Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell ; 185(21): 3857-3876, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36240739

ABSTRACT

The discovery of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway arose from investigations of how cells respond to interferons (IFNs), revealing a paradigm in cell signaling conserved from slime molds to mammals. These discoveries revealed mechanisms underlying rapid gene expression mediated by a wide variety of extracellular polypeptides including cytokines, interleukins, and related factors. This knowledge has provided numerous insights into human disease, from immune deficiencies to cancer, and was rapidly translated to new drugs for autoimmune, allergic, and infectious diseases, including COVID-19. Despite these advances, major challenges and opportunities remain.


Subject(s)
COVID-19 , Janus Kinases , Animals , Cytokines/metabolism , Humans , Interferons/metabolism , Janus Kinases/metabolism , Mammals/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction
2.
EMBO J ; 39(11): e101573, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32323871

ABSTRACT

High expression of 2',5'-oligoadenylate synthetase 1 (OAS1), which adds AMP residues in 2',5' linkage to a variety of substrates, is observed in many cancers as a part of the interferon-related DNA damage resistance signature (IRDS). Poly(ADP-ribose) (PAR) is rapidly synthesized from NAD+ at sites of DNA damage to facilitate repair, but excessive PAR synthesis due to extensive DNA damage results in cell death by energy depletion and/or activation of PAR-dependent programmed cell death pathways. We find that OAS1 adds AMP residues in 2',5' linkage to PAR, inhibiting its synthesis in vitro and reducing its accumulation in cells. Increased OAS1 expression substantially improves cell viability following DNA-damaging treatments that stimulate PAR synthesis during DNA repair. We conclude that high expression of OAS1 in cancer cells promotes their ability to survive DNA damage by attenuating PAR synthesis and thus preventing cell death.


Subject(s)
2',5'-Oligoadenylate Synthetase/biosynthesis , DNA Damage , Gene Expression Regulation, Enzymologic , Poly ADP Ribosylation , 2',5'-Oligoadenylate Synthetase/genetics , Adenosine Monophosphate/genetics , Adenosine Monophosphate/metabolism , Cell Death , Cell Line, Transformed , Humans
3.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34799452

ABSTRACT

Programmed death ligand 1 (PD-L1), an immune-checkpoint protein expressed on cancer cells, also functions independently of the immune system. We found that PD-L1 inhibits the killing of cancer cells in response to DNA damage in an immune-independent manner by suppressing their acute response to type I interferon (IFN; IFN-I). In addition, PD-L1 plays a critical role in sustaining high levels of constitutive expression in cancer cells of a subset of IFN-induced genes, the IFN-related DNA damage resistance signature (IRDS) which, paradoxically, protects cancer cells. The cyclic GMP-AMP synthase-stimulator of the IFN genes (cGAS-STING) pathway is constitutively activated in a subset of cancer cells in the presence of high levels of PD-L1, thus leading to a constitutive, low level of IFN-ß expression, which in turn increases IRDS expression. The constitutive low level of IFN-ß expression is critical for the survival of cancer cells addicted to self-produced IFN-ß. Our study reveals immune-independent functions of PD-L1 that inhibit cytotoxic acute responses to IFN-I and promote protective IRDS expression by supporting protective chronic IFN-I responses, both of which enhance the resistance of cancer cells to DNA damage.


Subject(s)
B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , DNA Damage/physiology , Interferon Type I/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Interferon Type I/genetics , Interferon-beta , Interferon-gamma/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Nucleotidyltransferases , Signal Transduction , Tumor Microenvironment
4.
Proc Natl Acad Sci U S A ; 114(52): 13792-13797, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29229854

ABSTRACT

Triple-negative breast cancer (TNBC), the deadliest form of this disease, lacks a targeted therapy. TNBC tumors that fail to respond to chemotherapy are characterized by a repressed IFN/signal transducer and activator of transcription (IFN/STAT) gene signature and are often enriched for cancer stem cells (CSCs). We have found that human mammary epithelial cells that undergo an epithelial-to-mesenchymal transition (EMT) following transformation acquire CSC properties. These mesenchymal/CSCs have a significantly repressed IFN/STAT gene expression signature and an enhanced ability to migrate and form tumor spheres. Treatment with IFN-beta (IFN-ß) led to a less aggressive epithelial/non-CSC-like state, with repressed expression of mesenchymal proteins (VIMENTIN, SLUG), reduced migration and tumor sphere formation, and reexpression of CD24 (a surface marker for non-CSCs), concomitant with an epithelium-like morphology. The CSC-like properties were correlated with high levels of unphosphorylated IFN-stimulated gene factor 3 (U-ISGF3), which was previously linked to resistance to DNA damage. Inhibiting the expression of IRF9 (the DNA-binding component of U-ISGF3) reduced the migration of mesenchymal/CSCs. Here we report a positive translational role for IFN-ß, as gene expression profiling of patient-derived TNBC tumors demonstrates that an IFN-ß metagene signature correlates with improved patient survival, an immune response linked with tumor-infiltrating lymphocytes (TILs), and a repressed CSC metagene signature. Taken together, our findings indicate that repressed IFN signaling in TNBCs with CSC-like properties is due to high levels of U-ISGF3 and that treatment with IFN-ß reduces CSC properties, suggesting a therapeutic strategy to treat drug-resistant, highly aggressive TNBC tumors.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Interferon-beta/pharmacology , Neoplasm Proteins/biosynthesis , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
6.
EMBO Rep ; 17(3): 367-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26882544

ABSTRACT

Signal transducer and activator of transcription 1 (STAT1) plays a pivotal role in the innate immune system by directing the transcriptional response to interferons (IFNs). STAT1 is activated by Janus kinase (JAK)-mediated phosphorylation of Y701. To determine whether STAT1 contributes to cellular responses without this phosphorylation event, we generated mice with Y701 mutated to a phenylalanine (Stat1(Y701F)). We show that heterozygous mice do not exhibit a dominant-negative phenotype. Homozygous Stat1(Y701F) mice show a profound reduction in Stat1 expression, highlighting an important role for basal IFN-dependent signaling. The rapid transcriptional response to type I IFN (IFN-I) and type II IFN (IFNγ) was absent in Stat1(Y701F) cells. Intriguingly, STAT1Y701F suppresses the delayed expression of IFN-I-stimulated genes (ISG) observed in Stat1(-/-) cells, mediated by the STAT2/IRF9 complex. Thus, Stat1(Y701F) macrophages are more susceptible to Legionella pneumophila infection than Stat1(-/-) macrophages. Listeria monocytogenes grew less robustly in Stat1(Y701F) macrophages and mice compared to Stat1(-/-) counterparts, but STAT1Y701F is not sufficient to rescue the animals. Our studies are consistent with a potential contribution of Y701-unphosphorylated STAT1 to innate antibacterial immunity.


Subject(s)
Immunity, Innate , Interferons/metabolism , Legionnaires' Disease/immunology , Listeriosis/immunology , Protein Processing, Post-Translational , STAT1 Transcription Factor/deficiency , Animals , Cell Line , Cells, Cultured , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mutation, Missense , Phosphorylation , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Second Messenger Systems
7.
Proc Natl Acad Sci U S A ; 112(33): 10443-8, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26216956

ABSTRACT

Up-regulation of IFN-stimulated genes (ISGs) is sustained in hepatitis C virus (HCV)-infected livers. Here, we investigated the mechanism of prolonged ISG expression and its role in IFN responsiveness during HCV infection in relation to unphosphorylated IFN-stimulated gene factor 3 (U-ISGF3), recently identified as a tripartite transcription factor formed by high levels of IFN response factor 9 (IRF9), STAT1, and STAT2 without tyrosine phosphorylation of the STATs. The level of U-ISGF3, but not tyrosine phosphorylated STAT1, is significantly elevated in response to IFN-λ and IFN-ß during chronic HCV infection. U-ISGF3 prolongs the expression of a subset of ISGs and restricts HCV chronic replication. However, paradoxically, high levels of U-ISGF3 also confer unresponsiveness to IFN-α therapy. As a mechanism of U-ISGF3-induced resistance to IFN-α, we found that ISG15, a U-ISGF3-induced protein, sustains the abundance of ubiquitin-specific protease 18 (USP18), a negative regulator of IFN signaling. Our data demonstrate that U-ISGF3 induced by IFN-λs and -ß drives prolonged expression of a set of ISGs, leading to chronic activation of innate responses and conferring a lack of response to IFN-α in HCV-infected liver.


Subject(s)
Endopeptidases/metabolism , Hepatitis C/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferons/immunology , Cell Line , HEK293 Cells , Hep G2 Cells , Hepacivirus , Hepatitis C/immunology , Hepatocytes/metabolism , Humans , Immunity, Innate , Lentivirus/metabolism , Liver/metabolism , Liver/virology , Phosphorylation , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Signal Transduction/drug effects , Tyrosine/chemistry , Ubiquitin Thiolesterase
8.
EMBO J ; 32(20): 2751-63, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24065129

ABSTRACT

A single high dose of interferon-ß (IFNß) activates powerful cellular responses, in which many anti-viral, pro-apoptotic, and anti-proliferative proteins are highly expressed. Since some of these proteins are deleterious, cells downregulate this initial response rapidly. However, the expression of many anti-viral proteins that do no harm is sustained, prolonging a substantial part of the initial anti-viral response for days and also providing resistance to DNA damage. While the transcription factor ISGF3 (IRF9 and tyrosine-phosphorylated STATs 1 and 2) drives the first rapid response phase, the related factor un-phosphorylated ISGF3 (U-ISGF3), formed by IFNß-induced high levels of IRF9 and STATs 1 and 2 without tyrosine phosphorylation, drives the second prolonged response. The U-ISGF3-induced anti-viral genes that show prolonged expression are driven by distinct IFN stimulated response elements (ISREs). Continuous exposure of cells to a low level of IFNß, often seen in cancers, leads to steady-state increased expression of only the U-ISGF3-dependent proteins, with no sustained increase in other IFNß-induced proteins, and to constitutive resistance to DNA damage.


Subject(s)
DNA Damage , Disease Resistance , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-beta/pharmacology , STAT1 Transcription Factor/genetics , STAT2 Transcription Factor/genetics , Virus Diseases/prevention & control , Animals , Antiviral Agents/pharmacology , Cells, Cultured , Chlorocebus aethiops , DNA Damage/drug effects , DNA Damage/genetics , Disease Resistance/drug effects , Disease Resistance/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Tumor Escape/drug effects , Tumor Escape/genetics , Vero Cells , Virus Diseases/genetics , Virus Diseases/immunology , Viruses/drug effects , Viruses/immunology , Viruses/pathogenicity
9.
Proc Natl Acad Sci U S A ; 110(42): 16975-80, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24082147

ABSTRACT

The activation of STAT3 by tyrosine phosphorylation, essential for normal development and for a normal inflammatory response to invading pathogens, is kept in check by negative regulators. Abnormal constitutive activation of STAT3, which contributes to the pathology of cancer and to chronic inflammatory diseases such as rheumatoid arthritis, occurs when negative regulation is not fully effective. SOCS3, the major negative regulator of STAT3, is induced by tyrosine-phosphorylated STAT3 and terminates STAT3 phosphorylation about 2 h after initial exposure of cells to members of the IL-6 family of cytokines by binding cooperatively to the common receptor subunit gp130 and JAKs 1 and 2. We show here that when the epidermal growth factor receptor (EGFR) is present and active, STAT3 is rephosphorylated about 4 h after exposure of cells to IL-6 or oncostatin M and remains active for many hours. Newly synthesized IL-6 drives association of the IL-6 receptor and gp130 with EGFR, leading to EGFR-dependent rephosphorylation of STAT3, which is not inhibited by the continued presence of SOCS3. This second wave of STAT3 activation supports sustained expression of a subset of IL-6-induced proteins, several of which play important roles in inflammation and cancer, in which both IL-6 secretion and EGFR levels are often elevated.


Subject(s)
Cytokine Receptor gp130/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation/physiology , Interleukin-6/biosynthesis , STAT3 Transcription Factor/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cytokine Receptor gp130/genetics , ErbB Receptors/genetics , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/genetics , Interleukin-6/pharmacology , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oncostatin M/metabolism , Oncostatin M/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Binding/physiology , STAT3 Transcription Factor/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
10.
Int J Cancer ; 136(4): E51-61, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25156627

ABSTRACT

The mechanism of multicellular drug resistance, defined as the reduced efficacy of chemotherapeutic drugs in solid tumors is incompletely understood. Here we report that colon carcinoma cells cultured as 3D microtissues (spheroids) display dramatic increases in the expression of a subset of type I interferon-(IFN)-stimulated genes (ISGs). A similar gene signature was associated previously with resistance to radiation and chemotherapy, prompting us to examine the underlying biological mechanisms. Analysis of spheroids formed by different tumor cell lines and studies using knock-down of gene expression showed that cell crowding leads to the induction of IFN regulatory factor-9 (IRF9) which together with STAT2 and independently of IFNs, is necessary for ISG upregulation. Increased expression of IRF9 alone was sufficient to induce the ISG subset in monolayer cells and to confer increased resistance to clinically used cytotoxic drugs. Our data reveal a novel mechanism of regulation of a subset of ISGs, leading to drug resistance in solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Apoptosis , Cell Communication , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferons/physiology , STAT2 Transcription Factor/metabolism , Transcriptional Activation
11.
Trends Cancer ; 9(1): 83-92, 2023 01.
Article in English | MEDLINE | ID: mdl-36216730

ABSTRACT

Acute exposure of cancer cells to high concentrations of type I interferon (IFN-I) drives growth arrest and apoptosis, whereas chronic exposure to low concentrations provides important prosurvival advantages. Tyrosine-phosphorylated IFN-stimulated gene (ISG) factor 3 (ISGF3) drives acute deleterious responses to IFN-I, whereas unphosphorylated (U-)ISGF3, lacking tyrosine phosphorylation, drives essential constitutive prosurvival mechanisms. Surprisingly, programmed cell death-ligand 1 (PD-L1), often expressed on the surfaces of tumor cells and well recognized for its importance in inactivating cytotoxic T cells, also has important cell-intrinsic protumor activities, including dampening acute responses to cytotoxic high levels of IFN-I and sustaining the expression of the low levels that benefit tumors. More thorough understanding of the newly recognized complex roles of IFN-I in cancer may lead to the identification of novel therapeutic strategies.


Subject(s)
Interferons , Neoplasms , Humans , Interferons/metabolism , Interferon-Stimulated Gene Factor 3/genetics , Interferon-Stimulated Gene Factor 3/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Signal Transduction , Tyrosine , Neoplasms/drug therapy , Neoplasms/genetics
12.
Proc Natl Acad Sci U S A ; 106(23): 9373-8, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19478064

ABSTRACT

In normal human cells treated with interferons (IFNs), the concentration of tyrosine-phosphorylated STAT1 (YP-STAT1), which drives the expression of a large number of genes, increases quickly but then decreases over a period of several hours. Because the STAT1 gene is activated by YP-STAT1, IFNs stimulate a large increase in the concentration of unphosphorylated STAT1 (U-STAT1) that persists for several days. To test the significance of high U-STAT1 expression, we increased its concentration exogenously in the absence of IFN treatment. In response, the expression of many immune regulatory genes (e.g., IFI27, IFI44, OAS, and BST2) was increased. In human fibroblasts or mammary epithelial cells treated with low concentrations of IFN-beta or IFN-gamma, the expression of the same genes increased after 6 h and continued to increase after 48 or 72 h, long after the concentration of YP-STAT1 had returned to basal levels. Consistent with its activity as a transcription factor, most U-STAT1 was present in the nuclei of these cells before IFN treatment, and the fraction in nuclei increased 48 h after treatment with IFN. We conclude that the nuclear U-STAT1 that accumulates in response to IFNs maintains or increases the expression of a subset of IFN-induced genes independently of YP-STAT1, and that many of the induced proteins are involved in immune regulation.


Subject(s)
Interferons/metabolism , STAT1 Transcription Factor/metabolism , Thionucleotides , Cell Line , Humans , Phosphorylation , STAT3 Transcription Factor/metabolism
13.
Article in English | MEDLINE | ID: mdl-28620095

ABSTRACT

Many cytokines and all interferons activate members of a small family of kinases (the Janus kinases [JAKs]) and a slightly larger family of transcription factors (the signal transducers and activators of transcription [STATs]), which are essential components of pathways that induce the expression of specific sets of genes in susceptible cells. JAK-STAT pathways are required for many innate and acquired immune responses, and the activities of these pathways must be finely regulated to avoid major immune dysfunctions. Regulation is achieved through mechanisms that include the activation or induction of potent negative regulatory proteins, posttranslational modification of the STATs, and other modulatory effects that are cell-type specific. Mutations of JAKs and STATs can result in gains or losses of function and can predispose affected individuals to autoimmune disease, susceptibility to a variety of infections, or cancer. Here we review recent developments in the biochemistry, genetics, and biology of JAKs and STATs.


Subject(s)
Cytokines/pharmacology , Interferons/pharmacology , Janus Kinases/physiology , STAT Transcription Factors/physiology , Animals , Chromatin Assembly and Disassembly , Gene Expression Regulation , Glycosylation , Humans , Janus Kinases/genetics , Mutation , Phosphorylation , STAT Transcription Factors/genetics
14.
Exp Mol Med ; 39(4): 524-34, 2007 Aug 31.
Article in English | MEDLINE | ID: mdl-17934341

ABSTRACT

2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a prototypic halogenated aromatic hydrocarbon (HAH), is known as one of the most potent toxicants. At least a part of its toxic effects appears to be derived from its ability to induce TNF-alpha production. However, the signaling pathway of TCDD that leads to TNF-alpha expression has not been elucidated. In this study, we investigated the signaling mechanism of TCDD-induced TNF-alpha expression in PMA-differentiated THP-1 macrophages. TCDD induced both mRNA and protein expression of TNF-alpha in a dose- and time-dependent manner. Alpha-naphthoflavone (NF), an aryl hydrocarbon receptor (AhR) inhibitor, prevented the TCDD-induced expression of TNF-alpha at both mRNA and protein levels. Genistein, a protein tyrosine kinase (PTK) inhibitor, and PD153035, an EGFR inhibitor, also blocked the increase of TNF-alpha expression by TCDD, indicating the role of EGFR in TCDD-induced TNF-alpha expression. On the other hand, PP2, a c-Src specific inhibitor, did not affect TCDD-induced TNF-alpha expression. EGFR phosphorylation was detected as early as 5 min after TCDD treatment. TCDD-induced EGFR activation was AhR-dependent since co-treatment with alpha-NF prevented it. ERK was found to be a downstream effector of EGFR activation in the signaling pathway leading to TNF-alpha production after TCDD stimulation. Activation of ERK was observed from 30 min after TCDD treatment. PD98059, an inhibitor of the MEK-ERK pathway, completely prevented the TNF-alpha mRNA and protein expression induced by TCDD, whereas inhibitors of JNK and p38 MAPK had no effect. PD153035, an EGFR inhibitor, as well as alpha-NF significantly reduced ERK phosphorylation, suggesting that ERK activation by TCDD was mediated by both EGFR and AhR. These results indicate that TNF-alpha production by TCDD in differentiated THP-1 macrophages is AhR-dependent and involves activation of EGFR and ERK, but not c-Src, JNK, nor p38 MAPK. A signaling pathway is proposed where TCDD induces sequential activation of AhR, EGFR and ERK, leading to the increased expression of TNF-alpha.


Subject(s)
Hazardous Substances/toxicity , Macrophages/metabolism , Polychlorinated Dibenzodioxins/toxicity , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Benzoflavones/pharmacology , Cell Differentiation , Cell Line, Tumor , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Genistein/pharmacology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Phosphorylation , Pyrimidines/pharmacology , Quinazolines/pharmacology , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Signal Transduction , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
15.
Exp Mol Med ; 37(3): 179-85, 2005 Jun 30.
Article in English | MEDLINE | ID: mdl-16000871

ABSTRACT

15-deoxy-delta(12,14)-PGJ(2)(15d-PGJ(2)) is a natural ligand that activates the peroxisome proliferators-activated receptor (PPAR) gamma, a member of nuclear receptor family implicated in regulation of lipid metabolism and adipocyte differentiation. Recent studies have shown that 15d-PGJ(2) is the potent anti-inflammatory agent functioning via PPARgamma-dependent and -independent mechanisms. Most postulated mechanisms for anti-inflammatory action of PPARgamma agonists are involved in inhibiting NF-kappaB signaling pathway. We examined the possibility that IL-6 signaling via the Jak-Stat pathway is modulated by 15d-PGJ(2) in lymphocytes and also examined whether the inhibition of IL-6 signaling is dependent of PPARgamma. 15d-PGJ(2) blocked IL-6 induced Stat1 and Stat3 activation in primary human lymphocytes, Jurkat cells and immortalized rheumatoid arthritis B cells. Inhibition of IL-6 signaling was induced rapidly within 15 min after treatment of 15d-PGJ(2). Other PPARgamma-agonists, such as troglitazone and ciglitazone, did not inhibit IL-6 signaling, indicating that 15d-PGJ(2) affect the IL-6-induced Jak-Stat signaling pathway via PPARgamma-independent mechanism. Although cycloheximide reversed 15d-PGJ(2)-mediated inhibition of Stat3 activation, actinomycin D had no effect on 15d-PGJ(2)-mediated inhibition of IL-6 signaling, indicating that inhibition of IL-6 signaling occur independent of de novo gene expression. These results show that 15d-PGJ(2) specifically inhibit Jak-Stat signaling pathway in lymphocytes, and suggest that 15d-PGJ(2) may regulate inflammatory reactions through the modulation of different signaling pathway other than NF-kappaB in lymphocytes.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-6/pharmacology , Lymphocytes/drug effects , Lymphocytes/metabolism , Prostaglandin D2/analogs & derivatives , Signal Transduction , Trans-Activators/metabolism , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Chromans/pharmacology , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Gene Expression Regulation , Humans , Hypoglycemic Agents/pharmacology , Jurkat Cells/metabolism , Jurkat Cells/pathology , Lymphocytes/cytology , NF-kappa B/metabolism , PPAR gamma/metabolism , Phosphorylation , Prostaglandin D2/pharmacology , Protein Synthesis Inhibitors/pharmacology , STAT1 Transcription Factor , STAT3 Transcription Factor , Thiazolidinediones/pharmacology , Troglitazone
16.
Cell Rep ; 12(2): 300-12, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26146080

ABSTRACT

The transcriptional response to infection with the bacterium Listeria monocytogenes (Lm) requires cooperative signals of the type I interferon (IFN-I)-stimulated JAK-STAT and proinflammatory NF-κB pathways. Using ChIP-seq analysis, we define genes induced in Lm-infected macrophages through synergistic transcriptional activation by NF-κB and the IFN-I-activated transcription factor ISGF3. Using the Nos2 and IL6 genes as prime examples of this group, we show that NF-κB functions to recruit enzymes that establish histone marks of transcriptionally active genes. In addition, NF-κB regulates transcriptional elongation by employing the mediator kinase module for the recruitment of the pTEFb complex. ISGF3 has a major role in associating the core mediator with the transcription start as a prerequisite for TFIID and RNA polymerase II (Pol II) binding. Our data suggest that the functional cooperation between two major antimicrobial pathways is based on promoter priming by NF-κB and the engagement of the core mediator for Pol II binding by ISGF3.


Subject(s)
Mediator Complex/metabolism , NF-kappa B/metabolism , STAT Transcription Factors/metabolism , Animals , Binding Sites , Histone Acetyltransferases/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Interferon-Stimulated Gene Factor 3/genetics , Interferon-Stimulated Gene Factor 3/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Listeria monocytogenes/physiology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Protein Binding , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Polymerase II/metabolism , Signal Transduction , Transcription Factor TFIID/metabolism , Transcriptional Activation
17.
Gene ; 293(1-2): 77-86, 2002 Jun 26.
Article in English | MEDLINE | ID: mdl-12137945

ABSTRACT

A novel alpha-tubulin gene (alpha6) was cloned from a genomic library of Naegleria gruberi strain NB-1 and characterized. The open reading frame of alpha6 contained 1359 nucleotides encoding a protein of 452 amino acids (aa) with a calculated molecular weight of 50.5 kDa. The nucleotide sequence of the open reading frame of alpha6 showed considerable divergence (68.4% identity) when compared with previously cloned N. gruberi alpha-tubulin genes, which share about 97% identity in DNA sequences. The deduced aa sequence of alpha6-tubulin was 61.9% identical to that of alpha13-tubulin, which was cloned from the same strain, and showed similar identities to those of alpha-tubulins from other species (54 approximately 62%). These data showed that alpha6-tubulin is one of the most divergent alpha-tubulins so far known. Alpha6-tubulin was found to be expressed in actively growing cells and repressed quickly when these cells were induced to differentiate. Immunostaining with an antibody against alpha6-tubulin showed that alpha6-tubulin is present in the nuclei and mitotic spindle-fibers but absent in flagellar axonemes or cytoskeletal microtubules. These data finally established the presence of an alpha-tubulin that is specifically utilized for spindle-fiber microtubules and distinct from the flagellar axonemal alpha-tubulins in N. gruberi, hence confirmed the multi-tubulin hypothesis in this organism.


Subject(s)
Naegleria/genetics , Tubulin/genetics , Amino Acid Sequence , Animals , Antibodies/immunology , Antibody Specificity , Base Sequence , Cell Differentiation/genetics , Cell Division/genetics , Cloning, Molecular , DNA, Protozoan/chemistry , DNA, Protozoan/genetics , Gene Expression , Genetic Variation , Mitosis/genetics , Molecular Sequence Data , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/immunology , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tubulin/immunology
18.
Semin Oncol ; 41(2): 156-73, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24787290

ABSTRACT

Constitutive expression of interferons (IFNs) and activation of their signaling pathways have pivotal roles in host responses to malignant cells in the tumor microenvironment. IFNs are induced by the innate immune system and in tumors through stimulation of Toll-like receptors (TLRs) and through other signaling pathways in response to specific cytokines. Although in the oncologic context IFNs have been thought of more as exogenous pharmaceuticals, the autocrine and paracrine actions of endogenous IFNs probably have even more critical effects on neoplastic disease outcomes. Through high-affinity cell surface receptors, IFNs modulate transcriptional signaling, leading to regulation of more than 2,000 genes with varying patterns of temporal expression. Induction of the gene products by both unphosphorylated and phosphorylated STAT1 after ligand binding results in alterations in tumor cell survival, inhibition of angiogenesis, and augmentation of actions of T, natural killer (NK), and dendritic cells. The interferon-stimulated gene (ISG) signature can be a favorable biomarker of immune response but, in a seemingly paradoxical finding, a specific subset of the full ISG signature indicates an unfavorable response to DNA-damaging interventions such as radiation. IFNs in the tumor microenvironment thus can alter the emergence, progression, and regression of malignancies.


Subject(s)
Interferons/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Tumor Microenvironment , Animals , Antineoplastic Agents/chemistry , Apoptosis , Cytokines/metabolism , DNA/metabolism , DNA Damage , Dendritic Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, p53 , Humans , Immunity, Innate , Mice , Mice, Transgenic , Neovascularization, Pathologic , Phosphorylation , Retroviridae/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction
19.
J Interferon Cytokine Res ; 31(1): 33-40, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21166594

ABSTRACT

The signal transducers and activators of transcription (STAT)1 and STAT3 genes are specifically activated by phosphorylated STATs 1 and 3, respectively, resulting in large and prolonged increases in the levels of unphosphorylated STATs (U-STATs) in response to interferons (for STAT1) or ligands that activate gp130, such as IL-6 (for STAT3). U-STATs 1 and 3 are transcription factors that drive gene expression by mechanisms distinct from those used by phosphorylated STATs. U-STAT3 drives expression of many proteins not induced by phospho-STAT3, including several that are important in tumorigenesis. U-STAT1 prolongs and increases expression of a subset of proteins induced initially in response to phospho-STAT1, leading to antiviral and immune responses that are long-lived. U-STAT1 levels are also high in some cancers, and the protein products of genes induced by U-STAT1 enhance resistance to DNA damage. Therefore, interferons not only drive short-term expression of proteins that inhibit growth and promote apoptosis and immune surveillance, but also promote long-term expression of proteins that facilitate tumor survival.


Subject(s)
Cytokines/metabolism , Gene Expression Regulation , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Gene Expression Regulation/drug effects , Humans , Immunity, Innate/drug effects , Interferons/pharmacology , Interferons/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , Virus Diseases/drug therapy , Virus Diseases/immunology , Virus Diseases/metabolism
20.
J Immunol ; 177(2): 1092-100, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16818766

ABSTRACT

In inflamed joints of rheumatoid arthritis, PGE(2) is highly expressed, and IL-10 and IL-6 are also abundant. PGE(2) is a well-known activator of the cAMP signaling pathway, and there is functional cross-talk between cAMP signaling and the Jak-STAT signaling pathway. In this study, we evaluated the modulating effect of PGE(2) on STAT signaling and its biological function induced by IL-10 and IL-6, and elucidated its mechanism in THP-1 cells. STAT phosphorylation was determined by Western blot, and gene expression was analyzed using real-time PCR. Pretreatment with PGE(2) significantly augmented IL-10-induced STAT3 and STAT1 phosphorylation, as well as suppressors of cytokine signaling 3 (SOCS3) and IL-1R antagonist gene expression. In contrast, PGE(2) suppressed IL-6-induced phosphorylation of STAT3 and STAT1. These PGE(2)-induced modulating effects were largely reversed by actinomycin D. Pretreatment with dibutyryl cAMP augmented IL-10-induced, but did not change IL-6-induced STAT3 phosphorylation. Misoprostol, an EP2/3/4 agonist, and butaprost, an EP2 agonist, augmented IL-10-induced STAT3 phosphorylation and SOCS3 gene expression, but sulprostone, an EP1/3 agonist, had no effect. H89, a protein kinase A inhibitor, and LY294002, a PI3K inhibitor, diminished PGE(2)-mediated augmentation of IL-10-induced STAT3 phosphorylation. In this study, we found that PGE(2) selectively regulates cytokine signaling via increased intracellular cAMP levels and de novo gene expression, and these modulating effects may be mediated through EP2 or EP4 receptors. PGE(2) may modulate immune responses by alteration of cytokine signaling in THP-1 cells.


Subject(s)
Adjuvants, Immunologic/physiology , Dinoprostone/physiology , Interleukin-10/physiology , Signal Transduction/immunology , Adjuvants, Immunologic/metabolism , Cell Line, Tumor , Cells, Cultured , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Cytokines/metabolism , Cytokines/physiology , Dinoprostone/metabolism , Gene Expression Regulation/immunology , Humans , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Receptors, Cytokine/biosynthesis , Receptors, Prostaglandin E/biosynthesis , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/physiology , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP3 Subtype , Receptors, Prostaglandin E, EP4 Subtype , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL