Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters










Publication year range
1.
ScientificWorldJournal ; 2022: 8454865, 2022.
Article in English | MEDLINE | ID: mdl-36330350

ABSTRACT

Background: Hot water extract of Sasa albomarginata (Kumazasa) leaves is commercially available and used as a dietary supplement or skincare cream. The extract possesses anti-inflammatory activity on the mouse atopic dermatitis model. To elucidate the mechanism of in vivo activity, we have studied the cellular anti-inflammatory and antioxidant activities of the extract and its constituents. Methods: Secretion of mouse and human IL-6 was measured by ELISA. ROS production was measured by a fluorescent reagent. Ultrahigh performance liquid chromatography (UHPLC)/MS was used for the ingredient analysis. Results: The Sasa albomarginata extract inhibited inflammatory mediators such as LPS-induced NO, IL-6, and ROS production in mouse monocyte leukemia RAW264.7 cells. It also inhibited iNOS, IL-6, and IL-1ß expressions. Moreover, it inhibited LPS-induced IL-6 expression and production in human monocyte leukemia THP-1 cells differentiated into macrophages. The HPLC analysis of the extract revealed the existence of coumaric acid, ferulic acid, and coumaric acid methyl ester. Coumaric acid methyl ester but not coumaric acid or ferulic acid inhibited LPS-induced NO, IL-6, and ROS production in RAW264.7 cells and IL-6 production in differentiated THP-1 cells. Conclusion: The hot water extract of Sasa albomarginata leaves and one of its constituents possess cellular anti-inflammatory and antioxidant activities.


Subject(s)
Leukemia , Sasa , Humans , Antioxidants/pharmacology , Lipopolysaccharides , Interleukin-6 , Esters , Reactive Oxygen Species , Plant Extracts/pharmacology , Plant Extracts/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Water , Nitric Oxide/metabolism
2.
Front Microbiol ; 13: 780534, 2022.
Article in English | MEDLINE | ID: mdl-35265056

ABSTRACT

Macrophages play a central role in the innate immune response to respiratory viral infections through pro-inflammatory factor secretion and phagocytosis. However, as a countermeasure, viral pathogens have evolved virulence factors to antagonize macrophage function. In our recent in vitro analyses of murine macrophage cell lines, Sendai virus (SeV) accessory protein C inhibited the secretion of pro-inflammatory factors, and C gene-knockout SeV (SeVΔC) caused drastic morphological changes in RAW264.7 macrophages, similar to those observed after stimulation with Lipid A, a well-known activator of actin-rich membrane ruffle formation and phagocytosis. Hence, we sought to determine whether the C protein limits phagocytosis in SeV-infected macrophages through the suppression of membrane ruffling. Phagocytosis assays indicated an upregulation of phagocytosis in both SeVΔC-infected and Lipid A-stimulated macrophages, but not in SeV WT-infected cells. Further, the observed membrane ruffling was associated with phagocytosis. RIG-I is essential for Lipid A-induced phagocytosis; its deficiency inhibited SeVΔC-stimulated phagocytosis and ruffling, confirming the essential role of RIG-I. Moreover, treatment with interferon (IFN)-ß stimulation and neutralizing antibodies against IFN-ß suggested that SeVΔC-induced phagocytosis and ruffling occurred in an IFN-ß-independent manner. A newly isolated SeVΔC strain that does not generate dsRNA further highlighted the importance of dsRNA in the induction of phagocytosis and ruffling. Taken together, the current results suggest that SeV C protein might limit phagocytosis-associated membrane ruffling in an RIG-I-mediated but IFN-independent manner via limiting the generation of intracellular dsRNA.

3.
Microbiol Immunol ; 66(3): 124-134, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34859490

ABSTRACT

Sendai virus (SeV) accessory protein C limits the generation of double-stranded RNAs, defective interfering RNAs, or both, during viral transcription and replication, thereby limiting interferon-ß production. Our recent in vitro analyses on murine macrophage cell lines demonstrated that this protein also contributes to restricting macrophage function, including the production of nitric oxide (NO) and inflammatory cytokines in addition to interferon-ß, in infected macrophages. This study showed that depletion of airway macrophages by clodronate-loaded liposomes led to the development of severe viral pneumonia in recombinant C gene-knockout SeV (SeV∆C)-infected mice, but did not modulate disease severity in wild-type SeV-infected mice. Furthermore, the severe disease observed in macrophage-depleted, SeV∆C-infected mice was associated with exacerbated virus replication in the lungs, leading to severe airway inflammation and pulmonary edema, indicating lung injury. These results suggested that the antimacrophage activity of SeV C protein might play a critical role in modulating lung injury and associated diseases caused by SeV.


Subject(s)
Respirovirus Infections , Sendai virus , Animals , Interferon-beta , Macrophages/metabolism , Mice , Sendai virus/metabolism , Severity of Illness Index
4.
Exp Ther Med ; 22(4): 1092, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34504546

ABSTRACT

Plasmacytoma is one of the most difficult types of leukemia to treat, and it often invades the bone down to the marrow resulting in the development of multiple myeloma. NF-κB is often constitutively activated, and promotes metastasis and drug resistance in neoplastic cells. The present study assessed the cellular anticancer activity of an NF-κB inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ), on mouse plasmacytoma SP2/0 cells. Cellular invasion was measured by Matrigel chamber assay, and apoptosis was assessed by detecting caspase-3 cleavage and by flow cytometric analysis with Annexin V. DHMEQ inhibited constitutively activated NF-κB at nontoxic concentrations. DHMEQ was also shown to inhibit cellular invasion of SP2/0 cells, as well as human myeloma KMS-11 and RPMI-8226 cells. The metastasis PCR array indicated that DHMEQ induced a decrease in KISS1 receptor (KISS1R) expression in SP2/0 cells. Knockdown of KISS1R by small interfering RNA suppressed cellular invasion, suggesting that KISS1R may serve an essential role in the invasion of SP2/0 cells. Furthermore, DHMEQ enhanced cytotoxicity of the anticancer agent melphalan in SP2/0 cells. Notably, DHMEQ inhibited the expression of NF-κB-dependent anti-apoptotic proteins, such as Bcl-XL, FLIP, and Bfl-1. In conclusion, inhibition of constitutively activated NF-κB by DHMEQ may be useful for future anti-metastatic and anticancer strategies for the treatment of plasmacytoma.

5.
Microbes Infect ; 22(8): 322-330, 2020 09.
Article in English | MEDLINE | ID: mdl-32032681

ABSTRACT

Sendai virus V protein is a known antagonist of RIG-I-like receptors (RLRs) RIG-I and MDA5, which activate transcription factors IRF3, leading to activation of ISGF3 and NF-κB. These transcription factors are known activators of inducible NO synthase (iNOS) and increase the production of nitric oxide (NO). By inhibiting ISGF3 and NF-κB, the V protein acts as an indirect negative regulator of iNOS and NO. Here we report that the V gene knockout Sendai virus [SeV V(-)] markedly enhanced iNOS expression and subsequent NO production in infected macrophages compared to wild-type SeV. The knockout of RIG-I in cells inhibited SeV V(-)-induced iNOS expression and subsequent NO production. To understand the underlying mechanism of the V protein-mediated negative regulation of iNOS activation, we transfected HEK293T cells with RIG-I and the RIG-I regulatory protein TRIM25. Our results demonstrated that the V protein inhibited iNOS activation via the RIG-I/TRIM25 pathway. Moreover, the V protein inhibited TRIM25-mediated K63-linked ubiquitination of RIG-I, as well as its CARD-dependent interaction with mitochondrial antiviral signaling (MAVS) molecules. These results suggest that the V protein downregulates iNOS activation and inhibits NO production by preventing the RIG-I-MAVS interaction, possibly through its effect on the ubiquitination status of RIG-I.


Subject(s)
DEAD Box Protein 58/metabolism , Macrophages/metabolism , Nitric Oxide/metabolism , Sendai virus/metabolism , Signal Transduction , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , DEAD Box Protein 58/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Macrophages/virology , Mice , Nitric Oxide Synthase Type II/metabolism , Protein Binding , RAW 264.7 Cells , Sendai virus/genetics , Transcription Factors/metabolism , Ubiquitination , Viral Proteins/genetics
6.
Mol Med Rep ; 18(6): 5279-5285, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30320338

ABSTRACT

PM2.5 is a particle with a diameter <2.5 µm that is often involved in air pollution. Nanoparticles <100 nm are thought to invade the trachea and lungs to cause inflammation, possibly through the activation of macrophages. On the other hand, titanium dioxide (TiO2) particles can be used in models of nano­micro­sized particles, as one can prepare the particles with such sizes. TiO2 particles are classified into Rutile, Anatase, and Brookite types by their crystal structure. Among them, Anatase­type TiO2 particles with a primary diameter of 50 nm (A50) were reported to induce interleukin (IL)­1ß production and secretion effectively in phorbol 12­myristate 13­acetate­treated human monocytic leukemia THP­1 cells (THP­1 macrophages). We previously designed and synthesized dehydroxymethyl­epoxyqinomicin (DHMEQ) as an inhibitor of NF­κB. The present study investigated whether the NF­κB inhibitor DHMEQ inhibits TiO2 nanoparticle­induced IL­1ß production in THP­1 macrophages, and determined the mechanism. As a result, DHMEQ inhibited A50­induced IL­1ß secretion in ELISA assays at nontoxic concentrations. It decreased the expression of IL­1ß mRNA, which was dependent on NF­κB. Although NLR family pyrin domain containing 3 (NLRP3)­inflammasome­caspase­1 activation is required for the maturation of IL­1ß, and DHMEQ reduced the NLRP3 mRNA expression and caspase­1 activity; a caspase­1 inhibitor did not influence the A50­induced IL­1ß production. Therefore, it is likely that inhibition of pro­IL­1ß expression by DHMEQ may be sufficient to inhibit mature IL­1ß production. Thus, DHMEQ may be useful for the amelioration of inflammation in the trachea and lungs caused by inhalation of PM2.5.


Subject(s)
Benzamides/pharmacology , Cyclohexanones/pharmacology , Interleukin-1beta/biosynthesis , NF-kappa B/antagonists & inhibitors , Nanoparticles , Titanium , Animals , Biomarkers , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Cell Line , Disease Models, Animal , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Models, Molecular , Nanoparticles/chemistry , Particulate Matter , Potassium/metabolism , Titanium/chemistry
7.
Innate Immun ; 24(7): 430-438, 2018 10.
Article in English | MEDLINE | ID: mdl-30189760

ABSTRACT

To suppress virus multiplication, infected macrophages produce NO. However, it remains unclear how infecting viruses then overcome NO challenge. In the present study, we report the effects of accessory protein C from Sendai virus (SeV), a prototypical paramyxovirus, on NO output. We found that in RAW264.7 murine macrophages, a mutant SeV without C protein (4C(-)) significantly enhanced inducible NO synthase (iNOS) expression and subsequent NO production compared to wild type SeV (wtSeV). SeV 4C(-) infection caused marked production of IFN-ß, which is involved in induction of iNOS expression via the JAK-STAT pathway. Addition of anti-IFN-ß Ab, however, resulted in only marginal suppression of NO production. In contrast, NF-κB, a primarily important factor for transcription of the iNOS gene, was also activated by 4C(-) infection but not wtSeV infection. Induction of NO production and iNOS expression by 4C(-) was significantly suppressed in cells constitutively expressing influenza virus NS1 protein that can sequester double-stranded (ds)RNA, which triggers activation of signaling pathways leading to activation of NF-κB and IRF3. Therefore, C protein appears to suppress NF-κB activation to inhibit iNOS expression and subsequent NO production, possibly by limiting dsRNA generation in the context of viral infection.


Subject(s)
Macrophages/physiology , Respirovirus Infections/immunology , Sendai virus/physiology , Viral Proteins/metabolism , Animals , Gene Expression Regulation , Interferon Regulatory Factor-3/metabolism , Janus Kinases/metabolism , Mice , Mutation/genetics , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , RNA, Double-Stranded/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Viral Nonstructural Proteins/metabolism , Viral Proteins/genetics
8.
J Virol ; 92(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30021903

ABSTRACT

Inflammasomes play a key role in host innate immune responses to viral infection by caspase-1 (Casp-1) activation to facilitate interleukin-1ß (IL-1ß) secretion, which contributes to the host antiviral defense. The NLRP3 inflammasome consists of the cytoplasmic sensor molecule NLRP3, adaptor protein ASC, and effector protein pro-caspase-1 (pro-Casp-1). NLRP3 and ASC promote pro-Casp-1 cleavage, leading to IL-1ß maturation and secretion. However, as a countermeasure, viral pathogens have evolved virulence factors to antagonize inflammasome pathways. Here we report that V gene knockout Sendai virus [SeV V(-)] induced markedly greater amounts of IL-1ß than wild-type SeV in infected THP1 macrophages. Deficiency of NLRP3 in cells inhibited SeV V(-)-induced IL-1ß secretion, indicating an essential role for NLRP3 in SeV V(-)-induced IL-1ß activation. Moreover, SeV V protein inhibited the assembly of NLRP3 inflammasomes, including NLRP3-dependent ASC oligomerization, NLRP3-ASC association, NLRP3 self-oligomerization, and intermolecular interactions between NLRP3 molecules. Furthermore, a high correlation between the NLRP3-binding capacity of V protein and the ability to block inflammasome complex assembly was observed. Therefore, SeV V protein likely inhibits NLRP3 self-oligomerization by interacting with NLRP3 and inhibiting subsequent recruitment of ASC to block NLRP3-dependent ASC oligomerization, in turn blocking full activation of the NLRP3 inflammasome and thus blocking IL-1ß secretion. Notably, the inhibitory action of SeV V protein on NLRP3 inflammasome activation is shared by other paramyxovirus V proteins, such as Nipah virus and human parainfluenza virus type 2. We thus reveal a mechanism by which paramyxovirus inhibits inflammatory responses by inhibiting NLRP3 inflammasome complex assembly and IL-1ß activation.IMPORTANCE The present study demonstrates that the V protein of SeV, Nipah virus, and human parainfluenza virus type 2 interacts with NLRP3 to inhibit NLRP3 inflammasome activation, potentially suggesting a novel strategy by which viruses evade the host innate immune response. As all members of the Paramyxovirinae subfamily carry similar V genes, this new finding may also lead to identification of novel therapeutic targets for paramyxovirus infection and related diseases.


Subject(s)
Inflammasomes/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Respirovirus Infections/metabolism , Sendai virus/metabolism , Viral Proteins/metabolism , Caspase 1/genetics , Caspase 1/metabolism , HEK293 Cells , Humans , Inflammasomes/genetics , Interleukin-1beta/genetics , Macrophages/pathology , Macrophages/virology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Protein Multimerization/genetics , Respirovirus Infections/genetics , Respirovirus Infections/pathology , Sendai virus/genetics , THP-1 Cells , Viral Proteins/genetics
9.
Int J Mol Sci ; 19(3)2018 Mar 03.
Article in English | MEDLINE | ID: mdl-29510517

ABSTRACT

We previously designed and synthesized dehydroxyepoxyquinomicin (DHMEQ) as an inhibitor of NF-κB based on the structure of microbial secondary metabolite epoxyquinomicin C. DHMEQ showed anti-inflammatory and anticancer activity in various in vivo disease models without toxicity. On the other hand, the process of cancer metastasis consists of cell detachment from the primary tumor, invasion, transportation by blood or lymphatic vessels, invasion, attachment, and formation of secondary tumor. Cell detachment from the primary tumor and subsequent invasion are considered to be early phases of metastasis, while tumor cell attachment to the tissue and secondary tumor formation the late phases. The assay system for the latter phase was set up with intra-portal-vein injection of pancreatic cancer cells. Intraperitoneal administration of DHMEQ was found to inhibit liver metastasis possibly by decreasing the expression of MMP-9 and IL-8. Also, when the pancreatic cancer cells treated with DHMEQ were inoculated into the peritoneal cavity of mice, the metastatic foci formation was inhibited. These results indicate that DHMEQ is likely to inhibit the late phase of metastasis. Meanwhile, we have recently employed three-dimensional (3D) culture of breast cancer cells for the model of early phase metastasis, since the 3D invasion just includes cell detachment and invasion into the matrix. DHMEQ inhibited the 3D invasion of breast cancer cells at 3D-nontoxic concentrations. In this way, DHMEQ was shown to inhibit the late and early phases of metastasis. Thus, DHMEQ is likely to be useful for the suppression of cancer metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cyclohexanones/pharmacology , NF-kappa B/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Benzamides/chemistry , Benzamides/therapeutic use , Cyclohexanones/chemistry , Cyclohexanones/therapeutic use , Humans , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Quinones/chemistry , Quinones/pharmacology
10.
Hum Cell ; 31(2): 95-101, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29249016

ABSTRACT

Increasing metabolic syndromes including type-2 diabetes mellitus, obesity, and steatohepatitis are serious problems in most countries in the world. Neurodegenerative diseases such as Alzheimer, Parkinson's, and Huntington's diseases are increasing in many countries. However, therapy for these diseases is not sufficient yet. Thus, effective chemotherapy for these diseases is being expected. Conophylline is an alkaloid isolated from the leaves of Ervatamia microphylla and related plants. It was found to induce beta-cell differentiation in the precursor pancreatic cells. Oral administration of this compound ameliorated type-2 diabetes mellitus model in mice and rats. Later, fibrosis of the pancreatic islets was found to be greatly reduced by conophylline in the pancreatic islets. It also inhibited chemically induced liver cirrhosis. Further study indicated that conophylline inhibited non-alcoholic steatohepatitis in the model mice. On the one hand, loss of autophagy often causes protein aggregation to give neural cell death. Conophylline was found to activate autophagy in cultured neural cells. Activation of autophagy ameliorated cellular models of Parkinson's and Huntington's diseases. Thus, conophylline is likely to be useful for the development of chemotherapy for metabolic and neurodegenerative diseases.


Subject(s)
Metabolic Syndrome/drug therapy , Neurodegenerative Diseases/drug therapy , Phytotherapy , Vinca Alkaloids/pharmacology , Vinca Alkaloids/therapeutic use , Animals , Autophagy/drug effects , Cell Differentiation/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Fibrosis , Humans , Islets of Langerhans/pathology , Mice , Molecular Targeted Therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Plant Leaves/chemistry , Tabernaemontana/chemistry , Vinca Alkaloids/isolation & purification
11.
Bioorg Med Chem Lett ; 27(3): 562-566, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28003138

ABSTRACT

(-)-Dehydroxymethylepoxyquinomicin ((-)-DHMEQ, 1) is a specific inhibitor of NF-κB. It binds to SH group in the specific cysteine residue of NF-κB components with its epoxide moiety to inhibit DNA binding. In the present research, we have designed and synthesized an epoxide-free analog called (S)-ß-salicyloylamino-α-exo-methylene-Æ´-butyrolactone (SEMBL, 3). SEMBL inhibited DNA binding of NF-κB component p65 in vitro. It inhibited LPS-induced NF-κB activation, iNOS expression, and inflammatory cytokine secretions. It also inhibited NF-κB and cellular invasion in ovarian carcinoma ES-2 cells. Moreover, its stability in aqueous solution was greatly enhanced compared with (-)-DHMEQ. Thus, SEMBL has a potential to be a candidate for a new anti-inflammatory and anticancer agent.


Subject(s)
4-Butyrolactone/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Design , NF-kappa B/antagonists & inhibitors , Salicylamides/pharmacology , 4-Butyrolactone/chemical synthesis , 4-Butyrolactone/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Molecular Structure , NF-kappa B/metabolism , RAW 264.7 Cells , Salicylamides/chemical synthesis , Salicylamides/chemistry , Structure-Activity Relationship
12.
Oncol Res ; 25(4): 479-483, 2017 Apr 14.
Article in English | MEDLINE | ID: mdl-27623760

ABSTRACT

A Wnt agonist, 2-amino-4-[3,4-(methylenedioxy)benzylamino]-6-(3-methoxyphenyl) pyrimidine, is a cell-permeable pyrimidine compound that has been shown to mimic the effect of Wnt. In this study, leukemic mouse cell lines, RAW 264.7 and J774.1, were incubated with the Wnt agonist. The Wnt agonist showed cell death in the concentration of 1-10 µM. The Wnt agonist did not show inhibition of GSK-3ß activity but induced ß-catenin accumulation in the nucleus. The Wnt agonist showed caspase-independent cell death, but no further involvement in cell death ER stress signaling. Here we discuss the possible mechanism of Wnt agonist-induced apoptotic cell death in RAW 264.7 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Pyrimidines/pharmacology , Wnt Signaling Pathway/drug effects , Animals , Caspases/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , Mice , beta Catenin/metabolism
13.
Immunopharmacol Immunotoxicol ; 38(4): 298-302, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27251848

ABSTRACT

IL-1ß is one of the inflammatory cytokines and is cleaved from pro-IL-1ß proteolytically by activated Caspase 1. For the activation of Caspase 1, inflammasome was formed by two signals, what is called, priming and triggering signals. In this study, it was found that mouse macrophage J774.1 cells, when treated by single large amount of lipopolysaccharide (LPS), produced a significant amount of IL-1ß. On the other hand, IL-1ß production was not detected when treated by a single, small amount of LPS. Then, focusing on endoplasmic reticulum (ER) stress response among stress responses induced by a large amount of LPS, when GSK2656157, a PERK inhibitor, was used for inhibition of ER stress, GSK2656157 reduced IL-1ß production dose-dependently. Next, when Thapsigargin, an ER stress reagent, was added with LPS, IL-1ß production increased more than by LPS alone. Thus, these results suggested that ER stress was involved in LPS-induced IL-1ß production. When the activation of Caspase 1 was examined by fluorescence activated cell sorter analysis, it was found that GSK2656157 inhibited LPS-induced Caspase 1 activation. Further, it was confirmed that GSK2656157 did not affect LPS-induced TNF-α production and activation of NF-κB and specifically inhibited the PERK/eIF-2α pathway. Therefore, it was found that GSK2656157 specifically inhibited ER stress induced by large amount of LPS and reduced LPS-induced IL-1ß production through inhibition of Caspase 1 activation.


Subject(s)
Adenine/analogs & derivatives , Caspase 1/immunology , Indoles/pharmacology , Interleukin-1beta/immunology , Lipopolysaccharides/toxicity , Macrophages/immunology , eIF-2 Kinase/antagonists & inhibitors , Adenine/pharmacology , Animals , Cell Line , Enzyme Activation/drug effects , Enzyme Activation/immunology , Mice
14.
Biomed Pharmacother ; 76: 1-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26653542

ABSTRACT

It has been known that LPS activates macrophages and induces IFN-ß production from macrophages. The endogenous IFN-ß produced by LPS stimulates the cells, which plays a role in innate immune. However, it was not elucidated yet if the signaling by exogenous IFN-ß was influenced by LPS stimulation. In this study, it was found pretreatment of LPS interrupted IFN-ß-induced JAK1/STAT1 phosphorylation. LPS pretreatment also reduced IFN-ß-induced ISG54, one of IFN-ß-inducible genes. Pretreatment with LPS for more than 2h shows inhibitory effect on IFN-ß-induced STAT1 phosphorylation but simultaneous treatment or post-treatment of LPS with IFN-ß did not show the inhibitory effect. The study using a neutralizing antibody to IFN-ß indicated that IFN-ß produced by LPS does not take part in the inhibitory effect of LPS. Furthermore, LPS did not affect the expression of IFN αß receptor. A previous report has shown that LPS-induced SOCS3 inhibited IFN-γ-induced STAT1 phosphorylation, likewise, it was also shown in this study that LPS induced SOCS3 expression and its expression inhibited IFN-ß-induced STAT1 phosphorylation which was confirmed by the knockdown study by the siRNA of SOCS3. The real-time PCR and immune-blot studies of SOCS3 indicated that LPS induced SOCS3 is independent of IL-6, IL-10, TNF-α and STAT3, and might depend on p38 activation by LPS. It was suggested that bacterial LPS rather interfere with IFN-ß actions, dependent on the timing of LPS stimulation.


Subject(s)
Interferon-beta/pharmacology , Lipopolysaccharides/pharmacology , STAT1 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cell Line , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Interferon-beta/administration & dosage , Macrophages/drug effects , Macrophages/metabolism , Mice , Phosphorylation/drug effects , RNA, Small Interfering/administration & dosage , Real-Time Polymerase Chain Reaction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Time Factors
15.
Innate Immun ; 21(7): 770-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26224488

ABSTRACT

The effect of TGF-ß1 on CpG DNA-induced type I IFN production was examined by reconstituting a series of signaling molecules in TLR 3 signaling. TGF-ß1 inhibited CpG DNA-induced IFN-α4 productivity in HeLa cells. Transfection of IFN regulatory factor (IRF)7 but not TNF receptor-associated factor (TRAF)6 and TRAF3 into cells triggered IFN-α4 productivity, and TGF-ß1 inhibited IRF7-mediated type I IFN production in the presence of TRAF6. TGF-ß1 induced ubiquitination of TRAF6, although CpG DNA did not induce it. Moreover, TGF-ß1 accelerated the ubiquitination of TRAF6 in the presence of CpG DNA. TGF-ß1 ubiquitinated TRAF6 at K63 but not K48. TGF-ß1 also induced ubiquitination of IRF7. Further, TGF-ß1 did not impair the interaction of IRF7 and TRAF6. CpG DNA induced the phosphorylation of IRF7 in the presence of TRAF6, whereas TGF-ß1 inhibited the IRF7 phosphorylation. Blocking of TRAF6 ubiquitination abolished the inhibition of CpG DNA-induced type I IFN production by TGF-ß. Taken together, TGF-ß was suggested to inhibit CpG DNA-induced type I IFN production transcriptionally via ubiquitination of TRAF6.


Subject(s)
Interferon-alpha/metabolism , Oligodeoxyribonucleotides/metabolism , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 6/metabolism , Transforming Growth Factor beta1/metabolism , DNA/genetics , HeLa Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon Regulatory Factor-7/metabolism , Phosphorylation , Protein Binding , Signal Transduction , Ubiquitination
16.
Int Immunopharmacol ; 26(1): 97-102, 2015 May.
Article in English | MEDLINE | ID: mdl-25817178

ABSTRACT

High-mobility group box 1 (HMGB1) is suggested to participate in development of local and systemic inflammatory disorders. Irbesartan (IRB), an angiotensin II type1 receptor blocker, is widely used for treatment of hypertension, especially in patients with diabetic nephropathy. The effect of IRB on lipopolysaccharide (LPS)-induced HMGB1 and nitric oxide (NO) production was examined using RAW 264.7 macrophage-like cells. IRB inhibited LPS-induced HMGB1 production. IRB also reduced LPS-induced expression of an inducible NO synthase, and inhibited LPS-induced NO production. The expression levels of IFN-ß protein and mRNA, which is a key molecule in MyD88-independent pathway of LPS signaling, were exclusively inhibited by IRB. Peroxisome proliferator-activated receptor-γ and angiotensin II type 1 receptor were not involved in the inhibitory action of IRB on LPS-induced HMGB1 and NO production. Collectively, IRB was suggested to inhibit LPS-induced HMGB1 production via downregulation of IFN-ß production in the MyD88-independent pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Biphenyl Compounds/pharmacology , HMGB1 Protein/biosynthesis , Interferon-beta/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Tetrazoles/pharmacology , Animals , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Down-Regulation , HMGB1 Protein/antagonists & inhibitors , Interferon-beta/antagonists & inhibitors , Irbesartan , Macrophages/immunology , Mice , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Real-Time Polymerase Chain Reaction
17.
Int Immunopharmacol ; 25(1): 162-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25617668

ABSTRACT

We have isolated 9-methylstreptimidone from microorganism as a new NF-κB inhibitor. Later, we designed 3-[(dodecylthiocarbonyl) methyl]-glutarimide (DTCM-glutarimide) as an analog of this compound, which shows anti-inflammatory activity in vivo. In the present research, we found that DTCM-glutarimide inhibited receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation of mouse bone marrow-derived macrophages and RANKL- or lipopolysaccharide (LPS)-induced osteoclast differentiation of RAW 264.7 cells without any toxicity. It also inhibited the RANKL-induced NFATc1 expression. Upstream signaling involving phosphorylation of Akt and GSK-3ß was induced by RANKL, of which the signaling was inhibited by DTCM-glutarimide. Then DTCM-glutarimide was confirmed to inhibit RANKL-induced NF-κB activity, possibly by inhibiting the Akt-mediated activation of IKK. Thus, DTCM-glutarimide inhibited osteoclastogenesis by blocking both the Akt-GSK3ß-NFATc1 and NF-κB-NFATc1 pathways. DTCM-glutarimide may be a candidate as a chemotherapeutic agent for severe bone resorption diseases.


Subject(s)
Bone Resorption/drug therapy , Macrophages/drug effects , NF-kappa B/antagonists & inhibitors , Osteoclasts/drug effects , Piperidones/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Down-Regulation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Lipopolysaccharides/metabolism , Macrophages/physiology , Mice , Mice, Inbred ICR , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Oncogene Protein v-akt/metabolism , Osteoclasts/physiology , Piperidones/chemical synthesis , RANK Ligand/metabolism , Signal Transduction/drug effects
18.
Microvasc Res ; 98: 68-73, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25582076

ABSTRACT

The effect of poly I:C on interferon (IFN)-γ-induced nitric oxide (NO) production in vascular endothelial cells was examined using murine aortic endothelial END-D cells. Poly I:C augmented IFN-γ-induced NO production although it alone did not induce the NO production. Poly I:C augmented the NO production via enhanced expression of an inducible NO synthase protein. Poly I:C did not affect the activation of Janus kinase (JAK) 1/2, and signal transducer and activator of transcription (STAT) 1 in IFN-γ signaling. Moreover, there was no significant difference in the IFN-γ-induced interferon regulatory factor (IRF) 1 expression between the presence and absence of poly I:C. Poly I:C led to the activation of IRF7 in END-D cells. Inhibition of poly I:C signaling by amlexanox, an inhibitor of TANK-binding kinase (TBK) 1 and IκB kinase (IKK) ε, abolished the augmentation of IFN-γ-induced NO production. Therefore, poly I:C was suggested to augment IFN-γ-induced NO production at the transcriptional level via enhanced IRF7 activation.


Subject(s)
Aorta/metabolism , Endothelial Cells/cytology , Interferon Regulatory Factor-7/metabolism , Interferon-gamma/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide/chemistry , Poly I-C/chemistry , Aminopyridines/chemistry , Animals , Cell Line , Cell Proliferation , Enzyme Inhibitors/chemistry , Flow Cytometry , Gene Expression Regulation/drug effects , Interferon Regulatory Factor-3/metabolism , Mice , Nitrites/chemistry , Phosphorylation , Signal Transduction
19.
Immunobiology ; 220(1): 136-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25172547

ABSTRACT

The effect of lipopolysaccharide (LPS) on the expression of p53 protein in RAW 264.7 macrophage cells was examined. LPS downregulated the expression of p53 protein 4-24 h after the stimulation. LPS-induced p53 inhibition was restored with pharmacological inhibitors of c-jun N-terminal kinase (JNK) and phosphatidylinositol 3-kinase (PI3K). It was also restored by inhibitors of MDM2 activation and proteasome. LPS-induced p53 inhibition corresponded to activation of MDM2. LPS-induced MDM2 activation was prevented by inhibitors of JNK and PI3K. The expression of p65 NF-κB at a late stage after LPS stimulation was downregulated in the presence of a MDM2 inhibitor. Nutlin-3 as a MDM2 inhibitor reduced LPS-induced production of nitric oxide but not tumor necrosis factor-α. Administration of LPS into mice downregulated the in vivo expression of p53 in the livers. Taken together, LPS was suggested to downregulate the expression of p53 via activation of MDM2 and enhance the activation of NF-κB at a late stage.


Subject(s)
Lipopolysaccharides/immunology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Imidazoles/pharmacology , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Liver/drug effects , Liver/immunology , Liver/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Nitric Oxide/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
20.
Innate Immun ; 21(2): 194-202, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24595208

ABSTRACT

Here we report that LPS induces osteoclast (OC) formation in murine RAW 264.7 macrophage cells in RPMI-1640 medium but not in α-minimum essential medium (α-MEM) as the original culture medium. LPS-induced OC formation in both media was examined to clarify the differential response. Receptor activator of NF-κB ligand induced OC formation in either α-MEM or RPMI-1640 medium. However, LPS-induced OC formation in RAW 264.7 cells maintained in RPMI-1640 medium, but not α-MEM, which was also supported by mouse bone marrow-derived macrophages, although they were less sensitive to LPS than RAW 264.7 cells. LPS augmented the expression of nuclear factor of activated T-cells (NFATc1) as a key transcription factor of osteoclastogenesis in cells maintained in RPMI-1640 medium, but reduced it in cells maintained in α-MEM. A high concentration of LPS was cytotoxic against cells maintained in α-MEM. Glutathione exclusively present in RPMI-1640 medium prevented LPS-induced cell death in α-MEM and augmented LPS-induced NFATc1 expression, followed by enhanced LPS-induced OC formation. LPS induced higher generation of reactive oxygen species in α-MEM than RPMI-1640 medium. An antioxidant enhanced LPS-induced OC formation, whereas a pro-oxidant reduced it. Taken together, redox balance in the culture condition was suggested to regulate in vitro LPS-induced OC formation.


Subject(s)
Culture Media/metabolism , Glutathione/metabolism , Lipopolysaccharides/metabolism , Macrophages/physiology , Osteoclasts/physiology , Animals , Cell Death , Cell Differentiation , Cell Line , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...