Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Eur Respir J ; 26(6): 1119-37, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16319345

ABSTRACT

Substantial experimental evidence now supports the notion that allergic diseases are characterised by a skewing of the immune system towards a T-helper cell type-2 (Th2) phenotype. Studies using both human and mouse model systems have provided key evidence for the role that Th2 cytokines play in driving many of the hallmarks of allergic inflammation. Furthermore, the signalling pathways by which Th2 cytokines exert their effects on airway target cells are rapidly being elucidated, and antagonists of the Th2 pathway are under active development. In this review, the current knowledge of the role of T-helper cell type-2 cells in asthma is summarised, focusing on how and where T-helper cell type-2 cells differentiate from naïve precursors. The signalling molecules and transcription factors involved in T-helper cell type-2 differentiation will be reviewed in detail, in an attempt to translate studies using genetically modified mice into meaningful insights about asthma and other allergic diseases.


Subject(s)
Asthma/immunology , Hypersensitivity/immunology , Th2 Cells/immunology , Animals , Asthma/diagnosis , Asthma/genetics , Cell Differentiation/physiology , Dendritic Cells/immunology , Dendritic Cells/physiology , Gene Expression Regulation , Humans , Hypersensitivity/genetics , Mice , Molecular Biology , Prognosis , Signal Transduction , Species Specificity
2.
J Biol Chem ; 276(52): 48871-8, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11687571

ABSTRACT

Interleukin-4 (IL-4) is a multifunctional cytokine that plays an important role in immune and inflammatory responses. Expression of the IL-4 gene is tightly controlled at the level of gene transcription by both positive and negative regulatory elements in the IL-4 promoter. Several constitutive nuclear factors have been identified that can interact with IL-4 promoter elements in DNA binding assays. Here we report that the zinc-finger protein YY-1 (Yin-Yang 1) can bind to multiple elements within the human IL-4 promoter. Cotransfection of Jurkat T cells with different IL-4 promoter/reporter constructs together with expression vectors encoding antisense, wild-type, or zinc finger-deleted mutant YY-1 suggested that YY-1 enhanced IL-4 promoter activity in a DNA-binding domain-dependent manner. Site-directed mutagenesis revealed that a proximal YY-1-binding site, termed Y0 ((-59)TCATTTT(-53)), was essential for YY-1-driven IL-4 promoter activity. In addition, cotransfected YY-1 enhanced both IL-4 promoter activity and endogenous IL-4 gene expression in nontransformed peripheral blood T cells. Thus, YY-1 positively regulates IL-4 gene expression in lymphocytes.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Interleukin-4/genetics , Promoter Regions, Genetic , Transcription Factors/metabolism , Calcimycin/pharmacology , DNA-Binding Proteins/genetics , Erythroid-Specific DNA-Binding Factors , Genes, Reporter , Humans , Interleukin-4/metabolism , Ionophores/pharmacology , Jurkat Cells , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Protein Binding , Transcription Factors/genetics , Transcriptional Activation , Transfection , YY1 Transcription Factor , Zinc Fingers/genetics
3.
Am J Respir Cell Mol Biol ; 24(6): 755-61, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11415942

ABSTRACT

Interleukin (IL)-13 is a T helper 2-derived cytokine that has recently been implicated in allergic airway responses. We hypothesized that IL-13 may regulate expression of eotaxin in airway epithelium. We found that IL-13 upregulated eotaxin messenger RNA and protein synthesis in the airway epithelial cell line BEAS-2B; this effect showed synergy with tumor necrosis factor (TNF)-alpha and also was inhibited by the glucocorticoid budesonide. To establish the mechanisms of eotaxin upregulation by IL-13, cells were transfected with an eotaxin promoter-luciferase reporter plasmid and transcription was activated by IL-13 (1.7-fold) and TNF-alpha (2.8-fold). The combination of IL-13 and TNF-alpha additively activated the promoter constructs (4.1-fold). Activation of signal transducer and activator of transcription (STAT) 6 by IL-13 was confirmed by nuclear protein binding to a DNA probe derived from the eotaxin promoter. Activation of eotaxin transcription by IL-13 and the additive effect with TNF-alpha were lost in plasmids mutated at a putative STAT6 binding site. Cotransfection with a wild-type STAT6 expression vector significantly enhanced activation of the eotaxin promoter after IL-13 stimulation (6-fold induction). A significant increase of eotaxin protein secretion in the supernatant of STAT6 wild-type-transfected cells was observed after IL-13 stimulation. Cotransfection with a dominant negative STAT6 mutant expression vector inhibited activation of the eotaxin promoter by IL-13. These results indicate that IL-13 stimulates eotaxin expression in airway epithelial cells and that STAT6 plays a pivotal role in this response.


Subject(s)
Chemokines, CC , Cytokines/biosynthesis , Interleukin-13/pharmacology , Respiratory Mucosa/metabolism , Trans-Activators/metabolism , Cell Line, Transformed , Chemokine CCL11 , Cytokines/genetics , Humans , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , STAT6 Transcription Factor , Signal Transduction , Transcriptional Activation , Up-Regulation
4.
Blood ; 97(6): 1742-9, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238116

ABSTRACT

Previous studies indicated that aspirin (acetylsalicylic acid [ASA]) can have profound immunomodulatory effects by regulating cytokine gene expression in several types of cells. This study is the first in which concentrations of ASA in the therapeutic range were found to significantly reduce interleukin (IL)-4 secretion and RNA expression in freshly isolated and mitogen-primed human CD4+ T cells. In contrast, ASA did not affect IL-13, interferon-gamma, and IL-2 expression. ASA inhibited IL-4, but not IL-2, promoter-driven chloramphenicol acetyltransferase expression in transiently transfected Jurkat T cells. The structurally unrelated nonsteroidal anti-inflammatory drugs indomethacin and flurbiprofen did not affect cytokine gene expression in T cells, whereas the weak cyclo-oxygenase inhibitor salicylic acid was at least as effective as ASA in inhibiting IL-4 expression and promoter activity. The inhibitory effect of ASA on IL-4 transcription was not mediated by decreased nuclear expression of the known salicylate target nuclear factor (NF)-kappaB and was accompanied by reduced binding of an inducible factor to an IL-4 promoter region upstream of, but not overlapping, the NF of activated T cells- and NF-kappaB-binding P1 element. It is concluded that anti-inflammatory salicylates, by means of a previously unrecognized mechanism of action, can influence the nature of adaptive immune responses by selectively inhibiting the expression of IL-4, a critical effector of these responses, in CD4+ T cells.


Subject(s)
Aspirin/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Interleukin-4/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Cyclooxygenase Inhibitors/pharmacology , Humans , Interleukin-4/genetics , Interleukin-4/metabolism , NF-kappa B/metabolism , NF-kappa B/pharmacology , Promoter Regions, Genetic/drug effects , Transcription, Genetic/drug effects
5.
Genomics ; 71(2): 246-51, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11161818

ABSTRACT

Genetic heterogeneity has been proposed as a hallmark feature of allergic disease. To test the hypothesis that total IgE levels are jointly influenced by a locus on chromosome 12q21.1-q21.31 and a locus on 17q11.2-q21.2, we conducted multipoint allele-sharing analyses using nonparametric linkage (NPL) methods on Afro-Caribbean families from Barbados to test for evidence of gene-gene interactions. Significant correlations were observed between NPL scores at D12S1052 and both D17S1293 and D17S1299 for a dichotomized phenotype of total IgE. An analysis of family-specific NPL scores revealed that evidence for interaction was being driven largely by one multiplex pedigree (NPL = 12.01, 12.23, and 12.16 at D12S1052, D17S1293, and D17S1299, respectively). Using the programs SIMWALK (v2.0) and GOLD, a different set of haplotypes in this influential family was observed around D12S1052 and the 17q loci compared to the other Barbados pedigrees. Our findings are a classic example of founder effect, provide evidence for sensitivity of this type of linkage analysis to unusual pedigrees, and highlight an element of genetic heterogeneity that has been given little attention in the study of complex traits.


Subject(s)
Genetic Heterogeneity , Genetic Linkage/genetics , Immunoglobulin E/genetics , Asthma/epidemiology , Barbados/epidemiology , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 17/genetics , Family Health , Female , Gene Expression Regulation/genetics , Genetic Markers , Genetic Predisposition to Disease , Haplotypes , Humans , Hypersensitivity/genetics , Immunoglobulin E/metabolism , Male , Pedigree , Phenotype
6.
J Immunol ; 165(6): 3384-92, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10975857

ABSTRACT

Infection of human epithelial cells with human rhinovirus (HRV)-16 induces rapid production of several proinflammatory cytokines, including IL-8, IL-6, and GM-CSF. We evaluated the role of NF-kappaB in HRV-16-induced IL-8 and IL-6 production by EMSA using oligonucleotides corresponding to the binding sites for NF-kappaB in the IL-6 and IL-8 gene promoters. Consistent with the rapid induction of mRNA for IL-8 and IL-6, maximal NF-kappaB binding to both oligonucleotides was detected at 30 min after infection. NF-kappaB complexes contained p65 and p50, but not c-Rel. The IL-8 oligonucleotide bound recombinant p50 with only about one-tenth the efficiency of the IL-6 oligonucleotide, even though epithelial cells produced more IL-8 protein than IL-6. Neither the potent glucocorticoid, budesonide (10-7 M), nor a NO donor inhibited NF-kappaB binding to either cytokine promoter or induction of mRNA for either IL-8 or IL-6. Sulfasalazine and calpain inhibitor I, inhibitors of NF-kappaB activation, blocked HRV-16-induced formation of NF-kappaB complexes with oligonucleotides from both cytokines, but did not inhibit mRNA induction for either cytokine. By contrast, sulfasalazine clearly inhibited HRV-16 induction of mRNA for GM-CSF in the same cells. Thus, HRV-16 induces epithelial expression of IL-8 and IL-6 by an NF-kappaB-independent pathway, whereas induction of GM-CSF is at least partially dependent upon NF-kappaB activation.


Subject(s)
Bronchi/immunology , Bronchi/metabolism , Cytokines/biosynthesis , Epithelial Cells/immunology , Epithelial Cells/metabolism , NF-kappa B/physiology , Rhinovirus/immunology , Bronchi/drug effects , Bronchi/virology , Budesonide/pharmacology , Calpain/antagonists & inhibitors , Cell Line , Cytokines/genetics , Epithelial Cells/drug effects , Epithelial Cells/virology , Glycoproteins/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interleukin-6/biosynthesis , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/biosynthesis , Interleukin-8/genetics , Interleukin-8/metabolism , NF-kappa B/biosynthesis , NF-kappa B/metabolism , Nitric Oxide Donors/pharmacology , Promoter Regions, Genetic/immunology , RNA, Messenger/biosynthesis , Rhinovirus/drug effects , Sulfasalazine/pharmacology , Time Factors
7.
J Biol Chem ; 275(47): 36605-11, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-10973979

ABSTRACT

Expression of cytokine genes in T cells is thought to result from a complex network of antigen- and mitogen-activated transcriptional regulators. CP2, a factor homologous to Drosophila Elf-1 and previously found to be a critical regulator of several viral and cellular genes in response to developmental signals, is rapidly activated in T helper (Th) cells in response to mitogenic stimulation. Here we show that overexpression of CP2 enhances interleukin (IL)-4 promoter-driven chloramphenicol acetyltransferase expression, while repressing IL-2 promoter activity, in transiently transfected Jurkat cells. A CP2-protected element, partially overlapping the nuclear factor of activated T cell-binding P2 sequence, was required for IL-4 promoter activation in CP2-overexpressing Jurkat cells. This CP2-response element is the site of a cooperative interaction between CP2 and an inducible heteromeric co-factor(s). Mutation of conserved nucleotide contacts within the CP2-response element prevented CP2 binding and significantly reduced constitutive and induced IL-4 promoter activity. Expression of a CP2 mutant lacking the Elf-1-homology region of the DNA-binding domain inhibited IL-4 promoter activity in a dominant negative fashion in transiently transfected Jurkat cells. Moreover, overexpressed CP2 markedly enhanced, while its dominant negative mutant consistently suppressed, expression of the endogenous IL-4 gene in the murine Th2 cell line D10. Taken together, these findings point to CP2 as a critical IL-4 transactivator in Th cells.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-4/genetics , Promoter Regions, Genetic , Transcription Factors/metabolism , Base Sequence , Consensus Sequence , DNA/metabolism , Gene Expression Regulation , Humans , Interleukin-2/genetics , Molecular Sequence Data , RNA-Binding Proteins , T-Lymphocytes/metabolism
8.
Leukemia ; 14(4): 629-35, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10764148

ABSTRACT

Interleukin 4 (IL-4) is a multifunctional cytokine that plays an important role in hematopoiesis, tumor cell growth, and cellular immune responses. Expression of the IL-4 gene is tightly controlled at the level of gene transcription, and many positive regulatory cis-elements have been identified in the proximal IL-4 promoter region. Relatively little is known about factors that downregulate IL-4 transcription. We performed a detailed deletional analysis of the proximal human IL-4 promoter and studied reporter gene activity in transiently transfected Jurkat T lymphoblasts. In this report, we characterize a novel negative regulatory element (termed P2 NRE) that is adjacent to a binding site for nuclear factor of activated T cells. Mutation of P2 NRE significantly enhanced the activity of a 175 base pair IL-4 promoter construct in transiently transfected Jurkat T lymphoblasts. Using nuclear extracts from Jurkat cells, we identify a candidate factor (termed Rep-1) that binds uniquely to the P2 NRE in DNA-binding assays. Rep-1 is not related to other factors previously shown to interact with the IL-4 promoter, and by UV cross-linking and SDS-PAGE analysis, we found that it migrates with a molecular mass of approximately 150 kDa. Characterizing the molecular mechanisms responsible for downregulating the IL-4 promoter should enhance our understanding of IL-4-gene dysregulation in disease states.


Subject(s)
Gene Expression Regulation , Interleukin-4/genetics , Nuclear Proteins , Promoter Regions, Genetic/genetics , Repressor Proteins/metabolism , Binding Sites , DNA Mutational Analysis , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Interleukin-4/biosynthesis , Jurkat Cells , Macromolecular Substances , Molecular Weight , NFATC Transcription Factors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Repressor Proteins/isolation & purification , Sequence Alignment , Sequence Deletion , Sequence Homology, Nucleic Acid , Transcription Factors/metabolism , Transcriptional Activation , Transfection
9.
Biochem Biophys Res Commun ; 270(3): 1016-23, 2000 Apr 21.
Article in English | MEDLINE | ID: mdl-10772943

ABSTRACT

Interleukin 4 (IL-4) gene expression is controlled at the level of transcription by the complex interactions of multiple factors that bind to a proximal promoter region. Nuclear factor of activated T cells (NFAT) can bind up to five purine-rich sequences in the IL-4 promoter termed the P elements (P0-P4). In this paper, we characterize a novel P element in the upstream region of the human IL-4 promoter that we term P5. P5 shares a core NFAT motif ((-353)GGAAA(-357)) and additional sequence similarity with the other P elements and supported strong interactions between the NFATp DNA-binding domain (DBD) and the AP-1 proteins cFos and cJun in DNA-binding assays. Inducibility of the IL-4 promoter was significantly impaired in a reporter construct in which the P5 element was mutated in the context of the full-length promoter. We conclude that P5 represents a novel IL-4 promoter P element that contributes to IL-4 promoter inducibility.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-4/genetics , Promoter Regions, Genetic , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Humans , Mice , Molecular Sequence Data , NFATC Transcription Factors , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides/chemistry , Sequence Alignment , Sequence Homology, Nucleic Acid , T-Lymphocytes/immunology , Transfection
10.
J Immunol ; 164(2): 825-32, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10623828

ABSTRACT

The mechanism by which glucocorticoids (GC) inhibit IL-4 gene expression is currently unknown. In T lymphocytes, IL-4 gene expression is regulated at the level of transcription by increases in intracellular calcium concentration and by the calcium-activated phosphatase calcineurin. In this paper we report that dexamethasone (Dex) inhibits calcium ionophore-induced activation of the human IL-4 promoter in transiently transfected Jurkat T cells. Inhibition of the promoter by Dex is dependent on expression of the GC receptor (GR), because it does not occur in GR-deficient cells. Dex also represses activation of the promoter induced by cotransfecting cells with a constitutively active mutant of calcineurin. Using a series of deletion constructs, we show that the proximal 95 bp of the IL-4 promoter contain a Dex-sensitive regulatory element. This region contains the P1 sequence, a proximal binding site for NF-AT. A calcium-induced but Dex-inhibited nuclear complex containing NF-AT binds to the P1 element in EMSA. Using immunoprecipitation under nondenaturing conditions, we found that the GRalpha isoform coprecipitates with NF-ATc in nuclear extracts of calcium ionophore- and Dex-treated cells. Taken together, our results show that GC inhibit IL-4 gene expression by interfering with NF-AT-dependent transactivation of the proximal human IL-4 promoter.


Subject(s)
Calcineurin/physiology , Calcium/physiology , Dexamethasone/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-4/genetics , Lymphocyte Activation/drug effects , Nuclear Proteins , Promoter Regions, Genetic/immunology , Base Composition , Calcineurin/metabolism , Calcineurin Inhibitors , Calcium/antagonists & inhibitors , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Activation/genetics , Enzyme Activation/immunology , Humans , Interleukin-4/metabolism , Jurkat Cells , NFATC Transcription Factors , Promoter Regions, Genetic/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , Response Elements/drug effects , Response Elements/immunology , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
11.
J Immunol ; 164(3): 1612-6, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10640782

ABSTRACT

Up-regulation of C-C chemokine expression characterizes allergic inflammation and atopic diseases. A functional mutation in the proximal promoter of the RANTES gene has been identified, which results in a new consensus binding site for the GATA transcription factor family. A higher frequency of this allele was observed in individuals of African descent compared with Caucasian subjects (p < 0.00001). The mutant allele was associated with atopic dermatitis in children of the German Multicenter Allergy Study (MAS-90; p < 0.037), but not with asthma. Transient transfections of the human mast cell line HMC-1 and the T cell line Jurkat with reporter vectors driven by either the mutant or wild-type RANTES promoter showed an up to 8-fold higher constitutive transcriptional activity of the mutant promoter. This is the first report to our knowledge of a functional mutation in a chemokine gene promoter. Our findings suggest that the mutation contributes to the development of atopic dermatitis. Its potential role in other inflammatory and infectious disorders, particularly among individuals of African ancestry, remains to be determined.


Subject(s)
Chemokine CCL5/genetics , Dermatitis, Atopic/genetics , Dermatitis, Atopic/immunology , Mutation/immunology , Alleles , DNA Mutational Analysis , Gene Frequency/immunology , Humans , Jurkat Cells , Polymorphism, Single-Stranded Conformational , Promoter Regions, Genetic/immunology , Tumor Cells, Cultured
12.
J Immunol ; 163(12): 6876-83, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10586089

ABSTRACT

The C-C chemokine eotaxin is a potent chemoattractant for eosinophils and probably plays an important role in the pathogenesis of asthma, although the mechanisms of its regulation are not well known. Airway epithelial cells express eotaxin mRNA and protein after stimulation with a variety of cytokines. We focused on the molecular mechanisms of eotaxin gene regulation by TNF-alpha and IL-4 in the airway epithelial cell line, BEAS-2B. Cells were transfected with luciferase reporter plasmids, which contained up to 1363 bp of the eotaxin promoter. Eotaxin promoter activity was increased by TNF-alpha (2.5-fold) and IL-4 (1.5-fold), respectively. The combination of TNF-alpha and IL-4 produced 3.6-fold activation of the eotaxin promoter. The eotaxin promoter contains overlapping consensus binding sites for transcription factors, NF-kappa B and STAT6, which are known to mediate responses to TNF-alpha and IL-4, respectively. Electrophoretic mobility shift assays revealed NF-kappa B binding after TNF-alpha stimulation and STAT6 binding after IL-4 stimulation using a DNA probe derived from the eotaxin promoter. Mutant plasmids were generated to define the roles of these transcription factors in eotaxin promoter activity. TNF-alpha stimulation, but not IL-4 stimulation, was lost in plasmids mutated at the NF-kappa B binding site, whereas IL-4 stimulation, but not TNF-alpha stimulation, was lost in plasmids mutated at the STAT6 binding site. When both sites were mutated, all transcriptional activation was lost. These results imply that TNF-alpha and IL-4 stimulate expression of the eotaxin gene by activating NF-kappa B and STAT6.


Subject(s)
Bronchi/metabolism , Chemokines, CC , Chemotactic Factors, Eosinophil/genetics , Cytokines/genetics , Epithelial Cells/metabolism , NF-kappa B/physiology , Signal Transduction/genetics , Trans-Activators/physiology , Transcriptional Activation/immunology , Base Sequence , Binding Sites/genetics , Binding Sites/immunology , Blotting, Western , Bronchi/cytology , Bronchi/immunology , Cell Line , Chemokine CCL11 , Chemotactic Factors, Eosinophil/isolation & purification , Cloning, Molecular , Cytokines/isolation & purification , Electrophoresis, Polyacrylamide Gel , Epithelial Cells/immunology , Humans , Molecular Sequence Data , NF-kappa B/metabolism , Promoter Regions, Genetic , STAT6 Transcription Factor , Signal Transduction/immunology , Trans-Activators/metabolism , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/pharmacology
14.
Blood ; 92(12): 4529-38, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9845517

ABSTRACT

The differentiation of naive T-helper (Th) cells into cytokine-secreting effector Th cells requires exposure to multiple signals, including exogenous cytokines. Interleukin-4 (IL-4) plays a major role in this process by promoting the differentiation of IL-4-secreting Th2 cells. In Th2 cells, IL-4 gene expression is tightly controlled at the level of transcription by the coordinated binding of multiple transcription factors to regulatory elements in the proximal promoter region. Nuclear factor of activated T cell (NFAT) family members play a critical role in regulating IL-4 transcription and interact with up to five sequences (termed P0 through P4) in the IL-4 promoter. The molecular mechanisms by which IL-4 induces expression of the IL-4 gene are not known, although the IL-4-activated transcription factor signal transducer and activator of transcription 6 (Stat6) is required for this effect. We report here that Stat6 interacts with three binding sites in the human IL-4 promoter by electrophoretic mobility shift assays. These sites overlap the P1, P2, and P4 NFAT elements. To investigate the role of Stat6 in regulating IL-4 transcription, we used Stat6-deficient Jurkat T cells with different intact IL-4 promoter constructs in cotransfection assays. We show that, whereas a multimerized response element from the germline IgE promoter was highly induced by IL-4 in Stat6-expressing Jurkat cells, the intact human IL-4 promoter was repressed under similar conditions. We conclude that the function of Stat6 is highly dependent on promoter context and that this factor promotes IL-4 gene expression in an indirect manner.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation , Interleukin-4/genetics , Nuclear Proteins , Promoter Regions, Genetic/genetics , T-Lymphocytes/chemistry , Trans-Activators/physiology , Binding Sites/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Genes, Reporter , Humans , Interleukin-4/pharmacology , Jurkat Cells , NFATC Transcription Factors , Receptors, Interleukin-4/physiology , STAT6 Transcription Factor , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Transcription, Genetic/drug effects , Transfection , Tumor Cells, Cultured
15.
J Leukoc Biol ; 64(5): 681-91, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9823775

ABSTRACT

The octamer motif (ATTTGCAT) present in several eukaryotic promoters and enhancers is now known to influence the transcription of several genes by interacting with members of a broad family of homeodomain proteins. The promoter of the human class II MHC gene HLA-DRA contains a conserved octamer element that can bind (among other proteins) the transcription factor Oct-2A and the high mobility group proteins (HMG) I/Y. We have previously determined that HMG I(Y) and Oct-2A cooperatively activate HLA-DRA gene expression, most likely due to the ability of HMG I(Y) to selectively recruit Oct-2A to the octamer motif. In this report, we present results of our investigations of the mechanisms of cooperative transactivation of HLA-DRA transcription by Oct-2A and HMG I(Y). We show that both the amino- and the carboxy-terminal domains of Oct-2A are required for HLA-DRA transactivation. Experiments using domain-swap chimeras of the Oct-1 and Oct-2A polypeptides indicate that cooperative activation of the DRA gene by HMG I(Y) and Oct-2A requires the carboxy-terminal domain (CTD) of Oct-2A. However, HMG I(Y) physically interacts with the conserved POU domains of both Oct-1 and Oct-2A. We therefore postulate that the nature of the CTD attached to the POU homeodomain influences the outcome of interaction with HMG I(Y). These studies support the view that HMG I(Y) is an important cofactor for HLA-DRA gene activation by Oct-2A and provide insights into its mechanism of action.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , HLA-DR Antigens/genetics , High Mobility Group Proteins/physiology , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription Factors/physiology , Transcriptional Activation , Binding Sites , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HLA-DR alpha-Chains , HMGA1a Protein , High Mobility Group Proteins/chemistry , Host Cell Factor C1 , Humans , Jurkat Cells , Macromolecular Substances , Models, Molecular , Octamer Transcription Factor-1 , Octamer Transcription Factor-2 , POU Domain Factors , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection
17.
Int Arch Allergy Immunol ; 114(3): 207-17, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9363900

ABSTRACT

In all mammalian species investigated so far, mast cells and basophils are the only cells that synthesize histamine and express plasma membrane receptors that bind IgE with high affinity (Fc epsilonRI). Human basophils and mast cells derive from distinct precursors that originate in the bone marrow and fetal liver and probably circulate in peripheral blood. There is extensive evidence that mast cells and basophils and their mediators are primary effectors of allergic inflammation. Immunologically activated human basophils release two cytokines: IL-4 and IL-13. Expression of several cytokines has been documented in a number of experimental models of human and rodent mast cells. However, to date few studies have analyzed the mechanisms of gene expression in human Fc epsilonRI+ cells. Some of these studies imply a role for NFAT and GATA family members in the IgE-mediated activation of cytokine gene transcription in basophils and mast cells. Studies of human basophils and mast cells isolated from different anatomic sites have established the different profiles of eicosanoids released by these cells. Recently, the characterization of arachidonic acid pools and the identification of novel enzymes involved in arachidonate remodeling and mobilization clarified in part how eicosanoid productions is regulated in mast cells and basophils. In addition to histamine, human mast cell secretory granules contain the neutral proteases tryptase, chymase and carboxypeptidase that possess several biochemical properties. In particular, tryptase may play a role as a fibrogenic factor and chymase might convert angiotensin I to angiotensin II. Mast cells are present in human heart and in human coronary arteries raising the possibility that local activation of cardiac mast cells might contribute to certain cardiovascular diseases. Recent evidence also suggests that mast cells and basophils can play a role during viral and bacterial infections. It is now evident that in man these two cells not only participate in inflammation associated with allergic disease, but also in chronic and fibrotic disorders affecting several organs and in host defense against bacterial and viral infections.


Subject(s)
Basophils/physiology , Mast Cells/physiology , Animals , Arachidonic Acids/metabolism , Cardiovascular Diseases/physiopathology , Cytokines/biosynthesis , Endopeptidases/metabolism , Gene Expression Regulation/physiology , Humans , Hypersensitivity/physiopathology , Molecular Biology
18.
Curr Opin Immunol ; 8(6): 796-803, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8994858

ABSTRACT

Several immunological disorders including allergic rhinitis, bronchial asthma, atopic dermatitis, food allergies, urticaria, nonhereditary angioedema, systemic anaphylaxis, and allergic conjunctivitis are associated with a positive family history, and share a positive response in the Prausnitz-Kuster (wheal and flare) reaction. Studies have shown that 20-30% of the population has a strong genetic predisposition for this condition, termed atopy, whose hallmark is a greatly elevated serum IgE concentration. A great deal is known about the cellular interactions that mediate the sensitization, immediate and late-phase reactions that follow encounters with allergen, as well as about the cell surface and signaling events that result in mediator release from inflammatory cells. Less is known of the genes that confer genetic predisposition for atopy; however, a worldwide effort to identify atopy genes is making significant progress.


Subject(s)
Hypersensitivity, Immediate/etiology , Hypersensitivity, Immediate/genetics , Animals , Humans
19.
J Immunol ; 156(2): 424-9, 1996 Jan 15.
Article in English | MEDLINE | ID: mdl-8543789

ABSTRACT

Atopy, which predisposes individuals to develop asthma, severe systemic anaphylaxis, and atopic dermatitis, is usually associated with dramatically elevated total serum IgE levels and is thought to be controlled by a major susceptibility gene and multiple minor susceptibility genes. A recent sib-pair analysis revealed a tight linkage between markers on 5q31.1 and a major susceptibility gene controlling total serum IgE levels. Due to its location within this cluster and its biologic role in Ig class switching and Th2 cell differentiation, the IL-4 gene has emerged as one major candidate for the atopy gene. In one model, polymorphisms within IL-4 regulatory elements might result in overexpression of the gene, amplifying Th2 cell differentiation and class switching to IgE. In support of this model, we report that the human IL-4 promoter exists in multiple allelic forms that exhibit distinct transcriptional activities in IL-4-positive T cells. A particular allele has an unusually high transcriptional activity. A nucleotide substitution within a recently described OAP40 element located just upstream of an NF-AT site (P sequence) appears to be largely responsible for the increased promotor strength of this particular allelic form of the IL-4 promoter. In EMSAs, this substitution results in a markedly enhanced affinity for sequence-specific complexes exhibiting an AP-1 specificity. The identification of allelic nucleotides, which results in overexpression of the IL-4 gene, provides specific targets for a comprehensive screening of atopic and nonatopic individuals and may provide a clue for genetic predisposition for atopy.


Subject(s)
Gene Expression Regulation , Hypersensitivity, Immediate/genetics , Interleukin-4/genetics , Promoter Regions, Genetic/genetics , Alleles , B-Lymphocytes , Base Sequence , Cell Line, Transformed , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Humans , Hypersensitivity, Immediate/immunology , Immunoglobulin E/biosynthesis , Leukemia, Basophilic, Acute/pathology , Leukemia-Lymphoma, Adult T-Cell/pathology , Molecular Sequence Data , Mutagenesis, Site-Directed , Sequence Alignment , Sequence Homology, Nucleic Acid , Th2 Cells/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic , Tumor Cells, Cultured
20.
Proc Natl Acad Sci U S A ; 92(25): 11623-7, 1995 Dec 05.
Article in English | MEDLINE | ID: mdl-8524816

ABSTRACT

Activation of individual CD4+ T cells results in differential lymphokine expression: interleukin 2 (IL-2) is preferentially produced by T helper type 1 (TH1) cells, which are involved in cell-mediated immune responses, whereas IL-4 is synthesized by TH2 cells, which are essential for humoral immunity. The Ca(2+)-dependent factor NF-ATp plays a key role in the inducible transcription of both these lymphokine genes. However, while IL2 expression requires the contribution of Ca(2+)- and protein kinase C-dependent signals, we report that activation of human IL4 transcription through the Ca(2+)-dependent pathway is diminished by protein kinase C stimulation in Jurkat T cells. This phenomenon is due to mutually exclusive binding of NF-ATp and NF-kappa B to the P sequence, an element located 69 bp upstream of the IL4 transcription initiation site. Human IL4 promoter-mediated transcription is downregulated in Jurkat cells stimulated with the NF-kappa B-activating cytokine tumor necrosis factor alpha and suppressed in RelA-overexpressing cells. In contrast, protein kinase C stimulation or RelA overexpression does not affect the activity of a human IL4 promoter containing a mouse P sequence, which is a higher-affinity site for NF-ATp and a lower-affinity site for RelA. Thus, competition between two general transcriptional activators, RelA and NF-ATp, mediates the inhibitory effect of protein kinase C stimulation on IL4 expression and may contribute to differential gene expression in TH cells.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Interleukins/biosynthesis , NF-kappa B/metabolism , Nuclear Proteins , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Base Sequence , Cell Differentiation , Genes, Reporter , Humans , Interleukin-2/biosynthesis , Interleukin-2/genetics , Interleukin-4/biosynthesis , Interleukin-4/genetics , Interleukins/genetics , Mice , Molecular Sequence Data , NFATC Transcription Factors , Promoter Regions, Genetic , Protein Kinase C , Species Specificity , T-Lymphocyte Subsets , Th1 Cells , Th2 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...