Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Blood Adv ; 8(8): 2032-2043, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38295282

ABSTRACT

ABSTRACT: Autophagy is an intracellular survival process that has established roles in the long-term survival and function of hematopoietic stem cells (HSC). We investigated the contribution of autophagy to HSC fitness during allogeneic transplantation and graft-versus-host disease (GVHD). We demonstrate in vitro that both tumor necrosis factor and IL-1ß, major components of GVHD cytokine storm, synergistically promote autophagy in both HSC and their more mature hematopoietic progenitor cells (HPC). In vivo we demonstrate that autophagy is increased in donor HSC and HPC during GVHD. Competitive transplant experiments demonstrated that autophagy-deficient cells display reduced capacity to reconstitute the hematopoietic system compared to wild-type counterparts. In a major histocompatibility complex-mismatched model of GVHD and associated cytokine dysregulation, we demonstrate that autophagy-deficient HSC and progenitors fail to establish durable hematopoiesis, leading to primary graft failure and universal transplant related mortality. Using several different models, we confirm that autophagy activity is increased in early progenitor and HSC populations in the presence of T-cell-derived inflammatory cytokines and that these HSC populations require autophagy to survive. Thus, autophagy serves as a key survival mechanism in HSC and progenitor populations after allogeneic stem cell transplant and may represent a therapeutic target to prevent graft failure during GVHD.


Subject(s)
Autophagy , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Animals , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Mice , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Disease Models, Animal , Transplantation, Homologous , Graft Rejection , Cytokines/metabolism
2.
Blood Adv ; 1(6): 341-351, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-29296949

ABSTRACT

T-helper 17 (Th17) cells have been widely implicated as drivers of autoimmune disease. In particular, Th17 cytokine plasticity and acquisition of an interleukin-17A+(IL-17A+)interferon γ(IFNγ)+ cytokine profile is associated with increased pathogenic capacity. Donor Th17 polarization is known to exacerbate graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (allo-SCT); however, donor Th17 cytokine coexpression and plasticity have not been fully characterized. Using IL-17 "fate-mapping" mice, we identified IL-6-dependent Th17 cells early after allo-SCT, characterized by elevated expression of proinflammatory cytokines, IL-17A, IL-22, granulocyte-macrophage colony-stimulating factor, and tumor necrosis factor. This population did not maintain lineage fidelity, with a marked loss of IL-17A and IL-22 expression late posttransplant. Th17 cells were further segregated based on IFNγ coexpression, and IL-17A+IFNγ+ Th17 displayed an enhanced proinflammatory phenotype. Th17 cytokine plasticity and IFNγ production were critically dependent upon donor-derived IL-12p40, and cyclosporine (CsA) treatment regulated this differentiation pathway. This observation was highly concordant with clinical samples from allo-SCT recipients receiving CsA-based immune suppression where although the IFNγ-negative-Th17 subset predominated, IFNγ+-Th17 cells were also present. In sum, Th17 polarization and ensuing differentiation are mediated by sequential inflammatory signals, which are modulated by immunosuppressive therapy, leading to distinct phenotypes within this lineage.

3.
JCI Insight ; 1(15): e86850, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27699243

ABSTRACT

Regulatory T cells (Tregs) play a crucial role in the maintenance of peripheral tolerance. Quantitative and/or qualitative defects in Tregs result in diseases such as autoimmunity, allergy, malignancy, and graft-versus-host disease (GVHD), a serious complication of allogeneic stem cell transplantation (SCT). We recently reported increased expression of autophagy-related genes (Atg) in association with enhanced survival of Tregs after SCT. Autophagy is a self-degradative process for cytosolic components that promotes cell homeostasis and survival. Here, we demonstrate that the disruption of autophagy within FoxP3+ Tregs (B6.Atg7fl/fl-FoxP3cre+ ) resulted in a profound loss of Tregs, particularly within the bone marrow (BM). This resulted in dysregulated effector T cell activation and expansion, and the development of enterocolitis and scleroderma in aged mice. We show that the BM compartment is highly enriched in TIGIT+ Tregs and that this subset is differentially depleted in the absence of autophagy. Moreover, following allogeneic SCT, recipients of grafts from B6.Atg7fl/fl-FoxP3cre+ donors exhibited reduced Treg reconstitution, exacerbated GVHD, and reduced survival compared with recipients of B6.WT-FoxP3cre+ grafts. Collectively, these data indicate that autophagy-dependent Tregs are critical for the maintenance of tolerance after SCT and that the promotion of autophagy represents an attractive immune-restorative therapeutic strategy after allogeneic SCT.


Subject(s)
Autophagy , Graft vs Host Disease/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow/physiopathology , Female , Hematopoietic Stem Cell Transplantation , Immune Tolerance , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
J Hepatol ; 65(4): 758-768, 2016 10.
Article in English | MEDLINE | ID: mdl-27262757

ABSTRACT

BACKGROUND & AIMS: Kupffer cells (KCs), the resident tissue macrophages of the liver, play a crucial role in the clearance of pathogens and other particulate materials that reach the systemic circulation. Recent studies have identified KCs as a yolk sac-derived resident macrophage population that is replenished independently of monocytes in the steady state. Although it is now established that following local tissue injury, bone marrow derived monocytes may infiltrate the tissue and differentiate into macrophages, the extent to which newly differentiated macrophages functionally resemble the KCs they have replaced has not been extensively studied. METHODS: We studied the two populations of KCs using intravital microscopy, morphometric analysis and gene expression profiling. An ion homeostasis gene signature, including genes associated with scavenger receptor function and extracellular matrix deposition, allowed discrimination between these two KC sub-types. RESULTS: Bone marrow derived "KCs" accumulating as a result of genotoxic injury, resemble but are not identical to their yolk sac counterparts. Reflecting the differential expression of scavenger receptors, yolk sac-derived KCs were more effective at accumulating acetylated low density lipoprotein, whereas surprisingly, they were poorer than bone marrow-derived KCs when assessed for uptake of a range of bacterial pathogens. The two KC populations were almost indistinguishable in regard to i) response to lipopolysaccharide challenge, ii) phagocytosis of effete red blood cells and iii) their ability to contain infection and direct granuloma formation against Leishmania donovani, a KC-tropic intracellular parasite. CONCLUSIONS: Bone marrow-derived KCs differentiate locally to resemble yolk sac-derived KC in most but not all respects, with implications for models of infectious diseases, liver injury and bone marrow transplantation. In addition, the gene signature we describe adds to the tools available for distinguishing KC subpopulations based on their ontology. LAY SUMMARY: Liver macrophages play a major role in the control of infections in the liver and in the pathology associated with chronic liver diseases. It was recently shown that liver macrophages can have two different origins, however, the extent to which these populations are functionally distinct remains to be fully addressed. Our study demonstrates that whilst liver macrophages share many features in common, regardless of their origin, some subtle differences in function exist. DATA REPOSITORY: Gene expression data are available from the European Bioinformatics Institute ArrayExpress data repository (accession number E-MTAB-4954).


Subject(s)
Bone Marrow , Humans , Kupffer Cells , Liver , Macrophages , Monocytes
5.
Blood ; 128(6): 794-804, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27338097

ABSTRACT

Chronic graft-versus-host disease (cGVHD) is a major cause of late mortality following allogeneic bone marrow transplantation (BMT) and is characterized by tissue fibrosis manifesting as scleroderma and bronchiolitis obliterans. The development of acute GVHD (aGVHD) is a powerful clinical predictor of subsequent cGVHD, suggesting that aGVHD may invoke the immunologic pathways responsible for cGVHD. In preclinical models in which sclerodermatous cGVHD develops after a preceding period of mild aGVHD, we show that antigen presentation within major histocompatibility complex (MHC) class II of donor dendritic cells (DCs) is markedly impaired early after BMT. This is associated with a failure of regulatory T-cell (Treg) homeostasis and cGVHD. Donor DC-restricted deletion of MHC class II phenocopied this Treg deficiency and cGVHD. Moreover, specific depletion of donor Tregs after BMT also induced cGVHD, whereas adoptive transfer of Tregs ameliorated it. These data demonstrate that the defect in Treg homeostasis seen in cGVHD is a causative lesion and is downstream of defective antigen presentation within MHC class II that is induced by aGVHD.


Subject(s)
Antigen Presentation , Bone Marrow Transplantation/adverse effects , Dendritic Cells/pathology , Graft vs Host Disease/pathology , T-Lymphocytes, Regulatory/pathology , Acute Disease , Adoptive Transfer , Animals , Chronic Disease , Dendritic Cells/immunology , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , Histocompatibility Antigens Class II/immunology , Lymphocyte Count , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation
6.
Am J Pathol ; 186(3): 524-38, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26762581

ABSTRACT

The interplay between the inflammatory infiltrate and tissue resident cell populations invokes fibrogenesis. However, the temporal and mechanistic contributions of these cells to fibrosis are obscure. To address this issue, liver inflammation, ductular reaction (DR), and fibrosis were induced in C57BL/6 mice by thioacetamide administration for up to 12 weeks. Thioacetamide treatment induced two phases of liver fibrosis. A rapid pericentral inflammatory infiltrate enriched in F4/80(+) monocytes co-localized with SMA(+) myofibroblasts resulted in early collagen deposition, marking the start of an initial fibrotic phase (1 to 6 weeks). An expansion of bone marrow-derived macrophages preceded a second phase, characterized by accelerated progression of fibrosis (>6 weeks) after DR migration from the portal tracts to the centrilobular site of injury, in association with an increase in DR/macrophage interactions. Although chemokine (C-C motif) ligand 2 (CCL2) mRNA was induced rapidly in response to thioacetamide, CCL2 deficiency only partially abrogated fibrosis. In contrast, colony-stimulating factor 1 receptor blockade diminished C-C chemokine receptor type 2 [CCR2(neg) (Ly6C(lo))] monocytes, attenuated the DR, and significantly reduced fibrosis, illustrating the critical role of colony-stimulating factor 1-dependent monocyte/macrophage differentiation and linking the two phases of injury. In response to liver injury, colony-stimulating factor 1 drives early monocyte-mediated myofibroblast activation and collagen deposition, subsequent macrophage differentiation, and their association with the advancing DR, the formation of fibrotic septa, and the progression of liver fibrosis to cirrhosis.


Subject(s)
Hepatitis, Animal/pathology , Liver Cirrhosis, Experimental/pathology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Animals , Chemokines/genetics , Chemokines/metabolism , Collagen Type I/metabolism , Disease Models, Animal , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation , Hepatitis, Animal/genetics , Hepatitis, Animal/metabolism , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Oligonucleotide Array Sequence Analysis , Thioacetamide
7.
Blood ; 126(13): 1609-20, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26206951

ABSTRACT

IL-17-producing cells are important mediators of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (SCT). Here we demonstrate that a distinct CD8(+) Tc17 population develops rapidly after SCT but fails to maintain lineage fidelity such that they are unrecognizable in the absence of a fate reporter. Tc17 differentiation is dependent on alloantigen presentation by host dendritic cells (DCs) together with IL-6. Tc17 cells express high levels of multiple prototypic lineage-defining transcription factors (eg, RORγt, T-bet) and cytokines (eg, IL-17A, IL-22, interferon-γ, granulocyte macrophage colony-stimulating factor, IL-13). Targeted depletion of Tc17 early after transplant protects from lethal acute GVHD; however, Tc17 cells are noncytolytic and fail to mediate graft-versus-leukemia (GVL) effects. Thus, the Tc17 differentiation program during GVHD culminates in a highly plastic, hyperinflammatory, poorly cytolytic effector population, which we term "inflammatory iTc17" (iTc17). Because iTc17 cells mediate GVHD without contributing to GVL, therapeutic inhibition of iTc17 development in a clinical setting represents an attractive approach for separating GVHD and GVL.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Graft vs Host Disease/pathology , Graft vs Leukemia Effect , Interleukin-17/immunology , Stem Cell Transplantation/adverse effects , Th17 Cells/pathology , Animals , Bone Marrow Transplantation/adverse effects , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Graft vs Host Disease/immunology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Th17 Cells/immunology
8.
J Exp Med ; 212(8): 1303-21, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26169940

ABSTRACT

The primacy of the gastrointestinal (GI) tract in dictating the outcome of graft-versus-host disease (GVHD) is broadly accepted; however, the mechanisms controlling this effect are poorly understood. Here, we demonstrate that GVHD markedly enhances alloantigen presentation within the mesenteric lymph nodes (mLNs), mediated by donor CD103(+)CD11b(-) dendritic cells (DCs) that migrate from the colon under the influence of CCR7. Expansion and differentiation of donor T cells specifically within the mLNs is driven by profound levels of alloantigen, IL-12, and IL-6 promoted by Toll-like receptor (TLR) and receptor for advanced glycation end products (RAGE) signals. Critically, alloantigen presentation in the mLNs imprints gut-homing integrin signatures on donor T cells, leading to their emigration into the GI tract where they mediate fulminant disease. These data identify a critical, anatomically distinct, donor DC subset that amplifies GVHD. We thus highlight multiple therapeutic targets and the ability of GVHD, once initiated by recipient antigen-presenting cells, to generate a profound, localized, and lethal feed-forward cascade of donor DC-mediated indirect alloantigen presentation and cytokine secretion within the GI tract.


Subject(s)
Antigens, CD/metabolism , Cell Movement/immunology , Colon/cytology , Dendritic Cells/metabolism , Graft vs Host Disease/physiopathology , Integrin alpha Chains/metabolism , Lymph Nodes/cytology , Analysis of Variance , Animals , Flow Cytometry , Interleukin-12/metabolism , Interleukin-6/metabolism , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Receptor for Advanced Glycation End Products , Receptors, CCR7/metabolism , Receptors, Immunologic/metabolism , T-Lymphocytes/immunology
9.
Blood ; 125(19): 2933-6, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25788702

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is widely used clinically to prevent neutropenia after cytotoxic chemotherapy and to mobilize hematopoietic stem cells (HSCs) for transplantation. Autophagy, a process of cytoplasmic component recycling, maintains cellular homeostasis and protects the cell during periods of metabolic stress or nutrient deprivation. We have observed that G-CSF activates autophagy in neutrophils and HSCs from both mouse and human donors. Furthermore, G-CSF-induced neutrophil and HSC mobilization is impaired in the absence of autophagy. In contrast, autophagy is dispensable for direct HSC mobilization in response to the CXCR4 antagonist AMD3100. Altogether, these data demonstrate an important role for G-CSF in invoking autophagy within hematopoietic and myeloid cells and suggest that this pathway is critical for ensuring cell survival in response to clinically relevant cytokine-induced stress. These findings have direct relevance to HSC transplantation and the increasing clinical use of agents that modulate autophagy.


Subject(s)
Autophagy , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Animals , Anti-HIV Agents/pharmacology , Antigens, CD34/genetics , Antigens, CD34/metabolism , Autophagy-Related Protein 5 , Benzylamines , Blotting, Western , Cells, Cultured , Cyclams , Flow Cytometry , Hematopoietic Stem Cells/pathology , Heterocyclic Compounds/pharmacology , Humans , Mice , Mice, Knockout , Microtubule-Associated Proteins/physiology , Neutrophils/drug effects , Neutrophils/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Autologous
10.
Blood ; 125(15): 2435-44, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25673640

ABSTRACT

Idiopathic pneumonia syndrome (IPS) is a relatively common, frequently fatal clinical entity, characterized by noninfectious acute lung inflammation following allogeneic stem cell transplantation (SCT), the mechanisms of which are unclear. In this study, we demonstrate that immune suppression with cyclosporin after SCT limits T-helper cell (Th) 1 differentiation and interferon-γ secretion by donor T cells, which is critical for inhibiting interleukin (IL)-6 generation from lung parenchyma during an alloimmune response. Thereafter, local IL-6 secretion induces donor alloantigen-specific Th17 cells to preferentially expand within the lung, and blockade of IL-17A or transplantation of grafts lacking the IL-17 receptor prevents disease. Studies using IL-6(-/-) recipients or IL-6 blockade demonstrate that IL-6 is the critical driver of donor Th17 differentiation within the lung. Importantly, IL-6 is also dysregulated in patients undergoing clinical SCT and is present at very high levels in the plasma of patients with IPS compared with SCT recipients without complications. Furthermore, at the time of diagnosis, plasma IL-6 levels were higher in a subset of IPS patients who were nonresponsive to steroids and anti-tumor necrosis factor therapy. In sum, pulmonary-derived IL-6 promotes IPS via the induction of Th17 differentiation, and strategies that target these cytokines represent logical therapeutic approaches for IPS.


Subject(s)
Acute Lung Injury/etiology , Cyclosporine/adverse effects , Immunosuppressive Agents/adverse effects , Interleukin-17/immunology , Interleukin-6/immunology , Lung/pathology , Stem Cell Transplantation/adverse effects , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Cyclosporine/therapeutic use , Female , Immunosuppressive Agents/therapeutic use , Interferon-gamma/immunology , Lung/drug effects , Lung/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Th17 Cells/drug effects , Th17 Cells/immunology , Transplantation, Homologous
11.
J Clin Invest ; 124(10): 4266-80, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25157821

ABSTRACT

Chronic GVHD (cGVHD) is the major cause of late, nonrelapse death following stem cell transplantation and characteristically develops in organs such as skin and lung. Here, we used multiple murine models of cGVHD to investigate the contribution of macrophage populations in the development of cGVHD. Using an established IL-17-dependent sclerodermatous cGVHD model, we confirmed that macrophages infiltrating the skin are derived from donor bone marrow (F4/80+CSF-1R+CD206+iNOS-). Cutaneous cGVHD developed in a CSF-1/CSF-1R-dependent manner, as treatment of recipients after transplantation with CSF-1 exacerbated macrophage infiltration and cutaneous pathology. Additionally, recipients of grafts from Csf1r-/- mice had substantially less macrophage infiltration and cutaneous pathology as compared with those receiving wild-type grafts. Neither CCL2/CCR2 nor GM-CSF/GM-CSFR signaling pathways were required for macrophage infiltration or development of cGVHD. In a different cGVHD model, in which bronchiolitis obliterans is a prominent manifestation, F4/80+ macrophage infiltration was similarly noted in the lungs of recipients after transplantation, and lung cGVHD was also IL-17 and CSF-1/CSF-1R dependent. Importantly, depletion of macrophages using an anti-CSF-1R mAb markedly reduced cutaneous and pulmonary cGVHD. Taken together, these data indicate that donor macrophages mediate the development of cGVHD and suggest that targeting CSF-1 signaling after transplantation may prevent and treat cGVHD.


Subject(s)
Graft vs Host Disease/pathology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/cytology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Antibodies, Monoclonal/chemistry , Cell Separation , Disease Models, Animal , Female , Flow Cytometry , Interleukin-17/metabolism , Lung/pathology , Lung Diseases/pathology , Macrophage Colony-Stimulating Factor/genetics , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/cytology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Signal Transduction , Skin/pathology , Stem Cell Transplantation
12.
J Immunol ; 192(11): 5426-33, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24790149

ABSTRACT

The stimulation of naive donor T cells by recipient alloantigen is central to the pathogenesis of graft-versus-host disease after bone marrow transplantation (BMT). Using mouse models of transplantation, we have observed that donor cells become "cross-dressed" in very high levels of recipient hematopoietic cell-derived MHC class I and II molecules following BMT. Recipient-type MHC is transiently present on donor dendritic cells (DCs) after BMT in the setting of myeloablative conditioning but is persistent after nonmyeloablative conditioning, in which recipient hematopoietic cells remain in high numbers. Despite the high level of recipient-derived alloantigen present on the surface of donor DCs, donor T cell proliferative responses are generated only in response to processed recipient alloantigen presented via the indirect pathway and not in response to cross-dressed MHC. Assays in which exogenous peptide is added to cross-dressed MHC in the presence of naive TCR transgenic T cells specific to the MHC class II-peptide combination confirm that cross-dressed APC cannot induce T cell proliferation in isolation. Despite failure to induce T cell proliferation, cross-dressing by donor DCs contributes to generation of the immunological synapse between DCs and CD4 T cells, and this is required for maximal responses induced by classical indirectly presented alloantigen. We conclude that the process of cross-dressing by donor DCs serves as an efficient alternative pathway for the acquisition of recipient alloantigen and that once acquired, this cross-dressed MHC can assist in immune synapse formation prior to the induction of full T cell proliferative responses by concurrent indirect Ag presentation.


Subject(s)
Antigen Presentation , Antigens/immunology , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Dendritic Cells/immunology , Immunological Synapses/immunology , Allografts , Animals , Histocompatibility Antigens Class II/immunology , Mice , Mice, Knockout , Peptides/immunology
13.
J Immunol ; 191(10): 5291-303, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24123683

ABSTRACT

Natural regulatory T cells (nTregs) play an important role in tolerance; however, the small numbers of cells obtainable potentially limit the feasibility of clinical adoptive transfer. Therefore, we studied the feasibility and efficacy of using murine-induced regulatory T cells (iTregs) for the induction of tolerance after bone marrow transplantation. iTregs could be induced in large numbers from conventional donor CD4 and CD8 T cells within 1 wk and were highly suppressive. During graft-versus-host disease (GVHD), CD4 and CD8 iTregs suppressed the proliferation of effector T cells and the production of proinflammatory cytokines. However, unlike nTregs, both iTreg populations lost Foxp3 expression within 3 wk in vivo, reverted to effector T cells, and exacerbated GVHD. The loss of Foxp3 in iTregs followed homeostatic and/or alloantigen-driven proliferation and was unrelated to GVHD. However, the concurrent administration of rapamycin, with or without IL-2/anti-IL-2 Ab complexes, to the transplant recipients significantly improved Foxp3 stability in CD4 iTregs (and, to a lesser extent, CD8 iTregs), such that they remained detectable 12 wk after transfer. Strikingly, CD4, but not CD8, iTregs could then suppress Teff proliferation and proinflammatory cytokine production and prevent GVHD in an equivalent fashion to nTregs. However, at high numbers and when used as GVHD prophylaxis, Tregs potently suppress graft-versus-leukemia effects and so may be most appropriate as a therapeutic modality to treat GVHD. These data demonstrate that CD4 iTregs can be produced rapidly in large, clinically relevant numbers and, when transferred in the presence of systemic rapamycin and IL-2, induce tolerance in transplant recipients.


Subject(s)
Graft vs Host Disease/immunology , Immune Tolerance/immunology , Interleukin-2/metabolism , Sirolimus/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow Transplantation , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation , Cytokines/biosynthesis , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/metabolism , Graft vs Host Disease/prevention & control , Immune Tolerance/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects
14.
Blood ; 121(17): 3511-20, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23430112

ABSTRACT

Donor T cells play pivotal roles in graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL) effects following bone marrow transplantation (BMT). DNAX accessory molecule 1 (DNAM-1) is a costimulatory and adhesion molecule, expressed mainly by natural killer cells and CD8(+) T cells at steady state to promote adhesion to ligand-expressing targets and enhance cytolysis. We have analyzed the role of this pathway in GVHD and GVL. The absence of DNAM-1 on the donor graft attenuated GVHD in major histocompatibility complex (MHC)-mismatched and MHC-matched BMT following conditioning with lethal and sublethal irradiation. In contrast, DNAM-1 was not critical for GVL effects against ligand (CD155) expressing and nonexpressing leukemia. The effects on GVHD following myeloablative conditioning were independent of CD8(+) T cells and dependent on CD4(+) T cells, and specifically donor FoxP3(+) regulatory T cells (Treg). The absence of DNAM-1 promoted the expansion and suppressive function of Treg after BMT. These findings provide support for therapeutic DNAM-1 inhibition to promote tolerance in relevant inflammatory-based diseases characterized by T-cell activation.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/physiology , Bone Marrow Transplantation , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect/immunology , Leukemia, Experimental/prevention & control , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/chemistry , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Female , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Leukemia, Experimental/etiology , Leukemia, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Transplantation Conditioning , Tumor Cells, Cultured , Whole-Body Irradiation
15.
Blood ; 119(24): 5898-908, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22538855

ABSTRACT

FoxP3(+) confers suppressive properties and is confined to regulatory T cells (T(reg)) that potently inhibit autoreactive immune responses. In the transplant setting, natural CD4(+) T(reg) are critical in controlling alloreactivity and the establishment of tolerance. We now identify an important CD8(+) population of FoxP3(+) T(reg) that convert from CD8(+) conventional donor T cells after allogeneic but not syngeneic bone marrow transplantation. These CD8(+) T(reg) undergo conversion in the mesenteric lymph nodes under the influence of recipient dendritic cells and TGF-ß. Importantly, this population is as important for protection from GVHD as the well-studied natural CD4(+)FoxP3(+) population and is more potent in exerting class I-restricted and antigen-specific suppression in vitro and in vivo. Critically, CD8(+)FoxP3(+) T(reg) are exquisitely sensitive to inhibition by cyclosporine but can be massively and specifically expanded in vivo to prevent GVHD by coadministering rapamycin and IL-2 antibody complexes. CD8(+)FoxP3(+) T(reg) thus represent a new regulatory population with considerable potential to preferentially subvert MHC class I-restricted T-cell responses after bone marrow transplantation.


Subject(s)
Bone Marrow Transplantation , CD8-Positive T-Lymphocytes/cytology , Forkhead Transcription Factors/metabolism , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/cytology , Animals , Antibodies/administration & dosage , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Epitopes/immunology , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Immune Tolerance/drug effects , Interleukin-2/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Phenotype , Sirolimus/administration & dosage , Sirolimus/pharmacology , Survival Analysis , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/pharmacology , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...