Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 12: 746469, 2021.
Article in English | MEDLINE | ID: mdl-34777358

ABSTRACT

Classical dendritic cells (cDCs) in mice have been divided into 2 major subsets based on the expression of nuclear transcription factors: a CD8+Irf8+Batf3 dependent (DC1) subset, and a CD8-Irf4+ (DC2) subset. We found that the CD8+DC1 subset can be further divided into CD8+DC1a and CD8+DC1b subsets by differences in surface receptors, gene expression, and function. Whereas all 3 DC subsets can act alone to induce potent Th1 cytokine responses to class I and II MHC restricted peptides derived from ovalbumin (OVA) by OT-I and OT-II transgenic T cells, only the DC1b subset could effectively present glycolipid antigens to natural killer T (NKT) cells. Vaccination with OVA protein pulsed DC1b and DC2 cells were more effective in reducing the growth of the B16-OVA melanoma as compared to pulsed DC1a cells in wild type mice. In conclusion, the Batf3-/- dependent DC1 cells can be further divided into two subsets with different immune functional profiles in vitro and in vivo.


Subject(s)
CD8 Antigens/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Transcriptome/immunology , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype
2.
Blood Adv ; 5(17): 3290-3302, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34432869

ABSTRACT

Replacement of failed organs followed by safe withdrawal of immunosuppressive drugs has long been the goal of organ transplantation. We studied changes in the balance of T cells and myeloid cells in the blood of HLA-matched and -mismatched patients given living donor kidney transplants followed by total lymphoid irradiation, anti-thymocyte globulin conditioning, and donor hematopoietic cell transplant to induce mixed chimerism and immune tolerance. The clinical trials were based on a conditioning regimen used to establish mixed chimerism and tolerance in mice. In preclinical murine studies, there was a profound depletion of T cells and an increase in immunosuppressive polymorphonuclear (pmn) myeloid-derived suppressor cells (MDSCs) in the spleen and blood following transplant. Selective depletion of pmn MDSCs in mice abrogated mixed chimerism and tolerance. In our clinical trials, patients given an analogous tolerance conditioning regimen developed similar changes, including profound depletion of T cells and a marked increase in MDSCs in blood posttransplant. Posttransplant pmn MDSCs transiently increased expression of lectin-type oxidized LDL receptor-1, a marker of immunosuppression, and production of the T-cell inhibitor arginase-1. These posttransplant pmn MDSCs suppressed the activation, proliferation, and inflammatory cytokine secretion of autologous T-cell receptor microbead-stimulated pretransplant T cells when cocultured in vitro. In conclusion, we elucidated changes in receptors and function of immunosuppressive myeloid cells in patients enrolled in the tolerance protocol that were nearly identical to those of MDSCs required for tolerance in mice. These trials were registered at www.clinicaltrials.gov as #NCT00319657 and #NCT01165762.


Subject(s)
Hematopoietic Stem Cell Transplantation , Animals , Clinical Trials as Topic , Humans , Immune Tolerance , Mice , Myeloid Cells , Transplant Recipients , Transplantation Conditioning
3.
Blood ; 129(12): 1718-1728, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28096089

ABSTRACT

The combination of total lymphoid irradiation and anti-T-cell antibodies safely induces immune tolerance to combined hematopoietic cell and organ allografts in humans. Our mouse model required host natural killer T (NKT) cells to induce tolerance. Because NKT cells normally depend on signals from CD8+ dendritic cells (DCs) for their activation, we used the mouse model to test the hypothesis that, after lymphoid irradiation, host CD8+ DCs play a requisite role in tolerance induction through interactions with NKT cells. Selective deficiency of either CD8+ DCs or NKT cells abrogated chimerism and organ graft acceptance. After radiation, the CD8+ DCs increased expression of surface molecules required for NKT and apoptotic cell interactions and developed suppressive immune functions, including production of indoleamine 2,3-deoxygenase. Injection of naive mice with apoptotic spleen cells generated by irradiation led to DC changes similar to those induced by lymphoid radiation, suggesting that apoptotic body ingestion by CD8+ DCs initiates tolerance induction. Tolerogenic CD8+ DCs induced the development of tolerogenic NKT cells with a marked T helper 2 cell bias that, in turn, regulated the differentiation of the DCs and suppressed rejection of the transplants. Thus, reciprocal interactions between CD8+ DCs and invariant NKT cells are required for tolerance induction in this system that was translated into a successful clinical protocol.


Subject(s)
Dendritic Cells/immunology , Graft Rejection/prevention & control , Immune Tolerance , Natural Killer T-Cells/immunology , Animals , Bone Marrow Transplantation , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Graft Rejection/immunology , Heart Transplantation , Mice
4.
Blood ; 119(6): 1581-9, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22174155

ABSTRACT

We used a model of combined bone marrow and heart transplantation, in which tolerance and stable chimerism is induced after conditioning with fractionated irradiation of the lymphoid tissues and anti-T-cell antibodies. Graft acceptance and chimerism required host CD4(+)CD25(+) Treg production of IL-10 that was in-turn enhanced by host invariant natural killer (NK) T-cell production of IL-4. Up-regulation of PD-1 on host Tregs, CD4(+)CD25(-) conventional T (Tcon) cells, and CD8(+) T cells was also enhanced by NKT cell production of IL-4. Up-regulated PD-1 expression on Tregs was linked to IL-10 secretion, on CD8(+) T cells was linked to Tim-3 expression, and on CD4(+) Tcon cells was associated with reduced IFNγ secretion. Changes in the expression of PD-1 were induced by the conditioning regimen, and declined after bone marrow transplantation. In conclusion, NKT cells in this model promoted changes in expression of negative costimulatory receptors and anti-inflammatory cytokines by Tregs and other T-cell subsets in an IL-4-dependent manner that resulted in tolerance to the bone marrow and organ grafts.


Subject(s)
Bone Marrow Transplantation/immunology , Heart Transplantation/immunology , Immune Tolerance/immunology , Natural Killer T-Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , Antilymphocyte Serum/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Cells, Cultured , Female , Fluorescent Antibody Technique , Hepatitis A Virus Cellular Receptor 2 , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , Programmed Cell Death 1 Receptor , Receptors, Virus/immunology , Receptors, Virus/metabolism , T-Lymphocytes, Regulatory/metabolism , Transplantation Conditioning/methods , Up-Regulation/drug effects
5.
J Infect Dis ; 196(6): 900-10, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17703422

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) infection depletes thymocytes and destroys thymic structure. Functional, tolerant human T cells develop in vivo in immunodeficient mice receiving porcine thymus and human fetal liver fragments under the kidney capsule. In this model, we evaluated the potential of porcine thymus to protect human thymocytes from the effects of HIV-1. Compared with that observed in control mice with human thymic grafts, porcine thymus attenuated human thymocyte depletion by the CCR5-tropic isolate JR-CSF without preventing thymocyte infection. Porcine thymus protected human thymocytes from infection and depletion by a CXCR4-tropic HIV-1 isolate without reducing peripheral blood viral loads or T cell infection. Human thymocytes from human but not porcine grafts showed decreased Bcl-2 expression and increased apoptosis after NL4.3 infection. Thus, porcine thymus protects human thymocytes from the cytopathic effect of HIV-1, suggesting a possible approach to achieving immune restoration in patients with acquired immunodeficiency syndrome who have incomplete responses to antiretroviral therapy. The model allows analysis of the mechanisms of HIV-mediated thymic dysfunction.


Subject(s)
Fetal Tissue Transplantation , HIV Infections/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/transplantation , Transplantation, Heterologous , Animals , Antigens, CD34/analysis , Apoptosis , Cytopathogenic Effect, Viral , Dendritic Cells/immunology , Dendritic Cells/virology , Fetus/cytology , Gene Expression , HIV Infections/pathology , Humans , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred NOD , Mice, SCID , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Swine , Swine, Miniature , T-Lymphocytes/virology , Thymus Gland/embryology , Thymus Gland/pathology , Viral Load
6.
J Immunol ; 173(3): 1744-56, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15265904

ABSTRACT

The pathogenesis of idiopathic pneumonia syndrome (IPS), a noninfectious pulmonary complication of allogeneic bone marrow transplantation (BMT), has not been fully elucidated. However, several contributing factors have been proposed, including lung injury caused by reactive oxygen and nitrogen intermediates during preconditioning and development of graft-vs-host disease (GVHD). Studies on the role of reactive oxygen and nitrogen intermediates in IPS have yielded conflicting results. We have described a murine model of IPS, in which the onset of lung inflammation was delayed by several weeks relative to GVHD. This study evaluated whether the delay in onset of IPS was due to slow turnover of NO-producing, immunosuppressive alveolar macrophages (AM) following BMT. The results indicated that AM were immunosuppressive due to synthesis of NO. However, NO production and immunosuppressive activity by AM did not decline after BMT, but rather remained elevated throughout the 12-wk development of GVHD and IPS. In a 14-day model of IPS, continuous inhibition of NO with aminoguanidine (AG) reduced signs of IPS/GVHD, but also led to higher mortality. When AG treatment was initiated after onset of IPS/GVHD, rapid mortality occurred that depended on the severity of IPS/GVHD. AG-enhanced mortality was not due to inhibition of marrow engraftment, elevated serum TNF-alpha, liver injury, or hypertensive responses. In contrast, T cells were involved, because depletion of CD4(+) lymphocytes 24 h before AG treatment prevented mortality. Thus, NO production following allogeneic BMT affords a protective effect that helps down-regulate injury caused by T cells during GVHD and IPS.


Subject(s)
Graft vs Host Disease/complications , Macrophages, Alveolar/physiology , Nitric Oxide/physiology , Pneumonia/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Animals , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/immunology , CD4-Positive T-Lymphocytes/immunology , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Guanidines/pharmacology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Nitric Oxide/antagonists & inhibitors , Nitrites/blood , Pneumonia/etiology , Pneumonia/immunology , Radiation Chimera , Spleen/cytology , Superoxides/metabolism , Syndrome , T-Lymphocytes/transplantation , Tumor Necrosis Factor-alpha/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...