Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 188(8): 3804-11, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22422880

ABSTRACT

The efficacy of allogeneic hematopoietic stem cell transplantation is limited by graft-versus-host disease (GVHD). Host hematopoietic APCs are important initiators of GVHD, making them logical targets for GVHD prevention. Conventional dendritic cells (DCs) are key APCs for T cell responses in other models of T cell immunity, and they are sufficient for GVHD induction. However, we report in this article that in two polyclonal GVHD models in which host hematopoietic APCs are essential, GVHD was not decreased when recipient conventional DCs were inducibly or constitutively deleted. Additional profound depletion of plasmacytoid DCs and B cells, with or without partial depletion of CD11b(+) cells, also did not ameliorate GVHD. These data indicate that, in contrast with pathogen models, there is a surprising redundancy as to which host cells can initiate GVHD. Alternatively, very low numbers of targeted APCs were sufficient. We hypothesize the difference in APC requirements in pathogen and GVHD models relates to the availability of target Ags. In antipathogen responses, specialized APCs are uniquely equipped to acquire and present exogenous Ags, whereas in GVHD, all host cells directly present alloantigens. These studies make it unlikely that reagent-based host APC depletion will prevent GVHD in the clinic.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Animals , B-Lymphocytes/pathology , CD11b Antigen/immunology , CD8-Positive T-Lymphocytes/pathology , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/pathology , Diphtheria Toxin , Female , Graft vs Host Disease/pathology , Humans , Isoantigens/immunology , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Radiation Chimera , Transplantation, Homologous
2.
Blood ; 118(24): 6426-37, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-21963602

ABSTRACT

Recipient antigen-presenting cells (APCs) initiate GVHD by directly presenting host minor histocompatibility antigens (miHAs) to donor CD8 cells. However, later after transplantation, host APCs are replaced by donor APCs, and if pathogenic CD8 cells continue to require APC stimulation, then donor APCs must cross-present host miHAs. Consistent with this, CD8-mediated GVHD is reduced when donor APCs are MHC class I(-). To study cross-presentation, we used hosts that express defined MHC class I K(b)-restricted miHAs, crossed to K(b)-deficient backgrounds, such that these antigens cannot be directly presented. Cross-priming was surprisingly efficient, whether antigen was restricted to the hematopoietic or nonhematopoietic compartments. Cross-primed CD8 cells were cytolytic and produced IFN-γ. CD8 cells were exclusively primed by donor CD11c(+) cells, and optimal cross-priming required that they are stimulated by both type I IFNs and CD40L. In studying which donor APCs acquire host miHAs, we made the surprising discovery that there was a large-scale transfer of transmembrane proteins from irradiated hosts, including MHC class I-peptide complexes, to donor cells, including dendritic cells. Donor dendritic cells that acquired host MHC class I-peptide complexes were potent stimulators of peptide-specific T cells. These studies identify new therapeutic targets for GVHD treatment and a novel mechanism whereby donor APCs prime host-reactive T cells.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , Autoantigens/adverse effects , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Graft vs Host Disease/immunology , Animals , Antigen-Presenting Cells/metabolism , Autoantigens/genetics , Bone Marrow Transplantation/adverse effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/metabolism , Histocompatibility Antigens Class I/adverse effects , Histocompatibility Antigens Class I/genetics , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Minor Histocompatibility Antigens/adverse effects , Minor Histocompatibility Antigens/genetics , Molecular Targeted Therapy , Peptide Fragments/adverse effects
3.
Blood ; 118(22): 5965-76, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-21917752

ABSTRACT

Donor T cells contribute to the success of allogeneic hematopoietic stem cell transplantation (alloSCT). Alloreactive donor T cells attack leukemia cells, mediating the GVL effect. Donor T cells, including the memory T cells (T(M)) that are generated after infection, also promote immune reconstitution. Nonetheless, leukemia relapse and infection are major sources of treatment failure. Efforts to augment GVL and immune reconstitution have been limited by GVHD, the attack by donor T cells on host tissues. One approach to augmenting GVL has been to infuse ex vivo-generated T cells with defined specificities; however, this requires expertise that is not widely available. In the present study, we tested an alternative approach, adoptive immunotherapy with CD8+ T(M) from donors vaccinated against a single minor histocompatibility antigen (miHA) expressed by leukemia cells. Vaccination against the miHA H60 greatly augmented T(M)-mediated GVL against mouse chronic-phase (CP-CML) and blast crisis chronic myeloid leukemia (BC-CML). T(M)-mediated GVL was antigen specific and was optimal when H60 expression was hematopoietically restricted. Even when H60 was ubiquitous, donor H60 vaccination had a minimal impact on GVHD. T(M) from lymphocytic choriomeningitis virus (LCMV)-immune and H60-vaccinated donors augmented GVL and protected recipients from LCMV. These data establish a strategy for augmenting GVL and immune reconstitution without elaborate T-cell manipulation.


Subject(s)
Graft vs Leukemia Effect/immunology , Immunologic Memory , Lymphocytic choriomeningitis virus/immunology , Minor Histocompatibility Antigens/immunology , T-Lymphocytes/immunology , Tissue Donors , Vaccination , Animals , Cell Proliferation , Hematopoiesis/immunology , Hematopoiesis/physiology , Immunologic Memory/physiology , Lymphocytes/cytology , Lymphocytes/physiology , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/physiology , Transplantation Immunology , Up-Regulation
4.
J Immunol ; 187(4): 1653-63, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21768400

ABSTRACT

Graft-versus-leukemia (GVL) against chronic-phase chronic myelogenous leukemia (CP-CML) is potent, but it is less efficacious against acute leukemias and blast-crisis chronic myelogenous leukemia (BC-CML). The mechanisms underlying GVL resistance are unknown. Previously, we found that alloreactive T cell targeting of GVL-sensitive bcr-abl-induced mouse CP-CML (mCP-CML) required TCR-MHC interactions and that multiple and redundant killing mechanisms were in play. To better understand why BC-CML is resistant to GVL, we performed a comprehensive analysis of GVL against mouse BC-CML (mBC-CML) induced by the retroviral transfer of the bcr-abl and NUP98/HOXA9 fusion cDNAs. Like human BC-CML, mBC-CML was GVL resistant, and this was not due to accelerated kinetics or a greater leukemia burden. To study T cell recognition and killing mechanisms, we generated a panel of gene-deficient leukemias by transducing bone marrow from gene-deficient mice. T cell target recognition absolutely required that mBC-CML cells express MHC molecules. GVL against both mCP-CML and mBC-CML required leukemia expression of ICAM-1. We hypothesized that mBC-CML would be resistant to some of the killing mechanisms sufficient to eliminate mCP-CML, but we found instead that the same mechanisms were effective against both types of leukemia, because GVL was similar against wild-type or mBC-CML genetically lacking Fas, TRAIL-R, Fas/TRAIL-R, or TNFR1/R2 or when donor T cells were perforin(-/-). However, mCP-CML, but not mBC-CML, relied on expression of programmed death-1 ligands 1 and 2 (PD-L1/L2) to resist T cell killing, because only GVL against mCP-CML was augmented when leukemias lacked PD-L1/L2. Thus, mBC-CML cells have cell-intrinsic mechanisms, distinct from mCP-CML cells, which protect them from T cell killing.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Blast Crisis/immunology , Graft vs Leukemia Effect/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Blast Crisis/genetics , Genes, abl/immunology , Graft vs Leukemia Effect/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/immunology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/immunology , Retroviridae , T-Lymphocytes/immunology , Transduction, Genetic
5.
Blood ; 117(2): 697-707, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20944073

ABSTRACT

Graft-versus-host disease (GVHD) is initiated and maintained by antigen-presenting cells (APCs) that prime alloreactive donor T cells. APCs are therefore attractive targets for GVHD prevention and treatment. APCs are diverse in phenotype and function, making understanding how APC subsets contribute to GVHD necessary for the development of APC-targeted therapies. Langerhans cells (LCs) have been shown to be sufficient to initiate skin GVHD in a major histocompatibility complex-mismatched model; however, their role when other host APC subsets are intact is unknown. To address this question, we used mice genetically engineered to be deficient in LCs by virtue of expression of diphtheria toxin A under the control of a BAC (bacterial artificial chromosome) transgenic hu-man Langerin locus. Neither CD8- nor CD4-mediated GVHD was diminished in recipients lacking LCs. Similarly, CD8- and CD4-mediated GVHD, including that in the skin, was unaffected if bone marrow came from donors that could not generate LCs, even though donor LCs engrafted in control mice. Engraftment of donor LCs after irradiation in wild-type hosts required donor T cells, with immunofluorescence revealing patches of donor and residual host LCs. Surprisingly, donor LC engraftment in Langerin-diphtheria toxin A (DTA) transgenic hosts was independent of donor T cells, suggesting that a Langerin(+) cell regulates repopulation of the LC compartment.


Subject(s)
Graft vs Host Disease/immunology , Langerhans Cells/immunology , Animals , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Mice, Transgenic
6.
J Immunol ; 186(1): 230-41, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21098219

ABSTRACT

Graft-versus-host disease (GVHD) is initiated by APCs that prime alloreactive donor T cells. In antipathogen responses, Ag-bearing APCs receive signals through pattern-recognition receptors, including TLRs, which induce the expression of costimulatory molecules and production of inflammatory cytokines, which in turn mold the adaptive T cell response. However, in allogeneic hematopoietic stem cell transplantation (alloSCT), there is no specific pathogen, alloantigen is ubiquitous, and signals that induce APC maturation are undefined. To investigate APC activation in GVHD, we used recipient mice with hematopoietic cells genetically deficient in pathways critical for APC maturation in models in which host APCs are absolutely required. Strikingly, CD8-mediated and CD4-mediated GVHD were similar whether host APCs were wild-type or deficient in MyD88, TRIF, or MyD88 and TRIF, which excludes essential roles for TLRs and IL-1ß, the key product of inflammasome activation. Th1 differentiation was if anything augmented when APCs were MyD88/TRIF(-/-), and T cell production of IFN-γ did not require host IL-12. GVHD was also intact when APCs lacked the type I IFNR, which amplifies APC activation pathways that induce type I IFNs. Thus in GVHD, alloreactive T cells can be activated when pathways critical for antipathogen T cell responses are impaired.


Subject(s)
Graft vs Host Disease/immunology , Hematopoiesis/immunology , Immunity, Innate , Signal Transduction/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/microbiology , Antigen-Presenting Cells/virology , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , Graft vs Host Disease/microbiology , Graft vs Host Disease/virology , Hematopoiesis/genetics , Host Specificity/genetics , Host Specificity/immunology , Immunity, Innate/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Radiation Chimera/immunology , Signal Transduction/genetics , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology
7.
Cell Res ; 16(2): 126-33, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16474424

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is an important hematopoietic growth factor and immune modulator. GM-CSF also has profound effects on the functional activities of various circulating leukocytes. It is produced by a variety of cell types including T cells, macrophages, endothelial cells and fibroblasts upon receiving immune stimuli. Although GM-CSF is produced locally, it can act in a paracrine fashion to recruit circulating neutrophils, monocytes and lymphocytes to enhance their functions in host defense. Recent intensive investigations are centered on the application of GM-CSF as an immune adjuvant for its ability to increase dendritic cell (DC) maturation and function as well as macrophage activity. It is used clinically to treat neutropenia in cancer patients undergoing chemotherapy, in AIDS patients during therapy, and in patients after bone marrow transplantation. Interestingly, the hematopoietic system of GM-CSF-deficient mice appears to be normal; the most significant changes are in some specific T cell responses. Although molecular cloning of GM-CSF was carried out using cDNA library of T cells and it is well known that the T cells produce GM-CSF after activation, there is a lack of systematic investigation of this cytokine in production by T cells and its effect on T cell function. In this article, we will focus mainly on the immunobiology of GM-CSF in T cells.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lymphocyte Activation , T-Lymphocytes/metabolism , Animals , Cancer Vaccines , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Inflammation/immunology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...