Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Blood Rev ; 32(6): 490-498, 2018 11.
Article in English | MEDLINE | ID: mdl-29691090

ABSTRACT

Lymphomas cause significant morbidity and mortality worldwide. A substantial number of patients ultimately relapse after standard treatment. However, the efficacy of these therapies can be counteracted by the patients' immune system, more specifically by myeloid-derived suppressor cells (MDSC). MDSC are a heterogeneous group of immature myeloid cells that suppress the innate and adaptive immune system via different mechanisms and accumulate under pathological conditions, such as cancer. MDSC play a role in the induction and progression of cancer and immune evasion. Increased numbers of MDSC have been reported in different lymphoma subtypes and are associated with a poor clinical outcome. This review aims to clarify the role of MDSC and their working mechanism in different lymphoma subtypes. Furthermore, the effect of MDSC on immunotherapies will be discussed.


Subject(s)
Lymphoma/etiology , Lymphoma/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Adaptive Immunity , Animals , Biomarkers , Combined Modality Therapy , Humans , Immunophenotyping , Immunotherapy , Lymphoma/pathology , Lymphoma/therapy , Molecular Targeted Therapy , Multiple Myeloma/etiology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Tumor Escape
2.
Bone Marrow Transplant ; 47(7): 985-92, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22041852

ABSTRACT

To date, myeloid-derived suppressor cells (MDSC) have been best studied in cancer, where they represent an escape mechanism for immune surveillance. MDSC are now also gaining interest in the context of transplantation. Suppressive CD11b(+) myeloid progenitor cells have been reported to expand endogenously during BM chimerism induction in mice; in particular, in irradiated MHC-matched BM chimeras and in parent-in-F1 BM chimeras. Myeloid cell expansion coincided with a time frame where donor lymphocyte infusion (DLI) therapy-mediated GVL effects without GVHD. Hypothesizing that regulatory myeloid cells may have a role in regulating post-transplant T-cell alloreactivity, we performed a detailed phenotypic and functional characterization of these cells in the parent-in-F1 C57BL/6 → [C57BL/6xDBA2] model. We found that transiently expanding CD11b(+) myeloid progenitor cells comprise the two phenotypically and functionally distinct mononuclear and polymorphonuclear MDSC subsets that were recently described in tumor-bearing mice. Both MDSC subsets suppressed in vitro and in vivo alloreactive T-cell proliferation. Also, both the subsets mediated enhanced in vitro suppression when harvested from chimeras, given a prior in vivo challenge with non-tolerant donor T cells, indicating that allo-activated T cells can activate MDSC in vivo. This study provides the basis to investigate the-potentially beneficial-role of expanding MDSC in influencing the risk of GVHD during chimerism induction.


Subject(s)
Bone Marrow Transplantation/immunology , Graft vs Leukemia Effect/immunology , Myeloid Cells/immunology , Animals , Female , Mice , Mice, Inbred C57BL , Mice, SCID , Myeloid Cells/pathology , Transplantation Chimera/immunology , Transplantation, Homologous
3.
Bone Marrow Transplant ; 46(4): 586-96, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20603621

ABSTRACT

GvHD is an important complication of allogeneic hematopoietic SCT. Parent-in-F1 models are frequently used to study GvHD immunobiology; the characteristics of parent-in-F1 GvHD vary between strain combinations and induction protocols. Here, we observed that a high-dose challenge of non-irradiated B6DBA2F1 and B6SJLF1 recipients with C57BL/6 splenocytes left the majority of recipients clinically healthy, while inducing progressive high-grade donor T-cell chimerism. We investigated this previously undescribed pattern of parent-in-F1 T-cell alloreactivity and studied the effect of serial parental splenocyte infusions on epithelial and lymphohematopoietic tissues. The majority of recipients of 4 weekly splenocyte infusions showed long-term survival with gradual establishment of high-grade donor chimerism and without any signs of epithelial-tissue GvHD. A minority of recipients showed BM failure type of GvHD and, respectively, graft rejection. Moreover, long-term F1 chimeras showed protracted pancytopenia, and in peripheral lymphoid tissues severe lymphopenia and near-complete eradication of APCs and dysfunction in antigen-presenting capacity in remaining APC. Hematopoiesis and lymphoid tissue composition recovered only after multilineage donor chimerism had established. In conclusion, we report on a novel type of parent-in-F1 hybrid GvHD, where a cumulative high dose of C57BL/6 parental splenocytes in non-irradiated F1 mice induces subclinical but severe hematolymphoid-tissue GvHD, causing prolonged immuno-incompetence.


Subject(s)
Cell Transplantation/adverse effects , Graft vs Host Disease/immunology , Lymphoid Tissue/pathology , Spleen/cytology , Animals , Antigen-Presenting Cells/pathology , Chimera , Graft Rejection , Immune System/pathology , Lymphoid Tissue/immunology , Mice , Mice, Inbred C57BL
4.
Leukemia ; 23(2): 340-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18987665

ABSTRACT

The role of graft-versus-malignancy reactivity in the effects of allogeneic hematopoietic stem cell transplantation and donor lymphocyte infusion (DLI) for myelodysplastic syndromes is as yet not well established. Clinical data are limited and animal models are scarce. Here, we report on the effects of allogeneic bone marrow transplantation (alloBMT) and DLI in a novel model of irradiation-induced murine myelodysplastic/myeloproliferation syndrome (MD/MPS). Total body irradiation with 8.5 Gy in SJL/J mice gave rise to a lethal wasting syndrome in 60% of mice, characterized by 1 degrees normocellular bone marrow with dysplastic features in erythroid, myeloid and megakaryocytic cell lineages, 2 degrees lymphosplenomegaly with spleens harboring a prominent extramedullary hematopoiesis with erythroid, myeloid and megakaryocytic lineages exhibiting dysplastic features, and foci of dysplastic hematomyelopoiesis in the liver, 3 degrees peripheral thrombocytopenia and 4 degrees evidence of disseminated infection or leukemic transformation in selected animals. This clinicopathological picture was consistent with a murine form of MD/MPS. Syngeneic or allogeneic (BALB/c) T cell-depleted BMT could not prevent the occurrence of lethal MD/MPS. In contrast, DLI at weeks 2-4 after BMT led to restoration of the dysbalanced hematomyelopoiesis. However, severe DLI-induced acute graft-versus-host disease occurred, precluding a survival advantage. We present evidence of the existence of a post-alloBMT DLI-induced graft-versus-MD/MPS effect in murine irradiation-induced MD/MPS.


Subject(s)
Bone Marrow Transplantation , Graft vs Leukemia Effect , Lymphocyte Transfusion , Myelodysplastic Syndromes/therapy , Animals , Disease Models, Animal , Mice , Myeloproliferative Disorders/therapy , Transplantation, Homologous , Treatment Outcome , Whole-Body Irradiation
5.
Leukemia ; 21(7): 1451-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17508005

ABSTRACT

We studied the effect of CTLA-4 blockade on graft-versus-leukemia and graft-versus-host responses in a mouse model of minor histocompatibility-mismatched bone marrow transplantation. Early CTLA-4 blockade induced acute graft-versus-host disease. Delayed CTLA-4 blockade resulted in a lethal condition with lymphosplenomegaly, but with stable mixed T-cell chimerism, unchanged alloreactive T-cell frequencies and absent anti-host reactivity in vitro. In contrast, multiorgan lymphoproliferative disease with autoimmune hepatitis and circulating anti-DNA auto-antibodies were documented. Splenic lymphocytes exhibited ex vivo spontaneous proliferation and a marked proliferative response against host-type dendritic cells pulsed with syngeneic (host-type) tissue-peptides. Both phenomena were exclusively mediated by host and not donor T cells, supporting an autoimmune pathogenesis. Selectively host-derived T-cell immune reactivity was equally documented against leukemia-peptide-pulsed dendritic cells, and this was paralleled by a strong in vivo antileukemic effect in anti-CTLA-4-treated and subsequently leukemia-challenged chimeras. In conclusion, delayed CTLA-4 blockade induced a host-derived antileukemic effect, occurring in the context of an autoimmune syndrome and strictly separated from graft-versus-host disease. Both antileukemic and autoimmune responses depended on the allogeneic component, as neither effect was seen after syngeneic bone marrow transplantation. Our findings reveal the potential of using CTLA-4 blockade to establish antileukemic effects after allogeneic hematopoietic stem cell transplantation, provided autoimmunity can be controlled.


Subject(s)
Antigens, CD/drug effects , Antigens, Differentiation/drug effects , Bone Marrow Transplantation , Graft vs Leukemia Effect , Transplantation Chimera , Animals , Antigens, CD/immunology , Antigens, Differentiation/immunology , Autoimmunity , CTLA-4 Antigen , Graft vs Host Disease , Histocompatibility , Leukemia/therapy , Mice , T-Lymphocytes/immunology , Treatment Outcome
6.
Transplant Proc ; 37(1): 516-7, 2005.
Article in English | MEDLINE | ID: mdl-15808695

ABSTRACT

UNLABELLED: To further study the interactions between innate and adaptive immunity in xenotransplantation, we explored the relative contribution of T-cell subsets in vascularized (heart) and cellular (islets) xenografts in a model with established xeno-non-reactivity of the innate system. MATERIALS: Specific innate xenotolerance was induced in xenoheart (hamster) recipients (nude rats) by a tolerizing regimen (TR), consisting of donor antigen infusion, temporary natural killer (NK)-cell depletion and a 4-week administration of leflunomide. Hamster pancreatic islets were transplanted either 1 week after heart transplantation or alone and syngeneic T-cell adoptive transfer was performed 10 days later. Purified CD3(+), CD4(+), and CD8(+) T cells were given 2 weeks after withdrawal of all drugs. At the day of rejection, xenografts were removed for histology. Serum was taken and IgM and IgG xenoantibody titers were measured by flow cytometry. RESULTS: Both heart and islet grafts were rejected after CD4(+) reconstitution. After CD8(+) T-cell adoptive transfer, cellular grafts were not rejected but vascularized grafts were rejected, although only after several months. Rejection in CD4(+) reconstituted nude rats was accompanied by the generation of predominantly IgG xenoantibodies. CONCLUSION: CD4(+) T lymphocytes are able to rapidly initiate the rejection of islet xenografts in the presence of a xenotolerant innate immune system either by breaking the "innate tolerance" (e.g., by activating macrophages and NK-cells) or through a mechanism without any involvement of the innate tolerance (e.g., T-dependent IgG antibody production). In contrast, CD8(+) T cells provoke a late rejection of only xenoheart grafts.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Heart Transplantation/immunology , Islets of Langerhans Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Cricetinae , Killer Cells, Natural/immunology , Lymphocyte Depletion , Rats
11.
Transplantation ; 71(2): 292-9, 2001 Jan 27.
Article in English | MEDLINE | ID: mdl-11213076

ABSTRACT

BACKGROUND: We have previously shown that allogeneic bone marrow (BM) chimeras preconditioned with total lymphoid irradiation and low-dose total body irradiation (TLI/TBI) develop a stronger graft-versus-leukemia (GVL) effect than chimeras preconditioned with high-dose total body irradiation only (TBI). Here, we report on the possible role of cytokines in the mechanism underlying this GVL effect. METHODS: Splenic mRNA levels of the cytokines interleukin (IL)-1, IL-2, IL-4, IL-6, IL-7, IL-10, IL-12, IL-15, interferon-gamma, tumor necrosis factor-alpha, and transforming growth factor-beta (TGF-beta), and of inducible nitric oxide synthetase were determined by reverse transcription-polymerase chain reaction in TLI/TBI- or TBI-conditioned C3H/AKR BM chimeras challenged with AKR-type BW5147.3 leukemia cells. Ex vivo TGF-beta protein production by splenocytes was determined using ELISA. The possibility that cytokines influence the GVL effect by modulating the activity of IL-2-activated lymphocytes (LAK cells) was investigated by in vitro assays on donor-type BM cells. RESULTS: Of all cytokine mRNA levels studied, those of TGF-beta and IL-7 were different between groups; both were significantly more elevated in TBI- than in TLI/ TBI-conditioned or normal mice. Differences were apparent after conditioning and were not influenced by additionally injected BM or leukemia cells. Cultured splenocytes of TBI-conditioned animals produced significantly more TGF-beta protein than those of TLI/TBI-conditioned ones or normal controls. r-TGF-beta but not r-IL-7 suppressed in vitro LAK activity of donor-type BM cells against BW5147.3 cells in a dose-dependent way. CONCLUSIONS: High-dose TBI-induced, host-derived splenic TGF-beta may inhibit generation of LAK cells from subsequently transplanted donor BM cells, suppressing their capacity to generate cytotoxicity upon injection of leukemia cells. The cytokine profile, induced by irradiation in host hematopoietic organs, can significantly modify posttransplant immunological processes such as the GVL effect and graft-versus-host disease (GVHD).


Subject(s)
Bone Marrow Cells/cytology , Cytotoxicity, Immunologic/drug effects , Killer Cells, Lymphokine-Activated/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Bone Marrow Cells/radiation effects , Cytokines/genetics , Graft vs Leukemia Effect/radiation effects , Interleukin-7/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Leukemia/immunology , Leukemia/pathology , Lymphotoxin-alpha/blood , Lymphotoxin-alpha/pharmacology , Mice , Mice, Inbred AKR , Mice, Inbred C3H , RNA, Messenger/metabolism , Radiation Chimera , Spleen/chemistry , Whole-Body Irradiation
12.
J Immunol ; 166(3): 1843-54, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11160231

ABSTRACT

Successful grafting of vascularized xenografts (Xgs) depends on the ability to reliably induce both T cell-independent and -dependent immune tolerance. After temporary NK cell depletion, B cell suppression, and pretransplant infusion of donor Ags, athymic rats simultaneously transplanted with hamster heart and thymus Xgs developed immunocompetent rat-derived T cells that tolerated the hamster Xgs but provoked multiple-organ autoimmunity. The autoimmune syndrome was probably due to an insufficient development of tolerance for some rat organs; for example, it led to thyroiditis in the recipient rat thyroid, but not in simultaneously transplanted donor hamster thyroid. Moreover, grafting a mixed hamster/rat thymic epithelial cell graft could prevent the autoimmune syndrome. These experiments indicate that host-type thymic epithelial cells may be essential for the establishment of complete self-tolerance and that mixed host/donor thymus grafts may induce T cell xenotolerance while maintaining self-tolerance in the recipient.


Subject(s)
Autoimmune Diseases/immunology , Thymus Gland/transplantation , Transplantation Tolerance , Transplantation, Heterologous/immunology , Animals , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , Cricetinae , Epithelial Cells/immunology , Epithelial Cells/pathology , Fetus/immunology , Graft Survival/genetics , Graft Survival/immunology , Heart Transplantation/immunology , Heart Transplantation/pathology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Lymphocyte Activation , Male , Rats , Rats, Inbred Strains , Rats, Nude , Species Specificity , T-Lymphocytes/immunology , Thymus Gland/embryology , Thymus Gland/pathology , Transplantation Tolerance/genetics , Transplantation, Heterologous/pathology , Transplantation, Homologous
13.
Diabetes ; 49(8): 1301-7, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10923629

ABSTRACT

Prevention of type 1 diabetes in NOD mice by 1,25-dihydroxyvitamin D3 [1alpha,25(OH)2D3] is accompanied by a T-helper (Th) 1/Th2 cytokine shift in the pancreas. The aim of this study was to investigate whether this immune shift also occurs outside of the pancreas and whether it is limited to autoantigen-specific immune responses. NOD mice treated with 1alpha,25(OH)2D3 (5 microg/kg every 2 days) or control vehicle were immunized with GAD65 (p524-543) or ovalbumin (OVA) in the rear footpads. First, we examined T-cell proliferation and cytokine production (via enzyme-linked immunosorbent assay) of draining lymph node cells in vitro with or without peptide rechallenge. Although no differences in proliferation were measured between control and 1alpha,25(OH)2D3-treated mice after in vitro GAD65 rechallenge, a marked shift in cytokine secretion profile was seen in 1alpha,25(OH)2D3-treated mice: interleukin-4 was increased (37 +/- 5 vs. 21 +/- 12 pg/ml in controls, P < 0.005), whereas gamma-interferon levels were decreased (6 +/- 3 vs. 9 +/- 3 ng/ml in controls, P < 0.05). This shift was absent in OVA-primed mice. Second, we measured cytokine profiles by reverse transcriptase-polymerase chain reaction in popliteal lymph nodes at different time points after priming with GAD65 or OVA in vivo. A marked Th1/Th2 shift occurred in 1alpha,25(OH)2D3-treated mice after in vivo priming with GAD65. Again, this shift was absent after OVA immunization. Finally, we measured cytokine profiles after rechallenge with a panel of autoantigens (GAD65, heat shock protein 65, insulin B-chain) and control antigens (OVA, keyhole limpet hemocyanine, myelin proteolipid protein, tetanus toxin) and confirmed the Th1/Th2 shift in autoantigen-injected mice but not in control antigen-injected mice. In conclusion, the immune deviation induced by 1alpha,25(OH)2D3 in NOD mice can also be induced in the peripheral immune system but is limited to pancreatic autoantigens.


Subject(s)
Autoantigens/immunology , Bacterial Proteins , Calcitriol/pharmacology , Cytokines/genetics , Glutamate Decarboxylase/immunology , Isoenzymes/immunology , Lymphocyte Activation , Peptide Fragments/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Amino Acid Sequence , Animals , Autoantigens/chemistry , Chaperonin 60 , Chaperonins/immunology , Diabetes Mellitus, Type 1/immunology , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/chemistry , Insulin/immunology , Interferon-gamma/genetics , Interleukin-4/genetics , Isoenzymes/chemistry , Lymph Nodes/immunology , Mice , Mice, Inbred NOD , Molecular Sequence Data , Peptide Fragments/chemistry , Th1 Cells/drug effects , Th2 Cells/drug effects , Transcription, Genetic
14.
Transplantation ; 70(1): 86-93, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10919580

ABSTRACT

BACKGROUND: The role of natural killer (NK) cells and macrophages (Møs) in the absence of T cell immunity in discordant xenograft (xg) rejection was investigated. METHODS: Guinea pig hearts were transplanted into athymic nude rats receiving no treatment or antixenoantibody (xAb), anticomplement (C), antinatural killer (NK) cell therapy (antiasialo GM1 [anti-ASGM1]) or their combinations. For anti-xAb therapy, natural xAbs were absorbed/neutralized by pretransplant guinea pig blood transfusion (pGPBT), followed by administration of the malononitriloamide MNA715. Cobra venom factor (CVF) was administered as anti-C therapy. FACScan analysis and a standard cytotoxicity assay determined NK cell number and cytotoxicity, respectively. ELISA and the CH50 assay measured titers of xAb and C activity, respectively. Rejected Xgs were examined by light microscopy and by immunohistochemistry. RESULTS: All hyperacutely (15+/-4 min) rejected Xgs from untreated rats showed deposits of C3 and IgM without cellular infiltrates. Combined anti-xAb and anti-C (pGPBT/MNA715/CVF) treatment significantly prolonged the survival of Xgs (3.7+/-0.6 days, P<0.001 vs. control group) showing NK cell and Mø infiltration without deposition of xAbs or C3. NK cell depletion (day -12) followed by exposure of recovering NK cells to the guinea pig antigens failed to induce specific NK cell nonresponsiveness and to further prolong xg survival in combined anti-xAb/anti-C group. In contrast, adding of continuous and repetitive depletion of NK cells significantly further prolonged xg survival to 7.4+/-0.5 days (P<0,001 vs. the anti-xAb/anti-C group) with rejected Xgs densely infiltrated by activated Møs without involvement of NK cells, C or xAbs. CONCLUSIONS: When xAb and C are suppressed in the discordant guinea pig-into-nude rat model, NK cells play an important role in xg rejection. When also NK cells are suppressed, activated Møs seem to reject discordant Xgs. The induction of specific NK cell nonresponsiveness fails in the guinea pig-into-rat combination.


Subject(s)
Antibodies, Heterophile/physiology , Complement System Proteins/physiology , Graft Rejection , Killer Cells, Natural/physiology , Macrophages/physiology , T-Lymphocytes/immunology , Transplantation, Heterologous/immunology , Animals , Guinea Pigs , Heart Transplantation/immunology , Immunohistochemistry , Male , Rats , Rats, Nude
15.
Transplantation ; 69(8): 1695-704, 2000 Apr 27.
Article in English | MEDLINE | ID: mdl-10836383

ABSTRACT

BACKGROUND: Pretransplant blood transfusions have beneficial effects on both clinical and experimental allograft survival. In the present study, we examined whether pretransplant hamster blood transfusions (pHBT) alone or together with peritransfusion immunosuppressive strategies designed to target B cells and/or natural killer (NK) cells, could modulate T cell-independent (T-I) xenoreactivity in athymic nude rats. METHODS: Hamster or mouse hearts were heterotopically xenotransplanted into untreated or treated athymic nude rats receiving either pHBT, anti-B cell or anti-NK cell therapy alone or their combinations. Xenoreactive antibodies (xAbs) and the percentage of NK cells were analyzed by FACScan analysis. NK cytotoxicity was measured by a standard 4 hr 51Cr release assay. Xenografts (Xgs) were examined by hematoxylin-eosin (H&E), by light microscopic method with Masson's trichrome and orcein staining, by immunofluorescent staining for immunoglobulin M and C3 deposition, and by immunohistochemical staining for infiltration of NK cells and macrophages (Mphis). RESULTS: In 1 of 6 rats given pHBT alone 2 weeks before receiving hamster xenografts, Xg survival was prolonged to 55 days compared with 3.0+/-1.2 days in the other 5 animals and with 3.0+/-0.6 days in untreated animals. In the 55 days, surviving Xg infiltration of Mphis and NK cells was seen together with severe signs of chronic rejection, such as fibrosis and obliterative vasculopathy. The addition of the anti-B cell immunosuppressant MNA715 (malononitriloamide x920715, 20 mg/kg/day) from day -14 to day +14 or of 100 microL of rabbit anti-asialo GM1 serum ([anti-ASGM1] an NK cell depleting antibody) on day -14 resulted in a significant and species-specific prolongation of the survival of hamster Xgs, respectively 59.8+/-9.6 days and 58.2+/-14.7 days (P<0.001 vs. control group), but not of mouse heart Xgs that were rejected in a normal tempo. All prolonged hamster Xgs were infiltrated with Mphis and NK cells and developed severe lesions of chronic rejection, such as fibrosis and obliterative vasculopathy. In contrast, MNA715 or anti-ASGM1 alone had no effect on Xg survival (4.8+/-1.7 days and 2.7+/-0.6 days, respectively). Combined MNA715/anti-ASGM1 treatment only moderately promoted Xg survival (10+/-5.0 days; P<0.001). A simultaneous administration of pHBT, MNA715, and anti-ASGM1 induced indefinite and species-specific Xg survival in all recipients. In vivo and in vitro studies demonstrated that both T-I B cell and NK cell species-specific xenotolerance were achieved. CONCLUSIONS: Pretransplant blood transfusion may have a species-specific immunomodulatory effect on T-I xenoreactivity. This effect is further enhanced by a temporary co-administration of MNA715 or by a single injection of anti-ASGM1. A combination of pHBT, MNA715, and anti-ASGM1 induces species-specific T-I xenotolerance.


Subject(s)
Blood Donors , Blood Transfusion , Immunity , Preoperative Care , T-Lymphocytes/physiology , Transplantation, Heterologous/immunology , Animals , Antibody Formation , Cricetinae , Graft Survival , Heart Transplantation/immunology , Killer Cells, Natural/physiology , Male , Mesocricetus/blood , Mice , Mice, Inbred BALB C , Rats , Rats, Nude , Species Specificity , Transplantation, Heterotopic
16.
Clin Exp Immunol ; 120(1): 139-46, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10759775

ABSTRACT

Receptors for 1,25(OH)2vitaminD3 are found in most immune cells and important immunological effects have been described in vitro, reflected by its capacity to prevent autoimmunity and to prolong graft survival. The aim of this study was to examine the presence and nature of the enzyme responsible for final activation of the molecule, 1-alpha-hydroxylase, in murine macrophages and to analyse its regulation and possible role in the immune system. Peritoneal macrophages from C57Bl/6 mice were incubated with lipopolysaccharide (LPS; 100 microg/ml), interferon-gamma (IFN-gamma; 500 U/ml) or a combination of both. By quantitative reverse transcriptase-polymerase chain reaction, using primers based on the murine renal cDNA sequence, low levels of 1-alpha-hydroxylase mRNA were detected in freshly isolated cells (18 +/- 7 x 10-6 copies/beta-actin copies). Analysis of the cDNA sequence of the gene revealed identical coding sequences for the macrophage and renal enzymes. mRNA levels rose three-fold with LPS (NS), but a six-fold increase was seen after IFN-gamma stimulation (P < 0.05). Combining LPS and IFN-gamma did not result in a major additional increase, but addition of cyclosporin A further increased levels 2.5-fold both in IFN-gamma- and combination-stimulated cells (P < 0.05). Time course analysis revealed that up-regulation of 1-alpha-hydroxylase was a late phenomenon, preceded by the up-regulation of activating macrophage products such as IL-1 and tumour necrosis factor-alpha. Finally, a defect in 1-alpha-hydroxylase up-regulation by immune stimuli was found in autoimmune non-obese diabetic mice. In conclusion, we propose that the up-regulation of 1-alpha-hydroxylase in activated macrophages, resulting in the synthesis of 1,25(OH)2D3, might be a negative feedback loop in inflammation. A defect in this system might be an additional element in tipping the balance towards autoimmunity.


Subject(s)
Macrophages, Peritoneal/enzymology , Steroid Hydroxylases/metabolism , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/genetics , Cell Line , Cholestanetriol 26-Monooxygenase , Cloning, Molecular , Cyclosporine/pharmacology , Cytokines/genetics , Female , Kidney/cytology , Kidney/enzymology , Kidney/immunology , Leukemia P388 , Macrophage Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Sequence Analysis, DNA , Steroid Hydroxylases/biosynthesis , Steroid Hydroxylases/genetics , Time Factors , Transcription, Genetic/immunology , Tumor Cells, Cultured , Up-Regulation/drug effects , Up-Regulation/immunology
18.
Transplantation ; 68(10): 1560-7, 1999 Nov 27.
Article in English | MEDLINE | ID: mdl-10589955

ABSTRACT

BACKGROUND: It has previously been demonstrated that adding T cell-depleted (TCD) host bone marrow (BM) to an MHC-mismatched BM inoculum allows for induction of long-term stable chimeras without graft-versus-host disease (GVHD) even when non-TCD allogeneic BM was used. AIMS: The present study was undertaken to investigate immune tolerance mechanisms in minor antigen-mismatched allogeneic BM chimeras when host-type BM was added to the BM inoculum. METHODS: C3H (H2k, Thy 1.2, Mls 2a) recipients were conditioned with 9.5 gray (Gy) of total body irradiation. To exclude any interference with possible subclinical GVHD, 5x10(6) TCD AKR (H2k, Thy 1.1, Mls 1a) BM cells were injected with (syn + allo) or without (allo) 5x 10(6) TCD C3H BM cells. Chimerism, clonal deletion, and T lymphocyte subsets were scored using FACS and anti-mouse Thy, Vbeta6, Vbeta3, CD3, CD4, or CD8 monoclonal antibodies. The stability of tolerance was studied by investigating mixed lymphocyte reaction and cytotoxic T cell induction in chimeras after immunization with host, donor, or third-party (BALB/c) splenocytes. Breaking of chimerism was attempted by injecting nontolerant 40x10(6) host-type splenocytes 2 months after BM transplantation. Cytokines and Valpha14 mRNA were assayed using real time quantitative reverse transcriptase-polymerase chain reaction at 4 and 48 hr, respectively, after injection of nontolerant host-type splenocytes. RESULTS: Both groups of mice became long-term stable mixed chimeras without any clinical sign of GVHD. Neither group was able to produce antihost nor antidonor cytotoxic T cells, even after immunization. The addition of syngeneic BM to the allogeneic inoculum reduced the overall level of allogeneic chimerism (from approximately 70% or approximately 85% in peripheral blood lymphocytes and spleen, respectively, in allo chimeras versus approximately 35% and approximately 60% in syn + allo chimeras). Moreover, it resulted in complete clonal deletion of both host-reactive (Vbeta3) and donor-reactive (Vbeta6) lymphocytes in syn + allo chimeras in contrast to in allo chimeras, in which only donor-reactive lymphocytes were completely deleted. After nontolerant C3H splenocyte injection, high levels of interleukin 2 mRNA were produced and chimerism decreased in syn + allo chimeras. In contrast, in allo chimeras, this maneuver was followed by the production of higher levels of interleukin 4 and interferon-gamma, and of Valpha14 mRNA, as well as by the proliferation of CD3+CD4-CD8- (double-negative) T cells and by an increase of donor chimerism. CONCLUSION: The addition of host-type BM to the allogeneic inoculum has an influence on the level of chimerism, the extent of clonal deletion, and the reaction of chimeras after the injection of nontolerant host-type splenocytes. In the latter phenomenon, cytokine production and proliferation of Valpha14+ CD3+CD4-CD8- (double-negative, natural killer T) lymphocytes may be involved.


Subject(s)
Bone Marrow Transplantation/immunology , Immunosuppression Therapy/methods , Killer Cells, Natural/immunology , Minor Histocompatibility Antigens/immunology , Transplantation Chimera , Animals , Antigens, Differentiation, T-Lymphocyte/immunology , Bone Marrow Transplantation/methods , Female , Flow Cytometry , Graft vs Host Disease/immunology , Histocompatibility Testing , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Transplantation, Homologous , Whole-Body Irradiation
19.
Transplantation ; 68(8): 1181-8, 1999 Oct 27.
Article in English | MEDLINE | ID: mdl-10551648

ABSTRACT

BACKGROUND: We previously described that a tolerogeneic regimen (TR) including (1) the infusion of a minced hamster heart suspension (MHH), (2) a single injection of an anti-natural killer (NK) cell serum (rabbit anti-asialo GM1 serum), and (3) a 4-week course of the B cell immunosuppressant leflunomide (20 mg/kg/ day) induced T cell-independent (T-I) B lymphocyte and NK cell tolerance for hamster xenoantigens in T-deficient athymic nude rats. In addition, the TR allowed for long-term hamster cardiac xenograft (Xg) survival when Xgs were transplanted 2 weeks (Day 0) after the initiation of the TR (started on Day - 14). The present study was undertaken to investigate some of the characteristics of this T-I xenotolerance in more detail. METHODS: To investigate the duration of the effect of the TR on the T-I xenotolerance, hamster Xgs were transplanted at various times after initiation of the TR. To investigate whether the maintenance of the T-I xenotolerance depended on the presence of the graft, tolerated Xgs were removed on Day +28, and the subsequent evolution of the T-I xenotolerance as well as of second hamster Xg was followed. In addition, the reversibility of NK cell nonresponsiveness by recombinant interleukin-2 was investigated in vitro. RESULTS: Xgs transplanted on day 0 or Day +7 showed long-term survival. However, all Xgs transplanted on Day +15, +30, and +60 were rapidly rejected. The latter rejection occurred in the absence of formation of anti-hamster immunoglobulin (Ig)M xenoreactive antibodies (xAbs) but correlated with the recovery of anti-hamster NK cell reactivity from day +14 on. Rejected Xgs showed infiltration of NK cells but absence of IgM xAbs or complement factor deposition. When tolerated first Xgs (transplanted on Day 0) were removed on Day +28, second hamster Xgs survived without treatment when transplanted 1 or 2 weeks later. However, second hamster Xgs transplanted 3 weeks after removal of the first Xgs were all rapidly rejected. Again, the latter rejection was characterized by the infiltration of the Xgs with NK cells and by the absence of anti-hamster IgM xAbs formation. Xenoreactive NK cell nonresponsiveness was not only shorter than xenoreactive B cell nonresponsiveness, but was also more fragile. This was evident from the fact that after addition of recombinant interleukin-2 in vitro, specific anti-hamster NK nonresponsiveness was easily broken. CONCLUSIONS: NK cell and T-I B cell xenotolerance can be induced in T-deficient rats. Compared with B cell xenotolerance, the maintenance of NK cell xenotolerance is much shorter, more dependent on the presence of the graft, and easily reversible in vitro.


Subject(s)
Antigens, Heterophile/immunology , B-Lymphocytes/physiology , Heart Transplantation/immunology , Immune Tolerance/physiology , Killer Cells, Natural/physiology , T-Lymphocytes/physiology , Transplantation Conditioning/methods , Animals , B-Lymphocytes/drug effects , Cricetinae , Graft Rejection/pathology , Graft Survival , Immune Tolerance/drug effects , Immunosuppressive Agents/pharmacology , Isoxazoles/pharmacology , Leflunomide , Mesocricetus , Mice , Mice, Inbred BALB C , Myocardium/pathology , Rats , Rats, Nude , Time Factors
20.
Transplantation ; 68(1): 130-6, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10428280

ABSTRACT

BACKGROUND: We have previously demonstrated that in a concordant hamster-to-nude rat cardiac transplant model, T-independent specific B-lymphocyte and natural killer (NK)-cell tolerance could be induced, leading to long-term xenograft (Xg) survival. Here, we investigated whether the same could be achieved in a clinically more relevant semi-discordant model involving hamster hearts transplanted into pre-sensitized, nude rats. METHODS: Sensitized, nude rats with high titers of anti-hamster immunoglobulin (Ig)M xenoantibodies (XAbs) were prepared by transplanting a first hamster heart without treatment. One week after rejection, a complete tolerizing regimen was given, including the following: a) an i.v. injection of hamster heart antigens; b) a 4-week administration of malononitriloamide; and c) a single injection of an anti-NK antiserum. Two weeks later, a second hamster heart was grafted. The isotype and level of XAb were examined by fluorescence-activated cell sorting. NK cytotoxicity was evaluated by a standard 4-hr 51Cr release assay. Hamster heart Xgs were examined by conventional histologic and immunohistochemical analysis. RESULTS: Untreated, presensitized, nude rats developing high titers of IgM XAb underwent hyperacute rejection within 4 hr (n=4) after transplantation of the second hamster heart. Immunohistochemical analysis showed intensive staining for IgM and C3 along the vascular endothelia in the rejected Xgs. In contrast, presensitized, nude rats receiving the complete tolerizing regimen had a rapid decrease in anti-hamster IgM XAb. The second hamster hearts were not rejected and showed long-term survival even after withdrawal of malononitriloamide (n=6). Moreover, tolerant rats showed specific B-lymphocyte tolerance and a specific continuous absence of anti-hamster NK-cell reactivity. CONCLUSION: T-independent B-lymphocyte and NK-cell xenotolerance can also be achieved in recipients with pre-existing IgM XAb.


Subject(s)
Heart Transplantation/immunology , Transplantation, Heterologous/immunology , Transplantation, Heterologous/pathology , Animals , Cricetinae , Graft Rejection/pathology , Immune Tolerance , Immunohistochemistry , Killer Cells, Natural/immunology , Male , Mesocricetus , Mice , Mice, Inbred BALB C , Models, Cardiovascular , Rats , Rats, Nude , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...