Blockade of Cd40-Cd154 Interferes with Human T cell Engraftment in Scid Mice

1998 ◽  
Vol 7 (1) ◽  
pp. 25-35 ◽  
Author(s):  
Teresa M. Foy ◽  
Melissa Mcilraith ◽  
Sally R. Masters ◽  
Jonathan J. Dunn ◽  
Aldo A. Rossini ◽  
...  

Antibodies to the ligand for CD40 (CD154) have been shown to exert profound effects on the development of cell-mediated immune responses in mice. The present study shows that an antibody to human CD154 (hCD40L) inhibits in vivo Tetanus toxoid (TT) specific secondary antibody responses in hu-PBL-scid mice, as well as the expansion of xenoreactive human T cells in the scid mice. A possible cause for the reduced expansion of xenoreactive, human T cells, was the decreased expression of murine B7.1 and B7.2 caused by the administration of anti-hCD40L. Therefore, it may be that defective maturation of murine antigen-presenting cells impeded the priming and expansion of human xenoreactive T cells.

Blood ◽  
1996 ◽  
Vol 88 (2) ◽  
pp. 721-730 ◽  
Author(s):  
H Segall ◽  
I Lubin ◽  
H Marcus ◽  
A Canaan ◽  
Y Reisner

Severe combined immunodeficient (SCID) mice are increasingly used as hosts for the adoptive transfer of human lymphocytes. Human antibody responses can be obtained in these xenogeneic chimeras, but information about the functionality of the human T cells in SCID mice is limited and controversial. Studies using human peripheral blood lymphocytes (PBL) injected intraperitoneally (IP) into SCID mice (hu-PBL-SCID mice) have shown that human T cells from these chimeras are anergic and have a defective signaling via the T-cell receptor. In addition, their antigenic repertoire is limited to xenoreactive clones. In the present study, we tested the functionality of human T cell in a recently described chimeric model. In this system, BALB/c mice are conditioned by irradiation and then transplanted with SCID bone marrow, followed by IP injection of human PBL. Our experiments demonstrated that human T cells, recovered from these hu-PBL-BALB mice within 1 month posttransplant, proliferated and expressed activation markers upon stimulation with anti-CD3 monoclonal antibody. A vigorous antiallogeneic human cytotoxic T-lymphocyte (CTL) response could be generated in these mice by immunizing them with irradiated allogeneic cells. Moreover, anti-human immunodeficiency virus type 1 (HIV-1) Net- specific human CTLs could be generated in vivo from naive lymphocytes by immunization of mouse-human chimeras with a recombinant vaccinia-nef virus. This model may be used to evaluate potential immunomodulatory drugs or cytokines, and could provide a relevant model for testing HIV vaccines, for production of antiviral T-cell clones for adoptive therapy, and for studying human T-cell responses in vivo.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 52-52 ◽  
Author(s):  
Peter Ebeling ◽  
Jan Duerig ◽  
Florian Grabellus ◽  
Ulrich Duehrsen ◽  
Siegfried Seeber ◽  
...  

Abstract In contrast to normal hematopoiesis and acute leukemias, research in CLL still is hampered by the lack of a reliable in vivo model for primary B-CLL. We here report highly reproducible engraftment of B-CLL cells, when 1x10^8 MNC derived from the peripheral blood of CLL patients were transplanted i.v and i.p. into NOD/SCID mice. So far, 14 different CLL samples were investigated in 41 mice. At weeks 4, 8 or 12 mice were sacrificed and bone marrow (BM), spleen, and peritoneal fluid (PF) were analyzed by FACS for human CD19/CD5/CD23/CD45 (B-CLL) cells and CD45/CD3/CD5 (T) cells. Additionally, HE- and immunostaining was performed on spleen sections. Analysis at week 4 revealed engraftment in NOD/SCID mice for 13/14 samples (spleen: 13/14, BM: 4/14, PF: 12/14). B-CLL cells were observed predominantly in the spleen (8.9±2.4% or 9.1±4.4x10^5 cells) and PF (19.0±4.4% or 3.4±1.8x10^5 cells) with much lower engraftment in BM (0.6±0.3% or 0.1±0.1x10^5 cells). Detection of B-CLL cells in peripheral blood could be obtained in 3/14 experiments. Also substantial engraftment of human T-cells was observed in 13/14 experiments (spleen: 13/14, BM: 8/14, PF: 11/14). T-cells engraftment was highest in the spleen (23.8±9.8% or 28.7±13.1x10^5 cells) and somewhat lower in PF (16.4±8.2% or 3.0±1.6x10^5 cells) and BM (7.3±3.8% or 2.9±1.1x10^5 cells). Subpopulation analysis revealed a CD4+ phenotype in 65, 59 and 72 % of T-cells within spleen, PF and BM, respectively. Noteworthy, immunohistological analysis of HE stained spleen sections of engrafted animals revealed a pseudofollicular infiltration with human CD45LCA+ cells along splenic arterioles. Within these pseudofollicles human B-CLL but also CD3+ T-cells were detected. Contribution of B-CLL and T-cells to individual follicles was highly variable ranging from 5–95% for both cell types. When engraftment was analysed separately for the i.p and the i.v. route, engraftment of transplanted cells in PF seemed to be depended on the i.p. route whereas splenic engraftment was obtained following i.v. as well as i.p. injection. Sustained B-CLL engraftment was seen after 8 weeks (spleen: 3.1±1.4% or 7.3±3.1x10^5 total cells; PF: 57.6±23.3% or 1.0±0.5x10^5 cells; n=3 mice) and 12 weeks (spleen: 1.4±1.3% or 0.3±0.3x10^5 cells; PF: 10.2±7.3% or 0.5±0.5x10^5 cells; n=2 mice). Thus, we have shown efficient engraftment of human B-CLL cells in the spleen and PF of NOD/SCID mice. This in vivo model should significantly help to understand B-CLL biology and to test novel therapeutic approaches. The observed pseudofolicular pattern of splenic infiltration supports the theory of T-cells creating a “microenvironment” sustaining the growth of the leukemic B cell clone.


Blood ◽  
2007 ◽  
Vol 110 (6) ◽  
pp. 1797-1805 ◽  
Author(s):  
Agnete Brunsvik Fredriksen ◽  
Bjarne Bogen

Abstract V regions of monoclonal Ig express an exquisite B-cell tumor–specific antigen called idiotype (Id). Id is a weak antigen and it is important to improve immunogenicity of Id vaccines. Chemokine receptors are expressed on antigen-presenting cells (APCs) and are promising targets for Id vaccines. Here we compare monomeric and dimeric forms of MIP-1α and RANTES that target Id to APCs in a mouse B lymphoma (A20) and a multiple myeloma model (MOPC315). MIP-1α was more potent than RANTES. The dimeric proteins were more potent than monomeric equivalents in short-term assays. When delivered in vivo by intramuscular injection of plasmids followed by electroporation, dimeric proteins efficiently primed APCs in draining lymph nodes for activation and proliferation of Id-specific CD4+ T cells. Good anti-Id antibody responses were obtained, and mice immunized only once were 60% to 80% protected in both tumor models. CD8+ T cells contributed to the protection. Antibody responses and tumor protection were reduced when the human Ig hinge = CH3 dimerization motif was replaced with syngeneic mouse counterparts, indicating that tumor-protective responses were dependent on xenogeneic sequences. The results suggest that bivalency and foreign sequences combine to increase the efficiency of chemokine-Id DNA vaccines.


Blood ◽  
1997 ◽  
Vol 89 (5) ◽  
pp. 1800-1810 ◽  
Author(s):  
Sergey Yurasov ◽  
Tobias R. Kollmann ◽  
Ana Kim ◽  
Christina A. Raker ◽  
Moshe Hachamovitch ◽  
...  

To develop an in vivo model wherein human hematopoiesis occurs, we transplanted severe combined immunodeficiency (SCID) mice with either human fetal bone marrow (HFBM) or human fetal liver (HFL). After transplantation of SCID mice with cultured HFBM (BM-SCID-hu mice) or HFL cells (Liv-SCID-hu mice), significant engraftment of the mouse bone marrow (BM) and population of the peripheral blood with human leukocytes was detected. Human colony-forming unit–granulocyte macrophage and burst forming unit-erythroid were detected in the BM of the BM-SCID-hu and Liv-SCID-hu mice up to 8 months after transplantation. When the HFBM or HFL cells were transduced with a retroviral vector before transplantation, integrated retroviral sequences were detected in human precursor cells present in the SCID mouse BM and in leukocytes circulating in the peripheral blood (PB) up to 7 months after transplantation. The PB of the BM-SCID-hu mice also became populated with human T cells after implantation with human thymic tissue, which provided a human microenvironment wherein human pre-T cells from the BM could mature. When the HFBM was retrovirally transduced before transplantation, integrated retrovirus was detected in sorted CD4+CD8+ double positive and CD4+ single positive cells from the thymic implant and CD4+ cells from the PB. Taken together, these data indicated that the BM of our BM-SCID-hu and Liv-SCID-hu mice became engrafted with retrovirally transduced human hematopoietic precursors that undergo the normal human hematopoietic program and populate the mouse PB with human cells containing integrated retroviral sequences. In addition to being a model for studying in vivo human hematopoiesis, these mice should also prove to be a useful model for investigating in vivo gene therapy using human stem/precursor cells.


Blood ◽  
2012 ◽  
Vol 119 (3) ◽  
pp. 696-706 ◽  
Author(s):  
De-Gang Song ◽  
Qunrui Ye ◽  
Mathilde Poussin ◽  
Gretchen M. Harms ◽  
Mariangela Figini ◽  
...  

AbstractThe costimulatory effects of CD27 on T lymphocyte effector function and memory formation has been confined to evaluations in mouse models, in vitro human cell culture systems, and clinical observations. Here, we tested whether CD27 costimulation actively enhances human T-cell function, expansion, and survival in vitro and in vivo. Human T cells transduced to express an antigen-specific chimeric antigen receptor (CAR-T) containing an intracellular CD3 zeta (CD3ζ) chain signaling module with the CD27 costimulatory motif in tandem exerted increased antigen-stimulated effector functions in vitro, including cytokine secretion and cytotoxicity, compared with CAR-T with CD3ζ alone. After antigen stimulation in vitro, CD27-bearing CAR-T cells also proliferated, up-regulated Bcl-XL protein expression, resisted apoptosis, and underwent increased numerical expansion. The greatest impact of CD27 was noted in vivo, where transferred CAR-T cells with CD27 demonstrated heightened persistence after infusion, facilitating improved regression of human cancer in a xenogeneic allograft model. This tumor regression was similar to that achieved with CD28- or 4-1BB–costimulated CARs, and heightened persistence was similar to 4-1BB but greater than CD28. Thus, CD27 costimulation enhances expansion, effector function, and survival of human CAR-T cells in vitro and augments human T-cell persistence and antitumor activity in vivo.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1160-1160
Author(s):  
Christina Hausl ◽  
Josenato Ilas ◽  
Christian Lubich ◽  
Rafi U. Ahmad ◽  
Eva M. Muchitsch ◽  
...  

Abstract Antibody responses against factor VIII (FVIII) are the major complication that arises when patients with hemophilia A are treated with factor VIII products. Therefore, understanding regulation of anti-FVIII immune responses is of outmost importance. Antibody responses are well established to result from differentiation of B cells into antibody-secreting plasma cells. B cells need help from activated CD4+ T cells to develop high-affinity antibody responses against protein antigens such as FVIII. Recently, naturally occurring CD4+CD25+ regulatory T cells have been shown to modulate antibody responses by either suppressing the function of CD4+ T helper cells or by directly acting on B cells. However, the potential importance of CD4+CD25+ T cells in regulating antibody responses to foreign protein antigens is controversial. Furthermore, the extent to which naturally occurring CD4+CD25+ T cells regulate antibody responses against exogenous proteins such as FVIII when these proteins are given to previously untreated patients is unclear. To obtain information on how important naturally occurring CD4+CD25+ T cells are under such conditions, we asked whether these cells regulate anti-FVIII antibody responses in murine hemophilia A. We studied E17 hemophilic mice with two different genetic backgrounds (C57BL/6J and Balb/c) and treated them with four intravenous doses of human FVIII given at weekly intervals. Before the first dose of FVIII, CD4+CD25+ T cells were depleted in vivo using an anti-CD25 antibody that has been shown to deplete naturally occurring CD4+CD25+ T cells in mice. In vivo depletion of regulatory T cells using the same antibody has been successfully applied in a variety of mouse studies to evaluate the significance of naturally occurring CD4+CD25+ T cells in different immunological systems. An isotype-matched control antibody was used as a negative control. A week after the second and the fourth dose of FVIII, plasma samples were taken and tested for anti-FVIII antibodies. We found differences in titers of anti-FVIII antibodies between mice treated with anti-CD25 antibodies and control mice in Balb/c mice but not in C57BL/6J mice. Hemophilic Balb/c mice that had been pre-treated with anti-CD25 antibodies developed higher titers of anti-FVIII antibodies than mice that had been pre-treated with an isotype-matched control antibody. Differences were seen as a statistical trend (p=0.091) after two doses of FVIII and reached statistical significance (p=0.024) after four doses of FVIII. No differences in antibody titers were observed in hemophilic C57BL/6J mice. Our results strongly indicate that the ability of naturally occurring regulatory T cells to modulate anti-FVIII antibody responses in hemophilic mice depends on the genetic background of these mice. Immunoregulatory factors such as cytokines or chemokines as well as differences in the number and functional activity of naturally occurring regulatory T cells that are found in secondary lymphoid organs are likely to determine the regulatory capacity of these cells. Based on our results we conclude that differences in number and functional activity of naturally occurring regulatory T cells should be considered in the search for risk factors associated with the development of FVIII inhibitors in patients.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2448-2448
Author(s):  
Ariane Brunk ◽  
Marion Nonn ◽  
Victoria Lang ◽  
Reinhard Henschler ◽  
Wolfgang Herr ◽  
...  

Abstract Abstract 2448 Poster Board II-425 Donor lymphocyte graft engineering to avoid graft-versus-host (GVH) reactivity while improving graft-versus-leukemia (GVL) immunity remains of central interest in allogeneic hematopoietic stem cell transplantation (HSCT). However, appropriate models to evaluate experimental concepts of donor lymphocyte allograft engineering in vivo are missing. We, therefore, established a human-murine chimeric transplantation model using immunodeficient NOD/SCID/IL2Rγcnull (NSG) mice to evaluate GVH reactivity of human T cell grafts in vivo. Moreover, since mechanisms of immune functions resembling human GVH immunity have not yet been addressed in detail in these humanized mice we started to analyse T cell trafficking and homing to lymphoid tissues across species barrier to examine the clinical relevance of T cell activity observed in this model. To this end, skin substitutes composed of human primary allogeneic fibroblasts embedded in a collagen-based matrix were subcutaneously implanted into NSG mice to detect alloreactive specificities within the implant post adoptive transfer of MHC-mismatched or haploidentical donor T lymphocytes. The skin substitutes revealed murine vascularisation two weeks after implantation as demonstrated by immunohistological studies. Following transfer of human HLA-mismatched or haploidentical T lymphocytes, up to 23% of the T cells migrated into skin substitutes explanted 21 days post injection. As this T cell migration and homing involves both murine and human adhesion molecules we further analyzed the specific adhesion mechanisms underlying the egress of human T cells from the murine bloodstream. Using laminar flow chamber experiments and real time video recordings we could first demonstrate that human anti-CD3/anti-CD28 preactivated T lymphocytes but not naive T cells bound and firmly adhered to the murine endothelial cell line bEND.3 (BEND3.EC) at shear stresses of up to 3.5 dynes/cm2. As controls, human umbilical vein endothelial cells (HUVEC) were used. Adhesion and transmigration was significally enhanced when both human and murine endothelial cells (ECs) were prestimulated with low doses of TNF-α (5-20ng/ml) to resemble an activated phenotype. Firm adhesion of activated T lymphocytes was suppressed following pretreatment with function-blocking anti-integrin-alpha 4 (CD49d, subunit of VLA-4) or anti-integrin-alpha L (CD11a, subunit of LFA-1) antibody (Ab) or when ECs were preincubated with anti-VCAM-1 (CD106). No inhibitory effects were observed when anti-Endoglin (CD105) Ab was included as specificity control suggesting that the integrin dimer VLA-4 and its counter-receptor VCAM-1 as well as the integrin dimer LFA-1 are required for the transmigration of human T cells across murine ECs. Primary ECs derived from murine aorta are currently used to confirm our results obtained with bEND.3 cells. As the same ligand-receptor pairs are described for human T-EC interaction these findings indicate a closely related mechanism of T cell extravasation in human and murine endothelium at least in our transplantation model. In conclusion these results suggest that intravenously transferred activated human T cells migrate into allogeneic skin substitutes involving VCAM-1 and integrin-alpha-4 for firm adhesion followed by transmigration. In vivo studies investigating the effects of the function-blocking antibodies against VCAM-1 and integrin-alpha-4 in our model to confirm the in vitro results are in progress and will be reported. In addition, our human-murine chimeric NSG transplantation model may represent a promising tool to study human GVH biology and to evaluate T cell graft engineering in allogeneic HSCT. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2326-2326
Author(s):  
Davide Bagnara ◽  
Matthew Kaufman ◽  
Carlo Calissano ◽  
Sonia Marsilio ◽  
Philip Chum ◽  
...  

Abstract Abstract 2326 Poster Board II-303 B-cell type chronic lymphocytic leukemia (B-CLL), an incurable disease of unknown etiology, results from the clonal expansion of a CD5+CD19+ B lymphocyte. Progress into defining the cell of origin of the disease and identifying a stem cell reservoir has been impeded because of the lack of a reproducible model for growing B-CLL cells in vivo. At least one possible cause for this is the murine microenvironment's inability to support B-CLL survival and proliferation. To overcome this barrier, we reconstituted NOD/SCID/γnull mice by intrabone (ib) or intravenous (iv) injection of 1 × 105 CD34+ cord blood cells along with ∼106 bone marrow-derived human mesenchymal stem cells (hMSCs) by ib injection. After human cellular engraftment, a total of 108 CFSE-labeled PBMCs from individual B-CLL patients were injected into the same bones or iv. Every two weeks thereafter, blood from the mice was examined for the presence of cells bearing CFSE, human CD45, and various human lineage markers by flow cytometry. In the presence of a human hematopoietic microenvironment derived from hHSCs, CFSE+CD5+CD19+ cells were readily detected in the blood of mice and many of these leukemic cells underwent at least 6 cell doublings. In contrast, the numbers of leukemic cells circulating in the blood of mice reconstituted with hMSCs without CD34+ cells was much less and these failed to proliferate. Thus, hMSCs were not essential in the model. Moreover, the percentage of leukemic cells expressing CD38 in the CFSE+CD5+CD19+ cell fraction was similar to that in the donor patient inoculum only in the mice in which B-CLL cell proliferation occurred. The percentage and intensity of CD38-expressing B-CLL cells was higher in the spleen and bone marrow (BM), far exceeding that in the blood and peritoneum. Of note, B-CLL cells formed follicular structures in the spleen that contained larger B cells expressing the same Ig H and Ig L chains as the CLL MNCs. CLL cells from these spleens exhibited the same IGHV/D/J rearrangement as in the donor leukemic cells, indicating their leukemic origin. These follicular structures are reminiscent of proliferation centers/pseudofollicles seen in patient lymph nodes and BM, in that they contained leukemic B cells of intermediate and large size. Finally, B-CLL cells adoptively transferred into these mice exhibit kinetics similar to those observed in patients in vivo, with birth rates calculated from CFSE-dilution data of (X-Y% per day). Robust T-cell expansion occurred in mice receiving CD34+ cells and occasionally in mice (10-20%) not receiving hCD34+ cells. Based on detailed SNP analyses, the expanded T cells were of B-CLL patient origin and not from hCD34+ cells. Notably when T cells were eliminated by injecting an anti-CD3 mAb (OKT3), B-CLL cell proliferation was inhibited. These latter two findings clearly indicate a need for autologous T lymphocytes in the successful adoptive transfer of B-CLL cells in this model and also highlight the fact that another cell type, derived from the normal allogeneic CD34+ cells, is needed. Because CD34+ cells could be substituted for by mature allogeneic monocytes or B lymphocytes from the blood of normal individuals, antigen-presenting cells may be the key cells that develop from the normal hHSCs. This would suggest that an allogeneic mixed lymphocyte reaction is needed for successful B-CLL cell survival and proliferation. This is consistent with finding that most of the mice with significant T-cell overexpansion died within 6 weeks of B-CLL cell injection from apparent graft vs. host disease. Finally, preliminary data using Rituxan (anti-CD20 mAb) to eliminate B-CLL cells from recipient mice suggest that this model is a good tool to perform pre-clinical studies on the efficacy of action of novel therapeutics. In summary, these studies indicate that allogeneic human antigen-presenting cells and autologous human T cells permit adoptive xenogeneic transfer and clonal expansion of B-CLL cells in immune deficient mice. This model will be useful in discovering and understanding non-genetic factors promoting B-CLL expansion because it recapitulates several features of human B-CLL. Finally, the model may help in the study of the basic biology of this disease, such as if leukemic stem cells exist, and also in conducting pre-clinical tests on possible new therapeutics. Disclosures: No relevant conflicts of interest to declare.


2016 ◽  
Vol 213 (2) ◽  
pp. 167-176 ◽  
Author(s):  
Yuwen Zhu ◽  
Alessandro Paniccia ◽  
Alexander C. Schulick ◽  
Wei Chen ◽  
Michelle R. Koenig ◽  
...  

T cell immunoglobulin and ITIM domain (TIGIT) and CD226 emerge as a novel T cell cosignaling pathway in which CD226 and TIGIT serve as costimulatory and coinhibitory receptors, respectively, for the ligands CD155 and CD112. In this study, we describe CD112R, a member of poliovirus receptor–like proteins, as a new coinhibitory receptor for human T cells. CD112R is preferentially expressed on T cells and inhibits T cell receptor–mediated signals. We further identify that CD112, widely expressed on antigen-presenting cells and tumor cells, is the ligand for CD112R with high affinity. CD112R competes with CD226 to bind to CD112. Disrupting the CD112R–CD112 interaction enhances human T cell response. Our experiments identify CD112R as a novel checkpoint for human T cells via interaction with CD112.


Blood ◽  
1996 ◽  
Vol 87 (7) ◽  
pp. 2930-2937 ◽  
Author(s):  
C Renner ◽  
S Bauer ◽  
U Sahin ◽  
W Jung ◽  
R van Lier ◽  
...  

Cure of a single established human Hodgkin's tumor growing subcutaneously in severe combined immunodeficient (SCID) mice can be achieved with a complex protocol using two bispecific monoclonal antibodies (Bi-MoAb) directed against the Hodgkin's associated CD30 antigen and the T-cell triggering molecules CD3 and CD28, respectively, together with human T cells prestimulated in vitro with Bi-MoAbs in the presence of CD30+ cells. To adapt this model to the clinical situation, disseminated tumors were established in SCID mice by intravenous injection of 2 x 10(7) cells of the Hodgkin's derived cell line L540CY. Treatment of SCID mice bearing disseminated CD30+ Hodgkin's tumors with the combination of CD3/CD30 and CD28/CD30 Bi-MoAbs and naive (ie, not in vitro prestimulated) human T cells resulted in the cure of all appropriately treated animals. T lymphocytes obtained from patients with advanced stage untreated Hodgkin's disease were as effective as lymphocytes from healthy controls. Treatment was effective even when delayed until 2 weeks after tumor inoculation, and application of Bi- MoAbs into SCID mice with circulating human T cells was as effective as injecting the Bi-MoAbs before the lymphocytes. Treatment results with isolated CD4+ and CD8+ human T cells suggest that both subsets are necessary for the Bi-MoAb mediated cure of xenografted human tumors in vivo. The efficacy and practicability of this preclinical immunotherapy protocol support and form the basis for the clinical evaluation of this approach in patients with Hodgkin's disease resistant to standard therapy.


Sign in / Sign up

Export Citation Format

Share Document