Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 366
Filter
1.
Sci Total Environ ; 799: 149432, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34365262

ABSTRACT

Metformin is one the most prescribed drug to treat type 2 diabetes. In wastewater treatment plants, this drug is bacterially transformed to guanylurea, which occurs at higher concentrations in the aquatic environments than its parent compound. Since there is a huge knowledge gap about the toxicity of this metabolite on aquatic organisms, we aimed to investigate the impact of guanylurea on the embryonic development and oxidative stress biomarkers of zebrafish (Danio rerio). For this effect, zebrafish embryos (4 h post fertilization) were exposed to 25, 50, 100, 200, 250, 25,000, 50,000, 75,000 µg/L guanylurea until 96 h post fertilization. Guanylurea led to a significant delay in the hatching process in all exposure groups. Furthermore, this transformation product affected the embryonic development of fish, inducing severe body alterations and consequently leading to their death. The most pronounced malformations were malformation of tail, scoliosis, pericardial edema, yolk deformation and craniofacial malformation. Concerning oxidative stress response, we demonstrated that guanylurea induced the antioxidant activity of superoxide dismutase, catalase, and glutathione peroxidase in zebrafish embryos. In addition, the levels of lipid peroxidation, protein carbonyl and hydroperoxide content were also increased in the embryos exposed to this transformation product. However, the integrated biomarker response (IBR) analysis carried out in this study demonstrated that oxidative damage biomarkers got more influence over the embryos than antioxidant enzymes. Thus, we can conclude that guanylurea induces oxidative stress in zebrafish embryos, and that this transformation product impair the normal development of this freshwater organism.


Subject(s)
Embryonic Development , Guanidine/analogs & derivatives , Oxidative Stress , Urea/analogs & derivatives , Water Pollutants, Chemical , Animals , Diabetes Mellitus, Type 2 , Embryo, Nonmammalian , Guanidine/toxicity , Urea/toxicity , Water Pollutants, Chemical/toxicity , Zebrafish
2.
Basic Res Cardiol ; 116(1): 24, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33844095

ABSTRACT

Omecamtiv mecarbil (OM) is a promising novel drug for improving cardiac contractility. We tested the therapeutic range of OM and identified previously unrecognized side effects. The Ca2+ sensitivity of isometric force production (pCa50) and force at low Ca2+ levels increased with OM concentration in human permeabilized cardiomyocytes. OM (1 µM) slowed the kinetics of contractions and relaxations and evoked an oscillation between normal and reduced intracellular Ca2+ transients, action potential lengths and contractions in isolated canine cardiomyocytes. Echocardiographic studies and left ventricular pressure-volume analyses demonstrated concentration-dependent improvements in cardiac systolic function at OM concentrations of 600-1200 µg/kg in rats. Administration of OM at a concentration of 1200 µg/kg was associated with hypotension, while doses of 600-1200 µg/kg were associated with the following aspects of diastolic dysfunction: decreases in E/A ratio and the maximal rate of diastolic pressure decrement (dP/dtmin) and increases in isovolumic relaxation time, left atrial diameter, the isovolumic relaxation constant Tau, left ventricular end-diastolic pressure and the slope of the end-diastolic pressure-volume relationship. Moreover, OM 1200 µg/kg frequently evoked transient electromechanical alternans in the rat in vivo in which normal systoles were followed by smaller contractions (and T-wave amplitudes) without major differences on the QRS complexes. Besides improving systolic function, OM evoked diastolic dysfunction and pulsus alternans. The narrow therapeutic window for OM may necessitate the monitoring of additional clinical safety parameters in clinical application.


Subject(s)
Action Potentials/drug effects , Arrhythmias, Cardiac/chemically induced , Cardiotonic Agents/toxicity , Hypotension/chemically induced , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Urea/analogs & derivatives , Ventricular Dysfunction, Left/chemically induced , Ventricular Function, Left/drug effects , Adult , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Blood Pressure/drug effects , Calcium Signaling/drug effects , Diastole , Dogs , Dose-Response Relationship, Drug , Female , Heart Rate/drug effects , Humans , Hypotension/metabolism , Hypotension/physiopathology , Kinetics , Male , Myocytes, Cardiac/metabolism , Rats, Inbred WKY , Systole , Urea/toxicity , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
3.
Biomed Phys Eng Express ; 7(2): 025006, 2021 01 30.
Article in English | MEDLINE | ID: mdl-33522495

ABSTRACT

Carbon-encapsulated magnetic nanoparticles are promising candidate materials for drug-delivery applications. However, due to their hydrophobic and aggregation properties, which indicate lower biocompatibility, proper surface modification of the carbon-based material is required. In the present study, we present the facile route to producing biocompatible magnetic nanocomposite iron oxide/carbon using the liquid medium arc-discharge method. The medium used was ethanol 50% with urea added in various concentrations. Using x-ray diffraction (XRD), the nanocomposite produced was confirmed to have a crystalline structure with distinctive peaks representing iron oxide, graphite, and urea. Fourier transform infrared spectroscopy (FTIR) analysis of the nanocomposite produced in ethanol/acetic acid or ethanol/urea medium shows several vibrations, including Fe-O, C-H, C-O, C=C, C-H, O-H, and C-N, which are intended to be the attached aromatic oxygen- and amine-containing functional groups. The nanocomposite particle was observed to have a core-shell structure that had an iron-compound core coated in a carbon shell possibly modified by polymeric urea groups. The presence of these groups suggested that the nanocomposite would be biocompatible with biological entities in the living body. Lastly, the prepared nanocomposite Fe3O4/C-urea underwent an in-vivo acute toxicity assay to confirm its toxicity. The highest dose of 2000 mg kg-1 BW in this study caused no deaths in the test animals even though cell damages were observed, especially in the liver. This highest dose is considered a maximum tolerable dose and is defined as practically non-toxic.


Subject(s)
Nanocomposites , Animals , Biocompatible Materials/toxicity , Carbon , Ethanol , Ferric Compounds , Humans , Magnetic Phenomena , Nanocomposites/toxicity , Patient Discharge , Urea/toxicity
4.
Article in English | MEDLINE | ID: mdl-33059086

ABSTRACT

We aimed to understand the toxic effects of two crop protecting agents, organophosphate pesticide, chlorpyrifos (CPF) and fertilizer, urea (U), and their binary mixtures at sublethal concentrations for 28-d in a freshwater fish Channa punctata with a battery of biochemical biomarkers in gill and liver. The study has practical value as such mixtures, so often present together in water in the agro-intensive areas, might be predicted to cause cocktail effects. Both CPF and U inhibited AChE, augmented SOD, CAT, GPx activities, and caused lipid peroxidation and depletion in tissue macromolecules in a concentration and duration-dependent manner. While U alone had less severe effects compared to CPF treatments, complex interactions were observed for three combination doses (1CPF + 1U, 2CPF + 1U, 1CPF + 2U). In their mutual effects, antagonism prevailed over other interactions when CPF and U were in equal proportion in the mixture, while synergism was observed for AchE and key antioxidant enzymes when more U was in the mixture. The present study concluded that urea in water bodies might impart adverse effects in combination with pesticides in non-target aquatic organisms such as fish, and there should be a restriction in its excessive usage.


Subject(s)
Acetylcholinesterase/metabolism , Antioxidants/metabolism , Chlorpyrifos/toxicity , Fishes/metabolism , Gills/drug effects , Liver/drug effects , Urea/toxicity , Animals , Catalase/metabolism , Drug Synergism , Fresh Water , Gills/enzymology , Gills/metabolism , Glutathione Peroxidase/metabolism , Liver/enzymology , Liver/metabolism , Malondialdehyde/metabolism , Oxidative Stress/drug effects , Pesticides/toxicity , Superoxide Dismutase/metabolism , Water Pollutants, Chemical/toxicity
5.
AAPS J ; 22(2): 55, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152888

ABSTRACT

Microsampling techniques enable the minimization of blood collection volume from animals and subsequent handling of the blood samples or their derived plasma or serum samples. This offers advantages over conventional large-volume sampling, such as eliminating the need for satellite animals and improving animal welfare aspects, and providing the opportunity for additional assessments in small animals where blood volume constraints limit endpoints. This study evaluated the feasibility of implementation of capillary microsampling (CMS) in a single-dose study in mice with the ultimate goal of enabling its use in toxicology studies. The focus was on the impact of microsampling on toxicokinetic assessment and on the subsequent hematology assessment in the same animal. A seventy (70)-µL blood collection via CMS from the tail vein had a minimal effect on the hematology parameters of mice (strain C57BL/6) in samples taken within 24 h of blood collection. TK parameters were similar in plasma samples collected via CMS and cardiac puncture sampling. A bioanalytical assay was developed which enabled the quantification of concentration of both the parent drug and a metabolite using only 5-µL plasma sample per analysis. Incurred sample reanalysis (ISR), unexpected event investigation, and re-assay were successfully performed on the limited samples (≤ 20 µL) collected from CMS. The results of this study confirmed the feasibility of implementing CMS in regulated mouse toxicity studies and demonstrated that it is possible to eliminate or reduce satellite animals.


Subject(s)
Blood Specimen Collection , Erythrocytes/drug effects , Hematologic Tests , Tail/blood supply , Toxicity Tests , Urea/analogs & derivatives , Valine/analogs & derivatives , Administration, Oral , Animals , Erythrocyte Count , Erythrocytes/metabolism , Feasibility Studies , Hematocrit , Hemoglobins/metabolism , Mice, Inbred C57BL , Toxicokinetics , Urea/administration & dosage , Urea/blood , Urea/toxicity , Valine/administration & dosage , Valine/blood , Valine/toxicity , Workflow
6.
Acta Biomater ; 106: 92-101, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32097711

ABSTRACT

Postoperative adhesions protect, repair, and supply nutrients to injured tissues; however, such adhesions often remain permanent and complicate otherwise successful surgeries by tethering tissues together that are normally separated. An ideal adhesion barrier should not only effectively prevent unwanted adhesions but should be easy to use, however, those that are currently available have inconsistent efficacy and are difficult to handle or to apply. A robust hydrogel film composed of alginate and a photo-crosslinkable hyaluronic acid (HA) derivative (glycidyl methacrylate functionalized hyaluronic acid (GMHA)) represents a solution to this problem. A sacrificial porogen (urea) was used in the film manufacture process to impart macropores that yield films that are more malleable and tougher than equivalent films produced without the sacrificial porogen. The robust mechanical behavior of these templated alginate/GMHA films directly facilitated handling characteristics of the barrier film. In a rat peritoneal abrasion model for adhesion formation, the polysaccharide films successfully prevented adhesions with statistical equivalence to the leading anti-adhesion technology on the market, Seprafilm®. STATEMENT OF SIGNIFICANCE: Postoperative adhesions often remain permanent and complicate otherwise successful surgeries by tethering tissues together that are normally separated and pose potentially significant challenges to patients. Therefore, the generation of adhesion barriers that are easy to deploy during surgery and effectively prevent unwanted adhesions is a big challenge. In this study robust hydrogel films composed of alginate and a photo-crosslinkable hyaluronic acid (HA) derivative (glycidyl methacrylate functionalized HA, GMHA) were fabricated and investigated for their potential to act as a solution to this problem using a rat peritoneal abrasion model for adhesion formation. We observed the polysaccharide films successfully prevented adhesions with statistical equivalence to the leading anti-adhesion technology on the market, Seprafilm®, suggesting that such films represent a promising strategy for the prevention of postoperative adhesions.


Subject(s)
Alginates/chemistry , Hyaluronic Acid/analogs & derivatives , Hydrogels/chemistry , Membranes, Artificial , Postoperative Complications/prevention & control , Tissue Adhesions/prevention & control , Alginates/toxicity , Animals , Anisotropy , Epoxy Compounds/chemistry , Epoxy Compounds/toxicity , Female , Fibroblasts/drug effects , Humans , Hyaluronic Acid/toxicity , Hydrogels/toxicity , Methacrylates/chemistry , Methacrylates/toxicity , Porosity , Rats, Sprague-Dawley , Tensile Strength , Urea/chemistry , Urea/toxicity
8.
Int J Mol Sci ; 20(12)2019 Jun 25.
Article in English | MEDLINE | ID: mdl-31242575

ABSTRACT

Elucidation of molecular and cellular mechanisms of the uremic syndrome is a very challenging task. More than 130 substances are now considered to be "uremic toxins" and represent a very diverse group of molecules. The toxicity of these molecules affects many cellular processes, and expectably, some of them are able to disrupt mitochondrial functioning. However, mitochondria can be the source of uremic toxins as well, as the mitochondrion can be the site of complete synthesis of the toxin, whereas in some scenarios only some enzymes of the pathway of toxin synthesis are localized here. In this review, we discuss the role of mitochondria as both the target and source of pathological processes and toxic compounds during uremia. Our analysis revealed about 30 toxins closely related to mitochondria. Moreover, since mitochondria are key regulators of cellular redox homeostasis, their functioning might directly affect the production of uremic toxins, especially those that are products of oxidation or peroxidation of cellular components, such as aldehydes, advanced glycation end-products, advanced lipoxidation end-products, and reactive carbonyl species. Additionally, as a number of metabolic products can be degraded in the mitochondria, mitochondrial dysfunction would therefore be expected to cause accumulation of such toxins in the organism. Alternatively, many uremic toxins (both made with the participation of mitochondria, and originated from other sources including exogenous) are damaging to mitochondrial components, especially respiratory complexes. As a result, a positive feedback loop emerges, leading to the amplification of the accumulation of uremic solutes. Therefore, uremia leads to the appearance of mitochondria-damaging compounds, and consecutive mitochondrial damage causes a further rise of uremic toxins, whose synthesis is associated with mitochondria. All this makes mitochondrion an important player in the pathogenesis of uremia and draws attention to the possibility of reducing the pathological consequences of uremia by protecting mitochondria and reducing their role in the production of uremic toxins.


Subject(s)
Mitochondria/metabolism , Urea/metabolism , Uremia/metabolism , Acute Kidney Injury/complications , Acute Kidney Injury/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Humans , Mitochondria/drug effects , Molecular Targeted Therapy , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Toxins, Biological/metabolism , Toxins, Biological/toxicity , Urea/toxicity , Uremia/blood , Uremia/drug therapy , Uremia/etiology
9.
Environ Toxicol Chem ; 38(5): 1023-1028, 2019 05.
Article in English | MEDLINE | ID: mdl-30835871

ABSTRACT

Metformin is currently thought to be the highest drug by weight released into the aquatic environment, as a direct result of its widespread use in the treatment of a number of human health disorders. The removal of metformin from wastewaters is directly related to the formation of guanylurea (metformin's only known persistent degradation product), which is generally present at higher concentrations in surface waters than the parent compound. With metformin use rising steadily, it is important to characterize the effects of guanylurea on nontarget aquatic organisms. We recently demonstrated the effects of developmental exposure to environmentally relevant concentrations of metformin on the growth of early life stage (ELS) medaka as well as effects on the body weight of adult male fish following full-life cycle exposures. In the present study, we describe similar effects of guanylurea exposure on these endpoints and life stages. Guanylurea led to effects on growth in a 28-d ELS assessment that were similar to those of metformin; however, these effects occurred at concentrations in the ng/L range compared with the µg/L range for metformin. A possible sex-dependent association with body weight changes was also observed in adults following a 165-d full-life cycle exposure to guanylurea alone or in a mixture with metformin. To our knowledge, the present is the first study to report the toxicity of guanylurea to nontarget aquatic organisms. Environ Toxicol Chem 2019;00:1-6. © 2019 SETAC.


Subject(s)
Guanidines/toxicity , Life Cycle Stages/drug effects , Metformin/toxicity , Oryzias/growth & development , Urea/analogs & derivatives , Animals , Body Weight/drug effects , Female , Guanidines/chemistry , Male , Metformin/chemistry , Oryzias/anatomy & histology , Urea/chemistry , Urea/toxicity , Water Pollutants, Chemical/toxicity
10.
Food Res Int ; 116: 241-248, 2019 02.
Article in English | MEDLINE | ID: mdl-30716942

ABSTRACT

Generation of uremic toxins p-cresylsulfate (p-CS), indoxyl sulfate (IS) and indole 3-acetic acid (IAA) in hemodialysis (HD) individuals may be associated with the gut flora and recognized markers of disease progression. This study investigated the effect of synbiotic meal on uremic toxins in HD individuals. We conducted randomized singleblind and placebo-controlled intervention study with 58 HD subjects (20F/38M, 63.1 ±â€¯10.9-old) who were randomly allocated in synbiotic group (SG, 40 g of extruded sorghum plus 100 mL of unfermented probiotic milk) or control group (CG, 40 g of extruded corn plus 100 mL of pasteurized milk), during 7-wk Metabolic markers and uremic toxins, fecal concentration of short chain fatty acid and pH value was determined. The SG group had decreased serum p-CS and IS, as well as decreased urea concentration (p < .05) compared to CG. SG showed higher fecal butyric acid and lower pH compared to baseline and SC (p < .05). In addition, serum p-CS and fecal pH were positively correlated to urea concentration in SG participants at the endpoint. The consumption of the synbiotic meal during 7-wk reduced colonic pH, and reduced serum uremic (p-CS and IS) toxins and urea in HD subjects.


Subject(s)
Meals , Renal Dialysis , Synbiotics , Urea/toxicity , Uremia/blood , Aged , Bifidobacterium longum , Biomarkers/blood , Brazil , Cresols , Female , Gastrointestinal Microbiome , Humans , Hydrogen-Ion Concentration , Indican , Indoleacetic Acids , Male , Middle Aged , Probiotics/therapeutic use , Renal Insufficiency, Chronic , Sulfuric Acid Esters , Urea/blood
11.
PLoS One ; 14(12): e0227123, 2019.
Article in English | MEDLINE | ID: mdl-31891646

ABSTRACT

BACKGROUND: End-stage renal disease (ESRD) patients usually suffer from a high prevalence of central nervous system abnormalities, including cognitive impairment and emotional disorders, which severely influence their quality of life. There have been many neuroimaging research developments in ESRD patients with brain function abnormalities; however, the dysfunction of the salience network (SN) of them has received little attention. The purpose of this study was to investigate the changes of global functional connectivity density (gFCD) in brains of ESRD patients undergoing hemodialysis using resting-state functional magnetic resonance imaging (re-fMRI). METHODS: re-fMRI data were collected from 30 ESRD patients undergoing hemodialysis (14 men, 38.33±7.44 years old) and 30 matched healthy controls (13 men, 39.17±5.7 years old). Neuropsychological tests including the Montreal Cognitive Assessment (MoCA) and Beck Depression Inventory (BDI) were used to evaluate the neurocognitive and psychiatric conditions of the subjects. Blood biochemistry tests, including hemoglobin level, serum albumin level, blood urea level, serum phosphate, serum calcium, and parathyroid hormone level, and dialysis-related indicators, including blood pressure fluctuations in dialysis, single-pool Kt/V(spKt/V), and ultrafiltration volume of dialysis were obtained from the ESRD patients. A two-sample t-test was used to examine the group differences in gFCD between ESRD patients and healthy controls after controlling for age, gender and education. RESULTS: Compared with healthy controls, ESRD patients exhibited a significantly increased gFCD in the salience network, including the bilateral insula, and dorsal anterior cingulated cortex (dACC), and there was no significant correlation between gFCD and the structural mean grey matter volume in patients for every cluster in the brain regions showing significant different gFCD between the two groups. Furthermore, there were significant negative correlations between the degree of connectivity in the right insula and spKt/V. CONCLUSION: Our findings revealed abnormal intrinsic dysconnectivity pattern of salience network-related regions in ESRD patients from the whole brain network perspective. The negative correlation between the right insula and spKt/V suggested that increased fractional removal of urea may reduce the pathological activity in the insula.


Subject(s)
Cerebral Cortex/physiopathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Gray Matter/physiopathology , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/therapy , Nerve Net/physiopathology , Adult , Female , Functional Neuroimaging , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Renal Dialysis , Urea/metabolism , Urea/toxicity
12.
Environ Pollut ; 246: 408-413, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30577009

ABSTRACT

An influx of chloride ions from road de-icing solutions can result in toxicological effects to organisms in terrestrial and aquatic environments. As such, "eco-friendly" de-icing alternatives are sought to mitigate environmental impacts of de-icing impervious surfaces, while maintaining human safety. While many alternative de-icers are economically impractical for municipal use, the residential commercial market is flooded with de-icing formulations claiming to be "eco-friendly". Given the little regulation and guidance that surrounds eco-labeling, the meaning of "eco-friendly" remains unclear in the context of biological systems. The objective of the current study was to determine the toxicity of three "eco-friendly" de-icing formulations to Chironomus dilutus using 10 d toxicity tests. The toxicity of these three formulations was compared to a traditional formulation composed entirely of chloride salts. Two of the "eco-friendly" de-icers demonstrated LC50s of 6.61 and 6.32 g/L, which were similar in toxicity to the traditional sodium chloride formulation with a LC50 6.29 g/L. The comparable toxicities of these formulations is likely due to the presence of chloride salts in each of the "eco-friendly" de-icers. The third "eco-friendly" formulation, a urea-based de-icer, demonstrated toxicity an order of magnitude higher than that of the traditional formulation with an LC50 of 0.63 g/L. While C. dilutus may not have been the intended endpoint in consideration when marketing these products as "eco-friendly", consideration of how eco-labeling is utilized and the role of environmental scientists in determining the meaning of such claims must be considered to ensure continued and future protection of the environment.


Subject(s)
Chironomidae/drug effects , Product Labeling/standards , Toxicity Tests/methods , Water Pollutants, Chemical/toxicity , Animals , Environmental Monitoring , Sodium Chloride/standards , Sodium Chloride/toxicity , Urea/toxicity
13.
Hemodial Int ; 22(S2): S29-S64, 2018 10.
Article in English | MEDLINE | ID: mdl-30457224

ABSTRACT

Hemodialysis for chronic renal failure was introduced and developed in Seattle, WA, in the 1960s. Using Kiil dialyzers, weekly dialysis time and frequency were established to be about 30 hours on 3 time weekly dialysis. This dialysis time and frequency was associated with 10% yearly mortality in the United States in 1970s. Later in 1970s, newer and more efficient dialyzers were developed and it was felt that dialysis time could be shortened. An additional incentive to shorten dialysis was felt to be lower cost and higher convenience. Additional support for shortening dialysis time was provided by a randomized prospective trial performed by National Cooperative Dialysis Study (NCDS). This study committed a Type II statistical error rejecting the time of dialysis as an important factor in determining the quality of dialysis. This study also provided the basis for the establishment of the Kt/Vurea index as a measure of dialysis adequacy. This index having been established in a sacrosanct randomized controlled trial (RCT), was readily accepted by the HD community, and led to shorter dialysis, and higher mortality in the United States. Kt/Vurea is a poor measure of dialysis quality because it combines three unrelated variables into a single formula. These variables influence the clinical status of the patient independent of each other. It is impossible to compensate short dialysis duration (t) with the increased clearance of urea (K), because the tolerance of ultrafiltration depends on the plasma-refilling rate, which has nothing in common with urea clearance. Later, another RCT (the HEMO study) committed a Type III statistical error by asking the wrong research question, thus not yielding any valuable results. Fortunately, it did not lead to deterioration of dialysis outcomes in the United States. The third RCT in this field ("in-center hemodialysis 6 times per week versus 3 times per week") did not bring forth any valuable results, but at least confirmed what was already known. The fourth such trial ("The effects of frequent nocturnal home hemodialysis") too did not show any positive results primarily due to significant subject recruitment issues leading to inappropriate selection of patients. Comparison of the value of peritoneal dialysis and HD in RCTs could not be completed because of recruitment problems. Randomized controlled trials have therefore failed to yield any meaningful information in the area of dose and or frequency of hemodialysis.


Subject(s)
Renal Dialysis/methods , Sodium/isolation & purification , Urea/metabolism , Blood Pressure , Hemodialysis, Home , Humans , Kidney Failure, Chronic/therapy , Prospective Studies , Randomized Controlled Trials as Topic , Regional Blood Flow , Renal Dialysis/mortality , Renal Dialysis/standards , Time Factors , Urea/toxicity
14.
Curr Environ Health Rep ; 5(2): 205-212, 2018 06.
Article in English | MEDLINE | ID: mdl-29713991

ABSTRACT

PURPOSE OF REVIEW: Urea has been utilized as a reductant in diesel fuels to lower emission of nitrogen oxides, igniting interest in probable human health hazards associated with exposure to exogenous urea. Here, we summarize and update key findings on potential health effects of exogenous urea, including carcinogenicity. RECENT FINDINGS: No definitive target organs for oral exposure were identified; however, results in animal studies suggest that the liver and kidney could be potential target organs of urea toxicity. The available human-subject literature suggests that the impact on lung function is minimal. Based on the literature on exogenous urea, we concluded that there was inadequate information to assess the carcinogenic potential of urea, or perform a quantitative assessment to derive reference values. Given the limited information on exogenous urea, additional research to address gaps for exogenous urea should include long-term cancer bioassays, two-generation reproductive toxicity studies, and mode-of-action investigations.


Subject(s)
Environmental Exposure/adverse effects , Urea/toxicity , Animals , Carcinogens/toxicity , Humans , Neoplasms/chemically induced
15.
Cell Biol Toxicol ; 34(2): 143-162, 2018 04.
Article in English | MEDLINE | ID: mdl-28815372

ABSTRACT

A recent hypothesis suggesting that the pharmacological target TRPV1 (transient receptor potential vanilloid subfamily, member 1) may function as a tumour suppressor, which potentially impacts the development of TRPV1 antagonist therapeutics for a range of conditions. However, little is known about the long-term physiologic effects of TRPV1 blockade in the skin. In vitro and in vivo studies suggested that the potent TRPV1 competitive antagonist AMG-9810 promoted proliferation in N/TERT1 cells (telomerase-immortalised primary human keratinocytes 1) and tumour development in mouse skin that was mediated through EGFR/Akt/mTOR signalling. We attempted to reproduce the reported in vitro and in vivo findings to further explore this hypothesis to understand the underlying mechanism and the risk associated with TRPV1 antagonism in the skin. In vitro proliferation studies using multiple methods and topical application with AMG-9810 and structurally similar TRPV1 antagonists such as SB-705498 and PAC-14028 were performed. Although we confirmed expression of TRPV1 in primary human epidermal keratinocytes (HEKn) and spontaneously immortalised human keratinocytes (HaCaT), we were unable to demonstrate cell proliferation in either cell type or any clear evidence of increased expression of proteins in the EGFR/Akt/mTOR signalling pathway with these molecules. We were also unable to demonstrate skin tumour promotion or underlying molecular mechanisms involved in the EGFR/Akt/mTOR signalling pathway in a single-dose and two-stage carcinogenesis mouse study treated with TRPV1 antagonists. In conclusion, our data suggest that inhibiting the pharmacological function of TRPV1 in skin by specific antagonists has not been considered to be indicative of skin tumour development.


Subject(s)
Cell Proliferation/drug effects , Keratinocytes/drug effects , Skin Neoplasms/metabolism , TRPV Cation Channels/antagonists & inhibitors , Acrylamides/toxicity , Animals , Anthracenes/toxicity , Bridged Bicyclo Compounds, Heterocyclic/toxicity , Capsaicin/analogs & derivatives , Capsaicin/toxicity , Cell Line , Cell Survival/drug effects , Cocarcinogenesis , Female , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Hairless , Piperidines/toxicity , Primary Cell Culture , Pyridines/toxicity , Pyrrolidines/toxicity , Risk , Skin Neoplasms/pathology , TRPV Cation Channels/genetics , Urea/analogs & derivatives , Urea/toxicity
16.
Clin Oral Investig ; 22(4): 1771-1781, 2018 May.
Article in English | MEDLINE | ID: mdl-29196947

ABSTRACT

OBJECTIVES: The aim of this study was to evaluate the cytotoxicity and the influence of bleaching agents on immunologically cell surface antigens of murine macrophages in vitro. MATERIALS AND METHODS: RAW 264.7 cells were exposed to bleaching gel extracts (40% hydrogen peroxide or 20% carbamide peroxide) and different H2O2 concentrations after 1 and 24-h exposure periods and 1-h exposure and 23-h recovery. Tests were performed with and without N-acetyl cysteine (NAC) and buthionine sulfoximine (BSO). Cell viability was determined by MTT assay. The expression of surface markers CD14, CD40, and CD54 with and without LPS stimulation was detected by flow cytometry, while the production of TNF-α was measured by ELISA. Statistical analysis was performed using the Mann-Whitney U test (α = 0.05). RESULTS: Extracts of bleaching agents were cytotoxic for cells after a 1-h exposure; cells could not recover after 24 h. This effect can be mitigated by the antioxidant NAC and increased by BSO, an inhibitor of glutathione (GSH) synthesis. LPS stimulated expression of all surface markers and TNF-α production. Exposure to bleaching agent extracts and H2O2 leads to a reduction of TNF-α, CD14, and CD40 expression, while the expression of CD54 was upregulated at non-cytotoxic concentrations. Whereas NAC reduced this effect, it was increased in the presence of BSO. CONCLUSIONS: Extracts of bleaching agents were irreversibly cytotoxic to macrophages after a 1-h exposure. Only the expression of CD54 was upregulated. The reactions are mediated by the non-enzymatic antioxidant GSH. CLINICAL RELEVANCE: The addition of an antioxidant can downregulate unfavorable effects of dental bleaching.


Subject(s)
Antigens, Surface/drug effects , Bleaching Agents/toxicity , Hydrogen Peroxide/toxicity , Peroxides/toxicity , Urea/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Antigens, Surface/immunology , Buthionine Sulfoximine/pharmacology , Carbamide Peroxide , Cell Survival/drug effects , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Macrophages/drug effects , Mice , Tooth Bleaching , Tumor Necrosis Factor-alpha/immunology , Urea/toxicity
17.
World J Gastroenterol ; 23(44): 7930-7938, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29209134

ABSTRACT

The underlying pathophysiology of liver dysfunction in urea cycle disorders (UCDs) is still largely elusive. There is some evidence that the accumulation of urea cycle (UC) intermediates are toxic for hepatocyte mitochondria. It is possible that liver injury is directly caused by the toxicity of ammonia. The rarity of UCDs, the lack of checking of iron level in these patients, superficial knowledge of UC and an underestimation of the metabolic role of fumaric acid, are the main reasons that are responsible for the incomprehension of the mechanism of liver injury in patients suffering from UCDs. Owing to our routine clinical practice to screen for iron overload in severely ill neonates, with the focus on the newborns suffering from acute liver failure, we report a case of citrullinemia with neonatal liver failure and high blood parameters of iron overload. We hypothesize that the key is in the decreased-deficient fumaric acid production in the course of UC in UCDs that causes several sequentially intertwined metabolic disturbances with final result of liver iron overload. The presented hypothesis could be easily tested by examining the patients suffering from UCDs, for liver iron overload. This could be easily performed in countries with a high population and comprehensive national register for inborn errors of metabolism. CONCLUSION: Providing the hypothesis is correct, neonatal liver damage in patients having UCD can be prevented by the supplementation of pregnant women with fumaric or succinic acid, prepared in the form of iron supplementation pills. After birth, liver damage in patients having UCDs can be prevented by supplementation of these patients with zinc fumarate or zinc succinylate, as well.


Subject(s)
Citrullinemia/metabolism , Hepatocytes/metabolism , Hyperammonemia/metabolism , Liver Diseases/etiology , Liver/physiopathology , Ammonia/blood , Ammonia/toxicity , Central Nervous System/physiopathology , Citrullinemia/blood , Citrullinemia/diagnosis , Citrullinemia/therapy , Enterobacter/isolation & purification , Fatal Outcome , Humans , Hyperammonemia/blood , Hyperammonemia/diagnosis , Hyperammonemia/therapy , Infant, Newborn , Lethargy/etiology , Lethargy/metabolism , Lethargy/physiopathology , Lethargy/therapy , Liver/cytology , Liver/metabolism , Liver Diseases/metabolism , Liver Diseases/physiopathology , Liver Diseases/therapy , Male , Sepsis/microbiology , Sepsis/therapy , Urea/blood , Urea/metabolism , Urea/toxicity
18.
Int J Toxicol ; 36(5): 395-402, 2017.
Article in English | MEDLINE | ID: mdl-28820005

ABSTRACT

Repeated developmental exposure to the organophosphate (OP) insecticide chlorpyrifos (CPF) inhibits brain fatty acid amide hydrolase (FAAH) activity at low levels, whereas at higher levels, it inhibits brain monoacylglycerol lipase (MAGL) activity. FAAH and MAGL hydrolyze the endocannabinoids anandamide (AEA) and 2-arachidonylglycerol (2-AG), respectively. Peripherally, AEA and 2-AG have physiological roles in the regulation of lipid metabolism and immune function, and altering the normal levels of these lipid mediators can negatively affect these processes. Exposure to CPF alters brain endocannabinoid hydrolysis activity, but it is unclear whether low-level exposure alters this activity in peripheral tissues important in metabolic and immune function. Therefore, rat pups were exposed orally from day 10 to 16 to 0.5, 0.75, or 1.0 mg/kg CPF or 0.02 mg/kg PF-04457845 (a specific FAAH inhibitor). At 12 hours postexposure, FAAH, MAGL, and cholinesterase (ChE) activities were determined. All treatments inhibited FAAH activity in brain, spleen, and liver. CPF inhibited ChE activity in spleen and liver (all dosages) and in brain (highest dosage only). CPF inhibited total 2-AG hydrolysis and MAGL-specific activity in brain and spleen (high dosage only). In liver, total 2-AG hydrolysis was inhibited by all treatments and could be attributed to inhibition of non-MAGL-mediated 2-AG hydrolysis, indicating involvement of other enzymes. MAGL-specific activity in liver was inhibited only by the high CPF dosage, whereas PF-04457845 slightly increased this activity. Overall, exposure to low levels of CPF and to PF-04457845 can alter endocannabinoid metabolism in peripheral tissues, thus potentially affecting physiological processes.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Arachidonic Acids/metabolism , Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , Endocannabinoids/metabolism , Glycerides/metabolism , Insecticides/toxicity , Polyunsaturated Alkamides/metabolism , Pyridazines/toxicity , Urea/analogs & derivatives , Animals , Brain/drug effects , Brain/metabolism , Cholinesterases/metabolism , Female , Liver/drug effects , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley , Spleen/drug effects , Spleen/metabolism , Urea/toxicity
19.
J Photochem Photobiol B ; 173: 545-550, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28692926

ABSTRACT

BACKGROUND: Cosmetic preservatives are used to protect cosmetic formulations and improve its shelf-life. However, these substances may exert phototoxic effects when used under sunlight. OBJECTIVE: To assess safety, efficacy and putative phototoxic effects of a sunscreen formulation SPF 30 and its excipients. MATERIALS/METHODS: Irradiation was performed with solar simulated light (SSL) and the sunscreen from the School of Pharmacy/UFRJ/Brazil. We used albino hairless mice in different groups (control (G1), only irradiated (G2), sunscreen plus irradiation (G3) and vehicle plus irradiation (G4) for morphological assessment and immunefluorescence detection to OKL38. In vitro analyses were with a Saccharomyces cerevisiae (SC) strain plus SSL in the presence of methylparaben, propylparaben, imidazolidinyl urea, aminomethyl propanol and their association. RESULTS: G3 and G4 displayed photosensitization leading to thickening of the epidermis and increased dermal cellularity. G4 displayed strong OKL38 labeling when compared with other groups. Aminomethyl propanol, methylparaben and propylparaben are endowed with phototoxic activity against SC. Propylparaben displayed the highest phototoxic effect, followed by excipients association. CONCLUSIONS: The sunscreen's vehicle is endowed with phototoxic activity. Propylparaben was the most phototoxic agent, increasing the overall phototoxicity of excipient association, pointing to a critical concern regarding vehicle associations intended to cosmetic purposes.


Subject(s)
Skin/drug effects , Sunscreening Agents/pharmacology , Animals , Cosmetics , Drug Compounding , Mice , Mice, Hairless , Microscopy, Fluorescence , Parabens/toxicity , Propanolamines/toxicity , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/radiation effects , Skin/pathology , Skin/radiation effects , Sunlight , Urea/analogs & derivatives , Urea/toxicity
20.
Ecotoxicol Environ Saf ; 144: 88-96, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28601521

ABSTRACT

Urea is a widespread organic pollutant, which can be a nitrogen source, playing different roles in the growth of submerged macrophytes depending on concentrations, while high cadmium (Cd) concentrations are often toxic to macrophytes. In order to evaluate the combined effect of urea and Cd on a submerged macrophyte, Cabomba caroliniana, the morphological and physiological responses of C. caroliniana in the presence of urea and Cd were studied. The results showed that high concentrations of urea (400mgL-1) and Cd (500µmolL-1) had negative effects on C. caroliniana. There were strong visible symptoms of toxicity after 4 days of exposure under Cd-alone, 400mgL-1 urea, and Cd+400mgL-1 urea treatments. In addition, 400mgL-1 urea and Cd had adverse effects on C. caroliniana's pigment system. Significant losses in chlorophyll fluorescence and photosynthetic rates, as well as Rubisco activity were also observed under Cd-alone, 400mgL-1 urea, and Cd+400mgL-1 urea treatments. 400mgL-1 urea markedly enhanced Cd toxicity in C. caroliniana, reflected by a sharp decrease in photosynthetic activity and more visible toxicity symptoms. The results of thiobarbituric acid reactive substances (TBARS) pointed to extreme oxidative stress in C. caroliniana induced under Cd or 400mgL-1 urea exposure. Exogenous ascorbate (AsA) protected C. caroliniana from adverse damage in 400mgL-1 urea, which further corroborated the oxidative stress claim under 400mgL-1 urea. However, results also demonstrated that lower urea concentration (10mgL-1) alleviated Cd-induced phytotoxicity by stimulating chlorophyll synthesis and photosynthetic activity, as well as activating the activity of catalase (CAT) and glutathione-S-transferase (GST), which may explain the alleviating effect of urea on C. caroliniana under Cd stress.


Subject(s)
Antioxidants/metabolism , Cadmium/toxicity , Magnoliopsida/drug effects , Photosynthesis/drug effects , Soil Pollutants/toxicity , Urea/toxicity , Ascorbic Acid/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Magnoliopsida/metabolism , Oxidative Stress/drug effects , Thiobarbituric Acid Reactive Substances/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...