Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Adv Exp Med Biol ; 959: 175-185, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28755195

RESUMO

The discovery that a natural product leptospermone had herbicidal activity formed the starting point for chemical synthesis to find more activity and selectivity. A series of molecules called triketones were found to possess good activity and 2-(2-nitro-4-trifluoromethylbenzoyl)-cyclohexane-1,3-dione (NTBC) was selected for toxicology testing. NTBC fed at low doses to rats and dogs caused keratopathy, which on cessation of the diet recovered. Mice, rabbits and monkeys fed NTBC did not show this response. Research discovered that NTBC caused tyrosinaemia which was due to inhibition of the enzyme 4-hydroxyphenylpyruvate dioxygenase in both mammals and plants thereby finding a novel target for killing plants. NTBC was also used sucessfully as a drug to treat a rare inborn error of metabolism, tyrosinaemia type I, in collaboration with Professor's Sven Lindstedt and Elisabeth Holme. Understanding the mechanism of toxicity of NTBC led to novel herbicide discovery and saved the lives of children with acute tyrosinaemia type I.


Assuntos
Cicloexanonas/farmacologia , Cicloexanonas/uso terapêutico , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Fígado/efeitos dos fármacos , Fígado/fisiologia , Nitrobenzoatos/farmacologia , Nitrobenzoatos/uso terapêutico , Tirosinemias/tratamento farmacológico , Animais , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Humanos , Tirosina/metabolismo , Tirosinemias/metabolismo
2.
Toxicol Appl Pharmacol ; 266(3): 345-55, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23220449

RESUMO

Parkinson disease (PD) is a debilitating neurodegenerative motor disorder, with its motor symptoms largely attributable to loss of dopaminergic neurons in the substantia nigra. The causes of PD remain poorly understood, although environmental toxicants may play etiologic roles. Solvents are widespread neurotoxicants present in the workplace and ambient environment. Case reports of parkinsonism, including PD, have been associated with exposures to various solvents, most notably trichloroethylene (TCE). Animal toxicology studies have been conducted on various organic solvents, with some, including TCE, demonstrating potential for inducing nigral system damage. However, a confirmed animal model of solvent-induced PD has not been developed. Numerous epidemiologic studies have investigated potential links between solvents and PD, yielding mostly null or weak associations. An exception is a recent study of twins indicating possible etiologic relations with TCE and other chlorinated solvents, although findings were based on small numbers, and dose-response gradients were not observed. At present, there is no consistent evidence from either the toxicological or epidemiologic perspective that any specific solvent or class of solvents is a cause of PD. Future toxicological research that addresses mechanisms of nigral damage from TCE and its metabolites, with exposure routes and doses relevant to human exposures, is recommended. Improvements in epidemiologic research, especially with regard to quantitative characterization of long-term exposures to specific solvents, are needed to advance scientific knowledge on this topic.


Assuntos
Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/epidemiologia , Tricloroetileno/intoxicação , Tricloroetileno/toxicidade , Animais , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Exposição Ambiental/efeitos adversos , Humanos , Doença de Parkinson Secundária/metabolismo , Doença de Parkinson Secundária/patologia , Solventes/intoxicação , Solventes/toxicidade , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , Substância Negra/patologia , Estudos em Gêmeos como Assunto
3.
Arch Toxicol ; 86(4): 571-89, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22124623

RESUMO

Ochratoxin A (OTA) is a widely studied compound due to its role in renal toxicity and carcinogenicity. However, there is still no consensus on the exact mechanisms of toxicity or carcinogenicity. In the current study, we analysed the effect of OTA on three human renal proximal tubular models (human primary, RPTEC/TERT1 and HK-2 cells) and two rat renal proximal tubular models (rat primary and NRK-52E cells). Global transcriptomics analysis at two exposure times was performed to generate a set of 756 OTA sensitive genes. This gene set was then compared in more detail across all models and additionally to a rat in vivo renal cortex model. The results demonstrate a well-conserved response across all models. OTA resulted in deregulation of a number of pathways including cytoskeleton, nucleosome regulation, translation, transcription, ubiquitination and cell cycle pathways. Interestingly, the oxidative stress activated Nrf2 pathway was not enriched. These results point to an epigenetic action of OTA, perhaps initiated by actin binding as the actin remodelling gene, advillin was the highest up-regulated in all models. The largest model differences were observed between the human and the rat in vitro models. However, since the human in vitro models were more similar to the rat in vivo model, it is more likely that these differences are model-specific rather than species-specific per se. This study demonstrates the usefulness of in vitro cell culture models combined with transcriptomic analysis for the investigation of mechanisms of toxicity and carcinogenicity. In addition, these results provide further evidence supporting a non-genotoxic mechanism of OTA-induced carcinogenicity.


Assuntos
Carcinógenos/toxicidade , DNA/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Micotoxinas/toxicidade , Ocratoxinas/toxicidade , Animais , Linhagem Celular , DNA/genética , Epigênese Genética/efeitos dos fármacos , Perfilação da Expressão Gênica , Humanos , Túbulos Renais Proximais/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Especificidade da Espécie , Testes de Toxicidade
4.
Arch Toxicol ; 86(11): 1741-51, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22760423

RESUMO

Potassium bromate (KBrO(3)) is an oxidising agent that has been widely used in the food and cosmetic industries. It has shown to be both a nephrotoxin and a renal carcinogen in in vivo and in vitro models. Here, we investigated the effects of KBrO(3) in the human and rat proximal tubular cell lines RPTEC/TERT1 and NRK-52E. A genome-wide transcriptomic screen was carried out from cells exposed to a sub-lethal concentration of KBrO(3) for 6, 24 and 72 h. Pathway analysis identified "glutathione metabolism", "Nrf2-mediated oxidative stress" and "tight junction (TJ) signalling" as the most enriched pathways. TJ signalling was less impacted in the rat model, and further studies revealed low transepithelial electrical resistance (TEER) and an absence of several TJ proteins in NRK-52E cells. In RPTEC/TERT1 cells, KBrO(3) exposure caused a decrease in TEER and resulted in altered expression of several TJ proteins. N-Acetylcysteine co-incubation prevented these effects. These results demonstrate that oxidative stress has, in conjunction with the activation of the cytoprotective Nrf2 pathway, a dramatic effect on the expression of tight junction proteins. The further understanding of the cross-talk between these two pathways could have major implications for epithelial repair, carcinogenesis and metastasis.


Assuntos
Bromatos/toxicidade , Túbulos Renais Proximais/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Proteínas de Junções Íntimas/metabolismo , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Túbulos Renais Proximais/citologia , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/genética , Ratos , Junções Íntimas/metabolismo , Testes de Toxicidade
5.
Metabolites ; 12(10)2022 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-36295804

RESUMO

This review briefly discusses the discovery of the mode of action of the triketone herbicide, 2-(2-nitro-4-trifluormethylbenzoyl)-1,3-cyclohexanedione and its use as a drug Nitisinone for the treatment of inborn errors of tyrosine metabolism. Nitisinone is a potent reversible tight-binding inhibitor of the enzyme 4-hydroxyphenylpyruvate dioxygenase, involved in the catabolism of the amino acid tyrosine. Nitisinone is used to treat the rare disease hereditary tyrosinaemia type 1 where the last enzyme in the breakdown of tyrosine, fumarylacetoacetase is deficient. Nitisinone is also used to treat patients with alkaptonuria where the enzyme homogentisic acid oxidase is deficient. Articles in this issue discuss metabolites of tyrosine catabolism in healthy patients and those with alkaptonuria.

6.
Toxicol Res (Camb) ; 9(5): 591-600, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33178419

RESUMO

Thiocarbamates are a major class of herbicides that were used extensively in the agricultural industry. Toxicological evaluation showed molinate caused reproductive impairment in male rats, whilst others produced behavioural effects at high doses. Rats dosed with molinate either as a single large oral dose of 100 mg/kg or as multiple doses of 50 mg/kg for 7 days produced inhibition of brain acetylcholinesterase (AChE). Molinate and other thiocarbamate herbicides undergo metabolism to form sulphoxides that can carbamoylate thiol's such as glutathione and proteins. We have chemically synthesised the sulphoxide and sulphone metabolites of six thiocarbamate herbicides and examined their ability to inhibit rat brain and human red cell AChE in vitro. Parent thiocarbamates were inactive, whilst the sulphoxides produced inhibition with IC50's in the 1-10 mM range, the sulphone metabolites were the most active with IC50's for molinate, pebulate, EPTC and vernolate in the µM range. Inhibition was both time- and dose-dependent with biomolecular rate constants for the inhibition of the human red cell enzyme of 0.3 × 102 and 2.0 × 102 M-1 min-1 for molinate sulphoxide and sulphone, respectively. No recovery of enzyme activity, with either enzyme, was seen following dilution of the inhibitor to a concentration that does not inhibit the enzyme for up to 24 h at 25°C at pH 7.4. The metabolites of these thiocarbamate herbicides are rather poor inhibitors of AChE when compared to the organophosphorus ester, paraoxon or the monomethylcarbamate, eserine. Unlike eserine the inhibition produced by the thiocarbamates is irreversible.

7.
Toxicol Res (Camb) ; 9(2): 117-126, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32440343

RESUMO

The industrial solvent trichloroethylene (TCE) and its two major metabolites trichloroethanol (TCE-OH) and trichloroacetic acid (TCA) cause formic aciduria in male F344 rats. Prior treatment of male F344 rats with 1-aminobenzotriazole a cytochrome P450 inhibitor, followed by TCE (16mk/kg, po), completely prevented formic aciduria, but had no effect on formic acid excretion produced by TCA (8 or 16 mg/kg, po), suggesting TCA may be the proximate metabolite producing this response. Dow and Green reported an increase in the concentration of 5-methyltetrahydrofolate (5-MTHF) in the plasma of rats treated with TCE-OH, suggesting a block in the cycling of 5-MTHF to tetrahydrofolate (THF). This pathway is under the control of the vitamin B12-dependent methionine salvage pathway. We therefore treated rats with three daily doses of methylcobalamin (CH3Cbl) or hydroxocobalamin (OHCbl), a cofactor for methionine synthase, or L-methionine, followed by TCE (16 mg/kg) to determine if they could alleviate the formic aciduria. These pretreatments only partially reduced the excretion of formic acid in the urine. Although prior treatment with S-adenosyl-L-methionine had no effect on formic acid excretion. Consistent with these findings, the activity of methionine synthase in the liver of TCE-treated rats was not inhibited. Transcriptomic analysis of the liver-identified nine differential expressed genes, of note, was downregulation of Lmbrd1 involved in the conversion of vitamin B12 into CH3Cbl, a cofactor for methionine synthase. Our findings indicate that the formic aciduria produced by TCE-OH and TCA may be the result of a block in the recycling of 5-MTHF to THF, the effect on the methionine salvage pathway being a secondary response following acute exposure.

8.
Toxicology ; 245(3): 163-6, 2008 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-18272278

RESUMO

The search for biomarkers and their evaluation by scientists and clinicians is of paramount importance if we are going to improve health care. In this paper we discuss the history of the discovery of biomarkers for renal and cardiac injury. We also summarize the use of biomarkers in preclinical evaluation in experimental animals and in patients to help diagnose or monitor a disease, predict outcome or to evaluate a therapeutic intervention. The need for validated biomarkers of tissue injury or disease that must be easy to analyse rapidly, preferably at the bedside, is essential if clinical decision making is to be optimally affected by their use.


Assuntos
Biomarcadores , Toxicologia/história , Toxicologia/tendências , Animais , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , História do Século XX , História do Século XXI , Humanos , Praguicidas/toxicidade
9.
Toxicology ; 229(1-2): 79-90, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17097791

RESUMO

Renal cell carcinoma is the most common neoplasm occurring in the kidney and is largely resistant to current chemotherapy. Understanding the mechanisms involved in renal carcinoma cell death may lead to novel and more effective therapies. In Cak(i)-1 renal cancer cells, using phosphatidylserine externalization as a marker of apoptosis, the anti-cancer drugs 5-fluorouracil (5-FU), and its pro-drugs, doxifluridine (Dox) and floxuridine (Flox) proceeds via a caspase-dependent mechanism. In contrast, phosphatidylserine externalization produced by staurosporine in the renal cancer cell lines Cak(i)-1 and A-498 proceeds via a caspase-independent mechanism. That is, the pan caspase inhibitor N-benzyloxycabonyl-Val-Ala-Asp-fluoromethylketone (ZVAD) did not ameliorate annexin V binding, cell shrinkage or changes in nuclear morphology. Subsequent experiments were conducted to determine mediators of phosphatidylserine externalization, using annexin V binding, when caspases were inhibited. Prior treatment of A-498 cells with cathepsin B (CA74 methyl ester), cathespsin D (pepstatin A) or calpain inhibitors (calpeptin, E64d) in the presence or absence of ZVAD did not ameliorate annexin V binding. The endonuclease inhibitor aurintricarboxylic acid (ATA), phospholipase A(2) inhibitor bromoenol lactone (BEL), protein synthesis inhibitor cycloheximide (CH) and chloride channel blockers niflumic acid (NFA) and 5-nitro-2-(3-phenylpropylamino) benzoic acid (NPPB) all had no effect on staurosporine-induced annexin V binding in A-498 cells either in the presence or absence of ZVAD. We also modulated sphingomyelin and the de novo pathways of ceramide synthesis and found no amelioration of staurosporine-induced annexin V binding in A-498 cells either in the presence or absence of ZVAD. These results indicate that 5-FU, Dox and Flox induce externalization of phosphatidylserine during apoptosis in Cak(i)-1 renal cancer cells primarily through a caspase-dependent mechanism and that externalization of phosphatidylserine during apoptosis produced by staurosporine in the renal cancer cell line A-498 is independent of many of the common signaling pathways known to be involved in this process.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Fosfatidilserinas/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Compostos de Anilina/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ácido Benzoico/farmacologia , Compostos de Benzilideno/farmacologia , Calpaína/antagonistas & inibidores , Catepsina B/antagonistas & inibidores , Catepsina D/antagonistas & inibidores , Linhagem Celular Tumoral , Tamanho Celular/efeitos dos fármacos , Ceramidas/antagonistas & inibidores , Cisplatino/farmacologia , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Fumonisinas/farmacologia , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Naftalenos/administração & dosagem , Naftalenos/farmacologia , Ácido Niflúmico/farmacologia , Nitrobenzoatos/farmacologia , Pironas/administração & dosagem , Pironas/farmacologia , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Estaurosporina/farmacologia , Triterpenos/farmacologia
10.
Toxicology ; 230(2-3): 234-43, 2007 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-17161896

RESUMO

The industrial solvent trichloroethylene (TCE) and its major metabolites have been shown to cause formic aciduria in male rats. We have examined whether chloral hydrate (CH) and trichloroacetic acid (TCA), known metabolites of TCE, produce an increase in formic acid in vitro in cultures of rat hepatocytes or human renal proximal tubule cells (HRPTC). The metabolism and cytotoxicity of CH was also examined to establish that the cells were metabolically active and not compromised by toxicity. Rat hepatocytes and HRPTC were cultured in serum-free medium and then treated with 0.3-3mM CH for 3 days or 0.03-3mM CH for 10 days, respectively and formic acid production, metabolism to trichloroethanol (TCE-OH) and TCA and cytotoxicity determined. No increase in formic acid production in rat hepatocytes or HRPTC exposed to CH was observed over and above that due to chemical degradation, neither was formic acid production observed in rat hepatocytes exposed to TCA. HRPTC metabolized CH to TCE-OH and TCA with a 12-fold greater capacity to form TCE-OH versus TCA. Rat hepatocytes exhibited a 1.6-fold and three-fold greater capacity than HRPTC to form TCE-OH and TCA, respectively. CH and TCA were not cytotoxic to rat hepatocytes at concentrations up to 3mM/day for 3 days. With HRPTC, one sample showed no cytotoxicity to CH at concentrations up to 3mM/day for 10 days, while in another cytotoxicity was seen at 1mM/day for 3 days. In summary, increased formic acid production was not observed in rat hepatocytes or HRPTC exposed to TCE metabolites, suggesting that the in vivo response cannot be modelled in vitro. CH was toxic to HRPTC at millimolar concentrations/day over 10 days, while glutathione derived metabolites of TCE were toxic at micromolar concentrations/day over 10 days [Lock, E.A., Reed, C.J., 2006. Trichloroethylene: mechanisms of renal toxicity and renal cancer and relevance to risk assessment. Toxicol. Sci. 19, 313-331] supporting the view that glutathione derived metabolites are likely to be responsible for nephrotoxicity.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Hidrato de Cloral/toxicidade , Formiatos/metabolismo , Hepatócitos/efeitos dos fármacos , Nefropatias/induzido quimicamente , Túbulos Renais Proximais/efeitos dos fármacos , Ácido Tricloroacético/toxicidade , Adolescente , Adulto , Animais , Cromatografia Gasosa , Etilenocloroidrina/análogos & derivados , Etilenocloroidrina/metabolismo , Hepatócitos/metabolismo , Humanos , Nefropatias/metabolismo , Túbulos Renais Proximais/metabolismo , Hepatopatias/metabolismo , Espectroscopia de Ressonância Magnética , Masculino , Ratos
11.
Toxicology ; 378: 76-85, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28063905

RESUMO

1, 1, 2-Trichloroethylene (TCE) is of environmental concern, due to evaporation while handling, chemical processing and leakage from chemical waste sites, leading to its contamination of ground water and air. For several decades there has been issues about possible long term health effects of TCE but recently the International Agency for Research on Cancer (IARC) and the US Environmental Protection Agency classified TCE as a human carcinogen. Links having been established between occupational exposures and kidney cancer and possible links to non-Hodgkin lymphoma and liver cancer, but there is more still more to learn. In male rats, TCE produces a small increase in the incidence of renal tubule tumours but not in female rats or mice of either sex. However, chronic renal injury was seen in these bioassays in both sexes of rats and mice. The mechanism of kidney injury from TCE is thought to be due to reductive metabolism forming a cysteine conjugate that is converted to a reactive metabolite via the enzyme cysteine conjugate ß-lyase. However, TCE also produces a marked and sustained formic aciduria in male rats and it has been suggested that long term exposure to formic acid could lead to renal tubule injury and regeneration. In this study we have determined if TCE produces formic aciduria in male mice following a single and repeat dosing. Male C57 Bl/6OlaHsd mice were dosed with 1000mg/kg by ip injection and urine collected overnight 24, 48, 72 and 96h after dosing. Formic acid was present in urine 24h after dosing, peaked around 48h at 8mg formic acid excreted/mouse, and remained constant over the next 24h and was not back to normal 96h after dosing. This was associated with a marked acidification of the urine. Plasma creatinine and renal pathology was normal. Plasma kinetics of formic acid showed it was readily cleared with an initial half-life of 2.42h followed by a slower rate with a half-life of 239h. Male mice were then dosed twice/week at 1000mg/kg TCE for 56days, as anticipated there was a marked and sustained formic aciduria over the duration of the study. This was associated with acidification of the urine, mild diuresis and a marked fall in urinary ammonia. Six biomarkers of renal injury KIM-1, NGAL, NAG, Cystatin-c, Albumin and IL-18 were measured in urine over time and they all showed a small increase at the later time points indicative of early markers of renal injury. However, there was no histological evidence of renal damage or renal tubule cell proliferation after 8 weeks' exposure to TCE. The concentration of formic acid in plasma at the end of the study was 1.05±0.61mM compared to control, 0.39±0.17mM. In the liver, formic acid was present at a concentration of 1mM in both control and treated mice while in the kidney it was higher at 2mM in both treated and controls. We also report that trichloroacetic acid (TCA) a metabolite of TCE also causes formic aciduria, at doses likely to arise in vivo after 1000mg/kg TCE namely 16 and 32mg/kg. Urinary formic acid peaked 24h after dosing at 4mg formic acid excreted/mouse. Thus, as in male and female rats (Yaqoob et al., 2013) male mice show a marked formic aciduria following TCE which after 8 weeks' exposure did not produce renal injury, but the small rise in renal biomarkers suggest renal damage may occur following longer exposure. Thus, TCE-induced formic aciduria may be a contributor factor in the chronic renal injury seen in male and female rats and mice.


Assuntos
Formiatos/urina , Solventes/toxicidade , Tricloroetileno/toxicidade , Amônia/urina , Animais , Formiatos/sangue , Rim/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL
12.
Toxicol Sci ; 91(2): 313-31, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16421178

RESUMO

1,1,2-Trichloroethylene (TCE) is an important solvent that is widespread in the environment. We have reviewed carcinogenicity data from seven bioassays with regard to renal injury and renal tumors. We report a consistent but low incidence of renal tubule carcinoma in male rats. Epidemiology studies on workers exposed to TCE (and other chlorinated solvents) indicate a weak association between high-level exposure and renal cancer. There appears to be a threshold below which no renal injury or carcinogenicity is expected to arise. TCE is not acutely nephrotoxic to rats or mice, but subchronic exposure to rats produces a small increase in urinary markers of renal injury. Following chronic exposure, pathological changes (toxic nephrosis and a high incidence of cytomegaly and karyomegaly) were observed. The basis for the chronic renal injury probably involves bioactivation of TCE. Based on the classification by E. A. Lock and G. C. Hard (2004, Crit. Rev. Toxicol. 34, 211-299) of chemicals that induce renal tubule tumors, we found no clear evidence to place TCE in category 1 or 2 (chemicals that directly or indirectly interact with renal DNA), category 4 (direct cytotoxicity and sustained tubule cell regeneration), category 5 (indirect cytotoxicity and sustained tubule cell regeneration associated with alpha2u-globulin accumulation), or category 6 (exacerbation of spontaneous chronic progressive nephropathy). TCE is best placed in category 3, chemicals that undergo conjugation with GSH and subsequent enzymatic activation to a reactive species. The implication for human risk assessment is that TCE should not automatically be judged by linear default methods; benchmark methodology could be used.


Assuntos
Neoplasias Renais/induzido quimicamente , Solventes/toxicidade , Tricloroetileno/toxicidade , Adenoma/induzido quimicamente , Animais , Carcinoma/induzido quimicamente , Humanos , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/patologia , Medição de Risco , Solventes/farmacocinética , Tricloroetileno/farmacocinética
13.
Genom Data ; 5: 254-6, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26484264

RESUMO

Aristolochic acids (AAs) are the active components of herbal drugs derived from Aristolochia species that have been used for medicinal purposes since antiquity. However, AAs have recently been discovered to be highly nephrotoxic and induced urothelial cancer in humans and malignant tumors in the kidney and urinary tract of rodents. In this study, we exposed rat renal proximal tubule cells in vitro to a sub-cytotoxic level of AAs at three different time points (6 h, 24 h and 72 h). We then analyzed the gene expression profile after the compound exposure. Functional analysis with Ingenuity Pathways Analysis and DAVID tools revealed that at the late time point (72 h) there are many significantly altered genes involved in cancer-related pathways such as p53 signaling. MIAMI-compliant microarray data are deposited in the NCBI GEO database under accession number GSE68687 and can be found at: http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE68687.

14.
Free Radic Biol Med ; 34(5): 607-15, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12614849

RESUMO

Despite extensive interest in the rodent nasal cavity as a target organ for toxicity, there is very limited information regarding nasal defenses against oxidative stress and xenobiotic-derived oxidants. Using immunohistochemistry, we have examined the distribution of Cu,Zn and Mn superoxide dismutase (SOD), catalase, glutathione (GSH) peroxidase, and DT-diaphorase in rat nasal tissues. In addition, we have determined the concentrations of ascorbate and alpha-tocopherol and the activities of SOD (combined Cu,Zn and Mn forms), catalase, GSH peroxidase, GSH reductase, and DT-diaphorase in nasal respiratory epithelium (RE), olfactory epithelium (OE), and in lung. Immunohistochemistry demonstrated that all four enzymes were similarly distributed, with the greatest staining intensity in dorsal-medial regions of the nasal cavity. In respiratory epithelium, ciliated columnar cells and subepithelial glands stained positively, while in olfactory tissue the enzymes were detected in the sustentacular cells and Bowman's glands. With the exception of SOD, enzyme activities were higher in RE than OE, while concentrations of ascorbate and alpha-tocopherol were higher in OE than RE. With the exception of catalase, nasal activities were either higher than or comparable to those of the lung. Thus, the rat nasal cavity appears to be well protected against oxidative damage.


Assuntos
Antioxidantes/metabolismo , Catalase/metabolismo , Glutationa Peroxidase/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , Cavidade Nasal/enzimologia , Superóxido Dismutase/metabolismo , Animais , Ácido Ascórbico/metabolismo , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Técnicas Imunoenzimáticas , Masculino , Cavidade Nasal/anatomia & histologia , Mucosa Olfatória/enzimologia , Mucosa Olfatória/metabolismo , Oxirredução , Estresse Oxidativo , Ratos , Ratos Wistar , alfa-Tocoferol/metabolismo
15.
Toxicol Sci ; 73(1): 195-206, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12657746

RESUMO

4-Aminophenol (4-AP), D-serine, and cisplatin are established rodent nephrotoxins that damage proximal tubules within the renal cortex. Using high throughput 2D gel proteomics to profile protein changes in the plasma of compound-treated animals, we identified several markers of kidney toxicity. Male F344 and Alpk rats were treated with increasing doses of 4-AP, D-serine, or cisplatin, and plasma samples were collected over time. Control groups received saline or nontoxic isomers, L-serine, and transplatin. Plasma proteins that displayed dose- and temporal-dependent regulation in each study were further characterized by mass spectrometry to elucidate the protein identity. Several isoforms of the rat-specific T-kininogen protein were identified in each study. T-kininogen was elevated in the plasma of 4-AP-, D-serine-, and cisplatin-treated animals at early time points, returning to baseline levels 3 weeks after treatment. The protein was not elevated in the plasma of control animals or those treated with nontoxic compounds. We propose that T-kininogen may be required to counteract apoptosis in proximal tubular cells in order to minimize tissue damage following a toxic insult. In addition, T-kininogen may be required to stimulate localized inflammation to aid tissue repair. We also identified several isoforms of the inter-alpha inhibitor H4P heavy chain in the 4-AP and D-serine studies. In each case, the protein expression levels in the blood samples paralleled the extent of kidney toxicity, highlighting the correlation between protein alterations and clinical chemistry endpoints. A further set of proteins correlating with kidney damage was found to be a component of the complement cascade and other blood clotting factors, indicating a contribution of the immune system to the observed toxicity. These observations underscore the value of proteomics in identifying new biomarkers and in the elucidation of mechanisms of toxicity.


Assuntos
Síndrome de Fanconi/induzido quimicamente , Síndrome de Fanconi/patologia , Proteoma/efeitos dos fármacos , 4-Aminopiridina/toxicidade , Animais , Antineoplásicos/toxicidade , Proteínas Sanguíneas/metabolismo , Cisplatino/toxicidade , Creatinina/sangue , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Processamento de Imagem Assistida por Computador , Testes de Função Renal , Cininogênios/biossíntese , Masculino , Espectrometria de Massas , Peso Molecular , Bloqueadores dos Canais de Potássio/toxicidade , Proteoma/química , Ratos , Ratos Endogâmicos F344 , Serina/toxicidade , alfa-Macroglobulinas/metabolismo
16.
Toxicol Sci ; 74(1): 215-27, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12730612

RESUMO

S-(1,2-Dichlorovinyl)-L-cysteine (DCVC), a model nephrotoxicant in mice, causes acute tubular necrosis and death at high doses. Our earlier studies revealed that renal tissue repair was critical for survival in mice with DCVC nephrotoxicity. The objective of this study was to investigate if increasing renal tissue repair could protect mice from the lethal outcome of DCVC. Male Swiss Webster (SW) mice were administered a low dose of DCVC (15 mg/kg, ip) 72 h before injection of a normally lethal dose of DCVC (75 mg/kg, ip); this resulted in 100% protection against the lethal effect of DCVC. Because DCVC caused approximately two fold decrease in cytosolic and mitochondrial beta-lyase activity, the possibility that DCVC protection may be caused by decreased bioactivation was examined. Mercuric chloride (HgCl2, 6 mg/kg), a nephrotoxicant with no effect on beta-lyase activity, was administered 96 h before a lethal dose of DCVC. This also resulted in 100% protection from the lethal effect of DCVC. In both studies total glutathione was unchanged at any time after the lethal dose of DCVC was administered, obviating the role of glutathione in protection. In both cases the augmented and sustained tissue repair induced by priming dose and documented by 3H-thymidine pulse labeling and immunocytochemistry for proliferating cell nuclear antigen resulted in 100% survival in spite of the extensive renal injury. These findings suggest that stimulation of renal tubular repair by the priming dose, through augmented cell division, and the resistance of new cells to mechanisms of progression of injury, underlies auto- and heteroprotection against DCVC. The molecular mechanisms may have potential application in pharmacotherapeutic intervention for treatment of acute renal failure.


Assuntos
Cisteína/análogos & derivados , Cisteína/toxicidade , Necrose Tubular Aguda/induzido quimicamente , Necrose Tubular Aguda/patologia , Animais , Nitrogênio da Ureia Sanguínea , Cisteína/urina , Citosol/metabolismo , DNA/biossíntese , Enzimas/sangue , Glutationa/metabolismo , Rim/enzimologia , Rim/metabolismo , Rim/patologia , Necrose Tubular Aguda/metabolismo , Masculino , Cloreto de Mercúrio/toxicidade , Camundongos , Oxirredução , Antígeno Nuclear de Célula em Proliferação/metabolismo , Fase S/efeitos dos fármacos , Sobrevida , Timidina/metabolismo , Urodinâmica/efeitos dos fármacos
17.
Toxicology ; 175(1-3): 143-52, 2002 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-12049843

RESUMO

The aim of this study was to examine the effect of haloalkene S-cysteine conjugates on cytosolic free Ca(2+) levels in renal epithelial cells using digital imaging fluorescence microscopy (DIFM). S-(1,2,3,4,4-pentachloro-1,3,-butadienyl)-L-cysteine (PCBC) and S-(1,2-dichlorovinyl)-L-cysteine (DCVC) were both cytotoxic to LLC-PK(1) cells in culture. Prior treatment of the cells with aminooxyacetic acid (AOAA), an inhibitor of the enzyme cysteine conjugate beta-lyase, afforded complete protection against the toxicity at concentrations of PCBC up to 100 microM and DCVC up to 500 microM. The cytotoxicity produced by PCBC (100 microM) was time dependent with no loss of lactate dehydrogenase (LDH) into the medium being observed until 4 h after exposure, while removal of calcium from the medium prevented the toxicity. Addition of PCBC (100 microM) to LLC-PK(1) cells produced a small progressive increase in intracellular calcium ([Ca(2+)](i)) from 72+/-6 to 126+/-11 nM following 10 min of exposure. At this time there was a marked cellular heterogeneity in the calcium response with some cells showing marked increases in [Ca(2+)](i), while others cycled between low and high values and some just maintained basal levels. Exposure to PCBC (100 microM) for 1 h produced a more marked increase in [Ca(2+)](I), 469+/-46 nM, with all cells responding. The elevation in [Ca(2+)](i) was concentration-related with increases seen at concentrations of 5 microM PCBC and above. The increase in [Ca(2+)](i) produced by PCBC (100 microM) was prevented by treatment with AOAA, and markedly reduced by a nominally calcium free medium or the addition of the calcium chelator EGTA. DCVC (500 microM) also markedly elevated [Ca(2+)](i) following exposure for 1 h, this was also prevented by AOAA and a nominal calcium free medium. These findings indicate that elevation in [Ca(2+)](i) produced by PCBC in renal epithelial cells, is an early event in the cascade of signalling changes leading to renal cell death. The major source of calcium appears to be from increased influx although a small component is released from intracellular stores which my trigger a stress protein response.


Assuntos
Butadienos/toxicidade , Cálcio/metabolismo , Cisteína/toxicidade , Fura-2/análogos & derivados , Homocisteína/análogos & derivados , Homocisteína/toxicidade , Tacrolimo/análogos & derivados , Ácido Amino-Oxiacético/farmacologia , Animais , Cisteína/análogos & derivados , Citosol/metabolismo , Ácido Egtázico/farmacologia , Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes/química , Fura-2/química , Processamento de Imagem Assistida por Computador , L-Lactato Desidrogenase/metabolismo , Células LLC-PK1 , Microscopia de Fluorescência , Suínos , Tacrolimo/metabolismo
18.
Neurotoxicology ; 25(6): 1031-40, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15474620

RESUMO

Previous studies have shown that L-2-chloropropionic acid (L-CPA) produces necrosis to cerebellar granule cells with some associated Purkinje cell damage in the rat. We have re-evaluated the neuropathology using the original sections and fresh sections from archived brain material from rats treated with L-CPA at different ages, times after dosing and the following prior treatment with the N-methyl-D-aspartate (NMDA) receptor antagonist, MK-801. In addition we have determined the lobular distribution of cerebellar granule cell necrosis produced by L-CPA. Using Fluoro-Jade staining to detect degenerating neurons, we have identified three new brain regions that show neuronal cell necrosis as a result of exposure to L-CPA, these are the medial habenular nucleus, pontine gray and inferior olivary nucleus. The neuronal cell degeneration was confirmed in conventional haematoxylin and eosin stained sections and in some cases by glial fibrillary acidic protein staining for reactive gliosis. The neuronal cell necrosis at these new sites was both time and dose dependent; young 22-day-old rats, which are refractory to L-CPA-induced cerebellar granule cell necrosis, did however show some neuronal cell degeneration in the medial habenular, pontine gray and inferior olivary nuclei. Treatment of rats with MK-801 30 min prior to L-CPA, afforded complete protection against the neuronal cell injury in the medial habenular, pontine gray and inferior olivary nuclei, similar to that previously reported for the cerebellum, supporting an excitotoxic mechanism of neuronal cell death. In the cerebellum the lobular distribution of the granule cell loss was not uniform, more severe granule cell loss occurring in lobules 1-4 and 9a + b. This localization exactly mirrors that seen previously in the cerebellum of rats given L-CPA and examined by magnetic resonance imaging (MRI). The basis for the neuronal cell loss in the medial habenular nucleus, pontine gray and inferior olivary nucleus, in addition to the major site in the cerebellum, and the sensitivity of particular cerebellar lobes is not currently understood. Anatomical connections between the sites of injury and their likely neurotransmitter use are discussed.


Assuntos
Encéfalo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Propionatos/toxicidade , Fatores Etários , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Hidrocarbonetos Clorados , Masculino , Neurônios/metabolismo , Neurônios/patologia , Ratos
19.
Toxicol Lett ; 140-141: 317-22, 2003 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-12676480

RESUMO

The US Environmental Protection Agency (EPA) in 1999 issued draft guidelines on carcinogen risk assessment, which included the use mode of action information in the risk assessment process. We have used the five stages of induction of toxicity as described by Aldridge to illustrate in the case of two drugs, tamoxifen and NTBC, how mode of action information played a key role in assessing the risk of cancer and target organ toxicity, respectively.


Assuntos
Carcinógenos/toxicidade , Cicloexanonas/toxicidade , Guias como Assunto , Nitrobenzoatos/toxicidade , Tamoxifeno/toxicidade , Animais , Testes de Carcinogenicidade , Cicloexanonas/farmacocinética , Meia-Vida , Humanos , Nitrobenzoatos/farmacocinética , Medição de Risco/métodos , Tamoxifeno/farmacocinética , Estados Unidos , United States Environmental Protection Agency
20.
Toxicology ; 323: 70-7, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-24923549

RESUMO

Trichloroethylene (TCE) is widely used as a cleaning and decreasing agent and has been shown to cause liver tumours in rodents and a small incidence of renal tubule tumours in male rats. The basis for the renal tubule injury is believed to be related to metabolism of TCE via glutathione conjugation to yield the cysteine conjugate that can be activated by the enzyme cysteine conjugate ß-lyase in the kidney. More recently TCE and its major metabolite trichloroethanol (TCE-OH) have been shown to cause formic aciduria which can cause renal injury after chronic exposure in rats. In this study we have compared the renal toxicity of TCE and TCE-OH in rats to try and ascertain whether the glutathione pathway or formic aciduria can account for the toxicity. Male rats were given TCE (500mg/kg/day) or TCE-OH at (100mg/kg/day) for 12 weeks and the extent of renal injury measured at several time points using biomarkers of nephrotoxicity and prior to termination assessing renal tubule cell proliferation. The extent of formic aciduria was also determined at several time points, while renal pathology and plasma urea and creatinine were determined at the end of the study. TCE produced a very mild increase in biomarkers of renal injury, total protein, and glucose over the first two weeks of exposure and increased Kim-1 and NAG in urine after 1 and 5 weeks exposure, while TCE-OH did not produce a consistent increase in these biomarkers in urine. However, both chemicals produced a marked and sustained increase in the excretion of formic acid in urine to a very similar extent. The activity of methionine synthase in the liver of TCE and TCE-OH treated rats was inhibited by about 50% indicative of a block in folate synthesis. Both renal pathology and renal tubule cell proliferation were reduced after TCE and TCE-OH treatment compared to controls. Our findings do not clearly identify the pathway which is responsible for the renal toxicity of TCE but do provide some support for metabolism via glutathione conjugation.


Assuntos
Etilenocloroidrina/análogos & derivados , Formiatos/urina , Rim/efeitos dos fármacos , Solventes/toxicidade , Tricloroetileno/toxicidade , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Acetilglucosaminidase/urina , Animais , Moléculas de Adesão Celular/urina , Etilenocloroidrina/toxicidade , Rim/patologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Masculino , Ácido Metilmalônico/urina , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Testes de Toxicidade Subcrônica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA