Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.027
Filter
1.
Front Immunol ; 15: 1369326, 2024.
Article in English | MEDLINE | ID: mdl-38953022

ABSTRACT

Objectives: Mast cell (MC) degranulation is a key process in allergic reactions and inflammatory responses. Aspartate aminotransferase 1 (AAT1)-derived endogenous sulfur dioxide (SO2) is an important regulator of MC function. However, the mechanism underlying its role in MC degranulation remains unclear. This study aimed to investigate the mechanism by which endogenous SO2 controlled MC degranulation. Methods: HMC-1 and Rat basophilic leukemia cell MC line (RBL-2H3) were used in the cell experiments. SO2 content was detected by in situ fluorescent probe. MC degranulation represented by the release rate of MC ß-hexosaminidase was determined using a colorimetric assay. Sulfenylation of galectin-9 (Gal-9) in MCs and purified protein was detected using a biotin switch assay. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to determine the exact sulfenylation sites of Gal-9 by SO2. Animal models of passive cutaneous anaphylaxis (PCA) and hypoxia-driven pulmonary vascular remodeling were used to investigate the effect of SO2 on mast cell activation in vivo. Site-directed mutation of Gal-9 was conducted to confirm the exact site of SO2 and support the significance of SO2/Gal-9 signal axis in the regulation of MC degranulation. Results: Degranulation was increased in AAT1-knockdowned MCs, and SO2 supplementation reversed the increase in MC degranulation. Furthermore, deficiency of endogenous SO2 contributed to IgE-mediated degranulation in vitro. Besides, SO2 inhibited IgE-mediated and hypoxia-driven MC degranulation in vivo. Mechanistically, LC-MS/MS analysis and site-directed mutation results showed that SO2 sulfenylated Gal-9 at cysteine 74. Sulfenylation of the 74th cysteine of Gal-9 protein was required in the SO2-inhibited MC degranulation under both physiological and pathophysiological conditions. Conclusion: These findings elucidated that SO2 inhibited MC degranulation via sulfenylating Gal-9 under both physiological and pathophysiological conditions, which might provide a novel treatment approach for MC activation-related diseases.


Subject(s)
Cell Degranulation , Cysteine , Galectins , Mast Cells , Sulfur Dioxide , Animals , Cell Degranulation/drug effects , Mast Cells/metabolism , Mast Cells/immunology , Mast Cells/drug effects , Cysteine/metabolism , Rats , Sulfur Dioxide/pharmacology , Sulfur Dioxide/metabolism , Humans , Galectins/metabolism , Mice , Male , Passive Cutaneous Anaphylaxis , Cell Line
2.
Cell Physiol Biochem ; 58(3): 212-225, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38852193

ABSTRACT

BACKGROUND/AIMS: Adrenaline quickly inhibits the release of histamine from mast cells. Besides ß2-adrenergic receptors, several in vitro studies also indicate the involvement of α-adrenergic receptors in the process of exocytosis. Since exocytosis in mast cells can be detected electrophysiologically by the changes in the membrane capacitance (Cm), its continuous monitoring in the presence of drugs would determine their mast cell-stabilizing properties. METHODS: Employing the whole-cell patch-clamp technique in rat peritoneal mast cells, we examined the effects of adrenaline on the degranulation of mast cells and the increase in the Cm during exocytosis. We also examined the degranulation of mast cells in the presence or absence of α-adrenergic receptor agonists or antagonists. RESULTS: Adrenaline dose-dependently suppressed the GTP-γ-S-induced increase in the Cm and inhibited the degranulation from mast cells, which was almost completely erased in the presence of butoxamine, a ß2-adrenergic receptor antagonist. Among α-adrenergic receptor agonists or antagonists, high dose prazosin, a selective α1-adrenergic receptor antagonist, significantly reduced the ratio of degranulating mast cells and suppressed the increase in the Cm. Additionally, prazosin augmented the inhibitory effects of adrenaline on the degranulation of mast cells. CONCLUSION: This study provided electrophysiological evidence for the first time that adrenaline dose-dependently inhibited the process of exocytosis, confirming its usefulness as a potent mast cell-stabilizer. The pharmacological blockade of α1-adrenergic receptor by prazosin synergistically potentiated such mast cell-stabilizing property of adrenaline, which is primarily mediated by ß2-adrenergic receptors.


Subject(s)
Cell Degranulation , Epinephrine , Exocytosis , Mast Cells , Prazosin , Animals , Mast Cells/drug effects , Mast Cells/metabolism , Mast Cells/cytology , Epinephrine/pharmacology , Rats , Prazosin/pharmacology , Cell Degranulation/drug effects , Male , Exocytosis/drug effects , Patch-Clamp Techniques , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Rats, Wistar
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 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 , Receptors, G-Protein-Coupled , TRPV Cation Channels , Animals , Humans , Male , Mice , Antipruritics/pharmacology , Antipruritics/therapeutic use , Calcium/metabolism , Cell Degranulation/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HEK293 Cells , Histamine/metabolism , Mast Cells/drug effects , Mast Cells/immunology , Mice, Inbred C57BL , Molecular Docking Simulation , Nerve Tissue Proteins/metabolism , Pruritus/drug therapy , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine/metabolism , TRPV Cation Channels/metabolism
5.
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
6.
J Immunotoxicol ; 21(1): 2345152, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38659406

ABSTRACT

The recent global resurgence of severe infections caused by the Group A streptococcus (GAS) pathogen, Streptococcus pyogenes, has focused attention on this microbial pathogen, which produces an array of virulence factors, such as the pore-forming toxin, streptolysin O (SOT). Importantly, the interactions of SOT with human neutrophils (PMN), are not well understood. The current study was designed to investigate the effects of pretreatment of isolated human PMN with purified SOT on several pro-inflammatory activities, including generation of reactive oxygen species (ROS), degranulation (elastase release), influx of extracellular calcium (Ca2+) and release of extracellular DNA (NETosis), using chemiluminescence, spectrophotometric and fluorimetric procedures, respectively. Exposure of PMN to SOT alone caused modest production of ROS and elastase release, while pretreatment with the toxin caused significant augmentation of chemoattractant (fMLP)-activated ROS generation and release of elastase by activated PMN. These effects of treatment of PMN with SOT were associated with both a marked and sustained elevation of cytosolic Ca2+concentrations and significant increases in the concentrations of extracellular DNA, indicative of NETosis. The current study has identified a potential role for SOT in augmenting the Ca2+-dependent pro-inflammatory interactions of PMN, which, if operative in a clinical setting, may contribute to hyper-activation of PMN and GAS-mediated tissue injury.


Subject(s)
Extracellular Traps , Neutrophils , Streptococcus pyogenes , Streptolysins , Humans , Bacterial Proteins/metabolism , Calcium/metabolism , Cell Degranulation/drug effects , Cells, Cultured , Extracellular Traps/immunology , Extracellular Traps/metabolism , Inflammation/immunology , Neutrophil Activation/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/drug effects , Pancreatic Elastase/metabolism , Reactive Oxygen Species/metabolism , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Streptolysins/metabolism
7.
Front Immunol ; 15: 1360615, 2024.
Article in English | MEDLINE | ID: mdl-38646521

ABSTRACT

Introduction: Malignant ascites indicates ovarian cancer progression and predicts poor clinical outcome. Various ascites components induce an immunosuppressive crosstalk between tumor and immune cells, which is poorly understood. In our previous study, imbalanced electrolytes, particularly high sodium content in malignant ascites, have been identified as a main immunosuppressive mechanism that impaired NK and T-cell activity. Methods: In the present study, we explored the role of high concentrations of ascites proteins and immunoglobulins on antitumoral NK effector functions. To this end, a coculture system consisting of healthy donor NK cells and ovarian cancer cells was used. The anti-EGFR antibody Cetuximab was added to induce antibody-dependent cellular cytotoxicity (ADCC). NK activity was assessed in the presence of different patient ascites samples and immunoglobulins that were isolated from ascites. Results: Overall high protein concentration in ascites impaired NK cell degranulation, conjugation to tumor cells, and intracellular calcium signaling. Immunoglobulins isolated from ascites samples competitively interfered with NK ADCC and inhibited the conjugation to target cells. Furthermore, downregulation of regulatory surface markers CD16 and DNAM-1 on NK cells was prevented by ascites-derived immunoglobulins during NK cell activation. Conclusion: Our data show that high protein concentrations in biological fluids are able to suppress antitumoral activity of NK cells independent from the mechanism mediated by imbalanced electrolytes. The competitive interference between immunoglobulins of ascites and specific therapeutic antibodies could diminish the efficacy of antibody-based therapies and should be considered in antibody-based immunotherapies.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Ascites , Killer Cells, Natural , Ovarian Neoplasms , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Ascites/immunology , Female , Antibody-Dependent Cell Cytotoxicity/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Cell Line, Tumor , Immunoglobulins/metabolism , Receptors, IgG/metabolism , Receptors, IgG/immunology , Cell Degranulation/immunology , Cell Degranulation/drug effects , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , Cetuximab/pharmacology
8.
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
9.
J Sci Food Agric ; 104(10): 5955-5963, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38415860

ABSTRACT

BACKGROUND: Food allergy has become a global public health problem. This study aimed to explore the possible anti-allergic effect of vitamin C (VC). A rat basophilic leukemia (RBL)-2H3 cell degranulation model was used to assess the effect of VC on degranulation in vitro, and an ovalbumin (OVA)-induced BALB/c mouse allergy model was used to assess the anti-allergy effect of VC in vivo. RESULTS: In vitro, VC significantly attenuated the release of ß-hexosaminidase, tryptase and histamine, and also reduced cytokine production (interleukins 4 and 6, tumor necrosis factor α) significantly (P < 0.05), with the inhibitory effect demonstrating a positive correlation with VC dose. In vivo, compared with the OVA group, the levels of serum immunoglobulins E and G1 of the VC low-dose (VCL) group (50 mg kg-1) and high-dose (VCH) group (200 mg·kg-1) were significantly reduced (P < 0.05). Furthermore, the plasma histamine level was also significantly decreased (P < 0.05). Moreover, TH2 cell polarization in mice of the VCL and VCH groups was significantly inhibited (P < 0.05), promoting the TH1/TH2 cell polarization balance. Additionally, VC treatment enhanced the expression of CD80 (P < 0.05) in spleen and small intestine tissues, while significantly inhibiting the expression of CD86 (P < 0.05); notably, high-dose VC treatment was more effective. CONCLUSION: VC exerted an anti-allergic effect through inhibiting degranulation and regulating TH1/TH2 cell polarization balance. © 2024 Society of Chemical Industry.


Subject(s)
Anti-Allergic Agents , Ascorbic Acid , Cell Degranulation , Food Hypersensitivity , Mice, Inbred BALB C , Th1 Cells , Th2 Cells , Animals , Th2 Cells/immunology , Th2 Cells/drug effects , Anti-Allergic Agents/pharmacology , Mice , Ascorbic Acid/pharmacology , Cell Degranulation/drug effects , Th1 Cells/immunology , Th1 Cells/drug effects , Rats , Food Hypersensitivity/drug therapy , Food Hypersensitivity/immunology , Immunoglobulin E/immunology , Humans , Female , Male , Ovalbumin/immunology , Ovalbumin/adverse effects , Cytokines/metabolism , Cytokines/immunology , beta-N-Acetylhexosaminidases/metabolism
10.
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
12.
J Ethnopharmacol ; 289: 115053, 2022 May 10.
Article in English | MEDLINE | ID: mdl-35104575

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Houttuynia cordata Thunb., a plant belonging to the family of Saururaceae, has been used as a traditional Chinese medicine for more than 1500 years. Because of its various pharmacological activities, it was widely used as antipyretic, detoxification, anti-inflammatory drugs. Houttuynia cordata (HC) injection was prepared using contemporary methods to extract effective components from H. cordata Thunb. However, the adverse event reports of HC injection are accumulating remarkably with the HC injection clinical applications increased. Previous studies demonstrated that the major side effects of HC injection were anaphylactoid reactions. Our work might shed the light on the role of Mas-related G-protein coupled receptor-X2 (MRGPRX2) in modulating drug-induced anaphylactoid reactions. AIM OF THE STUDY: We aimed to investigate the role of the mouse Mas-related G-protein coupled receptor B2 (Mrgprb2) (the orthologous gene of human MRGPRX2) in anaphylactoid reactions induced by HC injection. MATERIALS AND METHODS: Mrgprb2 related anaphylactoid reactions induced by HC injection were investigated by histamine/ß-hexosaminidase releasing, mast cell degranulation, and hind paw swelling assays by using a Mrgprb2 knockout mouse model. Furthermore, the transcriptomic profiles of the anaphylactoid reaction induced by HC injection was analyzed by RNA sequencing. RESULTS: Mice without Mrgprb2 exhibited significantly decreasing in mast cell degranulation, serum histamine release, and hind paw swelling degrees. The RNA sequencing results indicated that Mrgprb2 could play a pivotal role in HC injection induced anaphylactoid reaction mediated by mTOR/AMPK pathway. Intriguingly, our results showed that Mrgprb2 might involve in Compound 48/80 induced anaphylactoid reactions mediated by Reelin/E-cadherin axis, which suggested different roles of Mrgprb2 in anaphylactoid reactions induced by HC injection and C48/80. CONCLUSION: Our studies reported effects and underlying mechanisms of Mrgprb2 in the anaphylactoid reaction induced by HC injection.


Subject(s)
Anaphylaxis/etiology , Drugs, Chinese Herbal/toxicity , Houttuynia/chemistry , Receptors, G-Protein-Coupled/genetics , Anaphylaxis/genetics , Animals , Cell Degranulation/drug effects , Drugs, Chinese Herbal/administration & dosage , Female , Histamine Release/drug effects , Male , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , p-Methoxy-N-methylphenethylamine/toxicity
13.
Toxicol Lett ; 359: 10-21, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35114312

ABSTRACT

Organic anion transporters 1 (OAT1) and OAT3 are responsible for transporting adefovir (ADV) into renal tubular epithelial cells. Our previous research found that ADV accumulated in the renal interstitium and caused renal interstitial fibrosis when Oat1/3 were inhibited by OATs inhibitor probenecid for long-term. Mast cells (MCs) in the interstitial space are considered to be key drivers of renal fibrosis. The current work investigated the effect of ADV on MCs in vitro and during the development of interstitial fibrosis in rats. Results indicate that ADV triggers chymase release from cultured RBL-2H3 mast cells in a time-and concentration-dependent manner. Angiotensin II (Ang II) in renal interstitium is generated mainly by chymase, renin and other products released from MCs, and has a direct effect on fibrosis through the angiotensin receptor. The concentrations of Ang II and fibrosis was significantly increased after administration of ADV alone or with probenecid for 4 weeks. The MCs membrane stabilizer sodium cromoglycate (SCG) and the angiotensin receptor antagonist Valsartan (VAL) could ameliorate ADV-induced nephrotoxicity. Additionally, SCG or VAL could reduce the accumulation of ADV in the renal interstitium by upregulating the expression of Oat1/3 and multidrug resistance-associated protein 4. Therefore, ADV accumulation in the renal interstitium could promote the degranulation of interstitial MCs and drive the development of renal fibrosis. SCG or VAL could ameliorate ADV-associated fibrosis by decreasing degranulation of MCs and accelerating renal clearance of ADV.


Subject(s)
Adenine/analogs & derivatives , Adenine/toxicity , Cell Degranulation/drug effects , Fibrosis/chemically induced , Kidney Diseases/chemically induced , Mast Cells/drug effects , Organophosphonates/toxicity , Adenine/blood , Animals , Disease Models, Animal , Fibrosis/physiopathology , Humans , Kidney Diseases/physiopathology , Kidney Tubules/drug effects , Male , Organophosphonates/blood , Rats
14.
J Pharm Pharmacol ; 74(3): 397-408, 2022 Mar 03.
Article in English | MEDLINE | ID: mdl-34969089

ABSTRACT

OBJECTIVES: The ethyl acetate extraction of Artemisia ordosica Krasch (AOK) root showed anti-allergic rhinitis (AR) effect, while the active compounds and pharmacological targets were unknown. METHODS: The P815 degranulation was established by cell counting kit 8 assay, ß-hexosaminidase releasing assay and toluidine blue staining. The flavonoids were screened in vitro. Then toluidine blue staining and ELISA were carried out to investigate the anti-inflammatory effects of the active compound. Network pharmacology was implemented to explain the mechanisms of the active compound. iGEMDOCK was used to investigate the binding between active compound and hub targets. KEY FINDINGS: C48/80 was the optimum reagent in triggering P815 degranulation. Naringenin could significantly decrease P815 degranulation. Meanwhile, naringenin could remarkably increase the IL-4 and decrease the tumour necrosis factor-α. The effect of naringenin on AR was achieved by regulating multiple targets (e.g. AKT1, MAPK3, VEGFA) and pathways (e.g. pathways in cancer, VEGF signalling pathway). Nine hub proteins were obtained by topological analysis. Multiple hydrogen bonds and van der Waals forces were formed between the naringenin and the residues of hub proteins. CONCLUSIONS: Naringenin might be one of the effective ingredients of AOK against AR. And its effects could achieve through regulating multiple targets and pathways.


Subject(s)
Artemisia/chemistry , Flavanones/pharmacology , Mast Cells/drug effects , Rhinitis, Allergic/drug therapy , Acetates/chemistry , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Cell Degranulation/drug effects , Cell Line, Tumor , Flavanones/isolation & purification , Mast Cells/metabolism , Mice , Molecular Docking Simulation , Network Pharmacology , Plant Roots
16.
Toxicology ; 465: 153034, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34774977

ABSTRACT

Iodinated contrast media (ICM) is widely used in radiological examination and interventional therapy. In the commonly used ICM, iodixanol is considered to be the safer one. However, compared with other ICMs, it has a higher incidence of delayed cutaneous adverse reactions. The underlying mechanisms are unclear. In this study, mice with positive allergic reactions were selected based on the mouse clinical allergy symptom score and skin and blood samples taken 1, 6, 24, 48, and 72 h after ICMs (6 g iodine/kg) injection for histological and blood analyses. ICMs-induced pseudo-allergic reactions were investigated through in vivo intravital vascular imaging and passive cutaneous anaphylaxis (PCA) not mediated by IgE and through, calcium imaging degranulation of mast cells (MCs), and western blot assays in vitro. Results shows iodixanol-induced systemic anaphylaxis caused severe extravasation of plasma proteins and degranulation of skin MCs, and increased levels of plasma histamine, cytokines and inflammatory chemokines. Mechanistically, iodixanol increases degranulation of MCs and promotes the synthesis of inflammatory factors by activating PLC-γ and PI3K-related pathways. Trigonelline inhibit iodixanol-induced MC-related pseudo-allergic reactions in vitro and in vivo. These results suggest that mice in the iodixanol group had a higher incidence of delayed cutaneous reactions, characterized by cytokine release over time and delayed cutaneous MC degranulation. Iodixanol's delayed cutaneous adverse reactions may be due to a delayed phase of MC-related pseudo-allergic reactions. Trigonelline revealed anti-allergic activity in iodixanol-induced MC-related pseudo-allergic reactions.


Subject(s)
Cell Degranulation/drug effects , Contrast Media/toxicity , Edema/chemically induced , Hypersensitivity, Delayed/chemically induced , Mast Cells/drug effects , Mast Cells/immunology , Passive Cutaneous Anaphylaxis/drug effects , Skin/drug effects , Triiodobenzoic Acids/toxicity , Alkaloids/pharmacology , Animals , Anti-Allergic Agents/pharmacology , Calcium/metabolism , Cell Line , Cytokines/metabolism , Edema/immunology , Edema/metabolism , Edema/prevention & control , Humans , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/metabolism , Hypersensitivity, Delayed/prevention & control , Male , Mast Cell Stabilizers/pharmacology , Mast Cells/metabolism , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinase/metabolism , Phospholipase C gamma/metabolism , Signal Transduction , Skin/immunology , Skin/metabolism , Time Factors
17.
J Med Chem ; 65(1): 485-496, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34931831

ABSTRACT

Inhibitor cystine knot peptides, derived from venom, have evolved to block ion channel function but are often toxic when dosed at pharmacologically relevant levels in vivo. The article describes the design of analogues of ProTx-II that safely display systemic in vivo blocking of Nav1.7, resulting in a latency of response to thermal stimuli in rodents. The new designs achieve a better in vivo profile by improving ion channel selectivity and limiting the ability of the peptides to cause mast cell degranulation. The design rationale, structural modeling, in vitro profiles, and rat tail flick outcomes are disclosed and discussed.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/drug effects , Pain/drug therapy , Sodium Channel Blockers/chemical synthesis , Sodium Channel Blockers/pharmacology , Spider Venoms/chemical synthesis , Animals , Cell Degranulation/drug effects , Cystine/chemistry , Drug Design , Hot Temperature , Mast Cells/drug effects , Models, Molecular , Pain Measurement/drug effects , Rats , Spider Venoms/pharmacology
18.
Hum Exp Toxicol ; 40(12_suppl): S702-S710, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34792426

ABSTRACT

Allergic rhinitis (AR) is a common allergic inflammatory and chronic reactive disease caused by allergen-induced immunoglobulin E (IgE). Tanshinone IIA (Tan IIA) is one of the active ingredients in Salvia miltiorrhiza Bunge (Danshen) and plays a vital role in inhibiting inflammation. Thus, we hypothesized that Tan IIA has anti-allergic effects and studied the function of Tan IIA in mast cells and an AR animal model. We induced RBL-2H3 cell sensitization with monoclonal anti-2,4,6-dinitrophenyl-immunoglobulin (Ig) E/human serum albumin (DNP-IgE/HSA) and constructed an ovalbumin (OVA)-induced AR model in mice. The role of Tan IIA in AR progression was studied using the MTT assay, ELISA, western blot, toluidine blue staining, HE staining, and Alcian blue and safranin O (A&S) staining. Tan IIA treatment significantly increased IgE/HSA-induced cell viability. However, Tan IIA treatment markedly downregulated the expression levels of ß-hexosaminidase, histamine, tumor necrosis factor (TNF-α), interleukin 1ß (IL-1ß), IL-4, and IL-5 in IgE/HSA-induced cells. Furthermore, Tan IIA improved typical symptoms in the OVA-induced AR model mice by inhibiting the phospholipase Cγ1 (PLCγ1)/protein kinase C (PKC)/IP3R pathway. Additionally, Tan IIA effectively improved the degranulation of RBL-2H3 cells and OVA-induced AR in mice. Together, these results suggest that Tan IIA may be a potential drug for the treatment of AR in the future.


Subject(s)
Abietanes/pharmacology , Cell Degranulation/drug effects , Mast Cells/drug effects , Phospholipase C gamma/metabolism , Protein Kinase C/metabolism , Rhinitis, Allergic/drug therapy , Abietanes/therapeutic use , Animals , Cell Line , Disease Models, Animal , Mice , Ovalbumin/adverse effects , Rhinitis, Allergic/metabolism
19.
Front Immunol ; 12: 732209, 2021.
Article in English | MEDLINE | ID: mdl-34650557

ABSTRACT

Background: Perioperative hypersensitivity reaction (HR) is an IgE-FcϵRI-mediated hypersensitivity reaction with degranulation and activation of mast cells and basophils. Several studies have focused on assessing the degranulation and activation of mast cells and basophils to diagnose and predict the prognosis of drug induced HR. However, it is challenging to isolate sufficiently pure mast cells and basophils from human sources to investigate. Effective biomarkers to assess mast cells and basophils activation in vivo could potentially have high diagnostic and prognostic values. In the present study, we investigated EVs pelleted from serum in patients with succinylated gelatin induced HR. Methods: Extracellular vesicles (EVs) were isolated using a total exosome isolation kit and ultracentrifugation, characterized by Western blot, transmission electron microscopy, and nanoparticle tracking analysis. Basophils were isolated from fresh peripheral blood by negative selection using Basophil Isolation Kit II. Human mast cell line was stimulated with IL4. The expression levels of proteins related to the hypersensitive response were evaluated by Western blotting and flow Cytometer. Histamine and tryptase levels were tested using a commercial ELISA kit, and gene expression of inflammatory mediators was evaluated by qRT-PCR. The receiver operating characteristic (ROC) curve was used to evaluate the specificity and sensitivity of biomarker in predicting HR. Results: The concentration of EVs and protein expression level of CD63, FcϵRI, CD203c and tryptase were significantly (p< 0.05) increased in HR samples. The expression level of mast cell/basophil specific CD203c were significantly increased in EVs derived from serum and basophils of HR patients, and the CD203c+-EVs production in mast cells is dramatically increased in the presence of IL4, which positively correlated with histamine, tryptase and inflammatory mediators. Moreover, the ROC curve of EVs concentration and CD203c expression indicated that CD203c+-EVs had a strong diagnostic ability for HR. Conclusion: Serum CD203c+-EVs serves as a novel diagnostic and prognostic biomarker for HR.


Subject(s)
Drug Hypersensitivity/diagnosis , Extracellular Vesicles/metabolism , Gelatin/adverse effects , Phosphoric Diester Hydrolases/blood , Plasma Substitutes/adverse effects , Pyrophosphatases/blood , Succinates/adverse effects , Adult , Aged , Basophils/drug effects , Basophils/immunology , Basophils/metabolism , Biomarkers/blood , Case-Control Studies , Cell Degranulation/drug effects , Cell Line , Drug Hypersensitivity/blood , Drug Hypersensitivity/immunology , Female , Histamine/metabolism , Histamine Release/drug effects , Humans , Male , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Middle Aged , Perioperative Period , Predictive Value of Tests , Prognosis , Tryptases/metabolism
20.
Front Immunol ; 12: 730346, 2021.
Article in English | MEDLINE | ID: mdl-34566991

ABSTRACT

Mast cell activators are a novel class of mucosal vaccine adjuvants. The polymeric compound, Compound 48/80 (C48/80), and cationic peptide, Mastoparan 7 (M7) are mast cell activators that provide adjuvant activity when administered by the nasal route. However, small molecule mast cell activators may be a more cost-efficient adjuvant alternative that is easily synthesized with high purity compared to M7 or C48/80. To identify novel mast cell activating compounds that could be evaluated for mucosal vaccine adjuvant activity, we employed high-throughput screening to assess over 55,000 small molecules for mast cell degranulation activity. Fifteen mast cell activating compounds were down-selected to five compounds based on in vitro immune activation activities including cytokine production and cellular cytotoxicity, synthesis feasibility, and selection for functional diversity. These small molecule mast cell activators were evaluated for in vivo adjuvant activity and induction of protective immunity against West Nile Virus infection in BALB/c mice when combined with West Nile Virus envelope domain III (EDIII) protein in a nasal vaccine. We found that three of the five mast cell activators, ST101036, ST048871, and R529877, evoked high levels of EDIII-specific antibody and conferred comparable levels of protection against WNV challenge. The level of protection provided by these small molecule mast cell activators was comparable to the protection evoked by M7 (67%) but markedly higher than the levels seen with mice immunized with EDIII alone (no adjuvant 33%). Thus, novel small molecule mast cell activators identified by high throughput screening are as efficacious as previously described mast cell activators when used as nasal vaccine adjuvants and represent next-generation mast cell activators for evaluation in mucosal vaccine studies.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cell Degranulation/drug effects , Immunity, Mucosal/drug effects , Mast Cells/drug effects , West Nile Fever/prevention & control , West Nile Virus Vaccines/administration & dosage , West Nile virus/pathogenicity , Administration, Intranasal , Animals , Cell Line , Disease Models, Animal , Drug Discovery , Female , High-Throughput Screening Assays , Host-Pathogen Interactions , Immunity, Mucosal/genetics , Immunization , Immunogenicity, Vaccine , Mast Cells/immunology , Mast Cells/virology , Mice, Inbred BALB C , Proof of Concept Study , West Nile Fever/genetics , West Nile Fever/immunology , West Nile Fever/virology , West Nile virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...