Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Acta Biol Hung ; 54(2): 167-76, 2003.
Article in English | MEDLINE | ID: mdl-14535622

ABSTRACT

Psychological stress modulates the immune system through the hypothalamic-pituitary-adrenal axis, the sympatho-adrenomedullary axis and the opioid system. According to literature data, restraint stress increases the immune cell apoptosis, decreases the spleen and thymus cell content, the natural killer (NK) activity in the spleen, and it compromises the anti-tumor immune response in mice. We immobilized mice in two consecutive nights, and then determined the cell number, apoptosis, NK cell content, NK activity and the level of cytokine mRNAs (TNF-beta, TNF-alpha, IL-4, IL-5, IL-1alpha, IFN-gamma, IL-2, IL-6, IL-1beta and IL-3) in the thymus and spleen. No consistent changes were detected in any of the immune parameters either in C57Bl/6 or in DBA/2 mice. Stressed or control B6 mice were injected with B16 melanoma cells immediately after the immobilization or one week later. No significant differences were found in the growth of primary tumors and lung metastases in stressed and control animals. Taken together, our mice, kept in a general-purpose non-SPF animal house, seemed to be refractory to the stress-induced immunomodulation. Our interpretation is that stress-induced immunomodulation can occur only in mice isolated from any background stressors, or rather natural stimuli, throughout their life.


Subject(s)
Cytokines/metabolism , Immobilization , Melanoma, Experimental/immunology , Spleen/immunology , Stress, Physiological/immunology , Thymus Gland/immunology , Animals , Antibodies, Monoclonal , Cytokines/genetics , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasm Transplantation , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , Spleen/cytology , Spleen/physiology , Thymus Gland/cytology , Thymus Gland/physiology , Tumor Cells, Cultured
2.
J Immunol ; 165(11): 6314-21, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11086068

ABSTRACT

CD8(+) T cells are important effectors, as well as regulators, of organ-specific autoimmunity. Compared with Tc1-type CD8(+) cells, Tc2 cells have impaired anti-viral and anti-tumor effector functions, although no data are yet available on their pathogenic role in autoimmunity. Our aim was to explore the role of autoreactive Tc1 and Tc2 cells in autoimmune diabetes. We set up an adoptive transfer model in which the recipients were transgenic mice expressing influenza virus hemagglutinin (HA) specifically in their pancreatic ss islet cells (rat insulin promoter-HA mice) and islet-specific Tc1 and Tc2 cells were generated in vitro from HA-specific CD8(+) cells of TCR transgenic mice (CL4-TCR mice). One million Tc1 cells, differentiated in vitro in the presence of IL-12, transferred diabetes in 100% of nonirradiated adult rat insulin promoter-HA recipients; the 50% diabetogenic dose was 5 x 10(5). Highly polarized Tc2 cells generated in the presence of IL-4, IL-10, and anti-IFN-gamma mAb had a relatively low, but definite, diabetogenic potential. Thus, 5 x 10(6) Tc2 cells caused diabetes in 6 of 18 recipients, while the same dose of naive CD8(+) cells did not cause diabetes. Looking for the cause of the different diabetogenic potential of Tc1 and Tc2 cells, we found that Tc2 cells are at least as cytotoxic as Tc1 cells but their accumulation in the pancreas is slower, a possible consequence of differential chemokine receptor expression. The diabetogenicity of autoreactive Tc2 cells, most likely caused by their cytotoxic activity, precludes their therapeutic use as regulators of autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes/transplantation , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/immunology , Islets of Langerhans/immunology , T-Lymphocyte Subsets/transplantation , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Insulin/genetics , Islets of Langerhans/pathology , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Pancreas/immunology , Pancreas/pathology , Promoter Regions, Genetic/immunology , Rats , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Tumor Cells, Cultured
3.
J Immunol ; 165(1): 202-10, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861053

ABSTRACT

Insulin-dependent diabetes is an autoimmune disease targeting pancreatic beta-islet cells. Recent data suggest that autoreactive CD8+ T cells are involved in both the early events leading to insulitis and the late destructive phase resulting in diabetes. Although therapeutic injection of protein and synthetic peptides corresponding to CD4+ T cell epitopes has been shown to prevent or block autoimmune disease in several models, down-regulation of an ongoing CD8+ T cell-mediated autoimmune response using this approach has not yet been reported. Using CL4-TCR single transgenic mice, in which most CD8+ T cells express a TCR specific for the influenza virus hemagglutinin HA512-520 peptide:Kd complex, we first show that i.v. injection of soluble HA512-520 peptide induces transient activation followed by apoptosis of Tc1-like CD8+ T cells. We next tested a similar tolerance induction strategy in (CL4-TCR x Ins-HA)F1 double transgenic mice that also express HA in the beta-islet cells and, as a result, spontaneously develop a juvenile onset and lethal diabetes. Soluble HA512-520 peptide treatment, at a time when pathogenic CD8+ T cells have already infiltrated the pancreas, very significantly prolongs survival of the double transgenic pups. In addition, we found that Ag administration eliminates CD8+ T cell infiltrates from the pancreas without histological evidence of bystander damage. Our data indicate that agonist peptide can down-regulate an autoimmune reaction mediated by CD8+ T cells in vivo and block disease progression. Thus, in addition to autoreactive CD4+ T cells, CD8+ T cells may constitute targets for Ag-specific therapy in autoimmune diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/prevention & control , Islets of Langerhans/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/agonists , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Animals, Newborn/immunology , Apoptosis/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , CD8-Positive T-Lymphocytes/virology , Cytotoxicity, Immunologic , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Injections, Intravenous , Islets of Langerhans/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Transgenic , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Spleen/immunology , Spleen/pathology
4.
APMIS ; 107(10): 903-12, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10549587

ABSTRACT

Groups of CBA mice immunosuppressed with anti-thymocyte serum (ATS) treatment were xeno-transplanted with either HeLa human cervical carcinoma cells or genetically modified cells expressing the human tumor necrosis factor-alpha (TNF) gene (All cells). Both cell lines were highly resistant to the cytotoxic effects of TNF. If 3 x 10(6) tumor cells were inoculated s.c. into female mice, HeLa cells grew progressively into large tumors and killed 74% of the recipients, while TNF-expressing All cells caused fatal tumor growth only in 22% of the mice. 3 x 10(6) or 1.5 x 10(7). All cells produced progressive tumor growth and lethality in all male recipients. In sera of all the A11-cell-transplanted mice, biologically active TNF was detected shortly (4.5 h) after tumor inoculation (6 39 U/ml), decreasing to below detection level in the circulation by day 3. In recipients of 15 million A11 cells, circulating TNF reappeared and reached high levels (12-1000 U/ml) 3 to 7 weeks later, when the animals bore large tumors (14-23 mm). Generally, such mice became cachectic, severely anemic, hypothermic, and soon died. On account of calcium mobilization from bones, their serum Ca levels were high. Electron microscopy revealed severe liver damage, but there were no signs of chronic arthritis. These results suggest that ATS-treated mice xenotransplanted with TNF-gene-transfected A11 human tumor cells provide a new model for studying the pathophysiological and anti-tumor effects of TNF.


Subject(s)
Carcinoma/metabolism , Neoplasm Proteins/physiology , Neoplasm Transplantation , Tumor Necrosis Factor-alpha/physiology , Anemia/etiology , Animals , Antilymphocyte Serum , Body Temperature , Body Weight , Cachexia/etiology , Carcinoma/complications , Cytotoxicity, Immunologic , Female , HeLa Cells/metabolism , HeLa Cells/transplantation , Humans , Hypercalcemia/etiology , Hypothermia/etiology , Immunocompromised Host , L Cells/metabolism , L Cells/transplantation , Liver/ultrastructure , Male , Mice , Mice, Inbred CBA , Neoplasm Proteins/metabolism , Recombinant Fusion Proteins/physiology , T-Lymphocytes , Transfection , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/metabolism
5.
Immunol Rev ; 169: 81-92, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10450510

ABSTRACT

There is now convincing evidence that autoreactive CD8+ T cells can contribute to the pathogenesis of organ-specific autoimmune diseases. In the non-obese diabetic mouse, there is direct evidence that beta-islet cell-specific CD8+ cytotoxic T cells have a pathogenic effect. In human diseases such as autoimmune diabetes and multiple sclerosis, indirect evidence also suggests a role for CD8+ T cells in tissue damage, although their antigen specificity is unknown. Transgenic mouse models as well as the use of knockout mice have been instrumental in the identification of the role of autoreactive CD8+ T cells. Spontaneous models of CD8+ T-cell-mediated autoimmunity generated through transgenesis should help delineate the effector mechanisms leading to tissue destruction. The study of autoreactive CD8+ T cells and the characterization of their antigenic specificity should help unravel the pathophysiology of organ-specific autoimmune diseases, help identify exacerbating foreign antigens, and allow the design of antigen-specific immunotherapy targeting the pathogenic autoreactive T cells.


Subject(s)
Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Animals , Autoimmune Diseases/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Phenotype
6.
Br J Cancer ; 79(7-8): 1067-73, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10098738

ABSTRACT

The possibility of using interleukin 2 (IL-2)-activated natural killer cells (A-NK) to carry methoxymorpholinyl doxorubicin (MMDX; PNU 152243) to liver-infiltrating tumours was explored in mice bearing 2-day established M5076 reticulum cell sarcoma hepatic metastases. In vitro, MMDX was 5.5-fold more potent than doxorubicin against M5076 tumour cells. MMDX uptake by A-NK cells correlated linearly with drug concentration in the incubation medium [correlation coefficient (r) = 0.999]; furthermore, as MMDX incorporation was readily reproducible in different experiments, the amount of drug delivered by A-NK cells could be modulated. In vivo experiments showed that intravenous (i.v.) injection of MMDX-loaded A-NK cells exerted a greater therapeutic effect than equivalent or even higher doses of free drug. The increase in lifespan (ILS) following A-NK cell delivery of 53 microg kg(-1) MMDX, a dosage that is ineffective when administered in free form, was similar to that observed in response to 92 microg kg(-1) free drug, a dosage close to the 10% lethal dose (ILS 42% vs. 38% respectively). These results correlated with pharmacokinetic studies showing that MMDX encapsulation in A-NK cells strongly modifies its organ distribution and targets it to tissues in which IL-2 activated lymphocytes are preferentially entrapped after i.v. injection.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/analogs & derivatives , Immunotherapy, Adoptive/methods , Immunotoxins/administration & dosage , Interleukin-2 , Killer Cells, Lymphokine-Activated , Liver Neoplasms/therapy , Lymphoma, Large B-Cell, Diffuse/therapy , Adolescent , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Humans , Immunotoxins/pharmacokinetics , Liver Neoplasms/secondary , Mice , Tumor Cells, Cultured
7.
Rev Neurol (Paris) ; 154(8-9): 586-91, 1998 Sep.
Article in French | MEDLINE | ID: mdl-9809373

ABSTRACT

Experimental autoimmune encephalomyelitis is an induced inflammatory and demyelinating disease of the central nervous system widely used as an animal model for multiple sclerosis. New insights into its pathophysiology have been possible due to recent immunological concepts. New therapeutical approaches have been designed and tested in experimental autoimmune encephalomyelitis and are now entering the clinical setting.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Multiple Sclerosis/physiopathology , Animals , Demyelinating Diseases/physiopathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Humans , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Remission Induction
8.
Br J Cancer ; 77(4): 656-62, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9484826

ABSTRACT

In syngeneic mice, the H5V polyoma middle-T oncogene-transformed endothelioma cell line induces Kaposi's sarcoma-like cavernous haemangiomas that regress transiently, probably because of an anti-tumour immune response, but eventually grow progressively and kill the host. To evaluate the generation of tumour-specific cytotoxic T lymphocytes (CTLs), spleen cells of tumour-bearing mice were restimulated with irradiated H5V cells in mixed leucocyte-tumour cell cultures. Tumour-specific CTLs were demonstrable only when low numbers of H5V stimulator cells were used (<1 H5V cell per 50 splenocytes). We found that H5V cells secrete immunosuppressive mediators because CTL generation was blocked when H5V cells culture supernatants were added to allogeneic mixed leucocyte cultures. As numerous tumour-derived immunosuppressive mediators may interfere with interleukin 12 (IL-12) production, we tested whether IL-12 treatment of the tumour-bearing mice would augment their immune response and thus suppress tumour growth. Indeed, IL-12 inhibited tumour growth and prevented mortality, but did not increase anti-H5V CTL generation either in vitro or in vivo. Moreover, the anti-tumour activity in IL-12-treated mice was abrogated by anti-interferon (IFN)-gamma monoclonal antibody (MAb) co-administration. These results strongly suggest that the anti-tumour effect of IL-12 is principally mediated by IFN-gamma release that in turn blocks H5V cell proliferation and induces the release of factors that suppress angiogenesis.


Subject(s)
Hemangiosarcoma/immunology , Hemangiosarcoma/therapy , Interleukin-12/therapeutic use , Skin Neoplasms/immunology , Skin Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Monoclonal/pharmacology , Hemangiosarcoma/mortality , Immunity, Cellular , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin Neoplasms/mortality , Tumor Cells, Cultured
9.
Acta Chir Hung ; 36(1-4): 150-1, 1997.
Article in English | MEDLINE | ID: mdl-9408323

ABSTRACT

In A/J (H-2a) (A) mice, the neonatal i.v. injection of (B10 x A)F1 spleen cells (SC) induces partial transplantation tolerance (TT) to C57BL/10ScSn (H-2b) (B10) skin allografts, chronic host-versus-graft disease (HVGD) and lethal lymphoproliferative disorders (LPD). They produce anti-T-cell autoantibodies (ATA), and the proliferative responses of their SC to the T cell mitogen Con A are decreased. We found that, similar to ATA, the hyporeactivity of T cells developed earlier (at 1-2 weeks of age) than splenomegaly. The proportions of both CD4+ and CD8+ T cells were not reduced in the spleens of tolerized mice without manifest LPD. The supernatants (SN) of Con A-stimulated tolerized SC contained no, or only small amounts of interleukin-2 (IL-2). Thus, in the tolerized mice, ATA and T cell deficiency preceded the development of LPD. ATA and the decreased amount of the T cell growth factor IL-2 might play a role in the defective T cell activation.


Subject(s)
Antigens/immunology , Immune Tolerance/immunology , Lymphoproliferative Disorders/immunology , T-Lymphocytes/immunology , Transplantation Immunology , Age Factors , Animals , Animals, Newborn , Autoantibodies/immunology , CD4 Lymphocyte Count/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Division/drug effects , Cell Transplantation , Chronic Disease , Concanavalin A/pharmacology , Host vs Graft Reaction/immunology , Interleukin-2/analysis , Interleukin-2/immunology , Lymphocyte Activation/immunology , Lymphocyte Count/drug effects , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Inbred Strains , Mitogens/pharmacology , Skin Transplantation/immunology , Spleen/cytology , Splenomegaly/etiology , Splenomegaly/immunology , T-Lymphocytes/drug effects
11.
Scand J Immunol ; 41(3): 313-7, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7871394

ABSTRACT

Graft-versus-host disease (GVHD) due to allogeneic bone marrow transplantation can be prevented by depleting the T cells from the marrow graft in vitro. However, the elimination of the donor T cells results in a higher frequency of graft failure, secondary infections and, in case of leukaemia, relapse. We found, that, in contrast to normal spleen cells, spleen cells from A or B10 donor mice pretreated with xenogeneic antithymocyte serum (ATS) in vivo did not induce GVHD in non-irradiated (B10 x A)F1 hybrids. Spleen cells of ATS-pretreated A donors did not cause GVHD in allogeneic CBA mice made neonatally tolerant to the A donor strain either. Furthermore, spleen cells from ATS-treated donors did not cause GVHD in irradiated F1 hybrid recipients, moreover, they decreased the lethal effect of irradiation. The in vivo ATS pretreatment improved the repopulating capacity of spleen cells in irradiated syngeneic recipients, too. The effect of the ATS treatment does not rely solely upon the elimination of T cells, since flow cytofluorometric analysis revealed only a partial depletion of both the CD4+ and CD8+ T cells of the ATS-pretreated animals. These observations may also have clinical relevance.


Subject(s)
Antilymphocyte Serum/immunology , Bone Marrow Transplantation/immunology , Graft Enhancement, Immunologic , Graft vs Host Disease/immunology , Spleen/immunology , T-Lymphocytes/immunology , Animals , Animals, Newborn/immunology , Immune Tolerance , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Inbred CBA , Spleen/cytology , Whole-Body Irradiation
15.
Eur J Immunol ; 23(11): 3011-20, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8223878

ABSTRACT

We have reported that, in A/J (A) (H-2a) mice, a partial tolerance to C57BL/10ScSn (B10) (H-2b) skin allografts and a high incidence of lethal lymphoproliferative disorders (LPD) can be induced by the neonatal i.v. injection of 2 x 10(7) semiallogeneic (B10 x A)F1 spleen cells (SC) (Végh, P., Baranyi, L. and Jánossy, T., Cell. Immunol. 1990, 129: 56). In this study, we show that the incidence and mortality of LPD were continuously growing from 1 month of age until the end of the experiment at 1 year (64% and 36%, respectively). Based on histology, 27% of the diseased mice suffered from lymphoid malignancies. In the remaining cases (73%), reactive histopathological changes were seen in the spleen, lymph nodes (LN), liver and kidneys. The proportion of CD4+ T cells in the spleen and LN as well as that of splenic B cells decreased, while the percentages of mature and immature myeloid cells doubled. The total cell number of each (sub)population, however, was elevated in both lymphoid organs. The cells taking part in the lymphoproliferation were of host (A) and not of donor (F1) origin. Preceding the development of apparent LPD, the SC, LN cell and thymus cell suspensions of 1-month-old tolerized mice showed reduced in vitro proliferative responses to T cell and T cell-dependent B cell mitogens (Con A or PWM), while their reactivity to a T cell-independent B cell mitogen (lipopolysaccharide) was essentially unimpaired. This hyporeactivity seems to be functional, because neither histology nor immunophenotyping by flow cytometry revealed significant alterations in the spleen and thymus of such animals, apart from a slight reduction in the ratio of CD4+/CD8+ T cell subpopulations in the spleen. The in vivo T cell-mediated immune response of the tolerized mice was practically normal to third party CBA/Ca (H-2k) allografts. Antithymocyte autoantibodies (ATA) were detected in the sera of 76% of the tolerized mice at 1 month of age (i.e., even before the mass appearance of LPD). ATA as well as antinuclear Ab were present in 65% of the adult tolerized mice, independently of the presence of LPD. Taken together, in A mice neonatally injected with (B10 x A)F1 SC, a partial, specific allograft tolerance and a chronic host-vs.-graft disease-like syndrome developed. The latter is manifested in hyporeactivity to T cell mitogens, development of autoantibodies and, subsequently, in progressive LPD and lymphoid malignancies.


Subject(s)
Autoimmunity , Immune Tolerance , Lymphoproliferative Disorders/etiology , Animals , Animals, Newborn , Autoantibodies/blood , Graft Survival , Lymphoproliferative Disorders/pathology , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Inbred CBA , Mitogens/pharmacology , Phenotype , Skin Transplantation/immunology , Spleen/immunology , Spleen/transplantation , T-Lymphocyte Subsets/immunology , Transplantation Immunology
16.
Immunobiology ; 188(1-2): 172-93, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8406557

ABSTRACT

Two lymphomas were found in, and isolated from A (H-2a) mice in which permanent transplantation tolerance was induced to CBA (H-2k) histocompatibility antigens by the neonatal injection of (CBAxA)F1 spleen cells. They proved to be of recipient origin and were transferable to syngeneic A mice, growing as disseminated lymphomas (L33 and L46) and killing the recipients rapidly. Analysis of the cell surface antigens disclosed that both lymphomas had an immature T cell phenotype [Thy-1+, CD5+, CD3low, TCR alpha beta low, CD4low, CD8high, heat-stable antigen (HSA) positive, and CD44-, MHC class II-, CD45R-, sIg-, Gr-1-, CD11b-]. Intraperitoneal (i.p.) injection of syngeneic A mice with viable L33 lymphoma cells resulted in a dose-dependent, significant prolongation of the mean survival times of "specific" CBA and MHC-identical B10.BR skin allografts as compared to the survival of appropriate grafts in non-lymphoma-bearing controls. The survival times of third party MHC-incompatible B10 (H-2b) and B10.D2 (H-2d) allografts were only slightly prolonged in A mice inoculated with L33 cells. The graft-protective effect was not abrogated if the proliferative capacity of the L33 cells was blocked by in vitro mitomycin C (MMC) pretreatment. Furthermore, the inoculation of L33 lymphoma into A mice significantly inhibited their DTH response to the sensitizing CBA histocompatibility antigens. In contrast, the L46 lymphoma had no effect on the survival of CBA allografts and the DTH reactivity. These data suggest that the CD4+CD8+TCR alpha beta + L33 T cell lymphoma originating from a neonatally tolerant mouse has a specific immunosuppressive effect on the in vivo reactivity of syngeneic mice to the tolerance-inducing (MHC class I) alloantigens.


Subject(s)
Hypersensitivity, Delayed , Immune Tolerance , Lymphoma/immunology , Major Histocompatibility Complex , T-Lymphocytes/immunology , Animals , Animals, Newborn , CD4 Antigens/immunology , CD8 Antigens/immunology , Cell Division/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Immunophenotyping , Lymphoma/pathology , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Inbred CBA , Mitomycin/pharmacology , Neoplasm Transplantation/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Transplantation, Isogeneic , Tumor Cells, Cultured
17.
Cell Immunol ; 146(2): 431-7, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8174181

ABSTRACT

The possibility, conditions, and quantitative aspects of eliciting GVHD in CBA (H-2k) mice made neonatally tolerant to the alloantigens of the donor A (H-2a) strain were studied. The intravenous injection of different doses (10(7)-2 x 10(8)) of A spleen cells caused a severe, often fatal, systemic GVHD in 12-month-old tolerant mice. The GVHD was found to be specific: spleen cells of a third party strain (B10) did not induce any disease. The intensity and the mortality of the GVHD depended on the cell dose and on the age of the recipients. In contrast, unirradiated (CBA x A)F1 recipients proved to be resistant to the lethal disease. In spite of their different susceptibility to the systemic GVHD, the tolerant and F1 hybrid recipients showed equally strong local GVH reactivity in the popliteal lymph node enlargement assay. Neonatally tolerant mice offer a new, sensitive model for the induction of lethal GVHD without the need of immunosuppression or irradiation.


Subject(s)
Graft vs Host Disease/immunology , Immune Tolerance/physiology , Aging/immunology , Animals , Animals, Newborn/immunology , Cell Transplantation , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Inbred CBA , Spleen/cytology , Transplantation Immunology , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...