Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Aliment Pharmacol Ther ; 39(10): 1113-25, 2014 May.
Article in English | MEDLINE | ID: mdl-24628464

ABSTRACT

BACKGROUND: Safety of individual probiotic strains approved under Investigational New Drug (IND) policies in cirrhosis with minimal hepatic encephalopathy (MHE) is not clear. AIM: The primary aim of this phase I study was to evaluate the safety, tolerability of probiotic Lactobacillus GG (LGG) compared to placebo, while secondary ones were to explore its mechanism of action using cognitive, microbiome, metabolome and endotoxin analysis in MHE patients. METHODS: Cirrhotic patients with MHE patients were randomised 1:1 into LGG or placebo BID after being prescribed a standard diet and multi-vitamin regimen and were followed up for 8 weeks. Serum, urine and stool samples were collected at baseline and study end. Safety was assessed at Weeks 4 and 8. Endotoxin and systemic inflammation, microbiome using multi-tagged pyrosequencing, serum/urine metabolome were analysed between groups using correlation networks. RESULTS: Thirty MHE patients (14 LGG and 16 placebo) completed the study without any differences in serious adverse events. However, self-limited diarrhoea was more frequent in LGG patients. A standard diet was maintained and LGG batches were comparable throughout. Only in the LGG-randomised group, endotoxemia and TNF-α decreased, microbiome changed (reduced Enterobacteriaceae and increased Clostridiales Incertae Sedis XIV and Lachnospiraceae relative abundance) with changes in metabolite/microbiome correlations pertaining to amino acid, vitamin and secondary BA metabolism. No change in cognition was found. CONCLUSIONS: In this phase I study, Lactobacillus GG is safe and well-tolerated in cirrhosis and is associated with a reduction in endotoxemia and dysbiosis.


Subject(s)
Hepatic Encephalopathy/therapy , Lactobacillus , Liver Cirrhosis/therapy , Probiotics/therapeutic use , Aged , Diarrhea/epidemiology , Diarrhea/etiology , Endotoxemia/therapy , Female , Follow-Up Studies , Gastrointestinal Tract/microbiology , Humans , Inflammation/epidemiology , Male , Metabolome , Microbiota , Middle Aged , Probiotics/adverse effects , Tumor Necrosis Factor-alpha/therapeutic use
2.
Kidney Int ; 72(1): 45-52, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17396111

ABSTRACT

Recent evidence indicates that renin itself might be profibrotic, independent of angiotensin II; however, the signaling system by which renin exerts a direct effect is not known. We tested the hypothesis that renin receptor activation, in turn, activates the extracellular-signal regulated kinase 1 and 2 (ERK1/2) of the mitogen-activated protein kinase system in mesangial cells. Recombinant rat renin induced a rapid phosphorylation of ERK1/2 and subsequent cell proliferation in a dose- and time-dependent manner. ERK1/2 activation by renin addition was not altered by angiotensin-converting enzyme inhibition or angiotensin receptor blockade. An ERK kinase inhibitor significantly reduced the renin-induced ERK1/2 phosphorylation and the subsequent increase in transforming growth factor-beta1 (TGF-beta1) and plasminogen activator inhibitor-1 mRNA expression. A small-inhibiting RNA, siRNA, against the renin receptor completely blocked ERK1/2 activation by rat renin. We conclude that renin induces ERK1/2 activation though a receptor-mediated, angiotensin II-independent mechanism in mesangial cells. This renin-activated pathway triggers cell proliferation along with TGF-beta1 and plasminogen activator inhibitor-1 gene expression. This system may play an important role in the overall profibrotic actions of renin.


Subject(s)
Mesangial Cells/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Renin/pharmacology , Transforming Growth Factor beta1/metabolism , Angiotensin II/genetics , Angiotensin II/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Mesangial Cells/cytology , Mitogen-Activated Protein Kinase 3/genetics , Phosphorylation/drug effects , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Signal Transduction/physiology , Time Factors , Transforming Growth Factor beta1/genetics
3.
Kidney Int ; 70(3): 515-22, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16788698

ABSTRACT

Plasminogen activator inhibitor-type 1 (PAI-1) is thought to be profibrotic by inhibiting plasmin generation, thereby decreasing turnover of pathological extracellular matrix (ECM). A mutant, noninhibitory PAI-1 (PAI-1R) was recently shown by us to increase glomerular plasmin generation and reduce disease in anti-thy-1 nephritis. Here, in vitro and in vivo studies were performed to determine whether enhanced plasmin-dependent ECM degradation underlies the therapeutic effect of PAI-1R. 3H-labeled ECM was produced by rat mesangial cells (MCs). The effect of wild-type PAI-1 (wt-PAI-1) and PAI-1R on ECM degradation by newly plated MCs was measured by the release of 3H into medium. In vivo, anti-thy-1 nephritis was assessed in normal, untreated diseased and PAI-1R treated rats with or without the plasmin/plasminogen inhibitor, tranexamic acid (TA). wt-PAI-1 totally inhibited plasmin generation and reduced ECM degradation by 76% when exogenous plasminogen was added. Although PAI-1R alone had no effect, PAI-1R in the presence of wt-PAI-1 reversed the wt-PAI-1 inhibition of ECM degradation in a time- and dose-dependent manner (P<0.001). Plasmin activity and zymography were consistent with ECM degradation. Plasmin inhibitors: alpha2-antiplasmin, aprotinin, and TA completely blocked PAI-1R's ability to normalize ECM degradation (P<0.001). Consistent with the in vitro results, TA reversed PAI-1R-induced reductions in glomerular fibrin and ECM accumulation. Other measures of disease severity were either unaltered or partially reversed. PAI-1R reduces pathological ECM accumulation, in large part through effectively competing with native PAI-1 thereby restoring plasmin generation and increasing plasmin-dependent degradation of matrix components.


Subject(s)
Extracellular Matrix/metabolism , Fibrinolysin/metabolism , Glomerulonephritis/drug therapy , Glomerulonephritis/metabolism , Mesangial Cells/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Animals , Antifibrinolytic Agents/pharmacology , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/pathology , Fibrinolysin/antagonists & inhibitors , Fibronectins/genetics , Fibronectins/metabolism , Fluorescent Antibody Technique , Glomerulonephritis/pathology , In Vitro Techniques , Macrophages/pathology , Male , Mesangial Cells/pathology , Monocytes/pathology , Mutation , Plasminogen/antagonists & inhibitors , Plasminogen/pharmacology , Plasminogen Activator Inhibitor 1/genetics , Proteinuria/drug therapy , Proteinuria/metabolism , Proteinuria/pathology , Rats , Rats, Sprague-Dawley , Tranexamic Acid/pharmacology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tritium
4.
Kidney Int ; 69(10): 1713-4, 2006 May.
Article in English | MEDLINE | ID: mdl-16688190

ABSTRACT

Many approaches to blocking profibrotic TGFbeta overexpression are under way. Therapeutic targeting of TGFbeta-Smad signaling holds promise for slowing or halting progressive renal disease. In this issue, Fukasawa et al., using the unilateral ureteral obstruction model, provide a new target for therapeutic intervention by identifying loss of the Smad corepressors Ski and SnoN as a mechanism that amplifies the profibrotic actions of TGFbeta.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Animals , DNA-Binding Proteins/analysis , Fibrosis/pathology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/analysis , Smad2 Protein/metabolism , Ureteral Obstruction/etiology , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
5.
Kidney Int ; 69(1): 105-13, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16374430

ABSTRACT

Recent evidence suggesting a strong interplay between components of the renin-angiotensin system and key mediators of fibrosis led us to hypothesize that renin, independent of its enzymatic action to enhance angiotensin (Ang) II synthesis, directly increases production of the fibrogenic cytokine transforming growth factor (TGF)-beta. Human or rat mesangial cells (MCs) were treated with human recombinant renin (HrRenin) or rat recombinant renin (RrRenin) and the effects on TGF-beta1, plasminogen activator inhibitor-type 1 (PAI-1), fibronectin (FN) and collagen 1 mRNA and protein were investigated. Blockade of the rat MC renin receptor was achieved using siRNA. HrRenin or RrRenin, at doses shown to be physiologically relevant, induced marked dose- and time-dependent increases in TGF-beta1. These effects were not altered by adding an inhibitor of renin's enzymatic action (RO 42-5892), the Ang II receptor antagonist losartan or the Ang-converting enzyme inhibitor enalapril. RrRenin also induced PAI-1, FN and collagen 1 mRNA and PAI-1 and FN protein in a dose-dependent manner. Neutralizing antibodies to TGF-beta partially blocked these effects. Supernatant and cell lysate Ang I and Ang II levels were extremely low. MC angiotensinogen mRNA was undetectable both with and without added renin. Targeting of the rat renin receptor mRNA with siRNA blocked induction of TGF-beta1. We conclude that renin upregulates MC TGF-beta1 through a receptor-mediated mechanism, independent of Ang II generation or action. Renin-induced increases in TGF-beta1 in turn stimulate increases in PAI-1, FN and collagen I. Thus, renin may contribute to renal fibrotic disease, particularly when therapeutic Ang II blockade elevates plasma renin.


Subject(s)
Angiotensin II/physiology , Extracellular Matrix Proteins/biosynthesis , Glomerular Mesangium/drug effects , Receptors, Cell Surface/physiology , Renin/pharmacology , Transforming Growth Factor beta/biosynthesis , Vacuolar Proton-Translocating ATPases/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Fibronectins/biosynthesis , Glomerular Mesangium/metabolism , Humans , Plasminogen Activator Inhibitor 1/biosynthesis , RNA, Messenger/analysis , Rats , Recombinant Proteins/pharmacology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
7.
Kidney Int ; 59(6): 2146-55, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11380816

ABSTRACT

BACKGROUND: In addition to its well-known role in degrading fibrin, recent evidence suggests that plasmin degrades matrix proteins and activates prometalloproteinases. Plasmin is generated from plasminogen by tissue plasminogen activator (t-PA). We hypothesized that t-PA treatment increases plasmin generation in nephritic glomeruli and degrades pathological matrix leading to a therapeutic reduction in matrix accumulation. METHODS: Anti-Thy-1 nephritis was induced by injection of OX-7 antibody. Rats were given twice daily intravenous injections of saline (disease control group) or human recombinant t-PA (rt-PA; 1 mg/kg body weight) on days 3 through 5. Proteinuria, glomerular matrix protein staining, and glomerular mRNA levels for transforming growth factor-beta 1 (TGF-beta 1), fibronectin, and plasminogen activator inhibitor type 1 (PAI-1) were evaluated at day 6. Localization of rt-PA, plasmin generation by glomeruli in vitro, and glomerular production and content of active TGF-beta1 were also investigated. RESULTS: Compared with disease control animals, proteinuria and staining score for periodic acid-Schiff (2.75 +/- 0.17 vs. 1.41 +/- 0.09), fibronectin-EDA+ (19 +/- 2 vs. 14 +/- 1), laminin (35 +/- 2 vs. 25 +/- 2), type I collagen (33 +/- 1 vs. 21 +/- 3), and type IV collagen (27 +/- 2 vs. 23 +/- 1) were significantly reduced in treated rats (P < 0.01). Glomerular TGF-beta 1, fibronectin, and PAI-1 mRNA levels were unchanged. rt-PA colocalized with fibrin along glomerular capillary walls and in the mesangium. Nephritic glomeruli in vitro had decreased plasmin activity, which was elevated by an in vivo presacrifice injection of rt-PA. Glomerular production and content of active TGF-beta 1 were unchanged by the rt-PA injection. CONCLUSIONS: : These results show that injected rt-PA binds to fibrin in nephritic glomeruli, thus increasing plasmin generation and promoting pathological matrix degradation without activating latent TGF-beta. Agents that increase plasmin generation, such as t-PA, may have potential as antifibrotic therapies.


Subject(s)
Fibrinolysin/biosynthesis , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/metabolism , Kidney Glomerulus/metabolism , Plasminogen Activators/pharmacology , Tissue Plasminogen Activator/pharmacology , Animals , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrin/analysis , Fibrin/metabolism , Fibrinogen/analysis , Fibrinogen/metabolism , Fibrinolysin/metabolism , Fibronectins/genetics , Gene Expression/drug effects , Glomerulosclerosis, Focal Segmental/pathology , Kidney Glomerulus/chemistry , Kidney Glomerulus/pathology , Male , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activators/analysis , Proteinuria/drug therapy , Proteinuria/metabolism , Proteinuria/pathology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Thy-1 Antigens , Tissue Plasminogen Activator/analysis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
8.
Kidney Int ; 57(4): 1493-501, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10760085

ABSTRACT

BACKGROUND: Transforming growth factor-beta (TGF-beta) overexpression plays a key role in the accumulation of extracellular matrix in acute and chronic renal diseases. Recent studies have suggested that the degree of reduction in pathological TGF-beta overexpression can be used as a therapeutic index to evaluate the antifibrotic potential of pharmacological angiotensin II (Ang II) blockade in renal disease. Using this target, we found that treatment with the angiotensin I-converting enzyme inhibitor enalapril or the Ang II type 1 receptor antagonist losartan reduced TGF-beta overexpression more effectively at doses clearly higher than those required to control blood pressure. However, both forms of Ang II blockade were only partially effective in normalizing TGF-beta expression. This study investigated whether a greater antifibrotic, TGF-beta-reducing benefit can be achieved when Ang II blockade is combined with dietary protein restriction. METHODS: Mesangioproliferative glomerulonephritis was induced in male Sprague-Dawley rats on a normal-protein diet. Treatment with a low-protein diet and/or maximally effective doses of enalapril or losartan was started one day after disease induction. On the fifth day, 24-hour urine protein excretion was measured. On the sixth day, cortical kidney tissue was taken for periodic acid-Schiff staining. Isolated glomeruli were used for mRNA extraction or were placed in culture for determination of production of TGF-beta1, the matrix protein fibronectin, and the protease inhibitor plasmin activator inhibitor type 1 (PAI-1) by enzyme-linked immunosorbent assay. RESULTS: Compared with untreated nephritic animals on a normal-protein diet, a single treatment with enalapril, losartan, or low-protein diet significantly reduced glomerular TGF-beta production, albeit to a similar degree of approximately 45%. A moderate, but significant further reduction in pathological TGF-beta expression of a total of 65% for enalapril and 60% for losartan was achieved when these drugs were combined with low-protein feeding. This reduction in TGF-beta overexpression paralleled decreased proteinuria, glomerular matrix accumulation, and overproduction of fibronectin and PAI-1. CONCLUSIONS: Ang II blockade and low-protein diet have additive effects on disease reduction, suggesting that disease progression in humans with chronic renal failure may be slowed more effectively when Ang II blockade and low-protein diet are combined. Since maximal pharmacological Ang II inhibition was used, it is likely that dietary protein restriction further reduces pathological TGF-beta overexpression by mechanisms different from those of enalapril or losartan.


Subject(s)
Angiotensin II/antagonists & inhibitors , Diet, Protein-Restricted , Glomerulonephritis/therapy , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Enalapril/therapeutic use , Losartan/therapeutic use , Male , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Transforming Growth Factor beta/antagonists & inhibitors , Treatment Outcome
10.
Kidney Int ; 57(3): 992-1001, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10720952

ABSTRACT

BACKGROUND: Based upon the central role transforming growth factor-beta (TGF-beta) overexpression appears to play in renal fibrotic diseases, we have recently advocated reduction of TGF-beta as a therapeutic target. As part of efforts to determine the strength of this approach, we have undertaken studies to quantitate the effects of currently used and promising therapies in terms of their potential to reduce markers of disease in anti-thymocyte-serum (ATS)-glomerulonephritis in the rat. Here we assess the therapeutic effect of L-arginine supplementation, which has been shown to reduce fibrosis in a number of hypertensive models, given alone or in combination with low protein diet and started 24 hours after disease induction. METHODS: Glomerulonephritis was induced by intravenous injection of OX-7 monoclonal antibody into 200 g Sprague-Dawley rats. Twenty-four hours later animals were placed in groups that were either untreated, treated with 1% L-arginine in drinking water or 6% protein diets or both. On the fifth day of disease 24-hour urine specimens were collected and systemic blood pressure was measured. On the sixth day rats were anesthetized. Kidneys were perfused, tissue was taken for PAS staining and glomeruli were isolated. Aliquots of glomeruli were used for RNA preparation and for culture to determine 72-hour production of TGF-beta, fibronectin and plasminogen activator-type 1 (PAI-1), which were assayed by ELISA on culture supernatants. Measures of nitrate and nitrite (NOx) production included plasma NOx, urinary NOx and glomerular production of NOx in culture. RESULTS: All disease measures except proteinuria and including matrix accumulation, TGF-beta, fibronectin and PAI-1 production and mRNA expression for TGF-beta, fibronectin and PAI-1 were significantly and similarly reduced by about 50% in groups treated with L-arginine or with low protein diet. Proteinuria was reduced in low protein treated but not in L-arginine supplemented rats. Neither systemic blood pressure nor measures of NO synthesis showed differences between groups that could be attributed to L-arginine supplementation. In contrast, disease-related increases in glomerular production of NOx were markedly reduced by low protein. Combined therapy resulted in small, but statistically significant decreases in most measures of disease. CONCLUSIONS: L-arginine supplementation reduces fibrotic disease in ATS-induced glomerulonephritis if started after disease induction. The absence of evidence for increased NO production related to L-arginine supplementation suggests that L-arginine is acting here through different pathways from those demonstrated in hypertensive models of disease. The data support the ideas that TGF-beta reduction is a valid therapeutic target and that quantitation of TGF-beta reduction is a useful approach for comparing antifibrotic drug candidates.


Subject(s)
Arginine/pharmacology , Dietary Proteins/administration & dosage , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Kidney/pathology , Wound Healing/drug effects , Animals , Culture Techniques , Dietary Proteins/pharmacology , Fibrosis/prevention & control , Glomerulonephritis/metabolism , Kidney Glomerulus/metabolism , Male , Nitrates/blood , Nitrates/metabolism , Nitrates/urine , Nitrites/blood , Nitrites/metabolism , Nitrites/urine , Proteinuria/urine , Rats , Rats, Sprague-Dawley , Time Factors
11.
Nephrol Dial Transplant ; 14(7): 1640-50, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10435871

ABSTRACT

Experimental studies have shown both therapeutic and detrimental consequences of modifying dietary L-arginine intake in renal diseases which likely reflect the complexity of L-arginine metabolism. L-Arginine intake is semi-essential and provides substrate for a number of L-arginine metabolites involved in renal pathology. Dietary L-arginine restriction has been identified as a key mediator of the beneficial effects of low protein diets on human renal fibrosis. Supplementing dietary L-arginine in renal diseases with increased iNOS expression appears to be detrimental and thus, may be harmful in immune-mediated human kidney disorders. Increasing L-arginine intake is beneficial in experimental models of hypertensive renal disease. Based upon available data, we believe additional questions must be answered experimentally, not only to prevent an adverse outcome in humans, but to enhance our chances of human trials which will result in substantially better amelioration of disease than currently available.


Subject(s)
Arginine/physiology , Diet , Dietary Supplements , Kidney Diseases/diet therapy , Animals , Humans , Models, Biological , Nitric Oxide/physiology , Ornithine/physiology , Rats
13.
Kidney Int ; 55(6): 2264-73, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10354274

ABSTRACT

BACKGROUND: Mesangial cell lysis in the antithymocyte serum (ATS)-induced model of glomerulonephritis is dependent on the generation of cytotoxic nitric oxide (NO) through transient induction of NO synthase (iNOS). We hypothesized that increased availability of L-arginine (L-Arg) during mesangial cell lysis might provide iNOS with increased substrate leading to increased lysis, and that this increased lysis would be reflected in more severe fibrotic disease at day 6. METHODS: To ensure whole body equilibration with high L-Arg at the time of injury, rats were pretreated with 1% L-Arg in drinking water for one week prior to the administration of ATS. Animals were sacrificed six hours after ATS injection when previous experiments had indicated iNOS induction had occurred and at six days. At six hours, plasma was obtained for L-Arg levels and nitrite/nitrate (NOx) content. Renal tissues were taken for histological evaluation of glomerular cell counts, macrophage infiltration (ED-1), and iNOS expression. Glomeruli were isolated for detection of iNOS mRNA and placed in culture to study the dependence of NO production on L-Arg concentration. In rats sacrificed at six days, L-Arg supplementation was stopped 16 hours after ATS injection. Fibrotic disease was evaluated by urinary protein excretion, histological assessment of glomerular cell number, matrix accumulation, and production of transforming growth factor-beta1 and matrix components fibronectin and plasminogen activator inhibitor type-1 (PAI-1) by isolated glomeruli in culture. RESULTS: At six hours, the glomerular cell number was significantly reduced by ATS injection (P < 0.01) and further significantly (P < 0. 05) reduced by L-Arg feeding [normal control (NC) = 64.2 +/- 1, ATS = 53.4 +/- 0.7, ATS + L-Arg = 50.8 +/- 0.7]. Disease increased macrophage infiltration and iNOS protein and iNOS mRNA levels markedly (P < 0.01), whereas L-Arg feeding did not further increase these variables. Plasma L-Arg levels (nmol/ml) were reduced by disease (NC = 121 +/- 9, ATS = 84 +/- 13, P < 0.01) and elevated by L-Arg feeding (ATS + L-Arg = 166 +/- 12, P < 0.01). Plasma NOx was significantly increased by ATS and further increased by ATS + L-Arg (P < 0.05). Production of NOx by cultured glomeruli showed striking L-Arg concentration dependence in six hours but not in normal glomeruli. In the group sacrificed at day 6, day 2 proteinuria was higher in the ATS + L-Arg group compared with the ATS alone group (P < 0.05). Measures of fibrotic disease at day 6 all showed large increases over control with ATS alone (P < 0.01), and further small, but significant increases when L-Arg was combined with ATS (P < 0.05). CONCLUSIONS: The results indicate that if given during disease induction, L-Arg supplementation can enhance iNOS-dependent tissue injury by providing increased substrate. Although the increase in injury with L-Arg supplementation was small, it led to increased fibrosis at day 6. These data predict that in diseases with repeated iNOS-dependent tissue injury, L-Arg supplementation may produce cumulative increases in tissue fibrosis.


Subject(s)
Arginine/pharmacology , Glomerular Mesangium/drug effects , Glomerular Mesangium/pathology , Glomerulonephritis/pathology , Animals , Antilymphocyte Serum , Arginine/administration & dosage , Arginine/metabolism , Cell Count , Disease Models, Animal , Fibrosis , Glomerular Mesangium/metabolism , Glomerulonephritis/etiology , Glomerulonephritis/metabolism , Macrophages/pathology , Male , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Proteinuria/etiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , T-Lymphocytes/immunology
14.
Kidney Int ; 55(2): 465-75, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9987071

ABSTRACT

BACKGROUND: The evidence that transforming growth factor-beta (TGF-beta) is a key mediator in the pathogenesis of fibrotic diseases is now supported by several lines of investigation. This evidence provides a certain base for targeting TGF-beta as an antifibrotic agent. METHODS: We generated a chimeric cDNA, termed TGF beta RII/Fc, encoding an extracellular domain of the TGF-beta type II receptor fused to the IgG-Fc domain, and tested whether TGF beta RII/Fc could be a novel strategy for treating glomerular diseases. RESULTS: In cultured BNul-7 cells, recombinant TGF beta RII/Fc reversed the antiproliferative response induced by TGF-beta 1. In addition, TGF beta RII/Fc diminished the TGF-beta 1-induced production of EIIIA-positive fibronectin in cultured normal rat kidney cells. We then introduced the chimeric cDNA into the muscle of the nephritic rats by the hemagglutinating virus of Japan liposome-mediated gene transfer method in order to block the TGF-beta activity in nephritic glomeruli through systemic delivery of chimeric molecules. Treatment with TGF beta RII/Fc gene transfection could suppress the glomerular TGF-beta mRNA in nephritic rats with a comparable effect in the reduction of extracellular matrix accumulation. CONCLUSION: TGF beta RII/Fc successfully inhibited the action of TGF-beta in vitro and in vivo, and gene therapy by chimeric TGF beta RII/Fc might be feasible for the therapy of glomerulosclerosis.


Subject(s)
Extracellular Matrix/metabolism , Genetic Therapy , Glomerulonephritis/therapy , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Receptors, Transforming Growth Factor beta/genetics , Animals , Chimera/genetics , Feasibility Studies , Gene Expression/physiology , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, Transforming Growth Factor beta/metabolism
15.
Kidney Int ; 55(2): 579-92, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9987082

ABSTRACT

BACKGROUND: Human immunodeficiency virus-associated nephropathy (HIVAN) is a renal disease of unknown pathogenesis. Recent evidence suggests that the fibrogenic cytokine transforming growth factor-beta (TGF-beta) might be involved. We hypothesized that overproduction of TGF-beta in the kidney might be involved in the pathogenesis of HIVAN. METHODS: The mRNA and protein expression of TGF-beta isoforms, TGF-beta 1, TGF-beta 2, and TGF beta 3, deposition of matrix proteins induced by TGF-beta, and levels of HIV Tat protein were studied in HIVAN. Controls included normal and diseased kidneys from HIV-positive and -negative patients. The ability of Tat to induce production of TGF-beta and matrix proteins was also studied in human mesangial cells. RESULTS: Normal kidneys, thin basement membrane nephropathy, and minimal change disease were negative for the three TGF-beta isoforms and Tat. In HIVAN, levels of TGF-beta isoforms and Tat were significantly increased, along with the expression of TGF-beta mRNA and deposition of matrix proteins stimulated by TGF-beta. Increased levels of TGF-beta isoforms, but not Tat, were also found in other glomerular diseases characterized by matrix accumulation. HIV infection, in the absence of HIVAN, was not associated with an increase in TGF-beta or Tat expression. Tat stimulated the expression and production of TGF-beta 1 and matrix proteins by human mesangial cells. CONCLUSIONS: Our findings suggest that overproduction of TGF-beta is involved in the pathogenesis of HIVAN.


Subject(s)
AIDS-Associated Nephropathy/metabolism , Transforming Growth Factor beta/metabolism , AIDS-Associated Nephropathy/pathology , Fibronectins/metabolism , Gene Products, tat/metabolism , Gene Products, tat/pharmacology , Glomerular Mesangium/drug effects , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Humans , Immunohistochemistry , In Situ Hybridization , Plasminogen Activator Inhibitor 1/metabolism , RNA, Messenger/metabolism , Transforming Growth Factor beta/genetics , tat Gene Products, Human Immunodeficiency Virus
16.
Kidney Int ; 54(5): 1570-80, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9844133

ABSTRACT

BACKGROUND: Overproduction of transforming growth factor-beta (TGF-beta) is a key mediator of extracellular matrix accumulation in fibrotic diseases. We hypothesized that the degree of reduction of pathological TGF-beta expression can be used as a novel index of the antifibrotic potential of angiotensin II (Ang II) blockade in renal disease. METHODS: One day after induction of Thy 1.1 glomerulonephritis, rats were treated with increasing doses of the Ang I converting enzyme (ACE) inhibitor enalapril and/or the Ang II receptor blocker losartan in the drinking water. Six days after disease induction the therapeutic effect on glomerular TGF-beta overexpression was evaluated. RESULTS: Both enalapril and losartan reduced TGF-beta overproduction in a dose-dependent manner, showing a moderate reduction at doses known to control blood pressure in renal forms of hypertension. A maximal reduction in TGF-beta expression of approximately 45% was seen for both drugs starting at 100 mg/liter enalapril and 500 mg/liter losartan, with no further reduction at doses of enalapril up to 1000 mg/liter or losartan up to 2500 mg/liter. Co-treatment with both drugs was not superior to single therapy. Consistent with our hypothesis that reduction in TGF-beta expression is a valid target, other disease measures, including glomerular matrix accumulation, glomerular production and mRNA expression of the matrix protein fibronectin and the protease inhibitor plasminogen-activator-inhibitor type 1 (PAI-1) closely followed TGF-beta expression. CONCLUSIONS: The data suggest that these therapies act through very similar pathways and that, in order to more effectively treat renal fibrosis, these drugs must be combined with other drugs that act by different mechanisms.


Subject(s)
Angiotensin II/antagonists & inhibitors , Glomerulonephritis, Membranoproliferative/drug therapy , Kidney/pathology , Losartan/therapeutic use , Transforming Growth Factor beta/biosynthesis , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Eating/drug effects , Enalapril/therapeutic use , Fibronectins/biosynthesis , Fibronectins/genetics , Fibrosis , Glomerulonephritis, Membranoproliferative/pathology , Male , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Rats , Rats, Sprague-Dawley
18.
Hypertension ; 31(1 Pt 2): 181-8, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9453300

ABSTRACT

Overproduction of transforming growth factor-beta clearly underlies tissue fibrosis in numerous experimental and human diseases. Transforming growth factor-beta's powerful fibrogenic action results from simultaneous stimulation of matrix protein synthesis, inhibition of matrix degradation, and enhanced integrin expression that facilitates matrix assembly. In animals, overexpression of transforming growth factor-beta by intravenous injection, transient gene transfer, or transgene insertion has shown that the kidney is highly susceptible to rapid fibrosis. The same seems true in human disease, where excessive transforming growth factor-beta has been demonstrated in glomerulonephritis, diabetic nephropathy, and hypertensive glomerular injury. A possible explanation for the kidney's particular susceptibility to fibrosis may be the recent discovery of biologically complex interactions between the renin-angiotensin system and transforming growth factor-beta. Alterations in glomerular hemodynamics can activate both the renin-angiotensin system and transforming growth factor-beta. Components of the renin-angiotensin system act to further stimulate production of transforming growth factor-beta and plasminogen activator inhibitor leading to rapid matrix accumulation. In volume depletion, transforming growth factor-beta is released from juxtaglomerular cells and may act synergistically with angiotensin II to accentuate vasoconstriction and acute renal failure. Interaction of the renin-angiotensin system and transforming growth factor-beta has important clinical implications. The protective effect of inhibition of the renin-angiotensin system in experimental and human kidney diseases correlates closely with the suppression of transforming growth factor-beta production. This suggests that transforming growth factor-beta, in addition to blood pressure, should be a therapeutic target. Higher doses or different combinations of drugs that block the renin-angiotensin system or entirely new drug strategies may be needed to achieve a greater antifibrotic effect.


Subject(s)
Angiotensin II/physiology , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney/pathology , Transforming Growth Factor beta/physiology , Animals , Fibrinolysin/metabolism , Fibrosis , Gene Transfer Techniques , Humans , Integrins/physiology , Transforming Growth Factor beta/biosynthesis
19.
Semin Nephrol ; 17(5): 455-66, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9316214

ABSTRACT

A large body of data has now implicated elevations in the cytokine transforming growth factor-beta (TGF-beta) as a key mediator of tissue fibrosis. A number of mechanisms by which TGF-beta can be increased have been identified. Among them is the potent vasoconstrictor angiotensin II (ANG II). In vitro data indicate that ANG II, independent of blood pressure, increases synthesis and decreases degradation of pathological extracellular matrix components. These effects are largely, but not completely, mediated by ANG II induction of TGF-beta. In many models of renal fibrosis and in a number of human renal diseases, blockade of ANG II retards disease progression. Very recent studies indicate that ANG II blockade suppresses TGF-beta, whether the therapeutic agent is an angiotensin converting enzyme inhibitor or an ANG II type 1 receptor antagonist. These data suggest that an important antifibrotic, therapeutic effect of ANG II blockade is reduction of TGF-beta overexpression and raise the question of whether disease progression could be further retarded if ANG II blockade were optimized for maximal TGF-beta reduction rather than for normalization of systemic blood pressure.


Subject(s)
Angiotensin II/biosynthesis , Hypertension, Renal/physiopathology , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Transforming Growth Factor beta/biosynthesis , Angiotensin II/analysis , Angiotensin II/drug effects , Animals , Disease Models, Animal , Fibrosis/drug therapy , Fibrosis/etiology , Humans , Hypertension, Renal/drug therapy , Hypertension, Renal/etiology , Kidney Diseases/physiopathology , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/drug effects
20.
Curr Opin Nephrol Hypertens ; 6(4): 389-93, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9263690

ABSTRACT

Overexpression of the cytokine transforming growth factor-beta has been identified as the key mediator of chronic progressive kidney fibrosis in experimental and human kidney diseases. The renoprotective effects of angiotensin II blockade and low-protein diets have recently been linked to downregulation of transforming growth factor-beta production. Neutralizing the actions of transforming growth factor-beta by decorin gene therapy appears to be a highly effective new approach in the treatment of renal fibrosis.


Subject(s)
Kidney Glomerulus/injuries , Transforming Growth Factor beta/physiology , Angiotensin II/antagonists & inhibitors , Animals , Decorin , Extracellular Matrix Proteins , Genetic Therapy , Humans , Kidney Diseases/etiology , Kidney Diseases/physiopathology , Kidney Diseases/therapy , Proteoglycans/genetics , Proteoglycans/therapeutic use , Transforming Growth Factor beta/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...