Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Immunity ; 51(5): 885-898.e7, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31542340

ABSTRACT

Graft-versus-host disease (GVHD) in the gastrointestinal (GI) tract is the principal determinant of lethality following allogeneic bone marrow transplantation (BMT). Here, we examined the mechanisms that initiate GVHD, including the relevant antigen-presenting cells. MHC class II was expressed on intestinal epithelial cells (IECs) within the ileum at steady state but was absent from the IECs of germ-free mice. IEC-specific deletion of MHC class II prevented the initiation of lethal GVHD in the GI tract. MHC class II expression on IECs was absent from mice deficient in the TLR adaptors MyD88 and TRIF and required IFNγ secretion by lamina propria lymphocytes. IFNγ responses are characteristically driven by IL-12 secretion from myeloid cells. Antibiotic-mediated depletion of the microbiota inhibited IL-12/23p40 production by ileal macrophages. IL-12/23p40 neutralization prevented MHC class II upregulation on IECs and initiation of lethal GVHD in the GI tract. Thus, MHC class II expression by IECs in the ileum initiates lethal GVHD, and blockade of IL-12/23p40 may represent a readily translatable therapeutic strategy.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Gastrointestinal Microbiome/immunology , Graft vs Host Disease/etiology , Histocompatibility Antigens Class II/immunology , Intestinal Mucosa/immunology , Animals , Antigen-Presenting Cells/metabolism , Biomarkers , Cytokines/metabolism , Disease Susceptibility , Female , Gene Expression , Graft vs Host Disease/mortality , Histocompatibility Antigens Class II/genetics , Ileum/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Kaplan-Meier Estimate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Male , Mice , Mice, Transgenic , Prognosis , Promoter Regions, Genetic , Signal Transduction
2.
Blood ; 138(8): 722-737, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34436524

ABSTRACT

Immunopathology and intestinal stem cell (ISC) loss in the gastrointestinal (GI) tract is the prima facie manifestation of graft-versus-host disease (GVHD) and is responsible for significant mortality after allogeneic bone marrow transplantation (BMT). Approaches to prevent GVHD to date focus on immune suppression. Here, we identify interferon-λ (IFN-λ; interleukin-28 [IL-28]/IL-29) as a key protector of GI GVHD immunopathology, notably within the ISC compartment. Ifnlr1-/- mice displayed exaggerated GI GVHD and mortality independent of Paneth cells and alterations to the microbiome. Ifnlr1-/- intestinal organoid growth was significantly impaired, and targeted Ifnlr1 deficiency exhibited effects intrinsic to recipient Lgr5+ ISCs and natural killer cells. PEGylated recombinant IL-29 (PEG-rIL-29) treatment of naive mice enhanced Lgr5+ ISC numbers and organoid growth independent of both IL-22 and type I IFN and modulated proliferative and apoptosis gene sets in Lgr5+ ISCs. PEG-rIL-29 treatment improved survival, reduced GVHD severity, and enhanced epithelial proliferation and ISC-derived organoid growth after BMT. The preservation of ISC numbers in response to PEG-rIL-29 after BMT occurred both in the presence and absence of IFN-λ-signaling in recipient natural killer cells. IFN-λ is therefore an attractive and rapidly testable approach to prevent ISC loss and immunopathology during GVHD.


Subject(s)
Bone Marrow Transplantation , Cytokines/pharmacology , Gastrointestinal Diseases , Graft vs Host Disease , Interleukins/pharmacokinetics , Signal Transduction , Animals , Cytokines/immunology , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/immunology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Interleukins/immunology , Mice , Mice, Knockout , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Severity of Illness Index , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Transplantation, Homologous
3.
PLoS Pathog ; 16(10): e1008994, 2020 10.
Article in English | MEDLINE | ID: mdl-33049000

ABSTRACT

Inflammation is critical for controlling pathogens, but also responsible for symptoms of infectious diseases. IL-27 is an important regulator of inflammation and can limit development of IFNγ-producing Tbet+ CD4+ T (Th1) cells. IL-27 is thought to do this by stimulating IL-10 production by CD4+ T cells, but the underlying mechanisms of these immunoregulatory pathways are not clear. Here we studied the role of IL-27 signalling in experimental visceral leishmaniasis (VL) caused by infection of C57BL/6 mice with the human pathogen Leishmania donovani. We found IL-27 signalling was critical for the development of IL-10-producing Th1 (Tr1) cells during infection. Furthermore, in the absence of IL-27 signalling, there was improved control of parasite growth, but accelerated splenic pathology characterised by the loss of marginal zone macrophages. Critically, we discovered that IL-27 signalling limited glycolysis in Th1 cells during infection that in turn attenuated inflammation. Furthermore, the modulation of glycolysis in the absence of IL-27 signalling restricted tissue pathology without compromising anti-parasitic immunity. Together, these findings identify a novel mechanism by which IL-27 mediates immune regulation during disease by regulating cellular metabolism.


Subject(s)
Interleukins/metabolism , Leishmaniasis, Visceral/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Female , Glycolysis , Interferon-gamma/immunology , Interleukins/immunology , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology , Spleen/immunology
4.
Blood ; 134(23): 2092-2106, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31578204

ABSTRACT

Graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (alloSCT) is characterized by interleukin-6 (IL-6) dysregulation. IL-6 can mediate effects via various pathways, including classical, trans, and cluster signaling. Given the recent availability of agents that differentially inhibit these discrete signaling cascades, understanding the source and signaling and cellular targets of this cytokine is paramount to inform the design of clinical studies. Here we demonstrate that IL-6 secretion from recipient dendritic cells (DCs) initiates the systemic dysregulation of this cytokine. Inhibition of DC-driven classical signaling after targeted IL-6 receptor (IL-6R) deletion in T cells eliminated pathogenic donor Th17/Th22 cell differentiation and resulted in long-term survival. After engraftment, donor DCs assume the same role, maintaining classical IL-6 signaling-dependent GVHD responses. Surprisingly, cluster signaling was not active after transplantation, whereas inhibition of trans signaling with soluble gp130Fc promoted severe, chronic cutaneous GVHD. The latter was a result of exaggerated polyfunctional Th22-cell expansion that was reversed by IL-22 deletion or IL-6R inhibition. Importantly, inhibition of IL-6 classical signaling did not impair the graft-versus-leukemia effect. Together, these data highlight IL-6 classical signaling and downstream Th17/Th22 differentiation as important therapeutic targets after alloSCT.


Subject(s)
Dendritic Cells/immunology , Graft vs Host Disease/immunology , Interleukin-6/immunology , Signal Transduction/immunology , Skin Diseases/immunology , Stem Cell Transplantation , Allografts , Animals , Cell Differentiation/immunology , Dendritic Cells/pathology , Gene Deletion , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Graft vs Leukemia Effect/genetics , Graft vs Leukemia Effect/immunology , Interleukin-6/genetics , Interleukins/genetics , Interleukins/immunology , Mice , Mice, Transgenic , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/immunology , Signal Transduction/genetics , Skin Diseases/genetics , Skin Diseases/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Interleukin-22
5.
Blood ; 132(16): 1675-1688, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30154111

ABSTRACT

Autologous stem cell transplantation (SCT) remains a standard of care for multiple myeloma (MM) patients and prolongs progression-free survival. A small cohort of patients achieve long-term control of disease, but the majority of patients ultimately relapse, and the mechanisms permitting disease progression remain unclear. In this study, we used a preclinical model of autologous SCT for myeloma where the disease either progressed (MM relapsed) or was controlled. In the bone marrow (BM), inhibitory receptor expression on CD8+ T cells correlated strongly with myeloma progression after transplant. In conjunction, the costimulatory/adhesion receptor CD226 (DNAM-1) was markedly downregulated. Interestingly, DNAM-1- CD8+ T cells in MM-relapsed mice had an exhausted phenotype, characterized by upregulation of multiple inhibitory receptors, including T-cell immunoglobulin and ITIM domains (TIGIT) and programmed cell death protein 1 (PD-1) with decreased T-bet and increased eomesodermin expression. Immune checkpoint blockade using monoclonal antibodies against PD-1 or TIGIT significantly prolonged myeloma control after SCT. Furthermore, CD8+ T cells from MM-relapsed mice exhibited high interleukin-10 (IL-10) secretion that was associated with increased TIGIT and PD-1 expression. However, while donor-derived IL-10 inhibited myeloma control post-SCT, this was independent of IL-10 secretion by or signaling to T cells. Instead, the donor myeloid compartment, including colony-stimulating factor 1 receptor-dependent macrophages and an IL-10-secreting dendritic cell population in the BM, promoted myeloma progression. Our findings highlight PD-1 or TIGIT blockade in conjunction with SCT as a potent combination therapy in the treatment of myeloma.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, Differentiation, T-Lymphocyte/metabolism , CD8-Positive T-Lymphocytes/immunology , Interleukin-10/physiology , Multiple Myeloma/prevention & control , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , Cells, Cultured , Hematopoietic Stem Cell Transplantation/adverse effects , Mice , Mice, Knockout , Multiple Myeloma/etiology , Multiple Myeloma/pathology , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/immunology
6.
Blood ; 129(15): 2172-2185, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28137828

ABSTRACT

Donor T-cell-derived interleukin-17A (IL-17A) can mediate late immunopathology in graft-versus-host disease (GVHD), however protective roles remain unclear. Using multiple cytokine and cytokine receptor subunit knockout mice, we demonstrate that stem cell transplant recipients lacking the ability to generate or signal IL-17 develop intestinal hyper-acute GVHD. This protective effect is restricted to the molecular interaction of IL-17A and/or IL-17F with the IL-17 receptor A/C (IL-17RA/C). The protection from GVHD afforded by IL-17A required secretion from, and signaling in, both hematopoietic and nonhematopoietic host tissue. Given the intestinal-specificity of the disease in these animals, we cohoused wild-type (WT) with IL-17RA and IL-17RC-deficient mice, which dramatically enhanced the susceptibility of WT mice to acute GVHD. Furthermore, the gut microbiome of WT mice shifted toward that of the IL-17RA/C mice during cohousing prior to transplant, confirming that an IL-17-sensitive gut microbiota controls susceptibility to acute GVHD. Finally, induced IL-17A depletion peritransplant also enhanced acute GVHD, consistent with an additional protective role for this cytokine independent of effects on dysbiosis.


Subject(s)
Gastrointestinal Microbiome/immunology , Graft vs Host Disease , Interleukin-17/immunology , Intestinal Diseases , Acute Disease , Animals , Disease Models, Animal , Dysbiosis/genetics , Dysbiosis/immunology , Dysbiosis/pathology , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Interleukin-17/genetics , Intestinal Diseases/genetics , Intestinal Diseases/immunology , Intestinal Diseases/pathology , Lymphocyte Transfusion , Mice , Mice, Knockout , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/immunology
7.
Blood ; 129(5): 630-642, 2017 02 02.
Article in English | MEDLINE | ID: mdl-27927647

ABSTRACT

Allogeneic bone marrow transplantation (allo-BMT) is a curative therapy for hematological malignancies, but is associated with significant complications, principally graft-versus-host disease (GVHD) and opportunistic infections. Natural killer (NK) cells mediate important innate immunity that provides a temporal bridge until the reconstruction of adaptive immunity. Here, we show that the development of GVHD after allo-BMT prevented NK-cell reconstitution, particularly within the maturing M1 and M2 NK-cell subsets in association with exaggerated activation, apoptosis, and autophagy. Donor T cells were critical in this process by limiting the availability of interleukin 15 (IL-15), and administration of IL-15/IL-15Rα or immune suppression with rapamycin could restore NK-cell reconstitution. Importantly, the NK-cell defect induced by GVHD resulted in the failure of NK-cell-dependent in vivo cytotoxicity and graft-versus-leukemia effects. Control of cytomegalovirus infection after allo-BMT was also impaired during GVHD. Thus, during GVHD, donor T cells compete with NK cells for IL-15 thereby inducing profound defects in NK-cell reconstitution that compromise both leukemia and pathogen-specific immunity.


Subject(s)
Bone Marrow Transplantation/adverse effects , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Graft vs Host Disease/immunology , Immunity, Innate , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Leukemia/immunology , Animals , Autophagy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/pathology , Female , Graft vs Host Disease/complications , Graft vs Host Disease/etiology , Graft vs Host Disease/pathology , Humans , Interleukin-15/immunology , Leukemia/complications , Leukemia/pathology , Leukemia/therapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous/adverse effects
8.
Am J Transplant ; 18(4): 810-820, 2018 04.
Article in English | MEDLINE | ID: mdl-28941323

ABSTRACT

Graft-versus-host disease (GVHD) is the major cause of nonrelapse morbidity and mortality after allogeneic stem cell transplantation (allo-SCT). Prevention and treatment of GVHD remain inadequate and commonly lead to end-organ dysfunction and opportunistic infection. The role of interleukin (IL)-17 and IL-22 in GVHD remains uncertain, due to an apparent lack of lineage fidelity and variable and contextually determined protective and pathogenic effects. We demonstrate that donor T cell-derived IL-22 significantly exacerbates cutaneous chronic GVHD and that IL-22 is produced by highly inflammatory donor CD4+ T cells posttransplantation. IL-22 and IL-17A derive from both independent and overlapping lineages, defined as T helper (Th)22 and IL-22+ Th17 cells. Donor Th22 and IL-22+ Th17 cells share a similar IL-6-dependent developmental pathway, and while Th22 cells arise independently of the IL-22+ Th17 lineage, IL-17 signaling to donor Th22 directly promotes their development in allo-SCT. Importantly, while both IL-22 and IL-17 mediate skin GVHD, Th17-induced chronic GVHD can be attenuated by IL-22 inhibition in preclinical systems. In the clinic, high levels of both IL-17A and IL-22 expression are present in the skin of patients with GVHD after allo-SCT. Together, these data demonstrate a key role for donor-derived IL-22 in patients with chronic skin GVHD and confirm parallel but symbiotic developmental pathways of Th22 and Th17 differentiation.


Subject(s)
Graft vs Host Disease/etiology , Interleukin-17/metabolism , Interleukins/metabolism , Skin Diseases/etiology , Stem Cell Transplantation/adverse effects , Tissue Donors , Animals , Chronic Disease , Female , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Prognosis , Skin Diseases/metabolism , Skin Diseases/pathology , Transplantation, Homologous , Interleukin-22
9.
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
10.
Blood ; 126(12): 1503-14, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26130706

ABSTRACT

Viral infection is a common, life-threatening complication after allogeneic bone marrow transplantation (BMT), particularly in the presence of graft-versus-host disease (GVHD). Using cytomegalovirus (CMV) as the prototypic pathogen, we have delineated the mechanisms responsible for the inability to mount protective antiviral responses in this setting. Although CMV infection was self-limiting after syngeneic BMT, in the presence of GVHD after allogeneic BMT, CMV induced a striking cytopathy resulting in universal mortality in conjunction with a fulminant necrotizing hepatitis. Critically, GVHD induced a profound dendritic cell (DC) defect that led to a failure in the generation of CMV-specific CD8(+) T-cell responses. This was accompanied by a defect in antiviral CD8(+) T cells. In combination, these defects dramatically limited antiviral T-cell responses. The transfer of virus-specific cells circumvented the DC defects and provided protective immunity, despite concurrent GVHD. These data demonstrate the importance of avoiding GVHD when reconstructing antiviral immunity after BMT, and highlight the mechanisms by which the adoptive transfer of virus-specific T cells overcome the endogenous defects in priming invoked by GVHD.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Cytomegalovirus Infections/etiology , Cytomegalovirus/immunology , Dendritic Cells/pathology , Graft vs Host Disease/complications , Adoptive Transfer , Animals , Bone Marrow Transplantation/adverse effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/therapy , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
11.
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
12.
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
13.
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
14.
J Immunol ; 195(12): 5707-17, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26538396

ABSTRACT

Intracellular infections, such as those caused by the protozoan parasite Leishmania donovani, a causative agent of visceral leishmaniasis (VL), require a potent host proinflammatory response for control. IL-17 has emerged as an important proinflammatory cytokine required for limiting growth of both extracellular and intracellular pathogens. However, there are conflicting reports on the exact roles for IL-17 during parasitic infections and limited knowledge about cellular sources and the immune pathways it modulates. We examined the role of IL-17 in an experimental model of VL caused by infection of C57BL/6 mice with L. donovani and identified an early suppressive role for IL-17 in the liver that limited control of parasite growth. IL-17-producing γδ T cells recruited to the liver in the first week of infection were the critical source of IL-17 in this model, and CCR2(+) inflammatory monocytes were an important target for the suppressive effects of IL-17. Improved parasite control was independent of NO generation, but associated with maintenance of superoxide dismutase mRNA expression in the absence of IL-17 in the liver. Thus, we have identified a novel inhibitory function for IL-17 in parasitic infection, and our results demonstrate important interactions among γδ T cells, monocytes, and infected macrophages in the liver that can determine the outcome of parasitic infection.


Subject(s)
Interleukin-17/metabolism , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Liver/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Humans , Immunosuppression Therapy , Leishmania donovani/growth & development , Liver/parasitology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/parasitology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, CCR2/metabolism , Superoxide Dismutase/metabolism , T-Lymphocytes/parasitology
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL