Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Control Release ; 368: 329-343, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38431094

ABSTRACT

Rheumatoid arthritis (RA) is a prevalent autoimmune disease characterized by excessive inflammation in the joints. Glucocorticoid drugs are used clinically to manage RA symptoms, while their dosage and duration need to be tightly controlled due to severe adverse effects. Using dexamethasone (DEX) as a model drug, we explored here whether peptide-guided delivery could increase the safety and therapeutic index of glucocorticoids for RA treatment. Using multiple murine RA models such as collagen-induced arthritis (CIA), we found that CRV, a macrophage-targeting peptide, can selectively home to the inflammatory synovium of RA joints upon intravenous injection. The expression of the CRV receptor, retinoid X receptor beta (RXRB), was also elevated in the inflammatory synovium, likely being the basis of CRV targeting. CRV-conjugated DEX increased the accumulation of DEX in the inflamed synovium but not in healthy organs of CIA mice. Therefore, CRV-DEX demonstrated a stronger efficacy to suppress synovial inflammation and alleviate cartilage/bone destruction. Meanwhile, CRV conjugation reduced immune-related adverse effects of DEX even after a long-term use. Last, we found that RXRB expression was significantly elevated in human patient samples, demonstrating the potential of clinical translation. Taken together, we provide a novel, peptide-targeted strategy to improve the therapeutic efficacy and safety of glucocorticoids for RA treatment.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Humans , Mice , Animals , Glucocorticoids/therapeutic use , Arthritis, Rheumatoid/drug therapy , Inflammation , Arthritis, Experimental/drug therapy , Peptides/therapeutic use , Therapeutic Index
2.
Arterioscler Thromb Vasc Biol ; 44(4): 807-821, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38269589

ABSTRACT

BACKGROUND: Rheumatic heart disease is the major cause of valvular heart disease in developing nations. Endothelial cells (ECs) are considered crucial contributors to rheumatic heart disease, but greater insight into their roles in disease progression is needed. METHODS: We used a Cdh5-driven EC lineage-tracing approach to identify and track ECs in the K/B.g7 model of autoimmune valvular carditis. Single-cell RNA sequencing was used to characterize the EC populations in control and inflamed mitral valves. Immunostaining and conventional histology were used to evaluate lineage tracing and validate single-cell RNA-sequencing findings. The effects of VEGFR3 (vascular endothelial growth factor receptor 3) and VEGF-C (vascular endothelial growth factor C) inhibitors were tested in vivo. The functional impact of mitral valve disease in the K/B.g7 mouse was evaluated using echocardiography. Finally, to translate our findings, we analyzed valves from human patients with rheumatic heart disease undergoing mitral valve replacements. RESULTS: Lineage tracing in K/B.g7 mice revealed new capillary lymphatic vessels arising from valve surface ECs during the progression of disease in K/B.g7 mice. Unsupervised clustering of mitral valve single-cell RNA-sequencing data revealed novel lymphatic valve ECs that express a transcriptional profile distinct from other valve EC populations including the recently identified PROX1 (Prospero homeobox protein 1)+ lymphatic valve ECs. During disease progression, these newly identified lymphatic valve ECs expand and upregulate a profibrotic transcriptional profile. Inhibiting VEGFR3 through multiple approaches prevented expansion of this mitral valve lymphatic network. Echocardiography demonstrated that K/B.g7 mice have left ventricular dysfunction and mitral valve stenosis. Valve lymphatic density increased with age in K/B.g7 mice and correlated with worsened ventricular dysfunction. Importantly, human rheumatic valves contained similar lymphatics in greater numbers than nonrheumatic controls. CONCLUSIONS: These studies reveal a novel mode of inflammation-associated, VEGFR3-dependent postnatal lymphangiogenesis in murine autoimmune valvular carditis, with similarities to human rheumatic heart disease.


Subject(s)
Heart Valve Diseases , Lymphatic Vessels , Myocarditis , Rheumatic Heart Disease , Humans , Mice , Animals , Rheumatic Heart Disease/genetics , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/pathology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Lymphatic Vessels/metabolism , Heart Valve Diseases/pathology , Disease Progression , RNA
3.
Arterioscler Thromb Vasc Biol ; 43(6): 943-957, 2023 06.
Article in English | MEDLINE | ID: mdl-37021574

ABSTRACT

BACKGROUND: Inflammation is a key driver of cardiovascular pathology, and many systemic autoimmune/rheumatic diseases are accompanied by increased cardiac risk. In the K/B.g7 mouse model of coexisting systemic autoantibody-mediated arthritis and valvular carditis, valve inflammation depends on macrophage production of TNF (tumor necrosis factor) and IL-6 (interleukin-6). Here, we sought to determine if other canonical inflammatory pathways participate and to determine whether TNF signaling through TNFR1 (tumor necrosis factor receptor 1) on endothelial cells is required for valvular carditis. METHODS: We first asked if type 1, 2, or 3 inflammatory cytokine systems (typified by IFNγ, IL-4, and IL-17, respectively) were critical for valvular carditis in K/B.g7 mice, using a combination of in vivo monoclonal antibody blockade and targeted genetic ablation studies. To define the key cellular targets of TNF, we conditionally deleted its main proinflammatory receptor, TNFR1, in endothelial cells. We analyzed how the absence of endothelial cell TNFR1 affected valve inflammation, lymphangiogenesis, and the expression of proinflammatory genes and molecules. RESULTS: We found that typical type 1, 2, and 3 inflammatory cytokine systems were not required for valvular carditis, apart from a known initial requirement of IL-4 for autoantibody production. Despite expression of TNFR1 on a wide variety of cell types in the cardiac valve, deleting TNFR1 specifically on endothelial cells protected K/B.g7 mice from valvular carditis. This protection was accompanied by reduced expression of VCAM-1 (vascular cell adhesion molecule), fewer valve-infiltrating macrophages, reduced pathogenic lymphangiogenesis, and diminished proinflammatory gene expression. CONCLUSIONS: TNF and IL-6 are the main cytokines driving valvular carditis in K/B.g7 mice. The interaction of TNF with TNFR1 specifically on endothelial cells promotes cardiovascular pathology in the setting of systemic autoimmune/rheumatic disease, suggesting that therapeutic targeting of the TNF:TNFR1 interaction could be beneficial in this clinical context.


Subject(s)
Heart Valve Diseases , Receptors, Tumor Necrosis Factor, Type I , Animals , Mice , Autoantibodies , Cytokines , Endothelial Cells/metabolism , Inflammation , Interleukin-4 , Interleukin-6/genetics , Myocarditis/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha , Vascular Cell Adhesion Molecule-1/metabolism
4.
J Immunol ; 208(12): 2643-2651, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35867674

ABSTRACT

Systemic autoantibody-mediated diseases accelerate chronic cardiovascular disease in humans. In the K/B.g7 mouse model of spontaneous autoantibody-mediated inflammatory arthritis, valvular carditis arises in part because of Fc receptor-mediated activation of macrophages, leading to production of pathogenic TNF and IL-6. In this study, we explored whether impaired efferocytosis mediated by the interaction of CD47-expressing apoptotic cells with signal regulatory protein α (SIRPα) on macrophages contributes to disease progression in this model. CD47-expressing apoptotic cells and SIRPα+ macrophages were abundant in inflamed/rheumatic cardiac valves from both mice and humans. In vivo anti-CD47 blockade both prevented and treated valvular carditis in K/B.g7 mice. Blocking CD47 enhanced macrophage efferocytosis and reduced macrophage production of TNF and IL-6. These studies highlight the CD47:SIRPα interaction as a key driver of chronic cardiac valve inflammation and suggest these molecules as potential therapeutic targets to reduce cardiovascular disease risk in autoantibody-driven inflammatory diseases.


Subject(s)
CD47 Antigen/metabolism , Myocarditis , Animals , Antigens, Differentiation/metabolism , Humans , Inflammation/metabolism , Interleukin-6/metabolism , Macrophages , Mice , Myocarditis/pathology , Phagocytosis , Receptors, Immunologic/metabolism
5.
Sci Adv ; 8(16): eabj5227, 2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35452291

ABSTRACT

Here, we report that the LynB splice variant of the Src-family kinase Lyn exerts a dominant immunosuppressive function in vivo, whereas the LynA isoform is uniquely required to restrain autoimmunity in female mice. We used CRISPR-Cas9 gene editing to constrain lyn splicing and expression, generating single-isoform LynA knockout (LynAKO) or LynBKO mice. Autoimmune disease in total LynKO mice is characterized by production of antinuclear antibodies, glomerulonephritis, impaired B cell development, and overabundance of activated B cells and proinflammatory myeloid cells. Expression of LynA or LynB alone uncoupled the developmental phenotype from the autoimmune disease: B cell transitional populations were restored, but myeloid cells and differentiated B cells were dysregulated. These changes were isoform-specific, sexually dimorphic, and distinct from the complete LynKO. Despite the apparent differences in disease etiology and penetrance, loss of either LynA or LynB had the potential to induce severe autoimmune disease with parallels to human systemic lupus erythematosus (SLE).

6.
J Immunol ; 205(10): 2840-2849, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33008950

ABSTRACT

Polyunsaturated fatty acids (PUFAs) and their metabolites are potent regulators of inflammation. Generally, omega (n)-3 PUFAs are considered proresolving whereas n-6 PUFAs are classified as proinflammatory. In this study, we characterized the inflammatory response in murine peritonitis and unexpectedly found the accumulation of adrenic acid (AdA), a poorly studied n-6 PUFA. Functional studies revealed that AdA potently inhibited the formation of the chemoattractant leukotriene B4 (LTB4), specifically in human neutrophils, and this correlated with a reduction of its precursor arachidonic acid (AA) in free form. AdA exposure in human monocyte-derived macrophages enhanced efferocytosis of apoptotic human neutrophils. In vivo, AdA treatment significantly alleviated arthritis in an LTB4-dependent murine arthritis model. Our findings are, to our knowledge, the first to indicate that the n-6 fatty acid AdA effectively blocks production of LTB4 by neutrophils and could play a role in resolution of inflammation in vivo.


Subject(s)
Anti-Inflammatory Agents/metabolism , Arthritis, Experimental/immunology , Fatty Acids, Omega-6/metabolism , Fatty Acids, Unsaturated/metabolism , Peritonitis/immunology , Animals , Anti-Inflammatory Agents/analysis , Arachidonic Acid/metabolism , Arthritis, Experimental/pathology , Cells, Cultured , Fatty Acids, Omega-6/analysis , Fatty Acids, Unsaturated/analysis , Humans , Leukotriene B4/metabolism , Lipidomics , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Transgenic , Neutrophils/immunology , Neutrophils/metabolism , Peritoneal Lavage , Peritonitis/pathology , Primary Cell Culture , THP-1 Cells , Zymosan/administration & dosage , Zymosan/immunology
7.
Nat Commun ; 10(1): 951, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862842

ABSTRACT

Targeted noninvasive control of the nervous system and end-organs may enable safer and more effective treatment of multiple diseases compared to invasive devices or systemic medications. One target is the cholinergic anti-inflammatory pathway that consists of the vagus nerve to spleen circuit, which has been stimulated with implantable devices to improve autoimmune conditions such as rheumatoid arthritis. Here we report that daily noninvasive ultrasound (US) stimulation targeting the spleen significantly reduces disease severity in a mouse model of inflammatory arthritis. Improvements are observed only with specific parameters, in which US can provide both protective and therapeutic effects. Single cell RNA sequencing of splenocytes and experiments in genetically-immunodeficient mice reveal the importance of both T and B cell populations in the anti-inflammatory pathway. These findings demonstrate the potential for US stimulation of the spleen to treat inflammatory diseases.


Subject(s)
Arthritis, Experimental/physiopathology , Arthritis, Experimental/therapy , Spleen/innervation , Spleen/physiopathology , Ultrasonic Therapy/methods , Animals , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/physiopathology , Arthritis, Rheumatoid/therapy , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cholinergic Fibers/immunology , Cholinergic Fibers/physiology , Gene Expression , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/immunology , Neuroimmunomodulation/genetics , Spleen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vagus Nerve Stimulation/methods
8.
Arthritis Res Ther ; 20(1): 154, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30053832

ABSTRACT

BACKGROUND: Pain accompanies rheumatoid arthritis and other chronic inflammatory conditions and is difficult to manage. Although opioids provide potent analgesia, chronic opioid use can cause tolerance and addiction. Recent studies have demonstrated functional interactions between chemokine and opioid receptor signaling pathways. Reported heterodimerization of chemokine and opioid receptors led our group to develop bivalent compounds that bind both types of receptors, with the goal of targeting opioids to sites of inflammation. MCC22 is a novel bivalent compound containing a CCR5 antagonist and mu opioid receptor (MOR) agonist pharmacophores linked through a 22-atom spacer. We evaluated the efficacy of MCC22 in the K/B.g7 T-cell receptor transgenic mouse model of spontaneous inflammatory arthritis. METHODS: MCC22 or morphine was administered intraperitoneally at varying doses to arthritic K/B.g7 mice or nonarthritic control mice. Mechanical pain hypersensitivity was measured each day before and after drug administration, using the electronic von Frey test. The potency of MCC22 relative to that of morphine was calculated. Functional readouts of pain included grip strength and nesting behavior. A separate dosing regimen was used to determine whether the drugs induced pharmacologic tolerance. RESULTS: MCC22 provided ~ 3000-fold more potent analgesia than morphine in this model. Daily treatment with MCC22 also led to a cumulative analgesic effect, reducing the daily baseline pain level. MCC22 produced no observable analgesic effect in nonarthritic control mice. Importantly, repeated administration of MCC22 did not induce pharmacologic tolerance, whereas a similar regimen of morphine did. Both grip strength and nesting behaviors improved among arthritic mice treated with MCC22. Ankle thickness and arthritis scores were not affected by MCC22. The analgesic effect of MCC22 was abolished in K/B.g7 mice genetically lacking CCR5, demonstrating the receptor specificity of the antagonist pharmacophore. CONCLUSIONS: MCC22 is a novel bivalent ligand that targets CCR5 and MOR. Our findings demonstrate that MCC22 provides highly potent analgesia and improved functional outcomes in a model of inflammatory arthritis, without inducing typical opioid tolerance. These findings suggest that MCC22 or similar compounds could be used to treat the pain associated with inflammatory arthritis and related conditions, while minimizing the risks typically associated with chronic opioid use.


Subject(s)
Analgesics/pharmacology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Isoquinolines/pharmacology , Pain , Piperidines/pharmacology , Receptors, CCR5/drug effects , Receptors, Opioid, mu/agonists , Animals , Arthritis, Experimental/complications , Arthritis, Rheumatoid/complications , Drug Tolerance , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pain/etiology , Receptors, CCR5/agonists , Receptors, CCR5/metabolism
9.
Circulation ; 137(23): 2478-2493, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29386201

ABSTRACT

BACKGROUND: Valvular heart disease is common and affects the mitral valve (MV) most frequently. Despite the prevalence of MV disease (MVD), the cellular and molecular pathways that initiate and perpetuate it are not well understood. METHODS: K/B.g7 T-cell receptor transgenic mice spontaneously develop systemic autoantibody-associated autoimmunity, leading to fully penetrant fibroinflammatory MVD and arthritis. We used multiparameter flow cytometry, intracellular cytokine staining, and immunofluorescent staining to characterize the cells in inflamed K/B.g7 MVs. We used genetic approaches to study the contribution of mononuclear phagocytes (MNPs) to MVD in this model. Specifically, we generated K/B.g7 mice in which either CX3CR1 or CD301b/macrophage galactose N-acetylgalactosamine-specific lectin 2 (MGL2)-expressing MNPs were ablated. Using K/B.g7 mice expressing Cx3Cr1-Cre, we conditionally deleted critical inflammatory molecules from MNPs, including the Fc-receptor signal-transducing tyrosine kinase Syk and the cell adhesion molecule very late antigen-4. We performed complementary studies using monoclonal antibodies to block key inflammatory molecules. We generated bone marrow chimeric mice to define the origin of the inflammatory cells present in the MV and to determine which valve cells respond to the proinflammatory cytokine tumor necrosis factor (TNF). Finally, we examined specimens from patients with rheumatic heart disease to correlate our findings to human pathology. RESULTS: MNPs comprised the vast majority of MV-infiltrating cells; these MNPs expressed CX3CR1 and CD301b/MGL2. Analogous cells were present in human rheumatic heart disease valves. K/B.g7 mice lacking CX3CR1 or in which CD301b/MGL2-expressing MNPs were ablated were protected from MVD. The valve-infiltrating CD301b/MGL2+ MNPs expressed tissue-reparative molecules including arginase-1 and resistin-like molecule α. These MNPs also expressed the proinflammatory cytokines TNF and interleukin-6, and antibody blockade of these cytokines prevented MVD. Deleting Syk from CX3CR1-expressing MNPs reduced their TNF and interleukin-6 production and also prevented MVD. TNF acted through TNF receptor-1 expressed on valve-resident cells to increase the expression of vascular cell adhesion molecule-1. Conditionally deleting the vascular cell adhesion molecule-1 ligand very late antigen-4 from CX3CR1-expressing MNPs prevented MVD. CONCLUSIONS: CD301b/MGL2+ MNPs are key drivers of autoimmune MVD in K/B.g7 mice and are also present in human rheumatic heart disease. We define key inflammatory molecules that drive MVD in this model, including Syk, TNF, interleukin-6, very late antigen-4, and vascular cell adhesion molecule-1.


Subject(s)
Autoimmune Diseases/immunology , Heart Valve Diseases/immunology , Lectins, C-Type/immunology , Phagocytes/immunology , Allogeneic Cells , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Bone Marrow Transplantation , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/immunology , Fibrosis , Heart Valve Diseases/genetics , Heart Valve Diseases/pathology , Humans , Inflammation , Interleukin-6/genetics , Interleukin-6/immunology , Lectins, C-Type/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Phagocytes/pathology , Rheumatic Heart Disease/pathology , Transplantation Chimera/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
10.
J Immunol ; 199(1): 33-38, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28539428

ABSTRACT

Despite accounting for 10-30% of the T cell population in mice and humans, the role of dual TCR-expressing T cells in immunity remains poorly understood. It has been hypothesized that dual TCR T cells pose an autoimmune hazard by allowing self-reactive TCRs to escape thymic selection. We revisited this hypothesis using the NOD murine model of type 1 diabetes. We bred NOD mice hemizygous at both TCRα and ß (TCRα+/- ß+/-) loci, rendering them incapable of producing dual TCR T cells. We found that the lack of dual TCRα expression skewed the insulin-specific thymocyte population toward greater regulatory T (Treg) cell commitment, resulting in a more tolerogenic Treg to conventional T cell ratio and protection from diabetes. These data support a novel hypothesis by which dual TCR expression can promote autoimmunity by limiting agonist selection of self-reactive thymocytes into the Treg cell lineage.


Subject(s)
Autoimmunity , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Diabetes Mellitus, Type 1/immunology , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/physiology , Thymocytes/immunology
11.
Arthritis Rheumatol ; 68(8): 1849-55, 2016 08.
Article in English | MEDLINE | ID: mdl-26882006

ABSTRACT

OBJECTIVE: Th17 cells and interleukin-17 (IL-17) cytokine family members are implicated in the pathogenesis of many rheumatic diseases. Most studies in mouse models of inflammatory arthritis have demonstrated a key role for the proinflammatory cytokine IL-17A and its receptor, the IL-17 receptor (IL-17R) A/C heterodimer. The aim of this study was to use a rigorous genetic approach to evaluate the contribution of Th17 cells and IL-17 in the autoantibody-dependent KRN T cell receptor-transgenic mouse model of arthritis. METHODS: We bred KRN mice expressing the major histocompatibility complex class II molecule A(g7) (referred to as K/B/g7 mice) and genetically lacking the related cytokines IL-17A and IL-17F or their critical receptor subunit, IL-17RA. Using bone marrow transplantation, we generated mice in which hematopoietic cells from K/B/g7 donor mice lacked the key Th17-differentiating transcription factor, retinoic acid receptor-related orphan nuclear receptor γt (Rorγt). RESULTS: K/B/g7 mice lacking both IL-17A and IL-17F produced normal titers of pathogenic autoantibodies, and arthritis developed in a typical manner. Similarly, neither IL-17RA nor Rorγt expression by hematopoietic cells was required for disease development in this model. CONCLUSION: Despite prior reports suggesting that Th17 cells and IL-17A are crucially involved in the pathogenesis of arthritis in K/BxN mice, the results presented here provide genetic evidence that IL-17A and IL-17F, IL-17RA, and Rorγt expression by hematopoietic cells are dispensable for normal arthritis progression in the K/B/g7 mouse model system. We discuss potential explanations for the discrepancies between these 2 highly similar model systems. These findings plus those in other mouse models of arthritis provide insight regarding why therapeutic biologic agents targeting the Th17/IL-17 axis are beneficial in some human rheumatic diseases but not others.


Subject(s)
Interleukin-17/physiology , Receptors, Antigen, T-Cell/genetics , Th17 Cells/physiology , Animals , Mice , Mice, Transgenic
12.
PLoS One ; 10(12): e0145762, 2015.
Article in English | MEDLINE | ID: mdl-26693713

ABSTRACT

Dual TCRα-expressing T cells outnumber dual TCRß-expressing cells by ~10:1. As a result, efforts to understand how dual TCR T cells impact immunity have focused on dual TCRα expression; dual TCRß expression remains understudied. We recently demonstrated, however, that dual TCRß expression accelerated disease in a TCR transgenic model of autoimmune arthritis through enhanced positive selection efficiency, indicating that dual TCRß expression, though rare, can impact thymic selection. Here we generated mice hemizygous for TCRα, TCRß, or both on the C57BL/6 background to investigate the impact bi-allelic TCR chain recombination has on T cell development, repertoire diversity, and autoimmunity. Lack of bi-allelic TCRα or TCRß recombination reduced αß thymocyte development efficiency, and the absence of bi-allelic TCRß recombination promoted γδ T cell development. However, we observed no differences in the numbers of naïve and expanded antigen-specific T cells between TCRα+/-ß+/- and wildtype mice, and TCR repertoire analysis revealed only subtle differences in Vß gene usage. Finally, the absence of dual TCR T cells did not impact induced experimental autoimmune encephalomyelitis pathogenesis. Thus, despite more stringent allelic exclusion of TCRß relative to TCRα, bi-allelic TCRß expression can measurably impact thymocyte development and is necessary for maintaining normal αß/γδ T cell proportions.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Regulation/immunology , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/immunology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/microbiology , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/genetics
13.
Arthritis Rheumatol ; 66(4): 852-62, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24757138

ABSTRACT

OBJECTIVE: Arthritis and valvular carditis coexist in several human rheumatic diseases, including systemic lupus erythematosus, rheumatic fever, and rheumatoid arthritis. T cell receptor-transgenic K/BxN mice develop spontaneous autoantibody-associated arthritis and valvular carditis. The common Fc receptor γ (FcRγ) signaling chain is required for carditis to develop in K/BxN mice. FcRγ pairs with numerous receptors in a variety of cells. The aim of this study was to identify the FcRγ-associated receptors and Fcγ receptor (FcγR)-expressing cells that mediate valvular carditis in this model. METHODS: We bred K/BxN mice lacking the genes that encode activating Fcγ receptors (FcγRI, FcγRIII, and FcγRIV), and we assessed these mice for valvular carditis. We similarly evaluated complement component C3-deficient K/BxN mice. Immunohistochemistry, bone marrow transplantation, and macrophage depletion were used to define the key FcRγ-expressing cell type. RESULTS: Genetic deficiency of only one of the activating Fcγ receptors did not prevent carditis, whereas deficiency of all 3 activating Fcγ receptors did. Further analysis demonstrated that FcγRIII and FcγRIV were the key drivers of valve inflammation; FcγRI was dispensable. C3 was not required. FcRγ expression by radioresistant cells was critical for valvular carditis to develop, and further analysis indicated that macrophages were the key candidate FcγR-expressing effectors of carditis. CONCLUSION: FcγRIII and FcγRIV act redundantly to promote valvular carditis in K/BxN mice with systemic autoantibody-associated arthritis. Macrophage depletion reduced the severity of valve inflammation. These findings suggest that pathogenic autoantibodies engage Fcγ receptors on macrophages to drive valvular carditis. Our study provides new insight into the pathogenesis of cardiovascular inflammation in the setting of autoantibody-associated chronic inflammatory diseases.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Macrophages/immunology , Myocarditis/immunology , Receptors, IgG/metabolism , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Autoantibodies/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Myocarditis/genetics , Myocarditis/metabolism , Receptors, IgG/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
14.
Immunity ; 39(1): 111-22, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23871208

ABSTRACT

Immune cells sense microbial products through Toll-like receptors (TLR), which trigger host defense responses including type 1 interferons (IFNs) secretion. A coding polymorphism in the protein tyrosine phosphatase nonreceptor type 22 (PTPN22) gene is a susceptibility allele for human autoimmune and infectious disease. We report that Ptpn22 selectively regulated type 1 IFN production after TLR engagement in myeloid cells. Ptpn22 promoted host antiviral responses and was critical for TLR agonist-induced, type 1 IFN-dependent suppression of inflammation in colitis and arthritis. PTPN22 directly associated with TNF receptor-associated factor 3 (TRAF3) and promotes TRAF3 lysine 63-linked ubiquitination. The disease-associated PTPN22W variant failed to promote TRAF3 ubiquitination, type 1 IFN upregulation, and type 1 IFN-dependent suppression of arthritis. The findings establish a candidate innate immune mechanism of action for a human autoimmunity "risk" gene in the regulation of host defense and inflammation.


Subject(s)
Autoimmunity/immunology , Immunity/immunology , Interferon Type I/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Toll-Like Receptors/immunology , Animals , Arthritis/genetics , Arthritis/immunology , Autoimmunity/genetics , Cell Line , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Dextran Sulfate/immunology , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunity/genetics , Immunoblotting , Interferon Type I/genetics , Interferon Type I/metabolism , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology , TNF Receptor-Associated Factor 3/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Ubiquitination/immunology
15.
J Immunol ; 191(3): 1055-62, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23794629

ABSTRACT

The class A macrophage scavenger receptor Msr1 (SR-A, CD204) has been reported to participate in the maintenance of immunological tolerance. We investigated the role of Msr1 in a mouse model of autoantibody-dependent arthritis. Genetic deficiency of Msr1 in K/BxN TCR transgenic mice decreased the incidence and severity of arthritis because of decreased autoantibody production. Despite normal initial activation of autoreactive CD4(+) T cells, potentially autoreactive B cells in Msr1(-/-) K/BxN mice retained a naive phenotype and did not expand. This was not due to an intrinsic B cell defect. Rather, we found that macrophages lacking Msr1 were inefficient at taking up the key autoantigen glucose-6-phosphate isomerase and that Msr1-deficient mice had elevated serum concentrations of glucose-6-phosphate isomerase. Arthritis developed normally when bone marrow from Msr1(-/-) K/BxN mice was transplanted into hosts whose macrophages did express Msr1. Thus, Msr1 can regulate the concentration of a soluble autoantigen. In this model, the absence of Msr1 led to higher levels of soluble autoantigen and protected mice from developing pathogenic autoantibodies, likely because of altered cognate interactions of autoreactive T and B cells with impaired differentiation of follicular Th cells.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Autoimmunity , B-Lymphocytes/immunology , Glucose-6-Phosphate Isomerase/immunology , Scavenger Receptors, Class A/metabolism , Animals , Arthritis, Experimental/immunology , Autoantibodies/biosynthesis , Autoantigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Glucose-6-Phosphate Isomerase/blood , Glucose-6-Phosphate Isomerase/metabolism , Lymphocyte Activation , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/immunology
16.
Arthritis Res Ther ; 14(6): R269, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23237573

ABSTRACT

INTRODUCTION: The effector functions of immunoglobulin G (IgG) are mediated by interaction of its Fc region with Fc receptors (FcγRs) and/or the complement system. The three main pathways of complement activation converge at C3. However, C3-independent pathways can activate C5 and other downstream complement components during IgG-initiated inflammatory responses. These C3-independent pathways of C5 activation are triggered by activating FcγRs in some systems or can be activated by factors of the coagulation cascade such as thrombin. Here we studied the interplay of C3, C5, and activating FcγRs in a model of spontaneous autoantibody-driven arthritis. METHODS: We utilized the K/BxN TCR transgenic mouse model of arthritis. We bred K/BxN mice bearing targeted or naturally-occurring mutations in one or more of the genes encoding complement components C3, C5, and FcRγ, the cytoplasmic signaling chain shared by the activating FcγRs. We measured arthritis development, the production of arthritogenic autoantibodies, T cell activation status and cytokine synthesis. In addition, we treated mice with anti-C5 monoclonal antibodies or with the thrombin inhibitor argatroban. RESULTS: We have previously shown that genetic deficiency of C5 protects K/BxN mice from the development of arthritis. We found here that C3-deficient K/BxN mice developed arthritis equivalent in severity to C3-sufficient animals. Arthritis also developed normally in K/BxN mice lacking both C3 and FcRγ, but could be ameliorated in these animals by treatment with anti-C5 monoclonal antibody or by treatment with argatroban. Production of arthritogenic autoantibodies, T cell activation, and T cell cytokine production were not affected by the absence of C3, C5, and/or FcRγ. CONCLUSIONS: In K/BxN mice, C5-dependent autoantibody-driven arthritis can occur in the genetic absence of both complement C3 and activating FcγRs. Our findings suggest that in this setting, thrombin activates C5 to provoke arthritis.


Subject(s)
Arthritis/immunology , Autoantibodies/immunology , Complement C3/immunology , Complement C5/immunology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antithrombins/pharmacology , Arginine/analogs & derivatives , Arthritis/genetics , Arthritis/metabolism , Autoantibodies/blood , Blood Coagulation/drug effects , Blood Coagulation/immunology , Complement Activation/drug effects , Complement Activation/immunology , Complement C3/deficiency , Complement C3/genetics , Complement C5/deficiency , Complement C5/genetics , Female , Flow Cytometry , Glucose-6-Phosphate Isomerase/immunology , Immunohistochemistry , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Pipecolic Acids/pharmacology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, IgG/deficiency , Receptors, IgG/genetics , Signal Transduction/drug effects , Signal Transduction/immunology , Sulfonamides
17.
Eur J Immunol ; 42(9): 2354-62, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22706882

ABSTRACT

Allelic exclusion of antigen receptor loci is a fundamental mechanism of immunological self-tolerance. Incomplete allelic exclusion leads to dual T-cell receptor (TCR) expression and can allow developing autoreactive αß T lymphocytes to escape clonal deletion. Because allelic exclusion at the TCR-ß locus is more stringent than at the TCR-α locus, dual TCR-ß expression has not been considered a likely contributor to autoimmunity. We show here that incomplete TCR-ß allelic exclusion permits developing thymocytes bearing the autoreactive, transgene-encoded KRN TCR to be positively selected more efficiently, thereby accelerating the onset of spontaneous autoimmune arthritis. Our findings highlight dual TCR-ß expression as a mechanism that can enhance the maturation of autoreactive pathogenic T cells and lead to more rapid development of autoimmune disease.


Subject(s)
Alleles , Arthritis/genetics , Arthritis/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , CD4 Antigens/genetics , CD4 Antigens/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocarditis/genetics , Myocarditis/immunology , Self Tolerance/genetics , Self Tolerance/immunology , T-Lymphocyte Subsets/immunology , Thymocytes/immunology , Transgenes
18.
Arthritis Res Ther ; 14(1): R29, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22314006

ABSTRACT

INTRODUCTION: Neuromedin U (NMU) is a neuropeptide with pro-inflammatory activity. The primary goal of this study was to determine if NMU promotes autoantibody-induced arthritis. Additional studies addressed the cellular source of NMU and sought to define the NMU receptor responsible for its pro-inflammatory effects. METHODS: Serum containing arthritogenic autoantibodies from K/BxN mice was used to induce arthritis in mice genetically lacking NMU. Parallel experiments examined whether NMU deficiency impacted the early mast-cell-dependent vascular leak response induced by these autoantibodies. Bone-marrow chimeric mice were generated to determine whether pro-inflammatory NMU is derived from hematopoietic cells or stromal cells. Mice lacking the known NMU receptors singly and in combination were used to determine susceptibility to serum-transferred arthritis and in vitro cellular responses to NMU. RESULTS: NMU-deficient mice developed less severe arthritis than control mice. Vascular leak was not affected by NMU deficiency. NMU expression by bone-marrow-derived cells mediated the pro-arthritogenic effect. Deficiency of all of the known NMU receptors, however, had no impact on arthritis severity and did not affect the ability of NMU to stimulate intracellular calcium flux. CONCLUSIONS: NMU-deficient mice are protected from developing autoantibody-induced inflammatory arthritis. NMU derived from hematopoietic cells, not neurons, promotes the development of autoantibody-induced inflammatory arthritis. This effect is mediated by a receptor other than the currently known NMU receptors.


Subject(s)
Arthritis/immunology , Autoantibodies/immunology , Neuropeptides/immunology , Receptors, Neurotransmitter/immunology , Animals , Arthritis/genetics , Arthritis/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Calcium/immunology , Calcium/metabolism , Female , Male , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Neuropeptides/deficiency , Neuropeptides/genetics , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Neurotensin/deficiency , Receptors, Neurotensin/genetics , Receptors, Neurotensin/immunology , Receptors, Neurotransmitter/deficiency , Receptors, Neurotransmitter/genetics , Spleen/immunology , Spleen/metabolism , Spleen/pathology
19.
J Immunol ; 187(5): 2702-10, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21795599

ABSTRACT

The immunopathogenic mechanisms mediating inflammation in multiorgan autoimmune diseases may vary between the different target tissues. We used the K/BxN TCR transgenic mouse model to investigate the contribution of CD4(+) T cells and ß(2) integrins in the pathogenesis of autoimmune arthritis and endocarditis. Depletion of CD4(+) T cells following the onset of arthritis specifically prevented the development of cardiac valve inflammation. Genetic absence of ß(2) integrins had no effect on the severity of arthritis and unexpectedly increased the extent of cardiovascular pathology. The exaggerated cardiac phenotype of the ß(2) integrin-deficient K/BxN mice was accompanied by immune hyperactivation and was linked to a defect in regulatory T cells. These findings are consistent with a model in which the development of arthritis in K/BxN mice relies primarily on autoantibodies, whereas endocarditis depends on an additional contribution of effector T cells. Furthermore, strategies targeting ß(2) integrins for the treatment of systemic autoimmune conditions need to consider not only the role of these molecules in leukocyte recruitment to sites of inflammation, but also their impact on the regulation of immunological tolerance.


Subject(s)
Autoimmune Diseases/immunology , CD18 Antigens/immunology , Myocarditis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Autoimmune Diseases/pathology , CD4-Positive T-Lymphocytes/immunology , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocarditis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...