scholarly journals Immune Tolerance After Delivery of Dying Cells to Dendritic Cells In Situ

2002 ◽  
Vol 196 (8) ◽  
pp. 1091-1097 ◽  
Author(s):  
Kang Liu ◽  
Tomonori Iyoda ◽  
Marzena Saternus ◽  
Yukino Kimura ◽  
Kayo Inaba ◽  
...  

Peripheral immune tolerance is believed to be induced by the processing and presentation of self-tissues that die during physiologic tissue turnover. To examine the mechanism that mediates tolerance, we injected mice with dying syngeneic TAP−/− splenocytes loaded with small amounts of the protein antigen, ovalbumin (OVA). After ingestion and presentation of cell-associated OVA by the CD8+ subset of dendritic cells in situ, large numbers of antigen-reactive, CD8+ T cell receptor (TCR) transgenic T lymphocytes were driven into cell cycle, but then the T cells were deleted. The animals were also tolerant to challenge with OVA in complete Freund's adjuvant. An agonistic anti-CD40 monoclonal antibody was then administered together with the OVA-loaded splenocytes, so that the dendritic cells in the recipient mice would mature. In contrast to observations made in the steady state, the antigen-reactive T cells expanded in numbers for 1–2 wk and produced large amounts of interleukin 2 and interferon γ, while the animals retained responsiveness to antigen rechallenge. The specific tolerance that develops when dendritic cells process self tissues in the steady state should prevent or reduce the development of autoimmunity when dying cells are subsequently processed during infection.

2004 ◽  
Vol 200 (9) ◽  
pp. 1167-1177 ◽  
Author(s):  
Hiromitsu Hara ◽  
Christopher Bakal ◽  
Teiji Wada ◽  
Denis Bouchard ◽  
Robert Rottapel ◽  
...  

Carma1 (also known as caspase recruitment domain [CARD]11, Bimp3) is a CARD-containing membrane-associated guanylate kinase family protein that plays an essential role in antigen receptor–induced nuclear factor κB activation. We investigated the role of Carma1 in the assembly of signaling molecules at the immune synapse using a peptide-specific system. We report that Carma1 is essential for peptide-induced interleukin 2 and interferon γ production, but dispensable for proliferation in T cells. Recruitment and distribution of T cell receptor, lymphocyte function associated 1, lipid rafts, and protein kinase C (PKC)θ to central and peripheral immune synapse regions occur normally in Carma1−/− T cells. Carma1 controls entry of IκB kinase (IKK) into lipid raft aggregates and the central region of the immune synapse, as well as activation of IKK downstream of PKC. Our data provide the first genetic evidence on a new class of molecular scaffold that controls entry of defined signaling components, IKK, into the central supramolecular activation cluster at T cell–antigen-presenting cell interfaces without having any apparent effect on the overall organization and formation of immune synapses.


2001 ◽  
Vol 194 (6) ◽  
pp. 769-780 ◽  
Author(s):  
Daniel Hawiger ◽  
Kayo Inaba ◽  
Yair Dorsett ◽  
Ming Guo ◽  
Karsten Mahnke ◽  
...  

Dendritic cells (DCs) have the capacity to initiate immune responses, but it has been postulated that they may also be involved in inducing peripheral tolerance. To examine the function of DCs in the steady state we devised an antigen delivery system targeting these specialized antigen presenting cells in vivo using a monoclonal antibody to a DC-restricted endocytic receptor, DEC-205. Our experiments show that this route of antigen delivery to DCs is several orders of magnitude more efficient than free peptide in complete Freund's adjuvant (CFA) in inducing T cell activation and cell division. However, T cells activated by antigen delivered to DCs are not polarized to produce T helper type 1 cytokine interferon γ and the activation response is not sustained. Within 7 d the number of antigen-specific T cells is severely reduced, and the residual T cells become unresponsive to systemic challenge with antigen in CFA. Coinjection of the DC-targeted antigen and anti-CD40 agonistic antibody changes the outcome from tolerance to prolonged T cell activation and immunity. We conclude that in the absence of additional stimuli DCs induce transient antigen-specific T cell activation followed by T cell deletion and unresponsiveness.


Blood ◽  
2005 ◽  
Vol 105 (11) ◽  
pp. 4383-4389 ◽  
Author(s):  
Loïc Dupré ◽  
Grazia Andolfi ◽  
Stuart G. Tangye ◽  
Rita Clementi ◽  
Franco Locatelli ◽  
...  

Abstract The adaptor protein SAP regulates signaling through signaling lymphocytic activation molecule (SLAM)–family receptors expressed on T and natural killer (NK) cells. In patients affected by X-linked lymphoproliferative (XLP) disease, mutations in the SH2D1A gene result in defective lytic activity. However, the mechanism by which SAP controls cytotoxic activity remains unclear. T-cell–receptor (TCR) activation of CD8+ cytotoxic T cells (CTLs) results in down-regulation of SAP, suggesting that this protein is involved in early activation events. Here, we show that SAP-deficient CTLs from patients with XLP and hemophagocytic lymphohistiocytosis (HLH) display a specific lytic defect against autologous and allogeneic Epstein-Barr virus (EBV)–positive B cells. This defect is associated with the defective polarization of 2B4, perforin, and lipid rafts at the contact area of CTLs with EBV-positive targets. Blockade of 2B4 in normal CTLs reproduces the defects in lysis and polarization observed in SAP-deficient CTLs. Expression and regulation of the SLAM-family receptors SLAM, CD84, and 2B4, as well as the lytic effectors perforin and granzyme-B are normal in SAP-deficient CTLs. In addition, TCR stimulation leads to normal proliferation and production of interleukin 2 (IL-2), IL-4, and interferon-γ (IFN-γ). These results demonstrate that the SAP/2B4 pathway plays a key role in CTL lytic activity against EBV-positive targets by promoting the polarization of the lytic machinery.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3830-3830
Author(s):  
Yasmine Van Caeneghem ◽  
Glenn Goetgeluk ◽  
Sylvia Snauwaert ◽  
Fritz Offner ◽  
Reno Debets ◽  
...  

Abstract T cell therapy for the treatment of malignant diseases is based on the lenti- or retroviral introduction of an exogenous receptor in peripheral blood T cells. The exogenous receptor is either antibody based or T cell receptor (TCR) based. Chimeric antigen receptors (CAR) are antibody based receptors that can redirect T cells against membrane antigens expressed by malignant cells. CD19-specific CARs were reported to be very effective in the treatment of CD19+ acute leukemias. To redirect T cells based on cytoplasmic antigens, transduction of a TCR is required. However, this approach still faces technical problems, esp. interference of the endogenous TCR chains may cause loss of avidity and possibly induction of autoimmunity. We here present an alternative strategy, in which, not mature T cells but CD34+ hematopoietic precursor cells are transduced and subsequently differentiated to mature T cells after introduction of a wild type TCR or of a fusion TCR:CD3ζ with or without costimulator signal. When Wilms tumor 1 (WT1)/HLA-A2-specific T cell receptor α and β chain is introduced in CD34+ cells derived from human thymus, cord blood or adult mobilized precursor cells and subsequently induced to differentiate to T cells on OP9 stromal cells expressing Delta-like ligand 1(OP9-DL1) in the presence of stem cell factor, flt3 ligand and interleukin 7, massive proliferation is observed while the cells differentiate to CD4+CD8+double positive (DP) transduced TCR+ immature cells. Few mature T cells are generated in these cultures, but after addition of the specific peptide to HLA-A2+ cultures, DP cells rapidly differentiate to phenotypically mature naïve CD8 single positive T cells. Upon activation, these T cells specifically lyse WT1/HLA-A2 cell lines and produce interferon-γ. Microarray expression analysis revealed these culture-generated T cells to be similar to TCR-transduced peripheral blood T cells, except for 1) the expression of only one TCR α and β chain by the in vitro generated T cells and 2) the underexpression of costimulatory/inhibitory molecules such as CD28, CTLA-4 and PD-L1. The absence of CD28 on the cell membrane was confirmed by flow cytometry. Since it was shown that CD28 signaling is essential for in vivo functionality using CARs, we next generated fusion TCR constructs of a gp100/HLA-A2-specific TCR and the signaling cassettes of CD3ζ and CD28.The following constructs were introduced in CD34+ cells: wild type TCR, TCR:ζ or TCR:CD28ζ α and β chains. The α and β chain double-transduced cells were subsequently cultured on OP9-DL1 in the absence of the specific antigen. It was observed that TCR:ζ transduced precursors proliferated significantly less than wild type TCR transduced cells, but the majority of the cells differentiated towards DP TCR:ζ+ cells, which upon addition of the specific antigen differentiated to phenotypically mature T cells. TCR:CD28ζ transduced cells proliferated least of all and spontaneously matured to functional double negative T cells without passing through the DP stage. These observations are compatible with data obtained in mice showing that strong TCR activation during thymocyte differentiation inhibits the generation of DP cells. In all of these cultures, endogenous TCR rearrangements were suppressed, which resulted in single receptor tumoricidal cells. Functional analysis of these various cell populations showed similar proliferation on T cell growth factors and specific cytolytic activity of gp100+ HLA-A2+ tumor lines. However, the TCR:CD28ζ transduced cells produced significantly higher levels of TNFα and interferon-γ and were the only ones that produced interleukin-2 upon specific stimulation. In conclusion, we have shown that high numbers of polyfunctional single receptor TCR:CD28ζ+ cells can be generated in vitro from clinically relevant stem cell sources. These cells produce interleukin-2, TNFα and interferon-γ and specifically kill gp100/HLA-A2+ tumor cell lines. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2003 ◽  
Vol 101 (12) ◽  
pp. 4909-4915 ◽  
Author(s):  
Yixin Jin ◽  
Eirunn Knudsen ◽  
Ling Wang ◽  
Yenan Bryceson ◽  
Bassam Damaj ◽  
...  

Abstract Sphingosine 1-phosphate (S1P) is a pleiotropic lysosphingophospholipid stored and secreted by platelets. Using reverse transcription–polymerase chain reaction and flow cytometric analyses, we determined the expression of S1P receptors (S1P1, S1P3, S1P4, and S1P5) in peripheral blood T cells. T cells were induced to proliferate in the presence of phorbol 12-myristate 13-acetate (PMA) plus ionomycin, anti-CD3 plus anti-CD28, and allogeneic immature or mature dendritic cells. This activity was inhibited by the addition of S1P. Enhanced T-cell proliferation was observed when these cells were stimulated with the same stimuli, but were incubated in serum-free media (SFM). Addition of S1P to SFM inhibited the stimulation of T cells induced by T-cell stimuli, suggesting that S1P is an important inhibitory molecule present in the serum. T-cell proliferation was also inhibited by the addition of dihydrosphingosine 1-phosphate (DHS1P), sphingosine, and ceramide; however, the latter 2 sphingolipids required higher concentrations than S1P. Pretreatment of T cells with pertussis toxin (PTX) blocked the inhibitory effect of S1P on activation with PMA plus ionomycin, but not on activation with anti-CD3 plus anti-CD28. This is corroborated with the down-regulation of S1P1 in T cells stimulated with anti-CD3 plus anti-CD28. Similarly, PTX did not affect the inhibitory effect of S1P on T-cell proliferation when dendritic cells were used as stimuli. Further, S1P or DHS1P but not ceramide or sphingosine enhanced rather than decreased secretion of interleukin 2 and interferon γ by T cells stimulated with anti-CD3 plus anti-CD28. These results show differential effects of S1P on polyclonal T-cell proliferation and cytokine secretion.


2007 ◽  
Vol 204 (5) ◽  
pp. 1095-1106 ◽  
Author(s):  
Helena Soares ◽  
HaeNa Waechter ◽  
Nicholas Glaichenhaus ◽  
Evelyne Mougneau ◽  
Hideo Yagita ◽  
...  

Interferon (IFN)-γ, a cytokine critical for resistance to infection and tumors, is produced by CD4+ helper T lymphocytes after stimulation by cultured dendritic cells (DCs) that secrete a cofactor, interleukin (IL)-12. We have identified a major IL-12–independent pathway whereby DCs induce IFN-γ–secreting T helper (Th)1 CD4+ T cells in vivo. This pathway requires the membrane-associated tumor necrosis family member CD70 and was identified by targeting the LACK antigen from Leishmania major within an antibody to CD205 (DEC-205), an uptake receptor on a subset of DCs. Another major DC subset, targeted with 33D1 anti-DCIR2 antibody, also induced IFN-γ in vivo but required IL-12, not CD70. Isolated CD205+ DCs expressed cell surface CD70 when presenting antigen to T cell receptor transgenic T cells, and this distinction was independent of maturation stimuli. CD70 was also essential for CD205+ DC function in vivo. Detection of the IL-12–independent IFN-γ pathway was obscured with nontargeted LACK, which was presented by both DC subsets. This in situ analysis points to CD70 as a decision maker for Th1 differentiation by CD205+ DCs, even in Th2-prone BALB/c animals and potentially in vaccine design. The results indicate that two DC subsets have innate propensities to differentially affect the Th1/Th2 balance in vivo and by distinct mechanisms.


1998 ◽  
Vol 187 (1) ◽  
pp. 49-57 ◽  
Author(s):  
Yong Ke ◽  
Hakling Ma ◽  
Judith A. Kapp

The mechanisms that maintain memory in T cells are not completely understood. We have investigated the role of antigen and interleukin (IL)-2 in the growth and maintenance of CD8+ T cells using a cytolytic T cell line specific for ovalbumin (OVA)257-264 presented by H-2Kb. This line does not secrete IL-4 or IL-2; hence, stimulation with the OVA-transfected EL4 line (E.G7-OVA) does not induce proliferation without addition of exogenous growth factors. Furthermore, this line can be maintained continuously by weekly addition of irradiated, splenic filler cells and IL-2, with or without E.G7-OVA. Although IL-2 induced proliferation of these cytotoxic T lymphocytes (CTLs), production of interferon γ and tumor necrosis factor α required stimulation of the CTL with E.G7-OVA. The kinetics of lymphokine secretion after stimulation by E.G7-OVA were the same whether the CTL had been maintained with or without antigen (Ag). In addition, both CTL lines killed E.G7-OVA target cells within 4 h. Thus, the effector functions of these CTLs were rapidly induced by T cell receptor (TCR) occupancy. CTLs cultured with or without Ag also served as memory T cells when parked for 100 d in unirradiated, syngeneic recipients without OVA. In the absence of OVA, the precursor frequency was identical in spleens of normal and β2-microglobulin knockout recipients, but significantly less in IL-2 knockout mice. The decline of memory in the absence of IL-2 supports data from other investigators, suggesting that cell cycling is important to the maintenance of CD8+ T cell memory. These data also suggest that stimulation of OVA-specific CTLs by lymphokines seems to be more important to maintaining memory than stimulation of TCRs by cross-reactive peptides complexed to class I molecules.


Sign in / Sign up

Export Citation Format

Share Document