Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Immunology ; 168(4): 640-653, 2023 04.
Article in English | MEDLINE | ID: mdl-36281956

ABSTRACT

Age-associated B cells (ABCs) are an immune cell subset linked to autoimmunity, infection and ageing, and whose pathophysiological importance was recently highlighted using single cell synovial tissue profiling. To elucidate their pathophysiological relevance, peripheral blood (PB) ABCs from early rheumatoid arthritis (eRA) patients naïve to disease-modifying anti-rheumatic drugs (DMARDs) were compared with their synovial fluid (SF) counterparts, and to PB ABCs from psoriatic arthritis patients and healthy controls. PB and SF B-cell subsets were phenotyped by multi-parameter flow cytometry, sorted and subjected to gene expression profiling (NanoString nCounter® Immunology V2 Panel) and functional characterization (stimulated cytokine measurements by immunoassay). PB ABCs of eRA patients, which are transcriptionally distinct from those of control cohorts, express chemokine receptors and adhesion molecules, such as CXCR3, that favour homing to inflammatory sites over lymphoid tissue. These cells are an activated, class-switched B-cell subset expressing high levels of HLA-DR, co-stimulatory molecules and T-bet. Their secretion profile includes IL-12p70 and IL-23 but low levels of IL-10. High surface expression of FcRL family members, including FcRL3, furthermore suggests a role for these cells in autoimmunity. Finally, and unlike in the periphery where they are rare, ABCs are the predominant B-cell subsets in SF. These observations indicate the predilection of ABCs for inflammatory tissue in RA, where their propensity for antigen presentation and pro-inflammatory phenotype may support autoimmune pathology. Their potential as a therapeutic target therefore warrants further study.


Subject(s)
Arthritis, Psoriatic , Arthritis, Rheumatoid , Humans , Synovial Fluid/metabolism , HLA-DR Antigens/metabolism , Receptors, Chemokine/metabolism
3.
J Relig Health ; 62(6): 3995-4015, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37697218

ABSTRACT

Police members can be exposed to morally transgressive events with potential for lasting psychosocial and spiritual harm. Through interviews with police members and police chaplains across Australia and New Zealand, this qualitative study explores the current role that police chaplains play in supporting members exposed to morally transgressive events. The availability of chaplains across police services and the close alignment between the support they offer, and the support sought by police, indicates they have an important role. However, a holistic approach should also consider organizational factors, the role of leaders, and access to evidence-based treatment in collaboration with mental health practitioners.


Subject(s)
Pastoral Care , Stress Disorders, Post-Traumatic , Humans , Spirituality , Clergy/psychology , New Zealand , Police , Australia , Morals
4.
Immunity ; 38(5): 998-1012, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23684987

ABSTRACT

At sites of inflammation, certain regulatory T cells (Treg cells) can undergo rapid reprogramming into helper-like cells without loss of the transcription factor Foxp3. We show that reprogramming is controlled by downregulation of the transcription factor Eos (Ikzf4), an obligate corepressor for Foxp3. Reprogramming was restricted to a specific subset of "Eos-labile" Treg cells that was present in the thymus and identifiable by characteristic surface markers and DNA methylation. Mice made deficient in this subset became impaired in their ability to provide help for presentation of new antigens to naive T cells. Downregulation of Eos required the proinflammatory cytokine interleukin-6 (IL-6), and mice lacking IL-6 had impaired development and function of the Eos-labile subset. Conversely, the immunoregulatory enzyme IDO blocked loss of Eos and prevented the Eos-labile Treg cells from reprogramming. Thus, the Foxp3(+) lineage contains a committed subset of Treg cells capable of rapid conversion into biologically important helper cells.


Subject(s)
Carrier Proteins/metabolism , Ikaros Transcription Factor/metabolism , Interleukin-6/metabolism , Nerve Tissue Proteins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Differentiation/immunology , DNA-Binding Proteins , Down-Regulation , Forkhead Transcription Factors/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-6/genetics , Lymphocyte Activation/immunology , Mice , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymus Gland
5.
Am J Transplant ; 21(4): 1402-1414, 2021 04.
Article in English | MEDLINE | ID: mdl-32506663

ABSTRACT

Ex vivo normothermic machine perfusion (NMP) of donor kidneys prior to transplantation provides a platform for direct delivery of cellular therapeutics to optimize organ quality prior to transplantation. Multipotent Adult Progenitor Cells (MAPC® ) possess potent immunomodulatory properties that could minimize ischemia reperfusion injury. We investigated the potential capability of MAPC cells in kidney NMP. Pairs (5) of human kidneys, from the same donor, were simultaneously perfused for 7 hours. Kidneys were randomly allocated to receive MAPC treatment or control. Serial samples of perfusate, urine, and tissue biopsies were taken for comparison. MAPC-treated kidneys demonstrated improved urine output (P = .009), decreased expression of injury biomarker NGAL (P = .012), improved microvascular perfusion on contrast-enhanced ultrasound (cortex P = .019, medulla P = .001), downregulation of interleukin (IL)-1ß (P = .050), and upregulation of IL-10 (P < .047) and Indolamine-2, 3-dioxygenase (P = .050). A chemotaxis model demonstrated decreased neutrophil recruitment when stimulated with perfusate from MAPC-treated kidneys (P < .001). Immunofluorescence revealed prelabeled MAPC cells in the perivascular space of kidneys during NMP. We report the first successful delivery of cellular therapy to a human kidney during NMP. Kidneys treated with MAPC cells demonstrate improvement in clinically relevant parameters and injury biomarkers. This novel method of cell therapy delivery provides an exciting opportunity to recondition organs prior to transplantation.


Subject(s)
Kidney Transplantation , Reperfusion Injury , Cell- and Tissue-Based Therapy , Humans , Kidney , Kidney Transplantation/adverse effects , Organ Preservation , Perfusion , Reperfusion Injury/prevention & control
6.
Clin Otolaryngol ; 46(5): 919-934, 2021 09.
Article in English | MEDLINE | ID: mdl-34053179

ABSTRACT

BACKGROUND: Novel cancer immunotherapy seeks to harness the body's own immune system and tip the balance in favour of antitumour activity. The intracellular enzyme indoleamine 2,3-dioxygenase (IDO) is a critical regulator of the tumour microenvironment (TME) via tryptophan metabolism. The potential immunotherapeutic role of IDO in head and neck squamous cell carcinoma (HNSCC) requires further exploration. We aim to assess the evidence on IDO in HNSCC. METHODS: A systematic review of literature and clinical trials databases. RESULTS: We included 40 studies: seven involved cell lines: eight assessed tumour immunohistochemistry: ten measured IDO gene transcription: 15 reported on clinical trials. Increased cell line IDO expression was postulated to adversely affect tumour metabolism and apoptosis. Immunohistochemical IDO expression correlated with worse survival. Gene transcription studies associated IDO with positive PD-L1 and human papillomavirus (HPV) status. Phase I/II clinical trials showed (a) overall response (34%-55%) and disease control rates (62%-70%) for IDO1 inhibitor in combination with a PD-1 inhibitor, (b) similar safety profiles when both are used in combination therapy compared to each as monotherapies and (c) IDO gene expression as a predictive biomarker for response to PD-L1 therapy. CONCLUSIONS: IDO expression is increased in the TME of HNSCC, which correlates with poor prognosis. However, the exact mechanism of IDO-driven immune modulation in the TME is an enigma. Future translational studies should map IDO activity during HNSCC treatment and elucidate its precise role in the TME, such research will underpin the development of clinical trials establishing the efficacy of IDO inhibitors in HNSCC.


Subject(s)
Immunotherapy/methods , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Squamous Cell Carcinoma of Head and Neck/enzymology , Squamous Cell Carcinoma of Head and Neck/immunology , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Humans , Tumor Microenvironment
7.
J Chem Phys ; 152(2): 024709, 2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31941300

ABSTRACT

A novel method of measuring the core level binding energies of multiple sized nanoparticles on the same substrate is demonstrated using the early stage of Au nanoparticle growth on reduced r-TiO2(110). This method employed in situ scanning tunneling microscopy (STM) and microfocused X-ray photoemission spectroscopy. An STM tip-shadowing method was used to synthesize patterned areas of Au nanoparticles on the substrate with different coverages and sizes. Patterns were identified and imaged using a UV photoelectron emission microscope. The Au 4f core level binding energies of the nanoparticles were investigated as a function of Au nanoparticle coverage and size. A combination of initial and final state effects modifies the binding energies of the Au 4f core levels as the nanoparticle size changes. When single Au atoms and Au3 clusters are present, the Au 4f7/2 binding energy, 84.42 eV, is similar to that observed at a high coverage (1.8 monolayer equivalent), resulting from a cancellation of initial and final state effects. As the coverage is increased, there is a decrease in binding energy, which then increases at a higher coverage to 84.39 eV. These results are consistent with a Volmer-Weber nucleation-growth model of Au nanoparticles at oxygen vacancies, resulting in electron transfer to the nanoparticles.

8.
Immunology ; 158(4): 353-361, 2019 12.
Article in English | MEDLINE | ID: mdl-31557322

ABSTRACT

Reagents that activate the signaling adaptor stimulator of interferon genes (STING) suppress experimentally induced autoimmunity in murine models of multiple sclerosis and arthritis. In this study, we evaluated STING agonists as potential reagents to inhibit spontaneous autoimmune type I diabetes (T1D) onset in non-obese diabetic (NOD) female mice. Treatments with DNA nanoparticles (DNPs), which activate STING when cargo DNA is sensed, delayed T1D onset and reduced T1D incidence when administered before T1D onset. DNP treatment elevated indoleamine 2,3 dioxygenase (IDO) activity, which regulates T-cell immunity, in spleen, pancreatic lymph nodes and pancreas of NOD mice. Therapeutic responses to DNPs were partially reversed by inhibiting IDO and DNP treatment synergized with insulin therapy to further delay T1D onset and reduce T1D incidence. Treating pre-diabetic NOD mice with cyclic guanyl-adenyl dinucleotide (cGAMP) to activate STING directly delayed T1D onset and stimulated interferon-αß (IFN-αß), while treatment with cyclic diguanyl nucleotide (cdiGMP) did not delay T1D onset or induce IFN-αß in NOD mice. DNA sequence analyses revealed that NOD mice possess a STING polymorphism that may explain differential responses to cGAMP and cdiGMP. In summary, STING agonists attenuate T1D progression and DNPs enhance therapeutic responses to insulin therapy.


Subject(s)
DNA/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Insulin/therapeutic use , Membrane Proteins/agonists , Nanoparticles/therapeutic use , T-Lymphocytes/immunology , Animals , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , DNA/chemistry , Disease Models, Animal , Drug Synergism , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Nanoparticles/chemistry , Nucleotides, Cyclic/metabolism , Polymorphism, Genetic , Up-Regulation
9.
Trends Immunol ; 37(3): 193-207, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26839260

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) has immunoregulatory roles associated with tryptophan metabolism. These include counter-regulation (controlling inflammation) and acquired tolerance in T cells. Recent findings reveal that IDO can be triggered by innate responses during tumorigenesis, and also by attempted T cell activation, either spontaneous or due to immunotherapy. Here we review the current understanding of mechanisms by which IDO participates in the control of inflammation and in peripheral tolerance. Focusing on the tumor microenvironment, we examine the role of IDO in response to apoptotic cells and the impact of IDO on Treg cell function. We discuss how the counter-regulatory and tolerogenic functions of IDO can be targeted for cancer immunotherapy and present an overview of the current clinical progress in this area.


Subject(s)
Immune Tolerance , Immunosuppression Therapy , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Inflammation , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Tumor Microenvironment
10.
Immunity ; 33(6): 942-54, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21145762

ABSTRACT

Foxp3(+) regulatory T (Treg) cells can undergo reprogramming into a phenotype expressing proinflammatory cytokines. However, the biologic significance of this conversion remains unclear. We show that large numbers of Treg cells undergo rapid reprogramming into activated T helper cells after vaccination with antigen plus Toll-like receptor 9 (TLR-9) ligand. Helper activity from converted Treg cells proved essential during initial priming of CD8(+) T cells to a new cross-presented antigen. Help from Treg cells was dependent on CD40L, and (unlike help from conventional non-Treg CD4(+) cells) did not require preactivation or prior exposure to antigen. In hosts with established tumors, Treg cell reprogramming was suppressed by tumor-induced indoleamine 2,3-dioxygenase (IDO) and vaccination failed because of lack of help. Treg cell reprogramming, vaccine efficacy, and antitumor CD8(+) T cell responses were restored by pharmacologic inhibition of IDO. Reprogrammed Treg cells can thus participate as previously unrecognized drivers of certain early CD8(+) T cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cross-Priming , Melanoma, Experimental/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/metabolism , Adoptive Transfer , Animals , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Transdifferentiation/drug effects , Cells, Cultured , Cross-Priming/drug effects , Forkhead Transcription Factors/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Lymphocyte Activation/drug effects , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred Strains , Oligodeoxyribonucleotides/administration & dosage , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Toll-Like Receptor 9/immunology , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
11.
J Immunol ; 198(6): 2286-2301, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28193829

ABSTRACT

The transmembrane protein CD83, expressed on APCs, B cells, and T cells, can be expressed as a soluble form generated by alternative splice variants and/or by shedding. Soluble CD83 (sCD83) was shown to be involved in negatively regulating the immune response. sCD83 inhibits T cell proliferation in vitro, supports allograft survival in vivo, prevents corneal transplant rejection, and attenuates the progression and severity of autoimmune diseases and experimental colitis. Although sCD83 binds to human PBMCs, the specific molecules that bind sCD83 have not been identified. In this article, we identify myeloid differentiation factor-2 (MD-2), the coreceptor within the TLR4/MD-2 receptor complex, as the high-affinity sCD83 binding partner. TLR4/MD-2 mediates proinflammatory signal delivery following recognition of bacterial LPSs. However, altering TLR4 signaling can attenuate the proinflammatory cascade, leading to LPS tolerance. Our data show that binding of sCD83 to MD-2 alters this signaling cascade by rapidly degrading IL-1R-associated kinase-1, leading to induction of the anti-inflammatory mediators IDO, IL-10, and PGE2 in a COX-2-dependent manner. sCD83 inhibited T cell proliferation, blocked IL-2 secretion, and rendered T cells unresponsive to further downstream differentiation signals mediated by IL-2. Therefore, we propose the tolerogenic mechanism of action of sCD83 to be dependent on initial interaction with APCs, altering early cytokine signal pathways and leading to T cell unresponsiveness.


Subject(s)
Antigens, CD/metabolism , Immunoglobulins/metabolism , Membrane Glycoproteins/metabolism , Monocytes/immunology , T-Lymphocytes/immunology , Cell Proliferation , Cells, Cultured , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-10/metabolism , Interleukin-2/metabolism , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/immunology , Lymphocyte Activation , Lymphocyte Antigen 96/metabolism , Protein Binding , Signal Transduction , Toll-Like Receptor 4/metabolism , CD83 Antigen
12.
PLoS Pathog ; 12(5): e1005615, 2016 05.
Article in English | MEDLINE | ID: mdl-27168185

ABSTRACT

Increased pain sensitivity is a comorbidity associated with many clinical diseases, though the underlying causes are poorly understood. Recently, chronic pain hypersensitivity in rodents treated to induce chronic inflammation in peripheral tissues was linked to enhanced tryptophan catabolism in brain mediated by indoleamine 2,3 dioxygenase (IDO). Here we show that acute influenza A virus (IAV) and chronic murine leukemia retrovirus (MuLV) infections, which stimulate robust IDO expression in lungs and lymphoid tissues, induced acute or chronic pain hypersensitivity, respectively. In contrast, virus-induced pain hypersensitivity did not manifest in mice lacking intact IDO1 genes. Spleen IDO activity increased markedly as MuLV infections progressed, while IDO1 expression was not elevated significantly in brain or spinal cord (CNS) tissues. Moreover, kynurenine (Kyn), a tryptophan catabolite made by cells expressing IDO, incited pain hypersensitivity in uninfected IDO1-deficient mice and Kyn potentiated pain hypersensitivity due to MuLV infection. MuLV infection stimulated selective IDO expression by a discreet population of spleen cells expressing both B cell (CD19) and dendritic cell (CD11c) markers (CD19+ DCs). CD19+ DCs were more susceptible to MuLV infection than B cells or conventional (CD19neg) DCs, proliferated faster than B cells from early stages of MuLV infection and exhibited mature antigen presenting cell (APC) phenotypes, unlike conventional (CD19neg) DCs. Moreover, interactions with CD4 T cells were necessary to sustain functional IDO expression by CD19+ DCs in vitro and in vivo. Splenocytes from MuLV-infected IDO1-sufficient mice induced pain hypersensitivity in uninfected IDO1-deficient recipient mice, while selective in vivo depletion of DCs alleviated pain hypersensitivity in MuLV-infected IDO1-sufficient mice and led to rapid reduction in splenomegaly, a hallmark of MuLV immune pathogenesis. These findings reveal critical roles for CD19+ DCs expressing IDO in host responses to MuLV infection that enhance pain hypersensitivity and cause immune pathology. Collectively, our findings support the hypothesis elevated IDO activity in non-CNS due to virus infections causes pain hypersensitivity mediated by Kyn. Previously unappreciated links between host immune responses to virus infections and pain sensitivity suggest that IDO inhibitors may alleviate heightened pain sensitivity during infections.


Subject(s)
Hyperalgesia/enzymology , Hyperalgesia/etiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Virus Diseases/complications , Animals , Disease Models, Animal , Flow Cytometry , Kynurenine/metabolism , Mice , Polymerase Chain Reaction
13.
J Immunol ; 194(4): 2011-21, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25560408

ABSTRACT

In recent years, the immune-potentiating effects of some widely used chemotherapeutic agents have been increasingly appreciated. This provides a rationale for combining conventional chemotherapy with immunotherapy strategies to achieve durable therapeutic benefits. Previous studies have implicated the immunomodulatory effects of melphalan, an alkylating agent commonly used to treat multiple myeloma, but the underlying mechanisms remain obscure. In the present study, we investigated the impact of melphalan on endogenous immune cells as well as adoptively transferred tumor-specific CD4(+) T cells in tumor-bearing mice. We showed that melphalan treatment resulted in a rapid burst of inflammatory cytokines and chemokines during the cellular recovery phase after melphalan-induced myelodepletion and leukodepletion. After melphalan treatment, tumor cells exhibited characteristics of immunogenic cell death, including membrane translocation of the endoplasmic reticulum-resident calreticulin and extracellular release of high-mobility group box 1. Additionally, there was enhanced tumor Ag uptake by dendritic cells in the tumor-draining lymph node. Consistent with these immunomodulatory effects, melphalan treatment of tumor-bearing mice led to the activation of the endogenous CD8(+) T cells and, more importantly, effectively drove the clonal expansion and effector differentiation of adoptively transferred tumor-specific CD4(+) T cells. Notably, the combination of melphalan and CD4(+) T cell adoptive cell therapy was more efficacious than either treatment alone in prolonging the survival of mice with advanced B cell lymphomas or colorectal tumors. These findings provide mechanistic insights into melphalan's immunostimulatory effects and demonstrate the therapeutic potential of combining melphalan with adoptive cell therapy utilizing antitumor CD4(+) T cells.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , CD4-Positive T-Lymphocytes/transplantation , Immunotherapy, Adoptive/methods , Melphalan/administration & dosage , Neoplasms, Experimental/therapy , Animals , Blotting, Western , Combined Modality Therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Transgenic
14.
J Immunol ; 194(12): 5713-24, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25980011

ABSTRACT

Inflammatory kidney disease is a major clinical problem that can result in end-stage renal failure. In this article, we show that Ab-mediated inflammatory kidney injury and renal disease in a mouse nephrotoxic serum nephritis model was inhibited by amino acid metabolism and a protective autophagic response. The metabolic signal was driven by IFN-γ-mediated induction of indoleamine 2,3-dioxygenase 1 (IDO1) enzyme activity with subsequent activation of a stress response dependent on the eIF2α kinase general control nonderepressible 2 (GCN2). Activation of GCN2 suppressed proinflammatory cytokine production in glomeruli and reduced macrophage recruitment to the kidney during the incipient stage of Ab-induced glomerular inflammation. Further, inhibition of autophagy or genetic ablation of Ido1 or Gcn2 converted Ab-induced, self-limiting nephritis to fatal end-stage renal disease. Conversely, increasing kidney IDO1 activity or treating mice with a GCN2 agonist induced autophagy and protected mice from nephritic kidney damage. Finally, kidney tissue from patients with Ab-driven nephropathy showed increased IDO1 abundance and stress gene expression. Thus, these findings support the hypothesis that the IDO-GCN2 pathway in glomerular stromal cells is a critical negative feedback mechanism that limits inflammatory renal pathologic changes by inducing autophagy.


Subject(s)
Amino Acids/metabolism , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/metabolism , Autoantibodies/immunology , Autophagy/immunology , Animals , Anti-Glomerular Basement Membrane Disease/genetics , Anti-Glomerular Basement Membrane Disease/pathology , Cytokines/biosynthesis , Disease Models, Animal , Enzyme Activation , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Mice, Knockout , Podocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Stress, Physiological
15.
J Immunol ; 194(5): 2369-79, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25646302

ABSTRACT

During hematopoiesis, hematopoietic stem cells constantly differentiate into granulocytes and macrophages via a distinct differentiation program that is tightly controlled by myeloid lineage-specific transcription factors. Mice with a null mutation of IFN regulatory factor 8 (IRF8) accumulate CD11b(+)Gr1(+) myeloid cells that phenotypically and functionally resemble tumor-induced myeloid-derived suppressor cells (MDSCs), indicating an essential role of IRF8 in myeloid cell lineage differentiation. However, IRF8 is expressed in various types of immune cells, and whether IRF8 functions intrinsically or extrinsically in regulation of myeloid cell lineage differentiation is not fully understood. In this study, we report an intriguing finding that, although IRF8-deficient mice exhibit deregulated myeloid cell differentiation and resultant accumulation of CD11b(+)Gr1(+) MDSCs, surprisingly, mice with IRF8 deficiency only in myeloid cells exhibit no abnormal myeloid cell lineage differentiation. Instead, mice with IRF8 deficiency only in T cells exhibited deregulated myeloid cell differentiation and MDSC accumulation. We further demonstrated that IRF8-deficient T cells exhibit elevated GM-CSF expression and secretion. Treatment of mice with GM-CSF increased MDSC accumulation, and adoptive transfer of IRF8-deficient T cells, but not GM-CSF-deficient T cells, increased MDSC accumulation in the recipient chimeric mice. Moreover, overexpression of IRF8 decreased GM-CSF expression in T cells. Our data determine that, in addition to its intrinsic function as an apoptosis regulator in myeloid cells, IRF8 also acts extrinsically to repress GM-CSF expression in T cells to control myeloid cell lineage differentiation, revealing a novel mechanism that the adaptive immune component of the immune system regulates the innate immune cell myelopoiesis in vivo.


Subject(s)
Cell Lineage/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interferon Regulatory Factors/immunology , Myeloid Cells/immunology , Myelopoiesis/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Proliferation , Chimera , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Mice , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myelopoiesis/drug effects , Myelopoiesis/genetics , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/transplantation
16.
J Immunol ; 194(7): 3295-304, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25710911

ABSTRACT

Breakdown in immunological tolerance to self-Ags or uncontrolled inflammation results in autoimmune disorders. Dendritic cells (DCs) play an important role in regulating the balance between inflammatory and regulatory responses in the periphery. However, factors in the tissue microenvironment and the signaling networks critical for programming DCs to control chronic inflammation and promote tolerance are unknown. In this study, we show that wnt ligand-mediated activation of ß-catenin signaling in DCs is critical for promoting tolerance and limiting neuroinflammation. DC-specific deletion of key upstream (lipoprotein receptor-related protein [LRP]5/6) or downstream (ß-catenin) mediators of canonical wnt signaling in mice exacerbated experimental autoimmune encephalomyelitis pathology. Mechanistically, loss of LRP5/6-ß-catenin-mediated signaling in DCs led to an increased Th1/Th17 cell differentiation but reduced regulatory T cell response. This was due to increased production of proinflammatory cytokines and decreased production of anti-inflammatory cytokines such as IL-10 and IL-27 by DCs lacking LRP5/6-ß-catenin signaling. Consistent with these findings, pharmacological activation of canonical wnt/ß-catenin signaling delayed experimental autoimmune encephalomyelitis onset and diminished CNS pathology. Thus, the activation of canonical wnt signaling in DCs limits effector T cell responses and represents a potential therapeutic approach to control autoimmune neuroinflammation.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway , Animals , Cell Differentiation , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Deletion , Gene Knockout Techniques , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Male , Mice , Mice, Transgenic , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/cytology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Wnt Signaling Pathway/drug effects , Wnt3A Protein/metabolism , beta Catenin/metabolism
17.
Proc Natl Acad Sci U S A ; 111(11): 4215-20, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24591636

ABSTRACT

Tolerance to apoptotic cells is essential to prevent inflammatory pathology. Though innate responses are critical for immune suppression, our understanding of early innate immunity driven by apoptosis is lacking. Herein we report apoptotic cells induce expression of the chemokine CCL22 in splenic metallophillic macrophages, which is critical for tolerance. Systemic challenge with apoptotic cells induced rapid production of CCL22 in CD169(+) (metallophillic) macrophages, resulting in accumulation and activation of FoxP3(+) Tregs and CD11c(+) dendritic cells, an effect that could be inhibited by antagonizing CCL22-driven chemotaxis. This mechanism was essential for suppression after apoptotic cell challenge, because neutralizing CCL22 or its receptor, reducing Treg numbers, or blocking effector mechanisms abrogated splenic TGF-ß and IL-10 induction; this promoted a shift to proinflammatory cytokines associated with a failure to suppress T cells. Similarly, CCR4 inhibition blocked long-term, apoptotic cell-induced tolerance to allografts. Finally, CCR4 inhibition resulted in a systemic breakdown of tolerance to self after apoptotic cell injection with rapid increases in anti-dsDNA IgG and immune complex deposition. Thus, the data demonstrate CCL22-dependent chemotaxis is a key early innate response required for apoptotic cell-induced suppression, implicating a previously unknown mechanism of macrophage-dependent coordination of early events leading to stable tolerance.


Subject(s)
Apoptosis/immunology , Chemokine CCL22/immunology , Macrophages/immunology , Sialic Acid Binding Ig-like Lectin 1/immunology , Transplantation Tolerance/immunology , Animals , Cell Movement/physiology , Chemokine CCL22/metabolism , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Immunohistochemistry , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR4/genetics , Sialic Acid Binding Ig-like Lectin 1/metabolism , Transplantation Tolerance/genetics
18.
Trends Immunol ; 34(3): 137-43, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23103127

ABSTRACT

Sustained access to nutrients is a fundamental biological need, especially for proliferating cells, and controlling nutrient supply is an ancient strategy to regulate cellular responses to stimuli. By catabolizing the essential amino acid TRP, cells expressing the enzyme indoleamine 2,3 dioxygenase (IDO) can mediate potent local effects on innate and adaptive immune responses to inflammatory insults. Here, we discuss recent progress in elucidating how IDO activity promotes local metabolic changes that impact cellular and systemic responses to inflammatory and immunological signals. These recent developments identify potential new targets for therapy in a range of clinical settings, including cancer, chronic infections, autoimmune and allergic syndromes, and transplantation.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Animals , Communicable Diseases/immunology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Neoplasms/immunology , Tryptophan/metabolism
19.
J Immunol ; 193(8): 4203-13, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25210120

ABSTRACT

Dendritic cells (DCs) sense microbes via multiple innate receptors. Signals from different innate receptors are coordinated and integrated by DCs to generate specific innate and adaptive immune responses against pathogens. Previously, we have shown that two pathogen recognition receptors, TLR2 and dectin-1, which recognize the same microbial stimulus (zymosan) on DCs, induce mutually antagonistic regulatory or inflammatory responses, respectively. How diametric signals from these two receptors are coordinated in DCs to regulate or incite immunity is not known. In this study, we show that TLR2 signaling via AKT activates the ß-catenin/T cell factor 4 pathway in DCs and programs them to drive T regulatory cell differentiation. Activation of ß-catenin/T cell factor 4 was critical to induce regulatory molecules IL-10 (Il-10) and vitamin A metabolizing enzyme retinaldehyde dehydrogenase 2 (Aldh1a2) and to suppress proinflammatory cytokines. Deletion of ß-catenin in DCs programmed them to drive Th17/Th1 cell differentiation in response to zymosan. Consistent with these findings, activation of the ß-catenin pathway in DCs suppressed chronic inflammation and protected mice from Th17/Th1-mediated autoimmune neuroinflammation. Thus, activation of ß-catenin in DCs via the TLR2 receptor is a novel mechanism in DCs that regulates autoimmune inflammation.


Subject(s)
Autoimmunity/immunology , Dendritic Cells/immunology , T-Lymphocytes, Regulatory/cytology , Toll-Like Receptor 2/immunology , beta Catenin/metabolism , Adoptive Transfer , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase 1 Family , Animals , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Inflammation/prevention & control , Interleukin-10/biosynthesis , Lectins, C-Type/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/immunology , Retinal Dehydrogenase , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Transcription Factor 7-Like 2 Protein/immunology , Zymosan/immunology , Zymosan/pharmacology , beta Catenin/genetics
20.
J Immunol ; 192(12): 5571-8, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24799564

ABSTRACT

Cytosolic DNA sensing activates the stimulator of IFN genes (STING) adaptor to induce IFN type I (IFN-αß) production. Constitutive DNA sensing to induce sustained STING activation incites tolerance breakdown, leading to autoimmunity. In this study, we show that systemic treatments with DNA nanoparticles (DNPs) induced potent immune regulatory responses via STING signaling that suppressed experimental autoimmune encephalitis (EAE) when administered to mice after immunization with myelin oligodendrocyte glycoprotein (MOG), at EAE onset, or at peak disease severity. DNP treatments attenuated infiltration of effector T cells into the CNS and suppressed innate and adaptive immune responses to myelin oligodendrocyte glycoprotein immunization in spleen. Therapeutic responses were not observed in mice treated with cargo DNA or cationic polymers alone, indicating that DNP uptake and cargo DNA sensing by cells with regulatory functions was essential for therapeutic responses to manifest. Intact STING and IFN-αß receptor genes, but not IFN-γ receptor genes, were essential for therapeutic responses to DNPs to manifest. Treatments with cyclic diguanylate monophosphate to activate STING also delayed EAE onset and reduced disease severity. Therapeutic responses to DNPs were critically dependent on IDO enzyme activity in hematopoietic cells. Thus, DNPs and cyclic diguanylate monophosphate attenuate EAE by inducing dominant T cell regulatory responses via the STING/IFN-αß/IDO pathway that suppress CNS-specific autoimmunity. These findings reveal dichotomous roles for the STING/IFN-αß pathway in either stimulating or suppressing autoimmunity and identify STING-activating reagents as a novel class of immune modulatory drugs.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interferons/immunology , Membrane Proteins/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , DNA/genetics , DNA/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Immune Tolerance/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferons/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/toxicity , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Signal Transduction/genetics , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/pathology
SELECTION OF CITATIONS
SEARCH DETAIL