergy Clin North Am 2006; 26:387.
  43. Dwyer DF, Barrett NA, Austen KF, Immunological Genome Project Consortium. Expression profiling of constitutive mast cells reveals a unique identity within the immune system. Nat Immunol 2016; 17:878.
  44. Huang E, Nocka K, Beier DR, et al. 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.
  45. Anderson DM, Lyman SD, Baird A, et al. Molecular cloning of mast cell growth factor, a hematopoietin that is active in both membrane bound and soluble forms. Cell 1990; 63:235.
  46. Lammie A, Drobnjak M, Gerald W, et al. Expression of c-kit and kit ligand proteins in normal human tissues. J Histochem Cytochem 1994; 42:1417.
  47. Flanagan JG, Leder P. The kit ligand: a cell surface molecule altered in steel mutant fibroblasts. Cell 1990; 63:185.
  48. Zsebo KM, Williams DA, Geissler EN, et al. Stem cell factor is encoded at the Sl locus of the mouse and is the ligand for the c-kit tyrosine kinase receptor. Cell 1990; 63:213.
  49. Williams DE, Eisenman J, Baird A, et al. Identification of a ligand for the c-kit proto-oncogene. Cell 1990; 63:167.
  50. Copeland NG, Gilbert DJ, Cho BC, et al. Mast cell growth factor maps near the steel locus on mouse chromosome 10 and is deleted in a number of steel alleles. Cell 1990; 63:175.
  51. Li L, Li Y, Reddel SW, et al. Identification of basophilic cells that express mast cell granule proteases in the peripheral blood of asthma, allergy, and drug-reactive patients. J Immunol 1998; 161:5079.
  52. Dahlin JS, Ekoff M, Grootens J, et al. KIT signaling is dispensable for human mast cell progenitor development. Blood 2017; 130:1785.
  53. Boyce JA. Mast cells: beyond IgE. J Allergy Clin Immunol 2003; 111:24.
  54. Furumoto Y, Charles N, Olivera A, et al. PTEN deficiency in mast cells causes a mastocytosis-like proliferative disease that heightens allergic responses and vascular permeability. Blood 2011; 118:5466.
  55. Hamawy MM, Mergenhagen SE, Siraganian RP. Adhesion molecules as regulators of mast-cell and basophil function. Immunol Today 1994; 15:62.
  56. Gurish MF, Tao H, Abonia JP, et al. Intestinal mast cell progenitors require CD49dbeta7 (alpha4beta7 integrin) for tissue-specific homing. J Exp Med 2001; 194:1243.
  57. Abonia JP, Austen KF, Rollins BJ, et al. Constitutive homing of mast cell progenitors to the intestine depends on autologous expression of the chemokine receptor CXCR2. Blood 2005; 105:4308.
  58. Abonia JP, Hallgren J, Jones T, et al. Alpha-4 integrins and VCAM-1, but not MAdCAM-1, are essential for recruitment of mast cell progenitors to the inflamed lung. Blood 2006; 108:1588.
  59. Bianchine PJ, Burd PR, Metcalfe DD. IL-3-dependent mast cells attach to plate-bound vitronectin. Demonstration of augmented proliferation in response to signals transduced via cell surface vitronectin receptors. J Immunol 1992; 149:3665.
  60. Kitamura Y, Shimada M, Hatanaka K, Miyano Y. Development of mast cells from grafted bone marrow cells in irradiated mice. Nature 1977; 268:442.
  61. Crapper RM, Schrader JW. Frequency of mast cell precursors in normal tissues determined by an in vitro assay: antigen induces parallel increases in the frequency of P cell precursors and mast cells. J Immunol 1983; 131:923.
  62. Mwamtemi HH, Koike K, Kinoshita T, et al. An increase in circulating mast cell colony-forming cells in asthma. J Immunol 2001; 166:4672.
  63. Hallgren J, Jones TG, Abonia JP, et al. Pulmonary CXCR2 regulates VCAM-1 and antigen-induced recruitment of mast cell progenitors. Proc Natl Acad Sci U S A 2007; 104:20478.
  64. Bankova LG, Dwyer DF, Liu AY, et al. Maturation of mast cell progenitors to mucosal mast cells during allergic pulmonary inflammation in mice. Mucosal Immunol 2015; 8:596.
  65. Gruber BL, Marchese MJ, Kew RR. Transforming growth factor-beta 1 mediates mast cell chemotaxis. J Immunol 1994; 152:5860.
  66. Olsson N, Piek E, ten Dijke P, Nilsson G. Human mast cell migration in response to members of the transforming growth factor-beta family. J Leukoc Biol 2000; 67:350.
  67. Taub D, Dastych J, Inamura N, et al. Bone marrow-derived murine mast cells migrate, but do not degranulate, in response to chemokines. J Immunol 1995; 154:2393.
  68. Juremalm M, Olsson N, Nilsson G. Selective CCL5/RANTES-induced mast cell migration through interactions with chemokine receptors CCR1 and CCR4. Biochem Biophys Res Commun 2002; 297:480.
  69. Nilsson G, Mikovits JA, Metcalfe DD, Taub DD. Mast cell migratory response to interleukin-8 is mediated through interaction with chemokine receptor CXCR2/Interleukin-8RB. Blood 1999; 93:2791.
  70. Kushnir-Sukhov NM, Gilfillan AM, Coleman JW, et al. 5-hydroxytryptamine induces mast cell adhesion and migration. J Immunol 2006; 177:6422.
  71. Enerbäck L. Mast cells in rat gastrointestinal mucosa. I. Effects of fixation. Acta Pathol Microbiol Scand 1966; 66:289.
  72. Gentek R, Ghigo C, Hoeffel G, et al. Hemogenic Endothelial Fate Mapping Reveals Dual Developmental Origin of Mast Cells. Immunity 2018; 48:1160.
  73. Li Z, Liu S, Xu J, et al. Adult Connective Tissue-Resident Mast Cells Originate from Late Erythro-Myeloid Progenitors. Immunity 2018; 49:640.
  74. Xing W, Austen KF, Gurish MF, Jones TG. Protease phenotype of constitutive connective tissue and of induced mucosal mast cells in mice is regulated by the tissue. Proc Natl Acad Sci U S A 2011; 108:14210.
  75. Irani AM, Craig SS, DeBlois G, et al. Deficiency of the tryptase-positive, chymase-negative mast cell type in gastrointestinal mucosa of patients with defective T lymphocyte function. J Immunol 1987; 138:4381.
  76. Weidner N, Austen KF. Heterogeneity of mast cells at multiple body sites. Fluorescent determination of avidin binding and immunofluorescent determination of chymase, tryptase, and carboxypeptidase content. Pathol Res Pract 1993; 189:156.
  77. Dougherty RH, Sidhu SS, Raman K, et al. Accumulation of intraepithelial mast cells with a unique protease phenotype in T(H)2-high asthma. J Allergy Clin Immunol 2010; 125:1046.
  78. Takabayashi T, Kato A, Peters AT, et al. Glandular mast cells with distinct phenotype are highly elevated in chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2012; 130:410.
  79. Abonia JP, Blanchard C, Butz BB, et al. Involvement of mast cells in eosinophilic esophagitis. J Allergy Clin Immunol 2010; 126:140.
  80. Dwyer DF, Ordovas-Montanes J, Allon SJ, et al. Human airway mast cells proliferate and acquire distinct inflammation-driven phenotypes during type 2 inflammation. Sci Immunol 2021; 6.
  81. Bangert C, Villazala-Merino S, Fahrenberger M, et al. Comprehensive Analysis of Nasal Polyps Reveals a More Pronounced Type 2 Transcriptomic Profile of Epithelial Cells and Mast Cells in Aspirin-Exacerbated Respiratory Disease. Front Immunol 2022; 13:850494.
  82. Rönnberg E, Boey DZH, Ravindran A, et al. Immunoprofiling Reveals Novel Mast Cell Receptors and the Continuous Nature of Human Lung Mast Cell Heterogeneity. Front Immunol 2021; 12:804812.
  83. Friend DS, Ghildyal N, Austen KF, et al. 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.
  84. Laitinen LA, Laitinen A, Haahtela T. Airway mucosal inflammation even in patients with newly diagnosed asthma. Am Rev Respir Dis 1993; 147:697.
  85. Yu M, Tsai M, Tam SY, et al. Mast cells can promote the development of multiple features of chronic asthma in mice. J Clin Invest 2006; 116:1633.
  86. Gurish MF, Pear WS, Stevens RL, et al. Tissue-regulated differentiation and maturation of a v-abl-immortalized mast cell-committed progenitor. Immunity 1995; 3:175.
  87. Ruitenberg EJ, Elgersma A. Absence of intestinal mast cell response in congenitally athymic mice during Trichinella spiralis infection. Nature 1976; 264:258.
  88. Lantz CS, Boesiger J, Song CH, et al. Role for interleukin-3 in mast-cell and basophil development and in immunity to parasites. Nature 1998; 392:90.
  89. Madden KB, Urban JF Jr, Ziltener HJ, et al. Antibodies to IL-3 and IL-4 suppress helminth-induced intestinal mastocytosis. J Immunol 1991; 147:1387.
  90. Urban JF Jr, Schopf L, Morris SC, et al. Stat6 signaling promotes protective immunity against Trichinella spiralis through a mast cell- and T cell-dependent mechanism. J Immunol 2000; 164:2046.
  91. Brightling CE, Bradding P, Symon FA, et al. Mast-cell infiltration of airway smooth muscle in asthma. N Engl J Med 2002; 346:1699.
  92. Crisp AJ, Chapman CM, Kirkham SE, et al. Articular mastocytosis in rheumatoid arthritis. Arthritis Rheum 1984; 27:845.
  93. Balzar S, Fajt ML, Comhair SA, et al. Mast cell phenotype, location, and activation in severe asthma. Data from the Severe Asthma Research Program. Am J Respir Crit Care Med 2011; 183:299.
  94. Prussin C, Metcalfe DD. 5. IgE, mast cells, basophils, and eosinophils. J Allergy Clin Immunol 2006; 117:S450.
  95. Benditt EP, Lagunoff D. The mast cell: its structure and function. Prog Allergy 1964; 8:195.
  96. Castells MC. Extracutaneous mastocytoma. J Allergy Clin Immunol 2006; 117:1513.
  97. Lerner NB, Nocka KH, Cole SR, et al. Monoclonal antibody YB5.B8 identifies the human c-kit protein product. Blood 1991; 77:1876.
  98. Irani AM, Bradford TR, Kepley CL, et al. Detection of MCT and MCTC types of human mast cells by immunohistochemistry using new monoclonal anti-tryptase and anti-chymase antibodies. J Histochem Cytochem 1989; 37:1509.
  99. Irani AM, Goldstein SM, Wintroub BU, et al. Human mast cell carboxypeptidase. Selective localization to MCTC cells. J Immunol 1991; 147:247.
  100. Escribano L, Garcia Montero AC, Núñez R, et al. Flow cytometric analysis of normal and neoplastic mast cells: role in diagnosis and follow-up of mast cell disease. Immunol Allergy Clin North Am 2006; 26:535.
  101. Castells MC, Irani AM, Schwartz LB. Evaluation of human peripheral blood leukocytes for mast cell tryptase. J Immunol 1987; 138:2184.
  102. 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.
  103. Irani AA, Schechter NM, Craig SS, et al. Two types of human mast cells that have distinct neutral protease compositions. Proc Natl Acad Sci U S A 1986; 83:4464.
  104. Gordon JR, Galli SJ. Mast cells as a source of both preformed and immunologically inducible TNF-alpha/cachectin. Nature 1990; 346:274.
  105. Malaviya R, Ikeda T, Ross E, Abraham SN. Mast cell modulation of neutrophil influx and bacterial clearance at sites of infection through TNF-alpha. Nature 1996; 381:77.
  106. Galli SJ, Maurer M, Lantz CS. Mast cells as sentinels of innate immunity. Curr Opin Immunol 1999; 11:53.
  107. Metcalfe DD, Boyce JA. Mast cell biology in evolution. J Allergy Clin Immunol 2006; 117:1227.
  108. Leal-Berumen I, Conlon P, Marshall JS. IL-6 production by rat peritoneal mast cells is not necessarily preceded by histamine release and can be induced by bacterial lipopolysaccharide. J Immunol 1994; 152:5468.
  109. Supajatura V, Ushio H, Nakao A, et al. Protective roles of mast cells against enterobacterial infection are mediated by Toll-like receptor 4. J Immunol 2001; 167:2250.
  110. 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.
  111. Wershil BK, Wang ZS, Gordon JR, Galli SJ. Recruitment of neutrophils during IgE-dependent cutaneous late phase reactions in the mouse is mast cell-dependent. Partial inhibition of the reaction with antiserum against tumor necrosis factor-alpha. J Clin Invest 1991; 87:446.
  112. Arock M, Ross E, Lai-Kuen R, et al. Phagocytic and tumor necrosis factor alpha response of human mast cells following exposure to gram-negative and gram-positive bacteria. Infect Immun 1998; 66:6030.
  113. Caulfield JP, Hein A, Moser G, Sher A. Light and electron microscopic appearance of rat peritoneal mast cells adhering to schistosomula of Schistosoma mansoni by means of complement or antibody. J Parasitol 1981; 67:776.
  114. Aschenbach JR, Seidler T, Ahrens F, et al. Luminal salmonella endotoxin affects epithelial and mast cell function in the proximal colon of pigs. Scand J Gastroenterol 2003; 38:719.
  115. King CA, Anderson R, Marshall JS. Dengue virus selectively induces human mast cell chemokine production. J Virol 2002; 76:8408.
  116. Gommerman JL, Oh DY, Zhou X, et al. A role for CD21/CD35 and CD19 in responses to acute septic peritonitis: a potential mechanism for mast cell activation. J Immunol 2000; 165:6915.
  117. McNeil BD, Pundir P, Meeker S, et al. Identification of a mast-cell-specific receptor crucial for pseudo-allergic drug reactions. Nature 2015; 519:237.
  118. Serhan N, Basso L, Sibilano R, et al. House dust mites activate nociceptor-mast cell clusters to drive type 2 skin inflammation. Nat Immunol 2019; 20:1435.
  119. Meixiong J, Anderson M, Limjunyawong N, et al. Activation of Mast-Cell-Expressed Mas-Related G-Protein-Coupled Receptors Drives Non-histaminergic Itch. Immunity 2019; 50:1163.
  120. Marone G, de Paulis A, Florio G, et al. Are mast cells MASTers in HIV-1 infection? Int Arch Allergy Immunol 2001; 125:89.
  121. van Schaik SM, Tristram DA, Nagpal IS, et al. Increased production of IFN-gamma and cysteinyl leukotrienes in virus-induced wheezing. J Allergy Clin Immunol 1999; 103:630.
  122. Castleman WL, Sorkness RL, Lemanske RF Jr, McAllister PK. Viral bronchiolitis during early life induces increased numbers of bronchiolar mast cells and airway hyperresponsiveness. Am J Pathol 1990; 137:821.
  123. Di Nardo A, Vitiello A, Gallo RL. Cutting edge: mast cell antimicrobial activity is mediated by expression of cathelicidin antimicrobial peptide. J Immunol 2003; 170:2274.
  124. Metz M, Piliponsky AM, Chen CC, et al. Mast cells can enhance resistance to snake and honeybee venoms. Science 2006; 313:526.
  125. Schneider LA, Schlenner SM, Feyerabend TB, et al. Molecular mechanism of mast cell mediated innate defense against endothelin and snake venom sarafotoxin. J Exp Med 2007; 204:2629.
  126. Akahoshi M, Song CH, Piliponsky AM, et al. Mast cell chymase reduces the toxicity of Gila monster venom, scorpion venom, and vasoactive intestinal polypeptide in mice. J Clin Invest 2011; 121:4180.
  127. Abonia JP, Friend DS, Austen WG Jr, et al. Mast cell protease 5 mediates ischemia-reperfusion injury of mouse skeletal muscle. J Immunol 2005; 174:7285.
  128. Suto H, Nakae S, Kakurai M, et al. Mast cell-associated TNF promotes dendritic cell migration. J Immunol 2006; 176:4102.
  129. Huang C, De Sanctis GT, O'Brien PJ, et al. Evaluation of the substrate specificity of human mast cell tryptase beta I and demonstration of its importance in bacterial infections of the lung. J Biol Chem 2001; 276:26276.
  130. Watanabe N, Bruschi F, Korenaga M. IgE: a question of protective immunity in Trichinella spiralis infection. Trends Parasitol 2005; 21:175.
  131. Knight PA, Wright SH, Lawrence CE, et al. 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.
  132. Gurish MF, Bryce PJ, Tao H, et al. IgE enhances parasite clearance and regulates mast cell responses in mice infected with Trichinella spiralis. J Immunol 2004; 172:1139.
  133. Dessein AJ, Parker WL, James SL, David JR. IgE antibody and resistance to infection. I. Selective suppression of the IgE antibody response in rats diminishes the resistance and the eosinophil response to Trichinella spiralis infection. J Exp Med 1981; 153:423.
  134. Mazzoni A, Siraganian RP, Leifer CA, Segal DM. Dendritic cell modulation by mast cells controls the Th1/Th2 balance in responding T cells. J Immunol 2006; 177:3577.
  135. Lawrence CE, Paterson YY, Wright SH, et al. Mouse mast cell protease-1 is required for the enteropathy induced by gastrointestinal helminth infection in the mouse. Gastroenterology 2004; 127:155.
  136. Noli C, Miolo A. The mast cell in wound healing. Vet Dermatol 2001; 12:303.
  137. Garbuzenko E, Nagler A, Pickholtz D, et al. Human mast cells stimulate fibroblast proliferation, collagen synthesis and lattice contraction: a direct role for mast cells in skin fibrosis. Clin Exp Allergy 2002; 32:237.
  138. Hermes B, Feldmann-Böddeker I, Welker P, et al. Altered expression of mast cell chymase and tryptase and of c-Kit in human cutaneous scar tissue. J Invest Dermatol 2000; 114:51.
  139. Younan GJ, Heit YI, Dastouri P, et al. Mast cells are required in the proliferation and remodeling phases of microdeformational wound therapy. Plast Reconstr Surg 2011; 128:649e.
  140. Garfield RE, Irani AM, Schwartz LB, et al. Structural and functional comparison of mast cells in the pregnant versus nonpregnant human uterus. Am J Obstet Gynecol 2006; 194:261.
  141. 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; 272:29158.
  142. Lu LF, Lind EF, Gondek DC, et al. Mast cells are essential intermediaries in regulatory T-cell tolerance. Nature 2006; 442:997.
  143. Gounaris E, Erdman SE, Restaino C, et al. Mast cells are an essential hematopoietic component for polyp development. Proc Natl Acad Sci U S A 2007; 104:19977.
  144. Lundequist A, Tchougounova E, Abrink M, Pejler G. Cooperation between mast cell carboxypeptidase A and the chymase mouse mast cell protease 4 in the formation and degradation of angiotensin II. J Biol Chem 2004; 279:32339.
  145. Goldstein SM, Kaempfer CE, Kealey JT, Wintroub BU. Human mast cell carboxypeptidase. Purification and characterization. J Clin Invest 1989; 83:1630.
  146. Lefrançais E, Duval A, Mirey E, et al. Central domain of IL-33 is cleaved by mast cell proteases for potent activation of group-2 innate lymphoid cells. Proc Natl Acad Sci U S A 2014; 111:15502.
  147. 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; 106:22381.
  148. Houde M, Jamain MD, Labonté J, et al. Pivotal role of mouse mast cell protease 4 in the conversion and pressor properties of Big-endothelin-1. J Pharmacol Exp Ther 2013; 346:31.
  149. Williams CM, Galli SJ. The diverse potential effector and immunoregulatory roles of mast cells in allergic disease. J Allergy Clin Immunol 2000; 105:847.
  150. Mihm MC Jr, Soter NA, Dvorak HF, Austen KF. The structure of normal skin and the morphology of atopic eczema. J Invest Dermatol 1976; 67:305.
  151. Crimi E, Chiaramondia M, Milanese M, et al. Increased numbers of mast cells in bronchial mucosa after the late-phase asthmatic response to allergen. Am Rev Respir Dis 1991; 144:1282.
  152. Jeziorska M, McCollum C, Woolley DE. Mast cell distribution, activation, and phenotype in atherosclerotic lesions of human carotid arteries. J Pathol 1997; 182:115.
  153. Sun J, Sukhova GK, Wolters PJ, et al. Mast cells promote atherosclerosis by releasing proinflammatory cytokines. Nat Med 2007; 13:719.
  154. Lee-Rueckert M, Kovanen PT. Mast cell proteases: physiological tools to study functional significance of high density lipoproteins in the initiation of reverse cholesterol transport. Atherosclerosis 2006; 189:8.
  155. Secor VH, Secor WE, Gutekunst CA, Brown MA. Mast cells are essential for early onset and severe disease in a murine model of multiple sclerosis. J Exp Med 2000; 191:813.
  156. Lee DM, Friend DS, Gurish MF, et al. Mast cells: a cellular link between autoantibodies and inflammatory arthritis. Science 2002; 297:1689.
  157. Younan G, Suber F, Xing W, et al. The inflammatory response after an epidermal burn depends on the activities of mouse mast cell proteases 4 and 5. J Immunol 2010; 185:7681.
  158. Robertson I, Greaves MW. Responses of human skin blood vessels to synthetic histamine analogues. Br J Clin Pharmacol 1979; 5:319.
  159. Lemanske RF Jr, Kaliner MA. Late phase allergic reactions. Int J Dermatol 1983; 22:401.
Topic 3979 Version 22.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟