Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Anaesthesia ; 77(11): 1259-1267, 2022 11.
Article in English | MEDLINE | ID: mdl-36173018

ABSTRACT

There is clear evidence of a growing workforce gap and this is compounded by demographic data that show the current workforce is ageing. Within the current workforce, more doctors are taking voluntary early retirement and the loss of these experienced clinicians from departments can have wide-ranging effects. Older doctors are at risk of age-related health problems (e.g. sight, musculoskeletal, menopause) and are more susceptible to the effects of fatigue, which may increase the risk of error and or complaint. The purpose of this working party and advocacy campaign was to address concerns over the number of consultants retiring at the earliest opportunity and whether a different approach could extend the working career of consultant anaesthetists and SAS doctors. This could be viewed as 'pacing your career'. The earlier this is considered in a clinician's career the greater the potential mitigation on individuals.


Subject(s)
Anesthetics , Anesthetists , Aging , Anesthesiologists , Female , Humans , Workforce
2.
Aust Vet J ; 99(5): 152-162, 2021 May.
Article in English | MEDLINE | ID: mdl-33624285

ABSTRACT

BACKGROUND: There is little information on foal mortality and the epidemiology of diseases in the neonatal period in Australian equine breeding enterprises. METHODOLOGY: This was a prospective cohort study of 1219 foals on 15 breeding farms in south-eastern Australia to identify the proportion of foals recognised on farm as abnormal at birth or within the first 48 h postpartum, determine the prevalence and risk factors for neonatal disease and assess the subsequent performance of foals in the study population. RESULTS: Overall, 27 foals died within 6 weeks of birth in the study population (2.2%), 142 foals (11.6%) were reported as abnormal at birth, and 304 (25.3%) were regarded as abnormal in the first 48 h postpartum. Non-septic orthopaedic disease (NSOD) was the most common abnormality recognised. Premature foals and foals born after dystocia or abnormal parturition were more likely to have clinical abnormalities recognised, but the intensity of nursing care did not predict outcome. Prophylactic administration of antimicrobial drugs was associated with increased mortality and septic disease. Maternal periparturient problems, foal gender, abnormality at birth and the presence of septic disease or neonatal maladjustment were associated with decreased performance outcomes, whereas measures to assess and augment passive immune transfer were associated with improved athletic performance. CONCLUSIONS AND RELEVANCE: Information in the current study is important for the treatment and management decisions on farm and to identify industry welfare and production priorities. Although the incidence of all outcome variables was variable, factors recognised on farm in the peri-parturient period were predictive of subsequent athletic performance.


Subject(s)
Horse Diseases , Infant, Newborn, Diseases , Animals , Animals, Newborn , Australia/epidemiology , Female , Horse Diseases/epidemiology , Horses , Infant, Newborn , Infant, Newborn, Diseases/veterinary , Pregnancy , Prevalence , Prospective Studies , South Australia
3.
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
4.
Aliment Pharmacol Ther ; 34(9): 1123-32, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21929591

ABSTRACT

BACKGROUND: Cirrhotic patients have an impaired health-related quality of life (HRQOL), which is usually analysed using static paper-pencil questionnaires. The Patient Reported Outcomes Measurement Information System (PROMIS) computerised adaptive testing (CAT) are flexible, freely available, noncopyrighted, HRQOL instruments with US-based norms across 11 domains. CAT presents five to seven questions/domain depending on the patient's response, from large validated question banks. This provides brevity and precision equivalent to the entire question bank. AIM: To evaluate PROMIS CAT tools against 'legacy instruments' for cirrhotics and their informal caregivers. METHODS: A total of 200 subjects: 100 cirrhotics (70 men, 53% decompensated) and 100 caregivers were administered the PROMIS and legacy instruments [Sickness Impact Profile (SIP), Beck depression/anxiety inventories, Pittsburgh Sleep-Quality Index (PSQI) and Epworth Sleepiness scale (ESS)] concurrently. Both legacy and PROMIS results for patients were compared with caregivers and US norms. These were also compared between compensated and decompensated patients. Preference for SIP or PROMIS was inquired of a selected group (n = 70, 50% patients). Test - retest reliability was assessed in another group of 20 patients. RESULTS: Patients had significant impairment on all PROMIS domains apart from anger and anxiety compared with caregivers and US norms (P < 0.02 to <0.0001). Decompensated patients had significantly worse sleep, pain, social and physical function scores compared with compensated ones, similar to legacy instruments. There was a statistically significant correlation between PROMIS and their corresponding legacy instruments. The majority (71%) preferred PROMIS over SIP. PROMIS tools had significant test - retest reliability (ICC range 0.759-0.985) when administered 12 ± 6 days apart. CONCLUSION: PROMIS computerised adaptive testing tools had significant concurrent and discriminant validity, test - retest reliability and subject preference for assessing HRQOL in cirrhotic patients.


Subject(s)
Health Status Indicators , Liver Cirrhosis/psychology , Quality of Life/psychology , Sickness Impact Profile , Adult , Caregivers/psychology , Depressive Disorder/etiology , Depressive Disorder/psychology , Diagnosis, Computer-Assisted , Disability Evaluation , Female , Health Surveys , Humans , Male , Middle Aged , Reproducibility of Results , Surveys and Questionnaires
5.
Eur J Pharm Biopharm ; 72(2): 471-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19462483

ABSTRACT

Salvinorin A is an unregulated potent hallucinogen isolated from the leaves of Salvia divinorum. It is the only known non-nitrogenous kappa-opioid selective agonist, and rivals synthetic lysergic acid diethylamide (LSD) in potency. The objective of this study was to characterize the in vitro transport, in vitro metabolism, and pharmacokinetic properties of Salvinorin A. The transport characteristics of Salvinorin A were assessed using MDCK-MDR1 cell monolayers. The P-glycoprotein (P-gp) affinity status was assessed by the P-gp ATPase assay. In vitro metabolism studies were performed with various specific human CYP450 isoforms and UGT2B7 to assess the metabolic characteristics of Salvinorin A. Cohorts (n = 3) of male Sprague Dawley rats were used to evaluate the pharmacokinetics and brain distribution of Salvinorin A (10 mg/kg, intraperitoneal (i.p.) over a 240-min period. A validated UV-HPLC and LC/MS/MS method was used to quantify the hallucinogen concentrations obtained from the in vitro and in vivo studies, respectively. Salvinorin A displayed a high secretory transport in the MDCK-MDR1 cells (4.07 +/- 1.34 x 10(-)5 cm/s). Salvinorin A also stimulated the P-gp ATPase activity in a concentration (5 and 10 microM)-dependent manner, suggesting that it may be a substrate of (P-gp). A significant decrease in Salvinorin A concentration ranging from 14.7 +/- 0.80% to 31.1 +/- 1.20% was observed after incubation with CYP2D6, CYP1A1, CYP2C18, and CYP2E1, respectively. A significant decrease was also observed after incubation with UGT2B7. These results suggest that Salvinorin A maybe a substrate of UGT2B7, CYP2D6, CYP1A1, CYP2E1, and CYP2C18. The in vivo pharmacokinetic study showed a relatively fast elimination with a half-life (t1/2) of 75 min and a clearance (Cl/F) of 26 L/h/kg. The distribution was extensive (Vd of 47.1 L/kg); however, the brain to plasma ratio was 0.050. Accordingly, the brain half-life was relatively short, 36 min. Salvinorin A is rapidly eliminated after i.p. dosing, in accordance with its fast onset and short duration of action. Further, it appears to be a substrate for various oxidative enzymes and multi-drug resistant protein, P-gp.


Subject(s)
Diterpenes, Clerodane/pharmacokinetics , Hallucinogens/pharmacokinetics , Animals , Cell Line , Chromatography, High Pressure Liquid , Dogs , Half-Life , Male , Rats , Rats, Sprague-Dawley , Spectrophotometry, Ultraviolet
6.
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
7.
Kidney Int ; 70(7): 1279-86, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16900090

ABSTRACT

Although it is clear that genetic predispositions play a role in progressive glomerulosclerosis, identification of specific genes is difficult because of natural genetic heterogeneity among individuals. We have reported a differential susceptibility to progressive glomerulosclerosis after induction of experimental glomerulonephritis anti-Thy-1 nephritis in Lewis rat substrains. Glomerular lesions in Lewis/Møllegard rats resolve spontaneously, whereas Lewis/Maastricht (Lew/Maa) rats develop progressive glomerulosclerosis. This predisposition for progressive glomerulosclerosis is governed by unknown genes that are expressed by renal cells. Here, differential gene expression analysis using a rat complementary DNA micro array revealed neuronal activity-regulated pentraxin (Narp) as a candidate gene involved in the remodeling or progression of damaged glomeruli. Glomerular Narp mRNA expression was monitored during disease in both Lewis sub strains. Immunohistochemistry revealed that Narp protein is exclusively expressed in Lew/Maa glomeruli 7 and 14 days after induction of anti-Thy-1 nephritis. Double-immunofluorescent staining showed that proliferating mesangial cells and parietal epithelial cells (PECs) at sites of adhesion to podocytes are partially Narp-positive, whereas podocytes fail to express Narp. Immunohistochemistry in nephritic Wistar, unilaterally nephrectomized Wistar and Sprague-Dawley rats showed that Narp protein is present only in strains that develop progressive glomerulosclerosis but never in strains that show remodeling. We conclude that Narp is a predictor for anti-Thy-1 nephritis-induced glomerulosclerosis and its expression by PECs may be involved in the progression to glomerulosclerosis.


Subject(s)
C-Reactive Protein/genetics , Gene Expression , Genetic Predisposition to Disease , Glomerulonephritis/genetics , Isoantibodies , Kidney Glomerulus/pathology , Nephritis/genetics , Nephritis/immunology , Nerve Tissue Proteins/genetics , Animals , Antibodies, Monoclonal/immunology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Proliferation , Disease Models, Animal , Disease Progression , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fluorescent Antibody Technique , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Immunohistochemistry , Isoantibodies/immunology , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Kinetics , Mesangial Cells/cytology , Mesangial Cells/metabolism , Mesangial Cells/pathology , Microscopy, Confocal , Nephrectomy , Nephritis/metabolism , Nephritis/pathology , Podocytes/metabolism , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Rats, Wistar , Time Factors
8.
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
9.
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
10.
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
11.
Biopharm Drug Dispos ; 24(9): 397-407, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14689468

ABSTRACT

The present study describes the brain uptake, pharmacokinetics and metabolism of an anticonvulsant enaminone ester E121, which belongs to a new and active series of compounds with potential in vivo anticonvulsant activity in rodent models, in rats. A single dose of E121 was administered i.p. to male Sprague Dawley rats at 10 mg E121/kg body weight. Cohorts of animals (n=3) were killed at varying times over 0-24 h to collect plasma and brain samples. Urinary excretion of E121 was studied in a separate group of five rats at the same dose. A validated HPLC method was used to quantify E121 and its metabolites in plasma, brain and urine. LC-MS/MS was used to characterize the metabolites. The plasma and brain Cmax of 11.0+/-3.0 mg/l and 10.4+/-1.4 mg/kg, respectively, were observed for E121 at 15 min post dose and they declined in a mono-exponential fashion. The plasma Cl/F and t1/2 were 0.57 l/h/kg and 0.75 h, respectively. The brain uptake ratio of E121 was 0.9. Mass spectral analysis of urine showed two major metabolites (m/z 280) and one minor metabolite (m/z 236) that were consistent with initial hydrolysis of the compound to the acid followed by further decarboxylation and appears to be the major route of elimination of E121. The rapid and moderate brain uptake of E121 correlates well with its potential anticonvulsant activity (ED50 3.0 mg/kg p.o. in rats). The brain uptake, pharmacokinetic and metabolic profile of E121 supports the need to further evaluate this compound for its potential as an antiepileptic.


Subject(s)
Aniline Compounds/pharmacokinetics , Anticonvulsants/pharmacokinetics , Brain/metabolism , Cyclohexanecarboxylic Acids/pharmacokinetics , Aniline Compounds/blood , Aniline Compounds/urine , Animals , Anticonvulsants/blood , Anticonvulsants/urine , Area Under Curve , Chromatography, Liquid , Cyclohexanecarboxylic Acids/blood , Cyclohexanecarboxylic Acids/urine , Half-Life , Injections, Intraperitoneal , Male , Mass Spectrometry , Rats , Rats, Sprague-Dawley , Time Factors
12.
Semin Nephrol ; 21(6): 563-72, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11709804

ABSTRACT

Better understanding of the hemodynamic-independent actions of the renin-angiotensin system (RAS) may lead to improved therapies for heart, kidney, and liver fibrosis. The conventional view of the RAS is that its role is solely hemodynamic. Pharmacologic blockade of the RAS is beneficial in treating hypertension, as well as primary renal and cardiac diseases. Recent findings from clinical trials and several laboratories that used different experimental approaches have revealed a whole new dimension to the RAS that is beyond the realm of hemodynamics. The RAS is best viewed as part of a system of interconnected molecules biologically designed to be activated after tissue injury to promote tissue repair and, when in excess, tissue fibrosis. This new understanding of the RAS has important clinical implications. It predicts and explains why blockade of the RAS with angiotensin-converting enzyme inhibitors (ACEI), the newer receptor antagonists, or both together, will significantly slow the progression of fibrotic disease. However, it further suggests that higher doses and/or a combination of angiotensin II blockade with another agent or agents might truly halt progressive fibrosis.


Subject(s)
Angiotensin II/antagonists & inhibitors , Kidney Diseases/drug therapy , Renin-Angiotensin System/physiology , Angiotensin II/physiology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Fibrosis/drug therapy , Humans , Liver Cirrhosis/drug therapy , Pulmonary Fibrosis/drug therapy , Renin-Angiotensin System/drug effects , Transforming Growth Factor beta/physiology
14.
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
15.
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
17.
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
18.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...