Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Pharmacol ; 59(2): 302-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160867

ABSTRACT

Polycyclic aromatic hydrocarbons (PAH) are ubiquitous environmental chemicals that suppress the immune system at multiple levels, including at the level of B cell development in the bone marrow microenvironment. Specifically, PAH induce preB cell apoptosis in primary bone marrow cultures and in cocultures of an early preB cell line (BU-11) and a bone marrow stromal cell line (BMS2). Previous studies focused on the molecular mechanisms through which PAH induce stromal cells to deliver an apoptosis signal to adjacent preB cells. Apoptosis signaling within the preB cell itself was not investigated. Here, the role of NF-kappaB, a lymphocyte survival factor, in PAH-induced preB cell apoptosis was assessed. Analysis of DNA-binding proteins extracted from the nuclei of untreated BU-11 cells indicated DNA-binding complexes comprising NF-kappaB subunits p50, c-Rel, and/or Rel A. NF-kappaB down-regulation with previously described inhibitors induced BU-11 cell apoptosis, demonstrating that the default apoptosis pathway blocked by NF-kappaB is functional at this early stage in B cell development. Similarly, exposure of BU-11/BMS2 cocultures to 7,12-dimethylbenz[a]anthracene (DMBA), a prototypic PAH, down-regulated nuclear Rel A and c-Rel before overt apoptosis. Finally, ectopic expression of Rel A or c-Rel rescued BU-11 cells from DMBA-induced apoptosis. These results extend previous observations by demonstrating that 1) NF-kappaB is a survival factor at an earlier stage of B cell development than previously appreciated and 2) NF-kappaB down-regulation is likely to be part of the molecular mechanism resulting in PAH-induced preB cell apoptosis. These results suggest nonclonally restricted, PAH-mediated suppression of B lymphopoiesis.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/pharmacology , Apoptosis , B-Lymphocytes/cytology , NF-kappa B/physiology , Animals , Carcinogens/pharmacology , Cell Survival , Down-Regulation , Environmental Pollutants/pharmacology , Mice , NF-kappa B/biosynthesis , NF-kappa B/metabolism , Proto-Oncogene Proteins c-rel/biosynthesis , Proto-Oncogene Proteins c-rel/physiology , Transcription Factor RelA , Tumor Cells, Cultured
2.
Chem Biol Interact ; 126(2): 137-57, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10862814

ABSTRACT

Members of the Rel family of proteins have been identified in Drosophila, an echinoderm, Xenopus, birds and mammals. Dimers of Rel proteins form the transcription factor nuclear factor kappaB (NF-kappaB) that rapidly activates genes encoding cytokines, cell surface receptors, cell adhesion molecules and acute phase proteins. Evidence suggests that xenobiotic compounds also may alter the activation of NF-kappaB. This study had a dual objective of identifying members of the Rel family and examining their activation by xenobiotic compounds in a marine fish model, scup (Stenotomus chrysops). A DNA-protein crosslinking technique demonstrated that liver, kidney and heart each had at least three nuclear proteins that showed specific binding to an NF-kappaB consensus sequence, with molecular weights suggesting that the proteins potentially corresponded to mouse p50, p65 (RelA) and c-rel. In addition, an approximately 35kD NF-kappaB binding protein was evident in liver and kidney. The 50 kD protein was immunoprecipitated by mammalian p50-specific antibodies. The presence of Rel members in fish implied by those results was confirmed by RT-PCR cloning of a Rel homology domain (an apparent c-rel) from scup liver. NF-kappaB activation occurred in vehicle-treated fish, but this appeared to decrease over time. In fish treated with 0.01 or 1 mg 3,3',4,4', 5-pentachlorobiphenyl per kg, NF-kappaB activation in liver did not decrease, and there was a 6-8-fold increase in activation 16-18 days following treatment. Treatment with 10 mg benzo[a]pyrene/kg had no effect on NF-kappaB-DNA binding, either at 3 or 6 days following treatment. The data show that the Rel family of proteins is present in fish, represented at least by a p50/105 homologue, and support a hypothesis that some aryl hydrocarbon receptor agonists can activate NF-kappaB in vivo.


Subject(s)
Gene Expression Regulation , Microsomes, Liver/enzymology , NF-kappa B/metabolism , Polychlorinated Biphenyls/pharmacology , Proto-Oncogene Proteins c-rel/genetics , Proto-Oncogene Proteins c-rel/metabolism , Receptors, Aryl Hydrocarbon/agonists , Amino Acid Sequence , Animals , Base Sequence , Chickens , Consensus Sequence , Cytochrome P-450 Enzyme System/genetics , Fishes , Gene Expression Regulation/drug effects , Humans , Kidney/metabolism , Liver/metabolism , Mice , Molecular Sequence Data , Phylogeny , Sequence Alignment , Sequence Homology, Amino Acid , Turkeys , Xenobiotics/pharmacology , Xenopus laevis
3.
J Immunol ; 163(1): 269-77, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10384125

ABSTRACT

Members of the NF-kappa B/Rel family of transcription factors are involved in many aspects of B lymphocyte development and function. NF-kappa B is constitutively active in these cells, in contrast with most other cell types. In the inactive form, NF-kappa B/Rel proteins are sequestered in the cytoplasm by members of the I kappa B family of NF-kappa B inhibitors. When activated, NF-kappa B is translocated to the nucleus, a process that involves the phosphorylation and proteasomal degradation of I kappa B proteins. Thus, NF-kappa B activation is accompanied by the rapid turnover of I kappa B proteins. We show that while this "classical" mode of NF-kappa B activation is a uniform feature of IgM+ B cell lines, all IgG+ B cells analyzed contain nuclear NF-kappa B yet have stable I kappa B alpha, I kappa B beta, and I kappa B epsilon. Furthermore, I kappa beta epsilon levels are at least 10 times lower in IgG+ B cells than in IgM+ B cells, an additional indication that the regulation of constitutive NF-kappa B activity in these two types of B cells is fundamentally different. These data imply the existence of a novel mechanism of NF-kappa B activation in IgG+ B cells that operates independently of I kappa B degradation. They further suggest that different isoforms of the B cell receptor may have distinct roles in regulating NF-kappa B activity.


Subject(s)
B-Lymphocyte Subsets/metabolism , Cell Nucleus/metabolism , I-kappa B Proteins , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , B-Lymphocyte Subsets/drug effects , B-Lymphocyte Subsets/immunology , Biological Transport , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Immunoglobulin Class Switching , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Lipopolysaccharides/pharmacology , Lymphoma, B-Cell , Mice , NF-KappaB Inhibitor alpha , Proto-Oncogene Proteins/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor RelB , Tumor Cells, Cultured
4.
J Immunol ; 158(6): 2551-7, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9058786

ABSTRACT

Pre-B cells can express secretory mu (mu(s))- as well as membrane mu (mu(m))-chains. We evaluated the ability of mu(s)-chains to associate with surrogate light chains and assemble into a pre-B cell receptor (BCR) complex in pre-B cells, and explored whether mu(s)-chains could be exploited to generate a secreted soluble pre-BCR. We demonstrate that mu(s)-chains can associate with SLC internally. The mu(s)-containing complexes form higher order polymeric structures, but these are never assembled into completed covalent structures. Instead, the complexes are efficiently retained and rapidly degraded. Alteration of the intracellular redox state by incubation with 2-ME resulted in the secretion of mu(s)-chains, suggesting that they are retained by a thiol-mediated retention mechanism. To identify the sequences on mu(s)-chains responsible for their retention, we generated stable transfectants of a mu-negative pre-B cell line expressing either wild-type or mutant mu(s) constructs. Mutation of a single cysteine (Cys575) in the mu(s) tailpiece resulted in the release and secretion of the mu(s) H chains. These were associated with the surrogate light chain proteins lambda5 and VpreB, and thus appear to constitute an authentic secreted soluble pre-BCR. The soluble pre-BCR has a specificity distinct from Ab consisting of the same heavy chain V region paired with conventional light chains.


Subject(s)
B-Lymphocytes/metabolism , Receptors, Antigen, B-Cell/metabolism , Stem Cells/metabolism , Animals , Antibody Specificity , B-Lymphocytes/immunology , Biopolymers/metabolism , Cell Line , Cysteine/genetics , Cysteine/physiology , Immunoglobulin Light Chains/metabolism , Immunoglobulin M/metabolism , Immunoglobulin mu-Chains/metabolism , Mice , Solubility , Stem Cells/immunology
5.
J Biol Chem ; 271(14): 8321-7, 1996 Apr 05.
Article in English | MEDLINE | ID: mdl-8626528

ABSTRACT

Activation of T lymphocytes requires the combined signaling of the T cell receptor and costimulatory molecules such as CD28. The ability of T cells to produce interleukin-2 (IL-2) is a critical control point in T lymphocyte activation. The IL-2 enhancer contains a functional motif named CD28 response element (CD28RE) that serves a role as a target for mitogenic T cell activation signals. The CD28RE sequence reveals similarity to the consensus kappaB binding motif. Here we demonstrate that CD28RE binds an inducible protein with a molecular mass of approximately 35 kDa called nuclear factor of mitogenic-activated T cells (NF-MATp35) that is clearly different from the known NF- kappaB/Rel family members. Induction of NF-MATp35 was shown to depend on de novo protein synthesis and was restricted to T cells that received a mitogenic combination of T cell stimuli, not necessarily including CD28 signaling. Nonmitogenic T cell stimulation did not result in appearance of NF-MATp35. These results indicate that mitogenic combinations of T cell activation signals are integrated at the level of NF-MATp35 induction. Similar to its effect on IL-2 production, cyclosporin A inhibited the induction of NF-MATp35. Taken together, these data demonstrate that the nuclear appearance of NF-MATp35 shows excellent correlation with IL-2 production, which is a unique characteristic among nuclear factors implicated in the control of IL-2 gene expression.


Subject(s)
CD28 Antigens/metabolism , Enhancer Elements, Genetic , Interleukin-2/genetics , Nuclear Proteins/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Base Sequence , Consensus Sequence , Cross-Linking Reagents , Cycloheximide/pharmacology , Cyclosporine/pharmacology , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Gene Expression Regulation, Viral , HIV-1/genetics , Humans , Molecular Weight , NF-kappa B/metabolism , Tumor Cells, Cultured
6.
Proc Natl Acad Sci U S A ; 90(3): 1023-7, 1993 Feb 01.
Article in English | MEDLINE | ID: mdl-8430069

ABSTRACT

The long terminal repeat (LTR) of the type 1 human immunodeficiency virus (HIV-1) and the 5' regulatory region of the gene encoding the interleukin 2 receptor alpha subunit (IL-2R alpha) share functional kappa B enhancer elements involved in the regulation of these inducible transcription units during T-cell activation. These kappa B enhancer elements are recognized by a structurally related family of interactive proteins that includes p50, p65, and the product of the c-rel protooncogene (c-Rel). Recent biochemical studies have shown that p65 and p50 form the prototypical NF-kappa B complex, which is rapidly translocated from the cytoplasm to the nucleus during T-cell activation. This intracellular signaling complex potently stimulates kappa B-directed transcription from either the HIV-1 LTR or the IL-2R alpha promoter via the strong transactivation domain present in p65. We now demonstrate that nuclear expression of human c-Rel, which is induced by either phorbol ester or tumor necrosis factor alpha with delayed kinetics relative to p65, markedly represses p65-mediated activation of these transcription units. These inhibitory effects of c-Rel correlate with its DNA-binding activity but not with its ability to heterodimerize with p50, suggesting that c-Rel inhibition involves competition with p50/p65 for occupancy of the kappa B enhancer element. Together, these findings suggest that one function of c-Rel is as a physiologic repressor of the HIV-1 LTR and IL-2R alpha promoters, serving to efficiently counter the strong transcriptional activating effects of p65.


Subject(s)
Gene Expression Regulation, Viral/drug effects , HIV Long Terminal Repeat/genetics , HIV-1/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins/pharmacology , Repressor Proteins/pharmacology , Base Sequence , Chloramphenicol O-Acetyltransferase/genetics , DNA Mutational Analysis , Enhancer Elements, Genetic/genetics , Humans , Interleukin-2/genetics , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-rel , T-Lymphocytes , Transcription, Genetic/drug effects , Transfection
7.
Semin Immunol ; 4(5): 287-96, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1336679

ABSTRACT

Mouse mammary tumor proviruses (MMTV) use a common enhancer/promoter region to accommodate their transcription in two different cellular environments. In mammary tissue, transcription is regulated through the hormone response element located in the 5' LTR. In B cells, transcription is hormone independent, can be stimulated following B cell activation, and is distinct from the transcription of other known inducible genes, including immunoglobulin. The open reading frame (ORF) in the viral 3' LTR has at least two functions. Its gene product(s) acts as a viral superantigen, but also has autoregulatory properties, leading to MMTV transactivation. We propose a scheme suggesting that MMTV evolved to use the B cell as an intermediary in its viral life cycle.


Subject(s)
Antigens, Viral, Tumor/biosynthesis , B-Lymphocytes/immunology , Genes, Viral , Mammary Tumor Virus, Mouse/genetics , Proviruses , Animals , Gene Expression Regulation, Neoplastic , Mice , Open Reading Frames/physiology , Repetitive Sequences, Nucleic Acid , Transcription, Genetic , Transcriptional Activation
8.
Proc Natl Acad Sci U S A ; 89(5): 1875-9, 1992 Mar 01.
Article in English | MEDLINE | ID: mdl-1542686

ABSTRACT

Molecular cloning of the polypeptide component of the Rel-related human p75 nucleoprotein complex has revealed its identity with the 65-kDa (p65) subunit of NF-kappa B. Functional analyses of chimeric proteins composed of NF-kappa B p65 C-terminal sequences linked to the DNA-binding domain of the yeast GAL4 polypeptide have indicated that the final 101 amino acids of NF-kappa B p65 comprise a potent transcriptional activation domain. Transient transfection of human T cells with an expression vector encoding NF-kappa B p65, but not NF-kappa B p50, produced marked transcriptional activation of a basal promoter containing duplicated kappa B enhancer motifs from the long terminal repeat of type 1 human immunodeficiency virus. These stimulatory effects of NF-kappa B p65 were synergistically enhanced by coexpression of NF-kappa B p50 but were completely inhibited by coexpression of the v-rel oncogene product. Together, these functional studies demonstrate that NF-kappa B p65 is a transactivating subunit of the heterodimeric NF-kappa B complex and serves as one cellular target for v-Rel-mediated transcriptional repression.


Subject(s)
NF-kappa B/physiology , Repressor Proteins/physiology , Retroviridae Proteins, Oncogenic/physiology , Transcription Factors/physiology , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Enhancer Elements, Genetic , Gene Expression Regulation , Humans , Macromolecular Substances , Molecular Sequence Data , Molecular Weight , NF-kappa B/chemistry , Oligodeoxyribonucleotides/chemistry , Oncogene Proteins v-rel , Repressor Proteins/chemistry , Trans-Activators , Transcription Factors/chemistry , Transcription, Genetic
9.
Oncogene ; 6(4): 665-7, 1991 Apr.
Article in English | MEDLINE | ID: mdl-2030915

ABSTRACT

HIVEN86A is an inducible member of a set of cellular proteins that specifically bind to the kappa B enhancer (Franza et al., 1987; Franza, 1988; Franza, 1990; Ballard et al., 1989; Bohnlein et al., 1988). This enhancer motif has been detected in numerous cellular and viral transcription control domains (Boshart et al., 1985; Sen & Baltimore, 1986; Nabel & Baltimore, 1987). Recently, cDNAs have been cloned (Kieran et al., 1990; Baldwin & Sharp, 1987) that encode the 50 kD DNA binding subunit of murine NF-kappa B (for review: Leonardo & Baltimore, 1989) and the closely related human kappa binding factor (KBF-1) (Kimura et al., 1986; Baldwin & Sharp, 1987). A 350 amino acid domain at the N-terminus of these proteins was found to be homologous with the v-rel oncogene from the avian reticuloendotheliosis virus, strain T (REV-T), as well as a maternal effect gene, dorsal (Kieran et al., 1990; Ghosh et al., 1990). Dorsal is known to activate transcription of certain Drosophila genes (Rushlow et al., 1987). The v-Rel oncoprotein has been identified as a transcriptional activator (Gelinas & Temin, 1988; Hannink & Temin, 1989; Bull et al., 1990) in certain assay systems and shown to be induced by the tumor promoter, phorbol 12-myristate 13-acetate (PMA) in avian cells (for review: Rice & Gilden, 1988). HIVEN86A is also inducible by PMA (Franza et al., 1987; Franza, 1988; Franza, 1990). We now demonstrate that the protein product of the human c-rel proto-oncogene is structurally identical to HIVEN86A.


Subject(s)
DNA-Binding Proteins/analysis , Nuclear Proteins/analysis , Proto-Oncogene Proteins/analysis , Electrophoresis, Gel, Two-Dimensional , Humans , Phytohemagglutinins/pharmacology , Polymerase Chain Reaction , Proto-Oncogene Mas , Proto-Oncogene Proteins c-rel , T-Lymphocytes/drug effects , Tetradecanoylphorbol Acetate/pharmacology
10.
Proc Natl Acad Sci U S A ; 87(24): 10028-32, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2263603

ABSTRACT

A sensitive DNA-protein crosslinking approach has been used to characterize four inducible T-cell proteins (50 kDa, 55 kDa, 75 kDa, and 85 kDa) that specifically bind to kappa B enhancer elements. Partial proteolytic mapping revealed a distinct cleavage pattern for three of these proteins. These polypeptides are sequestered as inactive precursors in the cytosol of unstimulated T cells but can be converted into active forms in vivo by phorbol ester stimulation or in vitro by detergent treatment. The induction of these proteins by phorbol ester results in a strikingly biphasic pattern of nuclear expression with the 55-kDa and 75-kDa species appearing within minutes, whereas the 50-kDa and 85-kDa species appear only several hours after cellular stimulation. These data suggest that NF-kappa B-binding activity may not correspond to a single polypeptide but rather a family of at least four inducible and differentially regulated DNA-binding proteins that are expressed with distinct kinetics in human T lymphocytes.


Subject(s)
Enhancer Elements, Genetic , NF-kappa B/genetics , T-Lymphocytes/metabolism , Base Sequence , Cell Line , Cell Nucleus/metabolism , Cytosol/metabolism , Gene Expression/drug effects , HeLa Cells/metabolism , Humans , Molecular Sequence Data , Molecular Weight , NF-kappa B/biosynthesis , Oligonucleotide Probes , Peptide Mapping , Protein Binding , Tetradecanoylphorbol Acetate/pharmacology
11.
Cell ; 63(4): 803-14, 1990 Nov 16.
Article in English | MEDLINE | ID: mdl-2225078

ABSTRACT

Studies of NF-kappa B suggest that this enhancer binding activity corresponds to a family of at least four proteins (p50, p55, p75, and p85) differentially induced with biphasic kinetics during T cell activation. While p55 and p50 are closely related to the 50 kd DNA binding subunit of NF-kappa B, p75 and p85 exhibit DNA binding properties that distinguish them from this 50 kd polypeptide and its regulatory subunits I kappa B and p65. All four members of this kappa B-specific protein family are structurally related to the v-Rel oncoprotein and one, p85, appears identical to human c-Rel. v-Rel, but not nontransforming v-Rel mutants, binds to the kappa B enhancer and inhibits NF-kappa B-activated transcription from the IL-2 receptor alpha promoter and HIV-1 LTR. These findings suggest a Rel-related family of kappa B enhancer binding proteins and raise the possibility that the transforming activity of v-Rel is linked to its inhibitory action on cellular genes under NF-kappa B control.


Subject(s)
DNA-Binding Proteins/genetics , NF-kappa B/physiology , Oncogenes , Protein-Tyrosine Kinases/genetics , Retroviridae Proteins, Oncogenic/genetics , Base Sequence , DNA Probes , Enhancer Elements, Genetic , Genetic Vectors , HeLa Cells/enzymology , Humans , Molecular Sequence Data , NF-kappa B/antagonists & inhibitors , Oligonucleotide Probes , Oncogene Proteins v-rel , Peptide Mapping , Plasmids , Protein Biosynthesis , Retroviridae Proteins, Oncogenic/isolation & purification , Retroviridae Proteins, Oncogenic/metabolism , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...