Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
EMBO J ; 43(13): 2789-2812, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38811853

RESUMO

It has remained unknown how cells reduce cystine taken up from the extracellular space, which is a required step for further utilization of cysteine in key processes such as protein or glutathione synthesis. Here, we show that the thioredoxin-related protein of 14 kDa (TRP14, encoded by TXNDC17) is the rate-limiting enzyme for intracellular cystine reduction. When TRP14 is genetically knocked out, cysteine synthesis through the transsulfuration pathway becomes the major source of cysteine in human cells, and knockout of both pathways becomes lethal in C. elegans subjected to proteotoxic stress. TRP14 can also reduce cysteinyl moieties on proteins, rescuing their activities as here shown with cysteinylated peroxiredoxin 2. Txndc17 knockout mice were, surprisingly, protected in an acute pancreatitis model, concomitant with activation of Nrf2-driven antioxidant pathways and upregulation of transsulfuration. We conclude that TRP14 is the evolutionarily conserved enzyme principally responsible for intracellular cystine reduction in C. elegans, mice, and humans.


Assuntos
Caenorhabditis elegans , Cisteína , Cistina , Camundongos Knockout , Oxirredução , Proteoma , Tiorredoxinas , Animais , Humanos , Camundongos , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Cisteína/metabolismo , Cistina/metabolismo , Peroxirredoxinas/metabolismo , Peroxirredoxinas/genética , Proteoma/metabolismo , Tiorredoxinas/metabolismo , Tiorredoxinas/genética
2.
Artigo em Inglês | MEDLINE | ID: mdl-31454703

RESUMO

This research aimed to assess the influence of dietary addition of rutin on inflammation, apoptosis and antioxidative responses in muscle of silver catfish (Rhamdia quelen) challenged with Aeromonas hydrophila (A. hydrophila). Fish were split into four groups as follows: control, 0.15% rutin, A. hydrophila, 0.15% rutin + A. hydrophila. After 2 weeks of feeding with standard or rutin diets, fish were challenged or not with A. hydrophila for 1 week. Rutin-added diet abrogates A. hydrophila induced-hemorrhage and inflammatory infiltration. It decreases A. hydrophila induced-apoptosis through decreasing the ratio of Bax to Bcl-2 and increasing phospho-Akt to Akt ratio. It diminishes the A. hydrophila induced-rise in nitric oxide and superoxide anion levels and reestablishes superoxide dismutase activity as well. Although such diet is unable to recover the levels of reduced glutathione (GSH), cysteine and glutamate cysteine ligase, which are depleted as a result of A. hydrophila infection, it diminishes the oxidized glutathione (GSSG) content, thus decreasing GSSG to GSH ratio. It increases the levels of cysteine residues of proteins and diminishes those of thiol-protein mixed disulfides, which were changed after A. hydrophila challenge. Finally, it reduces A. hydrophila induced-lipid peroxidation, markedly elevates ascorbic acid and thus reestablishes total antioxidant capacity, whose levels were decreased after A. hydrophila challenge. In conclusion, the dietary addition of rutin at 0.15% impairs A. hydrophila-induced inflammatory response, inhibits A. hydrophila-induced apoptosis and promotes cell survival. It also reduces the A. hydrophila-induced oxidative stress and stimulates the antioxidative responses in muscle of A. hydrophila-infected silver catfish.


Assuntos
Peixes-Gato/imunologia , Doenças dos Peixes/metabolismo , Infecções por Bactérias Gram-Negativas , Músculos/metabolismo , Rutina/farmacologia , Aeromonas hydrophila , Ração Animal , Animais , Antioxidantes/farmacologia , Apoptose , Suplementos Nutricionais , Infecções por Bactérias Gram-Negativas/metabolismo , Infecções por Bactérias Gram-Negativas/veterinária , Estresse Oxidativo , Substâncias Protetoras/farmacologia
3.
J Pathol ; 247(1): 48-59, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30221360

RESUMO

Obesity is associated with local and systemic complications in acute pancreatitis. PPARγ coactivator 1α (PGC-1α) is a transcriptional coactivator and master regulator of mitochondrial biogenesis that exhibits dysregulation in obese subjects. Our aims were: (1) to study PGC-1α levels in pancreas from lean or obese rats and mice with acute pancreatitis; and (2) to determine the role of PGC-1α in the inflammatory response during acute pancreatitis elucidating the signaling pathways regulated by PGC-1α. Lean and obese Zucker rats and lean and obese C57BL6 mice were used first; subsequently, wild-type and PGC-1α knockout (KO) mice with cerulein-induced pancreatitis were used to assess the inflammatory response and expression of target genes. Ppargc1a mRNA and protein levels were markedly downregulated in pancreas of obese rats and mice versus lean animals. PGC-1α protein levels increased in pancreas of lean mice with acute pancreatitis, but not in obese mice with pancreatitis. Interleukin-6 (Il6) mRNA levels were dramatically upregulated in pancreas of PGC-1α KO mice after cerulein-induced pancreatitis in comparison with wild-type mice with pancreatitis. Edema and the inflammatory infiltrate were more intense in pancreas from PGC-1α KO mice than in wild-type mice. The lack of PGC-1α markedly enhanced nuclear translocation of phospho-p65 and recruitment of p65 to Il6 promoter. PGC-1α bound phospho-p65 in pancreas during pancreatitis in wild-type mice. Glutathione depletion in cerulein-induced pancreatitis was more severe in KO mice than in wild-type mice. PGC-1α KO mice with pancreatitis, but not wild-type mice, exhibited increased myeloperoxidase activity in the lungs, together with alveolar wall thickening and collapse, which were abrogated by blockade of the IL-6 receptor glycoprotein 130 with LMT-28. In conclusion, obese rodents exhibit PGC-1α deficiency in the pancreas. PGC-1α acts as selective repressor of nuclear factor-κB (NF-κB) towards IL-6 in pancreas. PGC-1α deficiency markedly enhanced NF-κB-mediated upregulation of Il6 in pancreas in pancreatitis, leading to a severe inflammatory response. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Interleucina-6/metabolismo , NF-kappa B/metabolismo , Obesidade/metabolismo , Pâncreas/metabolismo , Pancreatite/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/deficiência , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Animais , Ceruletídeo , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/complicações , Obesidade/genética , Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Fosforilação , Ratos Zucker , Transdução de Sinais , Ácido Taurocólico , Fator de Transcrição RelA/metabolismo , Regulação para Cima
4.
Redox Biol ; 16: 276-284, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29567616

RESUMO

p38α is a redox sensitive MAPK activated by pro-inflammatory cytokines and environmental, genotoxic and endoplasmic reticulum stresses. The aim of this work was to assess whether p38α controls the antioxidant defense in the liver, and if so, to elucidate the mechanism(s) involved and the age-related changes. For this purpose, we used liver-specific p38α-deficient mice at two different ages: young-mice (4 months-old) and old-mice (24 months-old). The liver of young p38α knock-out mice exhibited a decrease in GSH levels and an increase in GSSG/GSH ratio and malondialdehyde levels. However, old mice deficient in p38α had higher hepatic GSH levels and lower GSSG/GSH ratio than young p38α knock-out mice. Liver-specific p38α deficiency triggered a dramatic down-regulation of the mRNAs of the key antioxidant enzymes glutamate cysteine ligase, superoxide dismutase 1, superoxide dismutase 2, and catalase in young mice, which seems mediated by the lack of p65 recruitment to their promoters. Nrf-2 nuclear levels did not change significantly in the liver of young mice upon p38α deficiency, but nuclear levels of phospho-p65 and PGC-1α decreased in these mice. p38α-dependent activation of NF-κB seems to occur through classical IκB Kinase and via ribosomal S6 kinase1 and AKT in young mice. However, unexpectedly the long-term deficiency in p38α triggers a compensatory up-regulation of antioxidant enzymes via NF-κB activation and recruitment of p65 to their promoters. In conclusion, p38α MAPK maintains the expression of antioxidant genes in liver of young animals via NF-κΒ under basal conditions, whereas its long-term deficiency triggers compensatory up-regulation of antioxidant enzymes through NF-κΒ.


Assuntos
Envelhecimento/genética , Antioxidantes/metabolismo , Fígado/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Animais , Catalase/genética , Estresse do Retículo Endoplasmático/genética , Regulação Enzimológica da Expressão Gênica , Glutamato-Cisteína Ligase/genética , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , NF-kappa B/genética , Superóxido Dismutase/genética , Superóxido Dismutase-1/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Redox Biol ; 11: 701-707, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28187322

RESUMO

No-caloric sweeteners, such as aspartame, are widely used in various food and beverages to prevent the increasing rates of obesity and diabetes mellitus, acting as tools in helping control caloric intake. Aspartame is metabolized to phenylalanine, aspartic acid, and methanol. Our aim was to study the effect of chronic administration of aspartame on glutathione redox status and on the trans-sulphuration pathway in mouse liver. Mice were divided into three groups: control; treated daily with aspartame for 90 days; and treated with aspartame plus N-acetylcysteine (NAC). Chronic administration of aspartame increased plasma alanine aminotransferase (ALT) and aspartate aminotransferase activities and caused liver injury as well as marked decreased hepatic levels of reduced glutathione (GSH), oxidized glutathione (GSSG), γ-glutamylcysteine ​​(γ-GC), and most metabolites of the trans-sulphuration pathway, such as cysteine, S-adenosylmethionine (SAM), and S-adenosylhomocysteine ​​(SAH). Aspartame also triggered a decrease in mRNA and protein levels of the catalytic subunit of glutamate cysteine ligase (GCLc) and cystathionine γ-lyase, and in protein levels of methionine adenosyltransferase 1A and 2A. N-acetylcysteine prevented the aspartame-induced liver injury and the increase in plasma ALT activity as well as the decrease in GSH, γ-GC, cysteine, SAM and SAH levels and GCLc protein levels. In conclusion, chronic administration of aspartame caused marked hepatic GSH depletion, which should be ascribed to GCLc down-regulation and decreased cysteine levels. Aspartame triggered blockade of the trans-sulphuration pathway at two steps, cystathionine γ-lyase and methionine adenosyltransferases. NAC restored glutathione levels as well as the impairment of the trans-sulphuration pathway.


Assuntos
Aspartame/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Glutationa/metabolismo , Edulcorantes/efeitos adversos , Acetilcisteína/administração & dosagem , Animais , Aspartame/administração & dosagem , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/patologia , Cistationina gama-Liase/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato-Cisteína Ligase/genética , Humanos , Fígado/metabolismo , Fígado/patologia , Metionina Adenosiltransferase/genética , Camundongos , Edulcorantes/administração & dosagem
6.
Free Radic Biol Med ; 104: 75-103, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28062361

RESUMO

Redox signaling regulates physiological self-renewal, proliferation, migration and differentiation in gastrointestinal epithelium by modulating Wnt/ß-catenin and Notch signaling pathways mainly through NADPH oxidases (NOXs). In the intestine, intracellular and extracellular thiol redox status modulates the proliferative potential of epithelial cells. Furthermore, commensal bacteria contribute to intestine epithelial homeostasis through NOX1- and dual oxidase 2-derived reactive oxygen species (ROS). The loss of redox homeostasis is involved in the pathogenesis and development of a wide diversity of gastrointestinal disorders, such as Barrett's esophagus, esophageal adenocarcinoma, peptic ulcer, gastric cancer, ischemic intestinal injury, celiac disease, inflammatory bowel disease and colorectal cancer. The overproduction of superoxide anion together with inactivation of superoxide dismutase are involved in the pathogenesis of Barrett's esophagus and its transformation to adenocarcinoma. In Helicobacter pylori-induced peptic ulcer, oxidative stress derived from the leukocyte infiltrate and NOX1 aggravates mucosal damage, especially in HspB+ strains that downregulate Nrf2. In celiac disease, oxidative stress mediates most of the cytotoxic effects induced by gluten peptides and increases transglutaminase levels, whereas nitrosative stress contributes to the impairment of tight junctions. Progression of inflammatory bowel disease relies on the balance between pro-inflammatory redox-sensitive pathways, such as NLRP3 inflammasome and NF-κB, and the adaptive up-regulation of Mn superoxide dismutase and glutathione peroxidase 2. In colorectal cancer, redox signaling exhibits two Janus faces: On the one hand, NOX1 up-regulation and derived hydrogen peroxide enhance Wnt/ß-catenin and Notch proliferating pathways; on the other hand, ROS may disrupt tumor progression through different pro-apoptotic mechanisms. In conclusion, redox signaling plays a critical role in the physiology and pathophysiology of gastrointestinal tract.


Assuntos
Gastroenteropatias/metabolismo , Trato Gastrointestinal/metabolismo , NADPH Oxidases/metabolismo , Estresse Oxidativo , Proliferação de Células/genética , Gastroenteropatias/patologia , Trato Gastrointestinal/patologia , Humanos , Mucosa Intestinal/metabolismo , Intestinos/patologia , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Compostos de Sulfidrila/metabolismo , Via de Sinalização Wnt/genética
7.
J Immunol ; 197(10): 4137-4150, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27798150

RESUMO

Chromatin remodeling seems to regulate the patterns of proinflammatory genes. Our aim was to provide new insights into the epigenetic mechanisms that control transcriptional activation of early- and late-response genes in initiation and development of severe acute pancreatitis as a model of acute inflammation. Chromatin changes were studied by chromatin immunoprecipitation analysis, nucleosome positioning, and determination of histone modifications in promoters of proinflammatory genes in vivo in the course of taurocholate-induced necrotizing pancreatitis in rats and in vitro in rat pancreatic AR42J acinar cells stimulated with taurocholate or TNF-α. Here we show that the upregulation of early and late inflammatory genes rely on histone acetylation associated with recruitment of histone acetyltransferase CBP. Chromatin remodeling of early genes during the inflammatory response in vivo is characterized by a rapid and transient increase in H3K14ac, H3K27ac, and H4K5ac as well as by recruitment of chromatin-remodeling complex containing BRG-1. Chromatin remodeling in late genes is characterized by a late and marked increase in histone methylation, particularly in H3K4. JNK and p38 MAPK drive the recruitment of transcription factors and the subsequent upregulation of early and late inflammatory genes, which is associated with nuclear translocation of the early gene Egr-1 In conclusion, specific and strictly ordered epigenetic markers such as histone acetylation and methylation, as well as recruitment of BRG-1-containing remodeling complex are associated with the upregulation of both early and late proinflammatory genes in acute pancreatitis. Our findings highlight the importance of epigenetic regulatory mechanisms in the control of the inflammatory cascade.


Assuntos
Montagem e Desmontagem da Cromatina , Epigênese Genética , Regulação da Expressão Gênica , Pancreatite Necrosante Aguda/genética , Pancreatite Necrosante Aguda/imunologia , Ativação Transcricional , Acetilação , Células Acinares/efeitos dos fármacos , Animais , Imunoprecipitação da Cromatina , DNA Helicases/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Inflamação/genética , Metilação , Proteínas Nucleares/genética , Pancreatite Necrosante Aguda/induzido quimicamente , Pancreatite Necrosante Aguda/metabolismo , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Ratos , Ácido Taurocólico/farmacologia , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/farmacologia
8.
Inflamm Allergy Drug Targets ; 13(5): 312-22, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25510330

RESUMO

Under physiological conditions, the balance between ROS production and removal properly maintains the intracellular redox-sensitive signaling as well as the appropriate status of protein thiols and disulfides. However, inflammation among other factors can modify this balance causing a rapid increase in intracellular ROS levels and hence thiol oxidation, eventually leading to oxidative stress. In the case of acute pancreatitis, both redox signaling and oxidative stress seem to contribute to the progression of the severe form of the disease. In this review we will focus on the reversible oxidation of protein cysteines during the course of acute pancreatitis. We describe disulfide stress in an acute inflammatory process, which is characterized by thiol oxidation in proteins, particularly protein cysteinylation, without significant changes in the glutathione redox status.


Assuntos
Cisteína/metabolismo , Dissulfetos/metabolismo , Pancreatite/metabolismo , Doença Aguda , Animais , Humanos , Terapia de Alvo Molecular , Oxirredução , Estresse Oxidativo , Pancreatite/tratamento farmacológico , Transdução de Sinais
9.
Neurochem Res ; 39(9): 1681-90, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24970110

RESUMO

Long-term intake of aspartame at the acceptable daily dose causes oxidative stress in rodent brain mainly due to the dysregulation of glutathione (GSH) homeostasis. N-Acetylcysteine provides the cysteine that is required for the production of GSH, being effective in treating disorders associated with oxidative stress. We investigated the effects of N-acetylcysteine treatment (150 mg kg(-1), i.p.) on oxidative stress biomarkers in rat brain after chronic aspartame administration by gavage (40 mg kg(-1)). N-Acetylcysteine led to a reduction in the thiobarbituric acid reactive substances, lipid hydroperoxides, and carbonyl protein levels, which were increased due to aspartame administration. N-Acetylcysteine also resulted in an elevation of superoxide dismutase, glutathione peroxidase, glutathione reductase activities, as well as non-protein thiols, and total reactive antioxidant potential levels, which were decreased after aspartame exposure. However, N-acetylcysteine was unable to reduce serum glucose levels, which were increased as a result of aspartame administration. Furthermore, catalase and glutathione S-transferase, whose activities were reduced due to aspartame treatment, remained decreased even after N-acetylcysteine exposure. In conclusion, N-acetylcysteine treatment may exert a protective effect against the oxidative damage in the brain, which was caused by the long-term consumption of the acceptable daily dose of aspartame by rats.


Assuntos
Acetilcisteína/farmacologia , Aspartame/administração & dosagem , Encéfalo/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Glicemia/análise , Peso Corporal , Encéfalo/metabolismo , Masculino , Ratos , Ratos Wistar
10.
Neurosci Lett ; 569: 163-8, 2014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24704379

RESUMO

Since N-acetylcysteine (NAC) is a donor of cysteine, we studied the relationship between NAC and concentration of oxidized and reduced glutathione (GSH/GSSG ratio), and glutathione peroxidase (GPx) and glutathione-S-transferase (GST) activities in the lumbosacral spinal cord of rats with chronic constriction injury (CCI) of the sciatic nerve that received NAC (150mg/kg/day, i.p.) or 0.9% saline solution for 3 or 10 days. Hydrogen peroxide (H2O2) and nitric-oxide (NO) metabolites were also measured. Von Frey hair and hot-plate tests showed hyperalgesia at day 1 in CCI rats. Hyperalgesia persisted at all other times in saline-treated CCI rats, but returned to pre-injury values in NAC-treated CCI rats after 3 postoperative days. GST activity and the GSH/GSSG ratio increased in saline-treated CCI rats, while the NAC treatment increased GST and GPx activities at day 10, with no significant change in the GSH/GSSG ratio. NAC treatment did not affect H2O2 levels, but it reduced NO metabolites in CCI rats 3 days after the surgery. Thus, the anti-hyperalgesic effect of NAC appears not to involve its action as a cysteine precursor for GSH synthesis, but involves a decrease in NO.


Assuntos
Acetilcisteína/farmacologia , Analgésicos/farmacologia , Glutationa Peroxidase/metabolismo , Glutationa Transferase/metabolismo , Glutationa/metabolismo , Neuralgia/metabolismo , Medula Espinal/efeitos dos fármacos , Animais , Constrição Patológica , Temperatura Alta , Peróxido de Hidrogênio/metabolismo , Hiperalgesia/fisiopatologia , Região Lombossacral , Masculino , Neuralgia/fisiopatologia , Óxido Nítrico/metabolismo , Estimulação Física , Ratos Wistar , Nervo Isquiático/lesões , Medula Espinal/metabolismo , Tato
11.
Free Radic Biol Med ; 75 Suppl 1: S30, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26461335

RESUMO

Long-term intake of aspartame at the acceptable daily ingestion dose causes oxidative stress in the rat kidney through the dysregulation of glutathione homeostasis. N-acetylcysteine (NAC) provides the cystein required for the production of GSH, being effective in treating disorders associated with oxidative stress. The aim of this research was to investigate the effects of NAC on the aspartame-induced oxidative stress in the rat kidney. The animals received aspartame by gavage for six weeks (40mg/kg). From the 5th week, NAC (1mmol/kg, via intraperitoneal) was injected for two weeks. Then, they were anaesthetized for blood sample and euthanized for the kidney collection. The blood was centrifuged at 1800g for 15min and the serum was separated for creatinine measurement. The tissue was homogenized in 1.15% KCl buffer and centrifuged at 700g for 10min at 4°C. The supernatant fraction obtained was used to the measurements of oxidative stress biomarkers. The creatinine levels were enhanced in the serum of aspartame-treated rats. NAC caused a reduction in the thiobarbituric acid reactive substances, lipid hydroperoxides, carbonyl protein and hydrogen peroxide levels, which were increased in the kidney of aspartame-treated animals. Additionally, NAC caused an elevation in the glutathione peroxidase and glutathione reductase activities, total glutathione, ascorbic acid, and total reactive antioxidant potential levels, which were decreased in the kidney of aspartame-treated rats. In conclusion, NAC may be useful for the protection of the rat kidney against aspartame-induced oxidative stress.

12.
Arch Environ Contam Toxicol ; 60(2): 309-18, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20711719

RESUMO

The effects of sublethal cadmium concentrations on oxidative stress parameters were evaluated in Rhamdia quelen. The fish were exposed to 0.44, 236, and 414 µg l⁻¹ cadmium for 7 and 14 days, followed by the same time periods for recovery. Enzymes, such as catalase (CAT), superoxide dismutase (SOD), and glutathione S-transferase (GST), and indicators of oxidative stress, such as thiobarbituric acid-reactive species (TBARS) and protein carbonyl, were verified in fish tissues. In addition, the accumulation of cadmium was evaluated in these tissues. Our results indicate that CAT and GST levels decreased in gills after exposure periods associated with increased TBARS levels. In hepatic tissue, CAT, GST, TBARS, and protein carbonyl levels increased after 7 days of exposure, whereas SOD activity decreased after exposure for 14 days. In the kidney, TBARS levels decreased after exposure for 7 days and increased after exposure for 14 days. During the recovery periods, some variations persisted in gills, liver, and kidney. Cadmium accumulation was most significant in liver, followed by kidney and gills. These results indicate that cadmium concentrations studied invoke a stress response in silver catfish.


Assuntos
Cádmio/metabolismo , Cádmio/toxicidade , Peixes-Gato/metabolismo , Poluentes Químicos da Água/metabolismo , Poluentes Químicos da Água/toxicidade , Animais , Catalase/efeitos dos fármacos , Proteínas de Peixes/metabolismo , Brânquias/efeitos dos fármacos , Brânquias/metabolismo , Glutationa Transferase/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Carbonilação Proteica , Superóxido Dismutase/efeitos dos fármacos , Substâncias Reativas com Ácido Tiobarbitúrico/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA