1
|
Li G, Ju J, Weyand CM, Goronzy JJ. Age-Associated Failure To Adjust Type I IFN Receptor Signaling Thresholds after T Cell Activation. THE JOURNAL OF IMMUNOLOGY 2015; 195:865-74. [PMID: 26091718 DOI: 10.4049/jimmunol.1402389] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 05/20/2015] [Indexed: 12/23/2022]
Abstract
With increasing age, naive CD4 T cells acquire intrinsic defects that compromise their ability to respond and differentiate. Type I IFNs, pervasive constituents of the environment in which adaptive immune responses occur, are known to regulate T cell differentiation and survival. Activated naive CD4 T cells from older individuals have reduced responses to type I IFN, a defect that develops during activation and that is not observed in quiescent naive CD4 T cells. Naive CD4 T cells from young adults upregulate the expression of STAT1 and STAT5 after activation, lowering their threshold to respond to type I IFN stimulation. The heightened STAT signaling is critical to maintain the expression of CD69 that regulates lymphocyte egress and the ability to produce IL-2 and to survive. Although activation of T cells from older adults also induces transcription of STAT1 and STAT5, failure to exclude SHP-1 from the signaling complex blunts their type I IFN response. In summary, our data show that type I IFN signaling thresholds in naive CD4 T cells after activation are dynamically regulated to respond to environmental cues for clonal expansion and memory cell differentiation. Naive CD4 T cells from older adults have a defect in this threshold calibration. Restoring their ability to respond to type I IFN emerges as a promising target to restore T cell responses and to improve the induction of T cell memory.
Collapse
Affiliation(s)
- Guangjin Li
- Department of Medicine, Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304; and Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jihang Ju
- Department of Medicine, Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304; and Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Cornelia M Weyand
- Department of Medicine, Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304; and Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jörg J Goronzy
- Department of Medicine, Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304; and Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
2
|
Boivin N, Baillargeon J, Doss PMIA, Roy AP, Rangachari M. Interferon-β suppresses murine Th1 cell function in the absence of antigen-presenting cells. PLoS One 2015; 10:e0124802. [PMID: 25885435 PMCID: PMC4401451 DOI: 10.1371/journal.pone.0124802] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/19/2015] [Indexed: 01/08/2023] Open
Abstract
Interferon (IFN)-β is a front-line therapy for the treatment of the relapsing-remitting form of multiple sclerosis. However, its immunosuppressive mechanism of function remains incompletely understood. While it has been proposed that IFN-β suppresses the function of inflammatory myelin antigen-reactive T cells by promoting the release of immunomodulatory cytokines such as IL-27 from antigen-presenting cells (APCs), its direct effects on inflammatory CD4+ Th1 cells are less clear. Here, we establish that IFN-β inhibits mouse IFN-γ+ Th1 cell function in the absence of APCs. CD4+ T cells express the type I interferon receptor, and IFN-β can suppress Th1 cell proliferation under APC-free stimulation conditions. IFN-β-treated myelin antigen-specific Th1 cells are impaired in their ability to induce severe experimental autoimmune encephalomyelitis (EAE) upon transfer to lymphocyte-deficient Rag1-/- mice. Polarized Th1 cells downregulate IFN-γ and IL-2, and upregulate the negative regulatory receptor Tim-3, when treated with IFN-β in the absence of APCs. Further, IFN-β treatment of Th1 cells upregulates phosphorylation of Stat1, and downregulates phosphorylation of Stat4. Our data indicate that IFN-γ-producing Th1 cells are directly responsive to IFN-β and point to a novel mechanism of IFN-β-mediated T cell suppression that is independent of APC-derived signals.
Collapse
Affiliation(s)
- Nicolas Boivin
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
| | - Joanie Baillargeon
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
| | - Prenitha Mercy Ignatius Arokia Doss
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Graduate Programme in Microbiology and Immunology, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
| | - Andrée-Pascale Roy
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Graduate Programme in Microbiology and Immunology, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
| | - Manu Rangachari
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
- * E-mail:
| |
Collapse
|
3
|
Lee TH, Lee KO, Kim YS, Kim SM, Huh KC, Choi YW, Kang YW. Cryptococcal meningitis in a patient with chronic hepatitis C treated with pegylated-interferon and ribavirin. Korean J Intern Med 2014; 29:370-4. [PMID: 24851072 PMCID: PMC4028527 DOI: 10.3904/kjim.2014.29.3.370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 06/22/2012] [Accepted: 08/06/2012] [Indexed: 11/27/2022] Open
Abstract
Various adverse events have been reported during combination therapy with pegylated (PEG)-interferon-α and ribavirin, although opportunistic infections, especially cryptococcal meningitis, are very rare. A 61-year-old woman complained of headaches and a fever during treatment of a chronic hepatitis C virus (HCV) infection. She had been treated for 7 months. Her headaches were refractory to analgesics, and she developed subtle nuchal rigidity. The cerebral spinal fluid (CSF) revealed a white blood cell count of 205/mm(3), 51 mg/dL protein, 35 mg/dL glucose, and negative Cryptococcus antigen. The CSF culture resulted in no growth. Five days later, the CSF was positive for Cryptococcus antigen. We administered amphotericin B and flucytosine, followed by fluconazole. Approximately 2 months later, she was discharged. For the first time, we report a case of cryptococcal meningitis during the treatment of chronic HCV with PEG-interferon-α and ribavirin.
Collapse
Affiliation(s)
- Tae-Hee Lee
- Department of Internal Medicine, Konyang University College of Medicine, Daejeon, Korea
| | - Kee-Ook Lee
- Department of Neurology, Konyang University College of Medicine, Daejeon, Korea
| | - Yong-Seok Kim
- Department of Internal Medicine, Konyang University College of Medicine, Daejeon, Korea
| | - Sun-Moon Kim
- Department of Internal Medicine, Konyang University College of Medicine, Daejeon, Korea
| | - Kyu-Chan Huh
- Department of Internal Medicine, Konyang University College of Medicine, Daejeon, Korea
| | - Young-Woo Choi
- Department of Internal Medicine, Konyang University College of Medicine, Daejeon, Korea
| | - Young-Woo Kang
- Department of Internal Medicine, Konyang University College of Medicine, Daejeon, Korea
| |
Collapse
|
4
|
Sellebjerg F, Krakauer M, Limborg S, Hesse D, Lund H, Langkilde A, Søndergaard HB, Sørensen PS. Endogenous and recombinant type I interferons and disease activity in multiple sclerosis. PLoS One 2012; 7:e35927. [PMID: 22701554 PMCID: PMC3368920 DOI: 10.1371/journal.pone.0035927] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 03/24/2012] [Indexed: 11/18/2022] Open
Abstract
Although treatment of multiple sclerosis (MS) with the type I interferon (IFN) IFN-β lowers disease activity, the role of endogenous type I IFN in MS remains controversial. We studied CD4+ T cells and CD4+ T cell subsets, monocytes and dendritic cells by flow cytometry and analysed the relationship with endogenous type I IFN-like activity, the effect of IFN-β therapy, and clinical and magnetic resonance imaging (MRI) disease activity in MS patients. Endogenous type I IFN activity was associated with decreased expression of the integrin subunit CD49d (VLA-4) on CD4+CD26(high) T cells (Th1 helper cells), and this effect was associated with less MRI disease activity. IFN-β therapy reduced CD49d expression on CD4+CD26(high) T cells, and the percentage of CD4+CD26(high) T cells that were CD49d(high) correlated with clinical and MRI disease activity in patients treated with IFN-β. Treatment with IFN-β also increased the percentage of CD4+ T cells expressing CD71 and HLA-DR (activated T cells), and this was associated with an increased risk of clinical disease activity. In contrast, induction of CD71 and HLA-DR was not observed in untreated MS patients with evidence of endogenous type IFN I activity. In conclusion, the effects of IFN-β treatment and endogenous type I IFN activity on VLA-4 expression are similar and associated with control of disease activity. However, immune-activating effects of treatment with IFN-β may counteract the beneficial effects of treatment and cause an insufficient response to therapy.
Collapse
Affiliation(s)
- Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Hervas-Stubbs S, Riezu-Boj JI, Gonzalez I, Mancheño U, Dubrot J, Azpilicueta A, Gabari I, Palazon A, Aranguren A, Ruiz J, Prieto J, Larrea E, Melero I. Effects of IFN-α as a signal-3 cytokine on human naïve and antigen-experienced CD8(+) T cells. Eur J Immunol 2011; 40:3389-402. [PMID: 21108462 DOI: 10.1002/eji.201040664] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
IFN-α/β link innate and adaptive immune responses by directly acting on naïve CD8(+) T cells. This concept unveiled in mice remains unexplored in humans. To investigate that, human CD8(+) CD45RO(-) cells were stimulated with beads coated with anti-CD3 and anti-CD28 mAb, mimicking Ag (type-1) and co-stimulatory (type-2) signals, in the presence or absence of IFN-α and their transcriptional profiles were defined by cDNA-microarrays. We show that IFN-α provides a strong third signal directly to human CD8(+) T cells resulting in regulation of critical genes for their overall activation. This transcriptional effect was substantiated at the protein level and verified by functional assays. Interestingly, the biological effects derived from this stimulation vary depending on the CD8(+) T-cell population. Thus, whereas IFN-α increases the proliferative capacity of naïve CD8(+) T cells, it inhibits or does not affect the proliferation of Ag-experienced cells, such as memory and effector CTL, including CMV-specific lymphocytes. Cytolysis and IFN-γ-secretion of all these populations are enhanced by IFN-α-derived signals, which are critical in naïve CD8(+) T cells for acquisition of effector functions. Our findings in human CD8(+) T cells are informative to understand and improve IFN-α-based therapies for viral and malignant diseases.
Collapse
Affiliation(s)
- Sandra Hervas-Stubbs
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Libri V, Azevedo RI, Jackson SE, Di Mitri D, Lachmann R, Fuhrmann S, Vukmanovic-Stejic M, Yong K, Battistini L, Kern F, Soares MVD, Akbar AN. Cytomegalovirus infection induces the accumulation of short-lived, multifunctional CD4+CD45RA+CD27+ T cells: the potential involvement of interleukin-7 in this process. Immunology 2011; 132:326-39. [PMID: 21214539 DOI: 10.1111/j.1365-2567.2010.03386.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The relative roles that ageing and lifelong cytomegalovirus (CMV) infection have in shaping naive and memory CD4+ T-cell repertoires in healthy older people is unclear. Using multiple linear regression analysis we found that age itself is a stronger predictor than CMV seropositivity for the decrease in CD45RA+ CD27+ CD4+ T cells over time. In contrast, the increase in CD45RA⁻ CD27⁻ and CD45RA+ CD27⁻ CD4+ T cells is almost exclusively the result of CMV seropositivity, with age alone having no significant effect. Furthermore, the majority of the CD45RA⁻ CD27⁻ and CD45RA+ CD27⁻ CD4+ T cells in CMV-seropositive donors are specific for this virus. CD45RA+ CD27⁻ CD4+ T cells have significantly reduced CD28, interleukin-7 receptor α (IL-7Rα) and Bcl-2 expression, Akt (ser473) phosphorylation and reduced ability to survive after T-cell receptor activation compared with the other T-cell subsets in the same donors. Despite this, the CD45RA+ CD27⁻ subset is as multifunctional as the CD45RA⁻ D27+ and CD45RA⁻ CD27⁻ CD4+ T-cell subsets, indicating that they are not an exhausted population. In addition, CD45RA+ CD27⁻ CD4+ T cells have cytotoxic potential as they express high levels of granzyme B and perforin. CD4+ memory T cells re-expressing CD45RA can be generated from the CD45RA⁻ CD27+ population by the addition of IL-7 and during this process these cells down-regulated expression of IL-7R and Bcl-2 and so resemble their counterparts in vivo. Finally we showed that the proportion of CD45RA+ CD27⁻ CD4+ T cells of multiple specificities was significantly higher in the bone marrow than the blood of the same individuals, suggesting that this may be a site where these cells are generated.
Collapse
Affiliation(s)
- Valentina Libri
- Division of Infection and Immunity, University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Vukmanovic-Stejic M, Agius E, Booth N, Dunne PJ, Lacy KE, Reed JR, Sobande TO, Kissane S, Salmon M, Rustin MH, Akbar AN. The kinetics of CD4+Foxp3+ T cell accumulation during a human cutaneous antigen-specific memory response in vivo. J Clin Invest 2008; 118:3639-50. [PMID: 18924611 DOI: 10.1172/jci35834] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Accepted: 08/20/2008] [Indexed: 12/27/2022] Open
Abstract
Naturally occurring CD4(+)CD25(hi)Foxp3(+) Tregs (nTregs) are highly proliferative in blood. However, the kinetics of their accumulation and proliferation during a localized antigen-specific T cell response is currently unknown. To explore this, we used a human experimental system whereby tuberculin purified protein derivative (PPD) was injected into the skin and the local T cell response analyzed over time. The numbers of both CD4(+)Foxp3(-) (memory) and CD4(+)Foxp3(+) (putative nTreg) T cells increased in parallel, with the 2 populations proliferating at the same relative rate. In contrast to CD4(+)Foxp3(-) T cell populations, skin CD4(+)Foxp3(+) T cells expressed typical Treg markers (i.e., they were CD25(hi), CD127(lo), CD27(+), and CD39(+)) and did not synthesize IL-2 or IFN-gamma after restimulation in vitro, indicating that they were not recently activated effector cells. To determine whether CD4(+)Foxp3(+) T cells in skin could be induced from memory CD4(+) T cells, we expanded skin-derived memory CD4(+) T cells in vitro and anergized them. These cells expressed high levels of CD25 and Foxp3 and suppressed the proliferation of skin-derived responder T cells to PPD challenge. Our data therefore demonstrate that memory and CD4(+) Treg populations are regulated in tandem during a secondary antigenic response. Furthermore, it is possible to isolate effector CD4(+) T cell populations from inflamed tissues and manipulate them to generate Tregs with the potential to suppress inflammatory responses.
Collapse
|
8
|
Martín-Saavedra FM, González-García C, Bravo B, Ballester S. Beta interferon restricts the inflammatory potential of CD4+ cells through the boost of the Th2 phenotype, the inhibition of Th17 response and the prevalence of naturally occurring T regulatory cells. Mol Immunol 2008; 45:4008-19. [DOI: 10.1016/j.molimm.2008.06.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 06/05/2008] [Accepted: 06/08/2008] [Indexed: 10/21/2022]
|
9
|
Quigley M, Huang X, Yang Y. STAT1 signaling in CD8 T cells is required for their clonal expansion and memory formation following viral infection in vivo. THE JOURNAL OF IMMUNOLOGY 2008; 180:2158-64. [PMID: 18250422 DOI: 10.4049/jimmunol.180.4.2158] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent advances have shown that direct type I IFN signaling on T cells is required for their efficient expansion in response to viral infections in vivo. It is not clear which intracellular signaling molecule is responsible for this effect. Although STAT1 has been shown to mediate many of the type I IFN-dependent biological effects, its role in T cells remains uncertain in vivo. In this study, we demonstrated that STAT1 signaling in CD8 T cells was required for their efficient expansion by promoting the survival of activated CD8 T cells upon vaccinia viral infection in vivo, suggesting that the direct effect of type I IFNs on CD8 T cells is mediated by STAT1. Furthermore, effector CD8 T cells that lack STAT1 signaling did not survive the contraction phase to differentiate into long-lived memory cells. These results identify a critical role for type I IFN-STAT1 signaling in multiple stages of CD8 T cell response in vivo and suggest that strategies to activate type I IFN-STAT1 signaling pathway may enhance vaccine potency.
Collapse
Affiliation(s)
- Michael Quigley
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
10
|
Gröschel S, Piggott KD, Vaglio A, Ma-Krupa W, Singh K, Goronzy JJ, Weyand CM. TLR-mediated induction of negative regulatory ligands on dendritic cells. J Mol Med (Berl) 2008; 86:443-55. [PMID: 18253710 DOI: 10.1007/s00109-008-0310-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Revised: 12/21/2007] [Accepted: 01/07/2008] [Indexed: 12/31/2022]
Abstract
Dendritic cells (DCs) shape T-cell response patterns and determine early, intermediate, and late outcomes of immune recognition events. They either facilitate immunostimulation or induce tolerance, possibly determined by initial DC activation signals, such as binding Toll-like receptor (TLR) ligands. Here, we report that DC stimulation through the TLR3 ligand dsRNA [poly(I:C)] limits CD4 T-cell proliferation, curtailing adaptive immune responses. CD4+ T cells instructed by either lipopolysaccharide (LPS) or poly(I:C)-conditioned DCs promptly upregulated the activation marker CD69. Whereas LPS-pretreated DCs subsequently sustained T-cell clonal expansion, proliferation of CD4+ T cells exposed to poly(I:C)-pretreated DCs was markedly suppressed. This proliferative defect required DC-T cell contact, was independent of IFN-alpha, and was overcome by exogenous IL-2, indicating T-cell anergy. Coinciding with the downregulation, CD4+ T cells expressed the inhibitory receptor PD-1. Antibodies blocking the PD-1 ligand PD-L1 restored proliferation. dsRNA-stimulated DCs preferentially induced PD-L1, whereas poly(I:C) and LPS both upregulated the costimulatory molecule CD86 to a comparable extent. Poly(dA-dT), a ligand targeting the cytoplasmic RNA helicase pattern-recognition pathway, failed to selectively induce PD-L1 upregulation, assigning this effect to the TLR3 pathway. Poly(I:C)-conditioned DCs promoted accumulation of phosphorylated SHP-2, the intracellular phosphatase mediating PD-1 inhibitory effects. The ability of dsRNA to bias DC differentiation toward providing inhibitory signals to interacting CD4+ T cells may be instrumental in viral immune evasion. Conversely, TLR3 ligands may have therapeutic value in silencing pathogenic immune responses.
Collapse
Affiliation(s)
- Stefan Gröschel
- Kathleen B. and Mason I. Lowance Center for Human Immunology, Department of Medicine, Emory University School of Medicine, Room 1003 Woodruff Memorial Research Building, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Darveau ME, Jacques E, Rouabhia M, Hamid Q, Chakir J. Increased T-cell survival by structural bronchial cells derived from asthmatic subjects cultured in an engineered human mucosa. J Allergy Clin Immunol 2008; 121:692-9. [PMID: 18194810 DOI: 10.1016/j.jaci.2007.11.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Revised: 11/16/2007] [Accepted: 11/27/2007] [Indexed: 11/24/2022]
Abstract
BACKGROUND Interaction between lymphocytes and structural cells has been proposed as a key factor in regulating inflammation in asthma. OBJECTIVE This study was designed to investigate the effect of epithelial cells and fibroblasts on T-lymphocyte survival by using a 3-dimensional tissue-engineered model. METHODS Engineered human bronchial mucosal tissues were produced by using fibroblasts, epithelial cells, and autologous T cells from asthmatic and healthy donors. T-cell apoptosis and apoptotic marker expression by T cells were evaluated by using the terminal deoxynucleotidyl transferase biotinylated d-UTP nick end-labeling technique and immunofluorescence, respectively. Cytokines implicated in T-cell survival were measured by means of ELISA in culture supernatants. RESULTS We demonstrated histologically that we were able to generate a well-structured engineered bronchial mucosa by using epithelial cells, fibroblasts, and T cells cultured from healthy and asthmatic subjects. Structural cells from asthmatic subjects cultured in this model induced a significant decrease in the ability of T cells to undergo apoptosis represented by a decrease in DNA fragmentation and proapoptotic molecule expression (Bcl-2-associated X protein and Fas ligand). Structural cells from healthy control subjects have no effect. Among cytokines measured in the supernatants, only TGF-beta(1) was significantly increased in the model derived from cells of asthmatic subjects. CONCLUSION These results support the concept that bronchial structural cells might play a critical role in the regulation of inflammation in asthma by increasing the survival of T lymphocytes. The results also further validated the model as a tool for investigating the interaction between inflammatory and structural cells.
Collapse
Affiliation(s)
- Marie-Eve Darveau
- Centre de Recherche, Hôpital Laval, Institut Universitaire de Cardiologie et de Pneumology, Laval, Quebec, Canada
| | | | | | | | | |
Collapse
|
12
|
Rodriguez B, Lederman MM, Jiang W, Bazdar DA, Gàrate K, Harding CV, Sieg SF. Interferon-alpha differentially rescues CD4 and CD8 T cells from apoptosis in HIV infection. AIDS 2006; 20:1379-89. [PMID: 16791012 DOI: 10.1097/01.aids.0000233571.51899.ab] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To examine the effects of interferon-alpha (IFN-alpha) on T cell survival and activation in HIV infection. DESIGN The effects of IFN-alpha on spontaneous apoptosis and CD38 expression among T cell subsets were determined in vitro and studied in relation to CD4 cell counts, plasma HIV RNA levels and the age of the subjects. METHODS Peripheral blood mononuclear cells from 48 HIV-infected persons and 17 healthy donors were incubated in vitro overnight with or without the addition of IFN-alpha. Percentages of apoptotic cells (positive for annexin V) and CD38 cells were determined among T cell subsets. RESULTS IFN-alpha inhibited spontaneous apoptosis of CD4 and CD8 T lymphocytes. This protective activity was impaired in CD4 T cells from HIV-infected persons. The reduced protection of IFN-alpha among CD4 cells from HIV-infected persons was not related to the percentages of activated (CD38 or CD45RO+CD38+) cells. Surprisingly, IFN-alpha induced CD38 expression among CD8 T cells from HIV-infected persons, and the magnitude of this effect was directly related to circulating CD4 T cell count. The CD8 T cell subset that expressed CD38 in response to IFN-alpha was defined as CD28 negative, CD62 ligand (CD62L) intermediate/negative. CONCLUSIONS Heightened expression of IFN-alpha in HIV infection may contribute to the phenotypic activation state that characterizes chronic infection while a diminished responsiveness of CD4 T cells to the protective effect of this cytokine may contribute to differential survival of CD4 and CD8 T cells in HIV disease.
Collapse
Affiliation(s)
- Benigno Rodriguez
- Division of Infectious Diseases, Case Western Reserve University, Center foe AIDS Research, University Hospitals of Cleveland, Cleveland, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Aichele P, Unsoeld H, Koschella M, Schweier O, Kalinke U, Vucikuja S. CD8 T cells specific for lymphocytic choriomeningitis virus require type I IFN receptor for clonal expansion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 176:4525-9. [PMID: 16585541 DOI: 10.4049/jimmunol.176.8.4525] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of type I IFN signaling in CD8 T cells was analyzed in an adoptive transfer model using P14 TCR transgenic CD8 T cells specific for lymphocytic choriomeningitis virus (LCMV) but deficient in type I IFNR. In the present study, we demonstrate severe impairment in the capacity of P14 T cells lacking type I IFNR to expand in normal type I IFNR wild-type C57BL/6 hosts after LCMV infection. In contrast, following infection of recipient mice with recombinant vaccinia virus expressing LCMV glycoprotein, P14 T cell expansion was considerably less dependent on type I IFNR expression. Lack of type I IFNR expression by P14 T cells did not affect cell division after LCMV infection but interfered with clonal expansion. Thus, direct type I IFN signaling is essential for CD8 T cell survival in certain viral infections.
Collapse
Affiliation(s)
- Peter Aichele
- Institute for Medical Microbiology and Hygiene, Department of Immunology, University of Freiburg, Freiburg, Germany.
| | | | | | | | | | | |
Collapse
|
14
|
Fischer U, Utke K, Somamoto T, Köllner B, Ototake M, Nakanishi T. Cytotoxic activities of fish leucocytes. FISH & SHELLFISH IMMUNOLOGY 2006; 20:209-26. [PMID: 15939625 DOI: 10.1016/j.fsi.2005.03.013] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2004] [Revised: 03/03/2005] [Accepted: 03/03/2005] [Indexed: 05/02/2023]
Abstract
Like mammalian leucocytes, white blood cells of fish are able to kill altered (e.g. virus-infected) and foreign (allogeneic or xenogeneic) cells. The existence of natural killer (NK)-like and specific cytotoxic cells in fish was first shown using allogeneic and xenogeneic effector/target cell systems. In addition to in vivo and ex vivo studies, very important contributions were made by in vitro analysis using a number of different long-term cytotoxic cell lines established from channel catfish. In mammals, specific cell-mediated cytotoxicity (CMC) as part of the adaptive immune response requires a number of key molecules expressed on effector leucocytes and target cells. CD8+ T lymphocytes kill infected cells only, if their antigen receptor (TCR) matches the MHC class I with bound peptide of the target cell. Expression patterns of the fish gene homologues for TCR, CD8 and MHC class I, as well as related genes, are in agreement with similar function. Convenient systems for the analysis of specific CMC have only recently become available for fish with the combination of clonal fish with syngeneic or allogeneic but MHC class I matching cell lines. It was demonstrated that both, NK- and cytotoxic T (Tc) cells are involved in the killing of virus infected MHC class I matching and mismatching target cells. Analysis of these lymphocyte subsets is only starting for fish. There is also evidence that the different viral proteins trigger different subsets of killer cells. This review further discusses findings on fish CMC with regard to temperature/seasons and ontogeny.
Collapse
Affiliation(s)
- Uwe Fischer
- Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, D-17493 Greifswald-Insel Riems, Germany.
| | | | | | | | | | | |
Collapse
|
15
|
Schwarting A, Paul K, Tschirner S, Menke J, Hansen T, Brenner W, Kelley VR, Relle M, Galle PR. Interferon-β: A Therapeutic for Autoimmune Lupus in MRL-Faslpr Mice. J Am Soc Nephrol 2005; 16:3264-72. [PMID: 16221871 DOI: 10.1681/asn.2004111014] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Type I interferons are associated with lupus. Genes that are regulated by IFN-alpha are upregulated in pediatric lupus patients. Gene deletion of the IFN-alpha/beta receptor in experimental lupus-like NZB mice results in reduced disease activity. Conversely, IFN-beta is a well-established treatment in multiple sclerosis, another autoimmune disease. For determining whether IFN-beta treatment is harmful or beneficial in lupus, MRL-Fas(lpr) mice were injected with this type I IFN. Treatment was initiated in MRL-Fas(lpr) mice with mild and advanced disease. IFN-beta was highly effective in prolonging survival and ameliorating the clinical (renal function, proteinuria, splenomegaly, and skin lesions), serologic (autoantibodies and cytokines), and histologic parameters of the lupus-like disease in mice that had mild and advanced disease. Several underlying mechanisms of IFN-beta therapy involving cellular (decreased T cell proliferation and infiltration of leukocytes into the kidney) and humoral (decrease in IgG3 isotypes) immune responses and a reduction in nephrogenic cytokines were identified. In conclusion, IFN-beta treatment of lupus nephritis in MRL-Fas(lpr) mice is remarkably beneficial and suggests that IFN-beta may be an appealing therapeutic candidate for subtypes of human lupus.
Collapse
Affiliation(s)
- Andreas Schwarting
- First Department of Medicine, Johannes-Gutenberg University of Mainz, Langenbeckstrasse 1, Mainz 55131, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
Chronic myelogenous leukemia (CML) was the first human malignancy where a consistent chromosomal abnormality, the BCR-ABL translocation, was identified as the causative genetic aberration. There is a mounting body of evidence suggesting that CML cells are particularly good targets for immunological surveillance mechanisms, the most intriguing being the curative effect of allogeneic donor lymphocyte infusion given in relapsed disease after allogeneic bone marrow transplantation. Likewise, interferon alpha (IFN alpha), which has long been considered as the standard conservative therapy in CML, may exert its life-prolonging effect by activating immunological effector functions. This review will focus on the recent advances in the understanding of the contribution of IFN alpha in eliciting T-cell responses against self-antigens in CML.
Collapse
Affiliation(s)
- Andreas Burchert
- Klinikum der Philipps Universität Marburg, Klinik für Hämatologie, Onkologie und Immunologie, Marburg, Germany
| | | |
Collapse
|
17
|
Ina K, Kusugami K, Kawano Y, Nishiwaki T, Wen Z, Musso A, West GA, Ohta M, Goto H, Fiocchi C. Intestinal Fibroblast-Derived IL-10 Increases Survival of Mucosal T Cells by Inhibiting Growth Factor Deprivation- and Fas-Mediated Apoptosis. THE JOURNAL OF IMMUNOLOGY 2005; 175:2000-9. [PMID: 16034145 DOI: 10.4049/jimmunol.175.3.2000] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mucosal T cells are essential to immune tolerance in the intestine, an organ constantly exposed to large amounts of dietary and bacterial Ags. We investigated whether local fibroblasts affect mucosal T cell survival, which is critical for maintenance of immune tolerance. Coculture with autologous fibroblasts significantly increased viability of mucosal T cells by inhibiting IL-2 deprivation- and Fas-mediated apoptosis, an effect that was both contact- and secreted product-dependent. Investigation of anti-apoptotic factors in the fibroblast-conditioned medium (FCM) revealed the presence of IL-10 and PGE2, but not IFN-beta, IL-2, or IL-15. Although recombinant IFN-beta, but not PGE2, effectively prevented T cell apoptosis, neutralizing Ab studies showed that only IL-10 blockade significantly increased T cells apoptosis, whereas neutralizing IFN-beta or IFN-alpha failed to inhibit the anti-apoptotic effect of FCM. To confirm that fibroblast-derived IL-10 was responsible for preserving mucosal T cell viability, IL-10 mRNA was demonstrated in fibroblasts by Southern blotting and RT-PCR. When FCM was submitted to HPLC fractionation, only the peak matching rIL-10 contained the anti-apoptotic activity, and this was eliminated by treatment with an IL-10-neutralizing Ab. Finally, when fibroblasts were transiently transfected with IL-10 antisense oligonucleotides, the conditioned medium lost its T cell anti-apoptotic effect, whereas medium from fibroblasts transfected with IFN-beta antisense oligonucleotides displayed the same anti-apoptotic activity of medium from untransfected fibroblasts. These results indicate that local fibroblast-derived IL-10 is critically involved in the survival of mucosal T cells, underscoring the crucial importance of studying organ-specific cells and products to define the mechanisms of immune homeostasis in specialized tissue microenvironments like the intestinal mucosa.
Collapse
Affiliation(s)
- Kenji Ina
- Division of Medical Oncology, Nagoya Memorial Hospital, Nagoya University School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Ueta M, Hamuro J, Kiyono H, Kinoshita S. Triggering of TLR3 by polyI:C in human corneal epithelial cells to induce inflammatory cytokines. Biochem Biophys Res Commun 2005; 331:285-94. [PMID: 15845391 DOI: 10.1016/j.bbrc.2005.02.196] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Indexed: 01/22/2023]
Abstract
Epithelial cells of the ocular surface are key in the first-line defense as a part of the mucosal immune system against pathogens. We investigated whether polyI:C induces the production by human corneal epithelial cells (HCEC) of pro-inflammatory cytokines and IFN-beta, and whether Toll-like receptor (TLR)-3 expression is amplified by polyI:C. TLR3 was expressed on the surface of HCEC. Stimulation with polyI:C elicited the elevated production and mRNA expression of IL-6 and IL-8 in HCEC. While polyI:C induced IFN-beta, far stronger than human fibroblasts, and TLR3 gene expression in HCEC, LPS stimulation did not. Similarly, polyI:C, but not LPS, induced the gene expression of IkappaBalpha and MAIL, members of the IkappaB family, in HCEC. The innate immune response of HCEC is distinct from that of immune-competent cells, and we suggest that this is indicative of the symbiotic relationship between corneal epithelium and microbes inhabiting the ocular surface.
Collapse
Affiliation(s)
- Mayumi Ueta
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | | | | | | |
Collapse
|
19
|
Affiliation(s)
- Ludwig Kappos
- Department of Neurology, University Hospitals, Kantonsspital, CH-4031 Basel, Switzerland.
| |
Collapse
|
20
|
Vieira PL, Christensen JR, Minaee S, O'Neill EJ, Barrat FJ, Boonstra A, Barthlott T, Stockinger B, Wraith DC, O'Garra A. IL-10-Secreting Regulatory T Cells Do Not Express Foxp3 but Have Comparable Regulatory Function to Naturally Occurring CD4+CD25+ Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2004; 172:5986-93. [PMID: 15128781 DOI: 10.4049/jimmunol.172.10.5986] [Citation(s) in RCA: 483] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Regulatory T cells (T(Reg)) control immune responses to self and nonself Ags. The relationship between Ag-driven IL-10-secreting T(Reg) (IL-10-T(Reg)) and naturally occurring CD4(+)CD25(+) T(Reg) is as yet unclear. We show that mouse IL-10-T(Reg) obtained using either in vitro or in vivo regimens of antigenic stimulation did not express the CD4(+)CD25(+) T(Reg)-associated transcription factor Foxp3. However, despite the absence of Foxp3 expression, homogeneous populations of IL-10-T(Reg) inhibited the in vitro proliferation of CD4(+)CD25(-) T cells with a similar efficiency to that of CD4(+)CD25(+) T(Reg). This inhibition of T cell proliferation by IL-10-T(Reg) was achieved through an IL-10-independent mechanism as seen for CD4(+)CD25(+) T(Reg) and was overcome by exogenous IL-2. Both IL-10-T(Reg) and CD4(+)CD25(+) T(Reg) were similar in that they produced little to no IL-2. These data show that Foxp3 expression is not a prerequisite for IL-10-T(Reg) activity in vitro or in vivo, and suggest that IL-10-T(Reg) and naturally occurring CD4(+)CD25(+) T(Reg) may have distinct origins.
Collapse
Affiliation(s)
- Pedro L Vieira
- Division of Immunoregulation, The National Institute for Medical Research, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Adams VC, Hunt JRF, Martinelli R, Palmer R, Rook GAW, Brunet LR. Mycobacterium vaccae
induces a population of pulmonary CD11c+
cells with regulatory potential in allergic mice. Eur J Immunol 2004; 34:631-638. [PMID: 14991592 DOI: 10.1002/eji.200324659] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The hygiene hypothesis proposes that common, harmless microorganisms, present throughout our evolutionary history, have helped to develop immunoregulatory mechanisms that prevent inappropriate immune responses by the host. Using a mouse model of allergic pulmonary inflammation, we report that treatment with an ubiquitous saprophytic mycobacterium, Mycobacterium vaccae, significantly reduces allergic inflammation by decreasing type 2 responses such as eosinophilia and IL-4 expression. Rather than observing an increase in type-1 cytokine expression, we found elevated production of IL-10 in the lungs suggesting a role for regulatory T cells. Since induction of these cells may be dependent on APC, we investigated the effects of M. vaccae treatment on pulmonary CD11c+ cells. Increased levels of IL-10, TGF-beta and IFN-alpha mRNA were detected in CD11c+ cells from M. vaccae-treated allergic mice. We propose that M. vaccae-induced CD11c+ cells have a potential regulatory role at the site of inflammation through their secretion of immunomodulatory cytokines.
Collapse
Affiliation(s)
- Victoria C Adams
- University College London, Department of Medical Microbiology, Windeyer Institute of Medical Sciences, London, GB
| | | | | | | | - Graham A W Rook
- University College London, Department of Medical Microbiology, Windeyer Institute of Medical Sciences, London, GB
| | | |
Collapse
|
22
|
Zhou YW, Aritake S, Tri Endharti A, Wu J, Hayakawa A, Nakashima I, Suzuki H. Murine lymph node-derived stromal cells effectively support survival but induce no activation/proliferation of peripheral resting T cells in vitro. Immunology 2003; 109:496-503. [PMID: 12871215 PMCID: PMC1783011 DOI: 10.1046/j.1365-2567.2003.01693.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Little is known about the homeostatic mechanisms by which the levels of peripheral lymphocytes are maintained. The survival of naïve T cells in vivo must be maintained by some factors that have not been characterized in an in vitro culture system. In this study, we established a culture system of stromal cells derived from murine lymph nodes and investigated the action of the stromal cells in supporting the survival of resting T cells in vitro. Most of the T cells cocultured with the stromal cells did not die, and the supernatant of cultured stromal cells increase the viability of T cells. This T-cell survival-supporting activity was maintained for more than 7 days. Although interleukin (IL)-4, IL-6, IL-7, and interferon-beta also rescued peripheral T cells from spontaneous cell death, medium-soluble and heat-sensitive factor(s) derived from the stromal cells supported the survival of T cells more effectively and for a longer time than did these cytokines. T cells maintained in the culture system with the stromal cells appeared to remain in a resting G0/G1 state and did not show remarkable DNA synthesis. From these results, it is presumed that some soluble factor(s) other than the tested cytokines that have been identified as supporting T-cell survival are produced from lymph node stromal cells. These factor(s) play an important role in maintenance of resting T cells.
Collapse
Affiliation(s)
- Yan-Wen Zhou
- Departments of Immunology and Equipment Center for Research and Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
23
|
Akbar AN, Taams LS, Salmon M, Vukmanovic-Stejic M. The peripheral generation of CD4+ CD25+ regulatory T cells. Immunology 2003; 109:319-25. [PMID: 12807474 PMCID: PMC1782989 DOI: 10.1046/j.1365-2567.2003.01678.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2003] [Accepted: 03/28/2003] [Indexed: 01/24/2023] Open
Affiliation(s)
- Arne N Akbar
- Department of Immunology and Molecular Pathology, Windeyer Institute for Medical Sciences, Royal Free and University College Medical School, London, UK.
| | | | | | | |
Collapse
|
24
|
Abstract
Plasmacytoid dendritic cells (PDCs) or natural interferon-producing cells, function as the body's innate defense against viral infections. As discussed here, they may play additional roles in response to bacterial pathogens and may have the capacity to induce different type of T-cell responses depending on what signals they receive. The discovery of murine PDCs will allow for the design of models to study viral immunobiology in vivo and to determine their function in various diseases that involve plasmacytoid dendritic cells, such as selected leukemias, lymphomas, allergies, different autoimmune conditions, and their possible role in inducing and maintaining tolerance.
Collapse
Affiliation(s)
- Pia Björck
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
25
|
Tang X, Yocum DE, DeJonghe D, Nordensson K. Characterizing a soluble survival signal for activated lymphocytes from CD14+ cells. Immunology 2002; 107:56-68. [PMID: 12225363 PMCID: PMC1782776 DOI: 10.1046/j.1365-2567.2002.01463.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
T-cell activation requires at least two signals: antigen and a costimulatory signal. As antigen-presenting cells play an important role in this area, the role of CD14+ cells in T-cell activation, proliferation and activation-induced cell death (AICD) was investigated. Using phytohaemagglutinin (PHA) activation, it was found that CD14+ cell depletion resulted in significantly greater AICD, decreased lymphocyte growth and up-regulated interleukin-2 (IL-2) secretion. However, T-cell activation was delayed according to the expression of CD69 and CD25. Dynabeads conjugated with anti-CD14 monoclonal antibody (mAb) bound CD14+ cells and induced secretion of IL-1beta, tumour necrosis factor-alpha (TNF-alpha), transforming growth factor-beta (TGF-beta) and IL-6, but not IL-2, IL-12 or IL-15. Supernatants were collected from Dynabeads-activated CD14+ cell cultures and designated as 'CD14 cocktails'. Addition of CD14 cocktails to CD14+ cell-depleted mononuclear cell cultures reversed the increased AICD, decreased lymphocyte growth and increased IL-2 secretion. Depletion of IL-1beta and TNF-alpha in the CD14 cocktails by panning followed by blocking with the corresponding mAbs had no effect on the active AICD protection. TGF-beta was determined not to be the active factor owing to the presence of >1.0 ng of TGF-beta in the media for culturing both CD14+ and CD14- peripheral blood mononuclear cells (PBMC). The CD14 cocktails did not contain IL-12 and IL-15. Depletion of IL-6 with panning followed by blocking residual IL-6 with anti-IL-6 mAb significantly reduced the protective effect of the CD14 cocktails. Human recombinant IL-6 also partially reversed the effects of CD14+ cell depletion on AICD, lymphocyte growth and IL-2 secretion. The data suggest that IL-6 is one of the active factors in the survival signal from CD14+ cells.
Collapse
Affiliation(s)
- Xiaolei Tang
- The Department of Microbiology and Immunology, the Arizona Arthritis Center, the University of Arizona, Tucson, Arizona, USA.
| | | | | | | |
Collapse
|
26
|
Blancheteau V, Charron D, Mooney N. HLA class II signals sensitize B lymphocytes to apoptosis via Fas/CD95 by increasing FADD recruitment to activated Fas and activation of caspases. Hum Immunol 2002; 63:375-83. [PMID: 11975981 DOI: 10.1016/s0198-8859(02)00384-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human leukocyte antigen (HLA) class II heterodimers have a well defined role in peptide presentation to helper T lymphocytes. Moreover, engagement of HLA class II molecules leads to signal transduction in the antigen presenting cell. Signaling via HLA-DR increases CD95 mediated hypoploidy in B-cell blasts. Given the importance of CD95 for the homeostasis of lymphocyte populations, we examined the impact of the HLA-DR signal on the most proximal events in the CD95 apoptotic pathway. CD95 activation recruits the adapter molecule Fas-associated death domain protein (FADD) and thereby provides a scaffold for procaspase-8 activation. The HLA-DR signal increased both recruitment of FADD and activation of caspases-3 and -8 via CD95 activation. Sensitization was tightly controlled because neither FADD recruitment to CD95 nor caspase activation was induced via HLA-DR alone. In contrast, the HLA-DR signal and the CD95 signal both led to decreased mitochondrial membrane potential. Taken together, these data indicate that ligand interaction with HLA class II molecules preactivates the antigen presenting cell for death in the event of a subsequent interaction with the CD95 ligand. This would ensure termination of a specific immune response, particularly in cells with limited sensitivity to CD95 mediated apoptosis alone.
Collapse
Affiliation(s)
- Vincent Blancheteau
- INSERM U396, Immunogénétique Humaine, Institut Biomédical des Cordeliers, Paris, France
| | | | | |
Collapse
|
27
|
Masli S, Turpie B, Hecker KH, Streilein JW. Expression of thrombospondin in TGFbeta-treated APCs and its relevance to their immune deviation-promoting properties. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:2264-73. [PMID: 11859114 DOI: 10.4049/jimmunol.168.5.2264] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
APCs deployed within iris/ciliary body are responsible for promoting anterior chamber-associated immune deviation following injection of Ag into the eye. TGFbeta-2, a constituent of the ocular microenvironment, converts conventional APCs that are pulsed with Ag into cells that induce immune deviation when injected into naive mice. TGFbeta-2-treated APCs under-express IL-12 and CD40, and over-express active TGFbeta. We have examined transcriptional changes within macrophage hybridoma no. 59, which promotes Th1 cell differentiation, and TGFbeta-2-treated no. 59 as well as macrophage hybridoma no. 63, both of which induce immune deviation similar to anterior chamber-associated immune deviation. Immune deviation-inducing hybridomas up-regulated expression of thrombospondin, TGFbeta, IFN-alpha and beta, murine macrophage elastase, and macrophage-inflammatory protein-2 genes, while down-regulating expression of the genes for NF-kappaB and CD40. Based on the known properties of these gene products, a model is proposed in which these gene products, alone and through interacting signaling pathways, confer upon conventional APCs the capacity to create and surround themselves with an immunomodulatory microenvironment. The model proposes that the pleiotropic effects of thrombospondin are primarily responsible for creating this microenvironment that is stabile, rich in active TGFbeta and IFN-alpha and beta, deficient in IL-12, and chemoattractant via macrophage-inflammatory protein-2 for NK T cells. It is further proposed that presentation of Ag to T cells in this microenvironment leads to their differentiation into regulatory cells that suppress Th1 cell-dependent immunogenic inflammation.
Collapse
Affiliation(s)
- Sharmila Masli
- Department of Ophthalmology, Schepens Eye Research Institute, Harvard Medical School, 20 Staniford Street, Boston, MA 02114, USA
| | | | | | | |
Collapse
|
28
|
Pontoux C, Banz A, Papiernik M. Natural CD4 CD25(+) regulatory T cells control the burst of superantigen-induced cytokine production: the role of IL-10. Int Immunol 2002; 14:233-9. [PMID: 11809742 DOI: 10.1093/intimm/14.2.233] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In normal mice a subpopulation of CD4 T cells constitutively expresses the IL-2 receptor alpha chain (CD25). This natural CD4 CD25(+) subset is thymus-born, constitutively expresses IL-10 mRNA,does not produce IL-2 and is resistant to apoptosis. These cells behave as regulatory T cells in the control of self-tolerance, inflammatory reactions and T cell homeostasis. The mechanisms by which natural CD4 CD25(+) cells control the immune response is unclear. We examined CD25-deficient mice, which over-express various cytokines, including proinflammatory molecules, after bacterial superantigen stimulation in vivo. We observed that this abnormal cytokine production could be controlled by the injection of natural CD4 CD25(+) T cells and that IL-10 production is needed, as CD4 CD25(+) T cells from IL-10 knockout mice do not correct cytokine over-production in vivo. As the circulating IL-10 produced by CD25-deficient mice was ineffective, we deduced that the key source of IL-10 was the regulatory T cell population. IL-10 is also involved in the control of cytokine production by normal T cells. However, the target of IL-10 in this control is undefined. Whether it acts directly on the effector T cells or on the regulatory CD4 CD25(+) T cells themselves to induce their functional maturation has to be clarified.
Collapse
Affiliation(s)
- Christiane Pontoux
- INSERM U345, Institut Necker, 156 rue de Vaugirard, 75730 Paris Cedex 15, France
| | | | | |
Collapse
|
29
|
Yarovinsky TO, Hunninghake GW. Lung fibroblasts inhibit activation-induced death of T cells through PGE(2)-dependent mechanisms. Am J Physiol Lung Cell Mol Physiol 2001; 281:L1248-56. [PMID: 11597917 DOI: 10.1152/ajplung.2001.281.5.l1248] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Activation-induced cell death (AICD) is a regulatory mechanism eliminating excess activated T cells, mainly through a Fas/Fas ligand-dependent mechanism. The goal of this study was to determine whether mouse primary lung fibroblasts are capable of modulating AICD. Using T cell hybridoma DO11.10, we found that fibroblasts in coculture rescue T cells from AICD. Fibroblast-conditioned medium (FCM) also inhibited apoptosis of T cells activated with immobilized anti-CD3 antibody. The effects of lung fibroblasts are mediated, in part, by secreted prostaglandin E(2) (PGE(2)) because treatment of fibroblasts with indomethacin decreased antiapoptotic activity of FCM. Addition of exogenous PGE(2) to FCM from fibroblast cultures treated with indomethacin restored the inhibitory activity of FCM. Expression of Fas receptor and Fas ligand by anti-CD3-activated DO11.10 cells was not affected by PGE(2). However, the same concentrations of PGE(2) significantly downregulated activation of caspase-8 and caspase-3. These results demonstrate that lung fibroblasts inhibit the AICD of T cells by secreting PGE(2), which downregulates caspase activation and apoptosis.
Collapse
Affiliation(s)
- T O Yarovinsky
- Division of Pulmonary, Critical Care, and Occupational Medicine, Department of Internal Medicine, University of Iowa, Iowa City, Iowa 52242, USA.
| | | |
Collapse
|
30
|
Emilie D, Burgard M, Lascoux-Combe C, Laughlin M, Krzysiek R, Pignon C, Rudent A, Molina JM, Livrozet JM, Souala F, Chene G, Grangeot-Keros L, Galanaud P, Sereni D, Rouzioux C. Early control of HIV replication in primary HIV-1 infection treated with antiretroviral drugs and pegylated IFN alpha: results from the Primoferon A (ANRS 086) Study. AIDS 2001; 15:1435-7. [PMID: 11504966 DOI: 10.1097/00002030-200107270-00014] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
IFN alpha has both antiviral and immunostimulating properties. The ANRS086 Primoferon A Study evaluated in 12 patients with primary HIV infection the tolerance and efficacy of an early and transient administration of pegylated IFN alpha, in addition to highly active antiretroviral therapy. Tolerance was good, and this regimen allowed the early control of HIV replication and rapid decay of the viral reservoir. These results support the initiation of comparative studies with pegylated INF alpha in primary HIV infection.
Collapse
Affiliation(s)
- D Emilie
- Service de Médecine Interne et d'Immunologie Clinique, Hôpital Antoine Béclère, Institut Paris-Sud sur les Cytokines, Clamart, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Karp CL, van Boxel-Dezaire AH, Byrnes AA, Nagelkerken L. Interferon-β in multiple sclerosis: altering the balance of interleukin-12 and interleukin-10? Curr Opin Neurol 2001; 14:361-8. [PMID: 11371761 DOI: 10.1097/00019052-200106000-00016] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Interferon-beta is a remarkably pleiotropic molecule. Antiviral, pro- and antiproliferative, pro- and antiapoptotic, and complex immunoregulatory activities have all been described. The precise mechanism(s) that underlie the beneficial effects of interferon-beta in multiple sclerosis remain poorly understood; this has hindered progress in the search for more effective therapies. An increasing body of literature supports the hypothesis that interferon-beta-mediated changes in the production and activities of the immunoregulatory cytokines interleukin-12 and interleukin-10 are important to the therapeutic benefits of interferon-beta in multiple sclerosis. These data are reviewed here.
Collapse
Affiliation(s)
- C L Karp
- Molecular Immunology Section, Children's Hospital Research Foundation, University of Cincinnati, Cincinnati, Ohio, USA.
| | | | | | | |
Collapse
|
32
|
Taams LS, Smith J, Rustin MH, Salmon M, Poulter LW, Akbar AN. Human anergic/suppressive CD4(+)CD25(+) T cells: a highly differentiated and apoptosis-prone population. Eur J Immunol 2001; 31:1122-31. [PMID: 11298337 DOI: 10.1002/1521-4141(200104)31:4<1122::aid-immu1122>3.0.co;2-p] [Citation(s) in RCA: 325] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Anergic/suppressive CD4(+)CD25(+) T cells exist in animal models but their presence has not yet been demonstrated in humans. We have identified and characterized a human CD4(+)CD25(+) T cell subset, which constitutes 7-10 % of CD4(+) T cells in peripheral blood and tonsil. These cells are a CD45RO(+)CD45RB(low) highly differentiated primed T cell population that is anergic to stimulation. Depletion of this small subset from CD4(+) T cells significantly enhances proliferation by threefold in the remaining CD4(+)CD25(-) T cells, while the addition of isolated CD4(+)CD25(+) T cells to CD4(+)CD25(-) T cells significantly inhibits proliferative activity. Blocking experiments suggest that suppression is not mediated via IL-4, IL-10 or TGF-beta and is cell-contact dependent. Isolated CD4(+)CD25(+) T cells are susceptible to apoptosis that is associated with low Bcl-2 expression, but this death can be prevented by IL-2 or fibroblast-secreted IFN-beta. However, the anergic/suppressive state of these cells is maintained after cytokine rescue. These human regulatory cells are therefore a naturally occurring, highly suppressive, apoptosis-prone population which are at a late stage of differentiation. Further studies into their role in normal and pathological situations in humans are clearly essential.
Collapse
Affiliation(s)
- L S Taams
- Department of Clinical Immunology, Royal Free and University College Medical School, London, GB
| | | | | | | | | | | |
Collapse
|
33
|
Wilson K. IFN maintains anergic cell survival. Arthritis Res Ther 2000. [DOI: 10.1186/ar-2000-66857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
34
|
Akbar AN, Lord JM, Salmon M. IFN-alpha and IFN-beta: a link between immune memory and chronic inflammation. IMMUNOLOGY TODAY 2000; 21:337-42. [PMID: 10871875 DOI: 10.1016/s0167-5699(00)01652-2] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The majority of expanded T cells generated during an immune response are cleared by apoptosis. Prevention of death in some activated T cells enables the persistence of a memory T-cell pool. Here, observations that IFN-alpha and IFN-beta inhibit activated T-cell apoptosis are described. Although this enables memory T cells to persist without antigen, excessive IFN-alpha or IFN-gamma secretion might lead to chronic inflammation.
Collapse
Affiliation(s)
- A N Akbar
- Department of Clinical Immunology, Royal Free and University College Medical School, Hampstead, London, UK.
| | | | | |
Collapse
|