Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Trends Immunol ; 45(5): 371-380, 2024 May.
Article in English | MEDLINE | ID: mdl-38653601

ABSTRACT

Peripheral sensory neurons recognize diverse noxious stimuli, including microbial products and allergens traditionally thought to be targets of the mammalian immune system. Activation of sensory neurons by these stimuli leads to pain and itch responses as well as the release of neuropeptides that interact with their cognate receptors expressed on immune cells, such as dendritic cells (DCs). Neuronal control of immune cell function through neuropeptide release not only affects local inflammatory responses but can impact adaptive immune responses through downstream effects on T cell priming. Numerous neuropeptide receptors are expressed by DCs but only a few have been characterized, presenting opportunities for further investigation of the pathways by which cutaneous neuroimmune interactions modulate host immunity.


Subject(s)
Sensory Receptor Cells , Skin , Humans , Animals , Sensory Receptor Cells/immunology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Skin/immunology , Neuropeptides/metabolism , Neuropeptides/immunology , Dendritic Cells/immunology , Neuroimmunomodulation , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/immunology
2.
Int Immunopharmacol ; 102: 108389, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34920312

ABSTRACT

The G protein-coupled receptor MrgprX2 in mast cells is known to be a crucial receptor for pseudo-allergic reactions. MrgprX2 activation leads to elevated intracellular calcium levels and mast cell degranulation, but the underlying mechanism remains to be elucidated. Herein, we investigated the role of the phosphatidylinositol 3 kinase (PI3K)/serum-threonine kinase (AKT) signaling pathway and phospholipase C gamma (PLCγ) in mast cell degranulation mediated by MrgprX2 in LAD2 human-derived mast cells. The results showed that phosphorylated AKT (p-AKT) and PLCγ up-regulation were accompanied by an increase in intracellular calcium following activation of MrgprX2 by Compound 48/80, an inducer of mast cell degranulation. In contrast, p-AKT and PLCγ were down-regulated and intracellular calcium levels decreased after MrgprX2 knockdown. Mast cell degranulation was clearly suppressed; however, inhibiting PI3K and PLCγ phosphorylation did not influence MrgprX2 expression. The increase in calcium concentration was suppressed and mast cell degranulation was weakened. Furthermore, by inhibiting PI3K and PLCγ phosphorylation in animals, the allergic symptoms caused by C48/80 were obviously reduced. We deduced that during the mast cell degranulation observed in pseudoallergic reactions, MrgprX2 regulated intracellular calcium levels via the PI3K/AKT and PLCγ pathways.


Subject(s)
Hypersensitivity/immunology , Mast Cells/immunology , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Animals , Calcium/metabolism , Cell Degranulation , Cell Line , Humans , Hypersensitivity/metabolism , Mast Cells/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Phosphatidylinositol 3-Kinase/immunology , Phospholipase C gamma/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , Signal Transduction
3.
Front Immunol ; 12: 688930, 2021.
Article in English | MEDLINE | ID: mdl-34867939

ABSTRACT

Acute anaphylaxis to small molecule drugs is largely considered to be antibody-mediated with immunogloblin E (IgE) and mast cell activation being key. More recently, a role for drug-reactive immunoglobulin G (IgG) with neutrophil activation has also been suggested, at least in reactions to neuromuscular blocking agents (NMBAs). However, the mast cell receptor MRGPRX2 has also been highlighted as a possible triggering mechanism in acute anaphylaxis to many clinically used drugs. Significantly, MRGPRX2 activation is not dependent upon the presence of drug-recognising antibody. Given the reasonable assumption that MRGPRX2 is expressed in all individuals, the corollary of this is that in theory, anybody could respond detrimentally to triggering drugs (recently suggested to be around 20% of a drug-like compound library). But this clearly is not the case, as the incidence of acute drug-induced anaphylaxis is very low. In this mini-review we consider antibody-dependent and -independent mechanisms of mast cell activation by small molecule drugs with a focus on the MRGPRX2 pathway. Moreover, as a juxtaposition to these adverse drug actions, we consider how increased understanding of the role of MRGPRX2 in anaphylaxis is important for future drug development and can complement exploration of this receptor as a drug target in broader clinical settings.


Subject(s)
Anaphylaxis/immunology , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Anaphylaxis/etiology , Anaphylaxis/therapy , Drug Hypersensitivity/etiology , Drug Hypersensitivity/immunology , Drug Hypersensitivity/therapy , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/immunology , Drug-Related Side Effects and Adverse Reactions/therapy , Gene Expression , Humans , Mast Cell Activation Disorders/etiology , Mast Cell Activation Disorders/immunology , Mast Cell Activation Disorders/therapy , Mast Cells/drug effects , Mast Cells/immunology , Models, Immunological , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics
4.
Nature ; 600(7887): 164-169, 2021 12.
Article in English | MEDLINE | ID: mdl-34789875

ABSTRACT

In the clades of animals that diverged from the bony fish, a group of Mas-related G-protein-coupled receptors (MRGPRs) evolved that have an active role in itch and allergic signals1,2. As an MRGPR, MRGPRX2 is known to sense basic secretagogues (agents that promote secretion) and is involved in itch signals and eliciting pseudoallergic reactions3-6. MRGPRX2 has been targeted by drug development efforts to prevent the side effects induced by certain drugs or to treat allergic diseases. Here we report a set of cryo-electron microscopy structures of the MRGPRX2-Gi1 trimer in complex with polycationic compound 48/80 or with inflammatory peptides. The structures of the MRGPRX2-Gi1 complex exhibited shallow, solvent-exposed ligand-binding pockets. We identified key common structural features of MRGPRX2 and describe a consensus motif for peptidic allergens. Beneath the ligand-binding pocket, the unusual kink formation at transmembrane domain 6 (TM6) and the replacement of the general toggle switch from Trp6.48 to Gly6.48 (superscript annotations as per Ballesteros-Weinstein nomenclature) suggest a distinct activation process. We characterized the interfaces of MRGPRX2 and the Gi trimer, and mapped the residues associated with key single-nucleotide polymorphisms on both the ligand and G-protein interfaces of MRGPRX2. Collectively, our results provide a structural basis for the sensing of cationic allergens by MRGPRX2, potentially facilitating the rational design of therapies to prevent unwanted pseudoallergic reactions.


Subject(s)
Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Pruritus/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/metabolism , Allergens/immunology , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Consensus Sequence , Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Models, Molecular , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/ultrastructure , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/ultrastructure , Receptors, Neuropeptide/immunology , Receptors, Neuropeptide/ultrastructure
5.
Anal Biochem ; 633: 114392, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34597615

ABSTRACT

Mas-related G-protein-coupled receptor X2 (MRGPRX2) has recently been reported to be associated with anaphylaxis. Detection of MRGPRX2 levels in human peripheral blood might serve as a powerful tool for predicting the predisposition of patients to anaphylactic reactions. For rapid measurement of MRGPRX2, we established a paper-based double-antibody sandwich enzyme-linked immunosorbent assay (ELISA) using mouse monoclonal antibody and horseradish peroxidase (HRP)-labelled rabbit polyclonal antibody as capture antibody and detection antibody, respectively. We avoided chemical functionalization of the cellulose paper by introducing bovine serum albumin (BSA) to provide COOH and NH2 groups for covalent immobilization of the capture antibody. Through amide condensation, a two-layer immobilization strategy was applied with BSA-BSA and BSA-capture antibody networks as the first and second layers, respectively. This strategy improved the quantity, activity and stability of the immobilized antibody. We then established a paper-based ELISA to detect MRGPRX2 in human peripheral blood. Our method is less laborious, easier to implement, and more cost-effective than conventional ELISA, while offering similar sensitivity, specificity, and accuracy. Therefore, it could serve as an innovative clinical point-of-care diagnostic tool, especially in areas that lack advanced clinical equipment.


Subject(s)
Anaphylaxis/blood , Enzyme-Linked Immunosorbent Assay , Nerve Tissue Proteins/blood , Paper , Receptors, G-Protein-Coupled/blood , Receptors, Neuropeptide/blood , Anaphylaxis/immunology , Humans , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology
6.
Front Immunol ; 12: 689410, 2021.
Article in English | MEDLINE | ID: mdl-34248979

ABSTRACT

Pseudomonas aeruginosa is a frequent cause of hospital-acquired wound infection and is difficult to treat because it forms biofilms and displays antibiotic resistance. Previous studies in mice demonstrated that mast cells (MCs) not only contribute to P. aeruginosa eradication but also promote wound healing via an unknown mechanism. We recently reported that host defense peptides (HDPs) induce human MC degranulation via Mas-related G protein-coupled receptor-X2 (MRGPRX2). Small molecule HDP mimetics have distinct advantages over HDPs because they are inexpensive to synthesize and display high stability, bioavailability, and low toxicity. Murepavadin is a lipidated HDP mimetic, (also known as POL7080), which displays antibacterial activity against a broad panel of multi-drug-resistant P. aeruginosa. We found that murepavadin induces Ca2+ mobilization, degranulation, chemokine IL-8 and CCL3 production in a human MC line (LAD2 cells) endogenously expressing MRGPRX2. Murepavadin also caused degranulation in RBL-2H3 cells expressing MRGPRX2 but this response was significantly reduced in cells expressing missense variants within the receptor's ligand binding (G165E) or G protein coupling (V282M) domains. Compound 48/80 induced ß-arrestin recruitment and promoted receptor internalization, which resulted in substantial decrease in the subsequent responsiveness to the MRGPRX2 agonist. By contrast, murepavadin did not cause ß-arrestin-mediated MRGPRX2 regulation. Murepavadin induced degranulation in mouse peritoneal MCs via MrgprB2 (ortholog of human MRGPRX2) and caused increased vascular permeability in wild-type mice but not in MrgprB2-/- mice. The data presented herein demonstrate that murepavadin activates human MCs via MRGPRX2 and murine MCs via MrgprB2 and that MRGPRX2 is resistant to ß-arrestin-mediated receptor regulation. Thus, besides its direct activity against P. aeruginosa, murepavadin may contribute to bacterial clearance and promote wound healing by harnessing MC's immunomodulatory property via the activation of MRGPRX2.


Subject(s)
Anti-Bacterial Agents/pharmacology , Mast Cells/drug effects , Nerve Tissue Proteins/immunology , Peptides, Cyclic/pharmacology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Animals , Calcium/immunology , Cell Degranulation/drug effects , Cell Line , Cytokines/immunology , Female , Humans , Male , Mast Cells/immunology , Mice, Inbred C57BL , Mice, Knockout , Rats
7.
Allergol Int ; 70(4): 480-488, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34090787

ABSTRACT

BACKGROUND: We previously reported upregulation of expression of Mas-related G protein-coupled receptor X2 (MRGPRX2) on mast cells (MCs) in the skin of patients with severe chronic spontaneous urticaria (CSU). Serum levels of substance P (SP) were reportedly significantly elevated, in correlation with the severity of CSU. Hemokinin-1 (HK-1) reportedly induced histamine release from LAD2 cells via MRGPRX2. We aimed to investigate HK-1's role in CSU. METHODS: The concentrations of HK-1 and SP were measured using ELISAs. Skin- and synovium-derived cultured MCs were generated by culturing dispersed skin and synovial cells, respectively, with stem cell factor. MRGPRX2 expression in the MCs was reduced using a lentiviral shRNA silencing technique. RESULTS: Anti-SP Ab used in the SP ELISA showed 100% cross-reactivity to HK-1, but anti-HK-1 Ab showed 0% cross-reactivity to SP. The serum level of HK-1 was significantly lower in patients with CSU (n = 151) than in non-atopic healthy control (NC) subjects (n = 114). The EC50 of histamine release from MCs induced by HK-1 (5056 nM) was 12-fold higher than by SP (426 nM). Brief pretreatment of MCs with HK-1 at concentrations of 3.0-10 µM significantly reduced histamine release by 0.1 µM SP. However, brief incubation of MCs with HK-1 did not elicit rapid MRGPRX2 internalization. CONCLUSIONS: In NC subjects, high HK-1 concentrations may desensitize MGRPRX2-mediated MC activation, thereby preventing MC degranulation by SP.


Subject(s)
Chronic Urticaria/blood , Tachykinins/blood , Adolescent , Adult , Aged , Aged, 80 and over , Cells, Cultured , Chronic Urticaria/immunology , Female , Humans , Male , Mast Cells/immunology , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , RNA, Small Interfering/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/immunology , Skin/cytology , Synovial Membrane/cytology , Tachykinins/immunology , Young Adult
8.
J Allergy Clin Immunol ; 148(2): 293-308, 2021 08.
Article in English | MEDLINE | ID: mdl-33957166

ABSTRACT

Cutaneous mast cells (MCs) express Mas-related G protein-coupled receptor-X2 (MRGPRX2; mouse ortholog MrgprB2), which is activated by an ever-increasing number of cationic ligands. Antimicrobial host defense peptides (HDPs) generated by keratinocytes contribute to host defense likely by 2 mechanisms, one involving direct killing of microbes and the other via MC activation through MRGPRX2. However, its inappropriate activation may cause pseudoallergy and likely contribute to the pathogenesis of rosacea, atopic dermatitis, allergic contact dermatitis, urticaria, and mastocytosis. Gain- and loss-of-function missense single nucleotide polymorphisms in MRGPRX2 have been identified. The ability of certain ligands to serve as balanced or G protein-biased agonists has been defined. Small-molecule HDP mimetics that display both direct antimicrobial activity and activate MCs via MRGPRX2 have been developed. In addition, antibodies and reagents that modulate MRGPRX2 expression and signaling have been generated. In this article, we provide a comprehensive update on MrgprB2 and MRGPRX2 biology. We propose that harnessing MRGPRX2's host defense function by small-molecule HDP mimetics may provide a novel approach for the treatment of antibiotic-resistant cutaneous infections. In contrast, MRGPRX2-specific antibodies and inhibitors could be used for the modulation of allergic and inflammatory diseases that are mediated via this receptor.


Subject(s)
Mast Cells/immunology , Mutation, Missense , Nerve Tissue Proteins/immunology , Polymorphism, Single Nucleotide , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Skin Diseases , Skin/immunology , Animals , Anti-Infective Agents/therapeutic use , Biomimetic Materials/therapeutic use , Humans , Mast Cells/pathology , Mice , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Skin/pathology , Skin Diseases/drug therapy , Skin Diseases/genetics , Skin Diseases/immunology , Skin Diseases/pathology
9.
Int J Mol Sci ; 22(7)2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33808264

ABSTRACT

The IL-1 family cytokine IL-33 activates and re-shapes mast cells (MCs), but whether and by what mechanisms it elicits cytokines in MCs from human skin remains poorly understood. The current study found that IL-33 activates CCL1, CCL2, IL-5, IL-8, IL-13, and TNF-α, while IL-1ß, IL-6, IL-31, and VEGFA remain unaffected in cutaneous MCs, highlighting that each MC subset responds to IL-33 with a unique cytokine profile. Mechanistically, IL-33 induced the rapid (1-2 min) and durable (2 h) phosphorylation of p38, whereas the phosphorylation of JNK was weaker and more transient. Moreover, the NF-κB pathway was potently activated, as revealed by IκB degradation, increased nuclear abundance of p50/p65, and vigorous phosphorylation of p65. The activation of NF-κB occurred independently of p38 or JNK. The induced transcription of the cytokines selected for further study (CCL1, CCL2, IL-8, TNF-α) was abolished by interference with NF-κB, while p38/JNK had only some cytokine-selective effects. Surprisingly, at the level of the secreted protein products, p38 was nearly as effective as NF-κB for all entities, suggesting post-transcriptional involvement. IL-33 did not only instruct skin MCs to produce selected cytokines, but it also efficiently co-operated with the allergic and pseudo-allergic/neurogenic activation networks in the production of IL-8, TNF-α, CCL1, and CCL2. Synergism was more pronounced at the protein than at the mRNA level and appeared stronger for MRGPRX2 ligands than for FcεRI. Our results underscore the pro-inflammatory nature of an acute IL-33 stimulus and imply that especially in combination with allergens or MRGPRX2 agonists, IL-33 will efficiently amplify skin inflammation and thereby aggravate inflammatory dermatoses.


Subject(s)
Cytokines/immunology , Interleukin-33/pharmacology , Mast Cells/drug effects , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, IgE/immunology , Receptors, Neuropeptide/immunology , Skin/drug effects , p38 Mitogen-Activated Protein Kinases/immunology , Foreskin/cytology , Humans , Interleukin-33/immunology , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mast Cells/cytology , Mast Cells/immunology , Phosphorylation , Primary Cell Culture , Signal Transduction/drug effects , Skin/immunology
10.
J Allergy Clin Immunol ; 148(2): 506-522.e8, 2021 08.
Article in English | MEDLINE | ID: mdl-33617860

ABSTRACT

BACKGROUND: Mas gene-related G protein-coupled receptors (MRGPRs) are a G protein-coupled receptor family responsive to various exogenous and endogenous agonists, playing a fundamental role in pain and itch sensation. The primate-specific family member MRGPRX2 and its murine orthologue MRGPRB2 are expressed by mast cells mediating IgE-independent signaling and pseudoallergic drug reactions. OBJECTIVES: Our aim was to increase knowledge about the function and regulation of MRGPRX2/MRGPRB2, which is of major importance in prevention of drug hypersensitivity reactions and drug-induced pruritus. METHODS: To identify novel MRGPR (ant)agonists, we screened a library of pharmacologically active compounds by utilizing a high-throughput calcium mobilization assay. The identified hit compounds were analyzed for their pseudoallergic and pruritogenic effects in mice and human. RESULTS: We found a class of commonly used drugs activating MRGPRX2 that, to a large extent, consists of antidepressants, antiallergic drugs, and antipsychotics. Three-dimensional pharmacophore modeling revealed structural similarities of the identified agonists, classifying them as cationic amphiphilic drugs. Mast cell activation was investigated by using the 3 representatively selected antidepressants clomipramine, paroxetine, and desipramine. Indeed, we were able to show a concentration-dependent activation and MRGPRX2-dependent degranulation of the human mast cell line LAD2 (Laboratory of Allergic Diseases-2). Furthermore, clomipramine, paroxetine, and desipramine were able to induce degranulation of human skin and murine peritoneal mast cells. These substances elicited dose-dependent scratching behavior following intradermal injection into C57BL/6 mice but less so in MRGPRB2-mutant mice, as well as wheal-and-flare reactions following intradermal injections in humans. CONCLUSION: Our results contribute to the characterization of structure-activity relationships and functionality of MRGPRX2 ligands and facilitate prediction of adverse reactions such as drug-induced pruritus to prevent severe drug hypersensitivity reactions.


Subject(s)
Antidepressive Agents/adverse effects , Behavior, Animal/drug effects , Cell Degranulation/drug effects , Drug Hypersensitivity/immunology , Mast Cells/immunology , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Animals , Antidepressive Agents/pharmacology , Cell Line , Drug Hypersensitivity/pathology , Humans , Mast Cells/pathology , Mice , Nerve Tissue Proteins/agonists , Receptors, G-Protein-Coupled/agonists , Receptors, Neuropeptide/agonists
11.
J Allergy Clin Immunol ; 147(2): 456-469, 2021 02.
Article in English | MEDLINE | ID: mdl-33071069

ABSTRACT

The Mas-related G protein-coupled receptor X2 (MRGPRX2) is a multiligand receptor responding to various exogenous and endogenous stimuli. Being highly expressed on skin mast cells, MRGPRX2 triggers their degranulation and release of proinflammatory mediators, and it promotes multicellular signaling cascades, such as itch induction and transmission in sensory neurons. The expression of MRGPRX2 by skin mast cells and the levels of the MRGPRX2 agonists (eg, substance P, major basic protein, eosinophil peroxidase) are upregulated in the serum and/or skin of patients with inflammatory and pruritic skin diseases, such as chronic spontaneous urticaria or atopic dermatitis. Therefore, MRGPRX2 and its agonists might be potential biomarkers for the progression of cutaneous inflammatory diseases and the response to treatment. In addition, they may represent promising targets for prevention and treatment of signs and symptoms in patients with skin diseases or drug reactions. To assess this possibility, this review explores the role and relevance of MRGPRX2 and its activators in cutaneous inflammatory disorders and chronic pruritus.


Subject(s)
Chronic Urticaria/immunology , Dermatitis, Atopic/immunology , Nerve Tissue Proteins/immunology , Pruritus/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Animals , Chronic Urticaria/metabolism , Dermatitis, Atopic/metabolism , Humans , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Nerve Tissue Proteins/metabolism , Pruritus/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism
12.
Article in English | MEDLINE | ID: mdl-32727338

ABSTRACT

AIM: Investigating about the history of allergies and discovery of the histamine's role in the immune response through historical references, starting with ancient anecdotes, analysing the first immunization attempts on animals to understand its importance as the anaphylaxis mediator. Moreover, we shortly resume the most recent discoveries on mast cell role in allergic diseases throughout the latest updates on its antibody-independent receptors. METHODS: Publications, including reviews, treatment guidelines, historical and medical books, on the topic of interest were found on Medline, PubMed, Web of Knowledge, Web of Science, Google Scholar, Elsevier's (EMBASE.comvarious internet museum archives. Texts from the National Library of Greece (Stavros Niarchos Foundation), from the School of Health Sciences of the National and Kapodistrian University of Athens (Greece). We selected key articles which could provide ahistorical and scientific insight into histamine molecule and its mechanism of action's discovery starting with Egyptian, Greek and Chinese antiquity to end with the more recent pharmacological and molecular discoveries. RESULTS: Allergic diseases were described by medicine since ancient times, without exactly understanding the physio-pathologic mechanisms of immuno-mediated reactions and of their most important biochemical mediator, histamine. Researches on histamine and allergic mechanisms started at the beginning of the 20th century with the first experimental observations on animals of anaphylactic reactions. Histamine was then identified as their major mediator of many allergic diseases and anaphylaxis, but also of several physiologic body's functions, and its four receptors were characterized. Modern researches focus their attention on the fundamental role of the antibody-independent receptors of mast cells in allergic mechanisms, such as MRGPRX2, ADGRE2 and IL-33 receptor. CONCLUSION: New research should investigate how to modulate immunity cells activity in order to better investigate possible multi-target therapies for host's benefits in preclinical and clinical studies on allergic diseases in which mast cells play a major role.


Subject(s)
Allergens/immunology , Histamine/immunology , Hypersensitivity/immunology , Immunity, Cellular/immunology , Mast Cells/immunology , Allergens/metabolism , Anaphylaxis/drug therapy , Anaphylaxis/immunology , Anaphylaxis/metabolism , Animals , Histamine/metabolism , Histamine Antagonists/pharmacology , Histamine Antagonists/therapeutic use , History, 20th Century , History, 21st Century , History, Ancient , History, Medieval , Humans , Hypersensitivity/drug therapy , Hypersensitivity/metabolism , Immunity, Cellular/drug effects , Mast Cells/drug effects , Mast Cells/metabolism , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology
14.
Cell Immunol ; 358: 104239, 2020 12.
Article in English | MEDLINE | ID: mdl-33129497

ABSTRACT

Roxithromycin (ROX) is a macrolide antibiotic with a variety of immunological effects. Mast cells (MCs) play a key role in host defense, mediating hypersensitivity and pseudo-allergic reactions. Mas-related G protein-coupled receptor X2 (MrgprX2) is the main receptor related to pseudo-allergy. In this study, we investigated the anti-pseudo-allergy effect of ROX and its underlying mechanism. The effects of ROX on passive cutaneous anaphylaxis (PCA) and active systemic allergy were examined, degranulation, Ca2+ influx, and cytokine release were studied in vivo and in vitro. Interactions between ROX and MrgprX2 protein were also detected through surface plasmon resonance. The PCA and active systemic allergy induced by compound 48/80 were inhibited by ROX. An intermolecular interaction was detected between the ROX and MrgprX2 protein. In conclusion, ROX could inhibit pseudo-allergic reactions, and this effect involves the Ca2+/PLC/IP3 pathway of MrgprX2. This study provides new insight into the anti-pseudo-allergy effects of ROX.


Subject(s)
Hypersensitivity/drug therapy , Receptors, G-Protein-Coupled/metabolism , Roxithromycin/pharmacology , Anaphylaxis/chemically induced , Animals , Anti-Allergic Agents/pharmacology , Cell Degranulation/immunology , Cytokines/metabolism , Male , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/immunology , Passive Cutaneous Anaphylaxis/drug effects , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Roxithromycin/metabolism , p-Methoxy-N-methylphenethylamine/adverse effects , p-Methoxy-N-methylphenethylamine/metabolism
15.
Int J Mol Sci ; 21(21)2020 Oct 22.
Article in English | MEDLINE | ID: mdl-33105700

ABSTRACT

Members of neuropeptide B/W signaling system have been predominantly detected and mapped within the CNS. In the rat, this system includes neuropeptide B (NPB), neuropeptide W (NPW) and their specific receptor NPBWR1. This signaling system has a wide spectrum of functions including a role in modulation of inflammatory pain and neuroendocrine functions. Expression of NPB, NPW and NPBWR1 in separate heart compartments, dorsal root ganglia (DRG) and stellate ganglia was proven by RT-qPCR, Western blot (WB) and immunofluorescence. Presence of mRNA for all tested genes was detected within all heart compartments and ganglia. The presence of proteins preproNPB, preproNPW and NPBWR1 was confirmed in all the chambers of heart by WB. Expression of preproNPW and preproNPB was proven in cardiac ganglionic cells obtained by laser capture microdissection. In immunofluorescence analysis, NPB immunoreactivity was detected in nerve fibers, some nerve cell bodies and smooth muscle within heart and both ganglia. NPW immunoreactivity was present in the nerve cell bodies and nerve fibers of heart ganglia. Weak nonhomogenous staining of cardiomyocytes was present within heart ventricles. NPBWR1 immunoreactivity was detected on cardiomyocytes and some nerve fibers. We confirmed the presence of NPB/W signaling system in heart, DRG and stellate ganglia by proteomic and genomic analyses.


Subject(s)
Myocardium/metabolism , Neuropeptides/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Animals , Fluorescent Antibody Technique , Ganglia, Spinal/metabolism , Gene Expression , Male , Neuropeptides/immunology , Neuropeptides/metabolism , Rats, Zucker , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/immunology , Reproducibility of Results , Signal Transduction , Stellate Ganglion/metabolism
16.
Curr Opin Allergy Clin Immunol ; 20(4): 352-361, 2020 08.
Article in English | MEDLINE | ID: mdl-32590503

ABSTRACT

PURPOSE OF REVIEW: Glycopeptide antibiotics such as vancomycin are frequently utilized to treat resistant Gram-positive infections such as methicillin-resistant Staphylococcus aureus. The current literature on glycopeptide and lipoglycopeptide structure, hypersensitivity and potential cross-reactivity was reviewed, highlighting implications for safe prescribing. RECENT FINDINGS: Structurally similar, glycopeptides could theoretically cross-react. Immediate reactions to vancomycin include non-IgE-mediated reactions (e.g. red man syndrome) and IgE-mediated hypersensitivity (e.g. anaphylaxis), sharing clinical features. Vancomycin can activate mast cells via MAS-related G-protein-coupled receptor X2, an IgE-independent receptor implicated in non-IgE reactions. In-vivo and in-vitro testing for suspected IgE-mediated reactions to glycopeptides remain ill-defined. Vancomycin is increasingly recognized to cause severe cutaneous adverse reactions (SCAR), with drug reaction with eosinophilia and systemic symptoms (DRESS) predominantly reported. Vancomycin DRESS has been associated with HLA-A32:-01, with a number needed to prevent of 1 in 74. Data demonstrating cross-reactivity amongst glycopeptides and lipoglycopeptides is limited to case reports/series. SUMMARY: Further studies and in-vivo/in-vitro diagnostics are required for better differentiation between IgE and non-IgE glycopeptide reactions. Despite its association with vanomycin DRESS, utility of pharmacogenomic screening for HLA-A32: 01 is ill-defined. Although HLA-A32:01 has been associated with vancomycin DRESS, its utility for pharmacogenomic screening is ill defined. Further clinical and immunological cross-reactivity data for glycopeptide/lipoglycopeptide antibiotics is required.


Subject(s)
Anaphylaxis/immunology , Anti-Bacterial Agents/adverse effects , Drug Hypersensitivity Syndrome/immunology , Staphylococcal Infections/drug therapy , Vancomycin/adverse effects , Anaphylaxis/diagnosis , Anaphylaxis/genetics , Anaphylaxis/prevention & control , Cross Reactions/genetics , Drug Hypersensitivity Syndrome/diagnosis , Drug Hypersensitivity Syndrome/genetics , Drug Hypersensitivity Syndrome/prevention & control , Drug Prescriptions/standards , HLA-A Antigens/genetics , HLA-A Antigens/immunology , Humans , Immunoglobulin E/immunology , Mast Cells/immunology , Methicillin-Resistant Staphylococcus aureus/drug effects , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Pharmacogenomic Testing , Pharmacogenomic Variants , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/immunology , Receptors, Neuropeptide/metabolism , Staphylococcal Infections/microbiology
17.
Proc Natl Acad Sci U S A ; 117(11): 6067-6074, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123064

ABSTRACT

Ocular inflammation is a major cause of visual impairment attributed to dysregulation of the immune system. Previously, we have shown that the receptor for growth-hormone-releasing hormone (GHRH-R) affects multiple inflammatory processes. To clarify the pathological roles of GHRH-R in acute ocular inflammation, we investigated the inflammatory cascades mediated by this receptor. In human ciliary epithelial cells, the NF-κB subunit p65 was phosphorylated in response to stimulation with lipopolysaccharide (LPS), resulting in transcriptional up-regulation of GHRH-R. Bioinformatics analysis and coimmunoprecipitation showed that GHRH-R had a direct interaction with JAK2. JAK2, but not JAK1, JAK3, and TYK2, was elevated in ciliary body and iris after treatment with LPS in a rat model of endotoxin-induced uveitis. This elevation augmented the phosphorylation of STAT3 and production of proinflammatory factors, including IL-6, IL-17A, COX2, and iNOS. In explants of iris and ciliary body, the GHRH-R antagonist, MIA-602, suppressed phosphorylation of STAT3 and attenuated expression of downstream proinflammatory factors after LPS treatment. A similar suppression of STAT3 phosphorylation was observed in human ciliary epithelial cells. In vivo studies showed that blocking of the GHRH-R/JAK2/STAT3 axis with the JAK inhibitor Ruxolitinib alleviated partially the LPS-induced acute ocular inflammation by reducing inflammatory cells and protein leakage in the aqueous humor and by repressing expression of STAT3 target genes in rat ciliary body and iris and in human ciliary epithelial cells. Our findings indicate a functional role of the GHRH-R/JAK2/STAT3-signaling axis in acute anterior uveitis and suggest a therapeutic strategy based on treatment with antagonists targeting this signaling pathway.


Subject(s)
Epithelial Cells/pathology , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Signal Transduction/immunology , Uveitis/pathology , Animals , Cell Line , Ciliary Body/cytology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Humans , Janus Kinase 2/metabolism , Lipopolysaccharides/immunology , Male , Nitriles , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines , Rats , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/immunology , Receptors, Pituitary Hormone-Regulating Hormone/antagonists & inhibitors , Receptors, Pituitary Hormone-Regulating Hormone/immunology , STAT3 Transcription Factor/metabolism , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Sermorelin/therapeutic use , Signal Transduction/drug effects , Uveitis/drug therapy , Uveitis/immunology
18.
Immunity ; 52(2): 404-416.e5, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32049054

ABSTRACT

Mast cells are rare tissue-resident cells of importance to human allergies. To understand the structural basis of principle mast cell functions, we analyzed the proteome of primary human and mouse mast cells by quantitative mass spectrometry. We identified a mast-cell-specific proteome signature, indicative of a unique lineage, only distantly related to other immune cell types, including innate immune cells. Proteome comparison between human and mouse suggested evolutionary conservation of core mast cell functions. In addition to specific proteases and proteins associated with degranulation and proteoglycan biosynthesis, mast cells expressed proteins potentially involved in interactions with neurons and neurotransmitter metabolism, including cell adhesion molecules, ion channels, and G protein coupled receptors. Toward targeted cell ablation in severe allergic diseases, we used MRGPRX2 for mast cell depletion in human skin biopsies. These proteome analyses suggest a unique role of mast cells in the immune system, probably intertwined with the nervous system.


Subject(s)
Mast Cells/cytology , Mast Cells/immunology , Animals , Biomarkers/metabolism , Cell Degranulation , Cell Lineage , Cells, Cultured , Connective Tissue/immunology , Humans , Immunotherapy , Mast Cells/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Neuroimmunomodulation , Proteoglycans/biosynthesis , Proteome , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/immunology , Receptors, Neuropeptide/metabolism , Skin/immunology
19.
Phytomedicine ; 68: 153149, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32032836

ABSTRACT

BACKGROUND: Mast cells (MCs) are crucial effectors in allergic disorders by secreting inflammatory mediators. The Mas-related G-protein-coupled receptor X2 (Mrgprx2) was shown to have a key role in IgE-independent allergic reactions. Therefore, potential drug candidates that directly target Mrgprx2 could be used to treat pseudo-allergic diseases. Shikonin, an active ingredient derived from Lithospermum erythrorhizon Sieb. et Zucc has been used for its anti-inflammatory properties since ancient China. PURPOSE: To investigate the inhibitory effects of Shikonin on IgE-independent allergy both in vitro and in vivo, as well as the mechanism underlying its effects. METHODS/STUDY DESIGNS: The anti-anaphylactoid activity of Shikonin was evaluated in PCA and systemic anaphylaxis models, Calcium imaging was used to assess intracellular Ca2+ mobilization. The release of cytokines and chemokines was measured using enzyme immunoassay kits. Western blot analysis was conducted to investigate the molecules of PLCγ-PKC-IP3 signaling pathway. The analytical method of surface plasmon resonance was employed to study the interaction between Shikonin and potential target protein Mrgprx2. RESULTS: Shikonin can suppress compound 48/80 (C48/80)-induced PCA, active systemic anaphylaxis, and MCs degranulation in mice in a dose-dependent manner. In addition, Shikonin reduced C48/80-induced calcium flux and suppressed LAD2 cell degranulation via PLCγ-PKC-IP3 signaling pathway. Moreover, Shikonin was found to inhibit C48/80-induced Mrgprx2 expression in HEK cells, displaying specific interactions with the Mrgprx2 protein. CONCLUSION: Shikonin could be a potential antagonist of Mrgprx2, thereby inhibiting pseudo-allergic reactions through Ca2+ mobilization.


Subject(s)
Anaphylaxis/drug therapy , Hypersensitivity/drug therapy , Naphthoquinones/pharmacology , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Anaphylaxis/chemically induced , Animals , Calcium/metabolism , Cell Degranulation/drug effects , Cell Line , Chemokines/metabolism , Cytokines/metabolism , Humans , Hypersensitivity/immunology , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice, Inbred C57BL , Naphthoquinones/chemistry , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Phospholipase C gamma/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/metabolism , Secretagogues/toxicity , p-Methoxy-N-methylphenethylamine/toxicity
20.
J Leukoc Biol ; 107(5): 797-807, 2020 05.
Article in English | MEDLINE | ID: mdl-31922289

ABSTRACT

Recently a G-protein-coupled receptor, MAS Related GPR Family Member X2 (MRGPRX2), was identified as a specific receptor on human mast cells responsible for IgE independent adverse drug reactions (ADR). Although a murine homologue, Mrgprb2, has been identified for this receptor, its affinity for many ADR-causing drugs is poor making it difficult to undertake in vivo studies to examine mechanisms of ADR and to develop therapeutic strategies. Here, we have created humanized mice capable of generating MRGPRX2-expressing human MCs allowing for the study of MRGPRX2 MCs-mediated ADR in vitro as well as in vivo. Humanized mice were generated by hydrodynamic-injection of plasmids expressing human GM-CSF and IL-3 into NOD-scid IL2R-γ-/- strain of mice that had been transplanted with human hematopoietic stem cells. These GM/IL-3 humice expressed high numbers of tissue human MCs but the MRGPRX2 receptor expressed in MCs were limited to few body sites including the skin. Importantly, large numbers of MRGPRX2-expressing human MCs could be cultured from the bone marrow of GM/IL-3 humice revealing these mice to be an important source of human MCs for in vitro studies of MRGPRX2-related MCs activities. When GM/IL-3 humice were exposed to known ADR causing contrast agents (meglumine and gadobutrol), the humice were found to experience anaphylaxis analogous to the clinical situation. Thus, GM/IL-3 humice represent a valuable model for investigating in vivo interactions of ADR-causing drugs and human MCs and their sequelae, and these mice are also a source of human MRGPRX2-expressing MCs for in vitro studies.


Subject(s)
Disease Models, Animal , Drug Eruptions/immunology , Mast Cells/immunology , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Animals , Contrast Media/toxicity , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interleukin-3/genetics , Mast Cells/drug effects , Meglumine/toxicity , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Organometallic Compounds/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...