Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
1.
Biochim Biophys Acta Mol Cell Res ; 1869(3): 119203, 2022 03.
Article in English | MEDLINE | ID: mdl-34968576

ABSTRACT

Leukotriene (LT) C4 synthase (LTC4S) catalyzes the conversion from LTA4 to LTC4, which is a proinflammatory lipid mediator in asthma and other inflammatory diseases. LTC4 is metabolized to LTD4 and LTE4, all of which are known as cysteinyl (Cys) LTs and exert physiological functions through CysLT receptors. LTC4S is expressed in adipocytes. However, the function of CysLTs and the regulatory mechanism in adipocytes remain unclear. In this study, we investigated the expression of LTC4S and production of CysLTs in murine adipocyte 3T3-L1 cells and their underlying regulatory mechanisms. Expression of LTC4S and production of LTC4 and CysLTs increased during adipogenesis, whereas siRNA-mediated suppression of LTC4S expression repressed adipogenesis by reducing adipogenic gene expression. The CysLT1 receptor, one of the two LTC4 receptors, was expressed in adipocytes. LTC4 and LTD4 increased the intracellular triglyceride levels and adipogenic gene expression, and their enhancement was suppressed by co-treatment with pranlukast, a CysLT1 receptor antagonist. Moreover, the expression profiles of LTC4S gene/protein during adipogenesis resembled those of peroxisome proliferator-activated receptor (PPAR) γ. LTC4S expression was further upregulated by treatment with troglitazone, a PPARγ agonist. Promoter-luciferase and chromatin immunoprecipitation assays showed that PPARγ directly bound to the PPAR response element of the LTC4S gene promoter in adipocytes. These results indicate that the LTC4S gene expression was enhanced by PPARγ, and LTC4 and LTD4 activated adipogenesis through CysLT1 receptors in 3T3-L1 cells. Thus, LTC4S and CysLT1 receptors are novel potential targets for the treatment of obesity.


Subject(s)
Adipocytes/cytology , Adipogenesis , Glutathione Transferase/genetics , Leukotriene C4/pharmacology , Leukotriene D4/pharmacology , PPAR gamma/metabolism , Receptors, Leukotriene/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Gene Expression Regulation , Glutathione Transferase/metabolism , Mice , PPAR gamma/genetics , Promoter Regions, Genetic , Receptors, Leukotriene/genetics
2.
Neuroscience ; 422: 99-118, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31726033

ABSTRACT

Microglia activation plays a key role in regulating inflammatory and immune reaction during cerebral ischemia and it exerts pro-inflammatory or anti-inflammatory effect depending on M1/M2 polarization phenotype. Cysteinyl leukotriene 2 receptor (CysLT2R) is a potent inflammatory mediator receptor, and involved in cerebral ischemic injury, but the mechanism of CysLT2R regulating inflammation and neuron damage remains unclear. Here, we found that LPS and CysLT2R agonist NMLTC4 significantly increased microglia proliferation and phagocytosis, up-regulated the mRNA expression of M1 polarization markers (IL-1ß, TNF-α, IFN-γ, CD86 and iNOS), down-regulated the expression of M2 polarization markers (Arg-1, CD206, TGF-ß, IL-10, Ym-1) and increased the release of IL-1ß and TNF-α. CysLT2R selective antagonist HAMI3379 could antagonize these effects. IL-4 significantly up-regulated the mRNA expression of M2 polarization markers, and HAMI3379 further increased IL-4-induced up-regulation of M2 polarization markers expression. Additionally, LPS and NMLTC4 stimulated NF-κB p50 and p65 proteins expression, and promoted p50 transfer to the nucleus. Pre-treatment with HAMI3379 and NF-κB signaling inhibitor Bay 11-7082 could reverse the up-regulation of p50 and p65 proteins expression, and inhibited p50 transfer to the nucleus. The conditional medium of BV-2 cells contained HAMI3379 could inhibit SH-SY5Y cells apoptosis induced by LPS and NMLTC4. These results were further confirmed in primary microglia. The findings indicate that CysLT2R was involved in inflammation and neuronal damage by inducing the activation of microglia M1 polarization and NF-κB pathway, inhibiting microglia M1 polarization and promoting microglia polarization toward M2 phenotype which may exerts neuroprotective effects, and targeting CysLT2R may be a new therapeutic strategy against cerebral ischemia stroke.


Subject(s)
Cell Polarity/physiology , Inflammation/physiopathology , Microglia/physiology , NF-kappa B/physiology , Neurons/pathology , Receptors, Leukotriene/physiology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cyclohexanecarboxylic Acids/pharmacology , Down-Regulation/drug effects , Humans , Inflammation Mediators/metabolism , Leukotriene C4/analogs & derivatives , Leukotriene C4/antagonists & inhibitors , Leukotriene C4/pharmacology , Lipopolysaccharides/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , NF-kappa B p50 Subunit/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Nitriles/pharmacology , Phagocytosis/drug effects , Phthalic Acids/pharmacology , Primary Cell Culture , Rats , Receptors, Leukotriene/agonists , Receptors, Leukotriene/drug effects , Signal Transduction/physiology , Sulfones/pharmacology , Transcription Factor RelA/biosynthesis , Up-Regulation/drug effects
3.
Prostaglandins Other Lipid Mediat ; 143: 106336, 2019 08.
Article in English | MEDLINE | ID: mdl-31112752

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are expressed in bovine uterus, and their agonists are arachidonic acid (AA) metabolites. We hypothesised that silencing of PPAR genes in bovine endometrial stromal cells (ESC) would change the intracellular signalling through PPAR and affect apoptosis after cell treatment with different AA metabolites. The study's aims are detection of apoptosis and examining the influence of prostaglandins and leukotrienes on apoptosis occurring in physiological ESC and cells with silenced PPAR (α, δ, and γ) genes. Silencing the PPARα and PPARδ genes in cells resulted in increased DNA fragmentation and mRNA and protein expression of caspase (CASP) -3 and -8 (P < 0.05). Neither DNA fragmentation nor the mRNA and protein expression of CASP3 and -8 in cells with silenced PPARγ gene were changed compared to physiological cells (P > 0.05). Among PPARs, PPARα and PPARδ appear to inhibit apoptosis, and AA metabolites, as PPAR agonists, modify this process in bovine ESC.


Subject(s)
Apoptosis/genetics , Arachidonic Acid/metabolism , Endometrium/cytology , Gene Silencing , Peroxisome Proliferator-Activated Receptors/deficiency , Peroxisome Proliferator-Activated Receptors/genetics , Animals , Apoptosis/drug effects , Cattle , Dinoprost/pharmacology , Dinoprostone/pharmacology , Female , Leukotriene B4/pharmacology , Leukotriene C4/pharmacology
4.
Oncotarget ; 8(21): 35033-35047, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28402256

ABSTRACT

Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths worldwide. Cyclooxygenase-2, which plays a key role in the biosynthesis of prostaglandin E2 (PGE2), is often up-regulated in CRC and in other types of cancer. PGE2 induces angiogenesis and tumor cell survival, proliferation and migration. The tumor suppressor 15-hydroxyprostaglandin dehydrogenase (15-PGDH) is a key enzyme in PGE2 catabolism, converting it into its inactive metabolite 15-keto-PGE2, and is often down-regulated in cancer. Interestingly, CRC patients expressing high levels of the cysteinyl leukotriene 2 (CysLT2) receptor have a good prognosis; therefore, we investigated a potential link between CysLT2 signaling and the tumor suppressor 15-PGDH in colon cancer cells.We observed a significant up-regulation of 15-PGDH after treatment with LTC4, a CysLT2 ligand, in colon cancer cells at both the mRNA and protein levels, which could be reduced by a CysLT2 antagonist or a JNK inhibitor. LTC4 induced 15-PGDH promoter activity via JNK/AP-1 phosphorylation. Furthermore, we also observed that LTC4, via the CysLT2/JNK signaling pathway, increased the expression of the differentiation markers sucrase-isomaltase and mucin-2 in colon cancer cells and that down-regulation of 15-PGDH totally abolished the observed increase in these markers.In conclusion, the restoration of 15-PGDH expression through CysLT2 signaling promotes the differentiation of colon cancer cells, indicating an anti-tumor effect of CysLT2 signaling.


Subject(s)
Colonic Neoplasms/enzymology , Enzyme Activation , Hydroxyprostaglandin Dehydrogenases/metabolism , Leukotriene C4/pharmacology , Caco-2 Cells , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Receptors, Leukotriene/metabolism , Signal Transduction/drug effects
5.
Eur J Pharmacol ; 794: 147-153, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-27887950

ABSTRACT

CysLT1 receptors are known to be involved in the pathogenesis of asthma. However, the functional roles of CysLT2 receptors in this condition have not been determined. The purpose of this study is to develop an experimental model of CysLT2 receptor-mediated LTC4-induced lung air-trapping in guinea pigs and use this model to clarify the mechanism underlying response to such trapping. Because LTC4 is rapidly converted to LTD4 by γ-glutamyltranspeptidase (γ-GTP) under physiological conditions, S-hexyl GSH was used as a γ-GTP inhibitor. In anesthetized artificially ventilated guinea pigs with no S-hexyl GSH treatment, i.v. LTC4-induced bronchoconstriction was almost completely inhibited by montelukast, a CysLT1 receptor antagonist, but not by BayCysLT2RA, a CysLT2 receptor antagonist. The inhibitory effect of montelukast was diminished by treatment with S-hexyl GSH, whereas the effect of BayCysLT2RA was enhanced with increasing dose of S-hexyl GSH. Macroscopic and histological examination of lung tissue isolated from LTC4-/S-hexyl-GSH-treated guinea pigs revealed air-trapping expansion, particularly at the alveolar site. Inhaled LTC4 in conscious guinea pigs treated with S-hexyl GSH increased both airway resistance and airway hyperinflation. On the other hand, LTC4-induced air-trapping was only partially suppressed by treatment with the bronchodilator salmeterol. Although montelukast inhibition of LTC4-induced air-trapping was weak, treatment with BayCysLT2RA resulted in complete suppression of this air-trapping. Furthermore, BayCysLT2RA completely suppressed LTC4-induced airway vascular hyperpermeability. In conclusion, we found in this study that CysLT2 receptors mediate LTC4-induced bronchoconstriction and air-trapping in S-hexyl GSH-treated guinea pigs. It is therefore believed that CysLT2 receptors contribute to asthmatic response involving air-trapping.


Subject(s)
Air , Leukotriene C4/pharmacology , Lung/drug effects , Lung/physiology , Receptors, Leukotriene/metabolism , Animals , Bronchoconstriction/drug effects , Cyclohexanecarboxylic Acids/pharmacology , Glutathione/analogs & derivatives , Glutathione/pharmacology , Guinea Pigs , Lung/metabolism , Male , Phthalic Acids/pharmacology , Respiration, Artificial , Salmeterol Xinafoate/pharmacology
6.
Mol Cell ; 64(4): 746-759, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863227

ABSTRACT

Excitation-transcription coupling, linking stimulation at the cell surface to changes in nuclear gene expression, is conserved throughout eukaryotes. How closely related coexpressed transcription factors are differentially activated remains unclear. Here, we show that two Ca2+-dependent transcription factor isoforms, NFAT1 and NFAT4, require distinct sub-cellular InsP3 and Ca2+ signals for physiologically sustained activation. NFAT1 is stimulated by sub-plasmalemmal Ca2+ microdomains, whereas NFAT4 additionally requires Ca2+ mobilization from the inner nuclear envelope by nuclear InsP3 receptors. NFAT1 is rephosphorylated (deactivated) more slowly than NFAT4 in both cytoplasm and nucleus, enabling a more prolonged activation phase. Oscillations in cytoplasmic Ca2+, long considered the physiological form of Ca2+ signaling, play no role in activating either NFAT protein. Instead, effective sustained physiological activation of NFAT4 is tightly linked to oscillations in nuclear Ca2+. Our results show how gene expression can be controlled by coincident yet geographically distinct Ca2+ signals, generated by a freely diffusible InsP3 message.


Subject(s)
Calcium Signaling , Calcium/metabolism , Inositol Phosphates/metabolism , NFATC Transcription Factors/genetics , Recombinant Fusion Proteins/genetics , Animals , Basophils/cytology , Basophils/drug effects , Basophils/metabolism , Bronchi/cytology , Bronchi/drug effects , Bronchi/metabolism , Cell Line , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Leukotriene C4/pharmacology , NFATC Transcription Factors/metabolism , Protein Transport , Rats , Recombinant Fusion Proteins/metabolism , Thapsigargin/pharmacology , Transcription, Genetic
7.
Biol Pharm Bull ; 39(4): 556-63, 2016.
Article in English | MEDLINE | ID: mdl-27040628

ABSTRACT

Lung allergic diseases sometimes accompany pulmonary vaso- and broncho-constriction. Rats are currently used for the experimental study of lung allergies. However, their hemodynamic mechanisms are not fully understood. Therefore the effects of allergic mediators were determined systematically in vivo in rats in terms of pulmonary vascular resistance (PVR), airway pressure (AWP) and total peripheral resistance (TPR). We directly measured pulmonary arterial pressure, left atrial pressure, systemic arterial pressure, central venous pressure and aortic blood flow to determine PVR and TPR, as well as AWP, following injections of platelet-activating factor (PAF), histamine, serotonin, leukotriene (LT) C4, and prostaglandin (PG) D2 in anesthetized open-chest artificially ventilated Sprague-Dawley (SD) rats. PVR was dose-dependently increased by consecutive administration of PAF, LTC4, and PGD2, with the maximal responsiveness being PAF>LTC4>PGD2. However, neither histamine nor serotonin changed PVR. TPR was decreased by all agents except LTC4 which actually increased it. PAF and serotonin, but not the other agents, increased AWP. In conclusion, allergic mediators exert non-uniform actions on pulmonary and systemic circulation and airways in anesthetized SD rats: PAF, LTC4 and PGD2, but not histamine or serotonin, caused substantial pulmonary vasoconstriction; LTC4 yielded systemic vasoconstriction, while the others caused systemic vasodilatation; only two mediators, PAF and serotonin, induce airway constriction.


Subject(s)
Blood Circulation/drug effects , Inflammation Mediators/pharmacology , Lung/drug effects , Vascular Resistance/drug effects , Anesthesia , Animals , Arterial Pressure/drug effects , Histamine/pharmacology , Hypersensitivity/physiopathology , Leukotriene C4/pharmacology , Lung/physiology , Male , Platelet Activating Factor/pharmacology , Prostaglandin D2/pharmacology , Rats, Sprague-Dawley , Serotonin/pharmacology , Vasoconstriction/drug effects
8.
J Immunol ; 194(11): 5061-8, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25904552

ABSTRACT

Cysteinyl leukotrienes (cysLTs) are bronchoconstricting lipid mediators that amplify eosinophilic airway inflammation by incompletely understood mechanisms. We recently found that LTC4, the parent cysLT, potently activates platelets in vitro and induces airway eosinophilia in allergen-sensitized and -challenged mice by a platelet- and type 2 cysLT receptor-dependent pathway. We now demonstrate that this pathway requires production of thromboxane A2 and signaling through both hematopoietic and lung tissue-associated T prostanoid (TP) receptors. Intranasal administration of LTC4 to OVA-sensitized C57BL/6 mice markedly increased the numbers of eosinophils in the bronchoalveolar lavage fluid, while simultaneously decreasing the percentages of eosinophils in the blood by a TP receptor-dependent mechanism. LTC4 upregulated the expressions of ICAM-1 and VCAM-1 in an aspirin-sensitive and TP receptor-dependent manner. Both hematopoietic and nonhematopoietic TP receptors were essential for LTC4 to induce eosinophil recruitment. Thus, the autocrine and paracrine functions of thromboxane A2 act downstream of LTC4/type 2 cysLT receptor signaling on platelets to markedly amplify eosinophil recruitment through pulmonary vascular adhesion pathways. The findings suggest applications for TP receptor antagonists in cases of asthma with high levels of cysLT production.


Subject(s)
Aspirin/pharmacology , Blood Platelets/immunology , Cysteine/immunology , Leukotriene C4/immunology , Leukotrienes/immunology , Platelet Activation/immunology , Allergens/immunology , Animals , Asthma/drug therapy , Asthma/immunology , Bone Marrow Transplantation , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Eosinophilia/blood , Eosinophilia/immunology , Inflammation/immunology , Intercellular Adhesion Molecule-1/biosynthesis , Leukotriene Antagonists/pharmacology , Leukotriene C4/pharmacology , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Thromboxane A2/biosynthesis , Thromboxane A2/immunology , Vascular Cell Adhesion Molecule-1/biosynthesis
9.
Prostaglandins Other Lipid Mediat ; 120: 115-25, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25839425

ABSTRACT

We evaluated the autocrine activities of cysteinyl leukotrienes (cysteinyl-LTs) in HUVEC and studied the signaling and the pharmacological profile of the CysLT2 receptor (CysLT2R) expressed by ECs, finally assessing the role of the CysLT2R in permeability alterations in a model of isolated brain. Cysteinyl-LTs and their precursor LTA4 contracted HUVEC and increased permeability to macromolecules, increasing the formation of stress fibers through the phosphorylation of myosin light-chain (MLC) following Rho and PKC activation. Accordingly, in an organ model of cerebral vasculature with an intact intima, neutrophils challenge leaded to significant formation of cysteinyl-LTs and edema. Pretreatment with a selective CysLT2R antagonist prevented cytoskeleton rearrangement and HUVEC contraction, along with edema formation in the brain preparation, while leaving the synthesis of cysteinyl-LTs unaffected. We also demonstrate here that the CysLT1R antagonist zafirlukast, pranlukast, pobilukast and iralukast also possess CysLT2R antagonistic activity, which could help in reconsidering previous data on the role of cysteinyl-LTs in the cardiovascular system. The results obtained are further supporting a potential role for CysLT2R in cardiovascular disease.


Subject(s)
Autocrine Communication , Cysteine/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Leukotrienes/metabolism , Receptors, Leukotriene/metabolism , Signal Transduction , Animals , Autocrine Communication/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Leukotriene A4/pharmacology , Leukotriene C4/pharmacology , Myosin Light Chains/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Permeability/drug effects , Phosphorylation/drug effects , Protein Kinase C/metabolism , Rats , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/metabolism , rho GTP-Binding Proteins/metabolism
10.
Life Sci ; 127: 66-72, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25744397

ABSTRACT

AIMS: Aconitum alkaloids mainly contain highly toxic aconitine (AC), mesaconitine (MA), and hypaconitine (HA) and less toxic benzoylaconine (BAC), benzoylmesaconine (BMA), benzoylhypaconine (BHA), aconine, mesaconine, and hypaconine. The efflux transporters including P-glycoprotein (P-gp), breast cancer resistance protein (BCRP) and multidrug resistance-associated protein 2 (MRP2) can efflux toxicants to prevent poisoning. Our previous publication has proved that P-gp and BCRP contributed to the efflux of AC, MA and HA, which is demonstrated in the human colonic adenocarcinoma cell lines (Caco-2 cells), Mardin-Darby canine kidney cell lines transfected with MDR1 or BCRP (MDR1-MDCKII and BCRP-MDCKII cells). However, the role of MRP2 remains uncertain. MAIN METHODS: The MRP2-MDCKII cells were used to determine the efflux ratios (Er) and intracellular amounts of Aconitum alkaloids. In addition, the importance of MRP2 was further investigated with or without the MRP2 inhibitor, LTC4. KEY FINDINGS: The Er values of AC, MA, HA, BAC, BMA and BHA in MRP2-MDCKII cells (6.4 ± 0.3, 5.9 ± 0.5, 2.2 ± 0.2, 1.6 ± 0.3, 1.7 ± 0.2 and 1.9 ± 0.2 respectively) were significantly higher than those in MDCKII cells, which were close to 1. In the presence of LTC4, the Er values of AC, MA, HA, BAC, BMA and BHA were reduced to approximately 1 and their intracellular amounts were also significantly increased in MRP2-MDCKII cells. SIGNIFICANCE: MRP2 was involved in the efflux of AC, MA, HA, BAC, BMA and BHA, which would be useful for the safe application of these components or their herbs.


Subject(s)
Aconitum/chemistry , Alkaloids/metabolism , Multidrug Resistance-Associated Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Biological Transport, Active/drug effects , Caco-2 Cells , Dogs , Humans , Leukotriene C4/pharmacology , Madin Darby Canine Kidney Cells , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Transfection
11.
Eur J Pharmacol ; 754: 98-104, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25704617

ABSTRACT

Cysteinyl leukotrienes act through G-protein-coupled receptors termed cysteinyl leukotriene 1 (CysLT1) and cysteinyl leukotriene 2 (CysLT2) receptors. However, little is known about the pathophysiological role of CysLT2 receptors in asthma. To elucidate the possible involvement of CysLT2 receptors in bronchoconstriction and airway vascular hyperpermeability, we have established a novel guinea pig model of asthma. In vitro study confirmed that CHO-K1 cells, expressing guinea pig CysLT2 and CysLT1 receptors are selectively stimulated by LTC4 and LTD4, respectively. However, when LTC4 was intravenously injected to guinea pigs, the resulting bronchoconstriction was fully abrogated by montelukast, a CysLT1 receptor antagonist, indicating rapid metabolism of LTC4 to LTD4 in the lung. We found that treatment with S-hexyl glutathione (S-hexyl GSH), an inhibitor of gamma-glutamyl transpeptidase, significantly increased LTC4 content and LTC4/(LTD4 plus LTE4) ratio in the lung. Under these circumstances, LTC4-induced bronchoconstriction became resistant to montelukast, but sensitive to Compound A, a CysLT2 receptor antagonist, depending on the dose of S-hexyl GSH. Combination with montelukast and Compound A completely abrogated this spasmogenic response. Additionally, we confirmed that LTC4 elicits airway vascular hyperpermeability via CysLT2 receptors in the presence of high dose of S-hexyl GSH as evidenced by complete inhibition of LTC4-induced hyperpermeability by Compound A, but not montelukast. These results suggest that CysLT2 receptors mediate bronchoconstriction and airway vascular hyperpermeability in guinea pigs and that the animal model used in this study may be useful to elucidate the functional role of CysLT2 receptors in various diseases, including asthma.


Subject(s)
Bronchoconstriction/drug effects , Capillary Permeability/drug effects , Glutathione/analogs & derivatives , Leukotriene C4/pharmacology , Receptors, Leukotriene/physiology , Acetates/pharmacology , Animals , Bronchoconstriction/physiology , Calcium/pharmacology , Capillary Permeability/physiology , Cyclopropanes , Dose-Response Relationship, Drug , Drug Interactions , Glutathione/pharmacology , Guinea Pigs , Leukotriene C4/antagonists & inhibitors , Leukotriene C4/pharmacokinetics , Leukotriene D4/pharmacology , Lung/metabolism , Male , Quinolines/pharmacology , Receptors, Leukotriene/agonists , Receptors, Leukotriene/drug effects , Sulfides , Triazoles/pharmacology
12.
Theriogenology ; 83(8): 1328-37, 2015 May.
Article in English | MEDLINE | ID: mdl-25707327

ABSTRACT

The effect of leukotriene (LT) C4 (LTC4) and LTD4 on the contractility of an inflamed porcine uterus was investigated. On Day 3 of the estrous cycle (Day 0 of the study), either saline or Escherichia coli suspension was injected into each uterine horn. Although acute endometritis developed in all bacteria-inoculated gilts, a severe acute endometritis was noted more often on Day 8 than on Day 16. Myometrial and endometrial/myometrial strips were incubated with LTC4 or LTD4 alone, or together with a cysteinyl-LT receptor antagonist (BAY-u9773). Leukotriene C4 increased the contraction intensity in the saline- and bacteria-treated uteri on Day 8; however, its effect was lower in the myometrium of inflamed uteri. Contraction frequency was found to decrease in the saline-treated uteri as opposed to inflamed ones, in which it was elevated. On Day 16, contraction intensity increased in response to LTC4 in the saline-treated uteri but was reduced in the inflamed organs. The value of this parameter was lower in the inflamed uteri than that in the saline-treated ones. Leukotriene D4 (Days 8 and 16) augmented contractility in the saline-treated uteri, but despite increasing its intensity in the inflamed organs, it decreased contraction frequency. Leukotriene C4 or LTD4, added to BAY-u9773-pretreated saline- and bacteria-treated uteri on both days, decreased the contraction intensity. On Day 16 after treatment with BAY-u9773 and LTC4, contraction intensity in the endometrium/myometrium of the inflamed uteri was lower than that in the saline-treated organs. Data show that both LTC4 and LTD4 affect the contractility of inflamed porcine uteri, though LTC4 exerts a weaker contractile effect.


Subject(s)
Endometritis/veterinary , Leukotriene C4/pharmacology , Leukotriene D4/pharmacology , Swine Diseases/physiopathology , Uterine Contraction/drug effects , Animals , Endometritis/microbiology , Endometritis/physiopathology , Endometrium/physiopathology , Escherichia coli , Escherichia coli Infections/physiopathology , Escherichia coli Infections/veterinary , Female , Leukotriene C4/antagonists & inhibitors , Myometrium/physiopathology , SRS-A/analogs & derivatives , SRS-A/pharmacology , Sus scrofa , Swine
13.
J Biol Chem ; 289(51): 35314-25, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25371198

ABSTRACT

Calcium/voltage-gated, large conductance potassium (BK) channels control numerous physiological processes, including myogenic tone. BK channel regulation by direct interaction between lipid and channel protein sites has received increasing attention. Leukotrienes (LTA4, LTB4, LTC4, LTD4, and LTE4) are inflammatory lipid mediators. We performed patch clamp studies in Xenopus oocytes that co-expressed BK channel-forming (cbv1) and accessory ß1 subunits cloned from rat cerebral artery myocytes. Leukotrienes were applied at 0.1 nm-10 µm to either leaflet of cell-free membranes at a wide range of [Ca(2+)]i and voltages. Only LTB4 reversibly increased BK steady-state activity (EC50 = 1 nm; Emax reached at 10 nm), with physiological [Ca(2+)]i and voltages favoring this activation. Homomeric cbv1 or cbv1-ß2 channels were LTB4-resistant. Computational modeling predicted that LTB4 docked onto the cholane steroid-sensing site in the BK ß1 transmembrane domain 2 (TM2). Co-application of LTB4 and cholane steroid did not further increase LTB4-induced activation. LTB4 failed to activate ß1 subunit-containing channels when ß1 carried T169A, A176S, or K179I within the docking site. Co-application of LTB4 with LTA4, LTC4, LTD4, or LTE4 suppressed LTB4-induced activation. Inactive leukotrienes docked onto a portion of the site, probably preventing tight docking of LTB4. In summary, we document the ability of two endogenous lipids from different chemical families to share their site of action on a channel accessory subunit. Thus, cross-talk between leukotrienes and cholane steroids might converge on regulation of smooth muscle contractility via BK ß1. Moreover, the identification of LTB4 as a highly potent ligand for BK channels is critical for the future development of ß1-specific BK channel activators.


Subject(s)
Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Leukotriene B4/metabolism , Animals , Calcium/metabolism , Cerebral Arteries/cytology , Female , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Leukotriene A4/chemistry , Leukotriene A4/metabolism , Leukotriene A4/pharmacology , Leukotriene B4/chemistry , Leukotriene B4/pharmacology , Leukotriene C4/chemistry , Leukotriene C4/metabolism , Leukotriene C4/pharmacology , Leukotriene D4/chemistry , Leukotriene D4/metabolism , Leukotriene D4/pharmacology , Leukotriene E4/chemistry , Leukotriene E4/metabolism , Leukotriene E4/pharmacology , Membrane Potentials/drug effects , Microinjections , Models, Molecular , Molecular Structure , Muscle Cells/cytology , Muscle Cells/metabolism , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Patch-Clamp Techniques , Protein Binding , Protein Structure, Tertiary , RNA, Complementary/administration & dosage , RNA, Complementary/genetics , Rats , Xenopus laevis
14.
J Gen Physiol ; 143(3): 345-59, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24567509

ABSTRACT

Orai proteins contribute to Ca(2+) entry into cells through both store-dependent, Ca(2+) release-activated Ca(2+) (CRAC) channels (Orai1) and store-independent, arachidonic acid (AA)-regulated Ca(2+) (ARC) and leukotriene C4 (LTC4)-regulated Ca(2+) (LRC) channels (Orai1/3 heteromultimers). Although activated by fundamentally different mechanisms, CRAC channels, like ARC and LRC channels, require stromal interacting molecule 1 (STIM1). The role of endoplasmic reticulum-resident STIM1 (ER-STIM1) in CRAC channel activation is widely accepted. Although ER-STIM1 is necessary and sufficient for LRC channel activation in vascular smooth muscle cells (VSMCs), the minor pool of STIM1 located at the plasma membrane (PM-STIM1) is necessary for ARC channel activation in HEK293 cells. To determine whether ARC and LRC conductances are mediated by the same or different populations of STIM1, Orai1, and Orai3 proteins, we used whole-cell and perforated patch-clamp recording to compare AA- and LTC4-activated currents in VSMCs and HEK293 cells. We found that both cell types show indistinguishable nonadditive LTC4- and AA-activated currents that require both Orai1 and Orai3, suggesting that both conductances are mediated by the same channel. Experiments using a nonmetabolizable form of AA or an inhibitor of 5-lipooxygenase suggested that ARC and LRC currents in both cell types could be activated by either LTC4 or AA, with LTC4 being more potent. Although PM-STIM1 was required for current activation by LTC4 and AA under whole-cell patch-clamp recordings in both cell types, ER-STIM1 was sufficient with perforated patch recordings. These results demonstrate that ARC and LRC currents are mediated by the same cellular populations of STIM1, Orai1, and Orai3, and suggest a complex role for both ER-STIM1 and PM-STIM1 in regulating these store-independent Orai1/3 channels.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Calcium/metabolism , Membrane Glycoproteins/metabolism , Action Potentials , Animals , Arachidonic Acid/pharmacology , Cell Membrane/metabolism , Cells, Cultured , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Leukotriene C4/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , ORAI1 Protein , Rats , Stromal Interaction Molecule 1
15.
Chin J Integr Med ; 19(5): 380-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23494327

ABSTRACT

OBJECTIVE: Cheongseoikki-tang (CIT, Korean), also called Qingshu Yiqi decoction () and Seisho-ekki-to (Japanese), is well known as an effective traditional combination of herbs for treating cardiovascular diseases. This study was to research its effects on bone marrow-derived mast cell (BMMC)-mediated allergy and inflammation mechanisms. METHODS: In this study, the biological effect of Cheongseoikki-tang ethanol extract (CITE) was evaluated, focusing on its effects on the production of allergic mediators by phorbol 12-myristate 13-acetate (PMA) plus calcium ionophore A23187 (A23187)-stimulated BMMCs. These allergic mediators included interleukin-6 (IL-6), prostaglandin D2 (PGD2), leukotriene C4 (LTC4), and ß-hexosaminidase (ß-hex). RESULTS: Our data revealed that CITE inhibited the production of IL-6, PGD2, LTC4, and ß-hex induced by PMA plus A23187 (P<0.05). CONCLUSION: These findings indicate that CITE has the potential for use in the treatment of allergy.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Bone Marrow Cells/pathology , Drugs, Chinese Herbal/therapeutic use , Hypersensitivity/drug therapy , Mast Cells/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Calcimycin/pharmacology , Cell Degranulation/drug effects , Cell Survival/drug effects , Drugs, Chinese Herbal/pharmacology , Hypersensitivity/pathology , Interleukin-6/metabolism , Leukotriene C4/pharmacology , Male , Mast Cells/drug effects , Mast Cells/physiology , Mice , Mice, Inbred BALB C , Prostaglandin D2/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology , beta-N-Acetylhexosaminidases/metabolism
16.
Respirology ; 18(4): 674-81, 2013 May.
Article in English | MEDLINE | ID: mdl-23432979

ABSTRACT

BACKGROUND AND OBJECTIVE: Synthesis of cysteinyl leukotrienes (cys-LT) is thought to cause inflammatory disorders such as bronchial asthma and allergic rhinitis. Recent reports have suggested that leukotriene C4 (LTC4 ) is an important regulator of pulmonary fibrosis. This study examined the effect of LTC4 in LTC4 synthase-overexpressed transgenic mice with bleomycin-induced pulmonary fibrosis. The function of lung-derived fibroblasts from transgenic mice was also investigated. METHODS: Bleomycin was administrated to transgenic mice and wild-type (WT) mice by intratracheal instillation. Concentrations of interleukin (IL)-4 and -13, interferon-γ, and transforming growth factor (TGF)-ß1 in bronchoalveolar lavage fluid were measured 1, 3, 7 and 14 days after the administration of bleomycin. Lung tissue was examined histopathologically on day 14. In addition, lung-derived fibroblasts from transgenic and WT mice were cultured for 7 days. Expression of TGF-ß1 mRNA was measured by real-time polymerase chain reaction. RESULTS: Both the pathological scores for pulmonary fibrosis (3.8 ± 0.4 vs 2.0 ± 0.1, P < 0.05) and the levels of IL-4 (12.1 ± 2.3 vs <7.8 pg/mL, P < 0.05), IL-13 (26.5 ± 5.2 vs <7.8 pg/mL, P < 0.01) and TGF-ß1 (211.1 ± 30.2 vs 21.3 ± 1.2 pg/mL, P < 0.01) on day 14 were significantly greater in transgenic than in WT mice. Furthermore, the reduction of LTC4 by pranlukast hydrate, a cys-LT1 receptor antagonist, in fibroblasts from transgenic significantly (P < 0.05) decreased the expression of TGF-ß1 mRNA (by ∼50%) compared with those from WT mice. CONCLUSIONS: Overexpression of LTC4 , amplifies bleomycin-induced pulmonary fibrosis in mice. Our findings suggest a role for LTC4 in lung fibrosis.


Subject(s)
Bleomycin/adverse effects , Leukotriene C4/adverse effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Glutathione Transferase/deficiency , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Interferon-gamma/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Leukotriene C4/pharmacology , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Pulmonary Fibrosis/pathology
17.
BMC Vet Res ; 8: 160, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-22963524

ABSTRACT

BACKGROUND: Glucocorticoids have been proven to be effective in the therapy of recurrent airway obstruction (RAO) in horses via systemic as well as local (inhalative) administration. Elective analysis of the effects of this drug on bronchoconstriction in viable lung tissue offers an insight into the mechanism of action of the inflammatory cascade occurring during RAO which is still unclear. The mechanism of action of steroids in treatment of RAO is thought to be induced through classical genomic pathways. We aimed at electively studying the effects of the glucocorticoid beclomethasone dipropionate on equine precision-cut lung slices (PCLS).PCLS were used to analyze ex-vivo effects of beclomethasone on inhibiting bronchoconstriction in the horse. The inhibiting effect was measured through instillation of a known mediator of inflammation and bronchoconstriction, leukotriene C4. For this, the accessory lobes of 13 horses subjected to euthanasia for reasons unrelated to the respiratory apparatus were used to obtain viable lung slices. RESULTS: After 30 minutes of PCLS incubation, beclomethasone showed to significantly inhibit the contraction of the bronchioles after instillation with leukotriene C4. The EC50 values of the two contraction curves (LTC4 with and without BDP) differed significantly from each other (p = 0.002). The possibility of a non-genomic rapid mechanism of action seems likely since transcriptional activities require a longer lag period. CONCLUSIONS: In human neuroendocrinology, high levels of glucocorticoids have been proven to function via a non-genomic mechanism of membrane receptors. The concentration of beclomethasone used on the lung slices in our study can be considered as high. This allows speculation about similar rapid non-genomic mechanisms of high-dosage inhaled glucocorticoids in the lower airways of horses. However, further assessment on a molecular basis is needed to confirm this.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Beclomethasone/pharmacology , Bronchoconstriction/drug effects , Horses , Leukotriene C4/pharmacology , Lung/drug effects , Animals , Female , Lung/physiology , Male
18.
Toxicol Appl Pharmacol ; 265(3): 316-24, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22982073

ABSTRACT

Emodin is a poorly bioavailable but promising plant-derived anticancer drug candidate. The low oral bioavailability of emodin is due to its extensive glucuronidation in the intestine and liver. Caco-2 cell culture model was used to investigate the interplay between UDP-glucuronosyltransferases (UGTs) and efflux transporters in the intestinal disposition of emodin. Bidirectional transport assays of emodin at different concentrations were performed in the Caco-2 monolayers with or without multidrug resistance-associated protein (MRP) and breast cancer resistance protein (BCRP) efflux transporter chemical inhibitors. The bidirectional permeability of emodin and its glucuronide in the Caco-2 monolayers was determined. Emodin was rapidly metabolized to emodin glucuronide in Caco-2 cells. LTC4, a potent inhibitor of MRP2, decreased the efflux of emodin glucuronide and also substantially increased the intracellular glucuronide level in the basolateral-to-apical (B-A) direction. MK-571, chemical inhibitor of MRP2, MRP3, and MRP4, significantly reduced the efflux of glucuronide in the apical-to-basolateral (A-B) and B-A directions in a dose-dependent manner. However, dipyridamole, a BCRP chemical inhibitor demonstrated no effect on formation and efflux of emodin glucuronide in Caco-2 cells. In conclusion, UGT is a main metabolic pathway for emodin in the intestine, and the MRP family is composed of major efflux transporters responsible for the excretion of emodin glucuronide in the intestine. The coupling of UGTs and MRP efflux transporters causes the extensive metabolism, excretion, and low bioavailability of emodin.


Subject(s)
Emodin/pharmacokinetics , Glucuronosyltransferase/metabolism , Intestinal Mucosa/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Biological Availability , Biological Transport , Caco-2 Cells , Humans , Intestinal Absorption , Intestines/enzymology , Leukotriene C4/pharmacology , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Propionates/pharmacology , Quinolines/pharmacology
19.
Int Arch Allergy Immunol ; 159(4): 422-7, 2012.
Article in English | MEDLINE | ID: mdl-22846852

ABSTRACT

BACKGROUND: Leukotrienes are potent inflammatory mediators which modulate immune responses and induce bronchoconstriction in susceptible individuals. Montelukast (MK) is a leukotriene receptor (CysLT1) antagonist that has been shown to prevent exacerbation of asthma. Considering the plethora of potential cellular targets for MK, specific mechanisms for its therapeutic action are still not fully understood. In vitro, we determined whether human dendritic cell function could be affected by leukotriene C(4) (LTC(4)) treatment and whether MK had potential in modulating this response. We also studied the effect of LTC(4) in the context of response to an airway virus (respiratory syncytial virus, RSV). METHODS: Human monocyte-derived dendritic cells (moDCs) exposed to LTC(4), MK, or both, were cocultured with autologous T cells, with or without RSV. The effects of LTC(4) and MK on cell function were determined by ELISA and proliferation assays. RESULTS: Both moDCs and their precursors--monocytes--express LTC(4) receptor CysLT1, making them potential targets for MK. moDCs cultured with LTC(4) release the eosinophil chemoattractant RANTES (CCL5) and induce greater T cell proliferation. Both were blocked by the presence of MK. MK treatment, albeit anti-inflammatory, did not interfere with the moDC-dependent T cell-proliferative responses induced by RSV. CONCLUSIONS: LTC(4), chronically present in the airways of asthma patients, could induce an exaggerated inflammatory response to airway infection via dendritic cell activation, which would be prevented by MK. Our study provides additional insight into the mechanisms of action of this leukotriene receptor antagonist.


Subject(s)
Acetates/pharmacology , Anti-Asthmatic Agents/pharmacology , Dendritic Cells/drug effects , Leukotriene C4/immunology , Quinolines/pharmacology , T-Lymphocytes/drug effects , Asthma/drug therapy , Asthma/immunology , Asthma/pathology , Cell Proliferation/drug effects , Chemokine CCL5/immunology , Chemokine CCL5/metabolism , Coculture Techniques , Cyclopropanes , Dendritic Cells/immunology , Dendritic Cells/virology , Humans , Leukotriene C4/metabolism , Leukotriene C4/pharmacology , Primary Cell Culture , Receptors, Leukotriene/immunology , Receptors, Leukotriene/metabolism , Respiratory Syncytial Viruses/immunology , Sulfides , T-Lymphocytes/immunology , T-Lymphocytes/virology
20.
Proc Natl Acad Sci U S A ; 109(18): 6969-74, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22509043

ABSTRACT

Stimulation of cells with physiological concentrations of calcium-mobilizing agonists often results in the generation of repetitive cytoplasmic Ca(2+) oscillations. Although oscillations arise from regenerative Ca(2+) release, they are sustained by store-operated Ca(2+) entry through Ca(2+) release-activated Ca(2+) (CRAC) channels. Here, we show that following stimulation of cysteinyl leukotriene type I receptors in rat basophilic leukemia (RBL)-1 cells, large amplitude Ca(2+) oscillations, CRAC channel activity, and downstream Ca(2+)-dependent nuclear factor of activated T cells (NFAT)-driven gene expression are all exclusively maintained by the endoplasmic reticulum Ca(2+) sensor stromal interaction molecule (STIM) 1. However, stimulation of tyrosine kinase-coupled FCεRI receptors evoked Ca(2+) oscillations and NFAT-dependent gene expression through recruitment of both STIM2 and STIM1. We conclude that different agonists activate different STIM proteins to sustain Ca(2+) signals and downstream responses.


Subject(s)
Calcium Signaling/drug effects , Calcium Signaling/genetics , Animals , Calcium Channels/metabolism , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Gene Expression/drug effects , Gene Knockdown Techniques , Leukotriene C4/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , NFATC Transcription Factors/metabolism , Rats , Receptors, Leukotriene/metabolism , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2
SELECTION OF CITATIONS
SEARCH DETAIL
...