Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.172
Filter
1.
J Sep Sci ; 47(11): e2300924, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38819784

ABSTRACT

Mas-related G protein-coupled receptor X2 (MrgprX2) is acknowledged as a mast cell-specific receptor, playing a crucial role in orchestrating anaphylactoid responses through mast cell degranulation. It holds promise as a target for regulating allergic and inflammatory diseases mediated by mast cells. Polygonum cuspidatum (PC) has shown notable anti-anaphylactoid effects, while its pharmacologically active components remain unclear. In this study, we successfully utilized MrgprX2 high-expressing cell membrane chromatography (CMC), in conjunction with liquid chromatography-mass spectrometry (LC-MS), to identify active anti-anaphylactoid components in PC. Our study pinpointed polydatin, resveratrol, and emodin-8-O-ß-d-glucoside as potential anti-anaphylactoid compounds in PC. Their anti-anaphylactoid activities were evaluated through ß-aminohexosidase and histamine release assays, demonstrating a concentration-dependent inhibition for both ß-aminohexosidase and histamine release. This approach, integrating MrgprX2 high-expression CMC with LC-MS, proves effective in screening potential anti-anaphylactoid ingredients in natural herbal medicines. The findings from this study illuminated the anti-anaphylactoid properties of specific components in PC and provided an efficient method for the drug development of natural products.


Subject(s)
Fallopia japonica , Receptors, G-Protein-Coupled , Receptors, Neuropeptide , Receptors, G-Protein-Coupled/metabolism , Fallopia japonica/chemistry , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/antagonists & inhibitors , Humans , Mass Spectrometry , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/chemistry , Chromatography, Liquid , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Mast Cells/drug effects , Mast Cells/metabolism , Plant Extracts/pharmacology , Plant Extracts/chemistry , Glucosides/pharmacology , Glucosides/chemistry , Glucosides/analysis , Molecular Structure , Liquid Chromatography-Mass Spectrometry
2.
Front Immunol ; 15: 1406438, 2024.
Article in English | MEDLINE | ID: mdl-38817611

ABSTRACT

Introduction: Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterised by itching, erythema, and epidermal barrier dysfunction. The pathogenesis of AD is complex and multifactorial; however,mast cell (MC) activation has been reported to be one of the crucial mechanisms in the pathogenesis of AD. The MC receptor Mas related G protein-coupled receptor-X2 (MRGPRX2) has been identified as a prominent alternative receptor to the IgE receptor in causing MC activation and the subsequent release of inflammatory mediators. The current study aimed to evaluate the therapeutic effect of a novel small molecule MRGPRX2 antagonist GE1111 in AD using in vitro and in vivo approaches. Methods: We developed an in vitro cell culture disease model by using LAD-2 MC, HaCaT keratinocytes and RAW 264.7 macrophage cell lines. We challenged keratinocytes and macrophage cells with CST-14 treated MC supernatant in the presence and absence of GE1111 and measured the expression of tight junction protein claudin 1, inflammatory cytokines and macrophage phagocytosis activity through immunohistochemistry, western blotting, RT-qPCR and fluorescence imaging techniques. In addition to this, we developed a DFNB-induced AD model in mice and evaluated the protective effect and underlying mechanism of GE1111. Results and Discussion: Our in vitro findings demonstrated a potential therapeutic effect of GE1111, which inhibits the expression of TSLP, IL-13, MCP-1, TNF-a, and IL-1ß in MC and keratinocytes. In addition to this, GE1111 was able to preserve the expression of claudin 1 in keratinocytes and the phagocytotic activity of macrophage cells. The in vivo results demonstrated that GE1111 treatment significantly reduced phenotypic changes associated with AD (skin thickening, scaling, erythema and epidermal thickness). Furthermore, immunohistochemical analysis demonstrated that GE1111 treatment preserved the expression of the tight junction protein Involucrin and reduced the expression of the inflammatory mediator periostin in the mouse model of AD. These findings were supported by gene and protein expression analysis, where GE1111 treatment reduced the expression of TSLP, IL-13, and IL-1ß, as well as downstream signalling pathways of MRGPRX2 in AD skin lesions. In conclusion, our findings provide compelling in vitro and in vivo evidence supporting the contribution of MRGPRX2-MC interaction with keratinocytes and macrophages in the pathogenesis of AD.


Subject(s)
Cytokines , Dermatitis, Atopic , Disease Models, Animal , Keratinocytes , Receptors, G-Protein-Coupled , Receptors, Neuropeptide , Skin , Animals , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Mice , Cytokines/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Humans , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/metabolism , Skin/pathology , Skin/drug effects , Skin/metabolism , Skin/immunology , Keratinocytes/drug effects , Keratinocytes/metabolism , HaCaT Cells , Macrophages/immunology , Macrophages/metabolism , Macrophages/drug effects , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , RAW 264.7 Cells , Inflammation Mediators/metabolism
3.
Int Immunopharmacol ; 134: 112256, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38744172

ABSTRACT

The incidence of allergic reactions has risen steadily in recent years, prompting growing interest in the identification of efficacious and safe natural compounds that can prevent or treat allergic diseases. Phellodendron amurense Rupr. has long been applied as a treatment for allergic diseases, whose primary component is phellodendrine. However, the efficacy of phellodendrine as a treatment for allergic diseases remains to be assessed. Mast cells are the primary effectors of allergic reactions, which are not only activated by IgE-dependent pathway, but also by IgE-independent pathways via human MRGPRX2, rat counterpart MRGPRB3. As such, this study explored the effect and mechanism of phellodendrine through this family receptors in treating allergic diseases in vitro and in vivo. These analyses revealed that phellodendrine administration was sufficient to protect against C48/80-induced foot swelling and Evans blue exudation in mice, and suppressed C48/80-induced RBL-2H3 rat basophilic leukemia cells degranulation, and ß-HEX, HIS, IL-4, and TNF-α release. Moreover, phellodendrine could reduce the mRNA expression of MRGPRB3 and responsiveness of MRGPRX2 by altering its structure. It was able to decrease Ca2+ levels, phosphorylation levels of CaMK, PLCß1, PKC, ERK, JNK, p38, and p65, and inhibit the degradation of IκB-α. These analyses indicate that berberine inhibits the activation of PLC and downregulates the release of Ca2+ in the endoplasmic reticulum by altering the conformation of MRGPRB3/MRGPRX2 protein, thereby inhibiting the activation of PKC and subsequently inhibiting downstream MAPK and NF-κB signaling, ultimately suppressing allergic reactions. There may thus be further value in studies focused on developing phellodendrine as a novel anti-allergic drug.


Subject(s)
Cell Degranulation , Hypersensitivity , Mast Cells , Receptors, G-Protein-Coupled , Animals , Rats , Mast Cells/drug effects , Mast Cells/immunology , Cell Degranulation/drug effects , Mice , Humans , Hypersensitivity/drug therapy , Hypersensitivity/immunology , Receptors, G-Protein-Coupled/metabolism , Anti-Allergic Agents/pharmacology , Anti-Allergic Agents/therapeutic use , Cytokines/metabolism , p-Methoxy-N-methylphenethylamine , Male , Phellodendron/chemistry , Cell Line, Tumor , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Mice, Inbred BALB C , NF-kappa B/metabolism , Signal Transduction/drug effects , Receptors, Neuropeptide
4.
Int J Biol Macromol ; 269(Pt 2): 132128, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723807

ABSTRACT

Selenium-rich tea polysaccharides (Se-TPS) were extracted via high hydrostatic pressure technology with a pressure of 400 MPa (200-500 MPa) for 10 min (3-20 min) at a material-to-solvent ratio of 1:40 (1:20-1:50). Subsequently, Se-TPS1-4 were isolated and purified, with Se-TPS3-4 as the main components. A spectral analysis proved that Se, which has antioxidant activity, existed. An in vitro study found that among Se-TPS, Se-TPS3-4 attenuated the release of ß-hexosaminidase, histamine, and interleukin (IL)-4. Furthermore, in vivo experiments revealed that treatment with Se-TPS downregulated IL-4 levels and upregulated TGF-ß and interferon-γ levels to improve imbalanced Th1/Th2 immunity in tropomyosin-sensitized mice. Moreover, Se-TPS promoted Lactobacillus and norank_f_Muribaculaceaek growth and upregulated metabolites such as genipin and coniferyl alcohol. Overall, these results showed the strong anti-allergy potential of Se-TPS by regulating mast cell-mediated allergic inflammatory responses and microbiota regulation, highlighting the potential of Se-TPS as a novel therapeutic agent to regulate allergy-associated metabolic disorders.


Subject(s)
Gastrointestinal Microbiome , Hydrostatic Pressure , Polysaccharides , Tea , Animals , Gastrointestinal Microbiome/drug effects , Polysaccharides/pharmacology , Polysaccharides/chemistry , Mice , Tea/chemistry , Mast Cells/metabolism , Mast Cells/drug effects , Mast Cells/immunology , Anti-Allergic Agents/pharmacology , Anti-Allergic Agents/chemistry , Anti-Allergic Agents/isolation & purification , beta-N-Acetylhexosaminidases/metabolism , Cytokines/metabolism , Male , Tropomyosin/metabolism , Tropomyosin/immunology
5.
Food Funct ; 15(11): 5895-5907, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38727519

ABSTRACT

In order to explore the in vivo anti-food allergy activity of Lactobacillus sakei subsp. sakei-fermented Eucheuma spinosum polysaccharides F1-ESP-3, an ovalbumin (OVA)-induced food allergy mouse model was established by ascites immunization and gavage. The weight, temperature, incidence of diarrhea, levels of allergic mediators and inflammatory factors in the serum of mice were analyzed. We analyzed the differentiation of mouse spleen lymphocytes and the proportion of sensitized mast cells by flow cytometry. The intestinal barrier status of mice was analyzed by intestinal pathological tissue sections and microbiota sequencing. The results showed that F1-ESP-3 could alleviate the food allergy symptoms of mice, such as hypothermia and loose stool; levels of OVA-specific immunoglobulin E, mast cell protease and histamine in the serum of sensitized mice and the proportion of dendritic cells and mast cells in mouse spleen were significantly reduced; in addition, F1-ESP-3 may protect the intestinal barrier and further improve the intestinal microenvironment of food-allergic mice by regulating the abundance of Bacteroidetes and Firmicutes. F1-ESP-3 can further improve the intestinal microenvironment of food-allergic mice by upregulating the levels of Lachnospiraceae, and may affect the signal pathways such as NOD-like receptor, MAPK, I kappa B and antigen processing and presentation.


Subject(s)
Food Hypersensitivity , Mice, Inbred BALB C , Polysaccharides , Animals , Mice , Food Hypersensitivity/drug therapy , Polysaccharides/pharmacology , Fermentation , Gastrointestinal Microbiome/drug effects , Female , Mast Cells/drug effects , Mast Cells/immunology , Disease Models, Animal , Immunoglobulin E/blood , Immunoglobulin E/immunology , Latilactobacillus sakei , Spleen/drug effects , Ovalbumin , Lactobacillus , Edible Seaweeds , Rhodophyta
6.
Int J Mol Sci ; 25(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38674081

ABSTRACT

Marrubiin is a diterpene with a long history of a wide range of biological activities. In this study, the anti-inflammatory effects of marrubiin were investigated using several in vitro and in vivo assays. Marrubiin inhibited carrageenan-induced peritoneal inflammation by preventing inflammatory cell infiltration and peritoneal mast cell degranulation. The anti-inflammatory activity was further demonstrated by monitoring a set of biochemical parameters, showing that the peritoneal fluid of animals treated with marrubiin had lower levels of proteins and lower myeloperoxidase activity compared with the fluid of animals that were not treated. Marrubiin exerted the most pronounced cytotoxic activity towards peripheral mononuclear cells, being the main contributors to peritoneal inflammation. Additionally, a moderate lipoxygenase inhibition activity of marrubiin was observed.


Subject(s)
Anti-Inflammatory Agents , Carrageenan , Diterpenes , Mast Cells , Animals , Carrageenan/adverse effects , Mice , Diterpenes/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Anti-Inflammatory Agents/pharmacology , Mice, Inbred C57BL , Peritonitis/chemically induced , Peritonitis/drug therapy , Peritonitis/metabolism , Peritonitis/pathology , Male , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/chemically induced , Inflammation/pathology , Cell Degranulation/drug effects , Peroxidase/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism
7.
Mol Immunol ; 170: 60-75, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38626622

ABSTRACT

Liver diseases caused by viral infections, alcoholism, drugs, or chemical poisons are a significant health problem: Liver diseases are a leading contributor to mortality, with approximately 2 million deaths per year worldwide. Liver fibrosis, as a common liver disease characterized by excessive collagen deposition, is associated with high morbidity and mortality, and there is no effective treatment. Numerous studies have shown that the accumulation of mast cells (MCs) in the liver is closely associated with liver injury caused by a variety of factors. This study investigated the relationship between MCs and carbon tetrachloride (CCl4)-induced liver fibrosis in rats and the effects of the MC stabilizers sodium cromoglycate (SGC) and ketotifen (KET) on CCl4-induced liver fibrosis. The results showed that MCs were recruited or activated during CCl4-induced liver fibrosis. Coadministration of SCG or KET alleviated the liver fibrosis by decreasing SCF/c-kit expression, inhibiting the TGF-ß1/Smad2/3 pathway, depressing the HIF-1a/VEGF pathway, activating Nrf2/HO-1 pathway, and increasing the hepatic levels of GSH, GSH-Px, and GR, thereby reducing hepatic oxidative stress. Collectively, recruitment or activation of MCs is linked to liver fibrosis and the stabilization of MCs may provide a new approach to the prevention of liver fibrosis.


Subject(s)
Carbon Tetrachloride , Cromolyn Sodium , Liver Cirrhosis , Liver , Mast Cells , Animals , Mast Cells/metabolism , Mast Cells/immunology , Mast Cells/drug effects , Carbon Tetrachloride/toxicity , Rats , Male , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/immunology , Liver Cirrhosis/chemically induced , Cromolyn Sodium/pharmacology , Liver/pathology , Liver/metabolism , Liver/drug effects , Transforming Growth Factor beta1/metabolism , Rats, Sprague-Dawley , Ketotifen/pharmacology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/immunology , Oxidative Stress/drug effects , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Vascular Endothelial Growth Factor A/metabolism
8.
Int Immunopharmacol ; 133: 112113, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38657498

ABSTRACT

BACKGROUND: Phytosphingosine and its derivative are known for their skin-protective properties. While mYG-II-6, a phytosphingosine derivative, has shown anti-inflammatory and antipsoriatic effects, its potential antipruritic qualities have yet to be explored. This study aimed to investigate mYG-II-6's antipruritic properties. METHODS: The calcium imaging technique was employed to investigate the activity of ion channels and receptors. Mast cell degranulation was confirmed through the ß-hexosaminidase assay. Additionally, in silico molecular docking and an in vivo mouse scratching behavior test were utilized. RESULTS: Using HEK293T cells transfected with H1R and TRPV1, we examined the impact of mYG-II-6 on histamine-induced intracellular calcium rise, a key signal in itch-mediating sensory neurons. Pretreatment with mYG-II-6 significantly reduced histamine-induced calcium levels and inhibited TRPV1 activity, suggesting its role in blocking the calcium influx channel. Additionally, mYG-II-6 suppressed histamine-induced calcium increase in primary cultures of mouse dorsal root ganglia, indicating its potential antipruritic effect mediated by histamine. Interestingly, mYG-II-6 exhibited inhibitory effects on human MRGPRX2, a G protein-coupled receptor involved in IgE-independent mast cell degranulation. However, it did not inhibit mouse MrgprB2, the ortholog of human MRGPRX2. Molecular docking analysis revealed that mYG-II-6 selectively interacts with the binding pocket of MRGPRX2. Importantly, mYG-II-6 suppressed histamine-induced scratching behaviors in mice. CONCLUSIONS: Our findings show that mYG-II-6 can alleviate histamine-induced itch sensation through dual mechanisms. This underscores its potential as a versatile treatment for various pruritic conditions.


Subject(s)
Cell Degranulation , Histamine , Mast Cells , Molecular Docking Simulation , Receptors, G-Protein-Coupled , TRPV Cation Channels , Animals , Mast Cells/drug effects , Mast Cells/immunology , Humans , TRPV Cation Channels/metabolism , Cell Degranulation/drug effects , HEK293 Cells , Histamine/metabolism , Receptors, G-Protein-Coupled/metabolism , Mice , Male , Pruritus/drug therapy , Calcium/metabolism , Antipruritics/pharmacology , Antipruritics/therapeutic use , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Neuropeptide/metabolism , Mice, Inbred C57BL
9.
Int Immunopharmacol ; 132: 111903, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38579561

ABSTRACT

Bruton's Tyrosine kinase (BTK) plays a pivotal role as the key mediator in B cell signaling. Recent research has revealed that it is also expressed in cells critical to asthma development, such as T cells, and eosinophils. This study aims to investigate the potential of BTK inhibitor in eosinophilic asthma mouse model. BALB/c mice were sensitized with ovalbumin (OVA) via intraperitoneal injections and followed by OVA nebulizations. The mice were treated with 250 µg/ml or 500 µg/ml of ibrutinib before the second intraperitoneal injection and the first nebulization. Two days after the last OVA challenge, airway hyperresponsiveness (AHR) was assessed with methacholine, and differential cell count in bronchoalveolar lavage fluid (BALF) was performed. The cytokines were measured in BALF, and serum OVA-specific IgE and IgG antibody levels were evaluated by ELISA. The inhibitory effect of ibrutinib was also evaluated in splenic mononuclear cells, mast cells, eosinophils, and T cells in vitro. Treatment with ibrutinib significantly attenuated AHR and airway inflammation, compared to the OVA-induced positive control. The treatment also reduced IL-4, IL-5, IL-13 and IFN-γ cytokine levels and suppressed OVA-specific IgE and IgG production compared to the OVA-induced positive control. Additionally, ibrutinib decreased beta-hexosaminidase release from mast cells, type 2 cytokine productions from mononuclear cells and T cells, and eosinophilic activation markers in vitro. The results of this study suggest that ibrutinib treatment could exert anti-allergic effects by inactivating B cells and other BTK-expressing cells. Further studies are needed to investigate the potential therapeutic effect of ibrutinib on allergic diseases.


Subject(s)
Adenine , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Asthma , Cytokines , Disease Models, Animal , Eosinophils , Immunoglobulin E , Mice, Inbred BALB C , Ovalbumin , Piperidines , Protein Kinase Inhibitors , Animals , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Asthma/drug therapy , Asthma/immunology , Piperidines/therapeutic use , Piperidines/pharmacology , Ovalbumin/immunology , Adenine/therapeutic use , Adenine/pharmacology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Cytokines/metabolism , Eosinophils/immunology , Eosinophils/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mice , Pyrimidines/therapeutic use , Pyrimidines/pharmacology , Female , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Immunoglobulin G/blood , Anti-Asthmatic Agents/therapeutic use , Anti-Asthmatic Agents/pharmacology , Cells, Cultured , Humans , Mast Cells/drug effects , Mast Cells/immunology
10.
J Ethnopharmacol ; 330: 118105, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38631485

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The traditional Chinese medicine (TCM) XYQFT is composed of 10 herbs. According to the NHIRD, XYQFT is one of the top ten most commonly used TCM prescriptions for asthma treatment. AIM OF THE STUDY: The aim of this study was to explore whether XYQFT reduces asthma symptoms in a mouse model of chronic asthma and determine the immunomodulatory mechanism of mast cells. MATERIALS AND METHODS: BALB/c mice were intratracheally (it) stimulated with 40 µL (2.5 µg/µL) of Dermatophagoides pteronyssinus (Der p) once a week for 6 consecutive weeks and orally administered XYQFT at 1 g/kg 30 min before Der p stimulation. Airway hypersensitivity, inflammatory cells in the BALF and total IgE in the blood were assessed in mice. In addition, RBL-2H3 cells (mast cells) were stimulated with DNP-IgE, after which different concentrations of XYQFT were added for 30 min to evaluate the effect of XYQFT on the gene expression and degranulation of DNP-stimulated RBL-2H3 cells. After the compounds in XYQFT were identified using LC‒MS/MS, the PBD method was used to identify the chemical components that inhibited the expression of the GM-CSF and COX-2 genes in mast cells. RESULTS: The airway hypersensitivity assay demonstrated that XYQFT significantly alleviated Der p-induced airway hypersensitivity. Moreover, cell counting and typing of bronchoalveolar lavage fluid revealed a significant reduction in Der p-induced inflammatory cell infiltration with XYQFT treatment. ELISA examination further indicated a significant decrease in Der p-induced total IgE levels in serum following XYQFT administration. In addition, XYQFT inhibited the degranulation and expression of genes (IL-3, IL-4, ALOX-5, IL-13, GM-CSF, COX-2, TNF-α, and MCP-1) in RBL-2H3 cells after DNP stimulation. The compounds timosaponin AIII and genkwanin in XYQFT were found to be key factors in the inhibition of COX-2 and GM-CSF gene expression in mast cells. CONCLUSION: By regulating mast cells, XYQFT inhibited inflammatory cell infiltration, airway hypersensitivity and specific immunity in a mouse model of asthma. In addition, XYQFT synergistically inhibited the expression of the GM-CSF and COX-2 genes in mast cells through timosaponin AIII and genkwanin.


Subject(s)
Asthma , Cyclooxygenase 2 , Drugs, Chinese Herbal , Granulocyte-Macrophage Colony-Stimulating Factor , Mast Cells , Animals , Male , Mice , Rats , Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunoglobulin E/blood , Mast Cells/drug effects , Mast Cells/metabolism , Mice, Inbred BALB C
11.
Cardiovasc Res ; 120(7): 681-698, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38630620

ABSTRACT

Mast cells are tissue-resident immune cells strategically located in different compartments of the normal human heart (the myocardium, pericardium, aortic valve, and close to nerves) as well as in atherosclerotic plaques. Cardiac mast cells produce a broad spectrum of vasoactive and proinflammatory mediators, which have potential roles in inflammation, angiogenesis, lymphangiogenesis, tissue remodelling, and fibrosis. Mast cells release preformed mediators (e.g. histamine, tryptase, and chymase) and de novo synthesized mediators (e.g. cysteinyl leukotriene C4 and prostaglandin D2), as well as cytokines and chemokines, which can activate different resident immune cells (e.g. macrophages) and structural cells (e.g. fibroblasts and endothelial cells) in the human heart and aorta. The transcriptional profiles of various mast cell populations highlight their potential heterogeneity and distinct gene and proteome expression. Mast cell plasticity and heterogeneity enable these cells the potential for performing different, even opposite, functions in response to changing tissue contexts. Human cardiac mast cells display significant differences compared with mast cells isolated from other organs. These characteristics make cardiac mast cells intriguing, given their dichotomous potential roles of inducing or protecting against cardiovascular diseases. Identification of cardiac mast cell subpopulations represents a prerequisite for understanding their potential multifaceted roles in health and disease. Several new drugs specifically targeting human mast cell activation are under development or in clinical trials. Mast cells and/or their subpopulations can potentially represent novel therapeutic targets for cardiovascular disorders.


Subject(s)
Cardiovascular Diseases , Mast Cells , Humans , Mast Cells/metabolism , Mast Cells/immunology , Mast Cells/drug effects , Mast Cells/pathology , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Signal Transduction , Phenotype , Myocardium/pathology , Myocardium/metabolism , Myocardium/immunology , Cardiovascular Agents/therapeutic use , Cardiovascular Agents/pharmacology , Cell Plasticity/drug effects , Inflammation Mediators/metabolism
12.
Inflamm Res ; 73(6): 945-960, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38587532

ABSTRACT

OBJECTIVE AND DESIGN: Mast cells (MCs), as the fastest immune responders, play a critical role in the progression of neuroinflammation-related diseases, especially in depression. Quercetin (Que) and kaempferol (Kae), as two major diet-derived flavonoids, inhibit MC activation and exhibit significant antidepressant effect due to their anti-inflammatory capacity. The study aimed to explore the mechanisms of inhibitory effect of Que and Kae on MC activation, and whether Que and Kae suppress hippocampal mast cell activation in LPS-induced depressive mice. SUBJECTS AND TREATMENT: In vitro assays, human mast cells (HMC-1) were pretreated with Que or Kae for 1 h, then stimulated by phorbol 12-myristate 13-acetate (PMA) and 2,5-di-t-butyl-1,4-benzohydroquinone (tBHQ) for 3 h or 12 h. In vivo assays, Que or Kae was administered by oral gavage once daily for 14 days and then lipopolysaccharide (LPS) intraperitoneally injection to induce depressive behaviors. METHODS: The secretion and expression of TNF-α were determined by ELISA and Western blotting. The nuclear factor of activated T cells (NFAT) transcriptional activity was measured in HMC-1 stably expressing NFAT luciferase reporter gene. Nuclear translocation of NFATc2 was detected by nuclear protein extraction and also was fluorescently detected in HMC-1 stably expressing eGFP-NFATc2. We used Ca2+ imaging to evaluate changes of store-operated calcium entry (SOCE) in HMC-1 stably expressing fluorescent Ca2+ indicator jGCamP7s. Molecular docking was used to assess interaction between the Que or Kae and calcium release-activated calcium modulator (ORAI). The  hippocampal mast cell accumulation and activation  were detected by toluidine blue staining and immunohistochemistry with ß-tryptase. RESULTS: In vitro assays of HMC-1 activated by PtBHQ (PMA and tBHQ), Que and Kae significantly decreased expression and secretion of TNF-α. Moreover, NFAT transcriptional activity and nuclear translocation of NFATc2 were remarkably inhibited by Que and Kae. In addition, the Ca2+ influx mediated by SOCE was suppressed by Que, Kae and the YM58483 (ORAI inhibitor), respectively. Importantly, the combination of YM58483 with Que or Kae had no additive effect on the inhibition of SOCE. The molecular docking also showed that Que and Kae both exhibit high binding affinities with ORAI at the same binding site as YM58483. In vivo assays, Que and Kae significantly reversed LPS-induced depression-like behaviors in mice, and inhibited hippocampal mast cell activation  in LPS-induced depressive mice. CONCLUSIONS: Our results indicated that suppression of SOCE/NFATc2 pathway-mediated by ORAI channels may be the mechanism of inhibitory effect of Que and Kae on MC activation, and also suggested Que and Kae may exert the antidepressant effect through suppressing hippocampal mast cell activation.


Subject(s)
Depression , Hippocampus , Kaempferols , Lipopolysaccharides , Mast Cells , NFATC Transcription Factors , Quercetin , Animals , Mast Cells/drug effects , Mast Cells/metabolism , NFATC Transcription Factors/metabolism , Kaempferols/pharmacology , Kaempferols/therapeutic use , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Male , Quercetin/pharmacology , Quercetin/therapeutic use , Depression/chemically induced , Depression/drug therapy , Depression/metabolism , Cell Line , Signal Transduction/drug effects , Mice , Calcium/metabolism , Calcium Channels/metabolism , Mice, Inbred C57BL , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use
13.
Clin Cancer Res ; 30(11): 2582-2597, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38578281

ABSTRACT

PURPOSE: To explore the cellular cross-talk of tumor-resident mast cells (MC) in controlling the activity of cancer-associated fibroblasts (CAF) to overcome tumor microenvironment (TME) abnormalities, enhancing the efficacy of immune-checkpoint inhibitors in sarcoma. EXPERIMENTAL DESIGN: We used a coculture system followed by further validation in mouse models of fibrosarcoma and osteosarcoma with or without administration of the MC stabilizer and antihistamine ketotifen. To evaluate the contribution of ketotifen in sensitizing tumors to therapy, we performed combination studies with doxorubicin chemotherapy and anti-PD-L1 (B7-H1, clone 10F.9G2) treatment. We investigated the ability of ketotifen to modulate the TME in human sarcomas in the context of a repurposed phase II clinical trial. RESULTS: Inhibition of MC activation with ketotifen successfully suppressed CAF proliferation and stiffness of the extracellular matrix accompanied by an increase in vessel perfusion in fibrosarcoma and osteosarcoma as indicated by ultrasound shear wave elastography imaging. The improved tissue oxygenation increased the efficacy of chemoimmunotherapy, supported by enhanced T-cell infiltration and acquisition of tumor antigen-specific memory. Importantly, the effect of ketotifen in reducing tumor stiffness was further validated in sarcoma patients, highlighting its translational potential. CONCLUSIONS: Our study suggests the targeting of MCs with clinically administered drugs, such as antihistamines, as a promising approach to overcome resistance to immunotherapy in sarcomas.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Mast Cells , Tumor Microenvironment , Humans , Mice , Animals , Mast Cells/drug effects , Mast Cells/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Sarcoma/drug therapy , Sarcoma/pathology , Sarcoma/immunology , Ketotifen/pharmacology , Ketotifen/therapeutic use , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Female , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/immunology
14.
Int Immunopharmacol ; 130: 111798, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38442583

ABSTRACT

Asthma is a serious global health problem affecting 300 million persons around the world. Mast cells (MCs) play a major role in airway hyperresponsiveness (AHR) and inflammation in asthma, their exact effector mechanisms remain unclear. Here, we aim to investigate the inhibitory effect of Bergapten (BER) on MRGPRX2-mediated MCs activation through asthma model. Mouse model of asthma was established to examine the anti-asthmatic effects of BER. Calcium (Ca2+) influx, ß-hexosaminidase and histamine release were used to assess MCs degranulation in vitro. RNA-Seq technique was conducted to study the gene expression profile. RT-PCR and Western Blotting were performed to examine targeting molecules expression. BER inhibited AHR, inflammation, mucous secretion, collagen deposition and lung MCs activation in asthma model. BER dramatically reduced levels of IL4, IL-5, and IL-13 in bronchoalveolar lavage fluid (BALF), as well as inflammatory cells. BER also reduced serum IgE levels. Pretreatment MCs with BER inhibited substance P (SP)-induced Ca2+ influx, degranulation and cytokines release from MCs. BER also reduced the phosphorylation levels of PKC, PLC, IP3R, AKT and ERK, which were induced by SP. Furthermore, RNA-seq analysis showed that SP up-regulated 68 genes in MCs, while were reversed by BER. Among these 68 genes, SP up-regulated NR4A1 expression, and this effect was inhibited by BER. Meanwhile, knockdown of NR4A1 significantly attenuated SP-induced MCs degranulation. In conclusion, NR4A1 plays a major role in MRGPRX2-mediated MCs activation, BER inhibited AHR and inflammation in asthmatic model by inhibiting MCs activation through MRGPRX2-NR4A1 pathway.


Subject(s)
5-Methoxypsoralen , Anti-Inflammatory Agents , Asthma , Mast Cells , Animals , Mice , 5-Methoxypsoralen/pharmacology , 5-Methoxypsoralen/therapeutic use , Asthma/drug therapy , Cell Degranulation , Inflammation/drug therapy , Lung/metabolism , Mast Cells/drug effects , Receptors, G-Protein-Coupled/metabolism , Substance P/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Mice, Inbred C57BL , Female
15.
Immunol Invest ; 53(4): 695-711, 2024 May.
Article in English | MEDLINE | ID: mdl-38504489

ABSTRACT

Mast cells play important role in acquired and natural immunity making these favorable therapeutic targets in various inflammatory diseases. Here we observed that, pentacyclic tri terpenoid betulinic acid (BA) treatment resulted in a significantly high number (9%) of cells positive for Hoechst and negative for annexin-V indicating that BA could interfere with plasma membrane integrity. The degranulation of both activated and non-activated mast cells was enhanced upon treatment with BA. The pre-treatment of BA had remarkable effect on calcium response in activated mast cells which showed increased calcium influx relative compared to untreated cells. The results also showed potentially less migration of BA treated mast cells signifying the possible effect of BA on cell membrane. BA treatment resulted in a significant increase in mRNA levels of IL-13 while as mRNA levels of other target cytokines, IL-6 and TNF-α seem to be not affected. Moreover, there was global Increase in phosphorylation of signaling proteins and no significant change in phosphorylation of FcεRI receptors indicating that the effect of BA was independent of signaling cascade or FcεRI receptor mediated mast cell aggregation. Overall, these results portray BA potentiates mast cell effector functions by compromising the membrane integrity and independent of FcεRI involvement.


Subject(s)
Betulinic Acid , Cell Degranulation , Cell Membrane , Mast Cells , Pentacyclic Triterpenes , Receptors, IgE , Triterpenes , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Pentacyclic Triterpenes/pharmacology , Cell Degranulation/drug effects , Receptors, IgE/metabolism , Cell Membrane/metabolism , Cell Membrane/drug effects , Triterpenes/pharmacology , Animals , Signal Transduction/drug effects , Calcium/metabolism , Rats , Phosphorylation/drug effects , Cytokines/metabolism
16.
Br J Pharmacol ; 181(13): 1993-2011, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38450758

ABSTRACT

BACKGROUND: The therapeutic potential of relaxin for heart failure and renal disease in clinical trials is hampered by the short half-life of serelaxin. Optimization of fatty acid-acetylated single-chain peptide analogues of relaxin culminated in the design and synthesis of R2R01, a potent and selective RXFP1 agonist with subcutaneous bioavailability and extended half-life. EXPERIMENTAL APPROACH: Cellular assays and pharmacological models of RXFP1 activation were used to validate the potency and selectivity of R2R01. Increased renal blood flow was used as a translational marker of R2R01 activity. Human mastocytes (LAD2 cells) were used to study potential pseudo-allergic reactions and CD4+ T-cells to study immunogenicity. The pharmacokinetics of R2R01 were characterized in rats and minipigs. KEY RESULTS: In vitro, R2R01 had comparable potency and efficacy to relaxin as an agonist for human RXFP1. In vivo, subcutaneous administration of R2R01 increased heart rate and renal blood flow in normotensive and hypertensive rat and did not show evidence of tachyphylaxis. R2R01 also increased nipple length in rats, used as a chronic model of RXFP1 engagement. Pharmacokinetic studies showed that R2R01 has a significantly extended terminal half-life. The in vitro assays with LAD2 cells and CD4+ T-cells showed that R2R01 had low potential for pseudo-allergic and immunogenic reactions, respectively. CONCLUSION AND IMPLICATIONS: R2R01 is a potent RXFP1 agonist with an extended half-life that increases renal blood flow in various settings including normotensive and hypertensive conditions. The preclinical efficacy and safety data supported clinical development of R2R01 as a potential new therapy for renal and cardiovascular diseases.


Subject(s)
Receptors, G-Protein-Coupled , Animals , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Humans , Rats , Swine , Male , Receptors, Peptide/agonists , Receptors, Peptide/metabolism , Swine, Miniature , Cardiovascular Diseases/drug therapy , Kidney Diseases/drug therapy , Rats, Sprague-Dawley , Peptides/pharmacology , Peptides/administration & dosage , Peptides/pharmacokinetics , Relaxin/pharmacology , Relaxin/administration & dosage , Relaxin/pharmacokinetics , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism
18.
Nat Nanotechnol ; 19(5): 698-704, 2024 May.
Article in English | MEDLINE | ID: mdl-38228804

ABSTRACT

Protein adsorption onto nanomaterials often results in denaturation and loss of bioactivity. Controlling the adsorption process to maintain the protein structure and function has potential for a range of applications. Here we report that self-assembled poly(propylene sulfone) (PPSU) nanoparticles support the controlled formation of multicomponent enzyme and antibody coatings and maintain their bioactivity. Simulations indicate that hydrophobic patches on protein surfaces induce a site-specific dipole relaxation of PPSU assemblies to non-covalently anchor the proteins without disrupting the protein hydrogen bonding or structure. As a proof of concept, a nanotherapy employing multiple mast-cell-targeted antibodies for preventing anaphylaxis is demonstrated in a humanized mouse model. PPSU nanoparticles displaying an optimized ratio of co-adsorbed anti-Siglec-6 and anti-FcεRIα antibodies effectively inhibit mast cell activation and degranulation, preventing anaphylaxis. Protein immobilization on PPSU surfaces provides a simple and rapid platform for the development of targeted protein nanomedicines.


Subject(s)
Mast Cells , Nanoparticles , Mast Cells/drug effects , Mast Cells/metabolism , Animals , Mice , Adsorption , Humans , Nanoparticles/chemistry , Nanomedicine/methods , Anaphylaxis , Polypropylenes/chemistry , Cell Degranulation/drug effects
19.
J Microbiol Biotechnol ; 34(4): 765-773, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38247218

ABSTRACT

Ozone, a highly reactive oxidant molecule, is widely used as a complementary therapy for various skin diseases, including wound healing, pressure ulcers, diabetic foot, and infections. However, there is limited research on the effectiveness of ozone for atopic dermatitis (AD). Ozonated sunflower oil (OSO) is an active ingredient obtained from partially ozonated sunflower oil (SO). OSO markedly reduced the LPS-induced increase in IL-1ß and nitric oxide (NO) levels in RAW 264.7 mouse macrophage cells. Oxazolone (OXZ) was applied to hairless mice to induce AD-like skin symptoms and immune response. OSO significantly alleviated the OXZ-induced increases in the number of infiltrating mast cells, epidermal thickness, AD symptoms, thymic stromal lymphopoietin (TSLP), and filaggrin, as well as the serum levels of NO, IgE, IL-1ß, and TNF-α. Furthermore, OSO inhibited the IL-4/STAT3/MAPK pathway and the expression of NF-κB. Our results suggest that OSO treatment could relieve AD-mediated skin damage through its anti-inflammatory and antioxidant activities. Therefore, it can be used as a therapeutic agent against AD-related skin diseases.


Subject(s)
Cytokines , Dermatitis, Atopic , Lipopolysaccharides , Nitric Oxide , Oxazolone , Ozone , Sunflower Oil , Animals , Mice , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , RAW 264.7 Cells , Cytokines/metabolism , Oxazolone/toxicity , Nitric Oxide/metabolism , Immunoglobulin E/blood , NF-kappa B/metabolism , Disease Models, Animal , Macrophages/drug effects , Macrophages/immunology , Interleukin-1beta/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , STAT3 Transcription Factor/metabolism , Skin/drug effects , Skin/pathology , Thymic Stromal Lymphopoietin , Inflammation/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Filaggrin Proteins , Interleukin-4/metabolism , Anti-Inflammatory Agents/pharmacology
20.
Int J Mol Sci ; 24(5)2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36902240

ABSTRACT

Mast cells (MCs) are tissue cells that are derived from bone marrow stem cells that contribute to allergic reactions, inflammatory diseases, innate and adaptive immunity, autoimmunity, and mental disorders. MCs located near the meninges communicate with microglia through the production of mediators such as histamine and tryptase, but also through the secretion of IL-1, IL-6 and TNF, which can create pathological effects in the brain. Preformed chemical mediators of inflammation and tumor necrosis factor (TNF) are rapidly released from the granules of MCs, the only immune cells capable of storing the cytokine TNF, although it can also be produced later through mRNA. The role of MCs in nervous system diseases has been extensively studied and reported in the scientific literature; it is of great clinical interest. However, many of the published articles concern studies on animals (mainly rats or mice) and not on humans. MCs are known to interact with neuropeptides that mediate endothelial cell activation, resulting in central nervous system (CNS) inflammatory disorders. In the brain, MCs interact with neurons causing neuronal excitation with the production of neuropeptides and the release of inflammatory mediators such as cytokines and chemokines. This article explores the current understanding of MC activation by neuropeptide substance P (SP), corticotropin-releasing hormone (CRH), and neurotensin, and the role of pro-inflammatory cytokines, suggesting a therapeutic effect of the anti-inflammatory cytokines IL-37 and IL-38.


Subject(s)
Cytokines , Mast Cells , Neuropeptides , Animals , Humans , Mice , Rats , Cytokines/physiology , Inflammation , Mast Cells/drug effects , Mast Cells/physiology , Substance P , Tumor Necrosis Factor-alpha , Neuropeptides/pharmacology , Neuropeptides/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...