Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2036 word(s) 2036 2021-11-25 06:54:32 |
2 update layout and reference Meta information modification 2036 2021-12-13 03:16:01 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Sato, N. Induction of Immune Tolerance in Islet Transplantation. Encyclopedia. Available online: https://encyclopedia.pub/entry/17002 (accessed on 27 July 2024).
Sato N. Induction of Immune Tolerance in Islet Transplantation. Encyclopedia. Available at: https://encyclopedia.pub/entry/17002. Accessed July 27, 2024.
Sato, Naoya. "Induction of Immune Tolerance in Islet Transplantation" Encyclopedia, https://encyclopedia.pub/entry/17002 (accessed July 27, 2024).
Sato, N. (2021, December 11). Induction of Immune Tolerance in Islet Transplantation. In Encyclopedia. https://encyclopedia.pub/entry/17002
Sato, Naoya. "Induction of Immune Tolerance in Islet Transplantation." Encyclopedia. Web. 11 December, 2021.
Induction of Immune Tolerance in Islet Transplantation
Edit

Allogeneic islet transplantation has become an effective treatment option for severe Type 1 diabetes with intractable impaired awareness due to hypoglycemic events. Although current immunosuppressive protocols effectively prevent the acute rejection associated with initial T cell activation in recipients, chronic rejection has remained an obstacle for achieving long-term allogeneic islet engraftment. 

islet transplantation tolerance induction

1. Introduction

Allogeneic pancreatic islet transplantation has been established as an effective option for severe Type 1 diabetes with intractable impaired awareness due to hypoglycemic events. Islet transplantation involves the intrahepatic delivery of donor-isolated islet cells that supplement insulin production, promoting the recovery of endogenous insulin secretion. Evidence has shown that the development of established immunosuppression protocol has improved outcomes of allogeneic islet transplantation [1]. However, the Collaborative Islet Transplant Registry data collected from islet transplant centers around the world reported 1- and 3-year insulin-independence rates of 71% and 24%, respectively [2]. A majority of islet transplant recipients return to some form of exogenous insulin usage within a few years of transplantation [3]. One factor associated with the long-term outcomes of transplanted islets is chronic rejection. Although current immunosuppression regimens effectively prevent acute rejection, which can suppress initial T cell priming by the donor antigen [4][5], no established immunosuppressive regimen has been effective in controlling chronic rejection.
When considering the adverse effects of immune suppression, some immunosuppressive drugs can be toxic to islet grafts [6], which could worsen the long-term functioning of the transplanted islets. Moreover, the long-term mediation of immunosuppressive drugs has also been associated with increased risk of infections [7], malignancies [8], cardiovascular disease [9][10], renal failure [11], de novo diabetes [12][13], and neurotoxicity [14]. With the increase in transplantation cases, a growing number of chronically suppressed transplant recipients struggle with such burdens.
The development of donor-specific immune tolerance to an allograft is the ultimate goal of any transplantation given its ability to possibly resolve chronic rejection and disregard the need for maintenance immunosuppression. Inducing donor-specific tolerance in animal transplantation models has been met with several challenges. Nonetheless, the accumulation of information has allowed the recent emergence of several key insights for operational tolerance induction, including the role of regulatory B cells (Bregs) for inducing or maintaining immunological tolerance.
As recently as 2019, Sigh et al. reported on a breakthrough in the tolerance induction protocol for allogeneic islet transplantation in non-human primate (NHP) models [15]. This approach for inducing donor-specific tolerance is unique in that it involves the strategic exposure of the recipient to donor antigens prior to transplantation. Several rodent models of allogeneic or xenogeneic transplantation have been evaluated on the impact of apoptotic donor lymphocyte infusion prior to transplantation on graft survival. The achievement by Sigh et al. represents a considerable step forward in the development of immune tolerance induction for human clinical applications.

2. General Understanding of the Relationship between the Immune System and Immunological Tolerance

The immune system can learn to discriminate between the self and nonself via a complex set of central and peripheral immune tolerance mechanisms. When considering immunological tolerance to the allograft, the high proportion of MHC alloreactive T cells, which generally range from 5 to 10% [16], has been considered as the main hindrance toward tolerance induction. Regarding T cell tolerance, central tolerance refers to the deletion of reactive clones within the thymus during negative selection. Peripheral T cell tolerance encompasses several mechanisms that take place outside the thymus, including peripheral deletion, anergy/exhaustion, and suppressive function of regulatory T cells (Tregs) [17]. The key lies in determining how to apply this mechanism of immunological tolerance, which is inherent in the body system, to induce donor-specific immune tolerance in transplantation.
During the immune destruction of the islet graft, the initial process of rejection is characterized by a rapid infiltration of innate immune cells to the grafts, which can be followed by an antigen-specific T cell response. The established immunosuppression protocol incorporating T cell depletion or anti-TNFa monoclonal antibodies could achieve T cell activation in allogeneic islet transplantation, which contributes to enhancing short-term graft survival [1]. However, the current protocol has been considered insufficient for controlling the humoral immune response, including antibody-mediated rejection, which is a significant mechanism of chronic allograft failure [18]. Indeed, B cells are known to mainly contribute to humoral immunity and boost cellular immunity. However, several experimental models have shown that B cell subsets ameliorate inflammation and autoimmunity disease, suggesting their capability for regulatory function, namely Bregs. Numerous bodies of evidence have indicated that B cells play essential roles in alloimmunity, including differentiation into antibody-producing plasma cells, sustaining long-term humoral immune memory, serving antigen-presenting cells (APCs), organizing the formation of tertiary lymphoid organs, and secreting pro- and anti-inflammatory cytokines (IL-10) [19]. Thus, therapeutic options targeting T cell–B cell interactions are of interest in the development of immunosuppressive protocols for transplantation [19]. Although a detailed description of B cell function in transplantation immunity is beyond the scope of the current paper, the following section summarizes exciting evidence obtained from islet transplantation animal models, demonstrating the significance of B cell functions in inducing and sustaining immunological tolerance.

3. Operative Tolerance Induction Using Low-Affinity TIM-1 mAb in an Islet Transplantation Model

TIM-1, which was initially reported to be a T cell costimulatory marker, is a member of the T cell immunoglobulin and mucin domain family of costimulatory molecules. However, an in vivo mice study by Ding et al. showed that TIM-1 was constitutively expressed on B cells rather than T cells and that 6–40% of TIM-1+ B cells express IL-10, including transitional, marginal zone, and follicular B cells [20]. Thus, reports have shown that TIM-1 is an inclusive marker of IL-10 + B cells. A mouse model of islet transplantation found that treatment with low-affinity anti-TIM-1, which has functional properties in the Breg development of recipients, leads to significantly prolonged islet allograft survival (approximately 30% of mice achieved long-term engraftment over 100 days) [20]. Interestingly, treating B cell-depleted recipients with anti-TIM-1 significantly enhanced IFN-g and ultimately prevented the commonly observed increase in Th2 cytokines. Therefore, B cells are required for anti-TIM-1-induced Th2 cytokine expression.
Moreover, a mouse islet allograft model found that anti-CD45RB antibody in combination with anti-TIM-1 antibody had a synergistic effect in inducing tolerance in all recipients. The dual antibody treatment significantly expanded regulatory B and T cells depending on the presence of recipient B cells with IL-10 activity [21]. Using B cell-deficient recipients or depleting B cells with anti-CD20 antibody abrogates the anti-CD45RB-induced tolerance following anti-TIM-1 dual antibody treatment. After exploring the reason why B cell depletion prevented the effects of dual antibody treatment on graft survival, the aforementioned study demonstrated that CD19 + CD5 + CD1d + B10 cells might play an important role only in early-stage transplantation tolerance induction following treatment. Additionally, the study also concluded that CD19 + TIM + B cells might play crucial roles in the whole process of tolerance induction and maintenance. These findings may explain why B cell depletion inhibited the effects of dual antibody treatment.

4. Important Evidence for Inducing Donor-Specific Tolerance to Preclinical Implementation

Although several promising approaches for tolerance induction in a rodent model of transplantation have been reported, as discussed earlier, very few have been translated to human or NHP transplantation models. Unlike laboratory mice, NHPs and humans already have a large collection of memory T and B cells at the time of transplantation. Heterologous immunity or the cross-reactive immune response has the potential to be alloreactive. For instance, up to 45% of anti-CMV T cell clones have been reported to be alloreactive [22]. Existing cross-reactive memory immune cells have been a significant hindrance to immunological tolerance in large animals or humans. Thus, tolerance induction in NHPs or humans would be more challenging than that in rodent models, partly due to cross-reactive memory T or B cells. Nevertheless, a few encouraging approaches have led us to believe that immunological tolerance can eventually be achieved in humans, such as mixed chimerism using hematopoietic cell transplantation [23][24] or ADL exposure [15].
One of the most advanced approaches for sustained tolerance is the use of hematopoietic cell transplantation to achieve durable chimerism, where the donor and recipient hematopoietic cells coexist at levels detectable by flow cytometry [24][25][26]. The central feature of the mixed chimerism-based transplant tolerance involves the intrathymic deletion of donor-reactive T cells by irradiation and/or anti-thymocyte globulin [27]. One study on kidney transplantation in NHPs reported that this protocol successfully promoted operationally sustained mixed chimerism in conditioning recipient and long-term engraftment [23]. Additionally, this approach has been successfully translated to both human leukocyte antigen-matched and -mismatched human renal transplant recipients with immunosuppression-free graft survival [24][28]. These results encourage attempts at extending this regimen to islet transplantation. Oura et al. reported the results of the mixed chimerism-based tolerance induction in islet transplantation in an NHP model, in which recipients were treated with a nonmyeloablative condition regimen that included total body irradiation, horse anti-thymocyte globulin anti-CD154 monoclonal antibody, and cyclosporine or anti-CD8 antibody (calcineurin inhibitor-free regimen) [29]. Accordingly, transient chimerism did not induce tolerance of islet graft survival, with islet function having been lost soon after the disappearance of chimerism [29]. Similar to islet transplantation, the induction of transient chimerism did not promote the tolerance of a heart allograft [30].
Table 1 summarizes the available evidence regarding the effectiveness of ADL in inducing immunological tolerance in islet transplantation. Studies using rodent islet transplantation models have demonstrated the efficacy of the peritransplant infusion of ECDI-fixed donor splenocytes in not only allogeneic islet transplantation [15][31] but also xenogeneic islet transplantation [32][33]. Focusing on evidence that promotes clinical implementation, recent data regarding the NHP model reported by Singh et al. can be considered a breakthrough achievement [15]. Their protocol involves peritransplant infusions of MHC-DRB allele-matched apoptotic donor leukocytes under short-term immune suppressions, including antagonistic anti-CD40 antibody 2C10R4, rapamycin, soluble tumor necrosis factor receptor, and anti-interleukin 6 receptor antibody.
Table 1. Summary of previous studies on tolerance induction using ECDI-donor splenocytes in islet transplantation animal experiments.
Summary of Previous Studies on Tolerance Induction Using ECDI-Donor Splenocytes in Islet Transplantation Animal Experiments
Year Authors Tx Model Induction Treatments Tx Outcome Mechanisms
2008 Luo et al. mouse-to-mouse
(allogeneicTx model)
ECDI-SPs 64% graft survival (>100 days) Depletion of alloantigen-specific T cells
CD4+CD25+ Tregs are required for tolerance induction by infusion of ECDI-treated donor splenocytes.
PD-1/PD-L1 signaling pathway is associated with donor-specific tolerance induction by ECDI-fixed SPs
2013 Wang et al. rat-to-mouse
(xenogeneic Tx model)
ECDI-SPs MST 48 days (18days for non-treated) Anti-donor antibody: rat ECDI-SPs induced anti-rat IgGs (High levels of anti-rat IgG were detectable by day 14)
C4d deposition: observed 14 days and 28 days in rat islet xenograft from recipients treated with rat ECDI-SPs.
B cell activation: upregulated expression of costimulatory molecules CD80, CD86, CD40, and OX40L
B cell infiltration: observed in ECDI-SPs recipients 2 or 4 weeks after Tx.
ECDI-SPs with B cell depletion 100% graft survival (>100 days) Anti-donor antibody: no production of anti-rat antibodies of all IgG subclasses at 14 days after ECDI-SPs
C4d deposition: negative
B cell activation: N.A
B cell infiltration: minimal infiltration of B220 cells
xeno-specific T-cell priming: suppressed
memory T cell generation: suppressed
rebound B cells: xeno-donor-specific B cell unresponsiveness
2017 Kang et al. pig-to-mouse
(xenogeneic Tx model)
no treatment acute rejection (by day 7–26 post-transplantation) B cell infiltration to grafts; prominent (cf. minimal infiltration of B cells to graft in alloislet Tx)
High expression of IL-17 on CD4 and CD8T cell from rejected mice (cf. high levels of IFN-r on T cell from rejected mice in alloislet Tx.)
ECDI-SPs only no prolongation of graft survival N.A
ECDI-SPs with B cell depletion prolongation of graft survival(40% graft >100 days) N.A

ECDI-SPs with B cell depletion and transient rapamycin
1. prolongation of graft survival(65% graft >100 days)
2. late rejection between day 100 and 200 post-transplantation (B cell reconstitution)
initial phase(day 21–70 post transplantation)
anti-pig IL-17 response: suppressed
rejection phase
B cell infiltration to graft: aggressive infiltration of B cells to graft
anti-pig antibody production: minimal
anti-pig INFr response; observed in indirect donor stimulation, but not direct donor stimulation
2019 A.Sigh HNP-to-NHP
(MHC lassⅡ matched)
(allogeneic Tx model)
ECDI-SPs and transient ISs
(anti-CD40, anti-IL-6R, anti-TNFaR, rapamycin)
long-term tolerance (100%) antigen-specific regulatory networks: Tr1, Breg, B10, MDSC
One DRB-matched ECDI-SPs; expanded alloantigen-specific regulation
2020 Dangi et al. mouse(B6)-to-mouse(Balb/c)
(allogeneic islet transplantation)
ECDI-SPs only no prolongation of graft survival  
ECDI-SPs and transient ISs
(anti-CD40 and rapamycin)
MST 35 days donor-specific graft-infiltrating T cells; inhibited
expansion of donor-specific memory B cells; inhibited
infiltrating B cells in late rejected islets with high expression of CD40 and CD86
ECDI-SPs and transient ISs (anti-CD40 and rapamycin)B cell depletion islet survival of >180 days in ~80% of recipients

References

  1. Bellin, M.D.; Barton, F.B.; Heitman, A.; Harmon, J.V.; Kandaswamy, R.; Balamurugan, A.N.; Sutherland, D.E.; Alejandro, R.; Hering, B.J. Potent induction immunotherapy promotes long-term insulin independence after islet transplantation in type 1 diabetes. Am. J. Transpl. Off. J. Am. Soc. Transpl. Surg. 2012, 12, 1576–1583.
  2. Alejandro, R.; Barton, F.B.; Hering, B.J.; Wease, S. Collaborative islet transplant registry, I. 2008 update from the collaborative islet transplant registry. Transplantation. 2008, 86, 1783–1788.
  3. Shapiro, A.M.; Ricordi, C.; Hering, B.J.; Auchincloss, H.; Lindblad, R.; Robertson, R.P.; Secchi, A.; Brendel, M.D.; Berney, T.; Brennan, D.C. International trial of the Edmonton protocol for islet transplantation. New Engl. J. Med. 2006, 355, 1318–1330.
  4. Chi, H. Regulation and function of mTOR signalling in T cell fate decisions. Nat. Rev. Immunol. 2012, 12, 325–338.
  5. Lewis, R.S. Calcium signaling mechanisms in T lymphocytes. Annu. Rev. Immunol 2001, 19, 497–521.
  6. Vallabhajosyula, P.; Hirakata, A.; Shimizu, A.; Okumi, M.; Tchipashvili, V.; Hong, H.; Yamada, K.; Sachs, D.H. Assessing the effect of immunosuppression on engraftment of pancreatic islets. Transplantation 2013, 96, 372–378.
  7. van Doesum, W.B.; Gard, L.; Bemelman, F.J.; de Fijter, J.W.; Homan van der Heide, J.J.; Niesters, H.G.; van Son, W.J.; Stegeman, C.A.; Groen, H.; Riezebos-Brilman, A.; et al. Incidence and outcome of BK polyomavirus infection in a multicenter randomized controlled trial with renal transplant patients receiving cyclosporine-, mycophenolate sodium-, or everolimus-based low-dose immunosuppressive therapy. Transpl. Infect. Dis. 2017, 19, e12687.
  8. Domhan, S.; Zeier, M.; Abdollahi, A. Immunosuppressive therapy and post-transplant malignancy. Nephrol. Dial. Transpl. 2009, 24, 1097–1103.
  9. Miller, L.W. Cardiovascular toxicities of immunosuppressive agents. Am. J. Transpl. Off. J. Am. Soc. Transpl. Surg. 2002, 2, 807–818.
  10. Lentine, K.L.; Brennan, D.C.; Schnitzler, M.A. Incidence and predictors of myocardial infarction after kidney transplantation. J. Am. Soc. Nephrol. 2005, 16, 496–506.
  11. Paramesh, A.S.; Roayaie, S.; Doan, Y.; Schwartz, M.E.; Emre, S.; Fishbein, T.; Florman, S.; Gondolesi, G.E.; Krieger, N.; Ames, S.; et al. Post-liver transplant acute renal failure: Factors predicting development of end-stage renal disease. Clin. Transpl. 2004, 18, 94–99.
  12. Rodriguez-Rodriguez, A.E.; Porrini, E.; Hornum, M.; Donate-Correa, J.; Morales-Febles, R.; Khemlani Ramchand, S.; Molina Lima, M.X.; Torres, A. Post-transplant diabetes mellitus and prediabetes in renal transplant recipients: An update. Nephron 2021, 145, 317–329.
  13. Mizrahi, N.; Braun, M.; Ben Gal, T.; Rosengarten, D.; Kramer, M.R.; Grossman, A. Post-transplant diabetes mellitus: Incidence, predicting factors and outcomes. Endocrine 2020, 69, 303–309.
  14. Wijdicks, E.F. Neurotoxicity of immunosuppressive drugs. Liver Transpl. 2001, 7, 937–942.
  15. Singh, A.; Ramachandran, S.; Graham, M.L.; Daneshmandi, S.; Heller, D.; Suarez-Pinzon, W.L.; Balamurugan, A.N.; Ansite, J.D.; Wilhelm, J.J.; Yang, A.; et al. Long-term tolerance of islet allografts in nonhuman primates induced by apoptotic donor leukocytes. Nat. Commun. 2019, 10, 3495.
  16. Suchin, E.J.; Langmuir, P.B.; Palmer, E.; Sayegh, M.H.; Wells, A.D.; Turka, L.A. Quantifying the frequency of alloreactive T cells in vivo: New answers to an old question. J. Immunol. 2001, 166, 973–981.
  17. Anderton, S.; Burkhart, C.; Metzler, B.; Wraith, D. Mechanisms of central and peripheral T-cell tolerance: Lessons from experimental models of multiple sclerosis. Immunol. Rev. 1999, 169, 123–137.
  18. Lodhi, S.A.; Lamb, K.E.; Meier-Kriesche, H.U. Solid organ allograft survival improvement in the United States: The long-term does not mirror the dramatic short-term success. Am. J. Transpl. Off. J. Am. Soc. Transpl. Surg. 2011, 11, 1226–1235.
  19. Karahan, G.E.; Claas, F.H.; Heidt, S. B Cell immunity in solid organ transplantation. Front. Immunol. 2016, 7, 686.
  20. Ding, Q.; Yeung, M.; Camirand, G.; Zeng, Q.; Akiba, H.; Yagita, H.; Chalasani, G.; Sayegh, M.H.; Najafian, N.; Rothstein, D.M. Regulatory B cells are identified by expression of TIM-1 and can be induced through TIM-1 ligation to promote tolerance in mice. J. Clin. Investig. 2011, 121, 3645–3656.
  21. Lee, K.M.; Kim, J.I.; Stott, R.; Soohoo, J.; O’Connor, M.R.; Yeh, H.; Zhao, G.; Eliades, P.; Fox, C.; Cheng, N.; et al. Anti-CD45RB/anti-TIM-1-induced tolerance requires regulatory B cells. Am. J. Transpl. Off. J. Am. Soc. Transpl. Surg. 2012, 12, 2072–2078.
  22. D’Orsogna, L.J.; van der Meer-Prins, E.M.; Zoet, Y.M.; Roelen, D.L.; Doxiadis, I.I.; Claas, F.H. Detection of allo-HLA cross-reactivity by virus-specific memory T-cell clones using single HLA-transfected K562 cells. Methods Mol. Biol. 2012, 882, 339–349.
  23. Kawai, T.; Sogawa, H.; Boskovic, S.; Abrahamian, G.; Smith, R.N.; Wee, S.L.; Andrews, D.; Nadazdin, O.; Koyama, I.; Sykes, M.; et al. CD154 blockade for induction of mixed chimerism and prolonged renal allograft survival in nonhuman primates. Am. J. Transpl. Off. J. Am. Soc. Transpl. Surg. 2004, 4, 1391–1398.
  24. Kawai, T.; Cosimi, A.B.; Spitzer, T.R.; Tolkoff-Rubin, N.; Suthanthiran, M.; Saidman, S.L.; Shaffer, J.; Preffer, F.I.; Ding, R.; Sharma, V.; et al. HLA-mismatched renal transplantation without maintenance immunosuppression. New Engl. J. Med. 2008, 358, 353–361.
  25. Scandling, J.D.; Busque, S.; Dejbakhsh-Jones, S.; Benike, C.; Millan, M.T.; Shizuru, J.A.; Hoppe, R.T.; Lowsky, R.; Engleman, E.G.; Strober, S. Tolerance and chimerism after renal and hematopoietic-cell transplantation. N. Engl. J. Med. 2008, 358, 362–368.
  26. Leventhal, J.; Abecassis, M.; Miller, J.; Gallon, L.; Ravindra, K.; Tollerud, D.J.; King, B.; Elliott, M.J.; Herzig, G.; Herzig, R.; et al. Chimerism and tolerance without GVHD or engraftment syndrome in HLA-mismatched combined kidney and hematopoietic stem cell transplantation. Sci. Transl. Med. 2012, 4, 124ra28.
  27. Tomita, Y.; Khan, A.; Sykes, M. Role of intrathymic clonal deletion and peripheral anergy in transplantation tolerance induced by bone marrow transplantation in mice conditioned with a nonmyeloablative regimen. J. Immunol. 1994, 153, 1087–1098.
  28. Kawai, T.; Sachs, D.H.; Sprangers, B.; Spitzer, T.R.; Saidman, S.L.; Zorn, E.; Tolkoff-Rubin, N.; Preffer, F.; Crisalli, K.; Gao, B.; et al. Long-term results in recipients of combined HLA-mismatched kidney and bone marrow transplantation without maintenance immunosuppression. Am. J. Transpl. Off. J. Am. Soc. Transpl. Surg. 2014, 14, 1599–1611.
  29. Oura, T.; Ko, D.S.; Boskovic, S.; O’Neil, J.J.; Chipashvili, V.; Koulmanda, M.; Hotta, K.; Kawai, K.; Nadazdin, O.; Smith, R.N.; et al. Kidney Versus Islet Allograft Survival After Induction of Mixed Chimerism With Combined Donor Bone Marrow Transplantation. Cell Transpl. 2016, 25, 1331–1341.
  30. Kawai, T.; Cosimi, A.B.; Wee, S.L.; Houser, S.; Andrews, D.; Sogawa, H.; Phelan, J.; Boskovic, S.; Nadazdin, O.; Abrahamian, G.; et al. Effect of mixed hematopoietic chimerism on cardiac allograft survival in cynomolgus monkeys. Transplantation 2002, 73, 1757–1764.
  31. Luo, X.; Pothoven, K.L.; McCarthy, D.; DeGutes, M.; Martin, A.; Getts, D.R.; Xia, G.; He, J.; Zhang, X.; Kaufman, D.B.; et al. ECDI-fixed allogeneic splenocytes induce donor-specific tolerance for long-term survival of islet transplants via two distinct mechanisms. Proc. Natl. Acad. Sci. USA 2008, 105, 14527–14532.
  32. Kang, H.K.; Wang, S.; Dangi, A.; Zhang, X.; Singh, A.; Zhang, L.; Rosati, J.M.; Suarez-Pinzon, W.; Deng, X.; Chen, X.; et al. Differential Role of B Cells and IL-17 Versus IFN-gamma During Early and Late Rejection of Pig Islet Xenografts in Mice. Transplantation 2017, 101, 1801–1810.
  33. Wang, S.; Tasch, J.; Kheradmand, T.; Ulaszek, J.; Ely, S.; Zhang, X.; Hering, B.J.; Miller, S.D.; Luo, X. Transient B-cell depletion combined with apoptotic donor splenocytes induces xeno-specific T- and B-cell tolerance to islet xenografts. Diabetes 2013, 62, 3143–3150.
More
Information
Subjects: Allergy
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 326
Revisions: 2 times (View History)
Update Date: 13 Dec 2021
1000/1000
Video Production Service