Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Immunol ; 192(7): 3180-9, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24585878

ABSTRACT

The majority of allogeneic stem cell transplants are currently undertaken using G-CSF mobilized peripheral blood stem cells. G-CSF has diverse biological effects on a broad range of cells and IL-10 is a key regulator of many of these effects. Using mixed radiation chimeras in which the hematopoietic or nonhematopoietic compartments were wild-type, IL-10(-/-), G-CSFR(-/-), or combinations thereof we demonstrated that the attenuation of alloreactive T cell responses after G-CSF mobilization required direct signaling of the T cell by both G-CSF and IL-10. IL-10 was generated principally by radio-resistant tissue, and was not required to be produced by T cells. G-CSF mobilization significantly modulated the transcription profile of CD4(+)CD25(+) regulatory T cells, promoted their expansion in the donor and recipient and their depletion significantly increased graft-versus-host disease (GVHD). In contrast, stem cell mobilization with the CXCR4 antagonist AMD3100 did not alter the donor T cell's ability to induce acute GVHD. These studies provide an explanation for the effects of G-CSF on T cell function and demonstrate that IL-10 is required to license regulatory function but T cell production of IL-10 is not itself required for the attenuation GVHD. Although administration of CXCR4 antagonists is an efficient means of stem cell mobilization, this fails to evoke the immunomodulatory effects seen during G-CSF mobilization. These data provide a compelling rationale for considering the immunological benefits of G-CSF in selecting mobilization protocols for allogeneic stem cell transplantation.


Subject(s)
Granulocyte Colony-Stimulating Factor/immunology , Hematopoietic Stem Cell Mobilization/methods , Interleukin-10/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Benzylamines , Cell Proliferation/drug effects , Cyclams , Flow Cytometry , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Granulocyte Colony-Stimulating Factor/pharmacology , Heterocyclic Compounds/immunology , Heterocyclic Compounds/pharmacology , Interleukin-10/genetics , Interleukin-10/metabolism , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stem Cell Transplantation/methods , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcriptome/drug effects , Transcriptome/immunology
2.
Blood ; 119(24): 5918-30, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22415754

ABSTRACT

Alloreactivity after transplantation is associated with profound immune suppression, and consequent opportunistic infection results in high morbidity and mortality. This immune suppression is most profound during GVHD after bone marrow transplantation where an inflammatory cytokine storm dominates. Contrary to current dogma, which avers that this is a T-cell defect, we demonstrate that the impairment lies within conventional dendritic cells (cDCs). Significantly, exogenous antigens can only be presented by the CD8(-) cDC subset after bone marrow transplantation, and inflammation during GVHD specifically renders the MHC class II presentation pathway in this population incompetent. In contrast, both classic and cross-presentation within MHC class I remain largely intact. Importantly, this defect in antigen processing can be partially reversed by TNF inhibition or the adoptive transfer of donor cDCs generated in the absence of inflammation.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Graft vs Host Disease/immunology , Immunosuppression Therapy , Adoptive Transfer , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Marrow Transplantation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cross-Priming/immunology , Graft vs Host Disease/pathology , Histocompatibility Antigens Class II/immunology , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/metabolism , Isoantigens/immunology , Mice , Mice, Transgenic , Peptides/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/metabolism
3.
Blood ; 118(12): 3399-409, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21719602

ABSTRACT

Although the effects of type II-IFN (IFN-γ) on GVHD and leukemia relapse are well studied, the effects of type I-interferon (type I-IFN, IFN-α/ß) remain unclear. We investigated this using type I-IFN receptor-deficient mice and exogenous IFN-α administration in established models of GVHD and GVL. Type I-IFN signaling in host tissue prevented severe colon-targeted GVHD in CD4-dependent models of GVHD directed toward either major histocompatibility antigens or multiple minor histocompatibility antigens. This protection was the result of suppression of donor CD4(+) T-cell proliferation and differentiation. Studies in chimeric recipients demonstrated this was due to type I-IFN signaling in hematopoietic tissue. Consistent with this finding, administration of IFN-α during conditioning inhibited donor CD4(+) proliferation and differentiation. In contrast, CD8-dependent GVHD and GVL effects were enhanced when type I-IFN signaling was intact in the host or donor, respectively. This finding reflected the ability of type I-IFN to both sensitize host target tissue/leukemia to cell-mediated cytotoxicity and augment donor CTL function. These data confirm that type I-IFN plays an important role in defining the balance of GVHD and GVL responses and suggests that administration of the cytokine after BM transplantation could be studied prospectively in patients at high risk of relapse.


Subject(s)
Bone Marrow Transplantation/immunology , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Interferon-alpha , Leukemia/immunology , Receptor, Interferon alpha-beta/deficiency , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/drug effects , Female , Graft vs Host Disease/pathology , Graft vs Leukemia Effect/drug effects , Humans , Interferon-alpha/immunology , Interferon-alpha/pharmacology , Interferon-beta/immunology , Leukemia/mortality , Leukemia/pathology , Leukemia/therapy , Lymphocyte Activation/immunology , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens/immunology , Receptor, Interferon alpha-beta/immunology , Signal Transduction , Survival Rate , Transplantation, Homologous , Whole-Body Irradiation
4.
Blood ; 116(5): 819-28, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20435882

ABSTRACT

The recent shift to the use of stem cells mobilized by granulocyte colony-stimulating factor (G-CSF) for hematopoietic transplantation has increased chronic graftversus-host disease (GVHD), although the mechanisms of this are unclear. We have found that G-CSF invokes potent type 17 rather than type 1 or type 2 differentiation. The amplification of interleukin-17 (IL-17) production by G-CSF occurs in both CD4 and CD8 conventional T cells and is dependent on, and downstream of, G-CSF-induced IL-21 signaling. Importantly, donor IL-17A controls the infiltration of macrophages into skin and cutaneous fibrosis, manifesting late after transplantation as scleroderma. Interestingly, donor CD8 T cells were the predominant source of IL-17A after transplantation and could mediate scleroderma independently of CD4 T cells. This study provides a logical explanation for the propensity of allogeneic stem cell transplantation to invoke sclerodermatous GVHD and suggests a therapeutic strategy for intervention.


Subject(s)
Bone Marrow Transplantation/adverse effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Mobilization/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Interleukin-17/physiology , Interleukins/physiology , Scleroderma, Localized/etiology , T-Lymphocyte Subsets/transplantation , Animals , Cell Differentiation/drug effects , Crosses, Genetic , Cytokines/biosynthesis , Female , Fibrosis , Humans , Interleukin-17/metabolism , Macrophages/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Radiation Chimera , Signal Transduction , Skin/immunology , Skin/pathology , Transplantation, Homologous/adverse effects
5.
Blood ; 116(2): 287-96, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20435883

ABSTRACT

Suppressor of cytokine signaling-3 (SOCS3) is the main intracellular regulator of signaling by granulocyte colony-stimulating factor, an immune-modulatory cytokine used to mobilize stem cells for transplantation. We have therefore studied the contribution of SOCS3 to the spectrum of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (SCT). Grafts from SOCS3(-/Deltavav) donor mice in which SOCS3 deficiency is restricted to the hematopoietic compartment had an augmented capacity to induce acute GVHD. With the use of SOCS3(-/DeltaLysM) and SOCS3(-/Deltalck) donors in which SOCS3 deficiency was restricted to the myeloid or T-cell lineage, respectively, we confirmed SOCS3 deficiency promoted acute GVHD mortality and histopathology within the gastrointestinal tract by effects solely within the donor T cell. SOCS3(-/Deltalck) donor T cells underwent enhanced alloantigen-dependent proliferation and generation of interleukin-10 (IL-10), IL-17, and interferon-gamma (IFNgamma) after SCT. The enhanced capacity of the SOCS3(-/Deltalck) donor T cell to induce acute GVHD was dependent on IFNgamma but independent of IL-10 or IL-17. Surprisingly, SOCS3(-/Deltalck) donor T cells also induced severe, transforming growth factor beta- and IFNgamma-dependent, sclerodermatous GVHD. Thus, the delivery of small molecule SOCS3 mimetics may prove to be useful for the inhibition of both acute and chronic GVHD.


Subject(s)
Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Suppressor of Cytokine Signaling Proteins/immunology , T-Lymphocytes/immunology , Animals , Cell Separation , Cytokines/biosynthesis , Female , Flow Cytometry , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Mice , Mice, Inbred C57BL , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes/metabolism , Transplantation, Homologous
6.
Blood ; 115(1): 122-32, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-19789388

ABSTRACT

Tumor necrosis factor (TNF) is a key cytokine in the effector phase of graft-versus-host disease (GVHD) after bone marrow transplantation, and TNF inhibitors have shown efficacy in clinical and experimental GVHD. TNF signals through the TNF receptors (TNFR), which also bind soluble lymphotoxin (LTalpha3), a TNF family member with a previously unexamined role in GVHD pathogenesis. We have used preclinical models to investigate the role of LT in GVHD. We confirm that grafts deficient in LTalpha have an attenuated capacity to induce GVHD equal to that seen when grafts lack TNF. This is not associated with other defects in cytokine production or T-cell function, suggesting that LTalpha3 exerts its pathogenic activity directly via TNFR signaling. We confirm that donor-derived LTalpha is required for graft-versus-leukemia (GVL) effects, with equal impairment in leukemic clearance seen in recipients of LTalpha- and TNF-deficient grafts. Further impairment in tumor clearance was seen using Tnf/Lta(-/-) donors, suggesting that these molecules play nonredundant roles in GVL. Importantly, donor TNF/LTalpha were only required for GVL where the recipient leukemia was susceptible to apoptosis via p55 TNFR signaling. These data suggest that antagonists neutralizing both TNF and LTalpha3 may be effective for treatment of GVHD, particularly if residual leukemia lacks the p55 TNFR.


Subject(s)
Graft vs Host Disease/immunology , Lymphotoxin-alpha/immunology , Animals , Apoptosis , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Graft vs Host Disease/pathology , Inflammation Mediators/metabolism , Mice , Protein Multimerization , Receptors, Antigen, T-Cell/immunology , Receptors, Immunologic/administration & dosage , Receptors, Immunologic/immunology , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor/immunology , Solubility , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/immunology
7.
Blood ; 113(22): 5644-9, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19336758

ABSTRACT

We have quantified the relative contribution of donor antigen-presenting cell populations to alloantigen presentation after bone marrow transplantation (BMT) by using transgenic T cells that can respond to host-derived alloantigen presented within the donor major histocompatibility complex. We also used additional transgenic/knockout donor mice and/or monoclonal antibodies that allowed conditional depletion of conventional dendritic cells (cDCs), plasmacytoid DC (pDCs), macrophages, or B cells. Using these systems, we demonstrate that donor cDCs are the critical population presenting alloantigen after BMT, whereas pDCs and macrophages do not make a significant contribution in isolation. In addition, alloantigen presentation was significantly enhanced in the absence of donor B cells, confirming a regulatory role for these cells early after transplantation. These data have major implications for the design of therapeutic strategies post-BMT, and suggest that cDC depletion and the promotion of B-cell reconstitution may be beneficial tools for the control of alloreactivity.


Subject(s)
Antigen-Presenting Cells/immunology , Bone Marrow Transplantation/immunology , Dendritic Cells/immunology , Isoantigens/immunology , Animal Experimentation , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD11 Antigens/genetics , Dendritic Cells/physiology , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
8.
Blood ; 113(23): 5999-6010, 2009 Jun 04.
Article in English | MEDLINE | ID: mdl-19369232

ABSTRACT

Invariant natural killer T cells (iNKT cells) have pivotal roles in graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL) effects. iNKT cells are activated through their T-cell receptors by glycolipid moieties (typically the alpha-galactosylceramide [alpha-GalCer] derivative KRN7000) presented within CD1d. We investigated the ability of modified alpha-GalCer molecules to differentially modulate alloreactivity and GVL. KRN7000 and the N-acyl variant, C20:2, were administered in multiple well-established murine models of allogeneic stem cell transplantation. The highly potent and specific activation of all type I NKT cells with C20:2 failed to exacerbate and in most settings inhibited GVHD late after transplantation, whereas effects on GVL were variable. In contrast, the administration of KRN7000 induced hyperacute GVHD and early mortality in all models tested. Administration of KRN7000, but not C20:2, was found to result in downstream interleukin (IL)-12 and dendritic cell (DC)-dependent natural killer (NK)- and conventional T-cell activation. Specific depletion of host DCs, IL-12, or donor NK cells prevented this pathogenic response and the induction of hyperacute GVHD. These data demonstrate the ability of profound iNKT activation to modulate both the innate and adaptive immune response via the DC-NK-cell interaction and raise concern for the use of alpha-GalCer therapeutically to modulate GVHD and GVL effects.


Subject(s)
Galactosylceramides/administration & dosage , Natural Killer T-Cells/drug effects , Stem Cell Transplantation , Animals , Cytokines/biosynthesis , Female , Galactosylceramides/chemistry , Galactosylceramides/pharmacology , Graft vs Leukemia Effect/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Natural Killer T-Cells/immunology , Survival Rate , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Homologous , Treatment Outcome
9.
J Immunol ; 182(2): 912-20, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19124734

ABSTRACT

The role of Ag presenting cell subsets in graft-versus-host disease (GVHD) remains unclear. We have thus examined the ability of plasmacytoid dendritic cells (pDC) to modulate transplant outcome. Surprisingly, host pDC were exquisitely sensitive to total body irradiation and were depleted before transplantation, thus allowing us to focus on donor pDC [corrected]. The depletion of all pDC from bone marrow grafts resulted in an acceleration of GVHD mortality while the depletion of mature pDC from G-CSF mobilized splenic grafts had no effect. Thus, donor bone marrow pDC, but not mature pDC contained within stem cell grafts attenuate acute GVHD. In the presence of GVHD, donor pDC completely failed to reconstitute although a CD11clow120G8+ precursor DC reconstituted in an exaggerated and transient manner. These cells expressed Flt-3, the macrophage colony stimulating factor receptor and, consistent with a common dendritic cell (DC) precursor, were capable of differentiation into pDC and conventional DC in vivo in the absence of GVHD. These precursors were MHC class II+ and CD80/86+ but lacked CD40, were actively presenting host Ag and inhibited GVHD and T cell proliferation in a contact-dependent fashion. These data demonstrate that GVHD prevents the maturation of pDC and instead promotes the generation of a suppressive precursor DC, further contributing to the state of immune paralysis after transplantation.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , Cells, Cultured , Dendritic Cells/transplantation , Female , Graft vs Host Disease/diagnosis , Immunophenotyping , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Spleen/immunology , Spleen/pathology , Stem Cell Transplantation , Stem Cells/immunology , Stem Cells/pathology
10.
J Clin Invest ; 115(11): 3093-103, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16224535

ABSTRACT

NKT cells have pivotal roles in immune regulation and tumor immunosurveillance. We report that the G-CSF and FMS-like tyrosine kinase 3 ligand (Flt-3L) chimeric cytokine, progenipoietin-1, markedly expands the splenic and hepatic NKT cell population and enhances functional responses to alpha-galactosylceramide. In a murine model of allogeneic stem cell transplantation, donor NKT cells promoted host DC activation and enhanced perforin-restricted CD8+ T cell cytotoxicity against host-type antigens. Following leukemic challenge, donor treatment with progenipoietin-1 significantly improved overall survival when compared with G-CSF or control, attributable to reduced graft-versus-host disease mortality and paradoxical augmentation of graft-versus-leukemia (GVL) effects. Enhanced cellular cytotoxicity was dependent on donor NKT cells, and leukemia clearance was profoundly impaired in recipients of NKT cell-deficient grafts. Enhanced cytotoxicity and GVL effects were not associated with Flt-3L signaling or effects on DCs but were reproduced by prolonged G-CSF receptor engagement with pegylated G-CSF. Thus, modified G-CSF signaling during stem cell mobilization augments NKT cell-dependent CD8+ cytotoxicity, effectively separating graft-versus-host disease and GVL and greatly expanding the potential applicability of allogeneic stem cell transplantation for the therapy of malignant disease.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Killer Cells, Natural/drug effects , Leukemia, Experimental/drug therapy , T-Lymphocytes/drug effects , Animals , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/immunology , Colony-Stimulating Factors/pharmacology , Dendritic Cells/immunology , Female , Galactosylceramides/physiology , Graft vs Leukemia Effect/drug effects , Graft vs Leukemia Effect/immunology , Killer Cells, Natural/metabolism , Leukemia, Experimental/immunology , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/pharmacology , Signal Transduction/immunology , Stem Cell Transplantation , T-Lymphocytes/metabolism
11.
Nat Med ; 18(1): 135-42, 2011 Nov 29.
Article in English | MEDLINE | ID: mdl-22127134

ABSTRACT

The presentation pathways by which allogeneic peptides induce graft-versus-host disease (GVHD) are unclear. We developed a bone marrow transplant (BMT) system in mice whereby presentation of a processed recipient peptide within major histocompatibility complex (MHC) class II molecules could be spatially and temporally quantified. Whereas donor antigen presenting cells (APCs) could induce lethal acute GVHD via MHC class II, recipient APCs were 100-1,000 times more potent in this regard. After myeloablative irradiation, T cell activation and memory differentiation occurred in lymphoid organs independently of alloantigen. Unexpectedly, professional hematopoietic-derived recipient APCs within lymphoid organs had only a limited capacity to induce GVHD, and dendritic cells were not required. In contrast, nonhematopoietic recipient APCs within target organs induced universal GVHD mortality and promoted marked alloreactive donor T cell expansion within the gastrointestinal tract and inflammatory cytokine generation. These data challenge current paradigms, suggesting that experimental lethal acute GVHD can be induced by nonhematopoietic recipient APCs.


Subject(s)
Antigen-Presenting Cells/immunology , Bone Marrow Transplantation/immunology , CD4-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Histocompatibility Antigens Class II/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/cytology , Cytokines/immunology , Dendritic Cells/immunology , Hematopoietic System/immunology , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Transplant Donor Site , Transplantation, Homologous
12.
Nat Med ; 15(4): 436-41, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19330008

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is often used to hasten neutrophil recovery after allogeneic bone marrow transplantation (BMT), but the clinical and immunological consequences evoked remain unclear. We examined the effect of G-CSF administration after transplantation in mouse models and found that exposure to either standard G-CSF or pegylated-G-CSF soon after BMT substantially increased graft-versus-host disease (GVHD). This effect was dependent on total body irradiation (TBI) rendering host dendritic cells (DCs) responsive to G-CSF by upregulating their expression of the G-CSF receptor. Stimulation of host DCs by G-CSF subsequently unleashed a cascade of events characterized by donor natural killer T cell (NKT cell) activation, interferon-gamma secretion and CD40-dependent amplification of donor cytotoxic T lymphocyte function during the effector phase of GVHD. Crucially, the detrimental effects of G-CSF were only present when it was administered after TBI conditioning and at a time when residual host antigen presenting cells were still present, perhaps explaining the conflicting and somewhat controversial clinical studies from the large European and North American BMT registries. These data have major implications for the use of G-CSF in disease states where NKT cell activation may have effects on outcome.


Subject(s)
Bone Marrow Transplantation/immunology , Granulocyte Colony-Stimulating Factor/therapeutic use , Killer Cells, Natural/immunology , Neutrophils/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Humans , Interferon-gamma/physiology , Lymphocyte Activation/drug effects , Lymphocyte Transfusion , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Models, Animal , Neutrophils/drug effects , T-Lymphocytes/transplantation , Transplantation, Homologous/immunology , Whole-Body Irradiation
13.
Blood ; 109(11): 5049-57, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17327399

ABSTRACT

Antigen-presenting cells (APCs) are critical for the initiation of graft-versus-host disease (GVHD), although the responsible APC subset and molecular mechanisms remain unclear. Because dendritic cells (DCs) are the most potent APCs and the NF-kB/Rel family member RelB is associated with DC maturation and potent APC function, we examined their role in GVHD. Within 4 hours of total body irradiation, RelB nuclear translocation was increased and restricted to CD11c(hi) DCs within the host APC compartment. Furthermore, the transient depletion of CD11c(hi) donor DCs that reconstitute in the second week after transplantation resulted in a transient decrease in GVHD severity. By using RelB(-/-) bone marrow chimeras as transplant recipients or RelB(-/-) donor bone marrow, we demonstrate that the induction and maintenance of GVHD is critically dependent on this transcription factor within both host and donor APCs. Critically, RelB within APCs was required for the expansion of donor helper T cell type 1 (Th1) effectors and subsequent alloreactivity, but not the peripheral expansion or function of donor FoxP3(+) regulatory T cells. These data suggest that the targeted inhibition of nuclear RelB translocation within APCs represents an attractive therapeutic strategy to dissociate effector and regulatory T-cell function in settings of Th1-mediated tissue injury.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation , Graft vs Host Disease/immunology , T-Lymphocytes, Regulatory/immunology , Transcription Factor RelB/biosynthesis , Active Transport, Cell Nucleus , Animals , Antibodies, Monoclonal/chemistry , CD11c Antigen/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells , Transcription, Genetic
14.
Blood ; 108(7): 2485-92, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16788097

ABSTRACT

Host antigen-presenting cells (APCs) are known to be critical for the induction of graft-versus-host disease (GVHD) after allogeneic bone marrow transplantation (BMT), but the relative contribution of specific APC subsets remains unclear. We have studied the role of host B cells in GVHD by using B-cell-deficient microMT mice as BMT recipients in a model of CD4-dependent GVHD to major histocompatibility complex antigens. We demonstrate that acute GVHD is initially augmented in microMT recipients relative to wild-type recipients (mortality: 85% vs 44%, P < .01), and this is the result of an increase in donor T-cell proliferation, expansion, and inflammatory cytokine production early after BMT. Recipient B cells were depleted 28-fold at the time of BMT by total body irradiation (TBI) administered 24 hours earlier, and we demonstrate that TBI rapidly induces sustained interleukin-10 (IL-10) generation from B cells but not dendritic cells (DCs) or other cellular populations within the spleen. Finally, recipient mice in which B cells are unable to produce IL-10 due to homologous gene deletion develop more severe acute GVHD than recipient mice in which B cells are wild type. Thus, the induction of IL-10 in host B cells during conditioning attenuates experimental acute GVHD.


Subject(s)
B-Lymphocytes/metabolism , Bone Marrow Transplantation/methods , Graft vs Host Disease/metabolism , Interleukin-10/physiology , Transplantation, Homologous/methods , Animals , Antigen-Presenting Cells/chemistry , Cytokines/metabolism , Disease Models, Animal , Female , Gene Deletion , Interleukin-10/metabolism , Mice , Mice, Inbred BALB C , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL