Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Arthritis Rheumatol ; 67(7): 1868-80, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25777776

ABSTRACT

OBJECTIVE: Myeloperoxidase (MPO) locally contributes to organ damage in various chronic inflammatory conditions by generating reactive intermediates. The contribution of MPO in the development of experimental lupus is unknown. The aim of this study was to define the role of MPO in murine lupus nephritis (LN). METHODS: LN was induced in C57BL/6 wild-type (WT) and MPO knockout (MPO(-/-) ) mice by intraperitoneal injection of pristane. Autoimmunity and glomerulonephritis were assessed 20 and 40 weeks after pristane administration. Cell apoptosis, leukocyte accumulation, and cytokine levels in the peritoneal cavity of WT and MPO(-/-) mice were assessed 3 or 6 days after pristane injection. RESULTS: MPO(-/-) mice developed more severe nephritis than did WT mice 20 and 40 weeks after pristane injection, despite having reduced glomerular deposition of antibody and complement and diminished levels of markers of oxidative stress (oxidized DNA and glutathione sulfonamide). Enhancement of renal disease in MPO-deficient mice correlated with increased accumulation of CD4+ T cells and macrophages in glomeruli, which, in turn, was associated with augmented generation of CD4+ T cell responses and increased activation and migration of dendritic cells in secondary lymphoid organs. In addition, the enhanced renal injury in MPO(-/-) mice was associated with increased glomerular accumulation of neutrophils and deposition of neutrophil extracellular traps. MPO deficiency also increased early cell apoptosis, leukocyte accumulation, and proinflammatory cytokine expression in the peritoneum. CONCLUSION: MPO attenuates pristane-induced LN by inhibiting early inflammatory responses in the peritoneum and limiting the generation of CD4+ T cell autoimmunity in secondary lymphoid organs.


Subject(s)
Autoimmunity/physiology , CD4-Positive T-Lymphocytes/pathology , Lupus Nephritis/chemically induced , Lupus Nephritis/prevention & control , Peroxidase/physiology , Terpenes/adverse effects , Animals , Apoptosis/physiology , Cytokines/metabolism , Disease Models, Animal , Female , Injections, Intraperitoneal , Kidney Glomerulus/pathology , Lupus Nephritis/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/physiology , Peritoneum/metabolism , Peroxidase/deficiency , Peroxidase/genetics , Terpenes/administration & dosage
2.
Arthritis Rheumatol ; 66(4): 907-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24757143

ABSTRACT

OBJECTIVE: Myeloperoxidase (MPO) is implicated as a local mediator of tissue damage when released extracellularly in many chronic inflammatory diseases. The purpose of this study was to explore the role of endogenous MPO in experimental rheumatoid arthritis (RA). METHODS: K/BxN serum-transfer arthritis was induced in C57BL/6 wild-type (WT) and MPO knockout (MPO(-/-) ) mice, and disease development was assessed. MPO activity was measured in joint tissues from mice with or without K/BxN arthritis. Collagen-induced arthritis (CIA) was induced in WT and MPO(-/-) mice, and disease development and immune responses were examined. MPO expression was assessed in synovial biopsy samples from patients with active RA, and the effect of MPO on synovial fibroblasts was tested in vitro. RESULTS: MPO was up-regulated in the joints of mice with K/BxN arthritis, and MPO deficiency attenuated the severity of the disease without affecting circulating cytokine levels. In CIA, MPO(-/-) mice had enhanced CD4+ T cell responses and reduced frequency of regulatory T cells in the lymph nodes and spleen, as well as augmented interleukin-17A and diminished interferon-γ secretion by collagen-stimulated splenocytes, without an effect on circulating anticollagen antibody levels. Despite enhanced adaptive immunity in secondary lymphoid organs, CIA development was attenuated in MPO(-/-) mice. Intracellular and extracellular MPO was detected in the synovium of patients with active RA, and human MPO enhanced the proliferation and decreased the apoptosis of synovial fibroblasts in vitro. CONCLUSION: MPO contributes to the development of arthritis despite suppressing adaptive immunity in secondary lymphoid organs. This suggests distinct effects of local MPO on arthritogenic effector responses.


Subject(s)
Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Inflammation/pathology , Joints/pathology , Peroxidase/metabolism , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Disease Progression , Inflammation/immunology , Inflammation/metabolism , Joints/immunology , Joints/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/genetics , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology
3.
Blood ; 121(20): 4195-204, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23509155

ABSTRACT

Myeloperoxidase (MPO) is important in intracellular microbial killing by neutrophils but extracellularly causes tissue damage. Its role in adaptive immunity and T-cell-mediated diseases is poorly understood. Here, T-cell responses in lymph nodes (LNs) were enhanced by MPO deletion or in vivo inhibition, causing enhanced skin delayed-type hypersensitivity and antigen (Ag)-induced arthritis. Responses of adoptively transferred OT-II T cells were greater in MPO-deficient than wild-type (WT) recipients. MPO, deposited by neutrophils in LNs after Ag injection, interacted with dendritic cells (DCs) in vivo. Culture of murine or human DCs with purified MPO or neutrophil supernatant showed that enzymatically dependent MPO-mediated inhibition of DC activation occurs via MPO-generated reactive intermediates and involves DC Mac-1. Transfer of DCs cultured with WT, but not MPO-deficient, neutrophil supernatant attenuated Ag-specific immunity in vivo. MPO deficiency or in vivo inhibition increased DC activation in LNs after immunization. Studies with DQ-ovalbumin showed that MPO inhibits Ag uptake/processing by DCs. In vivo DC transfer and in vitro studies showed that MPO inhibits DC migration to LNs by reducing their expression of CCR7. Therefore, MPO, via its catalytic activity, inhibits the generation of adaptive immunity by suppressing DC activation, Ag uptake/processing, and migration to LNs to limit pathological tissue inflammation.


Subject(s)
Dendritic Cells/immunology , Inflammation/immunology , Neutrophils/enzymology , Peroxidase/physiology , T-Lymphocytes/physiology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Antigen Presentation/immunology , Cells, Cultured , Dendritic Cells/metabolism , Humans , Inflammation/etiology , Inflammation/genetics , Inflammation/metabolism , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Male , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/immunology , Metabolism, Inborn Errors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Peroxidase/genetics , Peroxidase/metabolism , T-Lymphocytes/metabolism
4.
J Am Soc Nephrol ; 23(12): 1955-66, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23138486

ABSTRACT

Mast cells contribute to the modulation of the immune response, but their role in autoimmune renal disease is not well understood. Here, we induced autoimmunity resulting in focal necrotizing GN by immunizing wild-type or mast cell-deficient (Kit(W-sh/W-sh)) mice with myeloperoxidase. Mast cell-deficient mice exhibited more antimyeloperoxidase CD4+ T cells, enhanced dermal delayed-type hypersensitivity responses to myeloperoxidase, and more severe focal necrotizing GN. Furthermore, the lymph nodes draining the sites of immunization had fewer Tregs and reduced production of IL-10 in mice lacking mast cells. Reconstituting these mice with mast cells significantly increased the numbers of Tregs in the lymph nodes and attenuated both autoimmunity and severity of disease. After immunization with myeloperoxidase, mast cells migrated from the skin to the lymph nodes to contact Tregs. In an ex vivo assay, mast cells enhanced Treg suppression through IL-10. Reconstitution of mast cell-deficient mice with IL-10-deficient mast cells led to enhanced autoimmunity to myeloperoxidase and greater disease severity compared with reconstitution with IL-10-intact mast cells. Taken together, these studies establish a role for mast cells in mediating peripheral tolerance to myeloperoxidase, protecting them from the development of focal necrotizing GN in ANCA-associated vasculitis.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/complications , Glomerulonephritis/immunology , Interleukin-10/metabolism , Mast Cells/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Cells, Cultured , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Humans , Immunomodulation , Lymph Nodes/pathology , Male , Mice , Mice, Inbred C57BL , Peripheral Tolerance , Peroxidase
5.
Am J Pathol ; 179(3): 1188-98, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21741931

ABSTRACT

T helper (Th)17 cells might contribute to immune-mediated renal injury. Thus, we sought to define the time course of IL-17A-induced kidney damage and examined the relation between Th17 and Th1 cells in a model of crescentic anti-glomerular basement membrane glomerulonephritis. Renal injury and immune responses were assessed in wild-type and in IL-17A-deficient mice on days 6, 14, and 21 of disease development. On day 6, when mild glomerulonephritis developed, IL-17A-deficient mice were protected from renal injury. On day 14, when more severe disease developed, protection from renal injury due to IL-17A deficiency was less evident. On day 21, when crescentic glomerulonephritis was fully established, disease was enhanced in IL-17A(-/-) mice, with increased glomerular T-cell accumulation and fibrin deposition, and augmented Th1 responses. Mice lacking the Th17-promoting cytokine, IL-23 (p19), also developed more severe disease than wild-type animals on day 21. In contrast, mice deficient in the key Th1-promoting cytokine, IL-12 (p35), had decreased Th1 and increased Th17 responses and developed less severe crescentic glomerulonephritis than wild-type animals. These studies show that IL-17A contributes to early glomerular injury, but it attenuates established crescentic glomerulonephritis by suppressing Th1 responses. They provide further evidence that Th1 cells mediate crescentic injury in this model and that Th1 and Th17 cells counterregulate each other during disease development.


Subject(s)
Acute Kidney Injury/immunology , Anti-Glomerular Basement Membrane Disease/immunology , Interleukin-17/deficiency , Th1 Cells/physiology , Animals , Cell Adhesion Molecules/metabolism , Cell Survival , Fibrin/metabolism , Immunity, Cellular/immunology , Immunoglobulins , Interferon-gamma/metabolism , Interleukin-12/deficiency , Interleukin-17/metabolism , Interleukin-23/deficiency , Interleukin-23/metabolism , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...