Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Exp Med ; 194(1): 79-88, 2001 Jul 02.
Article in English | MEDLINE | ID: mdl-11435474

ABSTRACT

Somatic cell mutagenesis is a powerful tool for characterizing receptor systems. We reported previously two complementation groups of mutant cell lines derived from CD14-transfected Chinese hamster ovary--K1 fibroblasts defective in responses to bacterial endotoxin. Both classes of mutants expressed a normal gene product for Toll-like receptor (TLR)4, and fully responded to stimulation by tumor necrosis factor (TNF)-alpha or interleukin (IL)-1 beta. We identified the lesion in one of the complementation groups in the gene for MD-2, a putative TLR4 coreceptor. The nonresponder phenotype of this mutant was reversed by transfection with MD-2. Cloning of MD-2 from the nonresponder cell line revealed a point mutation in a highly conserved region resulting in a C95Y amino acid exchange. Both forms of MD-2 colocalized with TLR4 on the cell surface after transfection, but only the wild-type cDNA reverted the lipopolysaccharide (LPS) nonresponder phenotype. Furthermore, soluble MD-2, but not soluble MD-2(C95Y), functioned to enable LPS responses in cells that expressed TLR4. Thus, MD-2 is a required component of the LPS signaling complex and can function as a soluble receptor for cells that do not otherwise express it. We hypothesize that MD-2 conformationally affects the extracellular domain of TLR4, perhaps resulting in a change in affinity for LPS or functioning as a portion of the true ligand for TLR4.


Subject(s)
Antigens, Surface/genetics , Drosophila Proteins , Endotoxins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigens, Surface/metabolism , CHO Cells/drug effects , Cell Line , Clone Cells , Cloning, Molecular , Cricetinae , DNA Mutational Analysis , Genetic Complementation Test , Humans , Interleukin-1/pharmacology , Interleukin-6/metabolism , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/toxicity , Lymphocyte Antigen 96 , Membrane Glycoproteins/genetics , Mutation , Receptors, Cell Surface/genetics , Signal Transduction , Toll-Like Receptor 4 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/pharmacology
2.
Int Immunopharmacol ; 1(3): 423-32, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11367527

ABSTRACT

Screening of 29 strains of Neisseria gonorrhoeae revealed that 16/21 serum resistant strains and 0/8 serum sensitive strains bound C4bp, suggesting that C4bp binding to gonococci could contribute to serum resistance. C4bp bound to gonococci retained cofactor (C4b-degrading) function. Using allelic exchange to construct strains with hybrid Por1A/B molecules, we demonstrate that the N-terminal loop (loop 1) of Por1A is required for C4bp binding. Serum resistant Por1B gonococcal strains also bind C4bp via their Por molecule. Using allelic exchange and site-directed mutagenesis, we have shown that loops 5 and 7 together form a negatively charged C4bp binding domain. C4bp-Por1B interactions are ionic in nature (inhibited by high salt as well as by heparin), while the C4bp-Por1A bond is hydrophobic. mAbs directed against SCR1 of the alpha-chain of C4bp inhibit C4bp binding to both Por1A and Por1B. Furthermore, only recombinant C4bp mutant molecules that contain alpha-chain SCR1 bind both Por1A and Por1B gonococci, confirming that SCR1 contains Por binding sites. C4bp alpha-chain monomers do not bind strains with either Por molecule, suggesting that the polymeric form of C4bp is required for binding to gonococci. Inhibition of C4bp binding to serum resistant Por1A and Por1B strains in a serum bactericidal assay using fAb fragments against C4bp SCR1 results in complete killing at 30 min of otherwise fully serum resistant strains in only 10% normal serum, underscoring the role of C4bp in mediating gonococcal serum resistance.


Subject(s)
Complement Inactivator Proteins , Glycoproteins , Neisseria gonorrhoeae/immunology , Porins/metabolism , Receptors, Complement/metabolism , Amino Acid Sequence , Binding Sites , Blood Bactericidal Activity/immunology , Complement C4/metabolism , Humans , Immunoglobulin M/metabolism , In Vitro Techniques , Molecular Sequence Data , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/pathogenicity , Phenotype , Porins/chemistry , Porins/genetics , Porins/immunology , Sequence Homology, Amino Acid
3.
J Exp Med ; 193(3): 281-95, 2001 Feb 05.
Article in English | MEDLINE | ID: mdl-11157049

ABSTRACT

We screened 29 strains of Neisseria gonorrhoeae and found 16/21 strains that resisted killing by normal human serum and 0/8 serum sensitive strains that bound the complement regulator, C4b-binding protein (C4bp). Microbial surface-bound C4bp demonstrated cofactor activity. We constructed gonococcal strains with hybrid porin (Por) molecules derived from each of the major serogroups (Por1A and Por1B) of N. gonorrhoeae, and showed that the loop 1 of Por1A is required for C4bp binding. Por1B loops 5 and 7 of serum-resistant gonococci together formed a negatively charged C4bp-binding domain. C4bp-Por1B interactions were ionic in nature (inhibited by high salt or by heparin), whereas the C4bp-Por1A bond was hydrophobic. Only recombinant C4bp mutant molecules containing the NH2-terminal alpha-chain short consensus repeat (SCR1) bound to both Por1A and Por1B gonococci, suggesting that SCR1 contained Por binding sites. C4bp alpha-chain monomers did not bind gonococci, indicating that the polymeric form of C4bp was required for binding. Using fAb fragments against C4bp SCR1, C4bp binding to Por1A and Por1B strains was inhibited in a complement-dependent serum bactericidal assay. This resulted in complete killing of these otherwise fully serum resistant strains in only 10% normal serum, underscoring the importance of C4bp in mediating gonococcal serum resistance.


Subject(s)
Complement C4b/immunology , Complement Inactivator Proteins , Glycoproteins , Neisseria gonorrhoeae/immunology , Porins/immunology , Receptors, Complement/immunology , Amino Acid Sequence , Base Sequence , Cell Line , Complement C4/immunology , Humans , Molecular Sequence Data , Peptide Fragments/immunology , Porins/genetics , Protein S/immunology , Receptors, Complement/genetics
4.
J Immunol ; 165(8): 4272-80, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035061

ABSTRACT

Gram-negative bacteria and the LPS constituent of their outer membranes stimulate the release of inflammatory mediators believed to be responsible for the clinical manifestations of septic shock. The GPI-linked membrane protein, CD14, initiates the signaling cascade responsible for the induction of this inflammatory response by LPS. In this paper, we report the generation and characterization of CD14-null mice in which the entire coding region of CD14 was deleted. As expected, LPS failed to elicit TNF-alpha and IL-6 production in macrophages taken from these animals, and this loss in responsiveness is associated with impaired activation of both the NF-kappaB and the c-Jun N-terminal mitogen-activated protein kinase pathways. The binding and uptake of heat-killed Escherichia coli, measured by FACS analysis, did not differ between CD14-null and wild-type macrophages. However, in contrast to the findings with LPS, whole E. coli stimulated similar levels of TNF-alpha release from CD14-null and wild-type macrophages at a dose of 10 bioparticles per cell. This effect was dose dependent, and at lower bacterial concentrations CD14-deficient macrophages produced significantly less TNF-alpha than wild type. Approximately half of this CD14-independent response appeared to be mediated by CD11b/CD18, as demonstrated by receptor blockade using neutrophil inhibitory factor. An inhibitor of phagocytosis, cytochalasin B, abrogated the induction of TNF-alpha in CD14-deficient macrophages by E. coli. These data indicate that CD14 is essential for macrophage responses to free LPS, whereas other receptors, including CD11b/CD18, can compensate for the loss of CD14 in response to whole bacteria.


Subject(s)
Escherichia coli/immunology , Gene Deletion , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , Animals , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Outer Membrane Proteins/physiology , CD18 Antigens/physiology , Cell Line , Cytokines/biosynthesis , Escherichia coli/physiology , Female , Lipopolysaccharide Receptors/blood , Macrophage Activation/genetics , Macrophage-1 Antigen/physiology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Insertional , Phagocytosis/immunology , Signal Transduction/immunology
5.
J Biol Chem ; 274(30): 21049-55, 1999 Jul 23.
Article in English | MEDLINE | ID: mdl-10409656

ABSTRACT

CD14-transfected Chinese hamster ovary K1 fibroblasts (CHO/CD14) respond to lipopolysaccharide (LPS) by metabolizing arachidonic acid and with translocation of NF-kappaB to the nucleus. Although previous experiments failed to identify the production of tumor necrosis factor-alpha and interleukin (IL)-1beta by CHO/CD14 cells, LPS did induce the expression of IL-6 mRNA and the subsequent release of the IL-6 protein. To identify additional LPS-inducible genes, a cDNA library derived from LPS-stimulated CHO/CD14 cells was screened by subtractive hybridization. Fourteen genes were found to be expressed differentially, and two were analyzed in detail: hop (Hsp70/Hsp90-organizing protein), which is the hamster homologue of the stress-inducible yeast gene, STI1, and clone H411, which encodes a novel LPS-inducible growth factor. In response to LPS, the expression of Hop mRNA was also increased in both the murine macrophage cell line, RAW 264.7, as well as in primary hamster macrophages. This suggested that the up-regulation of Hop expression is part of the macrophage stress response to LPS. Clone H411 encodes a protein in the epidermal growth factor-like repeat protein family. Overexpression of H411 cDNA in the RAW 264.7 macrophage cell line promoted an increased growth rate, suggesting that expression of H411 is part of the proliferative cell response to LPS. Both Hop and H411 represent novel gene products not previously recognized as part of the complex biological response to endotoxin.


Subject(s)
Fungal Proteins/genetics , Gene Expression Regulation/drug effects , Growth Substances/genetics , Heat-Shock Proteins/genetics , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/pharmacology , Molecular Chaperones , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Fungal Proteins/metabolism , Growth Substances/metabolism , Heat-Shock Proteins/metabolism , Humans , Mice , Molecular Sequence Data , Rats , Sequence Alignment
6.
J Immunol ; 162(12): 6971-5, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10358136

ABSTRACT

Toll-like receptor (TLR) 2 and TLR4 have been implicated in the responses of cells to LPS (endotoxin). CD14-transfected Chinese hamster ovary (CHO)-K1 fibroblasts (CHO/CD14) are exquisitely sensitive to endotoxin. Sequence analysis of CHO-TLR2, compared with human and mouse TLR2, revealed a single base pair deletion. This frameshift mutation resulted in an alternative stop codon, encoding a protein devoid of transmembrane and intracellular domains. CHO-TLR2 cDNA failed to enable LPS signaling upon transient transfection into human epithelial kidney 293 cells. Site-directed mutagenesis of CHO-TLR2 enabled expression of a presumed full-length hamster TLR2 that conferred LPS responsiveness in human epithelial kidney 293 cells. Genomic TLR2 DNA from primary hamster macrophages also contained the frameshift mutation found in CHO fibroblasts. Nevertheless, hamster peritoneal macrophages were found to respond normally to LPS, as evidenced by the induction of cytokines. These results imply that expression of TLR2 is sufficient but not essential for mammalian responses to endotoxin.


Subject(s)
Alleles , CHO Cells/metabolism , Drosophila Proteins , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Base Sequence , CHO Cells/immunology , Cloning, Molecular , Cricetinae , Female , Humans , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/deficiency , Mice , Molecular Sequence Data , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/deficiency , Sequence Deletion , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors
7.
J Biol Chem ; 274(20): 13993-8, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10318811

ABSTRACT

The activation of phagocytes by lipopolysaccharide (LPS) has been implicated in the pathogenesis of Gram-negative sepsis. Although the interaction between CD14 and LPS is a key event in the signaling cascade, the molecular mechanism by which cellular activation occurs remains obscure. We hypothesized that the main function of CD14 was to bind LPS and transfer it to a second receptor, which then initiates the subsequent signal for cellular activation. Thus, surface binding of LPS to the cell membrane would be the critical step that CD14 carries out. To test this hypothesis, we examined the activity of two other proteins known to bind LPS, lipopolysaccharide-binding protein and bactericidal/permeability-increasing protein. We found that when these normally soluble proteins were expressed in Chinese hamster ovary-K1 fibroblasts as glycosylphosphatidylinositol-anchored proteins, both could substitute for CD14 in initiating LPS signaling. Pharmacological studies with synthetic lipid A analogues demonstrated that these surface expressed LPS-binding proteins had characteristics that were qualitatively identical to membrane CD14. These data support the hypothesis that a receptor distinct from CD14 functions as the actual signal transducer and suggest that surface binding of LPS to the cell membrane is the crucial first step for initiating downstream signaling events.


Subject(s)
Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/pharmacology , Membrane Proteins/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides , Base Sequence , Blood Bactericidal Activity , Blood Proteins/metabolism , CD11 Antigens/metabolism , CHO Cells , Cricetinae , Fibroblasts/drug effects , Fibroblasts/metabolism , Gram-Negative Bacteria/metabolism , Humans , Molecular Sequence Data
8.
J Immunol ; 161(10): 5413-20, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9820516

ABSTRACT

The activation of phagocytes by the lipid A moiety of LPS has been implicated in the pathogenesis of Gram-negative sepsis. While two LPS receptors, CD14 and CD11/CD18, have been associated with cell signaling, details of the LPS signal transduction cascade remain obscure. CD14, which exists as a GPI-anchored and a soluble protein, lacks cytoplasmic-signaling domains, suggesting that an ancillary molecule is required to activate cells. The CD11/CD18 integrins are transmembrane proteins. Like CD14, they are capable of mediating LPS-induced cellular activation when expressed on the surface of hamster fibroblasts Chinese hamster ovary (CHO)-K1. The observation that a cytoplasmic deletion mutant is still capable of activating transfected CHO-K1 argues that CD11/CD18 also utilizes an associated signal transducer. We sought to identify further similarities between the signaling systems utilized by CD14 and CD11/CD18. LPS-binding protein, which transfers LPS to CD14, enhanced both LPS-induced cellular activation and binding of Gram-negative bacteria in CD11/CD18-transfected CHO-K1, thus implying that LPS-binding protein can also transfer LPS to CD11/CD18. When synthetic lipid A analogues were analyzed for their ability to function as LPS agonists, or antagonists, in the CHO transfectants, we found the effects were identical regardless of which LPS receptor was expressed. This supports the hypothesis that a receptor distinct from CD14 and CD11/CD18 is responsible for discriminating between the lipid A of LPS and the LPS antagonists. We propose that this receptor, which is the target of the LPS antagonists, functions as the true signal transducer in LPS-induced cellular activation for both CD14 and CD11/CD18.


Subject(s)
Acute-Phase Proteins , CD11 Antigens/physiology , CD18 Antigens/physiology , Lipid A/physiology , Lipopolysaccharide Receptors/physiology , Membrane Glycoproteins , Signal Transduction/immunology , Animals , CD11 Antigens/genetics , CHO Cells , Carrier Proteins/physiology , Cell Line , Cricetinae , Gram-Negative Bacteria/immunology , Humans , Lipid A/analogs & derivatives , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Mice , Mycobacterium/immunology , Peptidoglycan/pharmacology , Transfection/drug effects , Transfection/immunology , Tumor Cells, Cultured
9.
J Immunol ; 159(7): 3519-30, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9317151

ABSTRACT

Human phagocytes recognize bacterial LPS (endotoxin) through membrane CD14 (mCD14), a proinflammatory LPS receptor. This study tested the hypothesis that anti-LPS Abs neutralize endotoxin by blocking cellular uptake through mCD14. Ab-associated changes in the uptake and cellular distribution of FITC-LPS were assessed by flow cytometry and laser scanning confocal microscopy in human CD14-transfected Chinese hamster ovary fibroblasts (CHO-CD14 cells) and human peripheral blood monocytes. LPS core- and O-side chain-specific mAbs inhibited mCD14-mediated LPS uptake by both cell types in the presence of serum. O-side chain-specific mAb concurrently enhanced complement-dependent LPS uptake by monocytes through complement receptor-1 (CR1) and uptake by CHO-CD14 cells involving another heat-labile serum factor(s) and cell-associated recognition molecule(s). Core-specific mAb inhibited mCD14-mediated uptake of homologous and heterologous LPS, while producing less concurrent enhancement of non-mCD14-mediated LPS uptake. The modulation by anti-LPS mAbs of mCD14-mediated LPS uptake was associated with inhibition of LPS-induced nuclear factor-kappaB (NF-kappaB) translocation and TNF-alpha secretion in CHO-CD14 cells and monocytes, respectively, while mAb enhancement of non-mCD14-mediated LPS uptake stimulated these activities. LPS-specific Abs thus mediate anti-inflammatory and proinflammatory functions, respectively, by preventing target cell uptake of LPS through mCD14 and augmenting uptake through CR1 or other cell receptors.


Subject(s)
Antibodies, Bacterial/pharmacology , Antibodies, Monoclonal/pharmacology , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Monocytes/immunology , Monocytes/pathology , Adult , Animals , Antibodies, Bacterial/chemistry , Antibodies, Monoclonal/chemistry , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Biological Transport/immunology , CHO Cells , Cricetinae , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Inflammation/blood , Inflammation/etiology , Inflammation/immunology , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/blood , Lipopolysaccharides/chemistry , Monocytes/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Protein Conformation , Receptors, Complement 3b/immunology
10.
J Exp Med ; 186(12): 2051-6, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9396775

ABSTRACT

Gram-negative bacterial lipopolysaccharide (LPS) stimulates phagocytic leukocytes by interacting with the cell surface protein CD14. Cellular responses to LPS are markedly potentiated by the LPS-binding protein (LBP), a lipid-transfer protein that binds LPS aggregates and transfers LPS monomers to CD14. LBP also transfers LPS to lipoproteins, thereby promoting the neutralization of LPS. LBP present in normal plasma has been shown to enhance the LPS responsiveness of cells in vitro. The role of LBP in promoting LPS responsiveness in vivo was tested in LBP-deficient mice produced by gene targeting in embryonic stem cells. Whole blood from LBP-deficient animals was 1,000-fold less responsive to LPS as assessed by the release of tumor necrosis factor (TNF)-alpha. Blood from gene-targeted mice was devoid of immunoreactive LBP, essentially incapable of transferring LPS to CD14 in vitro, and failed to support cellular responses to LPS. These activities were restored by the addition of exogenous recombinant murine LBP to the plasma. Despite these striking in vitro findings, no significant differences in TNF-alpha levels were observed in plasma from wild-type and LBP-deficient mice injected with LPS. These data suggest the presence of an LBP-independent mechanism for responding to LPS. These LBP knockout mice may provide a tool for discovering the nature of the presumed second mechanism for transferring LPS to responsive cells.


Subject(s)
Acute-Phase Proteins/genetics , Carrier Proteins/physiology , Gene Deletion , Lipopolysaccharides/pharmacology , Membrane Glycoproteins , Animals , Carrier Proteins/genetics , Chimera , In Vitro Techniques , Kidney/chemistry , Kidney/drug effects , Lipopolysaccharide Receptors/metabolism , Liver/chemistry , Liver/drug effects , Mice , Mice, Knockout , Mice, Mutant Strains , Tumor Necrosis Factor-alpha/metabolism
11.
J Immunol ; 152(9): 4444-54, 1994 May 01.
Article in English | MEDLINE | ID: mdl-8157962

ABSTRACT

NF-beta A is a monocyte, neutrophil, and B cell-specific nuclear protein that is involved in regulation of the IL-1 beta gene. These studies further define the functional role of NF-beta A in RAW264.7 monocytic cells by using transient transfection analysis. We showed that NF-beta A was able to activate transcription from a heterologous promoter in a distance-independent and dose-dependent manner. NF-beta A also appeared to function in a positionally independent manner within the IL-1 beta cap-site proximal (CSP) promoter. NF-beta A was required for maximal IL-1 beta gene expression directed by the upstream LPS-inducible enhancer element. Deletion of the NF-beta A-binding sequence resulted in an 80% reduction in basal reporter gene activity and an 86% reduction in LPS-inducible reporter gene activity in constructs containing only the enhancer and CSP promoter. Other regulatory elements located between the enhancer and the cap site were not able to substitute functionally for the absence of NF-beta A. Recently, other investigators have reported that IL-1 beta CSP promoter function was decreased by introducing multiple mutations within both the NF-beta A-binding sequence, and a putative overlapping NF-IL-6-binding sequence. We have found that these mutations predominantly affect NF-beta A binding. Furthermore NF-beta A, and not NF-IL-6, was required for supporting basal and LPS-inducible transcription from a minimal IL-1 beta CSP promoter (positions -58 to +11). This promoter region did not appear to direct monocyte-specific IL-1 beta gene expression because reporter constructs containing the IL-1 beta CSP promoter were also active in transiently transfected HeLa cells.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-1/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins , Cell Line , DNA/genetics , DNA Probes/genetics , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/drug effects , Gene Expression , HeLa Cells , Humans , Lipopolysaccharides/pharmacology , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription, Genetic
12.
Mol Immunol ; 31(2): 139-51, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8309477

ABSTRACT

Interleukin 1 beta (IL-1 beta) is a proinflammatory cytokine that exhibits a wide variety of biological activities. Genomic sequences that mediate the induction of human IL-1 beta gene transcription by lipopolysaccharide and phorbol esters are located more than 2,700 bp upstream of the transcriptional start site (cap site). These upstream elements require additional cap site-proximal (CSP) sequences which are necessary for basal transcription of the human IL-1 beta gene. In addition, these CSP sequences have been shown to mediate both cell type-specific expression of this gene, and trans-activation by some viral proteins. In this study, we report the identification of a novel nuclear protein, termed NF beta C, that binds to a DNA sequence which spans the cap site of the human IL-1 beta gene (positions -12 to +8). We have also identified a second region (positions -305 to -280) containing a putative NF-kappa B binding site. We show here that this region can bind three distinct nuclear proteins. One protein is similar or identical to NF-kappa B, a second protein (termed NF beta B) binds a distinct sequence that substantially overlaps the 5' half of the NF kappa B binding sequence, and a third protein (termed NF beta D) binds a distinct sequence that substantially overlaps the 3' half of the NF kappa B binding sequence. Unlike NF kappa B, NF1 beta B and NF beta D are present in nuclear extracts prepared from unstimulated monocytic cells. Although the NF beta D and NF beta C binding sequences share no significant similarity, each sequence can specifically compete for the binding of either protein to DNA, whereas oligonucleotides containing only the NF kappa B or NF beta B motifs do not compete for the binding of NF beta C or NF beta D. This suggests that NF beta C and NF beta D can specifically interact in vitro, possibly through a common subunit.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-1/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Animals , Base Sequence , Binding, Competitive , Cells, Cultured , DNA/genetics , DNA/metabolism , Humans , Mice , Molecular Sequence Data , NF-kappa B/metabolism , Protein Binding
13.
Mol Cell Biol ; 12(8): 3439-48, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1630455

ABSTRACT

Prointerleukin 1 beta (IL-1 beta) is a cytokine that mediates a broad range of biological activities. Genomic sequences that regulate IL-1 beta transcription include both inducible regulatory elements located more than 2,700 bp upstream of the transcriptional start site (cap site) and proximal elements located near the TATA box of this gene. In this study, we focused on the identification and characterization of trans-acting nuclear regulatory proteins that bind to the cap site-proximal region of the human IL-1 beta gene. We identified a protein, termed NFIL-1 beta A (NF beta A), that binds to a highly conserved 12-bp DNA sequence (-49 to -38) located upstream of the TATA box motif in both the human and murine IL-1 beta genes. The IL-1 alpha gene, which lacks a TATA motif, does not possess an NF beta A-binding sequence within the promoter region, suggesting that NF beta A may selectively regulate IL-1 beta expression. Using electrophoretic mobility shift assays, we identified several distinct DNA-protein complexes that are expressed in a cell-type-specific manner. In monocytic cell lines, the relative abundance of these complexes varies rapidly following stimulation of the cells with phorbol esters or lipopolysaccharide. UV cross-linking analysis identified two distinct DNA-binding polypeptides that comprise distinct complexes. The functional role of NF beta A was assessed in transient transfection assays. These data indicate that NF beta A is required for both basal and inducible promoter activity in monocytic cells. Furthermore, the human cytomegalovirus immediate-early 1 gene product requires the presence of NF beta A in order to trans-activate the proximal IL-1 beta promoter in a monocytic cell line. We propose that NF beta A is a factor that mediates either direct or indirect activation by the immediate-early 1 gene product. The proximity of this essential factor to the TATA motif suggests a possible role in transcriptional initiation.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-1/genetics , Promoter Regions, Genetic , Protein Precursors/genetics , Transcription, Genetic , Transcriptional Activation , Animals , Base Sequence , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA Probes , Exons , Humans , Methylation , Mice , Molecular Sequence Data , Regulatory Sequences, Nucleic Acid , Restriction Mapping , Retroviridae Proteins, Oncogenic , Sequence Homology, Nucleic Acid , TATA Box , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...