Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Kidney Int ; 93(1): 110-127, 2018 01.
Article in English | MEDLINE | ID: mdl-28754552

ABSTRACT

Ubiquitin C-terminal hydrolase L1 (UCH-L1) is a major deubiquitinating enzyme of the nervous system and associated with the development of neurodegenerative diseases. We have previously shown that UCH-L1 is found in tubular and parietal cells of the kidney and is expressed de novo in injured podocytes. Since the role of UCH-L1 in the kidney is unknown we generated mice with a constitutive UCH-L1-deficiency to determine its role in renal health and disease. UCH-L1-deficient mice developed proteinuria, without gross changes in glomerular morphology. Tubular cells, endothelial cells, and podocytes showed signs of stress with an accumulation of oxidative-modified and polyubiquitinated proteins. Mechanistically, abnormal protein accumulation resulted from an altered proteasome abundance leading to decreased proteasomal activity, a finding exaggerated after induction of anti-podocyte nephritis. UCH-L1-deficient mice exhibited an exacerbated course of disease with increased tubulointerstitial and glomerular damage, acute renal failure, and death, the latter most likely a result of general neurologic impairment. Thus, UCH-L1 is required for regulated protein degradation in the kidney by controlling proteasome abundance. Altered proteasome abundance renders renal cells, particularly podocytes and endothelial cells, susceptible to injury.


Subject(s)
Glomerulonephritis/enzymology , Immune Complex Diseases/enzymology , Podocytes/enzymology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Glomerulonephritis/genetics , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Hypotension/enzymology , Hypotension/genetics , Immune Complex Diseases/genetics , Immune Complex Diseases/immunology , Immune Complex Diseases/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Knockout , Oxidation-Reduction , Podocytes/immunology , Podocytes/pathology , Proteinuria/enzymology , Proteinuria/genetics , Proteolysis , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/genetics , Ubiquitination
2.
J Pharmacol Exp Ther ; 353(2): 288-98, 2015 May.
Article in English | MEDLINE | ID: mdl-25698787

ABSTRACT

Small vessel vasculitis is a life-threatening condition and patients typically present with renal and pulmonary injury. Disease pathogenesis is associated with neutrophil accumulation, activation, and oxidative damage, the latter being driven in large part by myeloperoxidase (MPO), which generates hypochlorous acid among other oxidants. MPO has been associated with vasculitis, disseminated vascular inflammation typically involving pulmonary and renal microvasculature and often resulting in critical consequences. MPO contributes to vascular injury by 1) catabolizing nitric oxide, impairing vasomotor function; 2) causing oxidative damage to lipoproteins and endothelial cells, leading to atherosclerosis; and 3) stimulating formation of neutrophil extracellular traps, resulting in vessel occlusion and thrombosis. Here we report a selective 2-thiouracil mechanism-based MPO inhibitor (PF-1355 [2-(6-(2,5-dimethoxyphenyl)-4-oxo-2-thioxo-3,4-dihydropyrimidin-1(2H)-yl)acetamide) and demonstrate that MPO is a critical mediator of vasculitis in mouse disease models. A pharmacokinetic/pharmacodynamic response model of PF-1355 exposure in relation with MPO activity was derived from mouse peritonitis. The contribution of MPO activity to vasculitis was then examined in an immune complex model of pulmonary disease. Oral administration of PF-1355 reduced plasma MPO activity, vascular edema, neutrophil recruitment, and elevated circulating cytokines. In a model of anti-glomerular basement membrane disease, formerly known as Goodpasture disease, albuminuria and chronic renal dysfunction were completely suppressed by PF-1355 treatment. This study shows that MPO activity is critical in driving immune complex vasculitis and provides confidence in testing the hypothesis that MPO inhibition will provide benefit in treating human vasculitic diseases.


Subject(s)
Acetamides/pharmacology , Enzyme Inhibitors/pharmacology , Glomerular Basement Membrane/drug effects , Glomerulonephritis/prevention & control , Immune Complex Diseases/prevention & control , Peroxidase/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrimidinones/pharmacology , Vasculitis/prevention & control , Animals , Glomerular Basement Membrane/pathology , Glomerulonephritis/enzymology , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Humans , Immune Complex Diseases/enzymology , Immune Complex Diseases/immunology , Immune Complex Diseases/pathology , Lung/blood supply , Lung/drug effects , Lung/immunology , Mice , Neutrophil Infiltration/drug effects , Signal Transduction/drug effects , Vasculitis/enzymology , Vasculitis/immunology , Vasculitis/pathology
3.
Nephrology (Carlton) ; 15(5): 560-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20649877

ABSTRACT

AIM: Renal expression of matrix metalloproteinases (MMP) and tissue inhibitors of MMP (TIMP) contribute to the development of tubulointerstitial fibrosis characteristic of progressive forms of primary glomerulonephritis (GN). The aim of this study was to investigate the therapeutic effect of MMP inhibitor, doxycycline, administration in an experimental rat model of immune-complex nephritis (ICN). METHODS: The induction of immune-complex glomerulonephritis was carried out by the administration of an i.v. dose of 2 mg bovine serum albumin (BSA) daily for 28 days after 8 weeks of s.c. immunization with 1 mg of BSA in complete Freund's adjuvant. Doxycycline (30 mg/kg) was given daily (in groups 2 and 4) by gavage for 28 days. RESULTS: Animals treated with doxycycline showed significant reduction in glomerular area and cell proliferation than non-treated controls. Glomerular deposition of immunoglobulin (Ig)G and C3 was less intense in treated rats than non-treated controls. Although not statistically significant, interstitial inflammation was less intense in treated rats than non-treated controls. Glomerular expression of MMP-9 by immunoflourescence was significantly inhibited in the treated group. In addition pro-MMP-2 on gelatin zymography was importantly suppressed by doxycycline in ICN. CONCLUSION: Doxycycline, in addition to its antibiotic property, may, following further investigation, provide a possible survival benefit in proliferative glomerulonephritis.


Subject(s)
Cell Proliferation/drug effects , Doxycycline/pharmacology , Glomerulonephritis/prevention & control , Immune Complex Diseases/prevention & control , Kidney Glomerulus/drug effects , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/pharmacology , Animals , Collagen Type IV/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Freund's Adjuvant , Glomerulonephritis/chemically induced , Glomerulonephritis/enzymology , Glomerulonephritis/pathology , Immune Complex Diseases/chemically induced , Immune Complex Diseases/enzymology , Immune Complex Diseases/pathology , Immunohistochemistry , Kidney Glomerulus/enzymology , Kidney Glomerulus/pathology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/metabolism , Rats , Rats, Wistar , Serum Albumin, Bovine , Time Factors , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism
4.
J Immunol ; 185(1): 624-33, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20530261

ABSTRACT

Mast cells exert protective effects in experimental antiglomerular basement membrane-induced glomerulonephritis (GN), yet the responsible mediators have not been identified. In this study, we investigated the role of mouse mast cell protease (mMCP)-4, the functional homolog of human chymase, using mMCP-4-deficient mice. Compared with wild type animals, mMCP-4-deficient mice exhibited lower proteinuria, blood creatinine, and blood urea nitrogen levels, indicating an aggravating role of mMCP-4. Kidney histology confirmed less severe renal damage in mMCP-4-deficient mice with reduced deposits, glomerular and interstitial cellularity, and fibrosis scores. High amounts of mMCP-4 were detected in renal capsules, but not in the whole kidney, from wild type mice. Its expression in renal capsules was markedly decreased after GN induction, suggesting that locally released enzyme by degranulated mast cells could contribute to the functional and physiopathological hallmarks of GN. Supporting a proinflammatory role, glomerular and interstitial macrophage and T cell infiltration, levels of proinflammatory TNF and MCP-1 mRNA, and the expression of the profibrotic peptide angiotensin II together with type I collagen were markedly downregulated in kidneys of mMCP-4-deficient mice. We conclude that mMCP-4 chymase, contrary to the global anti-inflammatory action of mast cells, aggravates GN by promoting kidney inflammation. These results highlight the complexity of mast cell-mediated inflammatory actions and suggest that chymase inhibition may represent a novel therapeutic target in GN.


Subject(s)
Anti-Glomerular Basement Membrane Disease/enzymology , Anti-Glomerular Basement Membrane Disease/pathology , Immune Complex Diseases/enzymology , Immune Complex Diseases/pathology , Inflammation Mediators/physiology , Serine Endopeptidases/physiology , Animals , Anti-Glomerular Basement Membrane Disease/immunology , Cells, Cultured , Fibrosis , Immune Complex Diseases/immunology , Kidney Function Tests , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
5.
Immunol Res ; 32(1-3): 211-5, 2005.
Article in English | MEDLINE | ID: mdl-16106072

ABSTRACT

Neutrophil-derived serine proteases play an important role in fighting infections. However, emerging data suggest that these proteases also regulate noninfectious inflammatory responses. Loss-of-function mutations in serine proteases have provided critical insights into the significance of these serine proteases in initiating and amplifying immune complex-mediated diseases.


Subject(s)
Immune Complex Diseases/enzymology , Immune Complex Diseases/immunology , Neutrophils/enzymology , Neutrophils/immunology , Serine Endopeptidases/immunology , Animals , Cell Movement , Chemotaxis, Leukocyte , Cytoplasmic Granules/enzymology , Humans , Infections/enzymology , Infections/immunology , Inflammation/enzymology , Inflammation/immunology , Mice
6.
Lab Invest ; 82(4): 505-13, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11950907

ABSTRACT

Dipeptidyl peptidase IV (DPPIV) is widely expressed in many tissues; however, its precise biological function is poorly understood. One of its possible physiologic roles is an involvement in the immune system, which plays a pivotal role in the pathogenesis of glomerulonephritis. The present study focused on the involvement of DPPIV in immune complex-mediated glomerulonephritis. Experimental nephritis was induced by anti-Thy-1.1 monoclonal antibody E30 using Wistar or F344 rats. The application of a new monoclonal antibody against DPPIV, F16, completely suppressed E30-induced proteinuria and mesangial proliferation in Wistar rats, whereas these preventive effects of F16 were not observed in F344 rats, which spontaneously lack DPPIV protein. Treatment with F16 inhibited glomerular deposition of complement C3 and complement C4 after the binding of E30 to the mesangial cell surface. Because the preventive effect of F16 was attributable to suppression of the complement cascade, we examined its influences on complement-dependent mesangial cell lysis in vitro. We discovered that the complement cascade was markedly inactivated in F16-treated Wistar rat serum but not in F16-treated F344 rats. These results indicate that DPPIV may play a somewhat crucial role in regulating the complement cascade and that inhibition of DPPIV may serve as a new target for preventing complement-dependent tissue injury.


Subject(s)
Dipeptidyl Peptidase 4/biosynthesis , Glomerulonephritis/enzymology , Immune Complex Diseases/enzymology , Animals , Antibodies, Blocking/pharmacology , Antigen-Antibody Complex/immunology , Binding Sites/drug effects , Cell Division/drug effects , Complement C3/metabolism , Complement C4/metabolism , Complement Membrane Attack Complex/metabolism , Cytotoxicity, Immunologic , Dipeptidyl Peptidase 4/immunology , Glomerular Mesangium/cytology , Glomerular Mesangium/immunology , Glomerulonephritis/immunology , Immune Complex Diseases/immunology , Immune Complex Diseases/prevention & control , Isoantibodies/administration & dosage , Male , Proteinuria/prevention & control , Rats , Rats, Inbred F344 , Rats, Wistar , Species Specificity
7.
Am J Pathol ; 158(6): 2139-44, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11395391

ABSTRACT

Matrix metalloproteases (MMPs) are a group of zinc-dependent endopeptidases that can degrade every component of the extracellular matrix. Under normal circumstances, the levels of MMPs are tightly regulated at both transcriptional and posttranscriptional levels. However, they are up-regulated in pathological states such as inflammation. Previous investigations have suggested that MMP-12 (metalloelastase) may be an important mediator in the pathogenesis of chronic lung injury. In this study we investigated the role of metalloelastase in the pathogenesis of acute lung injury using mice containing a targeted disruption of the metalloelastase gene. Neutrophil influx into the alveolar space in metalloelastase-deficient animals was reduced to approximately 50% of that observed in parent strain mice following the induction of injury by immune complexes. In addition, lung permeability in metalloelastase-deficient mice was approximately 50% of that of injured parent strain animals with normal levels of metalloelastase and this was correlated with histological evidence of less lung injury in the metalloelastase-deficient animals. Collectively, the data suggest that metalloelastase is necessary for the full development of acute alveolitis in this model of lung injury. Further, the data suggest that reduced injury in metalloelastase-deficient mice is due in part to decreased neutrophil influx into the alveolar space.


Subject(s)
Immune Complex Diseases/enzymology , Lung Diseases/enzymology , Lung Diseases/immunology , Metalloendopeptidases/physiology , Acute Disease , Animals , Bronchoalveolar Lavage Fluid/cytology , Leukocyte Count , Lung/pathology , Lung Diseases/pathology , Matrix Metalloproteinase 12 , Matrix Metalloproteinase 2/physiology , Matrix Metalloproteinase 9/physiology , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Neutrophil Infiltration
8.
J Immunol ; 163(10): 5633-9, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10553093

ABSTRACT

The involvement of immune complexes during experimental arthritis in induction of metalloproteinases (MMP)-induced neoepitopes in aggrecan in cartilage, as well as the role of stromelysin-1 (SLN-1) in the induction of this neoepitope, was investigated. Passive immune complex arthritis was induced, and generation of the MMP-specific cleavage product (VDIPEN) was studied by immunolocalization. The role of SLN-1 was studied with use of SLN-1-deficient (SLN-1KO) mice. VDIPEN expression was studied in vitro by exposing the cartilage to IL-1 and subsequent activation of latent MMPs. Immune complex arthritis was characterized by an acute inflammation, with influx of mainly polymorphonuclear cells into the joint cavity. Expression of VDIPEN neoepitopes was consistently found in areas extensively depleted from proteoglycans. SLN-1KO mice did not show expression of the VDIPEN neoepitope, although inflammation and proteoglycan depletion was comparable to wild-type mice. In addition, erosions of cartilage were absent in SLN-1KO mice, but were present in wild-type mice, suggesting an important role for SLN-1 in cartilage destruction. In vitro studies showed that SLN-1 is also pivotally involved in IL-1-induced MMP activity. Stimulated polymorphonuclear neutrophils were able to activate latent MMPs present in the cartilage. Neutrophil elastase was also capable of activating IL-1-induced latent MMPs, which identifies elastase as a possible activator for latent VDIPEN-inducing MMPs. This study suggests that IC are important in the activation of latent MMPs in cartilage, possibly through polymorphonuclear neutrophil activation on the cartilage edge. SLN-1 is a pivotal enzyme in overall MMP-activity in cartilage during immune complex-mediated arthritis.


Subject(s)
Arthritis/enzymology , Arthritis/immunology , Cartilage, Articular/enzymology , Immune Complex Diseases/enzymology , Matrix Metalloproteinase 3/physiology , Animals , Arthritis/genetics , Arthritis/pathology , Cartilage, Articular/pathology , Enzyme Activation/genetics , Enzyme Activation/immunology , Epitopes/biosynthesis , Immune Complex Diseases/genetics , Immune Complex Diseases/immunology , Immune Complex Diseases/pathology , Interleukin-1/pharmacology , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/metabolism , Oligopeptides/biosynthesis , Peptide Fragments/biosynthesis
9.
Biochem Biophys Res Commun ; 247(1): 84-7, 1998 Jun 09.
Article in English | MEDLINE | ID: mdl-9636659

ABSTRACT

Previous studies have shown high arginase activity at inflammatory sites. Arginase converts L-arginine to L-ornithine, sharing a common substrate with nitric oxide synthase. It exists as two isoforms, AI and AII. While the function of liver arginase (AI) in ureagenesis has been defined, the role and isoform of arginase in cells without a complete urea cycle are unknown. We therefore determined arginase isoform mRNA expression in glomerular acute immune complex inflammation, and its cultured constituent cells. AI was induced in nephritic glomeruli, and in mesangial cells stimulated with IL-4 and cAMP, and was present in elicited neutrophils and macrophages. AII was constitutively expressed. Our data strongly suggest that AI, thought to be restricted to the liver, accounts for high arginase activity at inflammatory sites where it may limit high output nitric oxide production and generate polyamines and proline essential for cell proliferation and matrix production. This identification of AI in inflamed tissue is an important step for understanding the consequences of increased arginase activity.


Subject(s)
Arginase/biosynthesis , Immune Complex Diseases/enzymology , Immune Complex Diseases/pathology , Up-Regulation/immunology , Acute Disease , Animals , Glomerular Mesangium/enzymology , Glomerulonephritis/enzymology , Glomerulonephritis/pathology , Inflammation/enzymology , Inflammation/immunology , Isoenzymes/biosynthesis , Kidney Glomerulus/enzymology , Kidney Glomerulus/pathology , Macrophages/enzymology , Male , Neutrophils/enzymology , Rats , Rats, Inbred Lew
10.
Pathol Biol (Paris) ; 46(10): 779-86, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9922994

ABSTRACT

Kidney ectopeptidases play an important role in the metabolism of different peptides. They activate precursor proteins or inactivate peptides including hormones, cytokines, vasoactive peptides (angiotensin II, endothelin), neuroendocrine hormones, changing local concentration in active peptides. Kidney ectopeptidase regulate cell proliferation, adhesion, matrix synthesis, cell signaling, cell activation, differentiation and cell-cell communication. The role of four major ectopeptidases (aminopeptidase A and N, dipeptidylpeptidase IV, and neutral endopeptidase) is presented.


Subject(s)
Endopeptidases/physiology , Kidney/enzymology , Aminopeptidases/physiology , Animals , CD13 Antigens/physiology , Cytokines/physiology , Dipeptidyl Peptidase 4/physiology , Disease Models, Animal , Endopeptidases/chemistry , Enzyme Induction , Glomerulonephritis/enzymology , Glomerulonephritis/immunology , Glutamyl Aminopeptidase , Hormones/metabolism , Humans , Immune Complex Diseases/enzymology , Immune Complex Diseases/immunology , Kidney Diseases/enzymology , Lupus Nephritis/enzymology , Mice , Neprilysin/physiology , Peptides/metabolism , Protease Inhibitors/pharmacology , Protein Precursors/metabolism , Rats , Rats, Inbred Lew
11.
Curr Biol ; 7(11): 877-80, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9382799

ABSTRACT

Neutrophils play an important part in the development of acute inflammatory injury. Human neutrophils contain high levels of the serine protease elastase, which is stored in azurophilic granules and is secreted in response to inflammatory stimuli. Elastase is capable of degrading many components of extracellular matrix [1-4] and has cytotoxic effects on endothelial cells [5-7] and airway epithelial cells. Three types of endogenous protease inhibitors control the activity of neutrophil elastase, including alpha-1 protease inhibitor (alpha-1PI), alpha-2 macroglobulin and secreted leukoproteinase inhibitor (SLPI) [8-10]. A disturbed balance between neutrophil elastase and these inhibitors has been found in various acute clinical conditions (such as adult respiratory syndrome and ischemia-reperfusion injury) and in chronic diseases. We investigated the effect of NX21909, a selected oligonucleotide (aptamer) inhibitor of elastase, in an animal model of acute lung inflammatory disease [11-14]. This inhibitor was previously selected from a hybrid library of randomized DNA and a small-molecule irreversible inhibitor of elastase (a valine diphenyl ester phosphonate, Fig. 1), by the blended SELEX process [15]. We show that NX21909 inhibits lung injury and neutrophil influx in a dose-dependent manner, the first demonstration of efficacy by an aptamer in an animal disease model.


Subject(s)
Leukocyte Elastase/antagonists & inhibitors , Lung/enzymology , Lung/pathology , Neutrophils/enzymology , Oligodeoxyribonucleotides/therapeutic use , Oligonucleotides/therapeutic use , Serine Proteinase Inhibitors/therapeutic use , Valine/analogs & derivatives , Animals , Capillary Permeability/immunology , Cell Movement/drug effects , Cell Movement/immunology , Immune Complex Diseases/drug therapy , Immune Complex Diseases/enzymology , Immune Complex Diseases/physiopathology , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/physiopathology , Lung/physiopathology , Male , Neutrophils/immunology , Oligonucleotides/chemistry , Rats , Rats, Inbred Strains , Serine Proteinase Inhibitors/chemistry , Valine/chemistry , Valine/therapeutic use
12.
Am J Respir Crit Care Med ; 150(6 Pt 2): S114-7, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7952645

ABSTRACT

Passively released or actively secreted elastase from neutrophils has been linked to the pathologic processes of a variety of inflammatory diseases, including idiopathic pulmonary fibrosis, rheumatoid arthritis, adult respiratory distress syndrome, and cystic fibrosis. The serine proteinase has a broad substrate specificity and may attack a number of host proteins outside of the neutrophil, including lung elastin and fibronectin. Such a proteolysis may change the normal surrounding tissue and the protein pattern of an inflammatory focus. Additionally, it acts as a potent secretagogue in minute amounts. The reason that neutrophil elastase is present in considerable concentrations outside of the neutrophil during chronic inflammation and that the major endogenous serine proteinase inhibitor for neutrophil elastase, alpha 1-proteinase inhibitor, is easily inactivated by proteolytic and oxidative attack is unclear. Released neutrophil elastase may also be involved in regulating chronic inflammation. In a feedback mechanism, neutrophil elastase inhibits neutrophil stimulation and concomitant elastase release by cleavage of immunoglobulins, complement components, and complement receptor type 1 on neutrophils. Besides a number of harmful effects of neutrophil elastase in inflammation, the latter mechanism, although considerably impairing phagocytosis, may be beneficial particularly in the light of persistent bacterial pathogens in the human lung affected by cystic fibrosis.


Subject(s)
Inflammation/enzymology , Lung Diseases/enzymology , Pancreatic Elastase/physiology , Animals , Chronic Disease , Cystic Fibrosis/enzymology , Exocytosis , Humans , Immune Complex Diseases/enzymology , Leukocyte Elastase , Phagocytosis
13.
Br J Pharmacol ; 107(4): 1159-62, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1281719

ABSTRACT

1. The ability of analogues of L-arginine (N-iminoethyl-L-ornithine (L-NIO), NG-monomethyl-L-arginine (L-NMMA), NG-nitro-L-arginine methyl ester (L-NAME) and NG-nitro-L-arginine (L-NNA)) to protect against inflammatory injury induced by activated neutrophils was investigated in rats following intradermal or intrapulmonary deposition of immune complexes. 2. The descending order of potency for protective effects of these analogues was: L-NIO > L-NMMA > L-NNA = L-NAME. The approximate IC50 value for L-NIO in the dermal vasculitis model was 65 microM. For all other compounds, the IC50 values were > 5 mM. 3. The protective effect of L-NIO in the skin was reversed in a dose-dependent manner by the presence of L-arginine, but not by D-arginine. L-Arginine also reversed the protective effects of L-NIO in immune complex-induced lung injury. 4. The protective effects of L-NIO were not associated with reductions in neutrophil accumulation, as measured by extraction from tissues of myeloperoxidase. 5. These data demonstrate that L-NIO has the most potent protective effects against immune complex-induced vascular injury induced by activated macrophages. Furthermore, they indicate that this injury is dependent upon the generation of nitric oxide.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Immune Complex Diseases/prevention & control , Vasculitis, Leukocytoclastic, Cutaneous/prevention & control , Alveolitis, Extrinsic Allergic/enzymology , Alveolitis, Extrinsic Allergic/prevention & control , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Immune Complex Diseases/enzymology , Male , NG-Nitroarginine Methyl Ester , Nitric Oxide Synthase , Nitroarginine , Ornithine/analogs & derivatives , Ornithine/pharmacology , Peroxidase/metabolism , Rats , Vasculitis, Leukocytoclastic, Cutaneous/enzymology , omega-N-Methylarginine
14.
Inflammation ; 16(3): 259-71, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1386836

ABSTRACT

Activated leukocytes in the peritoneal cavity have been shown to release phospholipase A2(PLA2) activity and increased production of eicosanoids. It is not known whether a similar phenomenon occurs in inflammatory bowel diseases. We measured PLA2 activity in the peritoneal leukocytes and eicosanoid content in the peritoneal cavity of rats with experimental colitis induced by dinitrochlorobenzene, immune complex-mediated colitis, and acetic acid-induced colitis. In all models of colitis, proteins in the peritoneal cavity were increased by 60-260% above control, leukocyte number was elevated by 3- to 10-fold, the PLA2 activity by 4- to 32-fold, and anti-PLA2 activity was absent. Eicosanoid concentrations were 3- to 4-fold higher than controls. In vitro studies confirmed that the peritoneal leukocytes were the source of PLA2. Treatment of rats with dexamethasone and leukocytes in vitro reduced PLA2 significantly, but they were well above control rats without colitis. However, dexamethasone did not improve the histology. These studies suggest that peritoneal leukocytes may serve as an additional source of inflammatory mediators, which may further enhance the inflammatory response in the rat colon.


Subject(s)
Colitis/enzymology , Leukocytes/enzymology , Phospholipases A/analysis , Acetates/toxicity , Acetic Acid , Animals , Arthus Reaction , Colitis/chemically induced , Colitis/immunology , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Dinitrochlorobenzene , Disease Models, Animal , Eicosanoids/biosynthesis , Hypersensitivity, Delayed/complications , Immune Complex Diseases/enzymology , Inflammatory Bowel Diseases , Leukocyte Count , Male , Peritoneal Cavity/cytology , Phospholipases A2 , Rats , Rats, Inbred Strains
15.
J Comp Pathol ; 104(1): 65-76, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1826913

ABSTRACT

Biochemical changes in plasma and urine were monitored in six cats before and during the induction of immune complex-mediated glomerulonephritis (ICGN) by daily intravenous administration of human serum albumin (HSA). The earliest indication of renal dysfunction in the cats was hypoalbuminaemia, which occurred as early as 13 weeks before cats developed clinical signs of renal disease. Proteinuria occurred 2 to 3 weeks before clinical disease, but was sensitive in predicting renal pathology in two cats that did not develop clinical signs of disease. In addition, increased activities of several urinary enzymes were detected in affected cats, with measurement of N-acetyl-beta-D-glucosaminidase and gamma-glutamyl transferase providing the earliest and most sensitive indication of renal damage. These plasma and urine measurements correlated more closely with the renal pathology, observed at postmortem, than clinical assessment of disease. It was concluded that ICGN in the cat could be diagnosed earliest by measurement of plasma protein concentration, whilst disease progress could be effectively monitored by including assays to measure urine protein and urine enzymes.


Subject(s)
Cat Diseases/blood , Glomerulonephritis/veterinary , Immune Complex Diseases/veterinary , Animals , Cat Diseases/enzymology , Cat Diseases/urine , Cats , Glomerulonephritis/blood , Glomerulonephritis/enzymology , Glomerulonephritis/urine , Immune Complex Diseases/blood , Immune Complex Diseases/enzymology , Immune Complex Diseases/urine , Proteinuria/veterinary
16.
Clin Exp Immunol ; 64(3): 597-605, 1986 Jun.
Article in English | MEDLINE | ID: mdl-2947759

ABSTRACT

In sputa of 21 cystic fibrosis patients suffering from chronic Pseudomonas aeruginosa lung infections, activities of lysosomal proteases released from polymorphonuclear leukocytes (PMN) and immune complexes were quantitatively determined. The results showed that low immune complex values correlated with high protease activities and vice versa. In a longitudinal study fluctuating, reciprocal changes of the parameters were observed. Therefore, the hypothesis was investigated as to whether PMN elastase cleaves immune complexes and regulates inflammation. It was shown that treatment of immune complex-positive sputa with PMN elastase decreased IgG and IgA immune complex values and lead to cleavage of the immunoglobulins in the hinge region. PMN elastase-treated samples were not able to stimulate the oxidative burst of PMN. Cleavage of in vitro built immune complexes did not lead to liberation of bound antigen. Western blot analysis of one sputum sample revealed split products of the IgG heavy chain. The results suggest that PMN elastase splits off the Fc portion of immunoglobulins in immune complexes and thus regulates inflammation by a feedback mechanism leading to cyclic inflammatory states.


Subject(s)
Immune Complex Diseases/enzymology , Neutrophils/enzymology , Pancreatic Elastase/immunology , Antigen-Antibody Complex/immunology , Cystic Fibrosis/enzymology , Cystic Fibrosis/immunology , Humans , Pancreatic Elastase/analysis
18.
Clin Chim Acta ; 82(1-2): 205-7, 1978 Jan 02.
Article in English | MEDLINE | ID: mdl-201399

ABSTRACT

By means of a biological assay the collagenolytic activity of kidneys from rats with experimental immune complex glomerulonephritis and control animals was tested. We detected collagenolysis in both panels but found quantitative differences in the collagenase activity. The enzymatic collagenolysis was significantly increased (P less than 0.001) in the nephritic kidneys. From the fact that the chelating agent EDTA inhibited potentially the released enzyme it can be concluded that the origin is the polymorphonuclear granules.


Subject(s)
Glomerulonephritis/enzymology , Immune Complex Diseases/enzymology , Kidney/enzymology , Microbial Collagenase/metabolism , Animals , Edetic Acid/pharmacology , Glomerulonephritis/immunology , Rats , Serum Albumin, Bovine
19.
Br J Dermatol ; 96(4): 349-55, 1977 Apr.
Article in English | MEDLINE | ID: mdl-140699

ABSTRACT

The lysosomal enzyme beta glucuronidase was selectively released from leukocytes when these were incubated with insoluble immnue complexes and also from the leukocytes of patients with immune complex disease when these were incubated with their own plasma.


Subject(s)
Antigen-Antibody Complex , Glucuronidase/metabolism , Leukocytes/enzymology , Humans , Immune Complex Diseases/enzymology , Leukocytes/immunology , Skin Diseases/enzymology , Skin Diseases/immunology
20.
Article in English | MEDLINE | ID: mdl-143103

ABSTRACT

In characterizing the immunochemical and biochemical nature of middle ear effusions (MEE) of chronic serous otitis media (SOM), we have previously shown that MEE contain all major immunoglobulin classes, increased levels of several lysosomal enzymes, decreased total complement, but increased levels of C3. This report extends these observations by showing that MEE also contains C3 proactivator (C3PA) that can be activated by those organisms involved in acute otitis media, which confirms a functional alternate complement pathway. Furthermore, we have demonstrated the presence of soluble immune complexes in MEE using the Raji cell test. These data further support an immune complex mediated etiopathogenesis for chronic SOM.


Subject(s)
Immune Complex Diseases , Otitis Media/immunology , Adolescent , Adult , Antigen-Antibody Complex , Child , Child, Preschool , Chronic Disease , Complement C3/metabolism , Enzyme Activation , Exudates and Transudates/enzymology , Exudates and Transudates/immunology , Humans , Immune Complex Diseases/enzymology , Infant , Otitis Media/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL