Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Exp Med ; 168(3): 1169-74, 1988 Sep 01.
Article in English | MEDLINE | ID: mdl-3049904

ABSTRACT

We infused microgram quantities of active or inactive PMN elastase and cathepsin G into the renal arteries of rats. Both active and inactive elastase localized to the glomerular capillary wall equally, and in amounts that could be achieved physiologically in GN. However, elastase-perfused rats developed marked proteinuria (196 +/- 32 mg/24 h) compared with control rats receiving inactive elastase (19 +/- 2 mg/24 h, p less than 0.005). Similar results were seen with active and inactive cathepsin G. Neither elastase nor cathepsin G infusion was associated with histologic evidence of glomerular injury. We conclude that the PMN neutral serine proteinases elastase and cathepsin G can mediate marked changes in glomerular permeability in vivo due to their proteolytic activity, and thus, may contribute to the proteinuria observed in PMN-dependent models of GN.


Subject(s)
Cathepsins/physiology , Kidney Diseases/enzymology , Neutrophils/enzymology , Pancreatic Elastase/physiology , Animals , Cathepsin G , Fluorescent Antibody Technique , Kidney Diseases/pathology , Kidney Glomerulus/pathology , Proteinuria/etiology , Rats , Serine Endopeptidases
2.
J Clin Invest ; 66(1): 71-81, 1980 Jul.
Article in English | MEDLINE | ID: mdl-7400310

ABSTRACT

Quantitation of immune deposit formation in glomeruli and correlation with immunohistologic and functional changes has been accomplished only in models of anti-glomerular basement membrane antibody-induced nephritis, or indirectly in immune complex disease by measuring radiolabeled antigen deposition. The kinetics of subepithelial immune deposit formation and the relationship between the quantity of antibody deposited and proteinuria are defined here for the first time in an established model of membranous immune complex nephritis (passive Heymann nephritis) induced by a single intravenous injection of (125)I-labeled sheep immunoglobulin (Ig)G antibody to rat tubular brush border antigen (Fx1A). Measurement of antibody deposition in glomeruli (GAb) isolated from rats injected with 10 mg of anti-Fx1A demonstrated a mean of 12 mug GAb in 4 h, which increased linearly to 48 mug in 5 d. GAb represented only 20 and 44% of total kidney antibody binding at these times. Proteinuria occurred only after 4-5 d of antibody deposition in rats with total kidney antibody binding exceeding approximately 200 mug/2 kidneys. Steroid treatment and vasoactive amine blockade did not significantly alter the quantity or localization of immune deposits. It was also demonstrated that isolated rat glomeruli specifically bound nephritogenic quantities of anti-Fx1A in vitro within hours. Analysis of the quantitative aspects of glomerular antibody deposition in vivo and glomerular antibody binding in vitro provides additional evidence that subepithelial immune deposits in passive Heymann nephritis may form in situ by reaction of free antibody with antigenic constitutents of the normal rat glomerulus. The observed kinetics of deposit formation differ markedly from those in anti-glomerular basement membrane disease and suggest a role for factors in addition to antigen-antibody interaction in determining this unique pattern of glomerular immune deposit formation.


Subject(s)
Antigen-Antibody Complex , Glomerulonephritis/immunology , Kidney Glomerulus/immunology , Adrenal Cortex Hormones/pharmacology , Animals , Chlorpheniramine/pharmacology , Cyproheptadine/pharmacology , Immunoglobulin G/immunology , Kinetics , Male , Proteinuria/immunology , Rats , Sheep
3.
J Clin Invest ; 66(6): 1339-50, 1980 Dec.
Article in English | MEDLINE | ID: mdl-7440718

ABSTRACT

The only established role for complement in mediating immunologic renal disease involves elaboration of leukochemotactic factors and neutrophil-dependent glomerular injury. In the passive Heymann nephritis (PHN) model of experimental membranous nephropathy, rats injected with sheep antibody to rat proximal tubular brush border antigen (Fx1A) form subepithelial deposits of sheep IgG and rat complement (C3), and develop heavy proteinuria after 5 d without glomerular inflammatory changes. To study the role of complement in mediating proteinuria in PHN, 16 rats were treated daily with cobra venom factor from before antibody injection to maintain C3 levels at < 10% of pretreatment values and compared to 16 untreated controls. Proteinuria at 5 d was abolished in C3-depleted rats (4 +/- 1, controls 70 +/- 15 mg/d, P < 0.001), although renal deposition of 125I-labeled antibody ws the same in both groups (188 +/- 35 vs. 191 +/- 22 microgram IgG/2 kidneys, P > 0.5). Nephritogenic doses of both the noncomplement-fixing F(ab')2 portion and the gamma 2 subclass of anti-Fx1A IgG produced subepithelial deposits of immunoglobulin without C3, but proteinuria did not occur despite glomerular deposition of up to 70 microgram/2 kidneys of gamma 2. However, glomerular deposition of as little as 60 microgram of gamma 1 produced C3 fixation in vivo and heavy proteinuria. No neutrophil exudate could be detected histologically in PHN from the time of antibody injection through development of proteinuria. Proteinuria in five PHN rats depleted of neutrophils to < 200/mm3 with antineutrophil serum was not reduced compared to six controls with normal neutrophil counts (34 +/- 9.6 vs. 25 +/- 10.4 mg/d, P > 0.5). These results demonstrate that proteinuria in the PHN model of membranous nephropathy is complement-dependent and strongly suggest a neutrophil-independent mechanism. Thus a new role for the complement system in mediating immunologic glomerular injury is identified.


Subject(s)
Complement System Proteins/physiology , Nephritis/immunology , Neutrophils/immunology , Animals , Antigen-Antibody Complex , Complement C3/metabolism , Disease Models, Animal , Glomerulonephritis/immunology , Immunosuppression Therapy , Proteinuria/immunology , Rats
4.
J Clin Invest ; 77(3): 762-7, 1986 Mar.
Article in English | MEDLINE | ID: mdl-3005365

ABSTRACT

To explore possible mechanisms by which complement membrane attack complexes (MAC) that are deposited in the glomerular mesangium might be pathogenic, we stimulated rat glomerular mesangial cells grown in vitro with nascent MACs formed from the purified human complement components C5b6 and normal human serum and measured production of superoxide ion (O2-) and hydrogen peroxide (H2O2). Mesangial cells incubated with C5b6 + serum, which results in cell membrane interaction with the MAC, produce 0.9 +/- 0.15 nmol O2-/10(5) cells per 30 min, which was significantly greater than the amount produced by cells incubated with C5b6 alone, serum alone, or decayed MACs that can no longer interact with the cell membrane (0.3 +/- 0.2, 0.4 +/- 0.1, 0.3 +/- 0.2 nmol O2-/10(5) cells per 30 min, respectively; P less than 0.02). Production of O2- after stimulation with MACs increased during the first 20 min of incubation but then plateaued. Cells exposed to decayed MACs produced small amounts of O2-, which did not increase from 20 to 60 min. Production of H2O2 was also observed after stimulation with MACs, and continued to increase during 60 min of incubation (1.22 +/- 0.16 nmol H2O2/10(5) cells per 60 min), whereas H2O2 production could not be detected after exposure to decayed MACs. Cell viability was not adversely affected by exposure to nascent MACs as determined by trypan blue exclusion or chromium-51 release. These results demonstrate that glomerular mesangial cell membrane interaction with the MAC stimulates the production of the toxic oxygen metabolites O- and H2O2. Activation of the terminal complement pathway by mesangial immune deposits in vivo might lead to tissue injury by stimulation of local production of toxic oxygen-free radicals.


Subject(s)
Complement System Proteins/metabolism , Kidney Glomerulus/metabolism , Oxygen/metabolism , Animals , Cell Survival , Cells, Cultured , Complement Membrane Attack Complex , Free Radicals , Hydrogen Peroxide/metabolism , Male , Rats , Superoxides/metabolism
5.
J Clin Invest ; 82(4): 1225-35, 1988 Oct.
Article in English | MEDLINE | ID: mdl-2971672

ABSTRACT

Neutrophils and platelets are frequently present in glomeruli in immune glomerulonephritis (GN). No role for the platelet in acute neutrophil-mediated renal injury has been defined. We investigated a neutrophil-mediated model of subendothelial immune complex GN in the rat. Rats were platelet-depleted (mean platelet less than 10,000/microliter) with goat anti-platelet IgG before induction of GN by the renal artery perfusion of concanavalin A followed by anti-concanavalin A IgG. Platelet-depletion resulted in a significant reduction in albuminuria (7 +/- 2 vs. 55 +/- 10 mg/24 h) and fractional albumin excretion (0.045 +/- 0.01 vs. 0.410 +/- 0.09) compared with controls. The decrease in albuminuria was not due to differences in blood or glomerular neutrophil counts, complement, renal function, or glomerular antibody binding. Platelet-depleted rats had equivalent subendothelial deposits and glomerular endothelial cell injury but had minimal platelet infiltrates and fibrin deposition compared with controls. These studies demonstrate a role for platelets in mediating acute neutrophil-induced glomerular injury and proteinuria in this model of GN.


Subject(s)
Blood Platelets/pathology , Glomerulonephritis/etiology , Immune Complex Diseases/etiology , Neutrophils/pathology , Animals , Binding Sites, Antibody , Blood Platelets/immunology , Complement System Proteins/metabolism , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Immune Complex Diseases/pathology , Immune Complex Diseases/physiopathology , Immune Sera , Kidney Function Tests , Kidney Glomerulus/immunology , Kidney Glomerulus/ultrastructure , Male , Neutrophils/immunology , Proteinuria/etiology , Rats , Rats, Inbred Strains , Thrombocytopenia/etiology , Thrombocytopenia/immunology , Thrombocytopenia/pathology
6.
J Clin Invest ; 79(5): 1379-87, 1987 May.
Article in English | MEDLINE | ID: mdl-3033023

ABSTRACT

Reactive oxygen species, particularly hydrogen peroxide (H2O2), participate in neutrophil-mediated glomerulonephritis. However, the mechanism of H2O2 neptrotoxicity is unknown. Myeloperoxidase (MPO), a neutrophil cationic enzyme that localizes in glomeruli, can react with H2O2 and halides to form highly reactive products. We tested the hypothesis that the MPO-H2O2-halide system may induce glomerular injury by infusing MPO followed by H2O2 in a chloride-containing solution into the renal artery of rats. Controls received MPO or H2O2 alone. MPO-H2O2-perfused rats developed significant proteinuria, endothelial cell swelling, and epithelial cell foot process effacement, whereas control kidneys were normal. In the presence of free 125I, MPO-H2O2-perfused rats incorporated large amounts of 125I, localized to the glomerular basement membrane and mesangium by autoradiography, into glomeruli. Glomerular iodination was greatly decreased or absent in controls. The MPO-H2O2-halide system causes glomerular injury and may be important in neutrophil-mediated glomerulonephritis.


Subject(s)
Hydrogen Peroxide/toxicity , Kidney Glomerulus/drug effects , Peroxidase/metabolism , Animals , Autoradiography , Microscopy, Electron , Proteinuria/metabolism , Rats , Rats, Inbred Strains
7.
J Clin Invest ; 96(6): 2809-19, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8675651

ABSTRACT

Podocyte injury is believed to contribute to glomerulosclerosis in membranous nephropathy. To identify the factors involved, we investigated the effects of basic fibroblast growth factor (bFGF), a cytokine produced by podocytes, on rats with membranous nephropathy (passive Heymann nephritis [PHN]). All rats received a daily i.v. bolus of 10 microg bFGF or vehicle from days 3-8 after PHN induction. In proteinuric PHN rats on day 8, bFGF injections further increased proteinuria. Podocytes of bFGF-injected PHN rats showed dramatic increases in mitoses, pseudocyst formation, foot process retraction, focal detachment from the glomerular basement membrane, and desmin expression. bFGF injections in PHN rats did not alter antibody or complement deposition or glomerular leukocyte influx. bFGF-injected PHN rats developed increased glomerulosclerosis when compared with control PHN rats. Also, bFGF induced proteinuria and podocyte damage in rats injected with 10% of the regular PHN-serum dose. None of these changes occurred in bFGF-injected normal rats, complement-depleted PHN rats or rats injected with 5% of the regular PHN serum dose. These divergent bFGF effects were explained in part by upregulated glomerular bFGF receptor expression, induced by PHN serum. Thus, bFGF can augment podocyte damage, resulting in increased glomerular protein permeability and accelerated glomerulosclerosis. This bFGF action is confined to previously injured podocytes. Release of bFGF from glomerular sources (including podocytes themselves) during injury may represent an important mechanism by which podocyte damage is enhanced or becomes self sustained.


Subject(s)
Fibroblast Growth Factor 2/toxicity , Glomerulonephritis, Membranous/pathology , Glomerulonephritis/chemically induced , Kidney Glomerulus/pathology , Animals , Apoptosis , Desmin/biosynthesis , Epithelium/drug effects , Epithelium/pathology , Epithelium/ultrastructure , Extracellular Matrix Proteins/drug effects , Extracellular Matrix Proteins/metabolism , Glomerulonephritis/pathology , Kidney Glomerulus/drug effects , Kidney Glomerulus/ultrastructure , Male , Microscopy, Electron , Mitosis/drug effects , Proteinuria/chemically induced , Rats , Rats, Sprague-Dawley , Reference Values , Time Factors
8.
J Clin Invest ; 72(6): 1948-57, 1983 Dec.
Article in English | MEDLINE | ID: mdl-6227634

ABSTRACT

Our recent observations of a complement-mediated, cell-independent mechanism of altered glomerular permeability in rat membranous nephropathy suggested a possible role for the terminal complement pathway in the mediation of proteinuria in certain forms of glomerular disease. To directly determine whether the membranolytic terminal complement components (C5b-C9) are involved in glomerular injury, we studied the development of proteinuria in normal and C6-deficient (C6D) rabbits, in both of which a membranous nephropathy-like lesion develops early in the course of immunization with cationized bovine serum albumin (cBSA) (pI 8.9-9.2). C6 hemolytic activity of C6D was 0.01% that of control rabbits. After 1 wk of daily intravenous injections of cBSA, proteinuria developed in 71% of controls (median 154, range 1-3,010 mg/24 h, n = 24), whereas none of C6D were proteinuric (median 6, range 2-12 mg/24 h, n = 12, P less than 0.01). After 1 wk of cBSA, both groups had qualitatively identical glomerular deposits of BSA, rabbit IgG, and C3 on immunofluorescence microscopy, predominantly subepithelial electron-dense deposits on electron microscopy, and minimal glomerular inflammatory cell infiltration of glomeruli. Glomeruli were isolated from individual animals after 1 wk of cBSA and deposits of rabbit IgG antibody were quantitated by a standardized in vitro assay using anti-rabbit IgG-125I. Rabbit IgG deposits were found to be similar in control (29.8 +/- 13.2, range 12.7-48.6 micrograms anti-IgG/2,000 glomeruli, n = 6) and C6D rabbits (32.6 +/- 13.8, range 16.8-48.8 micrograms anti-IgG/2,000 glomeruli, n = 5, P greater than 0.05). After 2 wk, coincident with a prominent influx of mononuclear cells and neutrophils, proteinuria developed in C6D rabbits. These results document, for the first time, a requirement for a terminal complement component in the development of immunologic glomerular injury. Since the only known action of C6 is in the assembly of the membrane attack complex, these observations suggest that the membranolytic properties of complement may contribute to glomerular damage.


Subject(s)
Complement C6/immunology , Glomerulonephritis/immunology , Immune Complex Diseases/immunology , Animals , Binding Sites, Antibody , Complement C5/physiology , Complement C5b , Complement C6/deficiency , Complement C9/physiology , Complement Pathway, Alternative , Glomerulonephritis/pathology , Immune Complex Diseases/pathology , Immunoglobulin G/immunology , Kidney Glomerulus/immunology , Kidney Glomerulus/ultrastructure , Microscopy, Electron , Proteinuria/immunology , Rabbits , Serum Albumin, Bovine/immunology
9.
J Clin Invest ; 100(4): 786-94, 1997 Aug 15.
Article in English | MEDLINE | ID: mdl-9259576

ABSTRACT

We investigated the origin of the glomerular mesangial cell, a smooth muscle-like cell that provides structural support in the glomerulus. Injection of anti-Thy 1 antibody that binds the Thy 1 antigen on rat mesangial cells eliminated (> 95%) the mesangial population at 20-28 h, while Thy 1-positive cells in the juxtaglomerular apparatus (JGA) were sequestered from the circulation and survived. Single pulse labeling with [3H]thymidine at 36 h labeled Thy 1-positive cells in the JGA and hilus. Serial biopsies demonstrated the progressive migration (5-15 micron/d) and proliferation of these mesangial reserve cells until the entire glomerulus was repopulated. The regenerating mesangial population expressed contractile and migratory proteins preferentially at the leading edge of the migratory front. Single as well as multiple pulse labeling with [3H]thymidine confirmed that the entire mesangial cell repopulation originated from only a few mesangial reserve cells. These reserve cells resided in the extraglomerular mesangium in the JGA and were not renin-secreting cells, macrophages, smooth muscle cells, or endothelial cells. These studies document mesangial cell migration in the anti-Thy 1 model of mesangial proliferative glomerulonephritis and provide evidence for a new role for the juxtaglomerular apparatus in the maintenance of the mesangial cell population.


Subject(s)
Glomerular Mesangium/pathology , Glomerulonephritis/pathology , Juxtaglomerular Apparatus/pathology , Kidney Glomerulus/pathology , Animals , Antigens, Surface/metabolism , Cell Movement , Disease Models, Animal , Dose-Response Relationship, Drug , Glomerular Mesangium/metabolism , Immunohistochemistry , Isoantibodies/administration & dosage , Juxtaglomerular Apparatus/metabolism , Kidney Glomerulus/metabolism , Male , Rats , Rats, Wistar , Time Factors
10.
J Clin Invest ; 62(6): 1275-87, 1978 Dec.
Article in English | MEDLINE | ID: mdl-372233

ABSTRACT

The development of immune deposits on the subepithelial surface of the glomerular capillary wall was studied in isolated rat kidneys perfused at controlled perfusion pressure, pH, temperature, and flow rates with recirculating oxygenated perfusate containing bovine serum albumin (BSA) in buffer and sheep antibody to rat proximal tubular epithelial cell brush border antigen (Fx1A). Control kidney were perfused with equal concentrations of non-antibody immunoglobulin (Ig)G. Renal function was monitored by measuring inulin clearance, sodium reabsorption, and urine flow as well as BSA excretion and fractional clearance. Perfused kidneys were studied by light, immunofluorescence, and electron microscopy. All kidneys perfused with anti-Fx1A developed diffuse, finely granular deposits of IgG along the glomerular capillary wall by immunofluorescence. Electron microscopy revealed these deposits to be localized exclusively in the subepithelial space and slit pores. Similar deposits were produced in a nonrecirculating perfusion system, thereby excluding the formation of immune complexes in the perfusate caused by renal release of tubular antigen. Control kidneys perfused with nonantibody IgG did not develop glomerular immune deposits. Renal function and BSA excretion were the same in experimental and control kidneys. Glomerular deposits in antibody perfused kidneys were indistinguishable from deposits in rats injected with anti-Fx1A or immunized with Fx1A to produce autologous immune complex nephropathy. These studies demonstrate that subepithelial immune deposits can be produced in the isolated rat kidney by perfusion with specific antibody to Fx1A in the absence of circulating immune complexes. In this model deposits result from in situ complex formation rather than circulating immune complex deposition.


Subject(s)
Glomerulonephritis/immunology , Kidney Tubules, Proximal/immunology , Animals , Antibodies , Antigen-Antibody Complex , Capillaries/ultrastructure , Fluorescent Antibody Technique , Glomerulonephritis/pathology , Histocytochemistry , Immunoglobulin G , In Vitro Techniques , Kidney Glomerulus/blood supply , Kidney Glomerulus/immunology , Kidney Glomerulus/ultrastructure , Male , Microscopy, Electron , Microvilli/immunology , Perfusion , Rats
11.
J Clin Invest ; 97(4): 1094-101, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8613533

ABSTRACT

The role of the glomerular visceral epithelial cell in the physiologic turnover and pathologic breakdown of the glomerular extracellular matrix has remained largely unexplored. In this study a 98-kD neutral proteinase secreted by cultured rat visceral glomerular epithelial cells was shown to be a calcium, zinc-dependent enzyme secreted in latent form. In addition, the protein was heavily glycosylated and demonstrated proteolytic activity against Type I gelatin, Type IV collagen gelatin, and fibronectin. The similarity in molecular mass and substrate specificities to the 92-kD human matrix metalloproteinase-9 (MMP-9, or gelatinase B) suggested the identity of this activity, which was confirmed by immunoprecipitation and Northern blot analysis. The differences in molecular mass (98 vs. 92 kD) were not due to species-specific differences in glycosylation patterns, since cultured rat peritoneal macrophages secreted MMP-9 as a 92-kD enzyme. Furthermore, transfection of the human MMP-9 cDNA into rat glomerular epithelial cells yielded the 98-kD product. Using a specific monoclonal anti-MMP-9 antibody and in situ reverse transcription (ISRT) analysis of MMP-9 mRNA, the expression of this enzyme was evaluated in vivo. Normal rat glomeruli expressed little immunohistochemical or ISRT staining for MMP-9, while in rats with passive Heymann nephritis there was a major increase in MMP-9 protein and mRNA staining within the visceral epithelial cells. The temporal patterns of MMP-9 expression correlated with the period of proteinuria associated with this model, suggesting that a causal relationship may exist between GEC MMP-9 expression and changes in glomerular capillary permeability.


Subject(s)
Collagenases/metabolism , Glomerulonephritis/enzymology , Kidney Glomerulus/enzymology , Animals , Cells, Cultured , Epithelium/enzymology , Extracellular Matrix Proteins/metabolism , Fluorescent Antibody Technique, Indirect , Gene Expression , In Situ Hybridization , Matrix Metalloproteinase 9 , Polymerase Chain Reaction/methods , RNA, Messenger/genetics , Rats
12.
J Clin Invest ; 61(3): 561-72, 1978 Mar.
Article in English | MEDLINE | ID: mdl-147884

ABSTRACT

The effect of increased capillary permeability on glomerular immune complex localization was studied in rats immunized with proximal tubular antigen (Fx1A) to induce autologous immune complex nephropathy (AICN). AICN rats were made proteinuric by injection or unilateral renal perfusion with aminonucleoside of puromycin (PA) before developing subepithelial complex deposits. Control AICN kidneys developed diffuse granular deposits of IgG and Fx1A on the subepithelial surface of the glomerular basement membrane (GBM) at 3 wk by immunofluorescence and electron microscopy, and deposits increased in subsequent weekly biopsies. In contrast, PA-nephrotic AICN kidneys developed few or no GBM deposits and a significant increase in mesangial localization of IgG and Fx1A during the period of PA-induced proteinuria. These alterations in complex localization were documented both in rats with PA nephrosis and in unilaterally PA-nephrotic kidneys compared with contralateral controls in the same animals, thus excluding any effect of PA on the immunopathogenetic mechanism in AICN as an explanation for these findings. The absence of GBM deposits closely correlated with reduced staining for polyanionic glomerular sialoprotein in proteinuric kidneys, since PA-perfused kidneys studied 2 wk after resolution of proteinuria demonstrated return of normal staining for sialoprotein and development of subepithelial complex deposits similar to those in contralateral control kidneys. These studies demonstrate that properties of the glomerulus itself play an important role in determining the site of complex deposition in experimental AICN and suggest that electrophysical characteristics of the glomerular capillary wall may influence complex localization on the GBM.


Subject(s)
Antigen-Antibody Complex , Disease Models, Animal , Immune Complex Diseases/pathology , Nephritis/pathology , Nephrosis/physiopathology , Puromycin Aminonucleoside/pharmacology , Puromycin/analogs & derivatives , Animals , Antibody Formation , Basement Membrane/immunology , Basement Membrane/pathology , Capillary Permeability/drug effects , Fluorescent Antibody Technique , Immune Complex Diseases/immunology , Immunoglobulin G/isolation & purification , Male , Microscopy, Electron , Microscopy, Fluorescence , Microvilli/immunology , Nephritis/immunology , Nephrosis/chemically induced , Nephrosis/immunology , Nephrosis/pathology , Proteinuria/chemically induced , Proteinuria/immunology , Proteinuria/physiopathology , Rats
13.
J Clin Invest ; 97(11): 2499-508, 1996 Jun 01.
Article in English | MEDLINE | ID: mdl-8647942

ABSTRACT

The expression of the two cytoskeletal linking proteins, moesin and radixin, was examined in experimental mesangial proliferative nephritis in rats (anti-Thy1 model). Moesin and radixin mRNA and protein are constitutively expressed in all cell types of normal rat glomeruli, except podocytes. In the anti-Thy1 model the expression of moesin and radixin was increased in infiltrating macrophages and in activated, alpha-smooth muscle actin-positive mesangial cells and was concentrated in the cellular extensions of mesangial cells in areas of glomerular remodelling. Studies using neutralizing antibodies demonstrated that the increase in moesin and radixin expression by mesangial cells is mediated by PDGF, but not bFGF. The increase in these cytoskeletal proteins appears to be regulated primarily (radixin) or partially (moesin) posttranscriptionally. The data suggest that PDGF mediated upregulation of the cytoskeletal proteins, moesin and radixin, is important for cell migration and other changes that accompany the coordinated restoration of glomerular architecture after injury.


Subject(s)
Blood Proteins/biosynthesis , Cytoskeletal Proteins , Fibroblast Growth Factor 2/pharmacology , Gene Expression/drug effects , Glomerular Mesangium/metabolism , Glomerulonephritis, IGA/metabolism , Kidney Glomerulus/metabolism , Membrane Proteins/biosynthesis , Microfilament Proteins , Platelet-Derived Growth Factor/pharmacology , Protein Biosynthesis , Actins/biosynthesis , Animals , Antibodies , Cells, Cultured , Complement C3/analysis , Glomerular Mesangium/pathology , Glomerulonephritis, IGA/pathology , Immunoglobulin G , Kidney Glomerulus/pathology , Male , Platelet-Derived Growth Factor/immunology , RNA Probes , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Thy-1 Antigens/immunology , Time Factors
15.
Transplantation ; 24(5): 358-66, 1977 Nov.
Article in English | MEDLINE | ID: mdl-72440

ABSTRACT

Heterologous anti-immunoglobulin is a potent immunosuppressive agent that prolongs H-2- and H-3-incompatible skin graft survival. In conjunction with antithymocyte serum, anti-immunoglobulin promotes greater graft life than either antiserum used alone, without evidence of toxicity to recipient animals. Combination treatment with anti-immunoglobulin, antithymocyte serum, and donor spleen cells produces long-term allograft survival to the lifetime of the host and can result in antigen-specific immune unresponsiveness of sufficient strength to permit acceptance of a second-set graft. Anti-immunoglobulin complexes formed in treated mice appear to be localized primarily in the lymphatic reticuloendothelial system.


Subject(s)
Antibodies, Anti-Idiotypic , Antigen-Antibody Complex , Epitopes , Graft Survival , Skin Transplantation , Animals , Kidney Glomerulus/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mononuclear Phagocyte System/immunology , Rabbits , Time Factors
16.
Transplantation ; 22(3): 229-35, 1976 Sep.
Article in English | MEDLINE | ID: mdl-788250

ABSTRACT

Heterologous anti-immunoglobulin (AI) is a potent immunosuppressive agent which compares favorably to anti-thymocyte serum (ATS) in inhibiting E and EAC-rosette formation and diminishing the secondary antibody response to sheep erythrocytes and E. coli lipopolysaccharide. AI prolongs H-2 incompatible skin allograft survival than either antiserum used alone, without evidence of toxicity to recipient mice. AI does not cause significant immune complex glomerular or renovascular deposition and may act by interference with antigen binding.


Subject(s)
Antibodies, Anti-Idiotypic/therapeutic use , Antilymphocyte Serum/therapeutic use , Immunosuppressive Agents , T-Lymphocytes/immunology , Animals , Antigen-Antibody Complex , Binding Sites, Antibody , Erythrocytes/immunology , Escherichia coli/immunology , Hemolytic Plaque Technique , Immune Adherence Reaction , Immunosuppressive Agents/therapeutic use , Kidney/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , Rabbits , Sheep , Skin Transplantation , Time Factors , Transplantation, Homologous
17.
Am J Kidney Dis ; 35(5): E23, 2000 May.
Article in English | MEDLINE | ID: mdl-10793052

ABSTRACT

Antineutrophil cytoplasmic antibodies (ANCA) have proved to be useful serological markers for a subset of vasculitic diseases, including Wegener's granulomatosis, microscopic polyangiitis, and the Churg-Strauss syndrome. The pathogenesis of the ANCA vasculitides remains less clear, including what role, if any, genetic factors play in the expression of ANCA-associated diseases. Familial cases of systemic vasculitis have been reported, and a number of studies have addressed HLA associations of Wegener's and microscopic polyangiitis, but the results have been confusing and inconsistent. We report the first case of P-ANCA-positive vasculitis presenting in a Native American father and daughter. Both patients had systemic vasculitis and were P-ANCA positive with anti-myeloperoxidase (MPO) antibodies.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/blood , Glomerulonephritis/genetics , Vasculitis/genetics , Child , Female , Glomerulonephritis/immunology , Humans , Male , Orbital Diseases/immunology , Peroxidase/immunology , Vasculitis/immunology
18.
Am J Kidney Dis ; 38(6): 1385-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11728980

ABSTRACT

Very-low-density lipoprotein (VLDL) catabolism is impaired in the nephrotic syndrome, partly as a result of structural changes that impair endothelial binding in the presence of lipoprotein lipase. Previous results suggested that postsynthetic modification of VLDL by high-density lipoprotein (HDL) in nephrotic syndrome rats causes their failure to bind endothelia normally. It is unknown (1) whether the structure of secreted lipoproteins is normal before exposure to nephrotic syndrome serum and (2) whether the same structural or functional defects are imparted to chylomicrons (CMs) through their interaction with HDL from nephrotic syndrome rats. CMs were isolated from thoracic duct lymph from rats with passive Heymann's nephritis (HN) and normal controls. CMs from control rats were incubated with HDL from either HN or control rats and reisolated, and apolipoprotein E (apo E) content and endothelial binding were determined. We found that CMs secreted by HN and control rats had similar apo E/B-48 ratios. HDL from HN rats had significantly lower apo E/A-I ratios than controls. Incubation of nascent control CMs with control HDL resulted in a 4-fold increase in CM apo E content, but binding was unaffected. Incubation with HDL from HN resulted in only a 50% increase in CM apo E content but reduced binding of these treated CMs by 50% compared either with nascent control CMs or with CMs incubated with control HDL. HDL from rats with HN alters CM binding to lipoprotein lipase by a mechanism that does not involve reducing the content of apo E already present on CMs at the time of secretion.


Subject(s)
Chylomicrons/metabolism , Endothelium, Vascular/metabolism , Glomerulonephritis/metabolism , Lipoproteins, HDL/metabolism , Animals , Apolipoproteins E/metabolism , Glomerulonephritis/chemically induced , Heymann Nephritis Antigenic Complex , Male , Rats , Rats, Sprague-Dawley , Serum Albumin/metabolism
19.
Metabolism ; 36(5): 496-501, 1987 May.
Article in English | MEDLINE | ID: mdl-3574136

ABSTRACT

Nephrotic patients and rats with experimentally induced nephrotic syndrome have elevated plasma triglycerides and impaired triglyceride removal. This may be due to a defective interaction of chylomicrons and very low density lipoproteins with lipoprotein lipase. Since the glycosaminoglycan, heparan sulfate, was found to stimulate the lipoprotein lipase reaction in vitro, we investigated the plasma heparan sulfate content and measured the urinary excretion of heparan sulfate in control rats and rats with experimentally induced nephrotic syndrome. In addition, we studied the effect of heparan sulfate on the rate of removal of radiolabeled chylomicrons in nephrotic rats. Glycosaminoglycan concentrations in plasma were the same in control and nephrotic rats, although 35S incorporation in high charge glycosaminoglycans was markedly reduced. In addition, in nephrotic rats there is a marked reduction in the urinary excretion of heparan sulfate and chondroitin sulfate suggesting a markedly reduced turnover of these glycosaminoglycans. This was associated with increased plasma triglycerides in nephrotic rats. Nephrotic rats showed a reduced rate of clearance of injected chylomicrons. Intravenous administration of heparan sulfate completely and immediately corrected the chylomicron removal defect. We also noted a log-dose response effect of administered heparan sulfate on chylomicron removal. This effect was not due to a release of soluble lipoprotein lipase by heparan sulfate. These findings suggest that a rapidly turning over fraction of plasma heparan sulfate may play an important role in chylomicron clearance.


Subject(s)
Chylomicrons/metabolism , Glycosaminoglycans/metabolism , Nephrotic Syndrome/metabolism , Animals , Electrophoresis, Agar Gel , Heparitin Sulfate/metabolism , Heparitin Sulfate/physiology , Lipoprotein Lipase/metabolism , Male , Rats , Rats, Inbred Strains , Triglycerides/blood
20.
Kidney Int Suppl ; 42: S19-26, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8361123

ABSTRACT

This review summarizes current understanding of the mechanisms which mediate immune glomerular injury in glomerulonephritis. Non-inflammatory lesions resembling minimal change disease and membranous nephropathy, respectively, are induced by non-complement fixing antibodies to glomerular epithelial cell (GEC) membrane antigens (minimal change disease) and GEC membrane insertion of the C5b-9 membrane attack complex of complement (membranous nephropathy). The cellular mechanisms of these effects are unclear but may involve GEC activation and release of local mediators, such as proteases or oxidants, or GEC detachment from underlying basement membrane. Inflammatory types of glomerular lesions are mediated by circulating inflammatory cells (neutrophils, platelets, macrophages) or by resident glomerular cells (mesangial cells) or both. Neutrophil-mediated injury involves local release of GBM degrading proteases or GBM halogenation induced by the interaction of neutrophil-derived myeloperoxidase with H2O2 and a halide. Neutrophil induced glomerular injury is augmented by platelets. Recent evidence establishes that mesangial cell proliferation in glomerulonephritis is mediated by complement and platelets. Mesangial cell proliferation is accompanied by increased expression of PDGF and PDGF receptor proteins and the genes for these proteins resulting in an autocrine mechanism of cell proliferation. Mesangial cell proliferation is also accompanied by increased release of neutral proteinase in sites of basement membrane damage. Mesangial cell proliferation precedes up-regulation of genes for extracellular matrix components in several models and may be important in the pathogenesis of glomerulosclerosis.


Subject(s)
Glomerulonephritis/etiology , Animals , Autoantibodies/metabolism , Basement Membrane/immunology , Blood Platelets/physiology , Complement Membrane Attack Complex/metabolism , Glomerular Mesangium/physiology , Humans , Kidney Glomerulus/immunology , Kidney Glomerulus/injuries , Leukocytes/physiology , Macrophages/physiology
SELECTION OF CITATIONS
SEARCH DETAIL