Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 3299, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34083537

ABSTRACT

Bioenergetic perturbations driving neoplastic growth increase the production of reactive oxygen species (ROS), requiring a compensatory increase in ROS scavengers to limit oxidative stress. Intervention strategies that simultaneously induce energetic and oxidative stress therefore have therapeutic potential. Phenformin is a mitochondrial complex I inhibitor that induces bioenergetic stress. We now demonstrate that inflammatory mediators, including IFNγ and polyIC, potentiate the cytotoxicity of phenformin by inducing a parallel increase in oxidative stress through STAT1-dependent mechanisms. Indeed, STAT1 signaling downregulates NQO1, a key ROS scavenger, in many breast cancer models. Moreover, genetic ablation or pharmacological inhibition of NQO1 using ß-lapachone (an NQO1 bioactivatable drug) increases oxidative stress to selectively sensitize breast cancer models, including patient derived xenografts of HER2+ and triple negative disease, to the tumoricidal effects of phenformin. We provide evidence that therapies targeting ROS scavengers increase the anti-neoplastic efficacy of mitochondrial complex I inhibitors in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Phenformin/pharmacology , STAT1 Transcription Factor/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Synergism , Electron Transport Complex I/antagonists & inhibitors , Energy Metabolism/drug effects , Female , Glutathione/antagonists & inhibitors , Glutathione/biosynthesis , Humans , Interferon-gamma/administration & dosage , Interferon-gamma/deficiency , Interferon-gamma/metabolism , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/administration & dosage , Oxidative Stress/drug effects , Phenformin/administration & dosage , Poly I-C/administration & dosage , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor/agonists , Xenograft Model Antitumor Assays
2.
J Interferon Cytokine Res ; 38(6): 261-271, 2018 06.
Article in English | MEDLINE | ID: mdl-29762078

ABSTRACT

Interferon-α (IFN-α) resistance is a major hurdle in the treatment of hepatocellular carcinoma (HCC). Signal transducers and activators of transcription 1 (STAT1) play a key role in exerting the antiproliferative and proapoptotic effects of IFN-α on tumors. In this study, we aimed to investigate whether resveratrol can promote IFN-α-induced growth inhibition and the apoptosis on HCC cells through the SIRT/STAT1 pathway. We found that IFN-α induced growth inhibition and apoptosis of SMMC7721 cells, and the effects could be significantly enhanced and blocked by resveratrol and EX527, respectively. Resveratrol not only activated SIRT1 but also induced phosphorylation of STAT1. Further study revealed that ablation of STAT1 reduced the combined antitumor effects of IFN-α and resveratrol, lowered the rate of apoptosis, and improved the viability of SMMC7721 cells. Whereas STAT1 overexpression strengthened the combined antitumor effects of resveratrol and IFN-α. Our findings suggest a novel strategy of using resveratrol to enhance the response of HCC to IFN-α treatment through the SIRT/STAT1 pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Interferon-alpha/pharmacology , Resveratrol/pharmacology , STAT1 Transcription Factor/metabolism , Sirtuins/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , STAT1 Transcription Factor/agonists , Signal Transduction/drug effects
3.
Mol Med Rep ; 16(5): 6405-6411, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28901399

ABSTRACT

Macrophage polarization is flexible, and involves in different signaling pathways and various transcription factors. Suppressor of cytokine signaling (SOCS) is an important inhibitor of cytokine signaling pathways and also a key physiological regulator for natural and acquired immunity systems. Following transfection of SOCS1 short hairpin (sh)RNA into mouse macrophage cells, reverse transcription­quantitative polymerase chain reaction demonstrated that the mRNA levels of Janus kinase (JAK)1 and signal transducer and activator of transcription (STAT)1 increased significantly. In addition, western blotting indicated that JAK1, STAT1 and p­STAT1 expression was significantly enhanced. Fludarabine can inhibit phosphorylation of STAT1 and SOCS1 expression. When fludarabine was added and SOCS1 shRNA was transfected, the inhibition of fludarabine was weakened, and p­STAT1 expression was upregulated. Flow cytometry detection indicated that, following the downregulation of SOCS1 expression, M1­type cells significantly increased, but the proportion of M2­type cells did not change significantly. Fludarabine can reduce the effect of SOCS1 shRNA on promoting M1­type cell polarization, and macrophages can polarize into both M1 and M2 phenotypes. Further ELISA results presented that, when downregulating SOCS1 expression, interleukin (IL)­4 and IL­10 expression was both downregulated, and tumor necrosis factor (TNF)­α and interferon (IFN)­Î³ expression was significantly upregulated. When adding fludarabine or injecting with the traditional Chinese medicine Xuebijing, IL­4 and IL­10 expression was both significantly upregulated, and TNF­α and IFN­Î³ expression was significantly downregulated. When adding fludarabine and downregulating SOCS1, IL­4, IL­10, TNF­α and IFN­Î³ expression presented no significant changes. The above results indicated that, when SOCS1 expression is downregulated, it will activate the JAK1/STAT1 pathway, and thereby promote the polarization of macrophages into M1 type. The findings are of great importance for understanding occurrence, development and treatment of various immune­related diseases.


Subject(s)
Janus Kinase 1/immunology , Macrophages, Peritoneal/immunology , STAT1 Transcription Factor/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Cell Differentiation , Drugs, Chinese Herbal/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Janus Kinase 1/genetics , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Mice , Phosphorylation , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/genetics , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors , Suppressor of Cytokine Signaling 1 Protein/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vidarabine/analogs & derivatives , Vidarabine/antagonists & inhibitors , Vidarabine/pharmacology
4.
J Biol Chem ; 292(24): 10048-10060, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28432122

ABSTRACT

IFNs are effective in inhibiting angiogenesis in preclinical models and in treating several angioproliferative disorders. However, the detailed mechanisms of IFNα-mediated anti-angiogenesis are not completely understood. Stat1/2/3 and PML are IFNα downstream effectors and are pivotal regulators of angiogenesis. Here, we investigated PML's role in the regulation of Stat1/2/3 activity. In Pml knock-out (KO) mice, ablation of Pml largely reduces IFNα angiostatic ability in Matrigel plug assays. This suggested an essential role for PML in IFNα's anti-angiogenic function. We also demonstrated that PML shared a large cohort of regulatory genes with Stat1 and Stat3, indicating an important role of PML in regulating Stat1 and Stat3 activity. Using molecular tools and primary endothelial cells, we demonstrated that PML positively regulates Stat1 and Stat2 isgylation, a ubiquitination-like protein modification. Accordingly, manipulation of the isgylation system by knocking down USP18 altered IFNα-PML axis-mediated inhibition of endothelial cell migration and network formation. Furthermore, PML promotes turnover of nuclear Stat3, and knockdown of PML mitigates the effect of LLL12, a selective Stat3 inhibitor, on IFNα-mediated anti-angiogenic activity. Taken together, we elucidated an unappreciated mechanism in which PML, an IFNα-inducible effector, possess potent angiostatic activity, doing so in part by forming a positive feedforward loop with Stat1/2 and a negative feedback loop with Stat3. The interplay between PML, Stat1/Stat2, and Stat3 contributes to IFNα-mediated inhibition of angiogenesis, and disruption of this network results in aberrant IFNα signaling and altered angiostatic activity.


Subject(s)
Endothelium, Vascular/metabolism , Interferon-alpha/metabolism , Neovascularization, Pathologic/prevention & control , Promyelocytic Leukemia Protein/metabolism , STAT1 Transcription Factor/agonists , STAT2 Transcription Factor/agonists , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Cell Line , Cells, Cultured , Endopeptidases/chemistry , Endopeptidases/genetics , Endopeptidases/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interferon-alpha/genetics , Mice, Knockout , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic , Promyelocytic Leukemia Protein/antagonists & inhibitors , Promyelocytic Leukemia Protein/genetics , Protein Processing, Post-Translational , RNA Interference , Recombinant Proteins/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
5.
Mol Cancer Ther ; 14(9): 2103-11, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26141950

ABSTRACT

Cisplatin is a cytotoxic chemotherapeutic drug frequently used to treat many solid tumors, including head and neck squamous cell carcinoma (HNSCC). EGF receptor (EGFR) inhibitors have also shown efficacy as alternatives to cisplatin in some situations. However, large clinical trials have shown no added survival benefit from the use of these two drugs in combination. Possible explanations for this include overlapping downstream signaling cascades. Using in vitro studies, we tested the hypothesis that cisplatin and EGFR inhibitors rely on the activation of the tumor suppressor STAT1, characterized by its phosphorylation at serine (S727) or tyrosine (Y701) residues. Cisplatin consistently increased the levels of p-S727-STAT1, and STAT1 siRNA knockdown attenuated cisplatin-induced cell death. EGFR stimulation also activated p-S727-STAT1 and p-Y701-STAT1 in a subset of cell lines, whereas EGFR inhibitors alone decreased levels of p-S727-STAT1 and p-Y701-STAT1 in these cells. Contrary to our hypothesis, EGFR inhibitors added to cisplatin treatment caused variable effects among cell lines, with attenuation of p-S727-STAT1 and enhancement of cisplatin-induced cell death in some cells and minimal effect in other cells. Using HNSCC tumor specimens from a clinical trial of adjuvant cisplatin plus the anti-EGFR antibody panitumumab, higher intratumoral p-S727-STAT1 appeared to correlate with worse survival. Together, these results suggest that cisplatin-induced cell death is associated with STAT1 phosphorylation, and the addition of anti-EGFR therapy to cisplatin has variable effects on STAT1 and cell death in HNSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Cisplatin/pharmacology , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , STAT1 Transcription Factor/metabolism , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Cetuximab/pharmacology , Cisplatin/administration & dosage , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Gene Knockdown Techniques , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Panitumumab , Phosphorylation , Prognosis , RNA, Small Interfering/genetics , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/genetics , Squamous Cell Carcinoma of Head and Neck , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Diabetes ; 64(12): 4285-97, 2015 12.
Article in English | MEDLINE | ID: mdl-25858560

ABSTRACT

Activation of AMPK suppresses inflammation, but the underlying mechanisms remain poorly understood. This study was designed to characterize the molecular mechanisms by which AMPK suppresses vascular inflammation. In cultured human aortic smooth muscle cells, pharmacologic or genetic activation of AMPK inhibited the signal transducer and activator of transcription-1 (STAT1), while inhibition of AMPK had opposite effects. Deletion of AMPKα1 or AMPKα2 resulted in activation of STAT1 and in increases in proinflammatory mediators, both of which were attenuated by administration of STAT1 small interfering RNA or fludarabine, a selective STAT1 inhibitor. Moreover, AMPK activation attenuated the proinflammatory actions induced by STAT1 activators such as interferon-γ and angiotensin II (AngII). Mechanistically, we found that AMPK activation increased, whereas AMPK inhibition decreased, the levels of mitogen-activated protein kinase phosphatase-1 (MKP-1), an inducible nuclear phosphatase, by regulating proteasome-dependent degradation of MKP-1. Gene silencing of MKP-1 increased STAT1 phosphorylation and prevented 5-aminoimidazole-4-carboxyamide ribonucleoside-reduced STAT1 phosphorylation. Finally, we found that infusion of AngII caused a more severe inflammatory response in AMPKα2 knockout mouse aortas, all of which were suppressed by chronic administration of fludarabine. We conclude that AMPK activation suppresses STAT1 signaling and inhibits vascular inflammation through the upregulation of MKP-1.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Dual Specificity Phosphatase 1/metabolism , Muscle, Smooth, Vascular/metabolism , STAT1 Transcription Factor/metabolism , Vasculitis/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Angiotensin II/adverse effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aorta, Thoracic , Cells, Cultured , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/chemistry , Dual Specificity Phosphatase 1/genetics , Enzyme Activation/drug effects , Humans , Interferon-gamma/adverse effects , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA Interference , Random Allocation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , Vasculitis/chemically induced , Vasculitis/immunology , Vasculitis/pathology
7.
Am J Physiol Endocrinol Metab ; 307(9): E847-55, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25205818

ABSTRACT

Growth hormone (GH) plays a key role regulating somatic growth and in controlling metabolism and other physiological processes in humans and other animal species. GH acts by binding to the extracellular part of its transmembrane receptor, leading to induction of multiple intracellular signal transduction pathways that culminate in changes in gene and protein expression. A key agent in GH-stimulated growth is the latent transcription factor signal transducer and activator of transcription (STAT) 5B, one of four STAT proteins induced by the GH receptor in cultured cells and in vivo. As shown by genetic and biochemical studies, GH-activated STAT5B promotes transcription of the gene encoding the critical growth peptide, insulin-like growth factor-I (IGF-I), and natural null mutations of STAT5B in humans lead to growth failure accompanied by diminished IGF-I expression. Here we have examined the possibility that other GH-activated STATs can enhance IGF-I gene transcription, and thus potentially contribute to GH-regulated somatic growth. We find that human STAT5A is nearly identical to STAT5B in its biochemical and functional responses to GH but that STAT1 and STAT3 show a weaker profile of in vitro binding to STAT DNA elements from the IGF-I gene than STAT5B, and are less potent inducers of gene transcription through these elements. Taken together, our results offer a molecular explanation for why STAT5B is a key in vivo mediator of GH-activated IGF-I gene transcription and thus of GH-regulated somatic growth.


Subject(s)
Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , STAT1 Transcription Factor/agonists , STAT3 Transcription Factor/agonists , STAT5 Transcription Factor/agonists , Tumor Suppressor Proteins/agonists , Up-Regulation , Animals , Base Sequence , COS Cells , Cell Nucleus , Chlorocebus aethiops , Conserved Sequence , Humans , Insulin-Like Growth Factor I/genetics , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Response Elements , STAT1 Transcription Factor/chemistry , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/chemistry , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Sequence Homology, Amino Acid , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
Chem Pharm Bull (Tokyo) ; 61(10): 1002-8, 2013.
Article in English | MEDLINE | ID: mdl-24088692

ABSTRACT

Jasmonates are lipid-based stress hormones that are critical for the defense of plants against insects. Two naturally occurring jasmonates, jasmonic acid and methyl jasmonate, have recently been explored for their efficacy as anti-cancer agents. Furthermore, certain synthetic jasmonates (e.g., the cyclopentenone isoprostane J2) exert anti-inflammatory actions in lipopolysaccharide (LPS)-challenged murine macrophages via down-regulation of chemokines and other inflammatory mediators. Chemokines participate in the development and progression of many inflammatory disorders, such as atopic dermatitis (AD) and Crohn's disease, as exemplified by the role of macrophage-derived chemokine (MDC/CCL22) in the pathology of AD. The current study therefore investigated the impact of jasmonate derivatives (jasmonic acid and methyl jasmonate) and their synthetic analogues (J2 and J7) on the expression of MDC in interferon (IFN)-γ- and tumor necrosis factor (TNF)-α-stimulated HaCaT human keratinocytes, as well as the attendant mechanism of action. Jasmonic acid, methyl jasmonate, and J2 failed to inhibit the cytokine-stimulated production of MDC. By contrast, J7 suppressed the mRNA and protein expression levels of MDC in a dose-dependent manner. Moreover, J7 diminished the activation of signal transducers and activators of transcription 1 (STAT1), but had no inhibitory effect on the nuclear factor kappa B (NF-κB) or mitogen-activated protein kinase (MAPK) pathways. These results demonstrate that J7 impairs IFN-γ- and TNF-α-induced inflammatory chemokine production by targeting the STAT1 pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/toxicity , Chemokines/metabolism , Cyclopentanes/chemistry , Cyclopentanes/pharmacology , Down-Regulation/drug effects , Fatty Acids, Unsaturated/chemistry , Oxylipins/pharmacology , STAT1 Transcription Factor/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Antineoplastic Agents/chemistry , Cell Line , Cell Survival/drug effects , Chemokines/genetics , Fatty Acids, Unsaturated/pharmacology , Humans , Inflammation Mediators/metabolism , Interferon-gamma/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Lipopolysaccharides/toxicity , Macrophages/drug effects , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oxylipins/chemistry , RNA, Messenger/metabolism , STAT1 Transcription Factor/agonists , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
9.
Biochem Biophys Res Commun ; 426(2): 242-6, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22935418

ABSTRACT

Signal transducer and activator of transcription (STAT) proteins are key signaling molecules in response to cytokines and in regulating T cell biology. However, there are contradicting reports on whether STAT is involved in T-cell antigen receptor (TCR) signaling. To better define the role of STAT in TCR signaling, we activated the CD4/CD8-associated Lck kinase by co-crosslinking TCR and CD4/CD8 co-receptors in human peripheral blood T cells. Sequential STAT1, STAT3 and STAT5 activation was observed 1 h after TCR stimulation suggesting that STAT proteins are not the immediate targets in the TCR complex. We further identified interferon-γ as the key cytokine in STAT1 activation upon TCR engagement. In contrast to transient STAT activation in cytokine response, this autocrine/paracrine-induced STAT activation was sustained. It correlated with the absence of two suppressors of cytokine signaling (SOCS) proteins, SOCS3 and cytokine-inducible SH2 containing protein that are negative feedback regulators of STAT signaling. Moreover, enforced expression of SOCS3 inhibited tyrosine phosphorylation of zeta-associated protein kinase of 70 kD in TCR-stimulated human Jurkat T cells. This is the first report demonstrating delayed and prolonged STAT activation coordinated with the loss of SOCS expression in human primary T cells after co-crosslinking of TCR and CD4/CD8 co-receptors.


Subject(s)
Autocrine Communication/immunology , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Paracrine Communication/immunology , Receptors, Antigen, T-Cell/metabolism , STAT Transcription Factors/metabolism , T-Lymphocytes/immunology , Gene Expression , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Phosphorylation , Receptors, Antigen, T-Cell/agonists , STAT Transcription Factors/agonists , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/agonists , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/agonists , STAT5 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Tyrosine/metabolism
10.
Biochem Biophys Res Commun ; 391(3): 1358-62, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20026063

ABSTRACT

Beta-glucans are heterogeneous groups of glucose polymers found in the cell walls of fungi, plants and some bacteria. Our previous report showed that a novel beta-1,3/1,6-glucan produced from Paenibacillus (P.) polymyxa JB115 can induce nitric oxide (NO) production in RAW264.7 cells. In the present study, the beta-glucan significantly increased luciferase activity in cells transfected with NFkappaB or AP1, but not STAT1, reporter vector DNA, which contain their binding promoter site. All specific NFkappaB and MAPKs pathway inhibitors (pyrrolidine dithiocarbamate, AG490, U0126, SB203580 and SP600125) remarkably attenuated NO production induced by the beta-glucan. Furthermore, Western blot analysis revealed that the stimulation of Raw264.7 cells by beta-glucan induced phosphorylation of IkappaB and the consequent translocation of NFkappaB into the nucleus. Meanwhile, phosphorylation of ERK1/2, JNK/SAPK and p38 MAPKs in cytoplasm were also confirmed. All these results indicated that beta-glucan from P. polymyxa JB115 activates macrophages through MAPKs and NFkappaB signaling pathway.


Subject(s)
Macrophage Activation , Macrophages/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/agonists , Paenibacillus/chemistry , beta-Glucans/pharmacology , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Nucleus/metabolism , I-kappa B Proteins/metabolism , Macrophages/enzymology , Macrophages/immunology , Mice , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Phosphorylation , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/metabolism , Transcription Factor AP-1/agonists , Transcription Factor AP-1/metabolism , beta-Glucans/isolation & purification
11.
Immunology ; 128(1 Suppl): e816-25, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19740343

ABSTRACT

Interleukin-27 (IL-27) is a new IL-12-related heterodimeric cytokine comprising a novel p28 molecule and the Epstein-Barr-virus-induced gene 3 (EBI3) molecules. It augments initiation of T helper type 1-mediated immunity by enhancing the proliferation and cytokine production of T cells. In this study, we examined whether a secreted form of IL-27 subunits would inhibit IL-27-mediated immunological responses. COS-7 cells transduced with the mouse (m) p28 gene secreted a monomeric mp28 protein; however, those transduced with the mEBI3 gene did not detect a mEBI3 protein in the culture supernatants. The secreted mp28 prevented the IL-27-mediated signal transduction and activator of transcription 1 phosphorylation and subsequently inhibited the IL-27-mediated intercellular adhesion molecule-1 induction and interferon-gamma production in CD4(+) T cells. We generated mp28-expressing murine carcinoma Colon 26 cells and inoculated a mixture of the mp28- and mIL-27-expressing Colon 26 cells into syngeneic BALB/c mice. Simultaneous production of mp28 and mIL-27 from Colon 26 cells suppressed IL-27-mediated anti-tumour effects in the mice. We examined the p28-mediated immune suppression by inoculating mp28-expressing myoblasts into allogeneic mice. Forced production of mp28 suppressed the allogeneic cytotoxic T-lymphocyte induction and subsequently retarded the graft rejection. Furthermore, production of both mp28 and mp40, which inhibits the functions of IL-12 and IL-23, prolonged the graft survival longer than the grafts expressing either mp28 or mp40. We propose that p28 can be a regulatory subunit for IL-27-mediated cellular immune responses and a possible therapeutic agent to suppress unfavourable immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Graft Survival/immunology , Interleukin-17/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , COS Cells , Cell Adhesion Molecules/agonists , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Female , Interferon-gamma/biosynthesis , Interferon-gamma/drug effects , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Myoblasts/immunology , Myoblasts/metabolism , Phosphorylation , Protein Subunits/genetics , Protein Subunits/immunology , Protein Subunits/pharmacology , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , Receptors, Interleukin-12/agonists , Receptors, Interleukin-12/immunology , Receptors, Interleukin-12/metabolism , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Transduction, Genetic
12.
Immunology ; 128(1 Suppl): e905-18, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19740351

ABSTRACT

The relationship between the p53 signal pathway and the response of human peripheral blood mononuclear cells (PBMC) to interferon (IFN)-beta has hitherto not been examined. Using an oligonucleotide microarray, we found differential expression of at least 70 genes involved in the p53 signal pathway, including p53, which regulate cell proliferation and cell death following stimulation with IFN-beta. We verified our observations on a limited set of p53-regulated genes at the transcriptional and translational levels. We also examined the consequences of the activation of the p53 signal pathway by IFN-beta in PBMC. When cultured in the presence of T-cell mitogens, IFN-beta restricted the entry of lymphocytes from the G0/G1 phase to the S phase and reduced the number of cells in the G2 phase. The addition of IFN-beta alone did not increase apoptosis. However, in the presence of actinomycin D, a DNA-damaging agent, addition of IFN-beta enhanced the susceptibility of PBMC to apoptosis. These observations suggest that, in spite of the activation of a number of mutually overlapping pathways mediating cell death, cell cycle arrest was the most evident consequence of IFN-beta signalling in PBMC.


Subject(s)
Gene Expression Regulation , Interferon-beta/metabolism , Leukocytes, Mononuclear/immunology , Tumor Suppressor Protein p53/metabolism , Adult , Apoptosis/genetics , Apoptosis/immunology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle/immunology , Female , Humans , Interferon-beta/pharmacology , Leukocytes, Mononuclear/drug effects , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/agonists , STAT2 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics
13.
Eur J Immunol ; 39(9): 2525-36, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19670379

ABSTRACT

IFN-beta currently serves as one of the major treatments for MS. Its anti-inflammatory mechanism has been reported as involving a shift in cytokine balance from Th1 to Th2 in the T-cell response against elements of the myelin sheath. In addition to the Th1 and Th2 groups, two other important pro-inflammatory cytokines, IL-17 and osteopontin (OPN), are believed to play important roles in CNS inflammation in the pathogenesis of MS. In this study, we examined the potential effects of IFN-beta on the regulation of OPN and IL-17 in MS patients. We found that IFN-beta used in vitro at 0.5-3 ng/mL significantly inhibited the production of OPN in primary T cells derived from PBMC. The inhibition of OPN was determined to occur at the CD4(+) T-cell level. In addition, IFN-beta inhibited the production of IL-17 and IL-21 in CD4(+) T cells. It has been described that IFN-beta suppresses IL-17 production through the inhibition of a monocytic cytokine, the intracellular translational isoform of OPN. Our further investigation demonstrated that IFN-beta also acted directly on the CD4(+) T cells to regulate OPN and IL-17 expression through the type I IFN receptor-mediated activation of STAT1 and suppression of STAT3 activity. Administration of IFN-beta to EAE mice ameliorated the disease severity. Furthermore, spinal cord infiltration of OPN(+) and IL-17(+) cells decreased in IFN-beta-treated EAE mice along with decreases in serum levels of OPN and IL-21. Importantly, decreased OPN production by IFN-beta treatment contributes to the reduced migratory activity of T cells. Taken together, the results from both in vitro and in vivo experiments indicate that IFN-beta treatment can down-regulate the OPN and IL-17 production in MS. This study provides new insights into the mechanism of action of IFN-beta in the treatment of MS.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Interferon-beta/pharmacology , Interleukin-17/antagonists & inhibitors , Multiple Sclerosis/immunology , Osteopontin/antagonists & inhibitors , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Movement/drug effects , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Glycoproteins/pharmacology , Humans , Interleukin-17/biosynthesis , Interleukins/antagonists & inhibitors , Interleukins/blood , Interleukins/metabolism , Male , Mice , Middle Aged , Myelin-Oligodendrocyte Glycoprotein , Osteopontin/biosynthesis , Osteopontin/blood , Peptide Fragments/pharmacology , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism
14.
Biochem Biophys Res Commun ; 387(4): 806-10, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19646959

ABSTRACT

Tumor hypoxia is associated with tumor promotion, malignant progression, and resistance to cancer therapy. The hypoxia-induced phenotypic changes in tumors result, at least partially, from the induction of hypoxia-responsive genes, such as chemokine receptor CXCR4. Hypoxia-inducible factor 1 (HIF-1) is a critical transcription factor involved in the transcriptional regulation of these genes. Although interferon-gamma (IFNgamma) exerts anti-tumor responses against various tumors, the effect of IFNgamma on HIF-1-dependent transcription remains to be determined. In this study, we examined the inhibitory effect of IFNgamma on hypoxia-induced CXCR4 gene expression in human glioblastoma cell lines and explored the mechanism of inhibition. Hypoxia (1% O(2)) and the iron chelator deferoxamine (DFX), a hypoxia mimetic, increased the levels of CXCR4 mRNA in A172 and T98G cells, and treatment with IFNgamma inhibited the expression of CXCR4 mRNA. Although hypoxia and DFX induced the expression of HIF-1alpha protein and its hypoxia response element (HRE) DNA-binding activity, IFNgamma failed to inhibit its expression or DNA-binding activity. The results of a luciferase reporter assay using a heterologous promoter construct containing the HRE sequence revealed that IFNgamma suppressed the hypoxia- and DFX-induced reporter activities. Lentivirus-mediated RNAi of signal transducer and activator of transcription 1 (STAT1) expression abolished the inhibitory effect of IFNgamma on hypoxia-induced reporter gene activity. Furthermore, this activity was not detected in a stable cell line expressing dominant-negative STAT1. These results indicate that IFNgamma-activated STAT1 functions as a negative regulator of HIF-1-dependent transcription in tumor cells.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interferon-gamma/pharmacology , Neoplasms/metabolism , Receptors, CXCR4/genetics , STAT1 Transcription Factor/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Neoplasms/genetics , STAT1 Transcription Factor/agonists , Transcription, Genetic/drug effects
15.
BMC Immunol ; 10: 46, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19712464

ABSTRACT

BACKGROUND: Umbilical cord blood (UCB) is enriched with transplantable CD34+ cells. In addition to CD34-expressing haematopoietic stem cells (HSC), human UCB contains a rare population of CD34-lineage- cells endowed with the ability to differentiate along the T/NK pathway in response to interleukin (IL)-15 and a stromal cell support. IL-21 is a crucial regulator of NK cell function, whose influence on IL-15-induced differentiation of CD34-lineage- cells has not been investigated previously. The present study was designed and conducted to address whether IL-21 might replace the stromal cell requirements and foster the IL-15-induced NK differentiation of human UCB CD34-lineage- cells. RESULTS: CD34-lineage- cells were maintained in liquid culture with Flt3-L and SCF, with the addition of IL-15 and IL-21, either alone or in combination. Cultures were established in the absence of feeder cells or serum supplementation. Cytokine-treated cells were used to evaluate cell surface phenotype, expression of molecular determinants of lymphoid/NK cell differentiation, secretion of IFN-gamma, GM-CSF, TNF-alpha and CCL3/MIP-1alpha, and cytolytic activity against NK-sensitive tumour cell targets. CD34-lineage- cells proliferated vigorously in response to IL-15 and IL-21 but not to IL-21 alone, and up-regulated phosphorylated Stat1 and Stat3 proteins. CD34-lineage- cells expanded by IL-21 in combination with IL-15 acquired lymphoid morphology and killer-cell immunoglobulin-like receptor (KIR)-CD56+CD16-/+ phenotype, consistent with pseudo-mature NK cells. IL-21/IL-15-differentiated cells expressed high levels of mRNA for Bcl-2, GATA-3 and Id2, a master switch required for NK-cell development, and harboured un-rearranged TCRgamma genes. From a functional standpoint, IL-21/IL-15-treated cells secreted copious amounts of IFN-gamma, GM-CSF and CCL3/MIP-1alpha, and expressed cell surface CD107a upon contact with NK-sensitive tumour targets, a measure of exocytosis of NK secretory granules. CONCLUSION: This study underpins a novel role for IL-21 in the differentiation of pseudo-mature lytic NK cells in a synergistic context with IL-15, and identifies a potential strategy to expand functional NK cells for immunotherapy.


Subject(s)
Cell Lineage/immunology , Fetal Blood/immunology , Interleukin-15/pharmacology , Interleukins/immunology , Killer Cells, Natural/immunology , Lysosomal-Associated Membrane Protein 1/metabolism , Cell Degranulation/drug effects , Cell Degranulation/physiology , Cell Differentiation , Chemokine CCL3/biosynthesis , Chemokine CCL3/immunology , Fetal Blood/cytology , Fetal Blood/drug effects , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Inhibitor of Differentiation Protein 2/immunology , Inhibitor of Differentiation Protein 2/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukins/pharmacology , Killer Cells, Natural/transplantation , Lysosomal-Associated Membrane Protein 1/immunology , Proto-Oncogene Proteins c-bcl-2/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/agonists , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism
16.
Clin Immunol ; 133(1): 61-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19631585

ABSTRACT

Interferon-gamma (IFN-gamma) plays a pathogenic role in systemic lupus erythematosus (SLE). Uncontrolled IFN-gamma signaling may result from a deficiency in the negative regulator, namely, suppressor of cytokine signaling-1 (SOCS-1). We investigated the activation status of IFN-gamma signaling pathway in SLE-afflicted (New-Zealand-BlackxNew-Zealand-White)F1 mice and determined its responsiveness when treating with a tolerogenic peptide, hCDR1, which ameliorates SLE. SOCS-1 was suppressed and pSTAT1 was enhanced in spleen-derived cells from SLE-affected mice as compared with healthy controls. Treatment with hCDR1 reversed the expression of these two molecules in association with clinical amelioration. In vitro stimulation with IFN-gamma resulted in elevated levels of SOCS-1 in cells from both vehicle and hCDR1-treated mice but this effect reached significance only in cells of the latter group, which also exhibited reduced levels of pSTAT1. Thus, SOCS-1 is diminished in SLE-affected mice, and treatment with hCDR1 results in its up-regulation thereby restoring control of IFN-gamma signaling pathway.


Subject(s)
Autoantigens/therapeutic use , Immunosuppression Therapy , Interferon-gamma/immunology , Lupus Erythematosus, Systemic/therapy , Nerve Tissue Proteins/therapeutic use , STAT1 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Disease Models, Animal , Down-Regulation/genetics , Down-Regulation/immunology , Female , Immune Tolerance/drug effects , Immune Tolerance/immunology , Lupus Erythematosus, Systemic/immunology , Mice , Peptides/therapeutic use , STAT1 Transcription Factor/agonists , Signal Transduction/drug effects , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/agonists , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Up-Regulation/genetics , Up-Regulation/immunology
17.
Clin Immunol ; 133(1): 27-44, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19559654

ABSTRACT

Interferon-beta is a current treatment for multiple sclerosis (MS). Interferon-beta is thought to exert its therapeutic effects on MS by down-modulating the immune response by multiple potential pathways. Here, we document that treatment of MS patients with interferon beta-1a (Rebif) results in a significant increase in the levels and function of the protein tyrosine phosphatase SHP-1 in PBMCs. SHP-1 is a crucial negative regulator of cytokine signaling, inflammatory gene expression, and CNS demyelination as evidenced in mice deficient in SHP-1. In order to examine the functional significance of SHP-1 induction in MS PBMCs, we analyzed the activity of proinflammatory signaling molecules STAT1, STAT6, and NF-kappaB, which are known SHP-1 targets. Interferon-beta treatment in vivo resulted in decreased NF-kappaB and STAT6 activation and increased STAT1 activation. Further analysis in vitro showed that cultured PBMCs of MS patients and normal subjects had a significant SHP-1 induction following interferon-beta treatment that correlated with decreased NF-kappaB and STAT6 activation. Most importantly, experimental depletion of SHP-1 in cultured PBMCs abolished the anti-inflammatory effects of interferon-beta treatment, indicating that SHP-1 is a predominant mediator of interferon-beta activity. In conclusion, interferon-beta treatment upregulates SHP-1 expression resulting in decreased transcription factor activation and inflammatory gene expression important in MS pathogenesis.


Subject(s)
Interferon-beta/therapeutic use , Multiple Sclerosis/drug therapy , NF-kappa B/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT1 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism , Adult , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Cytokines/blood , Female , Gene Silencing/immunology , Humans , Interferon beta-1a , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Middle Aged , Multiple Sclerosis/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/immunology , STAT6 Transcription Factor/antagonists & inhibitors , STAT6 Transcription Factor/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
18.
J Nutr Biochem ; 20(9): 726-34, 2009 Sep.
Article in English | MEDLINE | ID: mdl-18926686

ABSTRACT

Bacterial lipopolysaccharide (LPS) is a major inducer of systemic inflammatory reactions and oxidative stress in response to microbial infections and may cause sepsis. In the present study, we demonstrate that retinoic acid inhibits LPS-induced activation in transgenic reporter mice and human monoblasts through inhibition of nuclear factor kappaB (NF-kappaB). By using noninvasive molecular imaging of NF-kappaB luciferase reporter mice, we showed that administration of retinoic acid repressed LPS-induced whole-body luminescence, demonstrating in vivo the dynamics of retinoic acid's ability to repress physiologic response to LPS. Retinoic acid also inhibited LPS-induced NF-kappaB activity in the human myeloblastic cell line U937. Retinoic-acid-receptor-selective agonists mimicked - while specific antagonists inhibited - the effects of retinoic acid, suggesting the involvement of nuclear retinoic acid receptors. Retinoic acid also repressed LPS-induced transcription of NF-kappaB target genes such as IL-6, MCP-1 and COX-2. The effect of retinoic acid was dependent on new protein synthesis, was obstructed by a deacetylase inhibitor and was partly eliminated by a signal transducer and activator of transcription-1 (STAT1)/methyltransferase inhibitor, indicating that retinoic acid induces a new protein, possibly STAT1, that is involved in inhibiting NF-kappaB. This provides more evidence for retinoic acid's anti-inflammatory potential, which may have clinical implications in terms of fighting microbial infections.


Subject(s)
Lipopolysaccharides/immunology , Monocyte-Macrophage Precursor Cells/metabolism , NF-kappa B/metabolism , Tretinoin/pharmacology , Analysis of Variance , Animals , Cells, Cultured , Chemokine CCL2/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Genes, Reporter , Humans , Injections, Subcutaneous , Interleukin-6/genetics , Interleukin-6/metabolism , Luciferases , Luminescent Measurements , Mice , Mice, Transgenic , Models, Animal , Monocyte-Macrophage Precursor Cells/immunology , Protein Biosynthesis , RNA, Messenger/metabolism , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/metabolism , Recombinant Fusion Proteins , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/metabolism , Statistics, Nonparametric , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tretinoin/administration & dosage , Tretinoin/metabolism , U937 Cells , Whole Body Imaging
19.
Infect Immun ; 74(12): 6877-84, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17030574

ABSTRACT

Gamma interferon (IFN-gamma)-induced indoleamine dioxygenase (IDO), which inhibits chlamydial replication by reducing the availability of tryptophan, is up-regulated by interleukin-1beta (IL-1beta) and tumor necrosis factor alpha (TNF-alpha). The mechanisms by which this occurs include an increase in the synthesis of interferon regulatory factor-1 as well as a nuclear factor-kappaB (NF-kappaB)-dependent increase in the expression of IFN-gamma receptors (IFN-gammaR). Although Chlamydia is susceptible to IDO, it up-regulates IFN-gammaR expression to a greater degree than either IL-1beta or TNF-alpha, perhaps through interaction with Toll-like receptors (TLR). The purpose of this study was to determine the mechanism by which Chlamydia psittaci up-regulates IFN-gammaR expression and evaluate this effect on IDO induction. Infection of HEK 293 cells with C. psittaci increased IFN-gammaR expression only in cells expressing either TLR2 or TLR4 and the adaptor protein MD-2. In addition, up-regulation of IFN-gammaR expression in Chlamydia-infected HeLa cells could be blocked either by neutralizing TLRs with anti-TLR2 and/or anti-TLR4 or by inhibiting NF-kappaB transactivation with a proteasome inhibitor. Although the newly expressed IFN-gammaR in Chlamydia-infected cells were capable of binding IFN-gamma, they did not enhance IFN-gamma-induced IDO activity in a manner similar to those observed for IL-1beta and TNF-alpha. Instead, IDO activation in Chlamydia-infected cells was no different than that induced in uninfected cells, despite the increase in IFN-gammaR expression. Furthermore, the amount of IFN-gamma-induced signal transducer and activator of transcription 1 (STAT-1) activation in infected cells paralleled that observed in uninfected cells, suggesting that STAT-1 activation by these newly expressed receptors was impaired.


Subject(s)
Chlamydia/physiology , Receptors, Interferon/metabolism , STAT1 Transcription Factor/agonists , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology , Coculture Techniques , HeLa Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Lymphocyte Antigen 96/metabolism , NF-kappa B/metabolism , Phosphorylation , Receptors, Interferon/genetics , Signal Transduction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Up-Regulation , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...