Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 327(1): F103-F112, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38779750

ABSTRACT

α-1-Microglobulin (A1M) is a circulating glycoprotein with antioxidant, heme-binding, and mitochondrial protection properties. The investigational drug RMC-035, a modified therapeutic A1M protein, was assessed for biodistribution and pharmacological activity in a broad set of in vitro and in vivo experiments, supporting its clinical development. Efficacy and treatment posology were assessed in various models of kidney ischemia and reperfusion injury (IRI). Real-time glomerular filtration rate (GFR), functional renal biomarkers, tubular injury biomarkers (NGAL and KIM-1), and histopathology were evaluated. Fluorescently labeled RMC-035 was used to assess biodistribution. RMC-035 demonstrated consistent and reproducible kidney protection in rat IRI models as well as in a model of IRI imposed on renal impairment and in a mouse IRI model, where it reduced mortality. Its pharmacological activity was most pronounced with combined dosing pre- and post-ischemia and weaker with either pre- or post-ischemia dosing alone. RMC-035 rapidly distributed to the kidneys via glomerular filtration and selective luminal uptake by proximal tubular cells. IRI-induced expression of kidney heme oxygenase-1 was inhibited by RMC-035, consistent with its antioxidative properties. RMC-035 also dampened IRI-associated inflammation and improved mitochondrial function, as shown by tubular autofluorescence. Taken together, the efficacy of RMC-035 is congruent with its targeted mechanism(s) and biodistribution profile, supporting its further clinical evaluation as a novel kidney-protective therapy.NEW & NOTEWORTHY A therapeutic A1M protein variant (RMC-035) is currently in phase 2 clinical development for renal protection in patients undergoing open-chest cardiac surgery. It targets several key pathways underlying kidney injury in this patient group, including oxidative stress, heme toxicity, and mitochondrial dysfunction. RMC-035 is rapidly eliminated from plasma, distributing to kidney proximal tubules, and demonstrates dose-dependent efficacy in numerous models of ischemia-reperfusion injury, particularly when administered before ischemia. These results support its continued clinical evaluation.


Subject(s)
Alpha-Globulins , Kidney , Reperfusion Injury , Animals , Reperfusion Injury/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/drug therapy , Alpha-Globulins/metabolism , Alpha-Globulins/pharmacology , Male , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Disease Models, Animal , Glomerular Filtration Rate/drug effects , Mice, Inbred C57BL , Humans , Mice , Heme Oxygenase-1/metabolism , Rats , Rats, Sprague-Dawley , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Tissue Distribution
2.
Arthritis Res Ther ; 24(1): 130, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35650639

ABSTRACT

BACKGROUND: We report results from a phase II randomised placebo-controlled trial assessing zibotentan, a highly selective endothelin receptor antagonist (ERA), in chronic kidney disease (CKD) secondary to systemic sclerosis (SSc). METHODS: This trial included three sub-studies: ZEBRA 1-a randomised placebo-controlled, double-blind trial of zibotentan in SSc patients with CKD2 or CKD3 (and glomerular filtration rate (GFR) >45 ml/min) over 26 weeks; ZEBRA 2A-a 26-week placebo-controlled, single-blind trial of zibotentan in scleroderma renal crisis patients not requiring dialysis; and ZEBRA 2B-an open label pharmacokinetic study of zibotentan in patients on haemodialysis. RESULTS: Sixteen patients were screened for ZEBRA 1. Of these, 6 patients were randomised to zibotentan and 7 to placebo. In ZEBRA 1, there were 47 non-serious adverse events (AE) during the trial. Twenty-seven occurred in the placebo group and 20 in the zibotentan group. One serious adverse event (SAE) occurred during ZEBRA1, in the placebo arm. Descriptive statistics did not suggest an effect of study drug on serum sVCAM1. Estimated GFR numerically declined in patients treated with placebo at 26 weeks and 52 weeks. In contrast, average eGFR increased in zibotentan-treated cases. The 4 patients in ZEBRA 2A experienced 8 non-serious AEs, distributed equally between placebo and zibotentan. There was one SAE each in placebo and zibotentan groups, both unrelated to study medication. ZEBRA 2B recruited 8 patients, 6 completed first dosing, and 2 completed a second dosing visit. Pharmacokinetic analysis confirmed zibotentan levels within the therapeutic range. Three patients experienced 3 non-serious AEs. One SAE occurred and was unrelated to study drug. CONCLUSIONS: Zibotentan was generally well-tolerated. ZEBRA 1 did not show any effect of zibotentan on serum sVCAM-1 but was associated with numerical improvement in eGFR at 26 weeks that was more marked at 52 weeks. ZEBRA 2B suggested a feasible dose regimen for haemodialysis patients. TRIAL REGISTRATION: EudraCT no: 2013-003200-39 (first posted January 28, 2014) ClinicalTrials.gov Identifier: NCT02047708 Sponsor protocol number: 13/0077.


Subject(s)
Renal Insufficiency, Chronic , Scleroderma, Systemic , Humans , Pyrrolidines , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/complications , Scleroderma, Systemic/chemically induced , Scleroderma, Systemic/complications , Scleroderma, Systemic/drug therapy , Single-Blind Method
3.
BMC Nephrol ; 21(1): 242, 2020 06 29.
Article in English | MEDLINE | ID: mdl-32600374

ABSTRACT

BACKGROUND: Diabetic kidney disease (DKD) remains one of the leading causes of premature death in diabetes. DKD is classified on albuminuria and reduced kidney function (estimated glomerular filtration rate (eGFR)) but these have modest value for predicting future renal status. There is an unmet need for biomarkers that can be used in clinical settings which also improve prediction of renal decline on top of routinely available data, particularly in the early stages. The iBEAt study of the BEAt-DKD project aims to determine whether renal imaging biomarkers (magnetic resonance imaging (MRI) and ultrasound (US)) provide insight into the pathogenesis and heterogeneity of DKD (primary aim) and whether they have potential as prognostic biomarkers in DKD (secondary aim). METHODS: iBEAt is a prospective multi-centre observational cohort study recruiting 500 patients with type 2 diabetes (T2D) and eGFR ≥30 ml/min/1.73m2. At baseline, blood and urine will be collected, clinical examinations will be performed, and medical history will be obtained. These assessments will be repeated annually for 3 years. At baseline each participant will also undergo quantitative renal MRI and US with central processing of MRI images. Biological samples will be stored in a central laboratory for biomarker and validation studies, and data in a central data depository. Data analysis will explore the potential associations between imaging biomarkers and renal function, and whether the imaging biomarkers improve the prediction of DKD progression. Ancillary substudies will: (1) validate imaging biomarkers against renal histopathology; (2) validate MRI based renal blood flow measurements against H2O15 positron-emission tomography (PET); (3) validate methods for (semi-)automated processing of renal MRI; (4) examine longitudinal changes in imaging biomarkers; (5) examine whether glycocalyx and microvascular measures are associated with imaging biomarkers and eGFR decline; (6) explore whether the findings in T2D can be extrapolated to type 1 diabetes. DISCUSSION: iBEAt is the largest DKD imaging study to date and will provide valuable insights into the progression and heterogeneity of DKD. The results may contribute to a more personalised approach to DKD management in patients with T2D. TRIAL REGISTRATION: Clinicaltrials.gov ( NCT03716401 ).


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/diagnostic imaging , Kidney/diagnostic imaging , Renal Insufficiency, Chronic/diagnostic imaging , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Disease Progression , Humans , Kidney/blood supply , Kidney/pathology , Magnetic Resonance Imaging , Observational Studies as Topic , Oxygen Radioisotopes , Positron-Emission Tomography , Prognosis , Prospective Studies , Renal Circulation , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/pathology , Ultrasonography
4.
Diabetes Care ; 41(9): 1947-1954, 2018 09.
Article in English | MEDLINE | ID: mdl-29980527

ABSTRACT

OBJECTIVE: The decline of estimated glomerular filtration rate (eGFR) in patients with type 2 diabetes is variable, and early interventions would likely be cost-effective. We elucidated the contribution of 17 plasma biomarkers to the prediction of eGFR loss on top of clinical risk factors. RESEARCH DESIGN AND METHODS: We studied participants in PROVALID (PROspective cohort study in patients with type 2 diabetes mellitus for VALIDation of biomarkers), a prospective multinational cohort study of patients with type 2 diabetes and a follow-up of more than 24 months (n = 2,560; baseline median eGFR, 84 mL/min/1.73 m2; urine albumin-to-creatinine ratio, 8.1 mg/g). The 17 biomarkers were measured at baseline in 481 samples using Luminex and ELISA. The prediction of eGFR decline was evaluated by linear mixed modeling. RESULTS: In univariable analyses, 9 of the 17 markers showed significant differences in median concentration between stable and fast-progressing patients. A linear mixed model for eGFR obtained by variable selection exhibited an adjusted R2 of 62%. A panel of 12 biomarkers was selected by the procedure and accounted for 34% of the total explained variability, of which 32% was due to 5 markers. The individual contribution of each biomarker to the prediction of eGFR decline on top of clinical predictors was generally low. When included into the model, baseline eGFR exhibited the largest explained variability of eGFR decline (R2 of 79%), and the contribution of each biomarker dropped below 1%. CONCLUSIONS: In this longitudinal study of patients with type 2 diabetes and maintained eGFR at baseline, 12 of the 17 candidate biomarkers were associated with eGFR decline, but their predictive power was low.


Subject(s)
Biomarkers/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetic Nephropathies/diagnosis , Glomerular Filtration Rate , Aged , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/blood , Diabetic Nephropathies/physiopathology , Female , Humans , Kidney Function Tests , Longitudinal Studies , Male , Middle Aged , Risk Factors
5.
J Med Chem ; 57(11): 4661-76, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24785301

ABSTRACT

Oral phosphodiesterase 4 (PDE4) inhibitors, such as cilomilast and roflumilast, have been shown to be efficacious against chronic obstructive pulmonary disease (COPD). However, these drugs have been hampered by mechanism-related side effects such as nausea and emesis at high doses. Compounds administered by inhalation are delivered directly to the site of action and may improve the therapeutic index required to overcome side effects. This paper describes systematic and rational lead optimization to deliver highly potent, long-acting, and efficacious preclinical inhaled PDE4 inhibitors with low emetic potential.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Benzamides/chemical synthesis , Niacinamide/analogs & derivatives , Phosphodiesterase 4 Inhibitors/chemical synthesis , Pulmonary Disease, Chronic Obstructive/drug therapy , Thiazoles/chemical synthesis , Vomiting/chemically induced , Administration, Inhalation , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacology , Benzamides/adverse effects , Benzamides/pharmacology , Dogs , Ferrets , Humans , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/immunology , Lung/pathology , Neutrophils/pathology , Niacinamide/adverse effects , Niacinamide/chemical synthesis , Niacinamide/pharmacology , Phosphodiesterase 4 Inhibitors/adverse effects , Phosphodiesterase 4 Inhibitors/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship , Thiazoles/adverse effects , Thiazoles/pharmacology
6.
Toxicol Appl Pharmacol ; 272(2): 399-407, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23872097

ABSTRACT

Wnt activation by inhibiting glycogen synthase kinase 3 (GSK-3) causes bone anabolism in rodents making GSK-3 a potential therapeutic target for osteoporotic and osteolytic metastatic bone disease. To understand the wnt pathway related to human disease translation, the ability of 3 potent inhibitors of GSK-3 (AZD2858, AR79, AZ13282107) to 1) drive osteoblast differentiation and mineralisation using human adipose-derived stem cells (hADSC) in vitro; and 2) stimulate rat bone formation in vivo was investigated. Bone anabolism/resorption was determined using clinically relevant serum biomarkers as indicators of bone turnover and bone formation assessed in femurs by histopathology and pQCT/µCT imaging. GSK-3 inhibitors caused ß-catenin stabilisation in human and rat mesenchymal stem cells, stimulated hADSC commitment towards osteoblasts and osteogenic mineralisation in vitro. AZD2858 produced time-dependent changes in serum bone turnover biomarkers and increased bone mass over 28 days exposure in rats. After 7 days, AZD2858, AR79 or AZ13282107 exposure increased the bone formation biomarker P1NP, and reduced the resorption biomarker TRAcP-5b, indicating increased bone anabolism and reduced resorption in rats. This biomarker profile was differentiated from anabolic agent PTH1-34 or the anti-resorptive Alendronate-induced changes. Increased bone formation in cortical and cancellous bone as assessed by femur histopathology supported biomarker changes. 14 day AR79 treatment increased bone mineral density and trabecular thickness, and decreased trabecular number and connectivity assessed by pQCT/µCT. GSK-3 inhibition caused hADSC osteoblastogenesis and mineralisation in vitro. Increased femur bone mass associated with changes in bone turnover biomarkers confirmed in vivo bone formation and indicated uncoupling of bone formation and resorption.


Subject(s)
Bone Remodeling/drug effects , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Mesenchymal Stem Cells/drug effects , Osteoblasts/drug effects , Osteogenesis/drug effects , Animals , Biomarkers/blood , Bone Density/drug effects , Cell Differentiation/drug effects , Enzyme Inhibitors/chemistry , Female , Femur/drug effects , Femur/metabolism , Femur/pathology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Molecular Structure , Osteoblasts/cytology , Osteoblasts/enzymology , Rats , Rats, Sprague-Dawley
7.
Bioorg Med Chem Lett ; 22(21): 6694-9, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23021991

ABSTRACT

The discovery and optimisation of a series of zwitterionic CCR3 antagonists is described. Optimisation of the structure led to AZ12436092, a compound with excellent selectivity over activity at hERG and outstanding pharmacokinetics in preclinical species.


Subject(s)
Drug Discovery , Piperidines/chemistry , Piperidines/pharmacokinetics , Receptors, CCR3/antagonists & inhibitors , Animals , Humans , Inhibitory Concentration 50 , Molecular Structure , Rats
8.
Environ Health Perspect ; 116(1): 13-20, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18197293

ABSTRACT

BACKGROUND: Exposure to particulate matter (PM) has been associated with increased cardiovascular morbidity; however, causative components are unknown. Zinc is a major element detected at high levels in urban air. OBJECTIVE: We investigated the role of PM-associated zinc in cardiac injury. METHODS: We repeatedly exposed 12- to 14-week-old male Wistar Kyoto rats intratracheally (1x/week for 8 or 16 weeks) to a) saline (control); b) PM having no soluble zinc (Mount St. Helens ash, MSH); or c) whole-combustion PM suspension containing 14.5 microg/mg of water-soluble zinc at high dose (PM-HD) and d ) low dose (PM-LD), e) the aqueous fraction of this suspension (14.5 microg/mg of soluble zinc) (PM-L), or f ) zinc sulfate (rats exposed for 8 weeks received double the concentration of all PM components of rats exposed for 16 weeks). RESULTS: Pulmonary inflammation was apparent in all exposure groups when compared with saline (8 weeks > 16 weeks). PM with or without zinc, or with zinc alone caused small increases in focal subepicardial inflammation, degeneration, and fibrosis. Lesions were not detected in controls at 8 weeks but were noted at 16 weeks. We analyzed mitochondrial DNA damage using quantitative polymerase chain reaction and found that all groups except MSH caused varying degrees of damage relative to control. Total cardiac aconitase activity was inhibited in rats receiving soluble zinc. Expression array analysis of heart tissue revealed modest changes in mRNA for genes involved in signaling, ion channels function, oxidative stress, mitochondrial fatty acid metabolism, and cell cycle regulation in zinc but not in MSH-exposed rats. CONCLUSION: These results suggest that water-soluble PM-associated zinc may be one of the causal components involved in PM cardiac effects.


Subject(s)
Air Pollutants/toxicity , Heart Diseases/chemically induced , Particulate Matter/toxicity , Zinc/toxicity , Aconitate Hydratase/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , DNA Damage , DNA, Mitochondrial/genetics , Gene Expression Profiling , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Inflammation/chemically induced , Inflammation/pathology , Lung/drug effects , Lung/pathology , Male , Mitochondria, Heart/metabolism , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred WKY
9.
J Toxicol Environ Health A ; 69(22): 2011-32, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17074742

ABSTRACT

It was recently demonstrated that particulate matter (PM) containing water-soluble zinc produces cardiac injury following pulmonary exposure. To investigate whether pulmonary zinc exposure produces systemic metal imbalance and direct cardiac effects, male Wistar Kyoto (WKY) rats (12-14 wk age) were intratracheally (IT) instilled with saline or 2 micromol/kg zinc sulfate. Temporal analysis was performed for systemic levels of essential metals (zinc, copper, and selenium), and induction of zinc transporter-2 (ZT-2) and metallothionein-1 (MT-1) mRNA in the lung, heart, and liver. Additionally, cardiac gene expression profile was evaluated using Affymetrix GeneChips (rat 230A) arrays to identify zinc-specific effects. Pulmonary zinc instillation produced an increase in plasma zinc to approximately 20% at 1 and 4 h postexposure with concomitant decline in the lung levels. At 24 and 48 h postexposure, zinc levels rose significantly (approximately 35%) in the liver. At these time points, plasma and liver levels of copper and selenium also increased significantly, suggesting systemic disturbance in essential metals. Zinc exposure was associated with marked induction of MT-1 and ZT-2 mRNA in lung, heart, and liver, suggesting systemic metal sequestration response. Given the functional role of zinc in hundreds of proteins, the gene expression profiles demonstrated changes that are expected based on its physiological role. Zinc exposure produced an increase in expression of kinases and inhibition of expression of phosphatases; up- or downregulation of genes involved in mitochondrial function; changes in calcium regulatory proteins suggestive of elevated intracellular free calcium and increases in sulfotransferases; upregulation of potassium channel genes; and changes in free radical-sensitive proteins. Some of these expression changes are reflective of a direct effect of zinc on myocardium following pulmonary exposure, which may result in impaired mitochondrial respiration, stimulated cell signaling, altered Ca2+ homeostasis, and increased transcription of sulfotransferases. Cardiotoxicity may be an outcome of acute zinc toxicosis and occupational exposures to metal fumes containing soluble zinc. Imbalance of systemic metal homeostasis as a result of pulmonary zinc exposure may underlie the cause of extrapulmonary effects.


Subject(s)
Air Pollutants/toxicity , Gene Expression/drug effects , Heart/drug effects , Inhalation Exposure , Myocardium/metabolism , Zinc Sulfate/toxicity , Animals , Calcium/metabolism , Cation Transport Proteins/drug effects , Cation Transport Proteins/metabolism , Copper/blood , Down-Regulation , Enzyme Induction/drug effects , Gene Expression Profiling , Homeostasis , Inflammation , Male , Metallothionein/drug effects , Metallothionein/metabolism , Occupational Exposure , Phosphoric Monoester Hydrolases/metabolism , Phosphotransferases/metabolism , Rats , Rats, Wistar , Selenium/blood , Zinc/blood
10.
Toxicol Sci ; 94(1): 193-205, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16929007

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterized by airway obstruction, inflammation, and mucus hypersecretion, features that are common in bronchitis, emphysema, and often asthma. However, current rodent models do not reflect this human disease. Because genetically predisposed spontaneously hypertensive (SH) rats display phenotypes such as systemic inflammation, hypercoagulation, oxidative stress, and suppressed immune function that are also apparent in COPD patients, we hypothesized that SH rat may offer a better model of experimental bronchitis. We, therefore, exposed SH and commonly used Sprague Dawley (SD) rats (male, 13- to 15-weeks old) to 0, 250, or 350 ppm sulfur dioxide (SO(2)), 5 h/day for 4 consecutive days to induce airway injury. SO(2) caused dose-dependent changes in breathing parameters in both strains with SH rats being slightly more affected than SD rats. Increases in bronchoalveolar lavage fluid (BALF) total cells and neutrophilic inflammation were dose dependent and significantly greater in SH than in SD rats. The recovery was incomplete at 4 days following SO(2) exposure in SH rats. Pulmonary protein leakage was modest in either strain, but lactate dehydrogenase and N-acetyl glucosaminidase activity were increased in BALF of SH rats. Airway pathology and morphometric evaluation of mucin demonstrated significantly greater impact of SO(2) in SH than in SD rats. Baseline differences in lung gene expression pattern suggested marked immune dysregulation, oxidative stress, impairment of cell signaling, and fatty acid metabolism in SH rats. SO(2) effects on these genes were more pronounced in SH than in SD rats. Thus, SO(2) exposure in SH rats may yield a relevant experimental model of bronchitis.


Subject(s)
Bronchitis/metabolism , Rats, Inbred SHR/metabolism , Sulfur Dioxide/toxicity , Acetylglucosaminidase/metabolism , Administration, Inhalation , Animals , Bronchitis/chemically induced , Bronchitis/physiopathology , Bronchoalveolar Lavage Fluid/cytology , Chemokine CXCL2 , Chemokines, CXC/genetics , Cluster Analysis , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , L-Lactate Dehydrogenase/metabolism , Male , Mucus/metabolism , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR/genetics , Rats, Sprague-Dawley , Respiratory Mechanics/drug effects , Species Specificity , Sulfur Dioxide/administration & dosage , Time Factors , Tumor Necrosis Factor-alpha/genetics , Weight Loss/drug effects
11.
Toxicol Appl Pharmacol ; 211(1): 41-52, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16005037

ABSTRACT

Cardiovascular damage induced by pulmonary exposure to environmental chemicals can result from direct action or, secondarily from pulmonary injury. We have developed a rat model of pulmonary exposure to zinc to demonstrate cardiac, coagulative, and fibrinolytic alterations. Male Wistar Kyoto rats were instilled intratracheally with saline or zinc sulfate, 131 microg/kg (2 micromol/kg); the alterations were determined at 1, 4, 24, and 48 h postexposure. High-dose zinc enabled us to show changes in circulating levels of zinc above normal and induce significant pulmonary inflammation/injury such that cardiac impairments were likely. At 1-24 h postexposure, plasma levels of zinc increased to nearly 20% above the base line. Significant pulmonary inflammation and injury were determined by analysis of bronchoalveolar lavage fluid and histopathology in zinc-exposed rats at all time points. Starting at 4 h postexposure, pulmonary damage was accompanied by persistently increased gene expressions of tissue factor (TF) and plasminogen activator-inhibitor-1 (PAI-1), but not thrombomodulin (TM). Cardiac tissues demonstrated similar temporal increases in expressions of TF, PAI-1, and TM mRNA following pulmonary instillation of zinc. In contrast to extensive pulmonary edema and inflammation, only mild, and focal acute, myocardial lesions developed in a few zinc-exposed rats; no histological evidence showed increased deposition of fibrin or disappearance of troponin. At 24 and 48 h postexposure to zinc, increases occurred in levels of systemic fibrinogen and the activated partial thromboplastin time. These data suggest that cardiovascular blood coagulation impairments are likely following pulmonary zinc exposure and associated pulmonary injury and inflammation.


Subject(s)
Air Pollutants/toxicity , Blood Coagulation/drug effects , Heart/drug effects , Lung/drug effects , Zinc/toxicity , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Gene Expression Regulation/drug effects , Heart Diseases/chemically induced , Heart Diseases/immunology , Heart Diseases/metabolism , Intubation, Intratracheal , Lung Diseases/chemically induced , Lung Diseases/immunology , Lung Diseases/metabolism , Male , Myocardium/immunology , Myocardium/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , RNA, Messenger/analysis , Rats , Rats, Inbred WKY , Thrombomodulin/genetics , Thrombomodulin/metabolism , Thromboplastin/genetics , Thromboplastin/metabolism , Zinc/administration & dosage
12.
Environ Health Perspect ; 113(11): 1561-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16263512

ABSTRACT

Several studies have reported health effects of concentrated ambient particles (CAP) in rodents and humans; however, toxicity end points in rodents have provided inconsistent results. In 2000 we conducted six 1-day exposure studies where spontaneously hypertensive (SH) rats were exposed to filtered air or CAPs (< or = 2.5 microm, 1,138-1,765 microg/m3) for 4 hr (analyzed 1-3 hr afterward). In seven 2-day exposure studies in 2001, SH and Wistar Kyoto (WKY) rats were exposed to filtered air or CAP (< or = 2.5 microm, 144-2,758 microg/m3) for 4 hr/day times 2 days (analyzed 1 day afterward). Despite consistent and high CAP concentrations in the 1-day exposure studies, no biologic effects were noted. The exposure concentrations varied among the seven 2-day exposure studies. Except in the first study when CAP concentration was highest, lavageable total cells and macrophages decreased and neutrophils increased in WKY rats. SH rats demonstrated a consistent increase of lavage fluid gamma-glutamyltransferase activity and plasma fibrinogen. Inspiratory and expiratory times increased in SH but not in WKY rats. Significant correlations were found between CAP mass (microgram per cubic meter) and sulfate, organic carbon, or zinc. No biologic effects correlated with CAP mass. Despite low chamber mass in the last six of seven 2-day exposure studies, the levels of zinc, copper, and aluminum were enriched severalfold, and organic carbon was increased to some extent when expressed per milligram of CAP. Biologic effects were evident in those six studies. These studies demonstrate a pattern of rat strain-specific pulmonary and systemic effects that are not linked to high mass but appear to be dependent on CAP chemical composition.


Subject(s)
Air Pollutants/toxicity , Dust , Rats, Inbred SHR , Rats, Inbred WKY , Air Pollutants/analysis , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Carbon/analysis , Dust/analysis , Fibrinogen/metabolism , Lung/cytology , Lung/drug effects , Lung/physiology , Macrophages/cytology , Macrophages/drug effects , Male , Metals/analysis , Neutrophils/cytology , Neutrophils/drug effects , Particle Size , Pulmonary Ventilation/drug effects , Rats , Species Specificity , Sulfates/analysis , gamma-Glutamyltransferase/metabolism
13.
Inhal Toxicol ; 17(1): 29-42, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15764481

ABSTRACT

Based on epidemiologic observations, the issue of adverse health effects of inhaled ultrafine particles (UFP) is currently under intensive discussion. We therefore examined cardiovascular effects of UFP in a controlled animal exposure on young, healthy WKY rats. Short-term exposure (24 h) to carbon UFPs (38 nm, 180 microg m (-3)), generated by spark discharging, induced a mild but consistent increase in heart rate (18 bpm, 4.8%), which was associated with a significant decrease in heart-rate variability during particle inhalation. The timing and the transient character of these responses point to a particle induced alteration of cardiac autonomic balance, mediated by a pulmonary receptor activation. After 24 h of inhalation exposure, bronchoalveolar lavage revealed significant but low-grade pulmonary inflammation (clean air 1.9% vs. UFPs 6.9% polymorphonuclear cells) and on histopathology sporadic accumulation of particle-laden macrophages was found in the alveolar region. There was no evidence of an inflammation-mediated increase in blood coagulability, as UFP inhalation did not induce any significant changes in plasma fibrinogen or factor VIIa levels and there were no prothrombotic changes in the lung or the heart at both the protein and mRNA level. Histological analysis revealed no signs of cardiac inflammation or cardiomyopathy. This study therefore provides toxicological evidence for UFP-associated pulmonary and cardiac effects in healthy rats. Our findings suggest that the observed changes are mediated by an altered sympatho-vagal balance in response to UFP inhalation, but do not support the concept of an inflammation-mediated prothrombotic state by UFP.


Subject(s)
Air Pollutants/toxicity , Carbon/toxicity , Cardiovascular Diseases/etiology , Heart Rate/drug effects , Inhalation Exposure , Lung/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cardiovascular Diseases/veterinary , Inflammation , Lung/pathology , Macrophages , Male , Particle Size , Rats , Rats, Wistar
14.
Inhal Toxicol ; 16 Suppl 1: 5-18, 2004.
Article in English | MEDLINE | ID: mdl-15204789

ABSTRACT

Toll-like receptor 4 (TLR4) has been shown to play a role in cell signaling that results in neutrophilic inflammation in response to lipopolysaccharide and respiratory syncytial virus infection. TLR4 also interacts with CD14, which upon complex formation triggers TLR4-associated signaling pathways to produce a proinflammatory response. This mechanism results in the activation of NF-kappa B and subsequent inflammatory gene induction. In order to determine the effect of combustion source particle matter (PM), rich in zinc and nickel but with negligible endotoxin, on a possible activation of TLR4-mediated cell signaling and inflammation, we intratracheally (IT) instilled 3.3 mg/kg of PM into 12-w-old healthy male Wistar Kyoto (WKY) and susceptible spontaneously hypertensive (SH) rats. Inflammation, inflammatory-mediator gene expression, bronchoalveolar lavage fluid (BALF) protein and LDH, TLR4 and CD14 protein, and NF-kappa B activation in the lung were determined after 24 h. Dose-response data (0.0, 0.83, 3.33, and 8.3 mg/kg PM) for BALF LDH were obtained as a marker of lung cell injury in SH rats. BALF neutrophils, but not macrophages, were significantly increased in the PM-exposed WKY and SH rats. SH rats showed a greater PMN increase than WKY rats. Similarly, BALF protein and LDH levels were also increased following PM exposure but to a significantly greater extent in SH rats. Plasma fibrinogen was increased only in SH rats exposed to PM. The increased inflammation seen in PM-exposed SH rats was accompanied by a significant increase in TLR4 protein in the lung tissue, which was primarily localized in alveolar macrophages and epithelial cells. CD14 was also increased by PM exposure in both SH and WKY rats but was significantly greater in the SH rats. These increases were associated with greater translocation of NF-kappa B in the lungs of SH rather than WKY rats. This was accompanied by increased macrophage inhibitory protein (MIP)-2 mRNA expression at 24 h of exposure. These data suggest that the increased inflammation in the lungs of PM-exposed SH rats compared to WKY rats is accompanied by an increase in TLR4-mediated cell signaling. Thus, one of the mechanisms for greater susceptibility of SH rats to PM exposure may involve an increased activation of the TLR4 signaling pathway.


Subject(s)
Air Pollutants/toxicity , Hypertension/immunology , Lung/drug effects , Lung/immunology , Membrane Glycoproteins/immunology , Power Plants , Receptors, Cell Surface/immunology , Animals , Boston , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chemokine CXCL2 , Hypertension/blood , Hypertension/genetics , Immunohistochemistry , Incineration , Inflammation/etiology , L-Lactate Dehydrogenase/analysis , Lipopolysaccharide Receptors/analysis , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharides/analysis , Lung/pathology , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/biosynthesis , Metals/toxicity , Monokines/analysis , NF-kappa B/analysis , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Cell Surface/analysis , Receptors, Cell Surface/biosynthesis , Signal Transduction/immunology , Toll-Like Receptor 4 , Toll-Like Receptors , Trachea , Tumor Necrosis Factor-alpha/analysis
15.
Toxicol Appl Pharmacol ; 195(1): 35-44, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14962503

ABSTRACT

While environmental particles are associated with mortality and morbidity related to pulmonary and cardiovascular (CV) disease, the mechanisms involved in CV health effects are not known. Changes in systemic clotting factors have been associated with pulmonary inflammation. We hypothesized that inhaled ultrafine particles result in an inflammatory response which may stimulate systemic clotting factor release. Adult male Wistar rats were exposed to either fine or ultrafine carbon black (CB) for 7 h. The attained total suspended particle concentrations were 1.66 mg/m(3) for ultrafine CB and 1.40 mg/m(3) for fine CB. Particle concentration of ultrafine particles was more than 10 times greater than that of fine particles and the count median aerodynamic diameter averaged 114 nm for the ultrafine and 268 nm for the fine carbon particles. Data were collected immediately, 16 and 48 h following exposure. Only ultrafine CB caused an increase in total bronchoalveolar lavage (BAL) leukocytes, whereas both fine (2-fold) and ultrafine (4-fold) carbon particles caused an increase in BAL neutrophils at 16 h postexposure. Exposure to the ultrafine, but not fine, carbon was also associated with significant increases in the total numbers of blood leukocytes. Plasma fibrinogen, factor VII and von Willebrand factor (vWF) were unaffected by particle treatments as was plasma Trolox equivalent antioxidant status (TEAC). Macrophage inflammatory protein-2 mRNA was significantly increased in BAL cells 48 h following exposure to ultrafine CB. The data show that there is a small but consistent significant proinflammatory effect of this exposure to ultrafine particles that is greater than the effect of the same exposure to fine CB.


Subject(s)
Air Pollutants/toxicity , Blood Coagulation/drug effects , Carbon/toxicity , Lung/drug effects , Administration, Inhalation , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chemokine CXCL2 , Leukocyte Count , Lung/metabolism , Male , Monokines/biosynthesis , Neutrophils/cytology , Particle Size , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Time Factors
16.
Free Radic Biol Med ; 34(11): 1369-82, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12757847

ABSTRACT

This review focuses on the potential role that oxidative stress plays in the adverse effects of PM(10). The central hypothesis is that the ability of PM(10) to cause oxidative stress underlies the association between increased exposure to PM(10) and both exacerbations of lung disease and lung cancer. Pulmonary inflammation may also underlie the cardiovascular effects seen following increased PM(10), although the mechanisms of the cardiovascular effects of PM(10) are not well understood. PM(10) is a complex mix of various particle types and several of the components of PM(10) are likely to be involved in the induction of oxidative stress. The most likely of these are transition metals, ultrafine particle surfaces, and organic compounds. In support of this hypothesis, oxidative stress arising from PM(10) has been shown to activate a number of redox-responsive signaling pathways in lung target cells. These pathways are involved in expression of genes that play a role in responses relevant to inflammation and pathological change, including MAPKs, NF-kappaB, AP-1, and histone acetylation. Oxidative stress from particles is also likely to play an important role in the carcinogenic effects associated with PM(10) and hydroxyl radicals from PM(10) cause DNA damage in vitro.


Subject(s)
Calcium Signaling , Environmental Pollutants/adverse effects , Oxidative Stress , Signal Transduction , Animals , Calcium/metabolism , Humans , Inflammation , Reactive Oxygen Species
17.
Biochem Biophys Res Commun ; 302(4): 860-4, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12646250

ABSTRACT

Oxidants and inflammatory mediators such as tumour necrosis factor-alpha (TNF-alpha) activate transcription factors such as NF-kappa B. Interleukin-8 (IL-8) is a ubiquitous inflammatory chemokine that mediates a multitude of inflammatory events in the lung. Ergothioneine is a naturally occurring thiol compound, which possesses antioxidant property. The aim of this study was to determine whether ergothioneine can inhibit the hydrogen peroxide (H(2)O(2))- and TNF-alpha-mediated activation of NF-kappa B and the release of IL-8 in human alveolar epithelial cells (A549). Treatment of A549 cells with H(2)O(2) (100 microM) and TNF-alpha (10 ng/ml) significantly increased NF-kappa B activation using a reporter assay. Ergothioneine inhibited both H(2)O(2)- and TNF-alpha-mediated activation of NF-kappa B. Both H(2)O(2) and TNF-alpha significantly increased IL-8 release, which was inhibited by pre-treatment of A549 cells with ergothioneine compared to the control untreated cells. Ergothioneine also abolished the transcriptional activation of IL-8 in an IL-8-chloramphenicol acetyltransferase (CAT) reporter system, transfected into A549 cells. This indicates a molecular mechanism for the anti-inflammatory effects of ergothioneine.


Subject(s)
Antioxidants/pharmacology , Epithelial Cells/metabolism , Ergothioneine/pharmacology , Interleukin-8/metabolism , NF-kappa B/metabolism , Oxidative Stress , Tumor Necrosis Factor-alpha/metabolism , Antioxidants/metabolism , Epithelial Cells/cytology , Ergothioneine/metabolism , Genes, Reporter , Glutathione/metabolism , Humans , Hydrogen Peroxide/metabolism , Molecular Structure , Oxidants/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Tumor Cells, Cultured
18.
Am J Physiol Lung Cell Mol Physiol ; 284(3): L533-40, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12573991

ABSTRACT

Increases in the levels of environmental particulate matter with a diameter of <10 microm diameter (PM(10)) in the air are associated with a variety of adverse health effects, particularly chronic lung and cardiovascular diseases. The expression of many inflammatory genes involves the remodeling of the chromatin structure provided by histone proteins. Histone acetylation causes the unwinding of chromatin structure, therefore allowing transcription factor access to promoter sites. Acetylation is reversible and is regulated by histone acetyltransferases (HATs), which promote acetylation, and deacetylases, which promote deacetylation. PM(10) and H(2)O(2) increased IL-8 protein release from A549 cells after 24-h treatment, and this was enhanced by histone deacetylase inhibition by trichostatin A (cotreatment). PM(10) and H(2)O(2) treatment also increased HAT activity as well as the level of acetylated histone 4 (H4). PM(10) enhanced H4 acetylation that was mediated by oxidative stress as shown by thiol antioxidant inhibition. Acetylation of H4 mediated by PM(10) was associated with the promoter region of the IL-8 gene. These data suggest that remodeling of chromatin by histone acetylation plays a role in PM(10)-mediated responses in the lungs.


Subject(s)
Air Pollutants/pharmacology , Epithelial Cells/metabolism , Histones/metabolism , Interleukin-8/metabolism , Oxidative Stress/drug effects , Acetylation/drug effects , Cell Line , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Gene Expression Regulation/drug effects , Histone Deacetylase 2 , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Hydrogen Peroxide/pharmacology , Interleukin-8/genetics , NF-kappa B/metabolism , Oxidants/pharmacology , Oxidative Stress/physiology , Particle Size , Promoter Regions, Genetic , Protein Transport/drug effects , RNA, Messenger/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism
19.
Am J Respir Cell Mol Biol ; 28(1): 111-21, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12495939

ABSTRACT

Both acute and chronic exposure to particulates have been associated with increased mortality and morbidity from a number of causes, including chronic obstructive pulmonary disease and other chronic lung diseases. The current study evaluated the hypothesis that ultrafine carbon particles, a component of ambient particulates, could affect tissue repair. To assess this, the three-dimensional collagen gel contraction model was used. Ultrafine carbon black particles, but not fine carbon black, inhibited fibroblast-mediated collagen gel contraction. Although previous research has indicated that inflammatory effects of ultrafine carbon black particles are mediated by oxidant mechanisms, the current study suggests that ultrafine carbon black's inhibition of fibroblast gel contraction is mediated by the binding of both fibronectin and transforming growth factor (TGF)-beta to the ultrafine particles. Binding of TGF-beta was associated with a reduction in nuclear localization of Smads, indicative of inhibition of TGF-beta signal transduction. There was also a decrease in fibronectin mRNA, consistent with a decrease in TGF-beta-mediated response. Taken together, these results demonstrate the ability of ultrafine particles to contribute to altered tissue repair and extend the known mechanisms by which these biologically active particles exert their effects.


Subject(s)
Carbon/pharmacology , Collagen/metabolism , Lung/drug effects , Base Sequence , Culture Media , DNA Primers , Enzyme-Linked Immunosorbent Assay , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Humans , Immunohistochemistry , Lung/cytology , Lung/metabolism , RNA, Messenger/genetics , Transforming Growth Factor beta/metabolism
20.
Ann N Y Acad Sci ; 973: 278-83, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12485877

ABSTRACT

Sustained oxidative stress caused by cigarette smoking induces a chronic inflammatory response, resulting in the destruction of the alveolar cell wall characteristic of emphysema. The loss of tissue may involve the progressive depletion of epithelial cells through inhibition of proliferation leading to cell death. The cell cycle regulator p21(waf1/cip1) acts as a G(1)/S and G(2)/M phase checkpoint regulator. We hypothesize that cigarette smoke-induced oxidative stress and transforming growth factor beta 1 (TGF-beta(1)) may inhibit cellular proliferation by p21(waf1/cip1) in type II alveolar epithelial cells (A549). A significant increase was observed in p21(waf1/cip1) mRNA expression in A549 cells by cigarette smoke condensate, H(2)O(2), and TGF-beta(1). In conclusion, cigarette smoke-induced oxidative stress and TGF-beta(1) modulate expression of the cell cycle inhibitor p21(waf1/cip1). This may be important in the growth arrest and cell survival of alveolar type II cells in the G(1) phase.


Subject(s)
Cell Cycle/physiology , Cyclins/genetics , Oxidative Stress/physiology , Pulmonary Alveoli/physiology , Respiratory Mucosa/physiology , Smoke/adverse effects , Smoking/physiopathology , Transforming Growth Factor beta/pharmacology , Base Sequence , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , DNA Primers , Gene Expression Regulation , Humans , Hydrogen Peroxide/pharmacology , Kinetics , Polymerase Chain Reaction , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...