• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 2
  • 2
  • Tagged with
  • 12
  • 12
  • 7
  • 7
  • 5
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 3
  • 3
  • 3
  • 3
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Tolerance to allografts using recipient bone marrow cells trandsduced with a donor MHC gene

Wong, Wilson January 1997 (has links)
No description available.
2

Regulation of Alloreactive CD8 T Cell Responses by Costimulation and Inflammation

Jangalwe, Sonal 30 June 2017 (has links)
CD8 T lymphocytes are a crucial component of the adaptive immune system and mediate control of infections and malignancy, but also autoimmunity and allograft rejection. Given their central role in the immune system, CD8 T cell responses are tightly regulated by costimulatory signals and cytokines. Strategies targeting signals that are critical for T cell activation have been employed in a transplantation setting to impede alloreactive T cell responses and prevent graft rejection. The goal of my thesis is to understand how costimulatory signals and inflammation regulate alloreactive CD8 T cell responses and how to target these pathways to develop more effective tools to prevent graft rejection. Costimulation blockade is an effective approach to prolong allograft survival in murine and non-human primate models of transplantation and is an attractive alternative to immunosuppressants. I describe a novel murine anti-CD40 monoclonal antibody that prolongs skin allograft survival across major histocompatibility barriers and attenuates alloreactive CD8 T cell responses. I find that the pro-apoptotic proteins Fas and Bim function concurrently to regulate peripheral tolerance induction to allografts. Activation of the innate immune system by endogenous moIecules released during surgery or infections in transplant recipients can modulate T cell responses. However, the direct impact of inflammation on alloreactive CD8 T cell responses is not clear. Using a T cell receptor (TCR) transgenic mouse modeI, I demonstrate that inflammatory stimuli bacterial lipopolysaccharide (LPS) and the viral dsRNA mimetic poly(I:C) differentially regulate donor-reactive CD8 T cell responses by generating distinct cytokine milieus. Finally I demonstrate the role of pro-inflammatory cytokines stem cell factor (SCF), granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-3 (IL-3) in improving human B cell development in humanized NOD-scid IL2Rγnull (NSG) mice.
3

Nanoparticle use in the modulation of transplant rejection in a murine model

Kassis, Elias Noah 10 September 2010 (has links)
Solid organ transplant has emerged over the last half century as an important treatment for solid organ failure. Management has matured dramatically over the past two decades with improvements in acute rejection, but long-term graft survival has improved very little and current treatment is limited by the side-effects and toxicities of immunosuppressive medications. Nanoparticle delivery of therapeutics, improving transport characteristics and decreasing systemic and local toxicity has emerged as a dynamic treatment modality, but little work has been done using nanoparticles in transplantation. Our research examined the use of CD4-targeted nanoparticles encapsulated with mycophenolic acid (MPA), a commonly used immunosuppressant in organ transplantation. This work is the first to examine antigen-specific targeting of nanoparticles in any transplant model. MPA-loaded particles show a slow and continuous release profile and biodistribution suggested retention in the spleen. Targeting of nanoparticles to CD4 T cells was suggested using ex vivo and in vitro flow cytometry. In the fully allogeneic MHCII mismatch BALB/C to C57BL/6 mice we found improved graft survival in the non-targeted MPA group and even greater graft survival in the CD4-targeted group. Targeted and non-targeted particle groups showed equal delay in rejection in the less immunogenic single MHC mismatch B6.H-2bm12 to C57BL/6 model that we showed to be CD4 dependent. In both models, graft survival times were increased over free drug and controls with roughly one thousand fold lower dose of drug in the nanoparticles as compared with free MPA. Consistent with these findings were decreased proliferation with targeted and non-targeted MPA-nanoparticles using in vitro and ex vivo mixed lymphocyte reactions. We postulated that the similar rejection times in targeted and non-targeted groups was due to dendritic cell (DC) involvement and we found active uptake of nanoparticles in DCs, a decrease in inflammatory cytokine production and a decrease in treated DCs ability to stimulate T cells via mixed lymphocyte reactions. Furthermore we found a possible mechanism in the DC interaction with T cells through the upregulation of the inhibiting co-stimulatory molecules B7-DC and B7-H1 on DCs treated with MPA-nanoparticles. We also found possible upregulation of CD4+CD25+ Foxp3 expressing Tregs which may serve to increase graft acceptance. These results explore the involvement of dendritic cells in the process of nanoparticle-induced graft acceptance and suggest the feasibility of using nanoparticle drug vectors in clinical transplant.
4

Induction and Maintenance of Transplantation Tolerance by Treatment with a Donor Specific Transfusion and Anti-CD154 mAb: a Dissertation

Iwakoshi, Neal N. 05 September 2000 (has links)
A two-element protocol consisting of one donor-specific transfusion (DST) plus a brief course of anti-CD154 mAb greatly prolongs the survival of murine islet, skin, and cardiac allografts. To study the mechanisms involved in the induction of allograft survival, we determined the fate of tracer populations of alloreactive T cell receptor (TcR) transgenic CD8+ T cells circulating in a normal microenvironment. In the first portion of this thesis, we observed that DST plus anti-CD154 mAb prolonged allograft survival and deleted alloreactive TcR transgenic CD8+ T cells. Neither component alone did so. Skin allograft survival was also prolonged in normal recipients treated with anti-CD154 mAb plus a depleting anti-CD8 mAb and in C57BL/6-CD8 knockout mice treated with anti-CD154 mAb monotherapy. We conclude that, in the presence of anti-CD154 mAb, DST leads to an allotolerant state in part by deleting alloreactive CD8+ T cells. Consistent with this conclusion, blockade of CTLA4 and B7-l/2 by CTLA4-Ig, which is known to abrogate the effects of DST and anti-CD154 mAb, prevented the deletion of alloreactive TcR transgenic CD8+ T cells. Also in support of our hypothesis, depletion of CD4+ T cells, which is known to abrogate the effects of DST and anti-CD154 mAb, prevented the deletion of alloreactive TcR transgenic CD8+ T cells. We continued by examining the effects of IFN-γ, IL-10 and IL-4. None of these cytokines had any significant effect on the deletion of alloreactive TcR transgenic CD8+ T cells induced by co-stimulation blockade. The last part of this thesis studied the behavior of alloreactive TcR transgenic CD8+ T cells during the maintenance phase of allograft survival induced by our two-element protocol. Using a hematopoietic TcR transgenic chimera system, our results demonstrated that levels of alloreactive CD8+ T cells remained low throughout the maintenance phase. These results document for the first time that peripheral deletion of alloantigen-specific CD8+ T cells is an important mechanism through which allograft survival can be prolonged by co-stimulatory blockade. We propose a unifying mechanism to explain allograft prolongation by DST and blockade by co-stimulation blockade.
5

Essential amino acid depletion by embryonic stem cells as a mechanism of immune privilege

Ichiryu, Naoki January 2013 (has links)
Mouse embryonic stem cells (ESCs) are capable of differentiating into any somatic cell type and are known to display fragile immune privilege in vivo and in vitro. The extent to which the depletion of essential amino acids (EAAs) by ESCs contributes to this phenomenon was investigated. ESCs were found to express various enzymes capable of catabolising EAAs within the culture medium. In particular, depletion of threonine, valine and lysine was found to have significant impact on T cell proliferation and differentiation, biasing their polarisation towards a FoxP3<sup>+</sup> T regulatory (T<sub>reg</sub>) phenotype. Supplementing ESC conditioned medium with these three EAAs alone rescued normal T cell proliferation, whereas artificially limiting their availability was sufficient to induce T<sub>reg</sub> cell differentiation to a level equivalent to general EAA depletion. The pattern of EAA catabolism by mouse ESC was shared by induced pluripotent stem cells, while mouse melanoma cell lines and human ESCs displayed distinct patterns of EAA depletion. The cytosolic branched chain aminotransferase enzyme, Bcat1, catalyses the first step of branched chain amino acid catabolism (isoleucine, leucine and valine), and is highly expressed by both mouse and human ESCs. The contribution of this enzyme to the establishment of acquired immune privilege by ESC-derived tissues was, therefore, investigated. ESC lines were derived from mice lacking Bcat1 activity and were characterised. Bcat1<sup>−/−</sup> ESC lines displayed no difference to their wildtype counterparts (Bcat1<sup>LoxP</sup>) in terms of in vitro proliferation and their capacity to form teratomas in vivo. Furthermore, the loss of Bcat1 function had little impact on the inhibition of T cell proliferation in culture, ability to induce T<sub>reg</sub> cell commitment or their ability to prevent rejection by T cell receptor transgenic recipients, suggesting the minimal contribution of Bcat1 to the depletion of EAAs by ESCs. In conclusion, EAA depletion by mouse ESC may provide a mechanistic explanation for the previously described immune-suppressive capacity of ESC.
6

Everolimus-Induced Immune Effects after Heart Transplantation: A Possible Tool for Clinicians to Monitor Patients at Risk for Transplant Rejection

Klaeske, Kristin, Lehmann, Sven, Palitzsch, Robert, Büttner, Petra, Barten, Markus J., Jawad, Khalil, Eifert, Sandra, Saeed, Diyar, Borger, Michael A., Dieterlen, Maja-Theresa 05 May 2023 (has links)
Background: Patients treated with an inhibitor of the mechanistic target of rapamycin (mTORI) in a calcineurin inhibitor (CNI)-free immunosuppressive regimen after heart transplantation (HTx) show a higher risk for transplant rejection. We developed an immunological monitoring tool that may improve the identification of mTORI-treated patients at risk for rejection. Methods: Circulating dendritic cells (DCs) and regulatory T cells (Tregs) were analysed in 19 mTORI- and 20 CNI-treated HTx patients by flow cytometry. Principal component and cluster analysis were used to identify patients at risk for transplant rejection. Results: The percentages of total Tregs (p = 0.02) and CD39+ Tregs (p = 0.05) were higher in mTORI-treated patients than in CNI-treated patients. The principal component analysis revealed that BDCA1+, BDCA2+ and BDCA4+ DCs as well as total Tregs could distinguish between non-rejecting and rejecting mTORI-treated patients. Most mTORI-treated rejectors showed higher levels of BDCA2+ and BDCA4+ plasmacytoid DCs and lower levels of BDCA1+ myeloid DCs and Tregs than mTORI non-rejectors. Conclusion: An mTORI-based immunosuppressive regimen induced a sufficient, tolerance-promoting reaction in Tregs, but an insufficient, adverse effect in DCs. On the basis of patient-specific immunological profiles, we established a flow cytometry-based monitoring tool that may be helpful in identifying patients at risk for rejection.
7

MicroRNAs na tolerância operacional no transplante renal humano / MicroRNAs levels in operational tolerance (OT) in human transplantation tolerance

Silva, Amanda Cabral da 04 June 2018 (has links)
A tolerância operacional (TO) é um estado de estabilidade funcional do órgão transplantado, após a parada de drogas imunossupressoras, por um período maior ou igual a um ano. Os microRNAs são pequenas moléculas de RNA não codificantes que regulam, negativamente, a expressão gênica de seus alvos, incluindo de moléculas relacionadas ao sistema imune, desempenhando um papel crítico na manutenção da homeostase. Nosso objetivo foi determinar se há um perfil diferencial de microRNAs na TO no transplante renal humano, indicando sua potencial participação nos mecanismos de tolerância. Analisamos o perfil sérico dos níveis de microRNAs na tolerância operacional (TO) (n= 8), comparando com rejeição crônica (RC) (n= 5), estáveis em uso de imunossupressão convencional (EST) (n= 5) e indivíduos saudáveis (SAU) (n= 5), utilizando, inicialmente, um painel de primers para 768 miRNAs, pela tecnologia TLDA (TaqMan Low Density Array). Detectamos um perfil diferencial nos níveis de microRNAs entre TO e o seu desfecho clínico oposto - RC- sugerindo que microRNAs podem integrar a rede de mecanismos envolvidos na tolerância ao transplante. Nesse perfil diferencial, alguns tiveram níveis maiores na TO: miR-885-5p (p=0,031), miR-331 (p=0,009), miR-27a (p=0,033), em comparação com RC, enquanto outros, o miR-1233-3p (p=0,029), miR-572 (p=0,028), miR-1260a (p=0.017) e o miR-638 (p=0,047) apresentaram menores níveis na TO. Na comparação de TO com o estado fisiológico (SAU), 2 dos microRNAs do perfil diferencial TO x RC apresentaram níveis diminuídos na TO: miR-27a-5p, (p=0,033) miR-1260a (p=0,017) e, para os demais, não encontramos diferenças de níveis, indicando predomínio de preservação do perfil na TO, em relação ao estado fisiológico. Por bioinformática, foram preditas vias de sinalização e observamos que os genes alvos desses microRNAs tiveram, predominantemente, relação com morte celular, integrando as vias de granzima e receptor de morte. Considerando os diversos alvos dos microRNAs do perfil diferencial e seus potenciais efeitos de favorecer vida e morte, nessas duas vias, encontramos uma razão de vida/morte de 1,95 na TO e de 0,39 na RC, indicando o maior potencial de promoção de morte na RC e, de vida, na TO, pela ação desses microRNAs. Esses dados indicam que a regulação de vias relacionadas à morte celular pode integrar os mecanismos de tolerância imunológica no transplante renal humano. Selecionamos o miR-885-5p para validar os achados, em um número maior de amostras, e confirmamos maiores níveis na TO (n=8), em relação à RC (n=12; p=0,0063) e também em relação ao SAU (n=12; p=0,0035). Considerando que CASP3 é um dos alvos do miR-885, interpretamos que a sua ação, na TO, pode promover a menor expressão de CASP3 e, consequentemente, favorecer a sobrevivência celular. Concluímos que mecanismos epigenéticos podem integrar a rede de mecanismos da tolerância ao transplante, como por meio da ação de microRNAs regulando a expressão de genes envolvidos com sobrevida/morte celular / Operational tolerance (OT) is a state of functional stability of the transplanted organ, after stopping immunosuppressive drugs for a period of at least one year. MicroRNAs are small non-coding RNA that downregulate gene expression of their targets, including genes related to the immune system, playing a critical role in keeping homeostasis. Our objective was to determine whether there is a differential profile of microRNAs in OT in human renal transplantation, indicating their potential participation in the mechanisms of tolerance. We analyzed the serum profile of microRNAs levels in operational tolerance (OT) (n = 8), compared with chronic rejection (CR) (n=5), stable subjects using conventional immunosuppression (STA) (n = 5) and healthy individuals (HI) (n = 5), initially using a panel of primers for 768 miRNAs, by TaqMan Low Density Array (TLDA) technology. We detected a differential profile in the levels of microRNAs between OT and its opposing clinical outcome - CR - suggesting that the microRNAs can integrate the network of mechanisms involved in human transplantation tolerance. Within this differential profile, some microRNAs showed higher levels in OT: miR-885-5p (p = 0.031), miR-331 (p = 0.009), miR-27a (p = 0.033) compared to CR, while others, miR-1233-3p (p = 0.029), miR-572 (p = 0.028), miR-1260a (p=0.017) and miR-638 (p = 0.047) had lower levels in OT. Comparing OT with the physiological state (HI), 2 microRNAs of the differential profile presented decreased levels in OT: miR-27a-5p, (p = 0.033) miR-1260a (p = 0.017) but no differences for the others, indicating the predominance of preservation of the profile in OT, in relation to the physiological state. Using bioinformatics, we predicted signaling pathways and we found that the target genes of these microRNAs were, predominantly, related to cell death, integrating the granzyme and death receptor pathways. Considering the various targets of the microRNAs comprised in the differential profile and their potential life-and-death effects, in these two pathways, we found a life-to-death ratio of 1.95 in OT and 0.39 in CR, indicating the greater potential to promote death in CR, and life in OT, by the action of these microRNAs. These data indicate that the regulation of pathways related to cell death may integrate the mechanisms of immune tolerance in human renal transplantation. We selected miR-885-5p to validate the findings in a larger number of subjects, and confirmed higher levels in OT (n = 8), in relation to CR (n = 12, p = 0.0063) and in relation to HI (n = 12, p = 0.0035). Considering that CASP3 is a relevant target of miR-885, we interpret that its action in OT can promote a decrease in CASP3 expression and, consequently, favor the preservation of cell survival. We conclude that epigenetic mechanisms can integrate the network of mechanisms of transplantation tolerance, such as by the action of microRNAs, regulating the expression of genes involved in cell survival and death
8

Autoimmune Diabetes and Transplantation Tolerance Induced by Costimulation Blockade in NOD Mice: a Dissertation

Lambert, Julie 13 August 2007 (has links)
NOD mice model human type 1 diabetes and have been used to investigate tolerance induction protocols for islet transplantation in a setting of autoimmunity. Costimulation blockade-based tolerance protocols that induce prolonged skin and permanent islet allograft survival in non-autoimmune mice have failed in NOD mice. To investigate the underlying mechanisms, we generated NOD hematopoietic chimeras. We were able to show that dendritic cell maturation defects seen in NOD mice are partially corrected in mixed hematopoietic chimeras. Furthermore, skin allograft survival was dependent upon the phenotype of the bone marrow donor, demonstrating that in the NOD the resistance to tolerance induction resides in the hematopoietic compartment. In addition, we studied congenic NOD mice bearing insulin dependent diabetes (Idd) loci that reduce diabetes incidence. The incidence of diabetes is reduced in NOD.B6 Idd3 mice, and virtually absent in NOD.B6 Idd3Idd5 mice. Islet allograft survival in NOD.B6 Idd3 mice is prolonged as compared to NOD mice, and in NOD.B6 Idd3Idd5 mice islet allograft survival is similar to that achieved in C57BL/6 mice. Alloreactive CD8 T cell depletion in NOD mice treated with costimulation blockade is impaired, but is partially restored in NOD.B6 Idd3 mice, and completely restored in NOD.B6 Idd3Idd5 mice. Idd3 results from variations in Il2 gene transcription. We hypothesized insufficient levels of IL-2 in NOD mice contributes to impaired deletion of alloreactive CD8 T cells and shortened islet allograft survival. We observed using synchimeric mice that co-administration of exogenous IL-2 to NOD mice treated with costimulation blockade led to deletion of alloreactive CD8 T cells comparable to that in C57BL/6 mice and prolonged islet allograft survival. However, some Idd loci impaired the induction of transplantation tolerance. These data suggest that Idd loci can facilitate or impair induction of transplantation tolerance by costimulation blockade, and that Idd3 (IL-2) is critical component in this process.
9

MicroRNAs na tolerância operacional no transplante renal humano / MicroRNAs levels in operational tolerance (OT) in human transplantation tolerance

Amanda Cabral da Silva 04 June 2018 (has links)
A tolerância operacional (TO) é um estado de estabilidade funcional do órgão transplantado, após a parada de drogas imunossupressoras, por um período maior ou igual a um ano. Os microRNAs são pequenas moléculas de RNA não codificantes que regulam, negativamente, a expressão gênica de seus alvos, incluindo de moléculas relacionadas ao sistema imune, desempenhando um papel crítico na manutenção da homeostase. Nosso objetivo foi determinar se há um perfil diferencial de microRNAs na TO no transplante renal humano, indicando sua potencial participação nos mecanismos de tolerância. Analisamos o perfil sérico dos níveis de microRNAs na tolerância operacional (TO) (n= 8), comparando com rejeição crônica (RC) (n= 5), estáveis em uso de imunossupressão convencional (EST) (n= 5) e indivíduos saudáveis (SAU) (n= 5), utilizando, inicialmente, um painel de primers para 768 miRNAs, pela tecnologia TLDA (TaqMan Low Density Array). Detectamos um perfil diferencial nos níveis de microRNAs entre TO e o seu desfecho clínico oposto - RC- sugerindo que microRNAs podem integrar a rede de mecanismos envolvidos na tolerância ao transplante. Nesse perfil diferencial, alguns tiveram níveis maiores na TO: miR-885-5p (p=0,031), miR-331 (p=0,009), miR-27a (p=0,033), em comparação com RC, enquanto outros, o miR-1233-3p (p=0,029), miR-572 (p=0,028), miR-1260a (p=0.017) e o miR-638 (p=0,047) apresentaram menores níveis na TO. Na comparação de TO com o estado fisiológico (SAU), 2 dos microRNAs do perfil diferencial TO x RC apresentaram níveis diminuídos na TO: miR-27a-5p, (p=0,033) miR-1260a (p=0,017) e, para os demais, não encontramos diferenças de níveis, indicando predomínio de preservação do perfil na TO, em relação ao estado fisiológico. Por bioinformática, foram preditas vias de sinalização e observamos que os genes alvos desses microRNAs tiveram, predominantemente, relação com morte celular, integrando as vias de granzima e receptor de morte. Considerando os diversos alvos dos microRNAs do perfil diferencial e seus potenciais efeitos de favorecer vida e morte, nessas duas vias, encontramos uma razão de vida/morte de 1,95 na TO e de 0,39 na RC, indicando o maior potencial de promoção de morte na RC e, de vida, na TO, pela ação desses microRNAs. Esses dados indicam que a regulação de vias relacionadas à morte celular pode integrar os mecanismos de tolerância imunológica no transplante renal humano. Selecionamos o miR-885-5p para validar os achados, em um número maior de amostras, e confirmamos maiores níveis na TO (n=8), em relação à RC (n=12; p=0,0063) e também em relação ao SAU (n=12; p=0,0035). Considerando que CASP3 é um dos alvos do miR-885, interpretamos que a sua ação, na TO, pode promover a menor expressão de CASP3 e, consequentemente, favorecer a sobrevivência celular. Concluímos que mecanismos epigenéticos podem integrar a rede de mecanismos da tolerância ao transplante, como por meio da ação de microRNAs regulando a expressão de genes envolvidos com sobrevida/morte celular / Operational tolerance (OT) is a state of functional stability of the transplanted organ, after stopping immunosuppressive drugs for a period of at least one year. MicroRNAs are small non-coding RNA that downregulate gene expression of their targets, including genes related to the immune system, playing a critical role in keeping homeostasis. Our objective was to determine whether there is a differential profile of microRNAs in OT in human renal transplantation, indicating their potential participation in the mechanisms of tolerance. We analyzed the serum profile of microRNAs levels in operational tolerance (OT) (n = 8), compared with chronic rejection (CR) (n=5), stable subjects using conventional immunosuppression (STA) (n = 5) and healthy individuals (HI) (n = 5), initially using a panel of primers for 768 miRNAs, by TaqMan Low Density Array (TLDA) technology. We detected a differential profile in the levels of microRNAs between OT and its opposing clinical outcome - CR - suggesting that the microRNAs can integrate the network of mechanisms involved in human transplantation tolerance. Within this differential profile, some microRNAs showed higher levels in OT: miR-885-5p (p = 0.031), miR-331 (p = 0.009), miR-27a (p = 0.033) compared to CR, while others, miR-1233-3p (p = 0.029), miR-572 (p = 0.028), miR-1260a (p=0.017) and miR-638 (p = 0.047) had lower levels in OT. Comparing OT with the physiological state (HI), 2 microRNAs of the differential profile presented decreased levels in OT: miR-27a-5p, (p = 0.033) miR-1260a (p = 0.017) but no differences for the others, indicating the predominance of preservation of the profile in OT, in relation to the physiological state. Using bioinformatics, we predicted signaling pathways and we found that the target genes of these microRNAs were, predominantly, related to cell death, integrating the granzyme and death receptor pathways. Considering the various targets of the microRNAs comprised in the differential profile and their potential life-and-death effects, in these two pathways, we found a life-to-death ratio of 1.95 in OT and 0.39 in CR, indicating the greater potential to promote death in CR, and life in OT, by the action of these microRNAs. These data indicate that the regulation of pathways related to cell death may integrate the mechanisms of immune tolerance in human renal transplantation. We selected miR-885-5p to validate the findings in a larger number of subjects, and confirmed higher levels in OT (n = 8), in relation to CR (n = 12, p = 0.0063) and in relation to HI (n = 12, p = 0.0035). Considering that CASP3 is a relevant target of miR-885, we interpret that its action in OT can promote a decrease in CASP3 expression and, consequently, favor the preservation of cell survival. We conclude that epigenetic mechanisms can integrate the network of mechanisms of transplantation tolerance, such as by the action of microRNAs, regulating the expression of genes involved in cell survival and death
10

The Genetic Basis of Resistance to Transplantation Tolerance Induced by Costimulation Blockade in NOD Mice: a Dissertation

Pearson, Todd 17 March 2003 (has links)
The NOD mouse is a widely studied model of type 1 diabetes. The loss of self-tolerance leading to autoimmune diabetes in NOD mice involves at least 27 genetic loci. Curing type I diabetes in mice and humans by islet transplantation requires overcoming both allorejection and recurrent autoimmunity. This has been achieved with systemic immunosuppression, but tolerance induction would be preferable. In addition to their genetic defects in self-tolerance, NOD mice resist peripheral transplantation tolerance induced by costimulation blockade using donor-specific transfusion and anti-CDl54 antibody. Failure has been attributed to the underlying autoimmunity, assuming that autoimmunity and resistance to transplantation tolerance have a common basis. Hypothesizing that these two abnormalities might be related, we investigated whether they had a common genetic basis. Diabetes-resistant NOD and C57BL/6 stocks congenic for various reciprocally introduced Idd loci were assessed for their ability to be tolerized. Surprisingly, in NOD congenic mice that are almost completely protected from diabetes, costimulation blockade failed to prolong skin allograft survival. In reciprocal C57BL/6 congenic mice with NOD-derived Idd loci, skin allograft survival was readily prolonged by costimulation blockade. Unexpectedly, we observed that (NOD x C57BL/6)F1 mice, which have no diabetes, nonetheless resist induction of tolerance to skin allografts. Further analyses revealed that the F1 mice shared the dendritic cell maturation defects and abnormal CD4+ T cell responses of the NOD but had lost its defects in macrophage maturation and NK cell activity. Finally, using a genome wide scan approach, we have identified four suggestive markers in the mouse genome that control the survival of skin allografts following DST and anti-CD154 mAb therapy. We suggest that mechanisms controlling autoimmunity and transplantation tolerance in NOD mice are not completely overlapping and are potentially distinct, or that the genetic threshold for normalizing the transplantation tolerance defect is higher than that for preventing autoimmune diabetes. We conclude that resistance to allograft tolerance induction in the NOD mouse is not a direct consequence of overt autoimmunity and that autoimmunity and resistance to costimulation blockade-induced transplantation tolerance phenotypes in NOD mice are not under identical genetic control.

Page generated in 0.1503 seconds