Next Article in Journal
The Activation of the Fibrodysplasia Ossificans Progressiva-Inducing ALK2-R206H Mutant Depends on the Distinct Homo-Oligomerization Patterns of ACVR2B and ACVR2A
Previous Article in Journal
EZH2 Inhibition Sensitizes IDH1R132H-Mutant Gliomas to Histone Deacetylase Inhibitor
Previous Article in Special Issue
Choroidal Mast Cells and Pathophysiology of Age-Related Macular Degeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Beyond Allergies—Updates on The Role of Mas-Related G-Protein-Coupled Receptor X2 in Chronic Urticaria and Atopic Dermatitis

1
Institute of Allergology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
2
Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Allergology and Immunology, 12203 Berlin, Germany
*
Author to whom correspondence should be addressed.
Cells 2024, 13(3), 220; https://doi.org/10.3390/cells13030220
Submission received: 9 October 2023 / Revised: 14 December 2023 / Accepted: 19 January 2024 / Published: 25 January 2024
(This article belongs to the Special Issue New Insights into Mast Cells Biology)

Abstract

:
Mast cells (MCs) are an important part of the immune system, responding both to pathogens and toxins, but they also play an important role in allergic diseases, where recent data show that non-IgE-mediated activation is also of relevance, especially in chronic urticaria (CU) and atopic dermatitis (AD). Skin MCs express Mas-related G-protein-coupled receptor X2 (MRGPRX2), a key protein in non-IgE-dependent MC degranulation, and its overactivity is one of the triggering factors for the above-mentioned diseases, making MRGPRX2 a potential therapeutic target. Reviewing the latest literature revealed our need to focus on the discovery of MRGPRX2 activators as well as the ongoing vast research towards finding specific MRGPRX2 inhibitors for potential therapeutic approaches. Most of these studies are in their preliminary stages, with one drug currently being investigated in a clinical trial. Future studies and improved model systems are needed to verify whether any of these inhibitors may have the potential to be the next therapeutic treatment for CU, AD, and other pseudo-allergic reactions.

1. Introduction

Inflammatory skin diseases have an adverse impact on the physical, psychological, and social wellbeing of patients, thereby highly compromising their quality of life along with that of their caregivers [1,2,3]. Atopic dermatitis (AD) and chronic urticaria (CU) are examples of such diseases, with an increasing global prevalence [4,5,6]. The major challenge in the treatment of such diseases is that they are complex and heterogeneous with respect to their pathogenesis. Nevertheless, the adverse impact of these chronic inflammatory skin diseases on public health highlights the need for improved treatment approaches and novel therapeutic agents [3,7,8]. Understanding the underlying pathophysiology of these skin diseases is essential for the discovery of novel therapeutic and preventative strategies.
The skin, the body’s largest organ, serves as a critical site for a variety of inflammatory processes, including immunity against infections and allergy [9]. Mast cells (MCs) are located within the dermis of the skin [10,11], in close proximity to blood vessels and sensory nerves, constituting about 10% of all dermal immune cells [12]. MCs play an important role in the immune response by detecting external stimuli such as pathogens and toxins; however, dysregulation of their activity or functionality may contribute to hypersensitivity reactions and associated disorders [13,14]. The pathophysiology of CU is not well understood [15,16]; however, MC activation and subsequent degranulation result in the development of urticaria symptoms [17], including wheels and/or angioedema [18]. MC degranulation releases histamine and other mediators such as MC-specific proteases and low levels of cytokines, leading to sensory nerve stimulation, vasodilation, and plasma extravasation as well as to the recruitment of other immune cells to the urticaria site [15]. Abnormal regulation of MC activity may also contribute to AD [19], a chronic inflammatory skin disease which is characterized by recurrent eczematous skin lesions with intense pruritus and type-2 immunity-associated hypersensitivity [20]. It has been observed that the number of MCs in the skin lesions of AD patients are significantly associated with the disease’s severity [21]. The inter-individual differences in MCs among humans may well underlie the heterogeneity of complex inflammatory skin disorders [22]. However, the comprehensive modes of MC activation have not all been unraveled yet.
The activation of MCs occurs (i) via the immunoglobulin E (IgE)-dependent and (ii) IgE-independent pathways [23,24]. It is hypothesized that the highly expressed Mas-related G-protein-coupled receptor X2 (MRGPRX2) constitutes the missing link connecting MCs to AD and CU, at least in selected endotypes where the disorders are non-IgE-dependent [25]. There are many reports on the involvement of IgE-independent reactions, mainly to food additives and drugs, in CU, AD, and other inflammatory disease. However, it is not well understood yet if they act via the MRGPRX2 receptor [26,27,28,29,30,31,32]. This review focuses on MRGPRX2-activated non-IgE MC hypersensitivity, hereby referred to as pseudo-allergic reactions for simplicity [33,34].

2. MRGPRX2 Receptor

MRGPRX2, a 37-kDa G-coupled receptor with seven transmembrane domains, is highly expressed in human skin MCs [35]. It is a primate-specific protein with a 53% sequence similarity to its mouse ortholog, MrgprB2 [36,37].
The activation of MRGPRX2 is initiated by many exogenous and endogenous substances, including cationic drugs, neuropeptides, and host defense peptides. These agents are important for mounting antimicrobial defenses and mediating neurogenic inflammation but may also trigger pseudo-allergic reactions [36,38]. The activation of MRGPRX2 induces a signaling cascade leading to elevated cytosolic calcium levels, mediated through Ca++ channels, Gai, Gaq, ERK, PI3K/AKT, and PLCγ, culminating in MC degranulation [13,39]. This process ultimately results in the release of a variety of synthesized mediators [40].
It is noteworthy that MRGPRX2 has been shown to be a fundamental signaling protein for MC degranulation in pseudo-allergic reactions, and these reactions themselves may serve as triggering factors in the pathogenesis of conditions such as CU and AD [41].

3. Current Insights into MRGPRX2 in CU and AD

To gain a comprehensive understanding of existing knowledge regarding the role of MRGPRX2 in the context of CU and AD, a thorough literature search was conducted in PubMed utilizing the key words “atopic dermatitis” or “urticaria” and “MRGPRX2”, filtering only for reviews, as described in Figure 1. The findings up until 2023 are summarized in Table 1.
Previous research has identified and confirmed that there is a role for MRGPRX2 in inflammatory skin diseases. An increased proportion of MRGPRX2-positive MCs and increased levels of this receptor’s expression in MCs have been shown in the skin of CU [19,21,35,42] and AD patients [7,43]. However [15], whether there is really a difference in the MC numbers between patients and healthy controls is not yet certain given that some studies reported increased mast cell numbers in both lesioned and non-lesioned skin samples [44,45]. Other studies found no difference between the mast cell numbers in the skin of CU patients and those in the skin of healthy individuals [46,47,48]. Future studies are needed to resolve this question.
Furthermore, the MRGPRX2 agonist, substance P (SP), also showed elevated serum levels in CU and AD patients that correlated with disease severity [7,35,42,49]. Intradermal administration of SP induced greater wheal reactions in CU patients compared to normal controls, which may be due to increased MRGPRX2 expression in CU patients [21,42].
In summary, MRGPRX2 and its ligands are likely involved in the pathogenesis of AD and CU, although the exact dimension of their contribution and mechanisms of action are not known.
Table 1. MRGPRX2-related reviews regarding AD and CU.
Table 1. MRGPRX2-related reviews regarding AD and CU.
DiseaseIncreased Expression in PatientsIncreased Ligand Level (SP)Increase in MCs Number or ActivityBiomarker PotentialReview SourceOriginal Research Citations
ADYesYes (plasma)Yes, number and activityYes[7][50]
ADNot investigatedNot investigatedYes, numberNot investigated[43][51]
ADYesYes (skin)Yes, numberNot investigated[25][50,52,53,54,55,56]
CUYesNot investigatedYes, number and activityNot investigated[57][44,45,51,58,59,60]
CUYesNot investigatedYes, numberYes[21][51,61]
CUYesNot investigatedYes, activityCorrelated with disease severity[42][51,62,63,64]
CUYesNot investigatedNot investigatedNot investigated[19][51]
CU, ADYesYes (in AD, skin)Yes, number (AD)Not investigated[49][50,53,65,66,67]
CU, ADYesYes (serum and skin)Not investigatedYes[35][46,51,61,68]
CU, ADYesNot investigatedNot investigatedNot investigated[69][70,71]
Inflammatory skin diseasesNot investigatedNot investigatedNot investigatedNot investigated[37][51]

4. Recent Advances in MRGPRX2 Research

To have an overview of current MRGPRX2 research in relation to allergic diseases, we conducted a literature search in PubMed using key words “atopic dermatitis” or “urticaria” and “MRGPRX2”, following the search string as described in Figure 1, however, additionally filtering only for the latest research, from 2020 until 2023. These findings are summarized in Table 2.
The latest research on the role of MRGPRX2 in skin inflammatory diseases yielded compelling insights into these complex immunological processes. These studies have shown increased MRGPRX2 expression in the MCs of CU patients and in the sera of severe-CU patients [51,62]. Given the central role of MRGPRX2 in pseudo-allergic reactions, research has focused on the discovery of MRGPRX2 activators, which could unravel disease elicitors, inhibitors for pharmaceutical intervention discovery, as well as understanding of the MRGPRX2 signaling pathway which leads to MC degranulation.

4.1. MRGPRX2 Activators and Signaling

Further progress has been made regarding MRGPRX2-activating molecules. The high-throughput screening of a library of pharmacologically active compounds has uncovered a class of commonly used drugs that activate MRGPRX2/Mrgprb2, all belonging to the category of cationic amphiphilic drugs [72]. Moreover, new synthetic molecules in the morphine compound family have been shown to specifically activate MRGPRX2/Mrgprb2 [73]. Interestingly, given that more than 90% of AD patients present Staphylococcus aureus colonization in the affected skin areas, recent investigations have shown that the Staphylococcus δ-toxin provokes MC degranulation via MRGPRX2 and that the compound QWF, a MRGPRX2 antagonist, inhibits this activation [74]. Further elucidation of MRGPRX2’s interaction with SP revealed that SP is a balanced MRGPRX2 agonist. Their interaction induces both G-protein-dependent signaling for degranulation and G-protein-independent signaling for β-arrestin recruitment and MRGPRX2 internalization [75]. A highly conserved tyrosine residue within MRGPRX2, Tyr279 plays a crucial role in modulating this response, as the replacement of Tyr279 to alanine (Y279A) abolishes SP-induced modulation [75]. β-arrestins have been reported to be negative regulators of MRGPRX2 function in human skin MCs; therefore, strengthening the β-arrestin function could provide novel therapeutic approaches for MRGPRX2-mediated diseases [76,77].
Furthermore, in another study, it was described that MRGPRX2 activation led to microphthalmia-associated transcription factor (MITF) phosphorylation and increased MITF levels, while the silencing or inhibition of MITF resulted in decreased MRGPRX2-induced degranulation [78]. Thus, the modulation of MITF and MITF-dependent targets may be considered a therapeutic approach in the context of pseudo-allergic reactions [78].

4.2. MRGPRX2 Inhibitors—Potential Therapeutic Candidates

Following the recent discovery of the potential role of MRGPRX2 in pseudo-allergic reactions, there is ongoing vast research towards finding specific inhibitors of the MRGPRX2 signaling pathway for novel therapeutic approaches, and they are summarized in Table 2, while their mechanisms of action are illustrated in Figure 2.
The majority of the compounds currently under investigation have been so far only tested in vitro or in animal models. Among these, C9 stands out as a potent and selective inhibitor of only MRGPRX2-mediated MC degranulation, without affecting Mrgprb2-mediated MC degranulation [79]. Nonetheless, the toxicity and stability of this compound still need to be tested before proceeding with clinical trials.
Several compounds of plant origin have surfaced as MRGPRX2 inhibitory agents. Celastrol [80], for instance, has exhibited MRGPRX2 inhibition and has been shown to reduce MC production, histamine release, scratching levels, and inflammatory factor expression in mice. The inhibitory effect of Celastrol was reversed upon the overexpression of MRGPRX2 in a mouse model, supporting the notion that this effect may be mediated via MRGPRX2 rather than its orthologs [80]. In very high concentrations, Osthole, an aromatic compound, has displayed inhibitory effects on MGRPRX2 in two in vitro models (LAD2 cells and RBL-2H3 cells steadily expressing MRGPRX2), in MCs isolated from human skin tissue, and in vivo in a mouse model [81]. However, due to the high concentration of the compound needed to achieve said inhibitory effect, Osthole needs to be tested before being labelled as clinically safe. Additionally, the low water solubility of Osthole necessitates modification of the compound. However, it is uncertain whether these modifications preserve its inhibitory function. Two Chinese herb derivatives, Phenol [82] and Paeoniflorin [41], have been reported to decrease CU symptoms. In vitro studies in LAD2 cells and in vivo experiments in mice have confirmed them to be MRGPRX2 inhibitors.
The rest of the potential inhibitors that have been recently reported in research papers are quite different from one another, which may be due to the fact that the MRGPRX2 signaling pathway is not fully known. An example of this is Synta66, an inhibitor of the Orai channels, i.e., the calcium release-activated calcium channels in murine and human MCs, since, upon MRGPRX2-mediated activation of the inhibitor, an influx of Ca2+ is induced [13,83,84,85]. Taking an entirely different direction, the role of Hemokinin-1 (HK-1) in CU has also been investigated, as it was indicated to induce histamine release from LAD2 cells via MRGPRX2 in previous studies [86]. Higher serum levels of HK-1 were detected in healthy controls versus CU patients. Intriguingly, the brief incubation of MCs with HK-1 caused the inhibition of histamine release and did not elicit rapid MRGPRX2 internalization. This suggests that, in healthy controls, HK-1 may regulate and desensitize MRGPRX2-mediated MC activation, thereby preventing MC degranulation by SP [86].
Another approach was to examine an already existing medication. Clarithromycin, a well-known antibiotic, has been revisited for its potential in the treatment of skin inflammatory diseases. It has been shown to inhibit FcεRI- and MRGPRX2-mediated MCs activation in LAD2 cells. Furthermore, in a single-center, self-comparison clinical trial involving 28 CU patients who were not responsive to third-generation antihistamines [84], clarithromycin showed a significant reduction in the wheal and itch symptoms as well as a decrease in the serum cytokine levels (TNF-α, IL-13, IL-4, IL-6, and tryptase) in those patients. Interestingly, CD300f, a leukocyte mono-immunoglobulin-like receptor 3 and an inhibitory immune receptor which is expressed on the surface of neutrophil granulocytes and MCs [87], was shown to be involved in clarithromycin’s inhibitory effect, as the inhibition of said antibiotic decreased significantly after CD300f knockdown in LAD2 cells [84].
Ongoing research is dedicated to identifying specific MRGPRX2 inhibitors for potential therapeutic applications in pseudo-allergic reactions, encompassing diverse compounds. These candidates, which exhibit varying degrees of efficacy in in vitro and animal models, hold promise for addressing MRGPRX2-mediated immune responses in skin inflammatory diseases.
Table 2. Latest inhibitors research for potential novel therapeutic approaches.
Table 2. Latest inhibitors research for potential novel therapeutic approaches.
Inhibitor NameDiseaseMode of Action/MRGPRX2 Activation PathwayIn Vivo
Model
In Vitro
Model
Reference
CelastrolAD
  • Reduces MC number
  • Inhibits histamine release
  • Suppresses MRGPRX2/ORI
  • Celastrol effect was reversed by overexpression of MRGPRX2
Mouse modelN/A[80]
FisetinCU
  • MRGPRX2 binding
  • Suppresses calcium mobilization
  • Decreases phosphorylation of Akt, P38, NF-κB, and PLCγ
Mouse modelHEK293, LAD2 cells[88]
PaeonolCU
  • Reduces histamine chemokine release and calcium influx
  • Antagonist of Src kinase activity downstream of MRGPRX2
Mouse modelLAD2 cells[82]
Artemisinic acidCU
  • Not binding directly MRGPRX2
  • Lyn kinase antagonist
  • Inhibits MC activation
Mouse modelN/A[89]
C9CU, AD
  • Inhibits degranulation, β-arrestin recruitment internalization
Mouse modelRBL-2H3 cells, LAD2 cells, human skin-derived MCs[79]
Clarithromycin *CU
  • Inhibits MC activation
  • Reduces wheal, itch, and serum cytokine levels
Mouse model
CU patients (28)
LAD2 cells[84]
Synta66CU, AD
  • Orai channels inhibitor
  • Inhibits SP-induced Ca2+ mobilization, degranulation
  • Inhibits ERK1/2, Akt phosphorylation
Mouse modelLAD2 cells, human skin-derived MCs[83]
HK-1CU
  • Inhibits histamine release
  • Inhibits SP-induced MCs activation
---Cultured MCs,
human skin-derived MCs
[86,90]
α-Linolenic acidCU
  • Lyn kinase antagonist
  • Reduces SP-induced MCs degranulation, and histamine and chemokines release
Mouse modelLAD2 cells, HEK293 cells[90]
OstholeCU
  • Does not compete with the MRGPRX2 ligands
  • Reduces MRGPRX2 surface and intracellular expression
  • Inhibits Ca2+ mobilization, degranulation, and cytokine and chemokine production
Mouse modelLAD2 cells, RBL-2H3 cells, human skin-derived MCs[81]
PaeoniflorinAD
  • Inhibits MCs degranulation, histamine release
  • Reduce calcium influx,
  • Downregulates phospho-Erk1/2, P38, PKC, AKT
Mouse modelLAD2 cells[41]
* In clinical trial.

5. Discussion

The outlook of future therapeutic strategies for pseudo-allergic reactions aims to find specific inhibitors targeting MCs, including their activation via the MRGPRX2 receptor [37,49]. However, primarily, it should be confirmed which specific elicitors of MRGPRX2-mediated reactions occur in the context of inflammatory skin diseases such as AD and CU. Therefore, a comprehensive understanding of MRGPRX2’s function and regulation, including the discovery of specific MRGPRX2 regulators, is needed.
According to our findings, there are currently several potential MRGPRX2 inhibitors undergoing investigation. Most studies are in their preliminary stages, when MC-MRGPRX2 activation is prevented due to the addition of the compounds named previously. For some of the inhibitors, there is some (partial) explanation regarding their mechanism of action, whether this it is binding directly to MRGPRX2 (Fisetin [88]) or inhibiting protein activity downstream of MRGPRX2 (Paeoniflorin [41], Paeonol [82], and Synta66 [83]). Most inhibitors in our review were evaluated in in vitro and animal model experiments, and the antibiotic clarithromycin has been clinically tested [84]. Revisiting established antibiotics for the treatment of skin inflammatory diseases is an intriguing approach, as many antibiotics have an additional immunomodulating effect. For example, low-dose doxycycline is used to treat acne and other skin diseases involving an overactive immune system [91]. Some antibiotics, such as dapsone, have been used in the past to treat urticaria [92]. Therefore, it would be worthwhile to explore the old literature regarding the use of various antibiotics in the treatment of CU and AD [93]. Unfortunately, due to the lack of clinical trials meeting modern standards, these treatments have often been overlooked or classified in treatment guidelines as having an insufficient level of evidence [94]. Notably, antibiotics, especially those for H. pylori and dapsone, have demonstrated improved remission rates and symptom relief in CU cases, with few adverse events, warranting further studies [93].
One of the limitations in MRGPRX2 research is the lack of animal models for studying these primate-specific receptors [95]. Most in vivo research was conducted in the mouse model C57BL/6, using Mrgprb2, the ortholog of MRGPRX2. The work by Bawazir et al. [79] has raised a question regarding the applicability of MRGPRX2 inhibitor study results from mice, since the mouse ortholog has other variants with a lot of sequence similarities, potentially leading to cross-activation. These variants differ from MRGPRX2, challenging the direct translation of animal model-based findings to human conditions [79]. Nonetheless, to address this challenge, mouse models generating MRGPRX2-expressing MCs have been developed [96].
In reviewing the presented inhibitors, we highlight three molecules that show the highest therapeutic potential so far. Bawazir et al. [79] presented C9 as a potent and, importantly, selective inhibitor of only MRGPRX2-mediated MC degranulation. Yao C et al. [80] found that the improvements of AD caused by Celastrol were reversed by means of treatment with MRGPRX2 overexpression, indicating that Celastrol might affect AD via MRGPRX2. Clarithromycin already showed positive outcomes in a preliminary clinical trial in the treatment of CU. Future studies on these compounds are needed to elucidate their clinical significance.

6. Conclusions

In summary, the identification and characterization of MRGPRX2 have ushered in a significant paradigm shift in our understanding of mast cell (MC) biology, shedding light on non-IgE-mediated clinical manifestations which are mediated by MCs [25]. The emergence of high-affinity receptor inhibitors targeting MRGPRX2 presents a promising and innovative therapeutic approach for addressing MC-mediated diseases including CU and AD. Future studies encompassing clinical trials will be needed to verify the clinical efficacy and safety profiles of these inhibitors. Consequently, additional research endeavors are warranted to explore the potential therapeutic applications of these inhibitors not only in CU and AD but also in a broader spectrum of pseudo-allergic reactions.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

MCsMast cells
CUChronic urticaria
ADAtopic dermatitis
IgEImmunoglobulin E
MRGPRX2Mas-related G-protein-coupled receptor X2

References

  1. Finlay, A.Y. The burden of skin disease: Quality of life, economic aspects and social issues. Clin. Med. 2009, 9, 592–594. [Google Scholar] [CrossRef] [PubMed]
  2. Chu, S.; Mehrmal, S.; Uppal, P.; Giesey, R.L.; Delost, M.E.; Delost, G.R. Burden of skin disease and associated socioeconomic status in Europe: An ecologic study from the Global Burden of Disease Study 2017. JAAD Int. 2020, 1, 95–103. [Google Scholar] [CrossRef]
  3. Zuberbier, T.; Lotvall, J.; Simoens, S.; Subramanian, S.V.; Church, M.K. Economic burden of inadequate management of allergic diseases in the European Union: A GA(2) LEN review. Allergy 2014, 69, 1275–1279. [Google Scholar] [CrossRef] [PubMed]
  4. Langan, S.M.; Mulick, A.R.; Rutter, C.E.; Silverwood, R.J.; Asher, I.; García-Marcos, L.; Ellwood, E.; Bissell, K.; Chiang, C.-Y.; Sony, A.E.; et al. Trends in eczema prevalence in children and adolescents: A Global Asthma Network Phase I Study. Clin. Exp. Allergy 2023, 53, 337–352. [Google Scholar] [CrossRef]
  5. Goncalo, M.; Gimenez-Arnau, A.; Al-Ahmad, M.; Ben-Shoshan, M.; Bernstein, J.A.; Ensina, L.F.; Fomina, D.; Galvan, C.A.; Godse, K.; Grattan, C.; et al. The global burden of chronic urticaria for the patient and society. Br. J. Dermatol. 2021, 184, 226–236. [Google Scholar] [CrossRef] [PubMed]
  6. Fricke, J.; Avila, G.; Keller, T.; Weller, K.; Lau, S.; Maurer, M.; Zuberbier, T.; Keil, T. Prevalence of chronic urticaria in children and adults across the globe: Systematic review with meta-analysis. Allergy 2020, 75, 423–432. [Google Scholar] [CrossRef] [PubMed]
  7. Thapaliya, M.; Chompunud Na Ayudhya, C.; Amponnawarat, A.; Roy, S.; Ali, H. Mast Cell-Specific MRGPRX2: A Key Modulator of Neuro-Immune Interaction in Allergic Diseases. Curr. Allergy Asthma Rep. 2021, 21, 3. [Google Scholar] [CrossRef]
  8. Kulthanan, K.; Chusakul, S.; Recto, M.T.; Gabriel, M.T.; Aw, D.C.W.; Prepageran, N.; Wong, A.; Leong, J.L.; Foong, H.; Quang, V.T.; et al. Economic Burden of the Inadequate Management of Allergic Rhinitis and Urticaria in Asian Countries Based on the GA(2)LEN Model. Allergy Asthma Immunol. Res. 2018, 10, 370–378. [Google Scholar] [CrossRef]
  9. Richmond, J.M.; Harris, J.E. Immunology and skin in health and disease. Cold Spring Harb. Perspect. Med. 2014, 4, a015339. [Google Scholar] [CrossRef]
  10. Kabashima, K.; Honda, T.; Ginhoux, F.; Egawa, G. The immunological anatomy of the skin. Nat. Rev. Immunol. 2019, 19, 19–30. [Google Scholar] [CrossRef]
  11. Wilgus, T.A.; Wulff, B.C. The Importance of Mast Cells in Dermal Scarring. Adv. Wound Care. 2014, 3, 356–365. [Google Scholar] [CrossRef]
  12. Dwyer, D.F.; Barrett, N.A.; Austen, K.F.; Immunological Genome Project, C. Expression profiling of constitutive mast cells reveals a unique identity within the immune system. Nat. Immunol. 2016, 17, 878–887. [Google Scholar] [CrossRef]
  13. Wang, Z.; Franke, K.; Bal, G.; Li, Z.; Zuberbier, T.; Babina, M. MRGPRX2-Mediated Degranulation of Human Skin Mast Cells Requires the Operation of G(alphai), G(alphaq), Ca++ Channels, ERK1/2 and PI3K-Interconnection between Early and Late Signaling. Cells 2022, 11, 953. [Google Scholar] [CrossRef] [PubMed]
  14. Sobiepanek, A.; Kuryk, L.; Garofalo, M.; Kumar, S.; Baran, J.; Musolf, P.; Siebenhaar, F.; Fluhr, J.W.; Kobiela, T.; Plasenzotti, R.; et al. The Multifaceted Roles of Mast Cells in Immune Homeostasis, Infections and Cancers. Int. J. Mol. Sci. 2022, 23, 2249. [Google Scholar] [CrossRef] [PubMed]
  15. Church, M.K.; Kolkhir, P.; Metz, M.; Maurer, M. The role and relevance of mast cells in urticaria. Immunol. Rev. 2018, 282, 232–247. [Google Scholar] [CrossRef]
  16. Kolkhir, P.; Gimenez-Arnau, A.M.; Kulthanan, K.; Peter, J.; Metz, M.; Maurer, M. Urticaria. Nat. Rev. Dis. Primers 2022, 8, 61. [Google Scholar] [CrossRef]
  17. Bracken, S.J.; Abraham, S.; MacLeod, A.S. Autoimmune Theories of Chronic Spontaneous Urticaria. Front. Immunol. 2019, 10, 627. [Google Scholar] [CrossRef]
  18. Zuberbier, T.; Aberer, W.; Asero, R.; Abdul Latiff, A.H.; Baker, D.; Ballmer-Weber, B.; Bernstein, J.A.; Bindslev-Jensen, C.; Brzoza, Z.; Buense Bedrikow, R.; et al. The EAACI/GA2LEN/EDF/WAO guideline for the definition, classification, diagnosis and management of urticaria. Allergy 2018, 73, 1393–1414. [Google Scholar] [CrossRef] [PubMed]
  19. Olivera, A.; Beaven, M.A.; Metcalfe, D.D. Mast cells signal their importance in health and disease. J. Allergy Clin. Immunol. 2018, 142, 381–393. [Google Scholar] [CrossRef] [PubMed]
  20. Weidinger, S.; Novak, N. Atopic dermatitis. Lancet 2016, 387, 1109–1122. [Google Scholar] [CrossRef]
  21. Numata, T.; Harada, K.; Nakae, S. Roles of Mast Cells in Cutaneous Diseases. Front. Immunol. 2022, 13, 923495. [Google Scholar] [CrossRef]
  22. Babina, M.; Guhl, S.; Artuc, M.; Trivedi, N.N.; Zuberbier, T. Phenotypic variability in human skin mast cells. Exp. Dermatol. 2016, 25, 434–439. [Google Scholar] [CrossRef]
  23. Ferry, X.; Brehin, S.; Kamel, R.; Landry, Y. G protein-dependent activation of mast cell by peptides and basic secretagogues. Peptides 2002, 23, 1507–1515. [Google Scholar] [CrossRef]
  24. Metcalfe, D.D.; Baram, D.; Mekori, Y.A. Mast cells. Physiol. Rev. 1997, 77, 1033–1079. [Google Scholar] [CrossRef]
  25. Wang, Z.; Babina, M. MRGPRX2 signals its importance in cutaneous mast cell biology: Does MRGPRX2 connect mast cells and atopic dermatitis? Exp. Dermatol. 2020, 29, 1104–1111. [Google Scholar] [CrossRef]
  26. Zuberbier, T.; Chantraine-Hess, S.; Hartmann, K.; Czarnetzki, B.M. Pseudoallergen-free diet in the treatment of chronic urticaria. A prospective study. Acta Derm. Venereol. 1995, 75, 484–487. [Google Scholar] [CrossRef]
  27. Worm, M.; Ehlers, I.; Sterry, W.; Zuberbier, T. Clinical relevance of food additives in adult patients with atopic dermatitis. Clin. Exp. Allergy 2000, 30, 407–414. [Google Scholar] [CrossRef]
  28. Magerl, M.; Pisarevskaja, D.; Scheufele, R.; Zuberbier, T.; Maurer, M. Effects of a pseudoallergen-free diet on chronic spontaneous urticaria: A prospective trial. Allergy 2010, 65, 78–83. [Google Scholar] [CrossRef] [PubMed]
  29. Zuberbier, T.; Pfrommer, C.; Specht, K.; Vieths, S.; Bastl-Borrmann, R.; Worm, M.; Henz, B.M. Aromatic components of food as novel eliciting factors of pseudoallergic reactions in chronic urticaria. J. Allergy Clin. Immunol. 2002, 109, 343–348. [Google Scholar] [CrossRef] [PubMed]
  30. Reese, I.; Zuberbier, T.; Bunselmeyer, B.; Erdmann, S.; Henzgen, M.; Fuchs, T.; Jager, L.; Kleine-Tebbe, J.; Lepp, U.; Niggemann, B.; et al. Diagnostic approach for suspected pseudoallergic reaction to food ingredients. J. Dtsch. Dermatol. Ges. 2009, 7, 70–77. [Google Scholar] [CrossRef] [PubMed]
  31. Zuberbier, T. The role of allergens and pseudoallergens in urticaria. J. Investig. Dermatol. Symp. Proc. 2001, 6, 132–134. [Google Scholar] [CrossRef]
  32. Yu, R.J.; Krantz, M.S.; Phillips, E.J.; Stone, C.A., Jr. Emerging Causes of Drug-Induced Anaphylaxis: A Review of Anaphylaxis-Associated Reports in the FDA Adverse Event Reporting System (FAERS). J. Allergy Clin. Immunol. Pract. 2021, 9, 819–829.e2. [Google Scholar] [CrossRef]
  33. McNeil, B.D. MRGPRX2 and Adverse Drug Reactions. Front. Immunol. 2021, 12, 676354. [Google Scholar] [CrossRef]
  34. Tatemoto, K.; Nozaki, Y.; Tsuda, R.; Konno, S.; Tomura, K.; Furuno, M.; Ogasawara, H.; Edamura, K.; Takagi, H.; Iwamura, H.; et al. Immunoglobulin E-independent activation of mast cell is mediated by Mrg receptors. Biochem. Biophys. Res. Commun. 2006, 349, 1322–1328. [Google Scholar] [CrossRef]
  35. Kuhn, H.; Kolkhir, P.; Babina, M.; Dull, M.; Frischbutter, S.; Fok, J.S.; Jiao, Q.; Metz, M.; Scheffel, J.; Wolf, K.; et al. Mas-related G protein-coupled receptor X2 and its activators in dermatologic allergies. J. Allergy Clin. Immunol. 2021, 147, 456–469. [Google Scholar] [CrossRef]
  36. McNeil, B.D.; Pundir, P.; Meeker, S.; Han, L.; Undem, B.J.; Kulka, M.; Dong, X. Identification of a mast-cell-specific receptor crucial for pseudo-allergic drug reactions. Nature 2015, 519, 237–241. [Google Scholar] [CrossRef]
  37. Subramanian, H.; Gupta, K.; Ali, H. Roles of Mas-related G protein-coupled receptor X2 on mast cell-mediated host defense, pseudoallergic drug reactions, and chronic inflammatory diseases. J. Allergy Clin. Immunol. 2016, 138, 700–710. [Google Scholar] [CrossRef] [PubMed]
  38. Pundir, P.; Liu, R.; Vasavda, C.; Serhan, N.; Limjunyawong, N.; Yee, R.; Zhan, Y.; Dong, X.; Wu, X.; Zhang, Y.; et al. A Connective Tissue Mast-Cell-Specific Receptor Detects Bacterial Quorum-Sensing Molecules and Mediates Antibacterial Immunity. Cell Host Microbe 2019, 26, 114–122.e8. [Google Scholar] [CrossRef]
  39. Zhang, F.; Hong, F.; Wang, L.; Fu, R.; Qi, J.; Yu, B. MrgprX2 regulates mast cell degranulation through PI3K/AKT and PLCgamma signaling in pseudo-allergic reactions. Int. Immunopharmacol. 2022, 102, 108389. [Google Scholar] [CrossRef] [PubMed]
  40. Porebski, G.; Kwiecien, K.; Pawica, M.; Kwitniewski, M. Mas-Related G Protein-Coupled Receptor-X2 (MRGPRX2) in Drug Hypersensitivity Reactions. Front. Immunol. 2018, 9, 3027. [Google Scholar] [CrossRef] [PubMed]
  41. Wang, J.; Zhang, Y.; Wang, J.; Liu, R.; Zhang, G.; Dong, K.; Zhang, T. Paeoniflorin inhibits MRGPRX2-mediated pseudo-allergic reaction via calcium signaling pathway. Phytother. Res. 2020, 34, 401–408. [Google Scholar] [CrossRef] [PubMed]
  42. Quan, P.L.; Sabate-Bresco, M.; Guo, Y.; Martin, M.; Gastaminza, G. The Multifaceted Mas-Related G Protein-Coupled Receptor Member X2 in Allergic Diseases and Beyond. Int. J. Mol. Sci. 2021, 22, 4421. [Google Scholar] [CrossRef] [PubMed]
  43. Corbiere, A.; Loste, A.; Gaudenzio, N. MRGPRX2 sensing of cationic compounds-A bridge between nociception and skin diseases? Exp. Dermatol. 2021, 30, 193–200. [Google Scholar] [CrossRef] [PubMed]
  44. Haas, N.; Toppe, E.; Henz, B.M. Microscopic morphology of different types of urticaria. Arch. Dermatol. 1998, 134, 41–46. [Google Scholar] [CrossRef] [PubMed]
  45. Kay, A.B.; Ying, S.; Ardelean, E.; Mlynek, A.; Kita, H.; Clark, P.; Maurer, M. Elevations in vascular markers and eosinophils in chronic spontaneous urticarial weals with low-level persistence in uninvolved skin. Br. J. Dermatol. 2014, 171, 505–511. [Google Scholar] [CrossRef]
  46. Smith, C.H.; Kepley, C.; Schwartz, L.B.; Lee, T.H. Mast cell number and phenotype in chronic idiopathic urticaria. J. Allergy Clin. Immunol. 1995, 96, 360–364. [Google Scholar] [CrossRef]
  47. Caproni, M.; Giomi, B.; Volpi, W.; Melani, L.; Schincaglia, E.; Macchia, D.; Manfredi, M.; D’Agata, A.; Fabbri, P. Chronic idiopathic urticaria: Infiltrating cells and related cytokines in autologous serum-induced wheals. Clin. Immunol. 2005, 114, 284–292. [Google Scholar] [CrossRef]
  48. Caproni, M.; Volpi, W.; Macchia, D.; Giomi, B.; Manfredi, M.; Campi, P.; Cardinali, C.; D’Agata, A.; Fabbri, P. Infiltrating cells and related cytokines in lesional skin of patients with chronic idiopathic urticaria and positive autologous serum skin test. Exp. Dermatol. 2003, 12, 621–628. [Google Scholar] [CrossRef]
  49. Roy, S.; Chompunud Na Ayudhya, C.; Thapaliya, M.; Deepak, V.; Ali, H. Multifaceted MRGPRX2: New insight into the role of mast cells in health and disease. J. Allergy Clin. Immunol. 2021, 148, 293–308. [Google Scholar] [CrossRef] [PubMed]
  50. Nattkemper, L.A.; Tey, H.L.; Valdes-Rodriguez, R.; Lee, H.; Mollanazar, N.K.; Albornoz, C.; Sanders, K.M.; Yosipovitch, G. The Genetics of Chronic Itch: Gene Expression in the Skin of Patients with Atopic Dermatitis and Psoriasis with Severe Itch. J. Investig. Dermatol. 2018, 138, 1311–1317. [Google Scholar] [CrossRef]
  51. Fujisawa, D.; Kashiwakura, J.; Kita, H.; Kikukawa, Y.; Fujitani, Y.; Sasaki-Sakamoto, T.; Kuroda, K.; Nunomura, S.; Hayama, K.; Terui, T.; et al. Expression of Mas-related gene X2 on mast cells is upregulated in the skin of patients with severe chronic urticaria. J. Allergy Clin. Immunol. 2014, 134, 622–633.e9. [Google Scholar] [CrossRef] [PubMed]
  52. Kawakami, T.; Ando, T.; Kimura, M.; Wilson, B.S.; Kawakami, Y. Mast cells in atopic dermatitis. Curr. Opin. Immunol. 2009, 21, 666–678. [Google Scholar] [CrossRef]
  53. Jarvikallio, A.; Harvima, I.T.; Naukkarinen, A. Mast cells, nerves and neuropeptides in atopic dermatitis and nummular eczema. Arch. Dermatol. Res. 2003, 295, 2–7. [Google Scholar] [CrossRef]
  54. Jarvikallio, A.; Naukkarinen, A.; Harvima, I.T.; Aalto, M.L.; Horsmanheimo, M. Quantitative analysis of tryptase- and chymase-containing mast cells in atopic dermatitis and nummular eczema. Br. J. Dermatol. 1997, 136, 871–877. [Google Scholar] [CrossRef]
  55. Liu, F.T.; Goodarzi, H.; Chen, H.Y. IgE, mast cells, and eosinophils in atopic dermatitis. Clin. Rev. Allergy Immunol. 2011, 41, 298–310. [Google Scholar] [CrossRef] [PubMed]
  56. Guerra Júnior, G.; de Luca, I.M.; Leonardo, M.B.; Vilela, M.M. Mast cell quantification in the skin of children with atopic dermatitis: Its value in diagnosis and in assessing the effectiveness of therapy. Allergol. Immunopathol. 1995, 23, 160–163. [Google Scholar]
  57. Elieh-Ali-Komi, D.; Metz, M.; Kolkhir, P.; Kocaturk, E.; Scheffel, J.; Frischbutter, S.; Terhorst-Molawi, D.; Fox, L.; Maurer, M. Chronic urticaria and the pathogenic role of mast cells. Allergol. Int. 2023, 72, 359–368. [Google Scholar] [CrossRef]
  58. Terhorst, D.; Koti, I.; Krause, K.; Metz, M.; Maurer, M. In chronic spontaneous urticaria, high numbers of dermal endothelial cells, but not mast cells, are linked to recurrent angio-oedema. Clin. Exp. Dermatol. 2018, 43, 131–136. [Google Scholar] [CrossRef]
  59. Natbony, S.F.; Phillips, M.E.; Elias, J.M.; Godfrey, H.P.; Kaplan, A.P. Histologic studies of chronic idiopathic urticaria. J. Allergy Clin. Immunol. 1983, 71, 177–183. [Google Scholar] [CrossRef]
  60. Nettis, E.; Dambra, P.; Loria, M.P.; Cenci, L.; Vena, G.A.; Ferrannini, A.; Tursi, A. Mast-cell phenotype in urticaria. Allergy 2001, 56, 915. [Google Scholar] [CrossRef]
  61. Borici-Mazi, R.; Kouridakis, S.; Kontou-Fili, K. Cutaneous responses to substance P and calcitonin gene-related peptide in chronic urticaria: The effect of cetirizine and dimethindene. Allergy 1999, 54, 46–56. [Google Scholar] [CrossRef]
  62. Cao, T.B.T.; Cha, H.Y.; Yang, E.M.; Ye, Y.M. Elevated MRGPRX2 Levels Related to Disease Severity in Patients With Chronic Spontaneous Urticaria. Allergy Asthma Immunol. Res. 2021, 13, 498–506. [Google Scholar] [CrossRef]
  63. Vena, G.A.; Cassano, N.; Di Leo, E.; Calogiuri, G.F.; Nettis, E. Focus on the role of substance P in chronic urticaria. Clin. Mol. Allergy 2018, 16, 24. [Google Scholar] [CrossRef] [PubMed]
  64. Wedi, B.; Gehring, M.; Kapp, A. The pseudoallergen receptor MRGPRX2 on peripheral blood basophils and eosinophils: Expression and function. Allergy 2020, 75, 2229–2242. [Google Scholar] [CrossRef]
  65. Steinhoff, M.; Neisius, U.; Ikoma, A.; Fartasch, M.; Heyer, G.; Skov, P.S.; Luger, T.A.; Schmelz, M. Proteinase-activated receptor-2 mediates itch: A novel pathway for pruritus in human skin. J. Neurosci. 2003, 23, 6176–6180. [Google Scholar] [CrossRef] [PubMed]
  66. Toyoda, M.; Morohashi, M. Morphological assessment of the effects of cyclosporin A on mast cell--nerve relationship in atopic dermatitis. Acta Derm. Venereol. 1998, 78, 321–325. [Google Scholar] [CrossRef] [PubMed]
  67. Sugiura, H.; Maeda, T.; Uehara, M. Mast cell invasion of peripheral nerve in skin lesions of atopic dermatitis. Acta Derm. Venereol. Suppl. 1992, 176, 74–76. [Google Scholar]
  68. Irani, A.M.; Sampson, H.A.; Schwartz, L.B. Mast cells in atopic dermatitis. Allergy 1989, 44 (Suppl. S9), 31–34. [Google Scholar] [CrossRef]
  69. Babina, M. The pseudo-allergic/neurogenic route of mast cell activation via MRGPRX2: Discovery, functional programs, regulation, relevance to disease, and relation with allergic stimulation. Itch 2020, 5, e32. [Google Scholar] [CrossRef]
  70. Manorak, W.; Idahosa, C.; Gupta, K.; Roy, S.; Panettieri, R.; Ali, H. Upregulation of Mas-related G Protein coupled receptor X2 in asthmatic lung mast cells and its activation by the novel neuropeptide hemokinin-1. Respir. Res. 2018, 19, 1. [Google Scholar] [CrossRef]
  71. Aguilera-Lizarraga, J.; Florens, M.V.; Van Brussel, T.; Clevers, E.; Van Oudenhove, L.; Lambrechts, D.; Wouters, M.M.; Boeckxstaens, G.E. Expression of immune-related genes in rectum and colon descendens of Irritable Bowel Syndrome patients is unrelated to clinical symptoms. Neurogastroenterol. Motil. 2019, 31, e13579. [Google Scholar] [CrossRef]
  72. Wolf, K.; Kuhn, H.; Boehm, F.; Gebhardt, L.; Glaudo, M.; Agelopoulos, K.; Stander, S.; Ectors, P.; Zahn, D.; Riedel, Y.K.; et al. A group of cationic amphiphilic drugs activates MRGPRX2 and induces scratching behavior in mice. J. Allergy Clin. Immunol. 2021, 148, 506–522.e8. [Google Scholar] [CrossRef]
  73. Iio, K.; Kutsumura, N.; Nagumo, Y.; Saitoh, T.; Tokuda, A.; Hashimoto, K.; Yamamoto, N.; Kise, R.; Inoue, A.; Mizoguchi, H.; et al. Synthesis of unnatural morphinan compounds to induce itch-like behaviors in mice: Towards the development of MRGPRX2 selective ligands. Bioorganic Med. Chem. Lett. 2022, 56, 128485. [Google Scholar] [CrossRef]
  74. Azimi, E.; Reddy, V.B.; Lerner, E.A. Brief communication: MRGPRX2, atopic dermatitis and red man syndrome. Itch 2017, 2, e5. [Google Scholar] [CrossRef]
  75. Chompunud Na Ayudhya, C.; Amponnawarat, A.; Ali, H. Substance P Serves as a Balanced Agonist for MRGPRX2 and a Single Tyrosine Residue Is Required for beta-Arrestin Recruitment and Receptor Internalization. Int. J. Mol. Sci. 2021, 22, 5318. [Google Scholar] [CrossRef] [PubMed]
  76. Babina, M.; Wang, Z.; Roy, S.; Guhl, S.; Franke, K.; Artuc, M.; Ali, H.; Zuberbier, T. MRGPRX2 Is the Codeine Receptor of Human Skin Mast Cells: Desensitization through beta-Arrestin and Lack of Correlation with the FcepsilonRI Pathway. J. Investig. Dermatol. 2021, 141, 1286–1296.e4. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, Z.; Li, Z.; Bal, G.; Franke, K.; Zuberbier, T.; Babina, M. beta-arrestin-1 and beta-arrestin-2 Restrain MRGPRX2-Triggered Degranulation and ERK1/2 Activation in Human Skin Mast Cells. Front. Allergy 2022, 3, 930233. [Google Scholar] [CrossRef] [PubMed]
  78. Guo, Y.; Olle, L.; Proano-Perez, E.; Aparicio, C.; Guerrero, M.; Munoz-Cano, R.; Martin, M. MRGPRX2 signaling involves the Lysyl-tRNA synthetase and MITF pathway. Front. Immunol. 2023, 14, 1154108. [Google Scholar] [CrossRef] [PubMed]
  79. Bawazir, M.; Amponnawarat, A.; Hui, Y.; Oskeritzian, C.A.; Ali, H. Inhibition of MRGPRX2 but not FcepsilonRI or MrgprB2-mediated mast cell degranulation by a small molecule inverse receptor agonist. Front. Immunol. 2022, 13, 1033794. [Google Scholar] [CrossRef] [PubMed]
  80. Yao, C.; Ye, W.; Chen, M. Inhibition of Mast Cell Degranulation in Atopic Dermatitis by Celastrol through Suppressing MRGPRX2. Dis. Markers 2023, 2023, 9049256. [Google Scholar] [CrossRef] [PubMed]
  81. Callahan, B.N.; Kammala, A.K.; Syed, M.; Yang, C.; Occhiuto, C.J.; Nellutla, R.; Chumanevich, A.P.; Oskeritzian, C.A.; Das, R.; Subramanian, H. Osthole, a Natural Plant Derivative Inhibits MRGPRX2 Induced Mast Cell Responses. Front. Immunol. 2020, 11, 703. [Google Scholar] [CrossRef]
  82. Ding, Y.; Dang, B.; Zhang, Y.; Hu, S.; Wang, Y.; Zhao, C.; Zhang, T.; Gao, Z. Paeonol attenuates Substance P-induced urticaria by inhibiting Src kinase phosphorylation in mast cells. Cell. Immunol. 2023, 388–389, 104728. [Google Scholar] [CrossRef]
  83. Chaki, S.; Alkanfari, I.; Roy, S.; Amponnawarat, A.; Hui, Y.; Oskeritzian, C.A.; Ali, H. Inhibition of Orai Channel Function Regulates Mas-Related G Protein-Coupled Receptor-Mediated Responses in Mast Cells. Front. Immunol. 2021, 12, 803335. [Google Scholar] [CrossRef]
  84. Che, D.; Zhang, T.; Zhang, T.; Zheng, Y.; Hou, Y.; Geng, S.; He, L. Clarithromycin-treated chronic spontaneous urticaria with the negative regulation of FcepsilonRIota and MRGPRX2 activation via CD300f. Int. Immunopharmacol. 2022, 110, 109063. [Google Scholar] [CrossRef] [PubMed]
  85. Occhiuto, C.J.; Kammala, A.K.; Yang, C.; Nellutla, R.; Garcia, M.; Gomez, G.; Subramanian, H. Store-Operated Calcium Entry via STIM1 Contributes to MRGPRX2 Induced Mast Cell Functions. Front. Immunol. 2019, 10, 3143. [Google Scholar] [CrossRef]
  86. Nishimori, N.; Toyoshima, S.; Sasaki-Sakamoto, T.; Hayama, K.; Terui, T.; Okayama, Y. Serum level of hemokinin-1 is significantly lower in patients with chronic spontaneous urticaria than in healthy subjects. Allergol. Int. 2021, 70, 480–488. [Google Scholar] [CrossRef]
  87. Maehara, A.; Kaitani, A.; Izawa, K.; Shiba, E.; Nagamine, M.; Takamori, A.; Isobe, M.; Uchida, S.; Uchida, K.; Ando, T.; et al. Role of the Ceramide-CD300f Interaction in Gram-Negative Bacterial Skin Infections. J. Investig. Dermatol. 2018, 138, 1221–1224. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, Y.; Huang, Y.; Dang, B.; Hu, S.; Zhao, C.; Wang, Y.; Yuan, Y.; Liu, R. Fisetin alleviates chronic urticaria by inhibiting mast cell activation via MRGPRX2. J. Pharm. Pharmacol. 2023, 75, 1310–1321. [Google Scholar] [CrossRef]
  89. Ding, Y.; Dang, B.; Wang, Y.; Zhao, C.; An, H. Artemisinic acid attenuated symptoms of substance P-induced chronic urticaria in a mice model and mast cell degranulation via Lyn/PLC-p38 signal pathway. Int. Immunopharmacol. 2022, 113 Pt B, 109437. [Google Scholar] [CrossRef]
  90. Ding, Y.; Wang, Y.; Li, C.; Zhang, Y.; Hu, S.; Gao, J.; Liu, R.; An, H. alpha-Linolenic acid attenuates pseudo-allergic reactions by inhibiting Lyn kinase activity. Phytomedicine 2021, 80, 153391. [Google Scholar] [CrossRef] [PubMed]
  91. Miller, S.T.; Stevermer, J.J. Low-dose doxycycline moderately effective for acne. J. Fam. Pract. 2003, 52, 594+597. [Google Scholar] [PubMed]
  92. Liang, S.E.; Hoffmann, R.; Peterson, E.; Soter, N.A. Use of Dapsone in the Treatment of Chronic Idiopathic and Autoimmune Urticaria. JAMA Dermatol. 2019, 155, 90–95. [Google Scholar] [CrossRef] [PubMed]
  93. Watanabe, J.; Shimamoto, J.; Kotani, K. The Effects of Antibiotics for Helicobacter pylori Eradication or Dapsone on Chronic Spontaneous Urticaria: A Systematic Review and Meta-Analysis. Antibiotics 2021, 10, 156. [Google Scholar] [CrossRef] [PubMed]
  94. Sabroe, R.A.; Lawlor, F.; Grattan, C.E.H.; Ardern-Jones, M.R.; Bewley, A.; Campbell, L.; Flohr, C.; Leslie, T.A.; Marsland, A.M.; Ogg, G.; et al. British Association of Dermatologists guidelines for the management of people with chronic urticaria 2021. Br. J. Dermatol. 2022, 186, 398–413. [Google Scholar] [CrossRef]
  95. Al Hamwi, G.; Riedel, Y.K.; Clemens, S.; Namasivayam, V.; Thimm, D.; Muller, C.E. MAS-related G protein-coupled receptors X (MRGPRX): Orphan GPCRs with potential as targets for future drugs. Pharmacol. Ther. 2022, 238, 108259. [Google Scholar] [CrossRef]
  96. Mencarelli, A.; Gunawan, M.; Yong, K.S.M.; Bist, P.; Tan, W.W.S.; Tan, S.Y.; Liu, M.; Huang, E.K.; Fan, Y.; Chan, J.K.Y.; et al. A humanized mouse model to study mast cells mediated cutaneous adverse drug reactions. J. Leukoc. Biol. 2020, 107, 797–807. [Google Scholar] [CrossRef]
Figure 1. PubMed search strategy and selection of articles for inclusion in this review.
Figure 1. PubMed search strategy and selection of articles for inclusion in this review.
Cells 13 00220 g001
Figure 2. An illustration of the theoretical modes of action of the MRGPRX2 signaling pathway inhibitors. The inhibition symbol represents generalized inhibition including reduction in phosphorylation, reduction in expression, or direct inhibition.
Figure 2. An illustration of the theoretical modes of action of the MRGPRX2 signaling pathway inhibitors. The inhibition symbol represents generalized inhibition including reduction in phosphorylation, reduction in expression, or direct inhibition.
Cells 13 00220 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lerner, L.; Babina, M.; Zuberbier, T.; Stevanovic, K. Beyond Allergies—Updates on The Role of Mas-Related G-Protein-Coupled Receptor X2 in Chronic Urticaria and Atopic Dermatitis. Cells 2024, 13, 220. https://doi.org/10.3390/cells13030220

AMA Style

Lerner L, Babina M, Zuberbier T, Stevanovic K. Beyond Allergies—Updates on The Role of Mas-Related G-Protein-Coupled Receptor X2 in Chronic Urticaria and Atopic Dermatitis. Cells. 2024; 13(3):220. https://doi.org/10.3390/cells13030220

Chicago/Turabian Style

Lerner, Liron, Magda Babina, Torsten Zuberbier, and Katarina Stevanovic. 2024. "Beyond Allergies—Updates on The Role of Mas-Related G-Protein-Coupled Receptor X2 in Chronic Urticaria and Atopic Dermatitis" Cells 13, no. 3: 220. https://doi.org/10.3390/cells13030220

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