551
|
Heissmeyer V, Macián F, Im SH, Varma R, Feske S, Venuprasad K, Gu H, Liu YC, Dustin ML, Rao A. Calcineurin imposes T cell unresponsiveness through targeted proteolysis of signaling proteins. Nat Immunol 2004; 5:255-65. [PMID: 14973438 DOI: 10.1038/ni1047] [Citation(s) in RCA: 424] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2003] [Accepted: 01/22/2004] [Indexed: 12/12/2022]
Abstract
Sustained calcium signaling induces a state of anergy or antigen unresponsiveness in T cells, mediated through calcineurin and the transcription factor NFAT. We show here that Ca(2+)-induced anergy is a multistep program that is implemented at least partly through proteolytic degradation of specific signaling proteins. Calcineurin increased mRNA and protein of the E3 ubiquitin ligases Itch, Cbl-b and GRAIL and induced expression of Tsg101, the ubiquitin-binding component of the ESCRT-1 endosomal sorting complex. Subsequent stimulation or homotypic cell adhesion promoted membrane translocation of Itch and the related protein Nedd4, resulting in degradation of two key signaling proteins, PKC-theta and PLC-gamma1. T cells from Itch- and Cbl-b-deficient mice were resistant to anergy induction. Anergic T cells showed impaired calcium mobilization after TCR triggering and were unable to maintain a mature immunological synapse, instead showing late disorganization of the outer ring containing lymphocyte function-associated antigen 1. Our results define a complex molecular program that links gene transcription induced by calcium and calcineurin to a paradoxical impairment of signal transduction in anergic T cells.
Collapse
Affiliation(s)
- Vigo Heissmeyer
- Center for Blood Research and Department of Pathology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
552
|
Che T, You Y, Wang D, Tanner MJ, Dixit VM, Lin X. MALT1/paracaspase is a signaling component downstream of CARMA1 and mediates T cell receptor-induced NF-kappaB activation. J Biol Chem 2004; 279:15870-6. [PMID: 14754896 DOI: 10.1074/jbc.m310599200] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
T cell receptor (TCR) induces a series of signaling cascades and leads to activation of multiple transcription factors, including NF-kappaB. Although the mechanism of TCR-induced NF-kappaB activation is not fully understood, recent studies indicate that Bcl10 and CARMA1, two adaptor/scaffold proteins, play essential roles in mediating TCR-induced NF-kappaB activation. MALT1/paracaspase is a caspase-like protein that contains an N-terminal death domain, two Ig-like domains, and a C-terminal caspase-like domain. It binds to Bcl10 through its Ig-like domains and cooperates with Bcl10 to activate NF-kappaB. Recently, it has been shown that MALT1 is involved in mediating TCR signal transduction, leading to activation of NF-kappaB. In this study, we show that MALT1 is recruited into the lipid rafts of the immunological synapse following activation of the TCR and the CD28 coreceptor (CD3/CD28 costimulation). This recruitment of MALT1 is dependent on CARMA1 because CD3/CD28 costimulation failed to recruit MALT1 into lipid rafts in CARMA1-deficient T cells. In addition, we also found that MALT1 not only binds to Bcl10 directly, but also associates with CARMA1 in a Bcl10-independent manner. Therefore, MALT1, Bcl10, and CARMA1 form a trimolecular complex. Expression of a MALT1 deletion mutant containing only the N-terminal death domain and the two Ig-like domains completely blocked CD3/CD28 costimulation-induced, but not tumor necrosis factor-alpha-induced, NF-kappaB activation. Together, these results indicate that MALT1 is a crucial signaling component in the TCR signaling pathway.
Collapse
Affiliation(s)
- Tuanjie Che
- Department of Microbiology and Immunology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York 14214, USA
| | | | | | | | | | | |
Collapse
|
553
|
Wang D, Matsumoto R, You Y, Che T, Lin XY, Gaffen SL, Lin X. CD3/CD28 costimulation-induced NF-kappaB activation is mediated by recruitment of protein kinase C-theta, Bcl10, and IkappaB kinase beta to the immunological synapse through CARMA1. Mol Cell Biol 2004; 24:164-71. [PMID: 14673152 PMCID: PMC303359 DOI: 10.1128/mcb.24.1.164-171.2003] [Citation(s) in RCA: 180] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CARMA1 (also known as CARD11) is a scaffold molecule and contains a caspase-recruitment domain (CARD) and a membrane-associated guanylate kinase-like (MAGUK) domain. It plays an essential role in mediating CD3/CD28 costimulation-induced NF-kappaB activation. However, the molecular mechanism by which CARMA1 mediates costimulatory signals remains to be determined. Here, we show that CARMA1 is constitutively associated with the cytoplasmic membrane. This membrane association is essential for the function of CARMA1, since a mutant of CARMA1, CARMA1(L808P), that is defective in the membrane association cannot rescue CD3/CD28 costimulation-induced NF-kappaB activation in JPM50.6 CARMA1-deficient T cells. Although CD3/CD28 costimulation effectively induces the formation of the immunological synapse in CARMA1-deficient T cells, the recruitment of protein kinase C-theta (PKC-theta), Bcl10, and IkappaB kinase beta (IKKbeta) into lipid rafts of the immunological synapse is defective. Moreover, expression of wild-type CARMA1, but not CARMA1(L808P), restores the recruitment of PKC-theta, Bcl10, and IKKbeta into lipid rafts in CARMA1-deficient T cells. Consistently, expression of a mutant CARMA1, CARMA1(DeltaCD), that cannot associate with Bcl10 failed to restore CD3/CD28 costimulation-induced NF-kappaB activation in JPM50.6 cells, whereas expression of Bcl10-CARMA(DeltaCD) fusion protein effectively restored this NF-kappaB activation. Together, these results indicate that CARMA1 mediates CD3/CD28 costimulation-induced NF-kappaB activation by recruiting downstream signaling components into the immunological synapse.
Collapse
Affiliation(s)
- Donghai Wang
- Department of Microbiology and Immunology, School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, New York 14214, USA
| | | | | | | | | | | | | |
Collapse
|
554
|
Schaefer BC, Kappler JW, Kupfer A, Marrack P. Complex and dynamic redistribution of NF-kappaB signaling intermediates in response to T cell receptor stimulation. Proc Natl Acad Sci U S A 2004; 101:1004-9. [PMID: 14724296 PMCID: PMC327141 DOI: 10.1073/pnas.0307858100] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The central zone of the supramolecular activation cluster (c-SMAC) is a zone of T cell receptor (TCR) enrichment that forms at a T cell/antigen-presenting cell (APC) junction in response to antigen stimulation. We demonstrate that there is a surprisingly complex relocalization process that brings PKC and Bcl10, two intermediates in TCR activation of NF-kappaB, to the cytoplasmic face of the c-SMAC. TCR activation causes enrichment of PKC at the c-SMAC, followed by Bcl10 relocalization to punctate cytoplasmic structures, often at sites distant from the c-SMAC. These Bcl10 structures then undergo further relocalization, becoming enriched at the c-SMAC. TCR activation of NF-kappaB therefore involves the dynamic relocalization of multiple signaling intermediates, with distinct phases proximal to and distant from the c-SMAC.
Collapse
Affiliation(s)
- Brian C Schaefer
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | | | | | | |
Collapse
|
555
|
Tan SL, Parker PJ. Emerging and diverse roles of protein kinase C in immune cell signalling. Biochem J 2004; 376:545-52. [PMID: 14570590 PMCID: PMC1223826 DOI: 10.1042/bj20031406] [Citation(s) in RCA: 189] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2003] [Revised: 10/20/2003] [Accepted: 10/22/2003] [Indexed: 12/27/2022]
Abstract
Members of the protein kinase C (PKC) family are expressed in many different cell types, where they are known to regulate a wide variety of cellular processes that impact on cell growth and differentiation, cytoskeletal remodelling and gene expression in the response to diverse stimuli. The broad tissue distribution and redundancy of in vitro function have often hampered the identification of definitive roles for each PKC family member. However, recent in vivo studies of PKC isoenzyme-selective knockout and transgenic mice have highlighted distinct functions of individual PKCs in the immune system. These genetic analyses, along with biochemical studies utilizing PKC isoenzyme-specific cDNA (wild-type, constitutively active and dominant-negative), antisense oligonucleotides (ASO), RNA interference (RNAi), and pharmacological inhibitors, indicate that PKC-regulated signalling pathways play a significant role in many aspects of immune responses, from development, differentiation, activation and survival of lymphocytes to macrophage activation. The importance of PKCs in cellular immune responses suggests that improved understanding of the molecular events that govern their actions could point to new avenues for development of treatments for immune disorders.
Collapse
Affiliation(s)
- Seng-Lai Tan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | | |
Collapse
|
556
|
Vonakis BM, Sora R, Langdon JM, Casolaro V, MacDonald SM. Inhibition of cytokine gene transcription by the human recombinant histamine-releasing factor in human T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2004; 171:3742-50. [PMID: 14500674 DOI: 10.4049/jimmunol.171.7.3742] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human recombinant histamine-releasing factor (HrHRF) preincubation enhances the secretion of histamine, IL-4, and IL-13 from FcepsilonRI-stimulated human basophils. In GM-CSF-primed human eosinophils, HrHRF increases IL-8 production. Our recent experiments were designed to evaluate the effects of HrHRF on human T cell cytokine production. Purified T cells were preincubated with GST-tagged HrHRF, followed by stimulation with PMA and A23187 overnight. A partial inhibition of IL-2 and IL-13 production (30 and 75%, respectively) was detected compared with that in cells treated with PMA/A23187 alone. However, the production of IFN-gamma was similar in PMA/A23187 stimulated cells with or without HrHRF. The inhibition of cytokine protein production was dose dependent and specific to the HrHRF portion of GST-HrHRF. The inhibition was not due to endotoxin, since preincubation with polymyxin B and HrHRF gave similar results to that with HrHRF alone. The same pattern and specificity of cytokine regulation were replicated in the Jurkat T cell line as for primary T cells. The PMA/A23187-stimulated activity of a proximal promoter IL-13, IL-4, or IL-2 luciferase construct transfected into Jurkat cells was partially inhibited (60, 32, or 70%, respectively) upon GST-HrHRF preincubation, suggesting that HrHRF functions to inhibit cytokine production in Jurkat cells by preventing gene transcription. The inhibition of IL-2 promoter activation was specific to the HrHRF portion of GST-HrHRF. We conclude that HrHRF, in addition to functioning as a histamine-releasing factor, can differentially modulate the secretion of cytokines from human basophils, eosinophils, T cells, and murine B cells, suggesting that it may induce a complex array of responses at sites of allergic inflammation.
Collapse
Affiliation(s)
- Becky M Vonakis
- The Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
557
|
Schmitz ML, Bacher S, Dienz O. NF-kappaB activation pathways induced by T cell costimulation. FASEB J 2004; 17:2187-93. [PMID: 14656980 DOI: 10.1096/fj.02-1100rev] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Analysis of knockout mice and of T cells deficient for individual signaling proteins allowed the identification of novel members of the costimulation-induced NF-kappaB activation pathway while biochemical approaches started to unveil their functional mechanisms. These results show that NF-kappaB activation depends on an early wave of tyrosine phosphorylation that allows the inducible formation of multiprotein complexes containing several proteins required for NF-kappaB activation: adaptor proteins including Src homology 2 domain-containing leukocyte phosphoprotein 76 (SLP-76) and proteins with enzymatic activity, such as phospholipase C (PLC) gamma and the exchange factor Vav1. While Vav1 contributes to Rac-dependent reorganization of the actin cytoskeleton, activated PLCgamma1 generates the protein kinase C (PKC) activator diacylglycerol. In T cells, the novel PKC isoform PKCtheta is indispensable for NF-kappaB activation and its enzymatic activity depends on recruitment to the immunological synapse. Downstream from PKCtheta, the caspase recruitment domain (CARD) proteins CARD11/CARMA1 and Bcl10 relay T cell receptor-derived signals to the IkappaB kinase (IKK) complex. Many members of the NF-kappaB activation cascade, including the IKKs, are either constitutively or inducibly translocated to the lipid raft fraction, showing a highly organized spatial distribution of these NF-kappaB activating proteins.
Collapse
Affiliation(s)
- M Lienhard Schmitz
- University of Bern, Department of Chemistry and Biochemistry, Switzerland.
| | | | | |
Collapse
|
558
|
Ge B, Li O, Wilder P, Rizzino A, McKeithan TW. NF-kappa B regulates BCL3 transcription in T lymphocytes through an intronic enhancer. THE JOURNAL OF IMMUNOLOGY 2004; 171:4210-8. [PMID: 14530344 DOI: 10.4049/jimmunol.171.8.4210] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Exposure to soluble protein Ags in vivo leads to abortive proliferation of responding T cells. In the absence of a danger signal, artificially provided by adjuvants, most responding cells die, and the remainder typically become anergic. The adjuvant-derived signals provided to T cells are poorly understood, but recent work has identified BCL3 as the gene, of those tested, with the greatest differential transcriptional response to adjuvant administration in vivo. As an initial step in analyzing transcriptional responses of BCL3 in T cells, we have identified candidate regulatory regions within the locus through their evolutionary conservation and by analysis of DNase hypersensitivity. An evolutionarily conserved DNase hypersensitive site (HS3) within intron 2 was found to act as a transcriptional enhancer in response to stimuli that mimic TCR activation, namely, PHA and PMA. In luciferase reporter gene constructs transiently transfected into the Jurkat T cell line, the HS3 enhancer can cooperate not only with the BCL3 promoter, but also with an exogenous promoter from herpes simplex thymidine kinase. Deletional analysis revealed that a minimal sequence of approximately 81 bp is required for full enhancer activity. At the 5' end of this minimal sequence is a kappaB site, as confirmed by EMSAs. Mutation of this site in the context of the full-length HS3 abolished enhancer activity. Cotransfection with NF-kappaB p65 expression constructs dramatically increased luciferase activity, even without stimulation. Conversely, cotransfection with the NF-kappaB inhibitor IkappaBalpha reduced activation. Together, these results demonstrate a critical role for NF-kappaB in BCL3 transcriptional up-regulation by TCR-mimetic signals.
Collapse
Affiliation(s)
- Baosheng Ge
- Department of Internal Medicine, Section of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68132, USA
| | | | | | | | | |
Collapse
|
559
|
Sparatore B, Passalacqua M, Pedrazzi M, Ledda S, Patrone M, Gaggero D, Pontremoli S, Melloni E. Role of the kinase activation loop on protein kinase C theta activity and intracellular localisation. FEBS Lett 2003; 554:35-40. [PMID: 14596910 DOI: 10.1016/s0014-5793(03)01073-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multiple protein kinase C (PKC) theta species, identified in an erythroleukaemia cell line, have been characterised in terms of their molecular properties and intracellular distribution. PKCthetas localised in the detergent-soluble cell fraction have an Mr of 76 kDa (theta-76) and contain Thr538 or pThr538 in the kinase activation loop. In contrast, PKCthetas localised in the Golgi complex have an Mr of 85 kDa (theta-85) and, although unphosphorylated at Thr538, are catalytically active. Strikingly, only theta-76 species which are unphosphorylated at Thr538 can undergo autocatalytic conversion to theta-85. Moreover, a Thr538-->Ala PKCtheta mutant is constitutively localised in the Golgi complex, confirming that changes in the phosphorylation state of this residue play a pivotal role in the overall control of catalytic properties and localisation of this kinase.
Collapse
Affiliation(s)
- Bianca Sparatore
- Department of Experimental Medicine, Biochemistry Section, University of Genoa, Viale Benedetto XV, 16132 Genoa, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
560
|
Choi DS, Young H, McMahon T, Wang D, Messing RO. The mouse RACK1 gene is regulated by nuclear factor-kappa B and contributes to cell survival. Mol Pharmacol 2003; 64:1541-8. [PMID: 14645685 DOI: 10.1124/mol.64.6.1541] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Receptor for activated C kinase 1 (RACK1) is a multifunctional, WD motif-containing protein important in regulating several cell surface receptors and intracellular protein kinases. To better understand its function, we cloned the mouse RACK1 gene and found it contains eight exons and seven introns, and maps to mouse chromosome 11B1.2-1.3. Promoter analysis identified NF-kappaB as an important transcription factor for promoter activity. In PC-12 cells, nerve growth factor (NGF), which activates nuclear factor-kappaB (NF-kappaB), maintained RACK1 levels and promoted cell survival in serum-free medium. Inhibitors of NF-kappaB activation blocked NGF-stimulated survival and RACK1 expression, whereas transgenic expression of RACK1 promoted survival in cells deprived of serum and NGF. Thus, RACK1 gene expression is induced by NF-kappaB and RACK1 contributes to NF-kappaB-mediated cell survival.
Collapse
Affiliation(s)
- Doo-Sup Choi
- Ernest Gallo Clinic and Research Center, 5858 Horton St., Suite 200, Emeryville, CA 94608, USA
| | | | | | | | | |
Collapse
|
561
|
Jun JE, Goodnow CC. Scaffolding of antigen receptors for immunogenic versus tolerogenic signaling. Nat Immunol 2003; 4:1057-64. [PMID: 14586424 DOI: 10.1038/ni1001] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Lymphocyte antigen receptors are responsible for inducing the opposite responses of immunity or tolerance. How the correct polarity of antigen receptor signaling is encoded has been an enduring enigma. Here we summarize recent advances defining key scaffolding molecules, CARMA1 (also known as CARD11) and the Cbl family of ubiquitin ligases, required for either immunogenic or tolerogenic signaling by antigen receptors. These scaffolding proteins may determine the polarity of response to antigen by promoting assembly around antigen receptors of competing multiprotein signal complexes: immunosomes versus tolerosomes. Each of the factors that influence immunogenicity or tolerogenicity--stage of lymphocyte differentiation, concurrent engagement of inhibitory or costimulatory receptors, extent of receptor crosslinking, and prior antigen experience--may be integrated in lymphocytes through their capacity to influence the probability of assembling immunosomes versus tolerosomes.
Collapse
Affiliation(s)
- Jesse E Jun
- Australian Cancer Research Foundation Genetics Laboratory and Medical Genome Centre, John Curtin School of Medical Research, Australian National University, Canberra ACT 2601, Australia
| | | |
Collapse
|
562
|
Ruland J, Duncan GS, Wakeham A, Mak TW. Differential Requirement for Malt1 in T and B Cell Antigen Receptor Signaling. Immunity 2003; 19:749-58. [PMID: 14614861 DOI: 10.1016/s1074-7613(03)00293-0] [Citation(s) in RCA: 306] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The translocation t(11;18)(q21;q21) involving MALT1 is the most common chromosomal abnormality in lymphomas of mucosa-associated lymphoid tissue. Although the paracaspase MALT1 can bind to BCL10, the physiological function of MALT1 is unknown. Using mouse models, we show that Malt1 is essential for T cell activation, proliferation, and IL-2 production in response to TCR ligation and strictly required for signal-specific NF-kappaB activation induced by the TCR but not TNF-alpha or IL-1 signaling. Malt1 operates downstream of Bcl10, controls the catalytic activity of the canonical IKK complex, and regulates the signaling of Jnk and p38 MAP kinases. In contrast to Bcl10 disruption, however, inactivation of Malt1 has only mild effects on B cell activation and does not cause defects during neurodevelopment. Thus, Malt1 is an essential regulator of Bcl10 signaling that is differentially required depending on cellular context.
Collapse
MESH Headings
- Animals
- Caspases
- Lymphoma, B-Cell, Marginal Zone/genetics
- Lymphoma, B-Cell, Marginal Zone/metabolism
- Mice
- Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein
- NF-kappa B/metabolism
- Neoplasm Proteins/deficiency
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Signal Transduction/immunology
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Jürgen Ruland
- Advanced Medical Discovery Institute, Ontario Cancer Institute, University of Toronto, 620 University Avenue, Toronto, Ontario M5G 2C1, Canada.
| | | | | | | |
Collapse
|
563
|
Brooks DG, Arlen PA, Gao L, Kitchen CMR, Zack JA. Identification of T cell-signaling pathways that stimulate latent HIV in primary cells. Proc Natl Acad Sci U S A 2003; 100:12955-60. [PMID: 14569007 PMCID: PMC240726 DOI: 10.1073/pnas.2233345100] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2003] [Accepted: 09/02/2003] [Indexed: 11/18/2022] Open
Abstract
Eradication of HIV infection depends on the elimination of a small, but stable population of latently infected T cells. After the discontinuation of therapy, activation of latent virus can rekindle infection. To purge this reservoir, it is necessary to define cellular signaling pathways that lead to activation of latent HIV. We used the SCID-hu (Thy/Liv) mouse model of HIV latency to analyze a broad array of T cell-signaling pathways and show in primary, quiescent cells that viral induction depends on the activation of two primary intracellular signaling pathways, protein kinase C or nuclear factor of activated T cells (NF-AT). In contrast, inhibition or activation of other important T cell stimulatory pathways (such as mitogen-activated protein kinase, calcium flux, or histone deacetylation) do not significantly induce virus expression. We found that the activation of NF-kappaB is critical to viral reactivation; however, all pathways that stimulate NF-kappaBdonot reactivate latent virus. Our studies further show that inhibition of NF-kappaB does not prevent activation of HIV by NF-AT, indicating that these pathways can function independently to activate the HIV LTR. Thus, we define several molecular pathways that trigger HIV reactivation from latency and provide evidence that latent HIV infection is maintained by the functional lack of particular transcription factors in quiescent cells.
Collapse
Affiliation(s)
- David G Brooks
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California-Los Angeles, 10833 LeConte Avenue, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|
564
|
Ishaq M, DeGray G, Natarajan V. Protein kinase C theta modulates nuclear receptor-corepressor interaction during T cell activation. J Biol Chem 2003; 278:39296-302. [PMID: 12890684 DOI: 10.1074/jbc.m302767200] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transcriptional repression by nuclear receptor corepressors plays a critical role in T cell development. However, the role of these corepressors in T cell activation is poorly understood. We report that T cell activation silenced transcription driven by nuclear receptors retinoic acid receptor, retinoid X receptor, and thyroid hormone receptor and induced silencing mediator of retinoic acid and thyroid hormone receptors (SMRT)-receptor interaction. Whereas the expression of a dominant active mutant of protein kinase C theta(PKC theta) induced strong SMRT-receptor interaction in the absence of T cell activation, a dominant negative mutant of PKC theta decreased the interaction. Loss of PKC theta expression by induction of "RNA interference" resulted in the attenuation of basal and activation-induced SMRT-receptor interaction. We suggest that T cell activation silences nuclear receptor-dependent transactivation in part through PKC theta-dependent enhancement of SMRT-receptor interaction.
Collapse
Affiliation(s)
- Mohammad Ishaq
- Laboratory of Molecular Cell Biology, Science Applications International Corp., National Cancer Institute-Frederick, National Institutes of Health, Frederick, Maryland 21702, USA.
| | | | | |
Collapse
|
565
|
Trushin SA, Pennington KN, Carmona EM, Asin S, Savoy DN, Billadeau DD, Paya CV. Protein kinase Calpha (PKCalpha) acts upstream of PKCtheta to activate IkappaB kinase and NF-kappaB in T lymphocytes. Mol Cell Biol 2003; 23:7068-81. [PMID: 12972622 PMCID: PMC193945 DOI: 10.1128/mcb.23.19.7068-7081.2003] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
NF-kappaB is an ubiquitous transcription factor that is a key in the regulation of the immune response and inflammation. T-cell receptor (TCR) cross-linking leads to NF-kappaB activation, an IkappaB kinase (IKK)-dependent process. However, the upstream kinases that regulate IKK activity following TCR activation remain to be fully characterized. Herein, we demonstrate using genetic analysis, pharmacological inhibition, and RNA interference (RNAi) that the conventional protein kinase C (PKC) isoform PKCalpha, but not PKCbeta1, is required for the activation of the IKK complex following T-cell activation triggered by CD3/CD28 cross-linking. We find that in the presence of Ca(2+) influx, the catalytically active PKCalphaA25E induces IKK activity and NF-kappaB-dependent transcription; which is abrogated following the mutations of two aspartates at positions 246 and 248, which are required for Ca(2+) binding to PKCalpha and cell membrane recruitment. Kinetic studies reveal that an early phase (1 to 5 min) of IKK activation following TCR/CD28 cross-linking is PKCalpha dependent and that a later phase (5 to 25 min) of IKK activation is PKCtheta dependent. Activation of IKK- and NF-kappaB-dependent transcription by PKCalphaA25E is abrogated by the PKCtheta inhibitor rottlerin or the expression of the kinase-inactive form of PKCtheta. Taken together, our results suggest that PKCalpha acts upstream of PKCtheta to activate the IKK complex and NF-kappaB in T lymphocytes following TCR activation.
Collapse
Affiliation(s)
- Sergey A Trushin
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | |
Collapse
|
566
|
Browne EP, Shenk T. Human cytomegalovirus UL83-coded pp65 virion protein inhibits antiviral gene expression in infected cells. Proc Natl Acad Sci U S A 2003; 100:11439-44. [PMID: 12972646 PMCID: PMC208776 DOI: 10.1073/pnas.1534570100] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The initial interaction of human cytomegalovirus with fibroblasts triggers, and then partially blocks, an innate immune response pathway that leads to the induction of IFN-responsive genes and proinflammatory chemokines. Infection of fibroblasts with human cytomegalovirus inhibited their ability to respond to exogenous IFN. Consistent with the observation that the block did not depend on de novo viral protein synthesis, ectopic expression of the viral UL83-coded pp65, an abundant virion protein, inhibited IFN signaling. Furthermore, DNA array analysis showed that infection with a pp65-deficient mutant virus caused a much stronger induction of many IFN-response and proinflammatory chemokine RNAs than infection with wild-type virus. The nuclear DNA-binding activities of transcription factors NF-kappaB and IRF1 were induced to a much greater extent after infection with the pp65-deficient mutant than with wild-type virus. IFN-stimulated gene factor 3 DNA-binding was modestly enhanced, whereas IRF3 activity was not affected by mutation of pp65. Together, these results imply that pp65, which is delivered to newly infected cells in the virion, antagonizes a pathway that affects NF-kappaB and IRF1 and prevents the accumulation of mRNAs encoded by numerous cellular antiviral genes.
Collapse
Affiliation(s)
- Edward P Browne
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014, USA
| | | |
Collapse
|
567
|
Zhong XP, Hainey EA, Olenchock BA, Jordan MS, Maltzman JS, Nichols KE, Shen H, Koretzky GA. Enhanced T cell responses due to diacylglycerol kinase zeta deficiency. Nat Immunol 2003; 4:882-90. [PMID: 12883552 DOI: 10.1038/ni958] [Citation(s) in RCA: 195] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2003] [Accepted: 06/27/2003] [Indexed: 11/09/2022]
Abstract
Much is known about how T cell receptor (TCR) engagement leads to T cell activation; however, the mechanisms terminating TCR signaling remain less clear. Diacylglycerol, generated after TCR ligation, is essential in T cells. Its function must be controlled tightly to maintain normal T cell homeostasis. Previous studies have shown that diacylglycerol kinase zeta (DGKzeta), which converts diacylglycerol to phosphatidic acid, can inhibit TCR signaling. Here we show that DGKzeta-deficient T cells are hyperresponsive to TCR stimulation both ex vivo and in vivo. Furthermore, DGKzeta-deficient mice mounted a more robust immune response to lymphocytic choriomeningitis virus infection than did wild-type mice. These results demonstrate the importance of DGKzeta as a physiological negative regulator of TCR signaling and T cell activation.
Collapse
MESH Headings
- Animals
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Cell Division/immunology
- DNA-Binding Proteins/immunology
- DNA-Binding Proteins/metabolism
- Diacylglycerol Kinase/deficiency
- Diacylglycerol Kinase/immunology
- Flow Cytometry
- Guanine Nucleotide Exchange Factors
- Immunoblotting
- Lectins, C-Type
- Lymphocyte Activation/immunology
- Lymphocytic Choriomeningitis/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Phosphatidic Acids/immunology
- Phosphatidic Acids/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Interleukin-2/immunology
- Receptors, Interleukin-2/metabolism
- Signal Transduction/immunology
- T-Lymphocytes/cytology
- T-Lymphocytes/enzymology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Xiao-Ping Zhong
- The Signal Transduction Program, The Abramson Family Cancer Research Institute, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
568
|
Mora AL, Corn RA, Stanic AK, Goenka S, Aronica M, Stanley S, Ballard DW, Joyce S, Boothby M. Antiapoptotic function of NF-kappaB in T lymphocytes is influenced by their differentiation status: roles of Fas, c-FLIP, and Bcl-xL. Cell Death Differ 2003; 10:1032-44. [PMID: 12934078 DOI: 10.1038/sj.cdd.4401257] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Inducible protection from apoptosis in vivo controls the size of cell populations. An important question in this respect is how differentiation affects mechanisms of apoptosis regulation. Among mature T lymphocytes, the NF-kappaB/Rel transcription factors are coupled to receptors that control cell population sizes by concurrently regulating survival and multiplication. In the present study, we used a transgenic inhibitor of NF-kappaB/Rel signaling to investigate the role of this pathway in proliferation and death of mature T cells in vivo. The results indicate that NF-kappaB integrates two critical yet distinct molecular pathways preventing apoptosis affected by the death receptor Fas, coordinately regulating levels of FLIP and Bcl-x(L) in primary T cells. Surprisingly, NF-kappaB blockade preferentially impacted naive as compared to memory T cells. The Fas/FasL pathway was linked to these findings by evidence that the abnormalities imposed by NF-kappaB inhibition were ameliorated by Fas deficiency, particularly for the CD4(+) lineage. Moreover, levels of an inhibitor of Fas-mediated apoptosis, c-FLIP, were diminished in cells expressing the transgenic inhibitor. NF-kappaB was also linked to T cell survival in vivo by mediating induction of Bcl-x(L): restoration of Bcl-x(L) levels reversed the preferential deficit of naive T cells, differentially impacting the CD4 and CD8 subsets. These results show that promoting survival and effective multiplication are central roles for NF-kappaB in T lymphoid homeostasis in vivo, but this effect and its underlying mechanisms are influenced by the developmental state of the lymphocyte.
Collapse
Affiliation(s)
- A L Mora
- Department of Microbiology & Immunology, Vanderbilt University Medical School, Nashville, TN 37232-2363, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
569
|
Xue L, Morris SW, Orihuela C, Tuomanen E, Cui X, Wen R, Wang D. Defective development and function of Bcl10-deficient follicular, marginal zone and B1 B cells. Nat Immunol 2003; 4:857-65. [PMID: 12910267 DOI: 10.1038/ni963] [Citation(s) in RCA: 152] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2003] [Accepted: 07/10/2003] [Indexed: 01/16/2023]
Abstract
Bcl10 is an intracellular protein essential for nuclear factor (NF)-kappaB activation after lymphocyte antigen receptor stimulation. Using knockout mice, we show that absence of Bcl10 impeded conversion from transitional type 2 to mature follicular B cells and caused substantial decreases in marginal zone and B1 B cells. Bcl10-deficient B cells showed no excessive apoptosis. However, both Bcl10-deficient follicular and marginal zone B cells failed to proliferate normally, although Bcl10-deficient marginal zone B cells uniquely failed to activate NF-kappaB efficiently after stimulation with lipopolysaccharide. Bcl10-deficient marginal zone B cells did not capture antigens, and Bcl10-deficient (Bcl10-/-) mice failed to initiate humoral responses, leading to an inability to clear blood-borne bacteria. Thus, Bcl10 is essential for the development of all mature B cell subsets.
Collapse
Affiliation(s)
- Liquan Xue
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | | | | | |
Collapse
|
570
|
Abstract
A functional immune system requires the selection of T lymphocytes expressing receptors that are major histocompatibility complex restricted but tolerant to self-antigens. This selection occurs predominantly in the thymus, where lymphocyte precursors first assemble a surface receptor. In this review we summarize the current state of the field regarding the natural ligands and molecular factors required for positive and negative selection and discuss a model for how these disparate outcomes can be signaled via the same receptor. We also discuss emerging data on the selection of regulatory T cells. Such cells require a high-affinity interaction with self-antigens, yet differentiate into regulatory cells instead of being eliminated.
Collapse
Affiliation(s)
- Timothy K Starr
- Center for Immunology and the Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis 55455, USA.
| | | | | |
Collapse
|
571
|
Wang C, Mooney JL, Meza-Romero R, Chou YK, Huan J, Vandenbark AA, Offner H, Burrows GG. Recombinant TCR ligand induces early TCR signaling and a unique pattern of downstream activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:1934-40. [PMID: 12902496 DOI: 10.4049/jimmunol.171.4.1934] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recombinant TCR ligands (RTLs) consisting of covalently linked alpha(1) and beta(1) domains of MHC class II molecules tethered to specific antigenic peptides represent minimal TCR ligands. In a previous study we reported that the rat RTL201 construct, containing RT1.B MHC class II domains covalently coupled to the encephalitogenic guinea pig myelin basic protein (Gp-MBP(72-89)) peptide, could prevent and treat actively and passively induced experimental autoimmune encephalomyelitis in vivo by selectively inhibiting Gp-MBP(72-89) peptide-specific CD4(+) T cells. To evaluate the inhibitory signaling pathway, we tested the effects of immobilized RTL201 on T cell activation of the Gp-MBP(72-89)-specific A1 T cell hybridoma. Activation was exquisitely Ag-specific and could not be induced by RTL200 containing the rat MBP(72-89) peptide that differed by a threonine for serine substitution at position 80. Partial activation by RTL201 included a CD3zeta p23/p21 ratio shift, ZAP-70 phosphorylation, calcium mobilization, NFAT activation, and transient IL-2 production. In comparison, anti-CD3epsilon treatment produced stronger activation of these cellular events with additional activation of NF-kappaB and extracellular signal-regulated kinases as well as long term increased IL-2 production. These results demonstrate that RTLs can bind directly to the TCR and modify T cell behavior through a partial activation mechanism, triggering specific downstream signaling events that deplete intracellular calcium stores without fully activating T cells. The resulting Ag-specific activation of the transcription factor NFAT uncoupled from the activation of NF-kappaB or extracellular signal-regulated kinases constitutes a unique downstream activation pattern that accounts for the inhibitory effects of RTL on encephalitogenic CD4(+) T cells.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- CD3 Complex/metabolism
- Calcium Signaling/genetics
- Calcium Signaling/immunology
- DNA-Binding Proteins/metabolism
- Guinea Pigs
- HLA-DR2 Antigen/metabolism
- HLA-DR2 Antigen/physiology
- Histocompatibility Antigens/metabolism
- Histocompatibility Antigens/physiology
- Humans
- Hybridomas
- Interleukin-2/biosynthesis
- Ligands
- Lymphocyte Activation/genetics
- Mice
- Mitogen-Activated Protein Kinases/metabolism
- Molecular Sequence Data
- Myelin Basic Protein/metabolism
- Myelin Basic Protein/pharmacology
- NF-kappa B/metabolism
- NFATC Transcription Factors
- Nuclear Proteins
- Peptide Fragments/metabolism
- Peptide Fragments/pharmacology
- Phosphorylation
- Protein-Tyrosine Kinases/metabolism
- Rats
- Rats, Inbred Lew
- Receptors, Antigen, T-Cell/physiology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/physiology
- Recombinant Proteins/metabolism
- Recombinant Proteins/pharmacology
- T-Lymphocytes/enzymology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transcription Factors/metabolism
- Tumor Cells, Cultured
- ZAP-70 Protein-Tyrosine Kinase
Collapse
Affiliation(s)
- Chunhe Wang
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | | | | | | | | | | | | | | |
Collapse
|
572
|
Díaz-Flores E, Siliceo M, Martínez-A C, Mérida I. Membrane translocation of protein kinase Ctheta during T lymphocyte activation requires phospholipase C-gamma-generated diacylglycerol. J Biol Chem 2003; 278:29208-15. [PMID: 12738795 DOI: 10.1074/jbc.m303165200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein kinase C (PKC) is the only PKC isoform recruited to the immunological synapse after T cell receptor stimulation, suggesting that its activation mechanism differs from that of the other isoforms. Previous studies have suggested that this selective PKC recruitment may operate via a Vav-regulated, cytoskeletal-dependent mechanism, independent of the classical phospholipase C/diacylglycerol pathway. Here, we demonstrate that, together with tyrosine phosphorylation of PKC in the regulatory domain, binding of phospholipase C-dependent diacylglycerol is required for PKC recruitment to the T cell synapse. In addition, we demonstrate that diacylglycerol kinase alpha-dependent diacylglycerol phosphorylation provides the negative signal required for PKC inactivation, ensuring fine control of the T cell activation response.
Collapse
Affiliation(s)
- Ernesto Díaz-Flores
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Cantoblanco, E-28049 Madrid, Spain
| | | | | | | |
Collapse
|
573
|
Thome M, Tschopp J. TCR-induced NF-kappaB activation: a crucial role for Carma1, Bcl10 and MALT1. Trends Immunol 2003; 24:419-24. [PMID: 12909454 DOI: 10.1016/s1471-4906(03)00177-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- B-Cell CLL-Lymphoma 10 Protein
- Caspases
- Guanylate Kinases
- Humans
- Lymphocyte Activation
- Lymphoma, B-Cell, Marginal Zone/chemistry
- Lymphoma, B-Cell, Marginal Zone/immunology
- Lymphoma, B-Cell, Marginal Zone/metabolism
- Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein
- NF-kappa B/immunology
- NF-kappa B/metabolism
- Neoplasm Proteins/chemistry
- Neoplasm Proteins/immunology
- Neoplasm Proteins/metabolism
- Nucleoside-Phosphate Kinase/immunology
- Nucleoside-Phosphate Kinase/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Margot Thome
- Institute of Biochemistry, University of Lausanne, Chemin des Boveresses 153, CH-1066 Epalinges, Switzerland.
| | | |
Collapse
|
574
|
Hock MB, Brown MA. Nuclear factor of activated T cells 2 transactivation in mast cells: a novel isoform-specific transactivation domain confers unique FcepsilonRI responsiveness. J Biol Chem 2003; 278:26695-703. [PMID: 12738787 DOI: 10.1074/jbc.m301007200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Murine nuclear factor of activated T cells (NFAT)2.alpha/beta differ by 42 and 28 unique amino-terminal amino acids and are differentially expressed. Both isoforms share conserved domains that regulate DNA-binding and subcellular localization. A genetic "one-hybrid" assay was used to define two distinct transactivation (TA) domains: in addition to a conserved TAD present in both isoforms, a second, novel TAD exists within the beta-specific amino terminus. Pharmacologic inhibitors Gö6976 and rottlerin demonstrate that both conventional and novel protein kinase C (PKC) family members regulate endogenous mast cell NFAT activity, and NFAT2 TA. Overexpression of dominant active PKC (which has been implicated in immune receptor signaling) induces NFAT2.alpha/beta TA. Mutations within the smallest PKC-responsive transactivation domain demonstrate that the PKC effect is at least partially indirect. Significantly, the beta-specific domain confers greater ability to TA in response to treatment with phorbol 12-myristate 13-acetate/ionomycin or lipopolysaccharide, and unique sensitivity to FcepsilonRI signaling. Accordingly, overexpression of NFAT2.beta results in significantly greater NFAT- and interleukin-4 reporter activity than NFAT2.alpha. These results suggest that whereas NFAT2 isoforms may share redundant DNA-binding preferences, there are specialized functional consequences of their isoform-specific domains.
Collapse
Affiliation(s)
- M Benjamin Hock
- Department of Pathology and Graduate Program in Genetics and Molecular Biology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
575
|
Egawa T, Albrecht B, Favier B, Sunshine MJ, Mirchandani K, O'Brien W, Thome M, Littman DR. Requirement for CARMA1 in antigen receptor-induced NF-kappa B activation and lymphocyte proliferation. Curr Biol 2003; 13:1252-8. [PMID: 12867038 DOI: 10.1016/s0960-9822(03)00491-3] [Citation(s) in RCA: 216] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ligation of antigen receptors (TCR, BCR) on T and B lymphocytes leads to the activation of new transcriptional programs and cell cycle progression. Antigen receptor-mediated activation of NF-kappa B, required for proliferation of B and T cells, is disrupted in T cells lacking PKC theta and in B and T cells lacking Bcl10, a caspase recruitment domain (CARD)-containing adaptor protein. CARMA1 (also called CARD11 and Bimp3), the only lymphocyte-specific member in a family of membrane-associated guanylate kinase (MAGUK) scaffolding proteins that interact with Bcl10 by way of CARD-CARD interactions, is required for TCR-induced NF-kappa B activation in Jurkat T lymphoma cells. Here we show that T cells from mice lacking CARMA1 expression were defective in recruitment of Bcl10 to clustered TCR complexes and lipid rafts, in activation of NF-kappa B, and in induction of IL-2 production. Development of CD5(+) peritoneal B cells was disrupted in these mice, as was B cell proliferation in response to both BCR and CD40 ligation. Serum immunoglobulin levels were also markedly reduced in the mutant mice. Together, these results show that CARMA1 has a central role in antigen receptor signaling that results in activation and proliferation of both B and T lymphocytes.
Collapse
Affiliation(s)
- Takeshi Egawa
- Molecular Pathogenesis Program, Skirball Institute for Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | | | | | | | | | | | | | | |
Collapse
|
576
|
Serra C, Federici M, Buongiorno A, Senni MI, Morelli S, Segratella E, Pascuccio M, Tiveron C, Mattei E, Tatangelo L, Lauro R, Molinaro M, Giaccari A, Bouché M. Transgenic mice with dominant negative PKC-theta in skeletal muscle: a new model of insulin resistance and obesity. J Cell Physiol 2003; 196:89-97. [PMID: 12767044 DOI: 10.1002/jcp.10278] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Protein kinase C theta (PKC-theta) is the PKC isoform predominantly expressed in skeletal muscle, and it is supposed to mediate many signals necessary for muscle histogenesis and homeostasis, such as TGFbeta, nerve-dependent signals and insulin. To study the role of PKC-theta in these mechanisms we generated transgenic mice expressing a "kinase dead" mutant form of PKC-theta (PKC-thetaK/R), working as "dominant negative," specifically in skeletal muscle. These mice are viable and fertile, however, by the 6-7 months of age, they gain weight, mainly due to visceral fat deposition. Before the onset of obesity (4 months of age), they already show increased fasting and fed insulin levels and reduced insulin-sensitivity, as measured by ipITT, but normal glucose tolerance, as measured by ipGTT. After the 6-7 months of age, transgenic mice develop hyperinsulinemia in the fasting and fed state. The ipGTT revealed in the transgenic mice both hyperglycemia and hyperinsulinemia. At the molecular level, impaired activation of the IR/IRS/PI3K pathway and a significant decrease both in the levels and in insulin-stimulated activation of the serine/threonine kinase Akt were observed. Taken together these data demonstrate that over-expression of dominant negative PKC-theta in skeletal muscle causes obesity associated to insulin resistance, as demonstrated by defective receptor and post-receptorial activation of signaling cascade.
Collapse
Affiliation(s)
- C Serra
- Department of Histology and Medical Embryology, University of Rome La Sapienza, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
577
|
Li-Weber M, Krammer PH. Regulation of IL4 gene expression by T cells and therapeutic perspectives. Nat Rev Immunol 2003; 3:534-43. [PMID: 12876556 DOI: 10.1038/nri1128] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Interleukin-4 (IL-4) is crucial for the differentiation of naive T helper (T(H)) cells into the T(H)2 effector cells that promote humoral (antibody) immunity and provide protection against intestinal helminths. IL-4 also has a central role in the pathogenesis of allergic inflammation. Many transcription factors are involved in the regulation of expression of the gene encoding IL-4. Initiation of transcription of the gene encoding IL-4 in naive T(H) cells is regulated by the T(H)2-specific transcription factor GATA3, whereas acute expression of the gene encoding IL-4 in T(H)2 cells is mediated by inducible, ubiquitous transcription factors after antigen encounter. This review focuses on acute activation of the gene encoding IL-4 in T cells and discusses therapeutic perspectives at the transcriptional level.
Collapse
Affiliation(s)
- Min Li-Weber
- Tumour Immunology Programme D030, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany.
| | | |
Collapse
|
578
|
Sanui T, Inayoshi A, Noda M, Iwata E, Oike M, Sasazuki T, Fukui Y. DOCK2 is essential for antigen-induced translocation of TCR and lipid rafts, but not PKC-theta and LFA-1, in T cells. Immunity 2003; 19:119-29. [PMID: 12871644 DOI: 10.1016/s1074-7613(03)00169-9] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
DOCK2 is a mammalian homolog of Caenorhabditis elegans CED-5 and Drosophila melanogaster Myoblast City which are known to regulate actin cytoskeleton. DOCK2 is critical for lymphocyte migration, yet the role of DOCK2 in TCR signaling remains unclear. We show here that DOCK2 is essential for TCR-mediated Rac activation and immunological synapse formation. In DOCK2-deficient T cells, antigen-induced translocation of TCR and lipid rafts, but not PKC-theta and LFA-1, to the APC interface was severely impaired, resulting in a significant reduction of antigen-specific T cell proliferation. In addition, we found that the efficacy of both positive and negative selection was reduced in DOCK2-deficient mice. These results suggest that DOCK2 regulates T cell responsiveness through remodeling of actin cytoskeleton via Rac activation.
Collapse
Affiliation(s)
- Terukazu Sanui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
579
|
Abstract
Many studies have shown the central importance of the co-receptors CD28, inducible costimulatory molecule (ICOS) and cytotoxic T lymphocyte antigen 4 (CTLA4) in the regulation of many aspects of T-cell function. CD28 and ICOS have both overlapping and distinct functions in the positive regulation of T-cell responses, whereas CTLA4 negatively regulates the response. The signalling pathways that underlie the function of each of the co-receptors indicate their shared and unique properties and provide compelling hints of functions that are as yet uncovered. Here, we outline the shared and distinct signalling events that are associated with each of the co-receptors and provide unifying concepts that are related to signalling functions of these co-receptors.
Collapse
Affiliation(s)
- Christopher E Rudd
- Molecular Immunology Section, Department of Immunology, Division of Investigative Science, Faculty of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 ONN, UK.
| | | |
Collapse
|
580
|
Pfeifhofer C, Kofler K, Gruber T, Tabrizi NG, Lutz C, Maly K, Leitges M, Baier G. Protein kinase C theta affects Ca2+ mobilization and NFAT cell activation in primary mouse T cells. J Exp Med 2003; 197:1525-35. [PMID: 12782715 PMCID: PMC2193906 DOI: 10.1084/jem.20020234] [Citation(s) in RCA: 270] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Protein kinase C (PKC)theta is an established component of the immunological synapse and has been implicated in the control of AP-1 and NF-kappaB. To study the physiological function of PKCtheta, we used gene targeting to generate a PKCtheta null allele in mice. Consistently, interleukin 2 production and T cell proliferative responses were strongly reduced in PKCtheta-deficient T cells. Surprisingly, however, we demonstrate that after CD3/CD28 engagement, deficiency of PKCtheta primarily abrogates NFAT transactivation. In contrast, NF-kappaB activation was only partially reduced. This NFAT transactivation defect appears to be secondary to reduced inositol 1,4,5-trisphosphate generation and intracellular Ca2+ mobilization. Our finding suggests that PKCtheta plays a critical and nonredundant role in T cell receptor-induced NFAT activation.
Collapse
Affiliation(s)
- Christa Pfeifhofer
- Institute of Medical Biology and Human Genetics, University of Innsbruck, Schoepfstrasse 41, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | |
Collapse
|
581
|
Abstract
Recent technological developments have facilitated explorations of the sustained signalling pathways important for lymphocyte activation. One key response necessary for lymphocyte activation is the prolonged activation of the lipid kinase phosphatidylinositol-3 kinase (PI3K). It has also been shown that there is sustained activation of diverse serine/threonine kinases in activated lymphocytes.
Collapse
Affiliation(s)
- Doreen A Cantrell
- Division of Cell Biology and Immunology, School of Life Sciences, MSI/WTB Complex, University of Dundee, Dow Street, Dundee DD1 5EH, UK.
| |
Collapse
|
582
|
Abstract
Signaling leading to the survival or apoptosis of immune system cells must be balanced to ensure the normal mounting and extinguishing of immune responses. One of the essential regulators of immune cell survival is the transcription factor nuclear factor kappaB (NF-kappaB). NF-kappaB is critical for the activation of T and B lymphocytes and is a central coordinator of innate and adaptive immunity. Pathogen recognition, whether mediated via the Toll-like receptors or via the antigen-specific T- and B-cell receptors, initiates the activation of distinct signal transduction pathways that activate NF-kappaB. Activation of NF-kappaB by these pathways is necessary for lymphocyte activation, expansion, and effector function in response to infection. In addition, recent work has shown that the aberrant activation of NF-kappaB by these pathways can contribute to the development of autoimmunity, chronic inflammation, or lymphoid malignancy. There is thus an urgent need to understand the exact molecular details of these signal transduction cascades so that we may develop novel therapeutics. This article will review the specific signal transduction pathways that mediate NF-kappaB activation in response to antigen receptor ligation in T and B lymphocytes. These newly defined pathways, which are essential for adaptive immune responses, are built around the key adapter protein, Bcl-10. Bcl-10 is known to participate in chromosomal translocations in human mucosa-associated lymphoid tissue lymphomas.
Collapse
Affiliation(s)
- Jürgen Ruland
- Advanced Medical Discovery Institute, Ontario Cancer Institute and University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
583
|
Abstract
Lymphocyte activation via antigen receptors initiates adaptive immune responses. Two papers in this issue of Immunity demonstrate that CARMA-1, a CARD carrying member of the MAGUK family proteins, is essential for lymphocyte activation. CARMA-1 functions by coupling antigen receptor signals to NF-kappaB induction and JNK activation.
Collapse
Affiliation(s)
- Abul K Abbas
- Department of Pathology, University of California San Francisco School of Medicine, San Francisco, CA 94143, USA
| | | |
Collapse
|
584
|
Hara H, Wada T, Bakal C, Kozieradzki I, Suzuki S, Suzuki N, Nghiem M, Griffiths EK, Krawczyk C, Bauer B, D'Acquisto F, Ghosh S, Yeh WC, Baier G, Rottapel R, Penninger JM. The MAGUK family protein CARD11 is essential for lymphocyte activation. Immunity 2003; 18:763-75. [PMID: 12818158 DOI: 10.1016/s1074-7613(03)00148-1] [Citation(s) in RCA: 283] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Members of the MAGUK family proteins cluster receptors and intracellular signaling molecules at the neuronal synapse. We report that genetic inactivation of the MAGUK family protein CARD11/Carma1/Bimp3 results in a complete block in T and B cell immunity. CARD11 is essential for antigen receptor- and PKC-mediated proliferation and cytokine production in T and B cells due to a selective defect in JNK and NFkappaB activation. Moreover, B cell proliferation and JNK activation were impaired upon stimulation of TLR4 with lipopolysaccharide, indicating that CARD11 is involved in both the innate and adaptive immune systems. Our results show that the same family of molecules are critical regulators of neuronal synapses and immune receptor signaling.
Collapse
Affiliation(s)
- Hiromitsu Hara
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 7, A-1030, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
585
|
Grundström S, Cederbom L, Sundstedt A, Scheipers P, Ivars F. Superantigen-induced regulatory T cells display different suppressive functions in the presence or absence of natural CD4+CD25+ regulatory T cells in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5008-17. [PMID: 12734345 DOI: 10.4049/jimmunol.170.10.5008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Repeated exposures to both microbial and innocuous Ags in vivo have been reported to both eliminate and tolerize T cells after their initial activation and expansion. The remaining tolerant T cells have been shown to suppress the response of naive T cells in vitro. This feature is reminiscent of natural CD4(+)CD25(+) regulatory T cells. However, it is not known whether the regulatory function of in vivo-tolerized T cells is similar to the function of natural CD4(+)CD25(+) regulatory T cells. In this study, we demonstrate that CD4(+)CD25(+) as well as CD4(+)CD25(-) T cells isolated from mice treated with superantigen three consecutive times to induce tolerance were functionally comparable to natural CD4(+)CD25(+) regulatory T cells, albeit more potent. The different subpopulations of in vivo-tolerized CD4(+) T cells efficiently down-modulated costimulatory molecules on dendritic cells, and their suppressive functions were strictly cell contact dependent. Importantly, we demonstrate that conventional CD4(+)CD25(-) T cells could also be induced to acquire regulatory functions by the same regimen in the absence of natural regulatory T cells in vivo, but that such regulatory cells were functionally different.
Collapse
MESH Headings
- Abatacept
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, CD
- Antigens, Differentiation/biosynthesis
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CTLA-4 Antigen
- Cell Communication/genetics
- Cell Communication/immunology
- Cells, Cultured
- Clonal Anergy/genetics
- Clonal Anergy/immunology
- Clonal Deletion/genetics
- Clonal Deletion/immunology
- Cytokines/physiology
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- Dose-Response Relationship, Immunologic
- Down-Regulation/genetics
- Down-Regulation/immunology
- Drug Administration Schedule
- Enterotoxins/administration & dosage
- Enterotoxins/pharmacology
- Female
- Genes, T-Cell Receptor beta/immunology
- Immunity, Innate/genetics
- Immunoconjugates
- Injections, Intravenous
- Interleukin-10/antagonists & inhibitors
- Interleukin-10/metabolism
- Leukocyte Common Antigens/biosynthesis
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, SCID
- Mice, Transgenic
- Receptors, Interleukin-2/biosynthesis
- Staphylococcus aureus/immunology
- Superantigens/administration & dosage
- Superantigens/pharmacology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/physiology
Collapse
|
586
|
Koike T, Yamagishi H, Hatanaka Y, Fukushima A, Chang JW, Xia Y, Fields M, Chandler P, Iwashima M. A novel ERK-dependent signaling process that regulates interleukin-2 expression in a late phase of T cell activation. J Biol Chem 2003; 278:15685-92. [PMID: 12595531 DOI: 10.1074/jbc.m210829200] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Engagement of the T cell antigen receptor (TCR) rapidly induces multiple signal transduction pathways, including ERK activation. Here, we report a critical role for ERK at a late stage of T cell activation. Inhibition of the ERK pathway 2-6 h after the start of TCR stimulation significantly impaired interleukin-2 (IL-2) production, whereas the same treatment during the first 2 h had no effect. ERK inhibition significantly impaired nuclear translocation of c-Rel with a minimum reduction of NF-AT activity. Requirement for sustained ERK activation was also confirmed using primary T cells. To induce sustained activation of ERK, T cells required continuous engagement of TCR. Stimulation of T cells with soluble anti-TCR antibody resulted in activation of ERK lasting for 60 min, but failed to induce IL-2 production. In contrast, plate-bound anti-TCR antibody activated ERK over 4 h and induced IL-2. Furthermore, T cells treated with soluble anti-TCR antibody produced IL-2 when phorbol 12-myristate 13-acetate, which activates ERK, was present in the culture medium 2-6 h after the start of stimulation. Together, the data demonstrate the presence of a novel activation process following TCR stimulation that requires ERK-dependent regulation of c-Rel, a member of the NF-kappaB family.
Collapse
Affiliation(s)
- Toru Koike
- Program in Molecular Immunology, Institute of Molecular Medicine and Genetics, Medical College of Georgia, CA 2004, 1120 15th Street, Augusta, GA 30912-2600, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
587
|
Méndez-Samperio P, Ayala H, Vázquez A. NF-kappaB is involved in regulation of CD40 ligand expression on Mycobacterium bovis bacillus Calmette-Guérin-activated human T cells. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2003; 10:376-82. [PMID: 12738634 PMCID: PMC154977 DOI: 10.1128/cdli.10.3.376-382.2003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Interaction between CD40L (CD154) on activated T cells and its receptor CD40 on antigen-presenting cells has been reported to be important in the resolution of infection by mycobacteria. However, the mechanism(s) by which Mycobacterium bovis bacillus Calmette-Guérin (BCG) up-regulates membrane expression of CD40L molecules is poorly understood. This study was done to investigate the role of the nuclear factor kappaB (NF-kappaB) signaling pathway in the regulation of CD40L expression in human CD4(+) T cells stimulated with BCG. Specific pharmacologic inhibition of the NF-kappaB pathway revealed that this signaling cascade was required in the regulation of CD40L expression on the surface of BCG-activated CD4(+) T cells. These results were further supported by the fact that treatment of BCG-activated CD4(+) T cells with these pharmacological inhibitors significantly down-regulated CD40L mRNA. In this study, inhibitor kappaBalpha (IkappaBalpha) and IkappaBbeta protein production was not affected by the chemical protease inhibitors and, more importantly, BCG led to the rapid but transient induction of NF-kappaB activity. Our results also indicated that CD40L expression on BCG-activated CD4(+) T cells resulted from transcriptional up-regulation of the CD40L gene by a mechanism which is independent of de novo protein synthesis. Interestingly, BCG-induced activation of NF-kappaB and the increased CD40L cell surface expression were blocked by the protein kinase C (PKC) inhibitors 1-[5-isoquinolinesulfonyl]-2-methylpiperazine and salicylate, both of which block phosphorylation of IkappaB. Moreover, rottlerin a Ca(2+)-independent PKC isoform inhibitor, significantly down-regulated CD40L mRNA in BCG-activated CD4(+) T cells. These data strongly suggest that CD40L expression by BCG-activated CD4(+) T cells is regulated via the PKC pathway and by NF-kappaB DNA binding activity.
Collapse
Affiliation(s)
- Patricia Méndez-Samperio
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, IPN, Carpio y Plan de Ayala, México D. F. 11340, México.
| | | | | |
Collapse
|
588
|
Bommireddy R, Saxena V, Ormsby I, Yin M, Boivin GP, Babcock GF, Singh RR, Doetschman T. TGF-beta 1 regulates lymphocyte homeostasis by preventing activation and subsequent apoptosis of peripheral lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4612-22. [PMID: 12707339 DOI: 10.4049/jimmunol.170.9.4612] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TGF-beta1 plays an important role in the maintenance of immune homeostasis and self-tolerance. To determine the mechanism by which TGF-beta1 prevents autoimmunity we have analyzed T cell activation in splenic lymphocytes from TGF-beta1-deficient mice. Here we demonstrate that unlike wild-type splenic lymphocytes, those from Tgfb1(-/-) mice are hyporesponsive to receptor-mediated mitogenic stimulation, as evidenced by diminished proliferation and reduced IL-2 production. However, they have elevated levels of IFN-gamma and eventually undergo apoptosis. Receptor-independent stimulation of Tgfb1(-/-) T cells by PMA plus ionomycin induces IL-2 production and mitogenic response, and it rescues them from anergy. Tgfb1(-/-) T cells display decreased CD3 expression; increased expression of the activation markers LFA-1, CD69, and CD122; and increased cell size, all of which indicate prior activation. Consistently, mutant CD4(+) T cells have elevated intracellular Ca(2+) levels. However, upon subsequent stimulation in vitro, increases in Ca(2+) levels are less than those in wild-type cells. This is also consistent with the anergic phenotype. Together, these results demonstrate that the ex vivo proliferative hyporesponsiveness of Tgfb1(-/-) splenic lymphocytes is due to prior in vivo activation of T cells resulting from deregulated intracellular Ca(2+) levels.
Collapse
Affiliation(s)
- Ramireddy Bommireddy
- Department of Molecular Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | |
Collapse
|
589
|
Li ZW, Omori SA, Labuda T, Karin M, Rickert RC. IKK beta is required for peripheral B cell survival and proliferation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4630-7. [PMID: 12707341 DOI: 10.4049/jimmunol.170.9.4630] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
NF-kappaB activity in mammalian cells is regulated through the IkappaB kinase (IKK) complex, consisting of two catalytic subunits (IKKalpha and IKKbeta) and a regulatory subunit (IKKgamma). Targeted deletion of Ikkbeta results in early embryonic lethality, thus complicating the examination of IKKbeta function in adult tissues. Here we describe the role of IKKbeta in B lymphocytes made possible by generation of a mouse strain that expresses a conditional Ikkbeta allele. We find that the loss of IKKbeta results in a dramatic reduction in all peripheral B cell subsets due to associated defects in cell survival. IKKbeta-deficient B cells are also impaired in mitogenic responses to LPS, anti-CD40, and anti-IgM, indicating a general defect in the ability to activate the canonical NF-kappaB signaling pathway. These findings are consistent with a failure to mount effective Ab responses to T cell-dependent and independent Ags. Thus, IKKbeta provides a requisite role in B cell activation and maintenance and thus is a key determinant of humoral immunity.
Collapse
Affiliation(s)
- Zhi-Wei Li
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, Cancer Center, University of California-San Diego, La Jolla, CA 92093, USA
| | | | | | | | | |
Collapse
|
590
|
Lam TKT, Carpentier A, Lewis GF, van de Werve G, Fantus IG, Giacca A. Mechanisms of the free fatty acid-induced increase in hepatic glucose production. Am J Physiol Endocrinol Metab 2003; 284:E863-73. [PMID: 12676648 DOI: 10.1152/ajpendo.00033.2003] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The associations between obesity, insulin resistance, and type 2 diabetes mellitus are well documented. Free fatty acids (FFA), which are often elevated in obesity, have been implicated as an important link in these associations. Contrary to muscle glucose metabolism, the effects of FFA on hepatic glucose metabolism and the associated mechanisms have not been extensively investigated. It is still controversial whether FFA have substantial effects on hepatic glucose production, and the mechanisms responsible for these putative effects remain unknown. We review recent progress in this area and try to clarify controversial issues regarding the mechanisms responsible for the FFA-induced increase in hepatic glucose production in the postabsorptive state and during hyperinsulinemia.
Collapse
Affiliation(s)
- Tony K T Lam
- Department of Physiology and Medicine, Medical Science Building, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | | | |
Collapse
|
591
|
Bauer B, Jenny M, Fresser F, Uberall F, Baier G. AKT1/PKBalpha is recruited to lipid rafts and activated downstream of PKC isotypes in CD3-induced T cell signaling. FEBS Lett 2003; 541:155-62. [PMID: 12706837 DOI: 10.1016/s0014-5793(03)00287-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Protein kinase (PK) Ctheta and Akt/PKBalpha cooperate in T cell receptor/CD28-induced T cell signaling. We here demonstrate the recruitment of endogenous Akt1 and PKCtheta to lipid rafts in CD3-stimulated T cells. Further we show that Myr-PKCtheta mediates translocation of endogenous Akt1 to the plasma membrane as well as to lipid rafts, most likely explained by the observed complex formation of both protein kinases. In addition, in peripheral mouse T cells, the PKC inhibitor Gö6850 could partially block Akt1 activation in CD3-induced signaling, placing PKC isotype(s) upstream of Akt1. However, T cells derived from PKCtheta knockout mice were not impaired in CD3- or phorbol ester-induced Akt1 activity. Taken together, the results of this study give new insights into the functional link of Akt1 and PKCtheta in T cell signaling, demonstrating the co-recruitment of the two kinases and showing a novel pathway leading to Akt1 transactivation where PKC isotype(s) are involved but PKCtheta is not essential.
Collapse
Affiliation(s)
- Birgit Bauer
- Department of Medical Biology and Human Genetics, University of Innsbruck, Austria
| | | | | | | | | |
Collapse
|
592
|
Wells AD, Liu QH, Hondowicz B, Zhang J, Turka LA, Freedman BD. Regulation of T cell activation and tolerance by phospholipase C gamma-1-dependent integrin avidity modulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4127-33. [PMID: 12682243 DOI: 10.4049/jimmunol.170.8.4127] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Ag receptor engagement without costimulation induces a tolerant state in CD4(+) T cells termed anergy. Anergic CD4(+) T cells are primarily characterized by the inability to produce IL-2, but the biochemical basis for this functional defect is not completely understood. We demonstrate that primary CD4(+) T cells anergized by costimulatory blockade exhibit impaired TCR-coupled phospholipase C (PLC)gamma-1 activation. This defect is associated with the marked reduction of multiple downstream signaling events required for IL-2 transcription, including mobilization of intracellular Ca(2+) and activation of the mitogen-activated protein kinase cascade. We also found that primary anergic CD4(+) T cells fail entirely to modulate their integrin binding avidity in response to TCR stimulation. Integrin avidity modulation is required for full T cell activation and effector function, and as we show in this study, is completely dependent upon PLCgamma-1 activity. Finally, analogs that mimic the actions of diacylglycerol and inositol 1,4,5-triphosphate, the immediate products of PLCgamma-1 activity, restored integrin avidity modulation and IL-2 production by anergic T cells. Thus, deficient coupling of PLCgamma-1 to the TCR appears to be a central biochemical defect that could potentially account for the failure of multiple functional responses in primary anergic CD4(+) T cells.
Collapse
Affiliation(s)
- Andrew D Wells
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
593
|
Lee SJE, Hori Y, Chakraborty AK. Low T cell receptor expression and thermal fluctuations contribute to formation of dynamic multifocal synapses in thymocytes. Proc Natl Acad Sci U S A 2003; 100:4383-8. [PMID: 12671067 PMCID: PMC153563 DOI: 10.1073/pnas.0630563100] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2002] [Accepted: 01/29/2003] [Indexed: 11/18/2022] Open
Abstract
Mature T cell activation and selection of immature T cells (thymocytes) are both initiated by binding of T cell receptor (TCR) molecules on the surface of T cells to MHC peptide (MHCp) molecules on the surface of antigen-presenting cells. Recent experiments have shown that the spatial pattern of receptors and ligands in the intercellular junction (synapse) is different during thymocyte selection compared with mature T cell activation. Using a statistical mechanical model, we show that lower TCR expression in thymocytes contributes to effecting these differences. An analogy with the phase behavior of simple fluids helps clarify how, for low TCR expression, thermal fluctuations lead to the dynamic synapse patterns observed for thymocytes. We suggest that a different synapse pattern resulting from lower TCR expression, which could mediate differential signaling, may be the reason why TCR expression level is low in thymocytes.
Collapse
Affiliation(s)
- Sung-Joo E Lee
- Biophysics Graduate Group, University of California, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
594
|
Zheng Y, Vig M, Lyons J, Van Parijs L, Beg AA. Combined deficiency of p50 and cRel in CD4+ T cells reveals an essential requirement for nuclear factor kappaB in regulating mature T cell survival and in vivo function. J Exp Med 2003; 197:861-74. [PMID: 12668645 PMCID: PMC2193891 DOI: 10.1084/jem.20021610] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Signaling pathways involved in regulating T cell proliferation and survival are not well understood. Here we have investigated a possible role of the nuclear factor (NF)-kappaB pathway in regulating mature T cell function by using CD4+ T cells from p50-/- cRel-/- mice, which exhibit virtually no inducible kappaB site binding activity. Studies with these mice indicate an essential role of T cell receptor (TCR)-induced NF-kappaB in regulating interleukin (IL)-2 expression, cell cycle entry, and survival of T cells. Our results further indicate that NF-kappaB regulates TCR-induced expression of antiapoptotic Bcl-2 family members. Strikingly, retroviral transduction of CD4+ T cells with the NF-kappaB-inducing IkappaB kinase beta showed that NF-kappaB activation is not only necessary but also sufficient for T cell survival. In contrast, our results indicate a lack of involvement of NF-kappaB in both IL-2 and Akt-induced survival pathways. In vivo, p50-/- cRel-/- mice showed impaired superantigen-induced T cell responses as well as decreased numbers of effector/memory and regulatory CD4+ T cells. These findings provide the first demonstration of a role for NF-kappaB proteins in regulating T cell function in vivo and establish a critically important function of NF-kappaB in TCR-induced regulation of survival.
Collapse
Affiliation(s)
- Ye Zheng
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | | | | | | |
Collapse
|
595
|
Abstract
The distinct protein kinase C (PKC) multigene family (PKC gene module) is known to be the 'classic' intracellular receptor for mitogenic phorbol esters, and it is widely accepted in the scientific community that the 'PKC effect' is essential in activation, differentiation, adhesion and motility, as well as in cellular survival, of T cells. Nevertheless, the first concepts about PKC isotype heterogeneity of cellular localization and function emerged only recently, when the PKC-theta pathways were mapped to critical signaling networks that control T cell receptor (TCR)/CD3-dependent interleukin (IL)-2 production and proliferation in T lymphocytes. This review summarizes the current knowledge about T cell expressed PKC gene products, their known and/or suspected regulation and cellular effector pathways, as well as physiological functions in T lymphocytes (as determined by molecular cell biology and ongoing mouse genetic studies). Given PKCs integral role in T cell function but today's very fragmentary molecular understanding of directly PKC-mediated effector functions in transmembrane signaling, a 'molecular biosystematics' approach is suggested to resolve the isotype-selective functions of this PKC gene family. Such an approach has to be based not only on genomic/cytogenetic analysis to establish its genetic relationships but also on biochemical/cell biology and genetic studies to resolve its functional diversity and, ultimately, nonredundant roles in real T cell physiology.
Collapse
Affiliation(s)
- Gottfried Baier
- Institute of Medical Biology and Human Genetics, University of Innsbruck, Austria.
| |
Collapse
|
596
|
Abstract
T lymphocytes play a key role in immunity by distinguishing self from nonself peptide antigens and regulating both the cellular and humoral arms of the immune system. Acquired, antigen-specific unresponsiveness is an important mechanism by which T cell responses to antigen are regulated in vivo. Clonal anergy is the term that describes T cell unresponsiveness at the cellular level. Anergic T cells do not proliferate or secrete interleukin (IL)-2 in response to appropriate antigenic stimulation. However, anergic T cells express the IL-2 receptor, and anergy can be broken by exogenous IL-2. Anergy can be induced by submitogenic exposure to peptide antigen in the absence of a costimulatory signal provided by soluble cytokines or by interactions between costimulatory receptors on T cells and counter-receptors on antigen-presenting cells. The molecular events that mediate the induction and maintenance of T cell anergy are the focus of this review. The molecular consequences of CD28-B7 interaction are discussed as a model for the costimulatory signal that leads to T cell activation rather than the induction of anergy.
Collapse
Affiliation(s)
- Leonard J Appleman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | |
Collapse
|
597
|
Abstract
Adapter proteins regulate leukocyte signal transduction through recruitment of effector molecules to multiprotein complexes. Recent studies in Adhesion and Degranulation promoting Adapter Protein (ADAP)-deficient mice have established that the cytoplasmic phosphoprotein ADAP is required for optimal, mature T-cell proliferation. Furthermore, ADAP plays a key role in T-cell antigen receptor (TCR)-mediated 'inside out' signaling leading to integrin activation and to enhanced cellular adhesion to integrin ligands. ADAP associates physically with molecules known to play roles in the regulation of TCR-stimulated actin polymerization. These associations support the hypothesis that ADAP functions in actin cytoskeletal reorganization leading to cellular adhesion and activation.
Collapse
Affiliation(s)
- Erik J Peterson
- Division of Rheumatology, Department of Medicine and the Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
598
|
Trautmann A, Revy P, Donnadieu E, Bismuth G. [Immunological synapses and neuronal synapses]. Med Sci (Paris) 2003; 19:429-36. [PMID: 12836215 DOI: 10.1051/medsci/2003194429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The interface between two cells from the immune system has recently been coined "immunological synapse". The authors review recent findings concerning the structure of the synapse formed between T lymphocytes and antigen-presenting cells. T cells can be part of different synapses, depending on the antigen-presenting cell (B cell hybridoma, proteo-lipid bilayer, macrophage, dendritic cell). The synapse formed with dendritic cells is discussed in more details. A comparison is made with the synapses from the nervous system. Several parallel questions are discussed: how receptors can be clustered, what is the influence of synapse functioning on the structure of the synapse. It is suggested that in both cases two modes of communication exist in parallel: direct cell-cell contacts and soluble mediators, neurotransmitters in one case, putative immunotransmitters in the other.
Collapse
Affiliation(s)
- Alain Trautmann
- Département de Biologie Cellulaire, Institut Cochin, Inserm U.567, Cnrs UMR 8104, 22, rue Méchain, 75014 Paris, France.
| | | | | | | |
Collapse
|
599
|
Abstract
Much progress has been made in understanding the function of protein kinase C-theta (PKCtheta) in the immune system since this Ca2+-independent PKC isotype was isolated in 1993 as an enzyme that is highly expressed in T lymphocytes and in muscle cells. Biochemical and genetic approaches revealed that, while dispensable for T-cell development, PKCtheta is required for the activation of mature T cells and for interleukin (IL)-2 production. This deficiency results from impaired receptor-induced stimulation of the transcription factors AP-1 and NF-kappaB. PKCtheta integrates T-cell receptor (TCR)/CD28 costimulatory signals, which are essential for productive T-cell activation and, most likely, for prevention of T-cell anergy. A unique property of PKCtheta is its highly selective recruitment to the central supramolecular activation complex (cSMAC) region of the immunological synapse (IS) in antigen-stimulated T cells. Our work revealed that this highly selective localization is not entirely dependent on phospholipase C (PLC) activity and diacylglycerol (DAG) production. Instead, a novel signaling pathway that requires functional Vav1, phosphatidylinositol 3-kinase (PI3-K), the small GTPase Rac and actin cytoskeleton reorganization regulates the localization and, perhaps, activation of PKCtheta. PKCtheta also provides a survival signal, which protects T cells from apoptosis. Additional work is required to identify the immediate targets of PKCtheta and its immune functions in vivo. This work is likely to validate PKCtheta as an attractive drug target.
Collapse
Affiliation(s)
- Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, San Diego, CA 92121, USA.
| | | |
Collapse
|
600
|
Seminario MC, Wange RL. Lipid phosphatases in the regulation of T cell activation: living up to their PTEN-tial. Immunol Rev 2003; 192:80-97. [PMID: 12670397 DOI: 10.1034/j.1600-065x.2003.00013.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The initiating events associated with T activation in response to stimulation of the T cell antigen receptor (TCR) and costimulatory receptors, such as CD28, are intimately associated with the enzymatically catalyzed addition of phosphate not only to key tyrosine, threonine and serine residues in proteins but also to the D3 position of the myo-inositol ring of phosphatidylinositol (PtdIns). This latter event is catalyzed by the lipid kinase phosphoinositide 3-kinase (PI3K). The consequent production of PtdIns(3,4)P2 and PtdIns(3,4,5)P3 serves both to recruit signaling proteins to the plasma membrane and to induce activating conformational changes in proteins that contain specialized domains for the binding of these phospholipids. The TCR signaling proteins that are subject to regulation by PI3K include Akt, phospholipase Cgamma1 (PLCgamma1), protein kinase C zeta (PKC-zeta), Itk, Tec and Vav, all of which play critical roles in T cell activation. As is the case for phosphorylation of protein substrates, the phosphorylation of PtdIns is under dynamic regulation, with the D3 phosphate being subject to hydrolysis by the 3-phosphatase PTEN (phosphatase and tensin homolog deleted on chromosome 10), thereby placing PTEN in direct opposition to PI3K. In this review we consider recent data concerning how PTEN may act in regulating the process of T cell activation.
Collapse
Affiliation(s)
- Maria-Cristina Seminario
- Laboratory of Cellular and Molecular Biology, National Institutes on Aging/IRP/NIH/DHHS, Baltimore, MD 21224, USA.
| | | |
Collapse
|