Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 204
Filter
1.
Curr Top Microbiol Immunol ; 316: 119-54, 2007.
Article in English | MEDLINE | ID: mdl-17969446

ABSTRACT

Type II interferon-gamma (IFN-gamma) is a pleiotropic cytokine that regulates many different cellular functions. The major signaling pathway activated by IFN-gamma involves sequential phosphorylation of the tyrosine residues of the Janus kinase (JAK) and signal transducer and activator of transcription (STAT) proteins, providing the primary mechanism through which gene expression is induced. However, recent work has revealed that the responses are complex, as shown by the activation of kinases in addition to JAKs, differential patterns of activation of STAT1, STAT3, and STAT5 in different cells, and activation of transcription factors other than STATs. This complexity is used to regulate biological functions differentially in a cell type-specific manner, by activating different specific signals and patterns of gene expression.


Subject(s)
Interferon-gamma/metabolism , Animals , Humans , Janus Kinases/metabolism , Receptors, Interferon/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Interferon gamma Receptor
2.
Eur J Immunol ; 31(12): 3747-54, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11745395

ABSTRACT

Interleukin-18 (IL-18), a pleiotropic cytokine produced by activated macrophages, plays significant roles in the immune response, inducing the secretion of IFN-gamma, TNF-alpha and IL-2, enhancing NK cell activity and potentiating the differentiation of Th1 cells. The intercellular signal transduction pathways through which IL-18 functions have not been thoroughly defined. We have generated a mutant cell line, I1A, that lacks the IRAK protein. In this line which has low or no expression of the other known IRAK family members, we find that the IL-1 receptor-associated kinase (IRAK) is essential for the activation of NFkappaB and JNK in response to IL-18. Furthermore, the death domain, but not the kinase activity of IRAK, is necessary for NFkappaB activation in response to IL-18. Interestingly, the N-proximal undetermined region of IRAK is necessary for NFkappaB activation, but not for JNK activation in response to IL-18, indicating IRAK may be a branchpoint in IL-18 signaling. In addition to IRAK, we implicate two other components in IL-18 signaling, TAK1 (TGF-beta-activated kinase 1) and its activator and substrate TAB1. A dominant negative mutant of TAK1 inhibits the IL-18-mediated NFkappaB activation, while IL-18 stimulation leads to the phosphorylation of TAB1. Finally, analysis of IL-18 signaling in IL-1-unresponsive mutant cell lines suggests that the IL-1- and IL-18-mediated pathways are similar, but may not be identical.


Subject(s)
Adaptor Proteins, Signal Transducing , Interleukin-18/pharmacology , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase Kinases/physiology , Protein Kinases/physiology , Signal Transduction , Carrier Proteins/physiology , Cells, Cultured , Humans , Interleukin-1/pharmacology , Interleukin-1 Receptor-Associated Kinases , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation
3.
J Biol Chem ; 276(48): 44365-8, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11591697

ABSTRACT

Previous experiments have suggested that induction of the beta-R1 gene by interferon (IFN)-beta required transcription factor ISGF-3 (IFN-stimulated gene factor-3) and an additional component. We now provide evidence that nuclear factor-kappaB (NF-kappaB) can serve as this component. Site-directed mutagenesis of an NF-kappaB binding site in the beta-R1 promoter or over-expression of an IkappaBalpha super-repressor abrogated IFN-beta-mediated induction of a beta-R1 promoter-reporter. IFN-beta treatment did not augment abundance of NF-kappaB but did lead to phosphorylation of the p65 NF-kappaB subunit. It is proposed that IFN-beta-mediated enhancement of the transactivation competence of NF-kappaB components is required for inducible transcription of the beta-R1 promoter. These results provide a novel insight into the role of NF-kappaB in the transcriptional response to IFN-beta.


Subject(s)
Interferon-beta/metabolism , NF-kappa B/physiology , Binding Sites , DNA-Binding Proteins/metabolism , Enzyme Activation , Genes, Reporter , Humans , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, gamma Subunit , Mutagenesis, Site-Directed , NF-kappa B/metabolism , Phosphorylation , Precipitin Tests , Promoter Regions, Genetic , Time Factors , Transcription Factor RelA , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured
4.
J Biol Chem ; 276(36): 33361-8, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11438544

ABSTRACT

STAT1 must be phosphorylated on serine 727 to be fully active in transcription. We show that phosphatidylinositol 3-kinase (PI3K) and its effector kinase Akt play an important role in the serine phosphorylation of STAT1 and in the activation of gene expression in response to interferon-gamma (IFN gamma). IFN gamma activates PI3K as well as Akt in a variety of cell lines. Specific inhibition of PI3K abrogates IFN gamma-induced, but not interleukin-1- or tumor necrosis factor-alpha-induced, phosphorylation of STAT1 on serine and reduces STAT1-dependent transcription and gene expression by approximately 7-fold. Constitutively active forms of PI3K or Akt activate and their dominant-negative derivatives inhibit STAT1-driven transactivation in response to IFN gamma. In addition to PI3K and Akt, JAK1, JAK2, and the tyrosine 440 STAT1 docking residue of IFNGR1 are required for STAT1 to be phosphorylated on serine. Taken together, these results suggest that the following events lead to the activation of STAT1 upon IFN gamma stimulation: 1) PI3K and Akt are activated by the occupied receptor and Tyr-440 is phosphorylated by the activated JAKs; 2) STAT1 docks to Tyr-440; and 3) Tyr-701 is phosphorylated by the JAKs and Ser-727 is phosphorylated by a kinase downstream of Akt.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Interferon-gamma/metabolism , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases , Serine/chemistry , Trans-Activators/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Line , Chromones/pharmacology , Enzyme Activation , Genes, Dominant , Genes, Reporter , Janus Kinase 1 , Janus Kinase 2 , Mice , Morpholines/pharmacology , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , STAT1 Transcription Factor , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Necrosis Factor-alpha/metabolism , Tyrosine/chemistry
5.
EMBO Rep ; 2(8): 736-42, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11463746

ABSTRACT

Epithelial cells are refractory to extracellular lipopolysaccharide (LPS), yet when presented inside the cell, it is capable of initiating an inflammatory response. Using invasive Shigella flexneri to deliver LPS into the cytosol, we examined how this factor, once intracellular, activates both NF-kappaB and c-Jun N-terminal kinase (JNK). Surprisingly, the mode of activation is distinct from that induced by toll-like receptors (TLRs), which mediate LPS responsiveness from the outside-in. Instead, our findings demonstrate that this response is mediated by a cytosolic, plant disease resistance-like protein called CARD4/Nod1. Biochemical studies reveal enhanced oligomerization of CARD4 upon S. flexneri infection, an event necessary for NF-kappaB induction. Dominant-negative versions of CARD4 block activation of NF-kappaB and JNK by S. flexneri as well as microinjected LPS. Finally, we showed that invasive S. flexneri triggers the formation of a transient complex involving CARD4, RICK and the IKK complex. This study demonstrates that in addition to the extracellular LPS sensing system mediated by TLRs, mammalian cells also possess a cytoplasmic means of LPS detection via a molecule that is related to plant disease-resistance proteins.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Drosophila Proteins , Gene Expression Regulation/physiology , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Shigella flexneri/physiology , Signal Transduction/physiology , Carrier Proteins/genetics , Cell Line , Genes, Reporter , HeLa Cells , Humans , I-kappa B Kinase , Interleukin-1/pharmacology , JNK Mitogen-Activated Protein Kinases , Lipopolysaccharides/administration & dosage , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microinjections , Nod1 Signaling Adaptor Protein , Precipitin Tests , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteins/genetics , Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Shigella flexneri/pathogenicity , TNF Receptor-Associated Factor 2 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/pharmacology
6.
Oncogene ; 20(20): 2527-36, 2001 May 03.
Article in English | MEDLINE | ID: mdl-11420662

ABSTRACT

Activation of MAP kinase leads to the activation of p53-dependent pathways, and vice-versa. Although the amount of p53 protein increases in response to MAP kinase-dependent signaling, the basis of this increase is not yet fully understood. We have isolated the mutant cell line AP14, defective in p53 expression, from human HT1080 fibrosarcoma cells, which have an activated ras allele. The expression of p53 mRNA and protein is approximately 10-fold lower in AP14 cells than in the parental cells. The high constitutive phosphorylation and activities of the MAP kinases ERK1 and ERK2 in HT1080 cells are greatly reduced in AP14 cells, although the levels of these proteins are unchanged, suggesting that the defect in the mutant cells affects the steady-state phosphorylation of ERK1 and ERK2. Overexpression of ERK2 in AP14 cells restored both MAP kinase activity and p53 expression, and incubation of the mutant cells with the phosphatase inhibitor orthovanadate resulted in strong coordinate elevation of MAP kinase activity and p53 expression. The levels of expression of the p53-regulated gene p21 parallel those of p53 throughout, showing that basal p21 expression depends on p53. The levels of p53 mRNA increased by 5-8-fold when activated ras was introduced into wild-type cells, and the levels of the p53 and p21 proteins decreased substantially in wild-type cells treated with the MEK inhibitor U0216. We conclude that MAP kinase-dependent pathways help to regulate p53 levels by regulating the expression of p53 mRNA.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , MAP Kinase Kinase Kinase 1 , MAP Kinase Signaling System/physiology , Tumor Suppressor Protein p53/biosynthesis , ras Proteins/physiology , 3T3 Cells , Animals , Fibrosarcoma/enzymology , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins c-raf/physiology , Proto-Oncogene Proteins p21(ras)/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
7.
J Biol Chem ; 276(34): 31819-24, 2001 Aug 24.
Article in English | MEDLINE | ID: mdl-11431470

ABSTRACT

Previously, we found that the protein kinase C (PKC) inhibitor H7 stimulates p53 to accumulate in a form incapable of inducing transcription from p53-dependent promoters. We concluded that H7 inhibits constitutive C-terminal phosphorylation of p53, which regulates its turnover in unstressed cells. We now show that p53 and its inhibitor MDM2 (HDM2 in human cells) are together in the nuclei of H7-treated cells and can be co-immunoprecipitated. Despite this association of p53 with the ubiquitin ligase MDM2, ubiquitinated p53 was not detected in H7-treated cells. Furthermore, co-treatment with H7 and the proteosome inhibitor LLnL prevented the accumulation of ubiquitinated p53 that was observed in cells treated solely with LLnL. In addition, treatment of cells with the PKC activator phorbol ester stimulated the ubiquitination of p53 and reduced its ability to accumulate after stress. H7 did not induce the phosphorylation of human p53 on Ser-15 (Ser-18 in mouse protein), a modification that occurs in response to DNA damage and leads to the release of MDM2 and to transactivation by p53. We conclude that phosphorylation of the C-terminal domain of p53 by PKC increases its ubiquitination and degradation in unstressed cells.


Subject(s)
Nuclear Proteins , Tumor Suppressor Protein p53/metabolism , Ubiquitins/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Hydrolysis , Mice , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Serine/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Suppressor Protein p53/chemistry
8.
Proc Natl Acad Sci U S A ; 98(12): 6674-9, 2001 Jun 05.
Article in English | MEDLINE | ID: mdl-11390994

ABSTRACT

Although Stat1 is essential for cells to respond fully to IFN-gamma, there is substantial evidence that, in the absence of Stat1, IFN-gamma can still regulate the expression of some genes, induce an antiviral state and affect cell growth. We have now identified many genes that are regulated by IFN-gamma in serum-starved Stat1-null mouse fibroblasts. The proteins induced by IFN-gamma in Stat1-null cells can account for the substantial biological responses that remain. Some genes are induced in both wild-type and Stat1-null cells and thus are truly Stat1-independent. Others are subject to more complex regulation in response to IFN-gamma, repressed by Stat1 in wild-type cells and activated in Stat1-null cells. Many genes induced by IFN-gamma in Stat1-null fibroblasts also are induced by platelet-derived growth factor in wild-type cells and thus are likely to be involved in cell proliferation. In mouse cells expressing the docking site mutant Y440F of human IFN-gamma receptor subunit 1, the mouse Stat1 is not phosphorylated in response to human IFN-gamma, but c-myc and c-jun are still induced, showing that the Stat1 docking site is not required for Stat1-independent signaling.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation/drug effects , Immediate-Early Proteins , Interferon-gamma/pharmacology , Trans-Activators/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Cells, Cultured , Chemokines/genetics , DNA-Binding Proteins/biosynthesis , Early Growth Response Protein 1 , Genes, Immediate-Early , Genes, jun , Genes, myc , Humans , Mice , Platelet-Derived Growth Factor/physiology , Receptors, Interferon/physiology , STAT1 Transcription Factor , Transcription Factors/biosynthesis , Interferon gamma Receptor
9.
Proc Natl Acad Sci U S A ; 98(12): 6680-5, 2001 Jun 05.
Article in English | MEDLINE | ID: mdl-11390995

ABSTRACT

Although Stat1 is required for many IFN-dependent responses, recent work has shown that IFNgamma functions independently of Stat1 to affect the growth of tumor cells or immortalized fibroblasts. We now demonstrate that both IFNgamma and IFNalpha/beta regulate proliferative responses in cells of the mononuclear phagocyte lineage derived from Stat1-null mice. Using both representational difference analysis and gene arrays, we show that IFNgamma exerts its Stat1-independent actions on mononuclear phagocytes by regulating the expression of many genes. This result was confirmed by monitoring changes in expression and function of the corresponding gene products. Regulation of the expression of these genes requires the IFNgamma receptor and Jak1. The physiologic relevance of IFN-dependent, Stat1-independent signaling was demonstrated by monitoring antiviral responses in Stat1-null mice. Thus, the IFN receptors engage alternative Stat1-independent signaling pathways that have important physiological consequences.


Subject(s)
DNA-Binding Proteins/physiology , Interferons/pharmacology , Trans-Activators/physiology , Animals , Cell Division/drug effects , Gene Expression Regulation/drug effects , Humans , Janus Kinase 1 , Macrophages/metabolism , Mice , Protein-Tyrosine Kinases/physiology , Receptors, Interferon/physiology , STAT1 Transcription Factor , Interferon gamma Receptor
10.
Oncogene ; 20(15): 1803-15, 2001 Apr 05.
Article in English | MEDLINE | ID: mdl-11313928

ABSTRACT

p53 protects mammals from neoplasia by inducing apoptosis, DNA repair and cell cycle arrest in response to a variety of stresses. p53-dependent arrest of cells in the G1 phase of the cell cycle is an important component of the cellular response to stress. Here we review recent evidence that implicates p53 in controlling entry into mitosis when cells enter G2 with damaged DNA or when they are arrested in S phase due to depletion of the substrates required for DNA synthesis. Part of the mechanism by which p53 blocks cells at the G2 checkpoint involves inhibition of Cdc2, the cyclin-dependent kinase required to enter mitosis. Cdc2 is inhibited simultaneously by three transcriptional targets of p53, Gadd45, p21, and 14-3-3 sigma. Binding of Cdc2 to Cyclin B1 is required for its activity, and repression of the cyclin B1 gene by p53 also contributes to blocking entry into mitosis. p53 also represses the cdc2 gene, to help ensure that cells do not escape the initial block. Genotoxic stress also activates p53-independent pathways that inhibit Cdc2 activity, activation of the protein kinases Chk1 and Chk2 by the protein kinases Atm and Atr. Chk1 and Chk2 inhibit Cdc2 by inactivating Cdc25, the phosphatase that normally activates Cdc2. Chk1, Chk2, Atm and Atr also contribute to the activation of p53 in response to genotoxic stress and therefore play multiple roles. p53 induces transcription of the reprimo, B99, and mcg10 genes, all of which contribute to the arrest of cells in G2, but the mechanisms of cell cycle arrest by these genes is not known. Repression of the topoisomerase II gene by p53 helps to block entry into mitosis and strengthens the G2 arrest. In summary, multiple overlapping p53-dependent and p53-independent pathways regulate the G2/M transition in response to genotoxic stress.


Subject(s)
Biomarkers, Tumor , Exonucleases , G2 Phase , Mitosis , Neoplasm Proteins , Tumor Suppressor Protein p53/physiology , 14-3-3 Proteins , Alkaloids/pharmacology , Animals , CDC2 Protein Kinase/metabolism , Caffeine/pharmacology , Cyclin B/metabolism , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/physiology , DNA Damage , Exoribonucleases , Humans , Phosphorylation , Proteins/physiology , Staurosporine/analogs & derivatives
11.
Oncogene ; 20(9): 1076-84, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11314044

ABSTRACT

Changes in the phosphorylation state of p53 are important in increasing its half-life and potency as a transcription factor. To investigate their roles, serine residues 15 and 37 were mutated to alanines and the mutated proteins were expressed stably at low basal levels in Li-Fraumeni-derived p53-null human fibroblasts. The accumulation of p53 after DNA damage was analysed quantitatively in multiple clones. Mutation of serine 15, serine 37 or both impaired the accumulation of the protein after exposing the cells to ultraviolet radiation (50-100% increase for the mutant proteins, 500% increase for wild-type p53) but not after treatment with adriamycin. The diminished accumulation of mutant p53 protein is due to a reduction of basal HDM association. Analysis of p53-dependent transcription revealed that phosphorylation of serine 15 is required to maintain basal levels of p21 mRNA. These results provide new evidence for an important function of serine 37 phosphorylation, clearly distinguish the pathways of p53 activation in response to ultraviolet radiation or DNA damage inflicted by adriamycin, and reveal that serine 15 is crucial to support the p53-mediated basal expression of p21.


Subject(s)
Nuclear Proteins , Serine/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects , Antineoplastic Agents/pharmacology , Blotting, Northern , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA/metabolism , DNA Primers/chemistry , Doxorubicin/pharmacology , Fibroblasts/metabolism , Genetic Vectors , Humans , Intracellular Signaling Peptides and Proteins , Mutation , Phosphorylation , Precipitin Tests , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism , Ultraviolet Rays , GADD45 Proteins
12.
Proc Natl Acad Sci U S A ; 98(8): 4461-5, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11287640

ABSTRACT

Mutant I1A cells, lacking IL-1 receptor-associated kinase (IRAK) mRNA and protein, have been used to study the involvement of IRAK in NFkappaB and c-Jun N-terminal kinase (JNK) activation. A series of IRAK deletion constructs were expressed in I1A cells, which were then tested for their ability to respond to IL-1. Both the N-terminal death domain and the C-terminal region of IRAK are required for IL-1-induced NFkappaB and JNK activation, whereas the N-proximal undetermined domain is required for the activation of NFkappaB but not JNK. The phosphorylation and ubiquitination of IRAK deletion mutants correlate tightly with their ability to activate NFkappaB in response to IL-1, but IRAK can mediate IL-1-induced JNK activation without being phosphorylated. These studies reveal that the IL-1-induced signaling pathways leading to NFkappaB and JNK activation diverge either at IRAK or at a point nearer to the receptor.


Subject(s)
Interleukin-1/physiology , Mitogen-Activated Protein Kinases/metabolism , Protein Kinases/metabolism , Base Sequence , DNA Probes , Enzyme Activation , Humans , Interleukin-1 Receptor-Associated Kinases , JNK Mitogen-Activated Protein Kinases , NF-kappa B/metabolism , Phosphorylation , Protein Binding , Proteins/metabolism , Recombinant Proteins/metabolism , Sequence Deletion , Signal Transduction , TNF Receptor-Associated Factor 6
13.
Mol Cell Biol ; 21(7): 2475-84, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259596

ABSTRACT

Interleukin-1 (IL-1) is a proinflammatory cytokine that recognizes a surface receptor complex and generates multiple cellular responses. IL-1 stimulation activates the mitogen-activated protein kinase kinase kinase TAK1, which in turn mediates activation of c-Jun N-terminal kinase and NF-kappaB. TAB2 has previously been shown to interact with both TAK1 and TRAF6 and promote their association, thereby triggering subsequent IL-1 signaling events. The serine/threonine kinase IL-1 receptor-associated kinase (IRAK) also plays a role in IL-1 signaling, being recruited to the IL-1 receptor complex early in the signal cascade. In this report, we investigate the role of IRAK in the activation of TAK1. Genetic analysis reveals that IRAK is required for IL-1-induced activation of TAK1. We show that IL-1 stimulation induces the rapid but transient association of IRAK, TRAF6, TAB2, and TAK1. TAB2 is recruited to this complex following translocation from the membrane to the cytosol upon IL-1 stimulation. In IRAK-deficient cells, TAB2 translocation and its association with TRAF6 are abolished. These results suggest that IRAK regulates the redistribution of TAB2 upon IL-1 stimulation and facilitates the formation of a TRAF6-TAB2-TAK1 complex. Formation of this complex is an essential step in the activation of TAK1 in the IL-1 signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/physiology , Interleukin-1/physiology , MAP Kinase Kinase Kinases/physiology , Receptors, Interleukin-1/physiology , Signal Transduction , Cell Line , Enzyme Activation , Humans , MAP Kinase Signaling System
14.
J Biol Chem ; 276(3): 1998-2006, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11032828

ABSTRACT

p53 represses the transcription of cdc2 and cyclin B1, causing loss of Cdc2 activity and G(2) arrest. Here we show that the region -22 to -2 of the cdc2 promoter called the R box is required for repression by p53 but not for basal promoter activity. The R box confers p53-dependent repression on heterologous promoters and binds to p130/E2F4 in response to overexpression of p53. R box-dependent repression requires p21/waf1, and overexpression of p21/waf1 also represses the cdc2 promoter. These observations suggest that p53 represses the cdc2 promoter by inducing p21/waf1, which inhibits cyclin-dependent kinase activity, enhancing the binding of p130 and E2F4, which together bind to and repress the cdc2 promoter.


Subject(s)
CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/metabolism , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Base Sequence , Cell Line , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/genetics , DNA , E2F4 Transcription Factor , Molecular Sequence Data , Protein Binding , Protein Serine-Threonine Kinases/genetics , Repressor Proteins/metabolism , Transcription, Genetic
15.
Proc Natl Acad Sci U S A ; 97(19): 10489-93, 2000 Sep 12.
Article in English | MEDLINE | ID: mdl-10962024

ABSTRACT

Use of an NF-kappaB-dependent selectable marker facilitated the isolation of a cell line containing a cDNA encoding Act1, an NF-kappaB activator. Act1 associates with and activates IkappaB kinase (IKK), leading to the liberation of NF-kappaB from its complex with IkappaB. Many signaling pathways that liberate NF-kappaB also activate activating transcription factor (ATF) and activator protein 1 (AP-1) through Jun kinase (JNK). Act1 also activates JNK, suggesting that it might be part of a multifunctional complex involved in the activation of both NF-kappaB and JNK. Act1 fails to activate NF-kappaB in an IL-1-unresponsive mutant cell line in which all known signaling components are present, suggesting that it interacts with an unknown component in IL-1 signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins , NF-kappa B/metabolism , Trans-Activators/genetics , Amino Acid Sequence , Cell Line , DNA, Complementary , Enzyme Activation , Helix-Loop-Helix Motifs , Humans , Interleukin-1/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Recombinant Proteins/metabolism , Sequence Homology, Nucleic Acid , Trans-Activators/chemistry , Trans-Activators/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins
16.
EMBO J ; 19(15): 4111-22, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10921891

ABSTRACT

Analysis of mRNA levels in cells that express or lack signal transducers and activators of transcription 1 (Stat1) reveals that Stat1 mediates the constitutive transcription of many genes. Expression of the low molecular mass polypeptide 2 (LMP2), which requires Stat1, has been studied in detail. The overlapping interferon consensus sequence 2/gamma-interferon-activated sequence (ICS-2/GAS) elements in the LMP2 promoter bind to interferon regulatory factor 1 (IRF1) and Stat1 and are occupied constitutively in vivo. The point mutant of Stat1, Y701F, which does not form dimers involving SH2-phosphotyrosine interactions, binds to the GAS element and supports LMP2 expression. Unphosphorylated Stat1 binds to IRF1 directly and we conclude that this complex uses the ICS-2/GAS element to mediate constitutive LMP2 transcription in vivo. The promoter of the IRF1 gene, which also contains a GAS site but not an adjacent ICS-2 site, is not activated by Stat1 Y701F. The promoters of other genes whose constitutive expression requires Stat1 may also utilize complexes of unphosphorylated Stat1 with IRF1 or other transcription factors.


Subject(s)
Cysteine Endopeptidases , DNA-Binding Proteins/metabolism , Phosphoproteins/metabolism , Proteins/genetics , Trans-Activators/metabolism , Base Sequence , Binding Sites , Dimerization , Gene Expression Profiling , Gene Expression Regulation , Humans , Interferon Regulatory Factor-1 , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein Binding , STAT1 Transcription Factor , Transcription, Genetic
17.
Exp Eye Res ; 70(4): 485-91, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10865997

ABSTRACT

Defects in apoptosis have been noted in signal transducer and activator of transcription (Stat) 1-null cells in vitro. The purpose of this study was to analyse the keratocyte apoptosis response that occurs in vivo in response to corneal epithelial injury in Stat 1null compared with control mice and to determine whether Stat 1null corneal fibroblasts have a defective response to death receptor activation in vitro. Corneal epithelial scrape injuries were performed in Stat 1-null and wild-type mice. Keratocyte apoptosis was monitored with the quantitative TUNEL assay and confirmed using transmission electron microscopy. Corneal fibroblast apoptosis in response to tumor necrosis factor (TNF) alpha, with and without inhibitors of nuclear factor kappa B (NF-kappaB) activation, was monitored using DNA laddering and the methylene blue assay. Significantly less keratocyte apoptosis was noted in Stat 1-null mice compared with wild-type controls. TNF alpha-induced apoptosis only occurred in wild-type mice in the presence of inhibitors of NF-kappaB activation. Corneal fibroblast TNF alpha-induced apoptosis was defective in Stat 1null corneal fibroblasts whether NF-kappaB activation was blocked or not. Stat 1 has an important role in the keratocyte apoptosis that occurs in response to corneal epithelial injury. Previous studies suggest that the defect is due to a lack of constitutive expression of caspases. This study demonstrates that this defect in apoptosis in Stat 1-null mice is present in vivo in Stat 1-null mice and suggests that Stat 1 could be a therapeutic target for transient inhibition of keratocyte apoptosis to modulate corneal wound healing.


Subject(s)
Apoptosis/physiology , Epithelium, Corneal/injuries , Keratinocytes/pathology , Animals , Fibroblasts/physiology , In Situ Nick-End Labeling , Lymphocytes, Null , Methylene Blue , Mice , Mice, Inbred C57BL , Microscopy, Electron , NF-kappa B/antagonists & inhibitors , Signal Transduction/genetics , Transcription, Genetic/genetics , Tumor Necrosis Factor-alpha/physiology
18.
Oncogene ; 19(21): 2619-27, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10851061

ABSTRACT

Stat1 is a fascinating and complex protein with multiple, yet contrasting transcriptional functions. Upon activation, it drives the expression of many genes but also suppresses the transcription of others. These opposing characteristics also apply to its role in facilitating crosstalk between signal transduction pathways, as it participates in both synergistic activation and inhibition of gene expression. Stat1 is a functional transcription factor even in the absence of inducer-mediated activation, participating in the constitutive expression of some genes. This review summarizes the well studied involvement of Stat1 in IFN-dependent and growth factor-dependent signaling and then describes the roles of Stat1 in positive, negative and constitutive regulation of gene expression as well as its participation in crosstalk between signal transduction pathways. Oncogene (2000).


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Trans-Activators/metabolism , Animals , Cell Division , Genes, Tumor Suppressor/genetics , Growth Substances/metabolism , Humans , Immunologic Surveillance , Interferons/metabolism , Receptor Cross-Talk , Repressor Proteins/metabolism , STAT1 Transcription Factor , Signal Transduction
19.
J Biol Chem ; 275(27): 20406-11, 2000 Jul 07.
Article in English | MEDLINE | ID: mdl-10764778

ABSTRACT

The LMP2 gene, which encodes a protein required for efficient presentation of viral antigens, requires both unphosphorylated Stat1 and IRF1 for basal expression. LMP2 expression is down-regulated by the adenovirus protein E1A, which binds to Stat1 and CBP/p300, and by the mutant E1A protein RG2, which binds to Stat1 but not to CBP/p300, but not by the mutant protein Delta2-36, which does not bind to either Stat1 or CBP/p300. Stat1 and IRF1 associate in untreated cells and bind as a complex to the overlapping ICS-2/GAS element of the LMP2 promoter. E1A interferes with the formation of this complex by occupying domains of Stat1 that bind to IRF1. These results reveal how adenovirus infection attenuates LMP2 expression, thereby interfering with the presentation of viral antigens.


Subject(s)
Adenovirus E1A Proteins/pharmacology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral/genetics , Phosphoproteins/metabolism , Trans-Activators/metabolism , Viral Matrix Proteins/genetics , Adenoviridae/pathogenicity , Cell Line , Down-Regulation , Humans , Interferon Regulatory Factor-1 , Mutation , Promoter Regions, Genetic , Protein Binding , STAT1 Transcription Factor , Viral Matrix Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
20.
Mol Biol Cell ; 11(3): 873-86, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10712506

ABSTRACT

Gene amplification in eukaryotes plays an important role in drug resistance, tumorigenesis, and evolution. The Schizosaccharomyces pombe sod2 gene provides a useful model system to analyze this process. sod2 is near the telomere of chromosome I and encodes a plasma membrane Na(+)(Li(+))/H(+) antiporter. When sod2 is amplified, S. pombe survives otherwise lethal concentrations of LiCl, and >90% of the amplified sod2 genes are found in 180- and 225-kilobase (kb) linear amplicons. The sequence of the novel joint of the 180-kb amplicon indicates that it is formed by recombination between homologous regions near the telomeres of the long arm of chromosome I and the short arm of chromosome II. The 225-kb amplicon, isolated three times more frequently than the 180-kb amplicon, is a palindrome derived from a region near the telomere of chromosome I. The center of symmetry of this palindrome contains an inverted repeat consisting of two identical 134-base pair sequences separated by a 290-base pair spacer. LiCl-resistant mutants arise 200-600 times more frequently in strains deficient for topoisomerases or DNA ligase activity than in wild-type strains, but the mutant cells contain the same amplicons. These data suggest that amplicon formation may begin with DNA lesions such as breaks. In the case of the 225-kb amplicon, the breaks may lead to a hairpin structure, which is then replicated to form a double-stranded linear amplicon, or to a cruciform structure, which is then resolved to yield the same amplicon.


Subject(s)
Gene Amplification , Schizosaccharomyces/genetics , Sodium-Hydrogen Exchangers/genetics , Base Sequence , Cloning, Molecular , DNA Topoisomerases, Type I/genetics , DNA, Fungal/analysis , Ligases/genetics , Molecular Sequence Data , Nucleic Acid Conformation , Repetitive Sequences, Nucleic Acid/genetics , Sequence Homology, Nucleic Acid , Telomere/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...