Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Proc Natl Acad Sci U S A ; 98(15): 8554-9, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11438690

ABSTRACT

The intracellular levels of many proteins are regulated by ubiquitin-dependent proteolysis. One of the best-characterized enzymes that catalyzes the attachment of ubiquitin to proteins is a ubiquitin ligase complex, Skp1-Cullin-F box complex containing Hrt1 (SCF). We sought to artificially target a protein to the SCF complex for ubiquitination and degradation. To this end, we tested methionine aminopeptidase-2 (MetAP-2), which covalently binds the angiogenesis inhibitor ovalicin. A chimeric compound, protein-targeting chimeric molecule 1 (Protac-1), was synthesized to recruit MetAP-2 to SCF. One domain of Protac-1 contains the I kappa B alpha phosphopeptide that is recognized by the F-box protein beta-TRCP, whereas the other domain is composed of ovalicin. We show that MetAP-2 can be tethered to SCF(beta-TRCP), ubiquitinated, and degraded in a Protac-1-dependent manner. In the future, this approach may be useful for conditional inactivation of proteins, and for targeting disease-causing proteins for destruction.


Subject(s)
Aminopeptidases/metabolism , DNA-Binding Proteins/metabolism , I-kappa B Proteins , Metalloendopeptidases/metabolism , Peptide Synthases/metabolism , Ubiquitins/metabolism , Animals , Cell Extracts , Cell Line, Transformed , Humans , NF-KappaB Inhibitor alpha , Ovum/metabolism , Protein Binding , Recombinant Fusion Proteins/metabolism , SKP Cullin F-Box Protein Ligases , Xenopus laevis
2.
Proc Natl Acad Sci U S A ; 98(12): 6668-73, 2001 Jun 05.
Article in English | MEDLINE | ID: mdl-11381115

ABSTRACT

The transcription factor NF-kappaB regulates expression of genes that are involved in inflammation, immune response, viral infection, cell survival, and division. However, the role of NF-kappaB in hypertrophic growth of terminally differentiated cardiomyocytes is unknown. Here we report that NF-kappaB activation is required for hypertrophic growth of cardiomyocytes. In cultured rat primary neonatal ventricular cardiomyocytes, the nuclear translocation of NF-kappaB and its transcriptional activity were stimulated by several hypertrophic agonists, including phenylephrine, endothelin-1, and angiotensin II. The activation of NF-kappaB was inhibited by expression of a "supersuppressor" IkappaBalpha mutant that is resistant to stimulation-induced degradation and a dominant negative IkappaB kinase (IKKbeta) mutant that can no longer be activated by phosphorylation. Furthermore, treatment with phenylephrine induced IkappaBalpha degradation in an IKK-dependent manner, suggesting that NF-kappaB is a downstream target of the hypertrophic agonists. Importantly, expression of the supersuppressor IkappaBalpha mutant or the dominant negative IKKbeta mutant blocked the hypertrophic agonist-induced expression of the embryonic gene atrial natriuretic factor and enlargement of cardiomyocytes. Conversely, overexpression of NF-kappaB itself induced atrial natriuretic factor expression and cardiomyocyte enlargement. These findings suggest that NF-kappaB plays a critical role in the hypertrophic growth of cardiomyocytes and may serve as a potential target for the intervention of heart disease.


Subject(s)
Cardiomegaly/etiology , I-kappa B Proteins , NF-kappa B/physiology , Angiotensin II/pharmacology , Animals , Animals, Newborn , Atrial Natriuretic Factor/genetics , Cells, Cultured , DNA-Binding Proteins/metabolism , NF-KappaB Inhibitor alpha , Phenylephrine/pharmacology , Rats , Rats, Sprague-Dawley
3.
Cancer Res ; 61(9): 3810-8, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11325857

ABSTRACT

Nuclear factor-kappaB (NF-kappaB)/Rel transcription factors regulate genes that control cell proliferation, survival, and transformation. In normal breast epithelial cells, NF-kappaB/Rel proteins are mainly sequestered in the cytoplasm bound to one of the specific inhibitory IkappaB proteins, whereas in breast cancers they are activated aberrantly. Human breast tumor cell lines, carcinogen-transformed mammary epithelial cells, and the majority of primary human or rodent breast tumor tissue samples express constitutively high levels of nuclear NF-kappaB/REL: To begin to understand the mechanism of this aberrant NF-kappaB/Rel expression, in this study we measured the activity of the major kinases implicated in regulation of IkappaB stability, namely IKKalpha, IKKbeta, and protein kinase, CK2 (formerly casein kinase II). Hs578T, D3-1, and BP-1 breast cancer cell lines displayed higher levels of IKKalpha, IKKbeta, and CK2 activity than untransformed MCF-10F mammary epithelial cells. Inhibition of IKK activity upon expression of dominant negative kinases or of CK2 activity by treatment with selective inhibitors decreased NF-kappaB/Rel activity in breast cancer cells. Inactivation of the IkappaB kinase complex in Hs578T cells via expression of a dominant negative IKKgamma/NF-kappaB essential modulator/IKK-associated protein 1 reduced soft agar colony growth. Thus, the aberrant expression of CK2 or IKK kinases promotes increased nuclear levels of NF-kappaB/Rel and transformation of breast cancer cells. Furthermore, primary human breast cancer specimens that displayed aberrant constitutive expression of NF-kappaB/Rel were found to exhibit increased CK2 and/or IKK kinase activity. These observations suggest these kinases play a similar role in an intracellular signaling pathway that leads to the elevated NF-kappaB/Rel levels seen in primary human mammary tumors and, therefore, represent potential therapeutic targets in the treatment of patients with breast cancer.


Subject(s)
Breast Neoplasms/enzymology , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Breast Neoplasms/pathology , Casein Kinase II , Cell Adhesion/physiology , Cell Division/physiology , Cell Survival/physiology , Cell Transformation, Neoplastic/metabolism , Enzyme Activation , Humans , I-kappa B Kinase , NF-kappa B/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Tumor Cells, Cultured
4.
Biochimie ; 83(3-4): 341-9, 2001.
Article in English | MEDLINE | ID: mdl-11295495

ABSTRACT

In most cases, target proteins of the ubiquitin system are completely degraded. In several exceptions, such as the first step in the activation of the transcriptional regulator NF-kappaB, the substrate, the precursor protein p105, is processed in a limited manner to yield the active subunit p50. p50 is derived from the N-terminal domain of p105, whereas the C-terminal domain is degraded. The mechanisms involved in this unique process have remained elusive. We have shown that a Gly-rich region (GRR) at the C-terminal domain of p50 is one important processing signal and that it interferes with processing of the ubiquitinated precursor by the 26S proteasome. Also, amino acid residues 441-454 are important for processing under non-stimulated conditions. Lys 441 and 442 serve as ubiquitination targets, whereas residues 446-454 may serve as a ligase recognition motif. Following IkappaB kinase (IKK)-mediated phosphorylation, the C-terminal domain of p105, residues 918-934, recruits the SCF(beta-TrCP) ubiquitin ligase, and ubiquitination by this complex leads to accelerated processing. The two sites appear to be recognized under different physiological conditions by two different ligases, targeting two distinct recognition motifs. We have shown that ubiquitin conjugation and processing of a series of precursors of p105 that lack the C-terminal IKK phosphorylation/TrCP binding domain, is progressively inhibited with increasing number of ankyrin repeats. Inhibition is due to docking of active NF-kappaB subunits to the ankyrin repeat domain in the C-terminal half of p105 (IkappaBgamma). Inhibition is alleviated by phosphorylation of the C-terminal domain that leads to ubiquitin-mediated degradation of the ankyrin repeat domain and release of the anchored subunits. We propose a model that may explain the requirement for two sites: a) a basal site that may be involved in co-translational processing prior to the synthesis of the ankyrin repeat domain; and b) a signal-induced site that is involved in processing/degradation of the complete molecule following cell activation, with rapid release of stored, transcriptionally active subunits.


Subject(s)
I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Peptide Hydrolases/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Ubiquitins/metabolism , Amino Acid Motifs , Ankyrins , DNA-Binding Proteins/metabolism , Glycine , Humans , I-kappa B Kinase , Multienzyme Complexes/metabolism , Peptide Synthases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , SKP Cullin F-Box Protein Ligases , Signal Transduction/physiology
5.
Cell Immunol ; 214(1): 54-9, 2001 Nov 25.
Article in English | MEDLINE | ID: mdl-11902829

ABSTRACT

IkappaB kinase (IKK) plays a key role in the regulation of nuclear factor kappaB (NF-kappaB). We previously demonstrated the expression of two kinases, IKK1 and IKK2, in fibroblast-like synoviocytes (FLS) and determined their functional consequences for inflammatory gene expression in vitro and in vivo. Recently, a novel inducible IkappaB kinase has been described, namely, IKK-i or IKK-epsilon, which is functionally and structurally distinct from constitutively expressed IKK1 and IKK2. Therefore, we investigated the expression and regulation of this novel kinase in FLS from patients with rheumatoid arthritis and osteoarthritis. Interestingly, constitutive gene expression and protein expression were observed in all cell lines examined. TNFalpha stimulation for 24 h increased IKK-i expression 7.2 +/- 1.8-fold in FLS (P < 0.02). IL-1 also significantly increased IKK-i gene expression. Time course experiments demonstrated that IKK-i gene expression increased within 3 h of TNFalpha stimulation and persisted for at least 24 h. Dose-response studies showed that as little as 1 ng/ml of TNFalpha increased IKK-i gene expression. Constitutive IKK-1 gene expression was also noted in rheumatoid arthritis, osteoarthritis, and normal synovium. This is the first report demonstrating constitutive expression and cytokine regulation of this novel kinase in primary human synovial cells.


Subject(s)
Fibroblasts/enzymology , Protein Serine-Threonine Kinases/biosynthesis , Synovial Membrane/enzymology , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/genetics , Cells, Cultured , Fibroblasts/drug effects , Humans , I-kappa B Kinase , Interleukin-1/pharmacology , Kinetics , Osteoarthritis/enzymology , Osteoarthritis/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/biosynthesis , Synovial Membrane/cytology , Transcription, Genetic , Transcriptional Activation , Tumor Necrosis Factor-alpha/pharmacology
7.
Mol Cell Biol ; 20(15): 5381-91, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10891479

ABSTRACT

NF-kappaB/Rel factors have been implicated in the regulation of liver cell death during development, after partial hepatectomy, and in hepatocytes in culture. Rat liver epithelial cells (RLEs) display many biochemical and ultrastructural characteristics of oval cells, which are multipotent cells that can differentiate into mature hepatocytes. While untransformed RLEs undergo growth arrest and apoptosis in response to transforming growth factor beta1 (TGF-beta1) treatment, oncogenic Ras- or Raf-transformed RLEs are insensitive to TGF-beta1-mediated growth arrest. Here we have tested the hypothesis that Ras- or Raf-transformed RLEs have altered NF-kappaB regulation, leading to this resistance to TGF-beta1. We show that classical NF-kappaB is aberrantly activated in Ras- or Raf-transformed RLEs, due to increased phosphorylation and degradation of IkappaB-alpha protein. Inhibition of NF-kappaB activity with a dominant negative form of IkappaB-alpha restored TGF-beta1-mediated cell killing of transformed RLEs. IKK activity mediates this hyperphosphorylation of IkappaB-alpha protein. As judged by kinase assays and transfection of dominant negative IKK-1 and IKK-2 expression vectors, NF-kappaB activation by Ras appeared to be mediated by both IKK-1 and IKK-2, while Raf-induced NF-kappaB activation was mediated by IKK-2. NF-kappaB activation in the Ras-transformed cells was mediated by both the Raf and phosphatidylinositol 3-kinase pathways, while in the Raf-transformed cells, NF-kappaB induction was mediated by the mitogen-activated protein kinase cascade. Last, inhibition of either IKK-1 or IKK-2 reduced focus-forming activity in Ras-transformed RLEs. Overall, these studies elucidate a mechanism that contributes to the process of transformation of liver cells by oncogene Ras and Raf through the IkappaB kinase complex leading to constitutive activation of NF-kappaB.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, ras , I-kappa B Proteins/metabolism , Liver/pathology , Retroviridae Proteins, Oncogenic/genetics , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Transformed , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology , I-kappa B Proteins/antagonists & inhibitors , I-kappa B Proteins/genetics , Liver/drug effects , NF-kappa B/metabolism , Oncogene Proteins v-raf , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Rats , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
8.
EMBO J ; 19(11): 2580-91, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10835356

ABSTRACT

Processing of the p105 precursor to form the active subunit p50 of the NF-kappaB transcription factor is a unique case in which the ubiquitin system is involved in limited processing rather than in complete destruction of the target substrate. A glycine-rich region along with a downstream acidic domain have been demonstrated to be essential for processing. Here we demonstrate that following IkappaB kinase (IkappaK)-mediated phosphorylation, the C-terminal domain of p105 (residues 918-934) serves as a recognition motif for the SCF(beta)(-TrCP) ubiquitin ligase. Expression of IkappaKbeta dramatically increases processing of wild-type p105, but not of p105-Delta918-934. Dominant-negative beta-TrCP inhibits IkappaK-dependent processing. Furthermore, the ligase and wild-type p105 but not p105-Delta918-934 associate physically following phosphorylation. In vitro, SCF(beta)(-TrCP) specifically conjugates and promotes processing of phosphorylated p105. Importantly, the TrCP recognition motif in p105 is different from that described for IkappaBs, beta-catenin and human immunodeficiency virus type 1 Vpu. Since p105-Delta918-934 is also conjugated and processed, it appears that p105 can be recognized under different physiological conditions by two different ligases, targeting two distinct recognition motifs.


Subject(s)
NF-kappa B/metabolism , Peptide Synthases/physiology , Protein Precursors/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/physiology , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Animals , COS Cells , Chlorocebus aethiops , HeLa Cells , Humans , I-kappa B Kinase , I-kappa B Proteins/metabolism , Macromolecular Substances , NF-kappa B p50 Subunit , Phosphorylation , Protein Structure, Tertiary , SKP Cullin F-Box Protein Ligases , Transcription, Genetic
9.
Oncogene ; 18(45): 6163-71, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10557108

ABSTRACT

A myriad of unrelated exogenous or endogenous agents that represent a threat to the organism are capable of inducing NF-kappaB activity, including viral infection, bacterial lipids, DNA damage, oxidative stress and chemotherapuetic agents. Likewise, NF-kappaB regulates the expression of an equally diverse array of cellular genes. These findings are indicative of the widespread significance of NF-kappaB as a mediator of cellular stress. Remarkably, the NF-kappaB pathway displays the capacity to activate, in a cell- and stimulus-specific manner, only a subset of the total repertoire of NF-kappaB-responsive genes. The seemingly promiscuous nature of NF-kappaB activation poses a regulatory quagmire as to how specificity is achieved at the level of gene expression. The review will summarize recent findings and explore how they further our understanding of the mechanism by which stimulus-specific activation of NF-kappaB is achieved in response to cellular stress.


Subject(s)
Gene Expression Regulation , NF-kappa B/metabolism , Oxidative Stress/physiology , Signal Transduction , Animals , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/metabolism
10.
Biochem Biophys Res Commun ; 260(3): 691-8, 1999 Jul 14.
Article in English | MEDLINE | ID: mdl-10403828

ABSTRACT

The NF-kappaB signal transduction pathway involves the interaction of several NF-kappaB and IkappaB family members that are activated by a diverse range of extracellular signals and that stimulate many different cellular responses. The biochemical regulation of this cascade can be studied by establishing a cell-free system using purified proteins. As a first step toward establishing an in vitro model incorporating multiple combinations of NF-kappaB and IkappaB proteins, we produced purified human p65 (RelA) and human IkappaBalpha proteins and tested their functionality. Full-length RelA and IkappaBalpha proteins were overproduced by coinfection of TN5-JE cells with two recombinant baculoviruses. RelA and IkappaBalpha formed a stable complex that could be purified to >95% homogeneity. Protein-protein interactions, protein-DNA binding, and protein phosphorylation were similar to the native proteins.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Proteins , NF-kappa B/metabolism , Animals , Baculoviridae/genetics , Cell Extracts , Cell Line , Chemical Precipitation , Chromatography, Affinity , DNA Probes , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , Enzyme Activation , HeLa Cells , Humans , I-kappa B Kinase , Moths/cytology , Moths/genetics , Moths/virology , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/isolation & purification , Peptide Mapping , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , Transcription Factor RelA
11.
Curr Opin Cell Biol ; 11(2): 226-32, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10209157

ABSTRACT

The recent identification of molecular components of the signal transduction pathway regulating activation of nuclear factor-kappaB (NF-kappaB) in response to cytokines such as tumor necrosis factor alpha and interleukin-1beta allows the evaluation of how other diverse stimuli impinge on the NF-kappaB activation pathway. These studies suggest a basis for specificity in activation of specific Rel-related family members and the genetic responses they promote.


Subject(s)
Gene Expression Regulation/physiology , NF-kappa B/physiology , Signal Transduction/physiology , Transcription, Genetic/physiology , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinases/physiology , DNA-Binding Proteins/physiology , Embryonic and Fetal Development , Extremities/embryology , I-kappa B Proteins , Inflammation/physiopathology , Mice , Mice, Mutant Strains , Models, Biological , Molecular Sequence Data , Morphogenesis , Osteoclasts/cytology , Protein Kinases/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Stress, Physiological/physiopathology
12.
Science ; 284(5412): 321-5, 1999 Apr 09.
Article in English | MEDLINE | ID: mdl-10195897

ABSTRACT

Phosphorylation of inhibitor of kappa B (IkappaB) proteins is an important step in the activation of the transcription nuclear factor kappa B (NF-kappaB) and requires two IkappaB kinases, IKK1 (IKKalpha) and IKK2 (IKKbeta). Mice that are devoid of the IKK2 gene had extensive liver damage from apoptosis and died as embryos, but these mice could be rescued by the inactivation of the gene encoding tumor necrosis factor receptor 1. Mouse embryonic fibroblast cells that were isolated from IKK2-/- embryos showed a marked reduction in tumor necrosis factor-alpha (TNF-alpha)- and interleukin-1alpha-induced NF-kappaB activity and an enhanced apoptosis in response to TNF-alpha. IKK1 associated with NF-kappaB essential modulator (IKKgamma/IKKAP1), another component of the IKK complex. These results show that IKK2 is essential for mouse development and cannot be substituted with IKK1.


Subject(s)
Liver/embryology , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Cell Line , DNA-Binding Proteins/metabolism , Embryonic and Fetal Development , Gene Targeting , I-kappa B Kinase , I-kappa B Proteins , Interleukin-1/pharmacology , Liver/cytology , Mice , NF-kappa B/metabolism , Phosphorylation , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Signal Transduction , Transcription Factor RelA , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology
13.
Mol Cell Biol ; 19(2): 1526-38, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9891086

ABSTRACT

Activation of the transcription factor NF-kappaB is controlled by the sequential phosphorylation, ubiquitination, and degradation of its inhibitory subunit, IkappaB. We recently purified a large multiprotein complex, the IkappaB kinase (IKK) signalsome, which contains two regulated IkappaB kinases, IKK1 and IKK2, that can each phosphorylate IkappaBalpha and IkappaBbeta. The IKK signalsome contains several additional proteins presumably required for the regulation of the NFkappaB signal transduction cascade in vivo. In this report, we demonstrate reconstitution of IkappaB kinase activity in vitro by using purified recombinant IKK1 and IKK2. Recombinant IKK1 or IKK2 forms homo- or heterodimers, suggesting the possibility that similar IKK complexes exist in vivo. Indeed, in HeLa cells we identified two distinct IKK complexes, one containing IKK1-IKK2 heterodimers and the other containing IKK2 homodimers, which display differing levels of activation following tumor necrosis factor alpha stimulation. To better elucidate the nature of the IKK signalsome, we set out to identify IKK-associated proteins. To this end, we purified and cloned a novel component common to both complexes, named IKK-associated protein 1 (IKKAP1). In vitro, IKKAP1 associated specifically with IKK2 but not IKK1. Functional analyses revealed that binding to IKK2 requires sequences contained within the N-terminal domain of IKKAP1. Mutant versions of IKKAP1, which either lack the N-terminal IKK2-binding domain or contain only the IKK2-binding domain, disrupt the NF-kappaB signal transduction pathway. IKKAP1 therefore appears to mediate an essential step of the NF-kappaB signal transduction cascade. Heterogeneity of IKK complexes in vivo may provide a mechanism for differential regulation of NF-kappaB activation.


Subject(s)
Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cloning, Molecular , HeLa Cells , Humans , I-kappa B Kinase , Intracellular Signaling Peptides and Proteins , Macromolecular Substances , Mice , Molecular Sequence Data , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Multiprotein Complexes , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Transcriptional Elongation Factors
14.
Nature ; 396(6711): 590-4, 1998 Dec 10.
Article in English | MEDLINE | ID: mdl-9859996

ABSTRACT

NF-kappaB, a ubiquitous, inducible transcription factor involved in immune, inflammatory, stress and developmental processes, is retained in a latent form in the cytoplasm of non-stimulated cells by inhibitory molecules, IkappaBs. Its activation is a paradigm for a signal-transduction cascade that integrates an inducible kinase and the ubiquitin-proteasome system to eliminate inhibitory regulators. Here we isolate the pIkappaBalpha-ubiquitin ligase (pIkappaBalpha-E3) that attaches ubiquitin, a small protein which marks other proteins for degradation by the proteasome system, to the phosphorylated NF-kappaB inhibitor pIkappaBalpha. Taking advantage of its high affinity to pIkappaBalpha, we isolate this ligase from HeLa cells by single-step immunoaffinity purification. Using nanoelectrospray mass spectrometry, we identify the specific component of the ligase that recognizes the pIkappaBalpha degradation motif as an F-box/WD-domain protein belonging to a recently distinguished family of beta-TrCP/Slimb proteins. This component, which we denote E3RSIkappaB (pIkappaBalpha-E3 receptor subunit), binds specifically to pIkappaBalpha and promotes its in vitro ubiquitination in the presence of two other ubiquitin-system enzymes, E1 and UBC5C, one of many known E2 enzymes. An F-box-deletion mutant of E3RS(IkappaB), which tightly binds pIkappaBalpha but does not support its ubiquitination, acts in vivo as a dominant-negative molecule, inhibiting the degradation of pIkappaBalpha and consequently NF-kappaB activation. E3RS(IkappaB) represents a family of receptor proteins that are core components of a class of ubiquitin ligases. When these receptor components recognize their specific ligand, which is a conserved, phosphorylation-based sequence motif, they target regulatory proteins containing this motif for proteasomal degradation.


Subject(s)
I-kappa B Proteins , Ligases/chemistry , Ubiquitins/metabolism , Amino Acid Sequence , Animals , Binding Sites , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins , Electrophoresis, Polyacrylamide Gel , GTP-Binding Proteins/isolation & purification , HeLa Cells , Humans , I-kappa B Kinase , Ligases/isolation & purification , Ligases/metabolism , Mass Spectrometry , Mice , Molecular Sequence Data , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Peptide Fragments/isolation & purification , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases , beta-Transducin Repeat-Containing Proteins
15.
J Biol Chem ; 273(46): 30410-4, 1998 Nov 13.
Article in English | MEDLINE | ID: mdl-9804806

ABSTRACT

Mononuclear phagocytes play a major role in immune and inflammatory responses. Bacterial lipopolysaccharide (LPS) induces monocytes to express a variety of genes by activating the NF-kappaB/Rel transcription factor family. Recently, we have reported that the tumor necrosis factor and interleukin 1 signaling pathways activate two kinases, IKK1 and IKK2. Phosphorylation of the IkappaB cytoplasmic inhibitors, IkappaBalpha, IkappaBbeta, and IkappaBepsilon, by these kinases triggers proteolytic degradation and the release of NF-kappaB/Rel proteins into the nucleus. At present, the role of the IKKs in LPS signaling has not been investigated. Here, we report that LPS induces IKK activity in human monocytes and THP-1 monocytic cells. The kinetics of activation of kinase activity in monocytic cells are relatively slow with maximal activity observed at 60 min, which coincides with the degradation of IkappaBs and the nuclear translocation of NF-kappaB. In transfection experiments, overexpression of wild type IKK1, a dominant negative mutant IKK1 (K44M), or wild type IKK2 did not affect LPS-induced kappaB-dependent transcription in monocytic cells. In contrast, a dominant negative mutant of IKK2 inhibited LPS induction of kappaB-dependent transcription in a dose-dependent manner. These results indicate that LPS induction of kappaB-dependent gene expression in human monocytic cells requires activation of IKK2.


Subject(s)
Lipopolysaccharides/pharmacology , Monocytes/enzymology , Protein Serine-Threonine Kinases/physiology , Cell Line , Enzyme Activation , Humans , I-kappa B Kinase , Kinetics , Monocytes/drug effects , NF-kappa B/metabolism , Promoter Regions, Genetic , Signal Transduction , Tumor Necrosis Factor-alpha/genetics
16.
Cell ; 93(5): 875-84, 1998 May 29.
Article in English | MEDLINE | ID: mdl-9630230

ABSTRACT

NF-kappaB, a key regulator of the cellular inflammatory and immune response, is activated by the HTLV-I transforming and transactivating protein Tax. We show that Tax binds to the amino terminus of the protein kinase MEKK1, a component of an IkappaB kinase complex, and stimulates MEKK1 kinase activity. Tax expression increases the activity of IkappaB kinase beta (IKKbeta) to enhance phosphorylation of serine residues in IkappaB alpha that lead to its degradation. Dominant negative mutants of both IKKbeta and MEKK1 prevent Tax activation of the NF-kappaB pathway. Furthermore, recombinant MEKK1 stimulates IKKbeta phosphorylation of IkappaB alpha. Thus, Tax-mediated increases in NF-kappaB nuclear translocation result from direct interactions of Tax and MEKK1 leading to enhanced IKKbeta phosphorylation of IkappaB alpha.


Subject(s)
Gene Products, tax/metabolism , Human T-lymphotropic virus 1 , I-kappa B Proteins , MAP Kinase Kinase Kinase 1 , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Binding Sites , COS Cells , DNA-Binding Proteins/metabolism , Enzyme Activation , Gene Expression Regulation , HeLa Cells , Humans , I-kappa B Kinase , Mutation , NF-KappaB Inhibitor alpha , Phosphorylation , Precipitin Tests , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Signal Transduction , Transfection
17.
Science ; 278(5339): 860-6, 1997 Oct 31.
Article in English | MEDLINE | ID: mdl-9346484

ABSTRACT

Activation of the transcription factor nuclear factor kappa B (NF-kappaB) is controlled by sequential phosphorylation, ubiquitination, and degradation of its inhibitory subunit IkappaB. A large multiprotein complex, the IkappaB kinase (IKK) signalsome, was purified from HeLa cells and found to contain a cytokine-inducible IkappaB kinase activity that phosphorylates IkappaB-alpha and IkappaB-beta. Two components of the IKK signalsome, IKK-1 and IKK-2, were identified as closely related protein serine kinases containing leucine zipper and helix-loop-helix protein interaction motifs. Mutant versions of IKK-2 had pronounced effects on RelA nuclear translocation and NF-kappaB-dependent reporter activity, consistent with a critical role for the IKK kinases in the NF-kappaB signaling pathway.


Subject(s)
Cell Cycle Proteins , NF-kappa B/metabolism , Phosphoprotein Phosphatases , Protein Serine-Threonine Kinases/metabolism , Cloning, Molecular , Dual Specificity Phosphatase 1 , Enzyme Activation , HeLa Cells , Helix-Loop-Helix Motifs , Humans , I-kappa B Kinase , Immediate-Early Proteins/metabolism , Leucine Zippers , Molecular Sequence Data , Phosphorylation , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
18.
EMBO J ; 16(21): 6486-94, 1997 Nov 03.
Article in English | MEDLINE | ID: mdl-9351830

ABSTRACT

Activation of the transcription factor NF-kappa B is a paradigm for signal transduction through the ubiquitin-proteasome pathway: ubiquitin-dependent degradation of the transcriptional inhibitor I kappa B in response to cell stimulation. A major issue in this context is the nature of the recognition signal and the targeting enzyme involved in the proteolytic process. Here we show that following a stimulus-dependent phosphorylation, and while associated with NF-kappa B, I kappa B is targeted by a specific ubiquitin-ligase via direct recognition of the signal-dependent phosphorylation site; phosphopeptides corresponding to this site specifically inhibit ubiquitin conjugation of I kappa B and its subsequent degradation. The ligase recognition signal is functionally conserved between I kappa B alpha and I kappa B beta, and does not involve the nearby ubiquitination site. Microinjection of the inhibitory peptides into stimulated cells abolished NF-kappa B activation in response to TNF alpha and the consequent expression of E-selectin, an NF-kappa B-dependent cell-adhesion molecule. Inhibition of NF-kappa B function by specific blocking of ubiquitin ligase activity provides a novel approach for intervening in cellular processes via regulation of unique proteolytic events.


Subject(s)
Cysteine Endopeptidases/metabolism , Ligases/antagonists & inhibitors , Multienzyme Complexes/metabolism , NF-kappa B/antagonists & inhibitors , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors , Transcription, Genetic/drug effects , Ubiquitins/metabolism , Amino Acid Sequence , Biological Transport , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , E-Selectin/biosynthesis , E-Selectin/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , HeLa Cells/drug effects , HeLa Cells/metabolism , Humans , Jurkat Cells/drug effects , Jurkat Cells/metabolism , Ligases/physiology , Molecular Sequence Data , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Proteasome Endopeptidase Complex , Signal Transduction/physiology , Transcription Factor RelB , Ubiquitin-Protein Ligases , Umbilical Veins
19.
Expert Opin Investig Drugs ; 6(5): 555-67, 1997 May.
Article in English | MEDLINE | ID: mdl-15989620

ABSTRACT

Advances in molecular medicine have revealed a key role for altered gene expression in the aetiology of many inflammatory diseases, including asthma, rheumatoid arthritis, inflammatory bowel disease and sepsis. Until recently, however, modulation of gene transcription has not been the subject of directed pharmaceutical research efforts. Notwithstanding, it is clear that the efficacy of several well-established anti-inflammatory therapeutics is mediated through their ability to modulate gene transcription. Understanding the mechanisms of action of these therapeutics and defining new gene regulatory pathways has stimulated a new wave of anti-inflammatory drug discovery. This update aims to cover our current understanding of transcription inhibitors in inflammation, including the mechanism of action of established therapeutics and the properties of new chemical entities recently described in the literature.

20.
Mol Cell Biol ; 16(4): 1295-304, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8657102

ABSTRACT

Extracellular stimuli that activate the transcription factor NF-kappaB cause rapid phosphorylation of the IkappaBalpha inhibitor, which retains NF-kappaB in the cytoplasm of nonstimulated cells. Phosphorylation of IkappaBalpha is followed by its rapid degradation, the inhibition of which prevents NF-kappaB activation. To determine the relationship between these events, we mapped the inducible phosphorylation sites of IkappaBalpha. We found that two residues, serines 32 and 36, were phosphorylated in response to either tumor necrosis factor, interleukin-1, or phorbol ester. Substitution of either serine blocks or slows down induction of IkappaBalpha degradation. Substitutions of the homologous sites in IkappaBbeta, serines 19 and 23, also prevent inducible IkappaBbeta degradation. We suggest that activation of a single IkappaB kinas e or closely related IkappaB kinases is the first cr itical step in NF-kappaB activation. Once phosphorylated, IkappaB is ubiquitinated. Unlike wild-type IkappaBalpha, the phosphorylation-defective mutants do not undergo inducible polyubiquitination. As substitution of a conserved lysine residue slows down the ubiquitination and degradation of IkappaBalpha without affecting its phosphorylation, polyubiquitination is required for inducible IkappaB degradation.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Proteins , NF-kappa B/antagonists & inhibitors , Ubiquitins/metabolism , 3T3 Cells , Animals , DNA-Binding Proteins/genetics , HeLa Cells , Humans , I-kappa B Kinase , Interleukin-1/pharmacology , Lysine/genetics , Mice , Mutation , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Peptide Mapping , Phosphorylation , Precipitin Tests , Protease Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Serine/genetics , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...