Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 456(1): 122-8, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25446109

ABSTRACT

The crosstalk of intracellular signaling pathways is extremely complex. Previous studies have shown that there is a potential crosstalk between MAPKs and NF-κB signaling pathways. It has been reported that JNK regulates cell survival under some conditions. But the molecular mechanism through which JNK regulates cell survival is still unclear. In the present study, we hypothesized that there was a crosstalk between JNK and NF-κB signaling pathway regulating cell survival and HSP27 phosphorylation mediates such a crosstalk. Our data showed that in HepG2 cells, suppression of JNK activation by a specific inhibitor or overexpression of JNK inactive mutant enhanced TNF-α-induced apoptosis. In addition, reduction of JNK activation attenuated HSP27 phosphorylation envoked by TNF-α, especially the phosphorylation of HSP27 at serine 78 residue. Our results also showed that suppression of JNK activation reduced the degradation of IκB-α, but did not affect IKK phosphorylation upon TNF-α stimulation. Co-immunoprecipitation experiments demonstrated that JNK regulated the degradation of IκB-α through promoting the formation of HSP27/IKK/IκB-α ternary complex in response to TNF-α. Suppression of JNK activation hindered HSP27 phosphorylation at Ser78 residue and subsequently reduced the interaction between IKK and IκB-α. Taken together, our study suggests that through modulation the phosphorylation of HSP27, JNK plays an important roles in cell survival via regulating NF-κB signaling pathway.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , MAP Kinase Kinase 4/metabolism , NF-kappa B/metabolism , Signal Transduction , Apoptosis , Enzyme Activation , Hep G2 Cells , Humans , I-kappa B Proteins/metabolism , Immunoprecipitation , JNK Mitogen-Activated Protein Kinases/metabolism , Mutation , NF-KappaB Inhibitor alpha , Phosphorylation , Protein Binding , Tumor Necrosis Factor-alpha/metabolism
2.
Inflamm Res ; 63(7): 597-608, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24691777

ABSTRACT

OBJECTIVE: Forsythin (FOR) is an active ingredient extracted from the fruit of the medicinal plant Forsythia suspensa (Thunb.) Vahl. Here, we investigated the effect of FOR on LPS-induced inflammatory response and the underlying molecular mechanisms in RAW264.7 macrophages. MATERIALS AND METHODS: RAW264.7 cells were pre-treated with or without FOR and then stimulated with or without LPS. The productions of TNF-α, IL-1ß, IL-6, PGE2 and NO were determined by ELISA and nitrite analysis, respectively. The expressions of nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) were measured by Western blotting and RT-PCR analysis. The activations of signaling molecules were detected by Western blotting using phosphorylation specific antibodies. Reactive oxygen species (ROS) production was determined by ROS assay. RESULTS: LPS-induced productions of IL-1ß, IL-6, TNF-α, NO and PGE2 were inhibited by FOR in a dose-dependent manner. FOR also suppressed the LPS-elevated expressions of iNOS and COX-2. Further investigations revealed that FOR significantly inhibited the LPS-induced activations of JAK-STATs and p38 MAPKs, but not of IKKα/ß in LPS-stimulated RAW264.7 cells. Additionally, FOR interfered with both JAK-STATs and p38 MAPKs signaling pathways to modulate the expressions of IL-1ß, IL-6, TNF-α, iNOS and COX-2. Furthermore, FOR reduced the LPS-induced ROS accumulation, validating that FOR serves as an antioxidant. CONCLUSIONS: Our data suggested that FOR exerts anti-inflammatory action, at least in part, via suppressing LPS-induced activation of JAK-STATs and p38 MAPKs signalings and production of ROS in macrophage cells.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Furans/pharmacology , Animals , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Lipopolysaccharides , Mice , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Reactive Oxygen Species/metabolism , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Int Immunopharmacol ; 18(1): 12-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24201081

ABSTRACT

Extracellular polysaccharides (EPSs) are high-molecular weight sugar-based polymers that are synthesized and secreted by many microorganisms. Recently, EPSs have attracted particular attention due to their multiple biological functions including anti-inflammation. However, studies rarely reported the molecular mechanisms underlying their functions. We previously purified an EPS from an oligotrophic bacteria (Bacillus sp. LBP32) found in Lop Nur Desert, which possesses a potent antioxidant activity, while the anti-inflammatory effects of EPS and signaling mechanisms underlying its action have not been clarified. In this study, we demonstrated that EPS significantly inhibited the LPS-induced release of pro-inflammatory mediators, such as nitric oxide (NO), IL-6 and TNF-α, without any significant cytotoxicity. EPS also downregulated the expression of nitric oxide synthase (iNOS) induced by LPS. Furthermore, activation of nuclear factor κB (NF-κB) was abrogated by EPS through inhibited the phosphorylation of IκB kinase (IKK). Activations of Mitogen-activated protein kinases (MAPKs), including p38 MAPK and c-Jun N-terminal kinase (JNK), were also found to be inhibited by EPS. In addition, the level of intracellular reactive oxygen species (ROS) was also significantly decreased with the treatment of EPS. In vivo experiments were conducted and showed that EPS could greatly improve the outcome of mice with LPS-induced endotoxic shock. Taken together, our data indicate that EPS prevents LPS-induced inflammatory response by inhibiting NF-κB and MAPKs activation and ROS production.


Subject(s)
Bacillus/immunology , Endotoxemia/immunology , Gram-Positive Bacterial Infections/immunology , Macrophages/immunology , Polysaccharides, Bacterial/metabolism , Virulence Factors/metabolism , Animals , Bacillus/pathogenicity , Cell Line , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Immunosuppression Therapy , Inflammation Mediators/metabolism , Macrophages/microbiology , Mice , NF-kappa B/metabolism , Polysaccharides, Bacterial/immunology , Reactive Oxygen Species/metabolism , Signal Transduction , Transcriptional Activation , Virulence Factors/immunology
4.
Cell Signal ; 26(3): 594-602, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24308965

ABSTRACT

Heat shock protein 27 (HSP27) regulates critical cellular functions such as development, differentiation, cell growth and apoptosis. A variety of stimuli induce the phosphorylation of HSP27, which affects its cellular functions. However, most previous studies focused on the role of HSP27 protein itself in apoptosis, the particular role of its phosphorylation state in signaling transduction remains largely unclear. In the present study, we reported that HSP27 phosphorylation modulated TRAIL-triggered pro-survival signaling transduction. In HeLa cells, suppression of HSP27 phosphorylation by specific inhibitor KRIBB3 or MAPKAPK2 (MK2) knockdown and by overexpression of non-phosphorylatable HSP27(3A) mutant demonstrated that hindered HSP27 phosphorylation enhanced the TRAIL-induced apoptosis. In addition, reduced HSP27 phosphorylation by KRIBB3 treatment or MK2 knockdown attenuated the TRAIL-induced activation of Akt and ERK survival signaling through suppressing the phosphorylation of Src. By overexpression of HSP27(15A) or HSP27(78/82A) phosphorylation mutant, we further showed that phosphorylation of HSP27 at serine 78/82 residues was essential to TRAIL-triggered Src-Akt/ERK signaling transduction. Co-immunoprecipitation and confocal microscopy showed that HSP27 interacted with Src and scaffolding protein ß-arrestin2 in response of TRAIL stimulation and suppression of HSP27 phosphorylation apparently disrupted the TRAIL-induced interaction of HSP27 and Src or interaction of HSP27 and ß-arrestin2. We further demonstrated that ß-arrestin2 mediated HSP27 action on TRAIL-induced Src activation, which was achieved by recruiting signaling complex of HSP27/ß-arrestin2/Src in response to TRAIL. Taken together, our study revealed that HSP27 phosphorylation modulates TRAIL-triggered activation of Src-Akt/ERK pro-survival signaling via interacting with ß-arrestin2 in HeLa cells.


Subject(s)
Apoptosis/genetics , Arrestins/metabolism , HSP27 Heat-Shock Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Anisoles/pharmacology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , HSP27 Heat-Shock Proteins/genetics , HeLa Cells , Heat-Shock Proteins , Humans , Intracellular Signaling Peptides and Proteins/genetics , Isoxazoles/pharmacology , MAP Kinase Signaling System , Molecular Chaperones , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering , beta-Arrestins , src-Family Kinases/metabolism
5.
Toxicol Lett ; 221(2): 73-82, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23792430

ABSTRACT

Chemotherapy resistance represents a major problem for the treatment of patients with lung carcinomas. Parthenolide (PN), a naturally occurring small molecule found in herb feverfew, has been used in clinical treatment. Although its importance in treating the chemotherapy resistance has been shown, the pharmacological benefits of PN for lung cancer with multidrug resistance are underappreciated. Using human lung epithelial carcinoma A549 and A549 derived DOX-resistant A549/DOX cell lines, we found that PN enhanced the apoptotic cytotoxicity of DOX in A549/DOX cells. PN inhibited P-glycoprotein (P-gp) up-regulation and promoted the intracellular accumulation of DOX in A549/DOX cells. PN also exhibited inhibitory effect on NF-κB activation in A549/DOX cells, suggesting that inhibition of NF-κB was involved in attenuating P-gp expression by PN. Moreover, we found that PN could also effectively inhibit the HSP70 up-regulation in A549/DOX cells. Further studies revealed a positive correlation between HSP70 and P-gp expression. Overexpression of HSP70 upregulated P-gp expression independently of NF-κB activation in A549 cells, and knockdown of HSP70 caused a reduced expression of P-gp in A549/DOX cells. RT-PCR experiments showed that HSP70 modulated the P-gp expression mainly at transcription level. Taken together, PN can reverse DOX resistance through suppressing P-gp expression by mechanisms involving attenuation of NF-κB activation and HSP70 up-regulation. Our results not only provide insight into potential use of PN in reversing P-gp mediated MDR to facilitate lung cancer chemotherapy, but also highlight a potential role of HSP70 in the development of drug resistance.


Subject(s)
Doxorubicin/pharmacology , Drug Resistance, Neoplasm , HSP70 Heat-Shock Proteins/metabolism , NF-kappa B/metabolism , Sesquiterpenes/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , HSP70 Heat-Shock Proteins/genetics , Humans , In Situ Nick-End Labeling , Lung Neoplasms/metabolism , Microscopy, Confocal , NF-kappa B/genetics , Signal Transduction , Up-Regulation
6.
Cell Signal ; 25(5): 1176-85, 2013 May.
Article in English | MEDLINE | ID: mdl-23357534

ABSTRACT

DNA damage activates p53 and its downstream target genes, which further leads to apoptosis or survival either by the cell cycle arrest or by DNA repair. In many tumors, the heat shock protein 27 (Hsp27) is expressed at high levels to provide protection against anticancer drugs. However, the roles of Hsp27 in p53-mediated cellular responses to DNA damage are controversial. Here, we investigated the interplay between the phosphorylation status of Hsp27 and p53 in kidney 293A (HEK293A) cells and found that over-expressing phosphorylated Hsp27 mimics (Hsp27-3D) activated p53/p21 in an ATM-dependent manner. In addition, incubation with doxorubicin (Dox), an anticancer drug, induced Hsp27 phosphorylation in human adenocarcinoma cells (MCF-7). In contrast, inhibition of Hsp27 phosphorylation retarded both p53 induction and p21 accumulation, and led to cell apoptosis. Furthermore, phosphorylated Hsp27 increased p53 nuclear importing and its downstream target gene expression such as p21 and MDM2, while de-phosphorylated Hsp27 impeded this procession. Taken together, our data suggest that Hsp27, in its phosphorylated or de-phosphorylated status, plays different roles in regulating p53 pathway and cell survival.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Doxorubicin/pharmacology , HSP27 Heat-Shock Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Checkpoints/drug effects , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation/drug effects , Drug Resistance, Neoplasm , HEK293 Cells , HSP27 Heat-Shock Proteins/antagonists & inhibitors , HSP27 Heat-Shock Proteins/genetics , Humans , MCF-7 Cells , Phosphorylation , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/genetics
7.
PLoS One ; 7(11): e49194, 2012.
Article in English | MEDLINE | ID: mdl-23145121

ABSTRACT

BACKGROUND: Abnormal activity of STAT3 is associated with a number of human malignancies. Hsp90 plays a central role in stabilizing newly synthesized proteins and participates in maintaining the functional competency of a number of signaling transducers involved in cell growth, survival and oncogenesis, such as STAT3. Hsp90 interacts with STAT3 and stabilizes Tyr-phosphorylated STAT3. It has been reported that luteolin possesses anticancer activity through degradation of Tyr(705)-phosphorylated STAT3. METHODOLOGY/PRINCIPAL FINDINGS: We found that overexpression of Hsp90 inhibited luteolin-induced degradation of Tyr(705)-phosphorylated STAT3 and luteolin also reduced the levels of some other Hsp90 interacting proteins. Results from co-immunoprecipitation and immunoblot analysis demonstrated that luteolin prevented the association between Hsp90 and STAT3 and induced both Tyr(705)- and Ser(727)-phosphorylated STAT3 degradation through proteasome-dependent pathway. The molecular modeling analysis with CHARMm-Discovery Studio 2.1(DS 2.1) indicated that luteolin could bind to the ATP-binding pocket of Hsp90. SPR technology-based binding assay confirmed the association between luteolin and Hsp90. ATP-sepharose binding assay displayed that luteolin inhibited Hsp90-ATP binding. CONCLUSIONS/SIGNIFICANCE: Luteolin promoted the degradation of Tyr(705)- and Ser(727)-phosphorylated STAT3 through interacting with Hsp90 and induced apoptosis of cancer cells. This study indicated that luteolin may act as a potent HSP90 inhibitor in antitumor strategies.


Subject(s)
Carcinoma , HSP90 Heat-Shock Proteins , Luteolin/pharmacology , Neoplasms , STAT3 Transcription Factor , Animals , Apoptosis/drug effects , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , HEK293 Cells , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Hep G2 Cells , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Protein Binding , Proteolysis , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Tyrosine/metabolism , Xenograft Model Antitumor Assays
8.
Int Immunopharmacol ; 11(2): 179-86, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21094287

ABSTRACT

Curcumin, a natural polyphenolic compound, has been reported to possess anti-inflammatory properties. Previous works showed that curcumin decreased lipopolysaccharide (LPS)-induced iNOS up-regulation at transcription level. However, whether curcumin could regulate iNOS at the post-translational level is still unclear. In the present study, we demonstrated that curcumin promoted the degradation of iNOS which is expressed under LPS stimulation in murine macrophage-like RAW 264.7 cells. Mechanically, such degradation of iNOS protein is due to ubiquitination and proteasome-dependency since it was almost completely blocked by N-benzoyloxycarbonyl-Leu-Leu-leucinal (MG132), a specific inhibitor of proteasome. Furthermore, curcumin decreased iNOS tyrosine phosphorylation through inhibiting ERK 1/2 activation and subsequently suppressed iNOS enzyme activity. In conclusion, our research displays a new finding that curcumin can promote the ubiqitination and degradation of iNOS after LPS stimulation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Curcumin/pharmacology , Macrophages/drug effects , Nitric Oxide Synthase Type II/antagonists & inhibitors , Animals , Cell Culture Techniques , Cell Survival/drug effects , Enzyme-Linked Immunosorbent Assay , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Macrophages/immunology , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Ubiquitination
9.
PLoS One ; 6(12): e29169, 2011.
Article in English | MEDLINE | ID: mdl-22216199

ABSTRACT

BACKGROUND: It was well known that the clinical use of chemotherapeutic drugs is restricted by severe adverse reactions and drug resistances. Thus it is necessary to figure out a strategy to increase the specific anti-tumor efficiency of chemotherapeutic drugs. Apigenin, a kind of flavonoids, has been reported to possess anticancer activities with very low cytotoxicity to normal tissue. METHODOLOGY/PRINCIPAL FINDINGS: Our results from cell viability assay, western-blots and TdT-mediated dUTP-biotin nick end labeling (TUNEL) assay demonstrated the synergistic pro-apoptotic effects of a low dose of apigenin and paclitaxel in human cancer cell lines. To analyze the underlying mechanism, we examined reactive oxygen species (ROS) staining after cells were treated with a combination of apigenin and paclitaxel, or each of them alone. Data from flow-cytometry showed that superoxides but not reduction of peroxides accumulated in HeLa cells treated with apigenin or a combination of apigenin and paclitaxel. Apigenin and paclitaxel-induced HeLa cell apoptosis was related to the level of ROS in cells. We further evaluated activity and protein level of superoxide dismutase (SOD). Apigenin significantly inhibited SOD activity but did not alter the SOD protein level suggesting that apigenin promoted ROS accumulation through suppressing enzyme activity of SOD. Addition of Zn(2+), Cu(2+) and Mn(2+) to cell lysates inhibited apigenin's effects on SOD activity. At the same time, data from caspase-2 over-expression and knocked-down experiments demonstrated that caspase-2 participated in apigenin and paclitaxel-induced HeLa cell apoptosis. CONCLUSIONS/SIGNIFICANCE: Taken together, our study demonstrated that apigenin can sensitize cancer cells to paclitaxel induced apoptosis through suppressing SOD activity, which then led to accumulation of ROS and cleavage of caspase-2, suggesting that the combined use of apigenin and paclitaxel was an effective way to decrease the dose of paclitaxel taken.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apigenin/pharmacology , Apoptosis/drug effects , Neoplasms/pathology , Paclitaxel/pharmacology , Base Sequence , Blotting, Western , DNA Primers , Drug Synergism , Flow Cytometry , HeLa Cells , Humans , In Situ Nick-End Labeling , Neoplasms/enzymology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism
10.
Apoptosis ; 15(7): 822-33, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20349136

ABSTRACT

Inducible heat shock protein70 (HSP70) is one of the most important HSPs for maintenance of cell integrity during normal cellular growth as well as pathophysiological conditions. Apoptosis signal-regulating kinase (ASK) 1, a mammalian MAPKKK, activates the JNK and p38 pathways. Here we report a novel function of HSP70 in regulating TNF-alpha-induced cell apoptosis. Our study demonstrated that HSP70 physically interacted with ASK1 and promoted the ubiquitin-dependent proteasomal degradation of ASK1. CHIP (carboxyl terminus of the HSC70-interacting protein) which acted as a co-chaperone of HSP70 cooperated with HSP70 in regulating ASK1. We also found that TNF-alpha stimulated HSP70/CHIP/ASK1 association and through cooperating with CHIP, HSP70 inhibits TNF-alpha-induced cell apoptosis both in over-expression and RNAi conditions. Structural analysis indicated that C-terminal domain of HSP70 was necessary for ASK1 degradation, and N- terminal domain of ASK1 was essential for its binding to HSP70. All these findings indicated that HSP70 and CHIP association is important for HSP70 in interacting with ASK1. Through forming the complex of HSP70/CHIP/ASK1, HSP70 promotes ASK1 proteasomal degradation and prevents TNF-alpha-induced cell apoptosis.


Subject(s)
Apoptosis , HSP70 Heat-Shock Proteins/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Humans , MAP Kinase Kinase Kinase 5/chemistry , Protein Structure, Tertiary , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitination
11.
Cell Immunol ; 258(1): 38-43, 2009.
Article in English | MEDLINE | ID: mdl-19362296

ABSTRACT

Inducible nitric oxide synthase (iNOS) is responsible for nitric oxide (NO) synthesis from l-arginine in response to inflammatory mediators. It is reported that iNOS is degraded mainly by the ubiquitin-proteasome pathway in RAW264.7 cells and human embryonic kidney (HEK) 293 cells. In this study, we showed that iNOS was ubiquitinated and degraded dependent on CHIP (COOH terminus of heat shock protein 70-interacting protein), a chaperone-dependent ubiquitin ligase. The results from overexpression and RNAi experiments demonstrated that CHIP decreased the protein level of iNOS, shortened the half-life of iNOS and attenuated the production of NO. Furthermore, CHIP promoted ubiquitination and proteasomal degradation of iNOS by associating with iNOS. These results suggest that CHIP plays an important role in regulation iNOS activity.


Subject(s)
Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Animals , Cell Line , Humans , Mice , Plasmids , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL