Skip to main content
Erschienen in: Acta Diabetologica 5/2019

13.02.2019 | Original Article

Combination therapy of an iNKT cell ligand and CD40–CD154 blockade establishes islet allograft acceptance in nonmyeloablative bone marrow transplant recipients

verfasst von: Taichi Kanzawa, Toshihito Hirai, Hironori Fukuda, Haruki Katsumata, Rumi Ishii, Masako Ikemiyagi, Yasuyuki Ishii, Kan Saiga, Masayoshi Okumi, Kazunari Tanabe

Erschienen in: Acta Diabetologica | Ausgabe 5/2019

Einloggen, um Zugang zu erhalten

Abstract

Aims

Islet transplantation is an effective therapeutic option for type 1 diabetes. Although maintenance immunosuppression therapy is required to prevent allogeneic rejection and recurrence of autoimmunity, long-term allograft survival has not yet been achieved partly because of its adverse effects. The induction of donor-specific immunotolerance is a promising approach for long-term allograft survival without maintenance immunosuppression therapy. We previously reported that combination therapy using a liposomal ligand for invariant natural killer T cells, RGI-2001, and anti-CD154 antibody established mixed hematopoietic chimerism for the induction of donor-specific immunotolerance. This study investigated whether the protocol could promote islet allograft acceptance in experimental diabetes.

Methods

Streptozotocin-induced diabetic BALB/c mice were transplanted with bone marrow cells from C57BL/6 donors and received combination therapy of RGI-2001 and anti-CD154 antibody after 3-Gy total body irradiation. 3 Weeks after bone marrow transplantation, islets isolated from C57BL/6 donors were transplanted under the kidney capsule.

Results

Mixed chimerism was established in diabetic mice receiving the tolerance induction protocol. After islet transplantation, blood glucose levels improved and normoglycemia persisted for over 100 days. Hyperglycemia recurred after islet grafts were removed. Histopathological examinations showed insulin-positive staining and absence of cellular infiltration in the islet grafts. T cells of recipients showed donor-specific hyporesponsiveness, and anti-donor antibodies were not detected.

Conclusions

The tolerance induction protocol with combination therapy of RGI-2001 and anti-CD154 antibody promoted islet allograft acceptance in a mouse diabetic model. This protocol may be clinically applied to islet transplantation for type 1 diabetes mellitus.
Literatur
2.
Zurück zum Zitat Robertson RP (2015) Islet transplantation for type 1 diabetes what have we learned from alloislet and autoislet successes? Diabetes Care 38:1030–1035CrossRef Robertson RP (2015) Islet transplantation for type 1 diabetes what have we learned from alloislet and autoislet successes? Diabetes Care 38:1030–1035CrossRef
3.
Zurück zum Zitat McCall M, Shapiro AM (2012) Update on islet transplantation. Cold Spring Harb Perspect Med 2012 2:a007823 McCall M, Shapiro AM (2012) Update on islet transplantation. Cold Spring Harb Perspect Med 2012 2:a007823
4.
Zurück zum Zitat Barton FB, Rickels MR, Alejandro R et al (2012) Improvement in outcomes of clinical islet transplantation: 1999–2010. Diabetes Care 35:1436–1445CrossRef Barton FB, Rickels MR, Alejandro R et al (2012) Improvement in outcomes of clinical islet transplantation: 1999–2010. Diabetes Care 35:1436–1445CrossRef
5.
Zurück zum Zitat Brooks AM, Walker N, Aldibbiat A et al (2013) Attainment of metabolic goals in the integrated UK islet transplant program with locally isolated and transported preparations. Am J Transpl 13:3236–3243CrossRef Brooks AM, Walker N, Aldibbiat A et al (2013) Attainment of metabolic goals in the integrated UK islet transplant program with locally isolated and transported preparations. Am J Transpl 13:3236–3243CrossRef
6.
Zurück zum Zitat Bensellam M, Laybutt DR, Jonas JC (2012) The molecular mechanisms of pancreatic β-cell glucotoxicity: recent findings and future research directions. Mol Cell Endocrinol 364:1–27CrossRef Bensellam M, Laybutt DR, Jonas JC (2012) The molecular mechanisms of pancreatic β-cell glucotoxicity: recent findings and future research directions. Mol Cell Endocrinol 364:1–27CrossRef
7.
Zurück zum Zitat Talchai C, Lin HV, Kitamura T, Accili D (2009) Genetic and biochemical pathways of β-cell failure in type 2 diabetes. Diabetes Obes Metab 11:38–45CrossRef Talchai C, Lin HV, Kitamura T, Accili D (2009) Genetic and biochemical pathways of β-cell failure in type 2 diabetes. Diabetes Obes Metab 11:38–45CrossRef
8.
Zurück zum Zitat Wallace M, Whelan H, Brennan L (2013) Metabolomic analysis of pancreatic beta cells following exposure to high glucose. Biochim Biophys Acta 1830:2583–2590CrossRef Wallace M, Whelan H, Brennan L (2013) Metabolomic analysis of pancreatic beta cells following exposure to high glucose. Biochim Biophys Acta 1830:2583–2590CrossRef
9.
Zurück zum Zitat Jonas JC, Bensellam M, Duprez J, Elouil H, Guiot Y, Pascal SM (2009) Glucose regulation of islet stress responses and β cell failure in type 2 diabetes. Diabetes Obes Metab 11:65–81CrossRef Jonas JC, Bensellam M, Duprez J, Elouil H, Guiot Y, Pascal SM (2009) Glucose regulation of islet stress responses and β cell failure in type 2 diabetes. Diabetes Obes Metab 11:65–81CrossRef
10.
Zurück zum Zitat Kikawa K, Sakano D, Shiraki N et al (2014) Beneficial effect of insulin treatment on islet transplantation outcomes in Akita mice. PLoS One 9(4):e95451CrossRef Kikawa K, Sakano D, Shiraki N et al (2014) Beneficial effect of insulin treatment on islet transplantation outcomes in Akita mice. PLoS One 9(4):e95451CrossRef
11.
Zurück zum Zitat Sweet IR, Gilbert M, Scott S et al (2008) Glucose-stimulated increment in oxygen consumption rate as a standardized test of human islet quality. Am J Transpl 8:183–192 Sweet IR, Gilbert M, Scott S et al (2008) Glucose-stimulated increment in oxygen consumption rate as a standardized test of human islet quality. Am J Transpl 8:183–192
12.
Zurück zum Zitat Matschinsky FM (1996) Banting Lecture 1995. A lesson in metabolic regulation inspired by the glucokinase glucose sensor paradigm. Diabetes 45:223–241CrossRef Matschinsky FM (1996) Banting Lecture 1995. A lesson in metabolic regulation inspired by the glucokinase glucose sensor paradigm. Diabetes 45:223–241CrossRef
13.
Zurück zum Zitat Mathew JM, Voss JH, McEwen ST et al (2018) Generation and characterization of alloantigen-specific regulatory T cells for clinical transplant tolerance. Sci Rep 8:1136CrossRef Mathew JM, Voss JH, McEwen ST et al (2018) Generation and characterization of alloantigen-specific regulatory T cells for clinical transplant tolerance. Sci Rep 8:1136CrossRef
14.
Zurück zum Zitat Shapiro AM (2013) Immune antibody monitoring predicts outcome in islet transplantation. Diabetes 62:1377–1378CrossRef Shapiro AM (2013) Immune antibody monitoring predicts outcome in islet transplantation. Diabetes 62:1377–1378CrossRef
15.
Zurück zum Zitat Jianming T, Shunliang Y, Jinquan C et al (2008) Simultaneous islet-kidney transplantation in seven patients of Type 1 diabetes with end-stage renal disease using a glucocorticoid-free immunosuppressive regimen with alemtuzumab induction. Diabetes 57:2666–2671CrossRef Jianming T, Shunliang Y, Jinquan C et al (2008) Simultaneous islet-kidney transplantation in seven patients of Type 1 diabetes with end-stage renal disease using a glucocorticoid-free immunosuppressive regimen with alemtuzumab induction. Diabetes 57:2666–2671CrossRef
16.
Zurück zum Zitat Danielle JB, Marc W, Carmen W et al (2014) Mesenchymal stromal cells improve transplanted islet survival and islet function in a syngenic mouse model. Diabetologia 57:522–531CrossRef Danielle JB, Marc W, Carmen W et al (2014) Mesenchymal stromal cells improve transplanted islet survival and islet function in a syngenic mouse model. Diabetologia 57:522–531CrossRef
17.
Zurück zum Zitat Hirai T, Ishii Y, Ikemiyagi M et al (2014) A novel approach inducing transplant tolerance by activated invariant natural killer T cells with costimulatory blockade. Am J Transpl 14:554–567CrossRef Hirai T, Ishii Y, Ikemiyagi M et al (2014) A novel approach inducing transplant tolerance by activated invariant natural killer T cells with costimulatory blockade. Am J Transpl 14:554–567CrossRef
18.
Zurück zum Zitat Hirai T, Ishii R, Miyairi S et al (2016) Clonal deletion established via invariant NKT cell activation and costimulatory blockade requires in vivo expansion of regulatory T cells. Am J Transpl 16:426–439CrossRef Hirai T, Ishii R, Miyairi S et al (2016) Clonal deletion established via invariant NKT cell activation and costimulatory blockade requires in vivo expansion of regulatory T cells. Am J Transpl 16:426–439CrossRef
19.
Zurück zum Zitat Miyairi S, Hirai T, Ishii R et al (2017) Donor bone marrow cells are essential for iNKT cell-mediated Foxp3+ Treg cell expansion in a murine model of transplantation tolerance. Eur J Immunol 47:734–742CrossRef Miyairi S, Hirai T, Ishii R et al (2017) Donor bone marrow cells are essential for iNKT cell-mediated Foxp3+ Treg cell expansion in a murine model of transplantation tolerance. Eur J Immunol 47:734–742CrossRef
20.
Zurück zum Zitat Ishii R, Hirai T, Miyairi S et al (2018) iNKT cell activation plus T-cell transfer establishes complete chimerism in a murine sublethal bone marrow transplant model. Am J Transpl 18:328–340CrossRef Ishii R, Hirai T, Miyairi S et al (2018) iNKT cell activation plus T-cell transfer establishes complete chimerism in a murine sublethal bone marrow transplant model. Am J Transpl 18:328–340CrossRef
21.
Zurück zum Zitat Chen YB, Efebera YA, Johnston L et al (2017) Increased Foxp3(+)Helios(+) regulatory T cells and decreased acute graft-versus-host disease after allogeneic bone marrow transplantation in patients receiving sirolimus and RGI-2001, an activator of invariant natural killer T cells. Biol Blood Marrow Transpl 23:625–634CrossRef Chen YB, Efebera YA, Johnston L et al (2017) Increased Foxp3(+)Helios(+) regulatory T cells and decreased acute graft-versus-host disease after allogeneic bone marrow transplantation in patients receiving sirolimus and RGI-2001, an activator of invariant natural killer T cells. Biol Blood Marrow Transpl 23:625–634CrossRef
22.
Zurück zum Zitat Duramad O, Laysang A, Li J, Ishii Y, Namikawa R (2011) Pharmacologic expansion of donor-derived, naturally occurring CD4(+)Foxp3(+) regulatory T cells reduces acute graft-versus-host disease lethality without abrogating the graft-versus-leukemia effect in murine models. Biol Blood Marrow Transpl 17:1154–1168CrossRef Duramad O, Laysang A, Li J, Ishii Y, Namikawa R (2011) Pharmacologic expansion of donor-derived, naturally occurring CD4(+)Foxp3(+) regulatory T cells reduces acute graft-versus-host disease lethality without abrogating the graft-versus-leukemia effect in murine models. Biol Blood Marrow Transpl 17:1154–1168CrossRef
23.
Zurück zum Zitat Lorenzo P, Matthew JE, Paola M et al (2013) Alloantibody and autoantibody monitoring predicts islet transplantation outcome in human type 1 diabetes. Diabetes 62:1656–1664CrossRef Lorenzo P, Matthew JE, Paola M et al (2013) Alloantibody and autoantibody monitoring predicts islet transplantation outcome in human type 1 diabetes. Diabetes 62:1656–1664CrossRef
24.
Zurück zum Zitat Brooks AMS, Carter V, Liew A et al (2015) De novo donor-specific HLA antibodies are associated with rapid loss of graft function following islet transplantation in type 1 diabetes. Am J Transpl 15:3239–3246CrossRef Brooks AMS, Carter V, Liew A et al (2015) De novo donor-specific HLA antibodies are associated with rapid loss of graft function following islet transplantation in type 1 diabetes. Am J Transpl 15:3239–3246CrossRef
25.
Zurück zum Zitat Li H, Inveradi L, Molano RD, Pileggi A, Ricordi C (2003) Nonlethal conditioning for the induction of allogeneic chimerism and tolerance to islet allografts. Transplantation 75:966–970CrossRef Li H, Inveradi L, Molano RD, Pileggi A, Ricordi C (2003) Nonlethal conditioning for the induction of allogeneic chimerism and tolerance to islet allografts. Transplantation 75:966–970CrossRef
26.
Zurück zum Zitat Luo B, Nanji SA, Schur CD, Pawlick RL, Anderson CC, Shapiro AM (2005) Robust tolerance to fully allogeneic islet transplants achieved by chimerism with minimal conditioning. Transplantation 80:370–377CrossRef Luo B, Nanji SA, Schur CD, Pawlick RL, Anderson CC, Shapiro AM (2005) Robust tolerance to fully allogeneic islet transplants achieved by chimerism with minimal conditioning. Transplantation 80:370–377CrossRef
27.
Zurück zum Zitat Liang Y, Huang T, Zhang C et al (2005) Donor CD8+ T cells facilitate induction of chimerism and tolerance without GVHD in autoimmune NOD mice conditioned with anti-CD3 mAb. Blood 105:2180–2188CrossRef Liang Y, Huang T, Zhang C et al (2005) Donor CD8+ T cells facilitate induction of chimerism and tolerance without GVHD in autoimmune NOD mice conditioned with anti-CD3 mAb. Blood 105:2180–2188CrossRef
28.
Zurück zum Zitat Racine J, Wang M, Zhang C et al (2011) Induction of mixed chimerism with MHC-mismatched but not matched bone marrow transplants results in thymic deletion of host-type autoreactive T-cells in NOD mice. Diabetes 60:555–564CrossRef Racine J, Wang M, Zhang C et al (2011) Induction of mixed chimerism with MHC-mismatched but not matched bone marrow transplants results in thymic deletion of host-type autoreactive T-cells in NOD mice. Diabetes 60:555–564CrossRef
29.
Zurück zum Zitat Wang M, Racine J, Zhang M et al (2014) MHC-mismatched chimerism is required for induction of transplantation tolerance in autoimmune nonobese diabetic recipients. J Immunol 193:2005–2015CrossRef Wang M, Racine J, Zhang M et al (2014) MHC-mismatched chimerism is required for induction of transplantation tolerance in autoimmune nonobese diabetic recipients. J Immunol 193:2005–2015CrossRef
30.
Zurück zum Zitat William FNC, Razavy H, Luo B et al (2008) Development of either split tolerance or robust tolerance along with humoral tolerance to donor and third-party alloantigens in nonmyeloablative mixed chimeras. J Immunol 180:5177–5186CrossRef William FNC, Razavy H, Luo B et al (2008) Development of either split tolerance or robust tolerance along with humoral tolerance to donor and third-party alloantigens in nonmyeloablative mixed chimeras. J Immunol 180:5177–5186CrossRef
31.
Zurück zum Zitat Al-Adra DP, Pawlick R, Shapiro AMJ, Anderson CC (2012) Targeting cells causing split tolerance allows fully allogeneic islet survival with minimal conditioning in NOD mixed chimeras. Am J Transpl 12:3235–3245CrossRef Al-Adra DP, Pawlick R, Shapiro AMJ, Anderson CC (2012) Targeting cells causing split tolerance allows fully allogeneic islet survival with minimal conditioning in NOD mixed chimeras. Am J Transpl 12:3235–3245CrossRef
32.
Zurück zum Zitat Beilhak GF, Scheffold YC, Weissman IL et al (2003) Purified allogeneic hematopoietic stem cell transplantation blocks diabetes pathogenesis in NOD mice. Diabetes 52:59–68CrossRef Beilhak GF, Scheffold YC, Weissman IL et al (2003) Purified allogeneic hematopoietic stem cell transplantation blocks diabetes pathogenesis in NOD mice. Diabetes 52:59–68CrossRef
33.
Zurück zum Zitat Nikolic B, Takeuchi Y, Leykin I, Fudaba Y, Smith RN, Sykes M (2004) Mixed hematopoietic chimerism allow cure of autoimmune diabetes through allogeneic tolerance and reversal of autoimmunity. Diabetes 53:376–383CrossRef Nikolic B, Takeuchi Y, Leykin I, Fudaba Y, Smith RN, Sykes M (2004) Mixed hematopoietic chimerism allow cure of autoimmune diabetes through allogeneic tolerance and reversal of autoimmunity. Diabetes 53:376–383CrossRef
34.
Zurück zum Zitat Kawai T, Andrews D, Colvin RB, Sachs DH, Cosimi AB (2000) Thromboembolic complications after treatment with monoclonal antibody against CD40 ligand. Nat Med 6:114CrossRef Kawai T, Andrews D, Colvin RB, Sachs DH, Cosimi AB (2000) Thromboembolic complications after treatment with monoclonal antibody against CD40 ligand. Nat Med 6:114CrossRef
35.
Zurück zum Zitat Kim SC, Wakwe W, Higginbotham LB et al (2017) Fc-Silent anti-CD154 domain antibody effectively prevents nonhuman primate renal allograft rejection. Am J Transpl 17:1182–1192CrossRef Kim SC, Wakwe W, Higginbotham LB et al (2017) Fc-Silent anti-CD154 domain antibody effectively prevents nonhuman primate renal allograft rejection. Am J Transpl 17:1182–1192CrossRef
Metadaten
Titel
Combination therapy of an iNKT cell ligand and CD40–CD154 blockade establishes islet allograft acceptance in nonmyeloablative bone marrow transplant recipients
verfasst von
Taichi Kanzawa
Toshihito Hirai
Hironori Fukuda
Haruki Katsumata
Rumi Ishii
Masako Ikemiyagi
Yasuyuki Ishii
Kan Saiga
Masayoshi Okumi
Kazunari Tanabe
Publikationsdatum
13.02.2019
Verlag
Springer Milan
Erschienen in
Acta Diabetologica / Ausgabe 5/2019
Print ISSN: 0940-5429
Elektronische ISSN: 1432-5233
DOI
https://doi.org/10.1007/s00592-019-01289-7

Weitere Artikel der Ausgabe 5/2019

Acta Diabetologica 5/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.