Next Article in Journal
Stratified Mucin-Producing Intraepithelial Lesion (SMILE) of the Uterine Cervix: High-Risk HPV Genotype Predominance and p40 Immunophenotype
Previous Article in Journal
Invadopodia, a Kingdom of Non-Receptor Tyrosine Kinases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Platelets, Not an Insignificant Player in Development of Allergic Asthma

The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510260, China
*
Author to whom correspondence should be addressed.
Cells 2021, 10(8), 2038; https://doi.org/10.3390/cells10082038
Submission received: 29 June 2021 / Revised: 26 July 2021 / Accepted: 6 August 2021 / Published: 10 August 2021
(This article belongs to the Section Cellular Immunology)

Abstract

:
Allergic asthma is a chronic and heterogeneous pulmonary disease in which platelets can be activated in an IgE-mediated pathway and migrate to the airways via CCR3-dependent mechanism. Activated platelets secrete IL-33, Dkk-1, and 5-HT or overexpress CD40L on the cell surfaces to induce Type 2 immune response or interact with TSLP-stimulated myeloid DCs through the RANK-RANKL-dependent manner to tune the sensitization stage of allergic asthma. Additionally, platelets can mediate leukocyte infiltration into the lungs through P-selectin-mediated interaction with PSGL-1 and upregulate integrin expression in activated leukocytes. Platelets release myl9/12 protein to recruit CD4+CD69+ T cells to the inflammatory sites. Bronchoactive mediators, enzymes, and ROS released by platelets also contribute to the pathogenesis of allergic asthma. GM-CSF from platelets inhibits the eosinophil apoptosis, thus enhancing the chronic inflammatory response and tissue damage. Functional alterations in the mitochondria of platelets in allergic asthmatic lungs further confirm the role of platelets in the inflammation response. Given the extensive roles of platelets in allergic asthma, antiplatelet drugs have been tested in some allergic asthma patients. Therefore, elucidating the role of platelets in the pathogenesis of allergic asthma will provide us with new insights and lead to novel approaches in the treatment of this disease.

1. Introduction

Asthma is a chronic and heterogeneous pulmonary disease which affects over 300 million people around the world [1]. Asthmatic patients can vary widely in terms of clinical presentation, severity, and pathophysiology, though they generally experience similar symptoms such as coughing, wheezing, breathlessness, and reversible airway obstruction. While its prevalence used to be reserved mainly for high-income countries, it has become a major public health challenge in China due to economic advances as well as rapid changes in environment and lifestyle [2]. Due to the complexity of the disease, there are several classification standards. Asthma phenotypes can be causatively grouped into allergic asthma and nonallergic asthma, or cellularly into eosinophilic, neutrophilic, paucigranulocytic, and mixed granulocytic asthma [3]. Furthermore, it is divided into Type 2 asthma and non-Type 2 asthma based on the inflammatory cytokine profiles [4,5].
Platelets are anucleate blood cells with a diameter of 2~4 μm generated from megakaryocytes. They contain a variety of secretory granules, such as α-granules, dense granules (δ-granules), and lysosomes, which not only contain coagulation-related factors, but also inflammatory mediators and protease [6]. Platelets are well known for their roles in hemostasis and thrombosis, and now increasing evidence has highlighted their immunological roles in inflammation (Figure 1). Platelet malfunctions can lead to atherosclerosis, stroke, myocardial infarction, and deep venous thrombosis as well as allergic diseases, such as atopic dermatitis [7,8,9,10].
The involvement of platelets in the pathogenesis of asthma has been known for many years, especially in the development of allergic asthma and aspirin-exacerbated respiratory disease (AERD) [11,12]. Allergic asthma, also called atopic asthma, is the most common type of asthma with similar pathological features including mucus overproduction, chronic airway inflammation, airway remodeling, and airway hyperresponsiveness (AHR). It has been characterized as infiltration of eosinophils, mast cells, and lymphocytes in the airways and features increased secretion of type 2 cytokines such as IL-4, IL-5, and IL-13 after allergen exposure [13]. The onset of allergic asthma is influenced by both genetic backgrounds and environmental factors, and is closely related to allergic rhinitis [1,14]. Allergens that trigger allergic asthma include house dust mites, pollens, dander from cats and dogs, mold spores, cockroaches [15], and even small molecules such as toluene diisocyanate (TDI) [16].
In this review, to provide a comprehensive understanding of the relationship between platelets and allergic asthma, we will elaborate on the following aspects: (i) the current understanding of platelets’ involvement in allergic asthma; (ii) the underlying mechanism of platelets’ role in the initiation of adaptive immunity and the pathogenic development of allergic asthma; (iii) the antiplatelet therapy in asthma treatment; and (iv) new insights on platelets in asthma.

2. Current Understanding of Platelets’ Involvement in Allergic Asthma

Platelet activation in allergic asthma has been highlighted by a great number of surveys (Table 1). Disturbed hemostatic balance in the lungs of allergic asthma has been evidenced by increased levels of cellular fibronectin, a marker of vascular injury in asthmatic patients [17,18]. It is said that the imbalances between the coagulation and anticoagulation system and the fibrinolytic system are jointly involved in asthma [18]. The platelet activation markers such as β-thromboglobulin (β-TG) and platelet factor 4 (PF4), which are increased in the plasma of atopic dermatitis patients, were found to be higher when patients were afflicted with concomitant asthma and allergic rhinitis [10,19]. By intrabronchially challenging house-dust-mite (HDM)-sensitive asthmatic patients with Dermatophagoides pteronysisnus (Dp) extract, it has been shown that allergen challenge is correlated with platelet activation in vivo, manifested as decreased levels of platelet count and increased plasma levels of β-TG and PF4 [20]. The involvement of platelets in allergic asthma can be indicated by the increased platelet counts in the bronchoalveolar lavage fluid (BALF) of the patients and animal models [21]. Consistently, the increased levels of PF4 and β-TG were also detected in BALF [22]. Furthermore, platelet deposition on interalveolar septum walls was also observed in bronchial biopsies and its number was significantly increased after allergen challenge [23,24]. Moreover, platelet-leukocyte conjugates were also observed in the peripheral blood of asthmatic patients after allergen exposure [25]. Platelet activity in patients with pollen-induced seasonal allergic rhinitis and asthma, using plasma PF4 as an indicator, was found to increase during the grass pollen season and decrease in the off season [26]. It has been shown that P-selectin in the nasal lavage fluid of asthmatic patients is positively correlated with the level of eosinophil cationic protein (ECP) [27]. Furthermore, eosinophil β1-integrin activation in asthma was found to be associated with activated platelets in a P-selectin-mediated manner [28,29]. Duarte, D. et al. found a significantly higher level of platelet-derived microparticles (PMPs) in the peripheral blood of patients with allergic asthma compared to healthy individuals [30].
The expression of high-affinity IgE receptor (FcεRI) and low-affinity IgE receptors (FcεRII/CD23) on platelets provides the structural basis for platelets’ involvement in allergic asthma. Monoclonal antibodies targeting IgE receptors (anti-FcεRI and anti-CD23) or IgE binding to the asthmatic patients’ platelets (anti-IgE) induced the RANTES release by platelets and cytotoxicity against schistosomula [31,34,35,37]. It has also been demonstrated that the allergen triggered platelet activation in an IgE-FcεR-dependent pathway and induced inflammatory mediators, such as serotonin (5-HT) and RANTES, from platelets [34,35]. Research conducted on humans revealed that the percentage of IgE+ platelets in atopic asthmatic patients was twice of that in humans with a normal IgE level, which is 10% [31]. This was also evidenced by the fact that the expression levels of FcεRI and FcεRII in platelets were significantly different between sham- and OVA-immunized mice [38]. However, others reported no distinct difference in the expression levels of FcεRI on the platelets in allergic patients and healthy controls [35].
In animal models, allergen exposure resulted in platelet migration to the airways, essential for eosinophil and lymphocyte recruitment and activation [25], and the critical role of P-selectin in platelets in the development of allergen-induced airway response was demonstrated in ovalbumin (OVA) and cockroach-induced murine asthmatic models [43,44]. Pitchford, S.C. et al. [38] showed that in OVA-sensitized mice, platelets migrated out of the blood vessels in an allergen-IgE-FcεRI pathway rather than due to hemorrhage after the allergen challenge. This migration of platelets occurred ahead of the infiltration of leukocytes into the lungs and was in single non-aggregated forms, which is different from the commonly observed aggregated state in thrombosis and hemostasis [38]. Due to the expression of chemokine receptors on the platelet surfaces, it is considered that they undergo chemotaxis in response to chemokines such as CCL11, CCL22, and CXCL12. Indeed, a recent study showed that CCR3 (CCL11 receptor) is essential for the recruitment of platelets into asthmatic lungs in the models of allergic inflammation. Under intravital microscopy, the rolling, adhesion, and extravascular migration of platelets in the HDM-sensitized murine models were markedly suppressed by SB328437, a CCR3 antagonist [36]. A study on platelet degranulation function in hemostasis and inflammation revealed that AHR and eosinophilic inflammation were significantly diminished in platelet-specific Munc13-4 KO mice, as the dense granule release was abrogated in these mice [45]. This evidence shows that platelets actively participate in the development of allergic asthma.
It is worth noting that while some have argued that allergens directly activate platelets based on the platelets’ anti-parasite cytotoxicity, others could not detect allergen-induced platelet aggregation and degranulation [39,46]. These contradicting results indicate that the underlying mechanism of platelets’ response to allergen is yet to be clarified. Interestingly, the work by Kasperska-Zajac, A. et al. found no significant differences in the plasma levels of PF-4 and β-TG in HDM-allergic patients and seasonal allergic rhinitis patients versus healthy non-atopic subjects [47,48]. Contrary to the notion that the vascular endothelial growth factor (VEGF) level is increased in plasma of allergic patients with asthma and atopic dermatitis, the work by Koczy-Baron, E. et al. showed that the free circulating VEGF level did not change in patients with chronic allergic rhinitis [49]. These discrepancies indicate that differences may exist in the platelet activity among patients with distinct clinical manifestations of atopy. This intriguing phenomenon was discussed in the review by Potaczek, D.P. [50].

3. Mechanisms of Platelets’ Role in the Pathogenesis of Allergic Asthma

It is now widely accepted that the intrapulmonary migration of platelets is a critical process for the development of allergic asthma because it not only promotes the recruitment of eosinophils and other leukocytes from the blood vessels into lung tissues but also serves as a reservoir of inflammatory mediators to promote the pathogenic process of allergic asthma [38]. In this section, we will discuss how intrapulmonary platelets contribute to the pathogenesis development of asthma.

3.1. Platelets in Adaptive Immune Response Induction

Type 2 immune response is recognized as an important mechanism in allergic diseases [51]. In recent years, the role of platelets in the induction of Type 2 immune response has attracted increasing attention. It is well established that dendritic cells (DCs) are the important antigen-presenting cells (APCs) which initiate and maintain Type 2 immune responses [52]. Amison, R.T. et al. demonstrated that platelets play a pivotal role in the sensitization stage of allergic asthma. They discovered that platelets rely on an IgE-FcεRI-dependent pathway to interact with pulmonary CD11c+ mononuclear cells (for example, CD11c+ DCs) which present antigen signals to T cells in bronchial lymph nodes and induce the formation of Type 2 immune response. When platelets were temporarily depleted during the sensitization period, the OVA-sensitized mice failed to develop an effective Type 2 immune response [53]. However, the underlying mechanism of the interaction between platelet and APC through the allergen-IgE-FcεRI axis remains obscure. On the other hand, elegant experiments by Dürk and colleagues ruled out the notion that platelets enhance the function of mature DCs in polarizing Th2 cells by secreting 5-HT and thus promoting the formation of Type 2 immunity [54,55]. It was reported that platelet activation promoted Th2 response and the development of asthma by upregulating the expression of CD40L (CD154) and partially inhibiting Foxp3+ regulatory T cell, resulting in a polarized Th2 response in allergic asthma [56]. Nakanishi, T. et al. put forward the idea that activated platelets participate in the Th2 immune response process in a CD154-independent fashion. They found that activated platelets could interact with thymic-stromal-lymphopoietin (TSLP)-stimulated myeloid DCs (TSLP-mDCs) by expressing RANK ligand (RANKL) to promote the maturation of TSLP-mDCs through the RANKL-RANK pathway. Mature TSLP-mDCs not only promoted the differentiation of naive T cells into Th2 cells but also drove the chemotaxis of memory Th2 cells to inflammatory sites by secreting chemokine CCL17, thereby maintaining the Th2-cell-mediated allergic condition [42].
Apart from Th2 cells, other mucosal innate immune cells were found to contribute to allergic asthma as well. Group 2 innate lymphoid cells (ILC2s), abundant in the lungs, intestines, and skin, serve as a source of type 2 cytokines alongside B and T cells [57]. Environmental allergens can damage the airway epithelial barrier and lead to the secretion of epithelial-derived alarmins such as IL-25, IL-33, and TSLP, which activate ILC2s. Takeda, T. et al. [58] confirmed that IL-33 is constitutively expressed on human platelets and megakaryocytes and could act as an essential regulator of eosinophilic inflammation in the airway. In the papain-induced murine airway eosinophilic inflammation model, depletion of platelets resulted in a remarkable decrease in the eosinophil counts in the BALF. Conversely, administration of wild-type platelets into IL-33–deficient mice significantly restored the eosinophil infiltration in an IL-33-dependent pathway. Based on these discoveries, they proposed that platelets might play an important role in airway Type 2 inflammation as an important cellular source of IL-33 [58]. In addition, platelets of asthmatic patients are thought to participate in tissue damage and repair via the Wnt signaling pathway. Chae, W.J. et al. [59] found that circulating Dickkopf-1 (DKK-1), which is mainly produced by platelets, inhibits the Wnt signaling pathway in type-2-cell-mediated inflammation. Upon the allergen challenge, the increase in circulating DKK-1 facilitated leukocyte migration and promoted Th2 cell differentiation and Type 2 cytokine production through the mTOR and MAPK signaling pathways [59].

3.2. Platelets in the Recruitment of Inflammatory Cells

Leukocyte infiltration in the airways during allergic asthma is initiated by chemokines from immune cell milieu or adhesion molecules expressed on cell surfaces. Platelets are involved in the pathogenesis of allergic asthma (Table 2). There are nearly 300 different proteins stored in platelet α-granules, such as CXCL1, PF4, β-TG, CXCL5, CXCL7, CXCL12, macrophage inflammatory protein-1α (MIP-1α), and RANTES (CCL5), which play an important role in leukocyte infiltration and inflammatory response [60]. In allergic diseases, PF4 induces the expression of IgG and IgE receptors, stimulates the release of histamine of basophils, activates eosinophils and other inflammatory cells, upregulates the expression of adhesion molecules, and facilitates adhesion between leukocytes and the endothelia, thus exacerbating allergic reactions [61]. RANTES (CCL5) binds to MIP-1α to recruit monocytes to the inflammatory site, attracts eosinophils and induces their degranulation and respiratory burst, promotes T cell activation and maturation, and recruits memory T cells to the lungs [62,63].
Among the adhesion molecules, P-selectin is the most well-studied glycoprotein and is found in α-granules of resting platelets and Weibel–Palade bodies of endothelial cells. It is a platelet activation surface maker induced by inflammatory or pro-aggregatory stimuli [66]. Platelets bind to eosinophils through the interaction between P-selectin and PSGL-1 and form platelet-leukocyte aggregates to mediate eosinophil migration into the lungs, thus triggering eosinophilic airway inflammation. This platelet and eosinophil interaction activates eosinophils to upregulate the expression of the integrins α4β1 (VLA-4) and αMβ2 (CD11b/CD18, Mac-1), which mediate the attachment of eosinophils to endothelia and thus promote recruitment of eosinophils to the inflammatory site [67].
Intriguingly, the formation of platelet-leukocyte aggregates influences the leukotriene synthesis in leukocytes as well. The role of cysteinyl leukotriene (CysLTs) and their receptors in the development of allergic asthma is highlighted in various studies. Activated platelets enhance the arachidonic acid metabolism, resulting in the increased production of CysLTs such as LTC4, LTD4, and LTE4. This process is considered to be an important mechanism in the pathogenesis of AERD [68]. AERD, accounting for a greater percentage of patients with severe asthma, is characterized by respiratory hyperreactions upon ingestion of COX-1 inhibitors and CysLTs overproduction, which are triggered by low doses of aspirin that inhibit COX-1 (cyclooxygenase-1) in platelets [69,70]. It represents a prototypical non-allergic endotype of asthma, along with a classic triad of symptoms (asthma, chronic rhinosinusitis with nasal polyposis, and hypersensitivity to aspirin and other cyclooxygenase-1 inhibitors) [71]. AERD is associated with increased CysLTs production and CysLTs receptor expression [72]. Elevated levels of platelet activation and the formation of platelet-leukocyte aggregates were observed in patients with AERD [69]. Leukocyte-adhering platelets convert the leukocyte-derived precursor leukotriene LTA4 to LTC4 through LTC4 synthase [68]. Prostaglandin E2 (PGE2), a downstream product of the COX-1 pathway which negatively regulates CysLT production by suppressing 5-lipoxygenase (5-LO) activity of leukocytes, was decreased by the COX-1 inhibitor and thus contributed to the CysLTs overproduction [72]. It is worth noting that platelets form aggregates with neutrophils and lymphocytes as well. Although allergic asthma is mostly eosinophilic, different degrees of neutrophil recruitment caused by allergen stimulation were observed in the airway of allergic asthma patients and asthmatic mouse models [73,74]. Activated platelets promoted the production of neutrophil superoxide anions in a P-selectin-dependent pathway [41]. The purine receptor P2Y1 on platelets also regulates leukocyte recruitment in allergic mice through the RhoA signaling pathway [65]. Moreover, myl9/12 secreted by platelets formed intravascular net-like structures that can bind to CD69+ CD4+ T cells [64].

3.3. The Roles of Platelets in Airway Hyperresponsiveness (AHR), Airway Remodeling, and Bronchoconstriction

Platelets can synthesize and release spasmogen, such as histamine, platelet activating factor (PAF), 5-HT, and thromboxane A2 (TxA2), that act on the airway smooth muscle cells to cause bronchoconstriction [75,76]. Depletion of platelets reduced allergen-induced airway hyperreactivity in allergic rabbits [77]. Studies on guinea pigs have shown that Bradykinin and capsaicin induced airways obstruction in a platelet-dependent manner [78]. PAF, a phospholipid derivative from various cells, including platelets, mast cells, basophils, eosinophils, and so on, is a strong activator of platelets and is well acknowledged for its role in bronchoconstriction, bronchial hyperresponsiveness, mucus hypersecretion, and gas exchange impairment [79]. TxA2, a potent airway smooth muscle contractile agent predominantly produced by platelets, is associated with airway inflammation and bronchial hyperreactivity [76]. Platelets act as a major reservoir and means of transport for brain-derived neurotrophic factor (BDNF). BDNF was found to contribute to airway obstruction and hyperresponsiveness in a model of allergic asthma [80]. Increased BDNF concentrations in platelets of asthmatic patients were found to correlate with the clinical parameters of airway dysfunction [32]. Recently, it was reported that the sphingolipid metabolism was altered in the patients allergic to house dust mites. Activated platelets are rich sources of sphingosine-1-phosphate (S1P), which acts directly on the smooth muscle cells to promote proliferation and AHR. The severity of allergen-induced bronchoconstriction is highly correlated with the plasma concentration of S1P [33].
Platelets are also rich in proteases, mitogens, and growth factors, which not only participate in the damage and repair process of allergic asthma, but also affect the phenotypes of airway epithelial cells, fibroblasts, and airway smooth muscle cells [81]. Metalloproteases (e.g., MMP2, MMP9), free radicals, and cationic proteins can degrade extracellular matrix, increase the vascular permeability of airway epithelia, and stimulate mucus secretion [60]. In murine models of chronic allergic inflammation, platelets are indispensable for the structural remodeling of the airway after chronic exposure to aerosolized allergens [23]. Platelet-derived growth factor (PDGF) promotes the proliferation of human airway smooth muscle cells as a mitogen, but is also involved in airway fibrosis and airway remodeling as a strong chemokine of fibroblasts [82,83,84]. In a mouse model with repeated exposure to allergens, PDGF overexpression and airway smooth muscle thickening were observed [85]. Vascular endothelial growth factor (VEGF) induces endothelial cell growth and angiogenesis and increases vascular permeability, and platelets secrete VEGF upon activation in vivo [86]. A study showed that VEGF levels are associated with the degree of vascularity and are inversely correlated with the airway caliber and level of AHR [87]. In allergic asthma, platelets inhibit eosinophil apoptosis by secreting granulocyte-macrophage colony-stimulating factor (GM-CSF), thus extending chronic inflammatory response and tissue damage [40]. In addition, GM-CSF enhances the 5-lipoxygenase (5-LO) activity in neutrophils and eosinophils and promotes the production of CysLTs, causing airway smooth muscle contraction, inflammatory cell recruitment, and tissue edema [68].
In conclusion, platelets interact with multiple inflammatory cells vital in the inflammatory process and development of allergic asthma occurs through direct cell-to-cell contact or inflammatory mediators, thus deeply involved in the pathogenesis of allergic asthma (Figure 2).

4. Antiplatelet Therapies for Asthma Control

Currently, the treatments for allergic asthma include various strategies, including anti-inflammatory agents, bronchodilators, allergen-specific immunotherapy, and biologics targeting eosinophil activation and cytokines production, such as anti-IL-5 therapy. However, these strategies often do not result in full resolution for most asthmatic endotypes [88]. Hence, a new therapy to treat allergic asthma is urgently needed. Given that platelets are extensively involved in the pathogenesis of allergic asthma, it is possible in principle to treat allergic asthma with antiplatelet drugs (Table 3). Common antiplatelet drugs include ADP receptor antagonists, thromboxane synthase inhibitors, thromboxane-prostanoid receptor (TP receptor) antagonists, 5-HT modifier, and cyclooxygenase (COX) inhibitors.
Platelets express four different purinergic receptors, P2Y1, P2Y12, P2Y14, and P2X1, on their surface and they play critical roles in hemostasis, thrombosis, and inflammation [98]. The G-protein-couple receptors (GPCRs) P2Y1 and P2Y12, when stimulated by ADP, induce platelet activation and aggregation. P2X1 is a ligand-gated ion channel and induces calcium mobilization and platelet shape change when activated by ATP [99]. The pyrimidine nucleotide-sensitive P2Y14 is a newly found P2 receptor whose function is still unknown [100]. Mechanistically, purine and pyrimidine nucleotides, released by damaged cells from inflammatory, traumatic, and ischemic conditions, interact with P2 receptors on platelets to assist in stabilizing platelet aggregation as well as recruiting and activating leukocytes. Blocking these receptors may prevent adverse cardiovascular events as well as secretion of proinflammatory mediators [101]. Clopidogrel, Prasugrel, and Ticagrelor are commonly used selective P2Y12 receptor antagonists and antithrombotic agents for various cardiovascular diseases [102,103]. P2Y12 receptor antagonists can also limit the release of platelets and the formation of platelet-leukocyte aggregates [89,90]. Clopidogrel inhibited eosinophilic inflammation and airway hyperreactivity in OVA-sensitized mice [91]. Combined use of Clopidogrel and Montelukast had a synergistic effect in asthmatic treatment [104]. However, Clopidogrel and other P2Y12 receptor antagonists, such as MRS2395 and AR-C66096, did not inhibit the infiltration of leukocytes in pulmonary inflammatory responses [65,105]. A proof-of-concept, randomized, controlled study found that Prasugrel only slightly reduced the burden of bronchial inflammation [92]. Therefore, larger-scale clinical studies are needed for P2Y12 receptor antagonists in the treatment of allergic asthma. Platelet P2Y1 receptors also participate in the pathological process of allergic asthma. Different ligands activate different P2Y1 receptor signaling pathways to promote inflammation by binding to different sites of P2Y1 receptors. The selective P2Y1 receptor antagonists MRS2179 and MRS2500 inhibited the recruitment of eosinophils and lymphocytes in the lungs of allergic inflammatory mice [65]. Therefore, the crystal structure of P2Y1 receptor is of great significance for designing a new generation of specific P2Y1 receptor antagonists to block the initiation or amplification of inflammatory signaling pathways [106]. Although the mechanisms of purinergic receptors in allergic asthma require further study, they are potential therapeutic targets for allergic asthma.
TxA2, a lipid metabolite produced by activated platelets through the arachidonic acid metabolic pathway, is an important proinflammatory mediator. In allergic asthma, TxA2 causes bronchial contraction, increased vascular permeability, tissue edema, and airway hyperresponsiveness [107]. Blocking TxA2 synthesis reduces the concentration of TxA2 during asthma attacks, but another bronchoconstrictor, prostaglandin, which acts together with TxA2 by binding to the TxA2 receptor (TP receptors), is generated due to the diverted arachidonic acid metabolism pathway [108]. Therefore, the combined use of TxA2 synthase inhibitor and TP receptor antagonist could be more effective in the treatment of allergic asthma. Indeed, the administration of Ozagrel (OKY-046, a selective TxA2 synthase inhibitor) and S-1452 (a TP receptor antagonist) inhibited the production of proinflammatory cytokines and prevented the infiltration of eosinophils into the airway [93]. Similarly, Seratrodast (AA-2414), a TP receptor antagonist, was reportedly able to reduce bronchial hyperresponsiveness by reducing airway inflammation [95]. ONO-1301, a novel prostacyclin agonist and TxA2 synthase inhibitor, was shown to suppress AHR and airway inflammation in asthma [94]. While TxA2 synthetase inhibitors and TP receptor antagonists were found to have positive effects on allergic asthma in Japan [109], studies from Western countries revealed no obvious curative effects. This discrepancy may be associated with the genetic polymorphism in TxA2 synthetase and TP receptors [108].
It has been demonstrated that platelets are an important source of 5-HT in peripheral blood. 5-HT is stored in the δ-granules of platelets after the uptake from intestinal enterochromaffin cells (ECs) and is released after platelet activation [110,111]. In patients with allergic asthma, the concentration of 5-HT in the lungs is closely correlated with the concentration of platelets in the blood. The increased concentration of 5-HT in the plasma is observed in patients with symptomatic asthma, and the level of free 5-HT is closely related to disease severity and pulmonary function [112]. Tianeptine enhances the uptake of free 5-HT by 5-HT-specific transporter in the resting platelets in peripheral blood for storage in δ-granules via 5-HT-specific transporters [96,113]. Tianeptine was effective in controlling asthma in two double-blind placebo cross-trials and in an open study covering 25,000 asthma patients over seven years [97].
It is noteworthy that aspirin, a member of NSAIDs that is commonly used to prevent thrombosis, is a selective COX-1 inhibitor which plays an antithrombotic role by inhibiting TxA2 synthesis in platelets [114]. Clinical studies have revealed that some patients with allergic asthma show intolerance to aspirin and may even develop AERD, mainly due to the significantly reduced PGE2 synthesis. Mast cell activation and the interaction between platelets and granulocytes lead to overexpression of CysLTs. All of these factors contribute to constriction of the bronchioles, acute exacerbation of asthma, and increased mucus production in AERD [115,116]. Therefore, aspirin should be used with caution when treating allergic asthma. Presently, AERD treatments include intravenous corticosteroids, anti-IgE, anti-IL-4/13, aspirin desensitization, and leukotriene-modifying drugs (leukotriene receptor antagonists and 5-lipooxygenase inhibitors) [70,71]. Due to the elevated levels of platelet activation and platelet-leukocyte aggregates in AERD, anti-platelet therapy would be an efficacious treatment, although clinical trials exploring the potential for platelet-targeted therapies in AERD are still in the early stages [11].
The application of antiplatelet drugs to allergic asthma needs to be further studied. Page CP pointed out in 1988 that platelets use different signaling pathways during hemostatic and inflammatory response. In an inflammatory response, inflammatory factors evoke platelet activation, and platelets participate in the inflammatory response through adhesion and secretion activities, but this generally does not include the aggregatory response [117]. Consistent with this notion, clinical studies found minor coagulation defects and prolonged bleeding time in patients with allergic asthma. Therefore, it is of great value to further study the underlying mechanism of platelets in the inflammatory process, which can provide a new direction for new therapeutics for allergic asthma.

5. New Insights of Platelets in Asthma

The complement system, an important part of innate immunity which acts quickly on the site of inflammation, has been found to play a role in the development of allergic asthma, especially the anaphylatoxins (C3a and 5a) [118]. For example, a study showed that C3 activates platelets to release 5-HT [119]. Growing evidence supports the hypothesis that the complement system connects inflammation and thrombosis [120,121]. Moreover, platelet activation leads to complement activation through different mechanisms, such as the binding of P-selectin with C3b [122], indicating a possible interaction between platelet and complement system in allergic asthma. However, studies on the relationship between the complement system and platelets are limited, so it will be very interesting to further explore this relationship.
Similarly, alveolar macrophages (AMs), residing in the epithelial surface of airways and lungs, are considered to be the key sensors and effectors for the changing environment [123]. Macrophage polarization is found to have a profound impact on asthma pathogenesis. Increased polarization and activation of M2 macrophage promote the pathogenesis of allergic asthma [124]. The interaction between platelets and macrophages has not been extensively explored so far. A recent article revealed that in bacteria-induced pulmonary inflammation, platelets educate AMs toward an anti-inflammatory phenotype (featured as CD68+ CD163+ AMs) that mediates remission [125].
Numerous studies have shown that mitochondrial dysfunction is an active contributor to the pathological condition and drives disease progression of asthma. The oxygen-rich environment and large contact areas of the alveoli render blood components, including platelets, sensitive to oxidative stress and damage [126]. Compared with healthy subjects, platelets from asthmatic patients are less dependent on glycolysis but are more dependent on TCA cycle which was evidenced by the increased activity of Krebs cycle enzymes [127]. Since the bioenergetics of platelets in asthmatic patients are altered, it was proposed that bioenergetics of circulating platelets could mirror those of airway epithelia in healthy and asthmatic individuals [126]. Thus, platelets are potential markers to monitor bioenergetic changes in asthmatic patients.
Up to now, research on the relationship between platelets and asthma mainly focused on Type 2 immunity. However, it is now generally believed that Type 2 immune-mediated response alone cannot explain the heterogeneity of asthma. Asthmatic patients showed increased neutrophils and Th17 cytokines in the BALFs [128], and platelet-derived mediators, such as CXCL1, CXCL4, and CCL5, promote the differentiation of Th17 cells [129,130]. Furthermore, platelets differentially regulate proliferation of CD4+ effector T cells (Th1, Th17, and Treg) and thus induce distinct dynamics of immune response [131].
The discovery of megakaryocytes (MKs), the precursors of platelets, in the lungs further confirmed the role of platelets in inflammation [132]. An article that was published in Nature in 2017 [133] first provided direct evidence that the lung acts as a major site of platelet biogenesis and as storage for hematopoietic progenitors. From their work, they visualized large numbers of megakaryocytes circulating through the lungs. These megakaryocytes migrated from extrapulmonary sites such as bone marrow and accounted for nearly 50% of the total platelet production. In addition, through RNA-seq analysis, they found that lung megakaryocytes were prone to an innate immunity function [133]. Yeung, A.K. et al. recently provided further evidence for this finding [134]. Resident lung MKs express higher levels of immune molecules compared to those in the bone marrow, supporting the notion that lung MKs have similar gene expression patterns as APCs. For example, lung MKs expressed higher levels of TLR2, TLR4, and MHC-II-associated molecules, which are involved in microbial surveillance and antigen presentation [134]. Lung MKs are also able to induce CD4+ T cells activation and modulate immune response in an MHC-II-dependent manner [135]. These data suggest that the platelets-megakaryocytes system may act as an integral part of the host defense system.
The mechanism by which platelets contribute to the development of allergic asthma is mainly conducted on the mouse models due to the similarity in the coagulation system between mouse and human [136]. However, there are also clear differences in the immune system between mouse and human [137,138]. Thus, developing humanized mouse models could serve as an important preclinical tool for asthma research.

6. Conclusions

Allergic asthma involves a variety of inflammatory cells and factors and its pathogenesis is complex. Platelets are anucleate cells and their numbers are more than 10 times that of white blood cells. They carry a variety of granules and special structures like open canalicular system (OCS) and dense tubular system (DTS). As a rich reservoir of inflammatory factors, they are highly reactive and secretory, and are of significant importance in the immune system. In allergic asthma, platelets can not only facilitate leukocytes to migrate into the lung, but also polarize adaptive immune response through a variety of possible ways: they promote differentiation and activation of Th2 and ILC2s, promote IgM to IgE switch, inhibit apoptosis of eosinophils in the lung, and induce allergy to innocuous allergens in the environment. Though a large number of studies suggest that platelets are involved in the development of allergic asthma, the exact molecular mechanisms are still not fully understood, especially the complex processes of allergen-specific activation of platelets and the induction of adaptive immune response as well as the development of AHR. Furthermore, the bioenergetic changes in platelets in asthma and the finding of platelet precursors, megakaryocytes, in lungs suggest the importance of studying the coagulation and hemostasis system in asthma. A powerful method for studying the pulmonary immune environment is now available with the advent of intravital imaging [139]. Therefore, exploring the role of platelets in the development of allergic asthma will not only enable us to have a more comprehensive understanding of the pathogenesis of allergic asthma, but will also provide new and better treatments for allergic asthma.

Author Contributions

Conceptualization, L.L. and J.Z.; writing—original draft preparation, L.L.; writing—review and editing, L.L., J.Z. and J.L.; supervision, A.T. and J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Science and Technology Major Project of China (2016ZX08011-005) and the National Natural Science Foundation of China (81871266).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Mandy Yang and Kuan-Hung Chen at Guangzhou Medical University for critical reading and scientific discussion.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5-LO5-Lipoxygenase
5-HTSerotonin
AHRAirway hyperresponsiveness
AERDAspirin-exacerbated respiratory disease
AMsAlveolar macrophages
APCsAntigen-presenting cells
BALFBronchoalveolar lavage fluid
BDNFBrain-derived neurotrophic factor
COXCyclooxygenase
cysLTsCysteinyl leukotriene
DCsDendritic cells
DKK-1Dickkopf-1
DTSDense tubular system
DpDermatophagoides pteronysisnus
ECPEosinophil cationic protein
ECsEnterochromaffin cells
GM-CSFGranulocyte-macrophage colony-stimulating factor
GPCRsG-protein-couple receptors
HDMHouse dust mite
ILC2Group 2 innate lymphoid cells
MIP-1αMacrophage inflammatory protein-1α
MKsMegakaryocytes
NSAIDsNon-steroidal anti-inflammatory drugs
OCSOpen canalicular system
OVAOvalbumin
PAFPlatelet-activating factor
PDGFPlatelet-derived growth factor
PEAPlatelet-eosinophil aggregate
PF4Platelet factor 4
PGsProstaglandins
PGE2Prostaglandin E2
PMPsPlatelet-derived microparticles
PNAPlatelet-neutrophil aggregate
RANKLRANK ligand
S1PSphingosine-1-phosphate
TDIToluene diisocyanate
TP receptorThromboxane-prostanoid receptor
TSLPThymic stromal lymphopoietin
TSLP-mDCTSLP-stimulated myeloid DCs
TxA2Thromboxane A2
VEGFVascular endothelial growth factor
β-TGβ-thromboglobulin

References

  1. Holgate, S.T.; Wenzel, S.; Postma, D.S.; Weiss, S.T.; Renz, H.; Sly, P.D. Asthma. Nat. Rev. Dis. Primers 2015, 1, 15025. [Google Scholar] [CrossRef]
  2. Huang, K.; Yang, T.; Xu, J.; Yang, L.; Zhao, J.; Zhang, X.; Bai, C.; Kang, J.; Ran, P.; Shen, H.; et al. Prevalence, risk factors, and management of asthma in China: A national cross-sectional study. Lancet 2019, 394, 407–418. [Google Scholar] [CrossRef]
  3. Radermecker, C.; Louis, R.; Bureau, F.; Marichal, T. Role of neutrophils in allergic asthma. Curr. Opin. Immunol. 2018, 54, 28–34. [Google Scholar] [CrossRef] [PubMed]
  4. Kuo, C.S.; Pavlidis, S.; Loza, M.; Baribaud, F.; Rowe, A.; Pandis, I.; Hoda, U.; Rossios, C.; Sousa, A.; Wilson, S.J.; et al. A Transcriptome-driven Analysis of Epithelial Brushings and Bronchial Biopsies to Define Asthma Phenotypes in U-BIOPRED. Am. J. Respir. Crit. Care Med. 2017, 195, 443–455. [Google Scholar] [CrossRef] [Green Version]
  5. Agache, I.; Rogozea, L. Endotypes in allergic diseases. Curr. Opin. Allergy Clin. Immunol. 2018, 18, 177–183. [Google Scholar] [CrossRef] [Green Version]
  6. Thon, J.N.; Italiano, J.E. Platelets: Production, morphology and ultrastructure. In Antiplatelet Agents. Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2012; pp. 3–22. [Google Scholar] [CrossRef]
  7. Pitchford, S.C. Defining a role for platelets in allergic inflammation. Biochem. Soc. Trans. 2007, 35, 1104–1108. [Google Scholar] [CrossRef] [Green Version]
  8. Gremmel, T.; Frelinger, A.L., 3rd; Michelson, A.D. Platelet Physiology. Semin. Thromb. Hemost. 2016, 42, 191–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Xu, X.R.; Zhang, D.; Oswald, B.E.; Carrim, N.; Wang, X.; Hou, Y.; Zhang, Q.; Lavalle, C.; McKeown, T.; Marshall, A.H.; et al. Platelets are versatile cells: New discoveries in hemostasis, thrombosis, immune responses, tumor metastasis and beyond. Crit. Rev. Clin. Lab. Sci. 2016, 53, 409–430. [Google Scholar] [CrossRef] [PubMed]
  10. Tamagawa-Mineoka, R.; Katoh, N.; Ueda, E.; Masuda, K.; Kishimoto, S. Elevated platelet activation in patients with atopic dermatitis and psoriasis: Increased plasma levels of beta-thromboglobulin and platelet factor 4. Allergol. Int. 2008, 57, 391–396. [Google Scholar] [CrossRef] [Green Version]
  11. Laidlaw, T.M.; Boyce, J.A. Platelets in patients with aspirin-exacerbated respiratory disease. J. Allergy Clin. Immunol. 2015, 135, 1407–1414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Idzko, M.; Pitchford, S.; Page, C. Role of platelets in allergic airway inflammation. J. Allergy Clin. Immunol. 2015, 135, 1416–1423. [Google Scholar] [CrossRef]
  13. Morianos, I.; Semitekolou, M. Dendritic Cells: Critical Regulators of Allergic Asthma. Int. J. Mol. Sci. 2020, 21, 7930. [Google Scholar] [CrossRef]
  14. Khan, D.A. Allergic rhinitis and asthma: Epidemiology and common pathophysiology. Allergy Asthma Proc. 2014, 35, 357–361. [Google Scholar] [CrossRef]
  15. Schatz, M.; Rosenwasser, L. The allergic asthma phenotype. J. Allergy Clin. Immunology. Pract. 2014, 2, 645–648. [Google Scholar] [CrossRef] [PubMed]
  16. Chen, S.; Deng, Y.; He, Q.; Chen, Y.; Wang, D.; Sun, W.; He, Y.; Zou, Z.; Liang, Z.; Chen, R.; et al. Toll-like Receptor 4 Deficiency Aggravates Airway Hyperresponsiveness and Inflammation by Impairing Neutrophil Apoptosis in a Toluene Diisocyanate-Induced Murine Asthma Model. Allergy Asthma Immunol. Res. 2020, 12, 608–625. [Google Scholar] [CrossRef]
  17. Bazan-Socha, S.; Kuczia, P.; Potaczek, D.P.; Mastalerz, L.; Cybulska, A.; Zareba, L.; Kremers, R.; Hemker, C.; Undas, A. Increased blood levels of cellular fibronectin in asthma: Relation to the asthma severity, inflammation, and prothrombotic blood alterations. Respir. Med. 2018, 141, 64–71. [Google Scholar] [CrossRef]
  18. De Boer, J.D.; Majoor, C.J.; Van ’t Veer, C.; Bel, E.H.; Van der Poll, T. Asthma and coagulation. Blood 2012, 119, 3236–3244. [Google Scholar] [CrossRef]
  19. Nastałek, M.; Potaczek, D.P.; Wojas-Pelc, A.; Undas, A. Plasma platelet activation markers in patients with atopic dermatitis and concomitant allergic diseases. J. Dermatol. Sci. 2011, 64, 79–82. [Google Scholar] [CrossRef] [PubMed]
  20. Kowal, K.; Pampuch, A.; Kowal-Bielecka, O.; DuBuske, L.M.; Bodzenta-Lukaszyk, A. Platelet activation in allergic asthma patients during allergen challenge with Dermatophagoides pteronyssinus. Clin. Exp. Allergy J. Br. Soc. Allergy Clin. Immunol. 2006, 36, 426–432. [Google Scholar] [CrossRef]
  21. Metzger, W.J.; Sjoerdsma, K.; Richerson, H.B.; Moseley, P.; Zavala, D.; Monick, M.; Hunninghake, G.W. Platelets in bronchoalveolar lavage from asthmatic patients and allergic rabbits with allergen-induced late phase responses. Agents Actions Suppl. 1987, 21, 151–159. [Google Scholar] [CrossRef] [PubMed]
  22. Averill, F.J.; Hubbard, W.C.; Proud, D.; Gleich, G.J.; Liu, M.C. Platelet activation in the lung after antigen challenge in a model of allergic asthma. Am. Rev. Respir. Dis 1992, 145, 571–576. [Google Scholar] [CrossRef] [PubMed]
  23. Pitchford, S.C.; Riffo-Vasquez, Y.; Sousa, A.; Momi, S.; Gresele, P.; Spina, D.; Page, C.P. Platelets are necessary for airway wall remodeling in a murine model of chronic allergic inflammation. Blood 2004, 103, 639–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Tutluoglu, B.; Gurel, C.B.; Ozdas, S.B.; Musellim, B.; Erturan, S.; Anakkaya, A.N.; Kilinc, G.; Ulutin, T. Platelet function and fibrinolytic activity in patients with bronchial asthma. Clin. Appl. Thromb. Hemost. 2005, 11, 77–81. [Google Scholar] [CrossRef] [PubMed]
  25. Pitchford, S.C.; Yano, H.; Lever, R.; Riffo-Vasquez, Y.; Ciferri, S.; Rose, M.J.; Giannini, S.; Momi, S.; Spina, D.; O’Connor, B.; et al. Platelets are essential for leukocyte recruitment in allergic inflammation. J. Allergy Clin. Immunol. 2003, 112, 109–118. [Google Scholar] [CrossRef]
  26. Kasperska-Zajac, A.; Brzoza, Z.; Rogala, B. Seasonal changes in platelet activity in pollen-induced seasonal allergic rhinitis and asthma. J. Asthma Off. J. Assoc. Care Asthma 2008, 45, 485–487. [Google Scholar] [CrossRef]
  27. Benton, A.S.; Kumar, N.; Lerner, J.; Wiles, A.A.; Foerster, M.; Teach, S.J.; Freishtat, R.J. Airway platelet activation is associated with airway eosinophilic inflammation in asthma. J. Investig. Med. Off. Publ. Am. Fed. Clin. Res. 2010, 58, 987–990. [Google Scholar] [CrossRef]
  28. Johansson, M.W.; Han, S.T.; Gunderson, K.A.; Busse, W.W.; Jarjour, N.N.; Mosher, D.F. Platelet activation, P-selectin, and eosinophil beta1-integrin activation in asthma. Am. J. Respir. Crit. Care Med. 2012, 185, 498–507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Johansson, M.W.; Mosher, D.F. Activation of beta1 integrins on blood eosinophils by P-selectin. Am. J. Respir. Cell Mol. Biol. 2011, 45, 889–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Duarte, D.; Taveira-Gomes, T.; Sokhatska, O.; Palmares, C.; Costa, R.; Negrao, R.; Guimaraes, J.T.; Delgado, L.; Soares, R.; Moreira, A. Increased circulating platelet microparticles as a potential biomarker in asthma. Allergy 2013, 68, 1073–1075. [Google Scholar] [CrossRef]
  31. Capron, M.; Jouault, T.; Prin, L.; Joseph, M.; Ameisen, J.C.; Butterworth, A.E.; Papin, J.P.; Kusnierz, J.P.; Capron, A. Functional study of a monoclonal antibody to IgE Fc receptor (Fc epsilon R2) of eosinophils, platelets, and macrophages. J. Exp. Med. 1986, 164, 72–89. [Google Scholar] [CrossRef]
  32. Lommatzsch, M.; Schloetcke, K.; Klotz, J.; Schuhbaeck, K.; Zingler, D.; Zingler, C.; Schulte-Herbruggen, O.; Gill, H.; Schuff-Werner, P.; Virchow, J.C. Brain-derived neurotrophic factor in platelets and airflow limitation in asthma. Am. J. Respir. Crit. Care Med. 2005, 171, 115–120. [Google Scholar] [CrossRef] [PubMed]
  33. Kowal, K.; Zebrowska, E.; Chabowski, A. Altered Sphingolipid Metabolism Is Associated With Asthma Phenotype in House Dust Mite-Allergic Patients. Allergy Asthma Immunol. Res. 2019, 11, 330–342. [Google Scholar] [CrossRef]
  34. Hasegawa, S.; Pawankar, R.; Suzuki, K.; Nakahata, T.; Furukawa, S.; Okumura, K.; Ra, C. Functional expression of the high affinity receptor for IgE (FcepsilonRI) in human platelets and its’ intracellular expression in human megakaryocytes. Blood 1999, 93, 2543–2551. [Google Scholar] [CrossRef]
  35. Hasegawa, S.; Tashiro, N.; Matsubara, T.; Furukawa, S.; Ra, C. A comparison of FcepsilonRI-mediated RANTES release from human platelets between allergic patients and healthy individuals. Int. Arch. Allergy Immunol. 2001, 125 (Suppl. 1), 42–47. [Google Scholar] [CrossRef]
  36. Shah, S.A.; Kanabar, V.; Riffo-Vasquez, Y.; Mohamed, Z.; Cleary, S.J.; Corrigan, C.; James, A.L.; Elliot, J.G.; Shute, J.K.; Page, C.P.; et al. Platelets Independently Recruit into Asthmatic Lungs and Models of Allergic Inflammation via CCR3. Am. J. Respir. Cell Mol. Biol. 2021, 64, 557–568. [Google Scholar] [CrossRef]
  37. Joseph, M.; Gounni, A.S.; Kusnierz, J.P.; Vorng, H.; Sarfati, M.; Kinet, J.P.; Tonnel, A.B.; Capron, A.; Capron, M. Expression and functions of the high-affinity IgE receptor on human platelets and megakaryocyte precursors. Eur. J. Immunol. 1997, 27, 2212–2218. [Google Scholar] [CrossRef]
  38. Pitchford, S.C.; Momi, S.; Baglioni, S.; Casali, L.; Giannini, S.; Rossi, R.; Page, C.P.; Gresele, P. Allergen induces the migration of platelets to lung tissue in allergic asthma. Am. J. Respir. Crit. Care Med. 2008, 177, 604–612. [Google Scholar] [CrossRef] [PubMed]
  39. Cardot, E.; Pestel, J.; Callebaut, I.; Lassalle, P.; Tsicopoulos, A.; Gras-Masse, H.; Capron, A.; Joseph, M. Specific activation of platelets from patients allergic to Dermatophagoides pteronyssinus by synthetic peptides derived from the allergen Der p I. Int. Arch. Allergy Immunol. 1992, 98, 127–134. [Google Scholar] [CrossRef]
  40. Raiden, S.; Schettini, J.; Salamone, G.; Trevani, A.; Vermeulen, M.; Gamberale, R.; Giordano, M.; Geffner, J. Human platelets produce granulocyte-macrophage colony-stimulating factor and delay eosinophil apoptosis. Lab. Investig. J. Tech. Methods Pathol. 2003, 83, 589–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Tsuji, T.; Nagata, K.; Koike, J.; Todoroki, N.; Irimura, T. Induction of superoxide anion production from monocytes an neutrophils by activated platelets through the P-selectin-sialyl Lewis X interaction. J. Leukoc. Biol. 1994, 56, 583–587. [Google Scholar] [CrossRef]
  42. Nakanishi, T.; Inaba, M.; Inagaki-Katashiba, N.; Tanaka, A.; Vien, P.T.; Kibata, K.; Ito, T.; Nomura, S. Platelet-derived RANK ligand enhances CCL17 secretion from dendritic cells mediated by thymic stromal lymphopoietin. Platelets 2015, 26, 425–431. [Google Scholar] [CrossRef] [PubMed]
  43. Pitchford, S.C.; Momi, S.; Giannini, S.; Casali, L.; Spina, D.; Page, C.P.; Gresele, P. Platelet P-selectin is required for pulmonary eosinophil and lymphocyte recruitment in a murine model of allergic inflammation. Blood 2005, 105, 2074–2081. [Google Scholar] [CrossRef]
  44. Lukacs, N.W.; John, A.; Berlin, A.; Bullard, D.C.; Knibbs, R.; Stoolman, L.M. E- and P-selectins are essential for the development of cockroach allergen-induced airway responses. J. Immunol. (Baltimore, Md.: 1950) 2002, 169, 2120–2125. [Google Scholar] [CrossRef] [Green Version]
  45. Cardenas, E.I.; Breaux, K.; Da, Q.; Flores, J.R.; Ramos, M.A.; Tuvim, M.J.; Burns, A.R.; Rumbaut, R.E.; Adachi, R. Platelet Munc13-4 regulates hemostasis, thrombosis and airway inflammation. Haematologica 2018, 103, 1235–1244. [Google Scholar] [CrossRef] [PubMed]
  46. Yoshida, A.; Ohba, M.; Wu, X.; Sasano, T.; Nakamura, M.; Endo, Y. Accumulation of platelets in the lung and liver and their degranulation following antigen-challenge in sensitized mice. Br. J. Pharmacol. 2002, 137, 146–152. [Google Scholar] [CrossRef] [Green Version]
  47. Kasperska-Zajac, A.; Rogala, B. Markers of platelet activation in plasma of patients suffering from persistent allergic rhinitis with or without asthma symptoms. Clin. Exp. Allergy 2005, 35, 1462–1465. [Google Scholar] [CrossRef] [PubMed]
  48. Kasperska-Zajac, A.; Rogala, B. Platelet activity measured by plasma levels of beta-thromboglobulin and platelet factor 4 in seasonal allergic rhinitis during natural pollen exposure. Inflamm. Res. 2003, 52, 477–479. [Google Scholar] [CrossRef]
  49. Koczy-Baron, E.; Grzanka, A.; Jochem, J.; Gawlik, R.; Kasperska-Zajac, A. Evaluation of circulating vascular endothelial growth factor and its soluble receptors in patients suffering from persistent allergic rhinitis. Allergy Asthma Clin. Immunol. Off. J. Can. Soc. Allergy Clin. Immunol. 2016, 12, 17. [Google Scholar] [CrossRef] [Green Version]
  50. Potaczek, D.P. Links between allergy and cardiovascular or hemostatic system. Int. J. Cardiol. 2014, 170, 278–285. [Google Scholar] [CrossRef]
  51. Lloyd, C.M.; Snelgrove, R.J. Type 2 immunity: Expanding our view. Sci. Immunol. 2018, 3, eaat1604. [Google Scholar] [CrossRef] [Green Version]
  52. Kumar, S.; Jeong, Y.; Ashraf, M.U.; Bae, Y.S. Dendritic Cell-Mediated Th2 Immunity and Immune Disorders. Int. J. Mol. Sci. 2019, 20, 2159. [Google Scholar] [CrossRef] [Green Version]
  53. Amison, R.T.; Cleary, S.J.; Riffo-Vasquez, Y.; Bajwa, M.; Page, C.P.; Pitchford, S.C. Platelets Play a Central Role in Sensitization to Allergen. Am. J. Respir. Cell Mol. Biol. 2018, 59, 96–103. [Google Scholar] [CrossRef] [PubMed]
  54. Page, C.; Pitchford, S. Platelets coming of age: Implications for our understanding of allergic inflammation. Am. J. Respir. Crit. Care Med. 2013, 187, 459–460. [Google Scholar] [CrossRef]
  55. Durk, T.; Duerschmied, D.; Muller, T.; Grimm, M.; Reuter, S.; Vieira, R.P.; Ayata, K.; Cicko, S.; Sorichter, S.; Walther, D.J.; et al. Production of serotonin by tryptophan hydroxylase 1 and release via platelets contribute to allergic airway inflammation. Am. J. Respir. Crit. Care Med. 2013, 187, 476–485. [Google Scholar] [CrossRef] [PubMed]
  56. Tian, J.; Zhu, T.; Liu, J.; Guo, Z.; Cao, X. Platelets promote allergic asthma through the expression of CD154. Cell. Mol. Immunol. 2015, 12, 700–707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Akdis, C.A.; Arkwright, P.D.; Bruggen, M.C.; Busse, W.; Gadina, M.; Guttman-Yassky, E.; Kabashima, K.; Mitamura, Y.; Vian, L.; Wu, J.; et al. Type 2 immunity in the skin and lungs. Allergy 2020, 75, 1582–1605. [Google Scholar] [CrossRef]
  58. Takeda, T.; Unno, H.; Morita, H.; Futamura, K.; Emi-Sugie, M.; Arae, K.; Shoda, T.; Okada, N.; Igarashi, A.; Inoue, E.; et al. Platelets constitutively express IL-33 protein and modulate eosinophilic airway inflammation. J. Allergy Clin. Immunol. 2016, 138, 1395–1403. [Google Scholar] [CrossRef] [Green Version]
  59. Chae, W.J.; Ehrlich, A.K.; Chan, P.Y.; Teixeira, A.M.; Henegariu, O.; Hao, L.; Shin, J.H.; Park, J.H.; Tang, W.H.; Kim, S.T.; et al. The Wnt Antagonist Dickkopf-1 Promotes Pathological Type 2 Cell-Mediated Inflammation. Immunity 2016, 44, 246–258. [Google Scholar] [CrossRef] [Green Version]
  60. Gomez-Casado, C.; Villasenor, A.; Rodriguez-Nogales, A.; Bueno, J.L.; Barber, D.; Escribese, M.M. Understanding Platelets in Infectious and Allergic Lung Diseases. Int. J. Mol. Sci. 2019, 20, 1730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Hayashi, N.; Chihara, J.; Kobayashi, Y.; Kakazu, T.; Kurachi, D.; Yamamoto, T.; Nakajima, S. Effect of platelet-activating factor and platelet factor 4 on eosinophil adhesion. Int. Arch. Allergy Immunol. 1994, 104 (Suppl. 1), 57–59. [Google Scholar] [CrossRef]
  62. Chihara, J.; Yasuba, H.; Tsuda, A.; Urayama, O.; Saito, N.; Honda, K.; Kayaba, H.; Yamashita, T.; Kurimoto, F.; Yamada, H. Elevation of the plasma level of RANTES during asthma attacks. J. Allergy Clin. Immunol. 1997, 100, S52–S55. [Google Scholar] [CrossRef]
  63. Bisset, L.R.; Schmid-Grendelmeier, P. Chemokines and their receptors in the pathogenesis of allergic asthma: Progress and perspective. Curr. Opin. Pulm. Med. 2005, 11, 35–42. [Google Scholar] [CrossRef] [PubMed]
  64. Hayashizaki, K.; Kimura, M.Y.; Tokoyoda, K.; Hosokawa, H.; Shinoda, K.; Hirahara, K.; Ichikawa, T.; Onodera, A.; Hanazawa, A.; Iwamura, C.; et al. Myosin light chains 9 and 12 are functional ligands for CD69 that regulate airway inflammation. Sci. Immunol. 2016, 1, eaaf9154. [Google Scholar] [CrossRef]
  65. Amison, R.T.; Momi, S.; Morris, A.; Manni, G.; Keir, S.; Gresele, P.; Page, C.P.; Pitchford, S.C. RhoA signaling through platelet P2Y(1) receptor controls leukocyte recruitment in allergic mice. J. Allergy Clin. Immunol. 2015, 135, 528–538. [Google Scholar] [CrossRef] [PubMed]
  66. Geng, J.G.; Chen, M.; Chou, K.C. P-selectin cell adhesion molecule in inflammation, thrombosis, cancer growth and metastasis. Curr. Med. Chem. 2004, 11, 2153–2160. [Google Scholar] [CrossRef] [Green Version]
  67. Johansson, M.W.; Mosher, D.F. Integrin activation States and eosinophil recruitment in asthma. Front. Pharmacol. 2013, 4, 33. [Google Scholar] [CrossRef] [Green Version]
  68. Laidlaw, T.M.; Kidder, M.S.; Bhattacharyya, N.; Xing, W.; Shen, S.; Milne, G.L.; Castells, M.C.; Chhay, H.; Boyce, J.A. Cysteinyl leukotriene overproduction in aspirin-exacerbated respiratory disease is driven by platelet-adherent leukocytes. Blood 2012, 119, 3790–3798. [Google Scholar] [CrossRef] [Green Version]
  69. Mitsui, C.; Kajiwara, K.; Hayashi, H.; Ito, J.; Mita, H.; Ono, E.; Higashi, N.; Fukutomi, Y.; Sekiya, K.; Tsuburai, T.; et al. Platelet activation markers overexpressed specifically in patients with aspirin-exacerbated respiratory disease. J. Allergy Clin. Immunol. 2016, 137, 400–411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Taniguchi, M.; Mitsui, C.; Hayashi, H.; Ono, E.; Kajiwara, K.; Mita, H.; Watai, K.; Kamide, Y.; Fukutomi, Y.; Sekiya, K.; et al. Aspirin-exacerbated respiratory disease (AERD): Current understanding of AERD. Allergol. Int. 2019, 68, 289–295. [Google Scholar] [CrossRef]
  71. Dominas, C.; Gadkaree, S.; Maxfield, A.Z.; Gray, S.T.; Bergmark, R.W. Aspirin-exacerbated respiratory disease: A review. Laryngoscope Investig. Otolaryngol. 2020, 5, 360–367. [Google Scholar] [CrossRef]
  72. Rusznak, M.; Peebles, R.S., Jr. Prostaglandin E2 in NSAID-exacerbated respiratory disease: Protection against cysteinyl leukotrienes and group 2 innate lymphoid cells. Curr. Opin. Allergy Clin. Immunol. 2019, 19, 38–45. [Google Scholar] [CrossRef]
  73. Lommatzsch, M.; Julius, P.; Kuepper, M.; Garn, H.; Bratke, K.; Irmscher, S.; Luttmann, W.; Renz, H.; Braun, A.; Virchow, J.C. The course of allergen-induced leukocyte infiltration in human and experimental asthma. J. Allergy Clin. Immunol. 2006, 118, 91–97. [Google Scholar] [CrossRef] [PubMed]
  74. Silvestri, M.; Oddera, S.; Sacco, O.; Balbo, A.; Crimi, E.; Rossi, G.A. Bronchial and bronchoalveolar inflammation in single early and dual responders after allergen inhalation challenge. Lung 1997, 175, 277–285. [Google Scholar] [CrossRef] [PubMed]
  75. Herd, C.M.; Page, C.P. Pulmonary immune cells in health and disease: Platelets. Eur. Respir. J. 1994, 7, 1145–1160. [Google Scholar] [PubMed]
  76. Yoshimi, Y.; Fujimura, M.; Myou, S.; Tachibana, H.; Hirose, T. Effect of thromboxane A2 (TXA2) synthase inhibitor and TXA2 receptor antagonist alone and in combination on antigen-induced bronchoconstriction in guinea pigs. Prostaglandins Other Lipid Mediat. 2001, 65, 1–9. [Google Scholar] [CrossRef]
  77. Coyle, A.J.; Page, C.P.; Atkinson, L.; Flanagan, R.; Metzger, W.J. The requirement for platelets in allergen-induced late asthmatic airway obstruction. Eosinophil infiltration and heightened airway responsiveness in allergic rabbits. Am. Rev. Respir. Dis. 1990, 142, 587–593. [Google Scholar] [CrossRef]
  78. Keir, S.D.; Spina, D.; Page, C.P. Bradykinin and capsaicin induced airways obstruction in the guinea pig are platelet dependent. Pulm. Pharmacol. Ther. 2015, 33, 25–31. [Google Scholar] [CrossRef]
  79. Kasperska-Zajac, A.; Brzoza, Z.; Rogala, B. Platelet activating factor as a mediator and therapeutic approach in bronchial asthma. Inflammation 2008, 31, 112–120. [Google Scholar] [CrossRef]
  80. Braun, A.; Lommatzsch, M.; Neuhaus-Steinmetz, U.; Quarcoo, D.; Glaab, T.; McGregor, G.P.; Fischer, A.; Renz, H. Brain-derived neurotrophic factor (BDNF) contributes to neuronal dysfunction in a model of allergic airway inflammation. Br. J. Pharmacol. 2004, 141, 431–440. [Google Scholar] [CrossRef] [Green Version]
  81. Barnes, P.J. New aspects of asthma. J. Intern. Med. 1992, 231, 453–461. [Google Scholar] [CrossRef]
  82. Zhang, X.Y.; Tang, X.Y.; Li, N.; Zhao, L.M.; Guo, Y.L.; Li, X.S.; Tian, C.J.; Cheng, D.J.; Chen, Z.C.; Zhang, L.X. GAS5 promotes airway smooth muscle cell proliferation in asthma via controlling miR-10a/BDNF signaling pathway. Life Sci. 2018, 212, 93–101. [Google Scholar] [CrossRef]
  83. Johnson, J.R.; Folestad, E.; Rowley, J.E.; Noll, E.M.; Walker, S.A.; Lloyd, C.M.; Rankin, S.M.; Pietras, K.; Eriksson, U.; Fuxe, J. Pericytes contribute to airway remodeling in a mouse model of chronic allergic asthma. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2015, 308, L658–L671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Wang, L.; Feng, X.; Hu, B.; Xia, Q.; Ni, X.; Song, Y. P2X4R promotes airway remodeling by acting on the phenotype switching of bronchial smooth muscle cells in rats. Purinergic Signal. 2018, 14, 433–442. [Google Scholar] [CrossRef] [PubMed]
  85. Yamashita, N.; Sekine, K.; Miyasaka, T.; Kawashima, R.; Nakajima, Y.; Nakano, J.; Yamamoto, T.; Horiuchi, T.; Hirai, K.; Ohta, K. Platelet-derived growth factor is involved in the augmentation of airway responsiveness through remodeling of airways in diesel exhaust particulate-treated mice. J. Allergy Clin. Immunol. 2001, 107, 135–142. [Google Scholar] [CrossRef] [PubMed]
  86. Wynendaele, W.; Derua, R.; Hoylaerts, M.F.; Pawinski, A.; Waelkens, E.; De Bruijn, E.A.; Paridaens, R.; Merlevede, W.; Van Oosterom, A.T. Vascular endothelial growth factor measured in platelet poor plasma allows optimal separation between cancer patients and volunteers: A key to study an angiogenic marker in vivo? Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 1999, 10, 965–971. [Google Scholar] [CrossRef]
  87. Hur, G.Y.; Broide, D.H. Genes and Pathways Regulating Decline in Lung Function and Airway Remodeling in Asthma. Allergy Asthma Immunol. Res. 2019, 11, 604–621. [Google Scholar] [CrossRef] [PubMed]
  88. Finotto, S. Resolution of allergic asthma. Semin. Immunopathol. 2019, 41, 665–674. [Google Scholar] [CrossRef]
  89. Thomas, M.R.; Storey, R.F. Effect of P2Y12 inhibitors on inflammation and immunity. Thromb. Haemost. 2015, 114, 490–497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Storey, R.F.; Judge, H.M.; Wilcox, R.G.; Heptinstall, S. Inhibition of ADP-induced P-selectin expression and platelet-leukocyte conjugate formation by clopidogrel and the P2Y12 receptor antagonist AR-C69931MX but not aspirin. Thromb. Haemost. 2002, 88, 488–494. [Google Scholar]
  91. Suh, D.H.; Trinh, H.K.; Liu, J.N.; Pham le, D.; Park, S.M.; Park, H.S.; Shin, Y.S. P2Y12 antagonist attenuates eosinophilic inflammation and airway hyperresponsiveness in a mouse model of asthma. J. Cell. Mol. Med. 2016, 20, 333–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Lussana, F.; Di Marco, F.; Terraneo, S.; Parati, M.; Razzari, C.; Scavone, M.; Femia, E.A.; Moro, A.; Centanni, S.; Cattaneo, M. Effect of prasugrel in patients with asthma: Results of PRINA, a randomized, double-blind, placebo-controlled, cross-over study. J. Thromb. Haemost. JTH 2015, 13, 136–141. [Google Scholar] [CrossRef] [Green Version]
  93. Shi, H.; Yokoyama, A.; Kohno, N.; Hirasawa, Y.; Kondo, K.; Sakai, K.; Hiwada, K. Effect of thromboxane A2 inhibitors on allergic pulmonary inflammation in mice. Eur. Respir. J. 1998, 11, 624–629. [Google Scholar] [PubMed]
  94. Hayashi, M.; Koya, T.; Kawakami, H.; Sakagami, T.; Hasegawa, T.; Kagamu, H.; Takada, T.; Sakai, Y.; Suzuki, E.; Gelfand, E.W.; et al. A prostacyclin agonist with thromboxane inhibitory activity for airway allergic inflammation in mice. Clin. Exp. Allergy J. Br. Soc. Allergy Clin. Immunol. 2010, 40, 317–326. [Google Scholar] [CrossRef]
  95. Fukuoka, T.; Miyake, S.; Umino, T.; Inase, N.; Tojo, N.; Yoshizawa, Y. The effect of seratrodast on eosinophil cationic protein and symptoms in asthmatics. J. Asthma Off. J. Assoc. Care Asthma 2003, 40, 257–264. [Google Scholar] [CrossRef]
  96. Chamba, G.; Lemoine, P.; Flachaire, E.; Ferry, N.; Quincy, C.; Sassard, J.; Ferber, C.; Mocaer, E.; Kamoun, A.; Renaud, B. Increased serotonin platelet uptake after tianeptine administration in depressed patients. Biol. Psychiatry 1991, 30, 609–617. [Google Scholar] [CrossRef]
  97. Lechin, F.; Van der Dijs, B.; Lechin, A.E. Treatment of bronchial asthma with tianeptine. Methods Find. Exp. Clin. Pharmacol. 2004, 26, 697–701. [Google Scholar] [CrossRef] [PubMed]
  98. Pitchford, S.; Cleary, S.; Arkless, K.; Amison, R. Pharmacological strategies for targeting platelet activation in asthma. Curr. Opin. Pharm. 2019, 46, 55–64. [Google Scholar] [CrossRef]
  99. Cattaneo, M. The platelet P2 receptors in inflammation. Hamostaseologie 2015, 35, 262–266. [Google Scholar] [CrossRef]
  100. Kunapuli, S.P.; Dorsam, R.T.; Kim, S.; Quinton, T.M. Platelet purinergic receptors. Curr. Opin. Pharmacol. 2003, 3, 175–180. [Google Scholar] [CrossRef]
  101. Mansour, A.; Bachelot-Loza, C.; Nesseler, N.; Gaussem, P.; Gouin-Thibault, I. P2Y(12) Inhibition beyond Thrombosis: Effects on Inflammation. Int. J. Mol. Sci. 2020, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Wang, D.; Yang, X.H.; Zhang, J.D.; Li, R.B.; Jia, M.; Cui, X.R. Compared efficacy of clopidogrel and ticagrelor in treating acute coronary syndrome: A meta-analysis. BMC Cardiovasc. Disord. 2018, 18, 217. [Google Scholar] [CrossRef]
  103. Spartalis, M.; Tzatzaki, E.; Spartalis, E.; Damaskos, C.; Athanasiou, A.; Moris, D.; Politou, M. The role of prasugrel in the management of acute coronary syndromes: A systematic review. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 4733–4743. [Google Scholar]
  104. Trinh, H.K.T.; Nguyen, T.V.T.; Choi, Y.; Park, H.S.; Shin, Y.S. The synergistic effects of clopidogrel with montelukast may be beneficial for asthma treatment. J. Cell. Mol. Med. 2019, 23, 3441–3450. [Google Scholar] [CrossRef]
  105. Amison, R.T.; Arnold, S.; O’Shaughnessy, B.G.; Cleary, S.J.; Ofoedu, J.; Idzko, M.; Page, C.P.; Pitchford, S.C. Lipopolysaccharide (LPS) induced pulmonary neutrophil recruitment and platelet activation is mediated via the P2Y1 and P2Y14 receptors in mice. Pulm. Pharmacol. Ther. 2017, 45, 62–68. [Google Scholar] [CrossRef] [Green Version]
  106. Zhang, D.; Gao, Z.G.; Zhang, K.; Kiselev, E.; Crane, S.; Wang, J.; Paoletta, S.; Yi, C.; Ma, L.; Zhang, W.; et al. Two disparate ligand-binding sites in the human P2Y1 receptor. Nature 2015, 520, 317–321. [Google Scholar] [CrossRef]
  107. Dogne, J.M.; De Leval, X.; Benoit, P.; Rolin, S.; Pirotte, B.; Masereel, B. Therapeutic potential of thromboxane inhibitors in asthma. Expert Opin. Investig. Drugs 2002, 11, 275–281. [Google Scholar] [CrossRef]
  108. Hernandez, J.M.; Janssen, L.J. Revisiting the usefulness of thromboxane-A2 modulation in the treatment of bronchoconstriction in asthma. Can. J. Physiol. Pharm. 2015, 93, 111–117. [Google Scholar] [CrossRef]
  109. Fujimura, M.; Sasaki, F.; Nakatsumi, Y.; Takahashi, Y.; Hifumi, S.; Taga, K.; Mifune, J.; Tanaka, T.; Matsuda, T. Effects of a thromboxane synthetase inhibitor (OKY-046) and a lipoxygenase inhibitor (AA-861) on bronchial responsiveness to acetylcholine in asthmatic subjects. Thorax 1986, 41, 955–959. [Google Scholar] [CrossRef] [Green Version]
  110. Mammadova-Bach, E.; Mauler, M.; Braun, A.; Duerschmied, D. Autocrine and paracrine regulatory functions of platelet serotonin. Platelets 2018, 29, 541–548. [Google Scholar] [CrossRef]
  111. Berger, M.; Gray, J.A.; Roth, B.L. The expanded biology of serotonin. Annu. Rev. Med. 2009, 60, 355–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Lechin, F.; Van der Dijs, B.; Orozco, B.; Lechin, M.; Lechin, A.E. Increased levels of free serotonin in plasma of symptomatic asthmatic patients. Ann. Allergy Asthma Immunol. Off. Publ. Am. Coll. Allergy Asthma Immunol. 1996, 77, 245–253. [Google Scholar] [CrossRef]
  113. McNicol, A.; Israels, S.J. Platelet dense granules: Structure, function and implications for haemostasis. Thromb. Res. 1999, 95, 1–18. [Google Scholar] [CrossRef]
  114. Badimon, L.; Vilahur, G.; Rocca, B.; Patrono, C. The Key Contribution of Platelet and Vascular Arachidonic Acid Metabolism To The Pathophysiology Of Atherothrombosis. Cardiovasc. Res. 2021. [Google Scholar] [CrossRef] [PubMed]
  115. Narayanankutty, A.; Resendiz-Hernandez, J.M.; Falfan-Valencia, R.; Teran, L.M. Biochemical pathogenesis of aspirin exacerbated respiratory disease (AERD). Clin. Biochem. 2013, 46, 566–578. [Google Scholar] [CrossRef] [PubMed]
  116. Le Pham, D.; Lee, J.H.; Park, H.S. Aspirin-exacerbated respiratory disease: An update. Curr. Opin. Pulm. Med. 2017, 23, 89–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Page, C.P. The involvement of platelets in non-thrombotic processes. Trends Pharmacol. Sci. 1988, 9, 66–71. [Google Scholar] [CrossRef]
  118. Laumonnier, Y.; Wiese, A.V.; Figge, J.; Karsten, C. Regulation and function of anaphylatoxins and their receptors in allergic asthma. Mol. Immunol. 2017, 84, 51–56. [Google Scholar] [CrossRef] [PubMed]
  119. Polley, M.J.; Nachman, R.L. Human platelet activation by C3a and C3a des-arg. J. Exp. Med. 1983, 158, 603–615. [Google Scholar] [CrossRef] [Green Version]
  120. Sauter, R.J.; Sauter, M.; Obrich, M.; Emschermann, F.N.; Nording, H.; Patzelt, J.; Wendel, H.P.; Reil, J.C.; Edlich, F.; Langer, H.F. Anaphylatoxin Receptor C3aR Contributes to Platelet Function, Thrombus Formation and In Vivo Haemostasis. Thromb. Haemost. 2019, 119, 179–182. [Google Scholar] [CrossRef]
  121. Subramaniam, S.; Jurk, K.; Hobohm, L.; Jäckel, S.; Saffarzadeplatelet activation and fibrin formatioh, M.; Schwierczek, K.; Wenzel, P.; Langer, F.; Reinhardt, C.; Ruf, W. Distinct contributions of complement factors to n in venous thrombus development. Blood 2017, 129, 2291–2302. [Google Scholar] [CrossRef] [Green Version]
  122. Martel, C.; Cointe, S.; Maurice, P.; Matar, S.; Ghitescu, M.; Théroux, P.; Bonnefoy, A. Requirements for membrane attack complex formation and anaphylatoxins binding to collagen-activated platelets. PLoS ONE 2011, 6, e18812. [Google Scholar] [CrossRef] [Green Version]
  123. Saradna, A.; Do, D.C.; Kumar, S.; Fu, Q.L.; Gao, P. Macrophage polarization and allergic asthma. Transl. Res. J. Lab. Clin. Med. 2018, 191, 1–14. [Google Scholar] [CrossRef]
  124. Jiang, Z.; Zhu, L. Update on the role of alternatively activated macrophages in asthma. J. Asthma Allergy 2016, 9, 101–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Rossaint, J.; Thomas, K.; Mersmann, S.; Skupski, J.; Margraf, A.; Tekath, T.; Jouvene, C.C.; Dalli, J.; Hidalgo, A.; Meuth, S.G.; et al. Platelets orchestrate the resolution of pulmonary inflammation in mice by T reg cell repositioning and macrophage education. J. Exp. Med. 2021, 218. [Google Scholar] [CrossRef] [PubMed]
  126. Winnica, D.; Corey, C.; Mullett, S.; Reynolds, M.; Hill, G.; Wendell, S.; Que, L.; Holguin, F.; Shiva, S. Bioenergetic Differences in the Airway Epithelium of Lean Versus Obese Asthmatics Are Driven by Nitric Oxide and Reflected in Circulating Platelets. Antioxid. Redox Signal. 2019, 31, 673–686. [Google Scholar] [CrossRef]
  127. Xu, W.; Cardenes, N.; Corey, C.; Erzurum, S.C.; Shiva, S. Platelets from Asthmatic Individuals Show Less Reliance on Glycolysis. PLoS ONE 2015, 10, e0132007. [Google Scholar] [CrossRef] [Green Version]
  128. Newcomb, D.C.; Peebles, R.S., Jr. Th17-mediated inflammation in asthma. Curr. Opin. Immunol. 2013, 25, 755–760. [Google Scholar] [CrossRef] [Green Version]
  129. Affandi, A.J.; Silva-Cardoso, S.C.; Garcia, S.; Leijten, E.F.A.; Van Kempen, T.S.; Marut, W.; Van Roon, J.A.G.; Radstake, T. CXCL4 is a novel inducer of human Th17 cells and correlates with IL-17 and IL-22 in psoriatic arthritis. Eur. J. Immunol. 2018, 48, 522–531. [Google Scholar] [CrossRef]
  130. Ponomarev, E.D. Fresh Evidence for Platelets as Neuronal and Innate Immune Cells: Their Role in the Activation, Differentiation, and Deactivation of Th1, Th17, and Tregs during Tissue Inflammation. Front. Immunol 2018, 9, 406. [Google Scholar] [CrossRef] [Green Version]
  131. Zhu, L.; Huang, Z.; Stalesen, R.; Hansson, G.K.; Li, N. Platelets provoke distinct dynamics of immune responses by differentially regulating CD4+ T-cell proliferation. J. Thromb. Haemost. JTH 2014, 12, 1156–1165. [Google Scholar] [CrossRef]
  132. Weyrich, A.S.; Zimmerman, G.A. Platelets in lung biology. Annu. Rev. Physiol. 2013, 75, 569–591. [Google Scholar] [CrossRef] [Green Version]
  133. Lefrançais, E.; Ortiz-Muñoz, G.; Caudrillier, A.; Mallavia, B.; Liu, F.; Sayah, D.M.; Thornton, E.E.; Headley, M.B.; David, T.; Coughlin, S.R.; et al. The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors. Nature 2017, 544, 105–109. [Google Scholar] [CrossRef]
  134. Yeung, A.K.; Villacorta-Martin, C.; Hon, S.; Rock, J.R.; Murphy, G.J. Lung megakaryocytes display distinct transcriptional and phenotypic properties. Blood Adv. 2020, 4, 6204–6217. [Google Scholar] [CrossRef]
  135. Pariser, D.N.; Hilt, Z.T.; Ture, S.K.; Blick-Nitko, S.K.; Looney, M.R.; Cleary, S.J.; Roman-Pagan, E.; Saunders, J., 2nd; Georas, S.N.; Veazey, J.; et al. Lung megakaryocytes are immune modulatory cells. J. Clin. Investig. 2021, 131, e137377. [Google Scholar] [CrossRef]
  136. Hogan, K.A.; Weiler, H.; Lord, S.T. Mouse models in coagulation. Thromb. Haemost. 2002, 87, 563–574. [Google Scholar] [CrossRef] [Green Version]
  137. Alessandrini, F.; Musiol, S.; Schneider, E.; Blanco-Pérez, F.; Albrecht, M. Mimicking Antigen-Driven Asthma in Rodent Models-How Close Can We Get? Front. Immunol 2020, 11, 575936. [Google Scholar] [CrossRef]
  138. Zschaler, J.; Schlorke, D.; Arnhold, J. Differences in innate immune response between man and mouse. Crit. Rev. Immunol. 2014, 34, 433–454. [Google Scholar] [CrossRef] [Green Version]
  139. Looney, M.R.; Headley, M.B. Live imaging of the pulmonary immune environment. Cell. Immunol. 2020, 350, 103862. [Google Scholar] [CrossRef]
Figure 1. Platelet-derived factors contributing to allergic asthma.
Figure 1. Platelet-derived factors contributing to allergic asthma.
Cells 10 02038 g001
Figure 2. Involvement of platelets in the pathogenesis of allergic asthma. Environmental allergens, anti-IgE, and/or anti-IgE receptors (anti-FcεRI and anti-FcεRII/CD23) activate resting platelets. (1) Activated platelets secrete IL-33 that acts on ILC2, or Dkk-1 and 5-HT to promote naive T helper cells’ differentiation into Th2 effectors. Moreover, they can influence the activity of TSLP-stimulated mDCs in a RANK-RANKL-dependent pathway or inhibit regulatory T cells’ differentiation by overexpressing CD40L (CD154). (2) Activated platelets are involved in the recruitment of inflammatory cells. Platelets form aggregates with leukocytes through P-selectin-PSGL-1 interaction after allergens stimulation. The platelet-eosinophil aggregates (PEA) activate eosinophils and upregulate the expression of αMβ2 and α4β1 integrins, through which PEA can adhere to the endothelial cells and migrate to the inflammatory sites. The platelet-neutrophil aggregates (PNA) activate the arachidonic acid pathway to generate CysLTs which contribute to ARED. The CD69-Myl9 system is a newly found mechanism mediating the infiltration of activated CD4+CD69+ T cells into the inflamed tissue. (3) Activated platelets produce spasmogen, such as TxA2, PF4, PAF, BDNF, and S1P, to induce bronchial smooth muscle contraction as well as airway hyperresponsiveness. They also release MMP2, MMP9, cationic proteins, and ROS to cause degradation of extracellular matrix. PDGF promotes proliferation of fibroblasts and airway smooth muscle cells. GM-CSF inhibits apoptosis of eosinophils and thus contributes to the chronic inflammatory response and tissue damage. GM-CSF also enhances the 5-LO activity in neutrophils and eosinophils to accelerate the production of CysLTs, causing airway smooth muscle contraction, inflammatory cell recruitment, and edema. All these processes together lead to chronic airway inflammation and airway remodeling.
Figure 2. Involvement of platelets in the pathogenesis of allergic asthma. Environmental allergens, anti-IgE, and/or anti-IgE receptors (anti-FcεRI and anti-FcεRII/CD23) activate resting platelets. (1) Activated platelets secrete IL-33 that acts on ILC2, or Dkk-1 and 5-HT to promote naive T helper cells’ differentiation into Th2 effectors. Moreover, they can influence the activity of TSLP-stimulated mDCs in a RANK-RANKL-dependent pathway or inhibit regulatory T cells’ differentiation by overexpressing CD40L (CD154). (2) Activated platelets are involved in the recruitment of inflammatory cells. Platelets form aggregates with leukocytes through P-selectin-PSGL-1 interaction after allergens stimulation. The platelet-eosinophil aggregates (PEA) activate eosinophils and upregulate the expression of αMβ2 and α4β1 integrins, through which PEA can adhere to the endothelial cells and migrate to the inflammatory sites. The platelet-neutrophil aggregates (PNA) activate the arachidonic acid pathway to generate CysLTs which contribute to ARED. The CD69-Myl9 system is a newly found mechanism mediating the infiltration of activated CD4+CD69+ T cells into the inflamed tissue. (3) Activated platelets produce spasmogen, such as TxA2, PF4, PAF, BDNF, and S1P, to induce bronchial smooth muscle contraction as well as airway hyperresponsiveness. They also release MMP2, MMP9, cationic proteins, and ROS to cause degradation of extracellular matrix. PDGF promotes proliferation of fibroblasts and airway smooth muscle cells. GM-CSF inhibits apoptosis of eosinophils and thus contributes to the chronic inflammatory response and tissue damage. GM-CSF also enhances the 5-LO activity in neutrophils and eosinophils to accelerate the production of CysLTs, causing airway smooth muscle contraction, inflammatory cell recruitment, and edema. All these processes together lead to chronic airway inflammation and airway remodeling.
Cells 10 02038 g002
Table 1. Various platelet-derived factors in the pathogenesis of allergic asthma in humans.
Table 1. Various platelet-derived factors in the pathogenesis of allergic asthma in humans.
ModelsSamplesIndicatorsSubjectsReferences
In vivoPeripheral bloodPlatelet-leukocyte conjugates↑ Asthmatic patients after allergen exposure[25]
Platelet-derived microparticles (PMPs) ↑Asthmatic patients [30]
Percentage of IgE+ platelets↑Asthmatic patients[31]
Eosinophil β1-integrin activation↑Asthmatic patients[28,29]
Platelet BDNF↑Patients with allergic asthma[32]
Plasmaβ-TG and PF4↑Atopic dermatitis patients with concomitant asthma and allergic rhinitis[19]
β-TG, PF4↑
Platelet count↓
House-dust-mite-sensitive asthmatic patients intrabronchially challenged with Dp extract[20]
PF4↑
(during the grass pollen season)
PF4↓(off season)
Patients with pollen-induced seasonal allergic rhinitis and asthma[26]
BDNF↑Patients with allergic asthma[32]
Phingosine-1-phosphate (SIP)↑House-dust-mite-allergic patients[33]
BALFIsolation of platelets Asthmatic patients[21]
5-HT↑Asthmatic patients[31,34,35]
β-TG, PF4↑Ragweed-allergic asthmatic subjects after challenge with ragweed antigen.[22]
Bronchial biopsiesPlatelet deposition on interalveolar septum walls
Platelet number↑
Asthmatic patients [23,24,36]
Nasal lavage fluidP-selectin positively correlated with ECP levelAsthmatic patients[27]
In vitroPlateletFcεRI and FcεRII/CD23 expressionHuman platelets and megakaryocyte[31,34,35,37,38]
RANTES release↑ Cytotoxicity against schistosomulaPlatelets treated by anti-FcεRI, anti-CD23, anti-IgE [34,35,37]
RANTES release↑Platelet from allergic patients stimulated with IgE and anti-IgE[35]
Allergen-specific cytotoxicity against schistosomula↑Patients allergic to Dermatophagoides pteronyssinus [39]
Allergen-specific platelet chemotaxisAllergic asthmatic patients[38]
GM-CSF↑
Eosinophils apoptosis↓
Human platelets and Eosinophils coculture[40]
P-selectin↑
Neutrophil superoxide anions↑
Human platelets and neutrophils coculture [41]
RANKL in platelets↑
CCL17 (Th2-attracting chemokine)↑
TRAP6-activated platelets with TSLP-stimulated DCs coculture [42]
Table 2. Evidence of platelets’ involvement in the pathogenesis of allergic asthma.
Table 2. Evidence of platelets’ involvement in the pathogenesis of allergic asthma.
Mechanism StudiedMethod/Animal ModelFindingsReferences
Platelet degranulationMunc13-4−/−micePlatelets Munc13-4 deficiency abolishes dense granule release to reduce AHR and eosinophilic inflammation.[45]
Platelet migration FcRγ−/−mice
Platelet depletion
Platelet transfusion
Allergen or anti-IgE antibody in vitro stimulation
Platelet migration is allergen-IgE-FcεRI dependent. [38]
CCR3 antagonistPlatelet rolling, adhesion, and extravascular migration are CCR3 dependent.[36]
Sensitization of allergic asthmaPlatelet depletion
FcRγ−/−mice
Platelets rely on an IgE-FcεRI-dependent pathway to induce Type 2 immune response formation during sensitization stage.[53]
Leukocyte recruitment Platelet depletion
Platelet transfusion
Depletion of platelets reduces, while transfusion of platelets restores the allergen-induced pulmonary leukocyte recruitment. [25]
Platelet depletion
Platelet transfusion
Platelets P-selectin is required for pulmonary eosinophils and lymphocytes recruitment.[43]
Selectin−/−miceP-selectin is critical in the development of allergen-induced airway response.[43,44]
Anti-Myl9/12 antibody
CD69-deficient mice
Platelet secreted myl9 upon activation and formed intravascular net-like structures to bind to CD69+ CD4+ T cells, regulating the pathological process of allergic asthma.[64]
P2Y1 antagonists
Platelet depletion
Platelet transfusion
Purine receptor P2Y1 on platelets regulates leukocyte recruitment in allergic mice through the RhoA signaling pathway. [65]
Type 2 immunity inductionTPH1−/−mice
BM chimera
Mast cell–deficient mice
Platelet transfusion
Platelets secrete 5-HT to enhance the ability of mature DCs to polarize Type 2 immunity formation.[55]
Platelet depletion
Platelet transfusion
Cd154−/−mice
Platelets inhibit Treg generation via CD154 to promote asthma development.[56]
Platelet depletion
Platelet transfusion
IL-33-deficient mice
IL-33 is essential for eosinophilic inflammation in the airway.[58]
Dkk1d/d miceDickkopf-1 (DKK-1) facilitated leukocyte migration and promoted Th2 cell differentiation and Type 2 cytokine production. [59]
Airway remodelingPlatelet depletionPlatelets are necessary for epithelial and smooth-muscle thickening and the deposition of reticular fibers in the extracellular matrix (ECM). [23]
Table 3. Antiplatelet drugs for treating allergic asthma.
Table 3. Antiplatelet drugs for treating allergic asthma.
CategoryTargetDrugMechanismReferences
ADP receptor antagonistsP2Y12 receptorClopidogrel
Prasugrel
Ticagrelor
Reduce the release of platelets and the formation of platelets-leukocyte aggregates, and inhibit eosinophilic inflammation and airway hyperreactivity.[89,90,91,92]
P2Y1 receptorMRS2179
MRS2500
Inhibit the recruitment of eosinophils and lymphocytes to the lung.[65]
TxA2 synthase inhibitorTxA2 synthaseOzagrel (OKY-46)
ONO-1301
Inhibit the production of proinflammatory cytokines and alleviate the eosinophil infiltration in the airways; suppress AHR and airway inflammation.[93,94]
TP receptor antagonistTP receptorSeratrodast (AA-2414)
S-1452
Reduce bronchial hyperresponsiveness by reducing airway inflammation.[93,95]
5-HT modifier5-HT-specific transporterTianeptineEnhance the uptake of free 5-HT in peripheral blood.[96,97]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Luo, L.; Zhang, J.; Lee, J.; Tao, A. Platelets, Not an Insignificant Player in Development of Allergic Asthma. Cells 2021, 10, 2038. https://doi.org/10.3390/cells10082038

AMA Style

Luo L, Zhang J, Lee J, Tao A. Platelets, Not an Insignificant Player in Development of Allergic Asthma. Cells. 2021; 10(8):2038. https://doi.org/10.3390/cells10082038

Chicago/Turabian Style

Luo, Liping, Junyan Zhang, Jongdae Lee, and Ailin Tao. 2021. "Platelets, Not an Insignificant Player in Development of Allergic Asthma" Cells 10, no. 8: 2038. https://doi.org/10.3390/cells10082038

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop