101
|
Freebern WJ, Haggerty CM, Montano I, McNutt MC, Collins I, Graham A, Chandramouli GVR, Stewart DH, Biebuyck HA, Taub DD, Gardner K. Pharmacologic profiling of transcriptional targets deciphers promoter logic. THE PHARMACOGENOMICS JOURNAL 2005; 5:305-23. [PMID: 16044165 DOI: 10.1038/sj.tpj.6500325] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The blueprint for cellular diversity and response to environmental change is encoded in the cis-acting regulatory sequences of most genes. Deciphering this 'cis-regulatory code' requires multivariate data sets that examine how these regions coordinate transcription in response to diverse environmental stimuli and therapeutic treatments. We describe a transcriptional approach that profiles the activation of multiple transcriptional targets against combinatorial arrays of therapeutic and signal transducing agents. Application of this approach demonstrates how cis-element composition and promoter context combine to influence transcription downstream of mitogen-induced signaling networks. Computational dissection of these transcriptional profiles in activated T cells uncovers a novel regulatory synergy between IGF-1 and CD28 costimulation that modulates NF-kappaB and AP1 pathways through signaling cascades sensitive to cyclosporin A and wortmannin. This approach provides a broader view of the hierarchical signal integration governing gene expression and will facilitate a practical design of combinatorial therapeutic strategies for exploiting critical control points in transcriptional regulation.
Collapse
Affiliation(s)
- W J Freebern
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, The Advanced Technology Center, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Kim J, Myers AC, Chen L, Pardoll DM, Truong-Tran QA, Lane AP, McDyer JF, Fortuno L, Schleimer RP. Constitutive and inducible expression of b7 family of ligands by human airway epithelial cells. Am J Respir Cell Mol Biol 2005; 33:280-9. [PMID: 15961727 PMCID: PMC2715317 DOI: 10.1165/rcmb.2004-0129oc] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Activated T cells have been implicated in chronic rhinosinusitis (CRS) and asthma and physically interact with epithelial cells in the airways. We now report that human airway epithelial cells display significant constitutive cell-surface expression of costimulatory ligands, B7-H1, B7-H2, B7-H3, and B7-DC. Expression of B7-H1 and B7-DC was selectively induced by stimulation of either BEAS2B or primary nasal epithelial cells (PNEC) with interferon (IFN)-gamma (100 ng/ml). The combination of IFN-gamma and tumor necrosis factor-alpha (100 ng/ml) selectively induced expression better than IFN-gamma alone. Fluticasone treatment (10(-7) M) reduced the baseline expression and inhibited the induction of B7-H1 and B7-DC in BEAS2B cells. In vitro exposure of PNEC to IFN-gamma also resulted in selective induction of B7-H1 and B7-DC. Monoclonal antibody blockade of B7-H1 or B7-DC enhanced IFN-gamma expression by purified T cells in co-culture experiments, suggesting that these two B7 homologs inhibit T cell responses at the mucosal surface. Immunohistochemical staining of human sinonasal surgical tissue confirmed the presence of B7-H1, B7-H2, and B7-H3 in the epithelial cell layer, especially in samples from patients diagnosed with Samter's Triad, a severe form of CRS. Real-time PCR analysis of sinonasal tissue revealed elevated levels of B7-H1 and B7-DC in CRS compared with controls. These results demonstrate that epithelial cells express functional B7 costimulatory molecules and that expression of selected B7 family members is inducible in vitro and in vivo. Epithelial B7 homologs could play a role in regulation of lymphocytic activity at mucosal surfaces.
Collapse
Affiliation(s)
- Jean Kim
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Rm 3A65A, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Cejas PJ, Carlson LM, Zhang J, Padmanabhan S, Kolonias D, Lindner I, Haley S, Boise LH, Lee KP. Protein kinase C betaII plays an essential role in dendritic cell differentiation and autoregulates its own expression. J Biol Chem 2005; 280:28412-23. [PMID: 15917249 DOI: 10.1074/jbc.m500345200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Dendritic cells (DC) arise from a diverse group of hematopoietic progenitors and have marked phenotypic and functional heterogeneity. The signal transduction pathways that regulate the ability of progenitors to undergo DC differentiation, as well as the specific characteristics of the resulting DC, are only beginning to be characterized. We have found previously that activation of protein kinase C (PKC) by cytokines or phorbol esters drives normal human CD34(+) hematopoietic progenitors and myeloid leukemic blasts (KG1, K562 cell lines, and primary patient blasts) to differentiate into DC. We now report that PKC activation is also required for cytokine-driven DC differentiation from monocytes. Of the cPKC isoforms, only PKC-betaII was consistently activated by DC differentiation-inducing stimuli in normal and leukemic progenitors. Transfection of PKC-betaII into the differentiation-resistant KG1a subline restored the ability to undergo DC differentiation in a signal strength-dependent fashion as follows: 1) by development of characteristic morphology; 2) the up-regulation of DC surface markers; 3) the induction of expression of the NFkappaB family member Rel B; and 4) the potent ability to stimulate allo-T cells. Most unexpectedly, the restoration of PKC-betaII signaling in KG1a was not directly due to overexpression of the transfected classical PKC (alpha, betaII, or gamma) but rather through induction of endogenous PKC-beta gene expression by the transfected classical PKC. The mechanism of this positive autoregulation involves up-regulation of PKC-beta promoter activity by constitutive PKC signaling. These findings indicate that the regulation of PKC-betaII expression and signaling play critical roles in mediating progenitor to DC differentiation.
Collapse
Affiliation(s)
- Pedro J Cejas
- Department of Microbiology and Immunology and Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, Miami, Florida 33156, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Kohm AP, Podojil JR, Williams JS, McMahon JS, Miller SD. CD28 regulates glucocorticoid-induced TNF receptor family-related gene expression on CD4+ T cells via IL-2-dependent mechanisms. Cell Immunol 2005; 235:56-64. [PMID: 16137665 DOI: 10.1016/j.cellimm.2005.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2004] [Revised: 06/30/2005] [Accepted: 07/14/2005] [Indexed: 11/19/2022]
Abstract
The glucocorticoid-induced TNF-related gene receptor (GITR) is the newest member of the costimulatory molecule family and is expressed on both resting CD4+CD25+ regulatory T (T(R)) cells and activated CD4+ T cells. We investigated the endogenous mechanisms that regulate GITR expression on both T(R) and CD4+ T cells, as well as the functional interaction between GITR and other costimulatory molecules. CD28 stimulation increased GITR expression on both T(R) and CD4+ T cells via IL-2-dependent mechanisms. In addition, ligation of GITR and/or CD28 increased the level of CD4+ T cell proliferation and effector function under both APC-dependent and -independent conditions, suggesting that these costimulatory molecules cooperate to regulate CD4+ T cell activation and function by directly signaling to the CD4+ T cell. Thus, GITR may serve opposing functional roles on CD4+ T(R) and effector cells and alterations in GITR expression and/or function may tip the balance between immune tolerance and effector function.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- CD28 Antigens/physiology
- Cells, Cultured
- Gene Expression Regulation/immunology
- Glucocorticoid-Induced TNFR-Related Protein
- Glucocorticoids/physiology
- Interleukin-2/genetics
- Interleukin-2/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Multigene Family/immunology
- RNA, Messenger/metabolism
- Receptors, Interleukin-2/deficiency
- Receptors, Interleukin-2/genetics
- Receptors, Interleukin-2/metabolism
- Receptors, Nerve Growth Factor/biosynthesis
- Receptors, Nerve Growth Factor/genetics
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/genetics
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/physiology
Collapse
Affiliation(s)
- Adam P Kohm
- Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
105
|
Nishiyama S, Manabe N, Kubota Y, Ohnishi H, Kitanaka A, Tokuda M, Taminato T, Ishida T, Takahara J, Tanaka T. Cyclosporin A inhibits the early phase of NF-κB/RelA activation induced by CD28 costimulatory signaling to reduce the IL-2 expression in human peripheral T cells. Int Immunopharmacol 2005; 5:699-710. [PMID: 15710339 DOI: 10.1016/j.intimp.2004.11.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2004] [Revised: 10/25/2004] [Accepted: 11/29/2004] [Indexed: 11/19/2022]
Abstract
Cyclosporin A (CsA) inhibits interleukin (IL)-2 production, activation and proliferation of human peripheral T cells (HPTC) costimulated with simultaneous engagement of T cell receptor (TCR)/CD3 and CD28. We demonstrated that 10 ng/ml CsA, which reduced the proliferation of HPTC costimulated with anti-CD3 and anti-CD28 by half, prevented NF-AT and NF-kappaB from migrating into the nucleus. Whereas CsA added even 30 min after the costimulation caused NF-AT to remain in the cytoplasm, the delayed addition of CsA could not prevent NF-kappaB from translocating into the nucleus. CsA, which was added to HPTC simultaneously with the engagement of both CD3 and CD28 or the 1-h-delayed engagement of CD28 after prior TCR/CD3-triggering, inhibited NF-kappaB p65/RelA from binding to the target DNA fragment, followed by reduction of HPTC proliferation in response to the costimulation. When CsA was added 30 min after the delayed engagement of CD28 following the prior engagement of TCR/CD3, these inhibitory effects were diminished. Antisense NF-kappaB p65/RelA oligonucleotides inhibited p65/RelA mRNA expression, diminished IL-2 mRNA expression in the costimulated HPTC and reduced HPTC proliferation to the same extent as CsA added simultaneously with the costimulation. The CsA- or antisense p65/RelA oligonucleotide-induced reduction in the proliferation of costimulated HPTC was overcome by the addition of exogenous IL-2. These findings indicate that the major effects of CsA on the early phase of CD28-mediated costimulation in the presence of TCR/CD3 signaling are to inhibit NF-kappaB/RelA from translocating into the nucleus and binding to the target DNA sequence in the IL-2 gene promoter region, which induces IL-2 expression leading to HPTC proliferation.
Collapse
Affiliation(s)
- Susumu Nishiyama
- First Department of Internal Medicine, Faculty of Medicine, Kagawa University, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Dhanireddy KK, Xu H, Mannon RB, Hale DA, Kirk AD. The clinical application of monoclonal antibody therapies in renal transplantation. Expert Opin Emerg Drugs 2005. [DOI: 10.1517/14728214.9.1.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
107
|
Zheng P, Liu Y. Co-stimulatory molecules B7-1 and B7-2 as experimental therapeutic targets. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.3.1.93] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
108
|
Minguillón J, Morancho B, Kim SJ, López-Botet M, Aramburu J. Concentrations of cyclosporin A and FK506 that inhibit IL-2 induction in human T cells do not affect TGF-beta1 biosynthesis, whereas higher doses of cyclosporin A trigger apoptosis and release of preformed TGF-beta1. J Leukoc Biol 2005; 77:748-58. [PMID: 15716327 DOI: 10.1189/jlb.0904503] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cyclosporin A (CsA) and FK506 suppress T cell activation by inhibiting calcineurin and the calcineurin-dependent transcription factors nuclear factor of activated T cells (NFATc), which are central regulators of T cell function. It was reported that CsA up-regulated the transcription of transforming growth factor-beta1 (TGF-beta1) in lymphocytes and other cells and activated its promoter in A549 lung carcinoma cells, but the mechanisms involved are poorly understood, and it is unclear whether calcineurin plays any role. We have studied the regulation of TGF-beta1 in normal human lymphocytes and cell lines. In Jurkat T cells, the TGF-beta1 promoter was activated by calcineurin and NFATc and inhibited by CsA and FK506. However, the promoter was insensitive to both drugs in A549 cells. In human T cells preactivated with phytohemagglutinin, biosynthesis of TGF-beta1, induced by the T cell receptor (TCR) or the TGF-beta receptor, was not substantially affected by CsA and FK506 concentrations (< or = 1 microM) that effectively inhibited interleukin-2 production. However, pretreatment of fresh lymphocytes with CsA or FK506 during primary TCR stimulation reduced their production of TGF-beta1 during secondary TCR activation. Finally, high concentrations of CsA (10 microM), in the range attained in vivo in experiments in rodents, caused apoptosis in human T cells and the release of preformed, bioactive TGF-beta1. These effects are unlikely to owe to calcineurin inhibition, as they were not observed with FK506. Our results indicate that CsA and FK506 are not general inducers of TGF-beta1 biosynthesis but can cause different effects on TGF-beta1 depending on the cell type and concentrations used.
Collapse
Affiliation(s)
- Jordi Minguillón
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Carrer Dr Aiguader 80, 08003 Barcelona, Spain
| | | | | | | | | |
Collapse
|
109
|
Nam KO, Kang H, Shin SM, Cho KH, Kwon B, Kwon BS, Kim SJ, Lee HW. Cross-Linking of 4-1BB Activates TCR-Signaling Pathways in CD8+ T Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2005; 174:1898-905. [PMID: 15699116 DOI: 10.4049/jimmunol.174.4.1898] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cross-linking of 4-1BB, a member of the TNFR family, increased tyrosine phosphorylation of TCR-signaling molecules such as CD3epsilon, CD3zeta, Lck, the linker for activation of T cells, and SH2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76). In addition, incubation of activated CD8+ T cells with p815 cells expressing 4-1BBL led to redistribution of the lipid raft domains and Lck, protein kinase C-theta;, SLP-76, and phospholipase C-gamma1 (PLC-gamma1) on the T cell membranes to the areas of contact with the p815 cells and recruitment of 4-1BB, TNFR-associated factor 2, and phospho-tyrosine proteins to the raft domains. 4-1BB ligation also caused translocation of TNFR-associated factor 2, protein kinase C-theta;, PLC-gamma1, and SLP-76 to detergent-insoluble compartments in the CD8+ T cells, and cross-linking of 4-1BB increased intracellular Ca2+ levels apparently by activating PLC-gamma1. The redistribution of lipid rafts and Lck, as well as translocation of PLC-gamma1, and degradation of IkappaB-alpha in response to 4-1BB were inhibited by disrupting the formation of lipid rafts with methyl-beta-cyclodextrin. These findings demonstrate that 4-1BB is a T cell costimulatory receptor that activates TCR-signaling pathways in CD8+ T cells.
Collapse
MESH Headings
- 4-1BB Ligand
- Animals
- Antibodies, Monoclonal/metabolism
- Antigens, CD
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/enzymology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Calcium/metabolism
- Cetomacrogol
- Cyclosporine/pharmacology
- Detergents
- Growth Inhibitors/pharmacology
- Intracellular Fluid/metabolism
- Ligands
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/immunology
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism
- Membrane Microdomains/enzymology
- Membrane Microdomains/metabolism
- Mice
- Mice, Inbred BALB C
- Phosphotyrosine/metabolism
- Protein Transport/immunology
- Pyrimidines/pharmacology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/physiology
- Receptors, Nerve Growth Factor/antagonists & inhibitors
- Receptors, Nerve Growth Factor/immunology
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Nerve Growth Factor/physiology
- Receptors, Tumor Necrosis Factor/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor/physiology
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Solubility
- TNF Receptor-Associated Factor 2/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 9
- Tumor Necrosis Factor-alpha/metabolism
- beta-Cyclodextrins/pharmacology
Collapse
Affiliation(s)
- Kyung-Ok Nam
- Immunomodulation Research Center and Graduate Program in Immunology and Biomedicine, University of Ulsan, Ulsan, Korea
| | | | | | | | | | | | | | | |
Collapse
|
110
|
Li Q, Ozer H, Lindner I, Lee KP, Kharfan-Dabaja MA. Protein Kinase C Blockade Inhibits Differentiation of Myeloid Blasts into Dendritic Cells by Calcium Ionophore A23187. Int J Hematol 2005; 81:131-7. [PMID: 15765781 DOI: 10.1532/ijh97.na0405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Direct differentiation of myeloid leukemia blasts into antigen-presenting dendritic cells (DCs) for use as cellular vaccines is unique in that identification of tumor-specific antigens may not be necessary because the antigens should already be endogenously expressed. We hypothesized that signaling through protein kinase C (PKC) is required for differentiation of HL-60 promyeloblasts into DCs upon stimulation with calcium ionophore A23187. To demonstrate the inhibitory effect of PKC blockade, we pretreated HL-60 myeloid blasts with the protein kinase inhibitor bisindolylmaleimide I (Bis-1) for 24 hours and then treated the cells with calcium ionophore A23187 for an additional 24 hours. Controls consisted of HL-60 blasts treated with A23187, Bis-1 alone, or media. We noted that blasts cultured in media, Bis-1, or Bis-1 then A23187 did not develop the morphologic and phenotypic DC characteristics, up-regulate Rel B, or activate allogeneic T-cells. Our findings suggested that PKC blockade inhibits morphologic, phenotypic, and functional differentiation of HL-60 promyeloblasts into antigen-presenting DCs. Our findings supported the role of PKC as an obligatory pathway for calcium ionophore A23187-induced differentiation of HL-60 myeloblasts into antigen-presenting DCs.
Collapse
Affiliation(s)
- Qian Li
- Section of Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | |
Collapse
|
111
|
Schleyer V, Landthaler M, Szeimies RM. Novel pharmacological approaches in the treatment of psoriasis. J Eur Acad Dermatol Venereol 2005; 19:1-20. [PMID: 15649186 DOI: 10.1111/j.1468-3083.2004.01070.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Progress in the understanding of psoriasis as a T-cell mediated inflammatory disease has led to the development of new immunomodulatory therapies. Currently the main focus is on the so-called biologics (or biological agents), including fusion proteins, monoclonal antibodies, cytokines and selective receptors. They mainly target single steps in the complex cascade of humoral and cellular inflammatory immuno-mechanisms that finally lead to the accelerated growth of epidermal and vascular cells in the psoriatic lesions. The most promising and advanced biological agents are discussed along with their influence on the critical pathophysiological steps in psoriasis, including depletion of T cells, blockade of initial T-cell activation and T-cell receptor (TCR) stimulation, blockade of costimulatory signals and T-cell proliferative signals as well as restoration of the T helper type 1 (Th1)/Th2 balance by diminishing type 1 cytokines and administration of type 2 cytokines. In addition to the biological agents, further development of 'classical' dermatological therapies, such as retinoids, or the discovery of new indications for non-dermatological agents contribute to the novel pharmacological approaches in the treatment of psoriasis.
Collapse
Affiliation(s)
- V Schleyer
- Department of Dermatology at the University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053 Regensburg, Germany
| | | | | |
Collapse
|
112
|
Kharfan-Dabaja MA, Ayala E, Lindner I, Cejas PJ, Bahlis NJ, Kolonias D, Carlson LM, Lee KP. Differentiation of acute and chronic myeloid leukemic blasts into the dendritic cell lineage: analysis of various differentiation-inducing signals. Cancer Immunol Immunother 2005; 54:25-36. [PMID: 15693136 PMCID: PMC11032788 DOI: 10.1007/s00262-004-0562-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2003] [Accepted: 04/09/2004] [Indexed: 11/26/2022]
Abstract
PURPOSE Ex vivo differentiation of myeloid leukemic blasts into dendritic cells (DCs) holds significant promise for use as cellular vaccines, as they may present a constellation of endogenously expressed known and unknown leukemia antigens to the immune system. Although variety of stimuli can drive leukemia --> DC differentiation in vitro, these blast-derived DCs typically have aberrant characteristics compared with DCs generated from normal progenitors by the same stimuli. It is not clear whether this is due to underlying leukemogenic mechanisms (e.g., specific oncogenes), genetic defects, stage of maturation arrest, defects in cytokine receptor expression or signal transduction pathways, or whether different stimuli themselves induce qualitatively dissimilar DC differentiation. METHODS To assess what factors may contribute to aberrant leukemic blast --> DC differentiation, we have examined how the same leukemic blasts (AML and CML) respond to different DC differentiation signals--including extracellular (the cytokine combination GM-CSF + TNF-alpha + IL-4) and intracellular (the protein kinase C agonist PMA, the calcium ionophore A23187, and the combination of PMA plus A23187) stimuli. RESULTS We have found that the same leukemic blasts will develop qualitatively different sets of DC characteristics in response to differing stimuli, although no stimuli consistently induced all of the characteristic DC features. There were no clear differences in the responses relative to specific oncogene expression or stage of maturation arrest (AML vs CML). Signal transduction agonists that bypassed membrane receptors/proximal signaling (in particular, the combination of PMA and A23187) consistently induced the greatest capability to activate T cells. Interestingly, this ability did not clearly correlate with expression of MHC/costimulatory ligands. CONCLUSIONS Our findings suggest that signal transduction may play an important role in the aberrant DC differentiation of leukemic blasts, and demonstrate that direct activation of PKC together with intracellular calcium signaling may be an effective method for generating immunostimulatory leukemia-derived DCs.
Collapse
MESH Headings
- Antigens, Surface/genetics
- Blast Crisis/genetics
- Blast Crisis/immunology
- Blast Crisis/metabolism
- Cell Differentiation/physiology
- Cell Line
- Cell Lineage/genetics
- Cell Lineage/immunology
- Cell Proliferation
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Phenotype
- Signal Transduction/genetics
- Signal Transduction/immunology
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Mohamed A. Kharfan-Dabaja
- Division of Hematology/Oncology, Department of Medicine, University of Miami, Miami, FL 33136 USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136 USA
| | - Ernesto Ayala
- Division of Hematology/Oncology, Department of Medicine, University of Miami, Miami, FL 33136 USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136 USA
| | - Inna Lindner
- Department of Microbiology and Immunology, University of Miami, Miami, FL 33136 USA
| | - Pedro J. Cejas
- Department of Microbiology and Immunology, University of Miami, Miami, FL 33136 USA
| | - Nizar J. Bahlis
- Division of Hematology/Oncology, Department of Medicine, University of Miami, Miami, FL 33136 USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136 USA
| | - Despina Kolonias
- Department of Microbiology and Immunology, University of Miami, Miami, FL 33136 USA
| | - Louise M. Carlson
- Department of Microbiology and Immunology, University of Miami, Miami, FL 33136 USA
| | - Kelvin P. Lee
- Division of Hematology/Oncology, Department of Medicine, University of Miami, Miami, FL 33136 USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136 USA
- Department of Microbiology and Immunology, University of Miami, Miami, FL 33136 USA
- Department of Microbiology and Immunology, University of Miami School of Medicine, Papanicolaou Bldg., Rm. 211, 1550 NW 10th Ave., Miami, FL 33136 USA
| |
Collapse
|
113
|
Why inhibitors of mammalian target of rapamycin will be important in organ transplantation. Curr Opin Organ Transplant 2004. [DOI: 10.1097/01.mot.0000146560.58398.e4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
114
|
Smith P, Walsh CM, Mangan NE, Fallon RE, Sayers JR, McKenzie ANJ, Fallon PG. Schistosoma mansoni worms induce anergy of T cells via selective up-regulation of programmed death ligand 1 on macrophages. THE JOURNAL OF IMMUNOLOGY 2004; 173:1240-8. [PMID: 15240716 DOI: 10.4049/jimmunol.173.2.1240] [Citation(s) in RCA: 159] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Infectious pathogens can selectively stimulate activation or suppression of T cells to facilitate their survival within humans. In this study we demonstrate that the trematode parasite Schistosoma mansoni has evolved with two distinct mechanisms to suppress T cell activation. During the initial 4- to 12-wk acute stages of a worm infection both CD4(+) and CD8(+) T cells are anergized. In contrast, infection with male and female worms induced T cell anergy at 4 wk, which was replaced after egg laying by T cell suppression via a known NO-dependent mechanism, that was detected for up to 40 wk after infection. Worm-induced anergy was mediated by splenic F4/80(+) macrophages (Mphi) via an IL-4-, IL-13-, IL-10-, TGF-beta-, and NO-independent, but cell contact-dependent, mechanism. F4/80(+) Mphi isolated from worm-infected mice were shown to induce anergy of naive T cells in vitro. Furthermore, naive Mphi exposed to live worms in vitro also induced anergy in naive T cells. Flow cytometry on in vivo and in vitro worm-modulated Mphi revealed that of the family of B7 costimulatory molecules, only programmed death ligand 1 (PD-L1) was selectively up-regulated. The addition of inhibitory mAb against PD-L1, but not PD-L2, to worm-modulated Mphi completely blocked the ability of these cells to anergize T cells. These data highlight a novel mechanism through which S. mansoni worms have usurped the natural function of PD-L1 to reduce T cell activation during early acute stages of infection before the subsequent emergence of egg-induced T cell suppression in the chronic stages of infection.
Collapse
Affiliation(s)
- Philip Smith
- Department of Biochemistry, Trinity College, Dublin, Ireland
| | | | | | | | | | | | | |
Collapse
|
115
|
Maus MV, Kovacs B, Kwok WW, Nepom GT, Schlienger K, Riley JL, Allman D, Finkel TH, June CH. Extensive replicative capacity of human central memory T cells. THE JOURNAL OF IMMUNOLOGY 2004; 172:6675-83. [PMID: 15153483 DOI: 10.4049/jimmunol.172.11.6675] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To characterize the replicative capacity of human central memory (T(CM)) CD4 T cells, we have developed a defined culture system optimized for the ex vivo expansion of Ag-specific CD4(+) T cells. Artificial APCs (aAPCs) consisting of magnetic beads coated with Abs to HLA class II and a costimulatory Ab to CD28 were prepared; peptide-charged HLA class II tetramers were then loaded on the beads to provide Ag specificity. Influenza-specific DR*0401 CD4 T(CM) were isolated from the peripheral blood of normal donors by flow cytometry. Peptide-loaded aAPC were not sufficient to induce resting CD4 T(CM) to proliferate. In contrast, we found that the beads efficiently promoted the growth of previously activated CD4 T(CM) cells, yielding cultures with >80% Ag-specific CD4 cells after two stimulations. Further stimulation with peptide-loaded aAPC increased purity to >99% Ag-specific T cells. After in vitro culture for 3-12 wk, the flu-specific CD4 T(CM) had surface markers that were generally consistent with an effector phenotype described for CD8 T cells, except for the maintenance of CD28 expression. The T(CM) were capable of 20-40 mean population doublings in vitro, and the expanded cells produced IFN-gamma, IL-2, and TNF-alpha in response to Ag, and a subset of cells also secreted IL-4 with PMA/ionomycin treatment. In conclusion, aAPCs expand T(CM) that have extensive replicative capacity, and have potential applications in adoptive immunotherapy as well as for studying the biology of human MHC class II-restricted T cells.
Collapse
Affiliation(s)
- Marcela V Maus
- Abramson Family Cancer Research Institute, University of Pennsylvania Cancer Center,University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Bryson JS, Zhang L, Goes SW, Jennings CD, Caywood BE, Carlson SL, Kaplan AM. CD4+ T cells mediate murine syngeneic graft-versus-host disease-associated colitis. THE JOURNAL OF IMMUNOLOGY 2004; 172:679-87. [PMID: 14688381 DOI: 10.4049/jimmunol.172.1.679] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Syngeneic graft-vs-host disease (SGVHD) develops following lethal irradiation, reconstitution with syngeneic bone marrow, and treatment with a 21-day course of the immunosuppressive agent cyclosporin A (CsA). Following cessation of CsA, this inducible disease is characterized by weight loss, diarrhea, and development of inflammation in the colon and liver. Although nonspecific effector cells and Th1 cytokines have been shown to participate in disease induction, the role of T cells has not been fully elucidated. Initial studies demonstrated significant increases in CD4+ T cells, but not other T cell populations in the colons of diseased animals relative to transplant control animals. To demonstrate a functional linkage between increases in colonic CD4+ T cells and disease induction, in vivo T cell depletion studies were performed. Beginning on the day of bone marrow transplantation, groups of control and CsA-treated animals were treated with mAb against either CD4 or CD8 for 21 days. Treatment with anti-CD4, but not anti-CD8, eliminated clinical symptoms and colon pathology. Interestingly, neither anti-CD4 nor anti-CD8 therapy affected the development of liver pathology associated with SGVHD. These findings demonstrated that CD4+ T cells initiate development of the intestinal inflammation associated with murine SGVHD.
Collapse
Affiliation(s)
- J Scott Bryson
- Department of Internal Medicine, Markey Cancer Center, Chandler Medical Center, University of Kentucky, Lexington, KY 40536, USA.
| | | | | | | | | | | | | |
Collapse
|
117
|
Yin M, Ochs RS. Mechanism for the paradoxical inhibition and stimulation of calcineurin by the immunosuppresive drug tacrolimus (FK506). Arch Biochem Biophys 2004; 419:207-13. [PMID: 14592464 DOI: 10.1016/j.abb.2003.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We examined the paradoxical inhibition and stimulation of calcineurin, the calcium-activated protein phosphatase, using the drug FK506 (tacrolimus) which acts as a complex together with its binding protein; the complex is designated here as FKC. We reproduced FKC inhibition with RIIp, a phosphorylated peptide substrate, and FKC stimulation with p-nitrophenylphosphate (pNPP) as substrate. The presence of RIIp in the pNPP assay caused inhibition. Yet, under these conditions, FKC still stimulated pNPP dephosphorylation to the same extent. The effects of Mn2+ were strikingly different for the two substrates when calcineurin was measured under otherwise identical conditions: Mn2+ stimulated pNPP dephosphorylation several fold, but only stimulated RIIp dephosphorylation by about 50%. When Pi was used as product inhibitor, FKC stimulation, but not calmodulin stimulation, was attenuated. We conclude that FKC enhances substrate binding to the enzyme. This would lead to inhibition with RIIp, known to bind calcineurin tightly, but stimulation with pNPP, known to bind calcineurin weakly. The result not only resolves the paradox but also elucidates the mechanism of action for this class of immunosuppressive drugs.
Collapse
Affiliation(s)
- Mali Yin
- Department of Chemistry, Sarah Lawrence College, Bronxville, NY 10708, USA
| | | |
Collapse
|
118
|
Acuto O, Michel F. CD28-mediated co-stimulation: a quantitative support for TCR signalling. Nat Rev Immunol 2004; 3:939-51. [PMID: 14647476 DOI: 10.1038/nri1248] [Citation(s) in RCA: 527] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Oreste Acuto
- Molecular Immunology Unit, Department of Immunology, Institut Pasteur, 25 Rue du Dr Roux, Cedex 15, 75724 Paris, France.
| | | |
Collapse
|
119
|
Shalaby WSW, Yeh H, Woo E, Corbett JT, Gray H, June CH, Shalaby SW. Absorbable microparticulate cation exchanger for immunotherapeutic delivery. ACTA ACUST UNITED AC 2004; 69:173-82. [PMID: 15116407 DOI: 10.1002/jbm.b.20040] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
An absorbable microparticulate cation exchanger was synthesized as a versatile carrier for biologically active proteins. In this work, acid-terminated polyglycolide (or polyglycolic acid) microparticulates (PG-MP) were surface modified for either sustained release of cytokines or as a platform for immunomodulation. The intended goal was to achieve in situ recruitment/maturation of dendritic cells and activation of T cells for tumor immunotherapy. PG-MP were prepared with a volume weighted mean diameter of 7.02 micro (range: 2.09-14.58 micro). Accessible carboxylic acid groups were determined to be 0.3 mmol/g with a corresponding zeta potential of -21.87 mV in phosphate-buffered saline. Under low magnification, scanning electron microscopy (SEM) revealed a highly textured surface due to processing from repetitive jet milling. However, a moderately porous architecture was noted at higher magnification. Electron spectroscopy for chemical analysis was used to characterize the PG-MP surface before and after adsorption of human granulocyte-macrophage colony stimulating factor (GM-CSF). Adsorption of GM-CSF on PG-MP (PG-GMCSF) resulted in a modest increase in the surface atomic concentration of nitrogen (0.97%). Pretreating the surface with poly-L-lysine (PG/Lys-GMCSF) prior to adding GM-CSF produced a nearly threefold increase in the surface nitrogen concentration (4.20% compared to 1.47%). This manipulation not only increased loading content, but also prolonged the release of GM-CSF released from 6 days to 26 days. ESCA on the post-release PG-MP samples (PG-GMCSF and PG/Lys-GMCSF) revealed a similar residual surface nitrogen concentration (2.26% vs. 2.35%). The observation was consistent with irreversibly adsorbed GM-CSF. It is postulated that irreversibly bound GM-CSF is released over time as a function of microparticulate degradation. Biological activity of released GM-CSF was confirmed by the proliferation of a GM-CSF-dependent cell line (TF-1) in the presence of microparticulates. PG-MP mediated activation of T cells was achieved through irreversible adsorption of either antimouse cd3 plus antimouse cd28 monoclonal antibodies (alpha-cd3/cd28-MP) or antihuman CD3 plus antihuman CD28 monoclonal antibodies (alpha-CD3/CD28-MP) on PG-MP. Irreversibly adsorbed antibodies were capable of activating both resting mouse and human T cells. Intracellular flow cytometry on mouse T cells revealed that nearly 50% of the activated cells produced interferon-gamma (IFN-gamma). This was consistent with a TH-1 or cell-mediated response. In vivo efficacy was evaluated in a mouse flank tumor model showing a significant antitumor effect both alone and in combination. Combination therapy was most effective at preventing tumor implantation (8/8 mice) and was able induce tumor regression (4/7 mice) and/or stable disease (3/7 mice) in a regression model. In these studies, immunohistochemistry was used to confirm local recruitment of dendritic cells. In conclusion, the PG-MP represents a novel absorbable cation exchanger that can be readily manipulated to deliver biologically active proteins for immunotherapy.
Collapse
Affiliation(s)
- Waleed S W Shalaby
- Division of Gynecologic Oncology, Christiana Care Health Services, Newark, Delaware 19713, USA.
| | | | | | | | | | | | | |
Collapse
|
120
|
Lee HW, Nam KO, Seo SK, Kim YH, Kang H, Kwon BS. 4-1BB cross-linking enhances the survival and cell cycle progression of CD4 T lymphocytes. Cell Immunol 2003; 223:143-50. [PMID: 14527512 DOI: 10.1016/s0008-8749(03)00169-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
4-1BB, a T cell co-stimulatory receptor, prolongs the survival and multiplication of CD4 T cells. Cross-linking 4-1BB stimulated expression of the anti-apoptotic genes bcl-XL and bcl-2, as well as of cyclins D2 and E, and inhibited expression of the cyclin-dependent kinase (cdk) inhibitor p27kip1. Ova-activated CD4 T cells of 4-1BB-deficient/DO11.10 TCR transgenic mice survived less well and underwent less expansion than cells of wild type DO11.10 TCR transgenic mice. These findings demonstrate that 4-1BB is a co-stimulatory molecule for CD4 T cell survival and expansion in vivo.
Collapse
MESH Headings
- Animals
- Antigens, CD
- Blotting, Western
- CD4-Positive T-Lymphocytes/physiology
- Cell Cycle/physiology
- Cell Cycle Proteins/biosynthesis
- Cell Cycle Proteins/drug effects
- Cell Survival/physiology
- Cross-Linking Reagents/pharmacology
- Cyclin D2
- Cyclin E/biosynthesis
- Cyclin E/drug effects
- Cyclin-Dependent Kinase Inhibitor p27
- Cyclins/biosynthesis
- Cyclins/drug effects
- Flow Cytometry
- Gene Expression Regulation/immunology
- Genes, bcl-2/drug effects
- Genes, bcl-2/physiology
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/physiology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Proto-Oncogene Proteins c-bcl-2/biosynthesis
- Proto-Oncogene Proteins c-bcl-2/drug effects
- Proto-Oncogene Proteins c-bcl-2/genetics
- RNA, Messenger/analysis
- Receptors, Antigen, T-Cell/genetics
- Receptors, Nerve Growth Factor/deficiency
- Receptors, Nerve Growth Factor/drug effects
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Tumor Necrosis Factor/deficiency
- Receptors, Tumor Necrosis Factor/drug effects
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- T-Lymphocyte Subsets/physiology
- Tumor Necrosis Factor Receptor Superfamily, Member 9
- Tumor Suppressor Proteins/biosynthesis
- Tumor Suppressor Proteins/drug effects
- bcl-X Protein
Collapse
Affiliation(s)
- Hyeon-Woo Lee
- The Immunomodulation Research Center and Graduate Program in Immunology and Biomedicine, University of Ulsan, Ulsan 680-749, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
121
|
Jung U, Foley JE, Erdmann AA, Eckhaus MA, Fowler DH. CD3/CD28-costimulated T1 and T2 subsets: differential in vivo allosensitization generates distinct GVT and GVHD effects. Blood 2003; 102:3439-46. [PMID: 12855580 DOI: 10.1182/blood-2002-12-3936] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adoptive T-cell therapy using CD3/CD28 co-stimulation likely requires in vivo generation of antigen specificity. Because CD28 promotes TH1/TC1 (T1) or TH2/TC2 (T2) differentiation, costimulation may generate donor T1 or T2 cells capable of differentially mediating allogeneic graft-versus-tumor (GVT) effects and graft-versus-host disease (GVHD). Costimulation under T1 or T2 conditions indeed generated murine TH1/TC1 cells secreting interleukin-2/interferon-gamma (IL-2/IFN-gamma) or TH2/TC2 cells secreting IL-4/IL-5/IL-10. In vivo, allogeneic T1 cells expanded, maintained T1 secretion, and acquired allospecificity involving IFN-gamma and IL-5. In contrast, allogeneic T2 cells expanded less and maintained T2 secretion but did not develop significant allospecificity.Allogeneic, but not syngeneic, T1 cells mediated a GVT effect against host-type breast cancer cells, as median survival time (MST) increased from 25.6 +/- 2.6 (tumor controls) to 69.2 +/- 5.9 days (P < 1.2 x 10(-9)). This T1-associated GVT effect operated independently of fasL because T1 cells from gld mice mediated tumor-free survival. In contrast, allogeneic T2 cells mediated a modest, noncurative GVT effect (MST, 29 +/- 1.3 days; P <.0019). T1 recipients had moderate GVHD (histologic score, 4 of 12) that contributed to lethality after bone marrow transplantation; in contrast, T2 recipients had minimal GVHD (histologic score, 1 of 12). CD3/CD28 co-stimulation, therefore, generates T1 or T2 populations with differential in vivo capacity for expansion to alloantigen, resulting in differential GVT effects and GVHD.
Collapse
Affiliation(s)
- Unsu Jung
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bldg 10, Rm 12N226, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
122
|
Wong SC, Oh E, Ng CH, Lam KP. Impaired germinal center formation and recall T-cell-dependent immune responses in mice lacking the costimulatory ligand B7-H2. Blood 2003; 102:1381-8. [PMID: 12714510 DOI: 10.1182/blood-2002-08-2416] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
B7-H2, which is expressed constitutively on B cells and binds the inducible costimulator (ICOS) on antigen-activated T cells, is a member of the B7 family of costimulatory ligands. We have inactivated B7-H2 in the mouse. B7-H2-/- mice generate normal populations of B and T cells in their various lymphoid organs but have lower basal levels of heavy chain class-switched antibodies in their sera. These mice are able to mount normal immune responses to both type I and type II T-cell-independent antigens. However, their pattern of responses to a T-cell-dependent antigen is altered, with greatly reduced production of antigen-specific heavy chain class-switched antibodies, the levels of which could not be elevated even with repeated immunizations. This suggests a critical role for B7-H2 in the recall phases of the immune response. Germinal center formation is also impaired in the mutant mice. While B cells from the mutant mice could response normally to anti-IgM, anti-CD40, and lipopolysaccharide stimulation, the production of T-helper-type II cytokines such as interleukin-4 (IL-4) and IL-10 by primed CD4+ T cells from mutant mice were reduced. This indicated that the defects in humoral responses and germinal center formation in B7-H2-deficient mice are due to the lack of T-cell-mediated help to the B cells. Hence, B7-H2 on B cells is important for recruiting T-cell help via its interaction with ICOS and plays a critical role in costimulating humoral immune responses.
Collapse
Affiliation(s)
- Siew-Cheng Wong
- Institute of Molecular and Cell Biology, Singapore, Republic of Singapore
| | | | | | | |
Collapse
|
123
|
Chueh SCJ, Lai MK, Liu IS, Teng FC, Chen J. Curcumin enhances the immunosuppressive activity of cyclosporine in rat cardiac allografts and in mixed lymphocyte reactions. Transplant Proc 2003; 35:1603-5. [PMID: 12826232 DOI: 10.1016/s0041-1345(03)00377-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Curcumin (CCM; diferuoylmethane) is a dietary pigment in curry with known antineoplastic and anti-inflammatory effects. The immunosuppressive effects of CCM were studied in (1) rat heterotopic cardiac transplant models, using Brown-Norway (BN, RT1(n)) hearts to WKY (RT1(u)) hosts or Buffalo (BUF, RT1(b)) hearts to Wistar-Furth (WF, RT1(u)) hosts, (2) reverse transcriptase-polymerase chain reaction analysis of cytokines from transplanted specimens, and (3) mixed lymphocyte reactions (MLR). In the BN-to-WKY model, CCM alone significantly increased the mean survival time (MST) to 20.5 to 24.5 days, as compared to 9.1 days among nontreated controls. The combination of CCM and subtherapeutic doses of CsA produced further prolongation of the MST to 28.5 to 35.6 days, better than that of CCM or CsA alone (P <.05). In a BUF-to-WF model, CCM alone did not increased the MST, unless it was combined with subtherapeutic doses of CsA, wherein two thirds of the grafts survived for more than 60 days (P <.05 as compared to either treatment group). Cytokine analysis revealed significantly reduced expression of interleukin-2 (IL-2), interferon-gamma (IFN-gamma) and granzyme B in the day 3 specimens of the CCM and CCM CsA-treated allografts compared with the nontreated allograft controls. MLRs using the two MHC-incompatible rat strains (BNxWKY) showed an effect of increasing concentrations of CCM and/or CsA, which by combination index (CI) analysis showed a synergistic effect (CI = 0.22 to 0.81). This study for the first time demonstrates the effectiveness of CCM as a novel adjuvant immunosuppressant with cyclosporine both in vivo and in vitro.
Collapse
Affiliation(s)
- S-C J Chueh
- Department of Urology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan.
| | | | | | | | | |
Collapse
|
124
|
Deenick EK, Gett AV, Hodgkin PD. Stochastic model of T cell proliferation: a calculus revealing IL-2 regulation of precursor frequencies, cell cycle time, and survival. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4963-72. [PMID: 12734339 DOI: 10.4049/jimmunol.170.10.4963] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The outcome of Ag exposure is dictated by complex regulation of T cell proliferation. The rates of proliferation and survival are altered by numerous signals that the cell receives and integrates to achieve a net response. We have illustrated previously how small changes in kinetic parameters can lead to large differences, even under conditions of saturating IL-2. In this study, we examine the effect of varying IL-2 concentration on T cell response and develop a model incorporating additional parameters of proliferation and survival. Strikingly, the proportion of cells that enter the first division, but not the time at which they enter, is dramatically altered by IL-2. Furthermore, the survival and average division time of cells in later divisions are also altered by IL-2 concentration. Together, the small simultaneous effects on these parameters result in large differences in total cell number. These results reveal how in vitro systems may exaggerate the contribution of IL-2, and thus how costimuli or additional helper cells that alter IL-2 concentration, even by relatively small amounts, will generate large in vitro differences in cell number and therefore appear obligatory. Furthermore, they illustrate how a quantitative model of T cell activation can clarify how complex signal integration is handled by T cells in situ, and therefore more appropriately aid development of a theory of behavior.
Collapse
Affiliation(s)
- Elissa K Deenick
- Immune Regulation Group, Centenary Institute of Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | | | | |
Collapse
|
125
|
Abstract
Sirolimus is the USAN-assigned generic name for the natural product rapamycin. Sirolimus is produced by a strain of Streptomyces hygroscopicus, isolated from a soil sample collected from Rapa Nui commonly known as Easter Island. Although sirolimus was isolated as an antifungal agent with potent anticandida activity, subsequent studies revealed impressive antitumor and immunosuppressive activities. Sirolimus demonstrates activity against several murine tumors, such as B16 43 melanocarcinoma, Colon 26 tumor, EM ependymoblastoma, and mammary and colon 38 solid tumors. Sirolimus is a potent inhibitor of antigen-induced proliferation of T cells, B cells, and antibody production. Demonstration of the potent immunosuppressive activity of sirolimus in animal models of organ transplantation led to clinical trials and subsequent approval by regulatory authorities for prophylaxis of renal graft rejection. Interest in sirolimus as an immunosuppressive therapy in organ transplantation derives from its unique mechanism of action, its unique side-effect profile, and its ability to synergize with other immunosuppressive agents. The molecular mechanism underlying the antifungal, antiproliferative, and immunosuppressive activities of sirolimus is the same. Sirolimus forms an immunosuppressive complex with intracellular protein, FKBP12. This complex blocks the activation of the cell-cycle-specific kinase, TOR. The downstream events that follow the inactivation of TOR result in the blockage of cell-cycle progression at the juncture of G1 and S phase.
Collapse
Affiliation(s)
- S N Sehgal
- Wyeth-Ayerst Research, Collegeville, Pennsylvania, USA
| |
Collapse
|
126
|
Tadaki DK, Williams A, Lee KP, Kirk AD, Harlan DM. Porcine CD80: cloning, characterization, and evidence for its role in direct human T-cell activation. Xenotransplantation 2003; 10:252-8. [PMID: 12694545 DOI: 10.1034/j.1399-3089.2003.02004.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Previous studies has shown that human anti-pig reactivity in mixed lymphocyte cultures require the indirect presentation of antigens by human antigen presenting cells (APC). Xenoreactivity was inhibited by blockade of human costimulatory molecules. We investigated the role of porcine costimulatory molecules in their ability to activate human T cells directly. Porcine CD80 was cloned from lipopolysaccharide (LPS)-activated porcine lymphocytes. Sequence analysis showed a high degree of conservation in residues involved in CD28/CTLA4. COS cells transfected with porcine CD80 was able to activate human T cells in a cyclosporine independent manner, demonstrating that porcine CD80 can costimulate human T cells. Tumor necrosis factor-alpha (TNF-alpha) activated porcine splenocytes have been shown to up-regulate B7s. In order to test the effect of costimulation blockade in a xeno system, activated splenocytes were cultured with purified CD4+ T cells. The results demonstrated that these cells were capable of activating human T cells and this activation can be blocked by using an antihuman CD80 antibody that demonstrated cross-reactivity to porcine CD80. Non-cross reactive antibodies had no effect, again suggesting direct activation of the human T cells. These data suggest that a reagent that can block both the direct and indirect activation is necessary for a discordant xenotransplant.
Collapse
Affiliation(s)
- D K Tadaki
- NIDDK/Navy Transplantation and Autoimmunity Branch, Naval Medical Research Center, Bethesda, MD 20889-5600, USA.
| | | | | | | | | |
Collapse
|
127
|
Kurosawa S, Myers AC, Chen L, Wang S, Ni J, Plitt JR, Heller NM, Bochner BS, Schleimer RP. Expression of the costimulatory molecule B7-H2 (inducible costimulator ligand) by human airway epithelial cells. Am J Respir Cell Mol Biol 2003; 28:563-73. [PMID: 12707012 DOI: 10.1165/rcmb.2002-0199oc] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Tissue structural cells are known in some situations to play a role in the presentation of antigen and in immunoregulation. We assessed the expression of B7 homologs, known to be involved in antigen presentation and lymphocyte costimulation, in human airway epithelial cells. Flow cytometry performed on the airway epithelial cell line BEAS-2B, as well as primary bronchial epithelial cells (PBEC), showed that B7-H2 was constitutively expressed on both BEAS-2B and PBEC, whereas B7-1 and B7-2 were undetectable on either epithelial cell type. B7-H2 expression was confirmed by Western blot using a specific antibody. Stimulation with various cytokines, including tumor necrosis factor-alpha, interferon-gamma, and interleukin-4, slightly downregulated B7-H2 expression detected by flow cytometry, but did not significantly alter the apparent level of protein as assessed by Western blotting. Northern blotting detected mRNA for B7-H2 and B7-1, but not B7-2. B7-H2 was cloned from BEAS-2B cells and the sequence verified. Expression of B7-H2 mRNA was detected by real-time reverse transcriptase-polymerase chain reaction in PBEC from three independent donors. Immunohistochemical analysis of airway derived from autopsies revealed expression of B7-H2 in human airway epithelial cells. These results demonstrate that airway epithelial cells express the costimulatory molecule B7-H2, and suggest the possibility that B7-H2 may participate in antigen presentation by epithelial cells.
Collapse
Affiliation(s)
- Shin Kurosawa
- Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Abstract
Programmed cell death is essential for the development and maintenance of cellular homeostasis of the immune system. The Bcl-2 family of proteins comprises both pro-apoptotic and anti-apoptotic members. A subset of pro-apoptotic members, called 'BH3-only' proteins, share sequence homology only in the minimal death domain, designated the Bcl-2 homology 3 (BH3) domain. BH3-only proteins operate as upstream sentinels, selectively sensing both intrinsic and extrinsic death stimuli. They communicate this information to the pro-apoptotic 'multidomain' members Bax or Bak--a process that is antagonized by anti-apoptotic members of the Bcl-2 family. The functional balance of pro-apoptotic versus anti-apoptotic influences, which operates at organelles, determines whether a lymphocyte will live or die. BH3-only molecules, often working in concert, compete for downstream multidomain pro- and anti-apoptotic BCL-2 members to control serial stages of lymphocyte development and homeostasis.
Collapse
Affiliation(s)
- Joseph T Opferman
- Howard Hughes Medical Institute, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
129
|
Hegde VR, Puar MS, Dai P, Patel M, Gullo VP, Chan TM, Silver J, Pramanik BN, Jenh CH. Condensed aromatic peptide family of microbial metabolites, inhibitors of CD28-CD80 interactions. Bioorg Med Chem Lett 2003; 13:573-5. [PMID: 12565974 DOI: 10.1016/s0960-894x(02)00919-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Three condensed aromatic peptides SCH79235 (1), SCH79236 (2), and SCH204698 (3) were isolated from the fermentation broth of a Streptomycete microorganism. The structure of SCH204698 (3) was established by extensive NMR spectral data. All these compounds exhibited good activity against CD28-CD80 binding with an IC(50) of 0.42, 0.38 and 0.22 microM, respectively.
Collapse
Affiliation(s)
- Vinod R Hegde
- Schering Plough Research Institute, 2015 Galloping Hill Road, Kenilworth, NJ 07033, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Härtel C, Hammers HJ, Schlenke P, Fricke L, Schumacher N, Kirchner H, Müller-Steinhardt M. Individual variability in cyclosporin A sensitivity: the assessment of functional measures on CD28-mediated costimulation of human whole blood T lymphocytes. J Interferon Cytokine Res 2003; 23:91-9. [PMID: 12744774 DOI: 10.1089/107999003321455480] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The quantitative analysis of cyclosporin A (CsA) effects might be helpful for optimizing immunosuppressive treatment after allogeneic organ transplantation in individual patients, as rejection can occur despite the existence of CsA blood levels within therapeutic ranges. Previous investigations found that costimulation of the CD28 pathway generally mediates CsA-resistant proliferation of T cell receptor (TCR)-activated T lymphocytes. However, here we describe considerable interindividual variation regarding the immunosuppressive effects of CsA (1000 microg/L) on anti-CD3/CD28 T cell costimulation in a human whole blood assay. In the in vitro study, we found a significant reduction of T cell proliferation, activation marker expression (CD25, CD69) on the T cell surface, and interleukin-2 (IL-2) protein expression in whole blood samples of all healthy subjects (n = 11). However, the investigation of cytokine mRNA profiles revealed variable results of in vitro CsA sensitivity. Whole blood samples of 3 of 11 healthy individuals demonstrated a marked suppression of IL-2 mRNA expression (>50%) and a partial inhibition of IL-4, interferon-gamma (IFN-gamma), and tumor necrosis factor-alpha (TNF-alpha) mRNA expression on addition of CsA. In contrast, the remaining 8 healthy individuals had cytokine mRNA expression levels that were unaffected or even increased when CsA was administered in vitro. In patients undergoing CsA monotherapy (ex vivo study, n = 9), we found a significant suppression of IL-2 mRNA levels in 4 of 9 patients ex vivo. Thus, we cannot confirm a universal CsA resistance of T cells on anti-CD3/CD28 costimulation. Instead, our results suggest an individual degree of CsA sensitivity that might be more consistent with clinical experience. Prospective studies are necessary to determine if individual degrees of CsA sensitivity correlate with clinical events and are associated with a low or high risk of transplant rejection.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal/immunology
- Antigens, CD/drug effects
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/drug effects
- Antigens, Differentiation, T-Lymphocyte/immunology
- Biomarkers
- Blood Cells/immunology
- CD28 Antigens/immunology
- Cell Division/drug effects
- Cells, Cultured
- Cyclosporine/blood
- Cyclosporine/pharmacology
- Cyclosporine/therapeutic use
- Dose-Response Relationship, Drug
- Female
- Gene Expression Regulation
- Genetic Variation
- Humans
- Immunosuppressive Agents/blood
- Immunosuppressive Agents/pharmacology
- Immunosuppressive Agents/therapeutic use
- Interferon-gamma/antagonists & inhibitors
- Interferon-gamma/drug effects
- Interleukin-2/biosynthesis
- Interleukin-2/blood
- Interleukin-2/genetics
- Interleukin-4/antagonists & inhibitors
- Interleukin-4/blood
- Interleukin-4/genetics
- Kidney Transplantation
- Lectins, C-Type
- Lymphocyte Activation
- Male
- RNA, Messenger/metabolism
- Receptors, Interleukin-2/drug effects
- Receptors, Interleukin-2/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/drug effects
Collapse
Affiliation(s)
- Christoph Härtel
- Institute of Immunology and Transfusion Medicine, University of Lübeck Medical School, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | | | | | | | | | | | | |
Collapse
|
131
|
Maus MV, Riley JL, Kwok WW, Nepom GT, June CH. HLA tetramer-based artificial antigen-presenting cells for stimulation of CD4+ T cells. Clin Immunol 2003; 106:16-22. [PMID: 12584046 DOI: 10.1016/s1521-6616(02)00017-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Current evidence suggests that immunotherapy for cancer or infectious diseases will require the activation of CD4(+) T cells in addition to the activation of cytotoxic CD8(+) T cells. To complement and overcome some of the limitations of dendritic-cell-based vaccines and ex vivo expansion of human T cells, we sought to engineer artificial antigen-presenting cells (aAPCs) for the stimulation of antigen-specific human CD4(+) T cells. We have designed a variety of aAPCs using magnetic beads as a scaffold on which to coat HLA-peptide tetrameric complexes along with costimulatory molecules such as anti-CD28. Here, we tested various forms of conjugation of the tetramers onto beads, characterized the relative concentration of antigen available on the surface of the beads, and evaluated the ability of different types of beads to promote activation of antigen-specific CD4(+) T cells. We find that an indirect coating of HLA-peptide tetramers on beads via an anti-Class II antibody provides specific stimulation of antigen-specific CD4(+) T cells.
Collapse
Affiliation(s)
- Marcela V Maus
- Abramson Family Cancer Research Institute, 557 BRB II/III, 421 Curie Boulevard, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | |
Collapse
|
132
|
Thomas AK, Maus MV, Shalaby WS, June CH, Riley JL. A cell-based artificial antigen-presenting cell coated with anti-CD3 and CD28 antibodies enables rapid expansion and long-term growth of CD4 T lymphocytes. Clin Immunol 2002; 105:259-72. [PMID: 12498807 DOI: 10.1006/clim.2002.5277] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We compared the ability of two genetically modified myeloid cells, K562 and U937, to serve as artificial antigen-presenting cells (aAPC). Both aAPC were stably transfected with the low-affinity Fcgamma receptor CD32 (K32/U32 cells). K32 cells loaded with anti-CD3 and anti-CD28 Ab (K32/CD3/28) induced more rapid CD4 T-cell expansion than CD3/28-coated beads. In contrast, U32/CD3/28 induced high levels of CD4 T-cell thymidine uptake but were unable to sustain long-term T-cell expansion. K32 cells, but not U32 cells, loaded with anti-CD3 alone also stimulated CD4 T-cell growth and IL-2 secretion, indicating the expression of additional costimulatory molecules on K32 cells. We found constitutive expression of B7-H3 and a strong upregulation of mRNA encoding for IL-15, PD-L1, and PD-L2 after coculture with CD4 T cells activated by K32/CD3/28 but not U32/CD3/28. We conclude that K32 aAPCs are a robust system for clinical scale ex vivo expansion of CD4 T cells.
Collapse
Affiliation(s)
- Anna K Thomas
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
133
|
Baumgartner R, Durant PJ, van Gessel Y, Chattopadhyay S, Beswick RL, Tadaki DK, Lasbury ME, Lee CH, Perrin PJ, Lee KP. Evidence for the requirement of T cell costimulation in the pathogenesis of natural Pneumocystis carinii pulmonary infection. Microb Pathog 2002; 33:193-201. [PMID: 12473434 DOI: 10.1006/mpat.2002.0528] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pneumocystis carinii pneumonia (PCP) is a frequent and serious opportunistic infection in immunocompromized patients. Although the pathogenesis of PCP-mediated lung injury is poorly understood, a central involvement of host inflammatory responses has been implicated. We have found that while the loss of specific T cell costimulatory signals increases susceptibility to the spontaneous pneumocystis infection, PCP-induced pulmonary injury (and subsequent morbidity and mortality) involves other intact costimulatory pathways. Mice that are genetically deficient for the costimulatory receptor CD154 (CD154 knockout (ko) mice) spontaneously developed PCP, consistent with the increased susceptibility of X-linked hyper IgM syndrome patients (caused by CD154 gene mutations) to P. carinii infection. In these mice PCP was manifested by progressive weight loss, dyspnea and death. In contrast, CD154 ko mice also genetically lacking ICAM1 (CD154 koxICAM1 ko) or CD28 (CD154 koxCD28 ko) costimulatory receptors had later onset of weight loss and significantly prolonged survival. Although onset of infection and age-matched P. carinii organism burden were equivalent, the CD154 single knockout mice had evidence of greater pulmonary inflammation vs. the double ko's. These findings suggest that costimulation-dependent T cell-mediated inflammation plays an important role in both susceptibility to and pathogenesis of PCP, and may identify potential molecular targets for novel immunomodulatory treatment approaches.
Collapse
|
134
|
Murphy LLS, Hughes CCW. Endothelial Cells Stimulate T Cell NFAT Nuclear Translocation in the Presence of Cyclosporin A: Involvement of the wnt/Glycogen Synthase Kinase-3β Pathway. THE JOURNAL OF IMMUNOLOGY 2002; 169:3717-25. [PMID: 12244165 DOI: 10.4049/jimmunol.169.7.3717] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cells resistant to the immunosuppressive drug cyclosporin A (CsA) may be important mediators of chronic graft rejection. We previously reported that T cells activated in the presence of endothelial cells (EC) develop resistance to CsA, and initiate IL-2 secretion within 8-12 h of triggering. CsA normally blocks the phosphatase, calcineurin, thus preventing nuclear translocation of the transcription factor, NFAT. We find that in the presence but not the absence of EC, NFAT1 can be detected in the nuclei of CsA-treated T cells within 8 h of triggering, reaching a maximal level of 60% of control by 24 h. Glycogen synthase kinase-3beta (GSK-3beta), which rephosphorylates NFAT and promotes nuclear export, is inhibited by EC costimulation. GSK-3beta is a component of the wnt signaling pathway, and EC express wnt-5a and T cells express frizzled-5, a wnt-5a receptor. Wnt-5a promotes T cell NFAT nuclear accumulation in the presence of CsA, an effect mimicked by Li(+), a potent inhibitor of GSK-3beta. The protein kinase C agonist PMA dramatically synergizes with both EC and wnt-5a in stimulating T cell IL-2 synthesis, and inhibition of either protein kinase C by Ro-31-8425 or G-proteins by pertussis toxin effectively blocks the actions of wnt-5a on T cells. Finally, a secreted, dominant-negative form of frizzled-5 blocks EC-mediated CsA resistance. Thus, EC promote CsA-resistant nuclear localization of NFAT and subsequent IL-2 synthesis through a noncanonical wnt-dependent pathway.
Collapse
Affiliation(s)
- Lisa L Salazar Murphy
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | | |
Collapse
|
135
|
Harlin H, Podack E, Boothby M, Alegre ML. TCR-independent CD30 signaling selectively induces IL-13 production via a TNF receptor-associated factor/p38 mitogen-activated protein kinase-dependent mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:2451-9. [PMID: 12193714 DOI: 10.4049/jimmunol.169.5.2451] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Initiation of T lymphocyte responses to most Ags requires concurrent stimulation through the TCR and costimulatory receptors such as CD28. Following initial activation, secondary receptors are up-regulated that can costimulate T cells in concert with TCR engagement. One such receptor is the TNFR family member CD30. In this study, we report that unlike CD28, ligation of CD30 on normal effector T cells induces IL-13 production in the absence of concurrent TCR engagement. TCR-independent CD30-mediated IL-13 release correlated with activation of c-Jun N-terminal kinase, p38 mitogen-activated protein kinase (MAPK), and NF-kappaB, and was completely inhibited by the expression of a TNFR-associated factor 2 (TRAF2) dominant-negative transgene (TRAF2.DN-Tg), but not by that of an I-kappaBalpha dominant-negative transgene. In parallel, expression of the TRAF2.DN-Tg selectively prevented the induction of c-Jun N-terminal kinase and p38 MAPK, but not that of NF-kappaB. Furthermore, IL-13 production was reduced in a dose-dependent manner by the p38 MAPK inhibitor SB203580. Together, these results suggest that TCR-independent CD30-mediated production of IL-13 is triggered by association of CD30 with TRAF family members and subsequent activation of p38 MAPK. Inasmuch as IL-13 can promote airway inflammation and cancer progression, production of IL-13 in a TCR-independent manner has important pathological implications in vivo.
Collapse
Affiliation(s)
- Helena Harlin
- Committee on Immunology and Department of Medicine, Section of Rheumatology, University of Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
136
|
Preston E, Kirk AD. Context-based therapy: A conceptual framework for transplantation tolerance. Transplant Rev (Orlando) 2002. [DOI: 10.1053/trre.2002.126011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
137
|
Hegde VR, Puar MS, Dai P, Patel M, Gullo VP, Pramanik B, Jenh CH. A microbial metabolite inhibitor of CD28–CD80 interactions. Tetrahedron Lett 2002. [DOI: 10.1016/s0040-4039(02)01029-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
138
|
Abstract
Typical immune responses lead to prominent clonal expansion of antigen-specific T and B cells followed by differentiation into effector cells. Most effector cells die at the end of the immune response but some of these cells survive and form long-lived memory cells. The factors controlling the formation and survival of memory T cells are reviewed.
Collapse
Affiliation(s)
- Jonathan Sprent
- Department of Immunology, IMM4, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA.
| | | |
Collapse
|
139
|
Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, Plas DR, Elstrom RL, June CH, Thompson CB. The CD28 signaling pathway regulates glucose metabolism. Immunity 2002; 16:769-77. [PMID: 12121659 DOI: 10.1016/s1074-7613(02)00323-0] [Citation(s) in RCA: 1122] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lymphocyte activation initiates a program of cell growth, proliferation, and differentiation that increases metabolic demand. Although T cells increase glucose uptake and glycolysis during an immune response, the signaling pathways that regulate these increases remain largely unknown. Here we show that CD28 costimulation, acting through phosphatidylinositol 3'-kinase (PI3K) and Akt, is required for T cells to increase their glycolytic rate in response to activation. Furthermore, CD28 controls a primary response pathway, inducing a level of glucose uptake and glycolysis in excess of that needed to maintain cellular ATP/ADP levels or macromolecular synthesis. These data suggest that CD28 costimulation functions to increase glycolytic flux, allowing T cells to anticipate energetic and biosynthetic needs associated with a sustained response.
Collapse
Affiliation(s)
- Kenneth A Frauwirth
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Pahl A, Zhang M, Török K, Kuss H, Friedrich U, Magyar Z, Szekely J, Horvath K, Brune K, Szelenyi I. Anti-inflammatory effects of a cyclosporine receptor-binding compound, D-43787. J Pharmacol Exp Ther 2002; 301:738-46. [PMID: 11961080 DOI: 10.1124/jpet.301.2.738] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
By virtue of its binding to cyclophilin, the cellular receptor for cyclosporine (CsA), we could identify a new compound D-43787 [N-[(1-tert-butyloxycarbonyl)-indolin-2-(S)-carbonyl]-indolin-2-(S)-carbonacid-[N-epsilon-benzyloxycarbonyl)-2-(S)-lysin methylester]-amide] exhibiting immunomodulating properties. It inhibited cell proliferation induced by 12-O-tetradecanoylphorbol-13-acetate (TPA)/ionomycin and anti-CD3/CD28 with an IC(50) of 0.3 microM. The protein phosphatase calcineurin, which is the target of the CsA-cyclophilin complex, is not inhibited by D-43787. It inhibited T helper cell (Th) 2 cytokines interleukin (IL)-4, -5, and -13 more effectively than the Th1 cytokine interferon (IFN)-gamma in human primary T cells. The IC(50) for IL-5 and IL-13 in TPA/ionomycin-stimulated peripheral blood mononuclear cells (PBMC) is 0.7 +/- 0.1 and 0.5 +/- 0.1 microM, respectively, whereas the IC(50) for IFN-gamma is 2.0 +/- 0.4 microM. When PBMC were stimulated with anti-CD3/CD28, the IC(50) for IL-4, -5, and -13 were 1.5 +/- 0.2, 1.8 +/- 0.2, and 1.9 +/- 0.4 microM, respectively. IFN-gamma was only partially inhibited under these conditions. This effect was even more pronounced in pure CD4(+) T cells. Pretreatment of human monocytes with D-43787 inhibited lipopolysaccharide-induced proinflammatory cytokines IL-6 and TNFalpha with an IC(50) of 1.2 +/- 0.1 and 4.7 +/- 0.9 microM, respectively. In vivo, D-43787 potently inhibited late-phase eosinophilia in actively sensitized and challenged guinea pigs (10 mg/kg, i.p.: 51%) and Brown-Norway rats (1 mg/kg, intrapulmonary: 66% 30 mg/kg, i.p.: 50%). In adjuvant-induced arthritis, D-43787 (10-40 mg/kg, b.i.d., i.p.) dose dependently reduced edema development on both hind paws. The potency of D-43787 was comparable with that of indomethacin and dexamethasone. In conclusion, we characterized a novel Th2 selective immunosuppressive drug with possible anti-asthmatic/anti-inflammatory effects. Its mode of action is distinct from that of CsA.
Collapse
Affiliation(s)
- Andreas Pahl
- Department of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Costa C, Barber DF, Fodor WL. Human NK cell-mediated cytotoxicity triggered by CD86 and Gal alpha 1,3-Gal is inhibited in genetically modified porcine cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3808-16. [PMID: 11937533 DOI: 10.4049/jimmunol.168.8.3808] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Delayed xenograft rejection is a major hurdle that needs to be addressed to prolong graft survival in pig-to-primate xenotransplantation. NK cell activation has been implicated in delayed xenograft rejection. Both Ab-dependent and independent mechanisms are responsible for the high susceptibility of porcine cells to human NK cell-mediated cytotoxicity. Previous reports demonstrated a role of Galalpha1,3-Gal Ag in triggering the Ab-independent responses. We hypothesize that expression of CD80 and/or CD86 on porcine cells may also play a role in NK cell activation as human NK cells express a variant of CD28. Our initial analysis showed that porcine endothelial cells and fibroblasts express CD86, but not CD80. Genetic engineering of these cells to express hCD152-hCD59, a chimeric molecule designed to block CD86 in cis, was accompanied by a reduction in susceptibility to human NK cell-mediated cytotoxicity. The use of a specific anti-porcine CD86-blocking Ab and the NK92 and YTS cell lines further confirmed the involvement of CD86 in triggering NK cell-mediated lysis of porcine cells. Maximal protection was achieved when hCD152-hCD59 was expressed in H transferase-transgenic cells, which show reduced Galalpha1,3-Gal expression. In this work, we describe two mechanisms of human NK cell-mediated rejection of porcine cells and demonstrate that genetically modified cells resist Ab-independent NK cell-mediated cytotoxicity.
Collapse
MESH Headings
- Abatacept
- Animals
- Animals, Genetically Modified/immunology
- Antigens, CD/biosynthesis
- Antigens, CD/physiology
- Antigens, Differentiation/biosynthesis
- Antigens, Differentiation/genetics
- Antigens, Differentiation/pharmacology
- B7-1 Antigen/biosynthesis
- B7-2 Antigen
- CD28 Antigens/biosynthesis
- CD59 Antigens/biosynthesis
- CD59 Antigens/genetics
- CD59 Antigens/pharmacology
- CTLA-4 Antigen
- Cell Line
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Cells, Cultured
- Cytotoxicity Tests, Immunologic
- Cytotoxicity, Immunologic/genetics
- Cytotoxicity, Immunologic/immunology
- Disaccharides/metabolism
- Disaccharides/physiology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Fibroblasts/immunology
- Fibroblasts/metabolism
- Fucosyltransferases/biosynthesis
- Fucosyltransferases/genetics
- Humans
- Immunity, Cellular/genetics
- Immunoconjugates
- Killer Cells, Natural/immunology
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/physiology
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/pharmacology
- Swine
- Transduction, Genetic
- Galactoside 2-alpha-L-fucosyltransferase
Collapse
Affiliation(s)
- Cristina Costa
- Department of Molecular Sciences, Alexion Pharmaceuticals, Cheshire, CT 06410, USA
| | | | | |
Collapse
|
142
|
Pahl A, Zhang M, Kuss H, Szelenyi I, Brune K. Regulation of IL-13 synthesis in human lymphocytes: implications for asthma therapy. Br J Pharmacol 2002; 135:1915-26. [PMID: 11959794 PMCID: PMC1573320 DOI: 10.1038/sj.bjp.0704656] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
1. IL-13 is an important mediator in inflammatory diseases such as asthma. IL-13 is mainly produced by T cells. However, signalling pathways leading to induction of this cytokine are not well-characterized. We analysed the regulation of IL-13 in human peripheral blood mononuclear cells and CD4(+) T cells. 2. Cyclosporine (CsA) and FK-506 inhibited IL-13 synthesis, when cells were stimulated by TPA/ionomycin. However, stimulation by alpha-CD3/alpha-CD28 led to an enhanced IL-13 synthesis. 3. NF-kappa B inhibitor N-tosyl-L-lysine chloromethylketone (TLCK) inhibited IL-13 synthesis more effectively after TPA/ionomycin stimulation. After alpha-CD3/alpha-CD28 stimulation, only 300 microM TLCK inhibited IL-13 synthesis. Dexamethasone inhibited IL-13 equally effective after alpha-CD3/alpha-CD28 and TPA/ionomycin stimulation. 4. p38 MAPK inhibitor SB203580 inhibited IL-13 synthesis only partially. MEK inhibitor U0126 inhibited TPA/ionomycin induced IL-13 synthesis very effectively, whereas alpha-CD3/alpha-CD28 stimulated IL-13 induction was resistant to this drug. 5. These results were confirmed in purified CD4(+) T cells. In difference to PBMCs alpha-CD3/alpha-CD28 stimulated IL-13 synthesis was effectively inhibited by CsA, FK-506 and U0126. 6. Therefore U0126 was tested in an animal model of allergic asthma. We could demonstrate for the first time that inhibition of the MEK - ERK cascade is a therapeutic option for asthma. Intraperitoneal administration of 10 mg kg(-1) U0126 reduced lung eosinophilia in ovalbumin-challenged Brown Norway rats by 44%. 7. These results demonstrate that different signalling pathways are involved in regulating IL-13 synthesis in primary human T cells. Characterizing highly potent inhibitors of IL-13 synthesis can be exploited to identify new drugs to treat immunological diseases such as asthma.
Collapse
Affiliation(s)
- Andreas Pahl
- Department of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nürnberg, Fahrstr. 17, D-91054 Erlangen, Germany.
| | | | | | | | | |
Collapse
|
143
|
|
144
|
Maus MV, Thomas AK, Leonard DGB, Allman D, Addya K, Schlienger K, Riley JL, June CH. Ex vivo expansion of polyclonal and antigen-specific cytotoxic T lymphocytes by artificial APCs expressing ligands for the T-cell receptor, CD28 and 4-1BB. Nat Biotechnol 2002; 20:143-8. [PMID: 11821859 DOI: 10.1038/nbt0202-143] [Citation(s) in RCA: 330] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The ex vivo priming and expansion of human cytotoxic T lymphocytes (CTLs) has potential for use in immunotherapy applications for cancer and infectious diseases. To overcome the difficulty in obtaining sufficient numbers of CTLs, we have developed artificial antigen-presenting cells (aAPCs) expressing ligands for the T-cell receptor (TCR) and the CD28 and 4-1BB co-stimulatory surface molecules. These aAPCs reproducibly activate and rapidly expand polyclonal or antigen-specific CD8(+) T cells. The starting repertoire of CD8+ T cells was preserved during culture. Furthermore, apoptosis of cultured CD8(+) T cells was diminished by this approach. This approach may have important therapeutic implications for adoptive immunotherapy.
Collapse
MESH Headings
- Antigen-Presenting Cells/immunology
- Antigens, CD
- Apoptosis
- CD28 Antigens/metabolism
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line
- Cell Separation
- Cells, Cultured
- Coculture Techniques
- Flow Cytometry
- Humans
- Immunotherapy, Adoptive/methods
- K562 Cells
- Ligands
- Protein Binding
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Tumor Necrosis Factor/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- Time Factors
- Tumor Necrosis Factor Receptor Superfamily, Member 9
Collapse
Affiliation(s)
- Marcela V Maus
- Abramson Family Cancer Research Institute, University of Pennsylvania Cancer Center, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
145
|
Abstract
Expanding the cytokine paradigm beyond the use of calcineurin inhibitors as baseline therapy provides new strategies in immunosuppression. Drugs such as FTY720 alter the sensitivity of lymphocytes to homing chemokines, and agents such as sirolimus (SRL) disrupt downstream cytokine signal transduction. Confirming studies in rodents and nonhuman primates, administration of either FTY720 or both of these drugs afford synergistic interactions with cyclosporine to renal transplant patients to rapidly and dramatically deplete peripheral blood lymphocytes (PBL) but neither granulocytes nor monocytes. Present information suggests that FTY720 facilitates lymphocyte homing mechanisms, leading to T and B cell sequestration in secondary lymphoid structures. Interestingly, FTY720 displays pharmacokinetic characteristics suggesting that therapeutic drug monitoring (TDM) will not be essential for clinical applications. In contrast, SRL is a critical-dose drug that requires TDM. SRL disrupts costimulatory and cytokine-stimulated T cell activation by inhibiting a multifunctional kinase, mammalian target of sirolimus (mTOR). Two pivotal trials including more than 1,300 patients demonstrated that addition of SRL to a CsA-based regimen reduces the incidence, time to onset, and severity of acute rejection episodes. When used alone, SRL seems therapeutically equivalent to CsA. In the coming decade, SRL is likely to be used in a variety of drug combination regimens both simultaneously and sequentially, not only to avert acute rejection episodes, but also to forestall chronic nephropathic processes. These two new agents are likely to usher in a new era of transplant therapy.
Collapse
Affiliation(s)
- Barry D Kahan
- University of Texas Medical School at Houston, Department of Surgery, Division of Immunology and Organ Transplantation, Houston, Texas 77030, USA.
| |
Collapse
|
146
|
Krueger JG. The immunologic basis for the treatment of psoriasis with new biologic agents. J Am Acad Dermatol 2002; 46:1-23; quiz 23-6. [PMID: 11756941 DOI: 10.1067/mjd.2002.120568] [Citation(s) in RCA: 380] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Psoriasis vulgaris is the most prevalent T-cell-mediated inflammatory disease in humans. The pathogenesis of psoriasis is linked to activation of several types of leukocytes that control cellular immunity and to a T-cell-dependent inflammatory process in skin that accelerates the growth of epidermal and vascular cells in psoriasis lesions. Critical steps in immunologic activation include Langerhans cell maturation (activation), T-cell activation, differentiation and expansion of type 1 T cells, selective trafficking of activated T cells to skin, and induction of an inflammatory cytokine and chemokine cascade in skin lesions. In turn, each of these steps offers an opportunity for intervention with engineered biologic therapeutics.
Collapse
Affiliation(s)
- James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY 10021-6399, USA.
| |
Collapse
|
147
|
Abstract
The regulation of gene expression by transcription factors is fundamental to the phenotype of all cells. The activated phenotype of cells engaged in inflammatory processes is characterized by induced expression of a diverse set of genes, including cytokines, enzymes and cell adhesion molecules. A relatively small number of inducible transcription factors, particularly NF-kappaB, AP-1, NFATs and STATs, are responsible for the expression of a wide variety of inflammatory phenotypic characteristics and therefore play a central role in the pathogenesis of rheumatic diseases. Each of these transcription factors can be modified by existing anti-rheumatic and anti-inflammatory drugs, although adverse effects and limited efficacy remain problems. The future development of therapeutic agents with specificity for transcription factors, especially NF-kappaB, might lead to safer and more effective treatment.
Collapse
Affiliation(s)
- M L Handel
- Garvan Institute of Medical Research & University of New South Wales, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
| | | |
Collapse
|
148
|
Kolber MA, Saenz MO. Enhancement of antigen driven lymphocyte proliferation secondary to GP41-induced B7 expression on adherent monocytes. Cell Immunol 2001; 214:35-44. [PMID: 11902827 DOI: 10.1006/cimm.2001.1882] [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: 11/22/2022]
Abstract
HIV-1 viral proteins are known to have immune regulatory effects. The interplay between these molecules and the host immune cells is complex. In this study the immune regulatory effects of gp41 on lymphocyte proliferation were evaluated as a function of the state of the monocyte. It is shown that monocyte adherence to tissue culture plates prevents suppression of lymphocyte proliferation to recall antigen in the presence of gp41. In addition, gp41 can enhance proliferation to low concentrations of Casta antigen when PBL are permitted time to adhere. It is shown that these effects are in part mediated through enhanced expression of the costimulatory molecules B7 and CD40. Cyclosporin A was not able to fully abrogate gp41-enhanced proliferation, indicating participation of a calcium-independent pathway. In addition, concentrations of anti-IL2 receptor antibody sufficient to inhibit maximal proliferation to antigen did not fully inhibit PBL proliferation to antigen that is augmented with gp41. Taken together these results suggest that modification of the monocyte state of activation or differentiation could mediate a response to gp41 that is immune enhancing.
Collapse
Affiliation(s)
- M A Kolber
- Department of Medicine, University of Miami School of Medicine, Miami, Florida 33101, USA.
| | | |
Collapse
|
149
|
Vanasek TL, Khoruts A, Zell T, Mueller DL. Antagonistic roles for CTLA-4 and the mammalian target of rapamycin in the regulation of clonal anergy: enhanced cell cycle progression promotes recall antigen responsiveness. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5636-44. [PMID: 11698435 DOI: 10.4049/jimmunol.167.10.5636] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD4(+) T cells that undergo multiple rounds of cell division during primary Ag challenge in vivo produce IL-2 on secondary Ag rechallenge, whereas cells that fail to progress through the cell cycle are anergic to restimulation. Anti-CTLA-4 mAb treatment during primary Ag exposure increases cell cycle progression and enhances recall Ag responsiveness; however, simultaneous treatment with rapamycin, an inhibitor of the mammalian target of rapamycin and potent antiproliferative agent, prevents both effects. The data suggest that cell cycle progression plays a primary role in the regulation of recall Ag responsiveness in CD4(+) T cells in vivo. CTLA-4 molecules promote clonal anergy development only indirectly by limiting cell cycle progression during the primary response.
Collapse
Affiliation(s)
- T L Vanasek
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
150
|
Fayen JD. Multiple cytokines sharing the common receptor gamma chain can induce CD154/CD40 ligand expression by human CD4+ T lymphocytes via a cyclosporin A-resistant pathway. Immunology 2001; 104:299-306. [PMID: 11722644 PMCID: PMC1783314 DOI: 10.1046/j.1365-2567.2001.01296.x] [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: 02/06/2023] Open
Abstract
Expression of CD154/CD40 ligand (CD154/CD40L), an important molecular component of CD4+ T-cell help, can be triggered by T-cell receptor (TCR) stimulation. Dephosphorylation of the transcriptional element Nuclear Factor of Activated T cells-1 (NFAT1) is a critical activation step in the TCR-initiated signal transduction cascade which promotes CD154/CD40L expression. Cyclosporin A (CsA), which interferes with NFAT1 activation, has been shown to be an effective inhibitor of TCR-triggered CD154/CD40L expression by resting T cells. We now report that recombinant interleukin-2 (rIL-2) is also capable of inducing CD154/CD40L on CD4+ T lymphoblasts via a pathway triggered independently of the CD3/TCR receptor complex. Recombinant IL-2-mediated CD154/CD40L expression, in contrast to that triggered by CD3/TCR stimulation, is only partially inhibited by CsA. The capacity of rIL-2 to induce CD154/CD40L expression by T lymphoblasts also extends to a restricted number of cytokines sharing the cytokine receptor common gamma chain, including IL-15, and, to a lesser extent, IL-7, but not IL-4. A similar CsA-resistant CD154/CD40L induction pathway can be triggered in primary T cells by the combination of anti-CD3 stimulation and recombinant lymphokines. In contrast to T lymphoblasts, the CsA-resistant CD154/CD40L induction in primary lymphocytes can be efficiently triggered by multiple cytokines which bind the common gamma chain receptor family. The data outline a novel pathway of CD154/CD40L induction which is, at least in part, independent of NFAT1 and resistant to CsA. A more complete understanding of the mechanisms governing CD154/CD40L expression may facilitate the rational design of specifically targeted immunotherapeutic agents.
Collapse
Affiliation(s)
- J D Fayen
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|