Roles and relevance of mast cells in infection and vaccination

2016-06-05 08:55YuFangZouXiang
THE JOURNAL OF BIOMEDICAL RESEARCH 2016年4期

Yu Fang, Zou Xiang

1Department of Microbiology and Immunology; Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550025, China;

2Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Research Center, Institute of Biomedicine, University of Gothenburg, Gothenburg 40530, Sweden.

Roles and relevance of mast cells in infection and vaccination

Yu Fang1, Zou Xiang2,✉

1Department of Microbiology and Immunology; Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550025, China;

2Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Research Center, Institute of Biomedicine, University of Gothenburg, Gothenburg 40530, Sweden.

In addition to their well-established role in allergy mast cells have been described as contributing to functional regulation of both innate and adaptive immune responses in host defense. Mast cells are of hematopoietic origin but typically complete their differentiation in tissues where they express immune regulatory functions by releasing diverse mediators and cytokines. Mast cells are abundant at mucosal tissues which are portals of entry for common infectious agents in addition to allergens. Here, we review the current understanding of the participation of mast cells in defense against infection. We also discuss possibilities of exploiting mast cell activation to provide adequate adjuvant activity that is needed in high-quality vaccination against infectious diseases.

mast cell, infection, host defense, vaccine, adjuvant

Introduction

Classically mast cells are considered critical effector cells in allergy by virtue of their potential to secrete a variety of allergic mediators. The number of mast cells is increased at sites of allergic inflammation, and there is a correlation between mast cell density in the tissue and the severity of allergic symptoms[1]. In allergy, plurivalent antigens bind and crosslink IgE molecules bound to the high-affinity IgE-receptor (FcεRI) expressed on mast cells, resulting in cell degranulation and release of proinflammatory mediators. Three major categories of mast cell mediators have been described:(1) preformed granule-associated mediators such as histamine and serotonin; (2) newly generated lipid mediators such as leukotrienes and prostaglandins; (3) de novo synthesized cytokines including chemokines. IgE-mediated activation of mast cells initiates the early phase of allergic responses, resulting in pathologies including greater epithelial permeability, mucus production, smooth muscle contraction, vasodilitation and neurogenic inflammation. The immediate response is followed by recruitment of a variety of other immune cells that participate in the late phase of the reaction, further exacerbating allergic pathology[1].

Mast cells are derived from hematopoietic progenitors in the bone marrow which migrate via blood to tissues all over the body where they further differentiate and mature into different phenotypes, depending on the local microenvironment. Stem cell factor (SCF), also known as steel factor, KIT ligand, or mast cell growth factor, is found to be the primary growth and differentiation factor for mast cells[2]. The cellular receptor for SCF is the product of the c-kitproto-oncogene. In addi-tion to SCF, mast cell growth and differentiation can be facilitated by several other cytokines including IL-3. For example, expansion of tissue mast cells upon nematode infection requires IL-3[3-4]. Immature mouse mast cells can be differentiatedin vitrofrom bone marrow precursor cells in the presence of IL-3 without SCF[5]. Mast cells are enriched in the skin, around blood vessels, and in mucosal membranes such as the respiratory and gastrointestinal tracts. Most notably, mast cells are highly enriched in the skin and mucosal barriers of the body, where they serve as a first line of defense. It is noteworthy that mature mast cells are capable of differentiating both phenotypically and functionally as a consequence of tissue-specific stimulation under defined microenvironmental conditions. For example, inflamed lungs are reported to have more tryptase/chymase-producing mast cells compared with non-inflamed lung tissue in which tryptase-producing mast cells are dominant[6-7].

Mast cell subtypes

Two major subtypes of rodent mast cells have been characterized, i.e. connective tissue mast cells (CTMC) and mucosal mast cells (MMC), based on their tissue localization[8-11]. For instance, skin mast cells and mast cells residing in the peritoneal cavity are CTMC, whereas mast cells located in the respiratory or gastrointestinal tracts are usually characterized as MMC. In addition to tissue localization, other properties such as protease and cytokine profiles, membrane receptor distribution, and growth factor requirements also distinguish these two types of mast cells. In addition to residing in connective and serosal tissues, CTMC in mice have been found in the submucosa of the stomach[12]and nasal tissue[13].

In contrast, human mast cells are usually grouped based on the expression pattern of two mast cell-specific proteases, i.e. tryptase and chymase. According to this classification, two major human mast cell subgroups have been proposed. Mast cells that contain only tryptase are referred to as MCT, whereas those that contain both tryptase and chymase are termed MCTC. In terms of correlation to their murine counterparts, MCTare found mainly in mucosal tissues, resembling mouse MMC, while MCTC, which reside in such sites as the skin and small intestinal submucosa, are more closely related to mouse CTMC[14], although the tissue localization is less stringent for human “CTMC” and “MMC”. Similar to mouse mast cells, human mast cells also differ in the requirement for growth and differentiation factors. Specifically, SCF is needed for the survival of both types, whereas IL-4 is indispensable for MCTC, but not for MCT[15].

Multitalented cells beyond allergy

In addition to IgE- and FcεRI-mediated cell activation, mast cells can be activated by a variety of other stimulators, such as IgG immune complexes, cytokines, complement components, neuropeptides, chemical agents, and physical stimuli, as mast cells express broad-ranging surface receptors including Fc receptors, complement receptors, and pathogen-associated molecular patterns (PAMP) such as Toll-like receptors (TLR). These observations, together with the description of a wide spectrum of mast cell mediators, provide a basis for proposals implicating mast cells in almost all aspects of immune responses. Therefore, mast cells have been postulated to be modulators of numerous physiological and pathological responses beyond their classically defined role in allergies mediated mainly through FcεRI. These multifunctional properties of mast cells have been more extensively reviewed elsewhere[16-17]. It has to be pointed out that the overwhelming research findings addressing the roles of mast cells have relied on the use of mast celldeficient, KIT mutant mice which have other phenotypic abnormalities in addition to mast cell deficiency. These data await further experimental verification using the KIT-independent mast cell-deficient models to eliminate the confounding elements as a result of KIT mutation[18].

The roles of mast cells in host defense

The earliest observation of a beneficial role of mast cells is their potential in defense against parasitic infection[19-20]. The MMC pool expands extensively during nematode infection, a process dependent on IL-3[3-4]. Both IgE and mouse mast cell protease-6 (mMCP-6) are required for chronic immune responses againstTrichinella spiralisinfections[21]. In a helminth infection model, mast cells contribute to pathogen clearance by migrating to the draining lymph nodes and producing IL-6 and IL-4[22]. Interestingly, mast cells have also been described to be critical for Th1 response-mediated defence against oral infection withToxoplasma gondii[23]. In addition to defense against helminth infections, mast cells have also been described to be protective in bacterial infections. One of the classic examples of mast cell-dependent anti-bacterial infection is demonstrated by the cecal ligation and puncture (CLP) model of acute peritonitis which is dependent on tumour necrosis factor (TNF)[24]and the ability of mast cells to lower neurotensin levels[25]. Mast cells harbour antimicrobial peptides including cathelicidin in their secretory granules[26]. Furthermore, β-hexosaminidase, which is abundantly contained in mast cell granules, has recently been reported to have bactericidal activity[27]. The roles andrelevance of mast cells in defense against viral and fungal infections have also been suggested[28-29].

Pathogen-mediated mast cell activation can be achieved through several mechanisms. Mast cells can be activated, through the equipped TLR, by direct recognition of microbial components such as bacterial lipopolysaccharide (LPS) and peptidoglycan resulting in distinct outcomes[30-32]. Mast cells can respond to microbial stimuli by surface proteins such as CD48[33-34]. Furthermore, mast cells can be stimulated by endogenous inflammatory factors such as cytokines and complement components secondary to infection[35-36]. Indirect interaction of mast cells with pathogens can also be achieved through the recognition of pathogen-antibody complexes by Fc receptors including FcεRI and Fcγ receptors expressed on mast cells[37-39]. Fc receptor-mediated mast cell activation may also be triggered in the presence of certain pathogen-derived proteins that can bind immunoglobulins in an antigen-independent manner. A classic example of such a bacteria-derived superantigen is protein A fromStaphylococcus aureuswhich can activate human and mouse tissue mast cells[40-42], as the FcεRI molecules on these mast cells are most likely to have already been occupied with IgE, resulting in crosslinking of FcεRI upon protein A binding. However, the pathophysiological roles of such superantigen-mediated mast cell activation in defence against infection await further clarification.

Similar to mast cell activation in other circumstances, the activation by pathogens is also believed to include both degranulation of pre-formed granular contents and selectivede novomediator production, for example, cytokines and lipid mediators, the patterns of which differ greatly depending on the stimulus encountered. These mast cell-associated products, such as TNF, IL-4, OX40 ligand and mMCP-6, are important for the recruitment and stimulation of other innate immune participants,e.g.neutrophils, macrophages, natural killer (NK) cells and eosinophils, contributing to the clearance of pathogens[21,30,43-44]. Mast cells not only interact with cells in the immediate vicinity where the infection first takes place but also influence distant targets,e.g.cells in lymph nodes through mediators that they release[45]. It is also reported that mast cells can kill bacteria by producing extracellular traps that contain antimicrobial mediators[46].

In addition to contributing to innate immune responses by virtue of their large spectrum of granular products, mast cells also form a link between innate and adaptive immunity. Mast cells modulate the phenotype and function of key players in adaptive immunity, such as dendritic cells (DC), B cells, and T cells. Mast cells have been shown to functionally interact with professional antigen presenting cells (APC) such as DC and regulate their function mainly through mast cell-derived granular pro ducts. For example, histamine is capable of regulating the chemotaxis of immature DC[47-48]and cross- presentation of extracellular antigens[49]. TNF produced from mast cells is critical for DC migration[50-52]. TLR7 ligand-mediated mast cell activation is effective for the migration and maturation of Langerhans cells[53]. Maturation and activation of immature DC by mast cell-DC direct contact results in the activation of T cells that release IFN-γ and IL-17 promoting Th1 and Th17 responses, respectively[54]. Mast cells provide essential signals such as IL-6 and CD40L to enhance proliferation of B cells and drive their differentiation toward IgA-secreting plasma cells[55]. Mast cells can enhance the activation of T cells by providing costimulatory signals and secreting TNF[56-58]. Mast cells also contribute to the recruitment of T cells to sites of viral infection by secreting chemotactic molecules[59-60].

One of the key processes in achieving successful adaptive immunity is the presentation of microbial antigens to T lymphocytes. Whether or not mast cells are capable of acting as antigen-presenting cells is still controversial[61-65]. This is largely because of the argument that mast cells under steady state do not seem to constitutively express major histocompatibility complex class II (MHC-II) or co-stimulatory molecules such as CD86[63-64]. In contrast, mast cells upregulate expression of MHC-II and costimulatory molecules following stimulation by inflammatory factors such as IFN-γ and LPS[63-64]. Therefore, mast cells may have the potential to directly present antigens to T cells at least under certain circumstances, for example, in inflamed tissues, to initiate adaptive immune responses. Mast cells have also been demonstrated to present antigen to and activate CD8+T cells through MHC-I molecules[66-67].

Alternatively, mast cells are reported as participating in antigen cross-presentation to T cells[68]. Cross-presentation refers to a process, most typically following intracellular microbial infection, during which professional APC ingest infected cells and display the antigens of the microbes originally engulfed by the infected cells for recognition by T lymphocytes[69]. This is an efficient mechanism for presenting the antigens of those microbes that have infected host cells that may not produce all the signals,e.g.MHC-II recognition and costimulation needed to initiate T cell activation. The professional APC that have ingested infected cells may present the microbial antigens to both CD4+and CD8+T lymphocytes depending on the processing and presentation routes. Morphological changes of the host cells as a result of,e.g.microbial infections, apoptosis, and tumourigenesis, will facilitate ingestion by APC. In principle, any type of cells that have internalized antigens can participate in cross-presentation upon ingestion by APC. Importantly, mast cells have been implicated in thepha gocytosis of various types of antigens[70-73]. Various mechanisms have been reported for mast cells to internalize bacterial pathogens[74-76]. Indeed, mast cells can serve as an antigen-reservoir and participate in antigen cross-presentation[68].In vitrocultured bone marrow-derived cultured mast cells (BMMC) can internalize IgE-bound chicken ovalbumin (OVA) protein, followed by engulfment by DC which process and present the OVA peptide to T cells that have specific receptors for the OVA peptide[68]. Induction of BMMC apoptosis is documented to be critical for efficient presentation by DC to T cells of the antigen originally phagocytosed by mast cells[68].

Owing to the fact that mast cells are capable of participating in both innate and adaptive immunity, and that they are enriched at the mucosal and skin barriers between the body and the external environment, mast cells, similar to skin Langerhans cells, tissue-resident DC and epithelial cells, are believed to be sentinel cells that are probably the first responders to a threat within seconds. Equipped with their immunologic armory of mediators, mast cells may possibly exert a pivotal role in the surveillance and elimination of pathogens by diversified mechanisms.

While people have been endowing mast cells with a more positive image in health, new findings also implicate mast cells or their released products negatively in infection. Although mast cell-associated TNF has been reported to be critical for a CLP model of acute peritonitis[24], it has to be pointed out that mast cell-derived TNF is not always protective in acute peritonitis, especially in models of severe CLP[77]. The detrimental effects of mast cells in severe peritonitis have also been ascribed to the release of IL-4 that inhibits the phagocytic potential of macrophages[78]. Mast cell degranulation may contribute to vascular leakage that may exacerbate dengue virus infection[38]. Even the potential of mast cells to recruit other immune effector cells during an infection is not always protective as this has been found to promoteChlamydia pneumoniaeinfection[79]. Interestingly, mMCP-4, the mouse counterpart of human mast cell chymase, can degrade TNF, thus dampening the severity of inflammation associated with sepsis and limiting the damage caused by TNF[80], suggesting antagonism between mast cell mediators, thus favouring protection. Therefore, the implication and relevance of mast cells in host defense is a complex issue and the net outcome may depend on many antagonistic factors.

The implication of mast cells in vaccination

A vaccine is a biological preparation that stimulates an immune response against specific antigens that either are derived from the pathogen itself or resemble the structure of the pathogen. Ever since the first documented vaccination attempt by Edward Jenner for the prevention of small pox in 1796, vaccines have played a crucial role in protecting people against many infectious diseases[81-82]. The eradication of smallpox and the effective control of polio represent two classic success stories of how vaccines can play a major role in improving global health. Nevertheless, the demand for better and more effective vaccines against many infectious diseases is still growing, especially when infections such as tuberculosis, HIV, dengue fever and malaria still present enormous global problems. From a societal point of view, vaccination remains the most effective intervention in the control of infectious diseases and for the improvement of global health.

There are two principal forms of vaccines: those that are live attenuated vaccines and those that are killed whole pathogens or subcomponent vaccines. An advantage of live attenuated vaccines is that they usually stimulate long-term immune responses similar to natural infection. However, live attenuated vaccines always come with a risk of reversion into more virulent organisms that could cause adverse reactions or more severe infections. In contrast, killed vaccines or subcomponent vaccines are more predictable and, therefore, safer. Furthermore, another concern that makes live attenuated vaccines less practical is the demand for a cold-chain for storing or transporting these vaccines. Therefore, killed vaccines are still much in use, even though they are weaker and usually do not promote as strong long-term memory responses.

To make killed vaccines more effective, we need adjuvants which are substances that enhance immune responses and stimulate long-lasting, robust protective immunity. An adjuvant that is included in the vaccine contributes greatly to the efficacy of the vaccination by affecting the immune responses both quantitatively and qualitatively. Importantly, protective immunity following vaccination may be generated with lower amounts of antigen and a reduced dosing frequency after addition of an adjuvant[83].

Of all currently available adjuvants, aluminium salts (alum) have the longest history in practical vaccination. Alum-based vaccines have a good safety record and are capable of inducing early, high-titer, long-lasting protective immunity. At present, alum is still the most widely used adjuvant in both veterinary and human vaccines. The mechanism of action has been proposed to depend on a depot effect, enabling physical adsorption of antigen onto the alum depots. Furthermore, alum is reported to have direct immunostimulating effects[84]. The relevance of mast cells in alum-mediated adjuvanticity hasbeen explored[84]. Interestingly, mast cells are found to respond to alum stimulation by releasing histamine and a panel of cytokines including IL-5 and IL-1β. Although by using the mast cell-deficientKitW/W-vmice it is demonstrated that mast cells are not required for the priming of endogenous CD4 and CD8 T cells[84], this does not formally exclude the contribution of mast cells to the adjuvant activity of alum in the wild-type mice as redundant pathways may exist.

However, alum does not seem to be effective for mucosal immunisation, a route that has appreciable advantages compared with routes that require needle injections, i.e. intramuscular or subcutaneous delivery of vaccines. Needle-free mucosal vaccination can be achievedviaoral, intranasal, sublingual, or intravaginal routes[85-86]. The obvious benefits of mucosal immunisation include avoidance of blood-borne contamination through re-use of syringes and needles as well as the fact that no trained professional personnel are required for vaccine delivery. Furthermore, mucosal immunisation can generate both systemic and mucosal immune responses[85-86]. Strikingly, mucosal immunisation can generate effective secretory IgA even at mucosal sites distant from where the vaccine is delivered[87-88]. For example, nasal immunisation can generate protective mucosal antibodies in the genital tract mucosa, which signifies the advantage of nasal vaccination. As most pathogens enter the body through mucosal surfaces, local mucosal immune responses are critically important in defense against invading pathogens. Therefore, how to achieve strong local protection has become one of the major goals of vaccine development.

As the mucosal route of vaccination, as opposed to the parenteral route, often results in immune tolerance development, potent adjuvants are much warranted. Therefore, the selection of a strong mucosal adjuvant for effective vaccination is vital and possibly as important as the vaccine antigens themselves[85]. A number of strategies are proposed to design mucosal adjuvants. TLR agonists have been tested and these include TLR4 ligand monophosphoryl lipid A[89-90], TLR9 ligand CpG oligodeoxynucleotides (ODN)[91]and the TLR5 ligand flagellin[92]. Bacterial enterotoxins which include cholera toxin (CT) andEscherichia coliheatlabile toxin (LT) constitute another major group of experimental mucosal adjuvants[93]. Both CT and LT are composed of five B-subunits (CTB and LTB) and a single copy of the A subunit (CTA or LTA)[94]. The CTA subunit is produced as a single polypeptide chain that is post-translationally modified through the action of aVibrio choleraeprotease to form two chains, CTA1 and CTA2, which remain linked by a disulphide bond. CTA1 is enzymatically active by ADP-ribosylating the cell membrane bound Gsα-protein, whereas CTB binds to GM1-gangliosides present on virtually all nucleated cells[95]. CTA2 is responsible for linking CTA into the CTB pentamer[96].

DC are believed to play a central role in the presentation of antigens to naïve T cells, which is a critical process for the development of adaptive immunity following natural infection[97]. As adjuvants are expected to mediate the same consequences as natural infections, quite a number of adjuvant studies are focused on the interaction of adjuvant with DC. Other types of cells have also been described to contribute to adjuvanticity. For example, B cells[98-99], macrophages[100], NK cells and NKT cells[101-103]have also been implicated as targets for vaccine adjuvants.

Given the accumulating evidence suggesting a functional interplay between mast cells and other immune cells such as DC, T cells and B cells in adaptive immune responses, also mast cells have been implicated in adjuvant functions. Indeed, mast cell activators such as c48/80 have been reported as exerting a mucosal adjuvant function[104]. More specifically, c48/80 is demonstrated to be an efficient adjuvant by mobilizing DC to the draining lymph nodes through production of TNF. Successful vaccinations of several animal infection models using c48/80 as adjuvant have now been reported[105-109]. Retention of c48/80 and antigen on mucosal surfaces by chitosan-based nanoparticles can further promote mucosal immunisation[110]. The IL-1 family cytokines such as IL-1, IL-18 and IL-33 have been shown to exert adjuvant function capable of augmenting protection against influenza virus infection[111]. Interestingly, the effect of IL-18 and IL-33 is suggested to be mast cell-dependent[111], which is not surprising as both cytokines can activate mast cells resulting in proinflammatory cytokine production. IL-18 together with IL-2 is potent in expanding the mucosal mast cell pool and the production of mMCP-1, which is critical for parasite expulsion[112]. IL-33 is described as a danger signal that can alert mast cells[113]and keratinocytederived IL-33 can stimulate mast cells to produce TNF and IL-6, cytokines critical for defence against herpes simplex virus infection[114]. Polymyxins which are clinically approved antibiotics can activate mast cells and boost immunisation[115]. In a QuilA-adjuvanted cattle vaccination model for protection against nematode infection, mast cells are most likely to be involved in the mechanism of adjuvanticity through the production of granzyme B and granulysin[116]. Synthetic particles harboring TNF, mimicking mast cell granules, have been reported to be powerful adjuvant in a mouse model of influenza[117]. Furthermore, it is suggested that the gold standard mucosal adjuvant CT may stimulate therelease of IL-6 from mast cells boosting humoral immune responses[118].

Although the bacterial enterotoxins have been demonstrated to be powerful mucosal adjuvants experimentally, these substances are precluded from clinical use because of their toxicity and, hence, they have very limited applicability in human vaccines[119-120]. Extensive studies have, however, focused on the detoxification of these molecules using various approaches. For example, site-directed mutagenesis has generated detoxified mutants, such as CT112K, LTG192, LTR72, or LTK63, with little or no enzymatic activity, but with retained adjuvant function in experimental models[121-124]. However, a drastically different approach was applied by Lycke and co-workers who developed an adjuvant based on the intact CTA1 molecule without the B-subunit. The CTA1 is linked genetically to a dimer of the D-fragment ofStaphylococcus aureusprotein A forming the CTA1-DD adjuvant. Thus, CTA1-DD has retained the adjuvant function while the molecule cannot bind to GM1-ganglioside, rendering the molecule nontoxic[125]. In contrast to CT, intranasal administration of CTA1-DD results in neither inflammation nor accumulation in nervous tissues as is found with CT or LT[126]. The adjuvanticity of CTA1-DD has been well documented in various infectious disease models, which includeChlamydia trachomatis, influenza, HIV,Mycobacterium tuberculosis,andHelicobacter pylori[127-133]. The ADP-ribosyltransferase activity is central to the adjuvant effect[134]. In addition, mechanistic studies have identified several mechanisms of action that may explain the adjuvanticity of CTA1-DDin vivo. As the DD domain binds to all immunoglobulins, CTA1-DD can target B cells through the B cell receptor, i.e. surface bound immunoglobulins, and promote B cell activation and germinal center development[135]. Moreover, the adjuvant also enhances T cell-independent immune responses[135]. Importantly, CTA1-DD stimulates germinal center formation effectively generating long-lived plasma cells and long-lived B memory cells[136]. Furthermore, also follicular DC and complement activation have been found to be essential elements for the function of this adjuvant[137].

In contrast to intactStaphylococcus aureusprotein A, which can activate mast cells[40,42], CTA1-DD fails to activate mast cells[138]. However, as the double D domains derived from protein A have binding sites for immunoglobulins, CTA1-DD can bind to all immunoglobulins including IgG[139-140]. We demonstrated that CTA1-DD and IgG may form complexes that are able to activate mast cells through Fcγ receptors, resulting in degranulation and the production of TNF and IL-6. Intranasal immunisation in the presence of CTA1-DD and IgG as an adjuvant can enhance antigen-specific immune responses compared with CTA1-DD alone. Importantly, this enhancement is dependent on mast cells[138]. Furthermore, we demonstrated that only CTMC, but not MMC, can be activated by immune complexes composed of CTA1-DD and IgG. This effect is mediated by FcγRIIIA, an activating receptor that is confirmed to be only expressed on CTMC. Indeed, CTMC are found in the nasal submucosa and these cells are demonstrated to express FcγRIIIA[13].

As MMC are not activated in response to stimulation by IgG immune complexes because of the lack of FcγRIIIA[13], it was intriguing to investigate whether or not MMC could contribute to adaptive immune responses somehow, perhaps using another mechanism. We have recently reported that IgG immune complex-primed MMC can mediate enhanced antigen-specific activation of T cells, possibly providing a cross-presentation mechanism to boost mucosal vaccination[141]. In practical immunisation, this may happen when IgG immune complex-containing vaccine formulations are used.

The development of adjuvants that enhance the potency of subunit vaccines formulated for administration through the mucosal routes is much desired. Dissecting and revealing the molecular mechanisms, through which mast cells precisely control adaptive immune responses to combat microbial infections, may have implications for rationally designing mucosal vaccine formulations. We propose that IgG immune complex-induced mast cell activation may be consideredas one of the components for mucosal vaccine adjuvants.Fig. 1summarizes the current knowledge regarding the strategies for the selection of vaccine formulations that target mast cells for enhancing immune responses.

Fig. 1Mast cell activation strategies that may be exploited to provide adjuvant activity. Mast cells can be activated by various factors including compound 48/80 (c48/80), IL-33, IL-18, cholera toxin (CT), QuilA, alum and IgG immune complexes (IgG IC). Upon activation, mast cells release TNF-α, IL-6, IL-5, IL-1β, histamine, mouse mast cell protease (mMCP), granzyme and granulysin that are critical in mobilizing immune responses. However, these are just examples of functional consequences of mast cell activation specific to the findings of those studies addressing roles of mast cells in vaccination cited in this review; the actual spectrum of mediators released by mast cells can be much more dynamic and complex.

One of the challenges associated with mast cellmediated immune enhancement, of course, lies in overcoming the complexity of safety issues for the clinical development of the vaccines. The constant threats posed by infectious diseases over millions of years may have driven evolutionary pressure to keep mast cells, despite their adverse properties,e.g.in causing allergy, in humans to exploit these cells’ beneficial functions in host defense. Our immune system has evolved mechanisms to balance the positive and negative contributions of mast cells to health. It is worth exploring strategies to make use of the adjuvant properties of mast cells to provide high-quality vaccination while minimizing any health-compromising factors.

Acknowledgements

This work was supported by Stiftelsen Clas Groschinskys Minnesfond; Konsul Berghs Stiftelse, Sweden. YF was supported by a postdoctoral start-up grant from the Affiliated Hospital of Guizhou Medical University, Guiyang, China.

[1] Galli SJ, Tsai M, Piliponsky AM. The development of allergic inflammation[J].Nature, 2008, 454(7203): 445-454.

[2] Wedemeyer J, Tsai M, Galli SJ. Roles of mast cells and basophils in innate and acquired immunity[J].Curr Opin Immunol, 2000, 12(6): 624-631.

[3] Lantz CS, Boesiger J, Song CH, et al. Role for interleukin-3 in mast-cell and basophil development and in immunity to parasites[J].Nature, 1998, 392(6671): 90-93.

[4] Madden KB, Urban JF, Jr., Ziltener HJ, et al. Antibodies to IL-3 and IL-4 suppress helminth-induced intestinal mastocytosis[J].J Immunol, 1991, 147(4): 1387-1391.

[5] Dvorak AM, Seder RA, Paul WE, et al. Effects of interleukin-3 with or without the c-kit ligand, stem cell factor, on the survival and cytoplasmic granule formation of mouse basophils and mast cells in vitro[J].Am J Pathol, 1994, 144(1): 160-170.

[6] Andersson CK, Mori M, Bjermer L, et al. Alterations in lung mast cell populations in patients with chronic obstructive pulmonary disease[J].Am J Respir Crit Care Med, 2010, 181(3): 206-217.

[7] Balzar S, Chu HW, Strand M, et al. Relationship of small airway chymase-positive mast cells and lung function in severe asthma[J].Am J Respir Crit Care Med, 2005, 171(5): 431-439.

[8] Enerback L. Mucosal mast cells in the rat and in man[J].Int Arch Allergy Appl Immunol, 1987, 82(3-4): 249-255.

[9] Kitamura Y. Heterogeneity of mast cells and phenotypic change between subpopulations[J].Annu Rev Immunol, 1989, 7: 59-76.

[10] Enerback L. Mast cells in rat gastrointestinal mucosa. 2. Dye-binding and metachromatic properties[J].Acta Pathol Microbiol Scand, 1966, 66(3): 303-312.

[11] Vliagoftis H, Befus AD. Rapidly changing perspectives about mast cells at mucosal surfaces[J].Immunol Rev, 2005, 206: 190-203.

[12] Gersch C, Dewald O, Zoerlein M, et al. Mast cells and macrophages in normal C57/BL/6 mice[J].Histochem Cell Biol, 2002, 118(1): 41-49.

[13] Fang Y, Zhang T, Lidell L, et al. The immune complex CTA1-DD/IgG adjuvant specifically targets connective tissue mast cells through FcgammaRIIIA and augments anti-HPV immunity after nasal immunization[J].Mucosal Immunol, 2013, 6(6): 1168-1178.

[14] 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].J Histochem Cytochem, 1989, 37(10): 1509-1515.

[15] Toru H, Eguchi M, Matsumoto R, et al. Interleukin-4 promotes the development of tryptase and chymase double-positive human mast cells accompanied by cell maturation[J].Blood, 1998, 91(1): 187-195.

[16] Moon TC, St Laurent CD, Morris KE, et al. Advances in mast cell biology: new understanding of heterogeneity and function[J].Mucosal Immunol, 2010, 3(2): 111-128.

[17] Kalesnikoff J, Galli SJ. New developments in mast cell biology[J].Nat Immunol, 2008, 9(11): 1215-1223.

[18] Rodewald HR, Feyerabend TB. Widespread immunological functions of mast cells: fact or fiction[J]?Immunity, 2012, 37(1): 13-24.

[19] Woodbury RG, Miller HR, Huntley JF, et al. Mucosal mast cells are functionally active during spontaneous expulsion of intestinal nematode infections in rat[J].Nature, 1984, 312(5993): 450-452.

[20] Nawa Y, Kiyota M, Korenaga M, et al. Defective protective capacity of W/Wv mice against Strongyloides ratti infection and its reconstitution with bone marrow cells[J].Parasite Immunol, 1985, 7(4): 429-438.

[21] Shin K, Watts GF, Oettgen HC, et al. Mouse mast cell tryptase mMCP-6 is a critical link between adaptive and innate immunity in the chronic phase of Trichinella spiralis infection[J].J Immunol, 2008, 180(7): 4885-4891.

[22] Liu AY, Dwyer DF, Jones TG, et al. Mast cells recruited to mesenteric lymph nodes during helminth infection remain hypogranular and produce IL-4 and IL-6[J].J Immunol, 2013, 190(4): 1758-1766.

[23] Cruz A, Mendes EA, de Andrade MV, et al. Mast cells are crucial in the resistance against Toxoplasma gondii oral infection[J].Eur J Immunol, 2014, 44(10): 2949-2954.

[24] Echtenacher B, Mannel DN, Hultner L. Critical protective role of mast cells in a model of acute septic peritonitis[J].Nature, 1996, 381(6577): 75-77.

[25] Piliponsky AM, Chen CC, Nishimura T, et al. Neurotensin increases mortality and mast cells reduce neurotensin levels in a mouse model of sepsis[J].Nat Med, 2008, 14(4): 392-398.

[26] Di Nardo A, Vitiello A, Gallo RL. Cutting edge: mast cell antimicrobial activity is mediated by expression of cathelicidin antimicrobial peptide[J].J Immunol, 2003, 170(5):2274-2278.

[27] Fukuishi N, Murakami S, Ohno A, et al. Does beta-hexosaminidase function only as a degranulation indicator in mast cells? The primary role of beta-hexosaminidase in mast cell granules[J].J Immunol, 2014, 193(4): 1886-1894.

[28] Saluja R, Metz M, Maurer M. Role and relevance of mast cells in fungal infections[J].Front Immunol, 2012, 3: 146.

[29] Abraham SN, St John AL. Mast cell-orchestrated immunity to pathogens[J].Nat Rev Immunol, 2010, 10(6):440-452.

[30] Vosskuhl K, Greten TF, Manns MP, et al. Lipopolysaccharide-mediated mast cell activation induces IFN-gamma secretion by NK cells[J].J Immunol, 2010, 185(1):119-125.

[31] Supajatura V, Ushio H, Nakao A, et al. Differential responses of mast cell Toll-like receptors 2 and 4 in allergy and innate immunity[J].J Clin Invest, 2002, 109(10):1351-1359.

[32] Dietrich N, Rohde M, Geffers R, et al. Mast cells elicit proinflammatory but not type I interferon responses upon activation of TLRs by bacteria[J].Proc Natl Acad Sci U S A, 2010, 107(19): 8748-8753.

[33] Munoz S, Hernandez-Pando R, Abraham SN, et al. Mast cell activation by Mycobacterium tuberculosis: mediator release and role of CD48[J].J Immunol, 2003, 170(11):5590-5596.

[34] Malaviya R, Gao Z, Thankavel K, et al. The mast cell tumor necrosis factor alpha response to FimH-expressing Escherichia coli is mediated by the glycosylphosphatidylinositol-anchored molecule CD48[J].Proc Natl Acad Sci U S A, 1999, 96(14): 8110-8115.

[35] Nilsson G, Johnell M, Hammer CH, et al. C3a and C5a are chemotaxins for human mast cells and act through distinct receptors via a pertussis toxin-sensitive signal transduction pathway[J].J Immunol, 1996, 157(4): 1693-1698.

[36] Sesti-Costa R, Silva GK, Proenca-Modena JL, et al. The IL-33/ST2 pathway controls coxsackievirus B5-induced experimental pancreatitis[J].J Immunol, 2013, 191(1):283-292.

[37] Brown MG, King CA, Sherren C, et al. A dominant role for FcgammaRII in antibody-enhanced dengue virus infection of human mast cells and associated CCL5 release[J].J Leukoc Biol, 2006, 80(6): 1242-1250.

[38] Syenina A, Jagaraj CJ, Aman SA, et al. Dengue vascular leakage is augmented by mast cell degranulation mediated by immunoglobulin Fcgamma receptors[J].Elife, 2015, 4:e05291.

[39] Matsumoto M, Sasaki Y, Yasuda K, et al. IgG and IgE collaboratively accelerate expulsion of Strongyloides venezuelensis in a primary infection[J].Infect Immun, 2013, 81(7): 2518-2527.

[40] Genovese A, Bouvet JP, Florio G, et al. Bacterial immunoglobulin superantigen proteins A and L activate human heart mast cells by interacting with immunoglobulin E[J].Infect Immun, 2000, 68(10): 5517-5524.

[41] Terada M, Tsutsui H, Imai Y, et al. Contribution of IL-18 to atopic-dermatitis-like skin inflammation induced by Staphylococcus aureus product in mice[J].Proc Natl Acad Sci U S A, 2006, 103(23): 8816-8821.

[42] Patou J, Gevaert P, Van Zele T, et al. Staphylococcus aureus enterotoxin B, protein A, and lipoteichoic acid stimulations in nasal polyps[J].J Allergy Clin Immunol, 2008, 121(1): 110-115.

[43] Malaviya R, Ikeda T, Ross E, et al. Mast cell modulation of neutrophil influx and bacterial clearance at sites of infection through TNF-alpha[J].Nature, 1996, 381(6577):77-80.

[44] Rodriguez AR, Yu JJ, Murthy AK, et al. Mast cell/IL-4 control of Francisella tularensis replication and host cell death is associated with increased ATP production and phagosomal acidification[J].Mucosal Immunol, 2011, 4(2): 217-226.

[45] Kunder CA, St John AL, Li G, et al. Mast cell-derived particles deliver peripheral signals to remote lymph nodes[J].J Exp Med, 2009, 206(11): 2455-2467.

[46] von Kockritz-Blickwede M, Goldmann O, Thulin P, et al. Phagocytosis-independent antimicrobial activity of mast cells by means of extracellular trap formation[J].Blood, 2008, 111(6): 3070-3080.

[47] Dawicki W, Jawdat DW, Xu N, et al. Mast cells, histamine, and IL-6 regulate the selective influx of dendritic cell subsets into an inflamed lymph node[J].J Immunol, 2010, 184(4): 2116-2123.

[48] Caron G, Delneste Y, Roelandts E, et al. Histamine induces CD86 expression and chemokine production by human immature dendritic cells[J].J Immunol, 2001, 166(10): 6000-6006.

[49] Amaral MM, Davio C, Ceballos A, et al. Histamine improves antigen uptake and cross-presentation by dendritic cells[J].J Immunol, 2007, 179(6): 3425-3433.

[50] Suto H, Nakae S, Kakurai M, et al. Mast cell-associated TNF promotes dendritic cell migration[J].J Immunol, 2006, 176(7): 4102-4112.

[51] Jawdat DM, Rowden G, Marshall JS. Mast cells have a pivotal role in TNF-independent lymph node hypertrophy and the mobilization of Langerhans cells in response to bacterial peptidoglycan[J].J Immunol, 2006, 177(3):1755-1762.

[52] Shelburne CP, Nakano H, St John AL, et al. Mast cells augment adaptive immunity by orchestrating dendritic cell trafficking through infected tissues[J].Cell Host Microbe, 2009, 6(4): 331-342.

[53] Heib V, Becker M, Warger T, et al. Mast cells are crucial for early inflammation, migration of Langerhans cells, and CTL responses following topical application of TLR7 ligand in mice[J].Blood, 2007, 110(3): 946-953.

[54] Dudeck A, Suender CA, Kostka SL, et al. Mast cells promote Th1 and Th17 responses by modulating dendritic cell maturation and function[J].Eur J Immunol, 2011, 41(7): 1883-1893.

[55] Merluzzi S, Frossi B, Gri G, et al. Mast cells enhance proliferation of B lymphocytes and drive their differentiation toward IgA-secreting plasma cells[J].Blood, 2010, 115(14): 2810-2817.

[56] Nakae S, Suto H, Iikura M, et al. Mast cells enhance T cell activation: importance of mast cell costimulatory molecules and secreted TNF[J].J Immunol, 2006, 176(4):2238-2248.

[57] Nakae S, Suto H, Kakurai M, et al. Mast cells enhance T cell activation: Importance of mast cell-derived TNF[J].Proc Natl Acad Sci U S A, 2005, 102(18): 6467-6472.

[58] McLachlan JB, Hart JP, Pizzo SV, et al. Mast cell-derived tumor necrosis factor induces hypertrophy of draining lymph nodes during infection[J].Nat Immunol, 2003, 4(12):1199-1205.

[59] Ebert S, Becker M, Lemmermann NA, et al. Mast cells expedite control of pulmonary murine cytomegalovirus infection by enhancing the recruitment of protective CD8 T cells to the lungs[J].PLoS Pathog, 2014, 10(4):e1004100.

[60] McAlpine SM, Issekutz TB, Marshall JS. Virus stimulation of human mast cells results in the recruitment of CD56(+) T cells by a mechanism dependent on CCR5 ligands[J].FASEB J, 2012, 26(3): 1280-1289.

[61] Frandji P, Oskeritzian C, Cacaraci F, et al. Antigen-dependent stimulation by bone marrow-derived mast cells of MHC class II-restricted T cell hybridoma[J].J Immunol, 1993, 151(11): 6318-6328.

[62] Fox CC, Jewell SD, Whitacre CC. Rat peritoneal mast cells present antigen to a PPD-specific T cell line[J].Cell Immunol, 1994, 158(1): 253-264.

[63] Kambayashi T, Allenspach EJ, Chang JT, et al. Inducible MHC class II expression by mast cells supports effector and regulatory T cell activation[J].J Immunol, 2009, 182(8): 4686-4695.

[64] Gaudenzio N, Espagnolle N, Mars LT, et al. Cell-cell cooperation at the T helper cell/mast cell immunological synapse[J].Blood, 2009, 114(24): 4979-4988.

[65] Suurmond J, van Heemst J, van Heiningen J, et al. Communication between human mast cells and CD4(+) T cells through antigen-dependent interactions[J].Eur J Immunol, 2013, 43(7): 1758-1768.

[66] Stelekati E, Bahri R, D'Orlando O, et al. Mast cellmediated antigen presentation regulates CD8+ T cell effector functions[J].Immunity, 2009, 31(4): 665-676.

[67] Malaviya R, Twesten NJ, Ross EA, et al. Mast cells process bacterial Ags through a phagocytic route for class I MHC presentation to T cells[J].J Immunol, 1996, 156(4): 1490-1496.

[68] Kambayashi T, Baranski JD, Baker RG, et al. Indirect involvement of allergen-captured mast cells in antigen presentation[J].Blood, 2008, 111(3): 1489-1496.

[69] Joffre OP, Segura E, Savina A, et al. Cross-presentation by dendritic cells[J].Nat Rev Immunol, 2012, 12(8):557-569.

[70] Spicer SS, Simson JA, Farrington JE. Mast cell phagocytosis of red blood cells[J].Am J Pathol, 1975, 80(3): 481-498.

[71] Malaviya R, Ross EA, MacGregor JI, et al. Mast cell pha gocytosis of FimH-expressing enterobacteria[J].J Immunol, 1994, 152(4): 1907-1914.

[72] Della Rovere F, Granata A, Monaco M, et al. Phagocytosis of cancer cells by mast cells in breast cancer[J].Anticancer Res, 2009, 29(8): 3157-3161.

[73] Sher A, Hein A, Moser G, et al. Complement receptors promote the phagocytosis of bacteria by rat peritoneal mast cells[J].Lab Invest, 1979, 41(6): 490-499.

[74] Shin JS, Gao Z, Abraham SN. Involvement of cellular caveolae in bacterial entry into mast cells[J].Science, 2000, 289(5480): 785-788.

[75] Munoz S, Rivas-Santiago B, Enciso JA. Mycobacterium tuberculosis entry into mast cells through cholesterol-rich membrane microdomains[J].Scand J Immunol, 2009, 70(3): 256-263.

[76] Wesolowski J, Caldwell V, Paumet F. A novel function for SNAP29 (synaptosomal-associated protein of 29 kDa) in mast cell phagocytosis[J].PLoS One, 2012, 7(11): e49886.

[77] Piliponsky AM, Chen CC, Grimbaldeston MA, et al. Mast cell-derived TNF can exacerbate mortality during severe bacterial infections in C57BL/6-KitW-sh/W-sh mice[J].Am J Pathol, 2010, 176(2): 926-938.

[78] Dahdah A, Gautier G, Attout T, et al. Mast cells aggravate sepsis by inhibiting peritoneal macrophage phagocytosis[J].J Clin Invest, 2014, 124(10): 4577-4589.

[79] Chiba N, Shimada K, Chen S, et al. Mast Cells Play an Important Role in Chlamydia pneumoniae Lung Infection by Facilitating Immune Cell Recruitment into the Airway[J].J Immunol, 2015, 194(8): 3840-3851.

[80] Piliponsky AM, Chen CC, Rios EJ, et al. The chymase mouse mast cell protease 4 degrades TNF, limits inflammation, and promotes survival in a model of sepsis[J].Am J Pathol, 2012, 181(3): 875-886.

[81] Nabel GJ. Designing Tomorrow’s Vaccines[J].N Engl J Med, 2013, 368(6): 551-560.

[82] Levitz SM, Golenbock DT. Beyond empiricism: informing vaccine development through innate immunity research[J].Cell, 2012, 148(6): 1284-1292.

[83] Coffman RL, Sher A, Seder RA. Vaccine adjuvants: putting innate immunity to work[J].Immunity, 2010, 33(4): 492-503.

[84] McKee AS, Munks MW, MacLeod MK, et al. Alum induces innate immune responses through macrophage and mast cell sensors, but these sensors are not required for alum to act as an adjuvant for specific immunity[J].J Immunol, 2009, 183(7): 4403-4414.

[85] Lycke N. Recent progress in mucosal vaccine development: potential and limitations[J].Nat Rev Immunol, 2012, 12(8): 592-605.

[86] Holmgren J, Svennerholm A-M. Vaccines against mucosal infections[J].Curr Opin Immunol, 2012, 24(3):343-353.

[87] Di Tommaso A, Saletti G, Pizza M, et al. Induction of antigen-specific antibodies in vaginal secretions by using a nontoxic mutant of heat-labile enterotoxin as a mucosal adjuvant[J].Infect Immun, 1996, 64(3): 974-979.

[88] Sundling C, Schon K, Morner A, et al. CTA1-DD adjuvant promotes strong immunity against human immunodeficiency virus type 1 envelope glycoproteins following mucosal immunization[J].J Gen Virol, 2008, 89(Pt 12): 2954-2964.

[89] Casella CR, Mitchell TC. Putting endotoxin to work for us: monophosphoryl lipid A as a safe and effective vac cine adjuvant[J].Cell Mol Life Sci, 2008, 65(20):3231-3240.

[90] Mata-Haro V, Cekic C, Martin M, et al. The vaccine adjuvant monophosphoryl lipid A as a TRIF-biased agonist of TLR4[J].Science, 2007, 316(5831): 1628-1632.

[91] Bode C, Zhao G, Steinhagen F, et al. CpG DNA as a vaccine adjuvant[J].Expert Rev Vaccines, 2011, 10(4):499-511.

[92] Mizel SB, Bates JT. Flagellin as an adjuvant: cellular mechanisms and potential[J].J Immunol, 2010, 185(10):5677-5682.

[93] Svennerholm AM. From cholera to enterotoxigenic Escherichia coli (ETEC) vaccine development[J].Indian J. Med. Res., 2011, 133(2): 188-196.

[94] Zhang RG, Scott DL, Westbrook ML, et al. The threedimensional crystal structure of cholera toxin[J].J Mol Biol, 1995, 251(4): 563-573.

[95] Sanchez J, Holmgren J. Cholera toxin - a foe & a friend[J].Indian J Med Res, 2011, 133: 153-163.

[96] Tomasi M, Battistini A, Araco A, et al. The role of the reactive disulfide bond in the interaction of cholera-toxin functional regions[J].Eur J Biochem, 1979, 93(3): 621-627.

[97] Steinman RM. Decisions about dendritic cells: past, present, and future[J].Annu Rev Immunol, 2012, 30: 1-22.

[98] Eriksson A, Lycke N. The CTA1-DD vaccine adjuvant binds to human B cells and potentiates their T cell stimulating ability[J].Vaccine, 2003, 22(2): 185-193.

[99] Campbell PA, Schuffler C, Rodriguez GE. Listeria cell wall fraction: a B cell adjuvant[J].J Immunol, 1976, 116(3): 590-594.

[100] Fevrier M, Birrien JL, Leclerc C, et al. The macrophage, target cell of the synthetic adjuvant muramyl dipeptide[J].Eur J Immunol, 1978, 8(8): 558-562.

[101] Kim S, Lalani S, Parekh VV, et al. Glycolipid ligands of invariant natural killer T cells as vaccine adjuvants[J].Expert Rev Vaccines, 2008, 7(10): 1519-1532.

[102] Hall LJ, Clare S, Dougan G. NK cells influence both innate and adaptive immune responses after mucosal immunization with antigen and mucosal adjuvant[J].J Immunol, 2010, 184(8): 4327-4337.

[103] Silk JD, Hermans IF, Gileadi U, et al. Utilizing the adju vant properties of CD1d-dependent NK T cells in T cell-mediated immunotherapy[J].J Clin Invest, 2004, 114(12): 1800-1811.

[104] McLachlan JB, Shelburne CP, Hart JP, et al. Mast cell activators: a new class of highly effective vaccine adjuvants[J].Nat Med, 2008, 14(5): 536-541.

[105] Meng S, Liu Z, Xu L, et al. Intranasal immunization with recombinant HA and mast cell activator C48/80 elicits protective immunity against 2009 pandemic H1N1 influenza in mice[J].PLoS One, 2011, 6(5): e19863.

[106] Staats HF, Fielhauer JR, Thompson AL, et al. Mucosal targeting of a BoNT/A subunit vaccine adjuvanted with a mast cell activator enhances induction of BoNT/A neutralizing antibodies in rabbits[J].PLoS ONE, 2011, 6(1): e16532

[107] McGowen AL, Hale LP, Shelburne CP, et al. The mast cell activator compound 48/80 is safe and effective when used as an adjuvant for intradermal immunization with Bacillus anthracis protective antigen[J].Vaccine, 2009, 27(27):3544-3552.

[108] Zeng L, Liu Y, Wang H, et al. Compound 48/80 acts as a potent mucosal adjuvant for vaccination against Streptococcus pneumoniae infection in young mice[J].Vaccine, 2015, 33(8): 1008-1016.

[109] Wang SH, Kirwan SM, Abraham SN, et al. Stable dry powder formulation for nasal delivery of anthrax vaccine[J].J Pharm Sci, 2012, 101(1): 31-47.

[110] Bento D, Staats HF, Goncalves T, et al. Development of a novel adjuvanted nasal vaccine: C48/80 associated with chitosan nanoparticles as a path to enhance mucosal immunity[J].Eur J Pharm Biopharm, 2015, 93: 149-164.

[111] Kayamuro H, Yoshioka Y, Abe Y, et al. Interleukin-1 family cytokines as mucosal vaccine adjuvants for induction of protective immunity against influenza virus[J].J Virol, 2010, 84(24): 12703-12712.

[112] Sasaki Y, Yoshimoto T, Maruyama H, et al. IL-18 with IL-2 protects against Strongyloides venezuelensis infection by activating mucosal mast cell-dependent type 2 innate immunity[J].J Exp Med, 2005, 202(5): 607-616.

[113] Enoksson M, Lyberg K, Moller-Westerberg C, et al. Mast cells as sensors of cell injury through IL-33 recognition[J].J Immunol, 2011, 186(4): 2523-2528.

[114] Aoki R, Kawamura T, Goshima F, et al. Mast cells play a key role in host defense against herpes simplex virus infection through TNF-alpha and IL-6 production[J].J Invest Dermatol, 2013, 133(9): 2170-2179.

[115] Yoshino N, Endo M, Kanno H, et al. Polymyxins as novel and safe mucosal adjuvants to induce humoral immune responses in mice[J].PLoS One, 2013, 8(4): e61643.

[116] Van Meulder F, Van Coppernolle S, Borloo J, et al. Granule exocytosis of granulysin and granzyme B as a potential key mechanism in vaccine-induced immunity in cattle against the nematode Ostertagia ostertagi[J].Infect Immun, 2013, 81(5): 1798-1809.

[117] St John AL, Chan CY, Staats HF, et al. Synthetic mast-cell granules as adjuvants to promote and polarize immunity in lymph nodes[J].Nat Mater, 2012, 11(3): 250-257.

[118] Leal-Berumen I, Snider DP, Barajas-Lopez C, et al. Cholera toxin increases IL-6 synthesis and decreases TNF-alpha production by rat peritoneal mast cells[J].J Immunol, 1996, 156(1): 316-321.

[119] Sears SD, Richardson K, Young C, et al. Evaluation of the human immune response to outer membrane proteins of Vibrio cholerae[J].Infect Immun, 1984, 44(2): 439-444.

[120] Fujihashi K, Koga T, van Ginkel FW, et al. A dilemma for mucosal vaccination: efficacy versus toxicity using enterotoxin-based adjuvants[J].Vaccine, 2002, 20(19-20):2431-2438.

[121] Dickinson BL, Clements JD. Dissociation of Escherichia coli heat-labile enterotoxin adjuvanticity from ADP-ribosyltransferase activity[J].Infect Immun, 1995, 63(5):1617-1623.

[122] Giuliani MM, Del Giudice G, Giannelli V, et al. Mucosal adjuvanticity and immunogenicity of LTR72, a novel mutant of Escherichia coli heat-labile enterotoxin with partial knockout of ADP-ribosyltransferase activity[J].J Exp Med, 1998, 187(7): 1123-1132.

[123] Hagiwara Y, Kawamura YI, Kataoka K, et al. A second generation of double mutant cholera toxin adjuvants:enhanced immunity without intracellular trafficking[J].J Immunol, 2006, 177(5): 3045-3054.

[124] Partidos CD, Salani BF, Pizza M, et al. Heat-labile enterotoxin of Escherichia coli and its site-directed mutant LTK63 enhance the proliferative and cytotoxic T-cell responses to intranasally co-immunized synthetic peptides[J].Immunol Lett, 1999, 67(3): 209-216.

[125] Lycke N. From toxin to adjuvant: basic mechanisms for the control of mucosal IgA immunity and tolerance[J].Immunol Lett, 2005, 97(2): 193-198.

[126] Eriksson AM, Schon KM, Lycke NY. The cholera toxin-derived CTA1-DD vaccine adjuvant administered intranasally does not cause inflammation or accumulate in the nervous tissues[J].J Immunol, 2004, 173(5): 3310-3319.

[127] Cunningham KA, Carey AJ, Lycke N, et al. CTA1-DD is an effective adjuvant for targeting anti-chlamydial immunity to the murine genital mucosa[J].J Reprod Immunol, 2009, 81(1): 34-38.

[128] Eliasson DG, El Bakkouri K, Schon K, et al. CTA1-M2e-DD: a novel mucosal adjuvant targeted influenza vaccine[J].Vaccine, 2008, 26(9): 1243-1252.

[129] Andersen CS, Dietrich J, Agger EM, et al. The combined CTA1-DD/ISCOMs vector is an effective intranasal adjuvant for boosting prior Mycobacterium bovisBCG immunity to Mycobacterium tuberculosis[J].Infect Immun, 2007, 75(1): 408-416.

[130] Sundling C, Schon K, Morner A, et al. CTA1-DD adjuvant promotes strong immunity against human immunodeficiency virus type 1 envelope glycoproteins following mucosal immunization[J].J Gen Virol, 2008, 89(Pt 12):2954-2964.

[131] Nedrud JG, Bagheri N, Schon K, et al. Subcomponent vaccine based on CTA1-DD adjuvant with incorporated UreB class II peptides stimulates protective Helicobacter pylori immunity[J].PLoS One, 2013, 8(12): e83321.

[132] Marks E, Helgeby A, Andersson JO, et al. CD4(+) T-cell immunity in the female genital tract is critically dependent on local mucosal immunization[J].Eur J Immunol, 2011, 41(9): 2642-2653.

[133] Eliasson DG, Helgeby A, Schon K, et al. A novel nontoxic combined CTA1-DD and ISCOMS adjuvant vector for effective mucosal immunization against influenza virus[J].Vaccine, 2011, 29(23): 3951-3961.

[134] Agren LC, Ekman L, Lowenadler B, et al. Adjuvanticity of the cholera toxin A1-based gene fusion protein, CTA1-DD, is critically dependent on the ADP-ribosyl-transferase and Ig-binding activity[J].J Immunol, 1999, 162(4): 2432-2440.

[135] Agren L, Sverremark E, Ekman L, et al. The ADP-ribosylating CTA1-DD adjuvant enhances T cell-dependent and independent responses by direct action on B cells involving anti-apoptotic Bcl-2 and germinal center-promoting effects[J].J Immunol, 2000, 164(12): 6276-6286.

[136] Bemark M, Bergqvist P, Stensson A, et al. A unique role of the cholera toxin A1-DD adjuvant for long-term plasma and memory B cell development[J].J Immunol, 2011, 186(3): 1399-1410.

[137] Mattsson J, Yrlid U, Stensson A, et al. Complement activation and complement receptors on follicular dendritic cells are critical for the function of a targeted adjuvant[J].J Immunol, 2011, 187(7): 3641-3652.

[138] Fang Y, Larsson L, Mattsson J, et al. Mast cells contribute to the mucosal adjuvant effect of CTA1-DD after IgG-complex formation[J].J Immunol, 2010, 185(5): 2935-2941.

[139] Agren LC, Ekman L, Lowenadler B, et al. Genetically engineered nontoxic vaccine adjuvant that combines B cell targeting with immunomodulation by cholera toxin A1 subunit[J].J Immunol, 1997, 158(8): 3936-3946.

[140] Agren L, Lowenadler B, Lycke N. A novel concept in mucosal adjuvanticity: the CTA1-DD adjuvant is a B cell-targeted fusion protein that incorporates the enzymatically active cholera toxin A1 subunit[J].Immunol Cell Biol, 1998, 76(3): 280-287.

[141] Ding J, Fang Y, Xiang Z. Antigen/IgG immune complexprimed mucosal mast cells mediate antigen-specific activation of co-cultured T cells[J].Immunology, 2014, 144:387-394.

✉Corresponding author: Zou Xiang, PhD, Department of Microbiology and Immunology, University of Gothenburg, Gothenburg 40530, Sweden. Tel: +46-31-7866322, E-mail: zou.xiang@gu.se.

© 2016 by the Journal of Biomedical Research. All rights reserved.

Received 26 February 2015, Revised 08 April 2015, Accepted 26 April 2015, Epub 29 June 2015

R392.12, Document code: A

The authors reported no conflict of interests.

10.7555/JBR.30.20150038