Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Heliyon ; 9(11): e21408, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38027705

ABSTRACT

In the past decade, there has been increasing interest in use of small molecules for immunomodulation. The affinity-based pull-down purification is an essential tool for target identification of small molecules and drug discovery. This study presents our recent efforts to investigate the cellular target(s) of Compound A, a small molecule with demonstrated immunomodulatory properties in human peripheral blood mononuclear cells (PBMCs). While we have previously observed the immunomodulatory activity of Compound A in PBMCs, the specific molecular targets underlying its effects remains elusive. To address this challenge, we synthesized a trifluoromethyl phenyl diazirine (TPD)-bearing trifunctional Probe 1 based on the chemical structure of Compound A, which could be used in a pull-down assay to efficiently bind to putative cellular targets via photoaffinity labelling. In this report, we utilized bovine serum albumin (BSA) as a model protein to establish a proof-of-concept in order to assess the suitability of Probe 1 for binding to an endogenous target. By the successful synthesis of Probe 1 and demonstrating the efficient binding of Probe 1 to BSA, we propose that this method can be used as a tool for further identification of potential protein targets of small molecules in living cells. Our findings provide a valuable starting point for further investigations into the molecular mechanisms underlying the immunomodulatory effects of Compound A.

2.
Comput Biol Med ; 152: 106442, 2023 01.
Article in English | MEDLINE | ID: mdl-36566625

ABSTRACT

SHP2 (Src homology-2 domain-containing protein tyrosine phosphatase-2) is a cytoplasmic protein -tyrosine phosphatase encoded by the gene PTPN11. It plays a crucial role in regulating cell growth and differentiation. Specifically, SHP2 is an oncoprotein associated with developmental pathologies and several different cancer types, including gastric, leukemia and breast cancer and is of great therapeutic interest. Given these roles, current research efforts have focused on developing SHP2 inhibitors. Allosteric SHP2 inhibitors have been shown to be more selective and pharmacologically appealing compared to competitive catalytic inhibitors targeting SHP2. Nevertheless, there remains a need for novel allosteric inhibitor scaffolds targeting SHP2 to develop compounds with improved selectivity, cell permeability, and bioavailability. Towards this goal, this study applied various computational tools to screen over 6 million compounds against the allosteric site within SHP2. The top-ranked hits from our in-silico screening were validated using protein thermal shift and biolayer interferometry assays, revealing three potent compounds. Kinetic binding assays were employed to measure the binding affinities of the top-ranked compounds and demonstrated that they all bind to SHP2 with a nanomolar affinity. Hence the compounds and the computational workflow described herein provide an effective approach for identifying and designing a generation of improved allosteric inhibitors of SHP2.


Subject(s)
Breast Neoplasms , Enzyme Inhibitors , Humans , Female , Molecular Docking Simulation , Enzyme Inhibitors/pharmacology , Cell Proliferation , Cell Differentiation
3.
Expert Opin Ther Targets ; 25(5): 347-363, 2021 05.
Article in English | MEDLINE | ID: mdl-34056985

ABSTRACT

Introduction:T cell functions are altered during chronic viral infections and tumor development. This is mainly manifested by significant changes in T cells' epigenetic and metabolic landscapes, pushing them into an 'exhausted' state. Reversing this T cell exhaustion has been emerging as a 'game-changing' therapeutic approach against cancer and chronic viral infection.Areas covered:This review discusses the cellular pathways related to T cell exhaustion, and the clinical development and possible cellular targets that can be exploited therapeutically to reverse this exhaustion. We searched various databases (e.g. Google Scholar, PubMed, Elsevier, and other scientific database sites) using the keywords T cell exhaustion, T cell activation, co-inhibitory receptors, and reversing T cell exhaustion.Expert opinion:The discovery of the immune checkpoints pathways represents a significant milestone toward understanding and reversing T cell exhaustion. Antibodies that target these pathways have already demonstrated promising activities in reversing T cell exhaustion. Nevertheless, there are still many associated limitations. In this context, next-generation alternatives are on the horizon. This includes the use of small molecules to block the immune checkpoints' receptors, combining them with other treatments, and identifying novel, safer and more effective immunotherapeutic targets.


Subject(s)
Neoplasms/therapy , T-Lymphocytes/immunology , Virus Diseases/therapy , Animals , Epigenesis, Genetic , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy/methods , Molecular Targeted Therapy , Neoplasms/immunology , Virus Diseases/immunology
4.
Sci Rep ; 10(1): 16262, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004839

ABSTRACT

Off-target interactions of drugs with the human ether-à-go-go related gene 1 (hERG1) channel have been associated with severe cardiotoxic conditions leading to the withdrawal of many drugs from the market over the last decades. Consequently, predicting drug-induced hERG-liability is now a prerequisite in any drug discovery campaign. Understanding the atomic level interactions of drug with the channel is essential to guide the efficient development of safe drugs. Here we utilize the recent cryo-EM structure of the hERG channel and describe an integrated computational workflow to characterize different drug-hERG interactions. The workflow employs various structure-based approaches and provides qualitative and quantitative insights into drug binding to hERG. Our protocol accurately differentiated the strong blockers from weak and revealed three potential anchoring sites in hERG. Drugs engaging in all these sites tend to have high affinity towards hERG. Our results were cross-validated using a fluorescence polarization kit binding assay and with electrophysiology measurements on the wild-type (WT-hERG) and on the two hERG mutants (Y652A-hERG and F656A-hERG), using the patch clamp technique on HEK293 cells. Finally, our analyses show that drugs binding to hERG disrupt and hijack certain native-structural networks in the channel, thereby, gaining more affinity towards hERG.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Computational Biology/methods , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/drug effects , Ether-A-Go-Go Potassium Channels/genetics , HEK293 Cells , Humans , Patch-Clamp Techniques , Structure-Activity Relationship
5.
Sci Rep ; 9(1): 12392, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31455818

ABSTRACT

Blockade of the programmed cell death 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) interaction has emerged as a powerful strategy in cancer immunotherapy. Recently, there have been enormous efforts to develop potent PD-1/PD-L1 inhibitors. In particular, Bristol-Myers Squibb (BMS) and Aurigene Discovery Technologies have individually disclosed several promising PD-1/PD-L1 inhibitors, whose detailed experimental data are not publicly disclosed. In this work, we report the rigorous and systematic in vitro characterization of a selected set of potent PD-1/PD-L1 macrocyclic peptide (BMSpep-57) and small-molecule inhibitors (BMS-103, BMS-142) from BMS and a peptidomimetic small-molecule inhibitor from Aurigene (Aurigene-1) using a series of biochemical and cell-based assays. Our results confirm that BMS-103 and BMS-142 are strongly active in biochemical assays; however, their acute cytotoxicity greatly compromised their immunological activity. On the other hand, Aurigene-1 did not show any activity in both biochemical and immunological assays. Furthermore, we also report the discovery of a small-molecule immune modulator, whose mode-of-action is not clear; however, it exhibits favorable drug-like properties and strong immunological activity. We hope that the results presented here will be useful in guiding the development of next-generation PD-1/PD-L1 small molecule inhibitors.


Subject(s)
B7-H1 Antigen/metabolism , Small Molecule Libraries/metabolism , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/chemistry , B7-H1 Antigen/genetics , Binding Sites , Cell Survival/drug effects , Genes, Reporter , Humans , Immunoassay , Interleukin-2/metabolism , Jurkat Cells , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Molecular Dynamics Simulation , Peptidomimetics , Protein Binding , Protein Structure, Tertiary , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
6.
Antiviral Res ; 171: 104566, 2019 11.
Article in English | MEDLINE | ID: mdl-31348951

ABSTRACT

As a part of innate immune defense, the role of mast cells during viral replication has been incompletely understood. In this study, we characterized and compared the responses of the human mast cell line, LAD2, and human lung epithelial cell line, Calu-3, against three influenza A virus strains; A/PR/8/34 (H1N1), A/WS/33 (H1N1) and A/HK/8/68 (H3N2). We found that there were strain-dependent mast cell responses, and different profiles of cytokine, chemokine and antiviral gene expression between the two cell types. All three strains did not induce histamine or ß-hexosaminidase release in LAD2. A/HK/8/68 induced release of prostaglandin D2 in LAD2, whereas A/PR/8/34 and A/WS/33 did not. We found that, among those examined, only CCL4 (by A/PR/8/34) was statistically significantly released from LAD2 cells. Furthermore, there was increased mRNA expression of viral recognition receptors (RIG-I and MDA5) and antiviral protein, viperin, but levels and kinetics of the expression were different among the cell types, as well as by the strains examined. Our findings highlight the variability in innate response to different strains of influenza A virus in two human cell types, indicating that further investigation is needed to understand better the role of mast cells and epithelial cells in innate immunity against influenza A viruses.


Subject(s)
Epithelial Cells/immunology , Epithelial Cells/virology , Host-Pathogen Interactions/immunology , Influenza A virus/immunology , Influenza, Human/immunology , Mast Cells/immunology , Mast Cells/virology , Biomarkers , Cell Line , Cytokines/metabolism , Disease Resistance/genetics , Epithelial Cells/metabolism , Humans , Influenza A virus/classification , Influenza, Human/genetics , Influenza, Human/metabolism , Influenza, Human/virology , Mast Cells/metabolism , Virus Release , Virus Replication
7.
Biochim Biophys Acta Gen Subj ; 1862(12): 2764-2778, 2018 12.
Article in English | MEDLINE | ID: mdl-30251665

ABSTRACT

BACKGROUND: CD28 and CTLA-4 are homologous T-cell receptors that bind with B7-1 and produce two opposing immunological signals required for T-cell activation and inactivation, respectively. It has been clinically proven that specific blockade of these key protein-protein interactions at the synapse can offer immunotherapeutic benefits for cancers and autoimmune treatments. Hence, there is a growing interest towards developing anti-CD28 and anti-CTLA-4 small molecule inhibitors. To achieve this goal, it is important to understand unique molecular level fingerprint interactions that stabilize CTLA-4/B7-1 and CD28/B7-1 complexes. However, until recently, the structure of the human CD28/B7-1 complex has not been resolved experimentally, which remains a significant setback in achieving specific inhibitors against CTLA-4 or CD28. METHODS: Here, we employed a combination of advanced molecular modelling and extensive molecular dynamics (MD) simulations to model the CD28/B7-1 complex and characterize the key interactions that stabilize the complex. RESULTS: Ensemble protein-protein docking and MD-based binding-free energy calculations were used to obtain a comprehensive structural model of the CD28/B7-1 complex, which was validated with various mutation-based experimental data from literature. Our CD28/B7-1 model has much weaker binding affinity than the CTLA-4/B7-1 complex, which is in agreement with the results from our binding assay experiments and previous studies. CONCLUSIONS: Per-residue energy decomposition of the binding affinities of the two complexes revealed the unique fingerprint hot-spot sites in CTLA-4/B7-1 and CD28/B7-1 complexes. GENERAL SIGNIFICANCE: The results presented in this work will, on a long-run, be useful to develop new generation of specific CD28 and CTLA-4 inhibitors for targeted immunotherapy.


Subject(s)
B7-1 Antigen/metabolism , CD28 Antigens/metabolism , CTLA-4 Antigen/metabolism , Molecular Dynamics Simulation , B7-1 Antigen/chemistry , CD28 Antigens/chemistry , Cluster Analysis , Humans , Immunotherapy , Ligands , Lymphocyte Activation , Protein Binding , Reproducibility of Results
8.
J Ethnopharmacol ; 169: 138-44, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25917838

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: 15,16-Dihydrotanshinone I (DHT-I), isolated from the dried root of Salvia miltiorrhiza Bung, which is traditionally used to treat cardiovascular and inflammatory diseases agent in Chinese medicine. DHT-I has been reported to have a broad range of biological activities, including antibacterial activity, and has been used to treat circulatory disorders, hepatitis, inflammation, cancer, and neurodegenerative diseases. AIM OF THE STUDY: The aim of this study was to evaluate the anti-allergic inflammatory effects of DHT-I on degranulation and on the generation of eicosanoids, such as, prostaglandin D2 (PGD2) and leukotriene C4 (LTC4), in IgE/Ag-stimulated bone marrow-derived mast cells (BMMCs). MATERIALS AND METHODS: The anti-allergic inflammatory activity of DHT-I was evaluated using BMMCs. The effects of DHT-I on mast cell activation were investigated by following degranulation and eicosanoid generation using ELISA and immunoblotting and immunoprecipitation techniques. RESULTS: DHT-I at a concentration of 20µM markedly inhibited degranulation and the generation of PGD2 and LTC4 in IgE/Ag-stimulated BMMCs (about 90% inhibitions, respectively). Analyses of FcεRI-mediated signaling pathways demonstrated that DHT-I inhibited the phosphorylations of spleen tyrosine kinase (Syk) and linker for activation of T cells (LAT), and inhibited downstream signaling process, including [Ca(2+)]i mobilization induced by the phosphorylation of phospholipase Cγ1 (PLCγ1), and the activations of mitogen-activated protein kinases (MAPKs) and the Akt-nuclear factor-κB (NF-κB) pathway. CONCLUSIONS: DHT-1 inhibits the release of allergic inflammatory mediators from IgE/Ag-stimulated mast cells by suppressing a FcεRI-mediated Syk-dependent signal pathway. This result suggests DHT-I offers a novel developmental basis for drugs targeting allergic inflammatory diseases.


Subject(s)
Bone Marrow Cells/drug effects , Immunoglobulin E/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mast Cells/drug effects , Phenanthrenes/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Salvia miltiorrhiza , Animals , Bone Marrow Cells/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Furans , Intracellular Signaling Peptides and Proteins/metabolism , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Phenanthrenes/isolation & purification , Plant Roots , Protein-Tyrosine Kinases/metabolism , Quinones , Syk Kinase
9.
Methods Mol Biol ; 1220: 339-45, 2015.
Article in English | MEDLINE | ID: mdl-25388261

ABSTRACT

The production of nitric oxide in mast cells has been difficult to measure due to the low amounts made by mast cells, as well as limitations in the specificity and sensitivity of the assays available. We present here a sensitive and specific 96-well plate-based method to directly measure NO using the cell-permeable fluorescent compound DAF-FM diacetate.


Subject(s)
Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Mast Cells/metabolism , Nitric Oxide/metabolism , Spectrometry, Fluorescence/methods , Animals , Cell Line , Humans , Mice
10.
Front Immunol ; 5: 569, 2014.
Article in English | MEDLINE | ID: mdl-25452755

ABSTRACT

Mast cells (MC) are widely distributed throughout the body and are common at mucosal surfaces, a major host-environment interface. MC are functionally and phenotypically heterogeneous depending on the microenvironment in which they mature. Although MC have been classically viewed as effector cells of IgE-mediated allergic diseases, they are also recognized as important in host defense, innate and acquired immunity, homeostatic responses, and immunoregulation. MC activation can induce release of pre-formed mediators such as histamine from their granules, as well as release of de novo synthesized lipid mediators, cytokines, and chemokines that play diverse roles, not only in allergic reactions but also in numerous physiological and pathophysiological responses. Indeed, MC release their mediators in a discriminating and chronological manner, depending upon the stimuli involved and their signaling cascades (e.g., IgE-mediated or Toll-like receptor-mediated). However, the precise mechanisms underlying differential mediator release in response to these stimuli are poorly known. This review summarizes our knowledge of MC mediators and will focus on what is known about the discriminatory release of these mediators dependent upon diverse stimuli, MC phenotypes, and species of origin, as well as on the intracellular synthesis, storage, and secretory processes involved.

11.
PLoS One ; 9(9): e108595, 2014.
Article in English | MEDLINE | ID: mdl-25268140

ABSTRACT

PGD2 has long been implicated in allergic diseases. Recent cloning of a second PGD2 receptor, DP2 (also known as CRTh2), led to a greater understanding of the physiological and pathophysiological implications of PGD2. PGD2 signaling through DP1 and DP2 mediates different and often opposite effects in many cell types of the immune system. Although mast cells (MC) are the largest source of PGD2 in the body, there is little information about their potential expression of DP2 and its functional significance. In this study, we show that tissue MC in human nasal polyps express DP2 protein, and that human MC lines and primary cultured human MC express mRNA as well as protein of DP2. By immunohistochemistry, we detected that 34% of MC in human nasal polyps expressed DP2. In addition, flow cytometry showed that 87% of the LAD2 human MC line and 98% of primary cultured human MC contained intracellular DP2. However, we could not detect surface expression of DP2 on human MC by single cell analysis using imaging flow cytometry. Blocking of endogenous PGD2 production with aspirin did not induce surface expression of DP2 in human MC. Two DP2 selective agonists, DK-PGD2 and 15R-15-methyl PGD2 induced dose-dependent intracellular calcium mobilization that was abrogated by pertussis toxin, but not by three DP2 selective antagonists. MC mediator release including degranulation was not affected by DP2 selective agonists. Thus, human MC express DP2 intracellularly rather than on their surface, and the function of DP2 in human MC is different than in other immune cells such as Th2 cells, eosinophils and basophils where it is expressed on the cell surface and induces Th2 cytokine and/or granule associated mediator release. Further studies to elucidate the role of intracellular DP2 in human MC may expand our understanding of this molecule and provide novel therapeutic opportunities.


Subject(s)
Gene Expression , Mast Cells/metabolism , Prostaglandin D2/biosynthesis , RNA, Messenger/genetics , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Aspirin/pharmacology , Calcium/metabolism , Cell Degranulation/drug effects , Cell Line , Cytosol/drug effects , Cytosol/metabolism , Humans , Ion Transport , K562 Cells , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mast Cells/cytology , Mast Cells/drug effects , Nasal Polyps/metabolism , Pertussis Toxin/pharmacology , Primary Cell Culture , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/antagonists & inhibitors , Prostaglandin D2/pharmacology , RNA, Messenger/agonists , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Receptors, Immunologic/agonists , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/metabolism
12.
J Allergy Clin Immunol ; 134(3): 714-721.e7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24948367

ABSTRACT

BACKGROUND: Extracellular signal-regulated kinases 1/2 (ERK1/2) make important contributions to allergic responses via their regulation of degranulation, eicosanoid production, and cytokine expression by mast cells, yet the mechanisms underlying their positive effects on FcεRI-dependent signaling are not fully understood. Recently, we reported that mast cell activation and anaphylaxis are negatively regulated by AMP-activated protein kinase (AMPK). However, little is known about the relationship between ERK1/2-mediated positive and the AMPK-mediated negative regulation of FcεRI signaling in mast cells. OBJECTIVE: We investigated possible interactions between ERK1/2 and AMPK in the modulation of mast cell signaling and anaphylaxis. METHODS: Wild-type or AMPKα2(-/-) mice, or bone marrow-derived mast cells obtained from these mice, were treated with either chemical agents or small interfering RNAs that modulated the activity or expression of ERK1/2 or AMPK to evaluate the functional interplay between ERK1/2 and AMPK in FcεRI-dependent signaling. RESULTS: The ERK1/2 pathway inhibitor U0126 and the AMPK activator 5-aminoimidazole-4-carboxamide-1-ß-4-ribofuranoside similarly inhibited FcεRI-mediated mast cell signals in vitro and anaphylaxis in vivo. ERK1/2-specific small interfering RNA also mimicked this effect on FcεRI signals. Moreover, AMPKα2 knockdown or deficiency led to increased FcεRI-mediated mast cell activation and anaphylaxis that were insensitive to U0126 or activator 5-aminoimidazole-4-carboxamide-1-ß-4-ribofuranoside, suggesting that the suppression of FcεRI signals by the inhibition of the ERK1/2 pathway relies largely on AMPK activation. ERK1/2 controlled AMPK activity by regulating its subcellular translocation. CONCLUSIONS: ERK1/2 ablated the AMPK-dependent negative regulatory axis, thereby activating FcεRI signals in mast cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anaphylaxis/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypersensitivity/immunology , Mast Cells/immunology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Anaphylaxis/etiology , Animals , Butadienes/pharmacology , Cell Degranulation/drug effects , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hypersensitivity/complications , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitriles/pharmacology , Receptors, IgG/metabolism , Ribonucleosides/pharmacology , Signal Transduction/drug effects
13.
Immunol Res ; 56(1): 32-43, 2013 May.
Article in English | MEDLINE | ID: mdl-23055084

ABSTRACT

Mast cells are important in innate immunity and protective against certain bacterial infections. However, there is limited evidence that mast cells respond to viruses. As mast cells are abundant in mucosal tissues of the lung, they are in a prime location to detect and respond to influenza virus. In this study, we characterized for the first time the replication cycle of influenza A virus in human mast cells by measuring influenza A virus transcription, RNA replication, protein synthesis, and formation of infectious virus as compared to the replication cycle in epithelial cells. We detected the presence of influenza A viral genomic RNA transcription, replication, and protein synthesis in human mast cells and epithelial cells. However, there was no significant release of infectious influenza A virus from mast cells, whereas epithelial cells produce ~100-fold virus compared with the inoculating dose. We confirmed that influenza A virus infects human mast cells, begins to replicate, but the production of new virus is aborted. Thus, mast cells may lack critical factors essential for productive infection or there are intrinsic or inducible anti-influenza A mechanisms in mast cells.


Subject(s)
Influenza A virus/physiology , Influenza, Human/immunology , Influenza, Human/virology , Mast Cells/virology , RNA, Viral/analysis , Virus Replication , Cells, Cultured , Epithelial Cells/immunology , Epithelial Cells/virology , Humans , Immunity, Innate , Influenza A virus/pathogenicity , Lung/pathology , Mast Cells/immunology , Transcription, Genetic , Virion/growth & development , Virion/immunology
14.
Ann Allergy Asthma Immunol ; 108(3): 172-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22374200

ABSTRACT

BACKGROUND: Nitric oxide (NO) has various roles in airway physiology and pathophysiology. Monitoring exhaled NO levels is increasingly common to measure airways inflammation and inhaled NO studied for its therapeutic value in premature infants and adult respiratory distress syndrome. NO is produced by 3 isoforms of NO synthase (NOS1, 2, 3), and each can play distinct and perhaps overlapping roles in the airways. However, the distribution, regulation, and functions of NOS in various cells in the upper airways, particularly in leukocytes, are incompletely understood. OBJECTIVE: To characterize the expression of NOS isoforms in leukocytes in normal middle turbinate tissues (MT) and in inflammatory nasal tissue (nasal polyps, NP). METHODS: Normal MT tissue was collected from surgical specimens that were to be discarded. The NP samples were from surgical tissue archives of 15 patients with chronic rhinosinusitis. Isoforms of NOS in cells were identified by double immunostaining using NOS isoform-specific and leukocyte-specific (mast cell, eosinophil, macrophage, neutrophil, or T cell) antibodies. RESULTS: The proportion of total cells below the epithelium that were positive for each isoform of NOS was higher in NP than in MT. Each isoform of NOS was found in all leukocyte populations studied, and there were significant differences in the percentage of leukocytes expressing NOS isoforms between MT and NP. CONCLUSION: All isoforms of NOS are expressed in leukocytes in MT and NP, and their expression varies among leukocyte types. Our data provide a basis to investigate the regulation, cell distribution, and distinct functions of NOS isoforms in normal and inflamed nasal tissues.


Subject(s)
Leukocytes/enzymology , Nasal Polyps/enzymology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type I/metabolism , Adult , Eosinophils/enzymology , Eosinophils/immunology , Female , Humans , Inflammation/immunology , Leukocytes/immunology , Macrophages/enzymology , Macrophages/immunology , Male , Mast Cells/enzymology , Mast Cells/immunology , Middle Aged , Nasal Mucosa/enzymology , Nasal Mucosa/immunology , Nasal Mucosa/pathology , Nasal Polyps/immunology , Nasal Polyps/pathology , Nitric Oxide/biosynthesis , Turbinates/enzymology
15.
J Leukoc Biol ; 91(4): 581-90, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22262798

ABSTRACT

In addition to its well-known role in relaxation of vascular smooth muscle, NO modulates immune responses in a concentration- and location-specific manner. For MC, it is well accepted that exogenous NO regulates their function. However, there are inconsistencies in the literature of whether MC express NOS and make NO. MC progenitors mature in peripheral tissues, but the factors that influence MC maturation and their specific phenotype, such as whether they express NOS, are not well understood. To study microenvironmental conditions that could be "permissive" for NOS expression, we cultured BMMC in various conditions--BMMC(IL-3), BMMC(SCF/IL-3), or BMMC(SCF/IL-4)-for >3 weeks and examined NOS expression. We detected Nos2 mRNA in BMMC(SCF/IL-4) but not BMMC(IL-3) or BMMC(SCF/IL-3). After stimulation with IFN-γ and/or LPS, NOS2 expression and NO production were detected in BMMC(SCF/IL-4) but rarely detected in BMMC cultured with other conditions. Confocal microscopic analysis showed that NOS2 expression induced by IFN-γ colocalized in CD117(+) BMMC. NO production, after activation with IFN-γ and LPS in BMMC(SCF/IL-4), was abrogated by pretreatment with the NOS2-specific inhibitor. In addition to NOS2 expression, BMMC(SCF/IL-4) were distinguished from BMMC(IL-3) in heparin and MMCP expression. Thus, MC progenitors that develop in SCF + IL-4 can be induced to express NOS2 after receiving appropriate signals, such as IFN-γ, and subsequently produce NO. Microenvironmental conditions during their development can influence whether MC are capable of NOS expression and of NO production.


Subject(s)
Bone Marrow Cells/immunology , Mast Cells/immunology , Nitric Oxide Synthase Type II/immunology , Nitric Oxide/immunology , Stem Cell Niche/immunology , Stem Cells/immunology , Animals , Bone Marrow Cells/cytology , Cytokines/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/immunology , Male , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Proto-Oncogene Proteins c-kit/immunology , Proto-Oncogene Proteins c-kit/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Time Factors
16.
J Immunol ; 185(1): 578-87, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20511553

ABSTRACT

NO is a short-lived free radical that plays a critical role in the regulation of cellular signaling. Mast cell (MC)-derived NO and exogenous NO regulate MC activities, including the inhibition of MC degranulation. At a molecular level, NO acts to modify protein structure and function through several mechanisms, including protein tyrosine nitration. To begin to elucidate the molecular mechanisms underlying the effects of NO in MCs, we investigated protein tyrosine nitration in human MC lines HMC-1 and LAD2 treated with the NO donor S-nitrosoglutathione. Using two-dimensional gel Western blot analysis with an anti-nitrotyrosine Ab, together with mass spectrometry, we identified aldolase A, an enzyme of the glycolytic pathway, as a target for tyrosine nitration in MCs. The nitration of aldolase A was associated with a reduction in the maximum velocity of aldolase in HMC-1 and LAD2. Nuclear magnetic resonance analysis showed that despite these changes in the activity of a critical enzyme in glycolysis, there was no significant change in total cellular ATP content, although the AMP/ATP ratio was altered. Elevated levels of lactate and pyruvate suggested that S-nitrosoglutathione treatment enhanced glycolysis. Reduced aldolase activity was associated with increased intracellular levels of its substrate, fructose 1,6-bisphosphate. Interestingly, fructose 1,6-bisphosphate inhibited IgE-mediated MC degranulation in LAD2 cells. Thus, for the first time we report evidence of protein tyrosine nitration in human MC lines and identify aldolase A as a prominent target. This posttranslational nitration of aldolase A may be an important pathway that regulates MC phenotype and function.


Subject(s)
Fructose-Bisphosphate Aldolase/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Nitric Oxide/physiology , Signal Transduction/immunology , Tyrosine/metabolism , Cell Degranulation/immunology , Cell Line, Tumor , Cells, Cultured , Fructose-Bisphosphate Aldolase/antagonists & inhibitors , Glycolysis/immunology , Humans , Immunophenotyping , Mast Cells/enzymology , Nitric Oxide Donors/metabolism , Protein Processing, Post-Translational/immunology , S-Nitrosoglutathione/metabolism , Substrate Specificity/immunology , Tyrosine/analogs & derivatives , Tyrosine/immunology
17.
Vascul Pharmacol ; 51(1): 13-20, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19013539

ABSTRACT

The matrix metalloproteinases (MMP-9 and MMP-2) in aortic smooth muscle cells (SMC) play key roles in the pathogenesis atherosclerosis. The SMC migration into the vascular wall via the bloodstream is directly linked with MMP-9 expression. Deoxypodophyllotoxin (DPT), a naturally occurring flavolignan with anti-inflammatory activity, was isolated from Anthriscus sylvestris Hoffm. and has been known inhibit the expression of MMP-9 in tumor necrosis factor-alpha (TNF-alpha) stimulated human aortic smooth muscle cells (HASMC). In this study, DPT was purified and demonstrated to inhibit the MMP-9/2 activities in TNF-alpha-induced HASMC. In addition, MMP-9 expression and migration was strongly inhibited by DPT in TNF-alpha-induced HASMC. To examine whether TNF-alpha-induced MMP-9 expressions are involved with migrations of HASMC, reverse transcription-polymerase chain reaction (RT-PCR) and luciferase-tagged promoter analysis were applied. These experiments revealed that DPT inhibited the mRNA transcription of MMP-9 gene expression. Furthermore, Western blot analysis indicated that the TNF-alpha-induced phosphorylation of extracellular signal regulated kinase 1 and 2 (ERK1/2), p38 and c-Jun N-terminal kinase (JNK) were strongly inhibited by DPT. From these results, it is concluded that DPT has an inhibitory activities on migration and MMP-2/9 activities, and MMP-9 transcription in HASMC.


Subject(s)
Aorta/enzymology , Apiaceae/physiology , Cell Migration Inhibition/drug effects , Lignans/pharmacology , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase Inhibitors , Myocytes, Smooth Muscle/enzymology , Podophyllotoxin/analogs & derivatives , Aorta/cytology , Aorta/drug effects , Apiaceae/chemistry , Cell Migration Inhibition/physiology , Cells, Cultured , Drugs, Chinese Herbal , Humans , Lignans/isolation & purification , MAP Kinase Signaling System/physiology , Matrix Metalloproteinase 9/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Roots , Podophyllotoxin/isolation & purification , Podophyllotoxin/pharmacology , Protease Inhibitors/isolation & purification , Protease Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/physiology
18.
Biol Pharm Bull ; 31(9): 1663-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18758056

ABSTRACT

Ym1 and Ym2 (Ym1/2) are chitinase-like proteins and we reported previously that IL-4 induced Ym1/2 in mouse bone marrow-derived mast cells. In the present study, ovalbumin-induced asthmatic mice were used to investigate the effect of glucocorticoids on Ym1/2 expression. Ym1/2 were highly induced in bronchoalveolar lavage fluid (BALF) and the lung. Ym1/2 expression was completely inhibited by dexamethasone (Dex) in BALF and weakly inhibited in the lung. Primary cultured macrophages were used to investigate the inhibition of Ym1/2 expression at the cellular level. Although Dex pretreatment inhibited the Ym1/2 expression level in an animal model, it did not reduce IL-4 induction of Ym1/2 expression in vitro. Next, we tested whether Dex blocks IL-4 induced STAT6 signaling and found that it had no inhibitory effect on the phosphorylation level of STAT6 in macrophages. The luciferase reporter assay also revealed that Dex did not inhibit IL-4 induction of Ym1/2 promoter activity. These results indicate that the inhibitory effect of Dex on Ym1/2 protein expression in the murine model of asthma does not involve the STAT6 signaling pathway.


Subject(s)
Chitinases/metabolism , Dexamethasone/pharmacology , Lectins/metabolism , STAT6 Transcription Factor/physiology , beta-N-Acetylhexosaminidases/metabolism , Animals , Blotting, Northern , Blotting, Western , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Chitinases/biosynthesis , Electrophoresis, Polyacrylamide Gel , Female , Indicators and Reagents , Lectins/biosynthesis , Luciferases/genetics , Luciferases/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Ovalbumin/metabolism , Phosphorylation , Plasmids/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , beta-N-Acetylhexosaminidases/biosynthesis
19.
J Cell Biochem ; 105(2): 524-33, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18773430

ABSTRACT

The matrix metalloproteinases (MMP-9 and MMP-2) production and smooth muscle cell (SMC) migration may play key roles in the pathogenesis of atherosclerotic lesions. In particular, the cancer cell invasion and SMC migration through vascular wall were shown to be directly associated with inducible MMP-9 expression. Previously, 3,4,5-trihydoroxybenzaldehyde (THBA) was purified from Geum japonicum and we demonstrated a direct inhibition effect of THBA on MMP-9 and MMP-2 activity in the supernatants of TNF-alpha-induced HASMCs. In addition, MMP-9 expression and migration was suppressed by THBA in the TNF-alpha-induced HASMCs. In this study, we also investigated whether TNF-alpha-induced MMP-9 expressions are involved with migrations of HASMCs by using cell signal inhibitors and MMP-9 inhibitors. An RT-PCR and luciferase-tagged promoter analysis revealed that THBA inhibits the transcription of MMP-9 mRNA. Moreover, an electrophoretic mobility shift assay (EMSA) exhibited that THBA also suppressed DNA binding of nuclear factor (NF)-kappaB and activator protein (AP)-1 transcription factors. Furthermore, Western blot analysis indicated TNF-alpha-induced phosphorylation of extracellular signal regulated kinase 1 and 2 (ERK1/2), p38 and c-Jun N-terminal kinase (JNK) were inhibited by THBA. Taken together, we suggest that THBA has inhibition effect to the migrations as well as MMP-2 and MMP-9 activities in HASMCs. Especially gelatinolytic activity was controlled by enzymatic inhibition of MMP-2 and MMP-9, and also down-regulated MMP-9 transcription via mitogen-activated protein kinase (MAPK) pathways in THBA treated HASMCs.


Subject(s)
Benzaldehydes/pharmacology , Geum/chemistry , Matrix Metalloproteinase Inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Benzaldehydes/isolation & purification , Gelatin/metabolism , Gene Expression Regulation/drug effects , Humans , MAP Kinase Signaling System , Matrix Metalloproteinase 9/genetics , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , Plants, Medicinal , Transcription, Genetic
20.
Int Immunopharmacol ; 8(10): 1395-400, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18687301

ABSTRACT

We evaluated the ability of saucerneol D (SD), a tetrahydrofuran-type sesquilignan isolated from Saururus chinensis, to regulate the expression of inducible nitric oxide synthase (iNOS) in lipopolysaccharide (LPS)-stimulated murine macrophage-like RAW264.7 cells. SD consistently inhibited nitric oxide (NO) production in a dose-dependent manner, with an IC(50) of 2.62 microM, and also blocked LPS-induced iNOS expression. SD potently suppressed both the reporter gene expression and DNA-binding activity of nuclear factor-kappaB (NF-kappaB). In addition, SD inhibited IkappaB-alpha degradation in a concentration- and time-dependent manner. SD also inhibited LPS-induced activation of various mitogen-activated protein kinases, including extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun NH(2)-terminal kinase (JNK). These findings suggest that SD may inhibit LPS-induced iNOS expression by blocking NF-kappaB and MAPK activation.


Subject(s)
Gene Expression/drug effects , Macrophages/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Animals , Cells, Cultured , Lignans/pharmacology , Lipopolysaccharides/toxicity , Macrophages/enzymology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...