Neonatal induction of allogeneic tolerance prevents T cell‐mediated autoimmunity in NOD mice

Diabetes in the NOD mouse strain is a genetically programmed T cell‐mediated autoimmune process that is directed against an as yet unknown antigen target(s) on pancreatic β cells. To investigate whether the course of the autoimmune disease could be altered by immune manipulations of the T cell repertoire, we have induced allogeneic tolerance by injecting F1 semiallogeneic spleen cells into NOD neonates. This procedure resulted in a significant protection against both insulitis and diabetes. However, although it requires the induction of tolerance, as shown by the failure of non‐tolerizing irradiated cells to prevent autoimmunity, protection appeared to be independent of the major histocompatibility complex haplotypes of the F1 spleen cells injected at birth, e.g. (C57BL/6 x NOD)F1, (CBA/Ca x NOD)F1 or (BALB/c x NOD)F1 cells. In addition, a similar degree of protection was induced, whether the tolerant state, as assessed by mixed lymphocyte reaction studies in vitro, was of short duration, ∼6 weeks, or lasted for more than 12 weeks. Putative veto or suppressor functions of chimeric T cells were ruled out, since mice tolerized with T cell‐depleted F1 spleen cells were equally protected.

[1]  P. Bedossa,et al.  Adoptive T cell transfer of autoimmune nonobese diabetic mouse diabetes does not require recruitment of host B lymphocytes. , 1988, Journal of immunology.

[2]  R. Zinkernagel,et al.  Intrathymic elimination of Mlsa-reactive (V beta 6+) cells during neonatal tolerance induction to Mlsa-encoded antigens , 1988, The Journal of experimental medicine.

[3]  L. Wicker,et al.  Both the Lyt-2+ and L3T4+ T cell subsets are required for the transfer of diabetes in nonobese diabetic mice. , 1988, Journal of immunology.

[4]  J. Streilein,et al.  Modification of the cytotoxic T cell repertoire in neonatal tolerance. Evidence for preferential survival of cells with low avidity for tolerogen. , 1987, Journal of immunology.

[5]  C. Boitard,et al.  Syngeneic transfer of autoimmune diabetes from diabetic NOD mice to healthy neonates. Requirement for both L3T4+ and Lyt-2+ T cells , 1987, The Journal of experimental medicine.

[6]  D. Coleman,et al.  Three recessive loci required for insulin-dependent diabetes in nonobese diabetic mice. , 1987, Science.

[7]  J. Streilein,et al.  Characterization of cytotoxic cells in mice rendered neonatally tolerant of MHC alloantigens: evidence for repertoire modification. , 1987, Journal of immunology.

[8]  L. Wicker,et al.  Genetic control of diabetes and insulitis in the nonobese diabetic (NOD) mouse , 1987, The Journal of experimental medicine.

[9]  S. Slavin,et al.  Alloantigen persistence in induction and maintenance of transplantation tolerance , 1987, The Journal of experimental medicine.

[10]  H. Mcdevitt,et al.  The first external domain of the nonobese diabetic mouse class II I-A beta chain is unique. , 1987, Proceedings of the National Academy of Sciences of the United States of America.

[11]  K. Yamamura,et al.  Prevention of autoimmune insulitis by expression of I–E molecules in NOD mice , 1987, Nature.

[12]  M. Dorf,et al.  The NOD mouse: recessive diabetogenic gene in the major histocompatibility complex. , 1986, Science.

[13]  P. Pereira,et al.  Establishment of idiotypic helper T-cell repertoires early in life , 1985, Nature.

[14]  J. Berzofsky,et al.  CORRELATION OF INTRATHYMIC TOLERANCE WITH INTRATHYMIC CHIMERISM IN NEONATALLY TOLERIZED MICE , 1985, Transplantation.

[15]  P. Pereira,et al.  Internal complementarities in the immune system: regulation of the expression of helper T-cell idiotypes. , 1984, Proceedings of the National Academy of Sciences of the United States of America.

[16]  S. Ishizaka,et al.  Early loss of precursors of CTL and IL 2-producing cells in the development of neonatal tolerance to alloantigens. , 1984, Journal of immunology.

[17]  J. Louis,et al.  Clonal analysis of T cell unresponsiveness to alloantigens induced by neonatal injection of F1 spleen cells into parental mice. , 1983, Journal of immunology.

[18]  W. Silvers,et al.  Spontaneous diabetes in rats: destruction of islets is prevented by immunological tolerance. , 1981, Science.

[19]  G. Nossal,et al.  Functional clonal deletion in immunological tolerance to major histocompatibility complex antigens. , 1981, Proceedings of the National Academy of Sciences of the United States of America.

[20]  R. Schwartz,et al.  Alloreactivity of an antigen-specific T-cell clone , 1980, Nature.

[21]  Y. Tochino,et al.  Breeding of a non-obese, diabetic strain of mice. , 1980, Jikken dobutsu. Experimental animals.

[22]  H. Hengartner,et al.  Fine specificity of a continuously growing killer cell clone specific for H‐Y antigen , 1979, European journal of immunology.

[23]  Ryser Je,et al.  Limiting Dilution Analysis of Alloantigen-Reactive T Lymphocytes I. Comparison of Precursor Frequencies for Proliferative and Cytolytic Responses , 1979 .

[24]  J. Klein,et al.  Neonatal tolerance induction across regions of H-2 complex. , 1977, Journal of immunology.

[25]  S. Dorsch,et al.  Recirculating, suppressor T cells in transplantation tolerance , 1977, The Journal of experimental medicine.

[26]  B. Chapuis,et al.  Neonatally induced transplantation tolerance: in vitro evidence supporting a clonal inactivation mechanism , 1975, European journal of immunology.

[27]  W. Silvers,et al.  Importance of chimerism in maintaining tolerance of skin allografts in mice. , 1973, Journal of immunology.

[28]  P. Medawar,et al.  In vitro reactivity of lymphoid cells from tolerant mice. , 1973, Transplantation proceedings.

[29]  P. Medawar,et al.  ‘Actively Acquired Tolerance’ of Foreign Cells , 1953, Nature.

[30]  P. Medawar,et al.  The Technique of Free Skin Grafting in Mammals , 1951 .