Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(2): e1012045, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38416790

ABSTRACT

Protein ubiquitination is essential for cellular homeostasis and regulation of several processes, including cell division and genome integrity. Ubiquitin E3 ligases determine substrate specificity for ubiquitination, and Cullin-RING E3 ubiquitin ligases (CRLs) make the largest group among the ubiquitin E3 ligases. Although conserved and most studied in model eukaryotes, CRLs remain underappreciated in Plasmodium and related parasites. To investigate the CRLs of human malaria parasite Plasmodium falciparum, we generated parasites expressing tagged P. falciparum cullin-1 (PfCullin-1), cullin-2 (PfCullin-2), Rbx1 (PfRbx1) and Skp1 (PfSkp1). PfCullin-1 and PfCullin-2 were predominantly expressed in erythrocytic trophozoite and schizont stages, with nucleocytoplasmic localization and chromatin association, suggesting their roles in different cellular compartments and DNA-associated processes. Immunoprecipitation, in vitro protein-protein interaction, and ubiquitination assay confirmed the presence of a functional Skp1-Cullin-1-Fbox (PfSCF) complex, comprising of PfCullin-1, PfRbx1, PfSkp1, PfFBXO1, and calcyclin binding protein. Immunoprecipitation, sequence analysis, and ubiquitination assay indicated that PfCullin-2 forms a functional human CRL4-like complex (PfCRL4), consisting of PfRbx1, cleavage and polyadenylation specificity factor subunit_A and WD40 repeat proteins. PfCullin-2 knock-down at the protein level, which would hinder PfCRL4 assembly, significantly decreased asexual and sexual erythrocytic stage development. The protein levels of several pathways, including protein translation and folding, lipid biosynthesis and transport, DNA replication, and protein degradation were significantly altered upon PfCullin-2 depletion, which likely reflects association of PfCRL4 with multiple pathways. PfCullin-2-depleted schizonts had poorly delimited merozoites and internal membraned structures, suggesting a role of PfCRL4 in maintaining membrane integrity. PfCullin-2-depleted parasites had a significantly lower number of nuclei/parasite than the normal parasites, indicating a crucial role of PfCRL4 in cell division. We demonstrate the presence of functional CRLs in P. falciparum, with crucial roles for PfCRL4 in cell division and maintaining membrane integrity.


Subject(s)
Plasmodium falciparum , Ubiquitin-Protein Ligases , Humans , Cell Division , Cullin Proteins/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
2.
Cells ; 12(22)2023 11 14.
Article in English | MEDLINE | ID: mdl-37998359

ABSTRACT

The skin presents a multifaceted microbiome, a balanced coexistence of bacteria, fungi, and viruses. These resident microorganisms are fundamental in upholding skin health by both countering detrimental pathogens and working in tandem with the skin's immunity. Disruptions in this balance, known as dysbiosis, can lead to disorders like psoriasis and atopic dermatitis. Central to the skin's defense system are mast cells. These are strategically positioned within the skin layers, primed for rapid response to any potential foreign threats. Recent investigations have started to unravel the complex interplay between these mast cells and the diverse entities within the skin's microbiome. This relationship, especially during times of both balance and imbalance, is proving to be more integral to skin health than previously recognized. In this review, we illuminate the latest findings on the ties between mast cells and commensal skin microorganisms, shedding light on their combined effects on skin health and maladies.


Subject(s)
Dermatitis, Atopic , Microbiota , Psoriasis , Humans , Mast Cells , Skin , Psoriasis/microbiology
3.
Front Allergy ; 4: 1109717, 2023.
Article in English | MEDLINE | ID: mdl-36970068

ABSTRACT

Sodium butyrate (NaBu) is a class I histone deacetylase inhibitor (HDACi) that can impede the proliferation of transformed cells. Although some HDACi downregulate the expression of the stem cell factor receptor (KIT/CD117), the effect of NaBu on KIT expression and human mast cell proliferation requires further elucidation. In this study, we examined the effects of NaBu on three transformed human mast cell lines, HMC-1.1, HMC-1.2 and LAD2. NaBu (100 µM) inhibited the proliferation and metabolic activity of all three cell lines without significantly affecting their viability, suggesting that although the cells had ceased to divide, they were not yet undergoing apoptosis. Cell cycle analysis using the cell-permeant dye, propidium iodide, indicated that NaBu significantly blocked the cell cycle progression of HMC-1.1 and HMC-1.2 from G1 to G2/M phases. Furthermore, NaBu downregulated the expression of C-KIT mRNA and KIT protein expression in all three cell lines, but this effect was most significant in the HMC-1.1 and HMC-1.2, both of which harbour activating mutations in KIT, which proliferate more rapidly than LAD2. These data support earlier observations showing that human mast cell lines are sensitive to histone deacetylase inhibition. However, our data presents the novel observation that inhibition of cell proliferation by NaBu was not associated with a loss in cell viability but rather an arrest of the cell cycle. Higher concentrations of NaBu led to modest increases in histamine content, tryptase expression, and granularity. In conclusion, NaBu treatment of human mast cell lines led to a modest enhancement of the hallmarks of mature mast cells.

4.
Immunity ; 55(9): 1645-1662.e7, 2022 09 13.
Article in English | MEDLINE | ID: mdl-35882236

ABSTRACT

Healthy skin maintains a diverse microbiome and a potent immune system to fight off infections. Here, we discovered that the epithelial-cell-derived antimicrobial peptides defensins activated orphan G-protein-coupled receptors (GPCRs) Mrgpra2a/b on neutrophils. This signaling axis was required for effective neutrophil-mediated skin immunity and microbiome homeostasis. We generated mutant mouse lines lacking the entire Defensin (Def) gene cluster in keratinocytes or Mrgpra2a/b. Def and Mrgpra2 mutant animals both exhibited skin dysbiosis, with reduced microbial diversity and expansion of Staphylococcus species. Defensins and Mrgpra2 were critical for combating S. aureus infections and the formation of neutrophil abscesses, a hallmark of antibacterial immunity. Activation of Mrgpra2 by defensin triggered neutrophil release of IL-1ß and CXCL2 which are vital for proper amplification and propagation of the antibacterial immune response. This study demonstrated the importance of epithelial-neutrophil signaling via the defensin-Mrgpra2 axis in maintaining healthy skin ecology and promoting antibacterial host defense.


Subject(s)
Bacterial Infections , Neutrophils , Receptors, G-Protein-Coupled , Animals , Mice , Anti-Bacterial Agents , Carrier Proteins , Defensins/genetics , Dysbiosis , Keratinocytes , Receptors, G-Protein-Coupled/metabolism , Staphylococcus aureus
5.
J Immunol ; 203(7): 1701-1714, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31484729

ABSTRACT

Intrathecal morphine infusion is often applied to treat chronic pain related to cancer and other conditions. However, persistent pain can be caused by nerve compression because of granuloma formation. In this study, a mouse model of morphine-induced granuloma formation by intrathecal catheterization morphine infusion into the atlanto-occipital membrane of the foramen magnum was established in wild-type mice, MrgprB2 mutant (MrgprB2-/-) mice, and in mast cell-deficient W-sash c-kit mutant (KitW-sh/W-sh) mice. Heat-related pain after surgery was performed to investigate the antipain effect of morphine. H&E staining and immunofluorescence staining of the spinal cord were applied to analyze the mechanism of granuloma formation. Morphine-induced mast cell degranulation was assessed by measuring the Ca2+ influx and mediator release. Anaphylactoid reactions were measured after s.c. morphine infusion to the paws. Chemokine release by mast cells was determined by Human XL Cytokine Array. Experiments with wild-type, MrgprB2 mutant, and mast cell-deficient W-sash c-kit mutant mice demonstrated that morphine activated mast cells and inflammatory cell aggregation through MrgprB2 in intrathecal infusion sites. The chemokine production of human mast cells demonstrated that granuloma formation is correlated with chemokines release. In addition, morphine activated mouse primary mast cells and de novo chemokine synthesis via the MRGPRX2 in human LAD2 cells. We concluded that granuloma formation during intrathecal morphine infusion was associated with MrgprB2/X2. Reducing MRGPRX2 potentially blocks morphine-induced side effects, including granuloma formation.


Subject(s)
Granuloma/immunology , Mast Cells/immunology , Morphine/adverse effects , Pain/immunology , Receptors, G-Protein-Coupled/immunology , Spinal Cord/immunology , Animals , Chemokines/genetics , Chemokines/immunology , Foramen Magnum/immunology , Foramen Magnum/pathology , Granuloma/pathology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Injections, Spinal , Male , Mast Cells/pathology , Mice , Mice, Knockout , Morphine/pharmacology , Pain/drug therapy , Pain/pathology , Receptors, G-Protein-Coupled/genetics , Spinal Cord/pathology
6.
Cell Host Microbe ; 26(1): 114-122.e8, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31278040

ABSTRACT

Quorum-sensing molecules (QSMs) are secreted by bacteria to signal population density. Upon reaching a critical concentration, QSMs induce transcriptional alterations in bacteria, which enable virulence factor expression and biofilm formation. It is unclear whether mammalian hosts can recognize QSMs to trigger responsive antibacterial immunity. We report that mouse mast-cell-specific G-protein-coupled receptor Mrgprb2 and its human homolog MRGPRX2 are receptors for Gram-positive QSMs, including competence-stimulating peptide (CSP)-1. CSP-1 activates Mrgprb2 and MRGPRX2, triggering mast cell degranulation, which inhibits bacterial growth and prevents biofilm formation. Such antibacterial functions are reduced in Mrgprb2-deficient mast cells, while wild-type mast cells fail to inhibit the growth of bacterial strains lacking CSP-1. Mrgprb2-knockout mice exhibit reduced bacterial clearance, while pharmacologically activating Mrgprb2 in vivo eliminates bacteria and improves disease score. These findings identify a host defense mechanism that uses QSMs as an "Achilles heel" and suggest MRGPRX2 as a potential therapeutic target for controlling bacterial infections.


Subject(s)
Bacterial Proteins/metabolism , Connective Tissue/immunology , Immunity, Innate , Mast Cells/immunology , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Streptococcus pneumoniae/immunology , Animals , Bacteriocins/metabolism , Enterococcus faecium/immunology , Humans , Mice , Mice, Knockout , Streptococcus pyogenes/immunology
7.
Neuron ; 101(3): 412-420.e3, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30686732

ABSTRACT

Mast cells can be found in close proximity to peripheral nerve endings where, upon activation, they release a broad range of pro-inflammatory cytokines and chemokines. However, the precise mechanism underlying this so-called neurogenic inflammation and associated pain has remained elusive. Here we report that the mast-cell-specific receptor Mrgprb2 mediates inflammatory mechanical and thermal hyperalgesia and is required for recruitment of innate immune cells at the injury site. We also found that the neuropeptide substance P (SP), an endogenous agonist of Mrgprb2, facilitates immune cells' migration via Mrgprb2. Furthermore, SP activation of the human mast cell led to the release of multiple pro-inflammatory cytokines and chemokines via the human homolog MRGPRX2. Surprisingly, the SP-mediated inflammatory responses were independent of its canonical receptor, neurokinin-1 receptor (NK-1R). These results identify Mrgprb2/X2 as an important neuroimmune modulator and a potential target for treating inflammatory pain.


Subject(s)
Cytokines/metabolism , Mast Cells/metabolism , Neuralgia/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Female , Humans , Male , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/genetics , Receptors, Neurokinin-1/metabolism , Substance P/metabolism
8.
Am J Physiol Endocrinol Metab ; 315(3): E357-E366, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29812984

ABSTRACT

Intraperitoneal administration of the melanocortin agonist melanotan II (MTII) to mice causes a profound, transient hypometabolism/hypothermia. It is preserved in mice lacking any one of melanocortin receptors 1, 3, 4, or 5, suggesting a mechanism independent of the canonical melanocortin receptors. Here we show that MTII-induced hypothermia was abolished in KitW-sh/W-sh mice, which lack mast cells, demonstrating that mast cells are required. MRGPRB2 is a receptor that detects many cationic molecules and activates mast cells in an antigen-independent manner. In vitro, MTII stimulated mast cells by both MRGPRB2-dependent and -independent mechanisms, and MTII-induced hypothermia was intact in MRGPRB2-null mice. Confirming that MTII activated mast cells, MTII treatment increased plasma histamine levels in both wild-type and MRGPRB2-null, but not in KitW-sh/W-sh, mice. The released histamine produced hypothermia via histamine H1 receptors because either a selective antagonist, pyrilamine, or ablation of H1 receptors greatly diminished the hypothermia. Other drugs, including compound 48/80, a commonly used mast cell activator, also produced hypothermia by both mast cell-dependent and -independent mechanisms. These results suggest that mast cell activation should be considered when investigating the mechanism of drug-induced hypothermia in mice.


Subject(s)
Histamine Agonists/pharmacology , Hypothermia/chemically induced , Mast Cells/drug effects , Peptides, Cyclic/pharmacology , alpha-MSH/analogs & derivatives , Animals , Histamine Release/drug effects , Histamine Release/genetics , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , alpha-MSH/pharmacology
9.
Biochem Pharmacol ; 146: 214-223, 2017 12 15.
Article in English | MEDLINE | ID: mdl-28987593

ABSTRACT

Mast cells are unique immunocytes that function as sentinel cells in host defense reactions such as immediate hypersensitivity responses and anaphylactic responses. The mast cell specific receptor MRGPRX2 (Mas-related G protein-coupled receptor X2) triggers mast cell degranulation-a key process in anaphylactic reactions. We sought to better understand anaphylactic reaction induced by sinomenine hydrochloride (SH). MRGPRX2-related pseudo-allergic reactions induced by SH were investigated using the hindpaw swelling and extravasation assay in vivo and mast cell degranulation assays in vitro. MrgprB2 knockout mice exhibit a reduced SH-induced inflammation effect. Furthermore, MRGPRX2 (the orthologous gene of MrgprB2) related human mast cells are activated by SH in a dose-dependent manner; however, MRGPRX2 knockdown mast cells showed reduced degranulation. The results showed a kind of mechanism that SH-induced anaphylactoid reactions were mediated by MRGPRX2 via activating PLC molecular signaling pathways to provoke mast cells Ca2+ mobilization and degranulation.


Subject(s)
Anaphylaxis/drug therapy , Mast Cells/metabolism , Morphinans/pharmacology , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Animals , Antirheumatic Agents/pharmacology , Calcium Signaling , Cell Line , Cells, Cultured , Gene Expression Regulation/physiology , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Peritoneum/cytology , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics
10.
Eur J Immunol ; 47(11): 1949-1958, 2017 11.
Article in English | MEDLINE | ID: mdl-28688196

ABSTRACT

Mast cells are unique immune cells that function as sentinels in host defence reactions, including immediate hypersensitivity responses and allergic responses. The mast cell-specific receptor named MAS-related G protein-coupled receptor X2 (MRGPRX2) triggers mast-cell degranulation, a key process in anaphylactoid reactions. It is widely observed that antimicrobials can induce pseudo-allergic reactions (i.e. IgE-independent mechanism) with symptoms ranging from skin inflammation to life-threatening systemic anaphylaxis. However, their direct involvement and the mechanisms underlying anaphylactoid reactions caused by antimicrobials have not been demonstrated. Structurally different antimicrobials were screened by Ca2+ imaging using MRGPRX2 overexpressing HEK293 cells. MRGPRX2 related anaphylactoid reactions induced by these components were investigated by body temperature drop and mast cell degranulation assays. We showed that MRGPRX2 is involved in allergic-like reactions to three types of antimicrobials in a dose-dependent manner. However, mast cells lacking the receptor show reduced degranulation. Furthermore, mice without MAS-related G protein-coupled receptor B2 (the orthologous gene of MRGPRX2) exhibited reduced substance-induced inflammation. Interestingly, ß-lactam and antiviral nucleoside analogues did not induce anaphylactic reactions, which were also observed in vitro. These results should alarm many clinicians that such drugs might induce anaphylactoid reactions and provide guidance on safe dosage of these drugs.


Subject(s)
Anaphylaxis/chemically induced , Anti-Infective Agents/toxicity , Cell Degranulation/drug effects , Drug Hypersensitivity/immunology , Mast Cells/drug effects , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Animals , Anti-Infective Agents/immunology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
J Immunol ; 195(6): 2774-87, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26283482

ABSTRACT

C5a generated during complement activation possesses proinflammatory and immunoregulatory properties critical for the development and modulation of allergic immune responses. In immune cells, C5a mediates its effects through binding to two G protein-coupled receptors, C5aR1 and C5aR2. Mast cells are key effectors in allergic reactions, and decades of research have suggested that the majority of C5a effects on mast cells are mediated through C5aR1, whereas the expression and function of C5aR2 have not been explored. We demonstrated that the human mast cell line Laboratory of Allergic Diseases 2 (LAD2) expresses surface C5aR2 but not C5aR1, whereas CD34(+) cell-derived primary mast cells do not express surface C5aR1 or C5aR2. Stem cell factor and IL-4 upregulated C5aR2 expression on LAD2 cells. Furthermore, C5a caused internalization of LAD2 cell-surface C5aR2. We therefore used LAD2 cells as a model to study C5a/C5aR2-induced biological responses and signaling in human mast cells. We found that whereas C5a was unable to induce degranulation, it stimulated GM-CSF, TNF, CXCL10, and CCL2 production. C5a caused ERK phosphorylation, a signaling molecule important in cytokine and chemokine generation. In addition, C5a stimulated adhesion and chemotaxis of mast cells. Wortmannin, an inhibitor of PI3K, and small interfering RNA against ß-arrestin-2 blocked C5a-induced adhesion. Silencing of C5aR2 using lentiviral short hairpin RNA rendered the cells unresponsive to C5a-induced adhesion, chemotaxis, and mediator release, as well as ERK phosphorylation. Overall, this study reveals a novel role for C5aR2 in C5a-mediated activation of mast cells and demonstrates that C5aR2 ligation initiates a ß-arrestin-2-, PI3K-, and ERK-dependent signaling pathway in these cells.


Subject(s)
Cell Movement/immunology , Complement C5a/immunology , Inflammation/immunology , Mast Cells/immunology , Receptors, Chemokine/metabolism , Androstadienes/pharmacology , Antigens, CD34/metabolism , Arrestins/genetics , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Degranulation/immunology , Cell Line , Chemokine CCL2/biosynthesis , Chemokine CXCL10/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Interferon-gamma/metabolism , Interleukin-4/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Chemokine/genetics , Stem Cell Factor/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Wortmannin , beta-Arrestin 2 , beta-Arrestins
12.
Nature ; 519(7542): 237-41, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25517090

ABSTRACT

Mast cells are primary effectors in allergic reactions, and may have important roles in disease by secreting histamine and various inflammatory and immunomodulatory substances. Although they are classically activated by immunoglobulin (Ig)E antibodies, a unique property of mast cells is their antibody-independent responsiveness to a range of cationic substances, collectively called basic secretagogues, including inflammatory peptides and drugs associated with allergic-type reactions. The pathogenic roles of these substances have prompted a decades-long search for their receptor(s). Here we report that basic secretagogues activate mouse mast cells in vitro and in vivo through a single receptor, Mrgprb2, the orthologue of the human G-protein-coupled receptor MRGPRX2. Secretagogue-induced histamine release, inflammation and airway contraction are abolished in Mrgprb2-null mutant mice. Furthermore, we show that most classes of US Food and Drug Administration (FDA)-approved peptidergic drugs associated with allergic-type injection-site reactions also activate Mrgprb2 and MRGPRX2, and that injection-site inflammation is absent in mutant mice. Finally, we determine that Mrgprb2 and MRGPRX2 are targets of many small-molecule drugs associated with systemic pseudo-allergic, or anaphylactoid, reactions; we show that drug-induced symptoms of anaphylactoid responses are significantly reduced in knockout mice; and we identify a common chemical motif in several of these molecules that may help predict side effects of other compounds. These discoveries introduce a mouse model to study mast cell activation by basic secretagogues and identify MRGPRX2 as a potential therapeutic target to reduce a subset of drug-induced adverse effects.


Subject(s)
Drug Hypersensitivity/immunology , Mast Cells/immunology , Mast Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Disease Models, Animal , Drug Hypersensitivity/genetics , Drug Hypersensitivity/prevention & control , Female , HEK293 Cells , Histamine Release , Humans , Inflammation/immunology , Inflammation/metabolism , Male , Mast Cells/drug effects , Mice , Mice, Knockout , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/metabolism
13.
Immunol Cell Biol ; 88(6): 632-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20309008

ABSTRACT

Antimicrobial peptides (AMPs) are ancient and essential elements of the host defense system, which are found in a wide variety of species. They show antimicrobial activity against a wide range of pathogenic microorganisms. In addition, AMPs are expressed by different immune cells and have a important function in host innate immune response against pathogens by mechanisms that are different from those involved in direct microbial cytolysis. One host innate immune response that is directly activated by AMPs involves induction of localized inflammation through interaction with mast cells. Activation of mast cells releases pre-formed mediators, cytokines, chemokines and eicosaniods, which influence recruitment, survival, phenotype and functions of many immune cells. Mast cells can respond to AMPs independent of antigen and Fc epsilon receptor 1 stimulation. One of these pathways involves G protein-coupled receptor signaling, which can lead to mast cell degranulation. Whether AMPs activate G proteins in mast cells through a receptor-dependent or a receptor-independent mechanism remains poorly understood and there are a great many questions that have yet to be answered. In this review, we will discuss the possible involvement and role of GPCRs in mast cells activation by AMPs and the gaps in our current understanding of this important interaction.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Mast Cells/immunology , Receptors, G-Protein-Coupled/immunology , Animals , Host-Pathogen Interactions , Humans , Immunity, Innate , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...