transforming-growth-factor-beta has been researched along with Glomerulonephritis--Membranous* in 14 studies
2 review(s) available for transforming-growth-factor-beta and Glomerulonephritis--Membranous
Article | Year |
---|---|
Mechanisms and consequences of TGF-ß overexpression by podocytes in progressive podocyte disease.
In patients with progressive podocyte disease, such as focal segmental glomerulosclerosis (FSGS) and membranous nephropathy, upregulation of transforming growth factor-ß (TGF-ß) is observed in podocytes. Mechanical pressure or biomechanical strain in podocytopathies may cause overexpression of TGF-ß and angiotensin II (Ang II). Oxidative stress induced by Ang II may activate the latent TGF-ß, which then activates Smads and Ras/extracellular signal-regulated kinase (ERK) signaling pathways in podocytes. Enhanced TGF-ß activity in podocytes may lead to thickening of the glomerular basement membrane (GBM) by overproduction of GBM proteins and impaired GBM degradation in podocyte disease. It may also lead to podocyte apoptosis and detachment from the GBM, and epithelial-mesenchymal transition (EMT) of podocytes, initiating the development of glomerulosclerosis. Furthermore, activated TGF-ß/Smad signaling by podocytes may induce connective tissue growth factor and vascular endothelial growth factor overexpression, which could act as a paracrine effector mechanism on mesangial cells to stimulate mesangial matrix synthesis. In proliferative podocytopathies, such as cellular or collapsing FSGS, TGF-ß-induced ERK activation may play a role in podocyte proliferation, possibly via TGF-ß-induced EMT of podocytes. Collectively, these data bring new mechanistic insights into our understanding of the TGF-ß overexpression by podocytes in progressive podocyte disease. Topics: Animals; Extracellular Signal-Regulated MAP Kinases; Glomerular Basement Membrane; Glomerulonephritis, Membranous; Glomerulosclerosis, Focal Segmental; Humans; Intercellular Signaling Peptides and Proteins; Kidney Glomerulus; Phosphatidylinositol 3-Kinases; Podocytes; Signal Transduction; Transforming Growth Factor beta | 2012 |
Pathogenic role of TGF-β in the progression of podocyte diseases.
In patients with progressive podocyte diseases, such as focal segmental glomerulosclerosis and membranous nephropathy, there is enhanced expression of transforming growth factor (TGF-β) in podocytes. Biomechanical strain in these diseases may cause overexpression of TGF-β and angiotensin II (Ang II) by podocytes. Oxidative stress induced by Ang II may activate the latent TGF-β. Increased TGF-β activity by podocytes may induce not only the thickening of the glomerular basement membrane (GBM), but also podocyte apoptosis and/or detachment from the GBM, initiating the development of glomerulosclerosis. Furthermore, mesangial matrix expansion frequently occurs in podocyte diseases in association with the development of glomerulosclerosis. This review examines open questions on the pathogenic role of TGF-β that links podocyte injury to GBM thickening, podocyte loss, mesangial matrix expansion and glomerulosclerosis in podocyte diseases. It also describes paracrine regulatory mechanisms of podocyte TGF-β on mesangial cells leading to increased matrix synthesis. Topics: Angiotensin II; Animals; Biomechanical Phenomena; Connective Tissue Growth Factor; Denys-Drash Syndrome; Diabetic Nephropathies; Glomerular Basement Membrane; Glomerulonephritis, Membranous; Glomerulosclerosis, Focal Segmental; Humans; Hypertension, Renal; Kidney Diseases; Models, Biological; Paracrine Communication; Podocytes; Signal Transduction; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A | 2011 |
1 trial(s) available for transforming-growth-factor-beta and Glomerulonephritis--Membranous
Article | Year |
---|---|
[Activity of cathepsin B and collagenase in urine and excretion of fibronectin and TGF-beta 1 in urine of patients with membranous glomerulonephritis].
In 30% cases nephrotic syndrome is due to membranous glomerulonephritis (MG). Fifty percent of patients reveal end stage renal disease in 15 years follow-up. The another 50% gain persistent remission. The pathogenesis of disease is not known. Protein accumulation in glomeruli leads to progressive loss of kidney structure and function in MG. Also the role of tissue proteolytic systems and growth factors in this process is not known. We aimed to estimate urine cathepsin B, collagenase activity and urine excretion of TGF-beta 1 and fibronectin in MG. MG patients revealed increased urine cathepsin B activity (10.58 +/- 8.73 pmol AMC/mg creatinine/min. vs. control 7.11 +/- 2.05 pmol AMC/mg creatinine/min. [p < 0.05]), urine collagenase activity (8.59 +/- 4.26 pmol AMC/mg creatinine/min. vs. control 3.84 +/- 2.09 pmol AMC/mg creatinine/min. [p > 0.02]) and increased urine excretion of fibronectin (214 +/- 335 ng/mg creatinine vs. control 12.7 +/- 6.7 ng/mg creatinine [p < 0.05]) and increased urine excretion of TGF-beta 1 (283.55 +/- 248.13 pg/ml vs. control 36.11 +/- 48.01 pg/ml [p < 0.05]). The results indicates on glomerular overproduction of TGF-beta 1 and urinary leak of proteolytic enzymes which may exacerbate glomerular proteolytic activity in MG. This may lead to glomerular protein accumulation and progressive loss of kidney function and structure in MG. Increased urine fibronectin excretion in MG patients seems to confirm the hypothesis. Topics: Adult; Cathepsin B; Collagenases; Female; Fibronectins; Glomerulonephritis, Membranous; Humans; Male; Transforming Growth Factor beta | 1997 |
11 other study(ies) available for transforming-growth-factor-beta and Glomerulonephritis--Membranous
Article | Year |
---|---|
Amelioration of anti-Thy1-glomerulonephritis by PPAR-gamma agonism without increase of endothelial progenitor cell homing.
Impaired glomerular endothelial integrity is pivotal in various renal diseases and depends on both the degree of glomerular endothelial injury and the effectiveness of glomerular endothelial repair. Glomerular endothelial repair is, in part, mediated by bone marrow-derived endothelial progenitor cells. Peroxisome proliferator activated receptor-gamma (PPAR-gamma) agonists have therapeutic actions independent of their insulin-sensitizing effects, including enhancement of endothelial progenitor cell function and differentiation. We evaluated the effect of PPAR-gamma agonist rosiglitazone (4 mg.kg(-1).day(-1)) on the course of anti-Thy1-glomerulonephritis in rats. Rosiglitazone limited the development of proteinuria and prevented plasma urea elevation (8.1 +/- 0.4 vs. 12.5 +/- 1.1 mmol/l, P = 0.002). Histologically, inflammatory cell influx was not affected, but rosiglitazone-treated rats did show fewer microaneurysmatic glomeruli on day 7 (26 +/- 3 vs. 41 +/- 5%, P = 0.01) and reduced activation of matrix production with reduced renal cortical transforming growth factor-beta, plasminogen activator inhibitor type 1, and fibronectin-1 mRNA expression. However, bone marrow-derived endothelial cell glomerular incorporation was not enhanced (3.1 +/- 0.4 vs. 3.6 +/- 0.3 cells/glomerular cross section; P = 0.31). Rosiglitazone treatment in nonnephritic rats did not influence proteinuria, urea, or renal histology. In conclusion, treatment with PPAR-gamma agonist rosiglitazone ameliorates the course of experimental glomerulonephritis in a nondiabetic model, but not through enhancing incorporation of bone marrow-derived endothelial cells in the glomerulus. Topics: Aneurysm; Animals; Blood Pressure; Bone Marrow Transplantation; Cell Movement; Disease Models, Animal; Endothelial Cells; Extracellular Matrix; Fibronectins; Gene Expression; Glomerulonephritis, Membranous; Hypoglycemic Agents; Isoantibodies; Kidney Cortex; Kidney Glomerulus; Male; Membrane Proteins; Plasminogen Activator Inhibitor 1; PPAR gamma; Proteinuria; Rats; Rats, Inbred BN; Rosiglitazone; Stem Cells; Thiazolidinediones; Transforming Growth Factor beta; Urea | 2008 |
Chronic renal failure and shortened lifespan in COL4A3+/- mice: an animal model for thin basement membrane nephropathy.
A heterozygous mutation in autosomal Alport genes COL4A3 and COL4A4 can be found in 20 to 50% of individuals with familial benign hematuria and diffuse glomerular basement membrane thinning (thin basement membrane nephropathy [TBMN]). Approximately 1% of humans are heterozygous carriers of mutations in the autosomal Alport genes and at risk for developing renal failure as a result of TBMN. The incidence and pathogenesis of renal failure in heterozygous COL4A3/4 mutation carriers is still unclear and was examined further in this study using COL4A3 knockout mice. In heterozygous COL4A3(+/-) mice lifespan, hematuria and renal function (serum urea and proteinuria) were monitored during a period of 3 yr, and renal tissue was examined by light and electron microscopy, immunohistochemistry, and Western blot. Lifespan of COL4A3(+/-) mice was found to be significantly shorter than in healthy controls (21.7 versus 30.3 mo). Persistent glomerular hematuria was detected starting in week 9; proteinuria of > 0.1 g/L started after 3 mo of life and increased to > 3 g/L after 24 mo. The glomerular basement membrane was significantly thinned (167 versus 200 nm in wild type) in 30-wk-old mice, coinciding with focal glomerulosclerosis, tubulointerstitial fibrosis, and increased levels of TGF-beta and connective tissue growth factor. The renal phenotype in COL4A3(+/-) mice resembled the clinical and histopathologic phenotype of human cases of TBMN with concomitant progression to chronic renal failure. Therefore, the COL4A3(+/-) mouse model will help in the understanding of the pathogenesis of TBMN in humans and in the evaluation of potential therapies. Topics: Aging; Animals; Collagen Type IV; Disease Models, Animal; Extracellular Matrix; Glomerular Basement Membrane; Glomerulonephritis, Membranous; Kidney; Kidney Failure, Chronic; Longevity; Mice; Mice, Transgenic; Nephritis, Hereditary; Phenotype; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2006 |
Renal angiotensin II up-regulation and myofibroblast activation in human membranous nephropathy.
The molecular mechanisms of renal injury and fibrosis in proteinuric nephropathies are not completely elucidated but the renin-angiotensin system (RAS) is involved. Idiopathic membranous nephropathy (MN), a proteinuric disease, may progress to renal failure. Our aim was to investigate the localization of RAS components in MN and their correlation with profibrotic parameters and renal injury.. Renal biopsies from 20 patients with MN (11 with progressive disease) were studied for the expression of RAS components [angiotensin-converting enzyme (ACE) and angiotensin II (Ang II)] by immunohistochemistry. Transforming growth factor-beta (TGF-beta) and platelet-derived growth factor (PDGF)-BB were studied by by in situ hybridization, and myofibroblast transdifferentiation by alpha-smooth muscle actin (alpha-SMA) staining.. ACE immunostaining was elevated in tubular cells and appeared in interstitial cells colocalized in alpha-actin-positive cells in progressive disease. Elevated levels of Ang II were observed in tubules and infiltrating interstitial cells. TGF-beta and PDGF mRNAs were up-regulated mainly in cortical tubular epithelial cells in progressive disease (P < 0.01) and correlated with the myofibroblast transdifferentiation (r = 0.8, P < 0.01 for TGF-beta; r = 0.6, P < 0.01 for PDGF). Moreover, in serial sections of progressive cases, the ACE and Ang II over-expression was associated with the tubular expression of these pro-fibrogenic factors, and with the interstitial infiltration and myofibroblast activation.. Intrarenal RAS is selectively activated in progressive MN. De novo expression of ACE at sites of tubulointerstitial injury suggests that the in situ Ang II generation could participate in tubular TGF-beta up-regulation, epithelial-myofibroblast transdifferentiation, and disease progression. These results suggest a novel role of Ang II in human tubulointerstitial injury. Topics: Angiotensin II; Becaplermin; Fibroblasts; Glomerulonephritis, Membranous; Humans; Kidney; Myocytes, Smooth Muscle; Platelet-Derived Growth Factor; Proto-Oncogene Proteins c-sis; Renin-Angiotensin System; Transforming Growth Factor beta; Up-Regulation | 2003 |
mRNA expression of glomerular basement membrane proteins and TGF-beta1 in human membranous nephropathy.
Immunogold densities for the 'classical' and 'novel' alpha chains of type IV collagen, laminin, and fibronectin are increased in the spikes in human membranous nephropathy (MN). To investigate the molecular mechanisms which underlie these changes in glomerular basement membrane (GBM) components, alpha1(IV) collagen, alpha4(IV) collagen, S-laminin, fibronectin, transforming growth factor (TGF)-beta1 and TGF-beta2 mRNA expression was examined in 12 renal biopsy specimens with MN and six renal biopsies with no detectable abnormality by RNA in situ hybridization. In controls, there were relatively low signals of alpha1(IV) collagen, alpha4(IV) collagen, S-laminin, and TGF-beta1 mRNAs, but there were no fibronectin or TGF-beta2 transcripts in glomerular cells. In MN, the number of alpha4(IV) collagen, alpha1(IV) collagen, S-laminin or TGF-beta1 mRNA-expressing cells per glomerular cross-section was significantly larger than in controls (p< 0.05), and fibronectin mRNA was occasionally expressed in glomerular visceral epithelial cells (GECs). No message for TGF-beta2 was seen in MN. The number of TGF-beta1 mRNA-expressing cells per glomerular cross-section significantly correlated with that of alpha1(IV) mRNA-expressing cells (p< 0.01). The MN patients with positivie signal for fibronectin mRNA exhibited more severe GBM thickening than those without (p< 0.05). These results indicate that the increased presence of GBM proteins in spikes of MN is associated with enhanced mRNA expression of these proteins. They also suggest that subepithelial deposits in MN stimulate GECs to produce TGF-beta1, which in turn could mediate the expression of GBM protein genes by GECs. Topics: Adolescent; Adult; Aged; Basement Membrane; Collagen; Female; Fibronectins; Gene Expression; Glomerulonephritis, Membranous; Humans; In Situ Hybridization; Kidney Glomerulus; Laminin; Male; Membrane Proteins; Middle Aged; RNA, Messenger; Transforming Growth Factor beta | 1999 |
Urine activity of cathepsin B, collagenase and urine excretion of TGF-beta 1 and fibronectin in membranous glomerulonephritis.
In 30% of cases nephrotic syndrome is caused by membranous glomerulonephritis (MG). Protein accumulation in glomeruli leads to progressive loss of kidney function and damage of structure in MG. The role of tissue proteolytic systems and growth factors in this process is not known. The purpose of the study was to estimate urine cathepsin B, collagenase activity and urine excretion of TGF-beta 1 and fibronectin in MG. Cathepsin B activity was greater in the urine of MG patients than in the control group (10.58 +/- 8.73 pmol AMC/mg creatinine per min-1 vs control 7.11 +/- 2.05 pmol AMC/mg creatinine per min-1; P < 0.05). Urine collagenase activity was higher in the group of patients than in the control group (8.59 +/- 4.26 pmol AMC/mg creatinine per min-1 vs control 3.84 +/- 2.09 pmol AMC/mg creatinine per min-1 P < 0.02). Urine excretion of fibronectin (45.60 ng/mg creatinine vs control 10.30 ng/mg creatinine; P < 0.04) and TGF-beta 1 levels in the urine were higher than in controls (283.55 +/- 248.13 pg/ml vs 36.11 +/- 48.01 pg/ml; P < 0.01). Results suggest glomerular overproduction of TGF-beta 1 and urinary leak of proteolytic enzymes (PE). This may result in decreased glomerular PE activity in MG and, with time, may lead to protein accumulation in renal glomeruli and to progressive loss of kidney function and damage of structures as the course of MG progresses. PE urine composition as well as ECM protein and cytokine urine excretion may allow noninvasive glomerulopathy course monitoring in humans in the future. Topics: Adult; Cathepsin B; Collagenases; Female; Fibronectins; Glomerulonephritis, Membranous; Humans; Male; Middle Aged; Transforming Growth Factor beta | 1998 |
The renoprotective properties of angiotensin-converting enzyme inhibitors in a chronic model of membranous nephropathy are solely due to the inhibition of angiotensin II: evidence based on comparative studies with a receptor antagonist.
In several models of renal disease progression, angiotensin-converting enzyme (ACE) inhibitors reduced proteinuria and limited glomerulosclerosis, which suggested that reduction of renal angiotensin II (Ang II) activity is crucial for the preservation of glomerular structure and function. However, it cannot be ruled out that other hormonal systems, including inhibition of the bradykinin breakdown, also play a role. We compared the effects of chronic treatment with the ACE inhibitor lisinopril with those of a specific Ang II receptor antagonist, L-158,809, on proteinuria and renal injury in passive Heymann nephritis (PHN), a model of immune renal disease that closely resembles human membranous nephropathy, with long-lasting proteinuria followed by tubulointerstitial damage and glomerulosclerosis. Passive Heymann nephritis was induced with 0.5 mL/100 g of rabbit anti-Fx1A antibody in 24 male Sprague-Dawley rats. The animals were divided into three groups of eight rats each, and were given the following in the drinking water on a daily basis: lisinopril (40 mg/L), L-158,809 (50 mg/L), or no therapy. Treatment started at day 7 (proteinuria was already present) and lasted 12 months. Eight normal rats were used as controls. Untreated PHN rats developed hypertension, while rats with PHN given lisinopril or L-158,809 all had systolic blood pressure values even lower than those of normal rats. Urinary protein excretion progressively increased with time in untreated PHN rats, who developed tubulointerstitial damage and glomerulosclerosis. Both lisinopril and L-158,809 exhibited a potent antiproteinuric effect and preserved glomerular and tubular structural integrity at a similar extent. Renal gene expression of transforming growth factor-beta and extracellular matrix proteins was also effectively reduced by the two treatments. These results indicate that ACE inhibitors and Ang II receptor antagonists are equally effective in preventing renal injury in PHN and suggest that the renoprotective effects of ACE inhibitors in this model are solely due to inhibition of Ang II. Topics: Angiotensin II; Angiotensin Receptor Antagonists; Angiotensin-Converting Enzyme Inhibitors; Animals; Blood Pressure; Chronic Disease; Extracellular Matrix Proteins; Glomerular Filtration Rate; Glomerulonephritis, Membranous; Imidazoles; Kidney; Lisinopril; Male; Proteinuria; Rats; Rats, Sprague-Dawley; Renal Plasma Flow; Tetrazoles; Transforming Growth Factor beta | 1997 |
Urinary transforming growth factor-beta 1 in membranous glomerulonephritis.
Human idiopathic membranous glomerulonephritis (MGN) has a highly variable clinical course and factors determining its outcome are poorly known. Since transforming growth factor-beta 1 (TGF-beta 1) has an essential role in renal fibrogenesis, we studied the possibility to use urinary excretion of TGF-beta 1 in the assessment of progression of the disease in patients with MGN.. Urinary TGF-beta 1 was determined in 41 patients with MGN, 25 healthy subjects, six non-proteinuric renal transplant patients, 10 patients with IgA glomerulonephritis, and seven proteinuric patients (with non-progressive diseases) using a novel, double antibody enzyme immunoassay. The results were compared with renal morphology and clinical indices of activity of MGN over 12 months.. The median urinary TGF-beta 1 excretion (pg/mg creatinine) was significantly higher (1730; range 60-16,970) in MGN patients than in the healthy controls (300; 30-1330; P < 0.0001). In renal allograft recipients the excretion was 840 (250-3440; P < 0.0001 vs healthy controls), in IgA GN it was 1130 (30-4910; P = 0.039), and in proteinuric patients it was 39 (29-165; P = NS). In MGN but not in the proteinuric controls or renal allograft recipients, urinary TGF-beta 1 correlated with urinary albumin excretion (r = 0.86, P < 0.0001) but no correlation with renal function or the duration of the disease was found. Urinary TGF-beta 1 at renal biopsy correlated with interstitial cellular inflammation and its excretion 1 year before the biopsy correlated with indices of sclerosis/fibrosis. Immunosuppressive therapy significantly decreased urinary TGF-beta 1 from 2800 (1610-16,960) to 840 (170-1600) pg/mg creatinine (P = 0.028). Patients with persistent nephrotic syndrome and/or declining renal function had a higher initial TGF-beta 1 excretion (median 3680; 1830-7420 pg/mg creatinine) than those entering partial or complete remission (1060; 60-1960; P = 0.003) within 12 months from sampling. Topics: Adult; Aged; Female; Glomerular Filtration Rate; Glomerulonephritis, Membranous; Humans; Immunosuppressive Agents; Kidney; Male; Middle Aged; Nephrotic Syndrome; Reference Values; Transforming Growth Factor beta | 1997 |
Differential expression of transforming growth factor-beta isoforms and receptors in experimental membranous nephropathy.
In membranous nephropathy (MN) overproduction of matrix by glomerular epithelial cells (GEC) is believed to be responsible for glomerular basement membrane thickening and spikes. We studied experimental MN in rats (passive Heymann nephritis, PHN) at 5, 10 and 30 days. PHN rats exhibited a marked increase in GEC immunostaining for TGF-beta 2 at all time points. TGF-beta 3 staining was increased at day 10 only, and TGF-beta 1 was unchanged. Glomerular mRNA for TGF-beta 2 and -beta 3 was increased by day 5 when urine protein increased, whereas TGF-beta 1 was not. TGF-beta 2 bioactivity was increased at day 5. There was also a marked increase in GEC immunostaining for TGF-beta receptor type I (T beta IR) and TGF-beta receptor type II (T beta IIR) at all time points in PHN. mRNA levels for both receptors increased at day 5. Increases in protein expression and mRNA levels for the TGF-beta 2 and -beta 3 isoforms, and T beta IR and T beta RII were prevented by complement depletion. We conclude that complement-mediated injury to the GEC in vivo is associated with the up-regulation of TGF-beta 2 and -beta 3 isoforms, an increase in TGF-beta 2 bioactivity, and an increase in T beta RI and T beta RII expression. This contrasts with changes in TGF-beta 1 reported in mesangial disease, suggesting that TGF-beta 2 and -beta 3 may be important in diseases of the GEC. The differential expression of TGF-beta isoforms and receptors may be important determinants of the GEC response to injury. Topics: Animals; Glomerulonephritis, Membranous; Kidney Glomerulus; Male; Rats; Rats, Sprague-Dawley; Receptors, Transforming Growth Factor beta; RNA, Messenger; Transforming Growth Factor beta | 1996 |
Increased excretion of urinary transforming growth factor beta in patients with focal glomerular sclerosis.
The urinary transforming growth factor beta (TGF-beta) excretion was measured in 33 patients including 10 with systemic lupus erythematosus (SLE), 8 with focal glomerular sclerosis (FGS), 9 with IgA nephropathy (IgAN), and 6 with membranous nephropathy (MN), and in 7 healthy subjects by enzyme-linked immunosorbent assay using a monoclonal antibody specific for TGF-beta 1 + 2 + 3. A significantly increased urinary TGF-beta excretion was observed in FGS patients (555.5 +/- 458.4 ng/mg Cr) as compared with normal controls (46.9 +/- 43.9 ng/mg Cr) (p < 0.05) and a relative increase in SLE patients (96.4 +/- 58.2 ng/mg Cr) and a decrease in MN patients (24.8 +/- 13.3 ng/mg Cr). In contrast, there was no difference in TGF-beta excretion between IgAN patients (54.1 +/- 37.4 ng/mg Cr) and normal controls. A correlation between the amount of proteinuria and TGF-beta was not found. As has been previously demonstrated in experimental studies, TGF-beta may play a similar role in human glomerular diseases. The results obtained in this study raised the possibility that extracellular matrix might be produced by glomerular cells in vivo under the control of TGF-beta and that TGF-beta might act as a stimulator for the development of glomerulosclerosis. Topics: Adolescent; Adult; Aged; Enzyme-Linked Immunosorbent Assay; Female; Glomerulonephritis, IGA; Glomerulonephritis, Membranous; Glomerulosclerosis, Focal Segmental; Humans; Lupus Erythematosus, Systemic; Male; Middle Aged; Proteinuria; Reference Standards; Transforming Growth Factor beta | 1994 |
Transforming growth factor beta 1 in renal allograft recipients.
Transforming growth factor beta (TGF beta 1) is a prosclerotic cytokine implicated in several disease processes. Recent reports have demonstrated a role for TGF beta 1 in experimental models of glomerulonephritis, focusing attention on the relevance of TGF beta to renal fibrogenesis in human disease. The study reported here was designed to investigate whether circulating, active TGF beta 1 could be detected in renal allograft recipients, and whether plasma levels correlated with episodes of rejection, a process involving both inflammation and fibrosis. We have developed an ELISA assay for active TGF beta 1 using commercially available antibodies, and measured plasma levels in 43 healthy controls, 11 patients with membranous nephropathy (MN) and impaired renal function, 17 transplant recipients with stable renal function, 27 patients with acute cellular rejection, 7 patients with chronic vascular rejection, and 10 patients with acute tubular necrosis and/or cyclosporine toxicity. In the last three groups diagnoses were biopsy-proved, and all samples were collected at the time of biopsy. TGF beta 1 was also measured in urine samples from 8, 11, 0, 9, 4, and 7 individuals, respectively, from each group. TGF beta 1 was not detected in plasma from any of the healthy controls or any of the MN patients, (detection limit of assay 0.1 ng/ml). By comparison, it was significantly increased in all groups of transplant recipients (unpaired t test, P < 0.01), but there were no significant differences between the transplant groups. Plasma TGF beta 1 level did not correlate with renal function (estimated by either serum creatinine or reciprocal creatinine), kidney donor age, recipient age, time since transplantation, or cyclosporine plasma trough level. TGF beta 1 was found in every urine sample tested from healthy controls, with a range from 1 ng/ml to 35 ng/ml. Among the 20 transplant patient urines tested, 2 were negative, 18 were positive but within the range determined for the healthy controls. There were no significant differences between the groups. Topics: Acute Disease; Adult; Aged; Aged, 80 and over; Biomarkers; Chronic Disease; Creatinine; Cyclosporine; Female; Glomerulonephritis, Membranous; Graft Rejection; Humans; Kidney Transplantation; Male; Middle Aged; Reference Values; Regression Analysis; Transforming Growth Factor beta; Transplantation, Homologous | 1994 |
Glomerular expression and cell origin of transforming growth factor-beta 1 in anti-glomerular basement membrane disease.
The glomerular expression (mRNA levels) of transforming growth factor-beta 1 (TGF-beta 1) was assessed in two forms of rat anti-glomerular basement membrane (GBM) disease, a macrophage-independent and a macrophage-dependent variant. After a single intravenous injection of rabbit anti-rat GBM immune serum, significant proteinuria and histopathologic changes developed in both variants. Increased TGF-beta 1 mRNA levels were found in isolated glomeruli of the macrophage-dependent variant only in which glomerular infiltration by macrophages also occurred. Macrophages isolated from glomeruli of animals with this variant demonstrated TGF-beta 1 mRNA levels comparable to those found in glomeruli isolated at the same time point after injection of the anti-GBM serum. The observations indicate that in anti-GBM disease, enhanced glomerular TGF-beta 1 expression occurs in the macrophage-dependent variant and suggest that infiltrating macrophages account for this event. Topics: Animals; Basement Membrane; Glomerulonephritis, Membranous; Kidney Glomerulus; Macrophages; Rabbits; Rats; Rats, Inbred Lew; RNA, Messenger; Transforming Growth Factor beta | 1994 |