scholarly journals Differential involvement of CD4+ cells in mediating skin graft rejection against different amounts of transgenic H-2K(b) antigen.

1991 ◽  
Vol 173 (1) ◽  
pp. 261-264 ◽  
Author(s):  
M Kawai ◽  
Y Obata ◽  
N Hamasima ◽  
T Takahashi ◽  
A Uenaka ◽  
...  

Differential involvement of CD4+ cells in mediating class I-disparate skin graft rejection was investigated using quantitatively different Kb transgenic mice as donors under conditions in which CD8+ cells were blocked in vivo by administration of anti-CD8 monoclonal antibody (mAb). Tg.H-2Kb-1 and -2 are C3H transgenic mice with 14 and 4 copies, respectively, of the H-2Kb gene. Cell surface expression of Kb antigen and the Kb antigenicity of skin for eliciting graft rejection with homozygous and heterozygous transgenic mice were correlated with the copy number. In vivo administration of anti-Lyt-2.1 (CD8) mAb markedly prolonged survival of heterozygous and homozygous C3H Tg.H-2Kb-2 skin grafted onto C3H mice, but prolonged survival of heterozygous Tg.H-2Kb-1 skin grafts much less and did not prolong survival of homozygous Tg.H-2Kb-1 grafts. Administration of anti-L3T4 (CD4) mAb alone did not have any effect on skin graft rejection. Administration of anti-L3T4 (CD4) mAb with anti-Lyt-2.1 (CD8) mAb blocked rejection in all combinations. These findings indicate that a quantitative difference of class I antigen caused differential activation of CD4+ cells under conditions in which CD8+ cells were blocked.

1987 ◽  
Vol 166 (4) ◽  
pp. 982-990 ◽  
Author(s):  
T Ichikawa ◽  
E Nakayama ◽  
A Uenaka ◽  
M Monden ◽  
T Mori

The cellular mechanisms of skin graft rejection with allelic H-2 class I differences were studied by examining the effect on graft survival of in vivo administration of anti-Lyt-2.2 mAb, anti-L3T4 mAb, or both to recipient mice. The injections of anti-Lyt-2.2 mAb and anti-L3T4 mAb caused selective depletions of Lyt-2+ cells and L3T4+ cells, respectively. Injection of anti-Lyt-2.2 mAb significantly prolonged graft survival in 7 of 12 combinations of H-2D-end difference, but did not prolong graft survival in 5 other combinations of H-2D-end difference, or in 2 combinations of H-2K-end difference. Injection of anti-L3T4 mAb did not prolong graft survival in any combinations with class I difference tested. Injection of anti-L3T4 mAb plus anti-Lyt-2.2 mAb markedly prolonged graft survival in the combinations with class I difference in which anti-Lyt-2.2 mAb had no effect and overcame the effect of anti-Lyt-2.2 mAb in those in which anti-Lyt-2.2 mAb had an effect in prolonging graft survival. These results indicated that in combinations in which anti-Lyt-2.2 mAb did not prolong graft survival, class I antigen stimulated L3T4+ effector cells when Lyt-2+ cells were blocked and Lyt-2+ effector cells when L3T4+ cells were blocked. On the other hand, in the combinations in which anti-Lyt-2.2 mAb prolong graft survival, these antigens initially caused preferential stimulation of Lyt-2+ but not L3T4+ effector cells, although delayed activation of L3T4+ effector cells occurred when Lyt-2+ cells were blocked. Furthermore, a significant correlation was found between the effect of anti-Lyt-2.2 mAb in prolonging graft survival and the failure of recipient mice to produce H-2 antibody. These results can be taken as evidence that L3T4+ effector cells are not involved in the initial phase of graft rejection in these combinations.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 50-50
Author(s):  
Gullu Gorgun ◽  
Tobias A.W. Holderried ◽  
Rifca Ledieu ◽  
David Zahrieh ◽  
John G. Gribben

Abstract Deregulation of the TCL1 pathway plays a crucial role in B-CLL pathogenesis and targeted expression of TCL1 results in the development in older mice of a B cell lymphoproliferative disorder resembling human B-CLL. CLL patients develop progressively impaired immunity and gene expression profiling of CD4 and CD8 T cells in B-CLL patients revealed defects in genes regulating critical pathways for T cell effector function. The onset of CLL in TCL1-transgenic mice also results in defects similar to those observed in CLL patients. Therefore, this murine model mimics the impact of CLL on the normal immune system, suggesting this may be an appropriate model to examine in vivo the impact of steps taken to repair T cell defects. In this study we examined whether infusion of CLL cells obtained from older mice induced similar changes in T cells of young mice, providing direct demonstration in vivo of interactions of CLL cells with the host immune system which result in development of immune deficiencies. Global gene expression profiling was performed using the Mouse 430_2 Affymetrix chip on highly purified CD4 and CD8 T cells from 6 non-transgenic mice and 16 TCL1 transgenic mice of different ages and at different stages in disease development and compared to that of cells from 6 TCL1 transgenic mice without CLL injected one week previously with 50 x 106 CLL cells. On unsupervised analysis using DNA-Chip Analyzer CD4 and CD8 T cells of young mice without CLL clustered with non-transgenic mice of different ages, whereas CD4 and CD8 cells from mice with developing or established CLL clustered with the young mice injected with CLL cells. Supervised analysis using Permax identified significant differences in expression for 628 genes (125 genes upregulated and 503 downregulated) in CD4 cells and 620 genes (320 genes upregulated and 300 genes downregulated) in CD8 cells in T cells from CLL bearing mice and CLL cell injected mice compared to non-transgenic mice and non-tumor bearing TCL1 mice. Comparison of pathways perturbed in the mice using GenMAPP finder compared to that observed in our previous studies in patients with CLL demonstrates similar alteration in many pathways, including regulation of cell proliferation and cell cycle control, cell differentiation, cytoskeleton formation, intracellular transportation and vesicle formation and transport. Examining these pathways functionally, we observed significantly decreased T cell proliferation, cytotoxicity and helper T cell function, increased numbers of CD4+CD25+CTLA4+ regulatory T cells and increased IL-4 amd IL-13 and decreased IL-12, IFNγ, sTNFRI, sTNFRII in CD4 cells and decreased IL-12p40, TIMP1 and TIMP2 in CD8 cells in both CLL bearing mice or mice injected with CLL cells compared to mice without CLL. These similar findings in human and murine CLL are in keeping with the hypothesis that interaction of the CLL cells with the normal immune function induces changes that result in decrease in T cell differentiation and effector function. It is intriguing to postulate that this effect diminishes autologous anti-tumor responses. We conclude that development of CLL in these transgenic mice induces T cell defects that mimic the defects that occur in CLL patients and that the TCL1 transgenic mouse model will serve as an ideal model to study steps to repair T cell function and their impact on CLL.


1991 ◽  
Vol 174 (1) ◽  
pp. 193-201 ◽  
Author(s):  
M Mieno ◽  
R Suto ◽  
Y Obata ◽  
H Udono ◽  
T Takahashi ◽  
...  

The generation of an in vitro major histocompatibility complex class I specific response of CD4-CD8- T cell receptor (TCR) alpha beta cytotoxic T lymphocytes (CTL) and their allogeneic tumor rejection were investigated. Inocula of BALBRL male 1 were rejected in C57BL/6 (B6) mice treated with minimum essential medium (MEM) (control), anti-L3T4 (CD4) monoclonal antibody (mAb) or anti-Lyt-2.2 (CD8) mAb and CTL against the tumor were generated in vitro. No rejection and no induction of CTL were observed in B6 mice treated with anti-L3T4 (CD4) plus anti-Lyt-2.2 (CD8) mAb. CTL with the classical Thy-1+ CD3+CD4-CD8+ TCR alpha beta phenotype were generated in mixed lymphocyte tumor cell culture (MLTC) spleen cells from B6 mice treated with MEM (control) or anti-L3T4 (CD4) mAb, whereas CTL with an unusual Thy-1+CD3+CD4-CD8- TCR alpha beta phenotype were generated in MLTC spleen cells from anti-Lyt-2.2 (CD8) mAb-treated B6 mice. Both types of CTL were reactive with both H-2Kd and Dd (Ld) class I antigen. These findings suggest that when CD4+ cells were blocked by anti-L3T4 (CD4) mAb, CD8+ CTL mediated rejection, and when CD8+ cells were blocked by anti-Lyt-2.2 (CD8) mAb, CD4+ cells were capable of mediating rejection, although less efficiently than CD8+ cells, by inducing CD4-CD8- TCR alpha beta CTL. The finding that adoptive transfer of CD4 and CD8-depleted MLTC spleen cells, obtained from anti-Lyt-2.2 (CD8) mAb-treated B6 mice that had rejected BALBRL male 1, resulted in rejection of BALBRL male 1 inoculated into B6 nu/nu mice confirmed the above notion. CTL clones with the CD4-CD8- TCR alpha beta phenotype specific for Ld were established.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4969-4969
Author(s):  
Claudia Niemand ◽  
Carina Conrads ◽  
Ramona Siemer ◽  
Mario Assenmacher

Abstract Several publications during the last few years have reported CD25hiCD4+ regulatory T cells (Tregs) to prevent or to reverse disease in different mouse models of experimental autoimmune encephalomyelitis (EAE), colitis, graft rejection and graft-versus-host-disease (GvHD). As mouse and human Tregs share many phenotypical and functional characteristics, Tregs could provide a promising therapeutic approach for various human autoimmune diseases and pathological alloresponses. Here we have shown that Tregs can be isolated from leukapheresis harvests by CD25 enrichment using the CliniMACS technology (n=13). By this procedure we obtained 2.32x108 (± 1.12x108, range 0.71–4.42x108) cells out of 1010 mononuclear cells with a mean purity of 52.12% (± 12.11%, range 25.48–66.61%) for CD25hiCD4+ cells. Around 90% of enriched cells were CD25+CD4+. Among contaminating CD4− cells most cells were CD25+ which were further characterized by counterstaining to be mainly CD19+ B cells and a few CD8+, CD56+ or CD123+ cells. It is possible to deplete the CD19+ or CD8+ cells by using CD19 Microbeads or CD8 Microbeads respectively with the CliniMACS Instrument before CD25 enrichment. Combined depletion of different cells, e.g. CD19+ and CD8+ cells is conceivable. Isolated cells were phenotypically and functionally characterized. The majority of the CD25hiCD4+ T cells expressed glucocorticoid-induced tumor necrosis factor receptor (GITR), CD62L and CD45RO. In addition, isolated cells were able to suppress the proliferation and activation of cocultured conventional CD4+ cells after polyclonal stimulation with anti-CD3 antibody. We conclude that the large-scale isolation of CD25hiCD4+ regulatory T cells for clinical applications (e.g. therapy of autoimmune diseases, graft rejection or GvHD) is possible by using the CliniMACS CD25 Reagent and the CliniMACS Instrument.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 5262-5262
Author(s):  
Emma Morris ◽  
Aristotle Tsallios ◽  
Gavin Bendle ◽  
Shao-an Xue ◽  
Hans Stauss

Abstract CD4 helper T cells play a critical role in the anti-tumour immune response. Cytokines secreted by CD4 T cells can have a direct effect on tumour cells and provide help for CTL priming and effector function. In this study we tested if it was possible to generate MHC class I-restricted helper T cells by retroviral TCR gene transfer into CD4 lymphocytes. Methods: We used a TCR (utilising V11) that recognises the influenza virus A nucleoprotein (NP366–379) peptide in the context of murine Db MHC class I. Murine splenocytes were isolated from C57BL/6 mice (H2b) and activated with conconavalin A and IL-7, and after 48 hours transduced with the pMX-TCR-IRES-TCR retroviral vector. The transduced splenocytes were then cultured in the presence of IL2 for a further 48 hours before staining with anti-murine CD4, CD8 and V11 antibodies and sorting into CD4+ V11+ and CD8+ V11+ populations. Sorted cells were expanded for a further 48–72 hours prior to functional assays. Functional Assays: Purified TCR-transduced (TCR-Td) CD8+ cells and purified TCR-Td CD4+ cells were tested for IFN secretion in response to dendritic cells (DCs) pulsed with NP peptide, an irrelevant peptide (pMDM100) or no peptide. Further experiments examined IFN secretion in response to peptide-loaded tumour cells (EL4 murine lymphoma cells) or transfected tumour cells expressing NP endogenously. Secretion of IFN was measured by ELISA. Results: (1) Antigen-specific IFN secretion was observed by both CD8+ (100% purity) and CD4+ cells (99.93% purity) expressing the class I-restricted TCR when incubated with peptide-loaded DCs. When tested with no peptide or irrelevant peptide, no IFN secretion was observed. The CD8+ cells were more sensitive, recognizing lower concentrations of peptide (10pM) than CD4+ cells (100pM). With peptide-coated EL4 tumour cells as stimulator cells, CD8+ cells showed a peptide-specific response. In contrast, the TCR-Td CD4+ cells were only able to elicit a weak peptide-specific response. Similarly, TCR-Td CD8+ cells were able to recognise NP transfected EL4 tumour cells (EL4NP68), whereas the CD4+ cells were unable to. However, the addition of syngeneic DCs restored the CD4+ cell response to NP transfected EL4 tumour cells to one equivalent to that seen with the TCR-Td CD8+ populations (Table 1). Summary: We have demonstrated that it is feasible to generate MHC class I-restricted CD4+ helper T cells, that are specific for peptide epitopes presented in the context of MHC class I. The CD4+ T cells can recognise antigen-expressing tumour cells in the presence of professional APC, such as DCs. The mechanism by which APC restore tumour recognition may involve trans-costimulation or cross presentation. The data suggest that class I-restricted CD4+ T cells may be able to contribute to enhanced anti-tumour immunity. αββββγγγγγβ γIFN Secretion (ng/ml) After Stimulation with DCs or Tumour Cells T Cell (Responder Cell) Stimulator Cell/s No Peptide NP (100nM) pMDM100 (100nM) Abbreviations: ND not done; DC, EL4 and EL4NP68 as indicated in text. TCR-Td CD8+ DCs 0.1 163.2 0.7 TCR-Td CD8+ EL4 0.1 19.9 0.2 TCR-Td CD8+ EL4NP68 16.6 ND ND TCR-Td CD8+ EL4NP68 + DCs 31.2 ND ND TCR-Td CD4+ DCs 0.1 163.9 0.2 TCR-Td CD4+ EL4 0.1 0.8 0.0 TCR-Td CD4+ EL4NP68 0.2 ND ND TCR-Td CD4+ EL4NP68 + DCs 25.3 ND ND


Blood ◽  
1996 ◽  
Vol 88 (3) ◽  
pp. 962-969 ◽  
Author(s):  
PJ Martin

Clinical trials and experimental studies have demonstrated that donor T cells can play a critical role in preventing allogeneic marrow graft rejection. Results of a previous study showed that donor T cells were most effective for preventing rejection when they recognize an alloantigen expressed by recipient T cells and can cause graft-versus- host disease (GVHD). The present study examined models where marrow graft rejection can be prevented by donor T cells that do not recognize host alloantigens and cannot cause GVHD. Donor T cells prevented rejection of major histocompatibility complex (MHC) class I and II- disparate F1 marrow in parental recipients prepared with > or = 800 cGy total body irradiation (TBI) but not in those prepared with < or = 750 cGy TBI. In recipients prepared with high TBI exposures, rejection was mediated entirely by host CD8 cells. With lower TBI exposures, rejection was mediated by host CD4 cells and CD8 cells. These observations suggested the hypothesis that donor T cells prevent rejection mediated by host effectors that recognize donor MHC class I alloantigens but do not prevent rejection mediated by host effectors that recognize donor class II alloantigens. Consistent with this hypothesis, further experiments showed that F1 donor T cells can prevent rejection of MHC class I-disparate marrow in irradiated parental recipients but have no detectable effect on rejection of MHC class II-disparate marrow. We propose that the expression of MHC class I molecules on donor T cells makes it possible for these cells to inactivate the host response against donor class I alloantigens through a veto mechanism, whereas the absence of MHC class II molecules on murine T cells explains why these cells cannot inactivate the host response against donor class II alloantigens. Finally, donor CD4 cells and CD8 cells were equivalently effective for preventing rejection of F1 marrow in parental recipients, suggesting that veto activity is not restricted solely to the CD8 subset of murine T cells. A veto mechanism could enable donor T cells to prevent allogeneic marrow graft rejection without causing GVHD.


1997 ◽  
Vol 186 (5) ◽  
pp. 645-653 ◽  
Author(s):  
Daniel E. Speiser ◽  
Renata Miranda ◽  
Arsen Zakarian ◽  
Martin F. Bachmann ◽  
Kim McKall-Faienza ◽  
...  

Induction and maintenance of cytotoxic T lymphocyte (CTL) activity specific for a primary endogenous tumor was investigated in vivo. The simian virus 40 T antigen (Tag) expressed under the control of the rat insulin promoter (RIP) induced pancreatic β-cell tumors producing insulin, causing progressive hypoglycemia. As an endogenous tumor antigen, the lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) was introduced also under the control of the RIP. No significant spontaneous CTL activation against GP was observed. However, LCMV infection induced an antitumor CTL response which efficiently reduced the tumor mass, resulting in temporarily normalized blood glucose levels and prolonged survival of double transgenic RIP(GP × Tag2) mice (137 ± 18 d) as opposed to control RIP-Tag2 mice (88 ± 8 d). Surprisingly, the tumor-specific CTL response was not sustained despite the facts that the tumor cells continued to express MHC class I and LCMV-GP–specific CTLs were present and not tolerized. Subsequent adoptive transfer of virus activated spleen cells into RIP(GP × Tag2) mice further prolonged survival (168 ± 11 d), demonstrating continued expression of the LCMV-GP tumor antigen and MHC class I. The data show that the tumor did not spontaneously induce or maintain an activated CTL response, revealing a profound lack of immunogenicity in vivo. Therefore, repetitive immunizations are necessary for prolonged antitumor immunotherapy. In addition, the data suggest that the risk for induction of chronic autoimmune diseases is limited, which may encourage immunotherapy against antigens selectively but not exclusively expressed by the tumor.


2021 ◽  
Vol 11 ◽  
Author(s):  
Florence Bettens ◽  
Zuleika Calderin Sollet ◽  
Stéphane Buhler ◽  
Jean Villard

In transplantation, direct allorecognition is a complex interplay between T-cell receptors (TCR) and HLA molecules and their bound peptides expressed on antigen-presenting cells. In analogy to HLA mismatched hematopoietic stem cell transplantation (HSCT), the TCR CDR3β repertoires of alloreactive cytotoxic CD8+ responder T cells, defined by the cell surface expression of CD137 and triggered in vitro by HLA mismatched stimulating cells, were analyzed in different HLA class I mismatched combinations. The same HLA mismatched stimulatory cells induced very different repertoires in distinct but HLA identical responders. Likewise, stimulator cells derived from HLA identical donors activated CD8+ cells expressing very different repertoires in the same mismatched responder. To mimic in vivo inflammation, expression of HLA class l antigens was upregulated in vitro on stimulating cells by the inflammatory cytokines TNFα and IFNβ. The repertoires differed whether the same responder cells were stimulated with cells treated or not with both cytokines. In conclusion, the selection and expansion of alloreactive cytotoxic T-cell clonotypes expressing a very diverse repertoire is observed repeatedly despite controlling for HLA disparities and is significantly influenced by the inflammatory status. This makes prediction of alloreactive T-cell repertoires a major challenge in HLA mismatched HSCT.


Sign in / Sign up

Export Citation Format

Share Document