Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Thyroid ; 15(2): 100-4, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15753666

ABSTRACT

In both medullary carcinoma and papillary carcinoma of the thyroid, altered expression of the RET gene is implicated in tumorigenesis. Recent studies suggest that loss of heterozygosity (LOH) at the G691S SNP may be associated with tumors from patients with a history of radiation exposure. We investigated LOH for three RET SNPs (G691S, S904S, and L769L) in tumor and normal tissue from 46 patients from Ukraine and Belarus who were exposed to radioactive fallout following the Chernobyl nuclear accident and were operated for papillary thyroid carcinoma between 1995 and 2000. Normal tissue from 28 patients was heterozygous for at least one SNP; DNA from the corresponding tumor samples was also heterozygous, indicating that no LOH had taken place. To assess SNP frequencies in a radiation-associated thyroid cancer cohort, we investigated a further 68 unpaired post-Chernobyl samples. For G691S, there was considerable deviation from Hardy-Weinberg equilibrium; more detailed analysis showed that this was linked to age at onset of disease. Among younger patients, the distribution of genotypes conformed to Hardy-Weinberg equilibrium; among older patients, we observed marked deviation (p = 0.0072), with significant over-representation of the rare S allele relative to the younger groups (Fisher's exact, p = 0.0233). This suggests that SNPs in the RET oncogene may play a role in sporadic papillary thyroid carcinoma.


Subject(s)
Carcinoma, Papillary/genetics , Loss of Heterozygosity , Neoplasms, Radiation-Induced/genetics , Oncogene Proteins/genetics , Polymorphism, Single Nucleotide , Receptor Protein-Tyrosine Kinases/genetics , Thyroid Neoplasms/genetics , Adolescent , Adult , Aged , Chernobyl Nuclear Accident , Child , Cohort Studies , Gene Frequency , Genetic Linkage , Humans , Middle Aged , Proto-Oncogene Proteins c-ret , Radioactive Fallout/adverse effects , Republic of Belarus , Ukraine
2.
Endocrinology ; 140(7): 3219-27, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385418

ABSTRACT

Tumor necrosis factor-alpha (TNFalpha) is a potential mediator of beta cell destruction in insulin-dependent diabetes mellitus. We have studied TNF-responsive pathways leading to apoptosis in beta cells. Primary beta cells express low levels of the type I TNF receptor (TNFR1) but do not express the type 2 receptor (TNFR2). Evidence for TNFR1 expression on beta cells came from flow cytometry using monoclonal antibodies specific for TNFR1 and TNFR2 and from RT-PCR of beta cell RNA. NIT-1 insulinoma cells similarly expressed TNFR1 (at higher levels than primary beta cells) as detected by flow cytometry and radio-binding studies. TNF induced NF-kappaB activation in both primary islet cells and NIT-1 cells. Apoptosis in response to TNFalpha was observed in NIT-1 cells whereas apoptosis of primary beta cells required both TNFalpha and interferon-gamma (IFNgamma). Apoptosis could be prevented in NIT-1 cells by expression of dominant negative Fas-associating protein with death domain (dnFADD). Apoptosis in NIT-1 cells was increased by coincubation with IFNgamma, which also increased caspase 1 expression. These data show that TNF-activated pathways capable of inducing apoptotic cell death are present in beta cells. Caspase activation is the dominant pathway of TNF-induced cell death in NIT-1 cells and may be an important mechanism of beta cell damage in insulin-dependent diabetes mellitus.


Subject(s)
Apoptosis/drug effects , Insulinoma/pathology , Islets of Langerhans/physiology , Pancreatic Neoplasms/pathology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/physiology , Caspase 1/metabolism , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , Female , Insulinoma/metabolism , Insulinoma/physiopathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred NOD , NF-kappa B/physiology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/physiopathology , Receptors, Tumor Necrosis Factor/metabolism , Tumor Cells, Cultured/drug effects
3.
Microbes Infect ; 3(11): 905-10, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11564438

ABSTRACT

Immunological tolerance to self is, in part, an active process mediated by a certain population of T lymphocytes. Our own work on the origin, phenotypic characterisation and mode of action of these regulatory T cells in rats forms the main focus of this review.


Subject(s)
Autoimmune Diseases/immunology , T-Lymphocytes/immunology , Thymus Gland/cytology , Animals , Disease Models, Animal , Down-Regulation , Humans , Organ Specificity , Rats , Self Tolerance
4.
Anticancer Res ; 22(6C): 3873-7, 2002.
Article in English | MEDLINE | ID: mdl-12553007

ABSTRACT

BACKGROUND: We have developed a PCR-based technique which has previously been shown to be capable of detecting DNA damage (Adducts, strand breaks) induced by carcinogens. We wanted to explore the possibility of detecting anti-carcinogens with this methodology through a corresponding reduction in DNA damage induction. MATERIALS AND METHODS: We have used a PCR-based system which relies on the fact that Taq polymerase cannot amplify damaged DNA efficiently. We immobilised DNA in microtitre plates and exposed them to UV-B light and hydrogen peroxide. In addition, we tested the capacity of sunscreens and ascorbate (vitamin C) to ameliorate the DNA damage caused by UV-B and hydrogen peroxide, respectively. RESULTS: We have shown that in the case of UV-B, sunscreens can effectively reduce the amount of DNA damage induced. This appears to be dependent upon the Sun Protection Factor of the sunscreen, with SPF 35 sunscreens protecting DNA particularly well. We found that both hydrogen peroxide and ascorbate were capable, at high doses, of inducing DNA damage. When mixed, ascorbate effectively increased the DNA damaging effect of hydrogen peroxide. CONCLUSION: We have shown that the PCR inhibition method may be suitable for untangling the complex carcinogenic/anticarcinogenic aspects of factors in the diet and environment. However, the DNA damaging effect of ascorbate must be viewed in the context of the high concentrations used here.


Subject(s)
Anticarcinogenic Agents/pharmacology , Ascorbic Acid/pharmacology , DNA Damage/drug effects , Reactive Oxygen Species/toxicity , Sunscreening Agents/pharmacology , Ultraviolet Rays/adverse effects , Animals , Antioxidants/adverse effects , Antioxidants/pharmacology , Ascorbic Acid/adverse effects , DNA/drug effects , DNA/metabolism , DNA/radiation effects , Dose-Response Relationship, Drug , Drug Synergism , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/toxicity , Mice , Polymerase Chain Reaction/methods , Reactive Oxygen Species/metabolism
5.
Int Immunol ; 7(12): 1885-95, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8746558

ABSTRACT

Expression of the co-stimulatory molecule B7-1 (CD80) on pancreatic beta cells can overcome peripheral T cell tolerance in transgenic models of autoimmune disease. This study aimed to determine if aberrant B7-1 or B7-2 (CD86) expression on pancreatic beta cells is involved in the pathogenesis of autoimmune diabetes in non-obese diabetic (NOD) mice. Immunohistochemical analysis of NOD pancreas sections revealed no evidence of B7-1 or B7-2 expression on pancreatic beta cells at any stage prior to the onset of either spontaneously arising or cyclophosphamide-accelerated diabetes. Likewise, the NOD-derived NIT-1 beta cell line did not express surface B7 or B7-1 mRNA either constitutively or following exposure to IFN-gamma and TNF-alpha, two cytokines known to be present in the insulitis lesion of NOD mice, or cAMP which can induce B7-1 expression on B cells. Both B7-1 and B7-2 were, however, highly expressed on the majority of islet-infiltrating inflammatory cells in NOD mice between days 7 and 12 after the administration of cyclophosphamide which results in accelerated beta cell destruction. Likewise B7-1 and B7-2 were extensively expressed on islet-infiltrating cells present at the time of diabetes onset in NOD SCID mice with adoptively transferred diabetes. By immunohistochemistry and flow cytometry, it was determined that the phenotype of B7+ cells in the pancreas of NOD mice 9 days after cyclophosphamide included a mixture of macrophages and both CD4+ and CD8+ T cells. B7-2 was also expressed on islet-infiltrating cells in the spontaneously occurring diabetes of female NOD mice, but the levels of B7-1 expression were low in comparison with the accelerated models of diabetes. RIP-IL-2 transgenic mice, which have extensive islet infiltration but no autoimmune beta cell destruction, also had virtually no B7-1 expression and a minority of B7-2-expressing inflammatory cells. Thus, the activation of beta cell-specific T cells in NOD mice does not appear to be a result of aberrant expression of B7 on the beta cells. Expression of B7-1 and B7-2 on islet-infiltrating cells is, however, associated with autoimmune beta cell destruction, suggesting a role for the B7-CD28 interaction in this process.


Subject(s)
B7-1 Antigen/metabolism , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , B7-1 Antigen/genetics , B7-2 Antigen , Base Sequence , Cell Line , DNA Primers/genetics , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/genetics , Female , Flow Cytometry , Gene Expression , Immunohistochemistry , Interleukin-2/genetics , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Molecular Sequence Data , Phenotype
6.
J Immunol ; 165(6): 3105-10, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10975823

ABSTRACT

Previously we have shown that autoimmune diabetes, induced in rats by a protocol of adult thymectomy and split-dose gamma irradiation, can be prevented by the transfer of a subset of CD4+ T cells with a memory phenotype (CD45RC-), as well as by CD4+CD8- thymocytes, from syngeneic donors. Further studies now reveal that in the thymus the regulatory cells are observed in the CD25+ subset of CD4+CD8- cells, whereas transfer of the corresponding CD25- thymocyte subset leads to acceleration of disease onset in prediabetic recipients. However, in the periphery, not all regulatory T cells were found to be CD25+. In thoracic duct lymph, cells that could prevent diabetes were found in both CD25- and CD25+ subsets of CD4+CD45RC- cells. Further, CD25- regulatory T cells were also present within the CD4+CD45RC- cell subset from spleen and lymph nodes, but were effective in preventing diabetes only after the removal of CD25- recent thymic emigrants. Phenotypic analysis of human thymocytes showed the presence of CD25+ cells in the same proportions as in rat thymus. The possible developmental relationship between CD25+ and CD25- regulatory T cells is discussed.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Receptors, Interleukin-2/physiology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Adoptive Transfer , Animals , Biomarkers/analysis , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , CD8 Antigens/biosynthesis , Cell Differentiation/immunology , Cell Movement/immunology , Cell Separation , Female , Immunologic Memory , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Depletion , Radiation Chimera/immunology , Rats , Receptors, Interleukin-2/biosynthesis , Spleen/cytology , Spleen/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , Thoracic Duct/cytology , Thoracic Duct/immunology , Thymectomy , Thymus Gland/cytology , Thymus Gland/metabolism , Thymus Gland/transplantation
7.
J Autoimmun ; 10(3): 293-8, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9218757

ABSTRACT

We have recently observed that inhibition of NF-kappaB in NIT-1 insulinoma cells protects them from tumour necrosis factor (TNF)-induced cell death in vitro, possibly because expression of interleukin-1 (IL-1)beta-converting enzyme (ICE), a member of the cysteine protease pathway of cell death, is decreased. In the current study we have examined the effect of the same inhibitor of NF-kappaB on class I major histocompatibility complex (MHC) protein expression in NIT-1 cells and shown that inhibition of NF-kappaB activation decreased basal and TNF-induced class I MHC levels. Although inducible nitric oxide synthase (iNOS) may also be inhibited by inhibition of NF-kappaB, this could not be demonstrated in NIT-1/delta sp cells because wild-type NIT-1 cells express very little iNOS. When NIT-1/delta sp12 cells, expressing high levels of the NF-kappaB inhibitor, are transplanted into immunodeficient NOD/scid mice, tumorigenesis and death by hypoglycemia proceed similarly to untransfected NIT-1 cells. Untransfected NIT-1 cells were killed by co-transfer of splenic T cells from diabetic but not non-diabetic NOD mice. NIT-1/delta sp12 cells were protected from killing in vivo by T cells from diabetic mice, in that tumours developed in four out of five mice and the kinetics of tumour development were not significantly delayed. NIT-1/delta sp12 cells were not protected from killing by T cells from mice previously primed with NIT-1 cells. In conclusion, inhibition of NF-kappaB is likely to suppress several different pathways of immune-mediated cell death in beta-cells and protects NIT-1 cells from immune attack by diabetogenic T cells in vivo. Inhibition of NF-kappaB is a potentially effective strategy for protection of pancreatic beta-cells in autoimmune diabetes.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/toxicity , Adoptive Transfer , Animals , Cell Death/immunology , Cell Line, Transformed , Cytotoxicity, Immunologic , Diabetes Mellitus, Type 1/prevention & control , Female , H-2 Antigens/biosynthesis , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Nitric Oxide Synthase/biosynthesis , T-Lymphocytes/transplantation , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/toxicity
8.
Eur J Immunol ; 31(4): 1247-54, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11298351

ABSTRACT

CD4(+)CD25(+) T cells in mice and rats are capable of transferring protection against organ-specific autoimmune disease and colitis and suppressing the proliferation of other T cells after polyclonal stimulation in vitro. Here we describe the existence in humans of CD4(+)CD25(+) T cells with the same in vitro characteristics. CD4(+)CD8(-)CD25(+) T cells are present in both the thymus and peripheral blood of humans ( approximately 10 % of CD4(+)CD8(-) T cells), proliferate poorly in response to mitogenic stimulation and suppress the proliferation of CD4(+)CD25(-) cells in co-culture. This suppression requires cell contact and can be overcome by the addition of exogenous IL-2. CD4(+)CD25(+) cells from thymus and blood were poor producers of IL-2 and IFN-gamma, and suppressed the levels of these cytokines produced by CD4(+)CD25(-) cells. However, CD4(+)CD25(+) PBL produced higher levels of IL-4 and similar amounts of IL-10 as CD4(+)CD25(-) cells. Regulatory CD4(+)CD25(+) T cells have an activated phenotype in the thymus with expression of CTLA-4 and CD122 (IL-2Rbeta). The fact that CD4(+)CD25(+) regulatory T cells are present with a similar frequency in the thymus of humans, rats and mice, suggests that the role of these cells in the maintenance of immunological tolerance is an evolutionarily conserved mechanism.


Subject(s)
CD4 Antigens/metabolism , Immune Tolerance/immunology , Receptors, Interleukin-2/metabolism , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Autoimmunity/immunology , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Communication/drug effects , Cell Division/drug effects , Cells, Cultured , Coculture Techniques , Flow Cytometry , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Ionomycin/pharmacology , Lymphocyte Activation/drug effects , Mitogens/immunology , Receptors, Interleukin-2/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Thymus Gland/drug effects , Thymus Gland/metabolism
9.
J Immunol ; 157(8): 3688-93, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8871671

ABSTRACT

Beta2m-deficient nonobese diabetic (NODbeta2mnull) do not develop insulitis or diabetes. Expression of a beta2m transgene controlled by the rat insulin promoter (RIP-beta2m) in NODbeta2mnull mice resulted in reconstitution of IFN-gamma-inducible cell surface MHC class I protein on pancreatic beta-cells. These mice developed insulitis, but did not develop diabetes. Transfer of T cells from diabetic NOD mice to NODbeta2mnull recipients resulted in insulitis, which took several months to progress to diabetes. In contrast, transgenic RIP-beta2m/NODbeta2mnull mice with islet MHC class I reconstitution developed diabetes rapidly after transfer of diabetic NOD spleen cells. Administration of cyclophosphamide, which accelerates diabetes in NOD mice, resulted in 43% of RIPbeta2m/NODbeta2mnull mice becoming diabetic compared with 75% of wild-type mice and 0% of NODbeta2mnull mice. Acceleration of diabetes by cyclophosphamide was prevented by anti-CD8 mAb treatment. FACS analysis of peripheral blood and lymphoid organs from transgene-bearing animals did not show an increase in the number of CD8+ T cells compared with that in NODbeta2mnull mice. In summary, beta-cell expression of beta2m in NODbeta2mnull mice resulted in a return of insulitis, but not spontaneous diabetes. These studies demonstrate that beta2m and cell surface MHC class I expression on beta-cells are essential for the initiation of diabetes in the NOD mouse and further confirm that efficient progression to diabetes requires both CD4+ and CD8+ T cells.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Insulin/genetics , Promoter Regions, Genetic , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Female , Histocompatibility Antigens Class I/metabolism , Immunohistochemistry , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphocyte Subsets/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Rats
10.
Mol Med ; 4(8): 495-501, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9742505

ABSTRACT

BACKGROUND: The non-obese diabetic (NOD) mouse is a model of human type 1 diabetes in which autoreactive T cells mediate destruction of pancreatic islet beta cells. Although known to be triggered by cytotoxic T cells, apoptosis has not been unequivocally localized to beta cells in spontaneously diabetic NOD mice. We created a model of accelerated beta-cell destruction mediated by T cells from spontaneously diabetic NOD mice to facilitate the direct detection of apoptosis in beta cells. MATERIALS AND METHODS: NOD.scid (severe combined immunodeficiency) mice were crossed with bm1 mice transgenically expressing the costimulatory molecule B7-1 (CD80) in their beta cells, to generate B7-1 NOD.scid mice. Apoptosis in islet cells was measured as DNA strand breakage by the TdT-mediated-dUTP-nick end labeling (TUNEL) technique. RESULTS: Adoptive transfer of splenocytes from spontaneously diabetic NOD mice into B7-1 NOD.scid mice caused diabetes in recipients within 12-16 days. Mononuclear cell infiltration and apoptosis were significantly greater in the islets of B7-1 NOD.scid mice than in nontransgenic NOD.scid mice. Dual immunolabeling for TUNEL and either B-7 or insulin, or the T cell markers CD4 and CD8, and colocalization by confocal microscopy clearly demonstrated apoptosis in beta cells as well in a relatively larger number of infiltrating T cells. The clearance time of apoptotic beta cells was estimated to be less than 6 min. CONCLUSIONS: B7-1 transgenic beta cells undergo apoptosis during their accelerated destruction in response to NOD mouse effector T cells. Rapid clearance implies that beta cells undergoing apoptosis would be detected only rarely during more protracted disease in spontaneously diabetic NOD mice.


Subject(s)
Apoptosis/immunology , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/pathology , Adoptive Transfer , Animals , B7-1 Antigen/physiology , Diabetes Mellitus, Type 1/immunology , Insulin/analysis , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Pancreas/immunology , Pancreas/pathology , Spleen/immunology , T-Lymphocytes/physiology
11.
Immunol Rev ; 182: 190-200, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11722634

ABSTRACT

Transfer of CD4+ T cells to immune-deficient mice in the absence of the CD25+ subset leads to the development of colitis, indicating that regulatory cells capable of controlling a bacteria-driven inflammatory response are present in normal mice. Cells with this function are present in the thymus as well as in the periphery of germ-free mice, suggesting they may be reactive with self-antigen. These cells resemble CD4+CD25+ cells that inhibit organ-specific autoimmunity, suggesting that a similar subset of regulatory T cells may control responses to self and foreign antigens. Development of colitis is dependent on accumulation of activated CD134L+ dendritic cells (DC) in the mesenteric lymph nodes, which is inhibited by CD4+CD25+ cells, indicating that regulatory T cells may control DC activation in vivo. Whilst inhibition of T-cell activation in vitro by CD4+CD25+ cells does not involve interleukin-10 and transforming growth factor-beta, these cytokines are required for the suppression of colitis. It may be that control of responses that activate the innate immune system requires multiple mechanisms of immune suppression. Recently, we identified CD4+CD25+ cells with immune suppressive activity in the thymus and peripheral blood of humans, raising the possibility that dysfunction in this mechanism of immune regulation may be involved in the development of autoimmune and inflammatory diseases.


Subject(s)
Antigens, CD , Colitis/immunology , Colitis/pathology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , T-Lymphocyte Subsets/immunology , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Animals , Antigens, Differentiation/immunology , Bacteria/pathogenicity , Dendritic Cells/immunology , Humans , Interleukin-10/immunology , Leukocyte Common Antigens/immunology , Membrane Glycoproteins , NAD+ Nucleosidase/immunology , Receptors, Interleukin-2/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Transforming Growth Factor beta/immunology
12.
Eur J Immunol ; 28(3): 949-60, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9541590

ABSTRACT

The concept that naive CD4+ and CD8+ T cells require co-stimulatory signals for activation and proliferation is well documented. Less clear is the need for co-stimulation during the effector phase of the T cell response. Here we examined the influence of B7-1 (CD80) during the effector phase of an autoimmune response to pancreatic islets using transgenic mouse lines which expressed B7-1 in either all or only some of their beta cells ("confluent" or "patchy" RIP-B7-1 mice). Transgenic expression of B7-1 in normal mouse islets that co-expressed the pro-inflammatory cytokine, IL-2, resulted in early spontaneous autoimmunity. Islets with IL-2 and "confluent" B7-1 expression were destroyed whereas islets with IL-2 and "patchy" B7-1 expression showed selective killing of the B7-1+ beta cells. Islet-reactive T cells, circulating in the RIP-B7-1/IL-2 mice, rejected syngeneic islet grafts, but only if these expressed B7-1. Introduction of the B7-1 transgene into the nonobese diabetic (NOD) genetic background likewise resulted in early spontaneous autoimmunity, but splenocytes from the diabetic animals could only transfer diabetes to NOD scid recipients that expressed B7-1 on their beta cells. In both these transgenic models, therefore, islet destruction required continuous B7-1 expression by target beta cells. Thus, although the normal repertoire contains T cells with potential islet reactivity, these T cells remain harmless because parenchymal cells like the beta cell cannot normally express B7-1. Our results also have implications for tumor immunotherapy in that the ability of T cells to kill poorly immunogenic targets may be dependent upon B7-1 expression by the target cell itself.


Subject(s)
Autoimmunity , B7-1 Antigen/physiology , T-Lymphocytes/immunology , Animals , Cytotoxicity, Immunologic , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Interleukin-2/metabolism , Islets of Langerhans/immunology , Lymphocyte Activation , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL