Isolation and characterization of human antigen-specific TCRαβ+ CD4-CD8- double-negative regulatory T cells

Blood ◽  
2005 ◽  
Vol 105 (7) ◽  
pp. 2828-2835 ◽  
Author(s):  
Karin Fischer ◽  
Simon Voelkl ◽  
Jana Heymann ◽  
Grzegorz K. Przybylski ◽  
Krishna Mondal ◽  
...  

AbstractDown-regulation of immune responses by regulatory T (Treg) cells is an important mechanism involved in the induction of tolerance to allo-antigens (Ags). Recently, a novel subset of Ag-specific T-cell receptor (TCR)αβ+ CD4-CD8- (double-negative [DN]) Treg cells has been found to be able to prevent the rejection of skin and heart allografts by specifically inhibiting the function of antigraft-specific CD8+ T cells. Here we demonstrate that peripheral DN Treg cells are present in humans, where they constitute about 1% of total CD3+ T cells, and consist of both naïve and Ag-experienced cells. Similar to murine DN Treg cells, human DN Treg cells are able to acquire peptide–HLA-A2 complexes from antigen-presenting cells by cell contact-dependent mechanisms. Furthermore, such acquired peptide-HLA complexes appear to be functionally active, in that CD8+ T cells specific for the HLA-A2–restricted self-peptide, Melan-A, became sensitive to apoptosis by neighboring DN T cells after acquisition of Melan-A–HLA-A2 complexes and revealed a reduced proliferative response. These results demonstrate for the first time that a sizable population of peripheral DN Treg cells, which are able to suppress Ag-specific T cells, exists in humans. DN Treg cells may serve to limit clonal expansion of allo-Ag–specific T cells after transplantation.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3306-3306
Author(s):  
Karin Fischer ◽  
Simon Voelkl ◽  
Grzegorz K. Przybylski ◽  
Christian A. Schmidt ◽  
Reinhard Andreesen ◽  
...  

Abstract Compelling evidence indicate that regulatory T (Treg) cells play an important role in the maintenance of immune tolerance to self and foreign antigens (Ag). Various subsets of T lymphocytes have been isolated in mice and humans that have the ability to down-regulate the proliferation of autoimmune effector cells. Recently, a novel subset of Ag-specific T-cell receptor (TCR)αβ+ CD4−CD8− (double negative, DN) Treg cells has been found to be able to prevent the rejection of skin and heart allografts by specifically inhibiting the function of anti-graft-specific CD8+ T cells. Here we demonstrate that peripheral DN Treg cells are present in humans, where they constitute about 1% of total CD3+ T cells, and consist of both naïve and Ag-experienced cells. Furthermore, analysis of T-cell receptor excision circles (TRECs) indicate that DN T cells are not recent thymic emigrants, but rather an expanded T-cell subset. MHC multimer staining revelaed a distinct population of DN T cells recognizing common MHC class I-restricted CMV and EBV antigens. DN T cells exhibited a strong proliferative response upon stimulation with allogeneic antigen presenting cells (APC) and secreted high amounts of IFN-γ but no IL-2, with some IL-5, and marginal levels of IL-4 and IL-10. Similar to murine DN Treg cells, human DN Treg cells are able to acquire peptide-HLA-A2 complexes from APCs by cell contact-dependent mechanisms. Furthermore, such acquired peptide-HLA complexes appear to be functionally active, in that CD8+ T cells specific for the HLA-A2-restricted self peptide, Melan-A, became sensitive to apoptosis by neighboring DN T cells after acquisition of Melan-A-HLA-A2 complexes and revealed a reduced proliferative response. These results demonstrate for the first time that a sizeable population of peripheral DN Treg cells exists in humans that are able to suppress Ag-specific T cells. DN Treg cells may serve to limit clonal expansion of allo-Ag-specific T cells after transplantation.


2001 ◽  
Vol 193 (11) ◽  
pp. 1303-1310 ◽  
Author(s):  
Detlef Dieckmann ◽  
Heidi Plottner ◽  
Susanne Berchtold ◽  
Thomas Berger ◽  
Gerold Schuler

It has been known for years that rodents harbor a unique population of CD4+CD25+ “professional” regulatory/suppressor T cells that is crucial for the prevention of spontaneous autoimmune diseases. Here we demonstrate that CD4+CD25+CD45RO+ T cells (mean 6% of CD4+ T cells) are present in the blood of adult healthy volunteers. In contrast to previous reports, these CD4+CD25+ T cells do not constitute conventional memory cells but rather regulatory cells exhibiting properties identical to their rodent counterparts. Cytotoxic T lymphocyte–associated antigen (CTLA)-4 (CD152), for example, which is essential for the in vivo suppressive activity of CD4+CD25+ T cells, was constitutively expressed, and remained strongly upregulated after stimulation. The cells were nonproliferative to stimulation via their T cell receptor for antigen, but the anergic state was partially reversed by interleukin (IL)-2 and IL-15. Upon stimulation with allogeneic (but not syngeneic) mature dendritic cells or platebound anti-CD3 plus anti-CD28 the CD4+CD25+ T cells released IL-10, and in coculture experiments suppressed the activation and proliferation of CD4+ and CD8+ T cells. Suppression proved IL-10 independent, yet contact dependent as in the mouse. The identification of regulatory CD4+CD25+ T cells has important implications for the study of tolerance in man, notably in the context of autoimmunity, transplantation, and cancer.


2007 ◽  
Vol 204 (11) ◽  
pp. 2747-2757 ◽  
Author(s):  
Pia P. Yachi ◽  
Carina Lotz ◽  
Jeanette Ampudia ◽  
Nicholas R.J. Gascoigne

T cells are extremely sensitive in their ability to find minute amounts of antigenic peptide in the midst of many endogenous peptides presented on an antigen-presenting cell. The role of endogenous peptides in the recognition of foreign peptide and hence in T cell activation has remained controversial for CD8+ T cell activation. We showed previously that in a CD8+ T cell hybridoma, nonstimulatory endogenous peptides enhance T cell sensitivity to antigen by increasing the coreceptor function of CD8. However, others were not able to detect such enhancement in naive and activated CD8+ T cells. Here, we show that endogenous peptides substantially enhance the ability of T cells to detect antigen, an effect measurable by up-regulation of activation or maturation markers and by increased effector function. This enhancement is most pronounced in thymocytes, moderate in naive T cells, and mild in effector T cells. The importance of endogenous peptides is inversely proportional to the agonist activity of the stimulatory peptide presented. Unlike for CD4+ T cells, the T cell receptor of CD8+ T cells does not distinguish between endogenous peptides for their ability to enhance antigen recognition.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1263-1263
Author(s):  
Jing-Zhou Hou ◽  
Jeanette Baker ◽  
Kelvin Sheehan ◽  
Robert S. Negrin

Abstract Naturally occurring CD4+CD25+ T regulatory cells (Tregs) are a subpopulation of CD4+ T cells vital to homeostasis and maintenance of tolerance. Tregs develop in the thymus and are characterized by expression of the FoxP3 transcription factor. Tregs suppress the proliferation and expansion of conventional CD25-CD4+ and CD8+ T cells through direct cell-cell contact. Defect in Tregs results in multi-organ autoimmune disorders. Adoptive transfer of Tregs cured diabetes in NOD mice and inflammatory bowel disease in murine models. Co-transplantation of Tregs with conventional CD4+ and CD8+ T cells has been shown to control acute graft versus host diseases (GVHD) without abrogating graft versus tumor (GVT) effects in murine bone marrow transplant models.Translating this novel strategy to suppress acute GVHD without loss of GVT holds great promise not only in clinical hematopoietic cell transplantation (HCT), but also in adoptive cellular therapy beyond HCT. Large scale isolation of sufficient human Tregs for therapeutic application remains challenging. Tregs comprise 2–10% of CD4+ T cells. In contrast to rodent Tregs, human CD4+CD25+ cells are more heterogeneous because CD25 is also expressed at lower level by activated non-suppressive conventional T cells and B cells. In addition, immunophenotype of human Tregs is less well studied. Here, we explore the isolation and characterization of human CD4+CD25+ Tregs from peripheral blood mononuclear cells of healthy blood donors and from CD34+ cell depleted fraction of allogenic donors. CD25+ cells were isolated by three different approaches: 1) CD4+ enrichment followed by CD25+ selection, 2) CD19 depletion followed by CD25+ selection, 3) CD25+ selection without prior enrichment or depletion, using magnetic beads. The purities of isolated CD4+CD25+ cells were 95%, 93%, and 93% respectively by FACS. The percentage of CD25 high expressing cells was also similar among the three approaches. More than 90% purity of CD4+CD25+ cells was consistently obtained by CD25+ selection alone using CliniMACS® CD25 microbeads via double column separation utilizing the AutoMACS®. The yield of CD25+ cells varied based upon the ratio of cells/buffer/microbeads. The isolated CD25+ cells were then sorted into CD4+CD25bright (top 25–30%) and CD4+CD25dim (bottom 25–30%) by FACS, fixed and stained for intracellular expression of FoxP3. More than 95% of CD4+CD25bright cells expressed FoxP3. In contrast, only 40% of CD4+CD25dim cells expressed FoxP3. No FoxP3 expression was detected in CD4+CD25− cells. Murine Tregs express a variety of co-stimulatory molecules, however, expression of these molecules is less well characterized on human CD4+CD25+ cells. Using multi-color FACS, we observed that human FoxP3+CD4+CD25bright cells express high levels of CD27 (98%), CD28 (100%), and CD30 (50%) and low levels (<10%) of CTLA-4, GITR, 4-1BB, PD-1 and ICOS on the cell surface. However, CTLA-4, GITR, PD-1 and ICOS were expressed intracellularly in more than 50% of FoxP3+CD4+CD25bright cells. OX40, TRAIL, ICOS ligand and FasL were not detected on the cell surface or in the cytoplasm of FoxP3+CD4+CD25bright cells. FoxP3+CD4+CD25bright cells also express CD62L and CCR5. In summary, highly purified CD4+CD25+ Treg cells can be isolated by magnetic beads and cell sorting. Importantly, the majority of the cells express FoxP3. Utilizing this technique, the clinical application of Tregs appears feasible.


2008 ◽  
Vol 5 (1) ◽  
pp. 47-53 ◽  
Author(s):  
Weijuan Gong ◽  
Mingchun Ji ◽  
Zhengfeng Cao ◽  
Liheng Wang ◽  
Yayun Qian ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 5117-5117
Author(s):  
Takeshi Nakamura ◽  
Miwako Narita ◽  
Akie Yamahira ◽  
Norihiro Watanabe ◽  
Nozomi Tochiki ◽  
...  

Abstract Abstract 5117 Although cellular immunotherapy based on antigen-specific cytotoxic T lymphocytes (CTLs) against tumors including leukemia and severe infections is a promising strategy, one of the pivotal issues is a hardship in constant supply of high quality antigen presenting cells for generating autologous CTLs against antigens associated with tumor cells or pathogens. We established a leukemic plasmacytoid dendritic cell (pDC) line (PMDC05) with potent antigen presenting capacity from leukemia cells of a HLA-A*0201/*2402 patient with pDC acute leukemia. We investigated whether PMDC05 could be efficiently used for generating CTLs specific for antigens of leukemia cells or pathogens and whether PMDC05 could be applicable for cellular immunotherapy for tumors and infections. PMDC05, which grew in the absence of feeder cells, was positive for CD4, CD56, CD33, HLA-DR, CD123 (IL-3Rα) and CD86 in the absence of lineage markers. mRNA of TLR1, TLR2, TLR4, TLR7 and TLR9 were clearly expressed and among these TLRs, TLR7 was prominent. Transcripts of preTα, SpiB, MX1, IRF1, IRF7 and IFN-α14 were markedly expressed in PMDC05 and those of λ-like 14.1, IL-12p35 or IL-12p40 were also expressed. By culturing PMDC05 with IL-3, CpG-B or LPS, the expression of CD1a, CD80, CD83, CD86 or HLA-DR was remarkably enhanced. Transcripts of IRF1, IRF7, IFN-α14, IL-12p35 and IL-12p40 were increased by culturing with CpG-A, CpG-B, influenza virus or LPS. Production of IFN-α and IL-12p75 was enhanced by the stimulation with CpG-A and LPS, respectively. PMDC05 possessed a considerable antigen presenting ability to naïve T cells, which was enhanced by culturing with IL-3 or influenza virus and especially by LPS, and the ability to uptake lucifer yellow. These findings revealed that PMDC05 is a unique leukemic pDC cell line with potent antigen presenting ability. Therefore, we tried to generate WT1 or CMV-specific CTLs by using PMDC05 pulsed with relevant antigen as antigen presenting cells. PMDC05, which was stimulated with 0.1 μg/ml LPS and loaded with 10 μg/mL WT1 peptides (HLA-A*2402-restricted, modified-type 9-mer peptide; CYTWNQMNL) or CMV pp65 peptides (HLA-A*2402-restricted, 9-mer peptide; QYDPVAALF) for 24 hours, was irradiated (60 Gy) and co-cultured with allogeneic CD8+ T cells purified from peripheral blood mononuclear cells (PB-MNCs) of HLA-A*2402+ healthy donor (PMDC05:CD8+ Tcells = 1:10). 50 U/ml IL-2 was added to the co-culture at day 2, and IL-2 as well as 10 ng/ml IL-7 were added every 3 days thereafter. Burkitt lymphoma cell line, Daudi, was used as control of PMDC05 in the same manner. PB-MNCs, which were presumed to contain regulatory T (Treg) cells, were also co-cultured with PMDC05 or Daudi. Induction of WT1 or CMV-specific CTLs was evaluated by flow cytometry analysis using HLA-A*2402 WT1 tetramer or CMV tetramer every week. Although Daudi pulsed with WT1 peptides could not induce WT1 tetramer+CD8+ T cells during the co-culture up to 7 weeks, PMDC05 began to induce WT1 tetramer+CD8+ T cells at week 4 and the percentage of WT1 tetramer+ T cells in CD8+ T cells rose to more than 75% at week 7. Likewise, CMV tetramer+CD8+ T cells were amplified in CD8+ T cells co-cultured with CMV peptide-pulsed PMDC05 but not in those with Daudi. Although CMV tetramer+CD8+ T cells were generated in MNCs co-cultured with PMDC05 pused with CMV peptides, WT1 tetramer+CD8+ T cells were not observed in in MNCs co-cultured with PMDC05. These data suggested that PMDC05 could be efficiently used for generating CTLs specific for antigens of tumors or pathogens and could be applicable for cellular immunotherapy for tumors and infections. In addition, it was suggested that generation of antigen-speific CTLs might be affected by co-existing Treg cells in some tumor specific antigens. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 4 (1) ◽  
pp. 36
Author(s):  
Naohiro Seo

Immune system is a precise mechanism for maintenance of homeostasis by lymphocyte-mediated elimination of extracellular and intercellular pathogens, and abnormal cells in cytokine-, chemokine-, antibody-, and cytotoxic granule-dependent manners. Extracellular vesicles, e.g. exosomes, released from multivesicular endosome in immune cells have been known to be a part of the immune system. Exosomes released by antigen-presenting cells (APCs) such as macrophages and dendritic cells (DCs) regulate natural killer (NK) cells, CD8+ T cells (Cytotoxic T lymphocytes [CTLs]), and CD4+ T cells (Th cells) including Th1, Th2, and regulatory T (Treg) cells. In the anti-tumor immune system, NK cells and CTLs are mainly involved in the elimination of tumor cells by direct interaction. Recently, we clarified that tumor-infiltrating CD8+ T cells prevent tumor invasion and metastasis by exosome-mediated destruction of tumor stroma consist of mesenchymal stem cells (MSCs) and cancer-associated fibroblasts (CAFs). In this review article, we describe the role of exosomes in controlling immune system and its clinical application.


Sign in / Sign up

Export Citation Format

Share Document