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 Immunol ; 164(10): 5352-61, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10799898

ABSTRACT

Virus infection of host cells activates a set of cellular genes, including cytokines, IFNs, and chemokines, involved in antiviral defense and immune activation. Previous studies demonstrated that virus-induced transcriptional activation of a member of the human CC-chemokine RANTES required activation of the latent transcription factors IFN-regulatory factor (IRF)-3 and NF-kappa B via posttranslational phosphorylation. In the present study, we further characterized the regulatory control of RANTES transcription during virus infection using in vivo genomic footprinting analyses. IRF-3, the related IRF-7, and NF-kappa B are identified as important in vivo binding factors required for the cooperative induction of RANTES transcription after virus infection. Using fibroblastic or myeloid cells, we demonstrate that the kinetics and strength of RANTES virus-induced transcription are highly dependent on the preexistence of IRFs and NF-kappa B. Use of dominant negative mutants of either I kappa B-alpha or IRF-3 demonstrate that disruption of either pathway dramatically abolishes the ability of the other to bind and activate RANTES expression. Furthermore, coexpression of IRF-3, IRF-7, and p65/p50 leads to synergistic activation of RANTES promoter transcription. These studies reveal a model of virus-mediated RANTES promoter activation that involves cooperative synergism between IRF-3/IRF-7 and NF-kappa B factors.


Subject(s)
Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation, Viral/immunology , NF-kappa B/physiology , Transcription Factors/physiology , Base Sequence , Binding Sites/genetics , Binding Sites/immunology , Cell Line , DNA-Binding Proteins/metabolism , Drug Synergism , Humans , Interferon Regulatory Factor-3 , Interferon Regulatory Factor-7 , Interferon Type I/physiology , Molecular Sequence Data , Promoter Regions, Genetic/immunology , Protein Binding/genetics , Protein Binding/immunology , Respirovirus/genetics , Respirovirus/immunology , Response Elements/immunology , Transcription Factors/metabolism , Transfection , U937 Cells
2.
Gene ; 237(1): 1-14, 1999 Sep 03.
Article in English | MEDLINE | ID: mdl-10524230

ABSTRACT

Interferons are a large family of multifunctional secreted proteins involved in antiviral defense, cell growth regulation and immune activation. Viral infection induces transcription of multiple IFN genes, a response that is in part mediated by the interferon regulatory factors (IRFs). The initially characterized members IRF-1 and IRF-2 are now part of a growing family of transcriptional regulators that has expanded to nine members. The functions of the IRFs have also expanded to include distinct roles in biological processes such as pathogen response, cytokine signaling, cell growth regulation and hematopoietic development. The aim of this review is to provide an update on the novel discoveries in the area of IRF transcription factors and the important roles of the new generation of IRFs--particularly IRF-3, IRF-4 and IRF-7.


Subject(s)
DNA-Binding Proteins/physiology , Interferons/genetics , Interferons/metabolism , Phosphoproteins/physiology , Transcription Factors/physiology , Amino Acid Sequence , Animals , Apoptosis/physiology , Cell Division/physiology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Humans , Immune System/metabolism , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Interferon Regulatory Factor-3 , Interferon Regulatory Factor-7 , Interferon Regulatory Factors , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, gamma Subunit , Leukemia, T-Cell/metabolism , Molecular Sequence Data , Phosphoproteins/chemistry , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
3.
Mol Cell Biol ; 19(9): 6140-53, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10454561

ABSTRACT

In unstimulated cells, NF-kappaB transcription factors are retained in the cytoplasm by inhibitory IkappaB proteins. Upon stimulation by multiple inducers including cytokines or viruses, IkappaBalpha is rapidly phosphorylated and degraded, resulting in the release of NF-kappaB and the subsequent increase in NF-kappaB-regulated gene expression. IkappaBalpha gene expression is also regulated by an NF-kappaB autoregulatory mechanism, via NF-kappaB binding sites in the IkappaBalpha promoter. In previous studies, tetracycline-inducible expression of transdominant repressors of IkappaBalpha (TD-IkappaBalpha) progressively decreased endogenous IkappaBalpha protein levels. In the present study, we demonstrate that expression of TD-IkappaBalpha blocked phorbol myristate acetate-phytohemagglutinin or tumor necrosis factor alpha-induced IkappaBalpha gene transcription and abolished NF-kappaB DNA binding activity, due to the continued cytoplasmic sequestration of RelA(p65) by TD-IkappaBalpha. In vivo genomic footprinting revealed stimulus-responsive protein-DNA binding not only to the -63 to -53 kappaB1 site but also to the adjacent -44 to -36 Sp1 site of the IkappaBalpha promoter. In vivo protection of both sites was inhibited by tetracycline-inducible TD-IkappaBalpha expression. Prolonged NF-kappaB binding and a temporal switch in the composition of NF-kappaB complexes bound to the -63 to -53 kappaB1 site of the IkappaBalpha promoter were also observed; with time after induction, decreased levels of transcriptionally active p50-p65 and increased p50-c-Rel heterodimers were detected at the kappaB1 site. Mutation of either the kappaB1 site or the Sp1 site abolished transcription factor binding to the respective sites and the inducibility of the IkappaBalpha promoter in transient transfection studies. These observations provide the first in vivo characterization of a promoter proximal transcriptional switch involving NF-kappaB and Sp1 that is essential for autoregulation of the IkappaBalpha promoter.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Switch , I-kappa B Proteins , NF-kappa B/metabolism , Podophyllin/analogs & derivatives , Base Sequence , Binding Sites/genetics , DNA/genetics , DNA/metabolism , DNA Footprinting , DNA Methylation , DNA Primers/genetics , DNA-Binding Proteins/metabolism , Gene Expression , Humans , Jurkat Cells , Molecular Sequence Data , NF-KappaB Inhibitor alpha , Podophyllin/metabolism , Podophyllotoxin/analogs & derivatives , Promoter Regions, Genetic , Transcription, Genetic , Transfection
4.
J Virol ; 73(4): 2694-702, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10074115

ABSTRACT

We have examined the consequences of overexpression of the IkappaBalpha and IkappaBbeta inhibitory proteins on the regulation of NF-kappaB-dependent beta interferon (IFN-beta) gene transcription in human cells after Sendai virus infection. In transient coexpression studies or in cell lines engineered to express different forms of IkappaB under tetracycline-inducible control, the IFN-beta promoter (-281 to +19) linked to the chloramphenicol acetyltransferase reporter gene was differentially inhibited in response to virus infection. IkappaBalpha exhibited a strong inhibitory effect on virus-induced IFN-beta expression, whereas IkappaBbeta exerted an inhibitory effect only at a high concentration. Despite activation of the IkappaB kinase complex by Sendai virus infection, overexpression of the double-point-mutated (S32A/S36A) dominant repressors of IkappaBalpha (TD-IkappaBalpha) completely blocked IFN-beta gene activation by Sendai virus. Endogenous IFN-beta RNA production was also inhibited in Tet-inducible TD-IkappaBalpha-expressing cells. Inhibition of IFN-beta expression directly correlated with a reduction in the binding of NF-kappaB (p50-RelA) complex to PRDII after Sendai virus infection in IkappaBalpha-expressing cells, whereas IFN-beta expression and NF-kappaB binding were only slightly reduced in IkappaBbeta-expressing cells. These experiments demonstrate a major role for IkappaBalpha in the regulation of NF-kappaB-induced IFN-beta gene activation and a minor role for IkappaBbeta in the activation process.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Viral , Interferon-beta/genetics , Respirovirus Infections/genetics , Respirovirus , Amino Acid Sequence , Cell Line , DNA-Binding Proteins/biosynthesis , Humans , I-kappa B Proteins , Interferon-beta/biosynthesis , Molecular Sequence Data , RNA, Messenger/analysis , RNA, Messenger/genetics , Transcription, Genetic , Transcriptional Activation
5.
J Interferon Cytokine Res ; 19(1): 1-13, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10048763

ABSTRACT

The interferon (IFN) regulatory factors (IRF) consist of a growing family of related transcription proteins first identified as regulators of the IFN-alpha/beta gene promoters, as well as the IFN-stimulated response element (ISRE) of some IFN-stimulated genes. IRF-3 was originally identified as a member of the IRF family based on homology with other IRF family members and on binding to the ISRE of the IFN-stimulated gene 15 (ISG15) promoter. Several recent studies have focused attention on the unique molecular properties of IRF-3 and its role in the regulation of IFN gene expression. IRF-3 is expressed constitutively in a variety of tissues, and the relative levels of IRF-3 mRNA do not change in virus-infected or IFN-treated cells. Following virus infection, IRF-3 is posttranslationally modified by protein phosphorylation at multiple serine and threonine residues, located in the carboxy-terminus of IRF-3. Phosphorylation causes the cytoplasmic to nuclear translocation of IRF-3, stimulation of DNA binding, and increased transcriptional activation, mediated through the association of IRF-3 with the CBP/p300 coactivator. The purpose of this review is to summarize recent investigations demonstrating the important role of IRF-3 in cytokine gene transcription. These studies provide the framework for a model in which virus-dependent phosphorylation of IRF-3 alters protein conformation to permit nuclear translocation, association with transcriptional partners, and primary activation of IFN and IFN-responsive genes.


Subject(s)
DNA-Binding Proteins/physiology , Interferons/metabolism , Transcription Factors/physiology , Amino Acid Sequence , Cytokines/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Interferon Regulatory Factor-3 , Molecular Sequence Data , Phosphorylation , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcriptional Activation
6.
Oncogene ; 14(21): 2575-83, 1997 May 29.
Article in English | MEDLINE | ID: mdl-9191057

ABSTRACT

Cyclin A transcription is cell cycle regulated and induced by cell proliferative signals. To understand the mechanisms underlined in this regulation in normal human cells, we have analysed in vivo protein-DNA interactions at the Cyclin A locus in primary T lymphocytes. Stimulation of purified T lymphocytes by a combination of monoclonal antibodies directed at CD2 and CD28 adhesion molecules gives rise to a long lasting proliferation in the absence of accessory cells. Cyclin A was observed after 4 days of costimulation with anti CD2 + CD28 whereas stimulation by anti CD2 or anti CD28 alone was not effective. In vivo genomic DMS footprinting revealed upstream of the major transcription initiation sites, the presence of at least three protein binding sites, two of which were constitutively occupied. They bind in vitro respectively ATF-1 and NF-Y proteins. The third site was occupied in quiescent cells or in cells stimulated by anti CD2 or anti CD28 alone. The mitogenic combination of anti CD2 + anti CD28 released the footprint as cells were committed to proliferation. Consistent with theses results, nuclear extracts prepared from quiescent cells formed a specific complex with this element, whereas extracts prepared from cells treated with anti CD2 + anti CD28 failed to do so after cells entered a proliferative state.


Subject(s)
CD2 Antigens/physiology , CD28 Antigens/physiology , Cyclins/genetics , Gene Expression Regulation , T-Lymphocytes/physiology , Activating Transcription Factor 1 , Antibodies, Monoclonal , Binding Sites , CCAAT-Enhancer-Binding Proteins , Cell Cycle/physiology , Cell Division/physiology , Cells, Cultured , Cyclins/metabolism , DNA Footprinting , DNA-Binding Proteins/metabolism , Humans , Lymphocyte Activation , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , T-Lymphocytes/metabolism , Time Factors , Transcription Factors/metabolism
8.
Mol Cell Biol ; 16(12): 6829-40, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8943338

ABSTRACT

The interleukin 2 receptor alpha-chain (IL-2R alpha) gene is a key regulator of lymphocyte proliferation. IL-2R alpha is rapidly and potently induced in T cells in response to mitogenic stimuli. Interleukin 2 (IL-2) stimulates IL-2R alpha. transcription, thereby amplifying expression of its own high-affinity receptor. IL-2R alpha transcription is at least in part controlled by two positive regulatory regions, PRRI and PRRII. PRRI is an inducible proximal enhancer, located between nucleotides -276 and -244, which contains NF-kappaB and SRE/CArG motifs. PRRII is a T-cell-specific enhancer, located between nucleotides -137 and -64, which binds the T-cell-specific Ets protein Elf-1 and HMG-I(Y) proteins. However, none of these proximal regions account for the induction of IL-2R alpha transcription by IL-2. To find new regulatory regions of the IL-2R alpha gene, 8.5 kb of the 5' end noncoding sequence of the IL-2R alpha gene have been sequenced. We identified an 86-nucleotide fragment that is 90% identical to the recently characterized murine IL-2-responsive element (mIL-2rE). This putative human IL-2rE, designated PRRIII, confers IL-2 responsiveness on a heterologous promoter. PRRIII contains a Stat protein binding site that overlaps with an EBS motif (GASd/EBSd). These are essential for IL-2 inducibility of PRRIII/CAT reporter constructs. IL-2 induced the binding of Stat5a and b proteins to the human GASd element. To confirm the physiological relevance of these findings, we carried out in vivo footprinting experiments which showed that stimulation of IL-2R alpha expression correlated with occupancy of the GASd element. Our data demonstrate a major role of the GASd/EBSd element in IL-2R alpha regulation and suggest that the T-cell-specific Elf-1 factor can serve as a transcriptional repressor.


Subject(s)
DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Interleukin-2/metabolism , Milk Proteins , Proteins/genetics , Receptors, Interleukin-2/genetics , T-Lymphocytes/metabolism , Trans-Activators/genetics , Animals , Base Sequence , Cell Line , Ephrin-A2 , Humans , Mice , Molecular Sequence Data , Receptors, Interleukin-2/metabolism , STAT5 Transcription Factor , Tumor Suppressor Proteins
9.
Nucleic Acids Res ; 24(9): 1682-7, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8649986

ABSTRACT

A wide variety of cells usefully but sometimes destructively produce nitric oxide via inducible nitric oxide synthase (iNOS). Data obtained by gel shift analysis and reporter assays have linked murine iNOS gene induction by cytokines and bacterial products with the binding of a number of proteins to a proximal promoter, as well as to a distal enhancer of the iNOS gene. Nevertheless, these techniques do not necessarily reflect protein occupation of sites in vivo. To address this, we have used dimethyl sulphate in vivo footprinting to determine binding events in the two murine iNOS transcription control regions, using a classical lipopolysaccharide induction of RAW 264.7 macrophages. Protein-DNA interactions are absent before activation. Exposure to lipopolysaccharide induces protection at a NF-kappaB site and hypersensitivity at a shared gamma-activated site/interferon-stimulated response element within the enhancer. Protections are seen at a NF-IL6, and an Oct site within the promoter. We also observe modulations in guanine methylation at two regions which do not correspond to any known putative binding elements. Furthermore, we confirm the probable involvement of interferon regulatory factor-1 (binding to its -901 to -913 site) and the binding of NF-kappaB to its proximal site. Our data demonstrate an abundance of hitherto-unrecognised protein-DNA binding events upon simple lipopolysaccharide activation of the iNOS gene and suggests a role for protein-protein interactions in its transcriptional induction.


Subject(s)
DNA Footprinting/methods , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/physiology , Nitric Oxide Synthase/genetics , Promoter Regions, Genetic/physiology , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Proteins , Cell Line , Cytokines/pharmacology , DNA/chemistry , DNA/metabolism , Enzyme Induction , Guanine/chemistry , Interferon Regulatory Factor-1 , Lipopolysaccharides/pharmacology , Macrophages , Methylation , Mice , Molecular Sequence Data , NF-kappa B/metabolism , Nitric Oxide Synthase/biosynthesis , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Polymerase Chain Reaction/methods , Transcription, Genetic
10.
J Immunol ; 156(1): 360-70, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-8598486

ABSTRACT

HIV-1 infection is associated with a progressive and functional decline in the CD4+ lymphoid Th1 subset. Here, we propose that the HIV nef gene product may function as a specific regulator of Th1 cytokine production. By use of a T cell-specific inducible expression system, we show that upon T cell activation, induced nef expression down-regulated both IL-2 and IFN-gamma production in a dose-dependent manner, whereas IL-4, IL-9, IL-13, IL-8, and TNF-alpha production remained unaffected. In addition to this, independent transfected clones expressing various nef genes, including nef sequences amplified directly from an HIV-1 primary clinical isolate, displayed a similar pattern of cytokine expression. The specific Th1 impairment induced by nef, therefore, seems to be an important and conserved feature of HIV-1 infection and may represent a significant function of this viral gene in AIDS pathogenesis.


Subject(s)
Down-Regulation/immunology , Gene Expression Regulation, Viral/immunology , Genes, nef/immunology , HIV-1/genetics , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Th1 Cells/metabolism , Base Sequence , Cell Culture Techniques , Down-Regulation/genetics , Humans , Interferon-gamma/genetics , Interleukin-2/genetics , Molecular Sequence Data , Transfection
11.
EMBO J ; 14(20): 5060-72, 1995 Oct 16.
Article in English | MEDLINE | ID: mdl-7588634

ABSTRACT

IL-2R alpha transcription is developmentally restricted to T cells and physiologically dependent on specific stimuli such as antigen recognition. To analyse the mechanisms used to activate IL-2R alpha transcription as well as those used to block it in non-expressing cells, we determined the protein-DNA interactions at the IL-2R alpha locus in three different cell types using the DMS/LMPCR genomic footprinting method. CD25/IL-2R alpha can be efficiently induced in primary human T cells since approximately 100% express this gene when receiving an appropriate combination of mitogenic stimuli. To understand why IL-2R alpha is not expressed in other haematopoietic cell types, we analysed BJAB B lymphoma cells which do not express the IL-2R alpha gene and contain constitutively active nuclear NF-kappa B. Primary fibroblasts from embryo and adult skin were selected to examine the mechanisms that may be used to keep the IL-2R alpha gene inactive in non-haematopoietic cells. The three main results are: (i) the stable in vivo occupancy of IL-2R alpha kappa B element in resting T cells, most probably by constitutive NF-kappa B p50 homodimer that could impair SRF binding to the flanking SRE/CArG box; (ii) its inducible occupancy by NF-kappa B p50-p65 associated with the binding of an SRE/CArG box DNA-binding factor upon mitogenic stimulation; and (iii) a correlation between the precommitment of T cells to activation and the presence of stable preassembled protein-DNA complexes in contrast with the bare IL-2R alpha locus in non-T cells.


Subject(s)
Gene Expression Regulation , NF-kappa B/metabolism , Receptors, Interleukin-2/genetics , T-Lymphocytes/metabolism , Transcription, Genetic , Antineoplastic Agents/pharmacology , Base Sequence , Biological Transport , Cell Compartmentation , Cell Differentiation , DNA Footprinting , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Humans , Lymphocyte Activation , Models, Genetic , Molecular Sequence Data , NF-kappa B p50 Subunit , Nuclear Proteins/metabolism , Proline/analogs & derivatives , Proline/pharmacology , Promoter Regions, Genetic , Protein Binding , Serum Response Factor , Thiocarbamates/pharmacology , Tumor Cells, Cultured
12.
Cell Growth Differ ; 4(11): 947-54, 1993 Nov.
Article in English | MEDLINE | ID: mdl-7905283

ABSTRACT

Stimulation of primary human T-lymphocytes via CD2 and CD28 adhesion molecules induces a long-lasting proliferation (> 3 weeks). This potent activation does not require accessory cells, such as monocytes, but depends on persistent interleukin 2 (IL-2) secretion and receptivity, which is associated with high and prolonged expression of the inducible CD25/IL-2 receptor alpha (IL-2R alpha) chain gene. The transcription factor NF-kappa B participates in the regulation of both IL-2 and IL-2R alpha genes, as well as multiple cellular genes involved in T-cell proliferation. To evaluate the role of NF-kappa B in human peripheral blood T-lymphocytes, we previously analyzed the activation of NF-kappa B-related complexes in response to CD2+CD28 costimulation. We demonstrated a long-term induction of p50/p65 heterodimer, a putative p65/c-Rel heterodimer, and a constitutive nuclear expression of KBF1/p50 homodimers. As the role of p50 remains unclear, we focused our present study on NF-kappa B1 (p50/p105) gene regulation. Using electrophoretic mobility shift assays and Western and Northern blot analyses, we studied NF-kappa B1 gene expression during T-cell stimulation via CD2+CD28. We observed a transient 4- to 5-fold increase of NF-kappa B1 gene expression at both the mRNA and protein levels, lasting for at least 24 h. p50 DNA-binding activity apparently stays highly controlled when p105 expression is enhanced by a physiological stimulus of peripheral blood T-cells. Partial inhibition of p50 and p105 expression by NF-kappa B1 antisense oligonucleotides significantly reduced T-cell proliferation and CD25/IL-2R alpha cell surface expression.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antigens, CD/immunology , Gene Expression Regulation/physiology , Lymphocyte Activation/immunology , NF-kappa B/genetics , T-Lymphocytes/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Base Sequence , Binding, Competitive , CD2 Antigens , CD28 Antigens/immunology , Cell Adhesion Molecules/immunology , Electrophoresis, Paper , Electrophoresis, Polyacrylamide Gel/methods , Humans , Molecular Sequence Data , Oligonucleotide Probes , Oligonucleotides, Antisense , Proteins/analysis , RNA, Messenger/analysis , Receptors, Immunologic/immunology , Receptors, Interleukin-2/biosynthesis
13.
Cell Growth Differ ; 4(4): 329-39, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8098618

ABSTRACT

Stimulation of highly purified human T-cells via CD2 and CD28 adhesion molecules induces and maintains proliferation for more than 3 weeks. This potent interleukin 2 (IL-2)-dependent activation does not require monocytes or accessory cells. Long-lasting IL-2 receptivity is associated with high-level expression of the inducible IL-2 receptor alpha chain (IL-2R alpha) gene that is regulated at both transcriptional and posttranscriptional levels. Increase of IL-2R alpha gene transcription involves the enhanced binding of the transcription factor NF-kappa B to its consensus sequence in the 5'-regulatory region of the IL-2R alpha gene. To dissect the molecular basis for the unusually persistent transcription of the IL-2R alpha gene, we analyzed nuclear NF-kappa B binding to a radiolabeled IL-2R alpha kappa B-specific oligonucleotide probe during the time course of CD2 + CD28 activation. Resting T-cell nuclear extracts contained KBF1/p50 homodimer. After stimulation, two new kappa B-specific complexes were identified as NF-kappa B p50-p65 heterodimer and putative c-Rel homodimer or c-Rel-p65 heterodimer. Both inducible complexes persisted for at least 3 weeks. Their relative levels were very similar for the duration of proliferation. In parallel, CD2 + CD28 activation triggered a significant intracellular thiol decrease, suggesting that oxygen radicals are involved in the signaling pathway of adhesion molecules. Finally, micromolar amounts of pyrrolidine dithiocarbamate, an oxygen radical scavenger that efficiently blocked the nuclear appearance of NF-kappa B in T-lymphocytes, also inhibited IL-2 secretion, IL-2R alpha cell surface expression, and T-cell proliferation. Together, these results suggest that NF-kappa B plays an important role in long-term activation of human primary T-lymphocytes via CD2 + CD28.


Subject(s)
Antigens, CD , Cell Adhesion Molecules , Cell Nucleus/metabolism , Lymphocyte Activation/physiology , NF-kappa B/biosynthesis , T-Lymphocytes/physiology , Antibodies, Monoclonal , Antigens, Differentiation, T-Lymphocyte , Base Sequence , CD2 Antigens , CD28 Antigens , Cell Division/drug effects , Cells, Cultured , Free Radical Scavengers , Humans , Molecular Sequence Data , NF-kappa B/physiology , Pyrrolidines/pharmacology , Receptors, Immunologic , Sulfhydryl Compounds/metabolism , Thiocarbamates/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...