scholarly journals Quantitative assessment of mouse skin transplant rejection using digital photography

2005 ◽  
Vol 39 (2) ◽  
pp. 209-214 ◽  
Author(s):  
F Schwoebel ◽  
J Barsig ◽  
A Wendel ◽  
J Hamacher

Mouse skin transplantation is an established in vivo model used to investigate the T-cell-mediated immune response of acute allograft rejection. The critical endpoint of this model is complete rejection of the allograft. However, visual judgement of this end stage of rejection is an arbitrary process and difficult to standardize. To overcome this problem, we established a monitoring method based on digital photography. Serial pictures from skin allografts of individual animals (C57BL/6 on BALB/c) were taken with a digital camera mounted on a microsurgical microscope. Thereby, the description and the correct assessment of early stages of rejection were possible due to the magnification of grafts by the microscope. Rejection scores were introduced to describe different stages from retained to completely rejected grafts. With cyclosporine A as a standard immunosuppressive treatment, we showed that early stages of skin rejection were unambiguously identified. This procedure allows the earlier termination of the experiment and reduction of animal distress, and it can be re-evaluated anywhere and any time after completion. This study demonstrates the suitability of monitoring experimental skin allograft rejection by digital photography, entailing several refinements in animal experimentation, both for the researcher and for the animal.

2021 ◽  
Vol 12 ◽  
Author(s):  
Heng Xu ◽  
Jizhang Yu ◽  
Jikai Cui ◽  
Zhang Chen ◽  
Xi Zhang ◽  
...  

Although studies in oncology have well explored the pharmacological effects of Birc5, little is known about its role in allogeneic T-cell responses. Therefore, the present study used a mouse model of acute heart allograft rejection to investigate the protective effect and mechanism of conditional knockout of Birc5 in T cells. Survivin (encoded by Birc5) was up-regulated in T cells activated in vivo and in vitro. Deletion of Birc5 in T cells attenuated acute heart allograft rejection by reducing the ratio of effector to naive T cells and Th1 to Tregs. In addition, deletion of Birc5 had no noticeable effect on proliferation but on apoptosis and the secretion of IFN-γ. The results revealed a significant increase in the percentage of Annexin V positive CD4+ T cells in the Birc5-/- group, compared to the WT. Moreover, there was significant increase in early apoptotic alloreactive T cells in Birc5-/- mice and this was partly mediated by caspase-3. Furthermore, treatment with YM155 inhibited acute heart allograft rejection in vivo and increased T-cell apoptosis in healthy human PBMCs in vitro. The results highlight a potential therapeutic target for the prevention and treatment of acute transplant rejection.


1999 ◽  
Vol 67 (7) ◽  
pp. S227
Author(s):  
N. Utku ◽  
M. Seifert ◽  
S. Fu ◽  
S. G. Tulius ◽  
T. Heinemann ◽  
...  

Blood ◽  
1998 ◽  
Vol 91 (1) ◽  
pp. 324-330 ◽  
Author(s):  
Liming Yang ◽  
Barb Du Temple ◽  
Qasim Khan ◽  
Li Zhang

Abstract Pretransplantation donor-specific transfusion (DST) can enhance allograft survival in man and animals. However, due to the lack of a specific marker to identify donor-reactive cells in vivo in man and normal (nontransgenic) animals, the underlying mechanism remains unknown. In this study, we use 2CF1 transgenic mice expressing a transgenic T-cell receptor (TCR) specifically recognizing Ld, a major histocompatibility complex (MHC) class I molecule, to delineate the role of DST in long-term skin allograft survival and its underlying mechanisms. Our main findings include: (1) in the absence of any other immunosuppressive treatment, a single dose pretransplantation infusion of viable splenocytes from an Ld+ donor is sufficient to induce permanent donor-specific skin allograft survival in 2CF1anti-Ld TCR transgenic mice; (2) DST leads to a deletion of the majority (>60%) of donor-reactive T cells in the periphery of the recipient. However, deletion does not necessarily result in tolerance; (3) remaining donor-reactive T cells from DST-treated mice are fully responsive to Ld in vitro, and can suppress the antidonor response of naive T cells in vitro only when exogenous interleukin (IL)-4 is provided; and (4) the sera level of IL-4 in DST-treated tolerant mice is significantly increased. These results suggest that the generation of a subset of T cells with the potential to specifically inhibit antidonor responses, together with promotion of IL-4 production in recipients, may be important mechanisms for the induction and maintenance of antigen-specific tolerance.


1990 ◽  
Vol 64 (01) ◽  
pp. 161-164 ◽  
Author(s):  
Rüdiger E Scharf

SummaryRenal transplant rejection is associated with platelet activation in vivo which may lead to partially α- and γ-granule-depleted platelets that continue to circulate. These “exhausted” platelets are hemostatically defective. Tb quantitate the extent of platelet granule depletion following kidney transplantation, we determined intraplatelet levels of β-thromboglobulin (βTG), platelet factor 4 (PF4), and serotonin (5-hydroxytryptamine, 5-HT) ex vivo in Tiiton X-1O0-treated platelet lysates. To explore biochemical alterations of partially depleted platelets, we studied platelet thromboxane A2 (TXA2) synthesis in citrated plateletrich plasma (PRP) upon stimulation with thrombin or collagen in 45 recipients of renal allografts and 10 healthy volunteers. The patients were divided into subjects with acute and chronic allograft rejection (N = 15), those with compensated renal failure after kidney transplantation but without evidence of allograft rejection (N = 15), and those with functioning renal transplant (N = 15). The mean intraplatelet content of βTG (38.6 ± 4.2 μE/109 platelets), PF4 (11.8 ± 1.8 μg/109 platelets), and 5-HT (274 ± 31 μg/109 platelets) in patients with acute or chronic renal allograft rejection was significantly lower than in other recipients off < idney transplants or healthy volunteers (βTG: 59.9±4.7 μgl 109 platelets; PF4: 20.4±2.3 ¼g/n platelets; s-rrr: 46lraB ngl 10e platelets; p < 0.ffi5 in all casls). Platelet TxB2 formation upon stimulation with thrombin (10 U/ml) or collagen (6.25 ¼g/ml) for 5 min was significantly reduced in patients with acute or chronic renal allograft rejection (2.25±0.29 and 0.641 0.08 nmoUl0e platelets for thrombin- and collagen-stimulated platelets, respectively) compared to that of healthy volunteers (4.72± 0.60 and 1.35 ± 0.12 nmol/109 platelets, respectively; p <0.05 in all cases). In contrast, platelet TXB2 formation of patients with functioning kidney transplant or those with compensated renal failure but without evidence of transplant rejection did not differ significantly from that of normals. These results confirm that platelets with reduced levels of α- and β-granular constituents are detectable in the circulation following kidney transplantation when acute or chronic renal allograft rejections occur. These platelets are incapable of forming normal amounts of thromboxane upon stimulation with thrombin and collagen in vitro. This dysfunction of thromboxane synthesis, due to alterations in the platelet arachidonate pathway, may reflect the previous activation of platelets in vivo associated with acute or chronic renal allograft rejection.


1997 ◽  
Vol 63 (7) ◽  
pp. 1040-1042 ◽  
Author(s):  
Vladimir L. Motin ◽  
Susan M. Kutas ◽  
Robert R. Brubaker

1987 ◽  
Vol 165 (5) ◽  
pp. 1296-1315 ◽  
Author(s):  
A S Rosenberg ◽  
T Mizuochi ◽  
S O Sharrow ◽  
A Singer

In the present study we used an adoptive transfer model with athymic nude mice to characterize the T cells involved in initiating and mediating skin allograft rejection. It was found that skin allograft rejection in nude mice required the transfer of immunocompetent T cells and that such reconstitution did not itself stimulate the appearance of T cells derived from the nude host. Reconstitution with isolated populations of Lyt-2+/L3T4- T cells resulted in the rapid rejection of MHC class I-disparate skin allografts, whereas reconstitution with isolated populations of L3T4+/Lyt-2- T cells resulted in the rapid rejection of MHC class II-disparate and minor H-disparate skin allografts. By correlating these rejection responses with the functional capabilities of antigen-specific T cells contained within the reconstituting Lyt-2+ and L3T4+ T cell populations, it was noted that skin allografts were only rejected by mice that, as shown by in vitro assessment, contained both lymphokine-secreting Th cells and lymphokine-responsive Tk cells specific for the alloantigens of the graft. The ability of two such functionally distinct T cell subsets to interact in vivo to reject skin allografts was directly demonstrated in H-Y-specific rejection responses by taking advantage of the fact that H-Y-specific Th cells are L3T4+ while H-Y specific Tk cells are Lyt-2+. Finally, the importance of in vivo interactions between functionally distinct Th/T-inducer cells and T killer (Tk)/T-effector cells in skin allograft rejection was demonstrated by the observation that normal B6 mice retain Qala and Kbm6 skin allografts because of a selective deficiency in antigen-specific Th cells, even though they contain T-effector cells that, when activated, are able to reject such allografts. Thus, the ability to reject skin allografts is neither unique to a specialized subset of T cells with a given Lyt phenotype, nor unique to a specialized subset of helper-independent effector T cells with so-called dual function capability. Rather, skin allograft rejection can be mediated by in vivo collaborations between T-inducer cells and T-effector cells, and the two interacting T cell subsets can express different Lyt phenotypes as well as different antigen specificities.


Blood ◽  
1998 ◽  
Vol 91 (1) ◽  
pp. 324-330
Author(s):  
Liming Yang ◽  
Barb Du Temple ◽  
Qasim Khan ◽  
Li Zhang

Pretransplantation donor-specific transfusion (DST) can enhance allograft survival in man and animals. However, due to the lack of a specific marker to identify donor-reactive cells in vivo in man and normal (nontransgenic) animals, the underlying mechanism remains unknown. In this study, we use 2CF1 transgenic mice expressing a transgenic T-cell receptor (TCR) specifically recognizing Ld, a major histocompatibility complex (MHC) class I molecule, to delineate the role of DST in long-term skin allograft survival and its underlying mechanisms. Our main findings include: (1) in the absence of any other immunosuppressive treatment, a single dose pretransplantation infusion of viable splenocytes from an Ld+ donor is sufficient to induce permanent donor-specific skin allograft survival in 2CF1anti-Ld TCR transgenic mice; (2) DST leads to a deletion of the majority (>60%) of donor-reactive T cells in the periphery of the recipient. However, deletion does not necessarily result in tolerance; (3) remaining donor-reactive T cells from DST-treated mice are fully responsive to Ld in vitro, and can suppress the antidonor response of naive T cells in vitro only when exogenous interleukin (IL)-4 is provided; and (4) the sera level of IL-4 in DST-treated tolerant mice is significantly increased. These results suggest that the generation of a subset of T cells with the potential to specifically inhibit antidonor responses, together with promotion of IL-4 production in recipients, may be important mechanisms for the induction and maintenance of antigen-specific tolerance.


Blood ◽  
2005 ◽  
Vol 105 (12) ◽  
pp. 4871-4877 ◽  
Author(s):  
Mahzuz Karim ◽  
Gang Feng ◽  
Kathryn J. Wood ◽  
Andrew R. Bushell

Abstract The importance of CD25+CD4+ regulatory T (Treg) cells in the control of immune responses is established, but their antigen specificity in vivo remains unclear. Understanding Treg-cell specificity requirements will be important if their potential is to be developed for immunotherapy. Pretreatment of recipient mice with donor alloantigen plus anti-CD4 antibody generates CD25+CD4+ Treg cells with the capacity to prevent skin allograft rejection in adoptive transfer recipients. Here we demonstrate that, although this regulation can be antigen-specific, reactivation with the original tolerizing alloantigen allows the Treg cells to suppress rejection of third-party allografts. Aware of the limitations of alloantigen pretreatment, we asked whether graft-protective Treg cells could be generated against unrelated, nongraft antigens. We demonstrate that bystander regulation also extends to CD25+CD4+ Treg cells generated in vivo by exposure to nominal antigens under anti-CD4 antibody cover. Providing these Treg cells are reexposed to the tolerizing antigens before adoptive transfer, they prevent the rejection of fully allogeneic skin grafts. That this might form the basis of a clinically relevant tolerance induction strategy is demonstrated by the fact that, when combined with subtherapeutic anti-CD8 antibody, Treg cells generated in response to nongraft antigens facilitate the acceptance of cardiac allografts in primary recipients. (Blood. 2005;105:4871-4877)


Sign in / Sign up

Export Citation Format

Share Document