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










Publication year range
1.
Int J Mol Sci ; 14(4): 7327-40, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23549267

ABSTRACT

Influenza viruses of avian origin continue to pose pandemic threats to human health. Some of the H5N1 and H9N2 virus subtypes induce markedly elevated cytokine levels when compared with the seasonal H1N1 virus. We previously showed that H5N1/97 hyperinduces tumor necrosis factor (TNF)-alpha through p38 mitogen activated protein kinase (MAPK). However, the detailed mechanisms of p38MAPK activation and TNF-alpha hyperinduction following influenza virus infections are not known. Negative feedback regulations of cytokine expression play important roles in avoiding overwhelming production of proinflammatory cytokines. Here we hypothesize that protein phosphatases are involved in the regulation of cytokine expressions during influenza virus infection. We investigated the roles of protein phosphatases including MAPK phosphatase-1 (MKP-1) and protein phosphatase type 2A (PP2A) in modulating p38MAPK activation and downstream TNF-alpha expressions in primary human monocyte-derived macrophages (PBMac) infected with H9N2/G1 or H1N1 influenza virus. We demonstrate that H9N2/G1 virus activated p38MAPK and hyperinduced TNF-alpha production in PBMac when compared with H1N1 virus. H9N2/G1 induced PP2A activity in PBMac and, with the treatment of a PP2A inhibitor, p38MAPK phosphorylation and TNF-alpha production were further increased in the virus-infected macrophages. However, H9N2/G1 did not induce the expression of PP2A indicating that the activation of PP2A is not mediated by p38MAPK in virus-infected PBMac. On the other hand, PP2A may not be the targets of H9N2/G1 in the upstream of p38MAPK signaling pathways since H1N1 also induced PP2A activation in primary macrophages. Our results may provide new insights into the control of cytokine dysregulation.


Subject(s)
Influenza, Human/enzymology , Influenza, Human/pathology , Protein Phosphatase 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Catalytic Domain , Cells, Cultured , Dual Specificity Phosphatase 1/metabolism , Enzyme Activation , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H9N2 Subtype/physiology , Influenza, Human/blood , Influenza, Human/virology , Macrophages/enzymology , Macrophages/pathology , Phosphorylation
2.
Molecules ; 16(4): 2802-16, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21455094

ABSTRACT

Panax ginseng is one of the most commonly used Chinese medicines in China, Asia and Western countries. The beneficial effects of ginseng have been attributed to the biological activities of its constituents, the ginsenosides. In this review, we summarize recent publications on the anti-inflammatory effects of ginseng extracts and ginsenosides on cellular responses triggered by different inducers including endotoxin, tumor necrosis factor-alpha (TNF-α), interferon-gamma and other stimuli. Proinflammatory cytokines, chemokines, adhesion molecules and mediators of inflammation including inducible nitric oxide synthase, cyclooxygenase-2 and nitric oxide orchestrate the inflammatory response. Ginseng extracts and ginsenosides including Rb1, Rd, Rg1, Rg3, Rh1, Rh2, Rh3 and Rp1 have been reported to have anti-inflammatory properties in different studies related to inflammation. Ginsenosides inhibit different inducers-activated signaling protein kinases and transcription factor nuclear factor-kappaB leading to decreases in the production of cytokines and mediators of inflammation. The therapeutic potential of ginseng on TNF-α-mediated inflammatory diseases is also discussed. Taken together, this summary provides evidences for the anti-inflammatory effects of ginseng extracts and ginsenosides as well as the underlying mechanisms of their effects on inflammatory diseases.


Subject(s)
Inflammation/prevention & control , Panax/chemistry , Plant Extracts/therapeutic use , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Line , Mice
3.
Cytokine ; 50(2): 210-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20171114

ABSTRACT

Expression of Epstein-Barr virus-encoded oncogenic latent membrane protein 1 (LMP1) has been substantially associated with tumorigenic transformation in the virus-infected cells. The pathogenic complexity of LMP1 is partly due to the cytokine dysregulation including IL-6 and IL-10 in perturbing the host immune responses. Here we have identified an important signaling event mediated by a dsRNA-dependent serine/threonine protein kinase, PKR, in regulating LMP1-induced IL-6 and IL-10 expression. We first demonstrated that PKR plays a significant role in mediating LMP1-induced cytokine expression by using a PKR inhibitor 2-aminopurine, and the specific role of PKR involved was confirmed by the use of siRNA oligos targeting PKR and/or a dominant-negative PKR mutant. We next revealed that PKR activity mediates LMP1-enhanced NF-kappaB nuclear translocation resulting in cytokine induction. We further demonstrated at the chromatin level that LMP1 can significantly elevate the phosphorylation of histone H3 on serine 10 (Ser 10), and the process was dependent on PKR activity. Our findings thus suggest that PKR plays an important role in mediating the cytokine gene expression induced by LMP1 through NF-kappaB activation and histone H3 Ser 10 phosphorylation.


Subject(s)
Interleukin-10/genetics , Interleukin-6/genetics , Viral Matrix Proteins/metabolism , eIF-2 Kinase/metabolism , Cell Line , Enzyme Activation/drug effects , Eukaryotic Initiation Factor-2B/metabolism , Histones/metabolism , Humans , Interleukin-10/metabolism , Interleukin-6/metabolism , Models, Biological , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
4.
J Med Chem ; 52(9): 2667-72, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19419201

ABSTRACT

Computational molecular docking provides an efficient and innovative approach to examine small molecule and protein interactions. We have utilized this method to identify potential inhibitors of the H5N1 neuraminidase protein. Of the 20 compounds tested, 4-(4-((3-(2-amino-4-hydroxy-6-methyl-5-pyrimidinyl)propyl)amino)phenyl)-1-chloro-3-buten-2-one (1) (NSC89853) demonstrated the ability to inhibit viral replication at a level comparable to the known neuraminidase inhibitor oseltamivir. Compound 1 demonstrated efficacy across a number of cell-lines assays and in both the H1N1 and H5N1 viruses. The predicted binding of 1 to the known H5N1 neuraminidase structure indicates a binding interface largely nonoverlapping with that of oseltamivir or another neuraminidase inhibitor zanamivir. These results indicate that 1 or similar molecules would remain effective in the presence of virus mutations conferring resistance to either oseltamivir or zanamivir and also vice versa.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H5N1 Subtype/drug effects , Influenza in Birds/virology , Models, Molecular , Neuraminidase/antagonists & inhibitors , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Birds , Cell Line , Computational Biology , Computer Simulation , Drug Evaluation, Preclinical , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/enzymology , Molecular Conformation , Neuraminidase/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/metabolism , Reproducibility of Results , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
5.
J Transl Med ; 7: 34, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19442267

ABSTRACT

BACKGROUND: Ginseng is believed to have beneficial effects against human diseases, and its active components, ginsenosides, may play critical roles in its diverse physiological actions. However, the mechanisms underlying ginseng's effects remain to be investigated. We hypothesize some biological effects of ginseng are due to its anti-inflammatory effects. METHODS: Human promonocytic U937 cells were used to investigate the immunomodulatory effects of ginseng following TNF-alpha treatment. A global gene expression profile was obtained by using genechip analysis, and specific cytokine expression was measured by quantitative RT-PCR and ELISA. HPLC was used to define the composition of ginsenosides in 70% ethanol-water extracts of ginseng. Activation of signalling kinases was examined by Western blot analysis. RESULTS: Seventy percent ethanol-water extracts of ginseng significantly inhibited the transcription and secretion of CXCL-10 following TNF-alpha stimulation. Nine ginsenosides including Rb1, Rb2, Rc, Rd, Re, Rf, Rg1, Rg3 and Rh1 were identified in our extract by HPLC. Seven out of nine ginsenosides could significantly inhibit TNF-alpha-induced CXCL-10 expression in U937 cells and give comparable inhibition of CXCL-10 transcription to those with the extract. However, the CXCL-10 suppressive effect of individual ginsenosides was less than that of the crude extract or the mixture of ginsenosides. The CXCL-10 suppression can be correlated with the inactivation of ERK1/2 pathways by ginseng. CONCLUSION: We showed ginseng suppressed part of the TNF-alpha-inducible cytokines and signalling proteins in promonocytic cells, suggesting that it exerts its anti-inflammatory property targeting at different levels of TNF-alpha activity. The anti-inflammatory role of ginseng may be due to the combined effects of ginsenosides, contributing in part to the diverse actions of ginseng in humans.


Subject(s)
Immunologic Factors/pharmacology , Panax/chemistry , Plant Extracts/pharmacology , Signal Transduction/drug effects , Chemokine CXCL10/metabolism , Chromatography, High Pressure Liquid , Culture Media , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Ginsenosides/analysis , Ginsenosides/pharmacology , Humans , Interleukin-8/metabolism , Mitogen-Activated Protein Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
6.
Blood ; 113(21): 5192-201, 2009 May 21.
Article in English | MEDLINE | ID: mdl-19279332

ABSTRACT

HIV infection remains a worldwide threat. HIV-1 transactivator protein Tat is one of the retroviral proteins identified as a key immunomodulator in AIDS pathogenesis. Although the primary function of Tat is to regulate HIV-1 replication in the infected cell, it also dysregulates cytokine production resulting in perturbation of the host immune response and enhancement of the retrovirus survival. Because interferon-gamma (IFNgamma) is a pleiotropic cytokine with potent antiviral and immunoregulatory effects, we investigated whether Tat interferes with the IFNgamma signal transduction in primary monocytes. We demonstrated that Tat impaired the IFNgamma-receptor signaling pathway at the level of STAT1 activation, possibly via Tat-dependent induction of suppressor of cytokine signaling-2 (SOCS-2) activity. We delineated the inhibitory role of SOCS-2 in IFNgamma signaling pathway by overexpression of exogenous SOCS-2 in HEK293 cell. The results showed that SOCS-2 suppressed the IFNgamma-activated STAT1 phosphorylation and consequent IFNgamma-regulated transcription of specific genes. To confirm the role of SOCS2 in the Tat-induced process, we demonstrated that SOCS-2 siRNA in human blood monocytes abrogated the Tat-dependent inhibition of IFNgamma signaling. Our data suggested a possible mechanism implicating the role of SOCS-2 in mediating HIV-1-induced immune evasion and dysregulation of IFNgamma signaling in primary human monocytes.


Subject(s)
HIV-1/pathogenicity , Interferon-gamma/metabolism , Monocytes/virology , Suppressor of Cytokine Signaling Proteins/genetics , Transcriptional Activation , tat Gene Products, Human Immunodeficiency Virus/physiology , Cells, Cultured , Humans , Immunity , STAT1 Transcription Factor/metabolism , Signal Transduction
7.
J Gen Virol ; 88(Pt 4): 1275-1280, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17374772

ABSTRACT

Pathogenesis of the highly pathogenic avian influenza virus A/Hong Kong/483/97 (H5N1/97) remains to be investigated. It was demonstrated recently that H5N1 dysregulation of proinflammatory cytokines in human macrophages is a p38-kinase-dependent process. The results indicated that macrophages may play a role in disease severity. To investigate cellular responses to H5N1 infection further, apoptosis and its related pathways were studied in primary blood macrophages. Here, it is shown that the H5N1/97 virus triggered apoptosis, including caspases and PARP activation, in infected macrophages with a delayed onset compared with H1N1 counterparts. Similar results were also found in human macrophages infected by precursors of the H5N1/97 virus. Thus, these results showed that the delay in apoptosis onset in macrophages infected by H5N1/97 and its related precursor subtypes may be a means for the pathogens to have longer survival in the cells; this may contribute to the pathogenesis of H5N1 disease in humans.


Subject(s)
Apoptosis , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Macrophages/cytology , Macrophages/virology , Animals , Caspases/biosynthesis , Cells, Cultured , Collagen Type XI/metabolism , Humans , Influenza, Human/virology , Macrophages/immunology , Macrophages/metabolism
8.
J Virol ; 81(1): 416-22, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17035307

ABSTRACT

Severe acute respiratory syndrome (SARS) is an emerging infectious disease caused by a novel coronavirus. Since its associated morbidity and mortality have been postulated to be due to immune dysregulation, we investigated which of the viral proteins is responsible for chemokine overexpression. To delineate the viral and cellular factor interactions, the role of four SARS coronavirus proteins, including nonstructural protein 1 (nsp-1), nsp-5, envelope, and membrane, were examined in terms of cytokine induction. Our results showed that the SARS coronavirus nsp-1 plays an important role in CCL5, CXCL10, and CCL3 expression in human lung epithelial cells via the activation of NF-kappaB.


Subject(s)
Chemokines/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Viral Nonstructural Proteins/metabolism , Chemokine CCL3 , Chemokine CCL5 , Chemokine CXCL10 , Chemokines/metabolism , Chemokines, CC/genetics , Chemokines, CC/metabolism , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Gene Expression Regulation , NF-kappa B/genetics , NF-kappa B/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/virology , Severe acute respiratory syndrome-related coronavirus/metabolism , Viral Envelope Proteins/physiology , Viral Matrix Proteins/physiology
9.
J Immunol ; 175(11): 7218-25, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16301626

ABSTRACT

Following infection of the host by Mycobacterium tuberculosis, induction of cytokines is a major defense mechanism to limit the pathogen invasion. Cytokines interact with each other to form an intertwined network of pathways. For example, IFN and TNF have been shown to interact through common pathways including IFN-inducible, dsRNA-activated serine/threonine protein kinase (PKR) induction. As a signal transducer, it has been conventionally known to regulate the induction of cytokine expression in response to virus infection through NF-kappaB. In light of the critical role of TNF in immunity and its cytotoxic effects mediated by PKR, we examined the role of the kinase in the regulation of immune response against M. tuberculosis using the interaction of bacillus Calmette-Guérin (BCG) and primary human blood monocytes as a model. Our results showed that BCG stimulates the induction of cytokine expression in human primary blood monocytes including TNF-alpha, IL-6, and IL-10. With the suppression of PKR by using PKR-mutant gene or 2-aminopurine as PKR inhibitor, we showed that the BCG-induced cytokine expression in human monocytes is regulated by the phosphorylation and activation of PKR. We also demonstrated that downstream of PKR induction is the activation of MAPK and translocation of NF-kappaB into the nucleus. NF-kappaB in turn mediates the transcription of specific cytokine genes. Taken together, PKR plays a critical role in the regulation of immune responses to mycobacterial infection and may serve as an important molecule in the innate antimycobacterial defense.


Subject(s)
Cytokines/biosynthesis , Monocytes/immunology , Monocytes/microbiology , Mycobacterium Infections/immunology , eIF-2 Kinase/metabolism , Animals , Blotting, Western , Electrophoretic Mobility Shift Assay , Gene Expression , Humans , MAP Kinase Signaling System/immunology , Mycobacterium bovis/immunology , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , U937 Cells , eIF-2 Kinase/immunology
10.
J Virol ; 79(16): 10147-54, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16051807

ABSTRACT

Avian influenza A virus subtype H5N1 can infect humans to cause a severe viral pneumonia with mortality rates of more than 30%. The biological basis for this unusual disease severity is not fully understood. We previously demonstrated that in contrast to human influenza A virus subtypes including H1N1 or H3N2, the H5N1 virus associated with the "bird flu" outbreak in Hong Kong in 1997 (H5N1/97) hyperinduces proinflammatory cytokines, including tumor necrosis factor alpha (TNF-alpha), in primary human macrophages in vitro. To delineate the molecular mechanisms involved, we analyzed the role of transcription factor NF-kappaB and cellular kinases in TNF-alpha dysregulation. H5N1 and H1N1 viruses did not differ in the activation of NF-kappaB or degradation of IkappaB-alpha in human macrophages. However, we demonstrated that unlike H1N1 virus, H5N1/97 strongly activates mitogen-activated protein kinase (MAPK), including p38 MAPK and extracellular signal-regulated kinases 1 and 2. Specific inhibitors of p38 MAPK significantly reduced the H5N1/97-induced TNF-alpha expression in macrophages. Taken together, our findings suggest that H5N1/97-mediated hyperinduction of cytokines involves the p38 MAPK signaling pathway. These results may provide insights into the pathogenesis of H5N1 disease and rationales for the development of novel therapeutic strategies.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza A virus/pathogenicity , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/metabolism , Membrane Glycoproteins/physiology , NF-kappa B/metabolism , Phosphorylation , Receptors, Cell Surface/physiology , Toll-Like Receptors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
11.
FEBS Lett ; 579(14): 3055-62, 2005 Jun 06.
Article in English | MEDLINE | ID: mdl-15907845

ABSTRACT

HIV Tat has been known to have multiple regulatory roles including replication of HIV and modulation of cellular kinases. We investigated whether signaling kinase PKR plays a critical role in mediating Tat-induced cytokine dysregulation. We showed Tat induction of IL-10 dysregulation is associated with PKR activation. To examine the mechanism involved, inhibition of PKR activity abrogated the Tat-induced cytokine induction. We next identified that the MAP kinases including ERK-1/2 and p38 are downstream of PKR in these Tat-induced pathways. Thus, PKR may play a critical role in mediating the subversive effects of HIV Tat resulting in IL-10 induction.


Subject(s)
Cytokines/genetics , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Products, tat/metabolism , MAP Kinase Signaling System , eIF-2 Kinase/immunology , eIF-2 Kinase/metabolism , CCAAT-Enhancer-Binding Protein-delta , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Cytokines/biosynthesis , Cytokines/immunology , Dose-Response Relationship, Drug , HIV-1 , Humans , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-10/metabolism , Leukocytes/metabolism , NF-kappa B/metabolism , Time Factors , Transcription Factors/metabolism , Up-Regulation , eIF-2 Kinase/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...