1
|
Srivastava M, Kaplan MH. Transcription Factors in the Development and Pro-Allergic Function of Mast Cells. FRONTIERS IN ALLERGY 2021; 2:679121. [PMID: 35387064 PMCID: PMC8974754 DOI: 10.3389/falgy.2021.679121] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/12/2021] [Indexed: 11/13/2022] Open
Abstract
Mast cells (MCs) are innate immune cells of hematopoietic origin localized in the mucosal tissues of the body and are broadly implicated in the pathogenesis of allergic inflammation. Transcription factors have a pivotal role in the development and differentiation of mast cells in response to various microenvironmental signals encountered in the resident tissues. Understanding the regulation of mast cells by transcription factors is therefore vital for mechanistic insights into allergic diseases. In this review we summarize advances in defining the transcription factors that impact the development of mast cells throughout the body and in specific tissues, and factors that are involved in responding to the extracellular milieu. We will further describe the complex networks of transcription factors that impact mast cell physiology and expansion during allergic inflammation and functions from degranulation to cytokine secretion. As our understanding of the heterogeneity of mast cells becomes more detailed, the contribution of specific transcription factors in mast cell-dependent functions will potentially offer new pathways for therapeutic targeting.
Collapse
Affiliation(s)
- Mansi Srivastava
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University-Purdue University, Indianapolis, IN, United States
| | - Mark H. Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Mark H. Kaplan
| |
Collapse
|
2
|
Mukai K, Tsai M, Saito H, Galli SJ. Mast cells as sources of cytokines, chemokines, and growth factors. Immunol Rev 2019; 282:121-150. [PMID: 29431212 DOI: 10.1111/imr.12634] [Citation(s) in RCA: 499] [Impact Index Per Article: 83.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mast cells are hematopoietic cells that reside in virtually all vascularized tissues and that represent potential sources of a wide variety of biologically active secreted products, including diverse cytokines and growth factors. There is strong evidence for important non-redundant roles of mast cells in many types of innate or adaptive immune responses, including making important contributions to immediate and chronic IgE-associated allergic disorders and enhancing host resistance to certain venoms and parasites. However, mast cells have been proposed to influence many other biological processes, including responses to bacteria and virus, angiogenesis, wound healing, fibrosis, autoimmune and metabolic disorders, and cancer. The potential functions of mast cells in many of these settings is thought to reflect their ability to secrete, upon appropriate activation by a range of immune or non-immune stimuli, a broad spectrum of cytokines (including many chemokines) and growth factors, with potential autocrine, paracrine, local, and systemic effects. In this review, we summarize the evidence indicating which cytokines and growth factors can be produced by various populations of rodent and human mast cells in response to particular immune or non-immune stimuli, and comment on the proven or potential roles of such mast cell products in health and disease.
Collapse
Affiliation(s)
- Kaori Mukai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Hirohisa Saito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health & Development, Tokyo, Japan
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
3
|
Fitzpatrick CJ, Morrow JD. Thalamic mast cell activity is associated with sign-tracking behavior in rats. Brain Behav Immun 2017; 65:222-229. [PMID: 28487202 PMCID: PMC5537013 DOI: 10.1016/j.bbi.2017.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 12/20/2022] Open
Abstract
Mast cells are resident immune cells in the thalamus that can degranulate and release hundreds of signaling molecules (i.e., monoamines, growth factors, and cytokines) both basally and in response to environmental stimuli. Interestingly, mast cell numbers in the brain show immense individual variation in both rodents and humans. We used a Pavlovian conditioned approach (PCA) procedure to examine whether mast cells are associated with individual variation in the attribution of incentive-motivational value to reward-related cues. During the PCA procedure, a lever response-independently predicts the delivery of a food pellet into a magazine, and over training sessions three conditioned responses (CRs) develop: sign-tracking (lever-directed CRs), goal-tracking (magazine-directed CRs), and an intermediate response (both CRs). In Experiment 1, we measured thalamic mast cell number/activation using toluidine blue and demonstrated that sign-trackers have increased degranulated (activated) but not granulated (inactive) mast cells. In Experiment 2, we infused the mast cell inhibitor, cromolyn (200µg/rat; i.c.v.), immediately before five daily PCA training sessions and demonstrated that mast cell inhibition selectively impairs the acquisition of sign-tracking behavior. Taken together, these results demonstrate that thalamic mast cells contribute to the attribution of incentive-motivational value to reward-related cues and suggest that mast cell inhibition may be a novel target for addiction treatment.
Collapse
Affiliation(s)
| | - Jonathan D Morrow
- Neuroscience Graduate Program, University of Michigan, 204 Washtenaw Ave, Ann Arbor, MI 48109, USA; Department of Psychiatry, University of Michigan, 4250 Plymouth Rd, Ann Arbor, MI 48109, USA.
| |
Collapse
|
4
|
Kong SK, Soo Kim B, Gi Uhm T, Soo Chang H, Sook Park J, Woo Park S, Park CS, Chung IY. Aspirin induces IL-4 production: augmented IL-4 production in aspirin-exacerbated respiratory disease. Exp Mol Med 2016; 48:e202. [PMID: 27534531 PMCID: PMC4686698 DOI: 10.1038/emm.2015.96] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 09/11/2015] [Indexed: 01/04/2023] Open
Abstract
Aspirin hypersensitivity is a hallmark of aspirin-exacerbated respiratory disease (AERD), a clinical syndrome characterized by the severe inflammation of the respiratory tract after ingestion of cyclooxygenase-1 inhibitors. We investigated the capacity of aspirin to induce interleukin-4 (IL-4) production in inflammatory cells relevant to AERD pathogenesis and examined the associated biochemical and molecular pathways. We also compared IL-4 production in peripheral blood mononuclear cells (PBMCs) from patients with AERD vs aspirin-tolerant asthma (ATA) upon exposure to aspirin. Aspirin induced IL-4 expression and activated the IL-4 promoter in a report assay. The capacity of aspirin to induce IL-4 expression correlated with its activity to activate mitogen-activated protein kinases, to form DNA-protein complexes on P elements in the IL-4 promoter and to synthesize nuclear factor of activated T cells, critical transcription factors for IL-4 transcription. Of clinical importance, aspirin upregulated IL-4 production twice as much in PBMCs from patients with AERD compared with PBMCs from patients with ATA. Our results suggest that IL-4 is an inflammatory component mediating intolerance reactions to aspirin, and thus is crucial for AERD pathogenesis.
Collapse
Affiliation(s)
- Su-Kang Kong
- Department of Bionano Technology, Hanyang University, Ansan, Republic of Korea
| | - Byung Soo Kim
- Department of Bionano Technology, Hanyang University, Ansan, Republic of Korea
| | - Tae Gi Uhm
- Department of Molecular and Life Sciences, College of Science and Technology, Hanyang University, Ansan, Republic of Korea
| | - Hun Soo Chang
- Soonchunhyang Medical Science Research Institute, College of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Asan, Chungcheongnam-do, Republic of Korea
| | - Jong Sook Park
- Division of Allergy and Respiratory, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Gyeonggi-do, Republic of Korea
| | - Sung Woo Park
- Division of Allergy and Respiratory, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Gyeonggi-do, Republic of Korea
| | - Choon-Sik Park
- Division of Allergy and Respiratory, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Gyeonggi-do, Republic of Korea
| | - Il Yup Chung
- Department of Bionano Technology, Hanyang University, Ansan, Republic of Korea
- Department of Molecular and Life Sciences, College of Science and Technology, Hanyang University, Ansan, Republic of Korea
| |
Collapse
|
5
|
Regulation of IL-4 Expression in Immunity and Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 941:31-77. [PMID: 27734408 DOI: 10.1007/978-94-024-0921-5_3] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IL-4 was first identified as a T cell-derived growth factor for B cells. Studies over the past several decades have markedly expanded our understanding of its cellular sources and function. In addition to T cells, IL-4 is produced by innate lymphocytes, such as NTK cells, and myeloid cells, such as basophils and mast cells. It is a signature cytokine of type 2 immune response but also has a nonimmune function. Its expression is tightly regulated at several levels, including signaling pathways, transcription factors, epigenetic modifications, microRNA, and long noncoding RNA. This chapter will review in detail the molecular mechanism regulating the cell type-specific expression of IL-4 in physiological and pathological type 2 immune responses.
Collapse
|
6
|
McLeod JJA, Baker B, Ryan JJ. Mast cell production and response to IL-4 and IL-13. Cytokine 2015; 75:57-61. [PMID: 26088754 PMCID: PMC4532630 DOI: 10.1016/j.cyto.2015.05.019] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 11/24/2022]
Abstract
IL-4 was identified as the first cytokine to be produced by mast cells and is responsible for promoting mast cell IL-13 production. IL-4 and IL-13 play a prominent role in stimulating and maintaining the allergic response. As closely related genes, IL-4 and IL-13 share a common receptor subunit, IL-4Rα, necessary for signaling. Here we summarize the literature on mast cell activation associated with IL-4 and IL-13 production, including downstream signaling. We also describe the positive and negative roles each cytokine plays in mast cell immunity and detail the differences that exist between mouse and human mast cell responses to IL-4 and IL-13.
Collapse
Affiliation(s)
- Jamie J A McLeod
- Department of Biology, Virginia Commonwealth University Richmond, VA 23284, United States.
| | - Bianca Baker
- Department of Biology, Virginia Commonwealth University Richmond, VA 23284, United States
| | - John J Ryan
- Department of Biology, Virginia Commonwealth University Richmond, VA 23284, United States
| |
Collapse
|
7
|
Junkins RD, MacNeil AJ, Wu Z, McCormick C, Lin TJ. Regulator of Calcineurin 1 Suppresses Inflammation during Respiratory Tract Infections. THE JOURNAL OF IMMUNOLOGY 2013; 190:5178-86. [DOI: 10.4049/jimmunol.1203196] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
8
|
Vihma H, Pruunsild P, Timmusk T. Alternative splicing and expression of human and mouse NFAT genes. Genomics 2008; 92:279-91. [PMID: 18675896 PMCID: PMC2577130 DOI: 10.1016/j.ygeno.2008.06.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Revised: 06/20/2008] [Accepted: 06/24/2008] [Indexed: 12/30/2022]
Abstract
Four members of the nuclear factor of activated T cells (NFAT) family (NFATC1, NFATC2, NFATC3, and NFATC4) are Ca(2+)-regulated transcription factors that regulate several processes in vertebrates, including the development and function of the immune, cardiovascular, musculoskeletal, and nervous systems. Here we describe the structures and alternative splicing of the human and mouse NFAT genes, including novel splice variants for NFATC1, NFATC2, NFATC3, and NFATC4, and show the expression of different NFAT mRNAs in various mouse and human tissues and brain regions by RT-PCR. Our results show that alternatively spliced NFAT mRNAs are expressed differentially and could contribute to the diversity of functions of the NFAT proteins. Since NFAT family members are Ca(2+)-regulated and have critical roles in neuronal gene transcription in response to electrical activity, we describe the expression of NFATC1, NFATC2, NFATC3, and NFATC4 mRNAs in the adult mouse brain and in the adult human hippocampus using in situ hybridization and show that all NFAT mRNAs are expressed in the neurons of the mouse brain with specific patterns for each NFAT.
Collapse
Affiliation(s)
| | | | - Tõnis Timmusk
- Department of Gene Technology, Tallinn University of Technology, Akadeemia Tee 15, Tallinn 19086, Estonia
| |
Collapse
|
9
|
Frossi B, Rivera J, Hirsch E, Pucillo C. Selective Activation of Fyn/PI3K and p38 MAPK Regulates IL-4 Production in BMMC under Nontoxic Stress Condition. THE JOURNAL OF IMMUNOLOGY 2007; 178:2549-55. [PMID: 17277164 DOI: 10.4049/jimmunol.178.4.2549] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mast cells have the ability to react to multiple stimuli, implicating these cells in many immune responses. Specific signals from the microenvironment in which mast cells reside can activate different molecular events that govern distinct mast cells responses. We previously demonstrated that hydrogen peroxide (H(2)O(2)) promotes IL-4 and IL-6 mRNA production and potentates FcepsilonRI-induced cytokine release in rat basophilic leukemia RBL-2H3 cells. To further evaluate the effect of an oxidative microenvironment (which is physiologically present in an inflammatory site) on mast cell function and the molecular events responsible for mast cell cytokine production in this environment, we analyzed the effect of H(2)O(2) treatment on IL-4 production in bone marrow-derived, cultured mast cells. Our findings show that nanomolar concentrations of H(2)O(2) induce cytokine secretion and enhance IL-4 production upon FcepsilonRI triggering. Oxidative stimulation activates a distinct signal transduction pathway that induces Fyn/PI3K/Akt activation and the selective phosphorylation of p38 MAP kinase. Moreover, H(2)O(2) induces AP-1 and NFAT complexes that recognize the IL-4 promoter. The absence of Fyn and PI3K or the inhibition of p38 MAPK activity demonstrated that they are essential for H(2)O(2)-driven IL-4 production. These findings show that mast cells can respond to an oxidative microenvironment by initiating specific signals capable of eliciting a selective response. The findings also demonstrate the dominance of the Fyn/p38 MAPK pathway in driving IL-4 production.
Collapse
Affiliation(s)
- Barbara Frossi
- Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, Udine, Italy
| | | | | | | |
Collapse
|
10
|
Ko CB, Lee BS, Cha SH, Sul D, Paik SG, Kang HS. Distinct role of IL-3 promoter and enhancer region in murine mast cells. Mol Immunol 2006; 44:1569-76. [PMID: 17027084 DOI: 10.1016/j.molimm.2006.08.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2006] [Revised: 08/14/2006] [Accepted: 08/22/2006] [Indexed: 12/31/2022]
Abstract
Crosslinking of Fcvarepsilon receptor on mast cells induces IL-3 gene expression with the concentration dependent of intracellular calcium, but its regulatory mechanism remains unclear. Here, we found that phorbol 12-myristate 13-acetate (PMA) alone did not induce IL-3 gene expression, but potentiated A23187-induced IL-3 gene expression. Interestingly, the A23187-induced IL-3 promoter activity was suppressed by PMA, but it was enhanced when IL-3 promoter contained enhancer region, a DH site. While IL-3 mRNA expression was increased by A23187 and PMA in a dose-dependent manner, the promoter activity appeared all or none in all doses of A23187 and PMA. IL-3 promoter region between -293 and -150bp was responsible for A23187-induced gene expression and PMA- or cyclosporin A (CsA)-mediated suppression. Taken together, IL-3 gene expression was primarily regulated at the transcriptional level, which was differentially controlled by a restricted promoter and enhancer region.
Collapse
Affiliation(s)
- Chang-Bo Ko
- School of Biological Sciences and Technology, Hormone Research Institute, Chonnam National University, 300 Yongbong-dong, Buk-gu, Gwangju 500-757, Republic of Korea
| | | | | | | | | | | |
Collapse
|
11
|
Ryzhov S, Goldstein AE, Biaggioni I, Feoktistov I. Cross-talk between G(s)- and G(q)-coupled pathways in regulation of interleukin-4 by A(2B) adenosine receptors in human mast cells. Mol Pharmacol 2006; 70:727-35. [PMID: 16707627 DOI: 10.1124/mol.106.022780] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human mast cells express functional A(2A) and A(2B) adenosine receptors. However, only stimulation of A(2B), not A(2A), leads to secretion of interleukin (IL)-4, an important step in adenosine receptor-mediated induction of IgE synthesis by B-cells. In this study, we investigate intracellular pathways that link stimulation of A(2B) receptors to IL-4 up-regulation in HMC-1 mast cells. Both A(2A) and A(2B) receptors couple to G(s) proteins and stimulate adenylate cyclase, but only A(2B) stimulates phospholipase Cbeta through coupling to G(q) proteins leading to activation of protein kinase C and calcium mobilization. Inhibition of phospholipase Cbeta completely blocked A(2B) receptor-dependent IL-4 secretion. The protein kinase C inhibitor 2-{8-[(dimethylamino)-methyl]-6,7,8,9-tetrahydropyrido[1,2-a]indol-3-yl}-3-(1-methyl-1H-indol-3-yl)maleimide (Ro-32-0432) had no effect on A(2B) receptor-mediated IL-4 secretion but inhibited phorbol 12-myristate 13-acetate-stimulated IL-4 secretion. In contrast, chelation of intracellular Ca(2+) inhibited both A(2B) receptor- and ionomycin-dependent IL-4 secretion. This Ca(2+)-sensitive pathway probably includes calcineurin and nuclear factor of activated T cells, because A(2B) receptor-dependent IL-4 secretion was blocked with cyclosporin A or 11R-VIVIT peptide. G(s)-linked pathways also play a role in the A(2B) receptor-dependent stimulation of IL-4 secretion; inhibition of adenylate cyclase or protein kinase A attenuated A(2B) receptor-dependent IL-4 secretion. Although stimulation of adenylate cyclase with forskolin did not increase IL-4 secretion on its own, it potentiated the effect of Pasteurella multocida toxin by 2-fold and ionomycin by 3-fold. Both forskolin and stimulation of A(2B) receptors up-regulated NFATc1 protein levels. We conclude that A(2B) receptors up-regulate IL-4 through G(q) signaling that is potentiated via cross-talk with G(s)-coupled pathways.
Collapse
Affiliation(s)
- Sergey Ryzhov
- Divisions of Cardiovascular Medicine, Vanderbilt University, Nashville, TN 37232-6300, USA
| | | | | | | |
Collapse
|
12
|
Monticelli S, Lee DU, Nardone J, Bolton DL, Rao A. Chromatin-based regulation of cytokine transcription in Th2 cells and mast cells. Int Immunol 2005; 17:1513-24. [PMID: 16199489 DOI: 10.1093/intimm/dxh329] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Th2 cells and mast cells are major sources of IL4, IL5 and IL13, cytokines that mediate immunity against parasites and are also central players in the pathophysiology of asthma, allergy and atopic disease. We asked whether Th2 cells and mast cells, which belong to the lymphoid and myeloid lineages, respectively, use different cis-acting regulatory regions to transcribe the cytokine genes. Comparison of DNase I hypersensitivity patterns at the RAD50/IL4/IL13 locus revealed that most hypersensitive sites (HSs) are common to Th2 and mast cells, but two regions [conserved non-coding sequence (CNS) 1 and mast cell HSs] show cell type-specific differences. CNS-1, one of the most highly conserved CNS regions in the RAD50/IL13/IL4 locus, displays two strong DNase I HSs in Th2 cells but is not DNase I hypersensitive in mast cells, explaining a previous finding that deletion of CNS-1 impairs cytokine expression in Th2 cells but not in mast cells. Conversely, two constitutive HSs (mast cell HSs) in the first intron of the IL13 gene are present in mast cells but not in Th2 cells; these sites develop early during mast cell differentiation and may have a role in maintaining accessibility of the IL13 locus to high-level transcription in stimulated cells.
Collapse
Affiliation(s)
- Silvia Monticelli
- Department of Pathology, Harvard Medical School, and CBR Institute for Biomedical Research, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
13
|
Gessner A, Mohrs K, Mohrs M. Mast Cells, Basophils, and Eosinophils Acquire Constitutive IL-4 and IL-13 Transcripts during Lineage Differentiation That Are Sufficient for Rapid Cytokine Production. THE JOURNAL OF IMMUNOLOGY 2005; 174:1063-72. [PMID: 15634931 DOI: 10.4049/jimmunol.174.2.1063] [Citation(s) in RCA: 221] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mast cells, basophils, and eosinophils are myeloid cells that are distinguished by their capability to produce IL-4 and IL-13. However, it is not clear how this potential is related to the lineage differentiation of these subsets. In the present study we used bicistronic IL-4 reporter (4get) mice to directly visualize IL-4 expression by nonlymphoid cells in vitro and in vivo at the single-cell level. Our data show that frequent expression of both Il4 alleles is initiated and maintained during ontogeny by an IL-4Ralpha- or Stat6-independent mechanism. Despite the constitutive presence of cytokine transcripts in differentiated cells under steady state conditions, cytokine production is not detectable in the absence of stimulation. Moreover, mature mast cells, basophils, and eosinophils also constitutively express IL-13. Both preformed IL-4 and IL-13 mRNAs are sufficient for rapid cytokine production upon stimulation. Our data show that mast cells, basophils, and eosinophils are programmed for IL-4 and IL-13 expression early in ontogeny. These novel findings have important implications for the prevention and therapeutic intervention of allergic and asthmatic diseases.
Collapse
Affiliation(s)
- André Gessner
- Institute of Clinical Microbiology, Immunology, and Hygiene University of Erlangen-Nurnberg, Erlangen, Germany
| | | | | |
Collapse
|
14
|
Masuda A, Yoshikai Y, Kume H, Matsuguchi T. The interaction between GATA proteins and activator protein-1 promotes the transcription of IL-13 in mast cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:5564-73. [PMID: 15494506 DOI: 10.4049/jimmunol.173.9.5564] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-13 is considered to be a key modulator in the pathogenesis of Th2-induced allergic inflammation, although little is known about the regulation of IL-13 transcription in mast cells. In T cells, involvement of GATA-3 in cell type-specific expression of the IL-13 gene has been reported. However, the mechanisms that induce rapid transactivation of the IL-13 gene in response to various types of stimulation have hitherto remained unknown. In this report, we describe our investigation of the promoter region necessary for IL-13 transcription; we have found that both AP-1 and GATA proteins are indispensable for IL-13 transcription in mouse mast cells. In our investigation, we focused on the functional interaction between GATA and AP-1 in the IL-13 promoter context. Transfection experiments have revealed that GATA-1 and GATA-2 proteins are able to associate with AP-1 proteins. We have also shown that overexpression of GATA-1 induced excess AP-1 binding to the IL-13 promoter as well as a significant increase in IL-13 production in mast cells. The results of the present study have shown that direct interaction between AP-1 and GATA proteins plays an important role in IL-13 transcription in mast cells.
Collapse
Affiliation(s)
- Akio Masuda
- Division of Host Defense, Center for Neural Disease and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | | | | | | |
Collapse
|
15
|
Koranteng RD, Swindle EJ, Davis BJ, Dearman RJ, Kimber I, Flanagan BF, Coleman JW. Differential regulation of mast cell cytokines by both dexamethasone and the p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580. Clin Exp Immunol 2004; 137:81-7. [PMID: 15196247 PMCID: PMC1809098 DOI: 10.1111/j.1365-2249.2004.02510.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Activated mast cells generate multiple cytokines but it is not known if these can be differentially regulated by pharmacological agents. We report here that the glucocorticoid dexamethasone (DEX) preferentially inhibited Ag-induced expression of IL-4 and IL-6 mRNA relative to TNF-alpha mRNA in RBL-2H3 cells. Likewise, the drug more readily inhibited release of IL-4 than TNF-alpha protein. SB203580, an inhibitor of p38 mitogen-activated protein kinase (MAPK), enhanced Ag-induced TNF-alpha mRNA expression without affecting IL-4 or IL-6 mRNA. At the protein level, SB203580 exerted little effect on TNF-alpha release but inhibited IL-4 release; notably, the ratio of TNF-alpha : IL-4 increased markedly with the concentration of SB203580, confirming the differential regulation of these cytokines. PD98059, an inhibitor of MAPK kinase (MEK), a component of the p44/42 MAPK pathway, partially inhibited Ag-induced expression of mRNA for all three cytokines while cyclosporin A inhibited Ag-induced IL-4 and IL-6 mRNA more readily than TNF-alpha mRNA. Ag activation of the cells led to phosphorylation of p38 and p44/42 MAPK but this was not influenced by DEX. In conclusion, mast cell cytokines can be differentially regulated pre- and post-translationally by DEX and SB203580 but there does not appear to be a direct mechanistic link between the actions of these two drugs.
Collapse
Affiliation(s)
- R D Koranteng
- Department of Pharmacology, University of Liverpool, Liverpool, UK
| | | | | | | | | | | | | |
Collapse
|
16
|
Kiani A, Habermann I, Haase M, Feldmann S, Boxberger S, Sanchez-Fernandez MA, Thiede C, Bornhäuser M, Ehninger G. Expression and regulation of NFAT (nuclear factors of activated T cells) in human CD34+cells: down-regulation upon myeloid differentiation. J Leukoc Biol 2004; 76:1057-65. [PMID: 15292278 DOI: 10.1189/jlb.0404259] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The calcineurin-dependent, cyclosporin A (CsA)-sensitive transcription factor nuclear factor of activated T cells (NFAT) represents a group of proteins, which is well-characterized as a central regulatory element of cytokine expression in activated T cells. In contrast, little is known about the expression or function of NFAT family members in myeloid cells; moreover, it is unclear whether they are expressed by hematopoietic stem/progenitor cells. Here, we show that NFATc2 (NFAT1) is expressed at high levels in CD34+ cells and megakaryocytes but not in cells committed to the neutrophilic, monocytic, or erythroid lineages. Cytokine-induced in vitro differentiation of CD34+ cells into neutrophil granulocytes results in the rapid suppression of NFATc2 RNA and protein. NFATc2 dephosphorylation/rephosphorylation as well as nuclear/cytoplasmic translocation in CD34+ cells follow the same calcineurin-dependent pattern as in T lymphocytes, suggesting that NFATc2 activation in these cells is equally sensitive to inhibition with CsA. Finally, in vitro proliferation, but not differentiation, of CD34+ cells cultured in the presence of fms-like tyrosine kinase 3 ligand (FLT3L), stem cell factor, granulocyte macrophage-colony stimulating factor (GM-CSF), interleukin-3, and G-CSF is profoundly inhibited by treatment with CsA in a dose-dependent manner. These results suggest a novel and unexpected role for members of the NFAT transcription factor family in the hematopoietic system.
Collapse
Affiliation(s)
- Alexander Kiani
- Department of Medicine I, University Hospital Carl Gustav Carus, University of Dresden Technical Center, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Lee MH, Kim E, Kim TS. Exposure to 4-tert-octylphenol, an environmentally persistent alkylphenol, enhances interleukin-4 production in T cells via NF-AT activation. Toxicol Appl Pharmacol 2004; 197:19-28. [PMID: 15126071 DOI: 10.1016/j.taap.2004.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2003] [Accepted: 02/10/2004] [Indexed: 11/16/2022]
Abstract
4-tert-Octylphenol (OP) is a representative endocrine disruptor that may have adverse effects on human health. The influence of this compound on allergic immune responses remains unclear. In this study, we have examined the effects of OP on production of interleukin-4 (IL-4), a pro-inflammatory cytokine closely associated with allergic immune responses. OP significantly enhanced IL-4 production in antigen-primed T cells in a dose-dependent manner. Treatment with OP in vivo resulted in significant increase of IL-4 production in T cells and of IgE levels in sera of antigen-primed mice. Furthermore, OP enhanced the activation of IL-4 gene promoter in EL4 T cells transiently transfected with IL-4 promoter/reporter constructs, and the enhancing effect mapped to a region in the IL-4 promoter containing binding sites for nuclear factor of activated T cell (NF-AT). Activation of T cells by phorbol-12-myristate-13-acetate (PMA) resulted in markedly enhanced binding activities to the NF-AT site, which significantly increased upon addition of OP, indicating that the transcription factor NF-AT was involved in the enhancing effect of OP on IL-4 production. The enhancement of IL-4 production by OP was blocked by FK506, a calcineurin inhibitor, but not by the estrogen receptor (ER) antagonist ICI 182780. FK506 inhibited the NF-AT-DNA binding activity and IL-4 gene promoter activity enhanced by OP in a dose-dependent manner. These findings demonstrate that OP enhances IL-4 production in T cells via the stimulation of calcineurin-dependent NF-AT activation.
Collapse
Affiliation(s)
- Mi H Lee
- Immunology Laboratory, College of Pharmacy, Chonnam National University, Kwangju 500-757, South Korea
| | | | | |
Collapse
|
18
|
Lee MH, Chung SW, Kang BY, Park J, Lee CH, Hwang SY, Kim TS. Enhanced interleukin-4 production in CD4+ T cells and elevated immunoglobulin E levels in antigen-primed mice by bisphenol A and nonylphenol, endocrine disruptors: involvement of nuclear factor-AT and Ca2+. Immunology 2003; 109:76-86. [PMID: 12709020 PMCID: PMC1782943 DOI: 10.1046/j.1365-2567.2003.01631.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bisphenol A (BPA) and p-nonylphenol (NP) are representative endocrine disruptors (EDs) that may have adverse effects on human health. The influence of these compounds on allergic immune responses remains unclear. In this study, we have examined the effects of BPA and NP on production of interleukin-4 (IL-4), a pro-inflammatory cytokine closely associated with allergic immune responses. Both BPA and NP significantly enhanced IL-4 production in keyhole limpet haemocyanin (KLH)-primed CD4+ T cells in a concentration-dependent manner. Treatment with BPA or NP in vivo resulted in significant increase of IL-4 production in CD4+ T cells and of antigen-specific immunoglobulin E (IgE) levels in the sera of KLH-primed mice. Furthermore, BPA and NP enhanced the activation of IL-4 gene promoter in EL4 T cells transiently transfected with IL-4 promoter/reporter constructs, and the enhancing effect mapped to a region in the IL-4 promoter containing binding sites for nuclear factor (NF)-AT. Activation of T lymphocytes by phorbol 12-myristate 13-acetate/ionomycin resulted in markedly enhanced binding activities to the NF-AT site, which significantly increased upon addition of BPA or NP, as demonstrated by the electrophoretic mobility shift assay, indicating that the transcription factor NF-AT was involved in the enhancing effect of BPA and NP on IL-4 production. The enhancement of IL-4 production by BPA or NP was significantly reduced by nitrendipine, a blocker of Ca2+ influx, and by FK506, a calcineurin inhibitor. FK506 inhibited the NF-AT-DNA binding activity and IL-4 gene promoter activity enhanced by BPA or NP. These results represent the first report describing possible enhancement of allergic response by EDs through increasing IL-4 production in CD4+ T cells and antigen-specific IgE levels in the sera via the stimulation of Ca2+/calcineurin-dependent NF-AT activation.
Collapse
Affiliation(s)
- Mee H Lee
- Immunology Laboratory, College of Pharmacy, Chonnam National University, Kwangju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
19
|
Kojima H, Tamura T, Okuda T, Kato C, Kinoshita Y, Honjo H. Expression of nuclear factor of activated T cells mRNA in maternal peripheral blood cells. Am J Reprod Immunol 2003; 49:139-48. [PMID: 12797520 DOI: 10.1034/j.1600-0897.2003.01165.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PROBLEM In T lymphocytes, nuclear factor of activated T cells (NF-AT) regulates the induction of cytokine genes upon antigenic stimulation. This study was designed to analyse the relationship between NF-AT and pregnancy. METHOD OF STUDY With informed consent, peripheral blood cells (PBCs) were obtained from non-pregnant (n = 114), pregnant (n = 604), and puerperal women (n = 52). The expression of NF-AT2 and NF-AT3 mRNAs in PBCs was measured by a quantitative reverse transcriptase-polymerase chain reaction method. RESULTS In the early pregnancy period, in successful pregnancy, both NF-AT2 and NF-AT3 mRNAs increased significantly, whereas in cases of spontaneous abortion they did not change significantly. After peaking, they decreased gradually and were re-elevated in the ninth and tenth gestational month. In the puerperal period, NF-AT3 mRNAs decreased, but NF-AT2 mRNA showed a comparatively high expression level. CONCLUSION These findings suggested that in humans NF-AT signal transduction might be involved in alloantigenic recognition and tolerance and play important roles, especially in the early and late period of pregnancy.
Collapse
Affiliation(s)
- H Kojima
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Kawaramachi Hirokouji, Kamigyo-Ku, Japan
| | | | | | | | | | | |
Collapse
|
20
|
Masuda A, Yoshikai Y, Aiba K, Matsuguchi T. Th2 cytokine production from mast cells is directly induced by lipopolysaccharide and distinctly regulated by c-Jun N-terminal kinase and p38 pathways. THE JOURNAL OF IMMUNOLOGY 2002; 169:3801-10. [PMID: 12244175 DOI: 10.4049/jimmunol.169.7.3801] [Citation(s) in RCA: 131] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Mast cells secrete multiple cytokines and play an important role in allergic inflammation. Although it is widely accepted that bacteria infection occasionally worsens allergic airway inflammation, the mechanism has not been defined. In this study, we show that LPS induced Th2-associated cytokine production such as IL-5, IL-10, and IL-13 from mast cells and also synergistically enhanced production of these cytokines induced by IgE cross-linking. LPS-mediated Th2-type cytokine production was abolished in mouse bone marrow-derived mast cells derived from C3H/HeJ mice, suggesting that Toll-like receptor 4 is essential for the cytokine production. Furthermore, we found that mitogen-activated protein kinases including extracellular signal-regulated kinase 1/2, c-Jun N-terminal kinase, and p38 kinase were activated by LPS stimulation in bone marrow-derived mast cells. Inhibition of extracellular signal-regulated kinase activation has little effect on LPS-mediated cytokine production. In contrast, inhibition of c-Jun N-terminal kinase activation significantly suppressed both IL-10 and IL-13 expression at both mRNA and protein levels. Interestingly, although inhibition of p38 did not down-regulate the mRNA induction, it moderately decreased all three cytokine productions by LPS. These results indicate that LPS-mediated production of IL-5, IL-10, and IL-13 was distinctly regulated by mitogen-activated protein kinases. Our findings may indicate a clue to understanding the mechanisms of how bacteria infection worsens the clinical features of asthma.
Collapse
Affiliation(s)
- Akio Masuda
- Laboratory of Host Defense and Germfree Life, Research Institute for Disease Mechanism and Control, Nagoya University School of Medicine, Nagoya, Japan.
| | | | | | | |
Collapse
|
21
|
Sherman MA, Powell DR, Brown MA. IL-4 induces the proteolytic processing of mast cell STAT6. THE JOURNAL OF IMMUNOLOGY 2002; 169:3811-8. [PMID: 12244176 DOI: 10.4049/jimmunol.169.7.3811] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
IL-4 is a potent, pleiotropic cytokine that, in general, directs cellular activation, differentiation, and rescue from apoptosis. However, in mast cells, IL-4 induces the down-regulation of activation receptors and promotes cell death. Mast cells have been shown to transduce IL-4 signals through a unique C-terminally truncated isoform of STAT6. In this study, we examine the mechanism through which STAT6 is processed to generate this isoform. We demonstrate that STAT6 processing in mast cells is initiated by IL-4-induced phosphorylation and nuclear translocation of full-length STAT6 and subsequent cleavage by a nuclear serine-family protease. The location of the protease in the nucleus ensures that the truncated STAT6 has preferential access to bind DNA. IL-4-responsive target genes in mast cells are identified by chromatin immunoprecipitation of STAT6, including the IL-4 gene itself. These results suggest a molecular explanation for the suppressive effects of IL-4 on STAT6-regulated genes in mast cells.
Collapse
Affiliation(s)
- Melanie A Sherman
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
22
|
Zabel MD, Wheeler W, Weis JJ, Weis JH. Yin Yang 1, Oct1, and NFAT-4 form repeating, cyclosporin-sensitive regulatory modules within the murine CD21 intronic control region. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3341-50. [PMID: 11907091 DOI: 10.4049/jimmunol.168.7.3341] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The murine complement receptor type 2 gene (Cr2/CD21) is expressed by murine B and follicular dendritic cells, but not murine T cells. We have previously shown that appropriate transcriptional control of the CD21 gene requires the CD21 promoter as well as intronic sequences. We have also demonstrated that altering chromatin structure by inhibiting histone deacetylases induces CD21 expression in murine T cells by increasing the accessibility of promoter and intronic regulatory elements. In this report, we identify seven distinct regulatory areas within the first intron of the murine CD21 gene that are conserved between mouse and human CD21 intronic sequences. EMSA competition and supershift analyses reveal the formation of multiple DNA-protein complexes at these sites that include Yin Yang 1, Oct1, and NFAT-4. NFAT-containing complexes were altered in B cells treated with the NFAT inhibitor cyclosporin A and correlated with a repression of CD21 gene transcription implicating NFAT transcriptional control. Functional data revealed that no single region conferred cell-specific reporter gene expression, but rather the entire CD21 regulatory element was required to confer cell-specific gene expression. Taken together, these data demonstrate the formation of repeating, overlapping regulatory modules, all of which are required to coordinately control the cell-specific expression of the murine CD21 gene. We propose a model in which Yin Yang 1 and Oct1 may recruit histone deacetylase to multiple sites in the CD21 intronic regulatory element in nonexpressing cells and NFAT either displaces this histone deacetylase or recruits a histone acetylase to allow the formation of a functional transcriptional complex in expressing cells.
Collapse
Affiliation(s)
- Mark D Zabel
- Department of Pathology, Division of Cell Biology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | | | | | |
Collapse
|
23
|
Schroeder JT, Miura K, Kim HH, Sin A, Cianferoni A, Casolaro V. Selective expression of nuclear factor of activated T cells 2/c1 in human basophils: evidence for involvement in IgE-mediated IL-4 generation. J Allergy Clin Immunol 2002; 109:507-13. [PMID: 11897999 DOI: 10.1067/mai.2002.122460] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND The nuclear factor of activated T cells (NFAT) family of transcription factors plays a key role in rapidly inducing IL4 gene expression in effector T cells. OBJECTIVE Because human basophils secrete high levels of IL-4, we have examined whether specific NFAT species are expressed in these cells and whether Fc(epsilon)RI-mediated activation affects their subcellular localization and transcriptional function. METHODS Intracellular NFAT protein was identified by using 2-color flow cytometry; gene expression was done with RT-PCR. Subcellular localization of NFAT was assessed by means of Western blotting. Electrophoretic mobility shift assays assessed NFAT involvement in IL-4 transcription. RESULTS Basophils constitutively expressed high levels of NFAT2. In contrast, NFAT1 (NFATp), which is found in most leukocytes, was not seen in basophils. Low-level staining for NFAT4 was detected but was variably expressed among donor cells. Likewise, NFAT2 mRNA was constitutively expressed in basophils, and message for NFAT4 was seen in 3 of 5 preparations, whereas that for NFAT1 was found in only 1 of 5 preparations. NFAT2 protein accumulated in the nuclei of basophils activated for 1 hour with anti-IgE, and this was inhibited with the addition of FK506. A protein-DNA complex was formed with nuclear lysates from basophils and an IL-4 promoter NFAT consensus probe, with greater binding intensities detected in lysates of activated cells. An antibody to NFAT2 reduced the formation of the complex, whereas no effects were seen with antibodies to NFAT1, NFAT4, or unrelated transcription factors. CONCLUSIONS The selective and specific expression of NFAT2 in basophils is unique among leukocytes. This transcription factor also appears to play a critical role in the Fc(epsilon)RI-mediated production of IL-4 in these cells.
Collapse
Affiliation(s)
- John T Schroeder
- Johns Hopkins Asthma and Allergy Center, Division of Clinical Immunology, Baltimore, MD 21224, USA
| | | | | | | | | | | |
Collapse
|
24
|
Bochkov VN, Mechtcheriakova D, Lucerna M, Huber J, Malli R, Graier WF, Hofer E, Binder BR, Leitinger N. Oxidized phospholipids stimulate tissue factor expression in human endothelial cells via activation of ERK/EGR-1 and Ca(++)/NFAT. Blood 2002; 99:199-206. [PMID: 11756172 DOI: 10.1182/blood.v99.1.199] [Citation(s) in RCA: 166] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Activation of endothelial cells by lipid oxidation products is a key event in the initiation and progression of the atherosclerotic lesion. Minimally modified low-density lipoprotein (MM-LDL) induces the expression of certain inflammatory molecules such as tissue factor (TF) in endothelial cells. This study examined intracellular signaling pathways leading to TF up-regulation by oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (OxPAPC), a biologically active component of MM-LDL. OxPAPC induced TF activity and protein expression in human umbilical vein endothelial cells (HUVECs). However, OxPAPC neither induced phosphorylation or degradation of I kappa B alpha nor DNA binding of nuclear factor-kappa B (NF-kappa B). Furthermore, OxPAPC-induced TF expression was not inhibited by overexpression of I kappa B alpha. These results strongly indicate that OxPAPC-induced TF expression is independent of the classical NF-kappa B pathway. However, OxPAPC stimulated phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 and expression of early growth response factor 1 (EGR-1). Inhibitors of mitogen-activated kinase/ERK (MEK) or protein kinase C (PKC) blocked elevation of both EGR-1 and TF. Furthermore, overexpression of NAB2, a corepressor of EGR-1, inhibited effects of OxPAPC. In addition, OxPAPC induced rapid and reversible elevation of free cytosolic Ca(++) levels and nuclear factor of activated T cells (NFAT)/DNA binding. Induction of TF expression by OxPAPC was partially inhibited by cyclosporin A, known to block calcineurin, a Ca(++)-dependent phosphatase upstream of NFAT. Treatment of OxPAPC with phospholipase A(2) destroyed its biologic activity and 1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphorylcholine was identified as one biologically active component of OxPAPC that induces TF expression. Together, the results demonstrate that OxPAPC induces TF expression in HUVECs through activation of PKC/ERK/EGR-1 and Ca(++)/calcineurin/NFAT pathways rather than by NF-kappa B-mediated transcription. Thus, oxidized phospholipids may contribute to inflammation by activating pathways alternative to the classical NF-kappa B pathway.
Collapse
Affiliation(s)
- Valery N Bochkov
- Department of Vascular Biology and Thrombosis Research, University of Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kehoe KE, Brown MA, Imani F. Double-stranded RNA regulates IL-4 expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:2496-501. [PMID: 11509588 DOI: 10.4049/jimmunol.167.5.2496] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
dsRNA, as genomic fragment, replicative intermediate, or stem and loop structure in cells infected by viruses, can act to signal the immune system of the presence of viral infections. Although most viral infections are associated with strong Th1 immune responses, Th2-type responses have also been observed. In this study, we characterize the effects of dsRNA on the induction of Th2 responses in human lymphocytes. We report that in addition to the well-known Th1-inducing capabilities of dsRNA, treatment of human lymphocytes with low concentrations of dsRNA (0.1-1 microg/ml) leads to the expression of the prototypic Th2 cytokine IL-4. This induction was accompanied with the concentration-dependent activation of NF-kappaB and NF-AT2 but not NF-AT1. In addition, dsRNA can directly activate an IL-4 promoter-driven chloramphenicol acetyltransferase reporter gene in transiently transfected Jurkat cells. These results are the first demonstration of a non-TCR-associated activator of NF-AT in human cells and suggest that dsRNA directly influences IL-4 gene expression through its effect on NF-AT activation. Our data provide support for the idea that dsRNA at low concentrations in vivo may induce a Th2-dominant response that is not optimal for protective immunity to the virus.
Collapse
Affiliation(s)
- K E Kehoe
- Division of Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Asthma and Allergy Center, Baltimore, MD 21224, USA
| | | | | |
Collapse
|
26
|
Biswas RS, Cha HJ, Hardwick JM, Srivastava RK. Inhibition of drug-induced Fas ligand transcription and apoptosis by Bcl-XL. Mol Cell Biochem 2001; 225:7-20. [PMID: 11716366 DOI: 10.1023/a:1012203110027] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Fas/Fas ligand system triggers apoptosis in many cell types. Bcl-XL overexpresion antagonizes Fas/Fas ligand-mediated cell death. The mechanism by which Bcl-XL influences Fas-mediated cell death is unclear. We have found that microtubule-damaging drugs (e.g. Paclitaxel) induce apoptosis in a Fas/FasL-dependent manner. Inhibition of Fas/FasL pathway by anti-FasL antibody, mutant Fas or a dominant negative FADD blocks paclitaxel-induced apoptosis. Paclitaxel induced apoptosis through activation of both caspase-8 and caspase-3. Overexpression of Bcl-XL leads to inhibition of paclitaxel-induced FasL expression and apoptosis. Bcl-XL prevents the nuclear translocation of NFAT (nuclear factor of activated T lymphocytes) by inhibiting the activation of calcineurin, a calcium-dependent phosphatase that must dephosphorylate NFAT for it to move to the nucleus. The loop domain in Bcl-XL can suppress the anti-apoptotic function of Bcl-XL and may be a target for regulatory post-translational modifications. Upon phosphorylation, Bcl-XL loses its ability to bind with calcineurin. Without NFAT nuclear translocation, the FasL gene is not transcribed. Thus, paclitaxel and other drugs that disturb microtubule function kill cells, at least in part, through the induction of FasL, and Bcl-XL-mediated resistance to these agents is related to failure to induce FasL expression.
Collapse
Affiliation(s)
- R S Biswas
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore 21201, USA
| | | | | | | |
Collapse
|
27
|
Huang C, Ding M, Li J, Leonard SS, Rojanasakul Y, Castranova V, Vallyathan V, Ju G, Shi X. Vanadium-induced nuclear factor of activated T cells activation through hydrogen peroxide. J Biol Chem 2001; 276:22397-403. [PMID: 11292823 DOI: 10.1074/jbc.m010828200] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the role of reactive oxygen species (ROS) in activation of nuclear factor of activated T cells (NFAT), a pivotal transcription factor responsible for regulation of cytokines, by vanadium in mouse embryo fibroblast PW cells or mouse epidermal Cl 41 cells. Exposure of cells to vanadium led to the transactivation of NFAT in a time- and dose-dependent manner. Scavenging of vanadium-induced H(2)O(2) with N-acety-L-cyteine (a general antioxidant) or catalase (a specific H(2)O(2) inhibitor) or the chelation of vanadate with deferoxamine, resulted in inhibition of NFAT activation. In contrast, an increase in H(2)O(2) generation by the addition of superoxide dismutase or NADPH enhanced vanadium-induced NFAT activation. This vanadate-mediated H(2)O(2) generation was verified by both electron spin resonance and fluorescence staining assay. These results demonstrate that H(2)O(2) plays an important role in vanadium-induced NFAT transactivation in two different cell types. Furthermore, pretreatment of cells with nifedipine, a calcium channel blocker, inhibited vanadium-induced NFAT activation, whereas and ionomycin, two calcium ionophores, had synergistic effects with vanadium for NFAT induction. Incubation of cells with cyclosporin A (CsA), a pharmacological inhibitor of the phosphatase calcineurin, blocked vanadium-induced NFAT activation. All data show that vanadium induces NFAT activation not only through a calcium-dependent and CsA-sensitive pathway but also involved H(2)O(2) generation, suggesting that H(2)O(2) may be involved in activation of calcium-calcineurin pathways for NFAT activation caused by vanadium exposure.
Collapse
Affiliation(s)
- C Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Cameron L, Hamid Q. Regulation of allergic airways inflammation by cytokines and glucocorticoids. Curr Allergy Asthma Rep 2001; 1:153-63. [PMID: 11899298 DOI: 10.1007/s11882-001-0083-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cytokines mediate the allergic inflammatory response of the airways, and glucocorticosteroids ameliorate allergy symptoms by regulating cytokine expression. Recent studies provide insight into the manner by which cytokines work together to mediate allergic airway disease. Real progress has also been gained in our understanding of subcellular mechanisms of allergic inflammation, particularly the role of transcription factors in regulating the expression of specific cytokine profiles and the differentiation of the TH2 subset. This article provides an update of recently reported findings in this field and highlights emerging concepts of allergic inflammation.
Collapse
Affiliation(s)
- L Cameron
- Respiratory Sciences Center, University of Arizona, Tucson, AZ, USA
| | | |
Collapse
|
29
|
Abstract
Until recently, mast cells have been viewed primarily as harmful because of their key role as effector cells of allergic and potentially lethal anaphylactic reactions. Their contribution to human health appeared instead to be limited to the elimination of parasites. There is, however, growing evidence for additional beneficial functions of mast cells, particularly regarding the initiation of acquired immune reactions. Thus, mast cells can phagocytize diverse particles, take up antigens, and express a number of receptors, particularly MHC class I and II antigens, ICAM-1 and -3, CD43, CD80, CD86 and CD40L which allow them to interact with T and B lymphocytes. They can also secrete numerous cytokines that induce and enhance recruitment and functions of lymphocytes. Finally, there is good evidence that mast cells present e.g. pollen and bacterial antigens, respond to bacterial superantigens, but fail to react to endogenously produced antigens or superantigens. Mast cells can also activate B cells directly to produce IgE, but this activity and the ability to produce IL-4 or IL-13 is restricted primarily to basophil leukocytes and mucosal mast cells. Finally, recent evidence attributes a pivotal role to the cells in natural immunity to bacteria. There is also emerging evidence that mast cells can downmodulate the immune response. While these data require further clarification, the basic ability of mast cells to initiate innate and acquired immune reactions can no longer be questioned.
Collapse
Affiliation(s)
- B M Henz
- Department of Dermatology, Humboldt University, Berlin, Germany.
| | | | | | | | | |
Collapse
|
30
|
Huang C, Mattjus P, Ma WY, Rincon M, Chen NY, Brown RE, Dong Z. Involvement of nuclear factor of activated T cells activation in UV response. Evidence from cell culture and transgenic mice. J Biol Chem 2000; 275:9143-9. [PMID: 10734048 PMCID: PMC2652749 DOI: 10.1074/jbc.275.13.9143] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Mammalian cells respond to UV radiation by signaling cascades leading to activation of transcription factors, such as activated protein 1, NFkappaB, and p53, a process known as the "UV response." Nuclear factor of activated T cells (NFAT) was first identified as an inducible nuclear factor in immune response and subsequently found to be expressed in other tissues and cells. To date, however, the regulation and function of NFAT in tissues and cells, other than the immune system, are not well understood. In this study, we demonstrate that UV radiation activates NFAT-dependent transcription through a calcium-dependent mechanism in mouse epidermal JB6 cell lines, as well as in the skin of NFAT-luciferase reporter transgenic mice. Exposure of JB6 cells to UV radiation leads to the transactivation of NFAT in a dose-dependent manner. A23187 had a synergistic effect with UV for NFAT induction, whereas pretreatment of cells with nifedipine, a calcium channel blocker, dramatically impaired the NFAT activity induced by either UV or UV plus A23187. Calcium-dependent activation of NFAT by UV was further confirmed by an in vivo study using NFAT-luciferase reporter transgenic mice. These results demonstrated that UV radiation is a strong activator for skin NFAT transactivation through calcium-dependent pathways, suggesting that NFAT activation may be a part of the UV response.
Collapse
Affiliation(s)
- Chuanshu Huang
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Peter Mattjus
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Wei-ya Ma
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Mercedes Rincon
- Program in Immunobiology, Department of Medicine, University of Vermont, Burlington, Vermont 05405
| | - Nan-yue Chen
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | | | - Zigang Dong
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| |
Collapse
|
31
|
Chen R, Burke TF, Cumberland JE, Brummet M, Beck LA, Casolaro V, Georas SN. Glucocorticoids inhibit calcium- and calcineurin-dependent activation of the human IL-4 promoter. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:825-32. [PMID: 10623828 DOI: 10.4049/jimmunol.164.2.825] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
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.
Collapse
Affiliation(s)
- R Chen
- Division of Pulmonary Medicine, Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21224, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Hirasawa N, Sato Y, Fujita Y, Ohuchi K. Involvement of a phosphatidylinositol 3-kinase-p38 mitogen activated protein kinase pathway in antigen-induced IL-4 production in mast cells. BIOCHIMICA ET BIOPHYSICA ACTA 2000; 1456:45-55. [PMID: 10611455 DOI: 10.1016/s0005-2728(99)00104-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We studied the involvement of phosphatidylinositol 3-kinase (PI3-kinase) in the antigen-induced IL-4 production in a rat mast cell line, RBL-2H3. The stimulation of IgE-sensitized RBL-2H3 cells by the antigen resulted in increased IL-4 mRNA levels followed by increased IL-4 production. Wortmannin and LY294002, PI3-kinase inhibitors, partially reduced both the antigen-induced increases in the IL-4 mRNA levels and IL-4 production in a concentration-dependent manner. Extracellular signal-regulated kinase, p38 mitogen-activated protein kinase (p38 MAPK) and c-Jun N-terminal kinase (JNK), which belong to the MAPK family, were activated by the antigen stimulation, and the activation of p38 MAPK in addition to JNK was suppressed markedly by wortmannin. The phosphorylation of endogenous activating transcription factor-2, a substrate of p38 MAPK, was also inhibited by wortmannin. The specific p38 MAPK inhibitor SB203580 partially inhibited the antigen-induced IL-4 production at mRNA levels, but the MEK-1 inhibitor PD98059 enhanced it. These findings suggest that the activation of PI3-kinase and p38 MAPK is partially responsible for the antigen-induced IL-4 production in RBL-2H3 cells.
Collapse
Affiliation(s)
- N Hirasawa
- Department of Pathophysiological Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan.
| | | | | | | |
Collapse
|
33
|
Reischl IG, Coward WR, Church MK. Molecular consequences of human mast cell activation following immunoglobulin E-high-affinity immunoglobulin E receptor (IgE-FcepsilonRI) interaction. Biochem Pharmacol 1999; 58:1841-50. [PMID: 10591138 DOI: 10.1016/s0006-2952(99)00226-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The cross-linking by immunoglobulin E of its high-affinity receptor, FcepsilonRI, on mast cells initiates a complex series of biochemical events leading to degranulation and the synthesis and secretion of eicosanoids and cytokines through the action of transcription factors, such as nuclear factor-kappaB. The initial activation involves the phosphorylation of FcepsilonRI beta- and gamma-subunits through the actions of the tyrosine kinases lyn and syk. For the purposes of description, the subsequent events may be grouped in three cascades characterized by the key proteins involved. First, the phospholipase C-inositol phosphate cascade activates protein kinase C and is largely responsible for calcium mobilization and influx. Second, activation of Ras and Raf via mitogen-activated protein kinase causes the production of arachidonic acid metabolites. Third, the generation of sphingosine and sphingosine-1-phosphate occurs through activation of sphingomyelinase. While the early signaling events tend to be specific for the cited cascades, there is an increasing overlap of activated proteins with the downstream propagation of the signal. It is the balanced interaction between these proteins that culminates in degranulation, synthesis, and release of eicosanoids and cytokines.
Collapse
|
34
|
Yoshida T, Ishikawa I, Ono Y, Imai T, Suzuki R, Yoshie O. An Activation-Responsive Element in Single C Motif-1/Lymphotactin Promoter Is a Site of Constitutive and Inducible DNA-Protein Interactions Involving Nuclear Factor of Activated T Cell. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.6.3295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Single C motif-1 (SCM-1)/lymphotactin is a C-type chemokine whose expression is activation dependent, cyclosporin A sensitive and restricted to CD8+ T cells, double-negative thymocytes, γδ-type T cells, and NK cells. In humans, there are two highly homologous genes encoding SCM-1α and SCM-1β. Here we examined the regulatory mechanism of the SCM-1 genes. The luciferase reporter gene under the control of the 5′ flanking region of 0.7 kb was strongly induced upon activation with anti-CD3 or PHA plus PMA only in SCM-1-producer T cell lines through a cyclosporin A-sensitive mechanism. An element termed E1 located at −108 to −95 nt relative to the major transcription start site was found to be critical for the promoter activity. In electrophoretic mobility shift assays using the E1 oligonucleotide as probe, nuclear extracts from unstimulated T and B cell lines formed a constitutive complex termed complex I, while nuclear extracts from stimulated SCM-1-producer T cell lines formed a higher mobility complex termed complex II with a concomitant decrease in complex I. The shift from complex I to complex II seen only in SCM-1-producer T cell lines upon activation was completely suppressed by cyclosporin A. Both complexes were critically dependent on the NF-AT core sequence TTTCC in the E1 element and were partially supershifted by anti-NF-ATp. One-hybrid assays in yeast isolated NF-ATp as an E1 binding protein, and transfection of NF-ATp into T and B cell lines strongly enhanced the activation-dependent SCM-1 promoter activity. Collectively, a unique mechanism involving NF-ATp appears to regulate the cell type-specific and activation-dependent expression of the SCM-1 genes.
Collapse
Affiliation(s)
- Tetsuya Yoshida
- *Shionogi Institute for Medical Science, Mishima, Settsu-shi, Osaka, Japan; and
| | - Izumi Ishikawa
- *Shionogi Institute for Medical Science, Mishima, Settsu-shi, Osaka, Japan; and
| | - Yuichi Ono
- *Shionogi Institute for Medical Science, Mishima, Settsu-shi, Osaka, Japan; and
| | - Toshio Imai
- *Shionogi Institute for Medical Science, Mishima, Settsu-shi, Osaka, Japan; and
- †Department of Bacteriology, Kinki University School of Medicine, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Ryuji Suzuki
- *Shionogi Institute for Medical Science, Mishima, Settsu-shi, Osaka, Japan; and
| | - Osamu Yoshie
- *Shionogi Institute for Medical Science, Mishima, Settsu-shi, Osaka, Japan; and
- †Department of Bacteriology, Kinki University School of Medicine, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| |
Collapse
|
35
|
Sherman MA, Nachman TY, Brown MA. Cutting Edge: IL-4 Production by Mast Cells Does Not Require c- maf. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.4.1733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Abstract
The c-maf transcription factor is selectively expressed in IL-4-producing Th2 cells. It has been implicated in IL-4 gene transcription based on its ability to directly activate the IL-4 gene in nonexpressing B cells and to promote IL-4-induced Th2 differentiation. However, it has not been definitively shown that IL-4 production by other cells is dependent on the presence of c-maf. Here, we show that IL-4-producing mast cells do not express the c-maf factor. Furthermore, mutation of a defined c-maf binding site within the proximal IL-4 promoter, which profoundly affects transcription in T cells, has no effect on expression of a reporter gene driven by the IL-4 promoter in mast cells. These results demonstrate that c-maf and its target binding site are not required for IL-4 production in all cell types and delineate additional cis- and trans-acting elements that contribute to the cell-type specific transcriptional regulation of IL-4.
Collapse
Affiliation(s)
- Melanie A. Sherman
- Emory University School of Medicine, Department of Experimental Pathology, Graduate Programs in Immunology and Molecular Pathogenesis and Genetics and Molecular Biology, Atlanta, GA 30322
| | - Tammy Y. Nachman
- Emory University School of Medicine, Department of Experimental Pathology, Graduate Programs in Immunology and Molecular Pathogenesis and Genetics and Molecular Biology, Atlanta, GA 30322
| | - Melissa A. Brown
- Emory University School of Medicine, Department of Experimental Pathology, Graduate Programs in Immunology and Molecular Pathogenesis and Genetics and Molecular Biology, Atlanta, GA 30322
| |
Collapse
|
36
|
Srivastava RK, Sasaki CY, Hardwick JM, Longo DL. Bcl-2-mediated drug resistance: inhibition of apoptosis by blocking nuclear factor of activated T lymphocytes (NFAT)-induced Fas ligand transcription. J Exp Med 1999; 190:253-65. [PMID: 10432288 PMCID: PMC2195578 DOI: 10.1084/jem.190.2.253] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Bcl-2 inhibits apoptosis induced by a variety of stimuli, including chemotherapy drugs and glucocorticoids. It is generally accepted that Bcl-2 exerts its antiapoptotic effects mainly by dimerizing with proapoptotic members of the Bcl-2 family such as Bax and Bad. However, the mechanism of the antiapoptotic effects is unclear. Paclitaxel and other drugs that disturb microtubule dynamics kill cells in a Fas/Fas ligand (FasL)-dependent manner; antibody to FasL inhibits paclitaxel-induced apoptosis. We have found that Bcl-2 overexpression leads to the prevention of chemotherapy (paclitaxel)-induced expression of FasL and blocks paclitaxel-induced apoptosis. The mechanism of this effect is that Bcl-2 prevents the nuclear translocation of NFAT (nuclear factor of activated T lymphocytes, a transcription factor activated by microtubule damage) by binding and sequestering calcineurin, a calcium-dependent phosphatase that must dephosphorylate NFAT to move to the nucleus. Without NFAT nuclear translocation, the FasL gene is not transcribed. Thus, it appears that paclitaxel and other drugs that disturb microtubule function kill cells at least in part through the induction of FasL. Furthermore, Bcl-2 antagonizes drug-induced apoptosis by inhibiting calcineurin activation, blocking NFAT nuclear translocation, and preventing FasL expression. The effects of Bcl-2 can be overcome, at least partially, through phosphorylation of Bcl-2. Phosphorylated Bcl-2 cannot bind calcineurin, and NFAT activation, FasL expression, and apoptosis can occur after Bcl-2 phosphorylation.
Collapse
Affiliation(s)
- R K Srivastava
- Laboratory of Immunology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224-6825, USA.
| | | | | | | |
Collapse
|
37
|
Song JS, Haleem-Smith H, Arudchandran R, Gomez J, Scott PM, Mill JF, Tan TH, Rivera J. Tyrosine Phosphorylation of Vav Stimulates IL-6 Production in Mast Cells by a Rac/c-Jun N-Terminal Kinase-Dependent Pathway. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.2.802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
This study investigates whether the guanine nucleotide exchange activity of Vav is linked to cytokine production in mast cells. Overexpression of Vav in the RBL-2H3 mast cell line resulted in the constitutive tyrosine phosphorylation and activation of Vav. We analyzed the functional effect of Vav overexpression on cytokine production. IL-2 and IL-6 mRNA levels were dramatically increased in Vav-overexpressing cells and correlated with increased NF-AT activity. Little or no effect was observed on the mRNA levels of IL-3, IL-4, GM-CSF, TNF-α, and TGF-β. FcεRI engagement did not further enhance IL-2 and IL-6 mRNA levels and only slightly enhanced NF-AT activity, but dramatically increased the mRNA levels of other tested cytokines. To understand the signal transduction required, we focused primarily on IL-6 induction by measuring mitogen-activated protein kinase activity and analyzing the effects of mutant or dominant negative forms of Vav, Rac1, and c-Jun N-terminal kinase-1 (JNK1). Vav overexpression resulted in the constitutive activation of JNK1 with little or no effect on p38 mitogen-activated protein kinase and ERK2. This was dependent on Vav-mediated activation of Rac1 as a Dbl domain-mutated Vav, inactive Rac N17, and inactive JNK1 down-regulated the Vav-induced JNK1 or IL-6 responses. Vav expression, but not expression of domain-mutated Vav, increased IL-6 secretion from nonimmortalized bone marrow-derived mast cells upon FcεRI engagement. We conclude that Vav phosphorylation contributes to IL-6 induction in mast cells.
Collapse
Affiliation(s)
- James S. Song
- *Section on Chemical Immunology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Hana Haleem-Smith
- *Section on Chemical Immunology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Ramachandran Arudchandran
- *Section on Chemical Immunology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jorge Gomez
- *Section on Chemical Immunology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Patricia M. Scott
- *Section on Chemical Immunology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - John F. Mill
- †Perinatal Research Facility, Department of Obstetrics and Gynecology PHC-3, Georgetown University School of Medicine, Washington, DC 20012; and
| | - Tse-Hua Tan
- ‡Department of Microbiology and Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Juan Rivera
- *Section on Chemical Immunology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
38
|
Altman A, Deckert M. The function of small GTPases in signaling by immune recognition and other leukocyte receptors. Adv Immunol 1999; 72:1-101. [PMID: 10361572 DOI: 10.1016/s0065-2776(08)60017-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- A Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, San Diego, California 92121, USA
| | | |
Collapse
|
39
|
Abstract
The acquisition of an IL-4-producing phenotype in Th2 cells requires IL-4 signaling through the STAT6 pathway during T cell differentiation. In this study we demonstrate that, unlike in naive T cells, IL-4 is not necessary for the development of an IL-4-producing phenotype in mast cells. Bone marrow-derived mast cell precursors from STAT6-/- mice can differentiate into mature cells that express IL-4 levels comparable to those of wild-type mast cells. In differentiated mast cells, activation in the presence of neutralizing anti-IL-4 antibodies or mutation of the consensus STAT6 sites does not diminish IL-4 promoter activity, indicating that IL-4 is not required for active transcription. Taken together, these data suggest that mast cell IL-4 production is not STAT6 dependent, providing evidence that these cells could generate IL-4 needed for the initiation and amplification of an effective Th2 immune response.
Collapse
Affiliation(s)
- M A Sherman
- Department of Pathology, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
40
|
Sherman MA, Powell DR, Weiss DL, Brown MA. NF-ATc Isoforms Are Differentially Expressed and Regulated in Murine T and Mast Cells. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.5.2820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
NF of activated T cells (NF-AT) denotes a family of transcription factors that regulate the activation-dependent expression of many immunologically important proteins. At least four distinct genes encode the various family members, and several isoforms of these have been identified as well. The overlapping expression patterns and similar in vitro binding and trans-activation activities on various promoter elements of NF-AT-regulated genes suggest some redundancy in the function of these proteins. However, the phenotypic analysis of NF-AT-deficient mice supports the idea that there are tissue- and gene-specific functions as well. In this study we have characterized the expression of NF-AT cDNAs in murine mast cells. The majority of clones identified correspond to two NF-ATc isoforms that differ only in their amino-terminal sequence. Despite minimal discrepancies in the coding region, there are striking tissue- and cell type-specific differences in isoform expression patterns. Detection of NF-ATc.α mRNA is strictly dependent on cell activation signals in both T and mast cell lines. In contrast, the β isoform is expressed at very low constitutive levels in both cell types but is only up-regulated in response to mast cell activation signals delivered through the FcεRI or via calcium ionophores. These results demonstrate another level of regulation within the NF-AT family that can contribute to cell type-specific gene expression.
Collapse
Affiliation(s)
| | | | - Deborah L. Weiss
- ‡Department of Chemistry, Williams College, Williamstown, MA 02167
| | - Melissa A. Brown
- *Department of Experimental Pathology, and
- †Graduate Program in Immunology and Molecular Pathogenesis and Genetics and Molecular Biology, Emory University School of Medicine, Atlanta, GA 30322; and
| |
Collapse
|
41
|
Armesilla AL, Lorenzo E, Gómez del Arco P, Martínez-Martínez S, Alfranca A, Redondo JM. Vascular endothelial growth factor activates nuclear factor of activated T cells in human endothelial cells: a role for tissue factor gene expression. Mol Cell Biol 1999; 19:2032-43. [PMID: 10022890 PMCID: PMC83996 DOI: 10.1128/mcb.19.3.2032] [Citation(s) in RCA: 160] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is a potent angiogenic inducer that stimulates the expression of tissue factor (TF), the major cellular initiator of blood coagulation. Here we show that signaling triggered by VEGF induced DNA-binding and transcriptional activities of nuclear factor of activated T cells (NFAT) and AP-1 in human umbilical vein endothelial cells (HUVECs). VEGF also induced TF mRNA expression and gene promoter activation by a cyclosporin A (CsA)-sensitive mechanism. As in lymphoid cells, NFAT was dephosphorylated and translocated to the nucleus upon activation of HUVECs, and these processes were blocked by CsA. NFAT was involved in the VEGF-mediated TF promoter activation as evidenced by cotransfection experiments with a dominant negative version of NFAT and site-directed mutagenesis of a newly identified NFAT site within the TF promoter that overlaps with a previously identified kappaB-like site. Strikingly, this site bound exclusively NFAT not only from nuclear extracts of HUVECs activated by VEGF, a stimulus that failed to induce NF-kappaB-binding activity, but also from extracts of cells activated with phorbol esters and calcium ionophore, a combination of stimuli that triggered the simultaneous activation of NFAT and NF-kappaB. These results implicate NFAT in the regulation of endothelial genes by physiological means and shed light on the mechanisms that switch on the gene expression program induced by VEGF and those regulating TF gene expression.
Collapse
Affiliation(s)
- A L Armesilla
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid, Facultad de Ciencias, Cantoblanco, Madrid 28049, Spain
| | | | | | | | | | | |
Collapse
|
42
|
Scholz D, Prieschl EE, Baumruker T. Mast cell signalling: a patent review. Expert Opin Ther Pat 1999. [DOI: 10.1517/13543776.9.1.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Annotated Patent Selections. Expert Opin Ther Pat 1999. [DOI: 10.1517/13543776.9.1.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
Bockamp EO, Fordham JL, Göttgens B, Murrell AM, Sanchez MJ, Green AR. Transcriptional regulation of the stem cell leukemia gene by PU.1 and Elf-1. J Biol Chem 1998; 273:29032-42. [PMID: 9786909 DOI: 10.1074/jbc.273.44.29032] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The SCL gene, also known as tal-1, encodes a basic helix-loop-helix transcription factor that is pivotal for the normal development of all hematopoietic lineages. SCL is expressed in committed erythroid, mast, and megakaryocytic cells as well as in hematopoietic stem cells. Nothing is known about the regulation of SCL transcription in mast cells, and in other lineages GATA-1 is the only tissue-specific transcription factor recognized to regulate the SCL gene. We have therefore analyzed the molecular mechanisms underlying SCL expression in mast cells. In this paper, we demonstrate that SCL promoter 1a was regulated by GATA-1 together with Sp1 and Sp3 in a manner similar to the situation in erythroid cells. However, SCL promoter 1b was strongly active in mast cells, in marked contrast to the situation in erythroid cells. Full activity of promoter 1b was dependent on ETS and Sp1/3 motifs. Transcription factors PU.1, Elf-1, Sp1, and Sp3 were all present in mast cell extracts, bound to promoter 1b and transactivated promoter 1b reporter constructs. These data provide the first evidence that the SCL gene is a direct target for PU.1, Elf-1, and Sp3.
Collapse
Affiliation(s)
- E O Bockamp
- University of Cambridge, Department of Haematology, Medical Research Council Centre, Hills Road, Cambridge CB2 2QH, United Kingdom
| | | | | | | | | | | |
Collapse
|
45
|
Nechushtan H, Razin E. Deciphering the early-response transcription factor networks in mast cells. IMMUNOLOGY TODAY 1998; 19:441-4. [PMID: 9785666 DOI: 10.1016/s0167-5699(98)01316-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- H Nechushtan
- Dept of Biochemistry, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | |
Collapse
|
46
|
Viola JP, Rao A. Role of the cyclosporin-sensitive transcription factor NFAT1 in the allergic response. Mem Inst Oswaldo Cruz 1998; 92 Suppl 2:147-55. [PMID: 9698927 DOI: 10.1590/s0074-02761997000800020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Proteins belonging to the NFAT (nuclear factor of activated T cells) family of transcription factors are expressed in most immune cell types, and play a central role in the transcription of cytokine genes, such as IL-2, IL-4, IL-5, IL-13, IFN-gamma, TNF-alpha, and GM-CSF. The activity NFAT proteins is regulated by the calcium/calmodulin-dependent phosphatase calcineurin, a target for inhibition by CsA and FK506. Recently, two different groups have described that mice lacking the NFAT1 transcription factor show an enhanced immune response, with tendency towards the development of a late Th2-like response. This review evaluates the possible role of NFAT proteins in the Th2 immune response and in the eosinophil-mediated allergic response.
Collapse
Affiliation(s)
- J P Viola
- Center for Blood Research, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
47
|
Boss V, Wang X, Koppelman LF, Xu K, Murphy TJ. Histamine induces nuclear factor of activated T cell-mediated transcription and cyclosporin A-sensitive interleukin-8 mRNA expression in human umbilical vein endothelial cells. Mol Pharmacol 1998; 54:264-72. [PMID: 9687567 DOI: 10.1124/mol.54.2.264] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The nuclear factor of activated T cells (NFAT) mediates a cyclosporin A (CsA)- and FK506-suppressible transcriptional program in lymphocytes after antigen-stimulated phospholipase C activation. Nonlymphoid cells also express NFAT isoforms, raising the possibility that these isoforms can be regulated by other extracellular stimuli. This study sought to determine whether histamine can trigger NFAT-mediated transcription in human umbilical vein endothelial cells (HUVEC), using a retrovirus-based luciferase reporter driven by a well characterized, NFAT-specific enhancer. Luciferase levels are induced up to 60-fold over basal levels after costimulation of HUVEC with Ca2+-mobilizing drugs and a phorbol ester, a response that is 20-fold greater than that observed when HUVEC are stimulated with either drug alone. These synergistic responses are inhibited in cells treated with CsA. CsA and FK506 also inhibit the luciferase response to histamine, indicating that histamine can induce NFAT-mediated transcription in HUVEC. To identify candidate genes in HUVEC that might be regulated by NFAT, the expression of several chemokine mRNAs was measured after histamine treatment. Of the mRNAs tested, only those encoding monocyte chemotactic protein-1 (approximately 2-fold over basal) and interleukin-8 (approximately 6-fold over basal) are induced by histamine; both of these responses are suppressed by CsA and FK506. The H1 histamine receptor antagonist chlorpheniramine, but not the H2 receptor antagonist ranitidine, blocks the effects of histamine in this preparation. These data provide the first evidence for a physiological inducer of NFAT-mediated transcription in endothelial cells and support the hypothesis that NFAT participates in H1 histamine receptor-induced interleukin-8 gene expression.
Collapse
Affiliation(s)
- V Boss
- Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
48
|
Boss V, Abbott KL, Wang XF, Pavlath GK, Murphy TJ. The cyclosporin A-sensitive nuclear factor of activated T cells (NFAT) proteins are expressed in vascular smooth muscle cells. Differential localization of NFAT isoforms and induction of NFAT-mediated transcription by phospholipase C-coupled cell surface receptors. J Biol Chem 1998; 273:19664-71. [PMID: 9677394 DOI: 10.1074/jbc.273.31.19664] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Expression of the antigen-regulated, cyclosporin A-sensitive nuclear factor of activated T cells (NFAT) is not restricted to lymphoid cells, as thought initially, but the physiological inducers of NFAT-mediated transcription in non-lymphoid cells are unknown. Here, cultured vascular smooth muscle cells (VSMC) are shown to express two isoforms of the NFAT family endogenously, which are localized differentially in cells under resting conditions. Using a retroviral NFAT-specific luciferase reporter, we show that VSMC support previously unrecognized complexities in NFAT-mediated transcription, including evidence for negative regulation by Ca2+ signaling and positive regulation through co-activation of adenylyl cyclase and Ca2+ mobilization. The VSMC mitogen platelet derived growth factor-BB (PDGF-BB) induces NFAT-mediated transcription in VSMC. Thrombin and angiotensin II, which activate Galphaq-coupled receptors, are significantly weaker inducers of NFAT-mediated luciferase expression than is PDGF-BB. However, co-stimulation studies show that Galphaq receptor agonists augment the NFAT-mediated transcriptional response to PDGF-BB. This synergy can be explained in part by augmented intracellular Ca2+ transients elicited by multiple agonist challenges. These data indicate that agonists for phospholipase C-coupled receptors stimulate NFAT-mediated transcription in VSMC differentially, and that NFAT can function to integrate co-activating signals in the extracellular environment.
Collapse
Affiliation(s)
- V Boss
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
49
|
Abstract
The Ig kappa 3' enhancer is required for high levels of Ig kappa gene expression. We now show that kappa 3' enhancer function increases five- to eightfold after stimulation of primary murine B cells with phorbol 12-myristate 13-acetate (PMA) and the calcium ionophore ionomycin. In the presence of cyclosporin A this induction is almost halved, suggesting that transcription factors of the NFAT family contribute to kappa 3' enhancer induction. Indeed, we identify a novel NFAT binding site which is required for full enhancer function. We find that this site is transcriptionally active in stimulated B cells, T cells and fibroblasts and that both PMA and ionomycin are required for maximal induction. Time course analysis of the components of the protein-DNA complex in primary lymphocytes reveals that both NFATp and NFATc are present in the complex after 15 min, while only NFATc is detectable after 4 h. This suggests that NFATc plays the dominant role in controlling long-term responses of this transcription factor family. Furthermore, JunB, JunD, FosB and cFos form part of the DNA-protein complex in Bal-17 B cells. Complex formation as well as transcriptional activity can also be induced by crosslinking of surface Ig. We have, thus, identified a unique NFAT complex in B cells that contributes to Ig kappa gene expression.
Collapse
Affiliation(s)
- K B Meyer
- The Wellcome/CRC Institute of Cancer and Developmental Biology, Department of Pathology, University of Cambridge, GB.
| | | |
Collapse
|
50
|
Regulation of Allergic Inflammation and Eosinophil Recruitment in Mice Lacking the Transcription Factor NFAT1: Role of Interleukin-4 (IL-4) and IL-5. Blood 1998. [DOI: 10.1182/blood.v91.7.2223.2223_2223_2230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transcription factors of the NFAT (nuclear factor of activated T cells) family regulate the expression of many genes encoding immunoregulatory cytokines and cell surface proteins during the immune response. The NFAT protein NFAT1 (NFATp) is expressed and functional in T cells, B cells, mast cells, and natural killer cells. Here we report a detailed analysis of the enhanced eosinophil responses of NFAT1-deficient mice, observed in an in vivo model of allergic inflammation. In addition to the pleural eosinophilia described previously, NFAT1−/− mice that have been sensitized with antigen display a significant increase, relative to wild-type mice, in the numbers of eosinophils in bone marrow and peripheral blood. After restimulation with antigen in vitro, antigen-responsive T cells from the draining lymph nodes of NFAT1−/− mice show increased expression of mRNA encoding the Th2 cytokines interleukin-4 (IL-4), IL-5, and IL-13. Consistent with this finding, there is a pronounced increase in the levels of IL-5 and IL-13 in the pleural cavities of sensitized NFAT1−/− mice after allergen challenge in vivo. Furthermore, development of eosinophilia depends on overexpression of IL-4 and IL-5, because it is strongly inhibited by administration of neutralizing antibodies to either of these cytokines. These results indicate that NFAT1-deficient mice are prone to develop a classically allergic phenotype characterized by eosinophilia and increased production of Th2 cytokines. Thus, the presence of NFAT1 might inhibit the allergic response, perhaps by interfering with the development of Th2 immune responses, and the lack or dysfunction of NFAT1 could potentially underlie certain cases of atopic disease.
Collapse
|