Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Int J Mol Sci ; 24(20)2023 Oct 12.
Article En | MEDLINE | ID: mdl-37894784

Idiopathic pulmonary fibrosis (IPF) is the most common and lethal form of the interstitial pneumonias. The cause of the disease is unknown, and new therapies that stop or reverse disease progression are desperately needed. Recent advances in next-generation sequencing have led to an abundance of freely available, clinically relevant, organ-and-disease-specific, single-cell transcriptomic data, including studies from patients with IPF. We mined data from published IPF data sets and identified gene signatures delineating pro-fibrotic or antifibrotic macrophages and then used the Enrichr platform to identify compounds with the potential to drive the macrophages toward the antifibrotic transcriptotype. We then began testing these compounds in a novel in vitro phenotypic drug screening assay utilising human lung macrophages recovered from whole-lung lavage of patients with silicosis. As predicted by the Enrichr tool, glitazones potently modulated macrophage gene expression towards the antifibrotic phenotype. Next, we assayed a subset of the NatureBank pure compound library and identified the cyclobutane lignan, endiandrin A, which was isolated from the roots of the endemic Australian rainforest plant, Endiandra anthropophagorum, with a similar antifibrotic potential to the glitazones. These methods open new avenues of exploration to find treatments for lung fibrosis.


Idiopathic Pulmonary Fibrosis , Thiazolidinediones , Humans , Australia , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Macrophages/metabolism , Thiazolidinediones/therapeutic use
2.
Sci Immunol ; 5(52)2020 Oct 23.
Article En | MEDLINE | ID: mdl-33097591

Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease in which airway macrophages (AMs) play a key role. Itaconate has emerged as a mediator of macrophage function, but its role during fibrosis is unknown. Here, we reveal that itaconate is an endogenous antifibrotic factor in the lung. Itaconate levels are reduced in bronchoalveolar lavage, and itaconate-synthesizing cis-aconitate decarboxylase expression (ACOD1) is reduced in AMs from patients with IPF compared with controls. In the murine bleomycin model of pulmonary fibrosis, Acod1−/− mice develop persistent fibrosis, unlike wild-type (WT) littermates. Profibrotic gene expression is increased in Acod1−/− tissue-resident AMs compared with WT, and adoptive transfer of WT monocyte-recruited AMs rescued mice from disease phenotype. Culture of lung fibroblasts with itaconate decreased proliferation and wound healing capacity, and inhaled itaconate was protective in mice in vivo. Collectively, these data identify itaconate as critical for controlling the severity of lung fibrosis, and targeting this pathway may be a viable therapeutic strategy.


Carboxy-Lyases/metabolism , Idiopathic Pulmonary Fibrosis/immunology , Macrophages, Alveolar/immunology , Succinates/metabolism , Administration, Inhalation , Adoptive Transfer/methods , Adult , Aged , Animals , Bleomycin/administration & dosage , Bleomycin/toxicity , Bronchoalveolar Lavage Fluid/immunology , Bronchoscopy , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts , Healthy Volunteers , Humans , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/therapy , Lung/cytology , Lung/immunology , Lung/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/transplantation , Male , Mice , Mice, Knockout , Middle Aged , Primary Cell Culture , Severity of Illness Index , Succinates/administration & dosage , Succinates/immunology
3.
J Exp Med ; 217(3)2020 03 02.
Article En | MEDLINE | ID: mdl-31917836

The ontogeny of airway macrophages (AMs) in human lung and their contribution to disease are poorly mapped out. In mice, aging is associated with an increasing proportion of peripherally, as opposed to perinatally derived AMs. We sought to understand AM ontogeny in human lung during healthy aging and after transplant. We characterized monocyte/macrophage populations from the peripheral blood and airways of healthy volunteers across infancy/childhood (2-12 yr), maturity (20-50 yr), and older adulthood (>50 yr). Single-cell RNA sequencing (scRNA-seq) was performed on airway inflammatory cells isolated from sex-mismatched lung transplant recipients. During healthy aging, the proportions of blood bronchoalveolar lavage (BAL) classical monocytes peak in adulthood and decline in older adults. scRNA-seq of BAL cells from lung transplant recipients indicates that after transplant, the majority of AMs are recipient derived. These data show that during aging, the peripheral monocyte phenotype is consistent with that found in the airways and, furthermore, that the majority of human AMs after transplant are derived from circulating monocytes.


Healthy Aging/physiology , Lung/physiology , Macrophages, Alveolar/physiology , Monocytes/physiology , Adult , Animals , Bronchoalveolar Lavage/methods , Child , Child, Preschool , Female , Humans , Leukocytes, Mononuclear/physiology , Male , Mice , Middle Aged , Young Adult
4.
JCI Insight ; 4(18)2019 09 19.
Article En | MEDLINE | ID: mdl-31487265

Autoimmune diseases resulting from MHC class II-restricted autoantigen-specific T cell immunity include the systemic inflammatory autoimmune conditions rheumatoid arthritis and vasculitis. While currently treated with broad-acting immunosuppressive drugs, a preferable strategy is to regulate antigen-specific effector T cells (Teffs) to restore tolerance by exploiting DC antigen presentation. We targeted draining lymph node (dLN) phagocytic DCs using liposomes encapsulating 1α,25-dihydroxyvitamin D3 (calcitriol) and antigenic peptide to elucidate mechanisms of tolerance used by DCs and responding T cells under resting and immunized conditions. PD-L1 expression was upregulated in dLNs of immunized relative to naive mice. Subcutaneous administration of liposomes encapsulating OVA323-339 and calcitriol targeted dLN PD-L1hi DCs of immunized mice and reduced their MHC class II expression. OVA323-339/calcitriol liposomes suppressed expansion, differentiation, and function of Teffs and induced Foxp3+ and IL-10+ peripheral Tregs in an antigen-specific manner, which was dependent on PD-L1. Peptide/calcitriol liposomes modulated CD40 expression by human DCs and promoted Treg induction in vitro. Liposomes encapsulating calcitriol and disease-associated peptides suppressed the severity of rheumatoid arthritis and Goodpasture's vasculitis models with suppression of antigen-specific memory T cell differentiation and function. Accordingly, peptide/calcitriol liposomes leverage DC PD-L1 for antigen-specific T cell regulation and induce antigen-specific tolerance in inflammatory autoimmune diseases.


Anti-Glomerular Basement Membrane Disease/drug therapy , Arthritis, Rheumatoid/drug therapy , Calcitriol/administration & dosage , Dendritic Cells/immunology , Immunodominant Epitopes/administration & dosage , Adoptive Transfer , Animals , Anti-Glomerular Basement Membrane Disease/diagnosis , Anti-Glomerular Basement Membrane Disease/immunology , Antigen Presentation/drug effects , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/immunology , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , CHO Cells , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cricetulus , Dendritic Cells/drug effects , Dendritic Cells/transplantation , Disease Models, Animal , Female , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , Humans , Immune Tolerance/drug effects , Immunodominant Epitopes/immunology , Immunologic Memory/drug effects , Injections, Subcutaneous , Liposomes , Lymph Nodes/cytology , Mice , Mice, Transgenic , Ovalbumin/administration & dosage , Peptide Fragments/administration & dosage , Phagocytosis/drug effects , Phagocytosis/immunology , Severity of Illness Index , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
5.
Front Immunol ; 9: 1092, 2018.
Article En | MEDLINE | ID: mdl-29872433

Polymorphisms impacting thymic function may decrease peripheral tolerance and hasten autoimmune disease. The NF-κB transcription factor subunit, RelB, is essential for the development and differentiation of medullary thymic epithelial cells (mTECs): RelB-deficient mice have reduced thymic cellularity and markedly fewer mTECs, lacking AIRE. The precise mechanism of this mTEC reduction in the absence of RelB is unclear. To address this, we studied mTECs and dendritic cells (DCs), which critically regulate negative selection, and thymic regulatory T-cells (tTreg) in RelB-/- mice, which have spontaneous multiorgan autoimmune disease. RelB-/- thymi were organized, with medullary structures containing AIRE- mTECs, DCs, and CD4+ thymocytes, but fewer tTreg. Granulocytes infiltrated the RelB-/- thymic cortex, capsule, and medulla, producing inflammatory thymic medullary atrophy, which could be treated by granulocyte depletion or RelB+ DC immunotherapy, with concomitant recovery of mTEC and tTreg numbers. These data indicate that central tolerance defects may be accelerated by autoimmune thymic inflammation where impaired RelB signaling impairs the medullary niche, and may be reversible by therapies enhancing peripheral Treg or suppressing inflammation.


Autoimmunity/genetics , Thymus Gland/immunology , Thymus Gland/metabolism , Transcription Factor RelB/deficiency , Animals , Atrophy , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Granulocytes/immunology , Granulocytes/metabolism , Mice , Mice, Knockout , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/pathology , Thyroiditis/etiology , Thyroiditis/metabolism , Thyroiditis/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , AIRE Protein
6.
Am J Respir Cell Mol Biol ; 58(3): 352-365, 2018 03.
Article En | MEDLINE | ID: mdl-28960101

RelB is a member of the NF-κB family, which is essential for dendritic cell (DC) function and maturation. However, the contribution of RelB to the development of allergic airway inflammation (AAI) is unknown. Here, we identify a pivotal role for RelB in the development of spontaneous AAI that is independent of exogenous allergen exposure. We assessed AAI in two strains of RelB-deficient (RelB-/-) mice: one with a targeted deletion and one expressing a major histocompatibility complex transgene. To determine the importance of RelB in DCs, RelB-sufficient DCs (RelB+/+ or RelB-/-) were adoptively transferred into RelB-/- mice. Both strains had increased pulmonary inflammation compared with their respective wild-type (RelB+/+) and heterozygous (RelB+/-) controls. RelB-/- mice also had increased inflammatory cell influx into the airways, levels of chemokines (CCL2/3/4/5/11/17 and CXCL9/10/13) and T-helper cell type 2-associated cytokines (IL-4/5) in lung tissues, serum IgE, and airway remodeling (mucus-secreting cell numbers, collagen deposition, and epithelial thickening). Transfer of RelB+/- CD11c+ DCs into RelB-/- mice decreased pulmonary inflammation, with reductions in lung chemokines, T-helper cell type 2-associated cytokines (IL-4/5/13/25/33 and thymic stromal lymphopoietin), serum IgE, type 2 innate lymphoid cells, myeloid DCs, γδ T cells, lung Vß13+ T cells, mucus-secreting cells, airway collagen deposition, and epithelial thickening. These data indicate that RelB deficiency may be a key pathway underlying AAI, and that DC-encoded RelB is sufficient to restore control of this inflammation.


Asthma/immunology , Dendritic Cells/immunology , Pneumonia/immunology , Th2 Cells/immunology , Transcription Factor RelB/genetics , Adoptive Transfer , Airway Remodeling/immunology , Animals , Asthma/pathology , Chemokines/blood , Dendritic Cells/transplantation , Female , Immunoglobulin E/blood , Male , Mice , Mice, Knockout
7.
Sci Transl Med ; 7(290): 290ra87, 2015 Jun 03.
Article En | MEDLINE | ID: mdl-26041704

In animals, immunomodulatory dendritic cells (DCs) exposed to autoantigen can suppress experimental arthritis in an antigen-specific manner. In rheumatoid arthritis (RA), disease-specific anti-citrullinated peptide autoantibodies (ACPA or anti-CCP) are found in the serum of about 70% of RA patients and are strongly associated with HLA-DRB1 risk alleles. This study aimed to explore the safety and biological and clinical effects of autologous DCs modified with a nuclear factor κB (NF-κB) inhibitor exposed to four citrullinated peptide antigens, designated "Rheumavax," in a single-center, open-labeled, first-in-human phase 1 trial. Rheumavax was administered once intradermally at two progressive dose levels to 18 human leukocyte antigen (HLA) risk genotype-positive RA patients with citrullinated peptide-specific autoimmunity. Sixteen RA patients served as controls. Rheumavax was well tolerated: adverse events were grade 1 (of 4) severity. At 1 month after treatment, we observed a reduction in effector T cells and an increased ratio of regulatory to effector T cells; a reduction in serum interleukin-15 (IL-15), IL-29, CX3CL1, and CXCL11; and reduced T cell IL-6 responses to vimentin(447-455)-Cit450 relative to controls. Rheumavax did not induce disease flares in patients recruited with minimal disease activity, and DAS28 decreased within 1 month in Rheumavax-treated patients with active disease. This exploratory study demonstrates safety and biological activity of a single intradermal injection of autologous modified DCs exposed to citrullinated peptides, and provides rationale for further studies to assess clinical efficacy and antigen-specific effects of autoantigen immunomodulatory therapy in RA.


Arthritis, Rheumatoid/therapy , Citrulline/chemistry , Dendritic Cells/immunology , HLA Antigens/genetics , Immunotherapy , Peptides/chemistry , Aged , Arthritis, Rheumatoid/immunology , Female , Humans , Male , Middle Aged
8.
Mol Nutr Food Res ; 57(9): 1550-6, 2013 Sep.
Article En | MEDLINE | ID: mdl-23495213

Type 2 diabetes is a chronic condition in which cells have reduced insulin signalling, leading to hyperglycemia and long-term complications, including heart, kidney and liver disease. Macrophages activated by dying or stressed cells, induce the transcription factor nuclear factor kappa-B leading to the production of pro-inflammatory cytokines including TNF and IL-6. These inflammatory macrophages in liver and adipose tissue promote insulin resistance, and medications which reduce inflammation and enhance insulin signalling improve glucose control. Curcumin is an anti-oxidant and nuclear factor kappa-B inhibitor derived from turmeric. A number of studies have shown that dietary curcumin reduces inflammation and delays or prevents obesity-induced insulin resistance and associated complications, including atherosclerosis and immune mediate liver disease. Unfortunately dietary curcumin is poorly absorbed by the digestive system and undergoes glucuronidation and excretion rather than being released into the serum and systemically distributed. This confounds understanding of how dietary curcumin exerts its beneficial effects in type 2 diabetes and associated diseases. New improved methods of delivering curcumin are being developed including nanoparticles and lipid/liposome formulations that increase absorption and bioavailability of curcumin. Development and refinement of these technologies will enable cell-directed targeting of curcumin and improved therapeutic outcome.


Curcumin/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Drug Delivery Systems/methods , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Curcuma/chemistry , Disease Models, Animal , Humans , I-kappa B Proteins/pharmacology , Inflammation/prevention & control , Insulin/blood , Insulin Resistance , Liver/drug effects , Liver/metabolism , Liver Diseases/etiology , Liver Diseases/prevention & control , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , Nanoparticles/chemistry , Obesity/complications , Obesity/drug therapy , Randomized Controlled Trials as Topic
9.
Proc Natl Acad Sci U S A ; 108(44): 18049-54, 2011 Nov 01.
Article En | MEDLINE | ID: mdl-22006331

Antigen-dependent interactions between T lymphocytes and dendritic cells (DCs) can produce two distinct outcomes: tolerance and immunity. It is generally considered that all DC subsets are capable of supporting both tolerogenic and immunogenic responses, depending on their exposure to activating signals. Here, we tested whether epidermal Langerhans cells (LCs) can support immunogenic responses in vivo in the absence of antigen presentation by other DC subsets. CD4 T cells responding to antigen presentation by activated LCs initially proliferated but then failed to differentiate into effector/memory cells or to survive long term. The tolerogenic function of LCs was maintained after exposure to potent adjuvants and occurred despite up-regulation of the costimulatory molecules CD80, CD86, and IL-12, but was consistent with their failure to translocate the NF-κB family member RelB from the cytoplasm to the nucleus. Commitment of LCs to tolerogenic function may explain why commensal microorganisms expressing Toll-like receptor (TLR) ligands but confined to the skin epithelium are tolerated, whereas invading pathogens that breach the epithelial basement membrane and activate dermal DCs stimulate a strong immune response.


Immune Tolerance , Langerhans Cells/immunology , Animals , Antigens, CD/immunology , Humans , Immunophenotyping , Mice
10.
J Immunol ; 187(8): 4018-30, 2011 Oct 15.
Article En | MEDLINE | ID: mdl-21900177

Costimulation-deficient dendritic cells (DCs) prevent autoimmune disease in mouse models. However, autoimmune-prone mice and humans fail to control expansion of peripheral autoreactive effector memory T cells (T(EMs)), which resist immunoregulation by costimulation-deficient DCs. In contrast, activation of DC costimulation may be coupled with regulatory capacity. To test whether costimulatory DCs control T(EMs) and attenuate established autoimmune disease, we used RelB-deficient mice, which have multiorgan inflammation, expanded peripheral autoreactive T(EMs), and dysfunctional Foxp3(+) regulatory T cells (Tregs) cells and conventional DCs. T(EMs) were regulated by Foxp3(+) Tregs when costimulated by CD3/CD28-coated beads or wild-type DCs but not DCs deficient in RelB or CD80/CD86. After transfer, RelB and CD80/CD86-sufficient DCs restored tolerance and achieved a long-term cure of autoimmune disease through costimulation of T(EM) and Foxp3(+) Treg IFN-γ production, as well as induction of IDO by host APCs. IDO was required for regulation of T(EMs) and suppression of organ inflammation. Our data challenge the paradigm that costimulation-deficient DCs are required to regulate established autoimmune disease to avoid T(EM) activation and demonstrate cooperative cross-talk between costimulatory DCs, IFN-γ, and IDO-dependent immune regulation. IFN-γ and IDO activity may be good surrogate biomarkers measured against clinical efficacy in trials of autoimmune disease immunoregulation.


Autoimmune Diseases/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Autoimmune Diseases/therapy , Dendritic Cells/transplantation , Flow Cytometry , Immune Tolerance/immunology , Immunomagnetic Separation , Inflammation/immunology , Inflammation/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Signal Transduction/immunology , Transcription Factor RelB/deficiency , Transcription Factor RelB/genetics , Transcription Factor RelB/immunology
11.
Thorax ; 65(12): 1053-60, 2010 Dec.
Article En | MEDLINE | ID: mdl-20965927

BACKGROUND: Infections with some bacteria, including Streptococcus pneumoniae, have been associated with a reduced incidence of asthma. Components of S pneumoniae may have the potential to modulate allergic inflammatory responses and suppress the development of asthma. OBJECTIVES: To determine if human S pneumoniae vaccines have the potential to suppress asthma by elucidating their effect on allergic airways disease (AAD) in mouse models. METHODS: AAD was induced in BALB/c mice by intraperitoneal sensitisation and intranasal challenge with ovalbumin. Pneumococcal conjugate or polysaccharide vaccines were administered at the time of sensitisation or during established AAD. Hallmark features of AAD were assessed. Levels of regulatory T cells (Tregs) were quantified by fluorescence-activated cell sorting, and their immunoregulatory capacity was assessed using proliferation assays and anti-CD25 antibody treatment. RESULTS: Intranasal administration of the conjugate vaccine, but not the polysaccharide vaccine, suppressed the hallmark features of AAD, including: eosinophilic and T helper 2-mediated inflammation; airway hyper-responsiveness; circulating immunoglobulin E (IgE) levels; and mucus hypersecretion. Intramuscular administration of the conjugate vaccine had limited protective effects. The conjugate vaccine increased Tregs in the lung-draining lymph nodes, lung and spleen. Furthermore, conjugate vaccine-induced Tregs had an enhanced capacity to suppress T effector responses. Anti-CD25 administration reversed the suppressive effects of the conjugate vaccine. CONCLUSIONS: A currently available human conjugate vaccine suppresses the hallmark features of AAD through the induction of Tregs. Thus targeted administration may provide a novel immunoregulatory treatment for asthma.


Asthma/prevention & control , Pneumococcal Vaccines/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Asthma/immunology , CD2 Antigens/immunology , Disease Models, Animal , Female , Immune Tolerance , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Th2 Cells/immunology , Vaccines, Conjugate/immunology
12.
J Immunol ; 180(5): 3166-75, 2008 Mar 01.
Article En | MEDLINE | ID: mdl-18292540

Dendritic cell (DC) differentiation is abnormal in type 1 diabetes mellitus (T1DM). However, the nature of the relationship between this abnormality and disease pathogenesis is unknown. We studied the LPS response in monocytes and monocyte-derived DCs isolated from T1DM patients and from non-T1DM controls. In T1DM patients, late LPS-mediated nuclear DNA binding by RelA, p50, c-Rel, and RelB was impaired as compared with type 2 DM, rheumatoid arthritis, and healthy subjects, associated with impaired DC CD40 and MHC class I induction but normal cytokine production. In TIDM monocytes, RelA and RelB were constitutively activated, and the src homology 2 domain-containing protein tyrosine phosphatase (SHP-1), a negative regulator of NF-kappaB, was overexpressed. Addition of sodium stibogluconate, a SHP-1 inhibitor, to DCs differentiating from monocyte precursors restored their capacity to respond to LPS in approximately 60% of patients. The monocyte and DC NF-kappaB response to LPS is thus a novel phenotypic and likely pathogenetic marker for human T1DM. SHP-1 is at least one NF-kappaB regulatory mechanism which might be induced as a result of abnormal inflammatory signaling responses in T1DM monocytes.


Dendritic Cells/immunology , Dendritic Cells/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Monocytes/immunology , Monocytes/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Adolescent , Adult , Aged , Antigen Presentation/immunology , Cell Differentiation/immunology , Cells, Cultured , Child , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Humans , Lymphocyte Activation/immunology , MAP Kinase Signaling System/immunology , Male , Middle Aged , Monocytes/metabolism , NF-kappa B/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
13.
J Immunother ; 29(6): 606-15, 2006.
Article En | MEDLINE | ID: mdl-17063123

Fever is an evolutionarily conserved mechanism to improve survival during infection. Previous studies have shown that feverlike temperatures directly enhance the function of murine bone marrow-derived dendritic cells (DCs). In the present study, we examined the response of human monocyte-derived DC to 39.5 degrees C hyperthermia. When primed with toll-like receptor agonists or bacterial extract but not proinflammatory cytokines, hyperthermia specifically enhanced secretion of interleukin (IL)-12p70 by DC, without altering the secretion of IL-10, tumor necrosis factor alpha or IL-1beta. These DC induced significantly higher levels of T-cell proliferation and interferon gamma production in assays of antigen presentation and MLR. Endogenous heat-sock protein 70 colocalized with CD40 in DC exposed to hyperthermic conditions. Recombinant CD40-Fc fusion protein blocked the increase in IL-12p70 secretion by DC primed with bacterial extract and hyperthermia. Thus, DC primed with toll-like receptor-agonists respond to hyperthermia with increased IL-12p70 secretion, mediated by heat-shock protein binding and activation of CD40. The data have important applications for clinical immunotherapy and the mechanism of fever.


CD40 Antigens/metabolism , Dendritic Cells/metabolism , Hot Temperature , Interleukin-12/metabolism , Toll-Like Receptors/agonists , Antigen Presentation/immunology , Antigens, Bacterial/pharmacology , Antigens, CD/metabolism , B7-2 Antigen/metabolism , CD40 Antigens/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Cytokines/metabolism , Cytokines/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HLA-DR Antigens/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunoglobulins/metabolism , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Membrane Glycoproteins/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Peptidoglycan/pharmacology , Poly I-C/pharmacology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tetanus Toxoid/immunology , CD83 Antigen
14.
J Immunol ; 176(12): 7278-87, 2006 Jun 15.
Article En | MEDLINE | ID: mdl-16751371

IL-1 is a key proinflammatory driver of several autoimmune diseases including juvenile inflammatory arthritis, diseases with mutations in the NALP/cryopyrin complex and Crohn's disease, and is genetically or clinically associated with many others. IL-1 is a pleiotropic proinflammatory cytokine; however the mechanisms by which increased IL-1 signaling promotes autoreactive T cell activity are not clear. Here we show that autoimmune-prone NOD and IL-1 receptor antagonist-deficient C57BL/6 mice both produce high levels of IL-1, which drives autoreactive effector cell expansion. IL-1beta drives proliferation and cytokine production by CD4(+)CD25(+)FoxP3(-) effector/memory T cells, attenuates CD4(+)CD25(+)FoxP3(+) regulatory T cell function, and allows escape of CD4(+)CD25(-) autoreactive effectors from suppression. Thus, inflammation or constitutive overexpression of IL-1beta in a genetically predisposed host can promote autoreactive effector T cell expansion and function, which attenuates the ability of regulatory T cells to maintain tolerance to self.


Cell Proliferation , Interleukin-1/physiology , Receptors, Interleukin-2/biosynthesis , Self Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Division/genetics , Cell Division/immunology , Cells, Cultured , Clonal Anergy/genetics , Clonal Anergy/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Forkhead Transcription Factors/biosynthesis , Genetic Predisposition to Disease , Immunologic Memory/genetics , Interferon-gamma/biosynthesis , Interleukin-1/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics , Self Tolerance/genetics , T-Lymphocytes, Regulatory/cytology
15.
J Immunol ; 173(3): 1671-80, 2004 Aug 01.
Article En | MEDLINE | ID: mdl-15265896

Paradoxically, while peripheral self-tolerance exists for constitutively presented somatic self Ag, self-peptide recognized in the context of MHC class II has been shown to sensitize T cells for subsequent activation. We have shown that MHC class II(+)CD86(+)CD40(-) DC, which can be generated from bone marrow in the presence of an NF-kappa B inhibitor, and which constitutively populate peripheral tissues and lymphoid organs in naive animals, can induce Ag-specific tolerance. In this study, we show that CD40(-) human monocyte-derived dendritic cells (DC), generated in the presence of an NF-kappa B inhibitor, signal phosphorylation of TCR zeta, but little proliferation or IFN-gamma in vitro. Proliferation is arrested in the G(1)/G(0) phase of the cell cycle. Surprisingly, responding T cells are neither anergic nor regulatory, but are sensitized for subsequent IFN-gamma production. The data indicate that signaling through NF-kappa B determines the capacity of DC to stimulate T cell proliferation. Functionally, NF-kappa B(-)CD40(-)class II(+) DC may either tolerize or sensitize T cells. Thus, while CD40(-) DC appear to "prime" or prepare T cells, the data imply that signals derived from other cells drive the generation either of Ag-specific regulatory or effector cells in vivo.


Dendritic Cells/physiology , Lymphocyte Activation , NF-kappa B/deficiency , T-Lymphocyte Subsets/immunology , Active Transport, Cell Nucleus , Antigen Presentation , Antigens/immunology , CD40 Antigens/analysis , Cells, Cultured/immunology , Clonal Anergy , Dendritic Cells/classification , Dendritic Cells/drug effects , HLA-D Antigens/analysis , Humans , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Membrane Proteins/metabolism , Monocytes/cytology , Monocytes/drug effects , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Sulfones/pharmacology , T-Lymphocyte Subsets/metabolism , Transcription Factor RelB , Transcription Factors/metabolism
16.
J Virol ; 76(4): 1914-21, 2002 Feb.
Article En | MEDLINE | ID: mdl-11799186

Previous studies have shown that Epstein-Barr virus-encoded latent membrane protein 1 (LMP1) is uniquely able to up-regulate the expression of the peptide transporters (referred to as TAP-1 and TAP-2) and major histocompatibility complex (MHC) class I in Burkitt's lymphoma (BL) cell lines. This up-regulation is often accompanied by a restoration of antigen-presenting function as measured by the ability of these cells to present endogenously expressed viral antigen to cytotoxic T lymphocytes. Here we show that the expression of LMP1 resulted in up-regulation and nuclear translocation of RelB that were coincident with increased expression of MHC class I in BL cells. Deletion of the C-terminal activator regions (CTARs) of LMP1 significantly impaired the abilities of LMP1 to translocate RelB into the nucleus and to up-regulate the expression of antigen-processing genes. Further analysis with single-point mutations within the CTARs confirmed that the residues critical for NF-kappa B activation directly contribute to antigen-processing function regulation in BL cells. This LMP1-mediated effect was blocked following expression of either dominant negative I kappa B alpha S32/36A, an NF-kappa B inhibitor, or antisense RelB. These observations indicate that upregulation of antigen-presenting function in B cells mediated by LMP1 is signaled through the NF-kappa B subunit RelB. The data provide a mechanism by which LMP1 modulates immunogenicity of Epstein-Barr virus-infected normal and malignant cells.


Antigen Presentation/genetics , B-Lymphocytes/virology , Gene Expression Regulation, Viral , Herpesvirus 4, Human/physiology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Viral Matrix Proteins/metabolism , Active Transport, Cell Nucleus , Antigen Presentation/physiology , B-Lymphocytes/immunology , Burkitt Lymphoma/virology , Cell Line, Transformed , Cell Nucleus/metabolism , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins/genetics , Transcription Factor RelB , Transcription Factors/genetics , Tumor Cells, Cultured , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics
...