Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 56(10): 2373-2387.e8, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37714151

ABSTRACT

Immunoglobulin A (IgA) maintains commensal communities in the intestine while preventing dysbiosis. IgA generated against intestinal microbes assures the simultaneous binding to multiple, diverse commensal-derived antigens. However, the exact mechanisms by which B cells mount broadly reactive IgA to the gut microbiome remains elusive. Here, we have shown that IgA B cell receptor (BCR) is required for B cell fitness during the germinal center (GC) reaction in Peyer's patches (PPs) and for generation of gut-homing plasma cells (PCs). We demonstrate that IgA BCR drove heightened intracellular signaling in mouse and human B cells, and as a consequence, IgA+ B cells received stronger positive selection cues. Mechanistically, IgA BCR signaling offset Fas-mediated death, possibly rescuing low-affinity B cells to promote a broad humoral response to commensals. Our findings reveal an additional mechanism linking BCR signaling, B cell fate, and antibody production location, which have implications for how intestinal antigen recognition shapes humoral immunity.


Subject(s)
B-Lymphocytes , Peyer's Patches , Mice , Humans , Animals , Antigens/metabolism , Receptors, Antigen, B-Cell/metabolism , Immunoglobulin A , Intestinal Mucosa
2.
Immunity ; 54(6): 1137-1153.e8, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34051146

ABSTRACT

Alterations in the cGAS-STING DNA-sensing pathway affect intestinal homeostasis. We sought to delineate the functional role of STING in intestinal inflammation. Increased STING expression was a feature of intestinal inflammation in mice with colitis and in humans afflicted with inflammatory bowel disease. Mice bearing an allele rendering STING constitutively active exhibited spontaneous colitis and dysbiosis, as well as progressive chronic intestinal inflammation and fibrosis. Bone marrow chimera experiments revealed STING accumulation in intestinal macrophages and monocytes as the initial driver of inflammation. Depletion of Gram-negative bacteria prevented STING accumulation in these cells and alleviated intestinal inflammation. STING accumulation occurred at the protein rather than transcript level, suggesting post-translational stabilization. We found that STING was ubiquitinated in myeloid cells, and this K63-linked ubiquitination could be elicited by bacterial products, including cyclic di-GMP. Our findings suggest a positive feedback loop wherein dysbiosis foments the accumulation of STING in intestinal myeloid cells, driving intestinal inflammation.


Subject(s)
Colitis/immunology , Dysbiosis/immunology , Immunity, Innate/immunology , Membrane Proteins/immunology , Myeloid Cells/immunology , Ubiquitination/immunology , Animals , Case-Control Studies , Female , Humans , Inflammation/immunology , Intestines/immunology , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology
3.
Immunity ; 48(5): 911-922.e7, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768176

ABSTRACT

Unc-93 homolog B1 (UNC93B1) is a key regulator of nucleic acid (NA)-sensing Toll-like receptors (TLRs). Loss of NA-sensing TLR responses in UNC93B1-deficient patients facilitates Herpes simplex virus type 1 (HSV-1) encephalitis. UNC93B1 is thought to guide NA-sensing TLRs from the endoplasmic reticulum (ER) to their respective endosomal signaling compartments and to guide the flagellin receptor TLR5 to the cell surface, raising the question of how UNC93B1 mediates differential TLR trafficking. Here, we report that UNC93B1 regulates a step upstream of the differential TLR trafficking process. We discovered that UNC93B1 deficiency resulted in near-complete loss of TLR3 and TLR7 proteins in primary splenic mouse dendritic cells and macrophages, showing that UNC93B1 is critical for maintaining TLR expression. Notably, expression of an ER-retained UNC93B1 version was sufficient to stabilize TLRs and largely restore endosomal TLR trafficking and activity. These data are critical for an understanding of how UNC93B1 can regulate the function of a broad subset of TLRs.


Subject(s)
Endosomes/immunology , Membrane Transport Proteins/immunology , Molecular Chaperones/immunology , Toll-Like Receptors/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , HEK293 Cells , Humans , Macrophages/immunology , Macrophages/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Stability , Protein Transport/immunology , Signal Transduction/genetics , Signal Transduction/immunology , THP-1 Cells , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
4.
Nat Immunol ; 15(8): 727-37, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24952505

ABSTRACT

Microbes or danger signals trigger inflammasome sensors, which induce polymerization of the adaptor ASC and the assembly of ASC specks. ASC specks recruit and activate caspase-1, which induces maturation of the cytokine interleukin 1ß (IL-1ß) and pyroptotic cell death. Here we found that after pyroptosis, ASC specks accumulated in the extracellular space, where they promoted further maturation of IL-1ß. In addition, phagocytosis of ASC specks by macrophages induced lysosomal damage and nucleation of soluble ASC, as well as activation of IL-1ß in recipient cells. ASC specks appeared in bodily fluids from inflamed tissues, and autoantibodies to ASC specks developed in patients and mice with autoimmune pathologies. Together these findings reveal extracellular functions of ASC specks and a previously unknown form of cell-to-cell communication.


Subject(s)
Apoptosis/immunology , Caspase 1/immunology , Cytoskeletal Proteins/immunology , Inflammation/immunology , Interleukin-1beta/immunology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antibodies/immunology , Apoptosis Regulatory Proteins , Autoantibodies/immunology , Autoimmune Diseases/immunology , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/genetics , Caspase Inhibitors/pharmacology , Cell Communication/immunology , Cytoskeletal Proteins/genetics , Humans , Inflammasomes/immunology , Lysosomes/pathology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Phagocytosis/immunology , Prions/chemistry , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Signal Transduction/immunology
5.
Proc Natl Acad Sci U S A ; 119(25): e2202327119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35696583

ABSTRACT

Pediatric patients with constitutively active mutations in the cytosolic double-stranded-DNA-sensing adaptor STING develop an autoinflammatory syndrome known as STING-associated vasculopathy with onset in infancy (SAVI). SAVI patients have elevated interferon-stimulated gene expression and suffer from interstitial lung disease (ILD) with lymphocyte predominate bronchus-associated lymphoid tissue (BALT). Mice harboring SAVI mutations (STING V154M [VM]) that recapitulate human disease also develop lymphocyte-rich BALT. Ablation of either T or B lymphocytes prolongs the survival of SAVI mice, but lung immune aggregates persist, indicating that T cells and B cells can independently be recruited as BALT. VM T cells produced IFNγ, and IFNγR deficiency prolonged the survival of SAVI mice; however, T-cell-dependent recruitment of infiltrating myeloid cells to the lung was IFNγ independent. Lethally irradiated VM recipients fully reconstituted with wild type bone-marrow-derived cells still developed ILD, pointing to a critical role for VM-expressing radioresistant parenchymal and/or stromal cells in the recruitment and activation of pathogenic lymphocytes. We identified lung endothelial cells as radioresistant cells that express STING. Transcriptional analysis of VM endothelial cells revealed up-regulation of chemokines, proinflammatory cytokines, and genes associated with antigen presentation. Together, our data show that VM-expressing radioresistant cells play a key role in the initiation of lung disease in VM mice and provide insights for the treatment of SAVI patients, with implications for ILD associated with other connective tissue disorders.


Subject(s)
Endothelial Cells , Lung Diseases, Interstitial , Membrane Proteins , T-Lymphocytes , Vascular Diseases , Animals , Child , Endothelial Cells/immunology , Endothelial Cells/radiation effects , Gain of Function Mutation , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lung Diseases, Interstitial/genetics , Lung Diseases, Interstitial/immunology , Lymphocyte Depletion , Lymphoid Tissue/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Radiation Tolerance , T-Lymphocytes/immunology , Vascular Diseases/genetics , Vascular Diseases/immunology
6.
Proc Natl Acad Sci U S A ; 116(16): 7941-7950, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30944222

ABSTRACT

Studies over the past decade have revealed a central role for innate immune sensors in autoimmune and autoinflammatory diseases. cGAS, a cytosolic DNA sensor, detects both foreign and host DNA and generates a second-messenger cGAMP, which in turn binds and activates stimulator of IFN genes (STING), leading to induction of type I interferons and inflammatory cytokines. Recently, gain-of-function mutations in STING have been identified in patients with STING-associated vasculopathy with onset in infancy (SAVI). SAVI patients present with early-onset systemic inflammation and interstitial lung disease, resulting in pulmonary fibrosis and respiratory failure. Here, we describe two independent SAVI mouse models, harboring the two most common mutations found in patients. A direct comparison of these strains reveals a hierarchy of immune abnormalities, lung inflammation and fibrosis, which do not depend on either IFN-α/ß receptor signaling or mixed lineage kinase domain-like pseudokinase (MLKL)-dependent necroptotic cell death pathways. Furthermore, radiation chimera experiments reveal how bone marrow from the V154M mutant mice transfer disease to the WT host, whereas the N153S does not, indicating mutation-specific disease outcomes. Moreover, using radiation chimeras we find that T cell lymphopenia depends on T cell-intrinsic expression of the SAVI mutation. Collectively, these mutant mice recapitulate many of the disease features seen in SAVI patients and highlight mutation-specific functions of STING that shed light on the heterogeneity observed in SAVI patients.


Subject(s)
Disease Models, Animal , Interferon Type I/metabolism , Vascular Diseases , Animals , Cell Death/immunology , Cytokines/metabolism , Gain of Function Mutation , Inflammation/immunology , Inflammation/physiopathology , Mice , Vascular Diseases/genetics , Vascular Diseases/immunology , Vascular Diseases/physiopathology
7.
J Immunol ; 2019 Sep 18.
Article in English | MEDLINE | ID: mdl-31534009

ABSTRACT

The AM14 BCR, derived from an autoimmune MRL/lpr mouse, binds autologous IgG2aa/j with low affinity, and as a result, AM14 B cells only proliferate in response to IgG2a immune complexes that incorporate DNA, RNA, or nucleic acid-binding proteins that serve as autoadjuvants. As such, AM14 B cells have served as a useful model for demonstrating the importance of BCR/TLR coengagement in the activation of autoreactive B cells. We now show that the same receptor recognizes an additional murine-encoded Ag, expressed by B6 splenocytes, with sufficient avidity to induce a TLR-independent proliferative response of BALB/c AM14 Vκ8 B cells both in vivo and in vitro. Moreover, detection of this cross-reactive Ag by B6 AM14 Vκ8 B cells promotes an anergic phenotype as reflected by suboptimal responses to BCR cross-linking and the absence of mature B cells in the bone marrow. The B6 Ag further impacts B cell development as shown by a dramatically expanded marginal zone compartment and extensive receptor editing in B6 AM14 Vκ8 mice but not BALB/c AM14 Vκ8 mice. Despite their anergic phenotypes, B6 AM14 Vκ8 B cells can respond robustly to autoantigen/autoadjuvant immune complexes and could therefore participate in both autoimmune responses and host defense.

8.
J Immunol ; 200(2): 737-748, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29212904

ABSTRACT

Necroptosis is a form of cell death associated with inflammation; however, the biological consequences of chronic necroptosis are unknown. Necroptosis is mediated by RIPK1, RIPK3, and MLKL kinases but in hematopoietic cells RIPK1 has anti-inflammatory roles and functions to prevent necroptosis. Here we interrogate the consequences of chronic necroptosis on immune homeostasis by deleting Ripk1 in mouse dendritic cells. We demonstrate that deregulated necroptosis results in systemic inflammation, tissue fibrosis, and autoimmunity. We show that inflammation and autoimmunity are prevented upon expression of kinase inactive RIPK1 or deletion of RIPK3 or MLKL. We provide evidence that the inflammation is not driven by microbial ligands, but depends on the release of danger-associated molecular patterns and MyD88-dependent signaling. Importantly, although the inflammation is independent of type I IFN and the nucleic acid sensing TLRs, blocking these pathways rescues the autoimmunity. These mouse genetic studies reveal that chronic necroptosis may underlie human fibrotic and autoimmune disorders.


Subject(s)
Autoimmunity , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunity , Inflammation/etiology , Inflammation/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Autoantibodies/immunology , Autoimmunity/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cytokines/metabolism , Disease Models, Animal , Fibrosis , Gene Expression Profiling , Inflammation/pathology , Inflammation/prevention & control , Lymphadenopathy/genetics , Lymphadenopathy/immunology , Lymphadenopathy/metabolism , Lymphadenopathy/pathology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Necrosis/genetics , Necrosis/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Toll-Like Receptors/metabolism
9.
J Neuroinflammation ; 16(1): 184, 2019 Sep 30.
Article in English | MEDLINE | ID: mdl-31570110

ABSTRACT

BACKGROUND: Glaucoma is a complex, multifactorial disease where apoptosis, microglia activation, and inflammation have been linked to the death of retinal ganglion cells (RGCs) and axon degeneration. We demonstrated previously that FasL-Fas signaling was required for axon degeneration and death of RGCs in chronic and inducible mouse models of glaucoma and that Fas activation triggered RGC apoptosis, glial activation, and inflammation. Here, we investigated whether targeting the Fas receptor with a small peptide antagonist, ONL1204, has anti-inflammatory and neuroprotective effects in a microbead-induced mouse model of glaucoma. METHODS: Intracameral injection of microbeads was used to elevate intraocular pressure (IOP) in Fas-deficient (Faslpr) mice and WT C57BL/6J mice that received an intravitreal injection of the Fas inhibitor, ONL1204 (2 µg/1 µl) (or vehicle only), on day 0 or day 7 after microbead injection. The IOP was monitored by rebound tonometry, and at 28 days post-microbead injection, Brn3a-stained RGCs and paraphenylenediamine (PPD)-stained axons were analyzed. The effects of ONL1204 on retinal microglia activation and the expression of inflammatory genes were analyzed by immunostaining of retinal flatmounts and quantitative PCR (qPCR). RESULTS: Rebound tonometry showed equivalent elevation of IOP in all groups of microbead-injected mice. At 28 days post-microbead injection, the RGC and axon counts from microbead-injected Faslpr mice were equivalent to saline-injected (no IOP elevation) controls. Treatment with ONL1204 also significantly reduced RGC death and loss of axons in microbead-injected WT mice when compared to vehicle-treated controls, even when administered after IOP elevation. Confocal analysis of Iba1-stained retinal flatmounts and qPCR demonstrated that ONL1204 also abrogated microglia activation and inhibited the induction of multiple genes implicated in glaucoma, including cytokines and chemokines (GFAP, Caspase-8, TNFα, IL-1ß, IL-6, IL-18, MIP-1α, MIP-1ß, MIP-2, MCPI, and IP10), components of the complement cascade (C3, C1Q), Toll-like receptor pathway (TLR4), and inflammasome pathway (NLRP3). CONCLUSIONS: These results serve as proof-of-principal that the small peptide inhibitor of the Fas receptor, ONL1204, can provide robust neuroprotection in an inducible mouse model of glaucoma, even when administered after IOP elevation. Moreover, Fas signaling contributes to the pathogenesis of glaucoma through activation of both apoptotic and inflammatory pathways.


Subject(s)
Glaucoma/pathology , Nerve Degeneration/pathology , Neuroprotective Agents/pharmacology , Peptides/pharmacology , Retinal Ganglion Cells/drug effects , fas Receptor/antagonists & inhibitors , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Cell Death/drug effects , Disease Models, Animal , Glaucoma/metabolism , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Nerve Degeneration/metabolism , Peptides/chemistry , Retinal Ganglion Cells/pathology
10.
Exp Eye Res ; 179: 47-54, 2019 02.
Article in English | MEDLINE | ID: mdl-30365944

ABSTRACT

Neutrophil-mediated inflammation plays a critical role in corneal damage following injury or infection. Previous studies demonstrated that membrane-bound FasL (mFasL) induces neutrophil chemokine production. However, the extracellular domain of mFasL is normally cleaved by matrix metalloproteinases to release a soluble form of FasL (sFasL) and sFasL antagonizes mFasL-mediated chemokine production. Therefore, we hypothesized that sFasL could be used to prevent neutrophil-mediated corneal inflammation associated with injury and bacterial keratitis. To test this hypothesis, GFP-only, sFasL-GFP, or mFasL-GFP were expressed in the corneal stroma of C57BL/6 mice, using intra-stromal injections of plasmid DNA or adenoviral vectors (AV) and the role of mFasL and sFasL in corneal inflammation was examined in models of corneal injury and LPS-induced keratitis. Our work addresses an important area of disagreement in the field of FasL, with regard to the mechanism by which sFasL regulates ocular inflammation. Herein, we demonstrate that an intrastromal injection of GFP-only, sFasL-GFP, or mFasL-GFP plasmid DNA resulted in GFP expression throughout the corneal stroma for up to two weeks with little to no evidence of inflammation in the GFP-only and sFasL-GFP groups and mild corneal inflammation in the mFasL-GFP group. Similarly, following epithelial debridement, corneas expressing GFP-only or sFasL-GFP showed no significant signs of corneal inflammation, with clear corneas at 15 days post debridement. By contrast, epithelial debridement of corneas expressing mFasL-GFP triggered persistent corneal inflammation and the development of central corneal opacities that was blocked by sFasL. Similar to the mFasL-GFP plasmid DNA, intrastromal injection of mFasL-GFP AV triggered mild corneal inflammation, but it was transient and resolved by day 10 with corneas remaining clear out to 30 days post injection. Nevertheless, intrastromal expression of mFasL-GFP AV exacerbated LPS-induced keratitis, corneal opacity, and neovascularization, while sFasL-GFP AV expression prevented LPS-induced keratitis, resulting in a clear cornea. Histological analysis of corneas with LPS-induced keratitis revealed a robust infiltration of macrophages and neutrophils and sFasL expression specifically blocked the neutrophil influx. Overall, our data demonstrate that stromal expression of mFasL is inflammatory, while sFasL is non-inflammatory, and opposes the effects of mFasL in mouse models of epithelial debridement and LPS-induced keratitis. These data demonstrate that a delicate balance between sFasL and mFasL regulates ocular inflammation. This study further identifies sFasL as a potent inhibitor of neutrophil-mediated corneal damage, and supports the potential use of sFasL in the treatment of neutrophil-mediated keratitis. These results strongly support the hypothesis that, in the immune privileged environment of the eye, the isoform of FasL regulates immune privilege and determines the extent of inflammation: mFasL promotes inflammation and sFasL blocks inflammation.


Subject(s)
Debridement , Disease Models, Animal , Epithelium, Corneal/surgery , Fas Ligand Protein/physiology , Keratitis/prevention & control , Adenoviridae/genetics , Animals , Blotting, Western , Corneal Neovascularization/etiology , Corneal Neovascularization/prevention & control , Corneal Stroma/metabolism , Female , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation/physiology , Genetic Vectors , Green Fluorescent Proteins/metabolism , Keratitis/etiology , Keratitis/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Plasmids/genetics
11.
J Immunol ; 197(12): 4626-4638, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27849168

ABSTRACT

Glaucoma is a multifactorial disease resulting in the death of retinal ganglion cells (RGCs) and irreversible blindness. Glaucoma-associated RGC death depends on the proapoptotic and proinflammatory activity of membrane-bound Fas ligand (mFasL). In contrast to mFasL, the natural cleavage product, soluble Fas ligand (sFasL) inhibits mFasL-mediated apoptosis and inflammation and, therefore, is an mFasL antagonist. DBA/2J mice spontaneously develop glaucoma and, predictably, RGC destruction is exacerbated by expression of a mutated membrane-only FasL gene that lacks the extracellular cleavage site. Remarkably, one-time intraocular adeno-associated virus-mediated gene delivery of sFasL provides complete and sustained neuroprotection in the chronic DBA/2J and acute microbead-induced models of glaucoma, even in the presence of elevated intraocular pressure. This protection correlated with inhibition of glial activation, reduced production of TNF-α, and decreased apoptosis of RGCs and loss of axons. These data indicate that cleavage of FasL under homeostatic conditions, and the ensuing release of sFasL, normally limits the neurodestructive activity of FasL. The data further support the notion that sFasL, and not mFasL, contributes to the immune-privileged status of the eye.


Subject(s)
Fas Ligand Protein/metabolism , Genetic Therapy , Glaucoma/therapy , Neuroprotection , Retinal Ganglion Cells/physiology , Acute Disease , Animals , Apoptosis , Cells, Cultured , Chronic Disease , Dependovirus/genetics , Disease Models, Animal , Fas Ligand Protein/genetics , Female , Glaucoma/genetics , Glaucoma/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Tumor Necrosis Factor-alpha/metabolism
12.
J Immunol ; 196(3): 1348-54, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26729810

ABSTRACT

Detection of endogenous nucleic acids by cytosolic receptors, dependent on STING, and endosomal sensors, dependent on Unc93b1, can provoke inflammatory responses that contribute to a variety of autoimmune and autoinflammatory diseases. In DNase II-deficient mice, the excessive accrual of undegraded DNA leads to both a STING-dependent inflammatory arthritis and additional Unc93b1-dependent autoimmune manifestations, including splenomegaly, extramedullary hematopoiesis, and autoantibody production. In this study, we use bone marrow chimeras to show that clinical and histological inflammation in the joint depends upon DNase II deficiency in both donor hematopoietic cells and host radioresistant cells. Additional features of autoimmunity in these mice, known to depend on Unc93b1 and therefore endosomal TLRs, also require DNase II deficiency in both donor and host compartments, but only require functional TLRs in the hematopoietic cells. Collectively, our data demonstrate a major role of both stromal and hematopoietic cells in all aspects of DNA-driven autoimmunity. These findings further point to the importance of cytosolic nucleic acid sensors in creating an inflammatory environment that facilitates the development of Unc93b1-dependent autoimmunity.


Subject(s)
Arthritis, Experimental/immunology , Autoimmunity/immunology , Hematopoietic Stem Cells/immunology , Stromal Cells/immunology , Animals , Disease Models, Animal , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/immunology , Flow Cytometry , Membrane Proteins/immunology , Membrane Transport Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation Chimera
13.
J Immunol ; 197(4): 1044-53, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27354219

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic, life-threatening autoimmune disorder, leading to multiple organ pathologies and kidney destruction. Analyses of numerous murine models of spontaneous SLE have revealed a critical role for endosomal TLRs in the production of autoantibodies and development of other clinical disease manifestations. Nevertheless, the corresponding TLR9-deficient autoimmune-prone strains consistently develop more severe disease pathology. Injection of BALB/c mice with 2,6,10,14-tetramethylpentadecane (TMPD), commonly known as pristane, also results in the development of SLE-like disease. We now show that Tlr9(-/-) BALB/c mice injected i.p. with TMPD develop more severe autoimmunity than do their TLR-sufficient cohorts. Early indications include an increased accumulation of TLR7-expressing Ly6C(hi) inflammatory monocytes at the site of injection, upregulation of IFN-regulated gene expression in the peritoneal cavity, and an increased production of myeloid lineage precursors (common myeloid progenitors and granulocyte myeloid precursors) in the bone marrow. TMPD-injected Tlr9(-/-) BALB/c mice develop higher autoantibody titers against RNA, neutrophil cytoplasmic Ags, and myeloperoxidase than do TMPD-injected wild-type BALB/c mice. The TMP-injected Tlr9(-/-) mice, and not the wild-type mice, also develop a marked increase in glomerular IgG deposition and infiltrating granulocytes, much more severe glomerulonephritis, and a reduced lifespan. Collectively, the data point to a major role for TLR7 in the response to self-antigens in this model of experimental autoimmunity. Therefore, the BALB/c pristane model recapitulates other TLR7-driven spontaneous models of SLE and is negatively regulated by TLR9.


Subject(s)
Cell Lineage , Lupus Nephritis/pathology , Myeloid Cells/pathology , Toll-Like Receptor 9/deficiency , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Flow Cytometry , Immunosuppressive Agents/toxicity , Lupus Nephritis/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Cells/immunology , Polymerase Chain Reaction , Terpenes/toxicity
14.
Proc Natl Acad Sci U S A ; 112(7): E710-7, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25646421

ABSTRACT

Cytosolic DNA-sensing pathways that signal via Stimulator of interferon genes (STING) mediate immunity to pathogens and also promote autoimmune pathology in DNaseII- and DNaseIII-deficient mice. In contrast, we report here that STING potently suppresses inflammation in a model of systemic lupus erythematosus (SLE). Lymphoid hypertrophy, autoantibody production, serum cytokine levels, and other indicators of immune activation were markedly increased in STING-deficient autoimmune-prone mice compared with STING-sufficient littermates. As a result, STING-deficient autoimmune-prone mice had significantly shorter lifespans than controls. Importantly, Toll-like receptor (TLR)-dependent systemic inflammation during 2,6,10,14-tetramethylpentadecane (TMPD)-mediated peritonitis was similarly aggravated in STING-deficient mice. Mechanistically, STING-deficient macrophages failed to express negative regulators of immune activation and thus were hyperresponsive to TLR ligands, producing abnormally high levels of proinflammatory cytokines. This hyperreactivity corresponds to dramatically elevated numbers of inflammatory macrophages and granulocytes in vivo. Collectively these findings reveal an unexpected negative regulatory role for STING, having important implications for STING-directed therapies.


Subject(s)
Autoimmunity/physiology , Membrane Proteins/physiology , Animals , Autoantibodies/biosynthesis , Dendritic Cells/immunology , Gene Expression Regulation/physiology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/physiology , Interferons/physiology , Lymphocyte Activation , Membrane Proteins/genetics , Mice
16.
J Immunol ; 195(8): 3507-12, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26432899

ABSTRACT

Distinct families of germline-encoded pattern recognition receptors can sense both microbial and endogenous nucleic acids. These DNA and RNA sensors include endosomal TLRs and cytosolic sensors upstream of stimulator of type I IFN genes (STING) and MAVS. The existence of overlapping specificities for both foreign and self nucleic acids suggests that, under optimal conditions, the activity of these receptors is finely tuned to effectively mediate host defense yet constrain pathogenic self-reactivity. This equilibrium becomes disrupted with the loss of either TLR9 or STING. To maintain immune protection, this loss can be counterbalanced by the elevated response of an alternative receptor(s). Unfortunately, this adjustment can lead to an increased risk for the development of systemic autoimmunity, as evidenced by the exacerbated clinical disease manifestations of TLR9-deficient and STING-deficient autoimmune-prone mice. These studies underscore the delicate balance normally maintained by tonic signals that prevent unchecked immune responses to nucleic acids released during infections and cellular duress or death.


Subject(s)
Autoimmune Diseases/immunology , DNA/immunology , Membrane Proteins/immunology , RNA/immunology , Toll-Like Receptor 9/immunology , Animals , Humans , Mice
17.
J Immunol ; 195(11): 5251-60, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26519529

ABSTRACT

Type 2 effector production of IL-13, a demonstrated requirement in models of fibrosis, is routinely ascribed to CD4(+) Th2 cells. We now demonstrate a major role for CD8(+) T cells in a murine model of sterile lung injury. These pulmonary CD8(+) T cells differentiate into IL-13-producing Tc2 cells and play a major role in a bleomycin-induced model of fibrosis. Differentiation of these Tc2 cells in the lung requires IL-21, and bleomycin treated IL-21- and IL-21R-deficient mice develop inflammation but not fibrosis. Moreover, IL-21R-expressing CD8(+) cells are sufficient to reconstitute the fibrotic response in IL-21R-deficient mice. We further show that the combination of IL-4 and IL-21 skews naive CD8(+) T cells to produce IL-21, which, in turn, acts in an autocrine manner to support robust IL-13 production. Our data reveal a novel pathway involved in the onset and regulation of pulmonary fibrosis and identify Tc2 cells as key mediators of fibrogenesis.


Subject(s)
Interleukin-13/biosynthesis , Interleukin-21 Receptor alpha Subunit/genetics , Interleukins/immunology , Pulmonary Fibrosis/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Bleomycin , Cell Differentiation/immunology , Cells, Cultured , Inflammation/immunology , Interleukin-13/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Interleukins/biosynthesis , Interleukins/genetics , Lung/cytology , Lung/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/cytology
18.
J Immunol ; 194(4): 1403-7, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25601924

ABSTRACT

In mice that fail to express the phagolysosomal endonuclease DNase II and the type I IFN receptor, excessive accrual of undegraded DNA results in a STING-dependent, TLR-independent inflammatory arthritis. These double-knockout (DKO) mice develop additional indications of systemic autoimmunity, including anti-nuclear autoantibodies and splenomegaly, that are not found in Unc93b1(3d/3d) DKO mice and, therefore, are TLR dependent. The DKO autoantibodies predominantly detect RNA-associated autoantigens, which are commonly targeted in TLR7-dominated systemic erythematosus lupus-prone mice. To determine whether an inability of TLR9 to detect endogenous DNA could explain the absence of dsDNA-reactive autoantibodies in DKO mice, we used a novel class of bifunctional autoantibodies, IgM/DNA dual variable domain Ig molecules, to activate B cells through a BCR/TLR9-dependent mechanism. DKO B cells could not respond to the IgM/DNA dual variable domain Ig molecule, despite a normal response to both anti-IgM and CpG ODN 1826. Thus, DKO B cells only respond to RNA-associated ligands because DNase II-mediated degradation of self-DNA is required for TLR9 activation.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Autoimmunity/immunology , B-Lymphocytes/immunology , Endodeoxyribonucleases/deficiency , Lymphocyte Activation/immunology , Animals , Antibody Specificity/immunology , DNA , Endodeoxyribonucleases/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Ligands , Mice , Mice, Knockout , Receptors, Antigen, B-Cell/immunology , Toll-Like Receptor 9/immunology
19.
J Immunol ; 194(3): 873-7, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25548216

ABSTRACT

Innate immune pattern recognition receptors sense nucleic acids from microbes and orchestrate cytokine production to resolve infection. Inappropriate recognition of host nucleic acids also results in autoimmune disease. In this study, we use a model of inflammation resulting from accrual of self DNA (DNase II(-/-) type I IFN receptor [Ifnar](-/-)) to understand the role of pattern recognition receptor-sensing pathways in arthritis and autoantibody production. Using triple knockout (TKO) mice deficient in DNase II/IFNaR together with deficiency in either stimulator of IFN genes (STING) or absent in melanoma 2 (AIM2), we reveal central roles for the STING and AIM2 pathways in arthritis. AIM2 TKO mice show limited inflammasome activation and, similar to STING TKO mice, have reduced inflammation in joints. Surprisingly, autoantibody production is maintained in AIM2 and STING TKO mice, whereas DNase II(-/-) Ifnar(-/-) mice also deficient in Unc93b, a chaperone required for TLR7/9 endosomal localization, fail to produce autoantibodies to nucleic acids. Collectively, these data support distinct roles for cytosolic and endosomal nucleic acid-sensing pathways in disease manifestations.


Subject(s)
Antibody Formation/immunology , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Autoantibodies/immunology , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/deficiency , Toll-Like Receptors/genetics , Animals , Arthritis, Experimental/diagnosis , Arthritis, Experimental/metabolism , Autoantigens/genetics , Autoantigens/immunology , Cluster Analysis , Cytokines/biosynthesis , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Immunity, Innate , Inflammation Mediators/metabolism , Male , Mice , Mice, Knockout , Phenotype , Signal Transduction , Toll-Like Receptors/metabolism
20.
J Immunol ; 194(6): 2504-12, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25681333

ABSTRACT

Endosomal TLRs play an important role in systemic autoimmune diseases, such as systemic erythematosus lupus, in which DNA- and RNA-associated autoantigens activate autoreactive B cells through TLR9- and TLR7-dependent pathways. Nevertheless, TLR9-deficient autoimmune-prone mice develop more severe clinical disease, whereas TLR7-deficient and TLR7/9-double deficient autoimmune-prone mice develop less severe disease. To determine whether the regulatory activity of TLR9 is B cell intrinsic, we directly compared the functional properties of autoantigen-activated wild-type, TLR9-deficient, and TLR7-deficient B cells in an experimental system in which proliferation depends on BCR/TLR coengagement. In vitro, TLR9-deficient cells are less dependent on survival factors for a sustained proliferative response than are either wild-type or TLR7-deficient cells. The TLR9-deficient cells also preferentially differentiate toward the plasma cell lineage, as indicated by expression of CD138, sustained expression of IRF4, and other molecular markers of plasma cells. In vivo, autoantigen-activated TLR9-deficient cells give rise to greater numbers of autoantibody-producing cells. Our results identify distinct roles for TLR7 and TLR9 in the differentiation of autoreactive B cells that explain the capacity of TLR9 to limit, as well as TLR7 to promote, the clinical features of systemic erythematosus lupus.


Subject(s)
B-Lymphocytes/immunology , Receptors, Antigen, B-Cell/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/immunology , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , Autoantigens/immunology , Autoimmunity/genetics , Autoimmunity/immunology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation/genetics , Cells, Cultured , Flow Cytometry , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Plasma Cells/immunology , Plasma Cells/metabolism , Receptors, Antigen, B-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rheumatoid Factor/immunology , Syndecan-1/immunology , Syndecan-1/metabolism , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/genetics , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Transcriptome/immunology
SELECTION OF CITATIONS
SEARCH DETAIL